You are on page 1of 302

3 BACTERIAL

DISEASES
D. J. Taylor, Editor
Actinobacillus pleuropneumoniae
26 D. J. Taylor

Actinobacillus pleuropneumoniae is the etiologic agent of ETIOLOGY


pleuropneumonia in pigs. The first observations of this
disease were made by Pattison et al. (1957), Matthews The etiologic agent A. pleuropneumoniae (formerly
and Pattison (1961), and Olander (1963). Shope (1964) Haemophilus pleuropneumoniae, H. parahaemolyticus),
later described an acute outbreak of a similar infection neotype strains Shope 4074 and ATCC 27088, is a small,
on a farm in Argentina. The name Haemophilus pleuro- gram-negative, encapsulated rod with typical coccobacil-
pneumoniae was given to the causal organism by Shope et lary morphology. The organism does not grow on blood
al. (1964) and White et al. (1964). The name was con- agar unless it is supplemented with NAD and shows
firmed by Kilian et al. (1978). The designation H. para- satellitism around colonies of staphylococci. Staphylo-
haemolyticus given to the California strains (Olander coccal streaks are normally required for primary isola-
1963; Biberstein et al. 1963) and the Swiss isolates (Nico- tion on this medium. The organism forms colonies 0.5–1
let and König 1966; Nicolet 1968) is considered a syn- mm after 24 hours’ incubation on blood agar in the pres-
onym. ence of staphylococcal colonies and is beta-hemolytic,
H. pleuropneumoniae was later transferred to the particularly when sheep red blood cells are used. Biovar
genus Actinobacillus and designated A. pleuropneumoniae 1 is NAD dependent, and biovar 2 is NAD independent
once DNA homology studies had demonstrated the close but requires the presence of specific pyridine nucleotides
relationship of H. pleuropneumoniae to A. lignieresii (Pohl or pyridine nucleotide precursors for its NAD biosynthe-
et al. 1983). The Pasteurella haemolytica–like organism sis (Niven and Levesque 1988). A. pleuropneumoniae pro-
described by Bertschinger and Seifert (1978) as a duces an increased zone of hemolysis within the zone of
causative agent of necrotizing pleuropneumonia is con- partial lysis surrounding a beta-toxinogenic Staphylococ-
sidered to be a nicotinamide adenine dinucleotide cus aureus (the CAMP phenomenon) (Nicolet 1970; Kil-
(NAD)-independent biovar of A. pleuropneumoniae (Pohl ian 1976) and this CAMP phenomenon has been shown
et al. 1983) and is known as A. pleuropneumoniae biovar to be related to the possession of the three cytolysins
2. Recent studies of isolates of V factor–dependent ApxI, ApxII, and ApxIII (Frey et al. 1994; Jansen et al.
strains from cases of disease (the “minor taxon”) have 1995). Additional detailed morphologic and biochemical
shown that they belong to three separate species: A. mi- characteristics may be found in the original reports
nor, A. porcinus, and A. indolicus (Moeller et al. 1996). (Shope 1964; Nicolet 1968; Kilian et al. 1978).
Pleuropneumonia is one of the important bacterial A number of related bacteria occur in pigs. A. pleu-
diseases of the respiratory tract of the pig and occurs in ropneumoniae can be distinguished from H. parasuis by
most pig-keeping countries. Its importance derives from the presence of hemolysis around staphylococcal streaks
the fact that it can cause pneumonia that results in death, on blood agar; from the NAD-dependent former
clinical disease that may become chronic, or subclinical Haemophilus sp. “minor group,” Haemophilus sp. “Taxon
disease in successive batches of pigs and causes losses C” (Rapp et al. 1985), now assigned to the actinobacilli as
from death, reduced production, and increased costs of A. minor, A. porcinus, and A. indolicus (Moeller et al.
medication or vaccination. One of the problems of this 1996) and Haemophilus sp. “Urease-negative” in the case
disease for the clinician is that animals may be treated of biovar 1; and from actinobacilli commonly isolated
with antimicrobials effective against the organism, but from pigs in the case of biovar 2 (Rapp et al. 1985). A. su-
they may not respond because of the severity of the ini- is is described in Chapter 44.
tial lesions and their nature. Both the organism and the A. pleuropneumoniae biovar 1 has been divided into 12
disease have been studied extensively and in consider- serovars. Serovars 1–5 were recognized by Kilian et al.
able detail, and the knowledge has been used to design (1978); however, serovar 5 was subdivided into subtypes
new vaccines and diagnostic methods, but the best and 5A and 5B (Nielsen 1986a). Subsequently, serovars 6, 7
most economical methods for control of the disease are (Rosendal and Boyd 1982), 8 (Nielsen and O’Connor
still a matter for discussion. 1984), 9 (Nielsen 1985b), 10 (Nielsen 1985c), 11 (Kamp

343
344 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

et al. 1987), and 12 (Nielsen 1986b) have been proposed. America, Japan, Korea, Taiwan, and Australia. Although
Serovar 10 was erroneously proposed by Kamp et al. some serovars are prevalent in certain countries (e.g.,
(1987) and was later referred to as serovar 11 (Nielsen serovar 2 in Switzerland, Denmark, and Sweden and
1986b). The serologic specificity is given by the capsular serovars 1 and 5 in the United States and Canada), sever-
polysaccharides and cellular lipopolysaccharides (LPS). al serovars may occur in the same country. Some serovars
However, some serovars show structural similarities or (e.g., serovar 3), considered to be of low virulence and of
have identical LPS O chains, thus explaining the cross-re- no epidemiologic importance in certain countries, may
actions observed between serovars 1–9 and 11; serovars be epidemic in others (Desrosiers et al. 1984; Brandreth
3, 6, and 8; and serovars 4 and 7 (Perry et al. 1990). and Smith 1985). Moeller et al. (1992) analyzed the mul-
Serotyping has been extended into biovar 2, where three tilocus enzyme electrophoresis (MEE) types of organ-
serotypes have been identified. isms in one restricted area and found that the elec-
A number of structures and proteins have been iden- trophoretic type (ET) was unrelated to the severity of the
tified in A. pleuropneumoniae. Some are involved in path- disease. The international relationship of the different
ogenicity and will be described in more detail below, and serovars is of special interest, for it points to a transmis-
many have been identified and their genes sequenced. sion through international exchange of animals. For fur-
Because those of importance in pathogenesis, diagnosis, ther information on the historic geographic distribution,
and protection are mentioned elsewhere in the text, they see the review of Sebunya and Saunders (1983). A series
are only reviewed briefly here. of papers have provided further information on the dis-
The cell wall is surrounded by a capsule as described tribution of serovars within countries (e.g., Austria,
above. The capsular polysaccharides of most serotypes where serovars 4–6 and 10 are most common; see Hofer
have been studied in considerable detail and their com- et al. 1996) or within regions of countries (e.g., Catalo-
position is known (Perry et al. 1990). Nonencapsulated nia, Spain, where 11 serovars, principally 1, 2, 4, 7, 9, and
forms have been produced (Inzana et al. 1993). The or- 11, have been identified; see Clota et al. 1996). McDowell
ganism produces fimbriae 0.5–2 nm in diameter and and Ball (1994) showed that serovars 1 and 4 were absent
60–450 nm in length, which have been identified in elec- from the British Isles, which may reflect the restrictions
tron microscopy (Utrera and Pijoan 1991). Outer-mem- on imports of live pigs to the U.K. from neighboring
brane proteins of MW 43 kDa have been identified countries of the European Union where these serovars
(O’Reilly et al. 1991; Hartmann et al. 1995) and may vary exist. Different serovars may also occur within farms.
with NAD availability. Iron-binding proteins have been The economic importance of the disease is principal-
identified (D’Silva et al. 1995) and characterized and the ly due to the mortality and production and medical costs
genes sequenced (Gonzalez et al. 1995). Enzymes such as in acute outbreaks. In chronically infected herds, Hunne-
superoxide dismutase have been identified and the gene man (1986) found that the rate of daily weight gain was
cloned (Langford et al. 1996), and secreted proteases not affected, although a study by Hartley et al. (1988)
have been demonstrated (Negrete-Abascal et al. 1994). showed that pleurisy at slaughter was associated with an
The best-known extracellular products are the three cy- increase of 1 day to slaughter, and clinical disease with
totoxins belonging to the RTX family of toxins and an increase of 8 days to slaughter. Rohrbach et al. (1993)
named by Frey et al. (1993). ApxI is a strong hemolysin of demonstrated that the presence of infection in a herd de-
105–110 kDa, is present in serotypes 1, 5, 9, 10, and 11, layed slaughter at 113.6 kg by 5.64 days.
and is encoded by the apx operon consisting of apxIC, A. pleuropneumoniae is a parasite of the respiratory
apxIIA, apxIB, and apxID for the activator, the structural tract with a high host specificity for pigs. In peracute and
gene, and the two secretion genes, respectively. ApxII is a acute infections, it can be found not only in pneumonic
hemolysin of 103–105 kDa found in all reference strains lesions or in septicemia but also in large numbers in
except for serotype 10 and is governed by similar genes, nasal discharges. Survivors of acute infections become
although secretion protein genes appear to be those of carriers, and the infectious agent is located mainly in
ApxI. ApxIII is a nonhemolytic cytotoxic protein of 120 necrotic lung lesions and/or in the tonsils, less frequent-
kDa found in serotypes 2, 3, 4, 6, and 8 and governed by ly in the nasal cavity (Kume et al. 1984).
the apxIII operon. The genes and many of the operons The incubation period can be quite variable. Experi-
governing them have been cloned and sequenced (e.g., mental infections show that an exposure to large num-
the apxIII operon; Jansen et al. 1994). bers of organisms leads to death within a few hours to a
few days. Exposure to low levels of infection may result
EPIDEMIOLOGY in subclinical disease.
All age categories are susceptible, but the presence of
Pleuropneumonia of the pig is widely distributed. It has toxin-neutralizing antibody can affect susceptibility in
become more important as pig production has become herds where the infection is endemic (Cruijsen et al.
more intensive. Outbreaks have been reported from 1995). The demonstration of antibody in sow colostrum
practically all European countries and from different in endemically infected herds and the persistence of
parts of the United States and Canada, Mexico, South colostral antibody in piglets may account for the com-
CHAPTER 26 ACTINOBACILLUS PLEUROPNEUMONIAE Taylor 345

mon development of the disease in pigs from 6 to 8 by aerosol or by contact, and experimental studies have
weeks of age. In the acute phase of the disease morbidi- shown that the organism can colonize the tonsil and ad-
ty is generally high. Depending on the virulence of the here to the alveolar epithelium. The same ET type may be
strain and on the particular environment, mortality may present in both sites in natural infections (Moeller et al.
vary but is generally high. Both morbidity and mortality 1992). Adhesion appears to be fimbrial in nature, al-
may be exacerbated by the presence of diseases such as though it is not clear whether this also applies to the ad-
Aujeszky’s disease and porcine reproductive and respira- hesion between the A. pleuropneumoniae capsule and the
tory syndrome (PRRS), although experimental studies receptors found in respiratory tract secretions (Belanger
have confirmed that combined infection with A. pleuro- et al. 1994). A. pleuropneumoniae organisms in the lung
pneumoniae and PRRS virus may not necessarily result in are rapidly phagocytosed by, or adhere to, alveolar
more severe disease (Pol et al. 1997). Extensive reviews of macrophages and produce the toxins ApxI, ApxII, and
epidemiologic features are given by Nielsen (1982), ApxIII. All are potentially toxic for alveolar
Nielsen and Mandrup (1977), and Rosendal and macrophages, endothelial cells, alveolar epithelial cells,
Mitchell (1983). and the endothelial cells of the capillaries of the alveolar
The main route of spread is airborne and the disease wall, although ApxIII is particularly active against alveo-
is transmitted mainly by direct contact from pig to pig or lar macrophages. The organisms appear to be protected
by droplets within short distances. In acute outbreaks from digestion by phagocytes by their capsules and also
the infection may not occur in every pen, suggesting the appear to be resistant to the action of complement
possible role of aerosols and air movement in the trans- (Rycroft and Cullen 1990). Infection is accompanied by
mission of the disease over longer distances within the elaboration of cytokines such as interleukin-1β and
buildings or the indirect transmission of contaminated IL-8 in the alveolar fluid, and tumor necrosis factor
exudate from acutely infected pigs by farm personnel. (TNF) is also present in the early tissue lesions (Baarsch
Transmission by small rodents or birds is doubtful, and et al. 1995). The damage from toxins and cytokines
humans are not common hosts of A. pleuropneumoniae. which accompanies infection is largely responsible for
Survival of the organism in the environment is consid- the lesions which develop (Bertram 1990; Fenwick 1990;
ered to be of short duration. When protected with mu- Inzana 1990; Nicolet 1990). The peracute and acute
cus or other organic matter, it can, however, survive for a forms of the disease were considered by Kiorpes et al.
few days and it can survive in clean water for periods of (1990) to produce a systemic effect similar to septic
up to 30 days at 4˚C. shock in humans. Differences in the virulence between
Transmission between herds occurs through the in- the serovars or even within the same serovar have often
troduction of carrier animals to populations without been observed (Desrosiers et al. 1984; Rosendal et al.
previous experience of the disease. Airborne transmis- 1985; Brandreth and Smith 1987). It is suggested that
sion from farm to farm must be taken into consideration such differences are due to capsular structure (Jacques et
but has not yet been demonstrated. Moving and mixing al. 1988), LPS composition (Jensen and Bertram 1986),
pigs increase the risk of pleuropneumonia. Factors such or type of hemolysin (Frey and Nicolet 1990). In general,
as crowding and adverse climatic conditions such as strains of serovars 1, 5, 9, 10, and 11 are found to be more
rapid changes in temperature and high relative humidity virulent than those of other serovars. Concomitant in-
coupled with insufficient ventilation encourage the de- fections with other pathogens of the respiratory tract
velopment and spread of the disease and, consequently, which aid the development of pleuropneumonia have
also affect morbidity and mortality. It is, therefore, not been described by Caruso and Ross (1990).
surprising that the highest incidence of outbreaks is ob- Lung lesions resulting from the toxic changes may be
served in growing and finishing pigs, mainly in seasons seen as early as 3 hours after experimental infection and
with adverse weather conditions. As a rule, large herds become progressively more obvious. The alveolar wall
which mix pigs frequently are more at risk than small becomes edematous and capillary congestion develops.
herds or herds with separate units. The lymphatics become dilated with edema fluid, fibrin,
Introduction of the disease by artificial insemination and inflammatory cells. Platelet aggregation and neu-
or embryo transfer is improbable, since the genital tract trophil accumulation may also be seen in the damaged
is not a common site of infection, and antimicrobials in alveolar wall, and both arteriolar thrombosis and wall
the diluent would prevent survival of the contaminating necrosis may develop to produce infarction. Micro-
organism. colonies of the organism may be seen in infected alveoli,
and bacteremia may occur. The edges of the lesions be-
PATHOGENESIS come filled with dead and damaged macrophages or de-
bris and can easily be demarcated from the surrounding
The pathogenesis of pleuropneumonia has been studied lung by 4 days postinfection. Purulent exudate contain-
extensively, both in terms of the development of the le- ing organisms is present in the bronchi. As the lesions
sions and in terms of the relationship of the organism to age, their centers become necrotic and healing occurs by
the tissues at a more molecular level. Infection is usually fibrosis.
346 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Experimental or natural infections stimulate an im- the extent of the lung lesions and the time of initiation of
mune response, and circulating antibodies can be detect- therapy. All stages of disease, from intermediate to fatal,
ed approximately 10–14 days postinfection. These anti- subacute, or chronic, may develop within an affected
bodies reach a maximum level within 4–6 weeks group.
postinfection and may persist at low levels for many The subacute and chronic forms develop after the dis-
months or even disappear after a bacteriologic cure appearance of acute signs. There is little or no fever, and
(Nicolet 1970; Nicolet et al. 1971; Bachmann 1972; a spontaneous or intermittent cough of varying intensi-
Nielsen 1982, 1988). Immune sows confer passive im- ty develops. Appetite may be reduced, and this may con-
munity on their offspring. Such colostral antibodies may tribute to the decreased rate of gain in body weight. Af-
persist for about 5–9 weeks (Bachmann 1972), but the fected animals can be identified by their intolerance of
protection may only last for as little as 3 weeks in some exercise. When moved, they lag behind the group and
cases (Nielsen 1975). The antibodies are directed against struggle only feebly when restrained. In chronically in-
a wide range of bacterial structures and products, in- fected herds there are often many subclinically diseased
cluding capsule, LPS antigens, toxins (which can be neu- animals. The clinical signs may be exacerbated by other
tralized), outer-membrane proteins, superoxide dismu- respiratory infections (mycoplasmal, bacterial, or viral).
tase, and iron-binding proteins. Both local IgA In primary outbreaks abortions may be observed (Wil-
antibodies and serum IgG antibodies are produced. son and Kierstead 1976), especially in specific-pathogen-
free (SPF) herds. Complications such as arthritis, endo-
CLINICAL SIGNS carditis, and abscesses in different sites may occur in
individual animals and were considered by Nicolet
Clinical signs vary with the age of the animals, their state (1970) to be due principally to serovar 3 of A. pleuro-
of immunity, the environmental conditions, and the de- pneumoniae, although other serovars have now been re-
gree of exposure to the infectious agent. The clinical ported from these sites. Middle-ear disease has recently
course can be peracute, acute, or chronic (Nicolet et al. been associated with A. pleuropneumoniae infection (Duff
1969; Nielsen 1982; Shope 1964). et al. 1996).
In the peracute form, one or more weaned pigs in the
same or different pens suddenly become very ill with LESIONS
fever to 106.7˚F (41.5˚C), apathy, and anorexia. There is
a short period of slight diarrhea and vomiting. The af- The gross pathological lesions are located mainly in the
fected animals lie on the floor without distinct respirato- respiratory tract. The pneumonia is mostly bilateral,
ry signs, the pulse rate increases very early, and cardiac with involvement of the cardiac and apical lobes, as well
and circulatory failure develop. The skin on the nose, as at least part of the diaphragmatic lobes, where pneu-
ears, legs, and later the whole body becomes cyanotic. In monic lesions are often focal and well demarcated (Fig.
the terminal phase, there is a severe dyspnea with mouth 26.1). In rapidly fatal cases the trachea and bronchi are
breathing, animals remain in a sitting posture, and rectal filled with a foamy, blood-tinged, mucous exudate and
temperatures decrease markedly. Shortly before death, few gross changes may be obvious. In slightly later pera-
there is usually a copious, foamy, blood-tinged discharge cute cases, the pneumonic areas appear dark and solid
through the mouth and nostrils. Death occurs within with little or no fibinous pleurisy; and the cut surface is
24–36 hours of the development of clinical signs. Occa- friable. Fibrinous pleurisy is very obvious in animals
sionally an animal may die suddenly without premonito- which die in the acute stage of the disease, at least 24
ry clinical signs or be found dead in a pen; experimental hours after infection, and the thoracic cavity contains a
studies have shown that the course of the disease may be blood-tinged fluid. As the lesions age, the fibrinous
as little as 3 hours from infection to death. In neonatal pleurisy over the affected areas of lung becomes fibrous
pigs the disease occurs as a septicemia with fatal results. and may adhere so strongly to the parietal pleura that
In the acute form, many pigs in the same or different lung parenchyma may remain attached to the parietal
pens are affected. Body temperature rises to 105–106˚F pleura when the lungs are removed at postmortem ex-
(40.5–41˚C), the skin may be reddened, and the animals amination. The uniform dark red or black of the early
are depressed, are reluctant to rise, refuse food, and are lung lesion becomes lighter in color and remains firm on-
reluctant to drink (Pijpers et al. 1990). Severe respiratory ly in the worst-affected areas. The lesions shrink in size as
symptoms with dyspnea, cough, and sometimes mouth resolution progresses, until in more chronic cases nod-
breathing are evident. Cardiac and circulatory failure are ules of different sizes remain, mostly in the diaphrag-
usually present, with congestion of the extremities. matic lobes. These abscess-like nodules are delimited by
There is a marked loss of condition, which is apparent a thick capsule of connective tissue (Fig. 26.2) and may
within 24 hours of the onset of the disease. The course of be associated with areas of adhesive fibrous pleurisy. In
the disease differs from animal to animal, depending on many cases the lung lesion resolves, and only a residual
CHAPTER 26 ACTINOBACILLUS PLEUROPNEUMONIAE Taylor 347

focus of adhesive fibrous pleurisy remains. A high preva-


lence of chronic pleuritis at slaughter is very suggestive
of pleuropneumonia.
In the early stages of the disease, the histopathologic
changes are characterized by necrosis, hemorrhage, neu-
trophil infiltration, macrophage and platelet activation,
vascular thrombosis, widespread edema, and fibrinous
exudate (Bertram 1985, 1986, 1988; Liggett and Harri-
son 1987). Following the acute response, macrophage in-
filtration, marked fibrosis around areas of necrosis, and
fibrous pleuritis are characteristic (Häni et al. 1973). Fur-
ther descriptions are given in early papers by Pattison et
al. (1957), Olander (1963), and Shope (1964).

DIAGNOSIS

Pleuropneumonia may be suspected clinically in acute


outbreaks. In such cases the presence of characteristic
lung lesions with pleurisy at postmortem examination
enhances suspicion, which is enforced by the histological
appearance of the lesions. The presence of an acute ex-
udative pneumonia with areas of necrosis surrounded by
palisades and whorls of neutrophil debris provides fur-
ther evidence for pleuropneumonia. In chronic infec-
tions the necropsy findings of firm, well-demarcated ab-
scesses associated with pleurisy and pericarditis are very
26.1. Gross lesions of a peracute case of pleuropneumo- suggestive. In view of the importance of this disease to
nia showing a well-circumscribed pneumonic area in herd health control programs and the potential for eco-
different lobes with fibrinous pleurisy. (Courtesy nomic loss, bacteriologic confirmation of the diagnosis
Professor H. König, Institute of Animal Pathology, should be carried out.
University of Berne.) It is easy to demonstrate the etiologic agent in
bronchial or nasal exudate and pneumonic lesions from
freshly dead animals. Gram-stained smears of lung le-
sions contain numerous gram-negative coccobacilli.
Their identity can be confirmed as A. pleuropneumoniae

26.2. Chronic pleuropneumo-


nia. Cross section of a well-
capsulated nodule in the
diaphragmatic lobe. (Courtesy
Professor H. König, Institute of
Animal Pathology, University of
Berne.)
348 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

by using a fluorescent antibody test (Nicolet 1970), by epidemiologic investigations (Nielsen 1988). Antibody
immunoperoxidase, by detection of serovar-specific anti- may be detected in a number of ways. Methods such as
gens in lung extracts with a coagglutination test (Mittal the complement fixation (CF) test or a 2-mercap-
et al. 1983), by using a latex agglutination test, or by toethanol (2-ME) tube agglutination (Mittal et al. 1984)
ELISA. Nucleic acid from bacteria may be detected by a are suitable for this purpose.
number of methods, including labeled DNA probes in Simple methods include ELISAs (Nicolet et al. 1981;
tissue and polymerase chain reaction (PCR). Direct con- Morrison et al. 1984), and antigens such as cell extracts
firmation by PCR of the presence of A. pleuropneumoniae or long-chain LPSs have been evaluated (Gottschalk et al.
in tissue is not yet routine. 1994). Blocking ELISAs (Nielsen 1995) and testing with
Primary isolation of A. pleuropneumoniae from tissues serovar-specific antigens allow the identification of anti-
and secretions may be carried out on 5% sheep blood body to a specific serovar, while detection of antibody to
agar with a cross-streak of Staphylococcus epidermidis or the toxins and, especially, neutralizing antibody to those
S. aureus. After aerobic incubation overnight, small toxins can indicate a measure of protection (Cruijsen et
colonies appear in the neighborhood of the streak (NAD al. 1995). Serum antibody determination can be used to
requirement) surrounded by a clear zone of complete he- determine antibody profiles of herds or to demonstrate
molysis. This allows a rapid presumptive bacteriologic the presence of colostral antibody (Nielsen 1995). In all
diagnosis. Altered blood agars (“chocolate agar”) allow serologic examinations, care should be taken to exclude
the growth of the organism, but it is less distinctive on antibodies generated by infection with A. suis and other
these media. Biochemical identification can be carried members of the Pasteurellaceae.
out by demonstrating the CAMP phenomenon, urease Hog cholera, erysipelas, and streptococcal infections
activity, and the fermentation of mannitol. In the case of must be considered in the possible differential diagnosis
mixed infections, particularly with P. multocida, or cont- of peracute and acute cases. In subacute and chronic in-
amination with other bacteria, the use of selective media fections, the lung lesions should be distinguished from
is recommended (Little and Harding 1971; Gilbride and those caused by pyogenic bacterial agents such as A. pyo-
Rosendal 1983). Jacobsen and Nielsen (1995) described a genes, S. aureus, diphtheroid rods, and Fusobacterium
selective medium capable of isolating the organism from necrophorum. The lung lesions caused by other porcine
tonsils, an important site of carriage. However, isolation actinobacilli may be indistinguishable from those of
may fail in very old chronic lesions or where treatment pleuropneumonia, and acute pasteurellosis may some-
has been given. In peracute cases it is possible to isolate times resemble pleuropneumonia.
the agent from other organs (septicemia).
Isolates may be confirmed as belonging to A. pleuro- TREATMENT
pneumoniae by PCR (Gram et al. 1996) or by serologic
tests using absorbed or monoclonal antibodies. They can A. pleuropneumoniae is particularly susceptible in vitro to
be identified to serovars using a PCR for the activator and penicillin, ampicillin, cephalosporin, chloramphenicol,
structural genes of the toxins (Frey et al. 1995) or, more tetracyclines, colistin, sulfonamide, cotrimoxazole
conventionally, using monoclonal antibodies to particu- (trimethoprim + sulfamethoxazole), and gentamicin, to
lar serovars (e.g., Lairini et al. 1995, for serovar 1; Ro- which it has low minimum inhibitory concentrations
driguez-Barbosa et al. 1995, for serovar 2). Serotyping (MIC). High MIC values are found for streptomycin,
can be routinely achieved by slide agglutination from a kanamycin, spectinomycin, spiramycin, and lincomycin
subculture on a medium enriched with serum (Nicolet (Nicolet and Schifferli 1982; Gilbride and Rosendal
1971) or by the coagglutination test (Mittal et al. 1987). 1984; Nadeau et al. 1988; Inoue et al. 1984). Sensitivities
In many cases the final identification can only be of A. pleuropneumoniae to antimicrobials have been re-
achieved by agar gel diffusion and by indirect hemagglu- viewed by Prescott and Baggot (1993).
tination. A critical evaluation of the earlier serotyping The emergence of resistance to ampicillin, strepto-
methods is given by Nicolet (1988). It is possible to mycin, sulfonamides, tetracyclines, and chlorampheni-
serotype organisms in tissue and as isolates with latex col is of serious concern. It seems to be frequent in
particle tests (Inzana 1995) using capsular polysaccha- serovars 1, 3, 5, and 7 (Gilbride and Rosendal 1984; Vail-
ride to sensitize the particles. The serotyping of isolates lancourt et al. 1988) but rare in other serovars, particu-
is recommended for rapid confirmation of the bacterio- larly serovar 2 (Nicolet and Schifferli 1982; Inoue et al.
logic diagnosis and is essential when vaccination policy is 1984). The antibiotic resistance is plasmid mediated
being considered. It demonstrates the local distribution (Hirsh et al. 1982; Huether et al. 1987; Wilson et al.
of serovars and allows the epidemiologic situation to be 1989).
evaluated and the performance of specific serologic tests The antimicrobial of first choice should be the one
to be monitored. with the lowest MIC and with the most satisfactory phar-
The detection of antibodies is of little diagnostic val- macokinetic properties. Consequently, the betalactams
ue in recent outbreaks but provides an important tool in (principally penicillin and cephalosporin), chloram-
CHAPTER 26 ACTINOBACILLUS PLEUROPNEUMONIAE Taylor 349

phenicol, cotrimoxazole, and, to some extent, tetracy- Once the infection has been established on a farm, it
clines are considered to be most active. Recently avail- is difficult to eliminate the infectious agent, although the
able substances such as the quinolone derivatives (en- herd may become clinically normal. Control programs
rofloxacin) (Kobisch et al. 1990) or the semisynthetic must take account of the epidemiologic features of pleu-
cephalosporin ceftiofur sodium (Stephano et al. 1990) ropneumonia. The first priority must be to control eco-
have been shown to be particularly effective after experi- nomic loss (mortality, clinical and subclinical disease)
mental challenge. Satisfactory results in the field have and then to consider the control or elimination of infec-
been reported with tiamulin (Anderson and Williams tion. Mortality can be controlled by the treatment of cas-
1990) and a combination of lincomycin and spectino- es and of infected groups using the methods and antimi-
mycin (Hsu 1990). Tilmicosin has been used in feed by crobials outlined above. Disease may be treated at an
Moore et al. (1996). The determination of an antibi- early stage by treating groups of animals into clean air-
ogram is recommended where problems are being expe- spaces and then maintaining them as a group in isolation
rienced with treatment. until slaughter. Where this may not be possible, control
Antibiotic therapy is effective in clinically affected an- of environmental factors such as temperature and venti-
imals only in the initial phase of the disease, when it can lation and use of solid partitions between pens may min-
reduce mortality. The nature of the lesions means that imize the development and severity of disease. Continu-
delay in treatment can result in a degree of infarction ous medication or pulse dosing may be practiced, but
and chronic damage which will leave the animal as a res- neither should be used for long, and the antimicrobial
piratory cripple even if it recovers. Antibiotics should be sensitivity of the organism should be monitored contin-
given parenterally (subcutaneously or intramuscularly) uously. Strategic medication should be targeted at peri-
and in high dosage, as affected animals may not eat or ods of risk, which can be identified by routine post-
drink (Pijpers et al. 1990). To ensure effective and mortem examinations, clinical examinations, and herd
durable blood concentrations, repeated injections may antibody profiles. The generalized methods used to con-
be required, depending on the pharmacokinetic proper- trol respiratory disease, such as the all-in/all-out system
ties of the antibiotic used. The success of therapy de- in fattening units, segregated early weaning, and large
pends mainly on early detection of clinical signs and on airspaces with separation, will all considerably reduce
rapid therapeutic intervention. Water treatment may be the risk of infection. Animals should be brought in only
used to treat members of the affected group which are from herds free from infection to avoid introducing new
still able to drink. Feed medicated with any of the above serovars or new antimicrobial resistances. In chronically
antimicrobials may be used successfully if all pigs have a infected herds, purchased seronegative animals should
normal food and water intake. Feed and water medica- be vaccinated before introduction to the herd.
tion can be used for the strategic treatment of infected A wide range of vaccines has been developed for this
groups on entry to an airspace. A combination of par- disease. They fall into two main groups: the killed organ-
enteral and peroral medication in a recent outbreak of- isms and the subunit vaccines. Vaccination with killed
ten yields the best results. In spite of apparent clinical organisms is serovar specific (Nielsen 1984), with possi-
success, it must be remembered that antibiotic therapy ble cross-immunity with cross-reacting serovars (Nielsen
does not eliminate infection in a herd. Chronic infections 1985a). The protection afforded has been extended by
in lung abscesses or on the tonsils of carriers persist to including all the serovars present in an area (e.g., 3, 6,
form an important source of infection for other animals. and 8 in the United Kingdom). The type of adjuvant used
Severely affected animals may not recover even after may affect efficacy, and care may also be necessary before
treatment and nursing and should be killed. using certain adjuvants in pigs intended for human con-
sumption, as some vaccines can produce undesirable
PREVENTION granulomatous lesions at the site of injection (Straw et
al. 1985). Vaccines in the second group are currently un-
Prevention and control of pleuropneumonia may be ac- der development or being marketed and consist of vary-
complished in a number of different ways. Farms free ing combinations of subunits such as toxins and outer-
from the disease and infection should maintain a policy membrane proteins. A wide range of antigens has been
of isolation coupled with the use of semen or embryos to found to be protective. They include the toxins (Haga et
introduce new genes. Any animals introduced should be al. 1997), combinations of Apx toxins and the outer-
hysterectomy derived or originate from herds known to membrane proteins (Valks et al. 1996), extracellular
be free from the disease and from infection. It may be products of A. pleuropneumoniae, and structural or func-
appropriate to hold them in quarantine prior to intro- tioning antigens such as the iron-binding proteins. Vac-
duction to the herd; developments in detecting the or- cines of this type are generally protective against all
ganism and in serology make it increasingly worthwhile serovars.
to test them for either infection or antibody prior to in- All the vaccines currently used are administered par-
troduction to the main herd. enterally, usually as a course of two injections, and are
350 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

intended to provide protection to the growing pig. Ex- under strict separation from the potentially infected
perimental vaccines consisting of attenuated organisms stock. These piglets, which are seronegative up to the age
(Inzana et al. 1993) and killed organisms or their sub- of 12 weeks, serve to restock the herd. Seropositive sows
units have been given by the aerosol and oral routes and are systematically eliminated until the entire breeding
have provided some protection. stock is seronegative. This program can take 6–12
Vaccines are generally given as maternal antibody months. During the elimination procedure, the whole
wanes and provide high levels of protection against mor- herd is protected from reinfection by medicated feed, for
bidity in experiments, reduce mortality, reduce the num- example, cotrimoxazole (trimethoprim + sulfamethoxa-
ber of treatments required, increase daily liveweight zole 1:20, 250 mg/kg feed). Certain reports suggest that
gain, and may improve feed conversion efficiency. The only partial success (Lariviere et al. 1990) or even failure
quality of the carcass is also improved, with fewer con- (Hunnemann 1986) has resulted from the application of
demnations for pneumonia and lower slaughtering costs such eradication programs, and the current require-
through reductions in pleurisy and pericarditis. The old- ments to avoid antimicrobial residues and to reduce an-
er generation of vaccines were not always effective in the timicrobial resistance may make them impractical.
field (Hunneman 1986). Vaccines do not always prevent
the carrier state but have been used as an aid in eradica- REFERENCES
tion programs. The decision to vaccinate should be care-
fully evaluated; the costs of mortality alone should not Anderson, M. D., and Williams, J. A. 1990. Effects of tia-
be the sole consideration, because the other effects on mulin base administered intramuscularly to pigs for
productivity listed above contribute to the benefits of treatment of pneumonia associated with Actinobacillus
vaccination. pleuropneumoniae. Proc Int Congr Pig Vet Soc 11:15.
Control of pleuropneumonia on a farm can be ac- Baarsch, M. J.; Scamurra, R. W.; Burger, K.; Foss, D. L.; Ma-
complished by combining treatment, vaccination, and heswaran, S. K.; and Murtaugh, M. P. 1995. Inflamma-
husbandry practices such as those outlined above. Disin- tory cytokine expression in swine experimentally infect-
fection should be included in any control program; the ed with Actinobacillus pleuropneumoniae. Inf and Imm
organism is sensitive to a wide range of commonly used 63:3587–3594.
disinfectants (Gutierrez et al. 1995). Bachmann, P. 1972. Beitrag zur Epidemiologie der Konta-
Control of pleuropneumonia in a region or breeding giosen Pleuropneumonie beim Schwein. Schweiz Arch
pyramid involves health schemes aimed at pleuropneu- Tierheilkd 114:362–382.
monia-free breeding and multiplying herds, serologic Belanger, M.; Dubreuil, D.; and Jacques, M. 1994. Proteins
monitoring, monitoring at slaughter and postmortem found within porcine respiratory tract secretions bind
examination of casualties, control of management, and lipopolysaccharides of Actinobacillus pleuropneumoniae
controlled pig traffic (serologic testing, quarantine). For capsular layer. Inf and Imm 62:868–873.
herds infected with A. pleuropneumoniae intending to join Bertram, T. A. 1985. Quantitative morphology of peracute
such a scheme, an eradication program is the method of pulmonary lesions in swine induced by Haemophilus
choice but requires careful evaluation of the economic pleuropneumoniae. Vet Pathol 22:598–609.
consequences. Depopulation and restocking with pigs ———. 1986. Intravascular macrophages in lungs of pigs
originating from certified pleuropneumonia-free herds is infected with Haemophilus pleuropneumoniae. Vet Pathol
the method of choice. This method is expensive and may 23:681–691.
lead to the loss of important bloodlines. These can be ———. 1988. Pathobiology of acute pulmonary lesions in
saved by a program of hysterectomy and separate rear- swine infected with Haemophilus (Actinobacillus) pleuro-
ing. pneumoniae. Can Vet J 29:574–577.
An extensive program of testing and evaluation is re- ———. 1990. Actinobacillus pleuropneumoniae: Molecular
quired before herds undergoing all other types of eradi- aspects of virulence and pulmonary injury. Can J Vet Res
cation program can be admitted to a pool of pleuro- 54:S53–S56.
pneumonia-free herds. Methods which have succeeded Bertschinger, H. U., and Seifert, P. 1978. Isolation of a Pas-
in the past include an eradication program in the existing teurella haemolytica–like organism from porcine necrotic
herd area but weaning at another farm, at the same time pleuropneumonia. Proc Int Congr Pig Vet Soc 5:Abstr
supported by a program of vaccination, medication, and M19.
culling and repopulation with disease-free gilts (Larsen Biberstein, E. L.; Mini, P. D.; and Gills, M. G. 1963. Action
et al. 1990). Breeding herds with a relatively low percent- of Haemophilus cultures on α-aminolevulinic acid. J Bac-
age of seropositive animals (up to 30%) have used the teriol 86:814–819.
“test-and-removal” of seropositive animals under med- Brandreth, S. R., and Smith, I. M. 1985. Prevalence of pig
ication (Nicolet 1970; Nielsen et al. 1976). The principle herds affected by pleuropneumonia associated with
is based on the serologic testing of sows shortly before Haemophilus pleuropneumoniae in eastern England. Vet
farrowing and by weaning the piglets at 2 weeks of age Rec 117:143–147.
CHAPTER 26 ACTINOBACILLUS PLEUROPNEUMONIAE Taylor 351

———. 1987. Comparative virulence of some English Gonzalez, G. C.; Yu, R. H.; Rosteck, P. R., Jr.; and Schryvers,
strains of Haemophilus pleuropneumoniae serotypes 2 A. B. 1995. Sequence, genetic analysis and expression of
and 3 in the pig. Res Vet Sci 42:187–193. transferrin receptor genes. Microbiology (Reading)
Caruso, J. P., and Ross, R. F. 1990. Effects of Mycoplasma hy- 141:2405–2416.
opneumoniae and Actinobacillus (Haemophilus) pleuro- Gottschalk, M.; Altman, E.; Charland, N.; Lasalle, F. De; and
pneumoniae infections on alveolar macrophage func- Dubreuil, J. D. 1994. Evaluation of a saline boiled ex-
tions in swine. Am J Vet Res 51:227–231. tract, capsular polysaccharides and long-chain
Clota, J.; Foix, A.; March, R.; Riera, P.; and Costa, L. 1996. lipopolysaccharides of Actinobacillus pleuropneumoniae
Caracterizacion serologica de cepas de Actinobacillus serotype 1 as antigens for the serodiagnosis of swine
pleuropneumoniae aisladas en Espana. Med Vet 13:17–22. pleuropneumonia. Vet Micro 42:91–104.
Cruijsen, T.; Leengoed, L. A. M. G. Van; Kamp, E. M.; Bar- Gram, T.; Ahrens, P.; and Nielsen, J. P. 1996. Evaluation of a
telse, A.; Korevaar, A.; and Verheijden, J. H. M. 1995. PCR for detection of Actinobacillus pleuropneumoniae in
Susceptibility to Actinobacillus pleuropneumoniae infec- mixed bacterial cultures from tonsils. Vet Micro
tion in pigs from an endemically infected herd is related 51:95–104.
to presence of toxin neutralising antibody. Vet Micro Gutierrez, C. B.; Barbosa, J. I. R.; Suarez, J.; Gonzalez, O. R.;
47:219–228. Tascon, R. I.; and Ferri, E. F. R. 1995. Efficacy of a variety
Desrosiers, R.; Mittal, K. R.; and Malo, R. 1984. Porcine of disinfectants against Actinobacillus pleuropneumoniae
pleuropneumonia associated with Haemophilus pleuro- serotype 1. Am J Vet Res 56:1025–1029.
pneumoniae serotype 3 in Quebec. Vet Rec 115:628–629. Haga, Y.; Ogino, S.; Ohashi, S.; Ajito, T.; Hashimoto, K.; and
D’Silva, C. G.; Archibald, F. S.; and Niven, D. F. 1995. Com- Sawada, T. 1997. Protective efficacy of an affinity puri-
parative study of iron acquisition by biotype 1 and bio- fied haemolysin vaccine against experimental swine
type 2 strains of Actinobacillus pleuropneumoniae. Vet Mi- pleuropneumonia. J Vet Med Sci 59:115–120.
cro 44:11–23. Häni, H.; König, H.; Nicolet, J.; and Scholl, E. 1973. Zur
Duff, J. P.; Scott, J. W.; Wilkes, M. K.; and Hunt, B. 1996. Haemophilus-Pleuropneumonie beim Schwein. V. Patho-
Otitis in a weaned pig: A new pathological role for Acti- morphologie. Schweiz Arch Tierheilkd 115:191–203.
nobacillus (Haemophilus) pleuropneumoniae. Vet Rec Hartley, P. E.; Wilesmith, J. W.; and Bradley, R. 1988. The in-
139:561–563. fluence of pleural lesions in the pig at slaughter on the
Fenwick, B. W. 1990. Virulence attributes of the duration of the fattening period: An on-farm study. Vet
lipopolysaccharides of the HAP group of organisms. Rec 123:208.
Can J Vet Res 54:S28–S32. Hartmann, L.; Schroeder, W.; and Luebke, A. 1995. Isola-
Frey, J., and Nicolet, J. 1990. Hemolysin patterns of Acti- tion of the major outer membrane proteins of Acti-
nobacillus pleuropneumoniae. J Clin Microbiol nobacillus pleuropneumoniae and Haemophilus parasuis. J
28:232–236. Vet Med (B) 42:59–63.
Frey, J.; Bosse, J. T.; Chang, Y. F.; Cullen, J. M.; Fenwick, B.; Hirsh, D. C.; Martin, L. D.; and Libal, M. C. 1982. Plasmid-
Gerlach, G. F.; Gygi, D.; Haesebrouck, F.; Inzana, T. J.; mediated antimicrobial resistance in Haemophilus pleu-
Jansen, R.; Kamp, E. M.; Macdonald, J.; Macinnes, J. I.; ropneumoniae. Am J Vet Res 43:269–272.
Mittal, K. R.; Nicolet, J.; Rycroft, A. N.; Segers, R. P. A. Hofer, J.; Schoder, J.; Buchner, A.; Vanek, E.; Lapan, G.;
M.; Smits, M. A.; Stenbaek, E.; Struck, D. K.; Bosch, J. F. Fuchs, K.; and Baumgartner, W. 1996. Serologischer
Van Den; Wilson, P. J.; and Young, R. 1993. Actinobacil- Querschnittsuntersuchung zu Verbreitung der Acti-
lus pleuropneumoniae RTX toxins: Uniform designation nobacillus pleuropneumoniae-Infektion in Österreich
of haemolysins, cytolysins pleurotoxin and their genes. J unter Berucksichtigung von epidemiologische und be-
Gen Microbiol 139:1723–1728. triebspezifischen Einflussfaktoren. Wiener Tierärtzl
Frey, J.; Kuhn, R.; and Nicolet, J. 1994. Association of the Monatsschr 83:80–90.
CAMP phenomenon in Actinobacillus pleuropneumoniae Hsu, F. S. 1990. Evaluation of lincospectin sterile solution
with the RTX toxins ApxI, ApxII and Apx III. FEMS Mi- and Linco-Spectin 44 Premix in the treatment of pleu-
crobiol Lett 124:245–251. ropneumonia. Proc Int Congr Pig Vet Soc 11:15.
Frey, J.; Beck, M.; Bosch, J. F. Van Den; Segers, R. P. A. M.; Huether, M. J.; Fedorka-Cray, P. J.; Pfannenstiel, M. A.; and
and Nicolet, J. 1995. Development of an efficient PCR Anderson, G. A. 1987. Plasmid profiles and antibiotic
method for toxin typing of Actinobacillus pleuropneumo- susceptibility of Haemophilus pleuropneumoniae serotype
niae strains. Mol and Cell Probes 9:277–282. 1, 3, 5 and 7. FEMS Microbiol Lett 48:179–182.
Gilbride, K. A., and Rosendal, S. 1983. Evaluation of a selec- Hunneman, W. A. 1986. Incidence, economic effects, and
tive medium for isolation of Haemophilus pleuropneumo- control of Haemophilus pleuropneumoniae infections in
niae. Can J Comp Med 47:445–450. pigs. Vet Q 8:83–87.
———. 1984. Antimicrobial susceptibility of 51 strains of Inoue, A.; Yamamoto, K.; Hirano, N.; and Murakami, T.
Haemophilus pleuropneumoniae. Can J Comp Med 1984. Drug susceptibility of Haemophilus pleuropneumo-
48:47–50. niae strains isolated from pigs. Jpn J Vet Sci 46:175–180.
352 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Inzana, T. J. 1990. Capsules and virulence in the HAP group Lairini, K.; Stenbaek, E.; Lacouture, S.; and Gottschalk, M.
of bacteria. Can J Vet Res 54:S22–S27. 1995. Production and characterisation of monoclonal
———. 1995. Simplified procedure for preparation of sen- antibodies against Actinobacillus pleuropneumoniae
sitised latex particles to detect capsular polysaccharide: serotype 1. Vet Micro 46:369–381.
Application to typing and diagnosis of Actinobacillus Langford, P. R.; Loynds, B. M.; and Kroll, J. S. 1996. Cloning
pleuropneumoniae. J Clin Micro 33:2297–2303. and molecular characterisation of copper-zinc superoxi-
Inzana, T. J.; Todd, J.; and Veit, H. P. 1993. Safety, stability dase dismutase from Actinobacillus pleuropneumoniae.
and efficacy of non-encapsulated mutants of Acti- Inf and Imm 64:5035–5041.
nobacillus pleuropneumoniae for use in live vaccines. Inf Lariviere, S.; D’Allaire, S.; De Lasalle, F.; Nadeau, M.;
and Imm 61:1682–1686. Moore, C.; and Ethier, R. 1990. Eradication of Acti-
Jacobsen, M. J., and Nielsen, J. P. 1995. Development and nobacillus pleuropneumoniae serotype 1 and 5 infections
evaluation of a selective and indicator medium for isola- in four herds. Proc Int Congr Pig Vet Soc 11:17.
tion of Actinobacillus pleuropneumoniae from tonsils. Vet Larsen, H.; Hogedahl Jorgensen, P.; and Szancer, J. 1990.
Micro 47:191–197. Eradication of Actinobacillus pleuropneumoniae from a
Jacques, M.; Foiry, B.; Higgins, R.; and Mittal, K. R. 1988. breeding herd. Proc Int Congr Pig Vet Soc 11:18.
Electron microscopic examination of capsular material Liggett, A. D., and Harrison, L. R. 1987. Sequential study of
from various serotypes of Actinobacillus pleuropneumoni- lesion development in experimental haemophilus pleu-
ae. J Bacteriol 170:3314–3318. ropneumonia. Res Vet Sci 42:204–221.
Jansen, R.; Briare, J.; Geel, A. B. M. Van; Kamp, E. M.; Little, T. W. A., and Harding, J. D. J. 1971. The comparative
Gielkens, A. L. J.; and Smits, M. A. 1994. Genetic map of pathogenicity of two porcine Haemophilus species. Vet
the Actinobacillus pleuropneumoniae RTX-toxin (apx) Rec 88:540–545.
operons: Characterisation of the apxIII operons. Inf and Matthews, P. R. J., and Pattison, I. H. 1961. The identifica-
Imm 62:4411–4418. tion of a Haemophilus-like organism associated with
Jansen, R.; Briare, J.; Kamp, E. M.; Gielkens, A. L. J.; and pneumonia and pleurisy in the pig. J Comp Pathol
Smits, M. A. 1995. The CAMP effect of Actinobacillus 71:44–52.
pleuropneumoniae is caused by Apx toxins. FEMS Micro- McDowell, S. W. J., and Ball, H. J. 1994. Serotypes of Acti-
biol Lett 126:139–143. nobacillus pleuropneumoniae isolated in the British Isles,
Jensen, A. E., and Bertram, T. A. 1986. Morphological and 1988–1993. Vet Rec 134:522–523.
biochemical comparison of virulent and avirulent iso- Mittal, K. R.; Higgins, R.; and Lariviere, S. 1983. Detection
lates of Haemophilus pleuropneumoniae serotype 5. Infect of type-specific antigens in the lungs of Haemophilus
Immun 51:419–424 pleuropneumoniae–infected pigs by coagglutination test.
Kamp, E. M.; Popma, J. K.; and Leengoed, L. A. M. G. Van. J Clin Microbiol 18:1355–1357.
1987. Serotyping of Haemophilus pleuropneumoniae in ———. 1987. An evaluation of agglutination and coagglu-
the Netherlands: With emphasis on heterogeneity with- tination techniques for serotyping of Haemophilus pleu-
in serotype 2 and (proposed) serotype 9. Vet Microbiol ropneumoniae isolates. Am J Vet Res 48:219–226.
13:249–257. Mittal, K. R.; Higgins, R.; Lariviere, S.; and Leblanc, D.
Kilian, M. 1976. The haemolytic activity of Haemophilus 1984. A 2-mercaptoethanol tube agglutination test for
species. Acta Pathol Microbiol Scand (B) 84:339–341. diagnosis of Haemophilus pleuropneumoniae infection in
Kilian, M.; Nicolet, J.; and Biberstein, E. L. 1978. Biochemi- pigs. Am J Vet Res 45:715–719.
cal and serological characterization of Haemophilus pleu- Moeller, K.; Nielsen, R.; Andersen, L. V.; and Killian, M.
ropneumoniae (Matthews and Pattison 1961) Shope 1964 1992. Clonal analysis of the Actinobacillus pleuropneumo-
and proposal of a neotype strain. Int J Syst Bacteriol niae population in a geographically-restricted area by
28:20–26. multilocus enzyme electrophoresis. J Clin Micro
Kiorpes, A. L.; MacWilliams, P. S.; Schenkman, D. I.; and 30:623–627.
Backstrom, L. R. 1990. Blood gas and hematological Moeller, K.; Fussing, V.; Grimont, P. A. D.; Paster, B. J.; De-
changes in experimental peracute porcine pleuropneu- whirst, F. E.; and Killian, M. 1996. Actinobacillus minor,
monia. Can J Vet Res 54:164–169. sp.nov.; Actinobacillus porcinus, sp.nov. and Actinobacil-
Kobisch, M.; Vannier, P.; Delaporte, S.; and Dellac, B. 1990. lus indolicus sp. nov.; three new V factor dependent
The use of experimental models to study in vivo the an- species from the respiratory tract of pigs. Int J Syst Bact
tibacterial activity of Enrofloxacin against Actinobacillus 46:951–956.
(Haemophilus) pleuropneumoniae and Mycoplasma hyo- Moore, G. M.; Basson, R. P.; and Tonkinson, L. V. 1996.
pneumoniae in combination with Pasteurella multocida. Clinical trials with tilmicosin phosphate in feed for the
Proc Int Congr Pig Vet Soc 11:16. control of naturally-acquired pleuropneumonia caused
Kume, K.; Nakai, T.; and Sawata, A. 1984. Isolation of by Actinobacillus pleuropneumoniae and Pasteurella multo-
Haemophilus pleuropneumoniae from the nasal cavities of cida in swine. Am J Vet Res 57:224–228.
healthy pigs. Jpn J Vet Sci 46:641–647. Morrison, R. B.; Bahdat, F.; Hilley, H. D.; Pijoan, C.; and
CHAPTER 26 ACTINOBACILLUS PLEUROPNEUMONIAE Taylor 353

Hill, H. T. 1984. An enzyme-linked immunosorbent as- ———. 1985b. Serological characterization of Haemophilus
say using heat-treated antigen for the detection of pleuropneumoniae (Actinobacillus pleuropneumoniae)
Haemophilus pleuropneumoniae antibodies. Proc Int Con- strains and proposal of a new serotype: Serotype 9. Acta
gr Pig Vet Soc 8:102. Vet Scand 26:501–512.
Nadeau, M.; Lariviere, S.; Higgins, R.; and Martineau, G. P. ———. 1985c. Serological characterization of Haemophilus
1988. Minimal inhibitory concentrations of antimicro- pleuropneumoniae (Actinobacillus pleuropneumoniae)
bial agents against Actinobacillus pleuropneumoniae. Can strains and proposal of a new serotype: Serotype 10. Ac-
J Vet Res 52:315–318. ta Vet Scand 26:581–585.
Negrete-Abascal, E.; Tenonio, V. R.; Serrano, J. J.; Garcia, C. ———. 1986a. Serology of Haemophilus (Actinobacillus)
C.; and Garza, M. de la. 1994. Secreted proteases in su- pleuropneumoniae serotype 5 strains: Establishment of
pernates degraded porcine gelatine, haemoglobin and subtypes A and B. Acta Vet Scand 27:49–58.
IgA. Can J Vet Res 58:83–86. ———. 1986b. Serological characterization of Actinobacil-
Nicolet, J. 1968. Sur l’hemophilose du porc. I. Identification lus pleuropneumoniae strains and proposal of a new
d’un agent frequent: Haemophilus parahaemolyticus. serotype: Serotype 12. Acta Vet Scand 27:453–455.
Pathol Microbiol 31:215–225. ———. 1988. Seroepidemiology of Actinobacillus pleuro-
———. 1970. Aspects microbiologiques de la pleuropneu- pneumoniae. Can Vet J 29:580–582.
monie contagieuse du porc. These d’habilitation, Berne. ———. 1995. Detection of antibodies against Actinobacil-
———. 1971. Sur l’hemophilose du porc. III. Differencia- lus pleuropneumoniae serotype 2 in porcine colostrum us-
tion serologique de Haemophilus parahaemolyticus. Zen- ing a blocking enzyme-linked immunosorbent assay. Vet
tralbl Bakteriol Hyg (Orig A) 216:487–495. Micro 43:277–281.
———. 1988. Taxonomy and serological identification of Nielsen, R., and Mandrup, M. 1977. Pleuropneumonia in
Actinobacillus pleuropneumoniae. Can Vet J 29:578–580. swine caused by Haemophilus parahaemolyticus: A study
———. 1990. Overview of the virulence attributes of the of the epidemiology of the infection. Nord Vet Med
HAP group of bacteria. Can J Vet Res 54:S12–S15. 29:465–473.
Nicolet, J., and König, H. 1966. Zur Haemophilus-Pleuro- Nielsen, R., and O’Connor, P. J. 1984. Serological character-
pneumonie beim Schwein: Bakteriologische, patholo- ization of 8 Haemophilus pleuropneumoniae strains and
gisch-anatomische und histologische Befunde. Pathol proposal of a new serotype: Serotype 8. Acta Vet Scand
Microbiol 29:301–306. 25:96–106.
Nicolet, J., and Schifferli, D. 1982. In vitro susceptibility of Nielsen, R.; Thomsen, A. D.; and Vesterlund, S. D. 1976.
Haemophilus pleuropneumoniae to antimicrobial sub- Pleuropneumonia caused by Haemophilus para-
stances. Proc Int Congr Pig Vet Soc 7:71. haemolyticus: An attempt to control the disease at two
Nicolet, J.; Konig, H.; and Scholl, E. 1969. Zur Haemophilus- progeny testing stations by serological blood testing fol-
Pleuropneumonie beim Schwein. II. Eine kontagiose lowed by removal of the seropositive animals and their
Krankheit von wirtschaftlicher Bedeutung. Schweiz litter mates. Nord Vet Med 28:349–352.
Arch Tierheilkd 111:166–174. Niven, D. F., and Levesque, M. 1988. V-factor-dependent
Nicolet, J.; de Meuron, P. A.; and Bachmann, P. 1971. IV. growth of Actinobacillus pleuropneumoniae biotype 2
L’épreuve de fixation du complément, un test de (Bertschinger 20008/76). Int J Syst Bacteriol
dépistage des infections à Haemophilus parahaemolyticus. 38:319–320.
Schweiz Arch Tierheilkd 113:191–200. Olander, H. J. 1963. A Septicaemic Disease of Swine and Its
Nicolet, J.; Paroz, P. H.; Krawinkler, M.; and Baumgartner, Causative Agent Haemophilus parahaemolyticus. Ph.D.
A. 1981. An enzyme-linked immunosorbent assay, using diss. Univ California.
an EDTA-extracted antigen for the serology of O’Reilly, T.; Niven, D. F.; and Brown, M. R. W. 1991. Pheno-
Haemophilus pleuropneumoniae. Am J Vet Res typic variation in the outer membrane protein composi-
42:2139–2142. tion of Actinobacillus (H.) pleuropneumoniae: Non-specif-
Nielsen, R. 1975. Colostral transfer of immunity to ic effect of exogenous pyridine supply. Vet Micro
Haemophilus parahaemolyticus in pigs. Nord Vet Med 29:159–172.
27:319–328. Pattison, I. H.; Howell, D. G.; and Elliott, J. 1957. A
———. 1982. Haemophilus pleuropneumoniae Infection in Haemophilus-like organism isolated from pig lung and
Pigs. Thesis. Copenhagen. the associated pneumonic lesions. J Comp Pathol
———. 1984. Haemophilus pleuropneumoniae serotypes— 67:320–329.
Cross protection experiments. Nord Vet Med Perry, M. B.; Altman, E.; Brisson, J. R.; Beynon, L. M.; and
36:221–234. Richards, J. C. 1990. Structural characteristics of the
———. 1985a. Haemophilus pleuropneumoniae (Actinobacil- antigenic capsular polysaccharides and lipopolysaccha-
lus pleuropneumoniae) serotypes 8, 3 and 6—Serological rides involved in the serological classification of Acti-
response and cross immunity in pigs. Nord Vet Med nobacillus (Haemophilus) pleuropneumoniae strains. Sero-
37:217–227. diagnosis and Immunotherapy in Inf Dis 4:299–308.
354 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Pijpers, A.; Vernooy, J. A. C. M.; Leengoed, L. A. M. G. Van; infection of swine. Am J Vet Res 51:1449–1453.
and Verheijden, J. H. M. 1990. Feed and water consump- Sebunya, T. N. K., and Saunders, J. R. 1983. Haemophilus
tion in pigs following an Actinobacillus pleuropneumoniae pleuropneumoniae infection in swine: A review. J Am Vet
challenge. Proc Int Congr Pig Vet Soc 11:39. Med Assoc 182:1331–1336.
Pohl, S.; Bertschinger, H. U.; Frederiksen, W.; and Man- Shope, R. E. 1964. Porcine contagious pleuropneumonia. I.
heim, W. 1983. Transfer of Haemophilus pleuropneumo- Experiment transmission, etiology and pathology. J Exp
niae and the Pasteurella haemolytica–like organism caus- Med 119:357–368.
ing porcine necrotic pleuropneumonia to the genus Shope, R. E.; White, D. C.; and Leidy, G. 1964. Porcine con-
Actinobacillus (Actinobacillus pleuropneumoniae comb. tagious pleuropneumonia. II. Studies of the pathogenic-
Nov.) on the basis of phenotypic and deoxyribonucleic ity of the etiological agent Haemophilus pleuropneumoni-
acid relatedness. Inst J Syst Bacteriol 33:510–514. ae. J Exp Med 119:369–375.
Pol, J. M. A.; Leengoed, L. A. M. G. Van; Stockhofe, N.; Kok, Stephano, A.; Navarro, R.; Rayo, C. D.; and Osorio, M. 1990.
G.; and Wensvoort, G. 1997. Dual infections of Effect of the use of ceftiofur sodium injectable (Excenel
PRRS/influenza or PRRSV/Actinobacillus pleuropneumo- sterile powder) for the treatment of induced Actinobacil-
niae in the respiratory tract. Vet Micro 55:259–264. lus pleuropneumoniae: Multiple dose titration study. Proc
Prescott, J. F., and Baggot, J. D. 1993. Antimicrobial Thera- Int Congr Pig Vet Soc 11:41.
py in Veterinary Medicine. Ames: Iowa State Univ Press. Straw, B. E.; MacLachlan, N. J.; Corbett, W. T.; Carter, P. B.;
Rapp, V. J.; Ross, R. F.; and Young, T. F. 1985. Characteriza- and Schey, H. M. 1985. Comparison of tissue reactions
tion of Haemophilus spp. isolated from healthy swine produced by Haemophilus pleuropneumoniae vaccines
and evaluation of cross-reactivity of complement-fixing made with six different adjuvants in swine. Can J Comp
antibiotics to Haemophilus pleuropneumoniae and Med 49:149–151.
Haemophilus taxon “Minor group.” J Clin Microbiol Utrera, V., and Pijoan, C. 1991. Fimbriae in Actinobacillus
22:945–950. pleuropneumoniae strains isolated from pig respiratory
Rodriguez-Barbosa, J. I.; Gutierrez, C. B.; Tascon, R. I.; tracts. Vet Rec 128:357–358.
Suarez, J.; Noronia, F. de; and Rodriguez-Ferri, E. F. Vaillancourt, J. P.; Higgins, R.; Martineau, G. P.; Mittal, K.
1995. Characterisation of monoclonal antibodies to O R.; and Lariviere, S. 1988. Changes in the susceptibility
antigen of lipopolysaccharide of Actinobacillus pleuro- of Actinobacillus pleuropneumoniae to antimicrobial
pneumoniae serotype 2 and their use in the classification agents in Quebec (1981–1986). J Am Vet Med Assoc
of field isolates. FEMS Immunol and Med Micro 193:470–473.
11:35–44. Valks, M. M. H.; Nell, T.; and Bosch, J. E. Van Den. 1996. A
Rohrbach, B. W.; Hall, R. F.; and Hitchcock, J. P. 1993. Effect clinical field trial in finishing pigs to evaluate the effica-
of subclinical infection with Actinobacillus pleuropneu- cy of a new APP subunit vaccine. Proc Int Congr Pig Vet
moniae in comingled feeder swine. J Am Vet Med Assoc Soc 14:208.
202:1095–1098. White, D. C.; Leidy, G.; Jamieson, J. D.; and Shope, R. E.
Rosendal, S., and Boyd, D. A. 1982. Serotyping of 1964. Porcine contagious pleuropneumonia. III. Interre-
Haemophilus pleuropneumoniae. J Clin Microbiol lationship of Haemophilus pleuropneumoniae to other
16:840–843. species of Haemophilus: Nutritional, metabolic, transfor-
Rosendal, S., and Mitchell, W. R. 1983. Epidemiology of mation and electron microscopy studies. J Exp Med
Haemophilus pleuropneumoniae infection in pigs: A sur- 120:1–12.
vey of Ontario pork producers, 1981. Can J Comp Med Wilson, P. J.; Deneer, H. G.; Potter, A.; and Albritton, W.
47:1–5. 1989. Characterization of a streptomycin-sulfonamide
Rosendal, S.; Boyd, D. A.; and Gilbride, K. A. 1985. Com- resistance plasmid from Actinobacillus pleuropneumoni-
parative virulence of porcine Haemophilus bacteria. Can ae. Antimicrob Agents Chemother 33:235–238.
J Comp Med 49:68–74. Wilson, R. W., and Kierstead, M. 1976. Haemophilus para-
Rycroft, A. N., and Cullen, J. M. 1990. Complement resis- haemolyticus associated with abortion in swine. Can Vet J
tance in Actinobacillus (Haemophilus) pleuropneumoniae 17:222.
Progressive and Nonprogressive
27 Atrophic Rhinitis
M. F. de Jong

A disease condition in pigs referred to as “atrophic rhini- rhinitis (AR) has been actively debated, with intermit-
tis” (or “infectious atrophic rhinitis”) and “chronic at- tent attempts at definition, for well over a century. Since
rophic rhinitis” has been recognized for nearly two cen- the 1930s, observations by Ratke (1938), Thunberg and
turies. A disease of the pig characterized by stunted Carlstrom (1940), and Philips (1946) have implied that
development or total disappearance of the nasal the disease was contagious. Shortly thereafter it was
turbinates (called “turbinate atrophy”) has been recog- demonstrated experimentally that turbinate atrophy was
nized for over 160 years and was first described as transmissible between pigs; when young pigs were inoc-
“Schnüffelkrankheit” in Germany (Franque 1830), where it ulated intranasally with crude material from atrophic
became prevalent in several areas. turbinates, they frequently developed turbinate atrophy
These conditions are now classified as either nonpro- (Jones 1947; MacNabb 1948; Philips et al. 1948; Gwatkin
gressive atrophic rhinitis (NPAR), caused by toxigenic et al. 1949, 1951; Terpstra and Akkermans 1960). Much
Bordetella bronchiseptica, or progressive atrophic rhinitis research has since been directed toward defining the pre-
(PAR), caused by toxigenic Pasteurella multocida, alone or cise microbiological agent(s) responsible. While several
in combination with other agents (e.g., B. bronchiseptica). management and husbandry factors can influence the
The characteristic lesion caused by both agents is a hy- severity and clinical expression of PAR, this disease is
poplasia of the nasal turbinate bones (conchal atrophy); now established primarily as an infectious disease de-
in moderate to severe outbreaks this is accompanied by spite attempts at one time to redefine it as a disorder fun-
degrees of facial distortion (including brachygnathia su- damentally of nutritional origin (Brown et al. 1966).
perior, lateral deviation of the snout, and septum devia- In 1956 Switzer suggested that turbinate atrophy may
tion) and nasal hemorrhage as a result of frequent sneez- be caused by several agents, including trichomonads
ing. Nasal hemorrhage is rare in NPAR but characteristic (Switzer 1951), filter-passing agents (Switzer 1953),
in PAR. viruses (Switzer and L’Ecuyer 1960; Edington et al.
In contrast to NPAR, PAR is of global economic sig- 1976), and mycoplasmas (Switzer 1955; Edington et al.
nificance to swine production, since in PAR, these signs 1976; Gois et al. 1977). Only special AR-toxigenic strains
are accompanied by poor growth of fattening pigs (Ped- of B. bronchiseptica and AR-toxigenic strains of P. multo-
ersen and Barfod 1981). Toxigenic P. multocida is still cida have consistently produced “turbinate atrophy”
spreading in the pig population, especially where disease when sufficient quantities of pure (broth) cultures have
control measurements are carried out ineffectively (Glat- been inoculated intranasally into young susceptible (e.g.,
tleider et al. 1996; Frymus et al. 1996). The PAR toxigenic colostrum-deprived specific-pathogen-free [SPF]) pigs.
P. multocida is also able to cause disease in other species, Despite these observations, however, clinical (and patho-
such as rabbits, goats, sheep, cattle, poultry, and turkeys. logical) disease cannot be attributed solely to infection
Even humans can become infected, and lesions some- with either one or both agents, since these infections can
what comparable to those in swine can result. Poultry, occur in the field in the (temporary) absence of clinical
sheep, and cattle, along with rats, cats, and dogs, are de- disease (the so-called subclinical stage of PAR). Interrup-
scribed as carriers (Avril et al. 1990; Donnio et al. 1991; tions between periods of clinical disease that can vary in
Nielsen and Frederiksen 1990). Toxigenic P. multocida duration from approximately 2 months to about 2 years
should therefore be considered a zoonosis and should be can occur in infected herds. Herd monitoring based on
of concern to governmental and human and veterinary clinical (and/or pathological) features, therefore, cannot
health organizations. guarantee the absence of infectious PAR in a pig herd.
Toxigenic B. bronchiseptica is widespread in swine Complementary monitoring of a herd or connected
production and considered to cause a minor or insignifi- (breeding) herds for over 1 year, based on bacteriologic
cant growth depression in diseased pigs and is therefore and/or serologic detection of the PAR-toxigenic P. multo-
called nonprogressive atrophic rhinitis. cida, may be necessary to obtain sufficient information
The individual and combined importance of both concerning the PAR-infected or PAR-free status of the
agents will be discussed in this chapter. herd.
The precise etiology of the “condition” atrophic

355
356 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

DEFINITION bronchiseptica produce turbinate hypoplasia only in pigs


until about the age of 6 weeks.
At this point it is important to define carefully the condi- The conditions for the growth and/or colonization of
tions described in this chapter. All diseases that cause P. multocida or B. bronchiseptica necessary to produce suf-
turbinate atrophy in swine are called infectious atrophic ficient amounts of toxin are influenced by bacteriologic
rhinitis by farmers. The term “infectious progressive at- and/or virologic damage to the mucosa and by certain
rophic rhinitis” should be reserved for the disease pro- environmental, management, and husbandry factors
duced by one etiologic agent, P. multocida. Pedersen and that create the (multifactorial) disease complex. When
Nielsen (1983) first recommended the use of this term as these factors are all present, very severe clinical PAR can
a result of the deliberations of a European Economic result. Since growth retardation and clinical PAR also oc-
Community commission of specialists on PAR. To ob- cur after parenteral dosing with the toxin of P. multocida,
tain a consensus in a worldwide forum, the proposal was colonization of toxigenic P. multocida in the nose may
repeated by Pedersen and coworkers in 1988. The first not be necessary for the development of the disease.
agreement among specialists in swine diseases from Eu- Tonsils and lungs also have to be considered as locations
rope, North and South America, and Asia was achieved for toxin production (Ackermann et al. 1994).
in 1988 (de Jong and Nielsen 1990). It was agreed to de-
fine PAR as a disease caused by infection with toxigenic Infectious Agents
P. multocida. In herds where suspicious manifestations BORDETELLA BRONCHISEPTICA. B. bronchiseptica is
such as sneezing, nose bleeding, snout deformation, a small, motile, gram-negative rod or coccobacillus mea-
growth retardation, turbinate atrophy, and septum devi- suring approximately 1.0 × 0.3 µm in size. The bacterium
ation are observed and where toxigenic P. multocida is de- is aerobic, does not ferment carbohydrates, utilizes cit-
tected (bacteriologically or serologically), the diagnosis rate, and splits urea.
of PAR can be confirmed. However, the disease may also B. bronchiseptica has been isolated widely from young
develop in or be transmitted by pigs from herds harbor- pigs with rhinitis and from pigs with pneumonia and al-
ing toxigenic P. multocida even though only slight or sub- so from animals in herds showing no clinical signs of res-
clinical disease is present. The advantage of an etiologic piratory disease. It is also a pathogen or potential
definition of PAR rests on the possibility of identifying, pathogen of many other mammals, including dogs, cats,
independently of actual clinical status, those herds that and rats.
are able to transmit or develop the severe clinical disease In the United States during the 1960s, B. bronchisepti-
(Bollwahn 1988). ca was said to be the principal cause of atrophic rhinitis
Estimates of the economic impact of PAR in swine (Switzer and Farrington 1975). After intranasal instilla-
have varied, but in moderate to severe outbreaks it can tion of pure cultures of B. bronchiseptica in colostrum-
be of considerable economic importance (Pedersen and deprived pigs a few days old, Cross and Claflin (1962)
Nielsen 1983; Glattleider et al. 1996). From the clinical were able to produce typical experimental turbinate at-
and pathological points of view, it is probably no longer rophy. This work was repeated by Ross et al. (1967), who
useful to employ the term “atrophic rhinitis” as denoting were able to reproduce turbinate atrophy with pure cul-
a single disease complex. Rather, bordetellosis and toxi- tures of AR-toxigenic B. bronchiseptica in 95% of pigs in-
genic pasteurellosis should be distinguished as NPAR oculated at 1–3 days of age but in only 66% of 4-week-old
and PAR. The infectious agents have completely different pigs. Brassinne et al. (1976) reported that only high num-
economic impacts in swine production and require dif- bers of AR-toxigenic B. bronchiseptica caused turbinate
ferent methods and strategies for prevention and treat- lesions. A toxigenic strain that caused 100% turbinate at-
ment. rophy in 3-week-old colostrum-deprived SPF pigs did not
cause typical atrophy when intranasally instilled during
ETIOLOGY 4 successive days in 6-week-old pigs (de Jong and Akker-
mans 1986). This indicated that between 3 and 6 weeks
Research suggests that toxigenic strains of both B. bron- of age, the sensitivity to a heavy infection with an AR-tox-
chiseptica and P. multocida are the primary infectious eti- igenic B. bronchiseptica strain dropped drastically. Dun-
ologic factors in NPAR and PAR, respectively. The sever- can et al. (1966a) had already stated that experimental
ity of the disease that develops in a pig is related to the infections with B. bronchiseptica did not cause severe pro-
amount of one or of both toxins absorbed by the animal gressive lesions. Pearce and Roe (1966) were unsuccess-
(Van Diemen et al. 1994a). The susceptibility of pigs to a ful in producing turbinate atrophy with cultures of B.
certain amount of toxin that causes turbinate bone re- bronchiseptica in naturally farrowed pigs but were able to
duction has been shown to be age related. Toxigenic produce the lesions when the culture was inoculated into
strains of P. multocida have been shown to produce se- colostrum-deprived pigs. This indicated protection
vere PAR, including extreme growth retardation, even in against AR lesions by colostral immunity.
pigs older than 3 months of age; toxigenic strains of B. Bacteriologic data from nasal swabs taken from pigs
CHAPTER 27 PROGRESSIVE AND NONPROGRESSIVE ATROPHIC RHINITIS de Jong 357

at different ages suggested that B. bronchiseptica infection (Rutter and Rojas 1982). More important, strains isolat-
starts to build up in conventional herds with and without ed from herds with or without progressive disease in the
clinical signs of PAR in the third week of age, when the United Kingdom all caused nonprogressive lesions of
nasal sensitivity for toxigenic B. bronchiseptica has al- similar severity (Rutter and Rojas 1982; Giles and Smith
ready started to decrease (Pedersen and Nielsen 1983; 1983). From observations in this laboratory, it appeared
de Jong 1985). This meant that, under natural (conven- that strains isolated in PAR-diseased herds and in herds
tional) conditions, the influence of B. bronchiseptica as a not suspected of PAR produced roughly the same
primary cause of PAR has been overestimated. Turbinate amount of toxin. Only a few strains differed from this
atrophy can occur in 2- to 3-month-old piglets in excep- pattern (de Jong and Akkermans 1986). Thus, there is
tional circumstances, such as after a primary B. bron- strong evidence, contrary to the opinions of Kielstein
chiseptica infection in SPF herds (Schöss 1982) and after (1983) and Nakai et al. (1986), that although there are
experimental infections in piglets free from antibodies. differences in the virulence of isolates of B. bronchisepti-
Partial or total regeneration of such atrophy has been de- ca, the severe lesions of clinical PAR cannot be attributed
scribed and such infections seem to result in only a limit- only to this organism. Pigs infected with B. bronchiseptica
ed and low percentage of transient clinical snout devia- in the deeper respiratory system may be more sensitive
tions; animals with lesions caused by B. bronchiseptica to other pulmonary infections. This means that B. bron-
did not develop significant growth depression (Pedersen chiseptica infections should not be neglected as a respira-
and Barfod 1982). tory pathogen.
Nearly all B. bronchiseptica strains from pigs pro-
duced the thermolabile AR toxin. B. bronchiseptica can al- PASTEURELLA MULTOCIDA. P. multocida is a non-
so affect the lower respiratory system, and toxin produc- motile, gram-negative rod or coccobacillus approximate-
tion from such regions may have some influence on ly 0.3 × 0.6 µm in size. The bacterium is aerobic, fer-
clinical symptoms of NPAR. ments glucose without gas, and produces indole. In fresh
Variations in virulence among porcine B. bronchisepti- smears the organism shows distinct bipolar staining.
ca strains are known to exist. Collings and Rutter (1985) The colonies of P. multocida type A are larger and more
determined that only those strains in phase 1 and isolat- mucoid than those of type D. On blood agar plates a
ed from pigs caused turbinate atrophy, and they estab- characteristic odor is generally produced.
lished that the ability to both colonize the nasal cavity in P. multocida and its subspecies have been isolated
large numbers and produce a cytotoxin were important widely from pigs with and without clinical symptoms of
virulence determinants. The role of the cytotoxin was rhinitis or pneumonia. Early studies (reviewed by
clearly established by Magyar et al. (1988), who also ex- Gwatkin 1959) established that P. multocida could exper-
amined the role of several other putative virulence deter- imentally produce turbinate atrophy in pigs and rabbits
minants, including a hemolysin, adenylate cyclase, and and that it was frequently but not always isolated from
an adhesin. By comparing the pathogenic effects of a field outbreaks.
porcine cytotoxic phase 1 strain with a noncytotoxic Several workers subsequently examined the ability of
phase 1 strain also of porcine origin, they established this organism to produce turbinate atrophy under con-
that it is the cytotoxin (which is probably the same as the trolled experimental conditions. Some strains studied
mouse lethal factor and dermonecrotic toxin) that is the produced a mild rhinitis but were unable to induce
key virulence determinant in the production of turbinate marked turbinate hypoplasia (Harris and Switzer 1968;
hypoplasia. Smith 1971; Koshimizu et al. 1973; Nakagawa et al. 1974;
In a comparison of three biological assays for the de- Edington et al. 1976), whereas in other studies, particu-
tection of toxigenic properties of B. bronchiseptica, dis- larly from Europe, cultures of P. multocida produced
crepancies were found between the guinea pig skin test, nasal deformity and turbinate atrophy (Dirks et al. 1973)
the mouse spleen atrophy assay, and the suckling mice and even severe PAR (Nielsen et al. 1976). In Germany
mortality assay (Mendoza 1993), suggesting that differ- and in the Netherlands particularly, P. multocida was
ences exist between these toxigenic properties. considered to be an important primary pathogen in PAR
Discrepancies in the results of studies obtained in dif- (Dirks et al. 1973). Medication and vaccination with bor-
ferent countries could arise from variation in virulence or detella vaccines in PAR herds reduced B. bronchiseptica
the amount of toxin produced by the organism con- successfully but failed to affect PAR. In these herds P.
cerned. This has been reported for isolates of B. bron- multocida was found to be the major pathogen. Reducing
chiseptica in the United States (Ross et al. 1967; Skelly et P. multocida in these herds decreased PAR (de Jong 1976-
al. 1980), Canada (Miniats and Johnson 1980), the Unit- 1979, 1980).
ed Kingdom (Collings 1983), and Hungary (Elias et al. A major step in resolving these conflicting opinions
1982). However, even the most virulent of 10 U.K. iso- began in the Netherlands when de Jong (1976-1979) and
lates did not cause progressive turbinate atrophy or sig- de Jong et al. (1980) began to test different P. multocida
nificant snout deformation in experimental infections strains isolated from herds with and without PAR in
358 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

colostrum-deprived SPF piglets, as described earlier by pigs aged 3, 6, 9, 12, and 16 weeks. In pigs infected with
Ross et al. (1967) for B. bronchiseptica. B. bronchiseptica, macroscopic turbinate lesions were no-
P. multocida grows in a semifluid mucus on the mu- ticed only in 3- and 6-week-old pigs, not in pigs of 9
cosal membrane of the nose rather than on the nasal ep- weeks and older. Toxigenic P. multocida still induced typ-
ithelium itself, so studies were redirected from cultures ical snout and turbinate alterations, including septal de-
washed off solid medium and resuspended to broth cul- viation, in pigs infected at 12 and at 16 weeks of age. The
tures. These broth cultures contained substances excret- severity of nose lesions decreased with increasing age
ed by the bacteria. After this change it was easy to repro- with the same dose of toxigenic P. multocida.
duce AR lesions with the same strain in 3-week-old pigs
(or older) instead of in 3-day-old pigs. Other Factors
Martineau et al. (1982) showed the importance of us- Despite the major role of infectious agents, other factors
ing broth cultures instead of cultures washed from solid contribute to the cause or at least the clinical expression
media, and explained that this could be a possible reason of AR, but they have proved difficult to evaluate and have
for the different findings of investigators (Nakai et al. been inadequately defined quantitatively. Most experi-
1986). Pure cultures of both dermonecrotic and nonder- enced clinicians have concluded that the severity of the
monecrotic type D and type A isolates of P. multocida es- disease is markedly influenced by extrinsic factors (Pen-
tablish themselves poorly in the nasal cavity of healthy ny 1977; Goodwin 1980). Smith (1983) provided a useful
conventional (Voets 1990), SPF (de Jong 1985), and gno- review of these noninfectious determinants.
tobiotic piglets (Rutter and Rojas 1982; Rutter 1983).
Nasal instillations of pure broth cultures of toxigenic P. NUTRITION. Although the role of the dietary calci-
multocida needed to be repeated for approximately 4 um:phosphorus imbalance is now discounted as a pri-
days to produce a severe P. multocida nasal infection that mary cause (Brown et al. 1966), nutritional deficiency of
resulted in PAR. Uninoculated pigs kept in contact with any kind may enhance the severity of infectious disease.
inoculated pigs became infected, but only mild lesions Feed consumption may be influenced by AR, since
were noticed 4 weeks later. Sneezing was sporadic in piglets with an acute rhinitis may accept feed less readily
these experiments with gnotobiotic and SPF pigs. In con- and become stunted and weak. Growing pigs with con-
trast, experimental infection with toxigenic P. multocida chal damage may also have reduced feed intake, thus
in conventional pigs pretreated with chemical irritants contributing to the reduced daily gain associated with
or with B. bronchiseptica resulted in sneezing, and PAR le- the condition.
sions also occurred in pigs in contact. The strains that
caused PAR lesions were called AR pathogenic. This GENETIC INFLUENCES. In the past it has been sug-
characteristic correlated with the ability to produce a gested that heredity played a major role, but heritability
thermolabile toxin. PAR could be reproduced complete- estimates have varied greatly, and attempts to control the
ly with bacteria-free filtrates of these unheated toxins. disease solely by genetic selection have failed. There is
Nasal infection by aerosol with 0.5 mL 5 and 13 µg probably a measure of genetically linked predisposition
toxin/mL per nostril in 4-week-old piglets induced sub- to AR, since breeds and strains that vary in susceptibility
clinical ventral turbinate hypoplasia (atrophy). Amounts to the disease and are susceptible to genetic pressure do
of 40 µg/mL induced severe lesions. During the 5-week occur. In the United Kingdom, for example, Large White
period after challenge, depression of weight gain started pigs are now generally considered more vulnerable than
in week 3 after challenge. In the 5, 13, 20, and 40 µg/mL Landrace pigs, although 30 years ago the reverse could
groups, the growth depressions during this period were have been the case. The subject has been reviewed by
32, 54, 52, and 96 grams/day/pig, respectively (Van Smith (1983), Voets et al. (1986a), and Martineau et al.
Diemen et al. 1994a). The severity of atrophy depended (1988).
on the amount of toxin administered into the nose of the
pig. MANAGEMENT, HOUSING, AND ENVIRONMENT.
The first publication explaining the role of toxigenic Severe growth-retarding AR is closely associated with in-
P. multocida in AR was by Ilina and Zasukhin (1975) in tensive methods of production; it is undoubtedly most
Russia and it encouraged the development of tests for se- severe when successive batches of pigs are housed in
lecting toxigenic P. multocida isolates. Instead of using a densely stocked, continuously occupied, poorly ventilat-
rabbit test, the guinea pig skin test was chosen because it ed buildings (Smith and Giles 1980). Penny (1977) has
also selected AR-pathogenic B. bronchiseptica strains identified several management factors that tend to pre-
(de Jong 1980; Blobel and Schliesser 1981; de Jong and dispose to an increased severity of AR (Table 27.1).
Akkermans 1986). Differences between infections with Instances have been observed where the disease was
toxigenic B. bronchiseptica and toxigenic P. multocida controlled, or at least reduced to economically accept-
strains were revealed when pure broth cultures were in- able levels, solely by the manipulation of housing and
stilled intranasally in groups of colostrum-deprived SPF environment and improved management. It is also a
CHAPTER 27 PROGRESSIVE AND NONPROGRESSIVE ATROPHIC RHINITIS de Jong 359

Table 27.1. Management factors influencing the may often be endemic because of infection passing later-
severity of atrophic rhinitis ally between different batches, particularly in systems
Increase Decrease where an all-in/all-out approach is not practiced.
B. bronchiseptica is primarily introduced by the intro-
Large herds, open herds Small herds, closed herds
Expanding herds Static herd size duction of carrier pigs (in SPF herds); recently purchased
High proportion of gilts Mainly old sows breeding stock are often held responsible.
Large farrowing unit Small or single farrowing unit Whether or not the sow is important in transmission,
(all-in/all-out) the recognized infectious agents pass readily between
Multiple suckling—piglets Single suckling
fostered between litters populations of young weaned pigs. Infection of pigs at
Large weaner pools More isolation, modular an early age may be vital, even when the clinical signs ap-
systems (all-in/all-out) pear late in the fattening period.
Large number in one airspace Small number in one airspace The chief mode of transmission of B. bronchiseptica
Frequent moving and mixing Little movement and mixing
from pig to pig is by aerosol droplet infection. The high
Intensive systems indoors Outdoor rearing
High stocking density Low stocking density prevalence of infection among growing pigs suggests
Poor ventilation and no Good ventilation and tempera- that transmission may occur at any age, but it is probably
temperature control ture control more common and more readily accomplished in sus-
Poor hygiene, little disinfection Good hygiene and disinfection ceptible young pigs, in which an active rhinitis with
Continual pig throughput Buildings rested
Dry-feeding, dusty atmosphere Wet-feeding, clean atmosphere sneezing develops. The infection can certainly spread
Mechanical food handling Hand delivery rapidly among populations of susceptible (nonimmune)
piglets (Smith et al. 1982).
Source: After Penny 1977.
B. bronchiseptica colonizes the ciliated mucosa of the
porcine respiratory tract very effectively; it is frequently
common belief that the disease may be more severe in a isolated from the tonsils, and large numbers (106/g) have
dusty atmosphere, particularly where dry and dusty feed been found in the intestinal contents of infected gnoto-
is delivered by automatic equipment. The influences of biotic pigs (Rutter 1985). Thus, direct-contact droplet in-
housing and feed, including the feed delivery system, on fection, and perhaps ingestion of fecal material, are like-
respiratory diseases of the pig have been reviewed by ly to be the main routes of transmission. The cycle of
Owen (1982) and Strang (1982), respectively. infection appears to be maintained by a small proportion
of breeding females. Litters within the farrowing house
EPIDEMIOLOGY become infected at an early age, but in the United King-
dom as well as in the Netherlands, the major spread
Bordetella bronchiseptica seems to occur after 2–3 weeks of age or after weaning,
B. bronchiseptica is widely prevalent in the pig population especially in large groups on flat decks, when 70–80% of
in countries with major swine-producing industries. The a group can become infected. The infection persists for
prevalence of B. bronchiseptica infection greatly exceeds several months, with a gradual reduction in the intensity
that of clinical AR or marked turbinate atrophy at and rate of infection. The age at which animals first be-
slaughter (Cameron et al. 1980), and although B. bron- come infected with B. bronchiseptica has an important ef-
chiseptica is frequently isolated from young pigs in out- fect on the development of lesions. The most severe le-
breaks of AR, the infection also occurs widely in herds sions occur in nonimmune animals infected during the
without the condition (Tornoe et al. 1976; Giles et al. first week of life (Duncan et al. 1966a). Animals infected
1980; Rutter 1981; Whittlestone 1982). at 4 weeks show less-severe lesions, while those infected
The dam has been considered a possible source of the at 9 weeks show virtually no lesions (de Jong and Akker-
important nasal infections for her suckling piglets, and mans 1986).
she has been reliably incriminated as a source of B. bron- The amount and type of immunity also influence the
chiseptica and P. multocida. Transmission also occurs be- epidemiology of bordetella infection. The presence of
tween sows and boars. Although traditionally viewed as passive antibody in the sera of piglets born to naturally
important, some observers have concluded that the infected dams (with toxigenic bordetellae) appeared to
sow’s role might be minor, since clinically NPAR-free provide protection against the development of turbinate
progeny resulted when sows from an affected herd were lesions (Rutter 1981) but not against infection (Kobisch
reared under improved conditions (Bercovich 1978). and Pennings 1989; Voets 1990). However, vaccination of
The degree of resistance to natural B. bronchiseptica sows appeared to delay infection in their piglets until
infection among sows does not appear to be marked, but 12–16 weeks of age compared to nonvaccinated herds, in
younger sows are more likely to be active shedders of the which litters became infected by 2 weeks of age (Rutter
organism. Although infection from the dam is probably et al. 1984).
the chief method of initiating the infection among pop- B. bronchiseptica shedding from vaccinated sows
ulations of suckling piglets, infection in weaner houses seems to be reduced in herds in which the sow popula-
360 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

tion has been vaccinated for some years. ly developed severe clinical signs. This led Braend and
B. bronchiseptica has been isolated from most domes- Flatla (1954) to conclude that “atrophic rhinitis was prac-
tic and wild animal species (Goodnow 1980), and be- tically unknown in Norway prior to [World War II,] after
cause it is a ubiquitous pathogen, there is always the risk which it has been rather common, most certainly be-
that infection could be introduced by nonporcine vec- cause of importation of pigs from Sweden where the dis-
tors. Most isolates from other species appear to be of low ease is common.” It was, therefore, assumed that a new
virulence in pigs, but it is possible that rodents might be- infectious organism had been imported. The disease was
come infected with pig strains and transmit them. Virtu- declared notifiable, and a slaughter policy was carried
ally every pig herd is infected with B. bronchiseptica, and out. This strategy was also followed in other European
variable amounts of brachygnathia superior (BS), mod- countries, such as in the United Kingdom until 1959 and
erately severe turbinate atrophy, and moderate signs of in the Netherlands until 1980. Similarly, the introduc-
septum deviation can be expected in all herds. For this to tion of AR into the United Kingdom was attributed to
occur, pigs from nonimmune dams must become infect- the importation of Swedish stock (Anon. 1954). There
ed within the first 4 weeks of life and develop lesions that are at least two possible explanations for these observa-
persist to slaughter, but in most, regeneration of tions: either B. bronchiseptica was introduced in uninfect-
turbinates begins approximately 4 weeks after the start ed animals and exacerbated existing infections with tox-
of infection, when the scrolls are not totally damaged. igenic P. multocida or, more likely, toxigenic P. multocida
Lesions such as septal deviation, turbinate bone hyper- was introduced with the imported stock.
plasia, and BS may be apparent at slaughter. The per- Estimates of the prevalence of PAR, whether from an
centage of pigs with a twisted snout generally does not individual farm or larger population, are usually con-
reach 1%. ducted by an examination of the heads of pigs after
In the field, however, the picture is likely to be more slaughter. Such surveys have indicated that macroscopic
complicated. For example, there are reports that nasal turbinate atrophy is widespread in pig popula-
Haemophilus parasuis in combined infections with type A tions. It occurred in about 40% of Danish and British
strains of P. multocida can produce mild turbinate lesions herds (Nielsen 1983) in the late 1970s, although a later
(Gois et al. 1983a). Others could not repeat these results. estimate from England showed a decline to 25%
B. bronchiseptica is killed in 30 minutes at 56˚C. It sur- (Cameron et al. 1980). Such a high level of turbinate at-
vives outside the body in dried droplets for up to 5 days rophy, however, does not reflect an equivalent level of
on glass, 3 days on cloth, and a few hours on paper. The clinical disease, since mild lesions are common in com-
survival time of B. bronchiseptica in soil is about 6 weeks. mercial fattening pigs, and mild or even moderate atro-
Low humidity and raised temperatures rapidly reduce phy in individuals may occur in herds without obvious
numbers of live bacteria. In liquid media B. bronchisepti- clinical disease or adverse economic effects. Today it is
ca survived for more than 8 weeks at 21˚C. In lake water believed that these lesions are caused by B. bronchiseptica
and in phosphate buffered saline solution (PBS), B. bron- (NPAR); growth retardation associated with more severe
chiseptica remained viable for at least 3 weeks (Porter et outbreaks has proved difficult to quantify, and some ob-
al. 1991). In a rotating aerosol chamber, the mean half- servers (Straw et al. 1983) have found no correlation be-
life at 21˚C and 76% relative humidity was 118.8 minutes tween the degree of turbinate atrophy and production
(Stehmann et al. 1992), and at 23˚C and 75% relative hu- parameters and, in these herds, now consider atrophy to
midity, it was 56.7 minutes (Müller et al. 1992). The or- result from infection with B. bronchiseptica (NPAR). Nev-
ganism is sensitive to common disinfectants (Stehmann ertheless, where clinically apparent disease occurs, there
et al. 1990). is frequently a reduction in average daily gain; this has
been conservatively estimated as 5–8% for pigs with se-
Pasteurella multocida vere atrophy (Nielsen 1983). In combination with
The introduction of a disease and its subsequent spread pleurisy and pneumonia this reduction can double.
within a population depend on numerous factors, in- The epidemiology of P. multocida infection in pigs is
cluding the nature of the etiologic agent, the host, and less well understood than that of B. bronchiseptica. The
the structure of the population. In intensive pig rearing, organism colonizes the tonsils, but some factor or fac-
many animals are in close contact and there is often tors, the mechanisms of which are not understood, are
widespread movement of breeding and replacement needed to assist colonization of the nasal mucosa. Non-
stock between herds. The main risks of introducing in- toxigenic and toxigenic type A strains can be isolated
fection are associated with purchased pigs, and disease from the lungs of pigs with pneumonia (Baekbo 1988),
may then spread rapidly within a seronegative herd. Ear- but P. multocida is much less effective than B. bronchisep-
ly reports that did not distinguish between PAR and tica in colonizing the trachea. In contrast to type A
NPAR clearly recognized that (P)AR was introduced by strains, toxigenic and nontoxigenic type D strains are iso-
carrier pigs. The disease appeared in many Norwegian lated less frequently in the lungs but more frequently in
herds that had imported pigs, several of which eventual- the nose. P. multocida has also been isolated from most
CHAPTER 27 PROGRESSIVE AND NONPROGRESSIVE ATROPHIC RHINITIS de Jong 361

animal species and is well recognized as an important The prevalence of toxigenic P. multocida may be relat-
pathogen in cattle, rabbits, poultry, and turkeys (Carter ed to the extent of clinical disease. The organism was iso-
1967). In some studies its distribution in pig herds was lated from 50% to 60% of young pigs sampled in a herd in
limited; only 9% were infected in one report (Harris and which almost 30% of fattening pigs had twisted snouts.
Switzer 1969), but such results may be attributable to the In less severely affected herds, larger numbers of young
presence of the commensal flora in the nasal cavity pigs had to be sampled before toxigenic strains were iso-
(Chanter and Rutter 1989). Today in most laboratories, lated (Rutter 1985; de Jong et al. 1988).
selective media and the technique of mouse passage can The distribution of toxigenic P. multocida in other
be used for primary isolation of the organism. Material species has still to be determined. Pedersen (1983),
from herds examined in this way (Rutter 1985) has yield- de Jong (1985), Rutter (1985), Baalsrud (1987), Ohkubo
ed toxigenic and nontoxigenic isolates of type A or D, et al. (1987), Kamp et al. (1990), and Frymus et al. (1996)
and mixed infections with these two types in the same reported that dermonecrotic strains occurred in cattle,
pig can occur. rabbits, dogs, cats, rats, poultry, goats, and sheep. A tox-
In contrast, the distribution of toxigenic isolates of P. igenic strain from pasteurellosis in turkeys produced se-
multocida appears to be limited to those herds with PAR vere turbinate atrophy in gnotobiotic pigs, but a toxi-
or a history of the disease (Rutter 1985). In Germany, the genic strain thought to have been isolated from ovine
Netherlands, and Denmark (Pedersen 1983), the picture pneumonia colonized the nasal cavity of pigs poorly and
is similar, indicating that the majority of herds infected did not produce significant lesions in combined infec-
with toxigenic P. multocida exhibit clinical signs of pro- tion with B. bronchiseptica (Rutter 1983, 1985). Toxigenic
gressive disease. In the Netherlands, however, toxigenic strains were isolated from humans suffering from tonsili-
P. multocida has been isolated from 15% of breeding tis, rhinitis, sinusitis, pleuritis, appendicitis, and sep-
herds with no history or clinical signs of progressive dis- ticemia and were pathogenic for pigs (Nielsen and Fred-
ease at the moment of the first detection of the toxigenic eriksen 1990; Donnio et al. 1991). This implies special
P. multocida. Most of these breeding herds were moni- risks for all persons who have contact with herds or ani-
tored and became clinically diseased within 2 years after mals infected with toxigenic P. multocida: farmers and
this detection. Only 5% of pigs 4–12 weeks of age were their families, farmhands, drivers, merchants, vets, con-
infected in these herds. A few herds remain clinically un- sultants, butchers, and employees of slaughterhouses.
affected for some years (de Jong 1983a; Goodwin et al. Wearing face masks will reduce the risk of becoming in-
1990), which indicates that toxigenic strains may be pres- fected and spreading the infection.
ent in some clinically unaffected herds, and these could The infectious agents are usually introduced into a
transmit progressive disease if infected stock were pur- previously unaffected herd by carrier pigs. Recently pur-
chased from them. chased breeding stock, gilts or boars, are commonly held
The main source of P. multocida infection for young responsible, although the evidence for their involvement
pigs appears to be pharyngeal transport of the organism is often circumstantial. The introduction of toxigenic
among breeding stock; 10–15% of sows in farrowing strains of P. multocida is the principal event preceding an
houses were infected with toxigenic isolates (de Jong outbreak. Poorly colonizing strains with a low toxin pro-
1983b), and some piglets were already infected with duction may represent an exception (Kavanagh 1994).
these strains within a week after birth. Toxigenic P. mul- Infection from other sources is rare but seems to become
tocida was also isolated from the vaginas of a few sows. more important if the spread of toxigenic P. multocida in
The age at which piglets first become infected with P. pigs is not stopped.
multocida affects the severity of the lesions produced, but P. multocida is easily destroyed by 60˚C in 10 minutes,
unlike B. bronchiseptica infection, older pigs may still de- by 0.5% phenol in 15 minutes, and by a 3.5% solution of
velop lesions. Significant turbinate atrophy occurred in cresol in a few minutes. In manure P. multocida remains
pigs infected with toxigenic P. multocida up to 16 weeks of infective for a month and in decomposing or frozen car-
age; Rutter et al. (1984) found that pigs that became nat- casses for 3 months. In a rotating aerosol chamber, the
urally infected between 12 and 16 weeks of age had mean half-life was 20.85 minutes at 23˚C and 75% rela-
turbinate lesions. It has been shown that apparently tive humidity.
healthy 3-month-old pigs can develop PAR when intro- The organism is susceptible to commonly used disin-
duced into a commercial production unit where severe fectants, including those of the following general cate-
disease is occurring (Nielsen et al. 1976). Injection of P. gories: quaternary ammonium compounds, phenolics,
multocida toxin (125 µg/kg) produced significant atro- sodium hypochlorite, iodophores, glutaraldehyde, and
phy in conventional pigs of 10 weeks of age (Rutter chlorhexidine.
1985). With 13 µg/mL instilled nasally, a subclinical PAR Formalin at a concentration of 0.2% or greater and
could be achieved in 4-week-old piglets necropsied 5 phenol at 0.5% will kill in less than 18 hours at 37˚C. On
weeks later. With 40 µg/mL, severe lesions were ob- stock culture agar P. multocida often survives for months
tained in the trials (Van Diemen et al. 1994a). or even years if kept at room temperature, but if stored
362 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

in the refrigerator, the bacteria may die in several days. of the nasal turbinate and is responsible for the osteopa-
Their viability can be maintained for many years in thy. The nature of this toxic factor has received much at-
blood or tissues in the frozen state at −20˚C or lower tention. Cell-free sonicated extracts from phase 1 B. bron-
(Blobel and Schliesser 1981). chiseptica were originally shown to contain a heat-labile
and dermonecrotic toxin, and it was assumed that this
PATHOGENESIS was probably an important factor in the pathogenesis.
Since then, such bacteria-free extracts containing high
Bordetella bronchiseptica levels of this toxin have been repeatedly inoculated in-
It is believed that B. bronchiseptica colonizes the nasal tranasally into piglets, where they produce nasal lesions
cavity by adhering to the nasal mucosa, where it proba- similar to those seen in naturally occurring AR (Hanada
bly preferentially attaches to the ciliated epithelial cells et al. 1979; Nakase et al. 1980; Magyar et al. 1988).
(Yokomizo and Shimizu 1979). This is followed by mul- The degree of severity of the hypoplastic lesions seen
tiplication at the mucosal surface and toxin production, in young pigs varies, and only rarely does severe hy-
leading to inflammatory, proliferative, and degenerative poplasia result (Figs. 27.1–27.3). The ventral scroll of the
changes in the nasal epithelium, including the loss of cil- ventral turbinate is the area most commonly and consis-
ia (Duncan et al. 1966a; Edington et al. 1976). The or- tently affected; grossly it varies in appearance from a
ganism is not believed to invade the deeper tissues. slightly shrunken and distorted scroll to virtually com-
It is assumed that the organism at the mucosal sur- plete absence. In the more severe cases the dorsal scrolls
face elaborates a toxin that diffuses into the osseous core of the ventral turbinate and the dorsal turbinate are also

27.1. Cross section of the snouts


of three uninfected 6-week-old pigs
showing normal anatomy of the
turbinate scrolls (conchae Kontr.
1-2-3). (Courtesy P. Schöss.)

27.2. Turbinate hypoplasia


in three pigs experimentally
infected at 5 and 10 days old
with B. bronchiseptica and
necropsied 40 days post-
infection (Va 6-7-8). (Courtesy
P. Schöss.)

27.3. Cross section of the snouts


of four pigs infected at 5 and 10
days old with B. bronchiseptica
and slaughtered at 6 months of
age. Presumed turbinate regenera-
tion and deformations of the
ventral turbinates and nasal bones
(os vomer) (Va 9-10-11-12).
(Courtesy P. Schöss.)
CHAPTER 27 PROGRESSIVE AND NONPROGRESSIVE ATROPHIC RHINITIS de Jong 363

usually affected. The important factors that affect the fied to a large extent (Becker et al. 1986; Williams et al.
severity of the hypoplasia are the degree of resistance of 1986; Doster et al. 1990; Dugal and Jacques 1990).
the pig to the infection (Smith et al. 1982) and the age P. multocida apparently colonizes the nasal cavity
when it was first acquired, since susceptibility to the poorly unless there is preexisting mucosal damage
damage the infection can produce declines as the pig gets (Elling and Pedersen 1983). Chemical irritants (e.g.,
older. acetic acid and B. bronchiseptica) induce different modifi-
The histologic changes in bordetella-induced hy- cations of the nasal epithelium, but both actions cause
poplastic rhinitis have been reported by Duncan et al. the production of nasal mucus, which results in a nasal
(1966a) and are detailed in Switzer and Farrington environment favorable to colonization by P. multocida
(1975). Briefly, there is a hyperplasia of the epithelium (Gagne and Martineau-Doize 1993). The different types
and, in places, a metaplasia, the epithelium becoming of mucins produced by the piglets’ nasal mucosa at dif-
more stratified in structure with polyhedral cells devoid ferent ages may contribute to an understanding of P.
of cilia. There is a degree of cellular infiltration (princi- multocida and B. bronchiseptica colonization (Martineau-
pally with neutrophils and mononuclear cells), a fibro- Doize et al. 1991a, b).
blastic proliferation in the lamina propria, and a reduc- Given this preconditioning, the organism will set up
tion in size and replacement fibrosis of the osseous core. a nasal infection and, if toxigenic, the toxin will be elab-
In more chronic stages there are increased numbers of orated. The nasal cavity, however, is not necessarily the
osteoblasts around the trabeculae, but osteoclasts are only possible site of toxin production. The toxin appears
rarely found. to be of crucial significance in the pathogenesis of PAR,
There is some variation in the toxigenicity of differ- since only toxigenic strains of P. multocida produce PAR
ent strains of B. bronchiseptica; porcine phase 1 strains lesions; furthermore, the toxin will produce progressive
are more toxigenic than phase 3 or nonporcine isolates snout shortening and turbinate atrophy when given to
(Collings and Rutter 1985). pigs intranasally (Ilina and Zasukhin 1975) and by a va-
riety of parenteral routes (Rutter and Mackenzie 1984).
TURBINATE REGENERATION. There is considerable The precise mechanism of action of the P. multocida
field and experimental evidence that the hypoplasia of toxin has not been clearly defined; but it will produce a
the turbinates produced by the uncomplicated infection variety of changes in the ventral turbinates, consisting of
of young pigs (up to about 8 weeks old) is capable of re- epithelial hyperplasia, atrophy of mucosal glands, in-
generation, which may sometimes be almost complete creasing volume of blood vessels, osteolysis, and a pro-
(Duncan et al. 1966a; Tornoe and Nielsen 1976; Rutter liferation of mesenchymal cells. These eventually will re-
1981; Smith et al. 1982). place the bone trabeculae and osteogenic and
A degree of turbinate hypoplasia in young pigs (Fig. osteoclastic tissues (Rutter and Mackenzie 1984). PAR
27.2), with a variable amount of subsequent regenera- therefore seems to result from a combination of early os-
tion as the pig grows to slaughter weight (Fig. 27.3), may teoblastic damage followed by a series of toxin-induced
thus occur in most herds infected with B. bronchiseptica. chronic changes that result in osteolysis and subsequent
This probably accounts for the high prevalence of mild replacement fibrosis. The toxins of B. bronchiseptica and
lesions of turbinate atrophy seen at slaughter in the P. multocida are different. Also, the ways in which the
many bordetella-infected herds free from obvious clini- turbinates are destroyed differ. The combination of both
cal AR, especially in cases where the nasal cavity remains toxins has detrimental effects on the turbinates and skull
infected with other species, particularly with nontoxi- bones (Martineau-Doize et al. 1990).
genic P. multocida or Haemophilus sp.
A reaction in the regeneration of the scrolls is the ir- CLINICAL SIGNS
regular increased (“hyperplastic”) bone structures in the
turbinate bones and other nose bones as well. Once in- Bordetella bronchiseptica
duced, the BS does not seem to regenerate and is difficult RHINITIS. The principal signs seen in bordetellosis
to separate from breed-associated BS. Only after elimina- are sneezing and snuffling in young pigs. This can occur
tion of B. bronchiseptica in such pig populations can the in piglets as young as 1 week but is frequently seen at 3–4
breed-associated BS be studied properly. weeks of age or about the time of weaning, which may be
related to both maternal colostral protection and the
Pasteurella multocida mixing of pigs at this stage.
The mechanisms of colonization by P. multocida and the Affected piglets sneeze, snuffle, and snort with a vari-
subsequent processes affecting the turbinate bone cells able degree of catarrhal rhinitis producing a variable
and leading to progressive atrophy and clinical PAR have amount of serous or frequently mucopurulent nasal dis-
been partially clarified. Furthermore, the mechanisms by charge, which may be observed by swabbing the nasal
which these chronic nasal changes and their associated cavity. Generally, the younger the piglet when initially af-
malfunctions cause growth retardation have been clari- fected, the more severe the clinical signs. The appetite is
364 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

usually only moderately to slightly impaired. The clinical forceful, violent sneezing. In gilts and sows the hemor-
signs increase in severity for a time, then tend to abate af- rhage in late gestation and farrowing can be life threat-
ter a few weeks, except in herds with clinical PAR, where ening to the dam and her piglets.
continued progressive turbinate damage causes frequent The most characteristic clinical signs of PAR are due
sneezing to continue. Uncomplicated B. bronchiseptica in- to disturbances of normal bone development of the
fections in older pigs produce only mild signs or remain nose; conspicuous deformities of the face may occur. The
clinically inapparent. most common is BS, in which the upper jaw is shortened
Not all sneezing in young pigs is attributable solely to in relation to the lower, as a result of growth depression
B. bronchiseptica, since infection with porcine cy- of the ossa nasales and maxillares, giving the face a
tomegalovirus or other agents may also produce or exac- “pushed-up” appearance. The skin and subcutis over the
erbate these signs. dorsum of the shortened snout are thrown into folds;
when the disturbance of bone growth affects one side of
BRONCHOPNEUMONIA. A more severe manifesta- the face more than the other, lateral deviation of the
tion of infection is bronchopneumonia, which is usually snout occurs (Figs. 27.4 and 27.5). This may vary in
seen as a primary condition in very young piglets (3–4 severity from a barely perceptible misalignment to severe
days). Although this type of disease is relatively uncom- twisting (possibly by as much as 50˚). These facial defor-
mon compared with the wide prevalence of nasal infec- mities reflect an underlying turbinate atrophy; in the
tion, B. bronchiseptica is an important pathogen in those case of lateral deviation the atrophy is more pronounced
cases of pneumonia with bronchitis that occur in young on the side of the deviation. The prevalence of facial dis-
pigs. The condition only affects young pigs and is most tortion varies among outbreaks, and not all pigs with sig-
common in winter (Whittlestone 1982). The major clin- nificant turbinate atrophy develop marked facial distor-
ical sign is coughing, perhaps with whooping and dysp- tion.
nea. Pyrexia is not usually marked (Switzer and Farring- Dirty streaks on the face radiating from the medial
ton 1975). Morbidity is high within litters, and mortality canthus of the eye, caused by tear staining and the en-
may be so in untreated cases. trapment of dust following occlusion of the nasolachry-
B. bronchiseptica is not infrequently isolated from mal duct, are common in PAR outbreaks (Fig. 27.4).
pneumonic lesions in older fattening pigs, but it is con- However, they are not diagnostic and may occur in the
sidered to be a secondary opportunist pathogen, and the absence of PAR.
clinical significance of its presence remains largely un- In moderate to severe herd outbreaks of PAR, the
known. clinical signs are frequently accompanied by growth re-
tardation and reduction in the efficiency of feed utiliza-
Pasteurella multocida tion. Feed utilization is particularly reduced in severely
Clinical signs of PAR are not usually seen in pigs until diseased pigs. The amount of P. multocida toxin may in-
about 4–12 weeks of age or later, depending on the sever- fluence growth performance (Doster et al. 1990; Van
ity of the outbreak, but sneezing and snuffling in baby Diemen et al. 1994a).
pigs are commonly recorded as the first signs. They are Some clinical parameters have been used in an at-
not, however, specific to or diagnostic of the condition, tempt to monitor and quantify disease levels. The preva-
since they frequently occur in the absence of subsequent lence of gross distortion among growing pigs is a crude
clinical PAR. Sneezing and snuffling in baby pigs is mere- measure of disease level but is not a sensitive index of
ly a reflection of an acute catarrhal rhinitis, which may turbinate atrophy. The prevalence and degree of BS in
be due to bordetellosis and/or infection with porcine cy- weaned pigs can provide useful information (Bercovich
tomegalovirus; other agents may possibly be involved, and de Jong 1976) but is not always diagnostic (Schöss
for example, Mycoplasma sp., Actinobacillus sp., and Au- 1983), and confusion can arise with breeds that are natu-
jeszky’s disease, influenza, and porcine reproductive and rally brachygnathic (e.g., Large White) (Van Groenland
respiratory syndrome (PRRS) viruses. In herds where 1984). Sneeze counts have been used successfully in an
subsequent infectious and other factors combine to attempt to assess the effects of treatment (Douglas and
cause progression to clinical AR, affected pigs will con- Ripley 1984; Kobisch and Pennings 1989).
tinue to sneeze, snuffle, and snort throughout the grow-
ing period; this is accompanied by a variable amount of LESIONS OF NPAR AND PAR
serous to mucopurulent nasal discharge. In severely af-
fected animals, sneezing may be pronounced and occa- Gross Lesions
sionally nasal bleeding may occur. Hemorrhage is usual- The gross lesions of PAR are restricted to the nasal cavi-
ly unilateral and varies in severity. Blood may be seen on ty and adjacent structures of the skull, although in severe
the walls of the pen or on the backs of the pigs; muco- cases the pig may also be stunted. At necropsy both BS
purulent material and even pieces of turbinate debris and facial distortion are observed in the intact head. The
may be expelled from the nose following episodes of dominant lesion is an atrophy of the ventral and dorsal
CHAPTER 27 PROGRESSIVE AND NONPROGRESSIVE ATROPHIC RHINITIS de Jong 365

mild to moderate cases the ventral scrolls of the


turbinates are by far the most consistently and severely
affected area; they vary from slightly shrunken to com-
plete atrophy (Figs. 27.7 and 27.8).
In more severe cases, atrophy of the dorsal scrolls of
the ventral turbinate and the dorsal and ethmoidal
turbinates may occur (Figs. 27.9 and 27.10); in the most
severe form there is a complete absence of all turbinate
structures (Fig. 27.11). In between these mild and severe
forms, a whole spectrum of atrophic changes may be ob-
served; occasionally the turbinates are bizarrely shaped
(Fig. 27.10), which may represent some degree of re-
growth of the conchae (Done 1985). Another gross
change that may be observed is bowing or buckling of
the nasal septum (Fig. 27.11); this is not uncommon and
is often associated with BS, facial distortion, and/or
asymmetrical atrophy. Irregular formation of the ossa
nasales and maxillares also occurs in PAR (de Jong 1985)
27.4. A 17-week-old pig with clinical PAR showing and should not be neglected (Figs. 27.10 and 27.11).
marked brachygnathia superior, wrinkling of the skin on Exudate may be found in the nasal cavity but is not a
the dorsum of the nose, and tear staining. constant finding. The amount and character depend on
the age of the lesion and the type of infection. The exu-
date consists of variable amounts of mucopurulent to
purulent material, possibly flecked with blood. The mu-
cosa lining the frontal sinus is sometimes inflamed, and
the sinus itself may contain mucopurulent material. The
bones surrounding the nasal cavity may have undergone
thinning or may be irregularly shaped to some extent.
Hyperplasia/hyperostosis can be observed in older pigs
(de Jong 1985).

ASSESSMENT OF TURBINATE ATROPHY. The vary-


ing severity of the atrophic changes has led to the devel-
opment of methods of quantitative assessment, but un-
fortunately, no one system has gained universal
acceptance. Subjective scoring of snouts (e.g., on a 0–5
scale as in the British system; Anon. 1978) has proved
very useful in evaluating treatment and monitoring
schemes. As well as wide variation between systems,
there is also considerable interobserver variation within
a system (D’Allaire et al. 1988). At least one reason for
this is probably the clinicians’ unwillingness to score a
herd as having PAR, despite a degree of mild atrophy,
when there has never been any clinical evidence of dis-
ease. Today, bacteriologic or serologic tests are necessary
to confirm downgrading of the health status of a herd
27.5. The head of a 15-week-old pig with clinical PAR (e.g., when toxigenic P. multocida is present). The cutoff
showing severe lateral deviation of the snout. The anato- point between normality and being affected is imprecise,
my of the skull is distinctly abnormal due to a failure of and the condition should not be regarded as a simple all-
normal bone development.
or-nothing phenomenon (Done 1985). The situation in
many herds with only mild atrophy probably represents
turbinates, and this can vary greatly in severity. The at- only the effects of a normally transient and often-resolv-
rophy is assessed by a cross section of the snout at the ing hypoplastic rhinitis without the progression charac-
level of the first/second upper premolar, at which point teristic of clinical PAR (see Definition above).
the conchae, dorsal and ventral, are symmetrically and Objective measures of assessing atrophy on a contin-
maximally developed in the normal pig (Fig. 27.6). In uous scale have also been developed, including a mor-
366 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

27.6. Cross section of the snout of an 18-week-old pig showing normal anatomy of the
turbinates.

27.7. Cross section of the snout of an 18-week-old pig. Slight distortion of the ventral scrolls of
the turbinates is present, a common finding.
CHAPTER 27 PROGRESSIVE AND NONPROGRESSIVE ATROPHIC RHINITIS de Jong 367

27.8. Cross section of the snout of an 18-week-old pig showing modest but definite turbinate
atrophy.

27.9. Cross section of the snout of an 18-week-old pig showing severe bilateral turbinate
atrophy.
368 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

27.10. Cross section of the snout of an 18-week-old pig; the atrophic turbinates have developed
into bizarre shapes.

27.11. Cross section of the snout of a 22-week-old pig showing total atrophy of all turbinate
structures, with severe bending of the nasal septum.
CHAPTER 27 PROGRESSIVE AND NONPROGRESSIVE ATROPHIC RHINITIS de Jong 369

phometric index (area of free space as a percentage of to- gen occur. Large alveolar macrophages are present in
tal cross-sectional area of snout section) (Done et al. some alveoli.
1984). Such systems provide parametric data suitable for
analysis, but as diagnostic tools they offer few advan- DIAGNOSIS
tages over subjective snout scoring (Collins et al. 1988,
1989). Bordetella bronchiseptica
Although the clinical signs are suggestive of infection, a
Histologic Changes definitive diagnosis of bordetellosis in pigs is only possi-
Depending on the type of process active at the time of ble by the bacteriologic examination of lung washings,
necropsy, acute, subacute, or chronic histologic changes lungs at postmortem, or nasal secretions. Nasal secre-
may be observed. tions are best collected on cotton-tipped swabs with ei-
The pathognomonic lesion of PAR by toxigenic P. ther metal or elastic plastic stems. Wooden-stemmed
multocida is a fibrous replacement of the bony plates of swabs and swabs with quick-breaking plastic stems
the ventral conchae (Done 1983a; Elling and Pedersen should be avoided, since sudden movement of the pig
1983; Martineau-Doize et al. 1990). Additionally, there may break the shaft. The live pig should be adequately re-
may be a variable metaplasia of the respiratory epitheli- strained and the external nares cleaned. Swabs are care-
um and inflammation of the mucosal lamina propria; fully inserted in a naris with a gentle rotating motion and
subacute cases of rhinitis in conventional pigs will show pushed carefully along the ventral meatus so as to avoid
various mixtures of degenerative, inflammatory, dys- trauma to the delicate turbinates. The swabs are then
trophic, and reparative processes. In SPF pigs infection submitted for laboratory examination, preferably in a
with toxigenic P. multocida does not induce a typical in- bacteriologic transport medium or a phosphate buffered
flammatory reaction but does induce toxic alterations. saline solution (PBS) under cool conditions (±4oC).
The histologic changes in pigs with PAR that show re- The organism grows well on blood agar or Mac-
tarded growth have been described by Yoshikawa and Conkey agar plus 1% glucose, but its isolation from field
Hanada (1981). specimens is often complicated by the overgrowth of
Lesions in parenchymatous organs may also be pres- other organisms (Smith and Baskerville 1979); hence,
ent in cases of severe infection with toxigenic P. multoci- culture on more selective media is recommended. Proce-
da (de Jong 1983a; Rutter 1983). Parenteral injections dures for isolating and identifying the organism from
with P. multocida toxin induced liver cirrhosis, renal fail- field specimens from pigs are given by Farrington and
ure, marked decrease of peripheral blood lymphocytes Switzer (1977), Smith and Baskerville (1979), and Rutter
without lysis, and growth retardation (Becker et al. 1986; (1981). Selective media to isolate both B. bronchiseptica
Williams et al. 1986; Cheville et al. 1988). and P. multocida on the same plate are discussed below
under the detection of P. multocida.
BRONCHOPNEUMONIA. Pneumonic lesions occur The serologic diagnosis of bordetella infection by the
principally in young pigs and have a characteristic scat- detection of agglutinating antibodies in the serum has
tered monolobular or bilobular distribution, mainly in been described. The various methods of antigen prepa-
the apical and cardiac lobes. Affected areas are initially ration, details of some of the tests employed, and their
dark red, become brown, then yellowish brown after a interpretation have been reviewed by Giles and Smith
time and develop a contracted appearance. The lesions (1983). Agglutinating antibodies to B. bronchiseptica are
are associated with the B. bronchiseptica element of the widespread in the pig population, but although their de-
complex. tection by serologic tests can be useful in making a herd
Histologically, the pneumonic lesions in bordetellosis diagnosis of bordetellosis, these are not commonly em-
are characteristic. Detailed descriptions of the ployed for routine diagnostic purposes, since, practically,
histopathology of experimental B. bronchiseptica bron- serologic tests offer few advantages over the culture of
chopneumonia are given by Duncan et al. (1966b) and nasal swabs.
Meyer and Beamer (1973) in conventional and germfree
swine, respectively. The lesions from these experimental Pasteurella multocida
cases are similar to those of field cases. Briefly, the most CLINICAL DIAGNOSIS. When the full range of clin-
severe cases affect the pneumonic vasculature, and there ical signs is present, a preliminary diagnosis of PAR on
are areas of extensive alveolar hemorrhage with necrosis clinical signs alone is possible, but none of the snout de-
and interlobular edema. In areas where hemorrhagic formations by themselves are pathognomonic for PAR.
changes are less extensive, there is an acute inflammato- Animals showing lateral deviation of the snout and/or
ry reaction with cellular infiltration, principally with marked BS, especially at an age of 10–12 weeks, almost
neutrophils. There is an accompanying bronchiolitis always have pronounced turbinate atrophy (Bercovich
with neutrophilic exudate. As the lesions age, vascular and de Jong 1976; de Jong 1985; Kobisch and Pennings
changes become less prominent and epithelialization of 1989). However, when these signs are not apparent or are
the alveoli, fibroblastic activity, and deposition of colla- of decreasing prevalence (e.g., following treatment), it is
370 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

not possible for even experienced observers to assess the CULTURAL AND SEROLOGIC DIAGNOSIS. Today,
extent of turbinate atrophy in the live animal. The pres- a definite diagnosis of PAR cannot be based solely on
ence of a few twisted snouts or sneezing alone is not suf- clinical and pathomorphologic observations but requires
ficient evidence to justify a diagnosis of PAR (see Defini- laboratory tests (Pedersen 1983). Detection of the two
tion). most significant bacterial pathogens is possible by the
culture of nasal and tonsillar swabs or tonsillar biopsies.
RADIOGRAPHIC DIAGNOSIS. Radiographic exami- The live pig should be adequately restrained and the ex-
nation of the snout has been developed to facilitate im- ternal nares cleaned; slender cotton-tipped swabs with
proved diagnosis of turbinate atrophy in the live animal; plastic or metal shafts should be inserted with slight ro-
a suitable procedure is described by Done (1976). In tation deep into both sides of the nasal cavity. Swabbing
some countries this method has enjoyed widespread the tonsillar surface or tonsillar biopsies can aid the iso-
popularity, but it is beset with technical difficulties and lation and differentiation of P. multocida and toxigenic P.
problems in interpretation of the radiographs. The multocida (Van Leengoed et al. 1986; de Jong et al. 1988;
method may not detect mild lesions reliably, and its val- Ackermann et al. 1994). Swabs should be transported to
ue has been questioned (Eikelenboom et al. 1978; Web- the laboratory within 24 hours, preferably in a transport
bon et al. 1978); furthermore, pigs must be sedated, medium under cooled conditions (4–8˚C). Nutrient
anesthetized, or physically immobilized, and the proce- transport media that support the growth of fast-growing
dure is costly and time-consuming. With experience, contaminants are best avoided, but sterile phosphate-
however, some observers consider radiographic exami- buffered saline is suitable (Pedersen 1983).
nation a useful aid (Schöss 1983). The same disadvan-
tages apply to rhinoscopy (Plonait et al. 1980). Modern Detection of P. multocida (and B. bronchiseptica).
methods such as computerized tomography, used as a di- Special selective media are described on which both B.
agnostic tool for PAR, facilitate the macroscopic grading bronchiseptica and P. multocida can grow (de Jong and
of the nasal structures in live pigs of any age (Jolie et al. Borst 1985; de Jong 1994; Moore 1994).
1990). The cultural isolation of P. multocida from nasal
swabs and the testing of their toxigenicity are detailed by
POSTMORTEM DIAGNOSIS. The prevalence and Pedersen (1983). When the nasal cavity is heavily infect-
severity of turbinate atrophy are best estimated by ex- ed with P. multocida, the organism can be recovered on
amination of snouts after slaughter. Snouts should be simple blood agars (Smith and Baskerville 1983). How-
transversely sectioned at the level of the first/second up- ever, field specimens frequently contain low numbers of
per premolar; sectioning cranial to this should be avoid- organisms, and the other nasal flora may mask the pres-
ed, since this will reveal a different pattern of turbinate ence of P. multocida on nonselective media. Mouse inoc-
development. Pigs of 4 weeks old or older that died dur- ulation greatly improves the recovery rate from field
ing weaning or prefattening can present turbinate atro- specimens, but a good in vitro method would be prefer-
phy at an early stage, and cross-sectioning can be carried able. Some selective media are described by Smith and
out with a simple iron saw by qualified local veterinari- Baskerville (1983), Rutter and Luther (1984), de Jong
ans during regular herd inspections. Material from ton- and Borst (1985), Leblanc et al. (1986a, b), Chanter and
sils, lungs, and the nose can be sent to laboratories to en- Rutter (1989), Avril et al. (1990), and de Jong (1994).
sure a proper diagnosis. To make a preliminary herd Evidence suggests that the tonsil is the preferred habitat
diagnosis, pigs have to be examined at slaughter at regu- for P. multocida in the pig, and improved detection rates
lar intervals. As many pigs as practical should be exam- may be achieved by the collection of tonsillar swabs or
ined; between 20 and 30 per time is suggested by Good- biopsies in combination with nasal swabs (de Jong et al.
win (1988). Atrophy is scored on subjective grading 1988). Tonsils and lungs can also be collected in the
systems (Bendixen 1971; Anon. 1978; Done 1983a, b; slaughterhouse and examined in the laboratory. Swabs
de Jong 1985). With low levels it is not possible to define from noses of pigs sampled after immersion in the hot-
a single cutoff point as representing freedom from PAR. water tank are unsatisfactory for the detection of toxi-
An acceptable level for an individual (toxigenic P. multo- genic P. multocida (Chanter and Rutter 1989).
cida–infected) herd is a matter of reasoned clinical judg- When clinical severe PAR is first detected in pigs, in-
ment but must be one in which the economic effects of fection by toxigenic P. multocida actually occurred weeks
the disease are minimal (Goodwin 1988). A monitoring or months earlier. The detection of the toxigenic P. mul-
scheme that could serve as a useful model with a dia- tocida from these pigs can be difficult. Therefore, it is rec-
grammatic representation of snout grading is described ommended to also examine clinically less severely affect-
by Goodwin (1980). Computerized versions have been ed pigs in such groups or pens.
developed by Collins et al. (1988), Barfod et al. (1990),
and Jolie and Thacker (1990). Detection of P. multocida Toxin. The central etio-
logic importance of toxigenic strains of P. multocida is
CHAPTER 27 PROGRESSIVE AND NONPROGRESSIVE ATROPHIC RHINITIS de Jong 371

that classification of field isolates into PAR toxin-positive gen, and antibodies to it take 3 months or longer to de-
or toxin-negative strains is necessary in understanding tect and only then in some pigs (Bording et al. 1990; Van
the epidemiology of the disease. The toxin is thermola- Diemen et al. 1994b). This means that serology may on-
bile, dermonecrotic in the guinea pig and lethal for the ly be of importance in detecting antibodies in the sow
mouse when administered intraperitoneally. All three population. A skin test has been described to detect anti-
tests give broadly comparable results; the methods are bodies in sow herds that have been infected (Schim-
described by de Jong (1980, 1985), Pedersen (1983), and melpfennig and Jahn 1988; Breuer and Schimmelpfennig
Rutter (1983), respectively. An in vitro system of detec- 1990).
tion by assessing the cytopathogenic effect in monolay- In the skin test the purified concentrated toxin is ap-
ers of Vero cells or embryonic bovine lung cells has been plied intradermally in one or different concentrations.
developed (Pennings and Storm 1984; Rutter and Luther Neutralization of the toxin indicates the presence of an-
1984). Today, enzyme-linked immunosorbent assays tibodies as a response to the infection or vaccination
(ELISAs) are replacing the earlier tests (Foged et al. with toxigenic P. multocida. Difficulties in application
1988). DNA probes have been developed to detect the and in the identification of the correct amount of toxin
gene responsible for toxin production in toxigenic P. mul- per dose have limited the use of the skin test in practice.
tocida (Andresen et al. 1990; Kamps et al. 1990; Lax and
Chanter 1990). The use of polymerase chain reaction DIFFERENTIAL DIAGNOSIS. Sneezing in young pigs
(PCR) tests is becoming of interest in diagnosis and in occurs in herds with active PAR, but it is not diagnostic
proposals for eradication of toxigenic P. multocida in itself, since it regularly occurs due to uncomplicated bor-
PAR-diseased herds (Nagai et al. 1994; de Jong 1994; detellosis or porcine cytomegalovirus infection (Rond-
de Jong et al. 1996). huis et al. 1980). Both agents are widely spread in the pig
industry and can cause severe mucous membrane dam-
P. multocida Serotypes. Determination of the cap- age, which is necessary for colonization by toxigenic P.
sular serotype of P. multocida is also often useful for epi- multocida. The frequency of severe sneezing can be used
demiological purposes; most toxin-positive strains are in clinical monitoring of PAR (Kobisch and Pennings
type D, although toxin-positive type A strains also occur. 1989). Influenza, PRRS, and pseudorabies are other
In some regions the toxigenic type D is prevalent, in oth- viruses causing damage to the nasal mucosa.
ers it seems to be type A (Cowart and Backstrom 1984; A variety of other conditions (reviewed by Done
Iwamatsu and Sawada 1988; Pijoan et al. 1988). The usu- 1977) may cause facial deformity in pigs; these are likely
al method of capsular serotyping is the indirect hemag- to cause confusion in clinical and postmortem diagnosis,
glutination test with rabbit antisera (Carter 1955). The since malformations of the turbinates can be observed.
hyaluronidase test (Carter and Rundell 1975) and acri- A localized bacterial infection entering via wounds may
flavine test (Carter and Subronto 1973) are simpler produce a paranasal abscess (bull nose) in young pigs.
methods for the detection of type A and D strains, re-
spectively, but not all porcine isolates are typeable by PROBLEMS WITH DIAGNOSIS. Should pigs from a
these methods (Pedersen 1983). Piliation, hemagglutina- herd with no clinically apparent disease that have no ob-
tion, and capsular serotypes did not show a correlation vious growth retardation but show mild turbinate atro-
with toxin production (Trigo and Pijoan 1988). An atyp- phy at slaughter be considered to be suffering from PAR
ical P. multocida strain producing a toxin similar to the or NPAR? It is suggested that when BS, lateral deviation,
dermonecrotic toxin of P. multocida subsp. multocida has and poor growth are obvious within the herd and atro-
been described in cattle (Kamp et al. 1990). Not only the phy at slaughter is marked, the herd should be suspected
capsule and somatic structure are of epidemiological in- of having clinical PAR. Conversely, where no turbinate
terest; the phage types and plasmid types are also inter- atrophy is seen in slaughtered pigs, the herd must be re-
esting tools with which to follow the distribution pattern garded as clinically free from PAR. However, defining the
of the different toxigenic strains (Lugtenberg et al. 1984; status of herds where mild atrophy occurs in the absence
Nielsen and Rosdahl 1988, 1990; Hoje et al. 1990, Rubies of clinical disease presents problems, since there is no
et al. 1996). satisfactory single cutoff point between affected and un-
affected herds (Goodwin 1988). These low levels of atro-
Serologic Tests. Although agglutinating antibodies phy have been regarded as representing degrees of sub-
to B. bronchiseptica can be detected in pig serum (Giles clinical PAR but are probably better viewed as indicating
and Smith 1983), this is of little diagnostic value. Sero- risk of developing clinical PAR. The level of atrophy
logic tests to detect antibodies against toxigenic P. multo- deemed satisfactory or acceptable for a given enterprise
cida resulting from vaccination or infection have been is often a matter of reasoned clinical judgment. With the
described (de Jong and Akkermans 1986; Bechmann help of bacteriologic and serologic investigations this
and Schöss 1990; Foged et al. 1990; Schimmelpfennig problem can be avoided (see Definition).
1990). The toxin in natural infection is a weak immuno- BS can occur as a breed-associated characteristic in
372 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

certain lines of the Large White/Yorkshire breed. Breed- significant disease, whether rhinitis, bronchopneumo-
associated BS increases with age and cannot be influ- nia, or infectious PAR. The mere presence of the infec-
enced by a medication program intended to combat the tion within an ordinary commercial herd is not, on its
influence of a bordetella and/or a pasteurella infection own, sufficient ground for initiating therapeutic mea-
in these pigs. The breed-associated level of BS can easily sures against it.
be assessed by such a medication method. All BS grading The sulfonamides were the first drugs to be used
higher than this lower genetic level may be a result of successfully in this respect (Switzer 1963) and are still
PAR or NPAR. A warning limit can be chosen (e.g., at an widely employed, either alone or in combination with
age of 8–12 weeks) to allow early selection of clinically antibiotics, or potentiated with trimethoprim. Bron-
diseased animals and to limit further damage in grower chopneumonia in piglets should be treated with par-
or fattening pigs as well as in the younger pigs that will enteral injections of sulfadoxine or sulfadiazine at 12.5
follow such groups. mg/kg with trimethoprim at 2.5 mg/kg daily for 3–5
Breed-associated BS is easily distinguished from PAR days.
by the absence of turbinate atrophy, except where regen- Alternatives to the sulfonamide drugs have been ex-
eration of the turbinates has occurred. amined. Most porcine isolates of B. bronchiseptica appear
Sows and gilts kept in stalls often bite, chew, or play to be sensitive to the tetracyclines (Sisak et al. 1978;
with bars or drinkers, and this can give rise to asymmet- Smith et al. 1980; Pijpers et al. 1989), and these drugs,
ric bone development causing protrusion of the lower particularly a long-acting formulation of oxytetracycline
jaw or mandibular misalignment. These conditions can given by parenteral injection to young pigs, appear to be
be confused with the facial deformity of AR, especially in suitable for the control of bordetellosis.
the older pig, but careful inspection should reveal that The new fluoroquinolones are also active against
the lower jaw is abnormally placed rather than that the porcine B. bronchiseptica (Hannan et al. 1989).
snout is shortened or laterally deviated. A useful tech-
nique is to draw an imaginary line between the center of Antibiotic Resistance
the ears and eyes and project it forward onto the snout. The in vitro activity of 12 sulfonamide drugs against B.
Some sows keep their snouts more to one side. This can bronchiseptica was compared by Mengelers et al. (1989),
cause misinterpretation. By pressing the molar teeth on who showed that the minimum inhibitory concentration
top of each other and comparing the diastema between (MIC)50 ranged from 0.5 to 8 µg/mL. Against a selection
the incisor teeth in the upper and lower jaw, distortion of the pathogenic respiratory bacteria of swine, sul-
can be noticed clinically at an early stage. This method famethoxazole had the highest antimicrobial activity,
can be carried out in combination with the BS-grading and sulfamezathine had an overall low activity. Isolates
method. With thin nasal swabs an increase in internasal of B. bronchiseptica from pigs have been shown to have
space can be observed clinically, but some experience is transmissible R factors that carry antibiotic resistance
necessary for the interpretation. (Terakado et al. 1974).

TREATMENT Medication
SOWS AND PIGLETS. To reduce the prevalence and
The effective treatment of an outbreak of NPAR or PAR severity of nasal infection acquired from dams, the feed
requires a selected combination of management, envi- of the sow can be medicated during the final month of
ronmental, chemotherapeutic, and vaccination proce- gestation. Sulfadimidine (sulfamethazine) (400–2000
dures. No one combination is equally applicable to all af- g/ton) and oxytetracycline (400–1000 g/ton) are the
fected herds. The overall aims of treatment are (1) to products most widely used.
reduce the prevalence and load of the important specific Suckling piglets are best medicated by strategic injec-
bacterial infections (bordetellosis and pasteurellosis) in tions of antibacterial agents in therapeutic dosages four
young pigs by sow vaccination, medication of feed, and to eight times during the first 3–4 weeks of life. The most
antibiotic treatment of piglets; (2) to treat growing pigs useful are potentiated sulfonamides, oxytetracycline,
with an acute rhinitis to reduce the weight of bacterial and penicillin/streptomycin.
infection and severity of the hypoplastic changes and If bordetellosis is the major infection in suckling
maintain efficient growth and feed utilization; and (3) to piglets, potentiated sulfonamides are the drugs of choice
manipulate housing, ventilation, and management to (12.5 mg/kg sulfadiazine or sulfadoxine + 2.5 mg/kg
improve the overall environment for the pigs (de Jong trimethoprim). Injections of oxytetracycline (20–80
and Bartelse 1980; Smith 1983). mg/kg) once or twice a week are also clinically effective
Management and therapeutic measures to control for PAR (de Jong and Oosterwoud 1977; Mefford et al.
bordetellosis in commercial pig herds are required or 1983). The long-acting formulation (20–80 mg/kg) may
usually deemed necessary by experienced clinicians only be the preferred product and is best given once or twice
in herds where this infection is associated with clinically a week during each of the first 3 or 4 weeks of life. If the
CHAPTER 27 PROGRESSIVE AND NONPROGRESSIVE ATROPHIC RHINITIS de Jong 373

organism is not resistant, the drug is effective against (220 g/ton) and sulfamethazine (550 g/ton); lincomycin,
pasteurellosis, since, experimentally, long-acting oxytet- spectinomycin, and amoxicillin trihydrate (10–20
racycline has reduced the prevalence of nasal infection g/ton).
and the severity of turbinate atrophy induced by P. mul- When a number of drugs are used in feed, a decrease
tocida (Gois et al. 1983b); although other researchers of bioavailability can occur, which may result from the
prefer doxycycline (Pijpers et al. 1988). In the Nether- amount of calcium, the feed processing, and the water
lands, intranasal spraying of oxytetracycline in a 5% so- ration given to the treated pigs (Counotte et al. 1984;
lution is used in piglets twice a week, when starting the Froe 1990; Sutter and Wanner 1990). The availability of
treatment. If effective after 2–3 months, a reduction some of these drugs and the regulations regarding their
from twice to once a week can be recommended (de Jong use in food-producing animals vary between countries.
1983b). This withdrawal of medication also depends on Selection of an appropriate antibiotic or combina-
the average antibody titer against the toxin of P. multoci- tion depends partly on cost, legislation, and clinical ex-
da in the dams resulting from the vaccination program. perience but should also be related to the antibiotic sen-
Other antibiotics to which P. multocida may be sensi- sitivity patterns of B. bronchiseptica and P. multocida
tive and that are frequently used in therapeutic concen- isolates and the established MICs (Pijpers et al. 1988;
trations against pneumonia caused by pasteurellosis in- Fales et al. 1990; Awad-Masalmeh et al. 1994). Differ-
clude penicillin/streptomycin (20,000 IU/10–25 ences in MIC between P. multocida type D and type A
mg/kg), tylosin (10–25 mg/kg), lincomycin/spectino- strains may occur in the same herd (Schimmelpfennig
mycin (50/100 mg/kg), ampicillin (10–20 mg/kg), 1990). In a severe outbreak, treatment should be direct-
amoxycillin (10–20 mg/kg), spiramycin (25 mg/kg), ed at pigs of all age groups other than those immediate-
quinolone derivatives (0.5–5 mg/kg), cephalosporins ly destined for slaughter; as the severity declines, reduc-
(1–5 mg/kg), and tiamulin (10–20 mg/kg) (Plonait and ing antibiotic use for the older fatteners should be the
Bickhardt 1988). The benefits of the treatments have not first priority. The appropriate withdrawal times before
been critically evaluated as far as nasal and pulmonary slaughter must always be adhered to. It is usually neces-
protection and/or elimination of the toxigenic P. multo- sary for pigs at risk to receive medicated feed for a mini-
cida are concerned. mum of 4–5 weeks and frequently for longer periods, de-
pending on the results of the vaccination program and
WEANERS AND GROWERS. The PAR in weaned pigs the improvement in housing, ventilation, and manage-
that leads eventually to marked turbinate atrophy at ment.
slaughter can be controlled to some extent by medica-
tion of weaner and/or grower rations or by the addition Vaccination
of antibiotics to the drinking water. Such medication al- SOWS. Vaccination of the sow induces a significant
so assists in the maintenance of growth and efficiency of degree of passive colostral protection against B. bron-
feed utilization in the face of active PAR, but as might be chiseptica in the serum of her suckling piglets
expected, medication is always much more effective (Koshimizu et al. 1973; Smith et al. 1982); in the field,
when the pigs’ environment is improved. Various an- this protection will often persist until about the time of
tibacterial agents alone or in combination are effective. weaning. The colostral protection afforded by sow vacci-
The sulfonamides are frequently included in rations be- nation is thus an effective aid in controlling B. bron-
cause of their known efficacy against bordetellosis. Their chiseptica infections among populations of young suck-
use and the problems of the development of drug resis- ling piglets; in herds where early piglet infection occurs
tance are of great concern. and rhinitis and/or bronchopneumonia develop in
Well-established drugs or combinations suitable for young pigs, sow vaccination should be recommended.
the control of PAR are (1) sulfadimidine (sulfameth- Initially two doses should be given 6 and 2 weeks before
azine) (400–2000 g/ton) in feed or sulfathiazole farrowing, followed by revaccination at 2 weeks before
(0.08–0.13 g/L) in the drinking water; (2) chlortetracy- each subsequent farrowing. Bordetella vaccines which in-
cline (165 g/ton), sulfadimidine (sulfamethazine) (165 duce toxin-neutralizing antibodies and pilus antibodies
g/ton), and penicillin G (83 g/ton) in feed; (3) tylosin are of interest.
(100 g/ton) and sulfadimidine (sulfamethazine) (100 In a prolonged vaccination scheme for gilts and the
g/ton) in feed; (4) carbadox (50 g/ton) and sulfadimi- sow and boar population, the number of B. bronchisepti-
dine (sulfamethazine) (100 g/ton) in feed; (5) oxytetracy- ca carriers is reduced. This in combination with the in-
cline in feed (400 g/ton) or in drinking water (0.18 g/L) creased colostrum protection and all-in/all-out proce-
(Giles 1986). Various other antibacterial agents, alone or dures in the farrowing and weaner sections may be
in combination, also have broadly similar beneficial ef- helpful in producing B. bronchiseptica–free offspring or in
fects on PAR lesions and help to maintain growth. For ex- reducing the B. bronchiseptica colonization in such popu-
ample, the following have been demonstrated as clinical- lations. These procedures also reduced bordetella pneu-
ly effective in feed: lincomycin (220 g/ton); lincomycin monia. In NPAR herds the use of combined bordetella
374 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

and pasteurella vaccines is not recommended, because able, since the main effects of the infection occur in
the antibodies against the pasteurella toxin lead to suspi- younger animals. Specific measures against B. bron-
cion of PAR in such herds. Downgrading the health sta- chiseptica infection should be directed toward preventing
tus could be the result. the infection from arising in suckling pigs (by manage-
Vaccination of the sow with a potent B. bronchiseptica ment, sow vaccination, or chemotherapy) or mitigating
vaccine is an effective way to reduce the prevalence and its effects in young pigs (by management and/or
severity of nasal bordetellosis in suckling and weaned chemotherapy).
piglets (de Jong 1985) but exerts only a limited effect on B. bronchiseptica vaccine has been widely employed.
clinical PAR (Giles and Smith 1983). Pathogenic deter- Although some observers have concluded that it has lit-
minants important in vaccines include the toxigenic tle benefit against clinical PAR and is generally less effec-
characters, the pilus-producing factor, and outer-mem- tive than sow vaccination (Giles and Smith 1982, 1983),
brane proteins. Lack of antibody to some of these prop- in some countries (notably the United States) the proce-
erties seems to influence the reduction of B. bronchisepti- dure nevertheless still enjoys fairly widespread use. B.
ca in sows and piglets. bronchiseptica/P. multocida bacterin vaccines are also
B. bronchiseptica/P. multocida vaccines have been eval- widely employed, but the composition of many of them
uated experimentally and in the field. In some countries means that they may be of little benefit in the field. In a
combined vaccines are available commercially. Such vac- study in which both sows and piglets were inoculated
cines have reduced the prevalence of clinical PAR with a combined vaccine, Mefford et al. (1983) demon-
(Schuller et al. 1980; Baars et al. 1982; de Jong et al. strated that vaccination alone did not influence turbinate
1984) but do not eliminate the condition. As might be atrophy and only marginally improved profitability.
expected, the thermolabile toxin for P. multocida appears Only vaccination of the piglets born of inadequately
to be an important determinant in eliciting protection, vaccinated or unvaccinated dams is of value in the case of
because experimental vaccination of sows with crude P. multocida toxoid vaccines. When sows are properly
toxin significantly protected their offspring against PAR vaccinated and produce good levels of antitoxic antibod-
(Baars et al. 1982, 1986; Pedersen and Barfod 1982). The ies, piglets may not respond to vaccination. If the dams
specific importance of the toxoid fraction has been eluci- show good titers, the colostral protection in pigs can last
dated (Nagy et al. 1986; Foged et al. 1989; Frymus et al. for 3–4 months. Vaccinations of the young breeding
1989; Chanter and Rutter 1990). stock can be started after this age. A high antitoxin titer
The antigens and mechanisms of protection against seems to reduce colonization by toxigenic P. multocida.
toxigenic P. multocida infection and associated disease The additional use of therapeutics (e.g., long-acting
have yet to be fully defined; reports show that toxoid oxytetracycline) in piglets significantly reduced turbinate
preparations of P. multocida produce an antitoxin re- atrophy at slaughter and markedly improved profitabili-
sponse, with an effect on colonization (Chanter and Rut- ty (Pejsak et al. 1990). Furthermore, some commercially
ter 1990). Claims made for B. bronchiseptica vaccines in available vaccines contain neither toxigenic strains nor P.
the control of AR were not fully substantiated in field use multocida toxoid, the manufacturers only claiming effica-
(Giles and Smith 1983); thus, a rush to develop further cy against pneumonic pasteurellosis.
combined vaccines without full appraisal of the required
antigens is undesirable. Housing and Husbandry
Some of the currently available combined B. bron- Medication and vaccination procedures should never be
chiseptica/P. multocida vaccines may be of benefit in con- introduced without concurrent attempts to improve
trolling bordetellosis and toxigenic pasteurellosis and in swine management and husbandry. Although the nonin-
reducing the prevalence and severity of PAR when com- fectious factors that contribute to the severity of PAR are
bined with housing and management changes (de Jong inadequately defined quantitatively, steps should always
et al. 1984). The marked reduction in toxigenic P. multo- be taken to reduce their influence. All-in/all-out systems
cida in the nose following vaccination with a potent tox- are favored for farrowing, weaner, and preferably fatten-
oid vaccine requires further evaluation to determine er management; the age of the sow herd can be allowed
whether the expression of high antibody levels can erad- to rise and the introduction of large numbers of infected
icate toxigenic P. multocida. Vaccination of sows with new gilts can be avoided; stocking density can be re-
combined vaccines can be as effective as piglet medica- duced; strict hygiene measures should be implemented;
tion. However, neither procedure constitutes a means of and correct ventilation rates should be maintained to re-
protection against the condition nor necessarily obviates duce the airborne concentration of bacterial pathogens,
the need for medication. Such a vaccination program, noxious gases, and dust. Steps should also be taken to re-
once started in an infected herd, has to be maintained, at duce factors that stress young pigs, including large tem-
least for many years. perature variations, chilling, and drafts. Replacement
breeding stock should not only be free from clinical signs
PIGLETS. Vaccination of older pigs undoubtedly pro- of disease but also be raised in herds free from infections
duces an active humoral response but its value is debat- with toxigenic P. multocida; introduced weaners should
CHAPTER 27 PROGRESSIVE AND NONPROGRESSIVE ATROPHIC RHINITIS de Jong 375

be free from active rhinitis and the associated sneezing adjuvanted vaccines were at one time widely employed in
and typical BS. Newly purchased stock should originate pig herds because several reports, mainly from the Unit-
from herds free from toxigenic P. multocida, be isolated ed States and Japan (e.g., Nakase et al. 1976; Goodnow
(quarantined), be tested bacteriologically or serological- 1977; Goodnow et al. 1979), indicated that such prod-
ly for freedom from toxigenic P. multocida, and be inte- ucts were highly effective in the control of field out-
grated slowly. Severely affected pigs with obvious and se- breaks of clinical PAR. In Europe, however, these bene-
vere growth retardation should be culled. Vaccination fits were much less obvious (Bercovich and Oosterwoud
programs should be started if breeding stock free from 1977; Pedersen and Barfod 1977; Giles and Smith 1982).
toxigenic P. multocida infection is brought into infected Later, further studies from the United States also con-
herds. Vaccination can reduce colonization by toxigenic cluded that such bordetella vaccines are of limited effica-
P. multocida when infection cannot be avoided. Such an cy in the overall control of PAR. A critical review of bor-
AR vaccination program needs to be carried out continu- detella vaccines (Giles and Smith 1983) concluded that,
ously in the whole sow population of the infected herd. as single antigens, their beneficial effects in the control of
Effective vaccination with potent vaccines in sows alone PAR are indeed limited and the previous claims for their
can also limit the economic damage in weaners. The risk usefulness overstated. In the light of new knowledge,
to breeders, multipliers, and fatteners of becoming in- combined vaccines have been developed consisting of B.
fected by toxigenic P. multocida can be limited by asking bronchiseptica/P. multocida bacterins or B. bronchiseptica
for or giving guarantees that the pigs bought or sold orig- bacterin combined with P. multocida toxoid. Such vac-
inate directly from herds certified free from the organ- cines are of use in the field, provided their limitations are
ism. This certification can be carried out under govern- fully realized by the clinician.
mental legislation. Only by means of such a system of Single bordetella vaccines should be used to limit the
enforced restrictions can the spread of toxigenic P. mul- influence of bordetellosis or NPAR but not in PAR-dis-
tocida via sales or auctions be prevented. eased herds.
PAR can be prevented effectively only by rearing
Depopulation swine free from the specific infections required for dis-
Depopulation and restocking with swine from a source ease to develop. The adoption of an SPF system of pro-
known to be free from toxigenic P. multocida are often the duction and the maintenance of an effective microbio-
only viable solution. Buildings should be thoroughly logic barrier are the only sure ways of achieving this.
cleaned and disinfected, then fumigated and left empty Medicated early weaning (Alexander et al. 1980) may
for 2 weeks to 2 months, depending on the efficacy and well be a viable alternative to the established methods of
quality of the hygiene program. Eradication of rats, producing SPF stock free from toxigenic P. multocida
mice, and birds must be carried out properly and contin- (James 1989; Blaha et al. 1990; Larsen et al. 1990). Tradi-
ually. Replacement pigs should be bought from sources tional vaccination or medication regimes applied to in-
known to be free from PAR and toxigenic P. multocida in- fected herds are not likely to create herds free from the
fection, based on clinical, abattoir, bacteriologic, and/or infections; thus, the infected herds pose a serious threat
serologic monitoring. to free herds in their neighborhood and also pose risks to
other branches of animal production, such as poultry,
PREVENTION rabbits, goats, sheep, and cattle (Nielsen et al. 1986; Fry-
mus et al. 1996). In a preventive disease control pro-
Since B. bronchiseptica is widely prevalent in the pig pop- gram, eradication schemes have to be carried out in all
ulation, its total exclusion from a herd is only possible by types of herds and in all kinds of animals infected with
the development of an SPF system or by medicated and toxigenic P. multocida. B. bronchiseptica appears to be
segregated early-weaning methods and the strict mainte- very widespread in the pig population, but the preva-
nance of an effective barrier. B. bronchiseptica is often one lence of infection with toxigenic strains of P. multocida is
of the first agents to infect an SPF herd. less well defined. A positive correlation exists between
the prevalence of toxigenic strains of P. multocida in a
Vaccination herd and the known occurrence of PAR (de Jong 1983b;
B. bronchiseptica vaccines have been developed and used Nielsen 1983; Pedersen 1983; Cowart and Backstrom
in several countries in an attempt to control both the in- 1984; Leblanc et al. 1986b; Bechmann and Schöss 1988;
fection and the clinical condition AR by immunologic Cowart et al. 1989), although the mere presence of this
methods. Killed whole-culture vaccines with aluminum infection does not always mean clinical disease. The po-
salt adjuvants were the first to be available commercially tential risk of clinical PAR developing in a herd could be
and have been licensed for use in several countries since eliminated simply by ensuring that pigs are free from
the 1970s. Other types of vaccine have also been investi- toxigenic P. multocida infection. Therefore, it is desirable
gated, including live avirulent strains (Krüger and to monitor breeding herds for this pathogen and to take
Horsch 1992) and subunit vaccines, but generally have steps to reduce its dissemination and introduction into
not been used widely in practice. Killed whole-culture unaffected herds. It is definitely beneficial to maintain
376 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

herds with no history of PAR and low scores of atrophy ease-monitoring systems may be effectively employed
at slaughter behind effective barriers or to bring in pigs without laborious clerical work (Collins et al. 1988). Be-
only from sources known to be free from the condition. cause the antibody titer against the toxin of P. multocida
Breeding companies that sell and export pigs from in- correlates with increased protection against turbinate at-
fected herds, inadequately monitored for infectious dis- rophy (Sorensen et al. 1990), serologic monitoring be-
eases like PAR, are involved more and more in financial comes of interest in determining a possible increase in
claims by new clients who will not accept the cost of PAR risk when a decrease in the titers occurs in herds
medication and vaccination and the degradation of the with a sow vaccination program. A program in which on-
health status of their own breeding herds. Because aero- ly sows are vaccinated can also protect the pigs during
genic spread has been described (Baekbo and Nielsen the fattening period. Recent investigations have shown a
1988; Stehmann et al. 1989) and spread of infection may relationship between antitoxin titers and some protec-
be possible from surrounding herds, special attention to tion against colonization by toxigenic pasteurellae
air filtration and decontamination systems (Rutter et al. (Chanter and Rutter 1990). Modern methods for the de-
1986; Voets et al. 1986b) could be necessary if distances tection of toxigenic P. multocida by DNA probes from
become too small (probably within 200–2000 m, de- samples of noses, tonsils, or lungs may soon become use-
pending on the size of the surrounding herds). PAR has ful in monitoring systems (Kamps et al. 1990). Positive
also been found in outdoor systems. Prevention of toxi- results have already been achieved by bacteriologic
genic P. multocida infection in outdoor systems can be (Schöss 1982; Schöss and Thiel 1984; de Jong et al. 1988)
difficult. Prevention by artificial insemination seems or serologic monitoring (de Jong et al. 1988; Bechmann
possible, but some risks exist when the antibiotics used and Schöss 1990; Foged et al. 1990; Schimmelpfennig
in the semen diluter do not eliminate the toxigenic pas- 1990).
teurellae (Overby 1990). Toxigenic P. multocida can be eliminated from infect-
ed breeding farms after intensive vaccination for a period
Monitoring of more than 5 years. These regularly vaccinated sows
Commercial producers should be aware of the current produce high anti–P. multocida toxin titers. During this
disease status of their herds. This applies to herds that period the replacement gilts and boars must be bought
have past or present evidence for the clinical condition, from herds free from toxigenic P. multocida. The replace-
as well as to producers who need to monitor the effects of ments should be introduced into the infected sow popu-
control measures or whose herds have remained free lation only after being vaccinated several times with an
from the condition and who appreciate early warning of atrophic rhinitis toxoid (ART) vaccine. The herd vaccina-
any change in herd status. Hence, ideally, herds need tion program needs to be continued until the last sow
monitoring systems that can quantify not only the pres- from the infected population has left the farm, which
ence but also the effects of PAR (Done 1983b), especially generally takes about 5 years. Methods to shorten such
in the case of an infected herd. Since this disease is not a periods by selecting and slaughtering the toxigenic P.
simple all-or-nothing condition, merely relying on the multocida carriers are being developed with the help of
presence of clinical disease disguises moderate to low the new PCR techniques. First attempts have been par-
levels of infection and recognizes it too late for the intro- tially successful (de Jong 1994; de Jong et al. 1994, 1996;
duction of prophylactic measures. de Jong and Braamskamp 1994).
Parameters that can be usefully measured are indirect Selecting carrier sows in nonvaccinating herds by
production or economic data, such as liveweight gain or bacteriological procedures and by the (DAKO) ELISA
efficiency of feed utilization, and clinical/pathological test or the PCR test and removing them have been de-
data related to PAR, including the amount of sneezing, scribed as successful in creating infection-free sow herds
the incidence of facial distortion, and the two most use- (Alt et al. 1996).
ful criteria, the prevalence and extent of BS in weaned
pigs and the prevalence and severity of turbinate atro- REFERENCES
phy at slaughter. The latter is the most popular in some
countries. In large, modern slaughterhouses the gather- Ackermann, M. R.; De Bey, M. C.; Register, K. B.; Larson, D.
ing of snouts and cross sections is difficult; in such situa- J.; and Kinyon, J. M. 1994. Tonsil and turbinate colo-
tions a scoring system on longitudinally opened snouts nization by toxigenic and non-toxigenic strains of Pas-
can be useful (Visser et al. 1988). Sophisticated methods teurella multocida in conventionally raised Iowa swine.
of monitoring have been developed, one of which is the Proc Int Congr Pig Vet Soc 13:162.
plotting of cumulative-sum charts with decision bound- Alexander, T. J. L.; Thornton, K.; Boon, G.; Lysons, R. J.; and
aries, since it gives early signals of deterioration or im- Gush, A. F. 1980. Medicated early weaning to obtain
provement. Done (1983b) has reviewed the methods of pigs free from pathogens endemic in the herd of origin.
monitoring PAR. Use of computer technology in slaugh- Vet Rec 106:114.
terhouses and on pig farms means that important dis- Alt, M.; Meyer, K.; and Schöss, P. 1996. Eradication of pro-
CHAPTER 27 PROGRESSIVE AND NONPROGRESSIVE ATROPHIC RHINITIS de Jong 377

gressive atrophic rhinitis by bacteriological and serolog- Bercovich, Z., and de Jong, M. F. 1976. Shortening of the up-
ical examination. Proc Int Congr Pig Vet Soc 14:246. per jaw (Brachygnathia superior) as a clinical feature of
Andresen, L. O.; Petersen, S. K.; Christiansen, C.; and atrophic rhinitis in approximately 8-week-old piglets. Ti-
Nielsen, J. P. 1990. Studies on the location of the Pas- jdschr Diergeneeskd 101:1011–1022.
teurella multocida toxin gene, tox A. Proc Int Congr Pig Bercovich, Z., and Oosterwoud, R. A. 1977. Vaccination
Vet Soc 11:60. with Bordetella bronchiseptica vaccine on a farm with at-
Anon. 1954. New pig disease in Britain. Vet Rec 66:316. rophic rhinitis: An evaluation of a field experiment.
———. 1978. Atrophic Rhinitis: A System of Snout Grad- Tijdschr Diergeneeskd 102:485.
ing. Ministry of Agriculture, Fisheries and Food Booklet Blaha, T.; Schimmel, D.; Erler, W.; and Burch, D. G. S. 1990.
LPD 51. Pinner, UK. Use of tiamulin for the production of pigs with healthy
Avril, J. L.; Donnio, P. Y.; and Pouedras, P. 1990. Selective lungs, suitable for research into respiratory diseases.
medium for Pasteurella multocida and its use to detect Proc Int Congr Pig Vet Soc 11:110.
oropharyngeal carriage in pig-breeders. J Clin Microbiol Blobel, H., and Schliesser, T. 1981. Handbuch der bak-
28:1438–1440. teriellen Infektionen bei Tieren. Band III V.E.B. Jena:
Awad-Masalmeh, M.; Kourouma, G.; Köfer, J.; and Schuh, Gustav Fischer Verlag.
M. 1994. Investigation on Pasteurella multocida lesions Bollwahn, W. 1988. Forensische Aspekte der Rhinitis at-
of slaughter swine suffering from chronic respiratory rophicans der Schweine. Tierärztl Prax (Suppl) 3:59–61.
disorders. Proc Int Congr Pig Vet Soc 13:172. Bording, A.; Petersen, S.; and Foged, N. T. 1990. Immuno-
Baalsrud, K. J. 1987. Atrophic rhinitis in goats in Norway. logical and pathological characterization of the Pas-
Vet Rec 121:350–353. teurella multocida toxin and its derivates. Proc Int Congr
Baars, J. C.; de Jong, M. F.; Storm, P. K.; Willems, H.; and Pig Vet Soc 11:62.
Pennings, A. 1982. Atrophic rhinitis and its control with Braend, M., and Flatla, J. L. 1954. Rhinitis infectiosa atrofi-
an adjuvant vaccine consisting of B. bronchiseptica and P. cans hos gris. Nord Vet Med 6:81–122.
multocida strains. Proc Int Congr Pig Vet Soc 7:121. Brassinne, M.; Dewaele, A.; and Gouffaux, M. 1976. In-
Baars, J. C.; Pennings, A.; and Storm, P. K. 1986. Challenge tranasal infection with Bordetella bronchiseptica in gnoto-
and field experiments with an experimental atrophic biotic piglets. Res Vet Sci 20:162–166.
rhinitis vaccine, containing Pasteurella multocida DNT- Breuer, J., and Schimmelpfennig, H. 1990. Skin testing for P.
toxoid and Bordetella bronchiseptica. Proc Int Congr Pig multocida antitoxic antibodies in breeding herds. Proc
Vet Soc 9:247. Int Congr Pig Vet Soc 11:51.
Baekbo, P. 1988. Pathogenic properties of Pasteurella multo- Brown, W. R.; Krook, L.; and Pond, W. G. 1966. Atrophic
cida in the lung of pigs. Proc Int Congr Pig Vet Soc 10:58. rhinitis in swine: Etiology, pathogenesis and prophylax-
Baekbo, P., and Nielsen, J. P. 1988. Airborne Pasteurella mul- is. Cornell Vet (Suppl 1) 56:1–107.
tocida in pig fattening units. Proc Int Congr Pig Vet Soc Cameron, R. D. A.; Giles, C. J.; and Smith, I. M. 1980. The
10:51. prevalence of Bordetella bronchiseptica and turbinate
Barfod, K.; Sorensen, V.; and Nielsen, J. P. 1990. Methods of (conchal) atrophy in English pig herds in 1978–1979.
evaluation of the degree of atrophic rhinitis. Proc Int Vet Rec 107:146.
Congr Pig Vet Soc 11:70. Carter, G. R. 1955. Studies on Pasteurella multocida. I. A
Bechmann, G., and Schöss, P. 1988. Untersuchungen über hemagglutination test for the identification of serologi-
das Vorkommen toxinbildener Pasteurella-multocida- cal types. Am J Vet Res 16:481–484.
Stamme in Tonsillen und Nasen von Ferkeln aus Rhini- ———. 1967. Pasteurellosis: Pasteurella multocida and Pas-
tis-atrophicans-unverdächtigen Zuchtbeständen. Dtsch teurella hemolytica. Adv Vet Sci Comp Med 11:321–379.
Tierärztl Wochenschr 95:257–312. Carter, G. R., and Rundell, S. W. 1975. Identification of type
———. 1990. Neutralizing activity against Pasteurella mul- A strains of P. multocida using staphylococcal
tocida toxin in sera of pigs with atrophic rhinitis. Proc hyaluronidase. Vet Rec 96:343.
Int Congr Pig Vet Soc 11:50. Carter, G. R., and Subronto, P. 1973. Identification of type
Becker, H. N.; Reed, P.; Woodard, J. C.; and White, E. C. D strains of Pasteurella multocida with acriflavine. Am J
1986. The effects of P. multocida type D toxin on Vet Res 34:293.
turbinates, body weight gains, and liver weight in piglets Chanter, N., and Rutter, J. M. 1989. Comparison of methods
when injected intramuscularly. Proc Int Congr Pig Vet for the sampling and isolation of toxigenic Pasteurella
Soc 9:249. multocida from the nasal cavity of pigs. Res Vet Sci
Bendixen, H. C. 1971. Om nysesyge hos svinet. Chronic dys- 47:355–358.
trophic s. Atrophic s. Infectious rhinitis in pigs. Vet Med ———. 1990. The role of osteolytic toxin in colonisation by
(Suppl 1) 23:177. toxigenic P. multocida. Proc Int Congr Pig Vet Soc 11:67.
Bercovich, Z. 1978. Contamination and age as factors in the Cheville, N. F.; Rimler, R. B.; and Thurston, J. R. 1988. A tox-
pathogenesis of atrophic rhinitis. Tijdschr Dierge- in from Pasteurella multocida type D causes acute hepat-
neeskd 103:833. ic necrosis in pigs. Vet Pathol 25:518–520.
378 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Collings, L. A. 1983. The Pathogenicity of Bordetella bron- de Jong, M. F., and Bartelse, A. 1980. The influence of man-
chiseptica in Porcine Atrophic Rhinitis. Ph.D. diss, Univ agement and housing on the isolation frequency of Bor-
Reading, UK. detella bronchiseptica in piglet populations. Proc Int Con-
Collings, L. A., and Rutter, J. M. 1985. Virulence of Bordetel- gr Pig Vet Soc 6:212.
la bronchiseptica in the porcine respiratory tract. J Med de Jong, M. F., and Borst, G. H. A. 1985. Selective medium
Microbiol 19:247. for the isolation of P. multocida and B. bronchiseptica. Vet
Collins, M. T.; Backstrom, L. R.; and Conrad, T. A. 1988. Rec 9:167.
Evaluation of semi-automatic digitizertablet snout mor- de Jong, M. F., and Braamskamp, J. 1994. Atrophic rhini-
phometry technique. Proc Int Congr Vet Soc 10:37. tis–herd monitoring: The investigation of toxigenic Pas-
Collins, M. T.; Backstrom, L.; and Brim, A. 1989. Turbinate teurella multocida from tonsils of culled sows collected in
perimeter ratio as an indicator of conchal atrophy for di- the slaughterhouse. Proc Int Congr Pig Vet Soc 13:161.
agnosis of atrophic rhinitis in pigs. Am J Vet Res de Jong, M. F., and Nielsen, J. P. 1990. Definition of pro-
50:421–424. gressive atrophic rhinitis. Vet Rec 27:93.
Counotte, G. H. M.; Eefting, T.; and Bosch, A. 1984. Stabili- de Jong, M. F., and Oosterwoud, R. A. 1977. Treatment with
ty and distribution of oxytetracycline hydrochloride oxytetracycline hydrochloride in the prevention of at-
during the manufacture and storage of pig-rearing pel- rophic rhinitis in baby pigs. Tijdschr Diergeneeskd
lets under field conditions. Tijdschr Diergeneeskd 102:266.
109(9):339–344. de Jong, M. F.; Oei, H. L.; and Tetenburg, G. J. 1980. AR
Cowart, R. P., and Backstrom, L. 1984. Prevalence of der- pathogenicity tests for Pasteurella multocida isolates.
monecrotic, toxin-producing Pasteurella multocida Proc Int Congr Pig Vet Soc 6:211.
strains in Illinois swine herds with varying levels of at- de Jong, M. F.; Oosterwoud, R. A.; and Bouwkamp, F. R.
rophic rhinitis and pneumonia. Proc Int Congr Pig Vet 1984. A field evaluation of the Nobi-Vac AR vaccine.
Soc 8:159. Proc Int Congr Pig Vet Soc 8:174.
Cowart, R. P.; Backstrom, L.; and Brim, T. A. 1989. Pas- de Jong, M. F.; Wellenberg, G.; Schaake, J.; and Frik, K.
teurella multocida and Bordetella bronchiseptica in atroph- 1988. Selection of pig breeding herds free from atroph-
ic rhinitis and pneumonia in swine. Can J Vet Res ic rhinitis by means of a bacteriological screening of
53(3):295–300. pigs on DNT producing Pasteurella multocida: A field
Cross, R. F., and Claflin, R. M. 1962. Bordetella bronchisepti- evaluation from 1981 until 1987. Proc Int Congr Pig Vet
ca induced porcine atrophic rhinitis. J Am Vet Med As- Soc 10:49.
soc 141:1467–1468. de Jong, M. F.; Kamp, E.; and Bokken, G. 1994. Selecting
D’Allaire, S.; Bigras-Poulin, M.; Paradis, M. A.; and Mar- sows harbouring AR toxigenic Pasteurella multocida by a
tineau, G. P. 1988. Evaluation of AR: Are the results re- PCR-test to eliminate progressive AR in a breeding herd.
peatable? Proc Int Congr Pig Vet Soc 10:38. Proc Int Congr Pig Vet Soc 13:167.
de Jong, M. F. 1976–1979. Atrofische Rhinitis onderzoek. de Jong, M. F.; Kamp, E.; Van Der Schoot, A.; and Von Ban-
Annual Report Central Veterinary Institute, C.D.I. Post- niseth, T. 1996. Elimination of AR toxinogenic pas-
bus 65, Lelystad, Netherlands. teurella from infected sow herds by a combination of
———. 1980. Some aspects of the study of atrophic rhini- ART vaccination and testing sows with a PCR and Elisa
tis in pigs. Tijdschr Diergeneesk 105:711–714. test. Proc Int Congr Pig Vet Soc 13:245.
———. 1983a. Atrophic rhinitis caused by intranasal or in- Dirks, C.; Schöss, P.; and Schimmelpfennig, H. 1973. Aetiol-
tramuscular administration of broth-culture and broth- ogy of atrophic rhinitis of swine. Dtsch Tierärztl
culture filtrates containing AR toxin of Pasteurella multo- Wochenschr 80:342.
cida. In Atrophic Rhinitis of Pigs. Ed. K. B. Pedersen and Done, J. T. 1976. Porcine atrophic rhinitis: Snout radiogra-
J. C. Nielsen. Comm Eur Communities Rep EUR 8643 phy as an aid to the diagnosis and detection of the dis-
EN. Luxembourg, p. 136. ease. Vet Rec 98:23.
———. 1983b. Atrophic Rhinitis of Pigs. Ed. K. B. Pedersen ———. 1977. Facial deformity in pigs. Vet Annu 17:96.
and J. C. Nielsen. Comm Eur Communities Rep EUR ———. 1983a. Atrophic rhinitis: Pathomorphological diag-
8643 EN. Luxembourg. nosis. In Atrophic Rhinitis of Pigs. Ed. K. B. Pedersen
———. 1985. Atrophic Rhinitis in Pigs. Thesis, Univ and J. C. Nielsen. Comm Eur Communities Rep EUR
Utrecht. Elinkwijk B.V. Utrecht. 8643 EN. Luxembourg, p. 3.
———. 1994. [Atrophic rhinitis: Problems of diagnosis.] ———. 1983b. Monitoring of atrophic rhinitis. In Atroph-
Rhinitis atrophicans: Problematik der Diagnostik. Prak- ic Rhinitis of Pigs. Ed. K. B. Pedersen and N. C. Nielsen.
tische Tierarzt 75 (Sondemummer): 82–83. Comm Eur Communities Rep EUR 8643 EN. Luxem-
de Jong, M. F., and Akkermans, J. P. W. M. 1986. I. Atroph- bourg, p. 193.
ic rhinitis caused by Bordetella bronchiseptica and Pas- ———. 1985. Porcine atrophic rhinitis—An update. Vet
teurella multocida and the meaning of a thermolabile tox- Annu 25:180–191.
in of P. multocida. Vet Q 8(3):204–214. Done, J. T.; Upcott, D. H.; Frewin, D. C.; and Hebert, C. N.
CHAPTER 27 PROGRESSIVE AND NONPROGRESSIVE ATROPHIC RHINITIS de Jong 379

1984. Atrophic rhinitis: Snout morphometry for quanti- Foged, N. T.; Nielsen, J. P.; and Barfod, K. 1990. The use of
tative assessment of conchal atrophy. Vet Rec 113:33. ELISA determination of Pasteurella multocida toxin anti-
Donnio, P. Y.; Avril, J. L.; Andre, P. M.; and Vaucel, J. 1991. bodies in the control of progressive atrophic rhinitis.
Dermonecrotic toxin production by strains of Pasteurel- Proc Int Congr Pig Vet Soc 11:49.
la multocida isolated from man. J Med Microbiol Franque. 1830. Was ist die Schnüffelkrankheit der
34:333–337. Schweine? Dtsch Z Gesammte Tierheilkd 1:75.
Doster, A. R.; Frantz, J. C.; Brown, A. L.; Huseman, B. R.; Froe, D. L., II. 1990. Oral tetracyclines: Factors affecting
and Hogg, A. 1990. Effects of Pasteurella multocida serum levels and efficacy. Proc Int Congr Pig Vet Soc
serotype D dermonecrotic toxin in swine. Proc Int Con- 11:353.
gr Pig Vet Soc 11:72. Frymus, T.; Muller, E.; Franz, B. A.; and Petzold, K. 1989.
Douglas, R. G. A., and Ripley, P. H. 1984. Sneeze counts as a Protection by toxoid-induced antibody of gnotobiotic
diagnostic aid in pig production. Vet Rec 114:321. piglets challenged with dermonecrotic toxin of Pas-
Dugal, F., and Jacques, M. 1990. Enhanced adherence of teurella multocida. J Vet Med (B) 36:674–680.
Pasteurella multocida to porcine tracheal rings pre-infect- Frymus, T.; Lésniewsky, S. F.; and Zurawski, A. 1996. Possi-
ed with Bordetella bronchiseptica. Proc Int Congr Pig Vet ble sources of progressive atrophic rhinitis: Toxigenic
Soc 11:73. pasteurella strains in species other than the pig. Proc Int
Duncan, J. R.; Ross, R. K.; Switzer, W. P.; and Ramsey, R. K. Congr Pig Vet Soc 14:247.
1966a. Pathology of experimental Bordetella bronchisep- Gagne, S., and Martineau-Doize, B. 1993. Nasal epithelial
tica infection in swine: Atrophic rhinitis. Am J Vet Res changes induced in piglets by acetic acid and by B. bron-
27:457–466. chiseptica. J Comp Pathol 109:71–81.
Duncan, J. R.; Ramsey, R. K.; and Switzer, W. P. 1966b. Giles, C. J. 1986. Atrophic rhinitis. In Diseases of Swine, 6th
Pathology of experimental Bordetella bronchiseptica in- ed. Ed. A. D. Leman, B. Straw, R. D. Glock, W. L. Men-
fection in swine: Pneumonia. Am J Vet Res 27:467. geling, R. H. C. Penny, and E. Scholl. Ames: Iowa State
Edington, N.; Smith, I. M.; Plowright, W.; and Watt, R. G. Univ Press, pp. 455–469.
1976. Relationship of porcine cytomegalovirus and Bor- Giles, C. J., and Smith, I. M. 1982. The value of vaccinating
detella bronchiseptica to atrophic rhinitis in gnotobiotic pigs with Bordetella bronchiseptica. Pig Vet Soc Proc 9:61.
piglets. Vet Rec 98:42. ———. 1983. Vaccination of pigs with Bordetella bron-
Eikelenboom, G.; Dik, K. J.; and de Jong, M. F. 1978. De chiseptica. Vet Bull 53:327.
waarde van het röntgenologisch onderzoek voor de di- Giles, C. J.; Smith, I. M.; Baskerville, A. J.; and Brothwell, E.
agnostiek van Atrofische Rhinitis. Tijdschr Dierge- 1980. Clinical, bacteriological and epidemiological ob-
neeskd 103:1002–1008. servations on infectious atrophic rhinitis of pigs in
Elias, B.; Kruger, M.; and Ratz, F. 1982. Epizootiologische southern England. Vet Rec 106:25.
Untersuchung der Rhinitis Atrophicans des Schweines. Glattleider, L.; Laval, A.; and Espeisse, O. 1996. Horizontal
II. Biologische Eigenschaften der von Schweinen transmission of atrophic rhinitis. Proc Int Congr Pig Vet
isolierten Bordetella bronchiseptica Stämme. Zentralbl Soc 14:251.
Veterinärmed (B) 29:619–635. Gois, M.; Kuksa, F.; and Sisak, F. 1977. Experimental infec-
Elling, E., and Pedersen, K. B. 1983. Atrophic rhinitis in pigs tion of gnotobiotic piglets with Mycoplasma hyorhinis
induced by a dermonecrotic type A strain of Pasteurella and Bordetella bronchiseptica. Zentralbl Veterinärmed
multocida. In Atrophic Rhinitis of Pigs. Ed. K. B. Peder- 34:89.
sen and N. C. Nielsen. Comm Eur Communities Rep Gois, M.; Barnes, H. J.; and Ross, R. F. 1983a. Potentiation
EUR 8643 EN. Luxembourg, p. 123. of turbinate atrophy in pigs by long-term nasal colo-
Fales, W. H.; Turk, J. R.; Miller, M. A.; Bean-Knudsen, C.; nization with Pasteurella multocida. Am J Vet Res 44:372.
Nelson, S. L.; Morehouse, G. L.; and Gossen, H. S. 1990. Gois, M.; Farrington, D. O.; Barnes, H. J.; and Ross, R. F.
Antimicrobial susceptibility of Pasteurella multocida 1983b. Long acting oxytetracycline for control of in-
type D from Missouri swine. J Vet Diagn Invest 2:80–81. duced Pasteurella multocida rhinitis in swine. J Am Vet
Farrington, D. O., and Switzer, W. P. 1977. Evaluation of Med Assoc 183:445.
nasal culturing procedures for the control of atrophic Goodnow, R. A. 1977. Control of atrophic rhinitis with Bor-
rhinitis caused by Bordetella bronchiseptica in swine. J detella bronchiseptica bacterin. Vet Med Small Anim Clin
Am Vet Med Assoc 170:34. 72:1210.
Foged, N. T.; Nielsen, J. P.; and Schrimer, A. L. 1988. Use of ———. 1980. Biology of Bordetella bronchiseptica. Microbi-
monoclonal antibodies in the diagnosis of atrophic ol Rev 44:722–738.
rhinitis. Proc Int Congr Pig Vet Soc 10:33. Goodnow, R. A.; Shade, F. J.; and Switzer, W. P. 1979. Effica-
Foged, N. T.; Nielsen, J. P.; and Jorsal, S. E. 1989. Protection cy of Bordetella bronchiseptica bacterin in controlling en-
against progressive atrophic rhinitis by vaccination with zootic atrophic rhinitis in swine. Am J Vet Res 40:58.
Pasteurella multocida toxin purified by monoclonal anti- Goodwin, R. F. W. 1980. Atrophic rhinitis of pigs. Vet Rec
bodies. Vet Rec 125:7–11. (Pract Suppl) 2:5.
380 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

———. 1988. Monitoring for atrophic rhinitis: The prob- Kamps, A. M. I. E.; Buys, W. E. C. M.; Kamp, E. M.; and
lem of higher snout scores. Vet Rec 123:566–568. Smits, M. A. 1990. Specificity of DNA probes for the de-
Goodwin, R. F. W.; Chanter, N.; and Rutter, J. M. 1990. De- tection of toxigenic Pasteurella multocida subsp. multoci-
tection and distribution of toxigenic Pasteurella multoci- da strains. J Clin Microbiol 28:1858–1861.
da in pig herds with different degrees of atrophic rhini- Kavanagh, N. T. 1994. Isolation of toxigenic P. multocida
tis. Vet Rec 126:452–456. type D from pigs in a herd free from progressive atroph-
Gwatkin, R. 1959. Rhinitis of swine. XII. Some practical as- ic rhinitis in Ireland. Proc Int Congr Pig Vet Soc 13:121.
pects of the rhinitis complex. Can J Comp Med Vet Sci Kielstein, P. 1983. Zur Bordetelleninfektion des Schweines
23:338. und die Bedeutung von Tiermodellen zum Nachweis
Gwatkin, R.; Plummer, P. J. G.; Byrne, J. L.; and Walker, R. protektiver Eigenschaften von Bordetella bronchiseptica.
V. L. 1949. Rhinitis of swine. III. Transmission to baby Monatsh Vet Med 38:504–509.
pigs. Can J Comp Med Vet Sci 13:15. Kobisch, M., and Pennings, A. 1989. An evaluation in pigs of
———. 1951. Rhinitis of swine. V. Further studies on the Nobi-Vac AR and an experimental atrophic rhinitis vac-
aetiology of infectious atrophic rhinitis. Can J Comp cine containing Pasteurella multocida DNT-toxoid and
Med Vet Sci 15:32. Bordetella bronchiseptica. Vet Rec 124:57–61.
Hanada, M.; Shimoda, K.; Tomita, S.; Nakase, Y.; and Koshimizu, K.; Kodama, Y.; Ogata, M.; Sanbyakuda, S.;
Nishiyama, Y. 1979. Production of lesions similar to nat- Otake, Y.; and Mimura, M. 1973. Studies on the etiology
urally occurring atrophic rhinitis by cell-free sonicated of infectious atrophic rhinitis of swine. V. Experimental
extract of Bordetella bronchiseptica. Jpn J Vet Sci 41:1. Bordetella bronchiseptica infection in conventional
Hannan, P. C. T.; O’Hanlon, P. J.; and Rogers, N. H. 1989. In piglets. Jpn J Vet Sci 35:223.
vitro evaluation of various quinolone antibacterial Krüger, M., and Horsch, F. 1992. Spezifische Stimulation
agents against veterinary mycoplasmas and porcine res- des Respirationstraktes von Ferkeln mit Bordetella bron-
piratory bacterial pathogens. Res Vet Sci 46:202. chiseptica-Lebendimpfstoffen. Mh Vet Med 47:75–78.
Harris, D. L., and Switzer, W. P. 1968. Turbinate atrophy in Larsen, S.; Hogedal Jorgensen, P.; and Nielsen, P. A. 1990.
young pigs exposed to Bordetella bronchiseptica, Pas- Elimination of specific pathogens in 3 to 4 week old
teurella multocida and combined inoculum. Am J Vet Res piglets by use of strategic medication. Proc Int Congr
29:777. Pig Vet Soc 11:387.
———. 1969. Nasal and tracheal resistance of swine Lax, A. J., and Chanter, N. 1990. Cloning of the toxin gene
against reinfection by Bordetella bronchiseptica. Am J Vet from Pasteurella multocida and its role in atrophic rhini-
Res 30:1161–1166. tis. Proc Int Congr Pig Vet Soc 11:61.
Hoje, S.; Norby, K.; and Falk, K. 1990. Plasmid profiles of se- Leblanc, R.; Denicourt, M.; Lariviera, S.; and Martineau, G.
lected strains of Pasteurella multocida from pneumonic P. 1986a. Comparison of isolation methods for the re-
lesions of slaughter weight swine. Proc Int Congr Pig Vet covery of Bordetella bronchiseptica and Pasteurella multo-
Soc 11:104. cida from the nasal cavities of piglets. Proc Int Congr Pig
Ilina, Z. M., and Zasukhin, M. I. 1975. Sib Nauch Rab, Sib Vet Soc 9:226.
Zonal’ Nauchno-Issled Vet Inst, Omsk 25:76 (cited in Vet Leblanc, R.; Lariviera, S.; Martineau, G. P.; and Mittal, K. R.
Bull 1977). 1986b. Characterization of Pasteurella multocida isolated
Iwamatsu, S., and Sawada, T. 1988. Relationship between from the nasal cavities of piglets from farms with or
serotypes, dermonecrotic toxin production of Pasteurel- without atrophic rhinitis. Proc Int Congr Pig Vet Soc
la multocida isolates and pneumonic lesions of porcine 9:225.
lung. Jpn J Vet Sci 50(6):1200–1206. Lugtenberg, B.; Boxtel, R. V.; and de Jong, M. F. 1984. At-
James, A. 1989. The manifestation and attempted eradica- rophic rhinitis in swine, correlation of Pasteurella multo-
tion of atrophic rhinitis in a newly established Duroc cida pathogenicity with membrane protein and
herd. Pig Vet J 23:113–121. lipopolysaccharide patterns. Infect Immun 46:48–54.
Jolie, R., and Thacker, B. 1990. Comparison of atrophic MacNabb, A. L. 1948. Rhinitis. Ont Vet Col Rep.
rhinitis morphometric measurements and macroscopic Magyar, T.; Chanter, N.; Lax, A. J.; Rutter, J. M.; and Hall, G.
grades of nasal cross sections on computerized tomog- A. 1988. The pathogenesis of turbinate atrophy in pigs
raphy scans in pigs. Proc Int Congr Pig Vet Soc 11:53. caused by Bordetella bronchiseptica. Vet Microbiol
Jolie, R.; De Roose, P.; and Tuyttens, N. 1990. Diagnosis of 18:135.
atrophic rhinitis by computerized tomography: A pre- Martineau, G. P.; Broes, A.; de Jong, M. F.; and Martineau-
liminary report. Vet Rec 126:591–594. Doize, B. 1982. Experimental reproduction of atrophic
Jones, T. L. 1947. Rhinitis in swine. Agric Inst Rev 2:274. rhinitis with Pasteurella multocida in gnotobiotic and
Kamp, E. M.; Ter Laak, E. A.; and de Jong, M. F. 1990. Atyp- conventional piglets. Proc Int Congr Pig Vet Soc 7:88.
ical pasteurella strains producing a toxin similar to the Martineau, G. P.; Denicourt, M.; Charette, P.; Lambert, J.;
dermonecrotic toxin of Pasteurella multocida subsp. mul- Desilets, A.; Sauvageau, R.; and Cousineau, G. 1988. Ret-
tocida. Vet Rec 126:434–437. rospective study on atrophic rhinitis and enzootic pneu-
CHAPTER 27 PROGRESSIVE AND NONPROGRESSIVE ATROPHIC RHINITIS de Jong 381

monia found in pigs at Quebec central testing station. Pt Nakai, T.; Kume, K.; Yoshikawa, H.; Oyamada, T.; and
2. Breed effect. Med Vet Quebec 18(4):175–179. Yoshikaw, T. 1986. Changes in the nasal mucosa of spe-
Martineau-Doize, B.; Frantz, J. C.; and Martineau, G. P. cific-pathogen-free neonatal pigs infected with Pasteurel-
1990. Cartilage and bone lesions: An explanation of the la multocida or Bordetella bronchiseptica. Jpn J Vet Sci
severity of conchal atrophy induced by Pasteurella mul- 48:693–701.
tocida type D dermonecrotic toxin. Proc Int Congr Vet Nakase, Y.; Kimiura, M.; and Shimoda, K. 1976. Efficacy of
Soc 11:68. inactivated Bordetella bronchiseptica vaccine for atrophic
Martineau-Doize, B.; Larochelle, R.; and Martineau, G. P. rhinitis under field conditions. Proc Int Congr Pig Vet
1991a. Nasal mucosa of the piglet: Risk factor for at- Soc 4:8.
rophic rhinitis. 23 Journées de la Recherche Porcine en Nakase, Y.; Kume, K.; Shimoda, K.; and Sawata, A. 1980. Ex-
France, Paris, 5, 6, 7, febr. Instituut Technique Porc. perimental atrophic rhinitis produced by cell-free ex-
MNE, pp. 175–178. tract of Bordetella bronchiseptica. Proc Int Congr Pig Vet
Martineau-Doize, B.; Trepanier, H.; Martineau, G. P. 1991b. Soc 6:202.
Histological changes in the nasal ventral conchae of Nielsen, J. P., and Frederiksen, W. 1990. Atrophic rhinitis in
piglets infected with Bordetella bronchiseptica. Can J Vet pigs caused by a human isolate of toxigenic Pasteurella
Res 55:42–49. multocida. Proc Int Congr Pig Vet Soc 11:75.
Mefford, D. E.; Vinson, R. A.; Swafford, W. S.; and Pinkston, Nielsen, J. P., and Rosdahl, V. T. 1988. Phage-typing of toxi-
M. L. 1983. The efficacy of long-acting oxytetracycline genic Pasteurella multocida. Proc Int Congr Pig Vet Soc
and/or Bordetella/Pasteurella bacterin in a swine herd 10:34.
with enzootic atrophic rhinitis. Vet Med Small Anim ———. 1990. Development and epidemiological applica-
Clin 78:1911. tion of a bacteriophage system for typing Pasteurella
Mendoza, S. 1993. Comparison of three biological assays multocida. J Clin Microbiol 28:103–107.
for the detection of toxigenic properties of Bordetella Nielsen, N. C. 1983. Prevalence and economic significance
bronchiseptica associated with atrophic rhinitis. Vet Mi- of atrophic rhinitis. In Atrophic Rhinitis of Pigs. Ed. K.
crobiol 36:215–219. B. Pedersen and N. C. Nielsen. Comm Eur Communities
Mengelers, M. J. B.; Van Klingeren, B.; and Van Miert, A. S. Rep EUR 8643 EN. Luxembourg, p. 35.
P. P. A. M. 1989. In vitro antimicrobial activity of sul- Nielsen, N. C.; Riising, H. J.; and Bille, N. 1976. Experimen-
fonamides against some porcine pathogens. Am J Vet tal reproduction of atrophic rhinitis in pigs reared to
Res 50:1022. slaughter weight. Proc Int Congr Pig Vet Soc 4:202.
Meyer, R. C., and Beamer, P. D. 1973. Bordetella bronchisep- Nielsen, N. C.; Bisgaard, M.; and Pedersen, K. B. 1986. Oc-
tica infections in germ-free swine: An experimental currence of toxin producing strains of Pasteurella multo-
pneumonia. Vet Pathol 10:550. cida in different mammalian and avian species. Proc Int
Miniats, O. P., and Johnson, J. A. 1980. Experimental at- Congr Pig Vet Soc 9:232.
rophic rhinitis in gnotobiotic pigs. Can J Comp Med Ohkubo, Y.; Hiramune, T.; and Kikuchi, N. 1987. Sero-types
44:358. and dermonecrotic activity of Pasteurella multocida iso-
Moore, M. K. 1994. A new selective enrichment procedure lates from nares of rabbits. J College Dairying, Jpn, Nat
for isolating Pasteurella multocida from avian and envi- Sci 12(1):279–285.
ronmental samples. Avian Dis 38:317–324. Overby, E. 1990. Determination of a possible survival rate of
Müller, W.; Schneider, J.; and Von Dossow, A. 1992. Survival Actinobacillus pleuropneumoniae bacteria in an EDTA se-
times of Pasteurella multocida (subsp. multocida) and men dilution experimentally infected. Proc Int Congr
Bordetella bronchiseptica strains in a rotating aerosol Pig Vet Soc 11:45.
chamber—A contribution to the epidemiology of at- Owen, J. E. 1982. The influence of buildings on respiratory
rophic rhinitis. Monatsh Vet Med 47(5):253–256. disease. Pig Vet Soc Proc 9:24.
Nagai, S.; Someno, S.; and Yagihashi, T. 1994. Differentia- Pearce, H. G., and Roe, C. K. 1966. Infectious atrophic rhini-
tion of toxigenic from nontoxigenic isolates of Pasteurel- tis: A review. Can Vet J 7:243.
la multocida by polymerase chain reaction. Proc Int Con- Pedersen, K. B. 1983. Cultural and serological diagnosis of
gr Pig Vet Soc 13:163. atrophic rhinitis in pigs. In Atrophic Rhinitis of Pigs. Ed.
Nagy, L. K.; Mackenzie, T.; and Scarnell, J. 1986. Serum an- K. B. Pedersen and J. C. Nielsen. Comm Eur Communi-
tibody values to Pasteurella multocida type D toxin and ties Rep EUR 8643 EN. Luxembourg, p. 22.
susceptibility of piglets to experimental challenge with Pedersen, K. B., and Barfod, K. 1977. Effect of vaccination of
toxigenic type D of P. multocida. Proc Int Congr Pig Vet sows with Bordetella bronchiseptica on the incidence of
Soc 9:224. atrophic rhinitis in swine. Nord Vet Med 29:369.
Nakagawa, M.; Shimizu, T.; and Motoi, Y. 1974. Pathology ———. 1981. The aetiological significance of Bordetella
of experimental atrophic rhinitis in swine infected with bronchiseptica and Pasteurella multocida in atrophic
Alcaligenes bronchisepticus or Pasteurella multocida. Natl rhinitis of swine. Nord Vet Med 33:513–522.
Inst Anim Health Q (Tokyo) 14:61. ———. 1982. Effect on the incidence of atrophic rhinitis of
382 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

vaccination of sows with a vaccine containing Pasteurel- Rubies, X.; Casal, J.; Fernandez, J.; and Pijoan, C. 1996.
la multocida toxin. Nord Vet Med 34:293. Transmission of Pasteurella multocida clones in a swine
Pedersen, K. B., and Nielsen, N. C., eds. 1983. Atrophic pyramid structure. Proc Int Congr Pig Vet Soc 14:243.
Rhinitis of Pigs. Comm Eur Communities Rep EUR 8643 Rutter, J. M. 1981. Quantitative observations on Bordetella
EN. Luxembourg, p. 205. bronchiseptica infection in atrophic rhinitis of pigs. Vet
Pedersen, K. B.; Nielsen, J. P.; Foged, N. T.; Elling, F.; Rec 108:451–454.
Nielsen, N. C.; and Willeberg, P. 1988. Atrophic rhinitis ———. 1983. Virulence of Pasteurella multocida in atrophic
in pigs: Proposal for a revised definition. Vet Rec 20:190. rhinitis of gnotobiotic pigs infected with Bordetella bron-
Pejsak, Z.; Hogg, A.; Foreman, K.; and Wasinska, B. 1990. chiseptica. Res Vet Sci 34:287.
The effect of terramycin LA in combination with a Bor- ———. 1985. Atrophic rhinitis in swine. Adv Vet Sci Comp
detella/Pasteurella vaccine in controlling atrophic rhini- Med 29:239–279.
tis in swine. Proc Int Congr Pig Vet Soc 11:76. Rutter, J. M., and Luther, P. D. 1984. Cell culture assay for
Pennings, A. M. M. A., and Storm, P. K. 1984. A test in Vero- toxigenic Pasteurella multocida from atrophic rhinitis of
cell monolayers for toxin production by strains of Pas- pigs. Vet Rec 114:393–396.
teurella multocida isolated from pigs suspected of having Rutter, J. M., and Mackenzie, A. 1984. Pathogenesis of at-
atrophic rhinitis. Vet Microbiol 9:503–508. rophic rhinitis in pigs: A new perspective. Vet Rec
Penny, R. H. C. 1977. The influence of management changes 114:89.
on the disease picture in pigs. Vet Annu 17:111. Rutter, J. M., and Rojas, X. 1982. Atrophic rhinitis in gnoto-
Philips, C. E. 1946. Infectious rhinitis in swine (bull nose). biotic piglets: Differences in the pathogenicity of Pas-
Can J Comp Med Vet Sci 10:33. teurella multocida in combined infection with Bordetella
Philips, C. E.; Longfield, H. F.; and Miltimore, J. E. 1948. bronchiseptica. Vet Rec 110:531.
Porcine infectious atrophic rhinitis experiments. Can J Rutter, J. M.; Taylor, R. J.; Crighton, W. G.; Robertson, I. B.;
Comp Med Vet Sci 12:268. and Bentson, A. J. 1984. Epidemiological study of Pas-
Pijoan, C.; Trigo, E.; and Hogg, A. 1988. Atrophic rhinitis in teurella multocida and Bordetella bronchiseptica in atroph-
pigs associated with a toxigenic strain of Pasteurella mul- ic rhinitis. Vet Rec 115:615–619.
tocida serotype A. Proc Int Congr Pig Vet Soc 10:32. Rutter, J. M.; Beard, M.; Carpenter, G. A.; and Fryer, J. T.
Pijpers, A.; Van Klingeren, B.; Schoevers, E. J.; Verheyden, J. 1986. The effect of air filtration on the performance and
H. M.; and Van Miert, A. S. P. 1988. The in vitro activity health of pigs. Proc Int Congr Pig Vet Soc 9:400.
of some antimicrobial agents against 4 porcine respira- Schimmelpfennig, H. 1990. Rhinitis atrophicans (Ra) des
tory tract pathogens. Proc Int Congr Pig Vet Soc 10:98. Schweines: Zur Neutralisation des Toxins von Pasteurel-
Pijpers, A.; Van Klingeren, B.; Schoevers, E. J.; Verheijden, J. la multocida. Dtsch Tierärztl Wochenschr 97:195–196.
H. M.; Van Miert, A. S. J. P. A. M. 1989. In vitro activity Schimmelpfennig, H., and Jahn, B. 1988. Rhinitis atrophi-
of five tetracyclines and some other antimicrobial cans (R.a.) des Schweines: Ein Hauttest zum Nachweis
agents against four porcine respiratory tract pathogens. antitoxischer Pasteurella multocida Antikorper. Dtsch
J Vet Pharmacol Therap 12:267–276. Tierärztl Wochenschr 95:285–286.
Plonait, H., and Bickhardt, K. 1988. Lehrbuch der Schöss, P. 1982. Bordetella bronchiseptica Infektionen in
Schweinekrankheiten. Berlin: Verlag Paul Parey. einem SPF Schweinebestand: Ein Beitrag zur Atiologie
Plonait, H.; Heinel, K. G.; and Bollwahn, W. 1980. Vergleich der Rhinitis atrophicans. Dtsch Tierärztl Wochenschr
von Endoskopie und Rontgenaufnahmen als Hilfsmittel 89:177–181.
zur Diagnose der Rhinitis atrophicans am lebenden ———. 1983. Clinical diagnosis of atrophic rhinitis. In At-
Schwein. Prakt Tierärztl 61:1056–1064. rophic Rhinitis of Pigs. Ed. K. B. Pedersen and N. C.
Porter, J. F.; Parton, R.; and Wardlaw, A. C. 1991. Growth Nielsen. Comm Eur Communities Rep EUR 8643 EN.
and survival of Bordetella bronchiseptica in natural wa- Luxembourg, p. 13.
ters and in buffered saline without added nutrients. Ap- Schöss, P., and Thiel, C. P. 1984. Occurrence of toxin-pro-
pl Environ Microbiol 57:1202–1206. ducing strains of Pasteurella multocida and Bordetella
Ratke, G. 1938. Untersuchungen über die Ursache und das bronchiseptica in pig herds with atrophic rhinitis and in
Wesen der Schuffelkrankheit des Schweines. Archiv unaffected herds. Proc Int Congr Pig Vet Soc 8:94.
Wiss Prakt Tierheild 72:371. Schuller, W.; Trubrich, H.; Kosztolich, O.; Flatscher, J.; and
Rondhuis P. R.; de Jong, M. F.; and Schep, J. 1980. Indirect Jahn, J. 1980. Vaccination against atrophic rhinitis in
fluorescence antibody studies in porcine cy- swine with a combined Bordetella bronchiseptica, Pas-
tomegalovirus infections in the Netherlands. Vet Q teurella multocida vaccine. Zentralbl Veterinärmed
2:65–68. 27:125.
Ross, R. F.; Switzer, W. P.; and Duncan, J. R. 1967. Compar- Sisak, F.; Gois, M.; and Kuksa, F. 1978. The sensitivity of the
ison of pathogenicity of various isolates of Bordetella strains of Bordetella bronchiseptica, Pasteurella multocida
bronchiseptica in young pigs. Can J Comp Med Vet Sci and Mycoplasma hyorhinis, isolated from pigs, to antibi-
31:53–57. otics and chemotherapeutics. Vet Med (Praha) 23:531.
CHAPTER 27 PROGRESSIVE AND NONPROGRESSIVE ATROPHIC RHINITIS de Jong 383

Skelly, B. J.; Pruss, M.; Pellegrino, R.; Andersen, D.; and Vet Med Assoc 123:45.
Abruzzo, G. 1980. Variation in degree of atrophic rhini- ———. 1955. Studies on infectious atrophic rhinitis. IV.
tis with field isolants of Bordetella bronchiseptica. Proc Characterization of a pleuropneumonia-like organism
Int Congr Pig Vet Soc 6:210. isolated from the nasal cavities of swine. Am J Vet Res
Smith, I. M. 1971. Studies on the Role of Some Microor- 16:540.
ganisms in Respiratory Infections of the Pig with Special ———. 1956. Studies on infectious atrophic rhinitis. V.
Reference to the Involvement of Pasteurella septica. Concept that several agents may cause turbinate atro-
Ph.D. diss., Univ London. phy. Am J Vet Res 17:478.
Smith, I. M., and Baskerville, A. J. 1979. A selective medium ———. 1963. Elimination of Bordetella bronchiseptica from
facilitating the isolation and recognition of Bordetella the nasal cavity of swine by sulphonamide therapy. Vet
bronchiseptica in pigs. Res Vet Sci 27:187. Med 58:571.
———. 1983. A selective medium for the isolation of Pas- Switzer, W. P., and Farrington, D. O. 1975. Infectious at-
teurella multocida in nasal specimens from pigs. Br Vet J rophic rhinitis. In Diseases of Swine, 4th ed. Ed. H. W.
139:476. Dunne and A. D. Leman. Ames: Iowa State Univ Press, p.
Smith, I. M., and Giles, C. J. 1980. Vaccines for atrophic 687.
rhinitis. Pig Farming Suppl (Oct.):83. Switzer, W. P., and L’Ecuyer, C. 1960. Detection of swine
Smith, I. M.; Oliphant, J.; Baskerville, A. J.; and Giles, C. J. nasal viruses in cell culture. Am J Vet Res 21:967.
1980. High prevalence of strains of Bordetella bron- Terakado, N.; Azechi, H.; Ninomiya, K.; Fukuyasu, T.; and
chiseptica resistant to potentiated sulphonamide in Eng- Shimizu, T. 1974. Incidence of R. factors in Bordetella
lish pig herds in 1978–1979. Vet Rec 106:462. bronchiseptica isolated from pigs. Jpn J Microbiol 18:45.
Smith, I. M.; Giles, C. J.; and Baskerville, A. J. 1982. The im- Terpstra, J. I., and Akkermans, J. P. W. M. 1960. Enkele aan-
munisation of pigs against experimental infection with tekeningen over Atrofische Rhinitis. Tijdschr Dierge-
Bordetella bronchiseptica. Vet Rec 110:488. neeskd 85:1222–1233.
Smith, W. J. 1983. Infectious atrophic rhinitis: Noninfec- Thunberg, E., and Carlstrom, B. 1940. Om nyssjuka hos svin
tious determinants. In Atrophic Rhinitis of Pigs. Ed. K. fran epizootisynpunkt. Skand Vet 30:711. Can J Comp
B. Pedersen and J. C. Nielsen. Comm Eur Communities Med Vet Sci 10:169.
Rep EUR 8643 EN. Luxembourg, p. 151. Tornoe, N., and Nielsen, N. C. 1976. Inoculation experi-
Sorensen, V.; Barfod, K.; Nielsen, J. P.; and Foged, N. T. ments with Bordetella bronchiseptica strains in SPF pigs.
1990. Effect of degree of atrophy and serum antitoxin Nord Vet Med 28:233.
titer on the daily weight gain and feed conversion. Proc Tornoe, N.; Nielsen, N. C.; and Svendsen, J. 1976. Bordetella
Int Congr Pig Vet Soc 11:57. bronchiseptica isolations from the nasal cavity of pigs in
Stehmann, R.; Mehlhorn, G.; and Neuparth, V. 1989. De- relation to atrophic rhinitis. Nord Vet Med 28:1.
tection of Bordetella bronchiseptica in the air of farrowing Trigo, E., and Pijoan, C. 1988. Effect of piliation, hemagglu-
and weaned piglet pens. Monatsh Vet Med 44:307–311. tination and capsular serotype of Pasteurella multocida
Stehmann, R.; Huster, A.; Mehlhorn, G.; and Neuparth, V. on the production of atrophic rhinitis in swine. Proc Int
1990. Survival and sensitivity to disinfectants of Borde- Congr Pig Vet Soc 10:31.
tella bronchiseptica, Haemophilus parasuis, and Acti- Van Diemen, P. M.; de Jong, M. F.; De Vries Reilingh, G.;
nobacillus pleuropneumoniae. Dtsch Vet Med Van Der Hel, W.; and Schrama, J. W. 1994a. Intranasal
Gesellschaft, pp. 315–320. administration of Pasteurella multocida toxin in a chal-
Stehmann, R.; Rottmayer, J.; Zschaubitz, K.; and Mehlhorn, lenge-exposure model used to induce subclinical signs of
G. 1992. Investigations on the survival of Bordetella atrophic rhinitis in pigs. Am J Vet Res 55:49–54.
bronchiseptica in the air. Zentralbl Veterinärmed (B) Van Diemen, P. M.; De Vries Reilingh, G.; Noordhuizen, J. P.
39:546–552. T. M and Parmentier, H. K. 1994b. Effect of Pasteurella
Strang, M. M. 1982. The influence of feed and feed delivery multocida-toxin on the immune system of piglets. Proc
systems on respiratory disease. Pig Vet Soc Proc 9:36. Int Congr Pig Vet Soc 13:120.
Straw, B. E.; Burgi, E. J.; Hilley, H. D.; and Leman, A. D. Van Groenland, G. J. 1984. Measuring the distance between
1983. Pneumonia and atrophic rhinitis in pigs from a tooth edges in piglets as a way to monitor breeding
test station. J Am Vet Med Assoc 182:607. farms for atrophic rhinitis. Proc Int Congr Vet Soc 8.
Sutter, M. T., and Wanner, M. 1990. Higher bioavailability Van Leengoed, L. A.; Kamp, E..; and Vecht, U. 1986. Tonsil
of tetracyclines given with liquid feed in weaned pigs. biopsy: A tool in epidemiological studies of atrophic
Proc Int Congr Pig Vet Soc 11:353. rhinitis and streptococcal meningitis in pigs. Proc Int
Switzer, W. P. 1951. Atrophic rhinitis and trichomonads. Vet Congr Pig Vet Soc 9:227.
Med 46:478. Visser, I. J. R.; Van den Ingh, T. S. G. A. M.; De Kruijf, J. M.;
———. 1953. Studies on infectious atrophic rhinitis of Tielen, M. J. M.; Urlings, H. A. P.; and Gruys, E. 1988.
swine. I. Isolation of a filterable agent from the nasal Atrofische rhinitis: The use of longitudinal sections of
cavity of swine with infectious atrophic rhinitis. J Am pigs’ heads in the diagnosis of atrophy of the turbinate
384 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

bones at the slaughter line. Tijdschr Diergeneeskd Whittlestone, P. 1982. Infectious agents associated with
113:1345–1355. porcine respiratory diseases. Pig Vet Soc Proc 9:71.
Voets, M. T. 1990. Evaluation of the challenge model to test Williams, P. R.; Hall, R. M.; and Rimler, R. B. 1986. Effect of
AR vaccine. Proc Int Congr Pig Vet Soc 11:56. purified Pasteurella multocida turbinate atrophy toxin on
Voets, M. T.; Tielen, M. J. M.; and Hunneman, W. 1986a. porcine peripheral blood lymphocytes in vivo and in vit-
The heritability of conchal atrophy in a slight AR-infec- ro. Proc Int Congr Pig Vet Soc 9:234.
tion. Proc Int Congr Pig Vet Soc 9:391. Yokomizo, Y., and Shimizu, T. 1979. Adherence of Bordetel-
———. 1986b. UV-ray treatment of air to prevent atrophic la bronchiseptica to swine nasal epithelial cells and its
rhinitis. Proc Int Congr Pig Vet Soc 9:404. possible role in virulence. Res Vet Sci 27:15.
Webbon, P. M.; Penny, R. H. C.; and Gray, J. 1978. Atrophic Yoshikawa, T., and Hanada, T. 1981. Histological studies on
rhinitis. The value of radiography for diagnosis in pigs with atrophic rhinitis showing retarded growth. Jpn
piglets. Br Vet J 134:193. J Vet Sci 43:221.
Brucellosis
28 A. P. MacMillan

Brucellosis of pigs is an infectious disease that has been caused by porcine brucellosis is of proportionately
recognized as a specific entity since 1914, when Traum greater significance than the risk from bovine brucellosis
(1914) isolated the organism from aborted porcine fetus- primarily because B. suis (biovars 1 and 3) appears to
es in Indiana, but for many years it was thought to be have a much higher degree of pathogenicity for humans
caused by an exceptionally pathogenic form of Brucella than B. abortus. There also tend to be higher numbers of
abortus until Huddleston (1929) named the infectious B. suis organisms in the tissues, providing a greater expo-
agent, B. suis, as a separate species. Brucellosis occurs in sure to persons who come in contact with infected pigs.
most countries throughout the world where pigs exist in As pigs do not produce dairy products, the incidence of
the wild or domesticated state. B. suis in humans is almost entirely occupational: in
In the United States porcine brucellosis was recog- farmers, veterinarians, and abattoir workers. Interest-
nized as a major disease, causing considerable economic ingly, although the infection of cattle with B. suis is rare,
loss during the 1920s-1950s. Since that time, changes in Cook and Noble (1984), working in Australia, reported
management combined with regulatory programs to several cases, probably contracted following contact with
eradicate the disease have gradually eliminated brucel- feral pigs. Persistent excretion in the milk may give rise
losis as a major disease problem from large areas of the to human epidemics (Borts et al. 1943). B. suis infection
country. Since 1989 all states have become participants of a ram has been reported by Paolicchi et al. (1993),
in the federal eradication program, and regions where who suggest that this may represent a public health haz-
the majority of pigs are raised appear to be virtually free ard.
of brucellosis. Pigs in the southeastern United States
seem to have the highest incidence of brucellosis, al- ETIOLOGY
though the herd infection rate there is less than 1.5%. Ac-
cording to USDA statistical information (Frye et al. The genus Brucella comprises six nomen species: B. abor-
1993), 0.5% of 1.6 million pigs tested on farms, at live- tus, B. melitensis, B. suis, B. neotomae, B. ovis, and B. canis
stock markets, and at slaughter establishments during (Brinley-Morgan and McCullough 1974; Alton et al.
fiscal year 1993 were serologic reactors. This information 1975). The genus appears to be genetically very homoge-
indicates a higher incidence than actually exists, since neous (Verger et al. 1985) but does not appear to be
single serologic reactors in noninfected herds are a fre- closely related to any other animal pathogens (de Ley et
quent occurrence. The actual number of herds classified al. 1987). The first three nomen species are further di-
as infected in 1993 was 83, and these were for the most vided into 8, 3, and 5 biovars, respectively. The principal
part small herds in endemic areas. hosts for B. melitensis are goats and sheep; for B. abortus,
The disease appears to be widespread in South Amer- cattle; for B. neotomae, desert wood rats; for B. ovis, sheep;
ica, where it is predominantly caused by biovar 1. In Eu- and for B. canis, dogs. The most common host for B. suis
rope (apart from Britain and Scandinavia, which are bru- biovars 1 and 3 is the pig, and these biovars are world-
cellosis free), there is a general low prevalence of porcine wide in distribution. B. suis biovar 2 occurs in Europe,
brucellosis. In Africa, the disease is reported by some where the hosts are pigs and the European hare (Lepus
countries, but the number of pigs on the continent is not capinensis), which can form a reservoir for occasional
large, and the true position is not entirely clear. Asia, par- outbreaks in both wild and domestic pigs. The disease in
ticularly Southeast Asia, seems to have a generally high pigs caused by biovar 2 differs slightly from that caused
prevalence of the disease, predominantly caused by bio- by biovars 1 and 3 in that miliary brucellosis of the
var 3 in South China and Singapore and by biovar 1 else- uterus is a feature, and unlike them, it does not appear to
where. In Australia the disease is confined to feral pigs in be pathogenic for humans. B. suis biovar 4 is enzootic in
Queensland (Alton 1990). reindeer and caribou (Rangifer spp.) in Siberia, Alaska,
Porcine brucellosis also has noteworthy public health and Canada and is apparently not pathogenic for pigs al-
implications. Until recently the source of the majority of though it causes many cases of human brucellosis. B. su-
human brucellosis has been Brucella suis–infected pigs is biovar 5 causes murine brucellosis.
(Fox and Kaufmann 1977). The public health hazard

385
386 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

B. suis is the only recognized Brucella species that frequently enhances the growth of brucellae, particular-
causes systemic or generalized infection leading to re- ly on primary isolation. Increased carbon dioxide ten-
productive failure in pigs. Pigs can be infected naturally sion does not enhance the growth of B. suis. A more com-
or experimentally with other Brucella species, but a char- plete discussion of biotyping procedures, biovars, and
acteristic of the infection is almost invariably a symp- formulation of growth media, as well as descriptive char-
tomless, self-limiting localized infection of lymph nodes acteristics of the entire genus, can be obtained from Al-
regional to the point of entry. It should be noted, howev- ton et al. 1988.
er, that the differentiation of biovars of Brucella must be In general, all biovars of B. suis have a noticeably
accomplished using methods only available in large ref- greater urease and catalase activity than other species of
erence centers, and this probably accounts for the fre- Brucella. Classification of B. suis into biovars is based on
quent reports before the early 1960s of the isolation of B. the combined findings of a variety of conventional and
melitensis from pigs (Alton 1990). specialized tests. Briefly, B. suis biovar 1 produces large
Bacteriological examination is often of great assis- amounts of hydrogen sulfide, whereas biovars 2, 3, 4,
tance in aiding a diagnosis, but it must be remembered and 5 produce little or none; growth of biovars 1, 2, and
that handling B. suis in the laboratory is extremely haz- 5 is inhibited to a greater degree by basic fuchsin than
ardous unless appropriate precautions are taken. The growth of biovars 3 and 4; B. suis is not lysed by routine
genus Brucella belongs to Hazard Group III and should test dilutions (RTD) of Tbilisi Brucella phage but may be
be handled in a Class I/III safety cabinet within Contain- partially lysed by 10,000 × RTD; using monospecific an-
ment Level III accommodation by staff with adequate tisera for the dominant A and M antigens, B. suis biovars
training and experience. 1, 2, and 3 are A dominant, while biovar 4 is AM (the on-
Primary isolations of B. suis, like the other species in ly distinguishing feature separating biovars 3 and 4), and
the genus, appear as small, convex, translucent colonies biovar 5 is M dominant (Table 28.1).
on the surface of agar media after incubation at 37˚C for As the oxidative metabolic characteristics of all B. su-
2–7 days. All Brucella species and biovars, except B. ovis is biovars are very similar, there are insufficient differ-
and B. canis, occur naturally with smooth colonial mor- ences for differentiation.
phology. B. ovis and B. canis always occur as rough forms, Among the B. suis, only biovars 1 and 3 are known to
even on primary isolation. All smooth forms of brucellae occur in pig-raising areas of the United States. Until 1946
may dissociate into intermediate, rough, or mucoid the only recognized cause of pig brucellosis in the Unit-
forms under certain artificially induced environmental ed States was the organism now known as B. suis biovar
conditions. This dissociation frequently occurs if cul- 1. At that time, B. suis biovar 3 (originally classified as
tures are left for a long period without being subcul- American B. melitensis) was first isolated from tissues of
tured, and it renders them incapable of being assigned to infected pigs by S. H. MacNutt (Borts et al. 1946). Since
species or biovar. Microscopically, Brucella organisms are the 1950s, reported isolations of B. suis biovar 1 have be-
small, gram-negative bacilli or coccobacilli and are non- come comparatively less frequent, while isolations of B.
motile and arranged singly. B. suis organisms from dif- suis biovar 3 have become more frequent. There is little
ferent sources may vary considerably in size but are gen- doubt that biovar 3 is now the predominant biovar in
erally 0.4–0.8 × 0.6–3.0 µm. porcine brucellosis in the United States.
Several commercially available agar media are suit-
able for isolation and propagation of B. suis; those more EPIDEMIOLOGY
commonly used include tryptose, trypticase-soy, Albimi,
serum dextrose, Farrell’s, and potato infusion. The addi- Most evidence indicates that most B. suis infection is
tion of serum to the media to a final concentration of 5% transmitted to susceptible animals through direct associ-

Table 28.1. Characteristics used for the differentiation of Brucella suis


Lysis by Lysis by
H2S Growth on Growth on Dominant Tb Phage Tb Phage
Production Thionin Fuchsin Antigen at RTD at 104 RTD
Biovar 1 + + −a A NLb Lc
Biovar 2 − + −a A NLb Lc
Biovar 3 − + +a A NLb Lc
Biovar 4 − + −d AM NLb Lc
Biovar 5 − + −d M NLb Lc
a
Agglutination with A and M monospecific serum.
b
No lysis.
c
Lysis.
d
Some strains may grow.
CHAPTER 28 BRUCELLOSIS MacMillan 387

ation with infected pigs. In this species, the most impor- sibly using strain RB51, a rough mutant strain recently
tant routes of infection are through the alimentary and introduced for routine use in cattle in the United States
genital tracts. The habits of pigs and usual character of (Enright 1995). There have been numerous instances of
the disease strongly suggest that the alimentary tract is B. suis infection or seropositivity in rodents or carnivo-
the most common portal of entry. Pigs of all ages may rous species trapped near areas where brucellosis in do-
eat food or drink fluid contaminated with discharges mesticated pigs has occurred. However, general indica-
from infected pigs. Piglets are frequently infected by tions are that these species acquired infection from the
nursing infected dams, and when breeding pigs are con- pigs and are terminal hosts of the infection. With few or
fined together, aborted fetuses and fetal membranes are no exceptions, epidemiological investigation of newly in-
readily consumed. Brucellosis is a venereal disease in fected pig herds has revealed the source as another herd
pigs, and sows are readily infected when mated with in- of domesticated pigs.
fected boars or when artificially inseminated with semen Experimental studies have shown that pigs can be in-
containing B. suis. Experimentally, pigs are readily infect- fected with B. suis biovar 4, B. abortus, B. melitensis, B. ca-
ed by conjunctival or intranasal exposure with suspen- nis, and B. neotomae. However, there has been no evi-
sions of B. suis. It is possible that organisms could also dence that these organisms invade the genital tract, are
enter through scarified or, possibly, intact skin. transmissible between pigs, or localize in any tissues oth-
The survival of brucellae under environmental con- er than lymph nodes draining the site of infection. All
ditions is a relatively important factor in transmission of available evidence indicates that these biovars or species
the disease. The survival rate of brucellae is similar to are not highly pathogenic for pigs, pigs are not likely to
that of other nonsporing gram-negative bacteria and as show clinical evidence of disease, and the infection is
such is extremely variable depending on prevailing con- self-limiting and usually persists less than 60 days. Nev-
ditions. Experimental evidence indicates that B. suis is ertheless, these infections do have importance for public
readily killed by pasteurization, 2–4 hours of direct sun- health. In particular, pigs are associated with B. abortus
light, and the most commonly used disinfectants. Bru- infection in packinghouse workers.
cellae can survive in organic matter at freezing or near- Pigs infected with B. suis biovars 1, 2, or 3 can serve as
freezing temperatures in excess of 2 years. Consequently, a source of infection for other domesticated animal
efforts to eliminate brucellosis from pig-raising premises species. B. suis infection can occur naturally in horses,
must include an effective sanitation program cattle, and dogs. Although the most common brucella in-
(Luchsinger et al. 1965). The most suitable methods for fection in horses is B. abortus, fistulous withers and other
preserving Brucella organisms for long periods are syndromes have been recorded as caused by B. suis when
lyophilization and/or storage at subfreezing tempera- horses were associated with infected pigs. Cattle are
tures. rarely infected with B. suis; when infection does occur,
There are few known reservoirs of B. suis infection the characteristic infection is mastitis, with B. suis organ-
other than infected domestic pigs. Only the European isms excreted in the milk representing a public health
hare and feral pigs have been established as significant risk. It can cause an acute infection in dogs, with preg-
potential reservoirs. The European hare was incriminat- nant bitches frequently aborting. There is a some evi-
ed as a natural host for B. suis biovar 2 as early as 1954 dence that B. canis, a significant pathogen in dogs,
(Bendtsen et al. 1954) and is apparently responsible for evolved from B. suis biovar 3.
periodic outbreaks of brucellosis of pigs in Europe. Feral
pigs in the southeastern United States, and to a much PATHOGENESIS
lesser extent elsewhere (Drew et al. 1992), have been dis-
covered to have a high rate of serologic reactors, with iso- Available literature and comparative studies indicate
lation of B. suis biovar 1 from some animals (Wood et al. that the pathogenesis of B. suis biovars 1, 2, and 3 is very
1976; Becker et al. 1978). Extensive studies conducted in similar (Thomsen 1934; Hutchings 1950; Hoerlein et al.
Florida showed that although only some populations are 1954; Deyoe 1967). Differences are generally related to
infected, the incidence within these groups is usually factors such as method of exposure, infecting dose, age
high (Leek et al. 1993). The epidemiological importance and breed of pigs, and possibly minor differences be-
of feral pigs in the maintenance of porcine brucellosis tween strains of the same biovar. However, the charac-
depends largely on the degree of contact between wild teristics of the disease produced are usually indistin-
and domestic pigs (Nettles 1991). If pig management guishable.
systems in regions where feral pigs exist prevent contact, Regardless of the route of infection, the organism
B. suis infection in feral pigs may be of greater public must be able to attach to and penetrate the mucosal ep-
health importance than a threat to the pig industry. The ithelium, although the mechanisms for this have not yet
vaccination of feral populations using dosed baits has been fully elucidated. Following the initial penetration,
been shown to be a feasible option to be considered in submucosal aggregations of lymphocytes and plasma
certain situations in the future (Fletcher et al. 1990), pos- cells form in response. Invading organisms are carried to
388 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

local lymph nodes, although it is not known whether killed at various intervals after exposure to B. suis and
they travel as free organisms or within phagocytes, and their tissues subjected to thorough bacteriological exam-
infected nodes become enlarged due to lymphoid and ination. Three-fourths of the pigs in each group of fe-
reticuloendothelial hyperplasia and infiltration. Brucella males had recovered from infection by 4–6 months or
organisms surviving regional node colonization enter a longer after exposure, whereas the recovery rate in males
phase of bacteremia, now protected from humoral im- never exceeded 50%. In contrast, Goode et al. (1952) and
mune mechanisms by their intracellular location within Manthei et al. (1952) isolated B. suis from only 12 of 474
neutrophils and macrophages. adult pigs exposed as suckling pigs. This information
In B. suis infection of pigs, bacteremia is an invariable demonstrates beyond doubt that the majority of pigs in-
finding in acute stages of the disease if frequent blood fected with B. suis will eventually recover spontaneously.
samples are collected and examined bacteriologically. In Nevertheless, sufficient numbers of permanently infect-
general, the onset of bacteremia ranges from 1 to 7 ed animals will remain to serve as a continual source of
weeks after exposure, with a mean of about 2 weeks post- infection.
exposure. Bacteremia persists an average of about 5
weeks and is generally continuous during that time. In- CLINICAL SIGNS
termittent bacteremia in individual pigs has been ob-
served to be as brief as 1 week to as long as 34 months. It Clinical evidence of B. suis infection varies considerably
is this bacteremia that is probably responsible for the in different herds. The majority of affected herds may
wide range of tissues secondarily infected during the have no signs of brucellosis recognizable by the herd
course of the disease. owner. The classic manifestations of pig brucellosis are
Within a short time after the bacteremia stage, B. su- abortion, infertility, orchitis, posterior paralysis, and
is can be isolated from a large number of sites in the lameness. Infected pigs fail to show any persisting or un-
body (Deyoe and Manthei 1967). The entire lymphatic dulating pyrexia. Clinical signs may be transient and
system is often affected for a period of time. With in- death is a rare occurrence.
creasing time after exposure, the sites of localization of Abortions may occur at any time during gestation
the organism tend to be reduced in number. Among and are influenced more by the time of exposure than by
lymph nodes, the most frequent sources of B. suis are the time of gestation. The rate of abortion is highest in
mandibular, gastrohepatic, internal iliac, and suprapha- sows or gilts exposed via the genital tract at the time of
ryngeal, in that order, depending essentially on the route breeding (Deyoe and Manthei 1969). Abortions have
of infection. Organs of the genital system containing been observed as early as 17 days following natural in-
high levels of erythritol, a sugar promoting the growth of semination by boars disseminating B. suis in the semen.
brucellae, become involved in many pigs and may re- Early abortions are usually overlooked under field condi-
main persistently infected. The placenta is a privileged tions, and the first indication is a large percentage of
site, and brucellae localize in the rough endoplasmic sows or gilts showing signs of estrus 30–45 days after the
reticulum of the chorionic trophoblasts. Despite severe service that resulted in conception. Little or no vaginal
placental infection, only mild inflammation of the en- discharge is observed with early abortions. Abortions
dometrium is observed. The spleen, liver, kidney, blad- that occur during the middle or late stages of gestation
der, mammary gland, and brain may be involved (Jubb are usually associated with females that acquire infection
et al. 1985), although not as regularly as lymph nodes. after pregnancy has advanced past 35 or 40 days. The
Other significant sources of B. suis, particularly in chron- persistence of genital infection in females varies consid-
ically infected pigs, are joint fluids and bone marrow. erably.
The response to invasion of B. suis becomes evident A small percentage of sows have been shown to shed
with the appearance of humoral antibody, activation of B. suis in vaginal discharges for as long as 30 months.
the cell-mediated immune system, and development of However, the majority ceased shedding organisms with-
microscopic lesions. These manifestations may occur si- in 30 days. A clinically apparent abnormal vaginal exu-
multaneously but usually are subsequent to the appear- date is seldom observed in sows that have uterine infec-
ance of detectable bacteremia; bacteremia may precede tion except just prior to and for a short time after
detectable antibody levels possibly by as much as 6–8 abortion. The majority of female pigs eventually recover
weeks. As pigs recover from B. suis infection (i.e., when from genital infection.
viable organisms are no longer present in their tissues), When genital infection in sows persists only a short
other manifestations, such as antibody levels, cellular hy- time after abortion, parturition, or breeding to an infect-
persensitivity, and microscopic lesions, recede and dis- ed boar and the sows are permitted two or three estrous
appear also. Unfortunately, many pigs remain perma- cycles of sexual rest, subsequent conception rates and re-
nently infected. productive capacity are usually very good.
In a series of experiments, infection was established Genital infection tends to be more persistent in boars
in 248 sexually mature pigs (Deyoe 1972a). They were than in sows. Some infected boars do not develop a lo-
CHAPTER 28 BRUCELLOSIS MacMillan 389

calized genital infection. However, boars that do develop DIAGNOSIS


genital infection seldom recover from it. Pathologic
changes in the male accessory glands or testes are gener- The most accurate and possibly the most sensitive
ally more extensive and irreversible than in the uterus. method of diagnosis of porcine brucellosis is isolation of
Infertility and lack of sexual drive may occur in infected Brucella organisms by direct culture methods. It has been
boars and are frequently associated with testicular in- shown that routine culture of a small sample of lymph
volvement. More commonly, however, boars have infec- nodes from carcasses will reveal as many positives as
tion in accessory genital glands and as a result dissemi- serologic diagnosis (Alton 1990; Rogers et al. 1989). This
nate large numbers of B. suis in their semen. These boars is a very practical survey strategy, as virtually all the pro-
do not necessarily have reduced fertility (Vandeplassche duce of the industry passes through abattoirs and the
et al. 1967). In most circumstances, clinically apparent material can be removed without damage to the carcass.
lesions of B. suis biovar 1, 2, or 3 infection in boars are Culture of other material that becomes available is often
seldom encountered. fruitful, such as vaginal swabs or products of abortion,
Clinical brucellosis in suckling and weaning pigs usu- semen samples or castrated testicles, the contents of
ally appears as spondylitis associated with posterior swollen joints, and blood samples. However, culture is
paralysis. These clinical signs are occasionally observed often not feasible because of inadequate or unavailable
in any age of pigs. laboratory facilities and trained personnel (Deyoe 1969).
B. suis can readily be grown on all the normal Brucella
LESIONS media in the absence of added CO2, the techniques being
fully described by Alton et al. (1988).
Macroscopic pathologic changes produced by B. suis in Detection of B. suis antigen in tissues of infected pigs
pigs are quite variable. Enough abscess formation may has been investigated, primarily using fluorescent anti-
occur in affected organs to result in necrosis and desqua- body (FA) techniques. The general conclusion has been
mation of a significant proportion of the mucous mem- that brucellae are seldom detectable in lymph node im-
brane. Generally, the histopathological changes consist pression smears with FA procedures because of the rela-
of uterine glands filled with leukocytes, cellular infiltra- tively low numbers of organisms typically present (Dey-
tion of the endometrial stroma, and hyperplasia of oe 1972b). Nevertheless, FA tests could probably be
periglandular connective tissue. Diffuse suppurative in- useful for examining aborted materials, since large num-
flammation is usually present in affected placentas. bers of B. suis are typical in such specimens. More re-
There also may be considerable necrosis of epithelium cently, potentially sensitive methods for the detection of
and diffuse hyperplasia of fibrous connective tissue. the presence of brucellae, such as the polymerase chain
Focal microscopic granulomatous lesions frequently reaction (PCR), are gradually being introduced into rou-
can be observed in livers of pigs with brucellosis, partic- tine use and may, in the future, prove a valuable method
ularly during bacteremic phases of the disease. These fo- of diagnosis in certain situations (Lealklevezas et al.
ci frequently are necrotic areas infiltrated with lympho- 1995).
cytes, macrophages, neutrophils, and giant cells, with Serologic procedures to detect antibodies against
sheets of histocytic and epithelioid cells with a central brucellae in infected pigs are generally the most practical
zone of caseous or coagulative necrosis. The lesions are and most common means of diagnosis, but the results
usually partially or completely enclosed by a fibrous cap- obtained are far from perfect. Market pig surveys have
sule. The necrotic portions of the granulomas are heavi- shown that as many as 18% of normal pigs may react at
ly infiltrated with neutrophils, and liquefaction and min- the 1:25 level when plate agglutination tests are used
eralization may occur (Enright 1990). (Deyoe 1969). On the other hand, some pigs produce lit-
These lesions are not specific for brucellosis, since tle or no antibody against brucellae. Because of variation
similar hepatic lesions are associated with other bacteri- in the stage of disease, an infected herd of pigs will near-
al infections. ly always contain some infected animals that have no de-
Microscopic lesions of bones are sometimes caused tectable brucella antibody. Some strains of B. suis appar-
by B. suis infection. These occur both in vertebrae and in ently do not stimulate antibody production as well as
long bones. The lesions are most frequently located adja- others (Deyoe 1967). Pigs exposed to a minimal infective
cent to the epiphyseal cartilage and usually consist of dose of B. suis generally have a prolonged incubation pe-
caseous centers surrounded by a zone of macrophages riod before significant quantities of antibody are pro-
and leukocytes and often by an outer zone of fibrous duced.
connective tissue. Because of the foregoing factors, current serologic
Focal areas of chronic lymphocytic and macrocytic tests are much less effective for the diagnosis of individ-
inflammation or focal abscesses are found infrequently ual pigs than they are in cattle. However, most serologic
in kidneys, spleen, brain, ovaries, adrenal glands, lungs, tests are entirely adequate as herd tests. Characteristical-
and other tissues of infected pigs (Deyoe 1968). ly, infected herds include a majority or large numbers of
390 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

infected individuals. Because of close contact between detect IgM brucella antibodies in pig serum; therefore,
animals and the tendency of brucellosis to spread rapid- they are highly specific. However, the relative sensitivity
ly through a herd, 50–80% is a common morbidity range of these methods is usually low in early stages of brucel-
(Spencer and Mattison 1975). When large herds have on- losis. By the time the antibody response peaks and there-
ly a single serologic reactor disclosed during a herd test, after in chronic stages, the Rivanol, mercaptoethanol,
it can generally be concluded that B. suis infection is not and CFT methods are generally as sensitive as the card
present. test. An evaluation of a range of serologic tests on cul-
Numerous serologic tests are available or have been ture-positive pigs was reported by Rogers et al. (1989).
investigated for use in diagnosis of porcine brucellosis The sensitivity of the RBT was 79%; of the CFT, 49%; and
(Alton et al. 1988). Many of these were developed for di- of the SAT, 51%. The specificity of the RBT was reported
agnosis of bovine brucellosis and have been adapted for to be 98% based on the results of testing over 30,000
testing pig sera. Most tests utilize B. abortus whole-cell serum samples. Other studies generally confirm the low
antigens. Since the commonly used antigen strains B. levels of diagnostic sensitivity achievable (Ferris et al.
abortus 1119-3 and S99 have the same or very similar sur- 1995; Payeur et al. 1990).
face lipopolysaccharide complexes as smooth B. suis, the Regardless of the serologic test used for diagnosis, de-
standardized antigens produced and distributed by the tection of 80–90% of infected pigs must be regarded as
Animal and Plant Health Inspection Service (APHIS) of the best that can be achieved at present.
the USDA and by the Central Veterinary Laboratory, Limited investigation of the enzyme-linked im-
Weybridge, United Kingdom, are equally useful for diag- munosorbent assay (ELISA) has been conducted, and it
nosis of both bovine and porcine brucellosis. This has appears that this method may be equal or slightly supe-
been confirmed by extensive laboratory testing of pig rior to other serologic procedures for diagnosis of
sera with both B. abortus and B. suis antigens. porcine brucellosis in the future (Office International des
The original test methods for the diagnosis of Epizooties 1997). Further investigation of this test for
porcine brucellosis were tube and plate agglutination use in eradication campaigns is warranted.
procedures. Interpretation of results was based on the Experimental studies reviewed by Corbel (1985) have
finding that most infected pig herds contained one or shown that infection with organisms of several other
more animals with more than 100 international units genera can produce antibodies reactive in brucellosis di-
(IU) of agglutination. It is now known that serum agglu- agnostic tests. These organisms include Escherichia coli
tination tests (SAT), although sensitive, are not suffi- serogroup O:157, Salmonella serobiovars of Kaufman-
ciently specific to be reliable diagnostic tools when used White group N, and most importantly Yersinia enteroco-
alone. Some of the inaccuracies can be overcome in situ- litica serogroup O:9. Infection of pigs with this latter or-
ations where frequent and repeated testing is practicable ganism has often been confirmed, and the cross-reaction
and the trend of antibody titers can be determined. that results (the dominant O polysaccharide antigen is
Reducing the pH of antigen-serum mixtures to 3–4 chemically identical to the A antigen present on the sur-
was also found to reduce nonspecific agglutination while face of all smooth brucellae) is highly significant. In
not affecting agglutination caused by serum from infect- some situations, yersiniosis poses a greater threat to the
ed animals. This led to the development of methods clas- agricultural industry than does brucellosis itself, due to
sified as buffered brucella antigen tests, in which stained the confusion with brucellosis in diagnosis and the con-
brucella antigen is buffered at pH 3.65. The buffered bru- sequent effect on the export trade. Great Britain has al-
cella antigen became the basis of the brucellosis card test ways been free from B. suis infection and enjoys a thriv-
and similar procedures such as buffered plate antigen ing export trade as a result of the generally high health
and Rose-Bengal tests (RBT). These tests are the most status of its stock. During the 7 years prior to 1988, the
practical method of diagnosis for porcine brucellosis at number of pigs tested for export certification giving a
present, are possibly still the preferred method for large- CFT reaction of greater than 20 international comple-
scale surveillance testing, and are “prescribed tests for in- ment-fixation test units (icftu) never exceeded 0.004%,
ternational trade” (Office International des Epizooties whereas the figures for 1988, 1989, and 1990 were 0.42%,
1997). They have a distinct advantage over standard ag- 0.70%, and 1.5%, respectively. Since 1988, at least 4% of
glutination tests because they are relatively unaffected by exporting herds have had more than 5% CFT positive re-
nonspecific agglutinins and are generally as sensitive as actions, with some herds reaching levels of more than
any other serologic test for diagnosis of porcine brucel- 50% of animals tested failing at this level. Y. enterocolitica
losis. O:9 has been isolated from many herds involved, and de-
Other tests, such as the Rivanol precipitation–serum spite extensive investigation, B. suis has not been recov-
agglutination, 2-mercaptoethanol, and complement fixa- ered (Wrathall et al. 1991).
tion tests (CFT), are frequently used for the diagnosis of Lymphocyte transformation tests have been used to
brucellosis in pigs and are very useful in confirming re- measure cell-mediated immune responses in infected
sults of card tests. The above tests very seldom or never pigs on a limited scale (Kaneene et al. 1978). There was
CHAPTER 28 BRUCELLOSIS MacMillan 391

high correlation between recovery of B. suis from tissues the disease. With the present incidence of porcine bru-
and detectable lymphocyte stimulation responses. How- cellosis in the United States, the cost of developing and
ever, the complexity of the method probably eliminates applying vaccination cannot be justified.
it from consideration as a diagnostic tool except in spe- There have been no basic studies specifically directed
cial cases. toward the mechanism of immunity against B. suis infec-
Tests for delayed hypersensitivity, using intradermal tion in pigs. Nevertheless, one must assume from the
injection of brucella allergens, have been studied, but re- overwhelming evidence accumulated in research on bru-
sults have not stimulated much enthusiasm in the United cellosis in other species that the fundamental mecha-
States. However, they are of similar accuracy to serologic nism is a cell-mediated immunity, with humoral immu-
tests and would be more appropriate for farm testing in nity having only a minor or nonexistent role.
some circumstances, although they are more difficult to Investigations into antibrucella immunity in pigs can
apply and read and would not be applicable in testing be summarized as follows:
programs for market pigs. In the face of cross-reactions
caused by Y. enterocolitica O:9, the use of such tests is per- 1. Some pigs are naturally resistant to brucella infec-
haps the most specific method of diagnosis. Skin tests tion, and this resistance could be enhanced markedly by
are used frequently for diagnosis of porcine brucellosis selective breeding programs (Cameron et al. 1941) if all
in many countries, particularly in Eastern Europe. other genetic factors could be ignored.
One of the most important aids to diagnosis is an ad- 2. The majority of pigs infected with virulent B. suis
equate herd history. Good records of clinical manifesta- will recover from the disease, but subsequent resistance
tions, movement of animals, additions to the herd, induced by the virulent infection may be transient, as
breeding records, and illnesses in persons working with most will be susceptible to reexposure probably within
the pigs provide invaluable information necessary to ar- 6–12 months after the initial infection.
rive at a diagnosis of brucellosis. Accurate epidemiologi- 3. Trials with attenuated B. suis or B. abortus strain 19
cal information is an essential supplement to laboratory have been unsuccessful in producing a persistent immu-
tests. nity with products considered to be safe for use.
4. Bacterins or extracts of killed B. suis have general-
TREATMENT ly been ineffective in stimulating immunity or else there
has been no conclusive evidence that the persistence of
No treatments, such as antibiotic therapy, dietary sup- immunity is adequate.
plements, or other chemotherapy, have been proven ef-
fective and economically feasible in curing pigs of bru- Control Measures
cellosis. Large doses of tetracyclines, streptomycin, or Experiences in control of porcine brucellosis indicate
sulfonamides given over relatively long periods have that eradication of the disease from pigs in the United
been investigated. In some trials these antibiotics alone States is desirable and feasible. Marked reduction in the
or in combination appeared promising. In general, how- incidence of the disease has occurred since 1950. One
ever, antibiotic therapy was effective in limiting the bac- significant factor in this reduction has been the tendency
teremic stage of the disease, but after therapy was dis- for pig production to become more specialized and less a
continued, viable B. suis was still present in tissues. part of diversified farming operations. Consequently,
Although treatments have not been effective in eliminat- the occurrence of reproductive disease in pigs has be-
ing all organisms from the host, chemotherapy in care- come proportionately more important, confinement sys-
fully selected circumstances could probably suppress tems and closed herds have eliminated many opportuni-
multiplication of B. suis in vivo sufficiently to alleviate ties for interfarm spread of disease, and larger units have
clinical manifestations and shedding of organisms. Even eliminated the “community boar” in most instances. An-
though such an approach may have limited practicabili- other important instrument in control of porcine bru-
ty, it could have beneficial effects in an infected herd and cellosis has been the establishment and maintenance of
should not be dismissed as useless. validated brucellosis-free herds, particularly purebred
herds or those selling breeding stock. Implementation of
PREVENTION effective surveillance programs such as identification
and testing of market pigs (sows and boars) has been in-
Immunity strumental in locating and eliminating large numbers of
Safe and reliable vaccines that produce serviceable im- infected herds. Finally, it has been found that whenever
munity against brucellosis in pigs have not been devel- recommended procedures to eradicate brucellosis from
oped. Significant resistance can be stimulated, but per- an individual herd or an enzootic area are conscientious-
sistence of immunity has been a limiting factor (Deyoe ly followed, there is very seldom any recurrence of the
1972a). Interest in vaccination of pigs in the United disease in that locality (e.g., Spencer and Mattison 1975).
States has declined along with the decline in incidence of The current brucellosis eradication program in the
392 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

United States is a joint state-federal and livestock indus- Brucella. In Bergey’s Manual of Determinative Bacteriol-
try program. The program is administered, supervised, ogy, 8th ed. Baltimore: Williams and Wilkins, pp.
and funded by cooperative efforts between state and fed- 278–282.
eral animal health regulatory agencies. The livestock in- Cameron, H. S.,; Gregory, P. W.; and Hughes, E. H. 1941.
dustries have input into procedures to be used through Studies on genetic resistance in swine to Brucella infec-
representation on advisory committees that ultimately tion. II. A bacteriological examination of resistant stock.
determine the Uniform Methods and Rules for Brucel- Cornell Vet 31:21–24.
losis Eradication, the principal guideline for conducting Cook, D. R., and Noble, J. W. 1984. Isolation of B. suis from
the program. These rules and guidelines are revised fre- cattle. Aust Vet J 61:263–264.
quently; therefore, current information regarding the Corbel, M. J. 1985. Effect of atrophic rhinitis vaccines on the
program as it applies in each state is always available reaction of pigs to serological tests for brucellosis. Vet
from each state veterinarian. Rec 117:150.
There are three acceptable alternative plans recom- de Ley, J.; Mannheim, W.; Segers, P.; Lievens, A.; Denijin,
mended for use when pigs herds are found to be, or sus- M.; Vanhouke, M.; and Gillis, M. 1987. Ribonucleic acid
pected of being, infected with B. suis. Plan 1 consists of cistron similarities and taxonomic neighbourhood of
depopulation of the entire herd, which is by far the most Brucella and CDC group Vd. Int J Syst Bacteriol
successful and the most economical in the long run. Plan 37:35–42.
2 is a procedure designed to salvage irreplaceable blood- Deyoe, B. L. 1967. Pathogenesis of three strains of Brucella
lines and basically consists of marketing the adult pigs suis in swine. Am J Vet Res 28:951–957.
for slaughter and retaining weanling pigs for breeding ———. 1968. Histopathologic changes in male swine with
stock, a plan that is not always successful and necessi- experimental brucellosis. Am J Vet Res 29:1215–1220.
tates considerable isolation and retesting requirements. ———. 1969. Diagnostic tests for swine brucellosis. Proc
Plan 3 consists of removing only serologic reactors and Annu Meet Livest Conserv 53:20–22.
retesting the herd as many times as necessary. This last ———. 1972a. Immunology and public health significance
procedure is rarely successful if the herd is actually in- of swine brucellosis. J Am Vet Med Assoc 160:640–643.
fected but is the plan of choice if the herd contains only ———. 1972b. Research findings applicable to eradication
a single reactor or if a very low proportion of animals are of swine brucellosis. Proc Annu Meet US Anim Health
reactors and there is reasonable doubt that brucellosis Assoc 76:108–114.
exists. Deyoe, B. L., and Manthei, C. A. 1967. Sites of localization
of Brucella suis in swine. Proc Annu Meet US Livest San-
REFERENCES it Assoc 71:102–108.
———. 1969. Swine brucellosis. In Proc 1967 Symp on Fac-
Alton, G. G. 1990. Brucella suis. In Animal Brucellosis. Boca tors Producing Embryonic and Fetal Abnormalities,
Raton, Fla.: CRC Press. Death, and Abortion in Swine. ARS 91-73, pp. 54–60.
Alton, G. G.; Jones, L. M.; and Pietz, D. E. 1975. Laboratory Drew, M. L.; Jessup, D. A.; Burr, A. A.; and Franti, C. E. 1992.
Techniques in Brucellosis, 2d ed. Geneva: World Health Serological survey for brucellosis in feral swine, wild ru-
Organisation. minants, and black bear of California. J Wildl Dis
Alton, G. G.; Jones, L. M.; Angus, R. D.; and Verger, J. M. 28:355–363.
1988. Techniques for the brucellosis laboratory. Paris: Enright, F. M. 1990. The pathogenesis and pathobiology of
INRA. Brucella infection in domestic animals. In Animal Bru-
Becker, H. N.; Belden, R. C.; Breault, T.; Burridge, M. J.; cellosis. Boca Raton, Fla.: CRC Press.
Frankenberger, W. B.; and Nicoletti, P. 1978. Brucellosis ———. 1995. Update on research with RB51 strain of Bru-
in feral swine in Florida. J Am Vet Med Assoc cella abortus. Proc Annu Meet US Anim Health Assoc
173:1181–1182. 99:647–648.
Bendtsen, H.; Christiansen, M.; and Thomsen, A. 1954. Bru- Ferris, R. A.; Schoenbaum, M. A.; and Crawford, R. P. 1995.
cella enzootics in swine herds in Denmark—Presumably Comparison of serological tests and bacteriologic cul-
with hare as a source of infection. Nord Vet Med ture for detection of brucellosis in swine from naturally
6:11–21. infected herds. J Am Vet Med Assoc 207:1332–1333.
Borts, I. H.; Harris, D. M.; Joynt, M. F.; Jennings, J. R.; and Fletcher, W. O.; Chreekmore, T. E.; Smith, M. S.; and Net-
Jordan, C. F. 1943. A milk borne epidemic of brucellosis tles, V. F. 1990. A field trial to determine the feasibility of
caused by the porcine biotype of Brucella (Brucella suis) delivering oral vaccines to wild swine. J Wildl Dis
in a raw milk supply. J Am Vet Med Assoc 121:319. 26:503–510.
Borts, I. H.; McNutt, S. H.; and Jordan, C. F. 1946. Brucella Fox, M. D., and Kaufmann, A. F. 1977. Brucellosis in the
melitensis isolated from swine tissues in Iowa. J Am Vet United States, 1965–1974. J Infect Dis 136:312–316.
Med Assoc 130:966. Frye, G. H.; Gilsdorf, M. J.; and Lenard, D. 1993. Status re-
Brinley-Morgan, W. J., and McCullough, N. B. 1974. Genus port—Fiscal year 1993. Cooperative State-Federal Bru-
CHAPTER 28 BRUCELLOSIS MacMillan 393

cellosis Eradication Program. Proc Annu Meet US Anim in feral swine. Proc Annu Meet US Anim Health Assoc
Health Assoc 97:138–154. 95:551–556.
Goode, E. R., Jr.; Manthei, C. A.; Blake, G. E.; and Amerault, Office International des Epizooties. 1997. Manual of Stan-
T. E. 1952. Brucella suis infection in suckling and wean- dards for Diagnostic Tests and Vaccines. 3rd Edition.
ling pigs. II. J Am Vet Med Assoc 121:456–464. Paris.
Hoerlein, A. B.; Hubbard, E. D.; Leith, T. S.; and Biester, H. Paolicchi, F. A.; Terzolo, H. R.; and Campero, C. M. 1993.
E. 1954. Swine Brucellosis. Vet Med Res Inst, Iowa State Isolation of Brucella suis from the semen of a ram. Vet
Coll. Rec 132:67.
Huddleston, I. F. 1929. The Differentiation of the Species of Payeur, J. B.; Miller, C. D.; Hennager, S. G.; and Ewalt, D. R.
the Genus Brucella. Bull Mich Agric Exp Stn no. 100. 1990. Comparison of five serologic tests and culture for
Hutchings, L. M. 1950. Swine brucellosis. In Brucellosis: brucellosis in swine experimentally infected with Brucel-
American Association for the Advancement of Science la suis biovar 1. Proc Annu Meet US Anim Health Assoc
Symposium. Washington, D.C.: Waverly Press, pp. 94:147–152.
188–198. Rogers, R. J.; Cook, D. R.; Kettlerer, P. J.; Baldock, F. C.;
Jubb, K. V. F.; Kennedy, P. C.; and Palmer, N. 1985. Patholo- Blackall, P. J.; and Stewart, R. W. 1989. An evaluation of
gy of Domestic Animals, vol. 3. Orlando: Academic three serological tests for antibody to Brucella suis in
Press, p. 349. pigs. Aust Vet J 66:77–80.
Kaneene, J. M.; Anderson, R. K.; Johnson, D. W.; Angus, R. Spencer, P. L., and Mattison, J. R. 1975. Pike County, Illi-
D.; Muscoplate, C. C.; Pietz, D. E.; Vanderwagon, L. C.; nois, swine brucellosis project. Proc Annu Meet US An-
and Sloane, E. E. 1978. Cell-mediated immune respons- im Health Assoc 79:86–91.
es in swine from a herd infected with Brucella suis. Am J Thomsen, A. 1934. Brucella infection in swine. Acta Pathol
Vet Res 39:1607–1611. Microbiol Scand (Suppl) 21.
Lealklevezas, D. S.; Martinez-Vazquez, I. O.; Lopez-Merino, Traum, J. 1914. Report to the Chief. Bureau of Animal In-
A.; and Martinez-Soriano, J. P. 1995. Single-step PCR for dustry, USDA, p. 30.
detection of Brucella spp. from blood and milk of infect- Vandeplassche, M.; Herman, J.; Spincemaille, J.; Bouters, R.;
ed animals. J Clin Microbiol 33:3087–3090. Dekeyser, P.; and Brone, E. 1967. Brucella suis infection
Leek, M. L. Van der; Becker, H. N.; Humphrey, P.; Adams, C. and infertility in swine. Meded Veeartsenijsch Rijksuniv
L.; Belden, R.C.; Frankenberger, W. B.; and Nicoletti, P. Gent 11:1–40.
L. 1993. Prevalence of Brucella sp. antibodies in feral Verger, J. M.; Greymont, F.; Gremont, P. A. D.; and Grayon,
swine in Florida. J Wildl Dis 29:410–415. M. 1985. Brucella, a monospecific genus as shown by de-
Luchsinger, D. W.; Anderson, R. K.; and Werring, D. F. oxyribonucleic acid hybridisation. Int J Syst Bacteriol
1965. A swine brucellosis epizootic. J Am Vet Med Assoc 35:292–295.
147:632–636. Wood, G. W.; Hendricks, J. B.; and Goodman, D. E. 1976.
Manthei, C. A.; Mingle, C. K.; and Carter, R. W. 1952. Bru- Brucellosis in feral swine. J Wildl Dis 12:579–582.
cella suis infection in suckling and weaning pigs. 1. J Am Wrathall, A. E.; Broughton, E. S.; Gill, K. P. W.; and Gold-
Vet Med Assoc 121:373–382. smith, G. P. 1991. Serological reactions to Brucella in
Nettles, V. F. 1991. Short- and long-term strategies for re- British pigs. Vet Rec 132:449–454.
solving problems of pseudorabies and swine brucellosis
Clostridial Infections
29 D. J. Taylor

A number of species of clostridia are involved in disease colonial morphology, by their biochemical characters, by
in swine. Clostridium perfringens (welchii) types A and C, the presence of specific antigens, and by the production
C. tetani, C. novyi, C. botulinum, and, to a lesser extent, C. of toxins. These toxins can also be used to distinguish be-
chauvoei and C. septicum are the causes of recognizable tween different varieties or types of a species. In some
clinical syndromes. The recognizable syndromes include cases the organisms can be identified in fixed smears by
C. perfringens type C enteritis, C. perfringens type A en- their morphology or antigenicity; or toxins can be identi-
teritis, tetanus, botulism, sudden death in sows associat- fied in pathological material, allowing confirmation of
ed with C. novyi, and blackleg (C. chauvoei and C. sep- diagnosis without isolation. Some clostridial species and
ticum). Some species are also involved as contaminants types can now be identified by DNA probes, polymerase
of wounds and lesions and may affect the clinical signs chain reaction (PCR), and sequencing technology in clin-
and the pathological and bacteriological findings in oth- ical specimens.
er diseases. Species which may invade existing lesions in- All of the clostridial species mentioned above cause
clude C. perfringens types A and C, C. novyi, C. septicum, disease by the elaboration of specific toxins or enzymes.
and C. chauvoei. Other species may be recorded in swine This fact is of vital importance when considering treat-
from time to time, and carcasses that are not chilled im- ment and prevention. Clostridia are sensitive to a wide
mediately after death are frequently invaded by range of antimicrobials, but these can only be effective in
clostridia from the gut. This invasion can be so rapid that treatment and prevention of disease when tissue de-
it may be difficult to identify the actual cause of death. struction is not very far advanced. Once toxins have been
The presence of clostridia in specific or nonspecific dis- produced and are fixed to tissues, antimicrobials can on-
ease in swine affects the prognosis of the disease and re- ly eliminate the bacteria and cannot reverse the damage.
quires specific treatment and supportive and preventive They may be of value in treatment, particularly in the
measures appropriate to the control of the organism(s) early stages of disease, but are of most use in short-term
present. prevention. Antitoxin may affect the course of the dis-
Clostridia are all large, gram-positive, spore-forming ease in some cases and protect in the short term, but
bacilli. All are anaerobic and can be grown in culture un- most recognizable clostridial diseases can be prevented
der appropriate conditions. Species can be identified by reliably by vaccinating the animal at risk or the sow to
the size and shape of the bacterial cell, by the presence or provide colostral protection.
absence of spores and their shape and position, by their

CLOSTRIDIUM PERFRINGENS TYPE C ENTERITIS


Fatal necrotic enteritis in nonimmune pigs is caused by (Matthias et al. 1968), the Netherlands (Plaisier 1971),
C. perfringens type C and is most common in piglets less Canada (Morin et al. 1983), and Japan (Azuma et al.
than 1 week of age. Affected animals may be found dead 1983). It has now been identified in most swine-rearing
or develop hemorrhagic diarrhea, which is rapidly fol- areas of the world.
lowed by death in acute cases. Subacute cases occur in
which mortality is less common. All are associated with a Etiology
necrotic enteritis caused by multiplication of the organ- C. perfringens is an encapsulated, gram-positive bacillus
ism in the small intestine. The disease was first identified measuring 1–1.5 µm by 4.8 µm. Spores are rarely visible
in 1955 in the United Kingdom (Field and Gibson 1955) but are ovoid to eccentric. The colonies are clearly visible
and Hungary (Szent-Iványi and Szabo 1955) but was sub- on horse or bovine blood agar after 24 hours’ incubation
sequently identified in the United States (Barnes and in anaerobic conditions and are 3–5 mm in diameter,
Moon 1964), Denmark (Høgh 1965), Germany grayish, circular, and surrounded by a variable zone of

395
396 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

beta hemolysis. Type C organisms produce alpha and be- Pathogenesis


ta toxins most consistently, as well as smaller amounts of Infection is oral and newborn piglets become infected
other toxins. C. perfringens type B has been recovered within minutes or hours of birth. The organisms multi-
from a syndrome resembling that described below ply in the gut to reach 108 –109 per gram of contents (Oh-
(Bakhtin 1956) and also produces beta toxin, but type D, nuna et al. 1992) and become attached to the jejunal ep-
which does not do so, has also been isolated (Harbola ithelial cells at the apexes of the villi (Arbuckle 1972;
and Khera 1990). Most isolates from the disease are iden- Walker et al. 1980). Desquamation of these epithelial
tified as belonging to type C. Type C organisms are not cells is accompanied by proliferation of the organisms
only primary causes of disease but can colonize the le- along the basement membrane and complete necrosis of
sions caused by diseases of piglets such as transmissible the lamina propria of the villi (Fig. 29.1). In peracute cas-
gastroenteritis (TGE). es hemorrhage accompanies the necrosis. The necrotic
zone later advances to involve the crypts, the muscularis
Epidemiology mucosae and submucosa, and occasionally the muscular
Outbreaks of C. perfringens type C enteritis have been layers. Some organisms penetrate the intestinal wall to
recorded in large numbers of herds in an area over a cause emphysema in the muscle layers and occasionally
short period of time, as in the epizootic reported from beneath the peritoneum and in the draining mesenteric
Minnesota and Iowa (Bergeland et al. 1966) when 41 lymph nodes. Thrombosis may develop in emphysema-
herds were affected. Once the disease has been identified tous areas. Most bacteria remain adherent to the necrot-
in a country or area, it becomes restricted to individual ic villi or are shed into the intestinal lumen along with
herds as outbreaks which may last for up to 2 months. In cell debris and blood and may sporulate there (Kubo and
a few units, the acute disease may occur over longer peri- Watase 1985).
ods, and in these cases nonimmune gilts or sows may The lethal, necrotizing beta toxin is the most impor-
have been introduced to an infected area or unit or tant factor in the pathogenesis of the disease. Strains iso-
piglets may not have received adequate levels of specific lated from piglets consistently produce alpha toxin
antibody in the colostrum. The age incidence of the dis- (lecithinase), beta toxin (Warrack 1963; Høgh 1967a),
ease is marked, as it normally occurs in pigs aged from 12 and a variety of the minor toxins, including the delta tox-
hours to 7 days, usually at around 3 days, although the in. Evidence for the importance of the toxin comes from
disease has been recorded in pigs aged 2–4 weeks (Berge- the widespread use of the toxoid alone in the prevention
land et al. 1966; Høgh 1974) and in weaned pigs of the disease. Experimental evidence is more equivocal.
(Meszaros and Pesti 1965; Matthias et al. 1968). Clinical- The disease was reproduced by the oral administration
ly affected pigs usually die, and mortality within affected of the toxin to pigs subsequently found to be carrying
litters may reach 100% in the progeny of nonimmune the organism (Field and Goodwin 1959; Bergeland 1965)
sows. When protective immunity develops in the sow as and by the inoculation of gut loops with whole-broth cul-
a consequence of disease in her litter, subsequent litters tures (Bergeland 1972), but no necrosis was produced by
are usually unaffected. Morbidity varies from herd to
herd according to the immune status of the sows in the
herd and the time period over which morbidity is mea-
sured. In 20 herds from Minnesota and Iowa, the inci-
dence of affected litters ranged from 9% to 100%, and the
total herd mortality ranged from 5% to 59%, with a mean
herd mortality of 26% (Bergeland et al. 1966). An aver-
age mortality of 54% was observed in 24 herds in Den-
mark (Høgh 1967b).
The organism can be transmitted as a vegetative or-
ganism from piglet to piglet in an infected farrowing
house or can be acquired from maternal feces. It can per-
sist in the environment as a vegetative organism or in the
spore form, and the disease can affect litters over long pe-
riods of time if vaccination is not practiced. Spores are
resistant to boiling for up to an hour and can persist for 29.1. Villus of the jejunum early in the course of
periods of at least a year in contaminated buildings. The C. perfringens type C infection in a 12-hour-old piglet.
purchase of carrier sows is the most likely source of the Numerous bacilli are present beneath the desquamated
disease for farms, but it can also be introduced in fecal epithelium in the area of the epithelial basement mem-
contamination on boots and clothing. brane (H&E).
CHAPTER 29 CLOSTRIDIAL INFECTIONS Taylor 397

the inoculation of beta toxin alone. The sensitivity of the feces are yellow-gray and mucoid, and the tail may be
toxin to trypsin may account for these results and for the coated with dried feces. Affected pigs may remain alert
occurrence of the disease in pigs of less than 4 days of and vigorous for 10 days or more, but their rate of
age, in which trypsin secretion is absent. growth is depressed. These pigs may eventually die after
Death results primarily from the effects of the intesti- several weeks or be killed because of their failure to gain
nal damage and to a lesser extent from the extraintestinal weight.
effects of the toxin. The toxin can cause sudden death fol-
lowing intravenous infusion in high doses and at lower Lesions
doses causes polioencephalomalacia, adrenal cortical Piglets that die from the peracute form are often in good
necrosis, nephrosis, and pulmonary edema (Bergeland condition and do not appear dehydrated. There may be
1965). It has been demonstrated both in the intestinal blackish discoloration of the abdominal skin even in an-
contents of affected pigs and in the peritoneal fluid. Hy- imals killed in extremis. The umbilical cord is usually
poglycemia occurs in field cases (Field and Goodwin still present. Reddish feces may be present on the per-
1959; Høgh 1967b) and may be a factor in mortality ineum. The abdominal wall is often edematous when cut.
along with secondary bacteremia due to C. perfringens or The most immediate and striking findings are the pres-
E. coli. ence of intensely hemorrhagic small intestines (Fig. 29.2)
and the presence of bloodstained fluid in the abdominal
Clinical Signs cavity. The lesions vary in extent, occurring in the je-
Clinical signs vary according to the immune status and junum but extending into the ileum. They may extend
the age of the piglets affected within a herd and from from 14 cm posterior to the pylorus to the cecum or af-
herd to herd. Peracute, acute, subacute, and chronic dis- fect only 2–3 cm of bowel. Lesions are reddish or black in
ease can be distinguished. The onset of clinical signs nor- color and there may be gas bubbles in the intestinal wall.
mally occurs within the first 2–3 days of life in all forms The mesenteric lymph nodes may be reddened. The con-
of the disease. tents of the affected area are hemorrhagic, and hemor-
rhagic contents may be found in the intestine distal to
PERACUTE FORM. Piglets may be found dead within the lesion, often as far as the rectum. The mucosa is in-
12–36 hours of birth. Affected piglets remain normal for tensely hemorrhagic and no villi can be distinguished.
the first 10 hours of life. They develop hemorrhagic diar- Microscopically, the villi in the affected portion of the je-
rhea in most cases and this may stain the perineum but junum are necrotic and covered by large gram-positive
can be overlooked. Piglets become weak, move with re- bacilli. The epithelium of the crypts may or may not be
luctance, rapidly become moribund, and may be crushed
by the sow. The rectal temperature falls to 35˚C and the
abdominal skin may blacken before death. Death may
occur in some animals without diarrhea being seen.

ACUTE FORM. Acute cases survive 2 days after the


onset of clinical signs and commonly die when 3 days
old. Throughout the course of the disease they have red-
dish-brown liquid feces that contain shreds of gray
necrotic debris. They lose condition and become gaunt
and weak, making only feeble attempts to suck on the
last day of life.

SUBACUTE FORM. These pigs have a persistent non-


hemorrhagic diarrhea and usually die when about 5–7
days of age. They remain active and alert and have a fair
appetite but become progressively more emaciated and
may be extremely thin and dehydrated at the time of
death. The feces tend to be yellow at first and then
change to a clear liquid containing flecks of necrotic de-
bris.

CHRONIC FORM. Chronic cases may have an inter- 29.2. Necrohemorrhagic jejunitis in a 1-day-old piglet
mittent or persistent diarrhea for 1 or more weeks. The with peracute C. perfringens type C enteritis.
398 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

necrotic, and there is profuse hemorrhage throughout


the mucosa and submucosa (Figs. 29.3 and 29.4). Large
numbers of gram-positive bacilli are present in smears of
the lesions.
Acute cases may be dehydrated or show loss of bodi-
ly condition, and there may be scalding of the perineum
and adherent reddish feces. The intestines are less fre-
quently reddened, and any reddish lesions may be local-
ized. Emphysema of a sharply demarcated portion of up
to 40 cm of the jejunum may be present from 30 cm pos-
terior to the pylorus, and this portion of the intestine
may be loosely adherent to adjacent segments by acute
fibrinous peritonitis (Fig. 29.5). The wall of the intestine
is usually thickened, and the contents are sometimes
bloodstained and often contain some necrotic debris.
The mucosa is yellow or grayish in color and covered
with loosely adherent necrotic debris. Microscopic le-
sions include the absence of most villi; those that survive
29.5. Acute C. perfringens type C enteritis in a 3-day-
are necrotic and covered with bacteria. The luminal sur- old piglet. An emphysematous segment of upper jejunum
face is covered with a necrotic membrane made of bacte- (left) is held together by acute peritonitis. Mucosal necro-
sis of the lower jejunum (right) is seen from the serosal
surface.

ria, shed epithelial cells, fibrin, and degenerating inflam-


matory cells lying directly over the submucosa. The sub-
mucosal vessels are necrotic and many contain thrombi.
Emphysema may be evident in the submucosa, tunica
muscularis, and under the serosa (Fig. 29.6). Large gram-
positive bacteria may be present in deeper layers of the
intestinal wall.
Subacute cases are usually in poor condition, and
there may be adhesions between affected areas of the
small intestine. The intestinal wall is thickened and fri-
able, and the mucosal surface is covered by a tightly ad-
herent necrotic membrane which may be seen from the
29.3. Jejunum of a piglet with peracute C. perfringens
serosal surface as longitudinal grayish-yellow bands (Fig.
type C enteritis. There is acute necrosis of the mucosa
and massive hemorrhage in the submucosa (H&E).
29.7).

29.6. Jejunum of a piglet with acute C. perfringens


29.4. The necrotic jejunal villi from a piglet with pera- type C enteritis. The mucosa is completely necrotic, and
cute C. perfringens type C infection are covered by nu- there is emphysema of the submucosa and tunica muscu-
merous gram-positive bacilli (Gram). laris (H&E).
CHAPTER 29 CLOSTRIDIAL INFECTIONS Taylor 399

C enteritis to be made. Hemorrhagic diarrhea in young


piglets, the pattern of mortality, and the presence of the
hemorrhagic, necrotic lesions in the small intestine are
the most important features on which to base this diag-
nosis. The presence of emphysematous sections of the
small intestine in freshly dead piglets is also significant.
Diagnosis of the chronic disease is more difficult and
may depend upon a history of previous infection in the
herd, the elimination of other causes of necrotic enteri-
tis, and laboratory confirmation of the presence of the
agent in the lesions. Coccidiosis (Isospora suis) and other
causes of villous atrophy such as rotavirus, TGE, and
porcine epidemic diarrhea may all cause lesions which
are colonized by bacteria, including C. perfringens type C
(Bergeland 1977). C. perfringens type A (see below) may
also cause clinical signs and lesions resembling those of
the subacute and chronic forms and may be distin-
guished from them only by laboratory means.
29.7. Subacute form of C. perfringens type C enteritis The diagnosis can be supported in the laboratory by
in a 6-day-old piglet. The entire jejunum is lined with a the examination of smears of the intestinal contents and
necrotic membrane. mucosal lesions and histological sections of the intesti-
nal wall for the presence of large, gram-positive rods.
The microscopic appearance of the hemorrhagic lesions
Chronically affected pigs may have lesions resem-
is characteristic and is almost pathognomonic. Diagno-
bling those described above, but they may not be obvi-
sis can be confirmed beyond doubt by the demonstration
ous from the serosal surface of the intestine. There may
of the beta toxin of C. perfringens type C in supernates of
be local thickening of the intestinal wall and local, well-
the hemorrhagic intestinal contents and sometimes in
defined areas to which necrotic membrane is adherent.
peritoneal fluid. Mouse inoculation and protection tests
These areas may only be 1–2 cm in length and consist of
using type-specific antisera have been used to detect the
areas in which the mucosa has been replaced by a necrot-
toxin, but ELISA tests using the beta toxin as antigen are
ic membrane with a variety of bacteria along the deep
now more frequently used (Havard et al. 1992). PCR
edge. Deeper layers of the intestinal wall show evidence
methods that use primers to detect alpha, beta, epsilon,
of chronic inflammation (Fig. 29.8).
and enterotoxins have also been described (Buogo et al.
1995) and reveal that, although many animals with clin-
Diagnosis
ical signs and postmortem findings of C. perfringens type
The clinical signs and gross postmortem findings are suf-
C contain organisms producing the beta toxin (types B
ficient for a presumptive diagnosis of C. perfringens type
and C), some contain only the alpha toxin, which may be
caused by type A.
In more chronic cases and in some acute ones the tox-
in cannot be identified, and less satisfactory confirma-
tion can be obtained by isolating C. perfringens type C
from scrapings of the intestinal mucosa by direct inocu-
lation of blood agar plates and identification and toxin
typing of the colonies isolated. This method of confir-
mation is less satisfactory because the organism has been
found as a secondary agent colonizing the lesions of
piglets with TGE and other virus diseases. When the dis-
ease is chronic, the organism may be impossible to iso-
late, and in those situations, earlier cases of the disease
must be sought in the herd to confirm a diagnosis.

29.8. Ileum of a piglet with chronic C. perfringens


Treatment and Prevention
type C enteritis. The mucosa is replaced by a necrotic There is little to be done once clinical signs are present,
membrane containing a variety of bacterial species. The because the intestinal damage is already extensive
submucosa, tunica muscularis, and serosa are infiltrated (Szabo and Szent-Iványi 1957; Høgh 1967b). Even when
by chronic inflammatory cells (H&E). the organism is eliminated, the animal still dies or re-
400 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

mains stunted. Prophylaxis is of more value than thera- identified in this species (Rood et al. 1985).
py. Prevention of the disease on a more permanent basis
The disease can be prevented by passive immuniza- relies on the vaccination of the sow with C. perfringens
tion with antitoxin to C. perfringens type C in litters of type C toxoid on two occasions, the first at service or
nonimmune sows in an outbreak. Parenteral injection of midgestation and the second 2–3 weeks before farrow-
antitoxin should be carried out as soon after birth as pos- ing. Provided the piglets suck colostrum, they will be
sible because the disease may already be occurring in protected (Ripley and Gush 1983). The level of passive
piglets only a few hours old. Oral antimicrobials such as antibody in the piglet is related to that in the sow at far-
ampicillin may also be given as soon after birth as possi- rowing. Levels can vary markedly from 4.5 IU/mL to 123
ble and should prevent the development of the disease. IU/mL (Matishek and McGrinley 1986). Booster injec-
Treatment should be repeated daily for the first 3 days of tions should be given about 3 weeks before subsequent
life. Recent studies suggest that resistance to antimicro- farrowings. Toxoid may also be of value in protecting
bials commonly used on a farm may develop in C. per- weaned pigs (Meszaros and Pesti 1965).
fringens, and tetracycline resistance plasmids have been

CLOSTRIDIUM PERFRINGENS TYPE A ENTERITIS


C. perfringens type A has been isolated from the intestines Epidemiology
of pigs for many years and has been considered to form The organism is ubiquitous in gut contents and in soil.
part of the normal flora (Mansson and Smith 1962). Ev- Spores can survive freezing and boiling for up to 10 min-
idence has gradually accumulated that it is involved in utes, but the vegetative cells are fully susceptible to heat.
enteric disease, both in neonatal piglets and in older pigs The epidemiology of C. perfringens type A is not com-
in the postweaning period (Jestin et al. 1985). The or- pletely known. The organism can be isolated on all
ganism is present in every pig and is distributed world- farms, and antibody is widespread in finishing pigs and
wide. Reports of its involvement in clinical disease or the in sows (Estrada Correa and Taylor 1989). Outbreaks of
reproduction of disease have come from Germany disease in litters and on farms appear to be associated
(Amtsberg et al. 1976), France (Ramisse et al. 1979), the with organisms with similar cultural characteristics, but
United Kingdom (Olubunmi 1982), the Netherlands no detailed studies have yet been carried out to deter-
(Nabuurs et al. 1983), and Romania (Secasiu 1984) in re- mine whether these are individual strains. It must be as-
cent years, and the syndrome is increasingly being recog- sumed that the major source of the organism for pigs
nized worldwide. housed intensively is pig feces, but the organism can be
demonstrated in some diets and in the environment.
Etiology Enterotoxigenic strains are capable of causing hu-
C. perfringens type A resembles C. perfringens type C, but man food poisoning, usually following growth in slowly
colonies are often surrounded by a double zone of he- cooling joints of meat following cooking. It is not known
molysis, the outer of which is caused by the alpha toxin whether food poisoning strains found in cooked pork are
and the inner by theta toxin. The organism does not pro- the same as those causing disease in the live animal, al-
duce beta toxin or the other major toxins used in typing, though it is considered that the enterotoxin gene is chro-
but some strains produce a powerful enterotoxin on mosomal in human strains and plasmid borne in animal
sporulation. An ability to sporulate is generally consid- strains.
ered a prerequisite for enterotoxin production. Spores
may require heat shocking to germinate. Enterotoxin- Pathogenesis
producing strains rarely produce large amounts of alpha Infection with C. perfringens type A occurs in most piglets
toxin, and there appear to be two separate syndromes as- within a few hours of birth, and the organism can be
sociated with the two types of C. perfringens type A. Both demonstrated in the first feces following the clearance of
sporulating and enterotoxin-producing strains of C. per- meconium. In nonimmune piglets infected with patho-
fringens type A have been demonstrated in pigs (Estrada genic strains, numbers build up in the ileum and je-
Correa and Taylor 1989). Damme-Jongsten et al. (1990) junum to levels of 108–109 per gram of contents. Similar
failed to demonstrate the gene in isolates of the organ- levels may be found in large-intestinal contents and fe-
ism, but it was demonstrated at low levels in normal fe- ces. Vegetative forms produce alpha toxin and possibly
ces (fewer than 10 cells per gram) by Miwa et al. (1997) other toxins which cause necrosis of the intestinal ep-
using a most-probable-number method combined with ithelium in experimental infections in vivo. Consistent
the nested PCR. changes were not produced by the inoculation of gut
CHAPTER 29 CLOSTRIDIAL INFECTIONS Taylor 401

loops with purified alpha toxin (Estrada Correa 1986), with superficial necrosis and accumulations of fibrin. Al-
but Johannsen et al. (1993b) detected slight edema of the though there is capillary dilatation, the hemorrhage seen
villi when 6-hour-old piglets were given 80–800 mouse in C. perfringens type C infections is absent. There may al-
lethal doses. Johannsen et al. (1993c) confirmed previous so be an inflammatory colitis. Gram-positive rods may
observations (Estrada Correa 1986) that invasion and at- be seen adjacent to the mucosa in Gram-stained sections
tachment did not appear in experimental cases of the but they do not appear to be attached to the mucosa (Jo-
disease. Sporulating forms produce enterotoxin, which hannsen et al. 1993c). In the weaned pig, there may be
causes dramatic villous necrosis and outpourings of flu- some villous atrophy in the ileum and superficial colitis,
id into the intestinal lumen. The toxin fixes to the cells of but in some cases the mucosa appears normal.
the colonic epithelium and may be responsible for failure Clostridia are obvious in smears of feces or gut con-
to resorb water. Antibody to both toxins is present in tents, and large numbers of spores are present when en-
colostrum, and disease associated with enterotoxin is terotoxin-producing strains are involved. C. perfringens
commonly seen in weaned pigs aged 5–7 weeks after ma- type A can be isolated from the small intestine in profuse
ternal antibody disappears (Estrada Correa 1986). culture in affected piglets and from the large-intestinal
C. perfringens type A also colonizes existing lesions. contents in weaned pigs. Sporulating bacteria can be iso-
lated by heating samples to 80˚C for 10 minutes before
Clinical Signs culture.
Piglets develop a creamy or pasty diarrhea within 48
hours of birth and lose condition in infections with veg- Diagnosis
etative forms of C. perfringens type A. Affected piglets ap- The clinical signs of disease in piglets resemble those of
pear hairy and have fecal staining of the perineum. mild C. perfringens type C infection. There is little or no
There is no fever and animals rarely die. If they do, there blood in the feces and there is little or no mortality. The
is often discoloration of the abdominal skin similar to condition can be confirmed by isolation of the organism
that seen in C. perfringens type C infections. Diarrhea in profuse culture, by the demonstration of toxin in gut
lasts for up to 5 days, and feces on the pen floor appear contents by ELISA, by the demonstration of the alpha
mucoid and may be pink in color. Recovered piglets re- toxin gene by PCR (Buogo et al. 1995), and by failure to
main in poor condition for some time. These clinical demonstrate the presence of other agents. In weaned
signs have been reproduced experimentally by Olubun- pigs the organism rarely occurs alone, and a diagnosis of
mi (1982), Estrada Correa (1986), and Johannsen et al. uncomplicated C. perfringens type A infection requires
(1993a), who also recorded submandibular edema. In- the failure to demonstrate other enteropathogens cou-
fections caused by the sporulating form cause a transient pled with the presence of high numbers of sporulating
watery diarrhea which may last for only 24–48 organisms and the demonstration of preformed entero-
hours. toxin in fecal filtrates by reverse passive agglutination
Weaned pigs 5–7 weeks of age develop diarrhea or tests or counter-immunoelectrophoresis (CIE). Toxin lev-
soft feces lasting 4–7 days. This diarrhea does not result els of 1:32 were considered significant by Jestin and
in death, but affected animals lose condition and become Popoff (1987) and Popoff and Jestin (1985), and maxi-
covered with feces. The rate of daily liveweight gain is mum levels of 1:160 were recorded.
depressed. Onset of the diarrhea can be delayed by the
use of antimicrobial feed additives that affect clostridia. Treatment
The clinical signs may form part of the “colitis” complex The disease in piglets can be treated by using antimicro-
(see Chapters 40 and 42). bials in the same way as with C. perfringens type C infec-
tion, but with more likelihood of success. Vaccination is
Lesions not possible, as current commercial clostridial vaccines
Piglets are usually dehydrated, with fecal staining of the rarely include alpha toxin or enterotoxin. Experimental
perineum. The small intestine is flaccid and thick-walled, vaccination of the sow has shown that vaccination is pos-
with pasty contents and no blood. Its mucosa is mildly sible, but the ubiquitous nature of the organism means
inflamed and necrotic material may be adherent to it. that it is usually sufficient to ensure that piglets ingest
The absence of villi is obvious using a hand lens. The sufficient colostrum. In weaned pigs it is usually suffi-
large intestine is distended with whitish, pasty contents. cient to include a growth promoter such as avoparcin
The mucosa is often normal or may be coated in necrot- (Taylor and Estrada Correa 1988) or salinomycin (Kyri-
ic debris. In weaned pigs there are few if any gross le- akis et al. 1995) in the ration at a level capable of inhibit-
sions in the mucosa, but there may be frothy, mucoid ing the organism. Madsen (1995) has described the use
contents in both the ileum and the cecum. of bacitracin methylene salicylate in the treatment of C.
Microscopic lesions in piglets include villous atrophy perfringens type A enteritis in piglets.
402 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

CELLULITIS AND GAS GANGRENE


Infected wounds in swine often contain bacteria of sev- development of pulmonary edema (Kellaway et al.
eral species. When the histotoxic clostridia become es- 1941). On the basis of this evidence, it could be speculat-
tablished in a wound, however, they rapidly become the ed that the pulmonary edema and the serofibrinous exu-
most predominant and significant pathogen. These dates found in the pericardial and pleural cavities of
clostridial wound infections are highly fatal and are some cases of malignant edema in swine are the result of
characterized by intense acute inflammation with abun- bacterial toxemia.
dant edema and varying amounts of gas and local tissue
necrosis. The inflammation spreads rapidly from the pri- Clinical Signs
mary invasion site, and there terminal generalized sepsis C. septicum infection has an acute course and is often fa-
usually occurs. tal in less than 24 hours. Gross swelling may be located
C. septicum, C. perfringens type A, C. novyi, and C. in any area of the body. Common sites include the in-
chauvoei are the common causes of clostridial cellulitis guinal and ventral abdominal region, the head and ven-
and gas gangrene in swine. tral cervical area, and the shoulder. The swelling spreads
rapidly from the primary site, but it usually remains con-
CLOSTRIDIUM SEPTICUM INFECTION fined to one general region of the body. If the limbs are
(MALIGNANT EDEMA) involved, there is reluctance to bear weight on the affect-
ed leg. The skin overlying the swollen area has a blotchy
Etiology reddish-purple discoloration. Palpation of the swollen
C. septicum appears to be the most common etiologic area reveals pitting edema, and crepitation may also be
agent of clostridial cellulitis and gas gangrene of swine. evident late in the course of the disease. In the terminal
The organism is an anaerobic gram-positive rod, ap- stage, affected swine lie in lateral recumbency and com-
proximately 0.6–0.8 µm wide and 3–8 µm long, which monly make a groaning noise during forced expiration.
forms oval subterminal spores. It is known to produce
four toxins, of which the alpha toxin is hemolytic, necro- Lesions
tizing, and lethal. In addition, hyaluronidase, deoxyri- There is conspicuous swelling in the general region of
bonuclease, and an oxygen-labile hemolysin are pro- the primary infection site (Fig. 29.9). Incision of the ery-
duced (Moussa 1958). thematous skin overlying the infection reveals subcuta-
neous edema that may be colorless with focal hemor-
Epidemiology rhages or may be uniformly sanguinous. Gas is present in
C. septicum is a common potential wound contaminant varying quantities. The adjacent skeletal muscle may be
because it is a widespread inhabitant of the soil edematous, with essentially normal color, or may have
(MacLennan 1962). The incidence of malignant edema the features of typical gas gangrene, in which there are
is particularly high on certain premises that have had black, dry, and crepitant areas (Fig. 29.10). Affected mus-
large populations of livestock for many years, suggesting cle may have a butyric odor indistinguishable from the
that there is a buildup of spore numbers in the environ- characteristic odor of C. chauvoei infection in the rumi-
ment of these farms. nant. The regional lymph nodes are enlarged and hem-
orrhagic and may be emphysematous. There is common-
Pathogenesis ly an acute fibrohemorrhagic peritonitis. The spleen is
Since C. septicum infection nearly always involves the only slightly enlarged. There is moderate pulmonary
skin and subcutis, it appears that most cases result from edema and congestion. Varying amounts of amber fluid
perforated wounds, even though evidence of a wound and fibrin may be found in the pleural cavity and peri-
may sometimes not be found. Tissue damage at the site cardial sac.
of inoculation favors the initial establishment of the in- Postmortem decomposition occurs rapidly, and sub-
fection. It is probable that the local lesion is largely the cutaneous gas accumulates progressively until the sub-
result of the necrotizing effect of the alpha toxin. It has cutis of the entire carcass is emphysematous. Focal post-
been proposed that the hyaluronidase produced by C. mortem lysis of the liver is commonly seen, with
septicum causes disappearance of the endomysium resulting grayish-tan foci being evident within several
(Aikat and Dible 1960), which may aid the spread of the hours after death. As postmortem decomposition pro-
infection through muscle. gresses, the foci become confluent, giving the liver a uni-
Toxemia undoubtedly is a major factor in causing form tan color with numerous gas bubbles.
death of the animal. Experimental intravenous infusion Microscopically, there is edema of the subcutis that
of C. septicum toxins in the cat causes a specific constric- contains large numbers of degenerating acute inflamma-
tion in the coronary and pulmonary circulations with the tory cells and bacteria. Septic thrombi in subcutaneous
CHAPTER 29 CLOSTRIDIAL INFECTIONS Taylor 403

29.11. Acute septic thrombophlebitis involving the sub-


cutaneous vein of a pig with C. septicum infection. The
thrombus contains many long slender rods and degener-
ating leukocytes (H&E).

29.9. C. septicum infection. The grossly swollen area


involves the entire left rear leg and extends cranially to
the umbilicus. The overlying skin has a blotchy reddish-
purple discoloration.

29.12. C. septicum infection of the rear leg of a pig.


Skeletal muscle fibers are undergoing fragmentation and
lysis. The adjacent connective tissues are edematous and
emphysematous and contain bacteria and degenerating
inflammatory cells (H&E).

29.10. Rear leg of a pig with C. septicum infection.


There is prominent subcutaneous edema. The infection the affected subcutis or muscle. Fluorescent-labeled anti-
extends into the ham, which has foci of black, dry body staining (Batty and Walker 1963) of direct impres-
necrotic muscle. sion smears of the local lesion is a rapid and accurate
method of positively identifying C. septicum. An alterna-
tive method is isolation of the organism by anaerobic
veins and lymphatics are commonly found (Fig. 29.11). culture and identification by biochemical tests; however,
Affected skeletal muscle fibers undergo coagulation this is more time-consuming and less reliable than the
necrosis with fragmentation and lysis, and bacteria are use of immunofluorescence (Martig 1966). The swarm-
readily found between the degenerating muscle fibers ing nature of C. septicum on commonly used media
(Fig. 29.12). means that small numbers of the organism may appear
following postmortem contamination and result in a
Diagnosis false-positive diagnosis.
A presumptive diagnosis is made by observation of typi-
cal gross lesions in the freshly dead animal. Laboratory Treatment and Prevention
confirmation is based on pathologic findings, together Treatment with antibiotics may be successful if given
with exclusion of other diseases, and identification of early in the course of the disease. There is little specific
the organism. Many bacteria are seen on direct smears of information available of the therapy of malignant edema
404 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

in swine. Zeller (1956) mentions the recovery of two pigs necrosis. Gangrene of the uterus and decomposition of
after treatment with oxytetracycline. Experimentally, the its contents may follow dystocia and attendant interfer-
prophylactic use of tetracyclines, penicillin, or chloram- ences in the sow. A foul-smelling, reddish, watery vulval
phenicol prevented the disease in mice; however, their ef- discharge may be seen in life, and the animal dies within
fectiveness was greater when administered locally at the 12–24 hours. The uterus and its contents are usually
inoculation site than when given systemically (Taylor dark green or black and contain gas bubbles which give
and Novak 1952). off a foul odor. There may be foul-smelling reddish fluid
Prevention of the disease involves good sanitation in the peritoneal cavity. Decomposition of the remainder
and the prevention of injuries. Sharp objects that may of the carcass is rapid, and lesions in other sites are rarely
cause perforating wounds should be removed from the identified.
environment. In many instances C. septicum infection is a The alpha toxin (lecithinase) of C. perfringens has
sequel of hypodermic injections in swine; therefore, ade- been shown to be almost solely responsible for the pro-
quate sanitary procedures should be followed when mak- duction of the local lesion (Aikat and Dible 1956). It is
ing injections or performing surgery. On premises where proposed that the lecithinase causes necrosis by action
there are recurrences of the disease, immunization with on lipoprotein complexes of cell membranes. In addi-
C. septicum bacterin could be considered; however, this is tion, there is evidence that the mu toxin (hyaluronidase)
seldom done in swine. causes separation of the muscle sarcolemma from the en-
domysium.
CLOSTRIDIUM PERFRINGENS TYPE A The diagnosis is based on clinical and pathologic
INFECTION (GAS GANGRENE) findings, together with the isolation and identification of
C. perfringens. In mixed infections, direct Gram-stained
C. perfringens is occasionally involved in wound infec- smears of the lesion are helpful in estimating the relative
tions in swine. The infection is acute and highly fatal and numbers of different bacteria present. C. perfringens is
usually of only sporadic incidence. evident as a robust gram-positive bacillus that seldom
A high herd incidence of C. perfringens gas gangrene contains spores. Isolation of the organism is easily ac-
is occasionally seen in young piglets as a complication of complished by anaerobic incubation for 18–24 hours of
injection of iron-containing preparations used for the blood agar and egg yolk agar plates streaked directly
prevention of nutritional anemia. Circumstantial evi- from the lesion.
dence suggests that injection of iron preparations creates Treatment of C. perfringens infections with antibiotics
a microenvironment in tissue that favors the growth of may be successful if instituted early in the course of the
C. perfringens. Jaartsveld et al. (1962) reported the occur- disease. Experimentally, penicillin injections given at the
rence of C. perfringens infection in two herds following same time as inoculation of C. perfringens in mice gave al-
intramuscular injection of an iron preparation. Twelve of most complete protection; however, a delay in penicillin
25 piglets in one herd died the morning following injec- injection of over 3 hours appreciably lowered the sur-
tion. It was assumed that contaminated hypodermic nee- vival rate (Hac and Habert 1943). Jaartsveld et al. (1962)
dles were the source of the infection. A similar problem mention the recovery of some clinically sick pigs follow-
was seen by Bergeland (Taylor and Bergeland 1992) in ing penicillin injection.
several herds in which C. perfringens occurred as a pure Prevention of C. perfringens gas gangrene in swine in-
infection in some cases and as a mixed infection with E. volves preventing the occurrence of deep, contaminated
coli and staphylococci in others. When the problem oc- wounds and prompt treatment of any such wounds with
curs, the herd incidence is usually high, with mortality systemic penicillins or antimicrobials other than amino-
approaching 50%. glycosides.
The affected piglets have marked swelling of the en-
tire rear limb that was injected, and the swelling extends CLOSTRIDIUM CHAUVOEI INFECTION
cranially to the umbilical area. The skin overlying the (BLACKLEG)
swollen area has a dark reddish-brown discoloration. In-
cision of the affected area reveals extensive edema, and The pig is generally considered to be quite resistant to C.
there may be a copious quantity of gas in the muscle and chauvoei infection, and historically there have been very
subcutis. The inflammatory exudate is brownish red, due few substantiated reports of this disease in swine. Sterne
largely to staining by the injected iron preparation. The and Edwards (1955) reported the occurrence of blackleg
lesion usually has a putrid odor. Postmortem decompo- in swine kept under very poor hygienic conditions in a
sition occurs rapidly, and the livers of pigs dead more lot where there previously had been high losses of cattle
than a few hours may have conspicuous gray foci of lysis from blackleg. Four pigs were examined, 2 of which were
that surround minute gas bubbles. Microscopically, found to be infected with C. chauvoei and 2 with C. sep-
acute thrombophlebitis may be evident, and affected ticum. Gualandi (1955) reported an outbreak of C. chau-
muscle fibers undergo fragmentation and liquefaction voei infection in which 15 of 34 pigs died in 2 days. Ede-
CHAPTER 29 CLOSTRIDIAL INFECTIONS Taylor 405

ma of the pharynx was a constant finding. Eggleston orrhage on the surface of the kidney. They reported the
(1950) described the losses of 3 pigs weighing 100–140 loss 4 days after farrowing of an adult sow that showed
pounds (45–63 kg) that had been fed a blackleg calf car- marked decomposition and the presence of massive
cass. There was swelling of the face and throat region, in- numbers of C. novyi in the internal organs. A further case
cluding the ears, and the odor of the lesions resembled report by Bourne and Kerry (1965) involved 4 cases of
rancid butter. Two large outbreaks were described from sudden death in sows on grass. Necropsy findings in-
Zimbabwe by Mavenyengwa and Matope (1995) where cluded rapid postmortem tympany; submandibular
34 pigs on one farm and 36 on another developed swellings; bloodstained fluid in the pleural, pericardial,
swollen and edematous limbs with reddish discoloration and peritoneal cavities; serosal hemorrhages; splenic en-
of the overlying skin, fever of 40–41˚C, and edema and largement; and marked degeneration and emphysema of
dark, hemorrhagic muscles in the affected legs. the liver. C. novyi was demonstrated in various tissues, in-
C. chauvoei is a pleomorphic, anaerobic, gram-posi- cluding heart blood. An obvious feature of pigs that had
tive rod, measuring 0.5–1 µm by 3–8 µm, that readily died from this disease was the bronze color of the liver
forms central to subterminal spores. It produces several and the presence of large numbers of small gas bubbles
exotoxins, of which the alpha toxin is lethal, necrotizing, in the substance of the organ when cut. These are partic-
and possibly hemolytic. In addition, deoxyribonuclease, ularly significant when the remainder of the carcass is
hyaluronidase, and an oxygen-labile hemolysin are pro- fresh. A detailed study of 17 cases of C. novyi infection in
duced (Moussa 1958). sows (Duran and Walton 1997) confirms many of these
The pathogenesis of C. chauvoei infection in pigs is features.
not completely understood. It is assumed that the organ- C. novyi is an anaerobic, spore-forming, gram-positive
ism usually has an oral portal rather than being a wound rod that varies in size but generally is the largest of the
infection. It is postulated that the bacteria sometimes clostridia encountered in swine. The organism produces
may lie dormant in various tissues until there is a favor- highly potent exotoxins. The lethal, necrotizing alpha
able microenvironment for their growth. Tissue damage toxin is considered to be the principal toxin of type A
such as bruising may then be the factor that triggers the and B strains. The type involved in swine infections was
disease. Once bacterial growth occurs, the disease ap- identified as type B by Itoh et al. (1987), although Duran
pears to be a manifestation of the effects of bacterial tox- and Walton (1997) report the isolation of type A alone in
ins and pathologically may closely resemble C. septicum 3 cases, type B in 4 cases, and a mixed culture in another.
infection. The disease affects large finishing pigs and breeding
Because of the pathologic similarity between C. sep- stock, principally sows, and was recorded more fre-
ticum and C. chauvoei infections and the apparent higher quently in spring and in older sows of parity greater than
incidence of C. septicum infection in swine, a diagnosis of 4 in moderate to good condition by Duran and Walton
blackleg can be made only by bacterial identification. (1997) and was characterized by sudden death.
The fluorescent antibody test (Batty and Walker 1963) A positive diagnosis of C. novyi infection in swine is
applied to direct impression smears of infected tissue is difficult, since suspect cases are usually found dead, and
a rapid and practical method of identification. Isolation some interval of time elapses between death and necrop-
by anaerobic culture may be difficult in decomposing sy. The organism is a common and early postmortem in-
specimens since C. chauvoei is relatively fastidious and vader, especially of adult swine in warm weather. There-
easily overgrown by other bacteria such as C. septicum. fore, a detailed examination to exclude other possible
The prevention of C. chauvoei infection involves min- causes of death must always be made. The disease
imizing exposure to the organism. Even though C. chau- should be suspected when there is a history of sudden
voei has not been demonstrated to be a common soil or- death, together with typical necropsy findings. Animals
ganism, circumstantial evidence from the few reports of that have died from C. novyi infection are generally in
the disease in pigs suggests that keeping swine on known good condition, with surface changes of congestion,
contaminated premises or allowing them to eat carcasses swelling, and tympany associated with the rapid decom-
of ruminants dead of blackleg are factors in its inci- position of the carcass. A serous or frothy exudate may
dence. be present at the nostrils. There is subcutaneous edema,
particularly in the cervical and inguinal regions. Pul-
CLOSTRIDIUM NOVYI INFECTION monary edema and tracheal froth, serofibrinous or
(SUDDEN DEATH) serosanguinous exudates in pericardial and pleural cavi-
ties, and unusually rapid decomposition with accumula-
C. novyi may cause sudden death in swine. Batty et al. tion of gas in the liver are all commonly found. The pres-
(1964) reported the sudden death of 12 pigs over a 3- ence of gas bubbles in the liver in an otherwise fresh
week period. Postmortem decomposition progressed un- carcass is particularly significant. The stomach is often
usually rapidly. The lungs were congested, the trachea full and there may be congestion of the spleen (Duran
contained bloodstained froth, and there was some hem- and Walton 1997).
406 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

The organism is rapidly identified by fluorescent an- swine awaits further study of the disease. The disease can
tibody staining of direct smears of infected tissue. Isola- be controlled by reducing the incidence of pneumonias,
tion and typing of the organism are difficult since it has metritis, and enteritis in affected groups of pigs, coupled
the most fastidious growth requirements of the with vaccination of finishing pigs and sows against the
clostridia commonly encountered in swine, but isolation disease using alum-adjuvanted vaccines such as those
has been described by Itoh et al. (1987) and Duran and readily available for sheep. Marco (1995) reported using
Walton (1997). Isolation may be improved by the use of zinc bacitracin at 200 ppm and twice daily feeding to re-
alcohol and heat treatment of specimens followed by cul- duce mortality in an affected sow population. The
ture on prereduced fastidious anaerobe agar. The organ- prompt disposal of carcasses by incineration or deep
ism may be recovered from the livers of some apparent- burial may reduce the contamination of the environ-
ly healthy animals on infected farms. ment by spores.
Information on pathogenesis and epidemiology in

TETANUS
Tetanus is a disease characterized by uncontrollable fect in the normal barriers to infection, usually via a deep
spasms of voluntary muscles caused by the toxin of C. skin wound. Since spores are commonly present in soil,
tetani elaborated at the site of a deep infection. Swine of any contaminated wound should be regarded as a possi-
all ages may be affected; however, the majority of cases ble site of C. tetani infection. Tetanus bacilli have little or
involve young pigs, usually as a complication of either no invasive ability and tend to remain localized at the
castration wound infection or umbilical infection. primary site of infection.
C. tetani is a slender, anaerobic, gram-positive rod C. tetani is a strict anaerobe; thus, its growth requires
measuring 0.3–0.6 µm by 2–8 µm that characteristically a microenvironment with lowered oxidation-reduction
forms terminal spores. Its major lethal toxin, tetanospas- potential. Deep foci of devitalized tissue provide a suit-
min, is a highly potent neurotoxic protein. An oxygen-la- able environment for growth of the organism. Such foci
bile hemolysin, tetanolysin, is also produced. may be formed by traumatic tissue injury, the presence of
foreign bodies, or infection by histotoxic bacteria intro-
Epidemiology duced during or subsequent to wounding. In swine the
C. tetani is a common inhabitant of the soil. Sergeeva and most commonly reported location of tetanus infection is
Matveev (1966) conducted a survey of 5338 soil samples castration wounds. Sakurai (1966) reported that 202 of
collected from various regions of the former USSR and 220 cases in Japan in which the infection site was known
found that the highest level of soil contamination was in were postcastration infections. Kaplan (1943) reported
the southern regions having fertile black soil, long peri- the loss of 60 of 250 young pigs from tetanus; 40 cases
ods of growth for plants and soil organisms, and highly were postcastration and 20 probably were umbilical in-
developed agriculture and cattle breeding. In some re- fections. Wounds inflicted by unclipped canine teeth as
gions the rate of isolation was as high as 40–62% of the well as infected dental alveoli during eruption of teeth
samples examined. In areas where the soil was not fertile, have also been suggested as possible sources of infection
regions with concentrated cattle raising and fields fertil- (Morrill 1964).
ized with animal manure had a significantly higher inci- The incubation period (interval between establish-
dence of soil contamination than regions without live- ment of the infection focus and onset of clinical signs)
stock. Tetanus morbidity in various areas of the former ranges from several days to several weeks. In general, cas-
USSR correlated directly with the prevalence of the or- es with a short incubation period run a more acute and
ganism in the soil. Sakurai (1966) also reported a high in- fulminating course with a higher fatality rate than cases
cidence of tetanus in areas of Japan where stock farms with a long incubation period.
and pastures had been located since early times. Toxin-containing vesicles pass by retrograde axonal
transport along the motor nerve fibers from neuromus-
Pathogenesis cular junctions at the site of the injury to act on the in-
The development of tetanus is dependent upon the pres- hibitory neurons in the ventral horn of the spinal cord to
ence of C. tetani in tissue in an environment that will per- inhibit their activity. Reflex arcs are potentiated and
mit its growth and toxin production, and the toxin tetany occurs. The toxin consists of a large molecule of
formed must subsequently reach the central nervous sys- 140–170 kDa that is split by protease to give a light chain
tem (CNS) in sufficient quantity to produce overt dis- that is enzymatic (a zinc-dependent endopeptidase) and
ease. The organism gains entrance to the tissue via a de- a heavy chain that binds to receptors. The L chain acts by
CHAPTER 29 CLOSTRIDIAL INFECTIONS Taylor 407

cleaving proteins involved in the exocytosis of neuro- such as a castration wound or umbilical abscess is appar-
transmitters by neurons. Tetanus toxin cleaves synapto- ent in many cases. Direct Gram-stained smears of exu-
brevin at the neuromuscular junction as well as having date from the lesion may reveal bacteria with typical C.
an effect in the CNS, but this peripheral action is not clin- tetani morphology, that is, slender gram-positive bacilli
ically apparent in affected pigs. with terminal spherical spores (“drumstick” forms)
Many factors may contribute to the cause of death. among the bacterial flora. The organism may be isolated
The consequences of prolonged recumbency and depri- by anaerobic culture or can be identified by immunoflu-
vation of nutrients may be factors in animals with a rela- orescence (Batty and Walker 1964); however, this is usu-
tively long survival time. In acute cases, respiratory fail- ally not necessary if there is adequate antemortem clini-
ure resulting from severe skeletal muscle spasms is likely cal observation of the affected animals.
to be the single most important factor. Whether or not
the toxin produces a specific metabolic lesion that is di- Treatment and Prevention
rectly involved in causing death in the pig is not known. The prognosis in affected swine is poor, and there is little
evidence that treatment by currently practical methods
Clinical Signs and Lesions is of real benefit. Mihaljevic (1966) reported that all 6
The clinical features of tetanus relate to spasms of skele- tetanus cases submitted to the clinic at Zagreb during
tal muscle which occur in the generalized form in swine. 1948–1965 succumbed to the disease. Kaplan (1943) de-
The earliest sign is a stiffened gait. The disease progress- scribed 4 recoveries of 60 affected pigs; however, the re-
es rapidly and usually is fully developed in 1–2 days. As covered cases may have been mild and were not neces-
the disease progresses, the pig has difficulty walking, the sarily associated with therapy. Only 11 of 240 cases
ears are erect, the tail tends to be extended straight out described in a report from Japan survived the disease
posteriorly, the head is slightly elevated, and there may (Sakurai 1966). Various suggested treatments include re-
be some protrusion of the nictitating membrane. Fur- opening castration wounds and flushing them with hy-
ther progression of the disease renders the pig incapable drogen peroxide, administration of antitoxin in an at-
of walking, and the skeletal muscles are very firm on pal- tempt to neutralize toxin not already fixed by nervous
pation. Ultimately the pig lies in lateral recumbency in tissue, administration of antibiotics, and the use of tran-
opisthotonus, with both thoracic and pelvic limbs ex- quillizers or barbiturates as muscle relaxants.
tended and directed posteriorly (Fig. 29.13). The tetanic Since there is no practical way to eliminate the spores
spasms are noticeably heightened by sudden sensory of C. tetani from the soil, control is directed toward the
stimuli such as noise, touch, or motion of a visible ob- prevention of wound contamination by soil or feces.
ject. Terminally, there are tachycardia and increased res- Good sanitation in the farrowing house, treatment of the
piration rate, and white froth may be present around the umbilical cord with antiseptics soon after birth, and
mouth and external nares. prompt clipping of the canine incisors are recommend-
No lesions specific for tetanus are found at necropsy. ed preventive measures against neonatal tetanus. Sharp
Conspicuous abrasions of the skin over pressure points objects that may cause skin wounds should be removed
may be seen, and there may be pulmonary congestion from the environment. Because most tetanus in swine
and edema. follows castration, particular emphasis should be placed
on proper surgical technique, with the provision of clean
Diagnosis quarters for the pigs after castration to prevent undue
The diagnosis of tetanus in swine is based on observa- contamination of the castration wound by soil or
tion of typical clinical signs. An obvious area of infection feces.
If reasonable preventive measures cannot be fol-
lowed or if valuable animals are wounded, passive im-
munization with tetanus antitoxin, the prophylactic use
of antibiotics, and/or active immunization with tetanus
toxoid may be indicated. Veronesi (1966) concluded that
the prophylactic use of large doses of long-acting peni-
cillin or tetracyclines (or repeated injections of short-act-
ing preparations of these drugs for 5 days) may be supe-
rior to antitoxin in preventing experimental tetanus in
mice if treatment is instituted within a few hours after in-
fection. An appreciable amount of active immunity may
be obtained from a single injection of alum-precipitated
29.13. Generalized tetanus in a 10-day-old pig that tetanus toxoid, and excellent protection for a year or
apparently resulted from umbilical infection. The ears more can be expected if three doses are given several
are erect and the limbs are rigidly extended. weeks apart (Morrill 1964).
408 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

BOTULISM
Botulism is a toxicosis characterized by rapidly progres- (Doiurtre 1967). Type C strains were incriminated in
sive flaccid paralysis caused by the toxins of C. botulinum. these cases.
The toxin is produced by the organism as it grows in de- The eating habits of nonconfined pigs should make
composing organic matter of animal or vegetable origin, them likely candidates for botulism. The very low inci-
and poisoning follows oral consumption of toxin-con- dence of the disease in swine is explained on the basis of
taining material by a susceptible animal. innate resistance. In 1919 Thom et al. reported that tox-
Swine are considered to be quite highly resistant to in produced by a strain isolated from spoiled canned as-
botulism, and there are few authentic reports of natural- paragus failed to affect a pig when administered orally.
ly occurring botulism in this species. The pig succumbed, however, when the toxin was inject-
ed subcutaneously. Dack and Gibbard (1926a) reported
Etiology similar findings. Two pigs fed 10 million and 7.5 million
The organism is an anaerobic, gram-positive bacillus, mouse lethal doses (LD) of type A toxin failed to develop
usually having a size range of 0.6–1.2 µm by 4–6 µm; signs. One of these pigs was later injected intraperi-
however, much longer forms may be found in some cul- toneally with a large dose of toxin and died 4 days later.
tures (Smith and Holdeman 1968). It forms oval, usually Dack and Gibbard indicated (1926b) that hog intestine
subterminal, spores. Growth requires rather strictly has a low permeability for the toxin. More recently,
anaerobic conditions and occurs within a wide range of Scheibner (1955) found young swine resistant to 1 mil-
temperatures up to body temperature but is perhaps op- lion mouse LD of types B, C, D, and E toxins given oral-
timal at about 30˚C. ly. A similar dose of type A toxin produced typical signs
The lethal toxin, a protein, is an extremely potent poi- and death; however, 60,000 mouse LD of type A toxin
son. There is variation among different strains of the or- had no effect.
ganism with respect to neurotoxin production. These dif- Smith et al. (1971) found weanling pigs to be moder-
ferences involve the antigenic properties of the toxin as ately susceptible to type B toxin when it was infused in-
well as the spectrum of animal species susceptible to the travenously, but they were highly resistant to the toxin
toxin. C. botulinum is currently divided into six types when given orally (oral:intravenous ratio = 16,700:1).
(A–F) on the basis of major toxin antigenic structure. The intravenous LD of type B toxin was approximately
Type C strains are further subdivided into types Ca and 180 mouse LD/kg body weight. This group of experi-
Cb, whose toxins are antigenically related but not identi- mental pigs was moderately resistant to intravenous in-
cal. Regardless of the type of toxin involved or the fusion of types A, Cb, E, and F toxins (LD = 4000–20,000
species of animal affected, however, all animals with bot- mouse LD/kg), at least moderately resistant to Ca toxin
ulism exhibit similar clinical signs. (LD = 18,000 mouse LD/kg), and highly resistant to type
D toxin (LD = 67,000 mouse LD/kg). They were highly
Epidemiology resistant to all types given orally.
C. botulinum is commonly present in soil throughout the
world. There are geographic variations in the incidence Pathogenesis
of the different types. The prevalence appears to be asso- Botulism is generally considered to be strictly a food poi-
ciated with the quantity of organic matter in the soil, and soning; that is, any significant quantity of the toxin is
factors such as fertilization with manure may increase elaborated in the foodstuff before it is eaten, and poi-
bacterial numbers. soning results from absorption of preformed toxin. The
The occurrence of botulism is dependent upon the bacteria are, of course, consumed along with the toxin,
consumption of a sufficient quantity of toxin by a sus- and it has been proposed that there may be further toxin
ceptible pig to produce disease, which implies that a po- production in the intestine.
tential foodstuff must provide an environment suitable Absorption of type A toxin was found to occur much
for growth and toxin elaboration by C. botulinum. The tis- more readily in the upper small intestine than in the
sues of dead animals, including crustaceans, fish, birds, ileum of rats, rabbits, and mice, and absorption from the
and mammals, that decompose during warm or hot stomach was poor in these species (May and Whaler
weather are the most common source of toxin for ani- 1958). These workers also found that the toxin was ab-
mals. Since botulism rarely occurs in swine, there are few sorbed by the lymphatics, with passage into the general
recorded sources of toxin for this species. The death of circulation by way of thoracic lymph rather than portal
five adult swine after eating dead fish from the edge of a blood.
partially dried-up lagoon was reported by Beiers and The toxin consists of a large molecule of 140–170
Simmons (1967). The loss of pigs being fed swill and de- kDa that is split by protease to give a light chain that is
composing brewers waste has also been reported enzymatic (a zinc-dependent endopeptidase) and a
CHAPTER 29 CLOSTRIDIAL INFECTIONS Taylor 409

heavy chain that binds to receptors. The L chain acts by mouse protection tests and employing type-specific diag-
cleaving proteins involved in the exocytosis of neuro- nostic antitoxins, but this does not appear to be the case
transmitters by neurons. Toxin types B, D, and F cleave in swine. On the 4 days following intravenous inocula-
synaptobrevin, types A and E act on SNAP-25, and type tion of a pig with 21,400 mouse LD of type A toxin/kg
C toxin acts on syntaxin, at the myoneural junction, pre- body weight, Smith et al. (1971) demonstrated 100, 100,
venting muscular contraction. Death is generally as- 30, and 10 mouse LD of toxin/mL serum. No toxin was
cribed to asphyxia resulting from paralysis of the mus- found in the serum of pigs 1 day after giving large doses
cles of respiration. of types B, Cb, or D toxin, and no toxin was demonstrat-
ed in urine from pigs inoculated with any of the types.
Clinical Signs Identification of type C toxin in filtrates of small-intes-
The latent period between consumption of toxic materi- tine content from a poisoned sow has been reported
al and onset of signs ranges from 8 hours to 3 days or (Beiers and Simmons 1967).
more. The clinical features are a manifestation of pro- The isolation and identification of C. botulinum may
gressive flaccid paralysis of voluntary muscles. The ini- also be of some value in establishing the diagnosis.
tial signs are weakness, incoordination, and staggering. Narayan (1967) reported recovery of C. botulinum from
Morrill and Bajwa (1964) stated that weakness often ap- swine that were silent carriers of the organism. Müller
pears in the forelegs of swine first, followed by involve- (1967) stated that type Cb organisms were isolated from
ment of the hindlegs. The paralysis may then progress to only 3–4% of the livers of healthy slaughtered cattle and
lateral recumbency with complete flaccidity. Smith et al. swine in Denmark, whereas this type was isolated from
(1971) observed an initial weakness of the pelvic limbs 90% of cattle and horses that had died from botulism.
and lumbar region, followed by general motor paralysis Yamakawa et al. (1992) found the organism to be wide-
and dilation of the pupils of the eyes. Other clinical signs spread in the livers, feces, and the environment of
include anorexia; lordosis; reduced vision or complete healthy pigs on infected farms. PCR using toxin gene se-
blindness; aphonia; excessive salivation; involuntary uri- quences has been used in other species but has not been
nation and defecation; and deep labored breathing reported in swine.
(Smintzis and Durin 1950; Beiers and Simmons 1967).
The time interval between onset of signs and death or Treatment and Prevention
recovery is variable and probably is largely determined If botulism is suspected, an effort should be made to find
by the amount of toxin consumed. Five adult swine that the toxin source and prevent further consumption of any
consumed decomposing fish died between 19 and 52 remaining suspect material by the herd.
hours after eating the fish, and 2 that staggered recov- The only specific treatment for botulism is the use of
ered later (Beiers and Simmons 1967). Smintzis and antitoxin. Antitoxins have been effective in reducing
Durin (1950) reported mortality on the sixth day of the mortality in humans when given after consumption of
disease. food suspected of containing toxin. It has been suggest-
No lesions specific for botulism are found at necrop- ed that antitoxin may be beneficial not only when given
sy. Significant findings might include the presence in the parenterally but when given orally as well in an attempt
stomach of the material suspected as the toxin source to neutralize toxin in the alimentary tract (Lamanna and
and the occurrence of aspiration pneumonia consequent Carr 1967). As pointed out by Burgen et al. (1949), the
to paralysis of the muscles of deglutition (Beiers and toxin appears to produce an irreversible neuromuscular
Simmons 1967). block; therefore, the principal benefit of antitoxin prob-
ably is to prevent additional fixation of toxin at my-
Diagnosis oneural junctions, thereby impeding the progressive
Because the pig apparently is quite highly resistant to severity of the disease. If antitoxins are to be of value,
botulism, a diagnosis should be made only after thor- they must contain antibodies to the specific type of tox-
ough investigation and exclusion of other possible diag- in involved. Therapy therefore indicates the use of poly-
noses. A presumptive diagnosis is based on observation valent antitoxins that incorporate the types most com-
of typical clinical signs, that is, a progressive flaccid monly present in a geographic area.
paralysis. Disclosure of a possible source such as spoiled Therapy aimed at reducing continued absorption of
canned goods or a decomposing animal carcass is also toxin from the intestine was suggested by Beiers and
helpful. Simmons (1967). They fed affected sows 1 gallon (4.5 L)
Methods of laboratory confirmation of the disease in of skim milk containing 4 ounces (120 g) of magnesium
swine are not clearly defined. Valuable confirmatory evi- sulfate, repeating this treatment three times at 12-hour
dence can be obtained by demonstrating the presence of intervals. Only 2 of 4 sows that were weak and staggering
the toxin in the suspected source material or in the poi- consumed the preparation, and both subsequently re-
soned animal’s gastrointestinal content or serum. Toxin covered, whereas the other 2 died. The use of sedatives
is readily detected in the serum of some species by also has been recommended.
410 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Prevention of botulism involves preventing the con- ———. 1967b. Necrotizing infectious enteritis in piglets
sumption of potentially toxic material such as spoiled caused by Clostridium perfringens type C. II. Incidence
garbage and decomposing animal tissue. Prophylactic and clinical features. Acta Vet Scand 8:301.
immunization with toxoids is not practical in swine be- ———. 1974. Porcine Infectious Necrotizing Enteritis
cause of the infrequent occurrence of the disease. Caused by Clostridium perfringens. Ph.D. diss. Royal Vet
Agric Univ, Copenhagen.
REFERENCES Kubo, M., and Watase, H. 1985. Electron microscopy of
Clostridium perfringens in the intestine of pigs with
Clostridium perfringens Type C Enteritis necrotic enteritis. Jap J Vet Sci 47:497–501.
Arbuckle, J. B. R. 1972. The attachment of Clostridium Matishek, P. H., and McGrinley, M. 1986. Colostral transfer
welchii (Cl. perfringens) type C to intestinal villi of pigs. J of Clostridium perfringens type C beta antitoxin in swine.
Pathol 106:65. Am J Vet Res 46:2147–2148.
Azuma, R.; Hamaoka, T.; Shioi, H.; Tanji, T.; Yamaguchi, H.; Matthias, D.; Illner, F.; and Bauman, G. 1968. Untersuchun-
and Shiga, K. 1983. Case report of necrotic enteritis in gen zur Pathogenese der Magen-Darm-Veranderungen
neonatal pigs caused by Clostridium perfringens type C. bei der infektiosen Gastroenteritis der Schweine. Arch
Nippon Juigaku Zasshi 45:135. Exp Veterinärmed 22:417.
Bakhtin, A. G. 1956. Dysentery of newborn piglets. Veteri- Meszaros, J., and Pesti, L. 1965. Studies on the pathogenesis
naiia (Moscow) 33:30. Abstr Vet Bull 26:562. of gastroenteritis in swine. Acta Vet Acad Sci Hung
Barnes, D. M., and Moon, H. W. 1964. Enterotoxemia in 15:465.
pigs due to Clostridium perfringens type C. J Am Vet Med Morin, M.; Turgeon, D.; Jolette, J.; Robinson, Y.; Phaneuf, J.
Assoc 144:1391. B.; Sauvageau, R.; Beauregard, M.; Teuscher, E.; Higgins,
Bergeland, M. E. 1965. Studies of a Porcine Enterotoxemia R.; and Lariviere, S. 1983. Neonatal diarrhea of pigs in
Caused by Clostridium perfringens Type C. Ph.D. diss. Quebec: Infectious causes of significant outbreaks. Can
Univ Minnesota. J Comp Med 47:11.
———. 1972. Pathogenesis and immunity of Clostridium Ohnuna, Y.; Kondo, H.; Saino, H.; Taguchi, M.; Ohno, A.;
perfringens type C enteritis in swine. J Am Vet Med Assoc and Matsuda, T. 1992. Necrotic enteritis due to Clostrid-
160:658. ium perfringens type C in newborn piglets. J Jpn Vet Med
———. 1977. Necrotic enteritis in nursing piglets. Proc Am Assoc 45:738–741.
Assoc Vet Lab Diagn 20:151. Plaisier, A. J. 1971. Enterotoxemia in piglets caused by
Bergeland, M. E.; Dermody, T. A.; and Sorensen, D. K. 1966. Clostridium perfringens type C. Tijdschr Diergeneeskd
Porcine enteritis due to Clostridium perfringens type C. I. 96:324.
Epizootiology and diagnosis. Proc US Livest Sanit Assoc Ripley, P. H., and Gush, A. F. 1983. Immunisation schedule
70:601. for the prevention of infectious necrotic enteritis caused
Buogo, C.; Capaul, S.; Haeni, H.; Frey, J.; and Nicolet, J. by Clostridium perfringens type C in piglets. Vet Rec
1995. Diagnosis of Clostridium perfringens type C enteri- 112:201.
tis in pigs using a DNA amplification technique (PCR). J Rood, J. I.; Buddle, J. R.; Wales, A. J.; and Sidhu, R. 1985.
Vet Med Ser B 42:51–58. The occurrence of antibiotic resistance in Clostridium
Field, H. L., and Gibson, E. A. 1955. Studies on piglet mor- perfringens from pigs. Aust Vet J 62:276–279.
tality. II. Cl. welchii infection. Vet Rec 67:31. Szabo, S., and Szent Iványi, T. 1957. Infectious necrotic en-
Field, H. L., and Goodwin, R. F. W. 1959. The experimental teritis in sucking pigs. II. Incidence and control of the
reproduction of enterotoxaemia in piglets. J Hyg (Camb) disease in Hungary. Acta Vet Acad Sci Hung 7:413.
57:81. Szent-Iványi, T., and Szabo, S. 1955. I. Clostridium welchii
Harbola, P. C., and Khera, S. S. 1990. Porcine enteritis due type C causing infectious necrotic enteritis in newborn
to Clostridium perfringens and its immunoprophylaxis. piglets. Magy Allatorv Lapja 10:403. Abstr Vet Bull
Proc Int Congr Pig Vet Soc 11:164. 26:259.
Havard, H. L.; Hunter, S. E. C.; and Titball, R. W. 1992. Walker, P. D.; Murrell, T. G. C.; and Nagy, L. K. 1980. Scan-
Comparison of the nucleotide sequence and develop- ning electron microscopy of the jejunum in enteritis
ment of a PCR test for the epsilon toxin gene of Clostrid- necroticans. J Med Microbiol 13:445.
ium perfringens type B and type D. FEMS Microbiol Lett Warrack, G. H. 1963. Some observations on the typing of
97:77–81. Clostridium perfringens. Bull Off Int Epizoot 59:1393.
Høgh, P. 1965. Enterotoksaemi hos pattegrise forarsaget of
Clostridium perfringens type C. Nord Vet Med 17:1. Clostridium perfringens Type A Enteritis
———. 1967a. Necrotizing infectious enteritis in piglets Amtsberg, G. W.; Bisping, W.; el-Sulkhon, S. N.;
caused by Clostridium perfringens type C. I. Biochemical Matthiesen, I.; and Krobischi, P. 1976. Clostridium per-
and toxigenic properties of the Clostridium. Acta Vet fringens type A infectie. Berl Münch Tierärztl Wochen-
Scand 8:26. schr 21:409–414.
CHAPTER 29 CLOSTRIDIAL INFECTIONS Taylor 411

Buogo, C.; Capaul, S.; Haeni, H.; Frey, J.; and Nicolet, J. Am J Vet Res 47:1132–1133.
1995. Diagnosis of Clostridium perfringens type C enteri- Ramisse, J.; Brement, A.; Poirier, J.; Rabreaud, C.; and Si-
tis in pigs using a DNA amplification technique (PCR). J monnet, P. 1979. Flore microbienne isolée au cours de
Vet Med Ser B 42:51–58. diarrhees neonatales mortelles chez le veau, l’agneau et
Damme-Jongsten, M. van; Haagsma, J.; and Notermans, S. le porcelet. Rev Vet Med 130:111–122.
1990. Enterotoxin gene not found in Clostridium perfrin- Secasiu, V. 1984. Diagnosticul bacteriologic welchiozei
gens isolates from pigs. Vet Rec 126:191–192. porcine (enterotoxiema anaeroba). Rev cresterea ani-
Estrada Correa, A. E. 1986. Studies of Clostridium perfringens malelor 2:38–45.
Type A Enteritis in the Pig. Ph.D. diss. Univ Glasgow. Taylor, D. J., and Estrada Correa, A. E. 1988. Avoparcin in
Estrada Correa, A. E., and Taylor, D. J. 1989. Clostridium per- the prevention of enterotoxigenic C. perfringens type A
fringens type A spores, enterotoxin and antibody to en- infections and diarrhoea in pigs. Proc Int Congr Pig Vet
terotoxin. Vet Rec 124:606–611. Soc 10:140.
Jestin, A., and Popoff, M. R. 1987. L’enterotoxine de
Clostridium perfringens de type A: Intérêt de son étude Cellulitis and Gas Gangrene
dans la pathologie digestive du porc a l’engrais. Rec de Aikat, B. K., and Dible, J. H. 1956. The pathology of
Med Vet 163:33–38. Clostridium welchii infection. J Pathol 71:461.
Jestin, A.; Popoff, M. R.; and Mahe, S. 1985. Epizootiologic ———. 1960. The local and general effects of cultures and
investigations of a diarrheic syndrome in fattening pigs. culture filtrates of Clostridium oedematiens, Cl. septicum,
Am J Vet Res 46:2149–2151. Cl. sporogenes, and Cl. histolyticum. J Pathol 79:277.
Johannsen, U.; Arnold, P.; Kohler, B.; and Selbitz, H. J. Batty, I., and Walker, P. D. 1963. Differentiation of Clostrid-
1993a. Untersuchungen zur experimentellen Clostridium ium septicum and Clostridium chauvoei by the use of fluo-
perfringens typ A enterotoxaemie der Saugferkel. rescent-labelled antibodies. J Pathol 85:517.
Monatsh für Veterinärmed 48:129–136. Batty, I.; Buntain, D.; and Walker, P. D. 1964. Clostridium
Johannsen, U.; Menger, S.; Arnold, P.; Kohler, B.; and oedematiens: A cause of sudden death in sheep, cattle
Selbitz, H. J. 1993b. Untersuchungen zur experi- and pigs. Vet Rec 76:1115.
mentellen Clostridium perfringens typ A enterotoxaemie Bourne, F. J., and Kerry, J. B. 1965. Clostridium oedematiens
der Saugferkel. Monatsh für Veterinärmed 48:267–273. associated with sudden death in the pig. Vet Rec
———. 1993c. Untersuchungen zur experimentellen 77:1463.
Clostridium perfringens typ A enterotoxaemie der Duran, C. O., and Walton, J. R. 1997. Clostridium novyi sud-
Saugferkel: Licht un elektronmikroscopischen Unter- den death in sows: Toxaemia or postmortem invader?
suchungen zur Pathologie und Pathogenesis der experi- Pig J 39:37–53.
mentellen Clostridium perfringens typ A Infektion. Eggleston, E. L. 1950. Blackleg in swine. Vet Med 45:253.
Monatsh für Veterinärmed 48:299–306. Gualandi, G. L. 1955. L’infezione da “Clost chauvoei” nel
Kyriakis, S. C.; Sarris, K.; Kritas, S. K.; Saolidis, K.; Tsinas, A. suino. Arch Vet Ital 6:57.
C.; and Tsiloyannis, V. K. 1995. The effect of salino- Hac, L. R., and Habert, A. C. 1943. Penicillin in treatment of
mycin on the control of Clostridium perfringens type A in- experimental Clostridium welchii infection. Proc Soc Exp
fection in growing pigs. J Vet Med Ser B 42:355–359. Biol Med 53:61.
Madsen, D. P. 1995. Managing management induced Itoh, H.; Uchida, M.; Sugiura, H.; Oguso, S.; and Yamakawa,
Clostridium perfringens type A infection in sucking pigs: a K. 1987. Outbreak of Clostridium novyi infection in swine
case study. Swine Health and Prod 3:207–208. and its rapid diagnosis. J Jpn Vet Med Assoc 40:365–369.
Mansson, I., and Smith, L. D. S. 1962. Atypical strains of Jaartsveld, F. H. J.; Janssens, F. T. M.; and Jobse, C. J. 1962.
Clostridium perfringens from swine. Acta Path Microbiol Clostridium infectie bij biggen. Tijdschr Diergeneeskd
Scand 55:342–348. 87:768.
Miwa, N.; Nishima, T.; Kubo, S.; and Atsumi, M. 1997. Most Kellaway, C. H.; Reid, G.; and Trethewie, E. R. 1941. Circu-
probable number method combined with nested poly- latory and other effects of the toxin of Cl. septique. Aust
merase chain reaction and enumeration of enterotoxi- J Exp Biol Med Sci 19:277.
genic C. perfringens in cattle, pigs and chickens. J Vet MacLennan, J. D. 1962. Histotoxic clostridial infections of
Med Sci 59:89–92. man. Bacteriol Rev 26:177.
Nabuurs, M.; Haagsma, J.; Molen, E.; and Heijden, J. 1983. Marco, E. 1995. Sudden deaths in sows. Pig J 35:157–163.
Diarrhoea in one to three week-old piglets associated Martig, J. 1966. Zur Differentialdiagnose zwischen
with Clostridium perfringens type A. Ann Rech Vet Rauschbrand und Pararauschbrand mit Hilfe der Im-
14:408–411. munofluoreszenz. Schweiz Arch Tierheilkd 108:303.
Olubunmi, P. A. 1982. Bacteria Associated with Inflamma- Mavenyengwa, M., and Matope, G. 1995. An outbreak of
tory Enteric Lesions in Pigs. Ph.D. diss. Univ Glasgow. quarter evil in weaner pigs in Zimbabwe. Zimbabwe Vet
Popoff, M. R., and Jestin, A. 1985. Enteropathogenicity of J 26:135–138.
purified Clostridium perfringens enterotoxin in the pig. Moussa, R. S. 1958. Complexity of toxins from Clostridium
412 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

septicum and Clostridium chauvoei. J Bacteriol 76:538. Burgen, A. S. V.; Dickens, F.; and Zatman, L. J. 1949. The ac-
Sterne, M., and Edwards, J. B. 1955. Blackleg in pigs caused tion of botulinum toxin on the neuromuscular junction.
by Clostridium chauvoei. Vet Rec 67:314. J Physiol 109:10.
Taylor, D. J., and Bergeland, M. E. 1992. Clostridial infec- Dack, G. M., and Gibbard, J. 1926a. Studies on botulinum
tions. In Diseases of Swine, 7th ed. Ed. A. D. Leman, B. toxin in the alimentary tract of hogs, rabbits, guinea
E. Straw, W. L. Mengeling, S. D’Allaire, and D. J. Taylor. pigs and mice. J Infect Dis 39:173.
Ames: Iowa State Univ Press, pp. 454–469. ———. 1926b. Permeability of the small intestine of rab-
Taylor, W. I., and Novak, M. 1952. Antibiotic prophylaxis of bits and hogs to botulinum toxin. J Infect Dis 39:181.
experimental clostridial infections. Antibiot Chemother Doiurtre, M. P. 1967. Botulism in animals in Senegal. Bull
2:639. Off Int Epizoot 67:1497.
Zeller, M. 1956. Enzootischer Pararauschbrand in einer Lamanna, C., and Carr, C. J. 1967. The botulinal, tetanal,
Schweinmastanastalt. Tierärztl Umsch 11:406. and enterostaphylococcal toxins: A review. Clin Pharma-
col Ther 8:286.
Tetanus May, A. J., and Whaler, B. C. 1958. The absorption of
Batty, I., and Walker, P. D. 1964. The identification of Clostridium botulinum type A toxin from the alimentary
Clostridium novyi (Clostridium oedematiens) and Clostrid- canal. Br J Exp Pathol 39:307.
ium tetani by the use of fluorescent labelled antibodies. J Morrill, C. C., and Bajwa, G. S. 1964. Botulism. In Diseases
Pathol 88:327. of Swine, 2d ed. Ed. H. W. Dunne. Ames: Iowa State
Kaplan, M. M. 1943. An unusual epizootic of tetanus in Univ Press, p. 605.
young pigs. Middx Vet 3:8. Müller, J. 1967. On the occurrence of Clostridium botulinum
Mihaljevic, K. 1966. A contribution to the study of lockjaw type C beta in the livers of slaughter animals in Den-
in animals. Vet Arh 36:152. mark. Bull Off Int Epizoot 67:1473.
Morrill, C. C. 1964. Clostridial infections. In Diseases of Narayan, K. G. 1967. Incidence of clostridia in pigs. Acta Vet
Swine, 2d ed. Ed H. W. Dunne. Ames: Iowa State Univ Acad Sci Hung 17:179.
Press, p. 378. Scheibner, G. von. 1955. Die Emfanglichkeit des Schweines
Sakurai, N. 1966. The relation between human and veteri- für Botulinus toxin der Typen A–E. Dtsch Tierärztl
nary tetanus in a Japanese prefecture. Principles on Wochenschr 62:355.
Tetanus. Proc 2d Int Conf Tetanus, p. 91. Smintzis, G., and Durin, D. 1950. Epizootie de botulisme
Sergeeva, T. I., and Matveev, K. I. 1966. Geographical distri- chez le porc. Bull Soc Sci Vet (Lyon), p. 71.
bution of Clostridium tetani in the soil of the U.S.S.R. Smith, L. D. S., and Holdeman, L. V. 1968. The Pathogenic
Principles on Tetanus. Proc 2d Int Conf Tetanus, Anaerobic Bacteria. Springfield, Ill.: Charles C. Thomas.
p. 77. Smith, L. D. S.; Davis, J. W.; and Libke, K. G. 1971. Experi-
Veronesi, R. 1966. Antibiotics versus antitetanic serum in mentally induced botulism in weanling pigs. Am J Vet
the prevention of human tetanus. Principles on Tetanus. Res 32:1327.
Proc 2d Int Conf Tetanus, p. 417. Thom, C.; Edmondson, R. B.; and Giltner, L. T. 1919. Botu-
lism from canned asparagus. J Am Med Assoc 73:907.
Botulism Yamakawa, K.; Kamya, S.; Yoshimura, T.; and Nakamura, S.
Beiers, P. R., and Simmons, G. C. 1967. Botulism in pigs. 1992. Clostridium botulinum type C in healthy swine in
Aust Vet J 43:270. Japan. Microbiol Immunol 36:29–34.
Eperythrozoonosis
30 K. Heinritzi

Eperythrozoonosis in swine is caused by the rickettsial velopmental stage (Zachary and Basgall 1985; Liebich
organism Eperythrozoon suis (Ristic and Kreier 1984). and Heinritzi 1992). These studies demonstrated that
the organism changes its size and shape during its devel-
HISTORY opment. They provide evidence that the former E.
parvum, considered to be nonpathogenic, is an immature
The disease was first reported by Doyle in Indiana in or intermediate form of E. suis. Free organisms are also
1932 as “a rickettsia-like or anaplasmosis-like disease in seen in the plasma. Those erythrocytes that have been
swine” (Doyle 1932). He described obvious icteroane- deformed by the organism are removed and then he-
mia, dyspnea, weakness, and fever up to 40.5˚C in 2- to molyzed by the spleen. In the acute stage, a generalized
8-month-old pigs, noted that the blood of the affected hemolytic icterus can occur. It is assumed that these
pigs was thin, that the red blood cells tend to agglutinate changes in the erythrocyte’s shape influence its fluidity
spontaneously, and that the serum was tinted an icterous and its osmotic resistance (Heinritzi and Plank 1992;
color. More recently, eperythrozoonosis has been ob- Zachary and Basgall 1985).
served in pigs of a wide age range, from piglets to preg- Eperythrozoonosis causes an autoimmune hemolytic
nant sows, all over the world. Splitter and Williamson anemia mediated by or associated with a cold agglutinin.
(1950) described the organism responsible for icteroane- The mechanisms that cause the hemolysis during the
mia in pigs as Eperythrozoon suis because of its similarity acute stage are not fully understood. Zachary and Smith
to known species of eperythrozoa: E. wenyoni of cattle (1985) assumed that during the interaction of E. suis and
and E. ovis of sheep. the erythrocyte membrane, the membrane structure
Eperythrozoon species have also been identified in oth- changes, thereby resulting in the freeing of masked anti-
er animals (Gothe and Kreier 1977). Pathogenicity, how- gens or the modification of existing antigens that are
ever, has only been demonstrated for E. suis, E. ovis then seen as foreign by the body’s own immune system.
(sheep), E. wenyoni (cattle), and E. coccoides (mice). After the organism adheres to the erythrocyte cell mem-
brane, autoantibodies are produced as part of the de-
ETIOLOGY AND PATHOGENESIS fense mechanism and attack the erythrocytes of the in-
fected animal. These antibodies are cold agglutinins of
E. suis are round to oval organisms with an average di- the M isotype that, depending on the temperature, are
ameter of 0.2–2 µm that adhere either individually or in deposited on the erythrocyte membrane to cause the ag-
chains to the outer erythrocyte membrane (Zachary and glutination of individual erythrocytes. At higher temper-
Basgall 1985; Liebich and Heinritzi 1992); they can also atures the cold agglutinin can be removed from the
encompass the entire erythrocyte. For a long time the erythrocyte membrane. At 0˚C it is tightly fixed to the
eperythrozoon organisms from pigs were divided into membrane (Zachary 1984; Jüngling et al. 1994).
two forms—E. suis and E. parvum—on the basis of their In the acute phase an increased bleeding potential is
size, form, pathogenicity, incubation period, therapeutic observed, caused by activated intravascular coagulation
resistance, filterability, and structure (Splitter 1950). with a consecutive consumptive coagulopathy. The
More recent studies indicate these differences are not a thromboplastin time is increased and the number of
question of two pathogenetically different organisms, thrombocytes decreased. The thromboelastogram shows
but rather that the E. suis organism changes its size and an increase in the reaction and coagulation time, as well
form as it matures (Zachary and Basgall 1985; Liebich as a decrease in the maximum amplitude. The larger the
and Heinritzi 1992). Scanning electron microscope stud- number of erythrocytes that are affected by E. suis, the
ies show that an infection with E. suis changes the more striking are the changes. Latent infection with E.
erythrocyte surface, causing a deformation (Zachary and suis has no influence on blood coagulation (Plank and
Basgall 1985; Liebich and Heinritzi 1992). During acute Heinritzi 1990). Acute E. suis infection produces a seri-
eperythrozoon attacks, immature and adult forms of the ous blood acidosis and hypoglycemia. In vivo and in vit-
organism are found next to each other on the erythro- ro experiments demonstrate that during an acute attack
cyte, representing the various sizes and forms of their de- of eprythrozoonosis, the amount of glucose used in-

413
414 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

creases strongly. This results from the metabolic activi- CLINICAL SIGNS
ties of E. suis (Heinritzi et al. 1990; Smith et al. 1990).
The acidosis results from an increase in the lactic acid Within an infected herd, eperythrozoon attacks occur in
concentration (metabolic component), as well as from a individual animals that are already suffering from re-
disturbance in the pulmonary gas exchange (respiratory duced resistance resulting from conditions likely to cause
component). The erythrocyte sedimentation rate is stress: after zootechnical procedures or dominance fights
markedly increased during an acute attack. or during parturition. The disease can also break out in a
It has not yet been possible to cultivate E. suis on a herd where the animals are already suffering from an-
cell-free medium. other chronic infectious disease.
Pigs of any age can be affected by E. suis infection.
EPIDEMIOLOGY Weaned piglets are particularly predisposed to infection,
as are piglets a few weeks after being castrated. The clin-
Eperythrozoa are species specific (Ristic and Kreier ical signs of an acute attack are pallor, fever up to 42˚C,
1984). Eperythrozoonosis caused by E. suis organisms occasional icterus, as well as cyanotic changes on the ex-
has only been observed in domestic pigs. Serologic tests tremities, especially on the margins of the ear cartilage.
from wild pigs have been uniformly negative for E. suis in The classic form of icteroanemia as described by Robb
the indirect hemagglutination (IHA) test. (1943) is now seldom seen. Anorexia, apathy, and dysp-
Transmission can occur directly via the oral uptake of nea may occur depending on the severity of the anemia.
blood and blood components: licking tail-docking Growth rate is depressed in those pigs that survive the
wounds, cannibalism, or the uptake of blood-contami- acute phase of the disease .
nated urine. Indirect transmission is by means of live Marbling (mottling) or red discoloration of the ear
vectors, such as ectoparasites (lice), and nonliving vec- cartilage can occur due to the impairment of the blood
tors, such as contaminated syringes or surgical instru- supply in the capillary vessels of the extremities. The
ments used for docking tails, tattooing, or castration. cold agglutinins that are present during an acute attack
Transmission is also possible via the use of a contami- are most likely responsible for the microagglutination
nated upper-jaw sling. Transmission during copulation is and thrombosis that occur in the capillaries of the ex-
only possible if the boar leaves blood-contaminated se- tremities, the temperature in these areas being lower
men in the vaginal area. Intrauterine transmission of E. than that of the core. Fine, light to dark red discol-
suis is considered unlikely (Heinritzi 1992). orations of the ear margins are characteristic. In some
In experimentally infected and splenectomized pigs, cases marked cyanotic discolorations of the complete ear
the incubation period lasts an average of 7 days (3–20). cartilage, the tail, and the distal parts of the extremities
Eperythrozoonosis is a so-called multifactorial dis- can be observed. Healing can be expected within a few
ease. The reason for this view is that under experimental days after therapy. Necrosis of the ear margins or large
conditions, it is not possible to produce an acute case in parts of the ear cartilage can occur if the eperythrozoon
a healthy pig that is kept under clean and correct condi- attack extends for a longer period of time, or if consecu-
tions solely by infecting it with the pathogen. If the host tive attacks occur. Since affected pigs do not develop a
has a functioning defense system, a balance develops be- strong immunity, reinfection is possible at any time.
tween itself and the pathogen; the number of eperythro- The chronic form of eperythrozoonosis causes un-
zoon organisms in the blood are kept at a constant low thriftiness, pallor, and sometimes allergic skin reactions
level. Clinically obvious signs of anemia, fever, and occa- in the form of urticaria or Morbus maculosus. It is impor-
sional icterus will occur only in especially stressed indi- tant to differentiate the clinical signs of Morbus maculo-
vidual animals within a herd. Stress in the form of over- sus, with its numerous petechiae and ecchymoses, from
stocking, bad climatic conditions, a change in housing or those of hog cholera.
food, or chronic general disease predisposes the pigs to Sows with eperythrozoonosis usually show clinical
clinical illness. signs 3–4 days after introduction to the farrowing ac-
The spread of the pathogen is strongly dependent on commodation or after farrowing. Acutely sick sows suffer
the potential for infection within an individual herd or from anorexia and fever to 42˚C, and mammary or vulval
between herds. Precise epidemiological studies are possi- edema may be visible for 1–3 days. These sows have a
ble only to a limited extent since serologic tests are at the depressed milk flow, and their maternal behavior is sub-
moment not dependable enough for the detection of la- normal or absent. Since the clinical signs of the E. suis in-
tent infected animals. Serologically negative animals fection can last until the puerperal period, differentiation
could still carry E. suis and transmit it to others. For this between eperythrozoonosis and puerperal disease can be
reason it is difficult to determine precisely the impor- very difficult (Bugnowski and Horsch 1988). Secondary
tance and spread of the pathogen within a definite pop- bacterial or viral infections and stressful factors, such as
ulation. mange, poor housing conditions, and lack of food, en-
courage the development of eperythrozoonosis. Finally,
CHAPTER 30 EPERYTHROZOONOSIS Heinritzi 415

the disease can lead to the so-called thin-sow syndrome be seen on the test tube wall and is specific for epe-
in affected pigs. rythrozoonosis. The phenomenon is enhanced by cool-
Reproductive problems have been described in sows ing the blood and almost vanishes when the blood is
infected with E. suis. Poor conception rates, anestrus, warmed to 37˚C.
abortion, and the birth of weak piglets have all been re- Eperythrozoonosis causes hemolytic anemia with the
ported. Brownback (1981) described a case where 65% of character of a normochromic and normocytic anemia.
the sows in an infected herd showed no signs of estrus The red cell status of sick animals shows an almost par-
within the first 7 days after weaning. Sisk et al. (1980) al- allel decrease in the number of erythrocytes, the hemo-
so identified reproductive problems, irregular cycles, globin concentration, and its hematocrit value. Capillary
abortions, small litters, premature births, and stillbirths, tubes containing blood from affected pigs are very strik-
as well as weak piglets, in serologically positive herds. ing when centrifuged because of the small percentage of
Schweighardt et al. (1986) described fertility problems in red blood cells, the broad leukocyte band, and the slight-
the form of anestrus, a return to estrus, and an increased ly icteric color of the plasma.
weaning to estrus interval for up to 60% of the sows ex- At the beginning of an attack, a few days before the
amined. fever reaches its highest point, single eperythrozoon or-
No noticeable influence on herd reproduction was ganisms can be demonstrated in blood smears. The high-
found in another study, however, where the effect of an er the number of eperythrozoon organisms in the blood,
E. suis infection on productivity and on newborn piglets the lower the hematocrit and hemoglobin value, and the
was investigated. The authors did not observe delayed es- lower the number of erythrocytes.
trus, embryonic death, or abortions (Zinn and Jesse Iron that is released during hemolysis is deposited at
1982; Zinn et al. 1983). Rosenkrans et al. (1984) found the site of phagocytosis. The amount of serum iron de-
no difference in the number of mummies, neonatal mor- creases slightly with increased number of organisms. If
tality, average birth weight, growth rate, and fertility be- the body has adequate stores of iron, the concentration
tween sows chronically infected and uninfected with E. of serum iron rises sharply after therapy, supporting the
suis. The authors postulated that in the course of time an massive erythropoesis that develops. This rapid stimula-
adaptation takes place and that the organism loses some tion of erythropoesis is associated with the sudden in-
of its virulence. Laboratory control studies have not crease in the red blood cell numbers, increased numbers
proven that the reproductive problems reported are due of immature cells, and a large increase in reticulocytes.
to eperythrozoonosis. In the acute form of hemolytic anemia, an increase in
Piglets of anemic sows are born anemic, without be- the amount of noncoupled (indirect) bilirubin can occur
ing infected themselves. The piglets are often pale and in the blood, as conjugation to glucuronic acid in the liv-
sometimes underweight and tend to develop disease eas- er is depressed. After treatment, there is a rapid decrease
ily. in the levels of serum bilirubin. This signals the end of
The acute clinical signs in feeder pigs usually develop erythrocyte destruction and indicates that a rapid
following cannibalism, overcrowding, poor housing, and change from hemolysis to hematopoesis has occurred. A
inadequate nutrition. Frequent injections for treatments definite leukocytosis is evident 1–2 days before or short-
and vaccinations also play a major role in the spread of ly after an attack of fever. The leukocyte response is seen
the organism and in subsequent reinfections. Cannibal- in terms of a neutrophilic leukocytosis.
ism is particularly important as a source of infection for
feeder pigs compared to other groups. Oral treatment DIAGNOSIS AND
with tetracycline used in the control of respiratory dis- DIFFERENTIAL DIAGNOSIS
eases can mask the clinical signs of eperythrozoonosis
during treatment. The affected pigs later show skin pal- The diagnosis of eperythrozoonosis can be based upon
lor, poor weight gain, and an increased sensitivity to oth- five criteria: clinical signs, hematological changes, direct
er secondary diseases. demonstration of the organism, indirect tests, and infec-
Morbidity due to the progress of subclinical infection tion in splenectomized pigs.
in an infected herd is difficult to measure. Mortality due
to eperythrozoonosis is less than 1%. Clinical Signs
The most important clinical signs for diagnosis are the
HEMATOLOGY AND METABOLISM occurrence of anemia with apathy and fever of 40˚C or
more. Additional clinical signs suggestive of eperythro-
Blood collected during the febrile stage is watery, has a zoonosis are discolorations of the ear margin and the
lacquered appearance, and does not stick to the test tube presence of allergic skin reactions. Weaned piglets short-
wall. When blood that was collected into a tube contain- ly after zootechnical procedures (castration) and feeder
ing an anticoagulant is poured after being cooled to pigs are most likely to be affected. Sows usually only suf-
room temperature, fine-grained microagglutination can fer from the subclinical form.
416 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Hematological Changes Howell-Jolly bodies are also stained. Live mycoplasmas


Blood taken during the febrile stage produces visible mi- present in the sample fluoresce with this acridine deriva-
croagglutination on blood films, and the normochromic, tive and appear as light yellow to light green fluorescent
normocytic character of a hemolytic anemia is proof of dots or disks, depending on the concentration of dye
the disease. The mean corpuscular hemoglobin concen- used and the degree of absorption (Stübner 1976). The
tration (MCHC), derived from the quotient of the hemo- erythrocytes differ from the mycoplasms in their size and
globin concentration and the hematocrit, indicates the position. Eperythrozoa stain more strongly, are almost
quantitative changes in the amount of hemoglobin in the twice the size, and are usually found on the erythrocyte
erythrocytes and confirms the presence of a or on its edge. Degenerating eperythrozoa appear small-
hypochromic anemia. Hypochromic conditions (MCHC er and are usually seen as green dots on the edge of the
under 300 g/L) occur when the production of new he- erythrocyte. The blood smear should, therefore, always
moglobin cannot match the rate of red cell production. be done on the day of sampling.
Other plasma biochemical effects include a fall in the
blood glucose level and a transient increase in the level of Indirect Serologic or PCR Tests
bilirubin. Serologic methods include the demonstration of anti-
bodies by the IHA test, complement fixation test (CFT),
Direct (Microscopic) Demonstration or ELISA. The cold agglutinins that occur during epe-
of the Organism rythrozoonosis are not specifically directed against the
Microscopic demonstration of the organism is best per- organism but are directed against erythrocyte mem-
formed during the acute febrile stage. Meticulous sam- branes that have been specifically damaged by E. suis
pling technique and preparation of a good-quality smear (Smith and Rahn 1975; Jüngling et al. 1994). The pro-
are essential for visualizing eperythrozoa by light mi- duction of antibodies is related temporally to the in-
croscopy. Before preparing a blood smear, the blood creasing number of organisms but not to the time of the
must be warmed to 38˚C; otherwise the erythrocytes ag- infection. This means that antibody production occurs in
glomerate due to the cold agglutinin, making the detec- a wavelike phase. Every new attack produces a new anti-
tion of eperythrozoa difficult. The size of the organism body response. In splenectomized pigs this undulating
varies and decreases with increasing duration of the phase of the antibody titer can easily be followed. Even
chronic disease. The organisms are visible on the sur- after several acute attacks, the antibody titer persists on-
faces of the erythrocytes as oval to round individual rings ly for 2–3 months, often falling below the threshold val-
or as chains completely encompassing the erythrocyte. ue. This means that false-negative titers are common.
Wright-Giemsa is the most common stain for demon- The booster effect, known from other infectious diseases,
strating eperythrozoa in fixed blood smears. Both this has not been demonstrated in E. suis infection in the pig.
stain and conventional quick stains may produce arti- In spite of this, the time interval between intermittent at-
facts (Giemsa precipitations) that adhere to the erythro- tacks becomes longer, and the intensity of the fever phas-
cyte membrane and that could be mistaken for epe- es weakens. Serologically negative pigs can be E. suis car-
rythrozoa. riers and infect other animals via blood contact. All
It is also possible to see individual eperythrozoa in serologic procedures are suitable only for herd diagnos-
their various forms under a fluorescent microscope with tics (Baljer et al. 1989; Schuller et al. 1990). Gwaltney et
acridine orange staining (Kreier and Gothe 1976). The al. (1993) describe a polymerase chain reaction (PCR)
complexity of this technique makes it suitable only for that for the moment seems to be the diagnostic aid of
diagnostic laboratories or clinics. One milliliter of blood choice. The results demonstrate the potential of PCR as
in EDTA, heparin, or acid citrate dextrose is required and a valuable tool for diagnosing and studying E. suis infec-
should be sent to the appropriate diagnostic laboratory tion in pigs.
(Heinritzi 1990). The advantage of this staining method
is that the bright orange organisms can easily be differ- Animal Infection in Splenectomized Pigs
entiated from the dark green background, even if their Ultimate confirmation of the presence of infection can
numbers are small. During the acute stage, eperythrozoa be accomplished by splenectomizing a pig considered to
appear as light to dark orange dots; in chronically infect- be infected or by transfusing blood from a suspected pig
ed pigs, however, the organisms are less distinctive, ap- to a splenectomized pig. Splitter (1950) used splenec-
pearing as small light yellow to light green dots that can tomized pigs for his experiment with E. suis, and diag-
be distinguished on the edge of the erythrocyte or are ly- nostic splenectomy is still considered the surest way of
ing free in the plasma (filter set 09-450-490/FT 510, LP diagnosing an E. suis infection. Between 3 and a maxi-
520). mum of 20 days after removing the spleen, the E. suis–in-
False positives are possible with this stain. Nuclei and fected pig develops an acute attack, which can then be
nuclear fragments can also fluoresce with the acridine or- easily diagnosed by demonstrating the organism in the
ange stain. This means that immature erythrocytes and blood smear.
CHAPTER 30 EPERYTHROZOONOSIS Heinritzi 417

TREATMENT negative or if the transfusion of 10 blood samples to a


splenectomized growing pig has no effect. The use of un-
The treatment of choice is the parenteral application of infected pigs minimizes the introduction of eperythro-
oxytetracycline in a dose of 20–30 mg/kg body mass zoonosis.
(BM). Pigs developing acute eperythrozoonosis with
fever must be treated parenterally, since they are unable REFERENCES
to eat sufficient medicated feed for therapy to be effec-
tive. Parenteral oxytetracycline can also be given to pigs Baljer, G.; Heinritzi, K.; and Wieler, L. 1989. Untersuchun-
on infected farms at times of extreme stress, such as dur- gen mit der indirekten Hämagglutination zum Eperyth-
ing rehousing or after zootechnical procedures. Howev- rozoon suis-Nachweis in experimentell und spontan in-
er, neither with parenteral nor with constant oral treat- fizierten Schweinen J. Vet Med B 36:417–423.
ment can the organism be eliminated. Oral Brownback, A. 1981. Eperythrozoonosis as a cause of infer-
chlortetracycline treatment can reduce the occurrence of tility in swine. Vet Med Small Anim Clin 76:375–378.
anemia. Continuous treatment with chlortetracycline Bugnowski, H., and Horsch, F. 1988. Zur Diagnostik der
may not be worthwhile, however, since splenectomized Eperythrozoonose des Schweines. Mh Vet-Med
pigs have developed acute eperythrozoonosis during oral 43:343–346.
therapy with chlortetracycline at 20 mg/kg BM in an ex- Claxton, M., and Kunesh, J. P. 1975. Eperythrozoonosis in
perimental study. Piglets and chronically ill pigs should swine. Iowa State Univ Vet 37:82–83.
also be given iron (200 mg iron dextran/piglet). Doyle, L. P. 1932. A rickettsia-like or anaplasmosis-like dis-
ease in swine. J Am Vet Med Ass 8:668–671.
PREVENTION Gothe, R., and Kreier, J. P. 1977. Aegyptianella, Eperythro-
zoon, and Haemobartonella. In Parasitic Protozoa, vol. 4.
Treatment should be supplemented by supportive and Ed. J. P. Kreier. New York: Academic Press, pp. 263–294.
prophylactic measures (Claxton and Kunesh 1975). It is Gwaltney, S. M.; Hays, M. P.; and Oberst, R. D. 1993. De-
important to stop the spread of infection and to prevent tection of Eperythrozoon suis using the polymerase chain
reinfection within a herd. There should be an ecto- and reaction. J Vet Diagn Invest 5(1):40–46.
endoparasite management program, and hygienic pre- Heinritzi, K. 1990. Zur Diagnostik der Eperythrozoon suis-In-
cautions should be taken during any procedure where fektion. Tierärztl Prax 18:477–481.
blood could be transmitted from one animal to the next. ———. 1992. Untersuchungen zur Übertragbarkeit von
Transmission by needles and by surgical instruments Eperythrozoon suis. Tierärztl Umschau 47:588–599.
contaminated with blood can be limited by changing Heinritzi, K., and Plank, G. 1992. Einfluß der Eperythrozoon
needles between injections and bleeding of sows. Sepa- suis-Infektion auf die osmotische Resistenz der Eryth-
rate sterile instruments should be used for each litter in rozyten. Berl Münch Tierärztl Wschr 105:380–383.
zootechnical procedures such as tooth resection, castra- Heinritzi, K.; Plank, G.; Peteranderl, W.; and Sandner, N.
tion, tattooing, tail docking, or even injections. This 1990. Untersuchungen zum Säure-Basen-Haushalt und
means that every unit must have at least two sets of sur- Kohlenhydratstoffwechsel bei der Infektion mit Eperyth-
gical instruments. The contaminated set can then be rozoon suis. J Vet Med B 37:412–417.
placed in a properly diluted disinfectant and used for al- Jüngling, A.; Erhard, M. H.; Heinritzi, K.; and Lösch, U.
ternate litters. 1994. Bedeutung und Verlauf eines Kälteagglutinins bei
Attacks may increase in frequency in infected herds, der Eperythrozoon suis-Infektion des Schweines. Berl
but a certain equilibrium is eventually reached between Münch Tierärztl Wschr 107:271–275.
the host and the organism. This also holds true for Kreier, J. P., and Gothe, R. 1976. Aegyptianellosis, Eperyth-
splenectomized animals. If this balance is disturbed, an rozoonosis, Grahamellosis and Haemobartonellosis. Vet
acute eperythrozoon attack can occur at any time, and Parasitol 2:83–95.
thus it is important to maintain good husbandry. Infec- Liebich, H., and Heinritzi, K. 1992. Licht- und elektronen-
tion with E. suis can become manifest only if other stress- mikroskopische Untersuchungen an Eperythrozoon suis.
ful factors are also affecting the pig. On the other hand, a Tierärztl Prax 20:270–274.
latent infection with E. suis is also a factor that can cause Plank, G., and Heinritzi, K. 1990. Disseminierte intravasale
the outbreak of an otherwise latent infection. The im- Gerinnung bei der Eperythrozoonose des Schweines.
portance of an E. suis infection does not lie in its prima- Berl Münch Tierärztl Wschr 103:13–18.
ry disorder but rather in the poor performances that it Ristic, M., and Kreier, J. 1984. Family III. Anaplasmataceae.
can cause. In Bergey’s Manual of Systematic Bacteriology, vol. 1.
There are no vaccines. If a herd is free from infection, Ed. N. R. Krieg and J. G. Holt. Baltimore: Williams and
new additions should be from eperythrozoon-free herds. Wilkins, p. 719.
Freedom from infection can be assumed if serologic or Robb, A. D. 1943. Ictero-anemia in growing swine. Vet Med
PCR tests of serum of 10 pigs in the farrowing house are 38:271–274.
418 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Rosenkrans, C., Jr.; Zinn, G. M.; and Jesse, G. W. 1984. Science 111:513–514.
Chlortetracycline as a treatment against the effects of Splitter, E. J., and Williamson, R. L. 1950. Eperythrozoono-
eperythrozoonosis on sow productivity. Proc Int Congr sis in swine: A preliminary report. J Am Vet Med Assoc
Pig Vet Soc 8:210. 116:360–364.
Schuller, W.; Heinritzi, K.; al-Nuktha, S.; Kilbl, S.; and Stübner, G. 1976. Mykoplasmennachweis durch Direktfluo-
Schuh, M. 1990. Serologische Verlaufsuntersuchungen rochromierung mit Acridinderivaten. Dtsch Med Wschr
mittels KBR und ELISA zum Nachweis von Antikörpern 101:1257–1258.
gegen die Eperythrozoon suis-Infektion des Schweines. Zachary, J. F. 1984. Proposed pathogenesis of porcine
Berl Münch Tierärztl Wschr 103:9–12. eperythrozoonosis: A possible animal model for cold ag-
Schweighardt, H.; Fellner, A.; Pechan, P.; and Lauermann, E. glutinin disease in man. Federation Proc 43,3:378.
1986. Eperythrozoonose beim Schwein—Ein Fall- Zachary, J. F., and Basgall, E. J. 1985. Erythrocyte membrane
bericht. Wien Tierärztl Mschr 73:250–253. alterations associated with the attachment and replica-
Sisk, D. B.; Cole, J. R.; and Pursell, A. R. 1980. Serologic in- tion of Eperythrozoon suis: A light and electron micro-
cidence of eperythrozoonosis in Georgia swine. In Proc scopic study. Vet Pathol 22:164–170.
13th Amer Assoc Vet Lab Diagnost, pp. 91–99. Zachary, J. F., and Smith, A. R. 1985. Experimental porcine
Smith, A. R., and Rahn, T. 1975. An indirect hemagglutina- eperythrozoonosis: T-lymphocyte suppression and mis-
tion test for the diagnosis of Eperythrozoon suis infection directed immune responses. Am J Vet Res 46:821–830.
in swine. Am J Vet Res 36:1319–1321. Zinn, G. M., and Jesse, G. W. 1982. Effect of eperythro-
Smith, J. E.; Cipriano, J. E.; and Hall, S. M. 1990. In vitro and zoonosis on sow productivity. Proc Int Congr Pig Vet Soc
in vivo glucose consumption in swine eperythrozoono- 7:141.
sis. J Vet Med B 37:587–592. Zinn, G. M.; Jesse, G. W.; and Dobson, A. W. 1983. Effect of
Splitter, E. J. 1950. Eperythrozoon suis n. sp. and Eperythro- eperythrozoonosis on sow productivity. J Am Vet Med
zoon parvum n. sp., two new blood parasites of swine. Assoc 182:369–371.
Erysipelas
31 R. L. Wood

Swine erysipelas (SE) or its equivalent in other lan- itive bacillus with a marked tendency to form elongated
guages—Schweinerotlauf, vlekziekte, rouget du porc, mal filaments.
rossino, entrace eresipelatoso, rozyca, and erisipela del
cerdo—is a disease caused by the bacterium Erysipelothrix Physicochemical Characteristics
rhusiopathiae (Sneath et al. 1986) and manifested by
acute or subacute septicemia and chronic proliferative le- MORPHOLOGY AND STAINING. The morphology
sions. The disease is worldwide in distribution and is of of E. rhusiopathiae is variable. In smears or cultures made
economic importance throughout Europe, Asia, and the directly from tissues in cases of acute infection, the or-
Australian and American continents. ganism appears as slender, straight or slightly curved
The identification of SE as a disease entity began in rods, 0.2–0.4 by 0.8–2.5 µm, occurring singly or in short
1878 when Koch isolated from an experimental mouse chains (Fig. 31.1). An occasional coccoid or clubbed form
an organism that he called “the bacillus of mouse sep- may be seen. Palisades and angular formations (“snap-
ticemia.” In 1882–83 Pasteur and Thuillier briefly de- ping division”) are common. The organism is nonmotile,
scribed the organism isolated from pigs with rouget. In non-spore-forming, and non-acid-fast. It stains readily
1886 Löffler published the first accurate description of with ordinary dyes and is gram-positive but is easily de-
the causative agent of Schweinerotlauf and described the colorized. After several subcultures on an artificial medi-
infection in swine. um, filamentous forms of the organism begin to appear.
In the United States the recorded history of SE began These forms may predominate in old cultures or in
when Smith (1885) isolated the causative organism from chronic lesions. Filamentous forms are somewhat thick-
a pig. The disease was not considered important, howev- ened, are greatly elongated (4–60 µm), and may form a
er, until serious outbreaks were reported in South Dako- mass resembling mycelia, especially in a liquid medium
ta in 1928; by 1959 acute SE had been reported in 44 (Fig. 31.1). The filamentous forms sometimes have a
states. Since that time the prevalence of SE apparently beaded appearance when Gram’s stain is used.
has decreased overall (Wood 1984). However, the disease
is still considered to be of economic importance, espe- GROWTH CHARACTERISTICS. Growth of E. rhu-
cially in the chronic form, and outbreaks of acute SE con- siopathiae at 37˚C in a nutrient broth appears at 24 hours
tinue to occur sporadically in endemic areas. as a faint turbidity with no odor or pellicle. Shaking typ-
E. rhusiopathiae occurs in most parts of the world, ically reveals a momentary appearance of rolling clouds.
and SE occurs in most areas where domestic swine are Slight sedimentation will be seen after 36–48 hours of in-
produced. The organism also causes polyarthritis of cubation. Growth is much heavier in broth enriched with
sheep and lambs and serious death losses in turkeys. It serum. In gelatin stabs incubated at 22˚C for 4–8 days,
has been isolated from body organs of many species of growth of the organism radiates out from the stab in all
wild and domestic mammals and birds as well as rep- directions, resembling a test-tube brush.
tiles, amphibians, and the surface slime of fish. Colonies of E. rhusiopathiae on agar media at 48
In humans E. rhusiopathiae causes erysipeloid, a local hours are tiny (less than 1 mm), are transparent, and
skin lesion that occurs chiefly as an occupational disease vary from smooth to rough, depending on cellular mor-
of persons engaged in handling and processing meat, phology (Fig. 31.1). Colonies of most strains have entire
poultry, and fish as well as of rendering-plant workers, edges, but some strains form colonies that are slightly
veterinarians, game handlers, leather workers, laborato- larger and have somewhat undulate edges. Granulelike
ry workers, and the like. The organism occasionally is structures usually appear under a colony just below the
isolated from cases of endocarditis in humans and rarely surface of the agar. Dissociation from smooth to rough
causes acute septicemic disease. form may occur during the development of a colony,
producing a sector (Fig. 31.1); the morphology of cells
ETIOLOGY from these intermediate forms will include a variety of
shapes from short, curved rods to short filaments.
E. rhusiopathiae, the causative agent of SE, is a gram-pos- Most strains of E. rhusiopathiae produce a narrow

419
420 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

SMOOTH INTERMEDIATE ROUGH


31.1. Cellular and colonial morphology of Erysipelothrix rhusiopathiae. Upper row: ×1200,
crystal violet; lower row: ×32. (Courtesy National Animal Disease Center, Ames, Iowa.)

zone of partial hemolysis on blood agar, usually with a Nørrung et al. 1987; Nørrung and Molin 1991). Strains
greenish color. Rough colonies do not induce hemolysis. that do not possess the specific antigen are referred to as
serovar N.
BIOCHEMICAL PROPERTIES. E. rhusiopathiae is rel- Immunizing antigen is discussed in the section on
atively inactive in commonly used tests of biochemical prevention.
activity (Cottral 1978). The organism produces acid but
no gas from certain fermentable carbon compounds and Biological Characteristics
produces hydrogen sulfide in triple-sugar iron agar GROWTH REQUIREMENTS. E. rhusiopathiae is fac-
(Vickers and Bierer 1958; White and Shuman 1961). ultatively anaerobic; some strains grow better in an at-
mosphere of reduced oxygen containing 5–10% carbon
ANTIGENIC STRUCTURE. Most, if not all, strains of dioxide. The organism will grow at temperatures of
E. rhusiopathiae have one or more common heat-labile 5–42˚C. Optimum growth occurs at 30–37˚C and at a pH
antigens, which are proteins or protein-saccharide-lipid range of 7.4–7.8. Growth is enhanced by serum, glucose,
complexes. The organism does not possess flagellar anti- protein hydrolysates, or surfactants such as Tween-80.
gens, since no flagella are present. The organism is fastidious; that is, a complex medium is
Heat-stable antigens consisting of peptidoglycan required, but specific nutrient requirements are not
fragments from the cell wall form the basis for identifica- known.
tion of various serovars (formerly referred to as
serotypes) within the species. The serovars are identified RESISTANCE. E. rhusiopathiae is relatively resistant to
by precipitin reactions with specific hyperimmune rab- adverse conditions for a non-spore-forming organism
bit sera, usually in a gel double-diffusion system. Most (see also the section on epidemiology). The organism is
isolates of the organism (75–80%) from swine fall into somewhat resistant to drying and can remain viable for
two major serovars designated 1 and 2 (Wood and Har- several months in animal tissues under a variety of con-
rington 1978; Takahashi et al. 1996). About 20% of iso- ditions. It can persist in frozen or chilled meat, decaying
lates make up a group of less-common serovars. Under a carcasses, dried blood, or fish meal. It is remarkably re-
numerical system introduced by Kucsera (1973), a total sistant to salting, pickling, and smoking and can survive
of 26 serovars have been described so far (Kucsera 1973; several months in cured and smoked hams. The organ-
Wood et al. 1978; Nørrung 1979; Xu et al. 1984, 1986; ism can survive in swine feces or fish slime for 1–6
CHAPTER 31 ERYSIPELAS Wood 421

months if temperatures remain below 12˚C. It is sensi- ly healthy swine inhabiting the pens. However, Wood
tive to penicillin and usually to the tetracyclines; it is (1973) found no evidence of growth or maintenance of
quite resistant to polymyxin B, neomycin, and the organism in test soils under various conditions of
kanamycin and is relatively resistant to streptomycin temperature, pH, moisture content, and organic matter
and the sulfonamides (see the section on treatment). It is content or in samples of swine-pen soils from which E.
killed readily by common disinfectants, heat (15 minutes rhusiopathiae had been isolated previously. Rapid death
at 60˚C), and gamma irradiation. curves were consistently demonstrated. This failure to
establish a stable population of the organism in soil is
Other Species within the Genus similar to results reported by other investigators since
Another species within the genus Erysipelothrix has been 1955. Present information indicates that soil that is more
proposed (Takahashi et al. 1987a). This species, desig- or less continually inoculated by infected animals pro-
nated E. tonsillarum, is distinguished from E. rhusiopathi- vides only a temporary medium for transmission of E.
ae by DNA homology values. Phenotypic characteristics rhusiopathiae.
of the two species are indistinguishable by the usual di-
agnostic bacteriologic methods. E. tonsillarum has been Factors of Susceptibility
isolated from a variety of sources, including the tonsils of
healthy swine. Reported isolates of E. tonsillarum have AGE AND GENETICS. Swine less than 3 months or
little or no virulence for swine; therefore, the species is more than 3 years of age are generally least predisposed
not considered significant in the etiology of SE. to SE. The relationship of age to susceptibility may be ex-
Classification of Erysipelothrix has challenged taxono- plained by naturally acquired passive immunity in the
mists throughout the recorded history of this ubiquitous young and active immunity following subclinical infec-
genus. It is likely that the use of current techniques in tion in older animals. Suckling pigs of immune sows are
bacterial genetic analysis will continue to generate pro- immune to infection for several weeks after birth. The
posals of additional species among isolates from various degree and duration of passive immunity are related to
animal hosts and other sources. the immune status of the sow.
There is no experimental evidence that susceptibility
EPIDEMIOLOGY to SE is related to genetics of the animal. Anecdotal ac-
counts of apparent resistance (or susceptibility) of cer-
Sources of Infection tain breeds or families of swine can most likely be ex-
The most important reservoir of E. rhusiopathiae is prob- plained by the presence of varying degrees of naturally
ably the domestic pig. It is estimated that 30–50% of ap- acquired passive or active immunity.
parently healthy or convalescent swine harbor the or-
ganism in their tonsils and other lymphoid tissues. These NATURAL ACTIVE IMMUNITY. For many decades
carriers can discharge the organism in their feces or after SE research was begun in the 1880s, a major prob-
oronasal secretions, creating an important source of in- lem was the inability to consistently induce acute SE in
fection. Swine affected with acute erysipelas shed E. rhu- swine by experimental infection with E. rhusiopathiae.
siopathiae profusely in feces, urine, saliva, and nasal se- The problem eventually was eliminated by use of specif-
cretions. ic-pathogen-free (SPF) pigs delivered aseptically by
Although secondary to swine in importance as surgery, deprived of colostrum, and raised in isolation.
sources of infection, the large variety of wild mammals This development revealed the importance of naturally
and birds known to harbor E. rhusiopathiae provides an acquired immunity as a factor in susceptibility to
extensive reservoir (Wood and Shuman 1981; Shuman erysipelas.
1971). Various species of domestic animals from which Naturally acquired active immunity is induced by
the organism has been isolated provide an additional po- previous infection with E. rhusiopathiae. It is well known
tential reservoir on swine-producing farms; however, that immunity to acute SE follows clinical disease. Less
with the possible exception of turkeys and sheep, their well recognized is the immunity that can be induced by
importance is doubtful. organisms of low virulence, which are capable of causing
The belief that E. rhusiopathiae can lead a saprophyt- mild unnoticed subacute disease or subclinical infection.
ic existence in the soil, living on dead and decaying or-
ganic material, has persisted for many years. However, PREDISPOSING FACTORS. SE can occur with other
available research data have consistently indicated that swine diseases, but the significance of preexisting infec-
the organism, like most other non-spore-forming patho- tious disease as a predisposing factor is uncertain. Para-
genic bacteria, finds an unfavorable environment in the sitic infestations have been reported to increase the
soil and dies out in a relatively short time, most likely be- severity of clinical SE. In addition, Cysewski et al. (1978)
cause of the action of protozoa. E. rhusiopathiae can be showed that the susceptibility of swine to acute SE can
found in the soil of swine pens and in feces of apparent- be enhanced by subclinical toxicity from aflatoxin in the
422 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

feed. This treatment also interfered with the induction of Acute SE


immunity to SE by vaccination. Acute systemic SE begins with bacteremia, which quick-
Environmental and stress factors such as nutrition, ly results in clinical signs of generalized infection (sep-
ambient temperature, and fatigue, particularly sudden ticemia). A nonsystemic infection consisting only of a lo-
changes in these conditions, have long been linked to the cal skin lesion may occur upon cutaneous exposure to a
appearance of SE. For example, acute disease has oc- strain of low virulence or when the pig is partly immune.
curred following sudden changes in diet such as acciden- In such cases the organism is eliminated without induc-
tal access to tankage or to a field of corn, feeding new ing septicemia and the lesion disappears. In the more
corn, or placing pigs on new pasture. Experimentally, typical systemic infection caused by virulent organisms,
sudden exposure to either excessive heat or cold or expo- bacteremia usually develops within 24 hours after expo-
sure to a sustained high temperature (30˚C) has been re- sure. The organism usually can no longer be cultured
ported to enhance susceptibility. Under natural condi- from blood or most body organs after a few days but may
tions, sudden changes in weather involving extreme persist, often for months, in the joints and in lymphoid
temperature changes may have the same effect. Varia- tissue such as tonsils, Peyer’s patches, and spleen.
tions in prevalence of SE believed to be related to yearly According to Schulz et al. (1975b, 1977), pathogene-
cycles or to season of the year have been reported, but sis of the early septicemic phase consists of changes in-
consistent patterns of prevalence corresponding to these volving capillaries and venules of most body organs, in-
factors have not emerged. Except for a possible relation- cluding synovial tissues. As early as 36 hours after
ship to temperature changes, the apparent variations re- subcutaneous exposure of swine to virulent E. rhu-
main unexplained. siopathiae, they observed swelling of endothelium, with
When attempting to evaluate factors that may be re- adherence of monocytes to vascular walls and evidence
lated to the occurrence of SE on the farm, it should be of widespread hyaline thrombosis. This process was re-
kept in mind that a variety of stimuli in the animal’s to- ferred to as a shocklike generalized coagulopathy leading
tal environment can affect the level of susceptibility ex- within 4 days to fibrinous thrombosis, diapedesis, inva-
isting at any given time. Sudden outbreaks of acute SE sion of vascular endothelium by bacteria, and deposition
may be the result of a combination of susceptibility of of fibrin in perivascular tissues. They stated that this
the animals and virulence of the causative organism, process leads eventually to connective-tissue activation
both of which are variable. in predisposed sites such as joints, heart valves, and
blood vessels.
PATHOGENESIS In severe acute SE, hemolysis is commonly observed.
Ischemic necrosis of perivascular tissues may occur,
Investigations using germfree pigs have demonstrated caused by interference with microcirculation. Drommer
that E. rhusiopathiae is the sole causative agent of SE and et al. (1970) observed a high incidence of encephaloma-
does not require the presence of any other infectious lacia in acute experimental SE and theorized that certain
agent for its disease-producing ability. strains of the organism are endotheliotropic and damage
the endothelial cell barrier in the central nervous system
Mode of Entry (CNS).
E. rhusiopathiae can gain entry to the body by a variety of Mild, delayed hypersensitivity responses to E. rhu-
routes. Infection through ingestion of contaminated feed siopathiae can be elicited and transferred by lymphoid
and water is considered a common mode. There is no in- cells. It is doubtful, however, that delayed hypersensitivi-
formation on specific areas within the digestive system ty has a significant part in the pathogenesis of acute SE.
where entry may occur, nor is it known whether the or-
ganism can invade normal mucosa. Early investigators Chronic SE
postulated it gained entrance through lesions produced Information on the pathogenesis of chronic SE is derived
by intestinal parasites; however, their presence is unnec- primarily from studies on development of the arthritic
essary. The organism can readily gain access to the body lesion, which has stimulated interest because of its ap-
through the palatine tonsils or other lymphoid tissue in parent similarity to the lesion of rheumatoid arthritis of
the wall of the digestive tract, but entrance is probably humans.
not limited to these areas. According to observations by Schulz et al. (1975a,
Natural infection no doubt can result from infected 1977), the joint lesion in chronic SE begins with acute
skin wounds, which may be concealed or too small to be synovitis that may occur as early as 4–10 days after expo-
readily noticeable. Experimental infection can be accom- sure to E. rhusiopathiae. Within 3 months, fibrinous exu-
plished easily by inoculation of scarified skin (Shuman dation, proliferation, and pannus formation occur, de-
1951); therefore, it is likely that infection in this manner veloping further into severe fibrosis and destruction of
from a contaminated environment is not uncommon. articular cartilage in 5–8 months. During this time, the
CHAPTER 31 ERYSIPELAS Wood 423

organism can be found sequestered within chondrocytes Mechanism of Pathogenicity


in addition to its presence in synovial tissue and fluid The mechanism by which E. rhusiopathiae incites disease
(Franz et al. 1995). The earliest changes in the synovial processes is not clearly understood. The organism is not
tissue are described as consisting of coagulopathy and known to produce toxins. Considerable evidence has ac-
fibrinous exudate into perivascular tissues. Fibrin de- cumulated indicating that neuraminidase, an enzyme
posited during the vascular phase, not bacterial colo- produced by a number of species of pathogenic bacteria,
nization, is believed to act as mediator of the subsequent is a factor in pathogenicity of E. rhusiopathiae. This en-
connective-tissue proliferation. zyme specifically cleaves alpha-glycosidic linkages in
Affected joints appear to become culture-negative af- neuraminic acid (sialic acid), a reactive mucopolysaccha-
ter 3–6 months, yet the arthritic lesions usually undergo ride on surfaces of body cells. The enzyme is produced
a progressive development that can continue at least 2 by E. rhusiopathiae during logarithmic growth, and the
years. The development of such lesions in the apparent amount of activity is reported to be less in avirulent
absence of the infectious agent has stimulated investiga- strains or strains of low virulence than in fully virulent
tion of the role of immunopathologic processes in strains (Müller 1981). Specific antibody activity against
chronic arthritis. Hypersensitivity may be a significant E. rhusiopathiae neuraminidase has been demonstrated
factor in the chronic proliferative and destructive in sera of swine with chronic SE, in commercial equine
changes but probably not in initiation of the lesion. antierysipelas serum, and in serum of rabbits hyperim-
There is evidence that the bacteria do not entirely dis- munized with a preparation of neuraminidase from the
appear from chronically affected joints, and the long- organism. This latter preparation also induced a low lev-
term progressive lesion may occur in response to the con- el of protection in mice to E. rhusiopathiae infection.
tinued presence of either whole bacterial cells or their Neuraminidase is not a toxin; it must be produced in
antigens. Schulz et al. (1977) reported that living E. rhu- large amounts to be pathogenically active. It can act on
siopathiae was occasionally isolated from such joints for substrates in cell membranes throughout the body, and
up to 2 years. Furthermore, they stated that E. rhu- no doubt its activity reaches high levels in an acute sep-
siopathiae antigen could be detected by immunofluores- ticemia. Therefore, its activity could be a major factor
cence and whole or fragmented bacteria could be seen mediating the widespread vascular damage, thrombosis,
with the electron microscope in culturally negative and hemolysis described (see the section on pathogene-
joints. Denecke and Trautwein (1986) reported detection sis of acute SE). The ability to adhere to cell surfaces may
of E. rhusiopathiae in arthritic joints microbiologically also play a role in the pathogenicity of E. rhusiopathiae,
and immunohistologically for up to 3 years. Specific an- and there is evidence that the process involves neu-
tibodies to the organism have been detected in synovial raminidase. Takahashi et al. (1987b) reported that viru-
fluid of chronically arthritic joints and apparently are lent strains of the organism adhered better to porcine
produced locally by plasma cells in the synovial tissue, kidney cells in vitro than did avirulent strains. Nakato et
which can assume a lymphoid function. It is not known al. (1987) reported that neuraminidase was essential for
whether the chronicity of the joint lesion is maintained adherence of E. rhusiopathiae bacteria to vascular en-
entirely by specific immune reactions against E. rhu- dothelial cells.
siopathiae antigen or whether superimposed autoim- Although neuraminidase activity may be largely re-
mune reactions are involved. sponsible for the pathogenicity of E. rhusiopathiae, it
A preponderance of evidence exists to indicate that does not explain virulence, which is the ability of an in-
erysipelatous arthritis is initiated by active infection of fectious agent to overcome the host’s defenses and initi-
the joint. Mild synovitis and arthritis have been induced ate the pathologic process. There is evidence that the vir-
in rabbits and rats by massive intravenous or intraarticu- ulence of E. rhusiopathiae, which is known to be variable,
lar injections with nonliving whole cells or fractions of is related to the organism’s ability to resist the action of
the organism (White et al. 1975; Hermanns et al. 1982), phagocytes (see also Mechanism of Immunity in the sec-
but the lesions were not as severe as those typically tion on prevention), and resistance to phagocytosis is
caused by infection. White et al. (1975) suggested that correlated with the presence of a protective capsulelike
the mild response induced by such antigens may predis- structure on the surface of virulent bacteria but not on
pose the joint to infection during a subsequent transient avirulent mutants (Shimoji et al. 1994).
septicemia.
Studies on the pathogenesis of endocarditis indicate Serovar and Clinical Form
that the valvular lesions begin with vascular inflamma- In the United States, reports of field cases frequently
tion and myocardial infarcts, possibly resulting from have described serovar 1 (usually subserovar 1a) as the
bacterial emboli. These processes, together with exuda- predominant isolate from acute septicemic disease and
tion of fibrin, lead to destruction of valvular endocardi- serovar 2 as the most common isolate from subacute and
um. chronic cases of SE. However, experimentally, all clinical
424 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

forms of SE can be induced readily in susceptible swine looked or so numerous it would be difficult to count
by strains of serovar 1 or 2. The less-common serovars (3 them all. An animal also may die before recognizable ur-
through 26; N) tend to have low virulence for swine, and ticarial lesions are evident. Individual lesions, by exten-
their clinical significance is doubtful. sion of the borders, assume a characteristic square or
rhomboid shape. In acute nonfatal erysipelas, these le-
CLINICAL SIGNS sions may spread considerably but will gradually disap-
pear within 4–7 days after their first appearance, with no
The clinical signs of SE can be divided into three general subsequent effect other than a superficial desquamation
classifications: acute, subacute, and chronic. In addition, to mark the site. The intensity of skin lesions has a direct
subclinical infection can occur in which no visible signs relationship to the outcome of the disease. Light pink to
of acute disease are evident but which can lead to chron- light purplish-red lesions are characteristic of acute non-
ic SE. fatal SE, whereas angry dark purplish-red lesions usually
precede death of the animal. In acute fatal disease, ex-
Acute SE tensive dark purplish discoloration often occurs over the
Acute SE is characterized by sudden onset, sometimes belly, ears, tail, posterior aspect of the thighs, and jowls.
with sudden death of one or more animals. Other ani- Infrequently, severely affected pigs do not die, and skin
mals in the herd may be noticeably sick, and some of necrosis may follow the severe cutaneous lesions. The ar-
these may subsequently die. Those visibly sick will have eas of necrotic skin are dark, dry, and firm and eventual-
temperatures of 104–108˚F (40–42˚C) and over, and ly become separated from the healing underlying tissue.
those with the higher temperatures may show signs of Affected areas, particularly the ears and tail, will eventu-
chilling. Some pigs may appear normal and yet have tem- ally slough. Healing may require many weeks as a result
peratures of around 106˚F (41˚C). In surviving pigs, tem- of secondary infection.
peratures usually return to normal within 5–7 days.
Affected animals withdraw from the herd and will be Subacute SE
found lying down. When approached, they resent being Subacute SE includes signs that are less severe in their
disturbed but usually will get up and move away. This manifestations than the acute form. The animals do not
usually is accompanied by squealing; when walking, they appear as sick; temperatures may not be as high or may
show a stiff, stilted gait. Upon stopping, they may be seen not persist as long; appetite may be unaffected; a few
to shift their weight in an apparent effort to ease the pain skin lesions may appear that may be easily overlooked;
in their legs. If left alone, they will soon lie down careful- and, if visibly sick, the animals will not remain so for as
ly. Pigs showing severe depression are nevertheless usu- long as those acutely ill. Some cases of subacute SE are so
ally aware of activities around them. They may show mild as to remain unnoticed.
some resentment at being disturbed but will make little
or no effort to rise. Upon being forced to get up, they may Chronic SE
stand for only a few moments before lying down again. Chronic SE may follow acute or subacute disease or sub-
While standing, the feet are carried well under them and clinical infection and is characterized most commonly by
the head is hung dejectedly, giving the back line a signs of arthritis. Signs of cardiac insufficiency may be
marked arched appearance. Others will not be able to
stand even when assisted.
Most affected animals will show partial or complete
inappetence. Bowel movements are usually retarded and
the feces firm and dry in pigs of market age and older, al-
though as the disease progresses, a diarrhea may appear
in younger animals. Abortion may occur in sows that
contract acute or subacute SE during pregnancy.
Cutaneous lesions (urticarial, or “diamond-skin” le-
sions) appear as early as the second and usually by the
third day after exposure to E. rhusiopathiae (Fig. 31.2).
On the light-skinned pig they can be seen as small, light
pink to dark purple areas that usually become raised, are
firm to the touch, and in most instances are easily pal-
pated. In animals with dark-pigmented skin, one must
rely mainly on palpation, although the weltlike lesions 31.2. Typical rhomboid urticarial lesions (“diamond-
may be detected by observing raised areas in the hair skin” lesions) of SE. (Courtesy National Animal Disease
coat. The lesions may be few in number and easily over- Center, Ames, Iowa.)
CHAPTER 31 ERYSIPELAS Wood 425

seen occasionally and will be most noticeable following lar response to infection by E. rhusiopathiae consists
exertion, sometimes causing sudden death. Chronic predominantly of mononuclear leukocytes and
arthritis results in joints that show various degrees of macrophages. Neutrophils may appear but do not pre-
stiffness and enlargement, sometimes as early as 3 weeks dominate. Purulent lesions are not characteristic of E.
after infection. Interference with locomotion ranges rhusiopathiae infection.
from a slight limp to complete refusal to put weight on Affected lymph nodes usually show acute hyperplas-
the limb, depending upon the extent of damage. Arthri- tic lymphadenitis, with hyperemia and hemorrhage. In
tis is the most important clinical manifestation of SE some nodes there may be evidence of thrombosis and
from an economic standpoint. The condition not only af- necrosis of small blood vessels and capillaries. Hemor-
fects growth rate but is responsible for significant losses rhagic nephritis with inflammatory changes in glomeruli
of prime cuts at the packing plant. may be seen occasionally. In addition, necrosis of renal
tubules with hyaline and granular casts has been report-
LESIONS ed. Focal accumulations of mononuclear cells may be
seen in subcapsular sinuses of the adrenal cortex. Le-
Rhomboid urticarial lesions (“diamond-skin” lesions) sions of skeletal muscle may occur, associated with vas-
are characteristic of acute SE, and when generalized (Fig. cular lesions. These consist of a segmented hyaline and
31.2), they are a reliable indicator of septicemia. This ob- granular necrosis of muscle fibers, which may be fol-
servation is important in meat inspection as well as in lowed by fibrosis, calcification, and regeneration. Le-
field diagnosis (see also the section on clinical diagnosis). sions of the CNS have been described, consisting of an-
giopathies with disturbances in permeability,
Acute SE degeneration of neurons, swelling of endothelial cells,
Most lesions of acute SE are similar to those of sep- and malacic foci in the cerebrum, brain stem, and spinal
ticemia caused by a variety of organisms. cord.

MACROSCOPIC LESIONS. In swine dead from acute CLINICAL PATHOLOGY. Leukocytosis may occur in
SE, evidence of diffuse cutaneous hemostasis is often field cases of SE that last for several days or possibly from
prominent, particularly in the skin of the snout, ears, mixed bacterial infection, but in uncomplicated acute SE
jowls, throat, abdomen, and thighs. The lungs may be a leukopenia accompanied by a relative lymphocytosis is
congested and edematous. Petechial and ecchymotic he- characteristic during the first 3–5 days. There may be a
morrhages may be seen on the epicardium and in the relative increase in the number of eosinophils.
musculature of the atria, particularly the left atrium. Ca- Hemoglobin and hematocrit values decrease during
tarrhal to hemorrhagic gastritis is common, and hemor- acute disease, followed later by the appearance of nucle-
rhage of the serosa of the stomach may be present. The ated erythrocytes. The sedimentation rate increases.
liver usually is congested. The appearance of the spleen Changes in plasma components during acute SE include
is of particular note, for it may be congested and a decrease in glucose and increases in glutamic ox-
markedly enlarged, particularly in animals affected for aloacetic transaminase activity, blood creatinine, and
several days. Petechial hemorrhages may be present in blood urea nitrogen.
the cortex of the kidneys. The appearance of the lymph
nodes will depend upon the degree of involvement in the Chronic SE
area they drain. There is some degree of enlargement The predominant lesion of chronic SE in swine is a pro-
with moderate to marked congestion; subcapsular hem- liferative, nonsuppurative arthritis, occurring most com-
orrhage of peripheral nodes may be seen after several monly in hock, stifle, elbow, and carpal joints. Spondyli-
days of illness. The mucosa of the urinary bladder usual- tis is occasionally seen. Vegetative proliferation on the
ly appears normal but may present areas of congestion. heart valves is less common.

MICROSCOPIC LESIONS. A histologic examination MACROSCOPIC LESIONS. Animals affected with


of skin lesions reveals damage to the capillaries and chronic arthritis have an enlargement of one or more
venules, with perivascular infiltration by lymphoid cells joints, most readily visible in hock and carpal joints. The
and fibroblasts. The pathologic changes occur in the joint capsule is thickened with fibrous connective tissue.
papillae and upper layers of the derma. Blood vessels of The joint cavity contains an excessive amount of serosan-
the papillae are congested and may contain microthrom- guinous synovial fluid, which may be slightly cloudy, in-
bi and bacteria. The papillae may also present focal dicating a small amount of purulent material. The pres-
necrotic areas as a result of circulatory stasis. Vascular le- ence of frank pus, however, is not characteristic of the
sions can be seen in the heart, kidney, lung, liver, nervous lesion. The synovial membrane presents varying degrees
system, skeletal muscle, and synovial membranes. Cellu- of hyperemia and proliferation (Fig. 31.3), which gives
426 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

the tissue a swollen, somewhat granular appearance, and DIAGNOSIS


often takes irregular forms, producing fringes (“tags”)
that project into the joint cavity. These fringes may be Clinical and bacteriologic examinations are the most re-
caught between the articulating surfaces and produce se- liable means of diagnosis of acute SE.
vere pain. The proliferating tissue also may extend across
the surface of articular cartilage, forming a pannus that Clinical Diagnosis
leads to destruction of the articular surface and eventu- Acute SE often cannot readily be differentiated clinically
ally to fibrosis and ankylosis of the joint. Lymph nodes from other septicemic diseases, such as Actinobacillus su-
associated with arthritic joints are usually enlarged and is septicemia (Miniats et al. 1989). Nevertheless, certain
edematous. clinical features of an outbreak in a herd are more
Vegetative endocarditis consists of proliferative gran- characteristic of SE than of other diseases if viewed in
ular growths on the heart valves and may be accompa- combination. For example, the following are presump-
nied by lesions resulting from cardiac insufficiency. Oth- tive of SE: a history of a few sudden deaths with no pri-
er internal organs may show chronic inflammatory or evidence of illness; several others sick with high tem-
changes such as infarcts of kidneys and spleen. Enlarge- peratures and apparent stiffness in legs; reluctance of
ment of the adrenal gland has been reported. sick pigs to move but unexpected vitality when aroused;
and clear, alert eyes. Other characteristic signs include a
MICROSCOPIC LESIONS. Lesions of the synovial fair appetite in some visibly sick animals; normal to dry
tissue may vary in severity, from slight perivascular accu- feces; death or recovery of sick animals within a few
mulation of mononuclear cells to an extensive prolifera- days; and, when present, the characteristic rhomboid
tive process. The typical synovial lesion in chronic SE is skin lesions. Marked improvement within 24 hours after
characterized by pronounced hyperplasia of the synovial treatment with penicillin supports the diagnosis. At
intima and subintimal connective tissue, with vascular- necropsy the presence of an enlarged spleen is sugges-
ization and accumulation of lymphoid cells and tive.
macrophages, forming a villous pad of inflammatory tis-
sue. Deposition and organization of fibrin may be seen. Bacteriologic Diagnosis
As the lesion progresses, proliferation of fibrous connec- Isolation of E. rhusiopathiae from the acutely affected an-
tive tissue becomes more prominent, and long fronds of imal provides a definite laboratory diagnosis of SE. He-
hyperplastic synovium may be seen. The surface lining moculture is a useful diagnostic aid in living animals, but
may become necrotic, with deposition of a fibrinous to specimens should be taken from several affected animals
fibrinopurulent exudate. Some tendency to follicle for- in the herd, as the presence of the organism in the blood
mation may be evident in the heavy accumulations of of an individual may be inconstant. At necropsy of a pig
lymphoid cells. There may be erosion of the articular car- that has died in the acute phase, the organism is easily
tilages along with periostitis and osteitis. In old lesions cultured from a variety of body organs (heart, lungs, liv-
ankylosis of the involved joint by fibrous adhesion may er, spleen, kidneys, joints). If the illness has persisted for
be accompanied by calcification. several days, however, the organism often can no longer
Vegetative growths on the heart valves are composed be cultured from internal organs but may still be found
of granulation tissue and superimposed masses of fib- in the joints. Under these conditions it is important to
rin. Connective-tissue proliferation occurs with addition- take several specimens of fluid and synovial tissue from
al fibrin formation, which can be the source of emboli. as many synovial sacs of a joint as possible, because the

31.3. Synovitis and arthritis


of chronic SE in a hock joint 8
weeks after exposure to E.
rhusiopathiae. Note hyperemia
and proliferation of synovial
tissue. (Courtesy National Animal
Disease Center, Ames, Iowa).
CHAPTER 31 ERYSIPELAS Wood 427

organisms may be present in small numbers and limited for acute erysipelas is administration of penicillin. E.
to certain areas. rhusiopathiae is highly sensitive to this antibiotic, and
Culture of E. rhusiopathiae from tissue specimens is treatment early in an acute outbreak usually results in
relatively simple and requires only basic laboratory dramatic response within 24–36 hours. Specific treat-
equipment and culture media such as tryptose or meat ment regimens generally involve giving penicillin alone
infusion media with or without blood or serum added. or in combination with other antibiotics or antiserum
Care should be taken to avoid accidental skin infection, (occasionally both) to provide a longer action. For exam-
as the organism is pathogenic for humans. Selective cul- ple, long-acting penicillin (available under various pro-
ture methods for isolation of the organism from conta- prietary names), consisting of a combination of 150,000
minated specimens are described elsewhere (Cottral units procaine penicillin G and 150,000 units benzathine
1978). penicillin G/cm3, may be given intramuscularly at a sin-
The use of immunofluorescence for rapid identi- gle dose of 5000–10,000 units/pound (454 g) to visibly
fication of E. rhusiopathiae has been reported; how- sick pigs (the entire herd may be treated with tetracycline
ever, the method may not be sufficiently specific and sen- in the drinking water: 500 mg/gallon, 132 mg/L) until 5
sitive for routine diagnostic purposes (Harrington et al. days after no sick pigs are observed. The entire herd may
1974). also be given antiserum if the outbreak is very severe. As
an alternative, long-acting penicillin may be given in se-
Serologic Diagnosis vere outbreaks, and procaine penicillin G in less severe
A variety of serologic tests have been used in attempts to cases. The use of antiserum for treatment of suckling
diagnose SE. These include plate, tube, and microtitra- pigs is a fairly common practice. Initiation of a vaccina-
tion agglutination; passive hemagglutination; hemagglu- tion program in previously unvaccinated herds where
tination inhibition; complement fixation; enzyme-linked outbreaks occur is strongly recommended.
immunosorbent assay (ELISA); and indirect immunoflu- Although penicillin has been consistently found to be
orescence. An agglutination test involving the use of the most effective antibiotic for treatment of acute SE,
growing culture as antigen was developed by Wellmann satisfactory results have been reported also with tetracy-
(1955). In this test, called the Wachstumsprobe or growth- clines (including chlortetracycline and oxytetracycline),
agglutination test, a culture of E. rhusiopathiae growing lincomycin, and tylosin. The organism is sensitive in
in liquid medium in the presence of sterile test serum ag- vitro to erythromycin, but this antibiotic has been re-
glutinates if sufficient specific antibody is present. ported to be relatively ineffective in vivo. Streptomycin,
No serologic test has proved useful for routine diag- dihydrostreptomycin, chloramphenicol, bacitracin,
nosis of acute infection or for differentiation between polymyxin B, neomycin, and sulfonamides are not effec-
immune and susceptible pigs. Serologic diagnosis may tive against SE. There have been no published reports of
have some value in detection of chronic infection, pri- development of resistance by E. rhusiopathiae to peni-
marily on a herd basis. Microtitration agglutination, cillin in the field since use of the antibiotic for treatment
growth agglutination, and ELISA are probably the most of SE was first reported in 1949. However, some isolates
reliable for this purpose but may be difficult to interpret. of the organism from swine have been found to be resis-
It can be concluded that serologic testing has limited tant to tetracyclines.
practical application in clinical diagnosis of SE in the There is no practical treatment for chronic SE. Exper-
field. The chief value of serologic procedures resides in imentally, the administration of antiinflammatory
research. agents has provided some alleviation of the effects of
chronic arthritis, and they may be used in treatment of
TREATMENT especially valuable individual animals.

The treatment of SE with hyperimmune serum, usually PREVENTION


obtained from horses, was introduced in 1899, several
years after it had been developed for use in conjunction Prevention of SE is best accomplished by sound prac-
with live-culture vaccination. Until the introduction of tices of herd health management, including a program of
antibiotics nearly 50 years later, the administration of immunization.
antiserum was the only worthwhile available form of
treatment. Although now considered obsolete by some General Management Practices
veterinarians, antiserum can still be useful. For maxi- Swine should be raised according to sound husbandry
mum effectiveness the serum must be given early in the practice relative to nutrition, housing, and condition of
course of the disease. The recommended therapeutic lots and pastures, and they should be observed regularly
dose, given subcutaneously, varies from 5 to 10 mL for for deviations from their usual attitude. Replacements
pigs weighing less than 50 pounds (23 kg) to 20–40 mL should be obtained from clean sources. The recent intro-
for pigs weighing more than 100 pounds (45 kg). duction of a new boar is a relatively common historical
It is generally accepted that the treatment of choice finding preceding acute outbreaks of SE in a herd. New-
428 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

ly purchased animals should be isolated for at least 30 ment for generating and distributing the aerosol is nec-
days. essary.
It is advisable to eliminate chronically affected swine Use of living vaccines leaves open the possibility,
from the herd, as they can remain carriers of the organ- however remote, of vaccinated animals becoming carri-
ism indefinitely. ers and disseminators of the organism, which conceiv-
Good sanitation is important in general herd man- ably could undergo increased virulence through serial
agement and is essential following the cessation of an passage. There is no experimental evidence, however,
outbreak. Walls and floors should be cleaned and disin- that attenuated SE vaccines can regain their virulence
fected. Phenolic, alkali, hypochlorite, or quaternary am- and pose a hazard to susceptible swine.
monium disinfectants are effective against the organism
but must be applied to clean surfaces. BACTERINS. Use of a bacterin consisting of a forma-
lin-killed whole culture adsorbed on aluminum hydrox-
Immunization ide gel was first reported in 1947. This type of product
A variety of biological products have been produced for has been used in the United States since 1953. It is typi-
the purpose of conferring immunity to SE in swine. The cally made from selected strains of serovar 2 that pro-
simultaneous or serum-culture method of immunization duce a soluble immunogenic product when grown in a
was introduced in 1893 and consists of concomitant in- complex liquid medium containing serum. This sub-
jections of virulent culture and antiserum. This method stance, most of which is released into the medium, has
was first used in the United States in 1938, and its use been described as a glyco-lipoprotein (White and Verwey
continued for about 20 years until safer products became 1970). It is considered by most investigators to be a nec-
available. The method is no longer used. Active immu- essary ingredient for stimulation of immunity by the
nization against SE is now carried out by the use of either bacterin. The most active component of the immuno-
attenuated (so-called avirulent) vaccines or nonliving genic substance has been identified as a protein fraction
products (bacterins). with a molecular weight of 64–66 kDa (Timoney and
Groschup 1993; Sato et al. 1995; Goodman 1996;
ATTENUATED VACCINES. Vaccines made from E. Zarkasie et al. 1996). The combination of the soluble im-
rhusiopathiae of reduced virulence were first licensed in munogenic product and whole killed bacteria, concen-
the United States in 1955. Attenuation of virulence has trated and adsorbed on aluminum hydroxide gel or oth-
been accomplished by passage through rabbits or chick- er suitable adjuvant, constitutes the basic features of an
en embryos, by air-drying, or by growth in media con- E. rhusiopathiae bacterin.
taining acridine dyes. Although these vaccines are com- Lysate bacterin, first reported in 1953, has been used
monly referred to as avirulent, they are in fact strains of in the United States since 1955. It is similar to whole-cul-
extremely low virulence for swine, often retaining some ture bacterin except that the bacterial cells have been
virulence for mice. They stimulate immunity in swine by lysed.
limited multiplication in the body; therefore, the re- Bacterins are given by subcutaneous or intramuscu-
sponse to vaccination is subject to such variables as sta- lar injection; a second (booster) injection in 3–5 weeks is
tus of passive or active immunity already existing in the generally recommended. Breeding animals should be
animal. In addition, there is some evidence that anti- given an additional booster injection annually.
serum given concomitantly may interfere with develop-
ment of immunity in response to attenuated vaccines. PASSIVE IMMUNITY. Temporary passive immunity
Manufacturers generally do not recommend use of can be induced by administration of commercially avail-
serum with their attenuated products except when im- able antiserum. Pigs given antiserum subcutaneously re-
mediate protection is necessary, as in the case of suckling ceive immediate passive protection, which persists for
pigs being given both vaccine and serum during a herd about 2 weeks. The preventive dose is half the therapeu-
outbreak. In this case, repeated vaccination at weaning is tic dose (see the section on treatment). Antiserum may
recommended. be useful during a herd outbreak for temporary protec-
Attenuated vaccines should not be given to swine be- tion of suckling pigs until they are old enough to be vac-
ing treated with antibiotics to which E. rhusiopathiae is cinated.
sensitive. Antibiotic treatment should be discontinued at
least 8–10 days before vaccination. MECHANISM OF IMMUNITY. The mechanism of
Attenuated vaccines are usually given by injection or immunity to E. rhusiopathiae infection is not clearly de-
administered orally in drinking water. Some manufac- fined, but there is little doubt that opsonization is a ma-
turers provide a product that can be given either way. In jor factor. Studies have shown that virulent E. rhu-
some parts of Europe and the former Soviet Union, vac- siopathiae bacteria opsonized with immune serum were
cination by aerosol has been practiced. Elaborate equip- readily eliminated by polymorphonuclear leukocytes
CHAPTER 31 ERYSIPELAS Wood 429

(Sawada et al. 1988) and by macrophages (Shimoji et al. Press, pp. 429–436, 671, 672, 679, 687.
1996). Nonopsonized virulent organisms were resistant Cysewski, S. J.; Wood, R. L.; Pier, A. C.; and Baetz, A. L.
to phagocytosis. 1978. Effects of aflatoxin on the development of ac-
quired immunity to swine erysipelas. Am J Vet Res
EFFICACY OF BIOLOGICS. No presently available 39:445–448.
immunizing product adequately fills the need for effec- Denecke, R., and Trautwein, G. 1986. Lokale Antigenpersis-
tive long-term protection against SE. Some veterinarians tenz und Chronizität der experimentellen Rotlauf-Pol-
consider living vaccines to be superior to bacterins, but yarthritis. Berl Münch Tierärztl Wochenschr
Shuman (1959) found no significant difference in their 99:200–208.
efficacies under experimental conditions. According to Drommer, W.; Schultz, L. C.; and Pohlenz, J. 1970. Experi-
most reports, vaccination generally can be expected to menteller Rotlauf beim Schwein: Permeabilitatsstorun-
induce immunity lasting 3–5 months. A second (boost- gen und Malazien im zentralen Nervensystem. Pathol
er) injection may increase this duration to 6 months and Vet 7:455–473.
is recommended, especially for bacterins. Development Franz, B.; Davies, M. E.; and Horner, A. 1995. Localization
of immunity in vaccinated swine may be adversely af- of viable bacterial antigens in arthritic joints of
fected by such environmental factors as overheating or Erysipelothrix rhusiopathiae-infected pigs. FEMS Im-
poor nutrition. munol Med Microbiol 12:137–142.
A serious deficiency of SE vaccination is its inability Goodman, S. A. 1996. USDA: Progress toward in vitro tests
to prevent the chronic form. Most investigators agree and other trends. Dev Biol Stand 86:41–47.
that vaccination has little effect on the incidence of Harrington, R., Jr.; Wood, R. L.; and Hulse, D. C. 1974. Com-
arthritis caused by E. rhusiopathiae, although this obser- parison of a fluorescent antibody technique and cultur-
vation is difficult to evaluate in the field, since SE vacci- al method for the detection of Erysipelothrix rhusiopathi-
nation is not universally practiced in the United States. It ae in primary broth cultures. Am J Vet Res 35:461–462.
is possible that vaccination reduces the overall preva- Hermanns, W.; Jessen, H.; Schulz, L. C.; Kerlen, G.; and
lence of arthritis by reducing the prevalence of acute Böhm, K. H. 1982. Über die Induktion einer chronischen
erysipelas. On the other hand, some believe vaccination Polyarthritis mit Bestandteilen von Rotlaufbakterien
actually causes an increase in arthritic lesions by initiat- (Erysipelothrix rhusiopathiae). II. Mitteilung: Versuche
ing a state of hypersensitivity to subsequent contact with zur Arthritis-Induktion bei Ratten. Zentralbl Veter-
the organism. An alternative explanation for the failure inärmed (B) 29:85–98.
of vaccination to prevent arthritis may exist, however. Kucsera, G. 1973. Proposal for standardization of the desig-
The organism may be carried to synovial tissues by nations used for serotypes of Erysipelothrix rhusiopathiae
loaded macrophages soon after exposure, thereby escap- (Migula) Buchanan. Int J Syst Bacteriol 23:184–188.
ing the opsonic effects of humoral immunity (Drommer Miniats, O. P.; Spinato, M. T.; and Sanford, S. E. 1989. Acti-
et al. 1970). Sequestration of the bacteria in chondro- nobacillus suis septicemia in mature swine: Two out-
cytes (Franz et al. 1995) might provide similar protection breaks resembling erysipelas. Can Vet J 30:943–947.
from immune mechanisms. Müller, H. E. 1981. Neuraminidase and other enzymes of
It is possible that certain uncommon serovars of E. Erysipelothrix rhusiopathiae as possible pathogenic fac-
rhusiopathiae may be refractory to the immunity induced tors. In Arthritis: Models and Mechanisms. Ed. H. De-
in mice and swine by standard SE vaccines. However, icher. Berlin: Springer-Verlag, p. 58.
such serovars are usually isolated from healthy carrier Nakato, H.; Shinomiya, K.; and Mikawa, H. 1987. Adhesion
pigs or nonporcine sources, and none have been directly of Erysipelothrix rhusiopathiae to cultured rat aortic en-
associated with cases of acute SE in the field. dothelial cells: Role of bacterial neuraminidase in the in-
Although vaccination against SE is not entirely effec- duction of arteritis. Path Res Pract 182:255–260.
tive in preventing the disease, it provides a worthwhile Nørrung, V. 1979. Two new serotypes of Erysipelothrix rhu-
means of control when used with other good manage- siopathiae. Nord Vet Med 31:462–465.
ment practices. A regular vaccination program for both Nørrung, V., and Molin, G. 1991. A new serotype of
breeding and market animals is recommended. Because Erysipelothrix rhusiopathiae isolated from pig slurry. Acta
of the ubiquity of E. rhusiopathiae, together with its Vet Hung 39:137–138.
poorly understood ability to exist in nature, the possibil- Nørrung, V.; Munch, B.; and Larsen, H. E. 1987. Occur-
ity of eradication of the organism seems remote. rence, isolation and serotyping of Erysipelothrix rhu-
siopathiae in cattle and pig slurry. Acta Vet Scand
REFERENCES 28:9–14.
Sato, H.; Hirose, K.; and Saito, H. 1995. Protective activity
Cottral, G. E. 1978. Manual of Standardized Methods for and antigenic analysis of fractions of culture filtrates of
Veterinary Microbiology. Ithaca, N.Y.: Cornell Univ Erysipelothrix rhusiopathiae. Vet Microbiol 43:173–182.
430 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Sawada, T.; Tamura, Y.; and Takahashi, T. 1988. Mechanism of Erysipelothrix rhusiopathiae strains of serovar 1a to tis-
of protection induced in mice against Erysipelothrix rhu- sue culture cells originated from porcine kidney and
siopathiae infection by treatment with porcine anti- their pathogenicity in mice and swine. Vet Microbiol
serum to the culture filtrate of an attenuated strain. Vet 13:57–64.
Microbiol 17:65–74. Takahashi, T.; Nagamine, N.; Kijima, M.; Suzuki, S.; Takagi,
Schulz, L. C.; Drommer, W.; Seidler, D.; Ehard, H.; Leim- M.; Tamura, Y.; Nakamura, M.; Muramatsu, M.; and
beck, R.; and Weiss, R. 1975a. Experimenteller Rotlauf Sawada, T. 1996. Serovars of Erysipelothrix strains isolat-
bei verschiedenen Spezies als Modell einer systemischen ed from pigs affected with erysipelas in Japan. J Vet Med
Bindegewebskrankheit. II. Chronische Phase mit beson- Sci 58:587–589.
derer Berucksichtigung der Polyarthritis. Beitr Pathol Timoney, J. F., and Groschup, M. M. 1993. Properties of a
154:27–51. protective protein antigen of Erysipelothrix rhusiopathi-
Schulz, L. C.; Drommer, W.; Seidler, D.; Ehard, H.; Von ae. Vet Microbiol 37:381–387.
Mickwitz, G.; Hertrampf, B.; and Böhm, K. H. 1975b. Vickers, C. L., and Bierer, B. W. 1958. Triple sugar iron agar
Experimenteller Rotlauf bei verschiedenen Spezies als as an aid in the diagnosis of erysipelas. J Am Vet Med As-
Modell einer systemischen Bindegewebskrankheit. I. soc 133:543–544.
Systemische vaskulare Prozesse bei der Organmanifesta- Wellmann, G. 1955. Die subklinische Rotlaufinfektion und
tion. Beitr Pathol 154:1–26. ihre Bedeutung für die Epidemiologie des Schweinerot-
Schulz, L. C.; Drommer, W.; Ehard, H.; Hertrampf, B.; Lei- laufs. Zentralbl Bakteriol (Orig A) 162:265–274.
bold, W.; Messow, C.; Mumme, J.; Trautwein, G.; Über- White, R. R., and Verwey, W. F. 1970. Solubilization and
schär, S.; Weiss, R.; and Winklemann, J. 1977. Patho- characterization of a protective antigen of Erysipelothrix
genetische Bedeutung von Erysipelothrix rhusiopathiae in rhusiopathiae. Infect Immun 1:387–393.
der akuten und chronischen Verlaufsform der Rotlaufar- White, T. G., and Shuman, R. D. 1961. Fermentation reac-
thritis. Dtsch Tierärzt Wochenschr 84:107–111. tions of Erysipelothrix rhusiopathiae. J Bacteriol
Shimoji, Y.; Yokomizo, Y.; Sekizaki, T.; Mori, Y.; and Kubo, 82:595–599.
M. 1994. Presence of a capsule in Erysipelothrix rhu- White, T. G.; Puls, J. L.; and Hargrave, P. 1975. Production
siopathiae and its relationship to virulence for mice. In- of synovitis in rabbits by fractions of a cell-free extract
fect Immun 62:2806–2810. of Erysipelothrix rhusiopathiae. Clin Immunol Im-
Shimoji, Y.; Yokomizo, Y.; and Mori, Y. 1996. Intracellular munopathol 3:531–540.
survival and replication of Erysipelothrix rhusiopathiae Wood, R. L. 1973. Survival of Erysipelothrix rhusiopathiae in
within murine macrophages: Failure of induction of the soil under various environmental conditions. Cornell
oxidative burst of macrophages. Infect Immun Vet 63:390–410.
64:1789–1793. ———. 1984. Swine erysipelas: A review of prevalence
Shuman, R. D. 1951. Swine erysipelas induced by skin scar- and research. J Am Vet Med Assoc 184:944–949.
ification. Proc Am Vet Med Assoc, p. 153. Wood, R. L., and Harrington, R., Jr. 1978. Serotypes of
———. 1959. Comparative experimental evaluation of Erysipelothrix rhusiopathiae isolated from swine and from
swine erysipelas bacterins and vaccines in weanling pigs, soil and manure of swine pens in the United States. Am
with particular reference to the status of their dams. Am J Vet Res 39:1833–1840.
J Vet Res 20:1002–1009. Wood, R. L., and Shuman, R. D. 1981. Erysipelothrix infec-
———. 1971. Erysipelothrix. In Infectious and Parasitic tion. In Infectious Diseases of Wild Mammals, 2d ed.
Diseases of Wild Birds. Ed. J. W. Davis, R. C. Anderson, Ed. J. W. Davis, L. H. Karstad, and D. O. Trainer,
L. H. Karstad, and D. O. Trainer. Ames: Iowa State Univ 297–305. Ames: Iowa State Univ Press.
Press, p. 141. Wood, R. L.; Haubrich, D. R.; and Harrington, R., Jr. 1978.
Smith, T. 1885. Second Annual Report of the Bureau of An- Isolation of previously unreported serotypes of
imal Industry. Washington, D.C.: U.S. Department of Erysipelothrix rhusiopathiae from swine. Am J Vet Res
Agriculture, p. 187. 39:1958–1961.
Sneath, P. H. A.; Mair, N. S.; Sharpe, M. E.; and Holt, J. G. Xu, K.; Hu, X.; Gao, C.; and Lu, Q. 1984. A new serotype of
1986. Bergey’s Manual of Systematic Bacteriology. Vol. Erysipelothrix rhusiopathiae. Anim Infect Dis 4:11–14.
2. Baltimore: Williams & Wilkins, pp. 1245–1249. Xu, K.; Gao, C.; and Hu, X. 1986. Study on a new serotype of
Takahashi, T.; Fujisawa, T.; Benno, Y.; Tamura, Y.; Sawada, Erysipelothrix rhusiopathiae isolated from marine fishes.
T.; Suzuki, S.; Muramatsu, M.; and Mitsuoka, T. 1987a. Anim Infect Dis 3:6–7, 48.
Erysipelothrix tonsillarum sp. nov. isolated from tonsils of Zarkasie, K.; Sawada, T.; Yoshida, T.; Takahashi, I.; and
apparently healthy pigs. Int J Syst Bact 37:166–168. Takahashi, T. 1996. Growth ability and immunological
Takahashi, T.; Hirayama, N.; Sawada, T.; Tamura, Y.; and properties of Erysipelothrix rhusiopathiae serotype 2. J
Muramatsu, M. 1987b. Correlation between adherence Vet Med Sci 58:87–90.
Escherichia coli Infections
32 H. U. Bertschinger and J. M. Fairbrother

INTRODUCTION
H. U. Bertschinger
The genus Escherichia is named after the German pedia- trum of antisera. This may be quite suitable, since in a
trician Theodor Escherich (1857–1911). It is classified given region, animal species, and organ, pathogenic
with the family Enterobacteriaceae, which belongs to the serotypes maintain their characteristic antigenic make-
gram-negative facultatively anaerobic rods. The species up. Thus one may deduce the complete serotype from a
Escherichia (E.) coli includes normal inhabitants of the simple slide agglutination with a living culture. Serotyp-
gastrointestinal tract and organisms causing a broad va- ing is diagnostically helpful in communicable types of
riety of intestinal and extraintestinal diseases in the disease caused by a limited number of serotypes, such as
porcine species. postweaning E. coli diarrhea and edema disease.

BACTERIOLOGY VIRULENCE FACTORS

E. coli are gram-negative, peritrichous flagellated rods of Bacterial traits involved in pathogenesis are called
variable length and with a diameter of about 1 µm. virulence factors. Much has been learned in this fast-
Colonies on solid media reach their full size within 1 day progressing field (Gyles 1994), but we are far from
of incubation. The appearance may vary from smooth to understanding every step in the development of the
rough or mucoid. There is a broad range of selective me- different kinds of disease caused by E. coli. Potent
dia available. Some strains produce hemolysins. Species exotoxins trigger the secretion of fluid into the gut
identification relies mainly on biochemical characters. It lumen in enterotoxigenic E. coli (ETEC) infections and
should be borne in mind that there are exceptions with are responsible for systemic pathology caused by entero-
every single biochemical character. Commercially avail- toxemic E. coli (ETEEC) strains. Endotoxin is present in
able identification kits therefore make use of up to 50 the outer membrane of most E. coli strains. Its signifi-
characters to achieve a high level of accuracy. The inter- cance is well documented only in extraintestinal infec-
pretation may be facilitated by computer-assisted pro- tions, such as septicemia, mastitis, and urinary tract in-
cessing of the data. The determination of DNA related- fections.
ness, the scientific base of discrimination between Many E. coli infections require the colonization of
species, is restricted to research laboratories. mucous membranes. With ETEC and ETEEC, adhesion
to the small intestine is mediated by extracellular pro-
SEROTYPING teinaceous appendages, which are called fimbriae or pili
and are highly host-specific. In some strains capsular
There are several ways to subdivide the species into polysaccharide has been shown to enhance the ability to
types. So far, serotypes have shown the best association colonize. Enteropathogenic E. coli (EPEC) colonizing the
with certain virulence traits. Complete serotyping in- lower gastrointestinal tract adhere by an attaching and
cludes determination of O (somatic), K (capsular or mi- effacing mechanism. In the pig, colonization of the uri-
crocapsular), H (flagellar), and F (fimbrial) antigens. Un- nary tract has received little attention so far.
like salmonellae, only a small percentage of E. coli Some E. coli utilize high-affinity iron-uptake systems
isolates are typeable with available antisera, since sero- to compete with the host for available iron. In extrain-
typing has been limited to isolates of proven or sus- testinal sites E. coli have to resist the natural bactericidal
pected pathogenicity. Presently, at least 173 O, 80 K, 56 activity of serum, a characteristic called serum resis-
H, and a fast-growing number of F antigens are officially tance. A given pathogenic strain may exhibit a whole set
recognized. of virulence factors, that is, more than one toxin and
In diagnostic laboratories serotyping is often reduced even more than three adhesion factors. Detection of
to one or two classes of antigens and to a limited spec- more virulence factors can be expected.

431
432 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

GENETICS OF VIRULENCE most extraintestinal infections in the pig as well.


The primary habitat of porcine E. coli is the gastroin-
Very specific sets of virulence factors are needed to cause testinal tract. The E. coli flora of individual pigs is ex-
a particular disease. Thus, strains causing enteric dis- tremely complex. When strains were distinguished by
eases are not associated with extraintestinal infections the combined application of O serogrouping, biotyping,
and vice versa. E. coli strains may be classed into patho- and resistogram typing, up to 25 strains were identified
types based on the production of particular virulence de- in the gastrointestinal tract of one individual (Hinton et
terminants. Many virulence factors examined so far are al. 1985). Numerically dominant strains change at inter-
plasmid determined. This applies in particular to he- vals from 1 day to several weeks, leading to successive
molysins, toxins, and adhesins of ETEC. In strains from waves of dominant strains (Katouli et al. 1995). Prolifer-
most extraintestinal infections, however, the genes en- ation of E. coli takes place mainly in the course of the
coding for fimbriae, cytotoxins, and hemolysin are chro- passage through the small intestine. Increase from the
mosomally located. In the laboratory, plasmids can easi- ileum to the rectum is minimal or absent (McAllister et
ly be transmitted from donor to recipient strains. al. 1979). Numbers in the large intestine fluctuate
However, such exchanges of genetic material do not ap- around 107 colony-forming units/g. However, E. coli con-
pear to play a major role in the field. The genetic makeup tribute less than 1% to the total bacterial count. When
of pathogenic E. coli strains is remarkably stable. This found elsewhere (feed, water, soil, etc.), E. coli are derived
may be because a whole set of virulence factors is in- from this habitat, usually by fecal contamination. Long
volved in the virulence of a particular strain, and certain survival times in the environment are promoted by low
recipient strains may not express transmitted plasmid- temperature and sufficient available water, among other
determined functions. The clinically important develop- factors. In an experiment with five slurry samples, a
ment of antimicrobial resistance is an exception to this porcine E. coli O139:K82(B) retained viability for be-
observation. tween 5 and over 11 weeks (Burrows and Rankin
The time-consuming and sometimes cumbersome as- 1970).
says for virulence factors are increasingly being replaced
by hybridization and polymerase chain reaction (PCR) REFERENCES
techniques. However, these techniques are not yet used
in all diagnostic laboratories. Awad-Masalmeh, M.; Baumgartner, W.; Passernig, A.; Sil-
ber, R.; and Hinterdorfer, F. 1990. Bakteriologische Un-
ECOLOGY tersuchungen bei an puerperaler Mastitis (MMA-Syn-
drom) erkrankten Sauen verschiedener Tierbestände
The particular ecology of pathogenic E. coli strains has Österreichs. Tierärztl Umschau 45:526–535.
been somewhat neglected. Intestinal infections caused Burrows, M. R., and Rankin, J. D. 1970. A further examina-
by ETEC and ETEEC are often contagious. The same tion of the survival of pathogenic bacteria in cattle slur-
strain is usually found in many sick pigs and often in con- ry. Br Vet J 126:32–34.
secutive batches of pigs. When edema disease spread Gyles, C. L. 1994. Escherichia coli in Domestic Animals and
through Denmark, 63% of the outbreaks were connected Humans. Wallingford, U.K.: CAB International.
by trade of pigs to one single infected breeding herd (Jor- Hinton, M.; Hampson, D. J.; Hampson E.; and Linton, A. H.
sal et al. 1996). On the other hand, clinical edema disease 1985. A comparison of the ecology of Escherichia coli in
occurred in no more than 5% of the herds with trading the intestine of healthy unweaned pigs and pigs after
contacts. These strains may sometimes appear in healthy weaning. J Appl Bact 58:471–478.
pigs without overt disease. They are usually shed in high Jorsal, S. E.; Aarestrup, F. M.; Ahrens, P.; Johansen, M.; and
numbers for a period of a few days only. The ensuing Baekbo, P. 1996. Oedema disease in Danish pig herds:
dramatic decrease seems to be due to the development of Transmission by trade of breeding animals. Proc Int
local immunity. Congr Pig Vet Soc 14:265.
Extraintestinal infections, however, do not behave Katouli, M.; Lund, A.; Wallgren, P.; Kühn, I., Söderlind, O.;
like communicable diseases. Individual pigs in a given and Möllby, R. 1995. Phenotypic characterization of in-
herd are affected most often by different strains. Mixed testinal Escherichia coli of pigs during suckling, post-
infections by more than one strain are frequent. In hu- weaning and fattening periods. Appl Environ Microbiol
mans, the fecal flora is obviously the reservoir of such 61:778–783.
pathogenic strains. In 18 out of 67 sows with mastitis, E. McAllister, J. S.; Kurtz, H. J.; and Short, E. C., Jr. 1979.
coli of the same O types present in samples of mammary Changes in the intestinal flora of young pigs with post-
secretion were isolated from the feces (Awad-Masalmeh weaning diarrhea or edema disease. J Anim Sci
et al. 1990). This underlines the endogenous character of 49:868–879.
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 433

NEONATAL ESCHERICHIA COLI DIARRHEA


J. M. Fairbrother

Diarrhea has become an economically important disease toxins STa (STI), STb (STII), or LT. Until recently, the
in pigs as a result of increasing intensification of farrow- most commonly observed ETEC in cases of neonatal di-
ing management. It may be classified into three main en- arrhea belonged to the classic serogroups O149, O8,
tities: neonatal diarrhea (within the first few days of O147, and O157, were F4-positive, and produced the en-
birth), young piglet diarrhea (from the first week of birth terotoxins LT and STb (Harel et al. 1991; Soderlind et al.
to weaning), and postweaning diarrhea. Escherichia coli 1988; Wilson and Francis 1986). An increasing number
is the most important etiologic agent of neonatal and of ETEC of serogroups such as O8, O9, O64, and O101
postweaning diarrhea. Etiologic agents of diarrhea in which are F5-, F6-, and/or F41-positive and mainly pro-
young piglets are more numerous and include transmis- duce the enterotoxin STa, or less often STb, are now be-
sible gastroenteritis virus, rotavirus, coccidia, and E. coli ing isolated. These ETEC cause diarrhea mainly in pigs
(Biehl and Hoefling 1986). E. coli are normal inhabitants aged from 0 to 6 days, and to a lesser extent in older pigs,
of the intestinal tract of animals but may also cause dis- whereas F4-positive ETEC are often isolated in diarrheic
ease. Pathogenic enteric E. coli belong to a restricted pigs from birth to weaning. Fewer F4-positive ETEC
number of serogroups and produce one or more viru- within the classic serogroups, especially O157, are now
lence factors, which are not usually found in nonpatho- being isolated.
genic E. coli. Over the last several years, our knowledge of Enteric colibacillosis complicated with shock also oc-
enteric E. coli virulence factors and their role in the curs in young pigs before and after weaning. E. coli asso-
pathogenesis of enteric disease has rapidly expanded. ciated with this disease either (1) are ETEC and com-
The terminology used to describe pathogenic E. coli has monly belong to serogroups O149, O157, and O8, which
greatly changed. Thus, E. coli may now be categorized by are F4-positive, produce STb and LT, but only occasion-
pathotype, based on production of virulence factors. ally produce Shiga-like toxin (SLT-IIe) (Faubert and Dro-
Specific E. coli pathotypes are associated with each of the let 1992), or (2) produce SLT-IIe and are associated with
main disease entities. A less frequently encountered edema disease. The latter will be discussed in a later sec-
manifestation of enteric E. coli infection is an acute shock tion.
syndrome which provokes gross lesions of hemorrhagic
gastroenteritis. Neonatal diarrhea in pigs has been re- EPIDEMIOLOGY
viewed by Alexander (1994).
The occurrence of E. coli diarrhea depends on an interac-
ETIOLOGY tion between the causative bacteria, environmental con-
ditions, and certain host factors. Only E. coli that carry
Neonatal diarrhea associated with E. coli is observed virulence factors as described in the previous section and
most commonly in pigs aged from 0 to 4 days. Causative are ingested in large numbers are able to cause diarrhea.
strains produce one or more enterotoxins and have been The newborn pig, on leaving the uterus and before reach-
designated enterotoxigenic E. coli (ETEC). ETEC adhere ing the teats of the sow, encounters the heavily contami-
to the small intestinal mucosa in neonatal pigs by means nated environment of the farrowing crate and the skin of
of one or more of the fimbrial adhesins F4 (K88), F5 the dam, resulting in ingestion of microbes from the in-
(K99), F6 (987P), or F41 (Table 32.1). They colonize the testinal flora of the sow. Thus, in conditions of poor hy-
small intestine and produce one or more of the entero- giene or in a continuous-farrowing system, a buildup of
pathogenic E. coli in the environment could lead to an
outbreak of neonatal E. coli diarrhea. The colostrum con-
Table 32.1. Predominant E. coli pathotypes associ- tains nonspecific bactericidal factors and specific anti-
ated with neonatal diarrhea in piglets body (IgA) that inhibit the adherence of pathogenic E.
Toxin(s) Fimbrial Adhesins O Serogroups coli in the intestine. If the dam has not been exposed to
the pathogenic E. coli present in the environment of the
STa F5, F5 + F41, F6, F6 + F41 O8, O9, O20, O64, O101 piglets, specific antibodies are not present in the
F41, F5 + 6, F5 + F6 + F41 O141
Sta + STb F4, F4 + 5, F5 + F6, F6, O20, O64, O141 colostrum, and the piglets are susceptible to infection.
not known Similarly, when individual piglets do not have access to
LT + STb F4, F4 + F6 O8, O147, O149, O157 colostrum, due to injury or inability to compete or due to
LT + Sta + agalactia or insufficient teats of the sow, they are more
STb F4, F4 + F6 O8, O147, O149, O157
susceptible to infection. Ambient temperature in the far-
STb F4, F6, not known O149
rowing house is also very important. In pigs kept at tem-
434 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

peratures of less than 25˚C, intestinal peristaltic activity mids (F4, F5, F6). Many fimbriae, such as F1 and F6, un-
is greatly reduced, and passage of bacteria and protective dergo phase variation and may be very poorly expressed
antibodies through the intestine is delayed (Sarmiento after several passages in culture conditions. Other fim-
1983). Increased numbers of pathogenic E. coli in the in- briae (F5 and F41) undergo quantitative variation and
testinal tract of these pigs result in a more severe diar- are only well expressed in culture media low in glucose or
rhea than in pigs kept at 30˚C. alanine such as Minca medium (Guinée et al. 1977).
Fimbriae adhere to specific receptors on the cell
PATHOGENESIS membrane of intestinal epithelial cells and to specific re-
ceptors or nonspecifically in the mucus coating the ep-
In the presence of the appropriate predisposing environ- ithelium. ETEC producing fimbriae F5, F6, and F41
mental conditions and host factors, pathogenic E. coli mostly colonize the posterior jejunum and ileum, where-
proliferate in the intestine and cause diarrhea by means as F4-positive ETEC tend to colonize the length of the je-
of specific virulence factors. Pathogenesis will be dis- junum and the ileum. Certain pigs do not have receptors
cussed with respect to the E. coli categories defined by the for the F4 adhesin on intestinal epithelial cells and are
production of these factors. thus resistant to infection by F4-positive ETEC (Sellwood
et al. 1975). This genetic resistance to infection is inher-
Enterotoxigenic E. coli ited in a simple Mendelian way, and the allele for the re-
Most pathogenic E. coli produce one or more fimbrial ad- ceptor is dominant. Subsequent studies have demon-
hesins that mediate their attachment to specific recep- strated at least five pig phenotypes, based on
tors on mucosal epithelial cells and in the adjacent susceptibility of brush borders of different pigs to ad-
mucous layer. These fimbriae (or pili) are hairlike ap- herence of isolates producing the different variants F4ab,
pendages extending from the bacterial cell and consist of F4ac, and F4ad (Bijlsma et al. 1982; Hu et al. 1993). The
structural protein subunits that, in many cases, act as a loci encoding porcine intestinal receptors for F4ab and
support for a separate adhesive protein found at the tips F4ac are closely linked on chromosome 13 (Edfors-Lilja
of the fimbriae. Fimbriae are classified by serologic reac- et al. 1995). A similar genetic resistance has not been ob-
tivity or by receptor specificity, the latter being manifest- served for the other fimbriae of neonatal porcine ETEC.
ed by agglutination of red blood cells from different ani- On the other hand, there appears to be an age resistance
mal species. The nomenclature for fimbriae has been to infection by F5-positive isolates, which is not observed
very diverse. For example, the first fimbrial adhesins for F4-positive isolates. Piglets are most susceptible to in-
demonstrated on porcine ETEC were thought to be cap- fection with F5-positive ETEC during the first several
sular antigens and were named K88 and K99. A more days of life and subsequently become more resistant.
standardized nomenclature based on serologic activity in This susceptibility could be related to a reduction of the
crossed immunoelectrophoresis and using an F designa- number of receptors present on intestinal epithelial cells
tion has been proposed (Orskov and Orskov 1983). The with age (Runnels et al. 1980).
latter nomenclature will be used in this chapter. ETEC adhering to the intestinal mucosa produce en-
Although an increasing number of fimbrial adhesins terotoxins which change the water and electrolyte flux of
have been described (more than 30), most fimbrial ad- the small intestine and may lead to diarrhea if the excess
hesins, with the exception of F1 (type 1) common fim- fluid from the small intestine is not absorbed in the large
briae, are associated with E. coli of particular serogroups intestine. Two major classes of enterotoxin are produced
isolated from specific animal species. F1 fimbriae are by porcine ETEC: heat-stable toxin (ST), which is resis-
found on most E. coli isolates and cause an agglutination tant to heat treatment at 100˚C for 15 minutes, and heat-
of guinea pig red cells which is inhibited by D-mannose. labile toxin (LT), which is inactivated at 60˚C for 15 min-
Their role in attachment of porcine ETEC to the intesti- utes (Guerrant et al. 1985). ST has been further divided
nal mucosa is still unclear (Jayappa et al. 1985; To et al. into STa and STb based on solubility in methanol and bi-
1984). ological activity (Burgess et al. 1978). The E. coli entero-
We have found only F1 and no other known fimbrial toxins have been reviewed in detail (Gyles 1994).
adhesins on certain diarrheagenic ETEC strains (Broes et LT is a high-molecular-weight toxin complex that
al. 1988). The four important fimbrial adhesins of consists of five B subunits able to bind to ganglioside re-
neonatal porcine ETEC are F4 (K88), F5 (K99), F6 ceptors on the intestinal epithelial cell surface and a bio-
(987P), and F41. F4 (K88) fimbriae have been divided in- logically active A subunit (Gill et al. 1981). After binding,
to three variants, F4ab, F4ac, and F4ad, based on sero- the latter activates adenylate cyclase, which stimulates
logic cross-reactions (Guinée and Jansen 1979). Many the production of cyclic AMP. High levels of cyclic AMP
ETEC isolates produce more than one fimbrial adhesin, in the cell result in increased secretion of Cl, Na, HCO3,
and common combinations are F5 and F6, F5 and F41, and water into the intestinal lumen. Excessive secretion
and F4 and F6. Production of fimbriae is controlled by will lead to dehydration, metabolic acidosis, and eventu-
genes on the bacterial chromosome (F1, F41) or on plas- ally death. Two subgroups of LT, LTI and LTII, have been
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 435

described (Holmes et al. 1986). Only LTI is neutralized In severe cases, clinical signs of dehydration, metabolic
by anticholera toxin. LT produced by porcine isolates be- acidosis, and death are observed. In certain cases, partic-
longs to the LTI subgroup. The LT produced by human ularly in young animals, the infection may be so rapid
and porcine ETEC has been designated LTh and LTp that death occurs before the development of diarrhea.
based on slight differences in the genes coding for the Neonatal diarrhea may first be observed 2–3 hours
toxin. after birth and may affect single pigs or whole litters. Gilt
STa (STI, ST1, and ST mouse) is a small, nonim- litters are more often affected than sow litters. A large
munogenic protein with a molecular weight (MW) of number of piglets in a farrowing house may be affected
2000 (Lallier et al. 1982). STa binds to a guanylyl cyclase and mortality may be very high in the first few days of
intestinal epithelial receptor (De-Sauvage et al. 1991) life. Diarrhea may be very mild with no evidence of de-
and activates guanylate cyclase, which stimulates pro- hydration or may be clear and watery. The feces vary in
duction of cyclic GMP. High levels of cyclic GMP in the color from clear to whitish or various shades of brown.
cell inhibit the Na/Cl cotransport system and reduce the The fecal material may just dribble from the anus down
absorption of electrolytes and water from the intestine the perineum and only be detected by close examination
(Dreyfus et al. 1984). STa is active in infant mice and of the perineal area. In very severe outbreaks, a small
young piglets of less than 2 weeks of age but is less active proportion of affected animals may vomit. In severe cas-
in older pigs. This could be due to differences in the con- es, 30–40% of total body weight may be lost as fluid into
centration of intestinal receptors with age (Cohen et al. the intestinal lumen and result in signs of dehydration.
1988). As with LT, STa produced by human and porcine The abdominal musculature is flaccid and atonic, the
ETEC has been designated STah and STap, based on dif- pigs are depressed and sluggish, the eyes may be sunken,
ferences in the genes coding for the toxin. and the skin may be bluish-gray in color and resemble
STb (STII, ST2, ST pig) is a small, 5000 MW protein parchment in texture. The loss of fluid and weight re-
that is antigenically and genetically unrelated to STa and sults in the exaggerated appearance of the bony promi-
is poorly immunogenic (Dubreuil 1997). STb stimulates nences. These animals usually die. In more chronic or
cyclic-nucleotide-independent fluid secretion in the gut less severely affected cases, the anus and perineum may
(Kennedy et al. 1984), which is independent of the cyclic be inflamed from contact with the alkaline fecal materi-
nucleotides but appears to be mediated by prostaglandin al. Pigs with less severe dehydration may continue to
E2 and possibly other secretagogues (Harville and Drey- drink and, if treated appropriately, may recover with on-
fus 1995; Peterson and Whipp 1995). STb is inactivated ly minimal long-term effects.
by trypsin and, in the presence of trypsin-inactivator, is Diarrhea in pigs from the neonatal to the postwean-
active in intestines of mice, rats, and calves (Whipp ing period is similar to that observed in neonatal piglets
1990). STb is found in 74% of all porcine ETEC isolates; but tends to be less severe. Morbidity may be the same as
33% of ETEC from older pigs with enteric colibacillosis is in the neonatal period but mortality is invariably lower.
only positive for STb (Moon et al. 1986). The role of STb The feces vary from grayish to whitish in unweaned
in the development of diarrhea is not yet known, al- piglets to brownish in recently weaned piglets. Enteric
though ETEC producing only STb can induce diarrhea in colibacillosis complicated by shock, when associated
experimentally infected newborn pigs (Fairbrother et al. with ETEC strains, occurs both in unweaned pigs from a
1989), and STb induces some villous atrophy in pig in- few days of age and in recently weaned pigs (Faubert and
testinal gut loops (Rose et al. 1987). Drolet 1992), in contrast to earlier reports. Apparently
healthy pigs die suddenly or decline rapidly with
Attaching and Effacing E. coli cyanosis of the extremities. A yellowish to brownish di-
Porcine attaching and effacing E. coli (AEEC) attach to arrhea is sometimes observed.
the intestinal mucosa and cause lesions similar to those
observed for enteropathogenic E. coli (EPEC) isolated LESIONS
from human infantile diarrhea (Hélie et al. 1990). They
attach intimately to the intestinal epithelial cell mem- Few specific pathological changes may be attributed to
brane by means of a bacterial outer-membrane protein enteric E. coli infection. Gross lesions that may be ob-
termed “intimin” or “EPEC attaching and effacing factor” served include dehydration, dilation of the stomach
(Eae), efface the microvilli, and invade the epithelial cells (which may contain undigested milk curd or feed in the
(Zhu et al. 1994). case of postweaning diarrhea), venous infarcts on the
greater curvature of the stomach, and dilation of the
CLINICAL SIGNS small intestine with some congestion of the small-in-
testinal wall. In cases of ETEC infection complicated by
Enteric E. coli infection is usually manifested by diarrhea, shock, characteristic lesions include marked congestion
the severity of which depends on the virulence factors of of the small-intestinal and stomach walls and blood-
the E. coli and the age and immune status of the piglets. tinged intestinal contents.
436 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Histologic lesions depend on the category of E. coli coli arranged in palisades with enterocyte degeneration
involved. In ETEC infections, layers of E. coli are ob- and light to moderate inflammation of the lamina pro-
served adhering to the mucosal epithelial cells of most of pria is observed, mostly in the ileum (Hélie et al. 1990).
the jejunum and ileum in the case of F4-positive ETEC Colonization is most intense in the duodenum and ce-
isolates, and of the posterior jejunum and/or the ileum cum, bacteria are sometimes observed in intracytoplas-
in the case of other ETEC isolates. Adhering bacteria may mic vacuoles in enterocytes, and there is a light to mod-
be found only in the crypts of Lieberkühn, or more often erate villous atrophy in the small intestine. On
covering the crypts and the tips of the villi. On transmis- transmission electronmicroscopy, bacteria are intimate-
sion electronmicroscopy, bacteria are usually located ap- ly attached to the cytoplasmic membrane of mature en-
proximately half a bacterial width away from the mi- terocytes and arranged in regular palisades, parallel to
crovilli, and fimbriae may sometimes be visualized the microvilli, with effacement of adjacent villi (Fig.
between the bacteria and the microvilli (Fig. 32.1). His- 32.2). The bacterial cell wall and the apical cell mem-
tological lesions, if observed, may include vascular con- brane of the enterocyte are separated by a narrow, regu-
gestion in the lamina propria with some hemorrhages in- lar gap of 10 nm at the cupping pedestal, and apical
to the intestinal lumen, increased numbers of dense regions are seen at attachment sites.
neutrophils and macrophages in the lamina propria and
migrating into the lumen, and some villous atrophy. In DIAGNOSIS
cases of ETEC enteric infection complicated by shock, E.
coli are found adhering to the mucosal epithelial cells of Enteric E. coli infection in young, unweaned pigs must be
the small intestine. Congestion, some hemorrhages, and differentiated from other common infectious causes of
in severe cases villous necrosis and microvascular fibri- diarrhea in pigs of this age group. These include Clostrid-
nous thrombi are observed in the lamina propria of the ium perfringens, transmissible gastroenteritis virus, ro-
stomach, small intestine, and colon. tavirus, and coccidia. More than one etiologic agent may
In pigs infected with AEEC isolates, a multifocal colo- be associated with a particular animal or outbreak. A
nization of the brush border of mature enterocytes by E. presumptive diagnosis may be made by determination of

32.1. Electron micrograph of attachment of E. coli 32.2. Electron micrograph of AEEC lesions.
987P positive strain in the intestine.
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 437

the fecal pH. Secretory diarrheic fluid as a result of en- the sensitivity of current gene probe techniques.
teric ETEC infection has an alkaline pH, whereas that However, the traditional approach for identification
from diarrheas associated with malabsorption as a result of pathogenic E. coli by serotyping will still be necessary,
of transmissible gastroenteritis virus or rotavirus infec- at least in reference laboratories, in order to monitor
tion are acid. changing trends and to identify new, emerging E. coli vir-
Diagnosis of enteric E. coli infection is based on clin- ulence determinants which could gain importance due to
ical signs, histopathological lesions, and the presence of the pressure of vaccination of sows against the currently
gram-negative organisms usually closely adhering to the predominant determinants.
small-intestinal mucosa (Wilson and Francis 1986). This
diagnosis is strengthened by the isolation of E. coli of the TREATMENT
appropriate serogroup or, more important, possessing
one or more of the above-mentioned virulence factors. Treatment of enteric E. coli infection should be aimed at
Detection of production of enterotoxins and cytotoxins removal of the pathogenic E. coli, correction of their
has been arduous. Until recently, it has been based on de- harmful effects, and provision of optimal environmental
tection of toxin biological activity. STa activity is deter- conditions. Therapy should be rapidly instituted to be as
mined in the infant mouse test, STb activity in pig and, effective as possible. It is important to confirm the diag-
more recently, in rat ligated gut loops, and LT and VT in nosis of E. coli infection by culture and to perform an-
cell culture assays. Fimbrial adhesins are detected by tibiotic sensitivity tests, because antibiotic sensitivity
serologic assays such as agglutination, immunofluores- varies greatly among E. coli isolates (Table 32.2). A
cence, and ELISA, using rabbit polyclonal antisera. How- broad-spectrum antibiotic treatment could be used ini-
ever, F5 and F41 are only produced when E. coli are tially until the results of antibiotic sensitivity are known.
grown on special minimal media, and F6 is often poorly In vitro resistance of E. coli isolates to a wide range of an-
produced in in vitro conditions. Alternatively, E. coli ad- timicrobial agents has greatly increased over the last sev-
hering to the intestinal mucosa may be demonstrated di- eral years. An alternative approach to the treatment of
rectly in infected pigs by examination of frozen sections enteric E. coli infection is the use of bacteriophages, an
using indirect immunofluorescence or by examination of approach that has been successful experimentally but
formalin-fixed, paraffin-embedded tissues using the im- has not been extensively applied in the field.
munoperoxidase technique. Fluid therapy, consisting of electrolyte replacement
Recent technological advances have greatly improved solutions containing glucose given orally, is useful for the
the detection of E. coli virulence factors (Woodward et al. treatment of dehydration and acidosis. Drugs which in-
1990, 1992; Wray and Woodward 1994). Use of mono- hibit the secretory effects of enterotoxin, such as chlor-
clonal antibodies has led to more specific, sensitive, and promazine and berberine sulfate, may be useful for the
reproducible assays for the detection of STa, F4, F5, F6, treatment of diarrhea, although many of these drugs
and F41. Such antibodies could be used in diagnostic kits have undesirable side effects. The use of such antisecre-
for the rapid detection and identification of pathogenic tory drugs as bencetimide and loperamide, alone or in
E. coli directly in the feces or intestinal contents of af- combination with antibacterial agents, has also been
fected piglets. Probes and the polymerase chain reaction suggested (Solis et al. 1993).
(PCR) have now been developed for the detection of the It is important to ensure that younger piglets are
genes coding for the fimbrial adhesins and enterotoxins maintained at a constant temperature of 30–34˚C and
of swine ETEC (Nagy et al. 1990; Woodward et al. 1990,
1992; Wray and Woodward 1994). There is a high corre-
lation between the results of the standard serologic and Table 32.2. Sensitivity to antimicrobial agents of E.
biological assays and those of gene probes for the detec- coli isolates from pigs aged from 0 to 7 days with
tion of fimbrial adhesins and enterotoxins of swine diarrhea in Quebec, 1994–96
ETEC (Harel et al. 1991). Probes are currently used in Antimicrobial Agent % Sensitive Isolates (n = 38)
many diagnostic laboratories for the identification of
Amikacin 100
ETEC isolates and for the detection of ETEC directly in Ampicillin 46
the feces of pigs with diarrhea (Harel et al. 1991; Monck- Apramycin 79
ton and Hasse 1988; Ojeniyi et al. 1994; Woodward et al. Ceftiofur 92
1990). Gene probe techniques often involve the use of ra- Cephalothin 70
Enrofloxacin 100
dioactivity and thus must be performed in controlled
Gentamycin 63
laboratory conditions. Nonradioactive gene probes are Neomycin 56
being developed and could feasibly be used in kits for the Spectinomycin 41
detection of pathogenic E. coli directly in the fecal or in- Tetracycline 7
testinal contents of diarrheic pigs by the veterinary prac- Tiamulin 5
Trimethoprim-sulfamethoxazole 71
titioner. Use of the PCR technique will greatly enhance
438 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

that recently weaned pigs are held in a draft-free envi- Immunity


ronment at a constant temperature of about 29.5˚C. Immunity to enteric E. coli infections is humoral and is
initially provided through the maternal colostrum, lacto-
PREVENTION genic antibodies in the milk of the sow, and subsequent-
ly by a local intestinal immune response. Specific anti-
A program for prevention of enteric E. coli infection bodies inhibit bacterial adherence to receptors on the
should be aimed at reduction of numbers of pathogenic intestinal epithelial cells and neutralize the activity of
E. coli in the environment by good hygiene, maintenance the enterotoxins or cytotoxins produced by E. coli.
of suitable environmental conditions, and provision of a Colostrum from the sow contains high levels of im-
high level of immunity. Because most pathogenic E. coli munoglobulin G (IgG), which rapidly decrease during
belong to a limited number of serogroups, enteric E. coli lactation and IgA becomes the main immunoglobulin
infection could be eliminated from some herds. class (Bourne 1976). The latter protects the gut against E.
coli infection. It appears that most IgA, IgM, and IgG in
Husbandry the milk of the sow is produced within the mammary
One of the most important factors in prevention of en- gland. During pregnancy, a proportion of the lympho-
teric E. coli infection is the maintenance of piglets at an cytes stimulated by antigens in the intestine migrate to
adequate environmental temperature (30–34˚C for un- the mammary gland and produce specific antibody
weaned pigs), free of drafts, and on a low-heat-conduct- against enteric pathogens. These antibodies are actively
ing floor. This is particularly true for piglets of below-av- transported to the colostrum and then the milk during
erage weight, who lose heat more rapidly because they lactation.
have a greater skin surface area per unit body weight. The newborn piglet begins to synthesize specific im-
Good hygiene in the farrowing area leads to a reduc- munoglobulin and develop intestinal immunity during
tion in the numbers of E. coli being presented to the the first week of life (Butler 1973). At first, IgM predom-
piglet to a level that it is able to control through its own inates, but after 2–3 weeks, it is replaced by IgA as the
defense mechanisms. most important immunoglobulin class in the intestine.
Farrowing-crate design is important because it affects Thus, during the first weeks of life, colostrum is the main
the position at which feces are deposited by the sow. In source of immunologic protection for the piglet.
crates that are too long, the feces are deposited over a Breakdown in the protection provided by colostrum
large area of the available floor space, thereby increasing may occur for several reasons. If the dam has not been
the heavily contaminated area. Ideally, the crate should exposed to ETEC present in the environment of the
be adjustable, allowing for a shorter crate for gilts than piglets, her colostrum will not contain the specific anti-
for sows. Crates on raised, perforated floors allow fecal bodies necessary for protection against adherence and
material to drop through and away from the piglets, and proliferation of ETEC. Also, any disease process causing
litters farrowed onto such floors have a noticeably lower agalactia in the sow will diminish transfer of colostrum.
incidence of diarrhea than those on solid concrete floors. Generalized systemic infection may cause a total reduc-
A dry, warm environment reduces the moisture avail- tion in colostrum production, whereas mastitis or in-
able to enhance the survival of E. coli. This is largely af- jured teats may affect production in one or several
fected by ventilation rates, although if room temperature glands. Piglets failing to receive colostrum due to defor-
is too high, sows tend to try and spread water over their mities, infection, small size, or damage at birth will also
lying area to cool themselves, thereby defeating other hy- be more susceptible to ETEC infection.
gienic procedures. The sow should be at a temperature of Maternal vaccination has been one of the most effec-
approximately 22˚C, necessitating a warmer creep area tive ways of controlling neonatal ETEC diarrhea in
for the piglets. piglets. Identification of virulence factors important in
Quarantine should be used to control the introduc- the pathogenesis of ETEC diarrhea and application of re-
tion of different E. coli pathotypes or other infectious combinant DNA technology have resulted in the produc-
agents into the herd. Animals in the herd will have little tion of more efficient vaccines over the last several years.
immunity to E. coli fimbrial antigens with which they One of the earliest vaccination techniques consisted of
have not had contact. Farrowing crates should be thor- taking the small-intestinal contents from a piglet with
oughly cleaned and disinfected between litters. An all- diarrhea, culturing it in milk, and feeding the culture to
in/all-out farrowing system with thorough disinfection pregnant sows, usually about a month before parturition
of the farrowing room between batches will reduce the E. (Kohler 1974). This technique is effective, conferring an
coli population in the environment. immunity lasting throughout the suckling period, and is
Diet may be modified in order to reduce colonization still used, particularly in the United States.
of the intestine by E. coli (Thomlinson and Lawrence Commonly used commercially available vaccines are
1981). Feeding of cultures of Streptococcus faecium to given parenterally and may be killed whole-cell bacterins
young pigs may reduce the incidence of diarrhea or purified fimbrial vaccines. Both of the latter types of
(Morkoc et al. 1984). vaccines appear to work equally well (Fahy 1987). Bac-
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 439

terins usually contain strains representing the most im- methanol-soluble, heat-stable Escherichia coli STb en-
portant serogroups and producing the fimbrial antigens terotoxin in infant mice, pigs, rabbits, and calves. Infect
F4, F5, F6, and F41 (Nagy 1986). They are usually given Immun 21:526–531.
parenterally at about 6 weeks and 2 weeks prior to par- Butler, J. E. 1973. Synthesis and distribution of im-
turition. Addition of the common fimbrial antigen F1 to munoglobulins. J Am Vet Med Assoc 163:795–800.
a fimbrial bacterin appeared to have been protective in Clarke, S.; Cahill, A.; Stirzaker, C.; Greenwood, P.; and Greg-
one study (Jayappa et al. 1985) but was not protective in son, R. 1985. Prevention by vaccination-animal bacteria.
another study (To et al. 1984). Recombinant DNA tech- In Infectious Diarrhea in the Young. Amsterdam: Elsevi-
nology has enabled the production of large quantities of er Science Publishers, p. 481.
purified fimbrial antigens for use in parenteral vaccines Cohen, M. B.; Guarino, A.; Shukla, R.; and Giannella, R. A.
for immunization of the dam (Clarke et al. 1985). 1988. Age-related differences in receptors for Escherichia
In cases where vaccination is ineffective, it is impor- coli heat-stable enterotoxin in the small and large intes-
tant to identify the serotypes involved for possible inclu- tine of children. Gastroenterology 94:367–373.
sion into an autogenous bacterin. Further characteriza- De-Sauvage, F. J.; Camerato, T. R.; and Goeddel, D. V. 1991.
tion of these isolates may identify new or variant Primary structure and functional expression of receptor
fimbrial adhesins important in the pathogenesis of for Escherichia coli heat-stable enterotoxin. J Biol Chem
ETEC diarrhea. An alternative approach to the problem 266:17912–17918.
of emerging ETEC negative for the known fimbriae has Dreyfus, L. A.; Jaso-Friedmann, L.; and Robertson, D. C.
been the use of vaccines containing the nontoxic form of 1984. Characterization of the mechanism of action of
the enterotoxin LT-B conjugated to the nonimmunogenic Escherichia coli heat stable enterotoxin. Infect Immun
STa (Klipstein 1986). Following conjugation, STa be- 44:493–501.
comes immunogenic, and this vaccine has given protec- Dubreuil, J. D. 1997. Escherichia coli STb enterotoxin: Re-
tion in experimental animals. Addition of these compo- view. Microbiology (in press).
nents to fimbrial vaccines will provide protection against Edfors-Lilja, I.; Gustafsson, U.; Duval-Iflah, Y.; Ellergren, H.;
emerging ETEC with new fimbrial antigens in neonates Johansson, M.; Juneja, R. K.; Marklund, L.; and Anders-
and against ETEC negative for the known fimbrial anti- son, L. 1995. The porcine intestinal receptor for Es-
gens and commonly found in older pigs. Isaacson (1994) cherichia coli K88ab, K88ac: Regional localization on
has recently reviewed the use of vaccines for the preven- chromosome 13 and influence of IgG response to the
tion of E. coli diseases. K88 antigen. Anim Genetics 26:237–242.
Finally, a novel approach to the prevention of ETEC Fahy, V. A. 1987. Preweaning colibacillosis. In Proc Conf
diarrhea in piglets could be the oral administration of ex- Australasian Pig Sci Assoc Albury, p. 177.
ogenous proteases such as bromelain (Mynott et al. Fairbrother, J. M.; Broes, A.; Jacques, M.; and Larivière, S.
1996). Such proteases can inhibit attachment of F4-pos- 1989. Pathogenicity of Escherichia coli O115:K“V165”
itive ETEC to the intestinal mucosa due to modification strains isolated from pigs with diarrhea. Am J Vet Res
of the receptor attachment sites. 50:1029–1036.
Faubert, C., and Drolet, R. 1992. Escherichia coli hemorrhag-
REFERENCES ic gastroenteritis in the piglet: Clinical, pathological and
microbiological findings. Can Vet J 33:251.
Alexander, T. J. L. 1994. Neonatal diarrhea in pigs. In Es- Gill, D. M.; Clements, J. D.; Robertson, D. C.; and Finkel-
cherichia coli in Domestic Animals and Humans. Ed. C. stein, R. A. 1981. Subunit number and arrangement in
L. Gyles. Wallingford, U.K.: CAB International, pp. Escherichia coli heat-labile enterotoxin. Infect Immun
151–170. 33:677–682.
Biehl, L. G., and Hoefling, D. C. 1986. Diagnosis and treat- Guerrant, R. L.; Holmes, R. K.; Robertson, C. C.; and Green-
ment of diarrhea in 7- to 14-day-old pigs. J Am Vet Assoc berg, R. N. 1985. Roles of enterotoxins in the pathogen-
188:1144–1146. esis of Escherichia coli diarrhea. In Microbiology—1985.
Bijlsma, I. G. W.; De Nijs, A.; Van Der Meer, C.; and Frik, J. Ed. L. Leive, P. F. Bonventre, J. A. Morello, S.
F. 1982. Different pig phenotypes affect adherence of Es- Schlesinger, S. D. Silver, and H. C. Wu. Washington,
cherichia coli to jejunal brush borders by K88ab, K88ac, D.C.: American Society for Microbiology, pp. 68–73.
or K88ad antigen. Infect Immun 37:891–894. Guinée, P. A. M., and Jansen, W. H. 1979. Behavior of Es-
Bourne, F. J. 1976. Humoral immunity in the pig. Vet Rec cherichia coli K antigens K88ab, K88ac, and K88ad in
98:499–501. immuno-electrophoresis, double diffusion, and hemag-
Broes, A.; Fairbrother, J. M.; Larivière, S.; Jacques, M.; and glutination. Infect Immun 23:700–705.
Johnson, W. M. 1988. Virulence properties of entero- Guinée, P. A. M.; Veldkamp, J.; and Jansen, J. 1977. Im-
toxigenic Escherichia coli O8:KX105 strains isolated from proved minca medium for the detection of K99 antigen
diarrheic piglets. Infect Immun 56:241–246. in calf enterotoxigenic strains of Escherichia coli. Infect
Burgess, M. N.; Bywater, R. J.; Cowley, C. M.; Mullan, N. A.; Immun 15:676–678.
and Newsome, P. M. 1978. Biological evaluation of a Gyles, C. L. 1994. Escherichia coli enterotoxins. In Escherichia
440 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

coli in Domestic Animals and Humans. Ed. C. L. Gyles. Morkoc, A.; Backstrom, L.; and Savage, D. 1984. Streptococ-
Wallingford, U.K.: CAB International, pp. 337–363. cus faecium in prevention of neonatal colibacillosis in
Harel, J.; Lapointe, H.; Fallara, A.; Lortie, L. A.; Bigras- piglets. Proc Int Congr Pig Vet Soc 8:76.
Poulin, M.; Larivière, S.; and Fairbrother, J. M. 1991. De- Mynott, T. L.; Luke, R. K.; and Chandler, D. S. 1996. Oral ad-
tection of genes for fimbrial antigens and enterotoxins ministration of protease inhibits enterotoxigenic Es-
associated with Escherichia coli serogroups from pigs cherichia coli receptor activity in piglet small intestine.
with diarrhea. J Clin Microbiol 29:745–752. Gut 38:28–32.
Harville, B. A., and Dreyfus, L. A. 1995. Involvement of 5- Nagy, B. 1986. Vaccines against toxigenic Escherichia coli dis-
hydroxytryptamine and prostaglandin E2 in intestinal ease in animals. In Development of Drugs and Vaccines
secretory action of Escherichia coli heat-stable enterotox- against Diarrhea. 11th Nobel Conf Stockholm 1985.
in B. Infect Immun 63:745–750. Lund: Stdentlitteratur, p. 53.
Hélie, P.; Morin, M.; Jacques, M.; and Fairbrother, J. M. Nagy, B.; Casey, T. A.; and Moon, H. W. 1990. Phenotype
1990. Experimental infection of newborn pigs with an and genotype of Escherichia coli isolated from pigs with
attaching and effacing Escherichia coli 045: K“E65” post-weaning diarrhea in Hungary. J Clin Microbiol
strain. Proc Int Congr Pig Vet Soc. Lausanne. 28:651–653.
Holmes, R. K.; Twiddy, E. M.; and Pickett, C. L. 1986. Pu- Ojeniyi, B.; Ahrens, P.; and Meyling, A. 1994. Detection of
rification and characterization of type II heat-labile en- fimbrial and toxin genes in Escherichia coli and their
terotoxin of Escherichia coli. Infect Immun 53:464–473. prevalence in piglets with diarrhoea: The application of
Hu, Z. L., Hasler-Rapacz, J.; Huang, S. C.; and Rapacz, J. colony hybridization assay, polymerase chain reaction
1993. Studies in swine on inheritance and variation in and phenotypic assays. J Vet Med B 41:49–59.
expression of small intestinal receptors mediating adhe- Orskov, I., and Orskov, F. 1983. Serology of Escherichia coli
sion of the K88 enteropathogenic Escherichia coli vari- fimbriae. Prog Allergy 33:80–105.
ants. J Heredity 84:157–165. Peterson, J. W., and Whipp, S. C. 1995. Comparison of the
Isaacson, R. E. 1994. Vaccines against Escherichia coli dis- mechanisms of action of cholera toxin and the heat-sta-
eases. In Escherichia coli in Domestic Animals and Hu- ble enterotoxins of Escherichia coli. Infect Immun
mans. Ed. C. L. Gyles. Wallingford, U.K.: CAB Interna- 63:1452–1461.
tional, pp. 629–647. Rose, R.; Whipp, S. C.; and Moon, H. W. 1987. Effects of
Jayappa, H. G.; Goodnow, R. A.; and Geary, S. J. 1985. Role Escherichia coli heat stable enterotoxin on small intesti-
of Escherichia coli type 1 pilus in colonization of porcine nal villi in pigs, rabbits, and lambs. Vet Pathol 24:
ileum and its protective nature as a vaccine antigen in 71–79.
controlling colibacillosis. Infect Immun 48:350–354. Runnels, P. L.; Moon, H. W.; and Schneider, R. A. 1980. De-
Kennedy, D. J.; Greenberg, R. N.; Dunn, J. A.; Abernathy, R.; velopment of resistance with host age to adhesion of
Ryerse, J. S.; and Guerrant, R.L. 1984. Effects of Es- K99+ Escherichia coli to isolated intestinal epithelial
cherichia coli heat stable enterotoxin STb on intestines of cells. Infect Immun 28:298–300.
mice, rabbits, and piglets. Infect Immun 46:639–643. Sarmiento, J. I. 1983. Environmental temperature: A predis-
Klipstein, F. A. 1986. Development of Escherichia coli vac- posing factor in the enterotoxigenic Escherichia coli–in-
cines against diarrheal disease in humans. In Develop- duced diarrhea of the newborn pig. M.S. thesis, Univ
ment of Drugs and Vaccines against Diarrhea. 11th No- Guelph, Ont.
bel Conf Stockholm 1985. Lund: Stdentlitteratur, p. 53. Sellwood, R.; Gibbons, R. A.; Jone, G. W.; and Rutter, J. M.
Kohler, E. M. 1974. Protection of pigs against neonatal en- 1975. Adhesion of enteropathogenic Escherichia coli to
teric colibacillosis with colostrum and milk from orally pig intestinal brush borders: The existence of two pig
infected sows. Am Vet J 35:331–338. phenotypes. J Med Microbiol 8:405–411.
Lallier, R.; Bernard, F.; Gendreau, M.; Lazure, C.; Seidah, N. Soderlind, O.; Thafvelin, B.; and Mollby, R. 1988. Virulence
G.; Chretien, M.; and St-Pierre, S. 1982. Isolation, pu- factors in Escherichia coli strains isolated from Swedish
rification, and structure of Escherichia coli heat stable pigs with diarrhea. J Clin Microbiol 26:879–884.
enterotoxin of porcine origin. Anal Biochem Solis, C. A.; Sumano, L. H.; and Marin, H. J. A. 1993. Treat-
127:267–275. ment of Escherichia coli induced diarrhoea in piglets
Monckton, R. P., and Hasse, D. 1988. Detection of entero- with antisecretory drugs alone or combined with an-
toxigenic Escherichia coli in piggeries in Victoria by DNA tibacterials. Pig Vet J 30:83–88.
hybridization using K88, K99, ST1, and ST2 probes. Vet Thomlinson, J. R., and Lawrence, T. J. L. 1981. Dietary ma-
Microbiol 16:273–281. nipulation of gastric pH in the prophylaxis of enteric
Moon, H. W.; Schneider, R. A.; and Mosely, S. L. 1986. Com- disease in weaned pigs: Some field observations. Vet Rec
parative prevalence of four enterotoxin genes among Es- 109:120–122.
cherichia coli isolated from swine. Am J Vet Res To, S. C.-M.; Moon, H. W.; and Runnels, P. L. 1984. Type 1
47:210–212. pili (F1) of porcine enterotoxigenic Escherichia coli: Vac-
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 441

cine trial and tests for production in the small intestine Woodward, M. J.; Carroll, P. J.; and Wray, C. 1992. Detec-
during disease. Infect Immun 43:1–5. tion of entero- and verocytotoxin genes in Escherichia
Whipp, S. C. 1990. Assay of enterotoxigenic Escherichia coli coli from diarrhoeal disease in animals using the poly-
heat-stable toxin b in rats and mice. Infect Immun merase chain reaction. Vet Microbiol 31:251–261.
58:930–934. Wray, C., and Woodward, M. J. 1994. Laboratory diagnosis
Wilson R. A., and Francis, D. H. 1986. Fimbriae and entero- of Escherichia coli infections. In Escherichia coli in Do-
toxins associated with Escherichia coli serogroups isolat- mestic Animals and Humans. Ed. C. L. Gyles. Walling-
ed from pigs with colibacillosis. Am J Vet Res ford, U.K.: CAB International, pp. 595–628.
47:213–217. Zhu, C.; Harel, J.; Jacques, M.; Desautels, C.; Donnenberg,
Woodward, M. J.; Kearsley, R.; Wray, C.; and Roeder, P. L. M. S.; Beaudry, M.; and Fairbrother, J. M. 1994. Viru-
1990. DNA probes for detection of toxin genes in Es- lence properties and attaching-effacing activity of Es-
cherichia coli isolated from diarrhoeal disease in cattle cherichia coli O45 from swine postweaning diarrhea. In-
and pigs. Vet Microbiol 22:277–290. fect Immun 62:4153–4159.

POSTWEANING ESCHERICHIA COLI DIARRHEA AND


EDEMA DISEASE
H. U. Bertschinger
Postweaning Escherichia coli diarrhea and edema disease about 20 kg. About 75% of the losses were associated
are treated in one section because they often both occur with PWECD. The study included nearly 49,000 litters.
in the same batch of pigs, and the causative bacteria In affected herds, losses in some batches of weaners may
share certain virulence factors. Treatment and preven- be as high as 50%. However, mortality due to ED is not as
tion are mainly the same for both diseases. great in North America.
Postweaning E. coli diarrhea (PWECD) is a communi- ED bears substantial similarities to the human dis-
cable diarrhea mediated by enterotoxins and observed eases caused by enterohemorrhagic strains of E. coli
mainly after weaning. It is also called postweaning en- (EHEC), which produce closely related, but not identical,
teric colibacillosis. Shiga-like toxins. However, the human EHEC strains col-
Edema disease (ED) is a communicable enterotox- onize the porcine digestive tract by a mechanism distinct
emia caused by certain E. coli colonizing the small intes- from enterotoxemic E. coli (ETEEC) (Tzipori et al. 1986).
tine and producing an exotoxin that gets to the blood Serotypes associated with ED are different from those of
and damages vessel walls. The names “edema disease,” EHEC.
“bowel edema,” and “gut edema” were coined because
edema of the submucosa of the stomach and the meso- ETIOLOGY
colon is often a prominent feature of the disease.
A clear-cut distinction between PWECD and ED is PWECD and ED are caused by strains of E. coli that pos-
not possible, because certain strains of E. coli are in- sess certain adhesion factors enabling the bacteria to col-
volved in both conditions. The early history of the two onize the small intestine and that produce one or several
diseases has been extensively reviewed by Sojka (1965). exotoxins. Nearly all of these E. coli are alpha-hemolytic.
E. coli is an important cause of death in weaned pigs. Most of them belong to a very limited number of
In Switzerland, PWECD or ED was diagnosed in 57% of serotypes. In a given area, the serotypes are closely asso-
3301 4- to 12-week-old pigs submitted for postmortem ciated with a rather constant set of adhesion factors and
examination. PWECD had a prevalence of 35% and ED of toxins. Thus, the serogroup O139 has been found world-
22% (Häni et al. 1976). Wegmann (1990) reported an es- wide to bear the fimbrial variant F18ab. Strains of this
sentially unchanged situation. A peak of postweaning serogroup from Australia always cause PWECD, whereas
deaths caused by E. coli was seen in 5- to 10-week-old those from Europe typically induce ED.
pigs. In the former East Germany, with 750,000 produc- The most comprehensive study on the prevalence of
tive sows, Jahn and Uecker (1987) reported a mortality in virulence factors with E. coli isolates from weaner pigs
weaned pigs of 1.8% due to E. coli. Direct and indirect an- with diarrhea was performed in Denmark (Ojeniyi et al.
nual losses were estimated at about 50 million East Ger- 1994). The fimbrial types F18 (earlier called F107, 2134P,
man marks. Svensmark et al. (1989) reported an overall 8813, and Av24) (Rippinger et al. 1995) and F4 (K88)
mortality of 2.4% in the period between weaning at 2–4 were detected with 44% and 36%, respectively, of the tox-
weeks and transfer to the fattening unit at a weight of igenic isolates (Table 32.3). In about 24% of the latter,
442 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Table 32.3. Fimbrial types and toxins detected in 184 E. coli isolates from 28- to 49-day-old pigs with diarrhea
Toxin(s)
SLT
LT SLT SLT SLT STa
Fimbrial Number LT STa STa SLT SLT STb STa STa STb Toxin
Group of Strains STb LT STb STb STb SLT STb LT LT LT STb LT Negative
F18 75 11 2 16 15 3 2 1 1 6 8 1 9
F18 + F6 4 4
F18 + F41 1 1
F18 + F4 5 5
F4 52 1 6 39 1 1 1 2 1
F4 + F6 1 1
F6 2 1 1
None 44 10 4 6 1 20 1 2
Total 184 22 12 62 18 9 23 2 1 13 1 10 2 9
Source: Modified from Ojeniyi et al. 1994.

genes coding for known fimbrial types were not detected. In a given pig population, strains with a certain anti-
More than one fimbrial type was found in about 6%. F6 genic makeup are often equipped with certain patterns
(987P) was most often present in association with either of virulence factors (Table 32.4). Identical serotypes
F4 or F18. In Canada, 11% of the toxigenic isolates were were detected in all continents, but their frequencies can
positive for F18, 9% for F4, 5% for F5, and none for F6 vary in different geographic regions.
(Fairbrother et al. 1994). The antigenic variants of F18
fimbriae were serologically determined using 380 iso- Enteropathogenic E. coli
lates obtained from fatal cases of PWECD and ED in Ger- An enteric syndrome distinct from classic PWECD was
many (Wittig et al. 1995). Variants F18ab and F18ac described by several investigators. It is characterized by
were found in 40% and 35%, and F4 in 14%, of the iso- attaching and effacing lesions caused by E. coli (AEEC).
lates. The remaining isolates were negative for F18 and The spontaneous disease is nonfatal. The AEEC do not
F4. possess any virulence factors of classic PWECD or ED
PWECD and ED are mediated by toxins. E. coli iso- strains (Zhu et al. 1994). Their virulence attributes are
lates from fatal cases are nearly always toxigenic (Im- dealt with in the subchapter on neonatal E. coli diarrhea.
berechts et al. 1994; Wittig et al. 1995), whereas fecal Experimental infection of gnotobiotic pigs allows repro-
samples from weaners with diarrhea less often give rise duction of the lesions (Fig. 32.2). A weaner diet contain-
to growth of toxigenic E. coli (Fairbrother et al. 1994; ing soya and field peas enhances bacterial colonization
Ojeniyi et al. 1994). This points to the multiple etiology and development of attaching and effacing lesions but
of postweaning diarrhea (see Chapter 57). It is notewor- causes no diarrhea (Neef et al. 1994). AEEC will not be
thy that strains producing SLT-IIe occur in pig popula- further dealt with in this chapter.
tions in the absence of typical ED. This was, for example, A comprehensive overview of E. coli and its role in
the case in Denmark (Table 32.3) before the recent inva- human and animal disease appeared in 1994 (Gyles
sion of strains of serogroup O139 (Aarestrup et al. 1994).
1997).
A more detailed description of the enterotoxins is EPIDEMIOLOGY
given in the preceding subchapter. The term “Shiga-like
toxin” (SLT-IIe) is a synonym of verotoxin, edema dis- The epidemiologies of PWECD and ED have many fea-
ease principle, neurotoxin, and vasotoxin. MacLeod and tures in common. The age group primarily affected by
Gyles (1990) developed a purification scheme resulting
in a homogeneous preparation of SLT-IIe free of endo-
Table 32.4. Examples of prevailing pathotypes of
toxin. As little as 3 ng of pure SLT-IIe per kilogram of E. coli in fatal cases of PWECD and ED in Switzerland
body weight administered intravenously to young pigs
Serotype Colonization Factor Toxin(s)
induces disease. Clinical signs and gross and microscop-
ic lesions are characteristic of ED, thus confirming that O139:K12:H1 F18ab SLT-IIe
SLT-IIe and EDP (edema disease principle) are identical. O141ab:H4 F18ac SLT-IIe, LT, STII
O149:K91:H10 F4ac LT, STII
Incubation time and severity of disease are directly relat-
ed to the toxin dose. Source: Modified from Imberechts et al. 1994.
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 443

PWECD and/or ED depends on the weaning age. There PATHOGENESIS


are some differences between E. coli with F4 and with
F18. The former can cause neonatal, preweaning, and For the sake of clarity, intestinal colonization and tox-
postweaning diarrhea, most often in the very first days emia will be presented separately. However, there may be
after weaning. The latter, however, more often cause dis- mutual interactions.
ease between 5 and 14 days after weaning (Svendsen et
al. 1974) or after introduction to fattening herds. Un- Colonization of the Small Intestine
weaned piglets can also be affected by PWECD and ED. An E. coli strain of O group 141 causing PWECD and ED
In suckling pigs, the severity of the disease depends on (Smith and Halls 1968) and two strains of O group 139
antibody titers in the milk of the sow (Sarmiento et al. devoid of enterotoxins (Bertschinger and Pohlenz 1983;
1988b). In adult pigs, ED is the more frequent cause of Bertschinger et al. 1990) reached high population densi-
central nervous system signs and a significant cause of ties throughout the intestinal tract, including the upper
mortality (Sydler et al. 1996). small intestine. Such colonization requires both mucosal
The morbidity in an affected herd is extremely vari- adhesion and proliferation. The degree of colonization
able. Within a particular litter it may be up to 80% or determines whether or not disease will result from infec-
more, but the average is 30–40%. With ED, the case fatal- tion. Adhering microcolonies or layers of bacteria were
ity rate ranges from 50% to over 90%. The course of the observed on the small-intestinal mucosae of pigs experi-
disease in the herd varies from 4 to 14 days, the average mentally infected with two strains of O group 139 (Fig.
being slightly under a week. The disease disappears as 32.3) (Bertschinger and Pohlenz 1983; Methiyapun et al.
abruptly as it appears. Recurrence on premises is com- 1984; Bertschinger et al. 1990). ETEEC adhere to the
mon (Kurtz et al. 1969). With PWECD the case fatality brush border similarly to enterotoxigenic E. coli (ETEC),
rate and the mortality tend to be lower. In untreated whereas other producers of SLTs show the “attaching
herds, the latter may attain 26% (Svendsen et al. 1974). and effacing” phenomenon. Since colonization depends
The environment of the weaner unit appears to be on a multiplicity of factors, it is not reliably repro-
the most likely source of pathogenic E. coli strains. Un- ducible. Some of the known factors are discussed here.
weaned pigs may acquire infection in the farrowing Brush border receptors for pathogenic E. coli are not
house, presumably from the same source, and carry it in- present in every pig. Genetic resistance resulting from
to the weaning unit. Routine cleaning and disinfection lack of receptors for F4 was first described by Sellwood
are usually insufficient to break the cycle of infection et al. (1975) (see section on neonatal colibacillosis). The
(Hampson et al. 1987). Under experimental conditions, receptor for F18 fimbriae is also controlled in a single lo-
however, transmission can be prevented by strict hygien- cus, and the presence of a receptor is dominant over ab-
ic measures (Smith and Halls 1968; Kausche et al. 1992). sence (Bertschinger et al. 1993). The receptor for F18 is
The minimal infectious dose is not known. In transmis- distinct from the receptor for F4. The genes for F4 were
sion experiments with a K88-positive ETEC strain, air- determined on chromosome 13 (Guérin et al. 1993),
borne transmission between pigs in wire cages 1.5 m whereas the genes for the F18 receptor were located on
apart was repeatedly observed (Wathes et al. 1989). Out- chromosome 6 close to the locus for stress susceptibility.
breaks tend to involve only one strain of E. coli at any one In a high proportion of the Swiss pig population, resis-
time. Occasionally, two potential pathogens are isolated, tance against stress is linked to susceptibility to adhesion
but one usually predominates. Multiple infections of of E. coli with F18 fimbriae (Vögeli et al. 1996). In view of
herds involving more than one serogroup were detected the low prevalence of stress-susceptible pigs, the fre-
in 47% of 84 herds (Awad-Masalmeh et al. 1988). quency of pigs with the F18 receptor must be high.
The spread of pathogenic E. coli is presumed to occur Fimbrial receptors are thought to be glycoconjugates.
via aerosols, feed, other vehicles, pigs, and possibly oth- The latter are subject to modulation by feed lectins such
er animals. Introduction of new pathogenic strains of E. as constituents of leguminous plants (Kelly et al. 1994).
coli into closed primary specific-pathogen-free (SPF) It may be speculated that feed-induced changes of the re-
herds with a high isolation standard was observed at in- ceptor are involved in the reduced susceptibility to colo-
tervals of 1–2 years. Once a site is contaminated with a nization by F18-positive E. coli in the very first days after
particular strain, it can remain so for an extended period. weaning (Bertschinger et al. 1993). Endogenous as well
The serotypes associated with postweaning diseases tend as orally administered proteases may reduce the receptor
to be similar in broad geographic areas. When ED en- activity for F4 fimbriae (Mynott et al. 1996). Receptors
tered Denmark in 1994, it spread from one SPF breeding for F4 are fully expressed from birth to adult age, where-
herd by trade of pigs to at least 37 other herds. The close as the F18 receptor is not yet fully expressed by piglets
clonal relationship of the causal strains was confirmed under about 20 days of age (Nagy et al. 1992). This ex-
(Aarestrup et al. 1997; Jorsal et al. 1996). Another 22 plains why E. coli with F18 fimbriae do not cause disease
herds became infected without known trading contact. in neonatal pigs. Constant expression of receptors may
444 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

32.3. Bacteria adhering patch-


wise to microvilli of small-intestinal
epithelium of a weaner 6 days after
oral inoculation with a culture of
E. coli O139:K12:H1.

underlie the earlier appearance after weaning of E. coli Bertschinger et al. (1978) using an ETEEC strain of
strains with F4 in herds where F4- and F18-positive serogroup O139:K12 for inoculation. The findings of
strains are endemic. Smith and Halls (1968) were confirmed. However, the
Sarmiento et al. (1988b) detected specific antibodies poor diets inhibited growth of the pigs. When these diets
in milk and suggested that milk antibody protects the were replaced by a conventional type of feed, coloniza-
piglets against colonization as long as they are nursing. tion and clinical disease developed. The inhibitory effect
A variety of viruses infect the porcine intestine and of the poor diet was abolished by supplementation with
may thereby change the bacterial environment. In an ex- fish meal but not with starch or fat. The precise mecha-
perimental postweaning diarrhea model, an F4-positive nism behind these phenomena remains to be elucidated.
ETEC strain colonizes weaned pigs in the absence of ro- Low room temperature in the weaner rooms was pro-
taviral infection (Sarmiento et al. 1988a). Dual infection posed as responsible for a more severe course of post-
of pigs with rotavirus and with an ETEC strain without weaning diarrhea (Wathes et al. 1989). Under experi-
F4 results in a more severe diarrhea than inoculation mental conditions, ED was not aggravated by cold stress
with either agent alone (Lecce et al. 1982). The investiga- (Kausche et al. 1992).
tors concluded that viral damage of the epithelium fa-
vors colonization by E. coli. Diarrhea
An acid environment has an inhibitory effect on E. The mechanisms by which enterotoxins induce diarrhea
coli. The pH of the stomach contents falls after weaning are described in the preceding section (neonatal E. coli
(Risley et al. 1992). Several investigators found that the diarrhea). Almost all E. coli involved in fatal cases of
pH of the jejunum cannot be reduced by acidification of PWECD produce LT (Imberechts et al. 1994). Pigs colo-
the feed. The pH close to the jejunal brush border is nized about 12 days after weaning with ETEC producing
slightly acid and highly regulated. It is not influenced by one or both heat-stable enterotoxins develop diarrhea
the pH of the chyme (McEwan et al. 1990). only exceptionally (Sarrazin and Bertschinger 1997).
Veterinary practitioners and farmers were convinced This difference between neonates and weaned pigs may
years ago that nutritious feed would play an important be explained at least in part by the marked increase of
role. Thus ED was named “protein intoxication.” Smith antisecretory factor beginning a few days after weaning
and Halls (1968) inoculated pigs on various feed regi- (Lange et al. 1993).
mens with an ETEEC strain of serogroup O141:K85a,c.
They found that severe feed restriction resulted in much Enterotoxemia
lower fecal numbers of the bacteria and absence of dis- Highly purified SLT-IIe induces a disease indistinguish-
ease. A similar effect was achieved by feeding pigs ad li- able from ED when administered intravenously to pigs
bitum on a diet extremely rich in fiber and low in nutri- (MacLeod and Gyles 1990). Transportation of the toxin
ents. The authors concluded that the physiological state into the circulation is still somewhat of a mystery. SLT-
of the intestinal epithelium might influence bacterial IIe binds preferentially to globotetraosyl ceramide and
adhesion. These experiments were extended by therefore to the red blood cells. Thus, vessels are subject-
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 445

ed to prolonged toxin exposure (Boyd et al. 1993). By im- ithelial necrosis secondary to necrosis of small arteries
munological methods the toxin can be detected in en- and arterioles may be responsible for luminal hemor-
dothelial cells of small blood vessels of the intestine and rhage.
in microvillous membranes of enterocytes at the base of
the villi (Waddell et al. 1996). CLINICAL SIGNS
The most consistent injury observed in natural cases,
after injection of partially purified toxin (Clugston et al. Postweaning E. coli Diarrhea
1974b; Gannon et al. 1989), and in pigs inoculated oral- In the spontaneous outbreaks caused by an E. coli strain
ly with live cultures (Methiyapun et al. 1984; Kausche et of O149 without F4 investigated by Svendsen et al.
al. 1992) is a degenerative angiopathy of small arteries (1974), the first manifestation of PWECD was sudden
and arterioles. The edema fluid found in various tissues death of one or several pigs as early as 2 days after wean-
is low in protein and could be the result of a mild in- ing. At the same time, feed consumption of the affected
crease in vascular permeability. Information on patho- batch of pigs dropped markedly, and a watery diarrhea
physiology of ED is scarce. Clugston et al. (1974a) ob- developed. At the onset, some pigs exhibited a character-
served an increase of blood pressure after intravenous istic quivering of the tail. The rectal temperature was
administration of EDP. Hypertension developed later normal. Affected pigs became dehydrated and de-
than clinical edema and was therefore thought to be the pressed. They ate irregularly, but even in the terminal
result of vascular injury rather than its cause. Hyperten- stage of the disease, they tried to drink. Many pigs
sion might exacerbate the lesions in the already damaged showed cyanotic discoloration of the tip of the nose, the
vessels. The development of injuries in the nervous sys- ears, and the abdomen. Even severely affected pigs tried
tem may be due to hypoxia resulting from impaired to move around with staggering, uncoordinated move-
blood flow (Clugston et al. 1974b). ments. The peak of mortality occurred 6–10 days after
A distinct type of ED is characterized by terminal weaning. Surviving pigs recovered well. Some pigs were
bloody diarrhea and hemorrhagic lesions in the cardiac completely spared from the disease.
region of the stomach, the ileum, and the large intestine In pigs experimentally infected with a strain with vir-
(Fig. 32.4) (Bertschinger and Pohlenz 1983). According ulence factors F4, LT, and STII, diarrhea started 1–2 days
to Gannon et al. (1989) and MacLeod and Gyles (1990), after inoculation and was fulminating and fatal in some
acute hemorrhagic gastroenteritis occurs in some of the of the pigs, whereas others survived after diarrhea of 3–4
pigs to which a high dose of SLT-IIv is administered. Ep- days’ duration (Sarmiento et al. 1988a). The pigs lost
some weight in the first 2 days after inoculation. From
the fifth day on, the growth curve of susceptible pigs re-
sumed a course parallel to that of genetically resistant
pigs. The latter were not colonized and remained free
from any symptoms.

Postweaning E. coli Diarrhea Combined


with Edema Disease
Smith and Halls (1968) described the disease experimen-
tally induced with a strain of serogroup O141:K85a,c,
obviously a producer of SLT-IIe and of enterotoxin(s).
The first sign was anorexia, which started at the onset of
shedding bacterial numbers above 109 colony-forming
units (CFU)/g of feces 3 (2–5) days postinoculation (PI)
and was observed in 20 out of 21 experimental pigs.
Anorexia lasted for several days in the 2 pigs that recov-
ered and until euthanasia in the 19 pigs killed when
death was imminent. Diarrhea appeared on day 4 (1–8)
PI. Usually it was severe but of short duration. It was fa-
tal in only 1 out of 17 diarrheic pigs. In most pigs diar-
rhea was no longer present when signs of nervous in-
volvement became apparent, that is, on day 6 (5–13) PI.
Swollen eyelids were seen at about the same time. Ataxia
32.4. Extended hemorrhages and minor mesenteric was accompanied with varying degrees of mental confu-
edema in the colon of a pig that developed bloody sion and was usually progressive. Affected pigs soon be-
diarrhea 5 days after inoculation with a culture of came completely recumbent. Severe dyspnea was usually
E. coli O139:K12:H1. present at this final stage. Nine out of 13 pigs with atax-
446 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

ia had to be killed on the day the nervous signs appeared. ma is rare. Affected pigs should be destroyed. Subclinical
Three others survived for 2–4 days. Pigs were moribund ED was observed in 28 of 29 pigs surviving for 2 weeks
7 (5–13) days PI. The rectal temperature always re- after inoculation with a strain of E. coli O139 positive for
mained within the normal range. SLT-IIe and STII. The pigs were clinically normal but de-
veloped vascular lesions. No pigs were allowed to survive
Edema Disease beyond the two weeks (Kausche et al. 1992).
A very similar disease was seen in pigs inoculated with a
nonenterotoxigenic ED strain of serotype O139:K12:H1 LESIONS
(Bertschinger et al. 1978). Discrepancies were that diar-
rhea was not associated with colonization and that more Postweaning E. coli Diarrhea
pronounced edema developed in some cases. In such cas- GROSS LESIONS. Pigs dead from PWECD are gener-
es ears, subcutaneous tissue over the frontal bones, nose, ally in good condition but severely dehydrated with
and lips were swollen (Fig. 32.5). In mild cases, subcuta- sunken eyes and some cyanosis. Lungs look pale and dry,
neous edema was accompanied by pruritus, which dis- like from well-bled pigs (Svendsen et al. 1974). The stom-
appeared after recovery. In some pigs with or without ach is often extended by dry feed. Variable hyperemia of
dyspnea, respiration was accompanied by a snoring the gastric mucosa is often noted in the fundic region.
sound. The small intestine is dilated, slightly edematous, and
Watery diarrhea with clots of fresh blood became ap- hyperemic. The contents vary from watery to mucoid,
parent in a few pigs at the terminal stage (Bertschinger with a characteristic smell. The mesentery is heavily con-
and Pohlenz 1983). Hemorrhagic colitis developed also gested. Contents of the large intestine most often look
in pigs to which high doses of toxin SLT-IIe were admin- light greenish or yellowish and are mucoid to watery.
istered (MacLeod et al. 1991). Pigs dying late in an outbreak look emaciated and exhib-
Chronic ED occurred in a variable, but mostly low, it a strong smell of ammonia. There are irregularly
proportion of pigs recovering from acute attacks of ED shaped superficial ulcerations in the fundic region of the
or PWECD (Bertschinger and Pohlenz 1974; Nakamura stomach and similar lesions of smaller size in the large
et al. 1982). The condition was called cerebrospinal an- intestine. The feces look yellow and pasty. The fluid from
giopathy before its association with ED became appar- the anterior chamber of the eye gives a positive reaction
ent. For periods varying from days to several weeks after for urea (P. Wegmann, Zurich, pers. comm.).
intestinal infection, growth stops, and sick pigs often Some authors use the terms “hemorrhagic gastroen-
show unilateral nervous disturbances such as circling teritis” or “colibacillary shock” to describe a form of E.
movements, twisting of the head, or atrophy of limb coli diarrhea characterized by severe congestion of the
muscles with progressive weakness. Subcutaneous ede- gastric fundus and the small intestine with or without
blood-tinged contents of the small intestine and some-
times the upper large intestine, but only exceptionally
with bloody feces (Faubert and Drolet 1992). This type of
lesion was always caused by E. coli with F4 fimbriae.

MICROSCOPIC LESIONS. Bacteria adhering to the


ileal and less consistently to the jejunal surface are always
seen. The bacterial layers are restricted to villi and look
patchy (Sarmiento et al. 1988a; Casey et al. 1992; Faubert
and Drolet 1992). The mucosa and the epithelium ap-
pear intact. Some investigators have reported increased
numbers of neutrophils in the superficial lamina pro-
pria. In pigs with so-called hemorrhagic gastroenteritis,
severe congestion of the gastric and small-intestinal mu-
cosae is commonly associated with microvascular fibri-
nous thrombi. Necrosis of villi with marked infiltration
of neutrophils occurs in severe cases. There is only occa-
sional hemorrhage in the lamina propria of the jejunum
and the ileum (Svendsen et al. 1974; Faubert and Drolet
1992).
32.5. Edematous swelling of eyelids, forehead, and
lips, breathing through open mouth, and inability to rise Edema Disease
in a weaner 4 days after oral inoculation with a culture of GROSS LESIONS. Pigs dead of ED are mostly in good
E. coli O139:K12:H1. condition, somewhat pale, and the bodies retain a fresh
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 447

appearance. Edema at specific sites is variable and may form of hemorrhagic gastroenteritis occurs which is
be absent in some animals. Subcutaneous edema may oc- quite different from that described with PWECD. In ad-
cur. Edema in the submucosa of the stomach is charac- dition to marked edema, the edematous submucosa of
teristic when present and is located in the region of the the cardiac region of the stomach and the mucosa of the
glandular cardia (Fig. 32.6). It may vary from being bare- lower small and upper large intestine show extensive he-
ly detectable to 2 cm or more in thickness. The edema morrhage. Watery diarrhea with clots of coagulated
fluid is usually serogelatinous in nature and occasionally blood occurs shortly before death in some of these pigs
may be bloodstained adjacent to the mucosa. If severe, (Bertschinger and Pohlenz 1983). Endothelial swelling,
the edema may extend into the fundic submucosa. In- vacuolation and proliferation, microthrombus forma-
flammatory edema associated with acute ulceration of tion, subendothelial fibrin, medial necrosis, and perivas-
the esophageal cardia must not be confused with that of cular edema were detected in such cases (Methiyapun et
ED. al. 1984). The similarity to human hemorrhagic colitis is
Edema of the gallbladder is sometimes seen. The striking.
mesocolon is a common site for edema. Careful inspec- If the causative strain of E. coli is also capable of pro-
tion of the pericardial, pleural, and peritoneal cavities ducing enterotoxin, lesions of postweaning diarrhea
may reveal occasional whitish fibrin strands and a slight may be added, and edema may be mild or absent.
increase in serous fluid. The mesenteric and colic nodes
vary in appearance from normal to being swollen, ede- MICROSCOPIC LESIONS. Patchy layers of adhering
matous, and congested. Typically, the stomach is full of bacteria are present throughout the small intestine early
dry, fresh-looking feed, and the small intestine is rela- in the course of ED (Bertschinger and Pohlenz 1983; Me-
tively empty. Colonic contents may be diminished in thiyapun et al. 1984; Bertschinger et al. 1990). Contrast-
amount. It may be inferred that this is a manifestation of ing with PWECD, the colonization has often disappeared
delayed gastric emptying, since some animals have a pe- when pigs with ED become moribund (Smith and Halls
riod of anorexia before death. Also, it has been shown ex- 1968).
perimentally that pigs with ED may eat very little for 48 The most important microscopic lesions are those of
hours before death and at necropsy have full stomachs a degenerative angiopathy affecting small arteries and ar-
(Smith and Halls 1968). The suggestion that some pigs terioles (Clugston et al. 1974b; Kausche et al. 1992). The
with ED are affected by constipation is also in accord lesions may occur in many organs and tissues. The dense
with these observations. arterial network in the mesocolon adjacent to the colic
The lungs may display varying degrees of edema and lymph nodes is frequently affected. The early acute lesion
a characteristic, patchy, sublobular congestion. In some is one of necrosis of smooth muscle cells in the tunica
cases this may be the only observable lesion. Cases with media characterized by pyknosis and karyorrhexis of nu-
laryngeal edema have also been observed. A few epicar- clei and hyaline change in cytoplasmic elements. In the
dial and endocardial petechiae may occur. This lesion walls of some affected vessels, fibrinoid material is de-
must not be confused with mulberry heart disease. posited (Fig. 32.7). Swelling of endothelial cells has also
In some pigs with spontaneous or experimental ED, a been observed. In acute experimental cases, edema of

32.6. Edematous swelling of


glandular mucosa of the cardiac
region and gelatinous submucosal
edema of the cut stomach wall from
a field case from which E. coli
O139:K12:H1 was isolated.
448 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

32.7. Arterioles in submucosa


of the urinary bladder: (left)
normal; (right) fibrinoid or
hyaline change, acute experi-
mental ED. (Clugston et al.
1974b.)

the leptomeninges and perivascular spaces has been plus the occurrence of eosinophilic, Periodic Acid Schiff
demonstrated. In older lesions, there may be prolifera- (PAS)–positive droplets around affected vessels. This an-
tion of adventitial and medial cells (Fig. 32.8). Vascular giopathy is considered to most likely be a manifestation
lesions may be difficult to detect in acute cases, but in of edema disease (Bertschinger and Pohlenz 1974).
surviving pigs or those affected subclinically, they are
more readily apparent (Kausche et al. 1992). Thrombosis DIAGNOSIS
is not usually a feature of uncomplicated naturally oc-
curring ED. Postweaning E. coli Diarrhea
In pigs that have recovered from natural outbreaks or Postweaning diarrhea is a very complex disease with
survived for several days following acute signs, there are multiple etiologies (see Chapter 57). Occurrence of diar-
lesions of focal encephalomalacia in the brain stem to- rhea early after weaning, marked dehydration, and at
gether with lesions in the small arteries and arterioles least some mortality are characteristics in the field allow-
(Kurtz et al. 1969; Kausche et al. 1992). These are ing a preliminary diagnosis of ETEC. The gross lesions,
thought to be the result of vascular injury leading to ede- including the characteristic smell, are also helpful. The fi-
ma and ischemia. A cerebrospinal angiopathy of pigs has nal diagnosis requires detection of ETEC, which are shed
been recognized as a clinicopathologic entity for some in high numbers. Hemolysis is not a valid criterion for
years. Its microscopic features are those described above identification of ETEC. Laboratories not equipped for
the determination of virulence factors should at least use
serotyping of living cultures with OK-antisera against
serotypes most prevalent in a given region.

Edema Disease
The diagnosis of acute ED is based on sudden appear-
ance and on clinical signs of neurologic disease in thriv-
ing pigs 1–2 weeks after weaning. In the live pig the most
important and constant diagnostic sign is partial ataxia
or a staggering gait. Subcutaneous edema in the palpe-
brae and over the frontal bones is also a cardinal sign
when present. At necropsy the characteristic lesions of
edema, when present, are helpful in confirming the diag-
nosis but may be absent in a significant number of cases,
especially when severe diarrhea has preceded ED. Diag-
nosis of ED in adult pigs may require additional efforts,
32.8. Submucosa of cardial gland region of the stom- such as histopathology and postmortem examination of
ach 17 days after inoculation with EDP (edema disease more than one pig.
principle containing SLT-IIe); arteriole with proliferative Bacteriologic examination of the small intestine and
arteriopathy. (Clugston et al. 1974b.) colon should yield nearly pure cultures of hemolytic E.
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 449

coli. However, bacterial numbers may already be drop- Table 32.5. Prevalences of sensitive isolates of
ping in more protracted cases (Bertschinger and Pohlenz typable E. coli from pigs over 2 weeks of age with
PWECD or ED from Switzerland
1983). After death the small numbers of organisms may
be overgrown by other enterobacteria. In contrast to Sensitive Isolates (%) in Serogroup
ETEC infections, a negative bacteriologic result therefore Antimicrobial O139 O141 O149
does not exclude the diagnosis of ED. Serologic identifi- Substance (n = 52) (n = 9) (n = 59)
cation of the common serotypes associated with ED is
Ampicillin 81 67 73
additional evidence. Serotyping is essential, because he- Amoxicillin/ 100 100 100
molytic E. coli not associated with other virulence factors clavulanic acid
are frequently encountered in the intestinal flora and Cefoxitin 100 100 100
may be present in high numbers. Streptomycin 35 44 37
Subacute or chronic ED is diagnosed by the demon- Spectinomycin 15 0 19
stration of arteriopathy and eventually lesions of focal Neomycin 90 100 81
encephalomalacia. Apramycin 92 89 93
Gentamicin 96 89 91
In cases of sudden death, differential diagnosis will
have to include microangiopathia dietetica and circulato- Tetracycline 36 44 47
ry failure, as seen after severe fighting. When pigs show Chloramphenicol 69 78 81
Enrofloxacin 100 100 100
nervous signs, viral encephalitis (enteroviral polioen- Colistina 87 89 100
cephalomyelitis, pseudorabies) and bacterial menin-
goencephalitis (Streptococcus suis, Haemophilus parasuis) Sulfonamide 31 11 15
Trimethoprim 77 44 73
as well as water deprivation should be considered. Furazolidone 88 44 100
a
TREATMENT Modified agar dilution technique (Bertschinger et al. 1996).

Much less is known about the treatment of these dis- Edema Disease
eases than about pathogenesis and about treatment of There is not much chance to save the lives of pigs with
neonatal colibacillosis. advanced signs such as severe subcutaneous edema, res-
piratory distress, or inability to rise. Evaluation of thera-
Antimicrobial Therapy py is difficult because the severity of the illness cannot be
Chemotherapeutic control of bacterial proliferation is quantified. Numerous remedies have been recommend-
therapeutically much more effective in PWECD than in ed in the past and have then been abandoned.
ED, because in the latter, toxin production in the gut is
nearly completed when clinical signs become visible. The PREVENTION
development of bacterial resistance against every active
principle hitherto used (Table 32.5) renders this ap- No universally effective prophylaxis is so far available. In
proach uncertain. It is not possible to give universal data view of the unpredictable, erratic occurrence of PWECD
on resistance, because the situation varies in different pig and ED in sequential batches of weaners and even in
populations depending on the substances preferentially contemporaneously weaned litters, efficacy of prophy-
used. lactic measures is hard to assess.
Sick pigs must be treated parenterally. They eat
and drink very little, even if they stand close to the Breeding of Resistant Pigs
creep and to the drinking nipple. Substances must be This is the approach to prevention that will be most
selected which reach the intestinal lumen, such as effective and economical in the long term. The selection
amoxicillin/clavulanic acid, fluoroquinolones, cephalo- of breeding stock is not yet feasible, because the tools for
sporins, or trimethoprim. Testing bacterial resistance is the identification of the genotype in living pigs are not
indispensable if there is a herd problem. yet available. Work in this direction is in progress. It will
be important to avoid coselection of unwanted traits
Supportive Therapy for PWECD closely linked with loci coding for the F4 and the F18
The supportive therapy has to counteract dehydration receptors. It cannot be predicted if additional types
and acidosis. Attractive rehydration fluid should be of- of adhesive fimbriae or new variants of known types
fered for spontaneous intake or injected intraperitoneal- will emerge which could bind to yet unidentified recep-
ly if the pig is anorectic. Such fluids may contain glucose, tors.
glycine, citric acid, and potassium dihydrogen phos-
phate in an isotonic solution (Bywater and Woode 1980). Prevention of Infection
Uptake should be equal to the loss (i.e., up to 25% of the The transmissible character of PWECD and ED is evi-
body weight). dent. In Denmark most of the recent spread of ED fol-
450 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

lowed the routes of pig trade (Jorsal et al. 1996). It was Active immunity against intravenous challenge with
logical to start an eradication program involving depop- SLT-IIe was induced in young pigs by a toxoid vaccine
ulation of affected farms and disinfection of the build- prepared from SLT-IIe by treatment with glutaraldehyde
ings (Johansen et al. 1996b). With one exception, the 15 (Dobrescu 1982). A similar toxoid was used for vaccina-
participating farms remained free of clinical disease for a tion of pigs 1 week before weaning (Awad-Masalmeh et
minimum of 4–7 months. They are under continuing al. 1989). The vaccine conferred highly significant pro-
surveillance. Some problems render the procedure risky. tection against ED after the pigs were orally challenged
The tools to prove the absence of pathogenic types of E. with ETEEC serogroup O139:K12. Vaccinated principals
coli from a given herd are not available. E. coli has a high shed lower numbers of the inoculated bacteria and had
tenacity in the environment. In our experience the isola- better weight gains than placebo-vaccinated littermates.
tion measures applied in the Swiss SPF nucleus herds A toxoid prepared by treatment of SLT-IIe with
were not suitable to keep out pathogenic E. coli. formaldehyde was not completely free of toxic activity.
Therefore, the toxin was modified by site-directed muta-
Immunoprophylaxis genesis. The genetically modified toxin was found to
Acquired immunity results in perfect protection against have no deleterious effect on the growth of vaccinated
intestinal colonization and/or against effects of the tox- pigs, and it prevented overt and subclinical ED when vac-
ins. Weaned pigs can be protected passively or actively. cinated pigs were challenged with an E. coli O139:F18
positive for SLT-IIe and STII (Bosworth et al. 1996). A
PASSIVE IMMUNITY. A daily dose of 525 mL, but different approach was chosen by MacLeod and Gyles
not of 270 mL, milk obtained from sows in late lactation (1991), who detoxified purified SLT-IIe with glutaralde-
fed to weaned pigs completely inhibited colonization, hyde. An adjuvanted experimental vaccine was evaluated
whereas pigs fed the same amount of cow’s milk shed in two herds infected by a strain of E. coli O139:F18, SLT-
high numbers of the ETEEC bacteria (Deprez et al. IIe. Mortality due to ED was significantly reduced, and
1986). Spray-dried porcine blood plasma fed at a dose of daily gain in the nursery was significantly improved.
90 g per pig/day had a similar inhibitory effect that last- Deaths caused by ETEC in one of the herds were not pre-
ed only as long as the plasma was fed, and the inhibitory vented (Johansen et al. 1996a; M. Johansen, Kjellerup,
effect could be improved by vaccination of the donor Denmark, pers. comm.). These toxoid vaccines are not
pigs (Deprez et al. 1990, 1996). Immune protection yet commercially available.
against colonization with F4- and F18-positive E. coli was
achieved by feeding eggs produced by vaccinated hens Chemoprophylaxis
(Deprez et al. 1992; Erhard et al. 1996). Such protection At present, preventive feed medication is practiced in a
was achieved against challenge strains which had only majority of the affected herds in most countries despite
the F18 fimbriae in common with the vaccine strains serious drawbacks such as nonacceptance by the con-
(Zúñiga et al. 1998). It remains to be shown if the anti- sumer, impaired buildup of immunity, and selection of
body-containing egg powder can be produced at an ac- resistant bacteria. Resistance is often induced within
ceptable price. days or a few weeks. Isolates from PWECD and ED show
the highest rate of resistance within porcine E. coli. Be-
ACTIVE IMMUNITY. Pigs that have been colonized sides the classes of substances mentioned above for par-
by an F18 ETEC producing STI and STII are protected enteral therapy, the aminoglycosides and colistin are
against recolonization by a heterologous ETEC sharing widely used. The latter has the advantages of high stabil-
no other antigens with the immunizing strain except F18 ity, low toxicity, absence of infectious resistance, and
fimbriae. There is complete cross-protection between slow development of resistance. With colistin, resistance
strains with fimbrial variants F18ab and F18ac (Sarrazin cannot reliably be detected by the agar diffusion tech-
and Bertschinger 1997). In this experimental model, 6 nique (Bertschinger et al. 1996). Investigators have re-
days after the onset of colonization, pigs are protected ported that oxytetracycline reduces the adhesion of E.
against recolonization (unpublished). Intestinal colo- coli at concentrations below the minimum inhibitory
nization leads to an increase of serum antibodies against concentration. Sarmiento and Moon (1988) reported
the fimbriae, especially of the IgA class. Partial protec- that PWECD induced by a tetracycline-resistant strain
tion against PWECD was reported by Fahy et al. (1992) takes an identical course in pigs eating feed with and
in pigs orally vaccinated before weaning with an attenu- without tetracycline.
ated strain of E. coli. Zinc oxide offers an alternative to antimicrobials.
Sera from neonatal and from weaned pigs from herds Feeds with contents between 2400 and 3000 ppm of zinc
with and without clinical ED do not contain neutralizing reduce diarrhea and mortality and improve growth. The
antibody against SLT-IIe (Gannon et al. 1988). However, activity is explained by an antibacterial effect (Holm and
in pigs that had survived an outbreak of ED, Wieler et al. Poulsen 1996). However, environmental considerations
(1995) found antibodies reacting in an ELISA to the B should be included in discussions of zinc oxide at such
subunit of SLT-IIe. high levels.
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 451

Dietary Measures new pens, etc. Wathes et al. (1989) observed a higher in-
Restriction of feed intake, high-fiber diets, or ad libitum cidence of scours and greater mortality due to PWECD
feeding of fiber have been reported as effective deter- in experimentally infected pigs kept at 15˚C than in con-
rents to the development of ED and postweaning diar- trols at higher temperatures. The experiment was not
rhea (Smith and Halls 1968; Bertschinger et al. 1978; perfectly conclusive because genetic resistance was ne-
Rantzer et al. 1996). The nutritive value of the feed may glected.
be reduced by increasing fiber content to 15–20% and re-
ducing crude protein and digestible energy to one-half of REFERENCES
the normal values (Bertschinger et al. 1978). The addi-
tion of fiber to normal diets may be beneficial. Dunne Aarestrup, M. F.; Jorsal, S. E.; Ahrens, P.; Jensen, N. E.; and
(1975) advocated feeding high-quality alfalfa coupled Meyling, A. 1997. Molecular characterization of Es-
with restriction of daily feed intake. To be effective, nu- cherichia coli strains isolated from pigs with edema dis-
trient intake has to be low enough to maintain daily gain ease. J Clin Microbiol 35:20–24.
below 1% of body weight in the 2 weeks after weaning. Awad-Masalmeh, M.; Reitinger, H.; Quakyi, E.; Hinterdor-
Such diets prevent colonization and impair the develop- fer, H.; Silber, R.; and Willinger, H. 1988. Observations
ment of immunity in the same way as antimicrobials. on the isolation and characterization of E. coli derived
Later outbreaks are frequently seen. from edema disease cases. Proc Int Congr Pig Vet Soc
A lower mortality due to E. coli enterotoxemia and 10:114.
improved weight gains were reported after introduction Awad-Masalmeh, M.; Schuh, M.; Köfer, J.; and Quakyi, E.
of rations with a reduced acid-binding capacity. A simi- 1989. Überprüfung der Schutzwirkung eines Tox-
lar effect is ascribed to organic acids. Others were unable oidimpfstoffes gegen die Ödemkrankheit des Absetzfer-
to reduce the mortality due to ED by the inclusion of a kels im Infektionsmodell. Dtsch Tierärztl Wochenschr
mixture of organic and inorganic acids in the feed (Jo- 96:397–432.
hansen et al. 1996b). This result is not surprising in view Bertschinger, H. U., and Pohlenz, J. 1974. Cerebrospinale
of the highly regulated pH close to the mucosal surface Angiopathie bei Ferkeln mit experimenteller Coli-En-
(McEwan et al. 1990). terotoxämie. Schweiz Arch Tierheilk 116:543–554.
Exogenous as well as endogenous proteases lower the ———. 1983. Bacterial colonization and morphology of
activity of intestinal F4 receptors. Bromelain, a protease the intestine in porcine Escherichia coli enterotoxemia
from pineapple stems, applied orally to pigs reduced the (edema disease). Vet Pathol 20:99–110.
binding of F4-positive ETEC to brush borders in a dose- Bertschinger, H. U.; Eggenberger, U.; Jucker, H.; and Pfirter,
dependent manner (Mynott et al. 1996). The efficacy in a H. P. 1978. Evaluation of low nutrient, high fibre diets
clinical situation remains to be demonstrated. for the prevention of porcine Escherichia coli enterotox-
Scandinavian workers have shown that antisecretory aemia. Vet Microbiol 3:281–290.
factor reverses secretory diarrhea induced by heat-labile Bertschinger, H. U.; Bachmann, M.; Mettler, C.; Pospischil,
enterotoxin. The levels of antisecretory factor in the A.; Schraner, E. M.; Stamm, M.; Sydler, T.; and Wild, P.
blood plasma can be increased by addition of glucose 1990. Adhesive fimbriae produced in vivo by Escherichia
and of some amino acids to the feed. Treated weaner pigs coli O139:K12(B):H1 associated with enterotoxaemia in
were reported to have a lower incidence of diarrhea and pigs. Vet Microbiol 25:267–281.
a greater weight gain (Göransson et al. 1993). Antisecre- Bertschinger, H. U.; Stamm, M.; and Vögeli, P. 1993. Inher-
tory factor probably exerts its effects on the enteric ner- itance of resistance to oedema disease in the pig: Exper-
vous system (Hansen and Skadhauge 1995). iments with an Echerichia coli strain expressing fimbriae
107. Vet Microbiol 35:79–89.
Bacterial Probiotics Bertschinger, H. U.; Mettler, C.; Blaser, C.; and Fritzsche, C.
In recent investigations Enterococcus faecium, Lactobacil- 1996. Colistin resistance of E. coli isolated from pigs
lus sp., and Bacillus cereus strain “toyoi” were fed to ex- with oedema disease and postweaning diarrhoea detect-
perimentally (De Cupere et al. 1992) and to naturally in- ed by a modified agar dilution method. Proc Int Congr
fected (Johansen et al. 1996b) pigs. No preventive effect Pig Vet Soc 14:271.
was recorded. Schulze (1977) confirmed earlier compar- Bosworth, B. T.; Samuel, J. E.; Moon, H. W.; O’Brien, A. D.;
ative quantitative studies of the gastrointestinal flora in Gordon, V. M.; and Whipp, S. C. 1996. Vaccination with
weaned and in unweaned pigs, where no evidence was genetically modified Shiga-like toxin IIe prevents edema
found for an interdependence between E. coli and the disease in swine. Infect Immun 64:55–60.
other components of the flora. Boyd, B.; Tyrrell, G.; Maloney, M.; Gyles, C.; Brunton, J.;
and Lingwood, C. 1993. Alteration of the glycolipid
General Management binding specificity of the pig edema toxin from globote-
Management of the weanling pig should minimize envi- traosyl to globotriaosyl ceramide alters in vivo tissue tar-
ronmental and other forms of stress such as unnecessary getting and results in a verotoxin 1–like disease in pigs. J
mixing of litters, chilling, transportation, assignment to Exp Med 177:1745–1753.
452 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Bywater, R. J., and Woode, G. N. 1980. Oral fluid replace- tis caused by Escherichia coli in piglets: Clinical, patho-
ment by a glucose glycine electrolyte formulation in E. logical and microbiological findings. Can Vet J
coli and rotavirus diarrhoea in pigs. Vet Rec 106:75–78. 33:251–256.
Casey, T. A.; Nagy, B.; and Moon, H. W. 1992. Pathogenici- Gannon, V. P. J.; Gyles, C. L.; and Friendship, W. 1988. Char-
ty of porcine enterotoxigenic Escherichia coli that do not acteristics of verotoxigenic Escherichia coli from pigs.
express K88, K99, F41, or 987P adhesins. Am J Vet Res Can J Vet Res 52:331–337.
53:1488–1492. Gannon, V. P. J.; Gyles, C. L.; and Wilcock, B. P. 1989. Effects
Clugston, R. E.; Nielsen, N. O.; and Roe, W. E. 1974a. Ex- of Escherichia coli Shiga-like toxins (verotoxins) in pigs.
perimental edema disease of swine (E. coli enterotox- Can J Vet Res 53:306–312.
emia). II. The development of hypertension after the in- Göransson, L.; Martinsson, K.; Lange, S.; and Lönnroth, I.
travenous administration of edema disease principle. 1993. Feed-induced lectins in piglets: Feed-induced
Can J Comp Med 38:29–33. lectins and their effect on post-weaning diarrhoea, daily
Clugston, R. E.; Nielsen, N. O.; and Smith, D. L. T. 1974b. weight gain and mortality. J Vet Med B 40:478–484.
Experimental edema disease of swine (E. coli enterotox- Guérin, G.; Duval-Iflah, Y.; Bonneau, M.; Bertaud, M.; Guil-
emia). III. Pathology and pathogenesis. Can J Comp Med laume, P.; and Ollivier, L. 1993. Evidence for linkage be-
38:34–43. tween K88ab, K88ac intestinal receptors to Escherichia
De Cupere, F.; Deprez, P.; Demeulenaere, D.; and Muylle E. coli and transferrin loci in pigs. Anim Genetics
1992. Evaluation of the effect of 3 probiotics on experi- 24:393–396.
mental Escherichia coli enterotoxaemia in weaned Gyles, C. L. 1994. Escherichia coli in Domestic Animals and
piglets. J Vet Med B 39:277–284. Humans. Wallingford, U.K.: CAB International.
Deprez, P.; Van den Hende, C.; Muylle, E.; and Oyaert, W. Hampson, D. J.; Fu, Z. F.; and Robertson, I. D. 1987. Investi-
1986. The influence of the administration of sow’s milk gation of the source of haemolytic Escherichia coli in-
on the post-weaning excretion of hemolytic E. coli in the fecting weaned pigs. Epidemiol Inf 99:149–153.
pig. Vet Res Commun 10:469–478. Häni, H.; Brändli, A.; Nicolet, J.; Von Roll, P.; Luginbühl, H.;
Deprez, P.; De Cupere, F.; and Muylle, E. 1990. The effect of and Hörning, B. 1976. Vorkommen und Bedeutung von
feeding dried plasma on experimental Escherichia coli Schweinekrankheiten: Analyse eines Sektionsguts
enterotoxemia in piglets. Proc Int Congr Pig Vet Soc (1971–1973). III. Pathologie des Digestionstraktes.
11:149. Schweiz Arch Tierheilk 118:13–29.
Deprez, P.; De Cupere, F.; Muylle, E.; and Zöbisch, H. 1992. Hansen, M. B.; and Skadhauge, E. 1995. New aspects of the
The use of egg powder antibodies in the prevention of pathophysiology and treatment of secretory diarrhoea.
post weaning colibacillosis. Proc Int Congr Pig Vet Soc Physiol Res 44:61–78.
12:251. Holm, A.; and Poulsen, H. D. 1996. Swine nutrition man-
Deprez, P.; Nollet, H.; Van Driessche, E.; and Muylle, E. agement update: Zinc oxide in treating E. coli diarrhea in
1996. The use of swine plasma components as adhesin pigs after weaning. Compend Cont Educ Pract Vet
inhibitors in the protection of piglets against Escherichia 18:S26–S48.
coli enterotoxemia. Proc Int Congr Pig Vet Soc 14:276. Imberechts, H.; Bertschinger, H. U.; Stamm, M.; Sydler, P.;
Dobrescu, L. 1982. Immunological studies in pigs using ede- Pohl, P.; De Greve, H.; Hernalsteens, J.-P.; Van Montagu,
ma disease principle (E. coli neurotoxin). Proc Int Congr M.; and Lintermans, P. 1994. Prevalence of F107 fimbri-
Pig Vet Soc 7:19. ae on Escherichia coli isolated from pigs with oedema dis-
Dunne, H. W. 1975. Colibacillosis and edema disease. In ease or postweaning diarrhoea. Vet Microbiol
Diseases of Swine, 4th ed. Ed. H. W. Dunne and A. D. 40:219–230.
Leman. Ames: Iowa State Univ Press, p. 674. Jahn, S., and Uecker, E. 1987. Ökonomische Untersuchun-
Erhard, M. H.; Bergmann, J.; Renner, M.; Hofmann, A.; and gen zur Kolienterotoxämie des Schweines. Monatsh Vet
Heinritzi, K. 1996. Prophylaktische Wirkung von spezi- Med 42:769–771.
fischen Dotterantikörpern bei Escherichia coli K88 (F4)- Johansen, M.; Andresen, L. O.; Jorsal, S. E.; Thomsen, L. K.;
bedingten Durchfallerkrankungen von Absatzferkeln. J Waddell, T.; and Gyles, C. L. 1996a. Vaccination against
Vet Med A 43:217–223. oedema disease. Proc Int Congr Pig Vet Soc 14:378.
Fahy, V. A.; Connaughton, I. D.; Driesen, S. J.; and Spicer, E. Johansen, M.; Baekbo, P.; and Thomsen, L. K. 1996b. Con-
M. 1992. Field trials with an oral E. coli vaccine (Auto- trol of edema disease in Danish pig herds. Proc Int Con-
vac) to prevent postweaning coliform enteritis. Proc Int gr Pig Vet Soc 14:256.
Congr Pig Vet Soc 12:255. Jorsal, S. E.; Aarestrup, F. M.; Ahrens, P.; Johansen, M.; and
Fairbrother, J. M.; Harel, J.; D’Allaire, S.; and Bonneau, M. Baekbo, P. 1996. Oedema disease in Danish pig herds:
1994. Characterisation of Escherichia coli isolated from Transmission by trade of breeding animals. Proc Int
postweaning piglets with and without diarrhea. Proc Int Congr Pig Vet Soc 14:265.
Congr Pig Vet Soc 13:212. Kausche, M.; Dean, E. A.; Arp, L. H.; Samuel, J. E.; and
Faubert, C., and Drolet, R. 1992. Hemorrhagic gastroenteri- Moon, H. W. 1992. An experimental model for subclin-
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 453

ical edema disease (Escherichia coli enterotoxemia) man- Rantzer, D.; Svendsen, J.; and Weström, B. 1996. Effects of
ifested as vascular necrosis in pigs. Am J Vet Res strategic feed restriction on pig performance and health
53:281–287. during the post-weaning period. Acta Agric Scand Sect
Kelly, D.; Begbie, R.; and King, T. P. 1994. Nutritional influ- A Anim Sci 46:219–226.
ences on interactions between bacteria and the small in- Rippinger, P.; Bertschinger, H. U.; Imberechts, H.; Nagy, B.;
testinal mucosa. Nutrition Res Rev 7:233–257. Sorg, I.; Stamm, M.; Wild, P.; and Wittig, W. 1995. Des-
Kurtz, H. J.; Bergeland, M. E.; and Barnes, D. M. 1969. ignations F18ab and F18ac for the related fimbrial types
Pathologic changes in edema disease of swine. Am J Vet F107, 2134P and 8813 of Escherichia coli isolated from
Res 30:791–806. porcine postweaning diarrhoea and from oedema dis-
Lange, S.; Martinsson, K.; Lönnroth, I.; and Göransson, L. ease. Vet Microbiol 45:281–295.
1993. Plasma level of antisecretory factor (ASF) and its Risley, C. R.; Kornegay, E. T.; Lindenmann, M. D.; Wood, C.
relation to post-weaning diarrhoea in piglets. J Vet Med M.; and Eigel, W. N. 1992. Effect of feeding organic
B 40:113–118. acids on selected intestinal content measurements of
Lecce, J. G.; Balsbaugh, R. K.; Clare, D. A.; and King, M. W. varying times postweaning in pigs. J Anim Sci
1982. Rotavirus and hemolytic enteropathogenic Es- 70:196–206.
cherichia coli in weanling diarrhea of pigs. J Clin Micro- Sarmiento, J. I., and Moon, H. W. 1988. Postweaning diar-
biol 16:715–723. rhea in swine: Effects of oxytetracycline on enterotoxi-
MacLeod, D. L., and Gyles, C. L. 1990. Purification and genic Escherichia coli infection. Am J Vet Res
characterization of an Escherichia coli Shiga-like toxin II 49:1160–1163.
variant. Infect Immun 58:1232–1239. Sarmiento, J. I.; Casey, T. A.; and Moon, H. W. 1988a. Post-
———. 1991. Immunization of pigs with a purified Shiga- weaning diarrhea in swine: Experimental model of en-
like toxin II variant toxoid. Vet Microbiol 29:309–318. terotoxigenic Escherichia coli infection. Am J Vet Res
MacLeod, D. L.; Gyles, C. L.; and Wilcock, B. P. 1991. Re- 49:1154–1159.
production of edema disease of swine with purified Shi- Sarmiento, J. I.; Dean, E. A.; and Moon, H. W. 1988b. Effects
ga-like toxin-II variant. Vet Pathol 28:66–73. of weaning on diarrhea caused by enterotoxigenic Es-
McEwan, G. T. A.; Schousboe, B.; and Skadhauge, E. 1990. cherichia coli in three-week-old pigs. Am J Vet Res
Direct measurement of mucosal surface pH of pig je- 49:2030–2033.
junum in vivo. J Vet Med A 37:439–444. Sarrazin, E., and Bertschinger, H. U. 1997. Role of fimbriae
Methiyapun, S.; Pohlenz, J. F. L.; and Bertschinger, H. U. F18 for actively acquired immunity against porcine en-
1984. Ultrastructure of the intestinal mucosa in pigs ex- terotoxigenic Escherichia coli. Vet Microbiol 54:133–144.
perimentally inoculated with an edema disease–produc- Schulze, F. 1977. Quantitative Magen-Darm-Flora-Analysen
ing strain of Escherichia coli (O139:K12:H1). Vet Pathol beim Ferkel vor und nach dem Absetzen unter Berück-
21:516–520. sichtigung der Pathogenese der Kolienterotoxämie.
Mynott, T. L.; Luke, R. K. J.; and Chandler, D. S. 1996. Oral Arch Exper Vet Med 31:299–316.
administration of protease inhibits enterotoxigenic Es- Sellwood, R.; Gibbons, R. A.; Jones, G. W.; and Rutter, J. M.
cherichia coli receptor activity in piglet small intestine. 1975. Adhesion of enteropathogenic Escherichia coli to
Gut 38:28–32. pig intestinal brush borders: The existence of two pig
Nagy, B.; Casey, T. A.; Whipp, S. C.; and Moon, H. W. 1992. phenotypes. J Med Microbiol 8:405–411.
Susceptibility of porcine intestine to pilus-mediated ad- Smith, H. W., and Halls, S. 1968. The production of oedema
hesion by some isolates of piliated enterotoxigenic Es- disease and diarrhoea in weaned pigs by the oral admin-
cherichia coli increases with age. Infect Immun istration of Escherichia coli: Factors that influence the
60:1285–1294. course of the experimental disease. J Med Microbiol
Nakamura, K.; Kubo, M.; Shoya, S.; Kashiwazaki, M.; Koizu- 1:45–59.
mi, S.; and Onai, M. 1982. Swine cerebrospinal angiopa- Sojka, W. J. 1965. Escherichia coli in Domestic Animals and
thy with demyelination and malacia. Vet Pathol Poultry. Farnham Royal, Bucks, England: Commonw
19:140–149. Agric Bur, pp. 104–156.
Neef, N. A.; McOrist, S.; Lysons, R. J.; Bland, A. P.; and Svendsen, J.; Larsen, J.; and Bille, N. 1974. Outbreaks of
Miller, B. G. 1994. Development of large intestinal at- post weaning Escherichia coli diarrhoea in pigs. Nord
taching and effacing lesions in pigs in association with Vet-Med 26:314–322.
the feeding of a particular diet. Infect Immun Svensmark, B.; Nielsen, K.; Willeberg, P.; and Jorsal S. E.
62:4325–4332. 1989. Epidemiological studies of piglet diarrhoea in in-
Ojeniyi, B.; Ahrens, P.; and Meyling, A. 1994. Detection of tensively managed Danish sow herds. Acta Vet Scand
fimbrial and toxin genes in Escherichia coli and their 30:55–62.
relevance in piglets with diarrhoea: The application of Sydler, T.; Buergi, E.; Bertschinger, H. U.; and Pospischil, A.
colony hybridization assay, polymerase chain reaction 1996. Edema disease in adult swine. Proc Int Congr Pig
and phenotypic assays. J Vet Med B 41:49–59. Vet Soc 14:272.
454 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Tzipori, S.; Wachsmuth, I. K.; Chapman, C.; Birner, R.; Brit- Wieler, L. H.; Franke, S.; Menge, C.; Rose, M.; Bauerfeind,
tingham, J.; Jackson, C., and Hogg, J. 1986. The patho- R.; Karch, H.; and Baljer, G. 1995. Investigations on the
genesis of hemorrhagic colitis caused by Escherichia coli immunoresponse during edema disease of piglets after
O157:H7 in gnotobiotic piglets. J Infect Dis weaning by using a recombinant B subunit of Shiga-like
154:712–716. toxin IIe. Dtsch Tierärztl Wochenschr 102:40–43.
Vögeli, P.; Bertschinger, H. U.; Stamm, M.; Stricker, C.; Hag- Wittig, W.; Klie, H.; Gallien, P.; Lehmann, S.; Timm, M.;
ger, C.; Fries, R.; Rapacz, J.; and Stranzinger, G. 1996. and Tschäpe, H. 1995. Prevalence of the fimbrial anti-
Genes specifying receptors for F18 fimbriated Es- gens F18 and K88 and of enterotoxins and verotoxins
cherichia coli, causing oedema disease and postweaning among Escherichia coli isolated from weaned pigs. Zbl
diarrhoea in pigs, map to chromosome 6. Anim Genet- Bakt 283:95–104.
ics 27:321–328. Zhu, C.; Harel, J.; Jacques, M.; Desautels, C.; Donnenberg,
Waddell, T. E.; Lingwood, C. A.; and Gyles, C. 1996. Inter- M. S.; Beaudry, M.; and Fairbrother, J. M. 1994. Viru-
action of verotoxin 2e with pig intestine. Infect Immuni- lence properties and attaching-effacing activity of Es-
ty 64:1714–1719. cherichia coli O45 from swine postweaning diarrhea. In-
Wathes, C. M.; Miller, B. G.; and Bourne, F. J. 1989. Cold fect Immun 62:4153–4159.
stress and post-weaning diarrhoea in piglets inoculated Zúñiga, A.; Yokoyama, H.; Albicker-Rippinger, P.; Eggen-
orally or by aerosol. Anim Prod 49:483–496. berger, E.; and Bertschinger H. U. 1998. Reduced intesti-
Wegmann, P. 1990. Pathology of swine—A portrait of eco- nal colonization with F18-positive enterotoxigenic Es-
nomic loss in pig production in Switzerland. Proc Int cherichia coli in weaned pigs fed chicken egg antibody
Congr Pig Vet Soc 11:295. against the fimbriae. FEMS Immunol Med Microbiol (in
press).

SYSTEMIC INFECTION
J. M. Fairbrother
E. coli may induce systemic infections such as septicemia lower numbers (2–4% each), but 49% of isolates were
or localized extraintestinal infections such as meningitis nontypeable. Not all E. coli isolates are able to cause sep-
or arthritis, resulting from bacteremia (Fairbrother and ticemia in colostrum-deprived piglets (Meyer et al. 1971;
Ngeleka 1994; Fairbrother et al. 1989; Morris and Sojka Murata et al. 1979).
1985). Septicemia due to E. coli may be primary, occur- The characteristics of E. coli involved in porcine sep-
ring predominantly in newborn to 4-day-old pigs, or sec- ticemia have not been greatly studied. However, sep-
ondary, when associated with diarrhea or other compro- ticemia-inducing strains may express virulence determi-
mising diseases in young pigs. nants which include fimbriae, polysaccharide capsule
and O-antigen capsule, lipopolysaccharide (LPS), the
ETIOLOGY aerobactin system, hemolysin, and other cytotoxins.
Fimbrial adhesins associated with E. coli isolates from
Only a relatively small number of E. coli serogroups have piglets with septicemia include the F1651, F1652 (Contre-
been reported in natural cases of septicemia. Serogroups pois et al. 1989; Fairbrother et al. 1986), and other fim-
O6, O8, O9, O11, O15, O17, O18, O20, O45, O60, O78, briae of the P, S, and F1C fimbrial families (Dozois et al.
O83, O93, O101, O112, O115, and O116 have most com- 1997).
monly been identified in isolates associated with sep- Enterotoxigenic E. coli may be associated with sec-
ticemia (Morris and Sojka 1985; Gyles 1986; Nielsen et ondary septicemia, particularly in older piglets. These
al. 1975a; Fairbrother et al. 1989). Other gram-negative isolates most frequently belong to the pathotypes LT +
bacteria such as Klebsiella spp. and Pseudomonas spp. STb, Sta + STb, or STb and may produce the F4 fimbriae
have been reported to be associated with systemic infec- (see Table 32.1).
tions in pigs (Nielsen et al. 1975b). In a 4-year study in The virulence determinants most frequently associat-
our laboratory from 1989 to 1992, the most commonly ed with isolates from primary septicemia in pigs or iso-
observed serogroups in isolates from cases of primary lates inducing septicemia in newborn colostrum-de-
septicemia were O9 (10%) and O20 (18%) (Fairbrother prived pigs are F1651, F1652, or other fimbriae of the P, S,
and Ngeleka 1994). Serogroups O1, O18, O60, O78, and F1C fimbrial families, production of colicin V, pro-
O101, O141, and O147 were isolated in relatively duction of the siderophore aerobactin, and resistance to
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 455

the bactericidal effects of serum (Fairbrother and Ngele- signs, due in part to the effect of bacterial endotoxin or
ka 1994). Isolates from cases of primary septicemia may cytotoxins or to the effects of inflammatory cytokines in-
occasionally produce cytotoxic necrotizing factor duced by these bacterial products (Nakajima et al. 1991;
(CNF1). Jesmok et al. 1992).
The role of some of the virulence determinants asso-
EPIDEMIOLOGY ciated with E. coli–inducing septicemia is only partially
understood. LPS, K capsule and O-antigen capsule, and
Primary septicemia is most often seen as sporadic cases production of siderophores such as aerobactin are
and rarely in the form of a small outbreak (Nielsen et al. thought to allow the bacteria to invade the host and es-
1975a). The disease may occur throughout the suckling cape its defense mechanisms. These determinants in-
period, with exceptional cases in pigs up to 80 days old. crease bacterial resistance to the bactericidal effect of
Epidemiology of the secondary systemic infection is de- complement and to phagocytosis and allow bacterial
termined by the underlying disease. growth in body fluids with low concentrations of free
iron (Crosa 1989; Ngeleka et al. 1992, 1993).
PATHOGENESIS Fimbriae appear to be important for the survival and
spread of bacteria within the host and subsequent bacte-
Primary neonatal septicemia occurs in piglets lacking rial pathogenicity, in part by promoting bacterial resis-
immunity, due either to an absence of ingested tance to the bactericidal effects of phagocytosis (Ngeleka
colostrum or to ingestion of colostrum lacking specific et al. 1992, 1993, 1994).
antibody. The disease may develop after bacterial inva-
sion of the respiratory or the gastrointestinal tract in the CLINICAL SIGNS
nonimmune host. Contamination of the umbilicus after
birth may also lead to colisepticemia. However, the intes- Clinical signs of infection include depression, lameness,
tine is considered as a major route of E. coli invasion reluctance to move, anorexia, rough hair coat, and la-
since the disease can be experimentally induced by oral bored respiration (Nielsen et al. 1975a). The affected
or intragastric administration of the organisms (Ngeleka piglets may show sternal recumbency and the abdomen
et al. 1993). may be somewhat distended. Sometimes piglets become
Secondary septicemia may develop after invasion of unconscious, with convulsions and paddling move-
the host by enterotoxigenic E. coli (ETEC), but in most ments; they may be in good bodily condition but
cases development of primary neonatal septicemia is as- cyanosis of the extremities may be observed. Some
sociated with intestinal permeability to macromolecules, piglets are found dead while others are comatose without
to some defect of the immune system (e.g., low levels of any sign of diarrhea. These clinical signs may develop
maternal colostrum), to low birth weight, and to sub- within 12 hours after birth and piglets can die within 48
lethal malformations (Gyles 1986; Murata et al. 1979). hours (Taylor 1989). In older piglets, the clinical signs
Bacteria may pass through the mucosa of the alimen- may include periodic scouring or other ailments which
tary tract, probably by endocytic uptake into intestinal precede the onset of acute septicemia with clinical signs
epithelial cells or through the intercellular spaces formed resembling those in the newborn pigs.
by lateral plasma membranes of adjacent epithelial cells,
to locate in the mesenteric lymph nodes before entering LESIONS
the bloodstream. This bacterial invasion may result in a
generalized infection (septicemia, polyserositis) with In acute primary septicemia, there may be no gross le-
bacteria disseminated in different extraintestinal organs sions other than congestion of the intestine, the mesen-
such as lung, liver, spleen, kidney, brain, and heart teric lymph nodes, and the extraintestinal organs. In sub-
blood, or in a localized infection (meningitis or arthritis) acute cases, subserous or submucosal hemorrhages and
(Morris and Sojka 1985). In the sow, a puerperal sepsis fibrinous polyserositis with gross signs of pneumonia
may be induced by an enteric E. coli soon after farrowing are usually observed, often accompanied by fibrinopuru-
(Sojka 1965). lent arthritis and meningitis (Morris and Sojka 1985;
Septicemia may be produced experimentally in Waxler and Britt 1972). Histological examination of the
colostrum-deprived piglets by intragastric inoculation lung reveals interalveolar interstitial pneumonia with
with isolates of porcine origin (Fairbrother and Ngeleka edema and neutrophilic infiltration, but alveoli are free
1994; Ngeleka et al. 1993). However, the disease can also of exudates. In secondary septicemia resulting from en-
be reproduced in piglets with isolates from other sources, teric colibacillosis, icterus, petechial hemorrhages in the
such as calves, cats, and poultry (Fairbrother et al. 1993; serosal membranes, and splenomegaly accompanied by
Murata et al. 1979; Meyer et al. 1971). Animals may de- severe diarrhea and dehydration can be observed in
velop fever, anorexia, diarrhea, dyspnea, or nervous some cases (Svendsen et al. 1975). In many cases of sec-
456 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

ondary systemic E. coli infection, presumably occurring mation of E. coli infection and antibiotic sensitivity test-
at a very late stage in the underlying disease, the changes ing. Meanwhile, parenteral or oral administration of
are slight or no lesions at all are recorded. broad-spectrum antibiotics to affected piglets is recom-
mended while waiting for results of diagnostic lab tests.
DIAGNOSIS This treatment may be useful in subacute cases of infec-
tion but is mostly ineffective after the appearance of clin-
Systemic colibacillosis is generally suspected with the ap- ical signs. However, the remaining piglets in the litter
pearance of the clinical signs described above. However, and affected piglets and littermates in adjacent litters
in the case of polyserositis, a differential diagnosis be- should be treated.
tween Mycoplasma hyorhinis and Hemophilus parasuis has
to be made. In the former infection, gross lesions can be REFERENCES
detected more than 6 days after infection. Mortality is
lower than in E. coli infection. In polyserositis due to E. Contrepois, M.; Fairbrother, J. M.; Kaura, Y. K.; and Gi-
coli, the exudates encountered in piglets are serofibri- rardeau, J. P. 1989. Prevalence of CS31A and F165 sur-
nous or fibrinopurulent, whereas in Hemophilus parasuis face antigens in Escherichia coli isolates from animals in
they tend to be serofibrinous. In the central nervous sys- France, Canada, and India. FEMS Microbiol Letts
tem and joints, these exudates are fibrinopurulent to pu- 59:319–324.
rulent (Waxler and Britt 1972). Infection due to H. para- Crosa, J. H. 1989. Genetics and molecular biology of
suis is rarely seen in the early suckling period but is more siderophore-mediated iron transport in bacteria. Micro-
common in piglets of 2–3 months of age. However, dif- biol Rev 53:517–530.
ferential diagnosis can be established after a careful mi- Dozois, C. M.; Clement, S.; Desautels, C.; and Fairbrother, J.
crobiological examination. In most of the cases, post- M. 1997. Expression of P, S, and F1C adhesins by cyto-
mortem examination and bacteriology are useful for toxic necrotizing factor I (CNF1)–producing Escherichia
identifying the infection. Diagnosis of primary systemic coli from septicaemic and diarrheic pigs. FEMS Microbi-
colibacillosis is strengthened by the isolation in pure cul- ol Letts (in press).
ture or by the predominance of E. coli in extraintestinal Fairbrother, J. M., and Ngeleka, M. 1994. Extraintestinal Es-
tissues, particularly E. coli of one of the above-men- cherichia coli infections in pigs. In Escherichia coli in Do-
tioned serogroups or, more important, possessing one or mestic Animals and Humans. Ed. C. L. Gyles. Walling-
more of the virulence factors, such as adhesins of the P, ford, U.K.: CAB International, pp. 221–236.
S, or F1C families, serum resistance, or production of Fairbrother, J. M.; Larivière, S.; and Lallier, R. 1986. New
aerobactin. Diagnosis of secondary systemic colibacillo- fimbrial antigen F165 on Escherichia coli serogroup O115
sis is strengthened by the isolation in pure culture or by strains isolated from piglets with diarrhea. Infect Im-
the predominance in extraintestinal tissues of E. coli pos- mun 51:10–15.
sessing one or more of the enterotoxins LT, STa, or STb Fairbrother, J. M.; Broes, A.; Jacques, M.; and Larivière, S.
and possibly one of the fimbrial adhesins associated with 1989. Pathogenicity of Escherichia coli O115:K“V165”
ETEC, particularly F4. strains isolated from pigs with diarrhea. Am J Vet Res
50:1029–1036.
PREVENTION AND TREATMENT Fairbrother, J. M.; Harel, J.; Forget, C.; Desautels, C.; and
Moore, J. 1993. Receptor binding specificity and patho-
Inadequate hygiene and poor environmental tempera- genicity of Escherichia coli F165-positive strains isolated
ture control increase the likelihood of infection. Thus, from piglets and calves and possessing pap related se-
prevention of infection should concentrate on reduction quences. Can J Vet Res 57:53–55.
or elimination of significant pathogenic E. coli popula- Gyles, C. L. 1986. Escherichia coli. In Pathogenesis of Bacter-
tions in the environment of the piglets and in providing ial Infections in Animals. Ed. C. L. Gyles and C. O.
a plentiful supply of colostrum at birth. Hygiene, espe- Thoen. Ames: Iowa State Univ Press, pp. 114–131.
cially washing and disinfection of the farrowing pens, Jesmok, G.; Lindsey, C.; Duerr, M.; Fournel, M.; and Emer-
will efficiently contribute to reduction of the infection. son, T. Jr. 1992. Efficacy of monoclonal antibody against
Young piglets should be maintained at an even tempera- human recombinant tumor necrosis factor in E.
ture of 35˚C for the first week of life. They must be kept coli–challenged swine. Am J Pathol 141:1197–1207.
dry and warm in clean surroundings. Affected piglets Meyer, R. C.; Saxena, S. P.; and Rhoades, H. E. 1971. Poly-
should be treated if necessary, and other litters of sus- serositis induced by Escherichia coli in gnotobiotic swine.
ceptible age watched. However, in the case of small out- Infect Immun 3:41–44.
breaks, careful monitoring of the causative serogroup(s) Morris, J. A., and Sojka, W. J. 1985. Escherichia coli as a
and autovaccination of the pregnant sows might be ben- pathogen in animals. In The Virulence of Escherichia
eficial. coli: Reviews and Methods. Ed. M. Sussman. London:
Treatment may be attempted after diagnostic confir- Academic Press, pp. 47–77.
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 457

Murata, H.; Yaguchi, H.; and Namioka, S. 1979. Relation- O115:K“V165” resist killing by porcine polymorphonu-
ship between the intestinal permeability to macromole- clear leukocytes in vitro: Role of F1651 fimbriae and
cules and invasion of septicemia-inducing Escherichia K “V165” O-antigen capsule. Infect Immun 62:398–
coli in neonatal piglets. Infect Immun 26:339–347. 404.
Nakajima, Y.; Ishikawa, Y.; Momotani, E.;, Takahashi, K.; Nielsen, N. C.; Bille, N.; Riising, H. J.; and Dam, A. 1975a.
Madarame, H.; Ito, A.; Ueda, H.; Wada, M.; and Taka- Polyserositis in pigs due to generalized Escherichia coli
hashi, H. 1991. A comparison of central nervous lesions infection. Can J Comp Med 39:421–426.
directly induced by Escherichia coli lipopolysaccharide in Nielsen, N. C.; Riising, H. J.; Larsen, J. L.; Bille, N.; and
piglets, calves, rabbits and mice. J Comp Pathol Svendsen, J. 1975b. Preweaning mortality in pigs: Acute
104:57–64. septicaemias. Nordisk Vet Med 27:129–139.
Ngeleka, M.; Harel, J.; Jacques, M.; and Fairbrother, J. M. Sojka, W. J. 1965. Escherichia coli in Domestic Animals and
1992. Characterization of a nonacidic polysaccharide Poultry. Farnham Royal, Bucks, England: Commonw
capsular antigen of septicemic Escherichia coli O115:K Agric Bur.
“V165”:F165 and evaluation of its role in pathogenicity. Svendsen, J.; Bille, N,; Nielsen, N. C.; Larsen, J. L.; and Riis-
Infect Immun 60:5048–5056. ing, H.-J. 1975. Preweaning mortality in pigs. 4. Diseases
Ngeleka, M.; Jacques, M.; Martineau-Doizé, B.; Harel, J.; of the gastrointestinal tract in pigs. Nord Vet Med
and Fairbrother, J. M. 1993. Pathogenicity of an Es- 27:85–101.
cherichia coli O115:K“V165” mutant negative for F1651 Taylor, D. J. 1989. Pig Diseases, 5th ed. Lennoxtown, Glas-
fimbriae in septicemia of gnotobiotic pigs. Infect Im- gow, pp. 171–172.
mun 61:836–843. Waxler, G. L., and Britt, A. L. 1972. Polyserositis and arthri-
Ngeleka, M.; Martineau-Doizé, B.; and Fairbrother, tis due to Escherichia coli in gnotobiotic pigs. Can J Comp
J. M. 1994. Septicemia-inducing Escherichia coli Med 36: 226–233.

COLIFORM MASTITIS
H. U. Bertschinger

The term “coliform mastitis” (CM) has been introduced hard to assess. The piglets suffer more than the affected
to denominate puerperal mastitis in the pig. The term dam. Bäckström et al. (1984) reported that the mortality
should end the confusing terminology in this field. In ad- of piglets nursing multiparous sows with MMA was
dition, this expression points out the parallels to CM in 55.8%, whereas that of piglets nursing healthy sows was
the cow. Other terms used to denominate CM and relat- 17.2%. The corresponding figures determined by Madec
ed conditions are discussed in Chapter 58 of this book. A et al. (1992) from 286 parturitions were 21% and 17%, re-
cumulative tabulation of necropsies performed on spectively. Mortality of piglets may result from length-
agalactic sows revealed that 59 of 72 sows (82%) had ened farrowing time, crushing by the sow, starvation,
gross lesions of mastitis (Ross et al. 1981). and impaired immunity to infectious agents because of
CM has a worldwide distribution. Hermansson et al. insufficient uptake of colostral immunoglobulins. The
(1978) disclosed an average incidence of agalactia post- average milk yield of three sows affected with CM on the
partum of 12.8%, with a variation among individual first 2 days after farrowing was about half the yield of
herds from 0.5% to 50%. In a Danish study covering more healthy sows, and the piglets of the sick sows lost some
than 72,000 farrowings on farms with a high manage- weight (Ross et al. 1975). Piglets sucking glands with
ment level, the incidence amounted to 9.5% of the far- mastitis of sows with subclinical CM had smaller weight
rowings (Jorsal 1986). In these studies, the true inci- gains only for days 1–4 postpartum (Bertschinger et al.
dence of CM was not determined. Wegmann et al. 1990).
(1986) examined samples of colostrum from 59 sows in
15 herds with a so-called mastitis-metritis-agalactia ETIOLOGY
(MMA) problem. No fewer than 83% of the sows were af-
fected with mastitis in at least one mammary complex. E. The term “coliform,” when used in the context of masti-
coli or Klebsiella pneumoniae were isolated from 79% of tis, covers the bacterial genera Escherichia, Klebsiella, En-
the 131 complexes with mastitis. terobacter, and Citrobacter. However, the methods used
Economic loss results from a number of factors and for identification in several studies were not adequate to
therefore is difficult to estimate. The death rate of affect- determine the genera and even less the species of the
ed sows is low. The cost of extra care and of treatment is bacterial isolates. E. coli was the organism most often
458 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

identified in either milk samples or affected mammary takes place. McDonald and McDonald (1975) found sig-
tissue (Ringarp 1960; Armstrong et al. 1968; nificant numbers of coliform bacteria in about one-
Bertschinger et al. 1977a; Ross et al. 1981; Wegmann et fourth of mammary glands cultured just before parturi-
al. 1986). Klebsiella, mostly K. pneumoniae, was prevalent tion. In a sequential examination of the mammary
in the cases investigated by Ross et al. (1975) and Jones secretions, E. coli was isolated from 30 glands. In 17 of
(1976). Staphylococcus epidermidis and a variety of strep- these glands, the bacteria were detected before the first
tococci were also found in the mammary glands of sows piglet was born (Bertschinger et al. 1990). New infec-
with signs of mastitis, either mixed with coliforms or as tions appeared not later than day 2 postpartum.
pure cultures. The noncoliform organisms were only ex- The bacteria are located in the ductular and alveolar
ceptionally associated with microscopic lesions of masti- lumina, either free or within phagocytic cells. Adhesion
tis (Bertschinger et al. 1977a; Ross et al. 1981). to surfaces is not prominent. At postmortem examina-
tion the causative bacteria are frequently isolated from
EPIDEMIOLOGY regional lymph nodes, whereas isolations from liver,
spleen, or kidney are rare (Armstrong et al. 1968;
CM of the sow appears to be noncontagious. Serologic Bertschinger et al. 1977a, b; Ross et al. 1981).
typing of isolates from mastitis revealed an extreme mul- Multiplication of bacteria in the mammary secretion
tiplicity of serologic types not only within a herd but al- is controlled by antimicrobial mechanisms, which so far
so within distinct glands of one sow. A significant pro- have been studied in the pig with respect to piglet enteri-
portion of subcomplexes harbor more than one type tis only. The antimicrobial activity of cow’s milk is due to
(Bertschinger et al. 1977a; Awad-Masalmeh et al. 1990). a variety of inhibitors acting in concert and conferring
The great variety of coliform bacteria associated with on the dry udder a nearly total resistance to coliform pro-
CM indicates an abundant reservoir of potentially path- liferation (Bramley 1976). Growth in vitro of a given
ogenic bacteria. The coliforms causing mastitis may orig- strain of E. coli in secretions of individual sows varies
inate from the flora of the sow as well as from the envi- enormously. The secretion of atrophic glands exerts a
ronment. In about one-third of the sows with mastitis, bactericidal effect (Wegmann 1985). CM is a self-curing
identical isolates were found in mastitic glands, the uter- disease. The bacteria generally disappear between 1 and
ine contents, and the urinary bladder (Bertschinger et al. 6 days after parturition (Wegmann and Bertschinger
1977a). The intestinal flora of the sow, the oral flora of 1984; Bertschinger et al. 1990). In severe cases, however,
the neonatal piglet, and environmental bacteria may sig- they persist in necrotic foci throughout lactation (Löpfe
nificantly contribute to contamination of the nipples. 1993).
Awad-Masalmeh et al. (1990) found identical O
serogroups of E. coli in mammary secretion and in feces Mammary Inflammation
of about one-fourth of 67 sows with CM. Muirhead CM in the sow is associated with massive accumulation
(1976) considered the bedding of the sow of paramount of neutrophils in the lumina of affected glands. Simulta-
importance. Dung and urine contaminate the udder. neous induction of CM in several mammary subcom-
Klebsiella spp. may also originate from wood shavings plexes of sows from which the piglets had been removed
used for bedding. Bertschinger et al. (1990) compared 12 resulted in severe leukopenia within 24 hours
farrowings each in conventional farrowing crates and in (Bertschinger et al. 1977b). Intracisternal instillation of
an experimental pen where the sows could lie down in a identical bacterial inocula following a highly standard-
clean resting area. Sows in the experimental pen had ized protocol led to a spectrum of reactions ranging from
much lower counts of coliform bacteria on their teat very severe local and general signs to subclinical mastitis
ends and an incidence of intramammary E. coli infec- (Löpfe 1993; Mossi 1995). Severe reaction is the conse-
tions 10 times lower than that of the sows in the crates. quence of massive and persistent multiplication of inoc-
ulated bacteria. In the experiments of Löfstedt et al.
PATHOGENESIS (1983) susceptibility to experimental infection was asso-
ciated with impaired function of circulating neutrophils.
Invasion of the Mammary Gland The cause of the impaired neutrophil function is still a
Mastitis was reproduced in the sow by intramammary mystery. Cytological findings in the secretion must be in-
instillation of not more than 120 organisms of a strain of terpreted with caution. Mammary glands not chosen by
K. pneumoniae. Following massive external contamina- a piglet undergo involution soon after parturition. Invo-
tion of the nipples with the same strain, the bacteria lution is accompanied by an increase in the total somatic
were recovered from 60 out of 142 subcomplexes exam- cell count as well as in the proportion of polymorphonu-
ined. External contamination of the nipples was as suc- clear (PMN) cells (Wegmann and Bertschinger 1984). In
cessful on gestation day 111 as 2 hours after completion some sows many glands show increases of total somatic
of farrowing (Bertschinger et al. 1977b). It is largely un- and of PMN cells in the absence of cultivable bacteria
known at what time spontaneous invasion of the cistern (Bertschinger et al. 1990).
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 459

Systemic Reaction floor. If access to the nipples is given by the sow, the pe-
The systemic signs of CM are brought about by the bac- riods of suckling are shortened. After suckling, the
terial endotoxin. An outline of the systemic changes is piglets stray about instead of resting in close contact
given in Chapter 58. CM in the absence of systemic reac- with their littermates.
tion is often revealed by methodical examination of Precise localization of mammary lesions is often not
mammary secretion (Wegmann 1985; Bertschinger et al. possible because reddening and heat of the skin extend
1990; Persson et al. 1996b). over several subcomplexes. The reliable clinical assess-
ment of the state of the actual mammary tissue is ren-
Immunity dered difficult by subcutaneous fat and considerable sub-
CM apparently does not result in protection against ho- cutaneous edema. If palpable, the mastitic tissue is
mologous reinfection (Bertschinger and Bühlmann firmer and palpation may cause pain. The red color of
1990). Ringarp (1960) reported a higher incidence in the skin is blanched by finger pressure, which causes a
sows than in gilts, as well as repeated occurrence in indi- depression of the tissue lasting for some time. Mere clin-
vidual sows of up to 10 times. Vaccination is not a ical examination will at best detect some of the affected
promising method for control of mastitis. Even when an subcomplexes (Persson et al. 1996b). The inguinal
autogenous vaccine was used, protection was unsatisfac- lymph nodes may be swollen.
tory (R. Ross, Ames, Iowa, unpublished data—1983). The fluid expressed from a nipple originates from
In the cow, there is considerable evidence that vacci- more than one subcomplex, because two or, rarely, three
nation with an R-mutant of E. coli results in a dramati- teat canals end in each nipple. Therefore, in samples tak-
cally reduced incidence of CM (Tyler et al. 1993). Vacci- en from a nipple, secretion from the unaffected, produc-
nation has little impact on the frequency of new tive subcomplex dominates. The exudate from inflamed
infections but decreases the incidence of overt disease. subcomplexes looks serous to creamy, like pus. It may
No reports have appeared so far on the efficacy of such contain clots of fibrin or blood. The pH is of limited di-
vaccines in the sow. agnostic value (Ross et al. 1981; Persson et al. 1996b), but
cytological examination allows differentiation between
CLINICAL SIGNS healthy and mastitic complexes at least during the first
48 hours after parturition (Wegmann and Bertschinger
Ross et al. (1975) described the clinical findings in sows 1984). Because mastitis is a local process, samples must
with proven CM and demonstrated changes quite similar be taken from individual complexes and not pooled. The
to those described earlier in sows with lactational failure. threshold value of the total cell count varies depending
Interpretation of clinical parameters is rendered difficult on the investigator. Bertschinger et al. (1990) suggested
by the presence of subclinical CM in apparently healthy 5 × 106 cells per mL and fewer than 70% PMN. In a recent
sows (Nachreiner and Ginther 1972; Middleton- study, Persson et al. (1996a) proposed 10 × 106 cells per
Williams et al. 1977; Persson et al. 1996b). mL. However, these authors did not distinguish between
The initial signs are most often detected on the first sucked and unsucked glands. Involution of some glands
or second day and more rarely on the third day after far- starts as early as 1 day after parturition (Wegmann and
rowing. However, they may be observed as early as dur- Bertschinger 1984). It leads to a significant increase of
ing parturition (Martin et al. 1967). The first symptoms the total cell count accompanied by a transient increase
are temperature response, listlessness, weakness, and of up to 60% of the proportion of PMN cells. As a con-
loss of interest in piglets. Affected sows prefer sternal re- sequence, cytological distinction between involution and
cumbency. In severe cases they become stiff and dizzy, mastitis may be difficult or impossible between 2 and 7
do not stand up, and may even become comatose. Con- days postpartum (Wegmann 1985). Bacteriological ex-
sumption of feed and water is either reduced or absent. amination of the secretion may be necessary in unclear
Body temperature is moderately elevated and only rarely cases. Infection persisting for several days is limited to
exceeds 42˚C. Afebrile cases have been reported; howev- severely affected complexes (Löpfe 1993).
er, the temperature was not taken continuously. On the
other hand, many normal sows will have rectal tempera- LESIONS
tures that exceed the 39.7˚C limit on the day of parturi-
tion and for 2 days thereafter (King et al. 1972). In af- Despite the high incidence of CM, there are not many re-
fected sows respiratory and heart rates are increased. In ports of necropsy findings (Martin et al. 1967; Jones
general the symptoms described do not last for more 1976; Middleton-Williams et al. 1977; Ross et al. 1981).
than 2–3 days. In general, lesions are confined to the mammary glands
The behavior of the piglets is very helpful in the early and regional lymph nodes. The subcutaneous tissue may
detection of lactational failure. Undernourished piglets be edematous over affected parts of the udder. For reli-
look gaunt. They frequently try to suck, move from nip- able demonstration of mastitis, Middleton-Williams et
ple to nipple, nibble at litter, and lick urine from the al. (1977) recommended a longitudinal section at the lev-
460 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

el of the nipples through each row of glands. Using this By histological examination additional lesions were
technique, irregularly scattered foci of mastitis were de- detected that had not been recognized at gross examina-
tected in 1–23 subcomplexes (Fig. 32.9). The appearance tion. In every case there was an acute purulent exudative
of affected mammary tissue varied from slightly in- mastitis with congestion. An extreme variability in sever-
creased firmness and grayish discoloration to sharply de- ity ranging from a small number of neutrophils in the
marcated, red-mottled, hard, and dry areas (Fig. 32.10). alveolar lumina to severe purulent infiltration with
The secretion was sparse and sometimes mixed with necrosis was obvious (Fig. 32.11). The severity of the le-
clots. sions varied not only between but also within subcom-
plexes, where unaffected tissue was found adjacent to se-
verely inflamed areas. Acute purulent lymphadenitis was
present in the inguinal and iliac lymph nodes (Middle-
ton-Williams et al. 1977). A sequential study of the mi-
croscopic lesions consequent to experimental intracister-
nal inoculation revealed in severe cases the persistence
throughout lactation of abscesslike large necrotic foci
surrounded by granulomatous connective tissue (Löpfe
1993). A predilection of microscopic lesions for certain
areas within a given complex was not evident. The mu-
cosa lining the cisternae was not affected.

DIAGNOSIS

Any hypogalactia at the beginning of lactation arouses


suspicion of CM. The diagnosis may be supported by
fever, anorexia, reluctance to stand up, lying on the
gland, and disinterest in the piglets. In severe cases some
affected glands may be reddened, swollen, and firm, and
the secretion may look abnormal. A reliable rapid test for
use on the farm is not available. Due to the higher cell
content of sow milk, tests developed for use with the cow
cannot be recommended. Bacteriological and cytological
examinations of the secretion make sense only if all
32.9. Distribution and intensity of histologic lesions in glands are sampled or if affected glands are known.
nine field cases of CM. (Modified from Middleton- The differential diagnosis of CM is reviewed in Chapter
Williams et al. 1977.) 58.

32.10. Acute CM of one subcom-


plex adjacent to unaltered glandular
tissue in longitudinal cut surface: (a)
subcutaneous edema, (b) demarcation
from adjacent subcomplex, (c) mot-
tled appearance of affected tissue.
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 461

32.11. Histopathology of a mammary gland 24 hours after experimental inoculation with a


culture of Klebsiella pneumoniae. (A) Low-power magnification within a single subcomplex
showing different types of mastitis next to secreting alveoli. (B) Dark area filled with densely
packed polymorphonuclear leukocytes and local destruction of epithelium. (C) Nearly empty-
looking alveoli containing low numbers of polymorphonuclear leukocytes. (D) Alveoli with
normal-looking secretion. (Institute of Veterinary Pathology, University of Zurich.)

TREATMENT er than the MIC (Oliel and Bertschinger 1990). Awad-


Masalmeh et al. (1990) tested 107 strains of E. coli isolat-
Therapeutic measures are usually not taken before the ed from sows with CM from 43 herds and found no re-
sow shows signs of dysgalactia. Thus, treatment may at sistance to enrofloxacin (Table 32.6). Therapeutic trials
best shorten the period of underfeeding of the piglets. are generally difficult to evaluate because the curing ef-
Chemotherapy is complicated by the heterogeneous fect is not quantified and often hardly distinguishable
pattern of antimicrobial susceptibility of individual iso- from spontaneous improvement. Options for supportive
lates not only within a herd but also within a sow. There- therapy are discussed in Chapter 58.
fore, sensitivity testing is of little value in individual cases. Much attention should be given to the piglets. They
The pharmacokinetics of antimicrobials has received may either be fostered by other sows or remain with their
only limited attention. One injection of 20 mg/kg body mother and receive a milk substitute. Sweetened con-
weight of a slow-release formulation of oxytetracycline densed milk diluted with water 1:1 can be used instead of
results in milk levels not surpassing 2 µg/mL, that is, just commercial products. Sterile 5% glucose solution in a
above the minimum inhibitory concentration (MIC) of dose of approximately 15 mL can be repeatedly injected
susceptible E. coli (Schoneweis et al. 1982). Enrofloxacin, intraperitoneally several hours apart, or a more concen-
a quinolone antibiotic, given at 2.5 mg/kg body weight trated solution may be applied intragastrically. When
orally every half day, is concentrated in colostrum and the pig obtains insufficient amounts of milk, protection
milk to mean levels of 1.2 µg/mL, which is 20 times high- against chilling is particularly important.
462 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Table 32.6. Sensitivity to antimicrobials of coliform isolates from mammary glands in Switzerland and Austria
Sensitive Isolates (%)
Bertschinger et al. 1977a Wegmann et al. 1986 Awad-Masalmeh et al. 1990
Substance (n = 80) (n = 107) (n = 107)
Ampicillin 90 86 74
Tetracycline 19 42 16
Chloramphenicol 95 81 64
Streptomycin 21 21 21
Neomycin 96 92 86
Gentamicin 100 100 100
Trimethoprim plus 100 84 51
sulfamethoxazole
Enrofloxacin not tested not tested 100

PREVENTION tion in sows against intramammary challenge with the


same strain used to prepare the bacterin (R. Ross, Ames,
Hygiene Measures Iowa, unpublished data—1982).
Muirhead (1976) and Jones (1979) suggested that pro-
tection of the teats from bacterial contamination might Hormones
be an effective prophylaxis of CM. Bertschinger et al. Some investigators found an extended period of gesta-
(1990) performed a prospective study of farrowing in tion in sows developing lactational failure. The length of
two types of pens. They concluded that the density of gestation can be controlled by use of prostaglandins.
the coliform flora on the teat apex reflects the degree of However, the prophylactic application led to conflicting
contamination of the lying area. Optimal prophylaxis is results. Studies focusing on CM are not known.
achieved by designing farrowing accommodation in such
a manner that the sow is prevented from lying down in Chemoprophylaxis
her own excreta. On the other hand, washing and disin- For the time being, chemoprophylaxis appears to be the
fection of the pen and of the newly housed sow con- most promising method of control where accommoda-
tribute much less to efficient prophylaxis. If cases of CM tion cannot be improved. The prevalence of drug resis-
accumulate, the bedding materials should be checked. tance (Table 32.6) and the wide variety of bacteria asso-
ciated with the disease in a given herd must be
Nutrition of the Sow considered when the drug is selected. Feed medication
Drastic reduction of the sow’s ration shortly before par- should be replaced by individual application of the drug
turition is a widespread practice. In a carefully designed in a small amount of feed because the feed consumption
long-term study using pairs of full sibs, the reduction of of the sow in the periparturient period is quite variable.
the daily feed allowance from 3.2 to 1.0 kg of a commer- Keeping the period of treatment as short as possible
cial-type feed lowered the incidence of agalactia from helps to postpone the emergence of drug resistance. In
26.6% to 14.4% (Persson et al. 1989). Udder changes were field trials, the morbidity from MMA was reduced from
registered in a high percentage of the agalactic sows, and 30% to 12% by giving 0.4 g trimethoprim, 1 g sulfadimi-
significant numbers of bacteria were found in more than dine, and 1 g sulfathiazole/150 kg body weight twice a
80% of the agalactic sows. The two feed regimens did not day. The treatment began on gestation day 112 and last-
influence the total cell count, the rate of PMN, or the pH ed for 4 days regardless of the day of farrowing (Boll-
of the secretion (Persson et al. 1996b). Plonait et al. wahn 1978). Six intramuscular injections of apramycin
(1986) reported a corresponding observation. Experi- (6.25 mg/kg) at 12-hour intervals reduced the severity of
mentally induced CM takes a similar course with sows on experimentally induced mastitis (Ross and Zimmer-
high and low rations (H. U. Bertschinger and A. mann 1982).
Bühlmann, unpublished data). This finding led to the Oliel and Bertschinger (1990) tested oral chemopro-
suggestion that feed reduction might act through re- phylaxis with enrofloxacin on experimentally induced
duced exposure of the teats due to the much smaller CM. Three glands of each sow were inoculated with E.
amounts of feces and urine contaminating the lying area. coli and three glands with K. pneumoniae. Eight sows were
not treated (treatment I), eight sows received 2.5 mg
Immunoprophylaxis (treatment II), and eight sows received 5.0 mg/kg body
Induction of specific immunity is hampered by the wide weight twice a day (treatment III). The inoculated bacte-
range of antigenic types of coliforms isolated from sows ria were reisolated in treatment I from 100%, in treat-
with CM. Use of an E. coli bacterin induced poor protec- ment II from 10%, and in treatment III from 2% of the in-
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 463

oculated glands. A beneficial effect on milk productivity Löpfe, P. J. 1993. Experimentelle Mastitis bei der Sau: Kor-
would be expected but could not be demonstrated be- relation der pathologisch-anatomischen und histologis-
cause the control sows did not develop systemic signs. chen Befunde mit den klinischen Befunden 4–30 Tage
nach der Ansteckung mit E. coli und Klebsiella pneumoni-
REFERENCES ae. DVM thesis, Zurich.
Löfstedt, J.; Roth, J. A.,; Ross, R. F.; and Wagner, W. C. 1983.
Armstrong, C. H.; Hooper, B. E.; and Martin, C. E. 1968. Mi- Depression of polymorphonuclear leukocyte function
croflora associated with agalactia syndrome of sows. associated with experimentally induced Escherichia coli
Am J Vet Res 29:1401–1407. mastitis in sows. Am J Vet Res 44:1224–1228.
Awad-Masalmeh, M.; Baumgartner, W.; Passernig, A.; Sil- Madec, F.; Miquet, J. M.; and Léon, E. 1992. La pathologie
ber, R.; and Hinterdorfer, F. 1990. Bakteriologische Un- de la parturition chez la truie: Étude épidémiologique
tersuchungen bei an puerperaler Mastitis (MMA-Syn- dans cinq élevages. Rec Méd Vét 168:341–349.
drom) erkrankten Sauen verschiedener Tierbestände Martin, C. E.; Hooper, B. E.; Armstrong, C. H.; and Amstutz,
Österreichs. Tierärztl Umschau 45:526–535. H. E. 1967. A clinical and pathologic study of the masti-
Bäckström, L.; Morkoc, A. C.; Connor, J.; Larson, R.; and tis-metritis-agalactia syndrome of sows. J Am Vet Med
Price, W. 1984. Clinical study of mastitis-metritis- Assoc 151:1629–1634.
agalactia in sows in Illinois. J Am Vet Med Assoc McDonald, T. J., and McDonald, J. S. 1975. Intramammary
185:70–73. infections in the sow during the peripartum period. Cor-
Bertschinger, H. U., and Bühlmann, A. 1990. Absence of nell Vet 65:73–83.
protective immunity in mammary glands after experi- Middleton-Williams, D. M; Pohlenz, J.; Lott-Stolz, G.; and
mentally induced coliform mastitis. Proc Int Congr Pig Bertschinger, H. U. 1977. Untersuchungen über das
Vet Soc 11:175. Mastitis-Metritis-Agalaktie-Syndrom (Milchfieber) der
Bertschinger, H. U.; Pohlenz, J.; and Hemlep, I. 1977a. Un- Sau. I. Pathologische Befunde bei Spontanfaellen.
tersuchungen über das Mastitis-Metritis-Agalaktie-Syn- Schweiz Arch Tierheilkd 119:213–222.
drom (Milchfieber) der Sau. II. Bakteriologische Be- Mossi, R. 1995. Experimentelle Kolimastitis bei der Sau:
funde bei Spontanfaellen. Schweiz Arch Tierheilkd Korrelation der pathologisch-anatomischen und histol-
119:223–233. ogischen mit den klinischen Befunden 6–72 Stunden
Bertschinger, H. U.; Pohlenz, J.; and Middleton-Williams, D. nach der Inokulation. DVM thesis, Zurich.
M. 1977b. Untersuchungen über das Mastitis-Metritis- Muirhead, M. R. 1976. Veterinary problems of intensive pig
Agalaktie-Syndrom (Milchfieber) der Sau. III. Galakto- husbandry. Vet Rec 99:288–292.
gene Erzeugung von Klebsiellen-Mastitis. Schweiz Arch Nachreiner, R. F., and Ginther, O. J. 1972. Porcine agalactia:
Tierheilkd 119:265–275. Hematologic, serum chemical and clinical changes dur-
Bertschinger, H. U.; Bürgi, E.; Eng, V.; and Wegmann, P. ing the preceding gestation. Am J Vet Res 33:799–809.
1990. Senkung der Inzidenz von puerperaler Mastitis Oliel, N., and Bertschinger, H. U. 1990. Prophylaxis of ex-
bei der Sau durch Schutz des Gesäuges vor Ver- perimentally induced coliform mastitis in the sow with
schmutzung. Schweiz Arch Tierheilkd 132:557–566. enrofloxacin (BAYTRIL®). Proc Int Congr Pig Vet Soc
Bollwahn, W. 1978. Effect of strategic TMP/S treatment on 11:186.
puerperium and conception in sows. Proc Int Congr Pig Persson, A.; Pedersen, A. E.; Göransson, L.; and Kuhl, W.
Vet Soc 5:KA16. 1989. A long term study on the health status and perfor-
Bramley, A. J. 1976. Variations in the susceptibility of lac- mance of sows on different feed allowances during late
tating and non-lactating bovine udders to infection pregnancy. I. Clinical observations, with special refer-
when infused with Escherichia coli. J Dairy Res ence to agalactia post partum. Acta Vet Scand 30:9–17.
43:205–211. Persson, A.; Pedersen Mörner, A.; and Kuhl, W. 1996a. A
Hermansson, I.; Einarsson, S.; Larsson, K.; and Bäckström, long term study on the health status and performance of
L. 1978. On the agalactia postpartum in the sow: A clin- sows on different feed allowances during late pregnancy.
ical study. Nord Vet Med 30:465–473. II. The total cell content and its percentage of polymor-
Jones, J. E. T. 1976. Bacterial mastitis and endometritis in phonuclear leucocytes in pathogen-free colostrum and
sows. Proc Int Congr Pig Vet Soc 4:E6. milk collected from clinically healthy sows. Acta Vet
———. 1979. Acute coliform mastitis in the sow. Vet Annu Scand 37:279–291.
19:97–101. ———. 1996b. A long term study on the health status and
Jorsal, S. E. 1986. Epidemiology of the MMA-syndrome, a performance of sows on different feed allowances dur-
field survey in Danish sow herds. Proc Int Congr Pig Vet ing late pregnancy. III. Escherichia coli and other bacte-
Soc 9:93. ria, total cell content, polymorphonuclear leucocytes
King, G. J.; Willoughby, R. A.; and Hacker, R. R. 1972. Fluc- and pH in colostrum and milk during the first 3 weeks of
tuations in rectal temperature of swine at parturition. lactation. Acta Vet Scand 37:293–313.
Can Vet J 13:72–74. Plonait, H.; Kump, A. W.-S.; and Schöning, G. 1986. Pro-
464 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

phylaxis of the MMA-syndrome by antibacterial med- Levels of oxytetracycline in plasma and milk in pig. Proc
ication and restricted feeding. Proc Int Congr Pig Vet Soc Int Congr Pig Vet Soc 7:291.
9:96. Tyler, J. W.,; Cullor, J. S.; and Ruffin, D. C. 1993. Immuniza-
Ringarp, N. 1960. Clinical and experimental investigation tion and immunotherapy for mastitis. Vet Clinics N Am:
into a post-parturient syndrome with agalactia in sows. Food Anim Pract 9:537–549.
Acta Agric Scand Suppl 7. Wegmann, P. 1985. Zur Pathogenese der Colimastitis beim
Ross, R. F., and Zimmermann, B. J. 1982. Control of Es- Mutterschwein. DVM thesis, Zurich.
cherichia coli–induced mastitis in sows with apramycin. Wegmann, P., and Bertschinger, H. U. 1984. Sequential cy-
Proc Int Congr Pig Vet Soc 7:14. tological and bacteriological examination of the secre-
Ross, R. F.; Zimmermann, B. J.; Wagner, W. C.; and Cox, D. tions from sucked and unsucked mammary glands with
1975. A field study of coliform mastitis in sows. J Am Vet and without mastitis. Proc Int Congr Pig Vet Soc 8:287.
Med Assoc 167:231–235. Wegmann, P.; Bertschinger, H. U.; and Jecklin, H. 1986. A
Ross, R. F.; Orning, A. P.; Woods, R. D.; Zimmermann, B. J.; field study on the prevalence of coliform mastitis
Cox, D. F.; and Harris, D. L. 1981. Bacteriologic study of (MMA) in Switzerland and the antimicrobial suscepti-
sow agalactia. Am J Vet Res 42:949–955. bility of the coliform bacteria isolated from the milk.
Schoneweis, D. A.; Hummels, S.; and Schulteis, L. 1982. Proc Int Congr Pig Vet Soc 9:92.

URINARY TRACT INFECTION


H. U. Bertschinger
Urinary tract infection (UTI) is present whenever any of ETIOLOGY
the typically sterile sections of the urinary tract are colo-
nized by microbes. UTI may or may not be accompanied Stirnimann and Tschudi (1985) gave a description of
by clinically manifest or subclinical disease. In the postmortem bacteriological findings in 12 sows with
pig, specific UTI caused by Actinobaculum (formerly acute urinary tract disease. A. suis was diagnosed in 9
Actinomyces) suis (Chapter 63) is distinguished from non- sows, 3 of them monoinfected, by means of an immuno-
specific UTI caused by a variety of microbes and dealt fluorescence technique. Two or three bacterial species
with in this chapter. According to Liebhold et al. (1995), were isolated from the 9 sows not monoinfected: E. coli (7
a nonspecific UTI often paves the way for A. suis. sows), Streptococcus sp. (5 sows), Staphylococcus epider-
Severe acute UTI is important to the veterinarian car- midis (2 sows), Klebsiella sp. (1 sow), Pseudomonas sp. (1
ing for breeding stock (Stirnimann 1984), and UTI is the sow), Aeromonas sp. and Bacteroides sp. (1 sow). Carr and
predominant cause of death in pigs over 1 year of age Walton (1993) reported that A. suis was detected in 21
(Häni et al. 1976). In a survey of culled sows, significant out of 23 cases of pyelonephritis. It was only exception-
bladder colonization was detected in 17% of the sows, ally present as a monoinfection.
and 80% of the colonized bladders exhibited histological Urine was obtained by catheter from 90 sows with se-
lesions of cystitis (Colman et al. 1988). vere acute urinary tract disease (Stirnimann 1984). A. su-
Many authors have suggested a relationship between is was again looked for by immunofluorescence but was
bacteriuria and reproductive disorders, including masti- detected in only 10 sows. Thirty-seven sows were
tis-metritis-agalactia (MMA). Sows developing MMA monoinfected. The bacterial spectrum was comparable
have a much higher prevalence of UTI in the preceding to that described above. In a German study, 13 out of 25
gestation period than sows with a normal puerperium sows with a significant bacteriuria but no pyelonephritis
(Miquet et al. 1990). According to Petersen (1983), ex- were infected with enterobacteria and streptococci but
amination of urine in late pregnancy allows recognition were free from A. suis (Liebhold et al. 1995).
of sows at risk to develop MMA at subsequent farrow- In aspirates taken at a slaughterhouse from 114 sow
ing. However, this view is not without controversy. Simi- bladders with significant colonization, Colman et al.
lar prevalences of UTI in herds with and without MMA (1988) identified E. coli (81 sows), diverse gram-negative
were reported by Becker et al. (1985). Potential patho- rods (4 sows), S. aureus (11 sows), S. hyicus (3 sows),
genetic relations between UTI and MMA were discussed E. faecalis (6 sows), E. faecium (3 sows), S. dysgalactiae
in detail by Berner (1988). (5 sows), and diverse gram-positive bacteria (7 sows).
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 465

A. suis was not detected by anaerobic culture. These fig- vesicoureteric reflux. The latter could be easily demon-
ures suggest a high prevalence of A. suis only in sows strated postmortem in cases of acute pyelonephritis.
with severe urinary tract disease. Furthermore, nonspe- Serum antibody against the infecting E. coli strain can
cific UTI cannot be regarded as a disease exclusively regularly be detected in sows with pyelonephritis, less of-
caused by E. coli. ten in sows with cystitis, and rather rarely in sows with
The urinary tract is a dynamic microbiological asymptomatic bacteriuria (Wagner 1990). E. coli strains
ecosystem. Dominant bacterial species change sponta- may persist in the urinary tract despite high antibody
neously in a significant proportion of sows surveyed over concentrations in the urine.
prolonged periods (Berner 1990). These changes become UTI predisposes to MMA in one or several ways
more frequent when sows are treated with antimicro- (Berner 1988). Ascending invasion of the uterus at par-
bials. turition and of the mammary glands from contamina-
tion of the lying area appears most likely. However, oth-
EPIDEMIOLOGY er routes cannot be ruled out. Identical OK serotypes of
E. coli were found in the urinary bladders and in the uteri
Nonspecific UTI behaves like a noncontagious infectious of 3 sows and in the bladder and mammary gland of 1
disease of endogenous origin. In the canine species, E. out of 9 sows with MMA killed for postmortem exami-
coli isolates from urine and from rectal samples of the nation (Bertschinger et al. 1977).
same dog show identity in extended phenotypic and
genotypic tests (Low et al. 1988). Corresponding studies CLINICAL SIGNS
with the pig are lacking. The fecal flora may achieve ac-
cess to the urinary tract more efficiently in females than In the vast majority of nonspecific UTI cases there are no
in males. Under intensive confinement conditions, sows’ clinical signs (Berner 1988). Akkermans and Pomper
vulvas are often placed in direct contact with feces (1980) concluded from an extended field study that sows
(Smith 1983). The dog-sitting position helps to force fe- with a significant bacteriuria tend to wean small litters,
cal material into the vagina. Sows resting for long peri- have increased intervals between litters, show a lower
ods void urine at longer intervals. However, housing con- fertility rate, and exhibit an inferior body condition. In
ditions have not yet been studied with respect to UTI. many sows with cystitis, careful observation reveals ab-
The age distribution of UTI favors the concept of normal urination (Becker et al. 1988). The sows stand in
continuous exposure to fecal contamination. The preva- one place before they void urine in small quantities with
lence of UTI increases from 18% in young sows with 1–3 straining. They are more often seen in a dog-sitting posi-
litters to 38% in old sows with 7 and more litters (Becker tion. Proteinuria, macrohemoturia, and pH increases are
et al. 1985). more prevalent in sows infected with A. suis than with E.
coli (Liebhold et al. 1995).
PATHOGENESIS Vulval discharge may appear as dried deposits
around the vulva, on the underside of the tail, or more
In humans and in dogs colonization of the lower genital often as a pool on the floor underneath the sows (Dial
tract and of the urinary tract by E. coli is greatly facilitat- and MacLachlan 1988a). The discharge may be mucoid,
ed by adhesive fimbriae. In the pig, however, only com- mucohemorrhagic, or purulent and is observed most of-
mon pili were detected on some of the E. coli isolates ex- ten during the final phase of urination. However, dis-
amined (Carr and Walton 1992). It is assumed that most charge may result from inflammation of any part of the
agents ascend through the urethra (Smith 1983). Inva- urogenital tract. Significant discharge is more often the
sion is favored by the short, wide urethra of the female consequence of endometritis than of UTI.
pig, the relaxation of the sphincter muscle in late preg- Severe pyelonephritis becomes clinically manifest
nancy and puerperium, trauma to the urethra and blad- during the first 2 weeks postpartum in 40% of the cases
der at coitus and parturition, abnormal bacterial colo- (Stirnimann 1984). Typical cases exhibit a rectal temper-
nization of the sinus urogenitalis and the genital organs, ature below 38.0˚C, a heart rate over 120, polypnoea,
incomplete closure of the vulva, and catheterization of cyanosis, ataxia, and more rarely generalized tremor
the bladder (Berner 1988). Repeated examination of in- (Stirnimann and Tschudi 1985). The blood concentra-
dividual sows led to the conclusion that asymptomatic tions of urea and creatinine are higher than normal.
bacteriuria may temporarily deteriorate to cystitis with
spontaneous remission (Berner 1988). Liebhold et al. LESIONS
(1995) assumed that nonspecific infection promotes col-
onization of the bladder by A. suis. Carr et al. (1990) pos- Berner (1981) examined 118 culled sows for bacteria and
tulate that bacterial colonization leads to shortening and lesions. Twenty-six out of 29 sows with a UTI presented
deformation of the ureteric valve and thereby promotes a cystitis and 12 sows an additional pyelonephritis.
466 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

The gross lesions of cystitis begin as focal or diffuse per 1980). Dip slides have the shortcoming that anaer-
mucosal hyperemia (Dial and MacLachlan 1988b). Sub- obes such as A. suis and slow growers will be missed.
sequently, there may be mucosal ulceration with fib- Catheterization of the sow is possible (Stirnimann
rinopurulent exudate over affected areas. The bladder 1984) but does not circumvent contamination and in-
wall becomes thickened. Similar lesions occur in the volves the risk of setting up a new UTI. Voiding can be in-
ureters and the renal pelvis if infection ascends the uri- duced by rousing the sows in the morning before feeding
nary tract. In pyelonephritis the inflammatory process time (Becker et al. 1985). When collecting midstream
extends into the renal parenchyma. Wedge-shaped foci urine, the attendant should avoid contact with the urine,
extend from the distorted pelvis to the cortex. Fibrosis of which may contain zoonotic agents such as leptospires.
the kidneys may occur with time. Diagnostic test strips are applicable to the urine of
Microscopic bladder lesions can be found even in pigs except for nitrite. The sensitivity of the latter test is
sows with nonspecific UTI and no proteinuria. They con- too low due to the low nitrite concentration in porcine
sist of a prominent goblet cell proliferation and of in- urine (Becker et al. 1985). The most useful parameters
traepithelial cysts containing a few granulocytes. The ep- are protein, hemoglobin, and pH. In cases due to A. suis,
ithelial layer is infiltrated with neutrophils, whereas the pH is strongly alkaline (greater than 8.5) (Carr et al.
mononuclear cells dominate in the lamina propria (Lieb- 1995). Cytological examination may allow discrimina-
hold et al. 1995). tion between bacteriuria, cystitis, and pyelonephritis.
The presence and concentrations of antibodies in the
DIAGNOSIS urine are not well correlated with the severity of the con-
dition (Wagner 1990). Test strips permit the rapid deter-
Mere clinical examination of the animal is of little value mination of blood urea (Liebhold et al. 1995). Concen-
in the diagnosis of UTI (Stirnimann 1984); urine must be trations greater than 10 mmol/L indicate uremia.
examined in most cases. Bacteriology of the urinary tract
is complicated by the normal flora colonizing the vagina TREATMENT
and the distal part of the urethra. Therefore, distinction
between contamination and infection is based on the Nearly all the treatments recommended in the literature
number of bacteria in the urine. A viable count of 105 are aimed at elimination of the bacteria by antimicro-
CFU/mL is interpreted as indicative of infection and 104 bials. The variable susceptibilities of the diverse bacteria
CFU/mL as suspicious. Dip slides (i.e., commercially involved and the frequent acquisition of R factors pose
available slides covered by bacterial culture media) give considerable problems (Table 32.7). With regard to the
satisfactory quantitative results (Akkermans and Pom- observed changes of infecting bacterial species or types

Table 32.7. Antimicrobial sensitivity of significant isolates from porcine UTI in Belgium and Switzerland
Rate of Sensitive Isolates (%)
Colman et al. 1988 Stirnimann 1988
Gram-negative Gram-positive E. coli
Substance (n = 62) (n = 18) (n = 21)
Penicillin G NT 44 NT
Penicillinase-stable penicillins NT 72 NT
Ampicillin 68 44 71
Amoxycillin and clavulanic acid 100 72 NT

Streptomycin 40 44 NT
Neomycin 98 61 86
Spectinomycin 85 72 NT
Gentamicin 100 61 100

Tetracycline 47 61 33
Chloramphenicol 82 83 67
Nitrofurane 95 94 14
Sulfonamide 37 50 52
Trimethoprim 76 61 NT
Trimethoprim and sulfonamide NT NT 76
Macrolide NT 61 NT
Lincomycin NT 72 NT
Note: NT = not tested.
CHAPTER 32 ESCHERICHIA COLI INFECTIONS Bertschinger, Fairbrother 467

in the course of antimicrobial treatments, Berner (1990) Becker, H.-A.; Kurtz, R.; and Von Mickwitz, G. 1985.
recommended the use of either broad-spectrum or com- Chronische Harnwegsinfektionen beim Schwein,
bined antimicrobials and suggested intensifying the Diagnose und Therapie (I). Prakt Tierarztl 66:1006–
search for alternative strategies. 1011.
Becker et al. (1988) treated 9 sows twice daily for 2 Becker, W.; Kurtz, R.; and Von Mickwitz, G. 1988. Chronis-
weeks via feed with sulfadimidine 1.0 g; sulfathiazole 1.0 che Harnwegsinfektionen beim Schwein, Diagnose und
g; and trimethoprim 0.4 g/sow. Significant bacteriuria Therapie (III). Prakt Tierarztl 69:41–45.
was present in 2 sows 1 week after the treatment and in 3 Berner, H. 1981. Untersuchungen zum Vorkommen von
sows 7 weeks later. The same substances applied over 4 Harnwegsinfektionen bei Muttersauen. 2. Mitteilung:
days gave much inferior results. Gentamicin, 2.5 mg/kg Harnwegsinfektionen bei Schlachtschweinen. Tierärztl
body weight, was injected intramuscularly to 15 sows on Umsch 36:250–255.
the first day, followed by 2.0 mg on the next 3 days. One ———. 1988. Cystitis in der MMA-Diagnostik. Prakt Tier-
week after this treatment, 10 sows were free of significant arztl 69:124–131.
bacteriuria. The authors concluded that prolonged treat- ———. 1990. Erregerwechsel als Ursache von Misserfolgen
ment should be preferred. Antimicrobial resistance was bei der Therapie bakteriell bedingter Krankheiten der
not checked. Urogenitalorgane des Schweines. Dtsch Tierärztl
Treatment of severely affected sows was reported by Wochenschr 97:20–24.
Stirnimann (1988). With 34 sows each, injection of Bertschinger, H. U.; Pohlenz, J.; and Hemlep, I. 1977. Unter-
ampicillin 3 g/sow daily for 4 days was compared with suchungen über das Mastitis-Metritis-Agalaktie-Syn-
the same antibiotic combined with novaminsulfon 10 drom (Milchfieber) der Sau. Schweiz Arch Tierheilkd
g/sow. The combined treatment led to a smaller number 119:223–233.
of emergency slaughters. Subclinical UTI persisted in Carr, J., and Walton, J. R. 1992. The characterization of Es-
about half of the successfully treated sows. cherichia coli isolates from the porcine urogenital tract.
Dial and MacLachlan (1988b) concluded that treat- Proc Int Congr Pig Vet Soc 12:262.
ment of urogenital infections of swine generally is frus- ———. 1993. Bacterial flora of the urinary tract of pigs as-
trating. sociated with cystitis and pyelonephritis. Vet Rec
132:575–577.
PREVENTION Carr, J.; Walton, J. R.; and Done, S. H. 1990. Observations
on the intravesicular portion of the ureter from healthy
Results of long-term prospective studies are not avail- pigs and those with urinary tract disease. Proc Int Congr
able. Berner (1988) recommended that all pregnant sows Pig Vet Soc 11:286.
be checked repeatedly for UTI and that positive sows be ———. 1995. Cystitis and ascending pyelonephritis in the
treated with antimicrobials shortly before parturition. sow. In Practice 17:71–79.
Smith (1983) suggested medicating all dry sows on prob- Colman, J.; Devriese, L.; and Verdonck, M. 1988. Bacteriuria
lem farms for 7–10 days at 6-week intervals. The interval and urinary tract infection in sows. Vlaams Diergeneesk
between treatments can be increased gradually as experi- Tijdschr 57:192–198.
ence dictates. Antibiotics such as tetracyclines or a suit- Dial, G., and MacLachlan, N. J. 1988a. Urogenital infections
able form of penicillin have been successfully used in the of swine. Part I. Clinical manifestations and pathogene-
diet. In view of the disappointing results reported by sis. Compend Contin Educ 10:63–71.
Berner (1990), these recommendations should be viewed ———. 1988b. Urogenital infections of swine. Part II.
with caution. Pathology and medical management. Compend Contin
According to Wagner (1990), immunoprophylaxis Educ 10:529–540.
holds little promise. Thus, Smith (1983) and Carr et al. Häni, H.; Brändli, A.; Luginbühl, H.; and König, H. 1976.
(1995) recommended the reduction of environmental Vorkommen und Bedeutung von Schweinekrankheiten:
exposure by improving fecal drainage and housing con- Analyse eines Sektionsgutes (1971–1973). Schweiz Arch
ditions. These factors as well as the role of water intake Tierheilkd 118:1–11.
should be further investigated. Frequency of urination Liebhold, M.; Wendt, M.; Kaup, F.-J.; and Drommer, W.
was increased by giving access to an exercise yard and by 1995. Clinical and light and electron microscopical find-
increasing water intake, which was achieved by a salt ings in sows with cystitis. Vet Rec 137:141–144.
content of 1% in the diet (Smith 1983). Low, D. A.; Braaten, B. A.; Ling, G. V.; Johnson, D. L.; and
Ruby, A. L. 1988. Isolation and comparison of Es-
REFERENCES cherichia coli strains from canine and human patients
with urinary tract infections. Infect Immun
Akkermans, J. P. W. M., and Pomper, W. 1980. The signifi- 56:2601–2609.
cance of a bacteriuria with reference to disturbances in Miquet, J. M.; Madec, F.; and Paboeuf, F. 1990. Epidemiolo-
fertility. Proc Int Congr Pig Vet Soc 6:44. gy of farrowing disorders in the sow: Preliminary results
468 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

of a prospective inquiry in 2 farms. Proc Int Congr Pig ———. 1988. Zur Behandlung der akuten Harn-
Vet Soc 11:472. wegsentzündung bei der Muttersau. Schweiz Arch Tier-
Petersen, B. 1983. Methods of early recognition of puerper- heilkd 130:605–611.
al and fertility disorders in the sow. Livest Prod Sci Stirnimann, J., and Tschudi, P. R. 1985. Beurteilung der
10:253–264. Nierenfunktion bei Muttersauen mit akuter Harn-
Smith, W. J. 1983. Cystitis in sows. Pig News Inform wegsentzündung. Schweiz Arch Tierheilkd
4:279–281. 127:575–582.
Stirnimann, J. 1984. Akute Harnwegsentzündung bei Wagner, S. 1990. Die Immunreaktion bei der durch Es-
der Muttersau. Schweiz Arch Tierheilkd 126:597– cherichia coli bedingten chronischen Harnwegsinfektion
605. des weiblichen Schweines. DVM thesis, Univ München.
Exudative Epidermitis
33 H. C. Wegener and E. W. Skov-Jensen

Exudative epidermitis (EE) has been known by its clinical blood agar after 24 hours’ incubation. It is coagulase neg-
signs for over 150 years (Spinola 1842). The classic dis- ative using the slide test and is heat-stable DNase, lipase,
ease occurs as an acute or peracute infection in suckling and hyaluronidase positive, and mannitol and acetoin
and newly weaned piglets in which a generalized epider- negative. These biochemical characteristics are of value
mitis may lead to dehydration and death. Early occur- in distinguishing the organism by conventional means
rence and distribution have been comprehensively re- from other staphylococci found in pigs (Devriese 1977).
viewed by Jones (1956), who provided a good description A selective indicator medium described by Devriese
of the disease and its effects on production. It has been (1977) which utilizes the lipase activity as the indicative
recorded in most pig-rearing countries in both piglets principle can be very useful for isolation of S. hyicus from
and weaners. Major studies have been carried out by pathological samples.
Sompolinsky (1953), Jones (1956), Underdahl et al. S. hyicus may be isolated from a range of other ani-
(1963, 1965), L’Ecuyer (1966), L’Ecuyer and Jericho mals, including ruminants and fowl (Devriese et al.
(1966), Hunter et al. (1970), and Wegener and Skov- 1978). Phenotypic and genotypic differences from
Jensen 1992. The condition is of sporadic occurrence in porcine isolates suggest that S. hyicus from other animals
all countries but may be of great importance in individ- may belong to separate ecovars (Devriese et al. 1978;
ual herds, especially in newly established or repopulated Schleifer 1986; Takeuchi et al. 1988). Their virulence
herds (Pepper and Taylor 1977). with regard to EE is not known.

ETIOLOGY EPIDEMIOLOGY

Staphylococcus hyicus is the causal agent of the general- EE has been described from all major pig-producing
ized form of the disease seen in piglets. Although it is countries, and the incidence has been reported to be in-
present in profuse culture in lesions in adult pigs, the creasing in some regions (Anon. 1991; Wegener 1992).
disease has not been reproduced in that age group. This increase may reflect changes in pig production to-
Strains of S. hyicus can be divided into virulent and avir- ward larger units, earlier weaning, and higher animal
ulent types with regard to the ability to reproduce EE ex- densities. The disease may occur sporadically and with
perimentally in piglets (Wegener et al. 1993). Both types low morbidity among litters in some pig herds, whereas
can be present simultaneously on the skin of diseased, as in others it reaches epidemic proportions affecting all lit-
well as healthy, piglets (Park and Kang 1986; Wegener et ters. This suggests that immunity plays an important
al. 1993). The production of an exfoliative toxin appears part in the cause of the disease in the individual animal
to be the single most important virulence determinant of as well as in the herd. The significance of immunity in re-
S. hyicus (Amtsberg 1979; Sato et al. 1991; Wegener et al. lation to EE has, however, not been thoroughly investi-
1993; Andresen et al. 1993; Andresen et al. 1997). How- gated.
ever, a range of other factors may also be necessary, but The disease occurs most commonly following the in-
not sufficient, to render S. hyicus capable of causing EE in troduction of carrier animals to a nonimmune herd and
piglets. affects successive litters of piglets, usually those born to
S. hyicus was first described by Sompolinsky (1953) as nonimmune sows. All litters in an affected herd may be
Micrococcus hyicus; it was then defined as a staphylococ- affected, and up to 70% of affected piglets may die. Out-
cus by Baird-Parker (1965). S. hyicus was separated into breaks are usually self-limiting and last for 2–3 months
S. hyicus subsp. hyicus and S. hyicus subsp. chromogenes, a but may persist or recur if nonimmune sows are brought
nonpathogen, by Devriese et al. (1978). S. hyicus subsp. into infected buildings or exposed to infected animals.
chromogenes was subsequently elevated to species S. chro- Outbreaks may start among the weaned piglets, possibly
mogenes by Hajek et al. (1986), making S. hyicus the taxo- as a result of mixing nonimmune litters and litters of im-
nomically correct designation for the causal agent of EE. mune carriers, and then spread to the farrowing section
The organism is a gram-positive coccus that forms 3–4 of the herd.
mm porcelain-white nonhemolytic colonies on sheep S. hyicus can frequently be recovered from the nasal

469
470 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

mucosa of healthy pigs, from the conjunctiva, the skin of 1990), and a capsule present in all virulent but not all
the snout or ear, and from the vagina in gilts and sows avirulent strains of S. hyicus inhibits phagocytosis by
(Hajsig et al. 1985; Wegener and Skov-Jensen 1992). S. neutrophils and macrophages (Wegener 1990). All
hyicus strains indistinguishable from those present in the porcine S. hyicus strains coagulate pig plasma, suggesting
vaginas of sows have been recovered from the skin of a potential for forming aggregates which may increase
their offspring, suggesting that colonization takes place protection of the bacterium against phagocytosis. In ad-
during passage through the birth canal (Wegener and dition, the production of catalase may protect the bac-
Skov-Jensen 1992). terium from being killed by the phagocytic cells. All of
S. hyicus is very resistant to adverse conditions (as these properties may contribute to overcoming the initial
most staphylococci are) and can persist in the environ- immune response of the piglet.
ment for long periods. The organism can persist for The most important factor in the pathogenesis is
weeks on fittings and surfaces, and it has been recovered probably the production of an exfoliative toxin of ap-
from the air of infected units at levels up to 2.5 × 104/m3, proximately 30 kDa. Crude or purified exfoliative toxin,
suggesting that airborne transmission is possible (We- which is demonstrable in culture supernates of the or-
gener 1992). Other species such as horses, dogs, cattle, ganism, can reproduce the skin alterations seen in clini-
goats, and chickens may be of little importance as cal EE when injected subcutaneously in piglet skin local-
sources of infection for pigs. ly (Wegener et al. 1993; Andresen et al. 1993; Sato et al.
1991). There are different antigenic variants of the toxin;
PATHOGENESIS however, they all seem to exert the same activity in pig
skin (Andresen et al. 1997). The effect of the purified tox-
Application of pure cultures of virulent S. hyicus to the in is separation of the cells in the epidermis, notably sep-
scarified skin of a nonimmune pig is sufficient to repro- aration of cells in the upper stratum spinosum, allowing
duce the disease (Underdahl et al. 1965; L’Ecuyer and for rapid intraepidermal spread of the bacteria (An-
Jericho 1966), but it can also be produced by subcuta- dresen et al. 1993). Exfoliation of the skin is accompa-
neous injection in specific-pathogen-free (SPF) piglets nied by excess sebaceous secretion and serous exudate. S.
(Underdahl et al. 1965; Wegener et al. 1993). Conven- hyicus is present in large numbers in the skin and may be
tional animals may be resistant to such applications, sug- isolated from the draining lymph nodes and blood. The
gesting that immunity may be an important protective mortality associated with this disease results from dehy-
factor. Studies indicate that other elements of the skin dration and possibly also septicemia.
flora, especially other staphylococci, may contribute to Exudative epidermitis shares many similarities with
this resistance to colonization (Allaker et al. 1988). Trau- the human infection called the staphylococcal scalded
ma from fighting, unclipped teeth, rough bedding, or skin syndrome, which is a Staphylococcus aureus infection
pen walls leading to exposure of dermis may allow the of the skin of neonates. The infection leads to a local or
organism to establish infection, although S. hyicus may a generalized exfoliation of the epidermis and excessive
also be able to penetrate the epidermis directly. sebaceous secretion and is caused by strains of S. aureus
The earliest changes are seen as skin reddening ac- capable of producing exfoliative toxins. Two variants of
companying the multiplication of the organism on the the toxin are known: ET-A, which is encoded by a gene lo-
skin surface and its growth between the corneocytes of cated on the chromosome, and ET-B, which is plasmid
the epidermis, where microcolonies develop. Inflamma- encoded. The exfoliative toxins of S. aureus and S. hyicus
tion, marked hyperplasia of the stratum corneum, and have different species specificity. ET-A and ET-B affect
invasion by neutrophils occur, with an increase in thick- the skin of humans and mice but not pigs, whereas the
ness of the epidermis, followed by its erosion. The stra- exfoliative toxin of S. hyicus affects pigs and chickens but
tum germinativum becomes disorganized and pene- not mice.
trates deeply into the dermis. Clinical signs develop in
gnotobiotic piglets when the number of organisms on CLINICAL SIGNS
the skin exceeds 105/cm2 (Allaker et al. 1988). S. hyicus
may adhere to fibronectin in the dermis by fibronectin- Piglets usually develop the disease between 4–6 days and
binding proteins on the bacterial surface (Lämmler et al. 5–6 weeks of age. Clinical signs begin with dejection and
1985). a reddish or coppery skin color. Thin, pale brown scales
The phagocyte-opsonin system is the pig’s first active of exudate develop in the axillae and groin and within
line of defense against the infection. Many S. hyicus 3–5 days spread to all parts of the body and rapidly be-
strains harbor determinants that may protect them from come dark in color and greasy in texture (Fig. 33.1). The
phagocytosis: protein A, present in the cell wall of most skin of affected piglets often feels hot, the hair coat is
porcine S. hyicus strains, reduces opsonization by bind- matted, and exudate may extend to the eyelashes. Ulcers
ing immunoglobulins at the Fc terminal (Takeuchi et al. may occur in the mouth, and separation of horn may oc-
CHAPTER 33 EXUDATIVE EPIDERMITIS Wegener, Skov-Jensen 471

33.1. Generalized exudative


epidermitis in a 3-week-old piglet.

cur at the bulbs of the heels. Anorexia and dehydration sion is marked in survivors, and productivity of the herd
are features of this disease. Severely affected piglets lose may be depressed by up to 35% during an outbreak and
weight rapidly and may die within 24 hours; death usu- up to 9% in the year following infection (Pepper and Tay-
ally occurs within 3–10 days. There is no pruritis, and lor 1977). Disease in adults varies in severity but occurs
fever is not common. as localized lesions on the back or flanks. Mild forms
Not all piglets in a litter are affected to the same ex- may appear as brownish areas of EE, but in some cases,
tent, and some individuals will suffer from chronic dis- there may be ulceration (Smith et al. 1990).
ease in which smaller areas of the body are involved (Fig.
33.2). Mildly affected piglets may have a yellowish skin, LESIONS
appear hairy, and have only a few flakes of exudate in the
axillae or groin or near facial scratches or damage on the Gross Lesions
knees or adjacent to badly clipped teeth. Growth depres- Early lesions of the infection include reddening of skin
and the presence of a clear exudate. The abdominal skin
can be peeled off by slight rubbing. Early lesions are usu-
ally present around the mouth, eyes, and ears as well as
on the abdomen. Later cases are covered by a thick
brownish, greasy, and odorous layer due to dirt and feces
sticking to the affected skin. During the recovery phase,
the skin is dry and crusted for a period of several days to
weeks. The carcasses of pigs that have died from EE are
dehydrated and emaciated. The superficial lymph nodes
are usually edematous and swollen. Most animals have
empty stomachs, and urate crystals may be seen in the
medulla of the kidney on section. There is often an accu-
mulation of mucoid or crystalline material in the pelvis
of the kidney, and pyelonephritis may be present.

Microscopic Lesions
Early changes of the epidermis are exfoliation, exocyto-
sis, crust formation, and formation of vesicles and pus-
tules classified as an “intraepidermal vesicular and pus-
tular dermatitis.” In the later stages, acanthosis
(hyperplasia of the epidermis) is observed (Fig. 33.3). In
the dermis, perivascular inflammation occurs (Andresen
33.2. A single piglet with generalized exudative et al. 1993). In histological sections of the skin, bacterial
epidermitis among only mildly affected or unaffected microcolonies may occur in the keratinized layer of the
littermates. epidermis.
472 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

33.3. Histological section of


skin of pig with generalized
exudative epidermitis (magnifica-
tion ×1200). Changes present in
the epidermis are exfoliation,
exocytosis, crust formation,
formation of vesicles and pustules
classified as an “intraepidermal
vesicular and pustular dermati-
tis,” and acanthosis (hyperplasia
of the epidermis). In the dermis,
perivascular inflammation can be
seen.

Bacteriology cal means (Devriese 1977) or by use of strip tests such as


S. hyicus can usually be isolated from the lesions, from the Staph-Zym test (Lämmler 1989). These have the ad-
the superficial lymph nodes, and frequently also from vantage of revealing the identity of non–S. hyicus staphy-
the liver and spleen of untreated cases but may be diffi- lococci.
cult to demonstrate on nonselective culture media if S. hyicus from pigs are very heterogeneous with re-
treatment has been given or if secondary infection by gard to phago-, sero-, and DNA fingerprinting types
Proteus sp. and Pseudomonas aeruginosa has occurred. (Wegener 1993a; Park and Kang 1987). Diagnosis is
The use of a selective indicative agar facilitates isolation complicated by the simultaneous presence of up to 8 dif-
of S. hyicus from pathological samples (Devriese 1977). ferent types of S. hyicus on diseased piglets. Wegener
Both virulent and avirulent strains of S. hyicus can be iso- (1993b) found that each diseased piglet on average har-
lated as mixed cultures from the skin, lymph nodes, and bored 1.9 different phage types and 2.3 different antibi-
organs of diseased piglets (Wegener 1992). Whether the otic resistance patterns among 10 randomly selected iso-
avirulent strains take any active part in the establish- lates of S. hyicus recovered from the skin. Only a slightly
ment or course of the infection remains unknown. lower diversity was observed for strains recovered from
the liver or the spleen of the animals. In the absence of
DIAGNOSIS simple methods to differentiate virulent from avirulent
strains in the diagnostic laboratory, all types of S. hyicus
The clinical signs are generally sufficient to reach a diag- should be regarded as potentially virulent. Thus, antimi-
nosis in young piglets. The lack of fever or of pruritis and crobials for therapy which affect all types present should
the generalized nature of the lesions, their appearance, be chosen. Similarly, autogenous vaccines should be pre-
and the variation in severity within an affected litter are pared from all types present on the diseased animals.
all features suggestive of the disease. Confirmation may Diagnosis is less easy when the lesions are mild, lo-
be obtained by histological and bacteriological means. It calized around predisposing lesions such as fight
may be necessary to confirm the identity of the staphy- wounds, or have been treated. The demonstration of S.
lococci isolated as S. hyicus by conventional bacteriologi- hyicus and response to antimicrobials may help confirm
CHAPTER 33 EXUDATIVE EPIDERMITIS Wegener, Skov-Jensen 473

uncomplicated disease of this type, but the organism REFERENCES


may be present in lesions caused by a number of initiat-
ing agents. Allaker, R. P.; Lloyd, D. H.; and Smith, L. M. 1988. Preven-
Other skin conditions that may be confused with EE tion of exudative epidermitis in gnotobiotic piglets by
include swine pox (localized lesions, rarely fatal), mange bacterial interference. Vet Rec 123:287–288.
(pruritis, demonstration of mites), ringworm (expand- Amtsberg, G. 1979. Nachweis von Exfoliation auslösenden
ing superficial lesions, isolation of fungus), pityriasis Substanzen in Kulturen von Staphylococcus hyicus des
rosea (expanding circles, nonfatal, lesions not greasy), Schweines und Staphylococcus epidermidis Biotyp 2 des
zinc deficiency (weaners, symmetrical dry lesions), der- Rindes. Zentralbl Veterinärmed (B) 26:257–272.
matosis vegetans (inherited in Landrace, fatal pneu- Andresen, L. O.; Wegener, H. C.; and Bille-Hansen, V. 1993.
monitis), and local wounds such as facial fight wounds Staphylococcus hyicus–skin reactions in piglets caused by
and abraded knees in piglets and crate injuries in adults. extracellular products and by partially purified exfolia-
The organism may be isolated from other pathologic tive toxin. Microbial Pathogen 15:217–225.
conditions such as arthritis in piglets (Noda and Fukui Andresen, L. O.; Bille-Hansen, V.; and Wegener, H. C. 1997.
1986) and cystitis in sows as well as from the skin of Staphylococcus hyicus exfoliative toxin: Purification and
healthy pigs. demonstration of antigenic diversity among toxins from
virulent strains. Microbial Pathogenesis (in press).
TREATMENT AND PREVENTION Anon. 1991 Greasy pig (disease review). Int Pigletter
11:30–32.
Treatment is most successful if carried out early in the Baird-Parker, A. C. 1965. The classification of staphylococci
disease; severely affected animals may not respond. The and micrococci from world-wide sources. J Gen Micro-
effect of systemic treatment is reduction in the severity biol 38:363–387.
of the skin lesions, development of only superficial le- Devriese, L. A. 1977. Isolation and identification of Staphy-
sions, and promotion of the healing process. S. hyicus is lococcus hyicus. Am J Vet Res 38:787–792.
frequently resistant to antibiotics. This resistance has Devriese, L. A.; Hajek, V.; Oeding, P.; Meyer, S. A.; and
been shown to be predominantly mediated by plasmids Schleifer, K. H. 1978. Staphylococcus hyicus (Sompolinsky
(Wegener and Schwarz 1993). Combinations of 1953) comb. nov. and Staphylococcus hyicus subsp. chro-
trimethoprim and sulfonamides or lincomycin and mogenes subsp. nov. Int J Syst Bacteriol 28:482–490.
spectinomycin have been shown to have good in vitro ac- Hajek, V.; Devriese, L. A.; Mordarski, M.; Goodfellow, M.;
tivity against S. hyicus (Wegener et al. 1994). Antimicro- Pulverer, G.; and Varaldo, P. E. 1986. Elevation of
bial treatment should be accompanied by the provision Staphylococcus hyicus subsp. chromogenes (Devriese et al.
of a fluid replacer or at least clean water for affected 1978) to species status: Staphylococcus chromogenes (De-
piglets and by local treatment with antibiotics or skin vriese et al. 1978) comb. nov. System Appl Microbiol
disinfectants such as cetrimide, hexocil, or Virkon X in 8:169–173.
order to speed recovery and prevent spread of the infec- Hajsig, D.; Babic, T.; and Madic, J. 1985. Exudative epider-
tion. Treatment may have to be continued for at least 5 mitis in piglets. II. Distribution of Staphylococcus hyicus
days, and clinically affected piglets may make a slow re- subsp. hyicus: Findings in healthy piglets. Vet Arh
covery or remain stunted. 55:45–51.
Vaccination of sows with autogenous bacterins made Hunter, D.; Todd, J. N.; and Larkin, M. 1970. Exudative epi-
from strains isolated on the affected farm may be of val- dermitis of pigs. Br Vet J 126:225–229.
ue in protecting the litters of newly purchased sows Jones, L. D. 1956. Exudative epidermitis of pigs. Am J Vet
when given before farrowing. Antibodies can effectively Res 17:179–193.
neutralize the effect of the exfoliative toxin in the skin. It Lämmler, C. 1989. Evaluation of the Staph-Zym system for
is possible that the toxin may be able to serve as a single identification of Staphylococcus hyicus and Staphylococcus
protective antigen; however, this has not yet been shown intermedius. J Vet Med (B) 36:180–184.
under field conditions. Therefore, autogenous vaccines Lämmler, C.; De Freitas, J. C.; Chatwal, G. S.; and Blobel, H.
should be prepared from both the bacterial cells and the 1985. Interaction of immunoglobulin G, fibrinogen, fi-
culture supernatant, which contains the exfoliative toxin. bronectin with Staphylococcus hyicus and Staphylococcus
The incidence of the disease may be reduced by clip- intermedius. Zbl Bakt Hyg (A) 260:232–237.
ping the teeth of litters at risk, by ensuring that pen sur- L’Ecuyer, C. 1966. Exudative epidermitis in pigs: Clinical
faces are not abrasive, and by providing soft and dry bed- studies and preliminary transmission trials. Can J Comp
ding, such as softwood sawdusts or chaffed straw. Sows Med 30:9–16.
entering farrowing accommodation should be washed L’Ecuyer, C., and Jericho, K. 1966. Exudative epidermitis in
and disinfected and placed in clean, disinfected, or fumi- pigs: Etiological studies and pathology. Can J Comp Med
gated pens. Prompt treatment of local lesions on both 30:94–101.
sows and piglets may also help. Noda, K., and Fukui, T. 1986. Outbreaks of pyogenic arthri-
474 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

tis in newborn piglets and stillbirth caused by S. hyicus Underdahl, N. R.; Grace, P. D.; and Young, G. A. 1963. Ex-
subsp. hyicus. Jpn J Vet Med Assoc 39:305–310. perimental transmission of exudative epidermitis of
Park, C. K., and Kang, B. K. 1986. Studies on exudative epi- pigs. J Am Vet Med Assoc 142:754–762.
dermitis in pigs. I. Isolation and some properties of Underdahl, N. R.; Grace, P. D.; and Twiehaus, M. J. 1965.
Staphylococcus hyicus subsp. hyicus from diseased and Porcine exudative epidermitis: Characterisation of bac-
healthy pigs. Korean J Vet Res 26:251–257. terial agent. Am J Vet Res 26:617–624.
———. 1987. Studies on exudative epidermitis in pigs. II. Wegener, H. C. 1990. Studies on Staphylococcus hyicus viru-
Serological typing of Staphylococcus hyicus subsp. hyicus lence in relation to exudative epidermitis in piglets. Proc
isolated from pigs. Korean J Vet Res 27:47–52. Int Congr Pig Vet Soc 11:197.
Pepper, T. A., and Taylor, D. J. 1977. The effect of exudative ———. 1992. Staphylococcus hyicus Epidemiology and Vir-
epidermitis on weaner production in a small pig herd. ulence in Relation to Exudative Epidermitis in Pigs.
Vet Rec 101:204–205. Ph.D. diss. Nat Vet Lab and Royal Vet Agric Univ, Copen-
Sato, H.; Tanabe, T.; Kuramoto, M.; Tanaka, K.; Hashimoto, hagen, Denmark.
T.; and Saito, H. 1991. Isolation of exfoliative toxin from ———. 1993a. Development of a phage typing system for
Staphylococcus hyicus subsp. hyicus and its exfoliative ac- Staphylococcus hyicus. Res Microbiol 144:237–244.
tivity in the piglet. Vet Microbiol 27:263–275. ———. 1993b. Diagnostic value of phage typing, antibi-
Schleifer, K. H. 1986. Micrococcaceae. In Bergey’s Manual for ogram typing and plasmid profiling of Staphylococcus
Determinative Bacteriology, 9th ed. Ed. P. H. A. Sneath. hyicus from piglets with exudative epidermitis. J Vet Med
Baltimore: William & Wilkins, pp. 1003–1035. (B) 40:13–20.
Smith, W. J.; Taylor, D. J.; and Penny, R. H. C. 1990. A Wegener, H. C., and Schwarz, S. 1993. Antibiotic resistance
Colour Atlas of Diseases and Disorders of the Pig. Lon- and plasmids in Staphylococcus hyicus isolated from pigs
don: Wolfe Publishing, pp. 113–114. with exudative epidermitis and from healthy pigs. Vet
Sompolinsky, D. 1953. De l’impetigo contagiosa suis. Microbiol 35:363–372.
Schweiz Arch Tierheilkd 95:302–309. Wegener, H. C., and Skov-Jensen, E. W. 1992. A longitudinal
Spinola, J. 1842. Die Krankheiten der Schweine. Berlin: Ver- study of Staphylococcus hyicus colonization of vagina of
lag Hirschwald, pp. 146–148. gilts and transmission to piglets. Epidemiol Infect
Takeuchi S.; Kobayashi, Y.; Morosumi, T.; and Mori, Y. 109:433–444.
1988. Protein A in Staphylococcus hyicus subsp. hyicus iso- Wegener, H. C.; Andresen, L. O.; and Bille-Hansen, V. 1993.
lated from pigs, chickens and cows. Jpn J Vet Sci Staphylococcus hyicus virulence in relation to exudative
50:153–157. epidermitis in the piglet. Can J Vet Res 57:119–125.
Takeuchi S.; Kobayashi, Y.; and Mori, Y. 1990. Assay of pro- Wegener, H. C.; Watts, L.; Salmon, S. A.; and Yancey, R. J.,
tein A in Staphylococcus hyicus subsp. hyicus by ELISA Jr. 1994. Antimicrobial susceptibility of Staphylococcus
and immunoelectron microscopy. Vet Microbiol hyicus isolated from exudative epidermitis in pigs. J Clin
25:297–302. Microbiol 32:793–795.
Haemophilus parasuis
34 V. J. Rapp-Gabrielson

Once considered a sporadic disease of young pigs com- further investigation (Kobisch and Desmettre 1980; Mo-
promised by stress, porcine polyserositis and arthritis rozumi and Nicolet 1986a; Kielstein 1991). Nicoti-
(Glasser’s disease), caused by Haemophilus parasuis, has namide adenine dinucleotide (NAD, or V factor) is re-
emerged as one of the significant bacterial diseases af- quired for growth and can be supplied by heated blood
fecting swine throughout the world. Adoption of new (chocolate agar) or by satellitic growth in the vicinity of a
production technologies for high-health-status herds streak of a staphylococcus strain. After 24–48 hours’
and the emergence of new respiratory syndromes have growth, colonies are small, translucent, and non-
contributed to an increase in prevalence and severity of hemolytic on blood agar.
the disease. Disease management with antibiotics, vacci- The existence of serovars was first reported by Bakos
nation, and other strategies is not always successful in et al. (1952). In subsequent years, expansion of this
countering production losses due to H. parasuis infec- serotyping scheme by other investigators led to several
tion. It has long been known that the immune status of a proposals for new serovars (Schimmel et al. 1985; Mo-
herd is a determinant of pathogenic outcome of infec- rozumi and Nicolet 1986b; Nicolet et al. 1986; Kielstein
tion (Nielsen and Danielsen 1975). However, the hetero- 1991; Rapp-Gabrielson and Gabrielson 1992). Presently,
geneity among H. parasuis strains is striking, and a better 15 serovars based on immunodiffusion are recognized
understanding of the association of these phenotypic (Kielstein and Rapp-Gabrielson 1992). The type-specific
and genotypic differences with virulence potential and antigen is heat-stable polysaccharide (Morozumi and
protective immunity is beginning to emerge. Nicolet 1986b) presumed to be capsule or lipopolysac-
charide (LPS). Serotyping of isolates from Japan, Ger-
ETIOLOGY many, the United States, Canada, and Australia shows
serovars 5, 4, and 13 to be most prevalent (Table 34.1).
Glässer (1910) first reported the association of a small A large percentage of isolates are nontypeable, in-
gram-negative rod with fibrinous serositis and pol- dicating some isolates may not express sufficient type-
yarthritis of swine. Initially, the causative agent was specific antigen or the probable existence of additional
identified as Haemophilus suis by Hjärre and Wramby serovars.
(1943) and as Haemophilus influenza suis by Lecce (1960).
The name was changed to H. parasuis based on demon-
Table 34.1. Prevalence of H. parasuis serovars
stration that the organism did not require X factor
(haemin or other porphyrins) for growth (Biberstein and Frequency (%)
White 1969; Kilian 1976). The taxonomic position of H. H. parasuis Canada and the
parasuis within the Pasteurellaceae is still uncertain, due Serovar Japana United States Germany Australia
to a lack of nucleic acid homology with other
1 2.5 2.2 4.1 2.4
Haemophilus species (De Ley et al. 1990; Dewhirst et al. 2 5.8 7.9 5.5 7.3
1992). Considerable genotypic heterogeneity has also 4 9.2 16.2 17.2 12.2
been demonstrated among H. parasuis strains (Smart et 5 14.2 23.3 23.8 31.7
al. 1988; Rapp-Gabrielson et al. 1992a; Blackall et al. 7 or 10b — 4.8 4.5 9.8
12 — 7.0 2.8 2.4
1997). It has been proposed that more than one bacteri- 13 — 11.0 4.5 17.1
al species may be represented by strains identified as 14 — 9.2 1.7 0
H. parasuis (Morozumi et al. 1986; Dewhirst et al. 3,6,8,9,11, or 15 — 4.3 10.3 1.0
1992). Nontypeable 68.3 14.1 26.2 12.2
Microscopically, H. parasuis cells are pleomorphic, Sources: Morikoshi et al. 1990; Rapp-Gabrielson and
varying from single coccobacilli to long, thin, filamen- Gabrielson 1992; Kielstein and Rapp-Gabrielson 1992; Blackall et
tous chains. A capsule can usually be demonstrated, but al. 1996.
a
expression is influenced by in vitro culture (Rapp- Only tested for H. parasuis serovars 1–5.
b
Some strains react to both serovars 7 and 10 and cannot be
Gabrielson et al. 1992b). Thus, the significance of re- distinguished by immunodiffusion (Rapp-Gabrielson, unpub-
ports associating lack of capsule with virulence needs lished data—1995; Blackall et al. 1996).

475
476 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

EPIDEMIOLOGY of H. parasuis strains occurring in the respiratory tract


that is capable of invading systemically and causing dis-
Haemophilus parasuis infects only swine. It is commonly ease (Rapp et al. 1986; Rapp-Gabrielson 1993).
isolated from nasal secretions of healthy swine An association between serovar and isolation from
(Bertschinger and Nicod 1970; Harris et al. 1969; Smart polyserositis was apparent in several studies (Bakos et al.
et al. 1989) and from the lungs of pigs with pneumonia, 1952; Morozumi and Nicolet 1986b; Kielstein 1991). Re-
but not generally from normal lungs (Little 1970; Møller cently, differences in virulence among serovars was
et al. 1993). In conventional herds, the organism is one of demonstrated by inoculation of SPF or cesarean-derived,
the earliest and most prevalent bacterial isolates from colostrum-deprived (CDCD) swine with strains repre-
nasal swabs of pigs at 1 week of age (Kott 1983). senting the 15 serovars (Kielstein and Rapp-Gabrielson
Historically, Glasser’s disease has been considered a 1992; Nielsen 1993; Amano et al. 1994; Rapp-Gabrielson
sporadic disease of young swine compromised by stress. et al. 1995). In these studies, strains representing some
However, the epizootiologic picture in specific-pathogen- serovars were highly virulent and strains representing
free (SPF) or high-health-status herds that represent an other serovars were avirulent. Virulence of field isolates
immunologically naive population is much different was consistent with that of the reference strain, indicat-
(Nielsen and Danielsen 1975; Baehler et al. 1974; ing a causal relationship between serovar and virulence
Menard and Moore 1990). Introduction of H. parasuis (Table 34.2). However, the demonstration that two
may result in systemic disease of high morbidity and serovar 14 strains differed in virulence for CDCD pigs in-
mortality, affecting swine at any stage of production. dicates that factors other than serovar contribute to the
Presently, H. parasuis is one of the most serious prob- virulence potential of a strain (Rapp-Gabrielson et al.
lems associated with mixing swine from different herds 1995).
or introduction of new breeding stock into a herd (Smart Sodium dodecyl sulfate-polyacrylamide gel elec-
et al. 1989; Menard and Moore 1990). trophoresis (SDS-PAGE) of whole-cell and outer-mem-
The role of H. parasuis in swine respiratory disease is brane (OM) proteins also demonstrates phenotypic het-
more problematic. Demonstration of purulent rhinitis erogeneity among strains (Morozumi and Nicolet 1986a;
associated with H. parasuis colonization supports a pos- Rapp et al. 1986; Morikoshi et al. 1990; Rosner et al.
sible role as a predisposing factor for other viral and bac- 1991; Rapp-Gabrielson et al. 1992a). These reports indi-
terial pathogens (Gois et al. 1983; Vahle et al. 1995, cate a possible association of virulence potential with
1997). In pneumonia, H. parasuis has been assumed to specific protein patterns, but the precise relationship be-
be an opportunistic secondary invader, causing disease tween protein pattern, serovar, and virulence potential
only in association with other viral or bacterial agents. remains to be defined. Heterogeneity of H. parasuis LPS
Such a relationship was evident after accidental infection has been demonstrated by SDS-PAGE patterns and im-
of pigs experimentally inoculated with pseudorabies munoblotting with monoclonal antibodies, but LPS pat-
virus with H. parasuis serovar 4 (Narita et al. 1994). How- terns have not been associated with virulence (Zucker et
ever, several recent reports indicate H. parasuis may be a al. 1994; Zucker et al. 1996). Filamentous structures pre-
primary agent in fibrinosuppurative bronchopneumonia sumed to be fimbriae have been demonstrated on some
(Pöhle et al. 1992; Barigazzi et al. 1994; Solano et al. H. parasuis strains, but their role in adhesion or
1998). Isolation of H. parasuis from pneumonia has in- pathogenicity remains to be defined (Münch et al.
creased substantially in recent years and is believed to be 1992).
associated with the increased prevalence of mycoplasma
pneumonia as well as viral respiratory pathogens such as
porcine reproductive and respiratory syndrome (PRRS)
virus, swine influenza virus, and porcine respiratory Table 34.2. Virulence of strains representing
H. parasuis serovars for SPF swine
coronavirus. H. parasuis, in combination with Mycoplas-
ma hyorhinis, was isolated from 51.2% of lungs from No. of
PRRS-infected swine (Kobayashi et al. 1996). However, H. parasuis Strains
Serovar Evaluated Virulencea
clinical impressions that PRRS is exacerbated by H. para-
suis infection have not been substantiated in experimen- 1, 5, 10, 12, 13, 14 10 Death within 96 hours
tal models (Solano et al. 1997; Cooper et al. 1995). 2, 4, 15 10 Severe polyserositis and arthritis
at necropsy
The pathogenic potential of H. parasuis strain(s) oc- 8 1 Mild clinical signs and gross
curring in a herd is also a factor in the severity and pro- lesions
gression of systemic disease. Serovars commonly isolat- 3, 6, 7, 9, 11 8 No clinical signs or gross lesions
ed from upper respiratory sites in swine include serovars Source: Kielstein and Rapp-Gabrielson 1992.
infrequently isolated from systemic sites (Bloch 1985; a
Swine inoculated intraperitoneally with 5 × 108 colony-
Rapp-Gabrielson 1993). There may be a subpopulation forming units.
CHAPTER 34 HAEMOPHILUS PARASUIS Rapp-Gabrielson 477

PATHOGENESIS CLINICAL SIGNS AND LESIONS

Experimental challenge models have been developed to Clinical presentation is dependent on the location of in-
investigate the pathogenesis of H. parasuis infection. flammatory lesions. In naive herds or pigs, onset is rapid,
Vahle et al. (1995) examined sequential events in infec- occurring a few days after exposure. Clinical signs in-
tion by inoculating CDCD pigs intranasally with a viru- clude pyrexia and apathy followed by inappetence and
lent strain of H. parasuis. Within 12 hours postinocula- anorexia. Dyspnea, pain (evidenced by squealing),
tion, H. parasuis was isolated from the nasal cavity and swollen joints, lameness, tremor, incoordination,
trachea; within 36 hours, from blood cultures; and at cyanosis, recumbency, and death may follow. Abortion
36–108 hours, from systemic tissues. Early colonization in gilts and chronic lameness in boars may be sequelae to
of the middle and caudal nasal cavity and trachea was al- acute infection. Even if infection of gilts is controlled by
so demonstrated by immunohistochemistry and trans- antibiotic treatment, pigs in subsequent farrowings may
mission electron microscopy (Vahle et al. 1997). Colo- experience severe disease (Menard and Moore 1990). In
nization was associated with purulent rhinitis, focal loss conventional herds, chronic infections in nursery pigs
of cilia, and acute cell swelling within the nasal and tra- may result in poorly performing pigs. Cough, dyspnea,
cheal mucosa. In vitro infection of nasal turbinate ex- weight loss, lameness, and rough hair coat are the pri-
plants also resulted in marked reduction in ciliary activi- mary clinical signs.
ty and damage to ciliated epithelial cells (Vahle 1996). Primary macroscopic lesions are a serofibrinous to
Bacterial cells were not closely associated with cilia or ep- fibrinopurulent exudate at single or multiple serosal sur-
ithelium, and the mechanism of colonization or cellular faces, including the peritoneum, pericardium, and pleu-
destruction was not defined. The observation by these ra; articular surfaces, particularly the carpal and tarsal
investigators that H. parasuis preferentially colonizes the joints, and the meninges may also be involved (Hjärre
nasal cavity and trachea, and not the tonsil, is in concor- 1958; Amano et al. 1994). Microscopically, the exudate
dance with the ability to isolate H. parasuis from the consists of fibrin, neutrophils, and lesser numbers of
nasal cavity, but not tonsil or lung specimens, from macrophages (Vahle et al. 1995). Less commonly, H.
slaughterhouse pigs (Møller et al. 1993). In contrast, it parasuis infection may result in acute septicemic disease
has been reported that H. parasuis antigen was detected in which cyanosis, subcutaneous and pulmonary edema,
in tonsillar tissue but not in the nasal cavity by im- and death can occur without the typical serosal inflam-
munoperoxidase stain and electron microscopic exami- mation (Riley et al. 1977; Peet et al. 1983; Desrosiers et
nation (Amano et al. 1994). al. 1986). Fasciitis and myositis (Hoefling 1991) and pu-
Mucosal injury may enhance invasion. Microbial and rulent rhinitis (Gois et al. 1983; Vahle et al. 1995) have al-
host factors involved with systemic infection are not so been reported.
known; however, the virulence of some strains is re-
markable. Intratracheal inoculation of less than 100 DIAGNOSIS
colony-forming units of strains representing several
serovars caused systemic disease and death in CDCD Diagnosis is usually based on herd history, clinical signs,
pigs within a few days (Rapp-Gabrielson et al. 1995). and necropsy. Bacterial isolation, necessary for confir-
Bacteremia is apparent in pigs in the early stages of in- mation, is not always successful. This is due in part to the
fection (Vahle et al. 1995). Septicemic lesions consist of fragility and fastidious growth requirements of H. para-
petechiae or ecchymoses in the liver, kidney, and suis relative to other bacteria that may also be present in
meninges; high levels of endotoxin are detected in plas- the specimen. Retrospective analysis of submissions to
ma, and fibrinous thrombi are present in many organs diagnostic laboratories in Ontario indicate that the true
(Amano et al. 1994). Subsequent replication at multiple incidence of disease may be 10-fold higher than report-
serosal surfaces produces the typical fibrinosuppurative ed, due in part to the inability to confirm the presence of
polyserositis, polyarthritis, and meningitis observed in H. parasuis from submitted specimens (Miniats et al.
field cases (Amano et al. 1994; Vahle et al. 1995). Pneu- 1986). Necropsies should be performed not only on pigs
monia was not prominent in one challenge model, even with severe clinical signs and lesions but also on pigs in
though H. parasuis was isolated from the lung (Vahle et the acute phase of disease, prior to administration of an-
al. 1997). Pneumonia was also not evident following in- tibiotics. The best chance for isolation is by culturing sev-
oculation of the reference strains of serovars 1, 4, or 5 eral serosal surfaces or exudates, cerebrospinal fluid, and
(Amano et al. 1994). Differing observations on the abili- heart blood, even if lesions are mild or not apparent. H.
ty of H. parasuis to produce pneumonia may be due to parasuis can be recovered from these fluids when inocu-
differences per se in the challenge models, the doses ad- lated into a transport medium prior to shipment to the
ministered, or pathogenic potential of the strains exam- diagnostic laboratory (Mendez-Trigo and Trigo 1996).
ined. Although somewhat laborious for routine diagnosis, spe-
478 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

cial dilution techniques followed by plating on selective can be ascertained only after identification of other viral
media containing antibiotics have been used successfully and bacterial pathogens that may be involved in the mul-
for culturing H. parasuis in high numbers from respira- tifactoral disease process.
tory specimens (Møller and Kilian 1990; Møller et al.
1993). Incubation in 5% CO2 or a candle jar does not en- TREATMENT
hance growth of H. parasuis. However, the use of defibri-
nated horse blood and tryptose blood agar base, rather Prophylactic use of antibiotics or therapeutic oral med-
than bovine or sheep blood and trypticase soy agar, does ication may be of little value in severe H. parasuis out-
appear to enhance growth. breaks (Madsen 1984; Wiseman et al. 1989; Menard and
Biochemical tests are required to distinguish H. para- Moore 1990). High antibiotic doses should be adminis-
suis from other NAD or V factor–dependent organisms tered parenterally as soon as clinical signs have manifest-
belonging to the family Pasteurellaceae that have been ed, and all pigs in the affected group, not just those show-
isolated from swine (Table 34.3; Møller et al. 1996). ing signs, should be treated (Desrosiers et al. 1986).
Occasionally misidentified as H. parasuis, these other Penicillin has been considered the drug of choice, but an
NAD-dependent organisms are present in high numbers increasing resistance to penicillin has been reported
in the nasal cavity, tonsils, or lungs and are believed to be (Kielstein and Leirer 1990). Most H. parasuis strains are
of low pathogenic potential (Møller and Kilian 1990; also sensitive in vitro to ampicillin, fluoroquinolone,
Møller et al. 1993). cephalosporins, gentamicin, spectinomycin, and poten-
It is not uncommon for several strains or serovars to tiated sulfas; higher numbers of strains are resistant to
be present in a herd, or even in different specimens from tetracycline, erythromycin, other aminoglycosides, and
a single pig (Smart et al. 1989; Rapp-Gabrielson and lincosamide (Kielstein 1985; Trigo et al. 1996).
Gabrielson 1992; Rapp-Gabrielson 1993). Thus, recov-
ery from systemic sites or gross lesions is the only assur- PREVENTION AND IMMUNITY
ance that the isolate obtained is involved in the disease
process. Serotyping, critical to an understanding of the Because nasal mucosa of baby pigs may be colonized be-
epizootiology of the disease outbreak and immune re- fore 1 week of age, elimination of H. parasuis by early
sponse to infection or vaccination, is available only at a weaning alone is unlikely to be successful. Clark et al.
few diagnostic laboratories. (1994) evaluated several medicated early-weaning strate-
Differential diagnosis should include septicemic bac- gies and found that H. parasuis could be eliminated only
terial infections caused by Streptococcus suis, when parenteral and oral administration of high doses of
Erysipelothrix rhusiopathiae, Actinobacillus suis, Salmonel- antibiotics to baby pigs was a part of the treatment.
la choleraesuis var. kunzendorf, and Escherichia coli. My- Elimination of H. parasuis from a herd may not be desir-
coplasma hyorhinis polyserositis and arthritis in 3- to 10- able, inasmuch as subsequent mixing of naive pigs with
week-old pigs produces lesions similar to H. parasuis. pigs harboring H. parasuis during later stages in produc-
The significance of H. parasuis in bronchopneumonia tion may result in a disease course with devastating eco-

Table 34.3. Differential biochemical reactions of swine NAD-dependent Pasteurellaceae


Other NAD-dependent Pasteurellaceae
Biochemical H. Actinobacillus Actinobacillus Haemophilus Actinobacillus Actinobacillus
Characteristic parasuis pleuropneumoniae minor Taxon C porcinus indolicus
Urease − + + − − −
Hemolysis − + − − − −
Indole − − − − − +
Fermentation of
Glucose + + + + +/− +
Lactose − − + − +/− +/−
Sucrose + + + + +/− +
Mannitol − + − − +/− +/−
Xylose − + +/− − +/− +/−
L-Arabinose − − − + +/− −
Raffinose − − + + +/− +
Sources: Møller and Kilian 1990; Rapp-Gabrielson and Gabrielson 1992; and Møller et al. 1996.
Note: A. minor was formerly known as Haemophilus taxon “Minor Group”; A. porcinus was formerly known as Haemophilus sp. taxons
D and E; A. indolicus was formerly known as Haemophilus sp. taxon F.
Key: + indicates greater than 90% of isolates are positive; −, less than 10% of isolates positive; +/−, variable reactions among isolates.
CHAPTER 34 HAEMOPHILUS PARASUIS Rapp-Gabrielson 479

nomic losses. Introduction of new breeding stock from a inate other respiratory pathogens, tighten weaning age
herd with a different health background should include and pig flow, and eliminate mixing of pigs at all stages of
isolation and acclimation periods long enough to allow production.
for development of protective immunity from either vac-
cination or natural exposure. REFERENCES
Maternal and natural immunity are critical factors
for controlling the disease process (Nielsen and Amano, H.; Shibata, M.; Kajio, N; and Morozumi, T. 1994.
Danielsen 1975). Swine previously exposed to nonpatho- Pathologic observations of pigs intranasally inoculated
genic H. parasuis strains develop resistance to subse- with serovar 1, 4, and 5 of Haemophilus parasuis using
quent challenge with virulent strains (Nielsen 1993). immunoperoxidase method. J Vet Med Sci 56:639–644.
Vaccination of gilts resulted in protective maternal im- Baehler, J. F.; Burgisser, H.; de Meuron, P. A.; and Nicolet, J.
munity for up to 4 weeks when piglets were challenged 1974. Infection à Haemophilus parasuis chez le porc.
with the same serovar contained in the bacterin (Solano Schweiz Arch Tierheilkd 116:183–188.
et al. 1998). Because of the septicemic nature of the dis- Bakos, K; Nilsson, A.; and Thal, E. 1952. Untersuchungen
ease, antibodies are probably the major protective im- über Haemophilus suis. Nord Vet-Med 4:241–255.
mune mechanism. Barigazzi, G.; Valenza, F.; Bollo, E.; Guarda, F.; Candotti, P.;
There are numerous reports of successful disease A. Raffo; and Foni, E. 1994. Anatomohistopathological
control by vaccination with commercial or herd-specific features related to Haemophilus parasuis infection in
bacterins (Nielsen and Danielsen 1975; Riising 1981; pigs. Proc Int Congr Pig Vet Soc 13:235.
Wiseman et al. 1989; Menard and Moore 1990; Schim- Bertschinger, H. U., and Nicod, B. 1970. Untersuchungen
mel et al. 1992). There are also instances where bacterins über die Nasenflora bei Schweinen Vergleich zwischen
are not efficacious, and these may be due to lack of cross- SPF-Herden und schwedisch sanierten Herden. Schweiz
protection for the strain or serovar involved in the dis- Arch Tierheilkd 112:493–499.
ease process. Cross-protection against other virulent Biberstein, E. L., and White, D. C.. 1969. A proposal for the
strains is not always evident in experimental challenge establishment of two new Haemophilus species. J Med
models (Miniats et al. 1991; Kielstein and Raßbach 1991; Microbiol 2:75–78.
Rapp-Gabrielson et al. 1997). Although cross-protection Blackall, P. J.; Rapp-Gabrielson, V. J.; and Hampson, D. J.
is primarily a concern for commercial bacterins, herd- 1996. Serological characterization of Haemophilus para-
specific bacterins may also lack efficacy because of the suis isolates from Australian pigs. Aust Vet J 73:93–95.
presence of more than one strain or serovar or the sub- Blackall, P. J.; Trott, D. J.; Rapp-Gabrielson, V.; and Hamp-
sequent introduction of a new strain into the herd. son, D. J. 1997. Analysis of Haemophilus parasuis by mul-
Demonstration that virulent strains may not protect tilocus enzyme electrophoresis. Vet Microbiol
against challenge with different strains of the same 56:125–134.
serovar, or even against challenge with the homologous Bloch, I. J. M. 1985. Beitrag zur Epidemiologie, Serologie
strain, indicates that the protective antigens may not be und Polyacrylamid-Gelelektrophorese von Haemophilus
identical to the virulence factors or the type-specific anti- parasuis. Diss. Univ Bern.
gens (Kielstein and Raßbach 1991; Rapp-Gabrielson et Clark, L. K.; Hill, M. A.; Kniffen, T. S.; Van Alstine, W.;
al. 1997). Stevenson, G.; Meyer, K. B.; Wu, C. C.; Scheidt, A. B.;
Efficacy against challenge with different strains of the Knox, K.; and Albregts, S. 1994. An evaluation of the
same serovar, as well as cross-protection against some components of medicated early weaning. Swine Health
heterologous serovars, has been demonstrated with a and Production 2:5–11.
bacterin containing H. parasuis serovars 4 and 5 (Rapp- Cooper, V. L.; Doster, A. R.; Hesse, R. A.; and Harris, N. B.
Gabrielson et al. 1996, 1997). However, in these same re- 1995. Porcine reproductive and respiratory syndrome:
ports, evaluation of this bacterin, as well as other com- NEB-1 PRRSV infection did not potentiate bacterial
mercial bacterins, demonstrated that none protected pathogens. J Vet Diagn Invest 7:313–320.
fully against the six most prevalent serovars associated De Ley, J.; Mannheim, W.; Mutters, R.; Piechulla, K.; Tytgat,
with disease in North America. Strains representing nine R.; Segers, P.; Bisgaard, M.; Frederiksen, W.; Hinz, K.-H.;
serovars, as well as nontypeable strains, have been and Vanhoucke, M. 1990. Inter- and intrafamilial simi-
demonstrated to be virulent. Because of the heterogene- larities of rRNS cistrons of the Pasteurellaceae. Int J Sys
ity of strains with pathogenic potential and the current Bacteriol 40:126–137.
lack of understanding of protective antigens and viru- Desrosiers, R.; Phaneuf, J. B.; and Broes, A. 1986. An out-
lence factors, it is unlikely that any bacterin will provide break of atypical Glasser’s disease in Quebec. Proc Int
cross-immunity against all strains of etiologic signifi- Congr Pig Vet Soc 9:277.
cance in the swine population. Control programs may in- Dewhirst, F. E.; Paster, B. J.; Olsen, I.; and Fraser, G. J. 1992.
clude vaccination and antibiotic treatments but also Phylogeny of 54 representative strains of species in the
should address management practices to reduce or elim- family Pasteurellaceae as determined by comparison of
480 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

16S rRNA sequences. J Bacteriol 174:2002–2013. Haemophilus influenzae suis. Ann NY Acad Sci
Glässer, K. 1910. Untersuchungen über die Schweineseuche 79:670–676.
mit besonderer Berücksichtigung ihrer Ätiologie and Little, T. W. A. 1970. Haemophilus infection in pigs. Vet Rec
Pathologie. Dtsch Tierärztl Wschr 18:729–733. 87:399–402.
Gois, M.; Barnes, H. J.; and Ross, R. F. 1983. Potentiation of Madsen P. 1984. Atypical outbreaks of Glässer’s disease in
turbinate atrophy in pigs by long-term nasal coloniza- Danish pig herds. Proc Int Congr Pig Vet Soc 8:107.
tion with Pasteurella multocida. Am J Vet Res Menard, J., and Moore, C. 1990. Epidemiology and man-
44:372–378. agement of Glasser’s disease in SPF herds. In Proc 21st
Harris, D. L.; Ross, R. F.; and Switzer, W. P. 1969. Incidence Annu Meet Am Assoc Swine Pract, Denver, pp.
of certain microorganisms in nasal cavities of swine in 187–200.
Iowa. Am J Vet Res 30:1621–1624. Mendez-Trigo, A. V., and Trigo, E. 1996. Viability of
Hjärre, A. 1958. Enzootic virus pneumonia and Glässer’s Haemophilus parasuis in tissues, exudates and pure cul-
disease of swine. Adv Vet Sci 4:235–263. ture swabs. Proc Int Congr Pig Vet Soc 14:314.
Hjärre, A., and Wramby, G. 1943. Über die fibrinöse Serosa- Miniats, O. P.; Smart, N. L.; and Metzger, K. 1986. Glasser’s
Gelenkentzündung (Glässer) beim Schwein. Z Infek- disease in southwestern Ontario. I. A retrospective
tionskr Parasitenkd Krankheit Hyg Haustiere 60:37–64. study. Proc Int Congr Pig Vet Soc 9:279.
Hoefling, D. C. 1991. Acute myositis associated with Miniats, O. P.; Smart, N. L.; and Rosendal, S. 1991. Cross
Haemophilus parasuis in primary SPF sows. J Vet Diagn protection among Haemophilus parasuis strains in im-
Invest 3:354–355. munized gnotobiotic pigs. Can J Vet Res 55:37–41.
Kielstein, P. 1985. Zur Glässerschen Krankheit und Møller, K., and Kilian, M. 1990. V factor–dependent mem-
Chemotherapeutika-Empfindichkeit ihres Erregers. Mh bers of the family Pasteurellaceae in the porcine upper
Vet-Med 40:801–809. respiratory tract. J Clin Microbiol 28:2711–2716.
———. 1991. Zur Glässerschen Krankheit des Schweines Møller, K.; Andersen, L. V.; Christensen, G.; and Kilian, M.
Untersuchungen über Zusammenhänge zwischen serol- 1993. Optimization of the detection of NAD dependent
ogischen Eigenschaften, Kapselausbildung und Virulenz Pasteurellaceae from the respiratory tract of slaughter-
von Haemophilus-parasuis-Stämmen. Mh Vet-Med house pigs. Vet Microbiol 36:261–271.
46:137–142. Møller, K.; Fussing, V.; Grimont, P. A. D.; Paster, B. J.;
Kielstein, P., and Leirer, R. 1990. Zur Glässerschen Dewhirst, F. E.; and Kilian, M. 1996. Actinobacillus minor
Krankheit des Schweines—Ätiologisch-epizootiologis- sp. nov., Actinobacillus porcinus sp. nov., and Actinobacil-
che Untersuchungen zum Erregerspektrum. Mh Vet- lus indolicus sp. nov., three new V factor–dependent
Med 45:577–582. species from the respiratory tract of pigs. Int J Syst Bac-
Kielstein, P., and Rapp-Gabrielson, V. J. 1992. Designation teriol 46:951–956.
of 15 serovars of Haemophilus parasuis based on im- Morikoshi, T.; Kobayashi, K.; Kamino, T.; Owaki, S.;
munodiffusion using heat-stable antigen extracts. J Clin Hayashi, S.; and Hirano S. 1990. Characterization of
Microbiol 30:862–865. Haemophilus parasuis isolated in Japan. Jpn J Vet Sci
Kielstein, P., and Raßbach, A. 1991. Serologische Typ- 52:667–669.
isierung und Nachweis immunogener Kreuzreaktionen Morozumi, T., and Nicolet, J. 1986a. Morphological varia-
von Haemophilus parasuis (Glässersche Krankheit). Mh tions of Haemophilus parasuis strains. J Clin Microbiol
Vet-Med 46:586–589. 23:138–142.
Kilian, M. 1976. A taxonomic study of the genus ———. 1986b. Some antigenic properties of Haemophilus
Haemophilus, with the proposal of a new species. J Gen parasuis and a proposal for serological classification. J
Microbiol 93:9–62. Clin Microbiol 23:1022–1025.
Kobayashi, H.; Morozumi, T.; Miyamoto, C.; Shimizu, M.; Morozumi, T.; Pauli, U.; Braun, R.; and Nicolet, J. 1986. De-
Yamada, S.; Ohashi, S.; Kubo, M,; Kimura, K.; Mitani, oxyribonucleic acid relatedness among strains of
K.; Ito, N.; and Yamamota, K. 1996. Mycoplasma hyorhi- Haemophilus parasuis and other Haemophilus spp. of
nis infection levels in lungs of piglets with porcine re- swine origin. Int J System Bacteriol 36:17–19.
productive and respiratory syndrome (PRRS). J Vet Med Münch, S.; Grund, S.; and Krüger, M. 1992. Fimbriae and
Sci 58:109–113. membranes on Haemophilus parasuis. Zentralbl Veterin-
Kobisch, M., and Desmettre, P. 1980. Hemophilose du porc: armed (B) 39:59–64.
Pouvoir pathogène expérimental de deux souches Narita, M.; Kawashima, K.; Matsuura, S.; Uchimura, A.; and
d’Haemophilus parasuis. Rec Méd Vét 156:219–224. Miura, Y. 1994. Pneumonia in pigs infected with
Kott, B. E. 1983. Chronological Studies of Respiratory Dis- pseudorabies virus and Haemophilus parasuis serovar 4. J
ease in Baby Pigs. M.S. thesis, Iowa State Univ, Ames. Comp Path 110:329–339.
Lecce, J. G. 1960. Porcine polyserositis with arthritis: Isola- Nicolet, J.; Morozumi, T.; and Bloch, I. 1986. Proposal for a
tion of a fastidious pleuropneumonialike organism and serological classification of H. parasuis. Proc Int Congr
CHAPTER 34 HAEMOPHILUS PARASUIS Rapp-Gabrielson 481

Pig Vet Soc 9:260. PAGE Proteinmustern von Haemophilus parasuis. Dtsch
Nielsen, R. 1993. Pathogenicity and immunity studies of Tierärztl Wschr 98:327–330.
Haemophilus parasuis serotypes. Acta Vet Scand Schimmel, D.; Kielstein, P.; and Hass, R. 1985. Zur serolo-
34:193–198. gische Typisierung von Haemophilus parasuis. Arch Exp
Nielsen, R., and Danielsen, V. 1975. An outbreak of Gläss- Vet Med, Leipzig, 39:944–947.
er’s disease: Studies on etiology, serology and the effect Schimmel, D.; Hogg, A.; and Uhlemann, J. 1992. Prevention
of vaccination. Nord Vet-Med 27:20–25. of Haemophilus parasuis infection with an inactivated
Peet, R. L.; Fry, J.; Lloyd, J.; Henderson, J.; Curran, J.; and vaccine. Proc Int Congr Pig Vet Soc 12:336.
Moir, D. 1983. Haemophilus parasuis septicaemia in pigs. Smart, N. L.; Miniats, O. P.; and MacInnes, J. I. 1988. Analy-
Aust Vet J 60:187. sis of Haemophilus parasuis isolates from southern On-
Pöhle, D.; Johannsen, U.; Kielstein, P.; Raßbach, A.; and tario swine by restriction endonuclease fingerprinting.
Wiegand, M. 1992. Investigations on pathology and Can J Vet Res 52:319–324.
pathogenesis of Haemophilus parasuis infection of Smart, N. L.; Miniats, O. P.; Rosendal, S.; and Friendship, R.
swine. Proc Int Congr Pig Vet Soc 12:335. 1989. Glasser’s disease and prevalence of subclinical in-
Rapp, V. J.; Ross, R. F.; and Nicolet, J. 1986. Characterization fection with Haemophilus parasuis in swine in southern
of the outer membrane proteins of Haemophilus parasu- Ontario. Can Vet J 30:339–343.
is. Proc Int Congr Pig Vet Soc 9:262. Solano, G.; Segales, J.; Collins, J.; Molitor, T.; and Pijoan, C.
Rapp-Gabrielson, V. J. 1993. Haemophilus parasuis: The as- 1997. Porcine reproductive and respiratory syndrome
sociation of serovar with prevalence, pathogenicity, and virus (PRRSv) interaction with Haemophilus parasuis. Vet
immunogenicity. Proc Allen D Leman Swine Conf, St. Microbiol 55:247–257.
Paul, 20:31–33. Solano, G.; Pijoan, C.; Rapp-Gabrielson, V.; Collins, J.; Car-
Rapp-Gabrielson, V. J., and Gabrielson, D. A. 1992. Preva- valho, L. F.; and Winkelman, N. 1998. Protective role of
lence of Haemophilus parasuis serovars among isolates maternal antibodies against Haemophilus parasuis infec-
from swine. Am J Vet Res 53:659–664. tion. Am J Vet Res. In press.
Rapp-Gabrielson, V. J.; Gabrielson, D. A.; and Musser, J. M. Trigo, E.; Mendez-Trigo, A. V.; and Simonson, R. 1996. An-
1992a. Phenotypic and genotypic diversity of timicrobial susceptibility profiles of Haemophilus para-
Haemophilus parasuis. Proc Int Congr Pig Vet Soc 12:334. suis: A retrospective study from clinical cases submitted
Rapp-Gabrielson, V. J.; Gabrielson, D. A.; and Schamber, G. during 1994 and 1995 to a veterinary diagnostic labora-
J. 1992b. Comparative virulence of Haemophilus parasu- tory. Proc Int Congr Pig Vet Soc 14:313.
is serovars 1 to 7 in guinea pigs. Am J Vet Res Vahle, J. L. 1996. Pathogenesis of Haemophilus parasuis in-
53:987–994. fection in swine. Ph.D. diss., Iowa State Univ, Ames.
Rapp-Gabrielson, V. J.; Kocur, G. J.; Clark, J. T.; and Muir, S. Vahle, J. L.; Harness, J. S.; and Andrews, J. J. 1995. Experi-
K. 1995. Virulence of different serovars of Haemophilus mental reproduction of Haemophilus parasuis infection
parasuis for cesarean-derived, colostrum-deprived pigs. in swine: Clinical, bacteriological, and morphologic
In Haemophilus, Actinobacillus, and Pasteurella. Ed. W. findings. J Vet Diagn Invest 7:476–480.
Donachie, F. A. Lainson, and J. C. Hodgson. New York: Vahle, J. L.; Haynes, J. S.; and Andrews, J. J. 1997. Interaction
Plenum Press, p. 204. of Haemophilus parasuis with nasal and tracheal mucosa
Rapp-Gabrielson, V. J.; Kocur, G.; Clark, J.; Muir, S.; and following intranasal inoculation of cesarean derived
White, R. 1996. Efficacy and duration of immunity of colostrum deprived (CDCD) swine. Can J Vet Res
the Haemophilus parasuis fractions of Suvaxyn® 61:200–206.
RespiFend® MH/HPS. Proc Int Congr Pig Vet Soc Wiseman, B.; Harris, D. L.; Glock, R. D.; and Wilkins, L.
14:300. 1989. Management of seedstock that is negative for
Rapp-Gabrielson, V. J.; Kocur, G. J.; Clark, J. T.; and Muir, S. Haemophilus parasuis. In Proc 20th Annu Meet Am As-
K. 1997. Haemophilus parasuis: Immunity in swine fol- soc Swine Pract, Des Moines, pp. 23–25.
lowing vaccination. Vet Med 92:83–90. Zucker, B.; Krüger, M.; Rehak, E.; and Horsch, F. 1994. Un-
Riising, H.-J. 1981. Prevention of Glässer’s Disease through tersuchungen zur Lipopolysaccharidestruktur von
Immunity to Haemophilus parasuis. Zentralbl Veterin- Haemophilus parasuis–Stämmen in der SDS-PAGE. Berl
armed (B) 28:630–638. Münch Tierärztl Wschr 107:78–81.
Riley, M. G. I.; Russell, E. G.; and Callinan, R. B. 1977. Zucker, B. A.; Baghian, A.; Truax, R.; O’Reilly, K. L.; and
Haemophilus parasuis infection in swine. J Am Vet Med Storz, J. 1996. Detection of strain-specific antigenic epi-
Assoc 171:649–651. topes of the lipo-oligosaccharide of Haemophilus parasu-
Rosner, H.; Kielstein, P.; Müller, W.; and Rohrmann, B. is by use of monoclonal and polyclonal antibodies. Am J
1991. Beziehung zwischen Serotyp, Virulenz und SDS- Vet Res 57:63–67.
Leptospirosis
35 W. A. Ellis

Leptospirosis is a cause of reproductive loss in breeding lope, which surrounds a cell wall or peptidoglycan com-
herds and has been reported in swine from all parts of plex, and two polar endoflagella (originating subtermi-
the world; however, knowledge of the incidence and eco- nally at each end).
nomic impact of the disease is largely confined to the in- The taxonomy of the leptospires has been through a
tensive pig industries of the Northern Hemisphere, Aus- period of change which continues to cause considerable
tralia, New Zealand, Argentina, and Brazil. confusion to those not intimately acquainted with the
Endemic infection in a herd of swine may produce lit- subject. Until recently a single genus Leptospira was rec-
tle evidence of clinical disease, but when it is first intro- ognized in the family Leptospiraceae. Two groupings were
duced into a susceptible breeding herd, or during peri- recognized within the genus: those found in animal
ods of waning herd immunity, it can cause very species (the parasitic strains) and those found in water
appreciable losses through abortion, the full-term birth (the saprophytic strains). These two groupings, which
of dead pigs or of weak pigs of reduced viability, or in- were referred to as the interrogans and biflexa complexes,
fertility. can be differentiated by their growth requirements and
Leptospires persist in the kidneys and genital tracts biochemical reactions. Only the parasitic strains are of
of carrier swine and are excreted in urine and genital flu- medical and veterinary interest.
ids. Survival outside the host is favored by warm, moist For taxonomic purposes and as an aid to epidemio-
conditions. Transmission is by direct or indirect contact logical studies, the parasitic leptospires were divided in-
with a carrier animal. to serogroups on the basis of antigenic relationships as
Interruption of transmission from an infected pig or determined by cross-agglutination reactions and were
other host to the pig is the critical factor in control. further subdivided into serovars by agglutination-ab-
Leptospirosis is an occupational zoonosis of those sorption patterns. About 23 serogroups are recognized,
who work with pigs. containing approximately 212 serovars.
The advent of genetic typing methods has provided
ETIOLOGY rapid, reproducible typing protocols. The current recom-
mendations on the taxonomy of leptospires (Ellis 1995)
Leptospirosis of swine is a disease caused by a variety of recognized eight species of pathogenic leptospires with-
morphologically similar, but antigenically and genetical- in the family Leptospiraceae. They are Leptospira interro-
ly distinct, small, motile, aerobic spirochetes belonging gans, L. borgpetersenii, L. inadai, L. kirschneri, L. noguchii,
to the genus Leptospira. They are thin, helical, motile, L. meyer, L. weilii, and L. santarosai.
gram-negative organisms that are often hooked at one or The species definition is based on a level of DNA-
both ends. They spin constantly on their long axis. They DNA homology of at least 70% and 5% or less divergence
range in length from about 6 to 20 µm, with an ampli- in DNA relatedness. Taxonomy at the subspecific level
tude of approximately 0.1–0.15 µm and a wavelength of continues to be based on serovars as defined by Dikken
about 0.5 µm. Under adverse nutritional conditions, lep- and Kmety (1978), but other valid methods which give
tospires may be greatly elongated, whereas under condi- results comparable to conventional serotyping can be
tions such as high salt concentrations or in aging culture used for their identification. Such methods include mon-
or tissues, leptospires may form coccoid forms of about oclonal antibody agglutination profiles, factor analysis,
1.5–2 µm in diameter (Faine 1994). They divide by bina- and analyses in which restriction-fragment length poly-
ry fission. They stain poorly with aniline dyes. Unstained morphisms or rRNA gene restriction patterns are used in
cells are visible only by dark-field microscopy. In a suit- pulsed-field gel electrophoresis analyses. The term “type”
able liquid environment, motility is accomplished by ro- is used to indicate strain differences at the subserovar
tating along the long axis, but this changes to an undu- level (Ellis 1995).
lating action in semisolid media. They require special
media containing mammalian serum or albumin for cul- Molecular Biology
tivation. The genus Leptospira is characterized by a guanine plus
The major structural components are an outer enve- cytosine (G + C) ratio of 35–41 mol% in its chromosomal

483
484 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

DNA, depending on species. The published genome size more, not all strains of serovar pomona are adapted to
has varied between 3100 and 5000 kb depending on the pigs nor are the other serovars of the Pomona serogroup,
techniques used to measure it and reflecting differences but they have rodent hosts (Sebek et al. 1983).
between strains. The Leptospira interrogans serovars In parts of North America, the prevalence of pomona
icterohaemorrhagiae and pomona have two circular chro- infection in pigs has fallen from the high levels observed
mosomes: the large (4400–4600 kb) and the small (350 in the 1950s and early 1960s: no carriers were detected in
kb) replicons are regarded as chromosomal because the a 1989 meat-plant survey carried out in Iowa (Bolin and
essential asd gene is located on the smaller unit. Leptospi- Cassells 1992). In contrast, Baker et al. (1989) recovered
ra interrogans strains contain two 23S and 16S rRNA serovar pomona (type kennewicki) from almost 10% of
genes but only one 5S rRNA gene. The 5S rRNA gene is pigs in a small survey in Canada.
highly conserved among the pathogenic leptospires. Leptospires have a particular affinity for the kidneys
There are differences in the global distribution of of infected pigs, where they persist, multiply, and are
some of the Leptospira species: L. interrogans, L. borg- voided in urine. This characteristic is very important in
petersenii, and L. kirschneri have a worldwide distribu- the transmission of infection.
tion, whereas L. noguchii and L. santarosai are found Infection is introduced into a susceptible herd by
mainly in North and South America, and L. weilii is three possible routes: the introduction of infected stock,
found mainly in China and eastern Asia. Strains that exposure to a contaminated environment, or contact
cause disease in pigs are found mainly in the L. interro- with an alternative infected animal vector (Hathaway
gans and L. borgpetersenii species. 1983). Carrier pigs are probably the most common route
of introduction. Replacement gilts (Edwards and Daines
EPIDEMIOLOGY 1979) or infected boars (Kemenes and Suveges 1976)
have been identified as important means of introducing
The epidemiology of swine leptospirosis is potentially infection.
very complicated, since swine can be infected by any of The importance of free-living species as possible
the pathogenic serovars. Fortunately, only a small num- sources of pomona infection of pigs depends on geo-
ber of serovars will be endemic in any particular region graphical location. In North America, the skunk has
or country. Furthermore, leptospirosis is a disease that been incriminated as a source of pomona outbreaks in
shows a natural nidality, and each serovar tends to be pigs (Mitchell et al. 1966).
maintained in specific-maintenance hosts. Therefore, in Once pomona has been introduced into a pig popula-
any region, pigs will be infected by serovars maintained tion, a high prevalence of infection is established. Only
by pigs or by serovars maintained by other animal low infective doses are required to transmit infection
species present in the area. The relative importance of (Chaudhary et al. 1966a, b). If direct contact is prevent-
these incidental infections is determined by the opportu- ed, indirect contact through contaminated effluent, wa-
nity that prevailing social, management, and environ- ter, or soil ensures transmission (Michna 1970; Buddle
mental factors provide for contact and transmission of and Hodges 1977; Kingscote 1986). The presence of
leptospires from other species to pigs. moisture is critical for indirect transmission; the organ-
Pigs act as maintenance hosts for serovars belonging isms cannot withstand dessication, but when infected
to the Pomona, Australis, and Tarassovi serogroups, urine is deposited in damp soil or water with a pH
while strains belonging to the Canicola, Icterohaemor- around or slightly on the alkaline side, the organisms
rhagiae, and Grippotyphosa serogroups are among the may survive for extended periods (Mitscherlich and
more commonly identified incidental infections in Marth 1984).
swine. During the initial herd infection, clinical disease may
occur in all ages of sows.
Pomona Infection Following the initial establishment of infection, an
Serovar pomona has been the most common serovar iso- endemic cycle typical of that in a maintenance host pop-
lated from pigs worldwide. Infection with this serovar ulation is set up (Hathaway 1981). Piglets are passively
has been extensively studied and it provides a suitable protected in the first weeks of life by colostrum-derived
model with which to illustrate general concepts of swine immunoglobulins from infected dams (Fish et al. 1963;
leptospirosis. Many strains of serovar pomona, especially Bolt and Marshall 1995a). The duration of this passive
those of type kennewicki found in the United States and protection depends primarily on the quantity of im-
Canada, are adapted to swine. Serovar pomona has been munoglobulins received in colostrum (Chaudhary et al.
the cause of widespread clinical disease in swine in 1966b). A study of grower pigs in New Zealand has
North and South America, Australia, New Zealand, parts shown that leptospiral infection becomes apparent in
of Asia, and Eastern and Central Europe and is endemic piglets from 12 weeks of age, and by slaughter up to 90%
in many of these regions. Such strains are apparently ab- may be infected. The intensity of leptospiruria is greatest
sent from the more westerly parts of Europe. Further- in the first 3–4 weeks of infection, after which it declines
CHAPTER 35 LEPTOSPIROSIS Ellis 485

and becomes intermittent (Bolt and Marshall 1995a, b). demically infected herds. In indoor sow units infected
Infection between groups of fattening pigs is often by with pig-adapted strains of bratislava, the prevalence of
urine-contaminated effluent from a common drainage sows with antibody titers of greater than 1:100 in the mi-
system (Buddle and Hodges 1977). croscopic agglutination test (MAT) is usually very low, al-
In herds with endemic infection, clinical disease is though many sows will have titers of less than 1:100.
usually restricted to gilts that either have been reared in This is thought to result from infection being primarily
isolation since weaning and reintroduced into the herd due to venereal transmission. In contrast, in units where
or, more commonly, have been brought in from an unin- the sows are kept outside, the seroprevalences (≥1:100)
fected herd. may be greater than 50%. This is thought to be due to the
sows being infected systemically as a result of exposure
Tarassovi Infection to infected rodent urine.
There is much less information available on the epidemi- Although the renal-carrier state does become estab-
ology of tarassovi infection in pigs. The pig acts as a lished, urinary excretion is poor compared with pomona
maintenance host for some strains of tarassovi found in excretion, and transmission within the fattening house is
Eastern Europe and Australia. In these regions, it does inefficient. Important additional carrier sites have been
not spread as rapidly in a pig population as does pomona identified, namely, the upper genital tracts of sows and
(Kemenes and Suveges 1976), but endemic infection is boars (Ellis et al. 1986b, c; Power 1991; Bolin and Cassells
readily maintained (Ryley and Simmons 1954b; 1992). Venereal transmission is thought to play an im-
Kemenes and Suveges 1976). portant role in the spread of bratislava infection.
Many strains of tarassovi have been recovered from
free-living animals (Anon. 1966, 1975) and these may Canicola Infection
give rise to incidental pig infections. For example, Although organisms belonging to the Canicola sero-
tarassovi has not been recovered from swine in the Unit- group have been recovered from swine in at least 11
ed States, but there is serologic evidence of infection in countries (Hanson and Tripathy 1986), little is known of
pigs (Cole et al. 1983) in the southeastern states, where it the epidemiology of canicola infection in pigs. The dog is
has been isolated from racoons, skunks, and opossums the recognized maintenance host for this serovar and is
(McKeever et al. 1958; Roth 1964). the probable vector whereby this serovar enters a pig-
gery, although a report from Peru (Paz-Soldan et al.
Australis Infection 1991) incriminates wildlife as the source of an outbreak
Serovar bratislava and to a lesser extent the closely relat- in sows. The long period of leptospiruria observed in in-
ed serovar muenchen have emerged as major swine-main- fected pigs (at least 90 days) (Michna 1962) and the abil-
tained leptospiral infections in the last few years. Sero- ity of canicola to survive for up to 6 days in undiluted pig
logic data have indicated widespread bratislava infection urine (Michna 1962) suggest that there would be an op-
in Germany (Weber and Fenske 1978), the United King- portunity for intraspecies transmission, but no studies
dom (Hathaway and Little 1981; Hathaway et al. 1981), have been done on this subject (Hathaway 1983).
Czechoslovakia (Propopcakova et al. 1981), the Nether-
lands (Bercovich et al. 1983), Sweden (Sandstedt and Icterohaemorrhagiae Infection
Engvall 1985), Denmark (Jensen and Binder 1989; Serologic evidence of Icterohaemorrhagiae serogroup in-
Nissen 1989), the United States (Hanson 1985, 1987), fection has been reported in many countries but few iso-
Canada (Kingscote 1988), Austria (Loimayr 1990), Aus- lations have been made from pigs (Hathaway 1985). It
tralia (Chappel et al. 1992), Brazil (Oliveira et al. 1994), appears that both serovars copenhageni and icterohaemor-
and South Africa (Potts et al. 1995). There is, as yet, no rhagiae may be involved. The maintenance host for these
information from Russia but it would be reasonable to serovars is the brown rat (Rattus norvegicus), and it is
assume that infection is now present in all major pig- probable that copenhageni and icterohaemorrhagiae are in-
rearing countries. troduced to susceptible stock via an environment conta-
Serovar bratislava was first recovered from a pig in minated with infected rat urine. Field investigation sug-
the Netherlands by Hartmann et al. (1975) (Ellis 1992) gests that transmission between swine is inefficient
and has now been recovered from pigs in the United (Hathaway 1985). Schnurrenberger et al. (1970) found
Kingdom (Ellis et al. 1986a, b, c), the United States (Ellis that urinary excretion lasted less than 35 days in natural-
and Thiermann 1986; Bolin and Cassells 1990, 1992), ly infected pigs, while Fennestad and Borg-Petersen
and Germany (Schonberg et al. 1992). (1966) failed to demonstrate leptospiruria in experimen-
The epidemiology of these strains is poorly under- tally infected pigs. Low prevalences of renal infection
stood. There are specific pig-adapted strains, strains that have been found in those microbiological surveys in
are maintained by pigs, dogs, horses, and hedgehogs, which Icterohaemorrhagiae strains have been recovered;
and strains that are found only in wildlife. Hathaway et al. (1981) reported a 0.7% prevalence in
Two very distinct serologic profiles may be seen in en- England, and McErlean (1973) found a 0.4% prevalence
486 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

in Ireland. It is believed, in the absence of supporting up to 3 weeks, after which a gradual decline occurs. Low
isolation data, that vaccine-induced antibodies are re- titers may be detectable for several years in many ani-
sponsible for the seroprevalences of icterohaemorrhagiae mals.
observed in the United States. Following the period of leptospiremia, the lep-
tospires localize in the proximal renal tubules, where
Grippotyphosa Infection they multiply and are voided in the urine. The duration
Serovar grippotyphosa infection is maintained by wildlife and intensity of urinary shedding vary from pig to pig
hosts, and incidental infection of pigs gives rise to low and with the infecting serovar. In the case of pomona in-
prevalences of antibodies in swine in various regions, fection, the intensity of excretion is highest during the
particularly Eastern and Central Europe and the United first month of shedding, when more than a million lep-
States. It has been recovered from pigs in the former tospires may be present in each milliliter of urine (Morse
USSR (Gorshanova 1964) and the United States (Hanson et al. 1958); leptospiruria is very constant during this pe-
et al. 1965, 1971). riod (Hodges et al. 1979). A variable period of intermit-
tent, low-intensity leptospiruria then ensues, and this
Sejroe Infection may last for up to 2 years in some cases (Ryley and Sim-
Serovar hardjo infection is maintained by cattle world- mons 1954a; Morse et al. 1958; Mitchell et al. 1966). Low
wide, and where cattle and pigs come in close contact, levels of antibody may be detected in the urine of pigs
the opportunity arises for infection in pigs to occur. (Morse et al. 1958), but the immunologic mechanism
There are now reports of the isolation of hardjo from whereby infection is ultimately eliminated from the kid-
pigs in the United Kingdom (Hathaway et al. 1983; Ellis neys is not known.
et al. 1986a) and the United States (Bolin and Cassells Leptospires also localize in the uterus of pregnant
1992). Persistence in renal tissue was not a feature of ex- sows, and abortion, production of stillborn pigs, and
perimental infection (Hathaway et al. 1983); therefore, neonatal disease frequently result from intrauterine in-
intraspecies transmission is unlikely. fections occurring in the last half of the gestation period.
Serovar sejroe, which is maintained by small rodents, Abortions and stillbirths usually occur 1–4 weeks follow-
has also been isolated from swine in Europe (Brandis ing infection of the gilt or sow (Hanson and Tripathy
1956; Fuzi et al. 1957; Combiesco et al. 1958), and an- 1986), by which time most sows have developed de-
other serovar in this group (serovar balcanica) has been tectable antibody titers. Since pig fetuses are capable of
recovered from swine in the former USSR (Matveeva et producing antibodies during the latter stages of gesta-
al. 1977). tion, some stillborn piglets will have detectable titers.
The pathogenesis of reproductive disease is poorly
PATHOGENESIS understood, but some authors believe that transplacen-
tal infection, occurring during the very limited period of
The most important route of natural infection has not maternal leptospiremia, is the sole cause (Fennestad and
been determined; however, it is thought to be via the mu- Borg-Petersen 1966). While this may be true for systemic
cous membranes of the eye, mouth, or nose (Alston and infections such as pomona, the low antibody titers detect-
Broom 1958; Alexander et al. 1964; Michna and Camp- ed in sows aborting bratislava-infected fetuses has led to
bell 1969). Infection via the vaginal route is also possible the hypothesis that infection occurs as a result of waning
(Ferguson and Powers 1956; Chaudhary et al. 1966a). uterine immunity being unable to prevent transplacental
Transmission of leptospires through milk from an in- infection by leptospires present in the genital tract. The
fected dam has been demonstrated experimentally (Tri- possibility of transplacental infection during lep-
pathy et al. 1981). A period of bacteremia, which may tospiremia appears to increase with the stage of preg-
last for a week, begins 1 or 2 days after infection. During nancy (Wrathall 1975). From midpregnancy onward, it
this period leptospires can be isolated from most organs is likely that the majority of fetuses in a litter at risk will
of the body and also from cerebrospinal fluid. This pri- become infected. Fennestad and Borg-Petersen (1966)
mary bacteremic phase ends with the appearance of cir- have suggested that horizontal transmission to litter-
culating antibodies, which are detectable usually after mates not infected during the period of maternal lep-
5–10 days (Hanson and Tripathy 1986). A secondary bac- tospiremia may also occur. Once the placental barrier is
teremic period (after 15–26 days) has been reported in breached, septicemia results in large numbers of lep-
experimental hardjo infection (Hathaway et al. 1983). tospires in all fetal tissues (Preston and Morter 1960). It
Antileptospiral agglutinins appear at detectable lev- is unlikely that placental insufficiency plays a role in fetal
els in the blood at approximately 5–10 days after infec- death (Wrathall 1975); abortion is probably initiated by
tion and reach maximum levels at around 3 weeks (Ryley toxic products released from dead and autolyzing fetus-
and Simmons 1954b; Ferguson and Powers 1956; Morse es.
et al. 1958). Peak titers vary considerably (1:1000 to An additional feature seen in bratislava infection but
1:100,000 in the MAT), and these may be maintained for not reported for the other swine leptospiral infections is
CHAPTER 35 LEPTOSPIROSIS Ellis 487

the persistence of leptospires in the oviduct and uterus pregnant sows aborted, while the number of dead
of nonpregnant sows (Ellis et al. 1986c; Ellis and Thier- piglets/sow rose from 8% prior to the outbreak to 28%
mann 1986; Bolin and Cassells 1992) and in the genital during the outbreak. Differences in strain pathogenicity
tracts of boars (Ellis et al. 1986b). also contribute to different prevalences of clinical abor-
tion in infected herds (Nagy 1993).
CLINICAL SIGNS A very high prevalence of serovars belonging to the
Australis serogroup has been observed in aborted pig lit-
The vast majority of swine leptospiral infections are sub- ters in part of the United Kingdom. Ellis et al. (1986a)
clinical. Two groups of pigs are most likely to experience isolated either serovar bratislava or muenchen from 71%
clinical infections: the young piglet and the pregnant of the litters they examined. Similar strains have also
sow. been recovered from aborted piglets in the United States
(Bolin and Cassells 1990), where a high prevalence in
Acute Leptospirosis aborted fetuses has also been noted (Bolin et al. 1991).
This phase usually coincides with the period of bac- Rehmtulla et al. (1992) reported fetal bratislava infection
teremia (Morse et al. 1958; Sleight and Lundberg 1961; following abortions in 16% of sows in a herd in Ontario.
Chaudhary et al. 1966a, b). In experimental infections, Egan (1995) reported fluorescent antibody test (FAT)–
many pigs exhibit transient anorexia, pyrexia, and list- positive prevalences ranging from 5% to 23% in diagnos-
lessness at this time (Hanson and Tripathy 1986). How- tic submissions in Ireland. Published experimental eval-
ever, the mild nature of these signs means that in natur- uations of the significance of such microbiological find-
al infections, especially in endemically infected herds ings are not available. There has, however, been an
where perhaps only one or two animals may be affected, absence of significant isolations of other abortifacient
this phase of infection usually goes unrecognized. agents from these cases, and the farrowing rate and the
There have been a few reports of jaundice and hemo- number of live piglets born/sow improve significantly
globinuria in naturally occurring outbreaks (Ferguson et following either bratislava vaccination (Frantz et al.
al. 1956), particularly in cases of infection in piglets un- 1989) or the use of an antibiotic medication program (El-
der 3 months of age by strains belonging to the Ictero- lis 1989). Van Til and Dohoo (1991) failed to demon-
haemorrhagiae serogroup (Klarenbeek and Winsser strate a relationship between positive serology and still-
1937; Field and Sellers 1951; Urban and Androsov 1976). births.
A high proportion of these undergo spontaneous recov- Following abortions due to pomona, there does not
ery within a week of when symptoms develop. The small appear to be any subsequent limitation on reproductive
number of such reports suggests that this more severe performance, even in pigs that remain infected for long
form of disease is rare. periods (Ferguson and Powers 1956; Mitchell et al. 1966;
Kemenes and Suveges 1976).
Chronic Leptospirosis Infertility is a feature of bratislava infection. An
Abortions, stillbirths, and the birth of weak piglets of re- analysis of serologic and clinical data by Hathaway and
duced viability are primary signs of chronic leptospiro- Little (1981) has shown a statistically significant relation-
sis, particularly pomona infection, in pigs (Bohl et al. ship between Australis serogroup titers and infertility in
1954; Fennestad and Borg-Petersen 1966) and it is this sows. Similar associations have been observed by Jensen
aspect of leptospirosis that can cause considerable eco- and Binder (1989) and Van Til and Dohoo (1991). Split-
nomic loss. Weak litters have been reported as a feature herd trials, carried out using a bratislava bacterin, have
of Icterohaemorrhagiae infection (Neto et al. 1997). demonstrated significant improvements in sow fertility
Information as to the importance of leptospirosis as (Frantz et al. 1989).
a cause of abortion in national swine herds is not avail-
able, and if it were, it must vary from country to country LESIONS
depending on prevalence, epidemiological, and manage-
ment factors, including the implementation of control The main pathological changes are essentially the same
measures. From the limited information available, it for all infections, with the primary lesion being damage
would appear that even in countries where vaccination to the membranes of the endothelial cells of small blood
has been widely practiced, leptospirosis is a common vessels.
cause of swine abortion. In Ontario, for example, 6% of In acute leptospirosis there are no pathognomonic
swine abortions were attributed to pomona infection gross changes. Pathological changes in acute pomona in-
(Anon. 1986). Endemic tarassovi infection was consid- fection are very limited, reflecting the mild nature of
ered to be the cause of a 3% abortion rate in herds in acute clinical disease. Hanson and Tripathy (1986) re-
Poland investigated by Wandurski (1982). Acute out- ported little gross or histopathological change in swine
breaks can still give rise to severe losses; Saravi et al. killed during the acute phase of leptospirosis. Burnstein
(1989) described an outbreak in a herd in which 19% of and Baker (1954) reported that petechial and ecchymot-
488 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

ic hemorrhages could be seen in the lungs of some pigs, eradication program on either a herd or a national basis;
and histological examinations have revealed minor renal (2) epidemiological studies; or (3) a determination of the
tubular damage, focal liver necrosis, lymphocytic infil- infectivity status of an individual animal to assess its
tration of the adrenal glands, and meningoencephalitis suitability for international trade or for introduction in-
with perivascular lymphocytic infiltration (Burnstein to an uninfected herd.
and Baker 1954; Sleight et al. 1960; Chaudhary et al. The mild, often inapparent, clinical signs of acute
1966a). leptospirosis make clinical diagnosis difficult; therefore,
In chronic leptospirosis, lesions are confined to the diagnosis is usually based on the results of laboratory
kidneys and consist of scattered small gray foci, often procedures.
surrounded by a ring of hyperemia. Microscopic exami- Laboratory diagnostic procedures for leptospirosis
nation shows these lesions to be a progressive focal in- fall into two groups. The first group consists of tests for
terstitial nephritis (Burnstein and Baker 1954; Langham antibody detection; the second contains the tests for the
et al. 1958; Cheville et al. 1980). The interstitial leukocyt- demonstration of leptospires in pig tissues. The selection
ic infiltrations, which consist mainly of lymphocytes, of tests to be carried out depends on the purpose for
macrophages, and plasma cells, may be extensive in which a diagnosis is to be made and the resources avail-
some areas. Focal damage may also involve glomeruli able.
and renal tubules. Some affected glomeruli are swollen,
some atrophic, and others are replaced by fibrosis. The Serologic Tests
Bowman’s capsule may be thickened, containing Serologic testing is the most widely used method for di-
eosinophilic granular material (Langham et al. 1958). agnosing leptospirosis, and the MAT (Cole et al. 1980;
Tubular changes involve atrophy, hyperplasia, and the Faine 1982) is the standard serologic test. The minimum
presence of necrotic debris in the lumen in some areas. antigen requirements are that the test should employ
Occasionally, petechial hemorrhages may be present in representative strains of all the serogroups known to ex-
interstitial spaces. ist in the particular country, plus those known to be
Older lesions mainly consist of fibrosis and intersti- maintained by pigs elsewhere.
tial infiltration. Chronic lesions with accompanying The MAT is used primarily as a herd test. To obtain
acute inflammatory changes are still noticeable as long useful information, at least 10 animals or 10% of the
as 14 months postinfection (Morter et al. 1960). herd, whichever is the greater (Cole et al. 1980), should
Experimental studies indicate leptospires can invade be tested. A retrospective diagnosis of both acute lep-
the mammary gland of pigs and produce a mild, focal tospirosis and pomona abortion may be made when the
nonsuppurative mastitis (Tripathy et al. 1981). majority of affected animals have titers of 1:1000 or
The gross pathology of fetuses aborted as a sequela of greater. Increasing the sample size and sampling a num-
pomona infection is nonspecific and includes edema of ber of different cohorts markedly improve epidemiolog-
various tissues, serous or bloodstained fluid in body cav- ical information, investigations of clinical disease, as-
ities, and sometimes petechial hemorrhages in the renal sessments of vaccination needs, and public health
cortex (Ryley and Simmons 1954b; Fennestad and Borg- tracebacks.
Petersen 1966; Wrathall 1975). These changes are proba- As an individual-animal test, the MAT is very useful
bly the result of intrauterine autolysis. Jaundice may be in diagnosing acute infection; rising antibody titers in
seen in some aborted piglets (Hathaway et al. 1983). Fo- paired acute and convalescent serum samples are diag-
cal necrosis, presenting as small grayish-white spots, is a nostic. The presence of antibody in fetal serum is diag-
frequent finding in the liver (Ryley and Simmons 1954b; nostic of leptospiral abortion.
Fish et al. 1963; Fennestad and Borg-Petersen 1966). His- The MAT has severe limitations in the diagnosis of
tological examination may reveal small foci of interstitial chronic infection in individual pigs, both in the diagno-
nephritis. sis of abortion and in the identification of renal or geni-
Placentas from aborted fetuses are grossly normal tal carriers. Infected animals may have MAT titers below
(Fish et al. 1963; Fennestad and Borg-Petersen the widely accepted minimum significant titer of 1:100
1966). (Ellis et al. 1986b, c).
Other serologic tests have been described for use in
DIAGNOSIS pigs, especially ELISA-based tests, but none of these have
gained widespread acceptance for use as diagnostic tests.
A diagnosis of leptospirosis in swine may be required
not only for the clinician to confirm leptospirosis as a Demonstration of Leptospires in Pig Tissues
cause of clinical disease but also for other reasons, such The isolation of leptospires from, or their demonstration
as (1) the assessment of the infection and/or the im- in, the internal organs (such as liver, lung, brain) and
mune status of a herd for the purposes of a control or body fluids (blood, cerebrospinal, thoracic, and peri-
CHAPTER 35 LEPTOSPIROSIS Ellis 489

toneal) of clinically affected animals gives a definitive di- tissue artifacts can be mistakenly identified as lep-
agnosis of acute clinical disease or, in the case of a fetus, tospires.
a diagnosis of leptospiral abortion and probable chronic The demonstration of leptospires by immunochemi-
infection of its mother. cal tests (immunofluorescence, immunoperoxidase, and
Their presence in the male or female genital tract, the immunogold) is more suited to most laboratory situa-
kidney, or urine, in the absence of evidence of general- tions; however, these tests are “number-of-organisms”
ized infection, is diagnostic of chronic infection. Failure dependent and lack the sensitivity of culture. They pro-
to demonstrate leptospires in the urine of a pig does not vide no information as to the infecting serovar (Ellis
rule out the possibility of the animal being a chronic re- 1990) and require high-IgG-titer antileptospire sera,
nal carrier; it merely indicates that the pig was not ex- which are not available commercially. Immunofluores-
creting detectable numbers of leptospires at the time of cence is the method of choice for the diagnosis of fetal
testing. leptospirosis. There have been a number of polymerase
chain reaction methods published, but so far these have
ISOLATION. Isolation, especially from clinical mater- failed to deliver the promised increase in sensitivity
ial, is difficult and time-consuming and is a job for labo- which the underlying technology should theoretically de-
ratories specializing in the identification of isolates. Iso- liver (Taylor et al. 1997).
lation from renal carriers is very useful in
epidemiological studies to determine which serovars are PREVENTION AND CONTROL
present in an animal species or in a particular group of
animals or geographic location. Interruption of transmission from an infected pig or oth-
The isolation of leptospires is the most sensitive er host to a pig is the critical factor in control. Control of
method of demonstrating their presence, provided that leptospirosis depends on the combined use of three
antibiotic residues are absent, that tissue autolysis is not strategies: antibiotic therapy, vaccination, and manage-
advanced, and that tissues for culture have been stored at ment. Unfortunately, not all these options are available
a suitable temperature (4˚C) and, in the case of urine, at in every country; for example, vaccines are not available
a suitable pH since collection. in many Western European countries, including the
Culture should be carried out in a semisolid United Kingdom, and problems of antibiotic residues
(0.1–0.2% agar) bovine serum-albumin medium contain- may make the use of antibiotic therapy difficult in other
ing either Tween 80 (Johnson and Harris 1967) or a com- situations. In the United States, the most useful antibiot-
bination of Tween 80 and Tween 40 (Ellis 1986), and ic for leptospiral control/treatment programs, strepto-
preferably a small amount of fresh rabbit serum mycin, is no longer available for veterinary use. Control
(0.4–2%) if the more fastidious leptospires such as programs must therefore be modified to meet local con-
bratislava are to be isolated. A dilution culture method ditions.
should be used (Ellis 1986). Contamination may be con- Vaccination induces immunity of relatively short du-
trolled by a variety of selective agents (e.g., 5-fluo- ration. Immunity to infection is probably never 100%
rouracil, nalidixic acid, fosfomycin, and a cocktail of ri- and, at best, lasts little more than 3 months (Kemenes
famycin, polymyxin, neomycin, 5-fluorouracil, and Suveges 1976; Ellis et al. 1989); immunity to clinical
bacitracin, and actidione). The use of selective agents disease is believed to last somewhat longer, although the
will reduce the chance of isolation where there are only exact duration is not known. Vaccination will markedly
small numbers of viable leptospires. Culture media con- reduce the prevalence of infection in a herd (Wrathall
taining 5-fluorouracil at levels between 200 and 500 1975; Kemenes and Suveges 1976) but will not eliminate
µg/mL should be used as transport media for the sub- infection (Hodges et al. 1976; Edwards and Daines 1979;
mission of samples (Ellis 1990). Cargill and Davos 1981). Given the widespread use of
Cultures should be incubated at 29–30˚C for at least tetracycline medication of feed in parts of Europe to con-
12 weeks, preferably for 26 weeks (Ellis 1986). They trol bratislava infection and all the attendant residue
should be examined by dark-ground microscopy every problems, there is an urgent need for the marketing of
1–2 weeks. an effective bratislava vaccine in Europe.
Antibiotics alone will not eliminate pig-maintained
OTHER METHODS OF DEMONSTRATING LEPTO- leptospiral infections from the individual carrier animal
SPIRES. Leptospires do not stain satisfactorily with or control infection in herds. Despite claims by some au-
the aniline dyes, and silver-staining techniques lack sen- thors that either systemic streptomycin at 25 mg/kg
sitivity and specificity (Baskerville 1986). Dark-ground body weight (Dobson 1974; Alt and Bolin 1996) or oral
microscopy of fetal fluids or urine has been widely used tetracyclines at levels of 800 g/ton of feed (Stalheim
in the diagnosis of leptospirosis and can be a useful tool 1967) will eliminate carriers, others have reported that
in the hands of an experienced diagnostician, but many these regimes do not work (Doherty and Baynes 1967;
490 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Hodges et al. 1979). Recent work into the use of alterna- ciation with multifocal interstitial nephritis in swine
tive antibiotic therapy regimes indicates that oxytetracy- at slaughter. Can J Vet Res 53:290–294.
cline (40 mg/kg for 3 or 5 days), tylosin (44 mg/kg for 5 Baskerville, A. 1986. Histological aspects of diagnosis of
days), or erythromycin (25 mg/kg for 5 days) may be ef- leptospirosis. In The Present State of Leptospirosis
fective in eliminating pomona from the kidneys of exper- Diagnosis and Control. Dordrecht, Netherlands:
imentally infected pigs (Alt and Bolin 1996). Martinus Nijhoff, pp. 33–43.
The main management factor in the control of lep- Bercovich, Z.; Spek, C. W.; and Comvalius-Adriaan, I.
tospirosis is the prevention of direct or indirect contact 1983. Occurrence of antibodies to various serotypes
with free-living vectors or other domestic stock. Strict of Leptospira interrogans among swine in the Nether-
biosecurity should be implemented and rodent control lands between 1975 and 1980. Tidjschr Diergeneeskd
programs instigated in and around the production com- 108:133–138.
plex. When faced with an outbreak of clinical disease, Bjoersdorff, A.; Korsgren, O.; Feinstein, R.; Andersson,
the best option is to treat both affected and at-risk stock A.; Tollemar, J.; Malmborg, A.-S.; Ehrnst, A.; and
with streptomycin at 25 mg/kg body weight, to immedi- Groth, C. G. 1995. Microbiological characterization
ately vaccinate the at-risk stock, and then to introduce a of porcine fetal islet-like cell clusters for intended
regular vaccination program. If vaccination is not an clinical xenografting. Xenotransplantation 2:26–31.
available option, then a feed medication program, using Bohl, E. H.; Powers, T. E.; and Ferguson, L. C. 1954.
either chlor- or oxytetracycline at 600–800 g/ton of feed, Abortions in swine associated with leptospirosis. J
should be introduced. This ration is fed either continu- Am Vet Med Assoc 124:262.
ously or on a 1-month-on/1-month-off basis. Alterna- Bolin, C. A., and Cassells, J. A. 1990. Isolation of Lep-
tively, it may be fed for two periods of 4 weeks in the tospira interrogans serovar bratislava from stillborn
year, preferably one in the spring and the other in the au- and weak pigs in Iowa. J Am Vet Med Assoc
tumn. 196:1601–1604.
The use of artificial insemination is an important ———. 1992. Isolation of Leptospira interrogans serovars
tool in the control of bratislava infection. bratislava and hardjo from swine at slaughter. J Vet Di-
ag Invest 4:87–89.
ZOONOSIS Bolin, C. A.; Cassells, J. A.; Hill, H. T.; Frantz, J. C.; and
Nielsen, J. N. 1991. Reproductive failure associated
Leptospirosis is an important occupational zoonosis of with Leptospira interrogans serovar bratislava infection
those who work with pigs, especially farmers and those of swine. J Vet Diag Invest 3:152–154.
involved in slaughtering pigs. Pigs are now being pro- Bolt, I., and Marshall, R. B. 1995a. The epidemiology of
duced for intended xeno grafting. It is important to re- Leptospira interrogans serovar pomona in grower pig
member the potential risk to recipients posed by pig-as- herds. NZ Vet J 43:10–15.
sociated leptospirosis (Bjoersdorff et al. 1995). ———. 1995b. The epidemiology of Leptospira interro-
gans serovar pomona in grower pig herds. NZ Vet J
REFERENCES 43:204.
Brandis, H. 1956. Über Leptospirosen durch den type L.
Alexander, A. D.; Yager, R. H.; and Keefe, T. J. 1964. Lep- saxkoebing. Klin Wochenschr 34:521.
tospirosis in swine. Bull Off Epizoot 61:273–304. Buddle, J. R., and Hodges, R. T. 1977. Observations on
Alston, J. M., and Broom, J. C. 1958. Leptospirosis in some aspects of the epidemiology of leptospirosis in
Man and Animals. Edinburgh: E. and S. Livingstone, a herd of pigs. NZ Vet J 25:56, 65–66.
pp. 65–75. Burnstein, T., and Baker, J. A. 1954. Leptospirosis in
Alt, D. P., and Bolin, C. A. 1996. Preliminary evaluation swine caused by Leptospira pomona. J Infect Dis
of antimicrobial agents for treatment of Leptospira 94:53–54.
interrogans serovar pomona infection in hamsters and Cargill, C. F., and Davos, D. E. 1981. Renal leptospirosis
swine. Am J Vet Res 57:59–62. in vaccinated pigs. Aust Vet J 57:236–238.
Anon. 1966. Zoonoses Surveillance: Leptospiral Chappel, R. J.; Ellis, W. A.; Adler, B.; Amon, L.; Millar, B.
Serotype Distribution Lists. USDHEW, Atlanta, Ga. D.; Zhu, S. S.; and Prime, R. W. 1992. Serological ev-
———. 1975. Zoonoses Surveillance: Leptospiral idence for the presence of Leptospira interrogans
Serotype Distribution Lists according to Host and serovar bratislava in Australian pigs. Aust Vet J
Geographic Area. July 1966–July 1973. USDHEW, At- 69:119–120.
lanta, Ga. Chaudhary, R. K.; Fish, N. A.; and Barnum, D. A. 1966a.
———. 1986. Diagnosis of abortions, V.L.S., Guelph. Experimental infection with Leptospira pomona in
Can Vet J 27:A20. normal and immune piglets. Can Vet J 7:106–112.
Baker, I. F.; McEwan, S. A.; Prescott, J. F.; and Meek, A. ———. 1966b. Protection of piglets from immunised
H. 1989. The prevalence of leptospirosis and its asso- sows via colostrum against experimental Leptospira
CHAPTER 35 LEPTOSPIROSIS Ellis 491

pomona infection. Can Vet J 7:121–127. tospires from genital tract and kidneys of aborted
Cheville, N. F.; Huhn, R.; and Cutlip, R. C. 1980. Ultra- sows. Vet Rec 118:294–295.
structure of renal lesions in pigs with acute lep- Ellis, W. A.; Montgomery, J. M.; and McParland, P. J.
tospirosis caused by Leptospira pomona. Vet Pathol 1989. An experimental study with a Leptospira inter-
17:338–351. rogans serovar bratislava vaccine. Vet Rec
Cole, J. R.; Ellinghausen, H. C.; and Rubin, H. L. 1980. 125:319–321.
Laboratory diagnosis of leptospirosis of domestic an- Faine, S. 1982. Guidelines for the control of leptospiro-
imals. Proc US Anim Health Assoc 83:189–199. sis. Offset Pub no. 67. Geneva: WHO.
Cole, J. R.; Hall, R. F.; Ellinghausen, H. C.; and Pursell, A. ———. 1994. Leptospira and Leptospirosis. Boca Raton:
R. 1983. Prevalence of leptospiral antibodies in Geor- CRC Press.
gia cattle and swine, with emphasis on Leptospira in- Fennestad, K. L., and Borg-Petersen, C. 1966. Experi-
terrogans serovar tarassovi. Proc US Anim Health As- mental leptospirosis in pregnant sows. J Infect Dis
soc 87:199–210. 116:57–66.
Combiesco, D.; Sturdza, N.; Sefer, M.; and Radu, I. 1958. Ferguson, L. C., and Powers, T. E. 1956. Experimental
Recherches sur les leptospirosis. Arch Roum Pathol leptospirosis in pregnant swine. Am J Vet Res
Exp Microbiol 17:245. 17:471–477.
Dikken, H., and Kmety, E. 1978. Serological typing meth- Ferguson, L. C.; Lococo, S.; Smith, H. R.; and Handy, A.
ods of leptospires. Methods in Microbiology H. 1956. The control and treatment of swine lep-
11:259–307. tospirosis during a naturally occurring outbreak. J
Dobson, K. J. 1974. Eradication of leptospirosis in com- Am Vet Med Assoc 129:263–265.
mercial pig herds. Aust Vet J 50:471. Field, H. I., and Sellers, K. C. 1951. Leptospira icterohaem-
Doherty, P. C., and Baynes, I. D. 1967. The effects of feed- orrhagiae infection in piglets. Vet Rec 63:78–81.
ing oxytetracycline on leptospira in pigs infected with Fish, N. A.; Ryu, E.; and Hulland, T. J. 1963. Bacteriolog-
L. pomona. Aust Vet J 43:135–137. ical and pathological studies of natural and experi-
Edwards, J. D., and Daines, D. 1979. A leptospirosis out- mental swine abortion due to Leptospira pomona. Can
break in a piggery. NZ Vet J 27:247–248. Vet J 4:317–327.
Egan, J. 1995. Porcine leptospirosis. Irish Vet J 48:399, Frantz, J. C.; Hanson, L. E.; and Brown, A. L. 1989. Effect
401–402. of vaccination with a bacterin containing Leptospira
Ellis, W. A. 1986. The diagnosis of leptospirosis in farm interrogans serovar bratislava on the breeding perfor-
animals. In The Present State of Leptospirosis Diag- mance of swine herds. Am J Vet Res 50:1044–1047.
nosis and Control. Dordrecht, Netherlands: Marti- Fuzi, M.; Alfoldy, Z.; Kiszel, J.; and Raditz, I. 1957. Die
nus Nijhoff, pp. 13–31. Leptospiren-Infection der Feldnagetiere in einem Ge-
———. 1989. Leptospira Australis infection in pigs. Pig biet von Westungarn. Acta Microbiol Acad Sci Hung
Vet J 22:83–92. 4:155–156.
———. 1990. Leptospirosis. In OIE Manual of Recom- Gorshanova, E. N. 1964. Domestic animals as a source of
mended Diagnostic Techniques and Requirements leptospirosis in Dagestan. J Microbiol (Moscow)
for Biological Products for List A and B Diseases, vol. 41:120.
2, sec. 7, pp. 1–11. Paris. Hanson, L. E. 1985. Report of the committee on lep-
———. 1992. Leptospirosis. In Diseases of Swine, 7th tospirosis. Proc US Anim Health Assoc 90:217.
ed. Ed. A. D. Leman, B. E. Straw, W. L. Mengeling, S. ———. 1987. Bratislava—A newly recognised leptospi-
D’Allaire, and D. J. Taylor. Ames: Iowa State Univ ral serovar in swine. Swine Consultant (Spring):1–8.
Press, pp. 529–535. Hanson, L. E., and Tripathy, D. N. 1986. Leptospirosis. In
———. 1995. International Committee on Systematic Diseases of Swine, 6th ed. Ed. A. D. Leman, B. Straw,
Bacteriology Subcommittee on the Taxonomy of Lep- R. D. Glock, W. L. Mengeling, R. H. C. Penny, and E.
tospira. Int J Systematic Bacteriol 45:872–874. Scholl. Ames: Iowa State Univ Press, pp. 591–599.
Ellis, W. A., and Thiermann, A. B. 1986. Isolation of Lep- Hanson, L. E.; Schnurrenberger, P. R.; Marshall, R. B.;
tospira interrogans serovar bratislava from sows in and Scherrick, G. W. 1965. Leptospiral serotypes in
Iowa. Am J Vet Res 47:1458–1460. Illinois cattle and swine. Proc US Livest Sanit Assoc
Ellis, W. A.; McParland, P. J.; Bryson, D. G.; and Cassells, 69:164.
J. A. 1986a. Prevalence of leptospira infection in Hanson, L. E.; Reynolds, H. A.; and Evans, L. B. 1971.
aborted pigs in Northern Ireland. Vet Rec 118:63–65. Leptospirosis in swine caused by serovar grippoty-
———. 1986b. Boars as carriers of leptospires of the phosa. Am J Vet Res 32:855.
Australis serogroup on farms with an abortion prob- Hartmann, E. G.; Brummelman, B.; and Dikken, H.
lem. Vet Rec 118:563. 1975. Leptospirae of serotype lora of the serogroup
Ellis, W. A.; McParland, P. J.; Bryson, D. G.; Thiermann, Australis isolated for the first time from swine in the
A. B.; and Montgomery, J. 1986c. Isolation of lep- Netherlands. Tidjschr Diergeneeskd 100:421–425.
492 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Hathaway, S. C. 1981. Leptospirosis in New Zealand: An tospires. Am J Trop Med Hyg 7:646.
ecological view. NZ Vet J 29:109–112. Michna, S. W. 1962. Abortion in the sow due to infection
———. 1983. Leptospirosis in Pigs in England. FRCVS by Leptospira canicola: A preliminary report. Vet Rec
thesis. Royal College Vet Surgeons, London. 74:917–919.
———. 1985. Porcine leptospirosis. Pig News Inf ———. 1970. Leptospirosis. Vet Rec 86:484–496.
6:31–34. Michna, S. W., and Campbell, R. S. F. 1969. Leptospiro-
Hathaway, S. C., and Little, T. W. A. 1981. Prevalence and sis in pigs: Epidemiology, microbiology and patholo-
clinical significance of leptospiral antibodies in pigs gy. Vet Rec 84:135–138.
in England. Vet Rec 108:224–228. Mitchell, D.; Robertson, A.; Corner, A. H.; and
Hathaway, S. C.; Little, T. W. A.; and Stevens, A. E. 1981. Boulanger, P. 1966. Some observations on the diag-
Serological and bacteriological survey of leptospiral nosis and epidemiology of leptospirosis in swine.
infection in pigs in southern England. Res Vet Sci Can J Comp Med 30:211–217.
31:169–173. Mitscherlich, E., and Marth, E. H. 1984. Microbial Sur-
Hathaway, S. C.; Ellis, W. A.; Little, T. W. A.; Stevens, A. vival in the Environment. Berlin: Springer-Verlag, pp.
E.; and Ferguson, H. W. 1983. Leptospira interrogans 202–218.
serovar hardjo in pigs: A new host parasite relation- Morse, E. V.; Bauer, D. C.; Langham, R. F.; Lang, R. W.;
ship in the United Kingdom. Vet Rec 113:153–154. and Ullrey, D. E. 1958. Experimental leptospirosis.
Hodges, R. T.; Stocker, R. P.; and Buddle, J. R. 1976. Lep- IV. Pathogenesis of porcine Leptospira pomona infec-
tospira interrogans serotype pomona infection and lep- tions. Am J Vet Res 19:388–394.
tospiruria in vaccinated pigs. NZ Vet J 24:37–39. Morter, E. V.; Morse, E. V.; and Langham, R. F. 1960. Ex-
Hodges, R. T.; Thompson, J.; and Townsend, K. G. 1979. perimental leptospirosis. VII. Re-exposure of preg-
Leptospirosis in pigs: The effectiveness of strepto- nant sows with Leptospira pomona. Am J Vet Res
mycin in stopping leptospiruria. NZ Vet J 21:95.
27:124–126. Nagy, G. 1993. Comparative pathogenicity study of Lep-
Jensen, A. M., and Binder, M. 1989. Serological reactions tospira interrogans serovar pomona strains. Acta Vet
for leptospires in reproductive failure in Danish pigs. Hung 41:315–324.
Is there a connection? A preliminary investigation. Neto, J. S. F.; Vasconelles, S. V.; Ito, F. H.; Moretti, A. S.
Dansk Vet 72:1181–1187. A.; Camargo, C. A.; Sakamoto, S. M.; Marangon
Johnson, R. C., and Harris, V. G. 1967. Differentiation of Turilli, C.; and Martini, M. 1997. Leptospira interro-
pathogenic and saprophytic leptospires. 1. Growth at gans serovar icterohaemorrhagiae seropositivity and
low temperatures. J Bacteriol 94:27–31. the reproductive performance of sows. Prev Vet Med
Kemenes, F., and Suveges, T. 1976. Leptospira-induced re- 31:87–93.
peated abortion in sows. Acta Vet Hung 26:395–403. Nissen, O. D. 1989. Leptospirosis in pigs: A review of the
Kingscote, B. F. 1986. Leptospirosis outbreak in a piggery literature and a study of the development of titres
in southern Alberta. Can Vet J 27:188–190. against L. bratislava in two herds. Dansk Vet
———. 1988. Leptospiral serovars in Canada. Can Vet J 72:619–635.
29:70–71. Oliveira, S. J. de; Borowski, S. M.; and Barcellos, D. E. S.
Klarenbeek, A., and Winsser, J. 1937. Ein Fall von spon- N. 1994. Serological evidence of infection by Lep-
taner Weilscher Krankheit bei Ferkeln. Dtsch tospira bratislava as the cause of reproductive disor-
Tierärztl Wochenschr 45:434–435. ders in pigs. Ciencia-Rural 24:345–348.
Langham, R. F.; Morse, E. V.; and Morter, R. L. 1958. Ex- Paz-Soldan, S. V.; Dianderas, M. T.; and Windsor, R. S.
perimental leptospirosis. V. Pathology of Leptospira 1991. Leptospira interrogans serovar canicola: A causal
pomona infection in swine. Am J Vet Res 19:395–400. agent of sow abortions in Arequipa, Peru. Trop Anim
Loimayr, V. 1990. Survey for antibodies to Leptospira in Hlth Prodn 23:233–240.
Austrian pigs. Wiener Tierarztl Monatsschr 77:60. Potts, A. D.; Lotter, C.; and Robinson, J. T. R. 1995. Sero-
Matveeva, A. A.; Sakharova, P. V.; Sharban, E. K.; and logical prevalence of leptospiral antibodies in pigs
Dragomir, A. V. 1977. [Etiology of leptospires in ani- in South Africa. Onderstepoort J Vet Res 62:281–
mals in the Moldavian Republic, USSR]. Veterinariya 284.
(Moscow) 1:61–63. Power, S. B. 1991. Diagnosing leptospira in pigs. Vet Rec
McErlean, B. A. 1973. The isolation of leptospirae from 128:43.
the kidneys of bacon pigs. Irish Vet J 27:185–186. Preston, K. S., and Morter, R. L. 1960. Rapid laboratory
McKeever, S.; Gorman, G. W.; Chapman, J. F.; Galton, confirmation of clinical diagnosis of Leptospira
M. M.; and Powers, D. K. 1958. Incidence of lep- pomona infection in pregnant sows. Allied Vet
tospirosis in wild mammals from southwestern Geor- 31:104–107.
gia with a report of new hosts of six serotypes of lep- Propopcakova, H.; Pospisil, R.; Cislakova, L.; and Kozak,
CHAPTER 35 LEPTOSPIROSIS Ellis 493

M. 1981. Determination of antibodies to leptospiro- Leptospira pomona in porcine tissues. J Am Vet Med
sis in animals and man in eastern Slovakia in the past Assoc 139:455–456.
ten years. Veterinarstvi 31:275–277. Sleight, S. D.; Langham, R. F.; and Morter, R. L. 1960. Ex-
Rehmtulla, A. J.; Prescott, J. F.; Nicholson, V. M.; and perimental leptospirosis: The early pathogenesis of
Bolin, C. A. 1992. Leptospira bratislava infection in Leptospira pomona infection in young swine. J Infect
aborted pigs in Ontario. Can Vet J 33:344–345. Dis 106:262–269.
Roth, E. E. 1964. Leptospirosis in wildlife in the United Stalheim, O. H. V 1967. Chemotherapy of renal lep-
States. Proc Am Vet Med Assoc 101:211. tospirosis in swine. Am J Vet Res 28:161–166.
Ryley, J. W., and Simmons, G. C. 1954a. Leptospira Taylor, M. J.; Ellis, W. A.; Montgomery, J. M.; Yan, K.-T.;
pomona as a cause of abortion and neonatal mortali- McDowell, S. W. J.; and Mackie, D. P. 1997. Magnet-
ty in swine. Queens J Agric Sci 11:61–74. ic immuno-capture PCR assay (MIPA): Detection of
———. 1954b. Leptospirosis of pigs with special refer- Leptospira borgpetersenii serovar hardjo. Vet Micro
ence to birth of dead pigs and neonatal mortality. 56:135–145.
Aust Vet J 30:203–208. Tripathy, D. N.; Hanson, L. E.; Mansfield, M. E.; and
Sandstedt, K., and Engvall, A. 1985. Serum antibodies to Thilsted, J. P. 1981. Pathogenesis of Leptospira
Leptospira bratislava in Swedish pigs and horses. pomona in lactating sows and transmission to piglets.
Nord Vet Med 37:312–313. Proc US Anim Health Assoc 85:188.
Saravi, M. A.; Molinar, R.; Soria, E. H.; and Barriola, J. L. Urban, V. P., and Androsov, V. A. 1976. [Titer of lep-
1989. Serological and bacteriological diagnosis, and tospiral antibodies as a function of time and course
reproductive consequences of an outbreak of porcine of infectious process.] Dokl Vses Acad Sci-Khoz
leptospirosis caused by a member of the Pomona Nauk 2:39–42.
serogroup. Res Sci Tech Off Int Epix 8:709–718. Van Til, L. D., and Dohoo, I. R. 1991. A serological survey
Schnurrenberger, P. R.; Hanson, L. E.; and Martin, R. J. of leptospirosis in Prince Edward Island swine herds
1970. Long term surveillance of leptospirosis on an and its association with infertility. Can J Vet Res
Illinois farm. Am J Epidemiol 92:223–239. 55:352–355.
Schonberg, A.; Hahnhey, B.; Kampe, U.; Schmidt, K., Wandurski, A. 1982. Effect of infections with various
and Ellis, W. A. 1992. The isolation and identification serotypes of Leptospira interrogans on the fertility of
of Leptospira interrogans serovar bratislava from a pig swine. Med Weter 38:218–220.
in Germany. J Vet Med Series B 39:362–368. Weber, B., and Fenske, G. 1978. Unterschungen zur Lep-
Sebek, Z.; Treml, F.; and Valova, M. 1983. Experimental tospira bratislava Infection beim Schwein. Monatsh
infection with the virulent central European murine Vet Med 33:652–656.
Leptospira pomona strain in the pig. Folia Parasitol Wrathall, A. E. 1975. Reproductive Disorders in Pigs. An-
30:269–275. im Health Rev Ser no. 11. Commonwealth Agricul-
Sleight, S. D., and Lundberg, A. M. 1961. Persistence of tural Bureaux.
Mycoplasmal Diseases
36 R. F. Ross

Mycoplasmal pneumonia of swine (MPS), or enzootic health-status swine and widespread availability of M. hy-
pneumonia, is caused by Mycoplasma hyopneumoniae and opneumoniae and PRRSV vaccines. Mycoplasma hyorhinis
is one of the most important contributors to disease-as- is another common inhabitant of the respiratory tract of
sociated loss in swine production. Recently, the infection swine, is not a primary cause of pneumonia, but occa-
has been attributed increased importance as part of a sionally causes polyserositis and arthritis in young pigs.
composite infection with porcine reproductive and respi- Another mycoplasma, Mycoplasma hyosynoviae, causes
ratory syndrome virus (PRRSV) and other agents in U.S. arthritis in growing-finishing swine. Other swine iso-
swine. This syndrome, known as the porcine respiratory lates, not known to be pathogenic or not common, in-
disease complex (PRDC), has emerged in spite of the clude M. flocculare, M. sualvi, M. hyopharyngis, and sever-
continuing adoption of strategies for rearing high- al species of acholeplasmas.

MYCOPLASMAL PNEUMONIA OF SWINE


Mare and Switzer (1965) and Goodwin et al. (1965) were mation concerning the biological and biochemical prop-
the first to report the isolation of a mycoplasma from erties of M. hyopneumoniae and methods for specific
pneumonic lung and the experimental reproduction of identification were reviewed by Ross and Whittlestone
the disease. The isolates, named M. hyopneumoniae and (1983). M. flocculare, a nonpathogen common in swine
M. suipneumoniae, were shown to be identical, and the lungs, has many morphological, growth, and antigenic
former name was given priority. Enzootic pneumonia similarities to M. hyopneumoniae. Finally, a very impor-
has been reported from many countries and has long tant attribute of mycoplasmas, with potential pathogen-
been believed to be one of the most common and eco- ic and control implications, is their marked propensity to
nomically important diseases occurring in swine. Loss switch surface antigens. Artiushin and Minion (1996) re-
associated with disease is the result of a complex interac- cently provided evidence of heterogeneity existing
tion between the mycoplasma and other infections, poor among isolates of M. hyopneumoniae.
management, and poor environmental conditions.
EPIDEMIOLOGY
ETIOLOGY
Field observations have strongly implicated carrier swine
Isolation of the organism is complicated by its extremely as a major source of infection with M. hyopneumoniae. In
fastidious growth requirements and the frequent pres- an early report Pullar (1948) summarized data on out-
ence of M. hyorhinis in the swine respiratory tract. The breaks of pneumonia in 190 Australian swine herds; 80%
most widely used growth medium is that described by of outbreaks were associated with introduction and
Friis (1975). Media and methods for isolation of M. hy- commingling of store (feeder) pigs with swine already
opneumoniae were reviewed by Ross and Whittlestone present on the farm, and 20% were associated with intro-
(1983). duction of adult breeding stock.
In primary broth cultures, M. hyopneumoniae grows Transmission of M. hyopneumoniae is apparently ef-
slowly, producing a faint turbidity and an acid color shift fected mainly by direct contact with respiratory tract se-
after 3–30 days’ incubation. Generally, cultures are pas- cretions from infected swine. Goodwin (1972a) demon-
saged several times in broth, then inoculated on agar strated that the organism could be isolated from nasal
medium and incubated in a 5–10% carbon dioxide at- samples from affected pigs. Farrington (1976) and
mosphere. Colonies of the organism become barely visi- Etheridge et al. (1979) demonstrated that transmission
ble after 2–3 days’ incubation, and they increase in size of the organism occurs among penmate swine; however,
to about 0.25–1 mm diameter in about 10 days. Infor- transmission does not always take place even among

495
496 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

commingled penmates (Goodwin 1972b). Because of the past 10 years, where improved control measures such as
fastidious nature of the organism and the large inoculum age-segregated rearing have been implemented, the over-
required for experimental transmission of the disease, all prevalence of pneumonia at slaughter may have de-
dogma has held that it could not be easily transmitted creased. In some areas, however, pulmonary disease has
between herds unless carrier swine were involved. How- increased in complexity and severity because of in-
ever, many breakdowns have occurred in pneumonia- creased involvement of viruses such as PRRSV with M.
free herds that were thought to be rather strictly isolated hyopneumoniae (Halbur et al. 1993).
(Whittlestone 1979). In Britain, Goodwin (1985) found The frequency of isolation of M. hyopneumoniae from
that breakdowns occurred most frequently when herds chronic swine pneumonia varies. In a survey of Iowa
were less than 3.2 km apart. Jorsal and Thomsen (1988) breeding swine of various ages, Young et al. (1983) found
found that reinfections occurred most frequently in Dan- that animals in 60% of 597 herds had complement-fixing
ish herds during the autumn and winter and when spe- (CF) antibodies to M. hyopneumoniae. In another survey
cific-pathogen-free (SPF) herds were close to non-SPF of Iowa swine, primarily slaughter-weight swine, Owen
herds. Transmission was especially likely when the latter (1990) found CF antibodies to the organism in sera from
contained more than 500 swine. Further work by the 80% of 88 herds and 32% of 2077 animals in these
same group (Thomsen et al. 1992) and work in Switzer- herds.
land (Stark et al. 1992) support the conclusion that air- Economic loss attributable to MPS has long been as-
borne transmission is a mechanism for reinfection of sociated with reduced rate of gain and feed efficiency
SPF herds with M. hyopneumoniae. (Switzer and Ross 1975). Pointon et al. (1985) reported
MPS is maintained in many herds by sow-to-pig that the growth rate of pigs held in contact with MPS-af-
transmission of M. hyopneumoniae. Once infection is es- fected pigs was reduced by 12.7% between 50 and 85 kg
tablished in a few pigs, transmission among penmates body weight. In another trial, they found that the growth
occurs, especially after animals are pooled together at rate of pigs exposed to infected dams was reduced by
weaning time. In continuous-production systems, M. hy- 15.9% (between 8 and 85 kg body weight), and feed con-
opneumoniae and a number of other important respira- version was decreased by 13.8% (between 10 and 25 kg
tory pathogens may be transmitted in large numbers body weight). Losses were estimated to be approximate-
from older to younger pigs. Overt signs of MPS usually ly $2.80 (Australian) for each pig produced.
are not seen until piglets are 6 weeks of age or older. Al- Much information about the prevalence of MPS and
though M. hyopneumoniae infections are commonly the related economic loss has been based on evaluation
thought to begin in the nursing pig, microbiological evi- of lungs at slaughter; however, caution must be exercised
dence that the organism is common in such lesions has when using data collected at slaughter. Scheidt et al.
not been presented. Kott (1983) found no evidence of M. (1990a) found no correlation between average daily gain
hyopneumoniae in lungs of 55 baby pigs with pneumonia. and severity of pneumonia at slaughter, and Noyes et al.
Predominant organisms isolated were Haemophilus para- (1990) found little correlation between extent of lesions
suis, M. hyorhinis, and Bordetella bronchiseptica. Pigs of detected at slaughter and those detected radiologically
various ages seem to be equally susceptible to the organ- earlier in life. In a study of pigs from two Danish con-
ism. Undoubtedly, the long incubation period, slow ventional farrow-to-finish herds, Paisley et al. (1993a,
spread of the organism in litters, increased animal den- 1993b) reported that mycoplasma-like pneumonia, as
sity, spread of other infectious agents, and environmen- well as several other respiratory lesions found at slaugh-
tal factors that develop following weaning contribute to ter, were related to reduced mean daily gain. However,
the peak prevalence of M. hyopneumoniae disease in they also concluded that lesions present at slaughter
growing-finishing swine. could be related to only 9–27% of the variation in mean
Surveys conducted in a variety of countries in the daily gain. They suggested that the remaining 20–90% of
past indicated that lesions typical of those seen with variation was due to factors such as environment, feed,
MPS occur in 30–80% of slaughter-weight swine (Whit- genetics, and management systems.
tlestone 1979; Switzer and Ross 1975). A more recent
Minnesota survey for gross lesions at slaughter indicated PATHOGENESIS
that 100% of 125 herds had typical “enzootic pneumo-
nia” lesions and 75% of the pigs were affected (Pointon et The incubation period of MPS was reported by Betts
al. 1990). Guerrero (1990) reported on surveys conduct- (1952) to be 10–16 days under natural conditions; how-
ed at slaughter in seven countries; the prevalence of ever, other reports have indicated considerable variabili-
pneumonia ranged from 38% to 100%. However, Tielen ty in duration. Onset of disease is probably dependent
(1995) indicated that the prevalence of pneumonia at on the intensity of infection with the organism on the
slaughter in the Netherlands has decreased from 23% in tracheal and bronchial mucosal surfaces. Disease has
1981 to 5.8% in 1990. It is conceivable that during the been reported in pigs 2 weeks of age (Holmgren 1974),
CHAPTER 36 MYCOPLASMAL DISEASES Ross 497

but it generally spreads slowly and many pigs do not evi- Virtually all naturally occurring cases of MPS are
dence disease until they are 3–6 months old. mixed infections involving mycoplasmas, bacteria, virus-
In the early stages of infection, large numbers of my- es, or nematodes. Experimentally induced M. hyopneu-
coplasmas have been detected by electron microscopy as moniae pneumonia has been shown to predispose swine
well as by immunofluorescence (IF) tests, primarily on to pneumonia caused by P. multocida (Ciprian et al. 1988;
the surfaces of the trachea, bronchi, and bronchioles. Amass et al. 1994) and Actinobacillus pleuropneumoniae
Very few organisms have been found in the small bron- (Yagihashi et al. 1984). The PRRSV, in interaction with
chioles and alveoli. Scanning (Mebus and Underdahl M. hyopneumoniae and other agents, has been strongly
1977) and transmission (Blanchard et al. 1992) electron implicated in the emergence of a widespread complicat-
microscopy of MPS-affected lungs revealed a close asso- ed pneumonia known as PRDC in many U.S. (Halbur et
ciation of mycoplasmas with the cilia as well as extensive al. 1993; Halbur 1996) and Canadian finishing units
loss of cilia. No evidence of a specialized attachment or (Moore 1996). The importance of environmental factors
orientation of the organism to the epithelial cells has in respiratory disease losses is discussed in Chapter 61.
been demonstrated. The microscopic changes, especially
peribronchial and perivascular lymphoreticular hyper- CLINICAL SIGNS
plasia, are widely thought to reflect significant involve-
ment of the immune response in lesion development. Betts (1952) described MPS as a chronic disease with a
Mycoplasma pathogens generally are known to uti- high morbidity and a low mortality. The principal clini-
lize a very complex virulence process; this involves at- cal sign is a chronic, nonproductive cough. Onset of the
tachment/colonization, cytotoxicity, competition for disease is gradual, with coughing continuing for a few
substrates, and evasion and modulation of the host im- weeks or even months, although some affected pigs evi-
mune response. Such processes are not measured by a dence little or no coughing. Intensity of coughing is of-
single effect but by the expression of a multitude of gene ten greatest in growing-finishing swine. Respiratory
products (adhesins, nutrient receptors, mitogens, poly- movements are normal unless extensive lung involve-
saccharide polymers, and metabolic intermediates). Re- ment, especially secondary bacterial infection, develops.
cent efforts have been directed toward elucidation of the Death loss associated with secondary bacterial infection
specific pathogenetic mechanisms utilized by M. hyo- and stress may occur at 4–6 months of age. Animals with
pneumoniae in induction of pneumonia. We have demon- this “secondary breakdown” may evidence inappetence,
strated that M. hyopneumoniae attaches to a variety of labored breathing or “thumping,” increased coughing,
porcine cells in vitro (Zielinski et al. 1990); however, the elevated temperatures, and prostration. Most pigs with
most meaningful was the demonstration of the attach- MPS evidence no malaise but appear unthrifty, their hair
ment of the organism to ciliary tufts of single ciliated tra- coat lacking a normal “bloom.” Growth may be retarded
cheal cells (Zielinski and Ross 1993). An in vitro mi- and stunting may occur, although appetites are usually
crotiter system for studying attachment of the organism normal.
to cilia was used to partially characterize the attachment
process and to demonstrate that at least two proteins, 97 LESIONS
and 145 kDa, in the organism’s membrane, were possibly
involved in the attachment process (Zhang et al. 1995). Gross lesions in lungs of swine with MPS consist of pur-
Two additional proteins of 28.5 and 57 kDa were shown ple to gray areas of consolidation. The lesions are virtu-
by Chen et al. (1996) to compete for binding of M. hyo- ally always in the ventral portions of the cranial and mid-
pneumoniae to porcine cilia. The attachment process in dle lobes, the accessory lobe, and the cranial portion of
M. hyopneumoniae disease is undoubtedly a multifaceted the caudal lobes of the lungs (Fig. 36.1). The gross ap-
and complex process. pearance of the involved lung resembles that of at-
Infected airways evidence ciliary and epithelial cell electatic lung, particularly during the chronic stages of
damage. Similar damage has been produced in vitro with disease. When the affected lung is incised, the consisten-
tracheal ring cultures inoculated with pneumonic lung cy is “meaty” but not excessively firm. In early- and mid-
homogenate containing M. hyopneumoniae by DeBey and dle-stage disease there is usually a catarrhal exudate in
Ross (1994). Another potentially important event in the the airways. Bronchial and mediastinal lymph nodes are
pathogenesis of the disease is the interaction of the my- often enlarged.
coplasma with lymphoid cells. Membranes of the organ- Microscopic changes in the lungs of pigs with exper-
ism were mitogenic for porcine lymphocytes in vitro imentally induced MPS have been described by Whittle-
(Messier and Ross 1991), and swine infected with the or- stone (1972). The lesions are very similar to those de-
ganism have altered alveolar macrophage function scribed in reports on the naturally occurring disease.
(Caruso and Ross 1990) and are immunosuppressed Early lesions consist of small accumulations of neu-
(Wannemuehler et al. 1988; Weng and Lin 1988). trophils in the lumina and around airways as well as in
498 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

36.1. The lung of a


pig with M. hyopneu-
moniae pneumonia.

the alveoli. Infiltrating lymphocytes are seen in adventi- cells in alveoli, and thickening of interalveolar septa.
tia of arterioles and venules and around airways. As the More advanced lesions, 17–40 days postinfection as de-
disease progresses there are increased numbers of lym- scribed by Whittlestone (1972), consist of extensively
phocytes in perivascular, peribronchial, and peribron- proliferated lymphoreticular tissue in perivascular and
chiolar tissues as well as in the lamina propria of the air- peribronchiolar areas (Fig. 36.3). In recovering lesions
ways. Alveoli may contain eosinophilic edema fluid and described by Whittlestone there were collapsed alveoli,
increased numbers of mononuclear, septal, and poly- alveolar emphysema, and rather extensive hyperplastic
morphonuclear cells (Fig. 36.2). By about 15–20 days lymphoid nodules (Fig. 36.4), especially in association
there are appreciable cuffing or lymphoid hyperplasia with the airways. MPS lesions are markedly influenced
around the airways, more extensive accumulations of by secondary bacterial infections, stress, poor air quality,
edema fluid, large mononuclear and other inflammatory and bad management (see Chapter 61).

36.2. The lung of a pig with M. hyopneumoniae 36.3. The lung of a pig with M. hyopneumoniae
disease. Early stage with mixed alveolar exudate and disease. More advanced lesion with resolving alveolar
some peribronchiolar lymphocytic infiltration (H&E). inflammation and peribronchiolar lymphocytic
hyperplasia (H&E; ×287).
CHAPTER 36 MYCOPLASMAL DISEASES Ross 499

ELISA antibodies develop in susceptible pigs by 3


weeks (Bereiter et al. 1990) after exposure to M. hyopneu-
moniae and persist for as long as 52 weeks postexposure
(Armstrong et al. 1983; Bereiter et al. 1990). In the study
by Bereiter et al., optical density (OD) values with the
Tween 20 ELISA peaked about 5–7 weeks postexposure,
and animals were still seropositive 1 year after exposure.
Using the Tween 20 ELISA, Zimmermann et al. (1986)
found 65% of animals positive between 4 and 8 months
of age, with numbers positive declining to 26–28% by
8–12 months of age. Yagihashi et al. (1993) found that
72% of sows from conventional herds were positive with
the Tween 20 ELISA; the seropositive rate declined with
increasing parity of sows. Utilizing sera from a large far-
row-to-finish herd with a history of respiratory disease,
we found that colostral antibodies declined from sample
1 (17 days of age) to sample 3 (52 days of age); mean
ODs then increased until sample 8 (132 days of age),
when a sharp increase occurred (Young and Ross, un-
published data—1993). Yagihashi et al. (1993) further
36.4. The lung of a pig with M. hyopneumoniae reported that in 3267 growing-finishing swine ranging in
disease. Advanced lymphocytic hyperplasia, including
age from 1 to 6 months, the seropositive rate increased
obliteration of bronchioles (H&E; ×287).
sharply from the age of 4 months to 6 months.
Zimmermann et al. (1989) and Zimmermann (1990)
reported that evaluation of colostrum was a useful strat-
DIAGNOSIS egy for monitoring infection in attempts to eradicate M.
hyopneumoniae from Swiss swine herds. Morris et al.
Clinical signs that are helpful and lead one to suspect (1994) reported that the median half-life of passively ac-
MPS include a chronic nonproductive cough, retarded quired ELISA antibodies against M. hyopneumoniae in
growth and stunting, low mortality, slow onset and suckling piglets was 15.8 days. They further indicated
spread, and repeated occurrence of the disease. Lesions that estimates of the time at which passive antibodies
of MPS, described in the previous section, are character- waned were 30, 45, and 63 days for piglets with initial an-
istic for MPS but are not specific in that similar lesions tibody concentrations classified as low, medium, or high,
may be seen in pneumonias caused by other agents. Re- respectively.
cently, Ross and Stemke (1996) reviewed traditional and Direct, as well as indirect, modifications of the IF test
new molecular approaches to diagnosis of MPS in swine. have been used to detect M. hyopneumoniae in lung tis-
Serologic methods found useful for diagnosis of MPS sues, and positive results obtained have correlated well
include indirect hemagglutination (IHA), CF, and en- with positive findings obtained by other methods. IF
zyme-linked immunosorbent assay (ELISA) (Ross and seems particularly suited for acute-stage disease, when
Whittlestone 1983). Other tests that have been evaluated larger numbers of mycoplasmal cells are present. The or-
include agglutination, latex agglutination, and indirect ganism can be found principally on the bronchial and
IF tests. bronchiolar lining of affected lung. M. hyopneumoniae
Nicolet et al. (1980) and Bommeli and Nicolet (1983) has also been detected in pneumonic lungs by use of an
developed an indirect ELISA using a Tween 20 extract for enzyme-linked immunoperoxidase technique (Brug-
detection of antibodies to M. hyopneumoniae. Cross-reac- gmann et al. 1977). Mattsson et al. (1995) and Blanchard
tions resulting from M. flocculare infections were mini- et al. (1996) reported very promising developments us-
mized using a slight modification of the Tween 20 ELISA ing the polymerase chain reaction (PCR) for rapid, spe-
(Bereiter et al. 1990). Additional tests for detection of cific detection of M. hyopneumoniae in nasal swabs and
antibody to M. hyopneumoniae, including a blocking tracheobronchial washings from infected pigs.
ELISA, have been reported in recent years (Feld et al. M. hyopneumoniae is one of the most difficult my-
1992); their reliability and usefulness on a wide scale coplasmas to isolate and identify. It grows slowly and is
need to be determined. In a comparison of ELISA tests, often overgrown by M. hyorhinis, a common secondary
Wallgren et al. (1996) found earlier detection with the in- invader in swine pneumonia. Although methods for iso-
direct (Tween 20) ELISA, whereas they found less cross- lation have been improved and more laboratories have
reactivity with other mycoplasmas with a blocking developed the capability to isolate the organism, diagno-
ELISA. sis by culture is not feasible in most situations.
500 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

TREATMENT pigs with experimentally induced MPS (Ross and Cox


1988). Two developments seem noteworthy with respect
Early evidence that M. hyopneumoniae disease could be to the issue of tiamulin and its value for treatment of en-
controlled at least in part by medication with tetracycline zootic pneumonia. Where acceptable, use of a combina-
antibiotics has been reviewed by Switzer and Ross tion of tiamulin with chlortetracycline or oxytetracycline
(1975). Tetracycline antibiotics do not prevent establish- may be beneficial in reducing the severity of the disease
ment of infection, and lesions apparently develop after (Burch et al. 1986; Koh et al. 1996). The second develop-
cessation of medication. Repeated administration of ment is that SDZ PMD 296 (Econor, Sandoz), a new
long-acting oxytetracycline during the suckling and early pleuromutilin derivative, appears to have exceptional ac-
nursery periods may have potential for reduction of tivity against M. hyopneumoniae (Hannan and Ripley
pneumonias occurring at later stages (Scheidt et al. 1996; Windsor et al. 1996), with promise that it will be
1990b). Inamoto et al. (1994), using an in vitro assay, re- more effective in treatment of enzootic pneumonia than
ported that a comparison of strains of M. hyopneumoni- tiamulin.
ae isolated in Japan between 1970–81 and 1989–90 indi- Newer quinolone antibiotics have good in vitro activ-
cated decreasing susceptibility to chlortetracycline. ity against M. hyopneumoniae (Hannan et al. 1989; Coop-
Feed medication with 1000 g/ton tylosin phosphate er et al. 1993). At least four quinolones—enrofloxacin
(Mare and Switzer 1966) and water medication with 2 (Simon et al. 1990), danofloxacin (Ross et al. 1990), nor-
g/gallon (530 g/L) tylosin tartrate (Huhn 1971) did not floxacin (Hannan and Goodwin 1990), and ofloxacin
prevent MPS. Goodwin (1972a) presented evidence that (Kuwano et al. 1992)—have been reported to be effective
intramuscular injection of 10 mg/kg tylosin daily, start- in treating the disease.
ing the day before exposure and continuing for 3 addi- Sulfonamides have long been known to have little in-
tional days, reduced the severity of the disease. However, fluence on M. hyopneumoniae, although they are widely
intramuscular inoculation with tylosin at 8.8 mg/kg for 5 used for control of bacterial infections responsible for a
days beginning 14 days after intratracheal challenge had great part of the economic loss associated with MPS.
no effect on incidence or severity of experimentally in- Penicillin, streptomycin, and erythromycin are of no val-
duced MPS (Ross, unpublished data—1981). In another ue in treatment of MPS.
study Ross and Skelly (unpublished data—1982) found
that tylosin phosphate at 100 g/ton fed for 8 weeks had PREVENTION
no effect on severity of lung lesions in pigs with natural-
ly acquired MPS. Early methods for prevention of MPS were reviewed by
Evidence has been presented that lincomycin fed at Switzer and Ross (1975). Effective control of the disease
200 g/ton for 3 weeks reduces the incidence and severity depends on providing an optimal environment, includ-
of MPS and results in improved performance (Van Bu- ing air quality, ventilation, and temperature, and proper
ren 1983). However, Ross (unpublished data—1985) stocking density. Use of a strict all-in/all-out production
found that continuous medication with lincomycin at scheme is possibly the most effective way to control the
500 g/ton of feed did not prevent transmission of M. hy- disease in infected herds (Clark et al. 1990; Scheidt et al.
opneumoniae from pigs with clinically overt MPS to in- 1990c). Clark et al. (1989) have recommended no more
contact susceptible pigs, nor did it reduce the severity of than 3 weeks’ variation in age in stocking a room. The
pneumonia in pigs with early-stage established disease. strategy of using repopulation to gain improved perfor-
Tiamulin at 200 ppm in feed for 10 days reduced the mance may be in part related to reduction in the severity
severity of experimentally induced (Schuller et al. 1977a) of, or complete elimination of, the retarding effects of
and naturally occurring (Martineau et al. 1980) MPS. MPS.
Tiamulin at 200 ppm in the feed for 10 days (Martineau Various health control schemes have been developed
et al. 1980) improved growth rates; given at 30 ppm con- which emphasized certification of herds free of MPS and
tinuously (Burch 1984b), it improved weight gains and other diseases. Methods used included on-farm inspec-
feed efficiency in the presence of MPS. Administration of tion, rigid isolation, and periodic examination of lungs
0.006% tiamulin in the drinking water (Johnson et al. from slaughter pigs reared in close confinement (Good-
1978) or parenterally at 15 mg/kg for 3 days (Burch win and Whittlestone 1967). Whittlestone (1979) has re-
1984a) was reported to result in clinical improvement viewed problems attendant to breakdowns in various
and improved growth in the presence of naturally occur- systems for maintenance of MPS-free herds.
ring MPS. However, in spite of the many published re- It has been known for many years that older sows that
ports on the efficacy of tiamulin, we were unable to de- have recovered from MPS are less likely to carry M. hyo-
tect a beneficial effect when the drug was administered in pneumoniae. A number of systems have been devised to
the drinking water at 60, 120, or 180 ppm for 10 days to take advantage of this natural adjunct to disease control.
CHAPTER 36 MYCOPLASMAL DISEASES Ross 501

Serotesting of older sows to detect those that are gram of clinical observation, commingling of progeny
seropositive, with the objective of culling them to estab- with known susceptible swine during growing and fin-
lish an MPS-free herd, has been attempted with variable ishing, slaughter examination of lungs, and milk and
success (Schuller et al. 1977b). Renewed efforts to use a blood serology with the Tween 20 ELISA.
serotest and cull strategy for controlling MPS should uti- Dee (1994) reported use of a modified MEW pro-
lize the Tween 20 ELISA discussed earlier. In work with gram to eliminate M. hyopneumoniae from two swine op-
the Tween 20 ELISA, Zimmermann et al. (1986) found erations. He reported that the program resulted in im-
substantially more positive animals when colostrum was provements in pig growth rate, reduced mortality,
tested rather than serum. reduced medication, and reduced vaccination costs per
The SPF pig program has found wide application in pig weaned. Clark et al. (1994) also assessed variations
the control of MPS. Producers that have repopulated ac- on the MEW approach and demonstrated that the pro-
cording to this system or with second-generation stock cedure appeared to eliminate M. hyopneumoniae com-
derived from primary herds have benefited greatly, pletely, along with several other important swine
although breakdowns have posed a problem. Monitor- pathogens. Similar results were obtained by Dritz et al.
ing for continued freedom from MPS should include (1996) using a segregated early-weaning procedure in
use of the most sensitive and specific tests, such as which no medication was used. Additional information
ELISA, for detection of subclinical M. hyopneumoniae in- regarding application of various high-health-status rear-
fections. ing strategies and their implications for M. hyopneumoni-
Alexander et al. (1980) developed the medicated ear- ae disease can be obtained in Dial et al. 1992.
ly-weaning (MEW) system, wherein intensive medica- Protective immunity develops in swine recovered
tion of the sow during late gestation and immediately from MPS. M. hyopneumoniae vaccines consisting of ad-
following parturition and of the newborn piglet is used juvanted whole-cell preparations have been shown to in-
to derive pigs free of M. hyopneumoniae. Pigs derived duce at least partial protection against development of
were free of MPS, and monitoring revealed no evidence gross lesions of experimentally induced MPS (Goodwin
of M. hyopneumoniae or Bordetella bronchiseptica in repre- and Whittlestone 1973; Ross et al. 1984). Commercial
sentative pigs examined from 5 weeks of age to slaugh- development of adjuvanted whole-cell vaccines has been
ter. Harris (1990) has devised a similar strategy reported recently (Dayalu and Ross 1990; Peterson et al.
(IsoweanTM) utilizing early weaning and rearing in three 1990; Kobisch et al. 1994). Field evaluations have been
isolated sites to prevent transmission of a variety of dis- carried out with these vaccines (Lium et al. 1994; Dayalu
eases, including M. hyopneumoniae, from the sow to the 1994). The utility of mycoplasma vaccines in the field de-
piglet. Pigs reared using the IsoweanTM strategy gained pends on clear evidence that the mycoplasma vaccine
substantially better than those reared in the traditional will significantly reduce losses caused by pneumonia in a
manner (Harris et al. 1990). Zimmermann et al. (1989) given swine herd. The PRDC has evolved in spite of the
reported that a control program based on removal of all use of mycoplasma vaccines. Whether current vaccines
young swine and gilts, coupled with feeding of tiamulin contain the appropriate mycoplasma antigens has not
or chlortetracycline/tylosin/sulfadimidine to the re- been determined. As demonstrated by Morrow et al.
maining older sows, resulted in elimination of M. hyo- (1994), use of M. hyopneumoniae vaccines should be
pneumoniae from 16 out of 17 Swiss herds. Continued based upon evaluation of the severity of disease in the
surveillance to ensure that the organism had not reen- herd; in their study, use of a M. hyopneumoniae vaccine in
tered the herd was accomplished with a four-prong pro- a herd with a low prevalance of infection had no demon-
strable effect on average daily gain.

M. HYORHINIS POLYSEROSITIS AND ARTHRITIS


Respiratory tract infection with M. hyorhinis has been re- ETIOLOGY
ported in swine from many different countries. It is usu-
ally the first mycoplasma isolated when an investigator Media and methods for isolation and growth of M. hy-
begins searching for these organisms in swine tissue. The orhinis have been summarized by Ross and Whittlestone
organism is an extremely common contaminant in cell (1983).
culture lines.
502 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

EPIDEMIOLOGY urally born pigs. It is possible that the pulmonary patho-


genicity of M. hyorhinis is strain variable. Although M.
M. hyorhinis infections are transmitted to young pigs hyorhinis is a frequent isolate from pneumonia with lym-
from infected sows or from older pigs in a nursery or phoreticular hyperplasia characteristic of that produced
grower unit. The organism can be isolated from the nasal by M. hyopneumoniae, it is also frequent in lesions of oth-
or sinus secretions of about 10% of sows (Ross and Spear er bronchopneumonias. No difference has been detected
1973) and from the nasal secretions of about 30–40% of in severity between pneumonias with or without M. hy-
weanling swine. The organism is also common in pneu- orhinis.
monic lungs of slaughter swine. Nasal and tracheo-
bronchial infections with M. hyorhinis appear to spread CLINICAL SIGNS
rapidly once one pig in a group becomes infected. Many
infected pigs evidence no clinical disease, although ex- Outbreaks of polyserositis generally occur in swine 3–10
perimental work with gnotobiotic pigs and a field study weeks of age, although occasionally the disease occurs in
by Kott (1983) suggest that it may play an important role young adult swine. The first clinical evidence of illness
in pneumonias of baby pigs. occurs 3–10 days after exposure or after a precipitating
stress. Acute-phase signs consist of a roughened hair
PATHOGENESIS coat, moderate temperature elevations, listlessness,
moderate inappetence, reluctance to move, labored
M. hyorhinis is very common in nasal and tracheo- breathing, tucked-up appearance, abdominal tender-
bronchial secretions of young swine. It seems likely that ness, lameness, and swollen joints. The duration and de-
other diseases such as pneumonia or stress facilitate gree of clinical change during the acute phase vary, de-
septicemic infection of pigs already colonized with the pending on the severity of the lesions. Acute-phase signs
organism. When this occurs, the organism may localize begin to abate 10–14 days after onset, and the main clin-
in the joints and the serous-membrane-lined body cavi- ical signs thereafter consist of lameness and swollen
ties, where it then elicits acute serofibrinous inflamma- joints. Joint involvement is most severe during the suba-
tion. cute stage of the disease. Lameness and joint swelling
M. hyorhinis can be isolated from acute-stage lesions may become less severe 2–3 months after onset, al-
of polyserositis or arthritis. During the subacute disease though some pigs are still lame 6 months later.
(i.e., 2 weeks to 3 months after onset) it can still be iso-
lated but is present in a decreasing number of sites. The LESIONS
organism persists for as long as 6 months in some affect-
ed joints. Acute-stage lesions consist of acute serofibrinous and
Some strains of the organism produce pneumonia in fibrinopurulent pericarditis, pleuritis, and to a lesser ex-
gnotobiotic pigs (Gois and Kuksa 1974). Less clear is its tent peritonitis (Fig. 36.5). Acute joint lesions consist of
capability to act as a primary cause of pneumonia in nat- swelling and hyperemia of the synovial membranes and

36.5. Lesions in a pig with


acute M. hyorhinis polyserositis
(12 days postinfection).
Fibrinopurulent exudate on the
liver and heart. Pericardium is
thickened. (By permission, New
York Academy of Sciences.)
CHAPTER 36 MYCOPLASMAL DISEASES Ross 503

increased amounts of serosanguineous synovial fluid. DIAGNOSIS


Subacute-phase serosal membrane lesions consist of
organized fibrous adhesions and thickened, cloudy Gross lesions of serofibrinous to fibrinopurulent poly-
serosal membranes. Synovial membranes may be quite serositis and arthritis in 3- to 10-week-old swine are sug-
thickened, and villous hypertrophy is evident. Synovial gestive of M. hyorhinis disease, although similar lesions
fluid is serosanguineous and increased markedly in are also caused by Haemophilus parasuis and other
amount. In later stages, 3–6 months, lesions appear less agents. The organism generally can be isolated from
active, although cartilage erosion and pannus formation acute- and early-subacute-phase lesions. Animals exam-
may be seen. Serosal lesions consist of organized fibrous ined for diagnosis should be killed in the acute stage of
adhesions. the disease. Autolytic changes in dead animals may re-
Japanese workers have demonstrated that M. hyorhi- duce the chances of isolating M. hyorhinis.
nis infection is very common in the eustachian tube of
young pigs. Morita et al. (1995) studied the pathology of TREATMENT
the ears of 479 pigs ranging in age from 1 day to 1 year.
Histological evidence of otitis was found in 76% of the Although M. hyorhinis is generally quite sensitive to ty-
pigs. Immunohistochemical examination revealed M. hy- losin or lincomycin in vitro, antibiotic therapy of swine
orhinis on the eustachian epithelia in 14 of 28 pigs exam- clinically ill with M. hyorhinis disease is not very satisfac-
ined. All pigs with this infection had an acute eustachitis. tory. The inflammatory response seems to either prevent
Mycoplasmas were also demonstrated among the cilia of antibiotic penetration or be self-perpetuating. Treatment
the eustachian tube using electron microscopy. The au- with tylosin or lincomycin on a herd basis may be bene-
thors suggested that porcine otitis media may be caused ficial.
primarily by M. hyorhinis infection.
In addition to eustachian tube infection, M. hyorhinis PREVENTION
has been associated with conjunctivitis in swine (Friis
1976). Rogers et al. (1991) demonstrated a mycoplasma- Control programs should emphasize control of other
like organism in association with conjunctivitis in swine conditions such as respiratory or enteric disease or stress
based upon ultrastructural examination but did not that might predispose to M. hyorhinis disease. No data
identify the mycoplasma. are available on the efficacy of prophylactic feed medica-
tion with tylosin, lincomycin, or other antibiotics.

M. HYOSYNOVIAE ARTHRITIS
Arthritis associated with M. hyosynoviae infection has EPIDEMIOLOGY
been reported principally in the United States, although
the disease also has been reported in England (Roberts et The organism is common in nasal and pharyngeal secre-
al. 1972; Blowey 1993) and Germany (Ross et al. 1977). tions of sows (Ross and Spear 1973), apparently persist-
Nielsen (1988) reported severe outbreaks of bursitis as- ing in such carrier swine indefinitely. Transmission from
sociated with M. hyosynoviae infection in Danish slaugh- adult carrier swine to young piglets generally does not
ter pigs. Buttenshon et al. (1995) reported isolation of occur until the pigs are 4–8 weeks of age. Shedding of
M. hyosynoviae from 8–9% of synovial fluid samples col- large numbers of the organism in mucosal secretions oc-
lected from nonpurulent arthritis of Danish slaughter curs during the acute stage of the disease.
pigs. Nasal and pharyngeal infection with M. hyosynoviae
occurs first in a few pigs at 4–8 weeks of age (Ross
ETIOLOGY and Spear 1973). The spread among penmates has
not been studied thoroughly; however, it seems that in
M. hyosynoviae can be isolated best in mycoplasma medi- some herds most pigs have experienced nasal and/or
um containing mucin (Ross and Karmon 1970). A selec- pharyngeal infection by 12 weeks of age. The rate of
tive procedure for isolation of M. hyosynoviae in the pres- spread might be related to population density and envi-
ence of M. hyorhinis has been reported (Friis et al. 1990). ronment.
504 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

PATHOGENESIS ing that, severity of lameness generally decreases. Many


animals recover and evidence no further lameness, or on-
M. hyosynoviae can be isolated from the blood 2–4 days ly stiffness. Some pigs may be lame for several weeks or
following intranasal exposure. The septicemia persists months; however, clinical signs in such animals are often
for 8–10 days, and during this time joint infection may a reflection of osteochondrosis as well. Morbidity of M.
occur. The duration and severity of joint infection are hyosynoviae arthritis varies from 1% to 50% in affected
variable. Many infected joints do not have lesions of herds. Mortality is very low, occurring only when affect-
arthritis and are infected for only a few days. Other joints ed animals cannot eat, are killed by penmates, or devel-
evidence acute arthritis; the organism probably grows to op complicating suppurative arthritis or bursitis
higher titers and persists longer in these joints. M. (Nielsen et al. 1996).
hyosynoviae has been isolated as long as 24 days postinoc-
ulation. LESIONS
Factors that contribute to development of lesions in
joints have not been determined. The arthritis is espe- Synovial membranes in acute M. hyosynoviae arthritis are
cially severe in heavily muscled swine that have poor leg swollen, edematous, and hyperemic (Fig. 36.6). Synovial
action and conformation (i.e., stilted gait with especially fluid is increased markedly in volume and is serofibri-
straight legs). Osteochondrosis, a degenerative disease of nous to serosanguineous. Periarticular tissues may be
the epiphyseal and subchondral osteoid tissue, is also edematous. Pigs with subacute disease have yellow to
extremely common in the same type of pig. It seems brown, thickened hyperemic membranes. Mild villous
likely that joint lesions developing as a result of osteo- hypertrophy may be seen. In chronic stages the mem-
chondrosis or trauma-induced bursal lesions (Nielsen branes are more thickened, and pannus formation may
1988) might predispose to arthritis caused by M. hyo- be seen. Articular cartilage changes may be seen, but
synoviae. these are usually focal and are very likely the result of os-
teochondrosis. The periarticular fibrosis seen in
CLINICAL SIGNS erysipelas is not observed.

Clinical evidence of M. hyosynoviae disease occurs princi- DIAGNOSIS


pally in pigs 12–24 weeks of age. Lameness appears sud-
denly and may occur in one or more limbs. Temperature An outbreak of acute lameness in 10- to 20-week-old
elevations generally are not detected. Severely affected swine that is not responsive to penicillin is suggestive of
pigs may have slight to moderate inappetence and loss of M. hyosynoviae arthritis. Definitive diagnosis of the dis-
weight. Animals with the disease usually have difficulty ease should be based on isolation of the organism from
rising or in some instances are not able to rise. Joint typical lesions. Animals examined should be representa-
swelling is usually not observed because joints covered tive of the herd problem, be in the acute stage of the dis-
with muscle are most often involved. Pseudocysts or cal- ease, and be unmedicated. Fluids may be aspirated from
luses on the cranial surface of the carpal joint or the joints of live animals or from slaughtered or necropsied
plantar and lateral surface of the tarsal joint may become animals (Nielsen et al. 1996). It is best to examine sever-
swollen (Nielsen 1988). al representative fluids, since some will be culture-nega-
Acute-phase signs persist for 3–10 days, and follow- tive.

36.6. Femorotibial joint of a pig


with M. hyosynoviae arthritis
(14 days postinfection). Synovial
membrane is darkened and
hypertrophied. Articular cartilage
is normal. (By permission, New
York Academy of Sciences.)
CHAPTER 36 MYCOPLASMAL DISEASES Ross 505

Antibodies detectable by means of the CF test devel- thor indicated some success through feed medication
op quickly during M. hyosynoviae disease (Zimmermann with 100 ppm of tiamulin hydrogen fumarate for 21 days
and Ross 1982). Many pigs have subclinical disease and or water medication with 8.8 mg/kg per day with tia-
develop antibodies to the organism; therefore, such tests mulin hydrogen fumarate. Pigs with bone and joint dam-
should be used with paired serums collected during the age caused by osteochondrosis will not respond well to
acute and early convalescent stages of the disease to as- any treatment.
certain that an antibody rise coincides with the clinical
problem. Serologic testing for M. hyosynoviae is not gen- PREVENTION
erally available.
Preventive measures should include selecting breeding
TREATMENT stock with good leg action and conformation, preventing
stress during the susceptible age, and allowing new
Successful treatment of M. hyosynoviae disease is best young breeding stock time to adjust to a new location.
achieved by use of injectable tylosin, lincomycin, or tia- Protective immunity results from infection with M.
mulin, possibly in combination with a corticosteroid. hyosynoviae. Previous speculation that pigs experiencing
Burch (1984a) reported that intramuscular treatment the infection prior to 12 weeks of age might be protected
with 10 mg/kg tiamulin or lincomycin for 3 days result- against the clinical disease at an older age has been dis-
ed in reduced lameness and improved gains in swine puted by Blowey (1993). The author found that CF titers
with naturally occurring mycoplasmal arthritis. Blowey to M. hyosynoviae increased with age and that serocon-
(1993) indicated that single injections of tiamulin hydro- version was not associated with onset of clinical disease.
gen fumarate (15 mg/kg) reduced severity of the disease Vaccines have been shown to be efficacious (Ross 1978)
but did not totally suppress the condition. The same au- but have not been made available commercially.

OTHER MYCOPLASMAS FROM SWINE


Switzer and Ross (1975) and Whittlestone (1979) have with members of the genus Mycoplasma. They differ in
reviewed information on mycoplasmas isolated from having a larger genome and being capable of growth in
swine other than M. hyorhinis, M. hyopneumoniae, and M. medium not containing sterols. These organisms are oc-
hyosynoviae. Isolates reported include mycoplasmas that casionally isolated from respiratory disease in swine,
normally are isolated from some other animal species; from nasal and pharyngeal secretions of swine, or from
previously unreported strains that represent new species environmental sources such as fecal material or recycled
but are not yet associated with disease in swine; and effluent used to remove manure from swine facilities.
acholeplasmas, saprophytic organisms that are common Convincing evidence that these organisms are important
in a wide variety of animal and plant sources. in naturally occurring swine respiratory disease has not
M. flocculare is a species that has been isolated from appeared. Gois et al. (1969) demonstrated that achole-
lungs and nasal cavities of swine in Denmark (Meyling plasmas may be the only mycoplasmas isolated from res-
and Friis 1972), England, and the United States (Arm- piratory tracts of swine in respiratory-disease-free herds.
strong and Friis 1981). Friis (pers. comm.—1978) indi-
cated that it is widespread in the upper respiratory tracts REFERENCES
of Danish swine, including SPF swine. Friis (1973) pre-
sented evidence that M. flocculare produced lymphocytic Alexander, T. J. L.; Thornton, K.; Boon, G.; Lysons, R. J.; and
infiltration in the nasal mucosa and peribronchial areas Gush, A. F. 1980. Medicated early weaning to obtain
of gnotobiotic pigs inoculated with the organism. Evi- pigs free from pathogens endemic in the herd of origin.
dence that it is of significance in naturally occurring res- Vet Rec 106:114–119.
piratory disease has not been presented. Amass, S. F.; Clark, L. K.; Van Alstine, W. G.; Bowersock, T.
The genitourinary tract is a common site of infection L.; Murphy, D. A.; Knox, K. E.; and Albregts, S. R. 1994.
with mycoplasmas in several animal species. Evidence Interaction of Mycoplasma hyopneumoniae and Pasteurel-
for a consistent mycoplasma flora in the genitourinary la multocida infections in swine. J Am Vet Med Assoc
tracts of swine has not been presented. 204:102–107.
Other mycoplasmas reported as occasional or inci- Armstrong, C. H., and Friis, N. F. 1981. Isolation of My-
dental isolates from swine include M. sualvi, M. coplasma flocculare from swine in the United States. Am
hyopharyngis, M. arginini, M. bovigenitalium, M. buccale, J Vet Res 42:1030–1032.
M. gallinarum, M. iners, M. mycoides, and M. salivarium. Armstrong, C. H.; Freeman, M. J.; Sands-Freeman, L.;
Acholeplasmas have many characteristics in common Lopez-Osuna, M.; Young, T.; and Runnels, L. J. 1983.
506 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Comparison of the enzyme-linked immunosorbent as- P114 from Mycoplasma hyopneumoniae. Proc 11th Int
say and the indirect hemagglutination and complement Congr Int Org Mycoplasmol 4:131.
fixation tests for detecting antibodies to Mycoplasma hy- Ciprian, A.; Pijoan, C.; Cruz, T.; Camacho, J.; Tortora, J.; Col-
opneumoniae. Can J Comp Med 47:464–470. menares, G.; Lopez-Revilla, R.; and de la Garza, M.
Artiushin, S., and Minion, F. 1996. Arbitrarily primed PCR 1988. Mycoplasma hyopneumoniae increases the suscepti-
analysis of Mycoplasma hyopneumoniae field isolates bility of pigs to experimental Pasteurella multocida pneu-
demonstrates genetic heterogeneity. Int J Syst Bacteriol monia. Can J Vet Res 52:434–438.
46:324–328. Clark, L. K.; Scheidt, A. B.; Mayrose, V. B.; Armstrong, C. H.;
Bereiter, M.; Young, T. F.; Joo, H. S.; and Ross, R. F. l990. Cline, T. R.; and Knox, K. 1989. Methodology to reduce
Evaluation of the ELISA, and comparison to the comple- days to market in swine herds with enzootic pneumonia.
ment fixation test and radial immunodiffusion enzyme Proc Iowa Vet Med Assoc 107:129–135.
assay for detection of antibodies against Mycoplasma hy- Clark, L. K.; Scheidt, A. B.; Mayrose V. B.; Armstrong, C. H.;
opneumoniae in swine serum. Vet Microbiol 25:177–192. and Knox, K. 1990. Prevention of the development of
Betts, A. O. 1952. Respiratory diseases of pigs. V. Some clin- enzootic pneumonia within an infected swine herd. Proc
ical and epidemiological aspects of virus pneumonia of Int Congr Pig Vet Soc 11:91.
pigs. Vet Rec 64:283–288. Clark, L. K.; Hill, M. A.; Kniffen, T. S.; VanAlstine, W.;
Blanchard, B.; Vena, M. M.; Cavalier, A.; Le Lannic, J.; Stevenson, G.; Meyer, K. B.; Wu, C. C.; Scheidt, A. B.;
Gouranton, J.; and Kobisch, M. 1992. Electron micro- Knox, K.; and Albregts, S. 1994. An evaluation of the
scopic observation of the respiratory tract of SPF piglets components of medicated early weaning. Sw Hlth Prod
inoculated with Mycoplasma hyopneumoniae. Vet Micro 2:5–11.
30:329–341. Cooper, A. C.; Fuller, J. R.; Fuller, M. K.; Whittlestone, P.;
Blanchard, B.; Kobisch, M.; Bove, J. M.; and Saillard, C. and Wise, D. R. 1993. In vitro activity of danofloxacin,
1996. Polymerase chain reaction for Mycoplasma hyo- tylosin and oxytetracycline against mycoplasmas of vet-
pneumoniae detection in tracheobronchiolar washings erinary importance. Res Vet Sci 54:329–334.
from pigs. Mol Cell Prob 10:15–22. Dayalu, K. I. 1994. Beneficial effects of a Mycoplasma hyo-
Blowey, R. W. 1993. Mycoplasma hyosynoviae arthritis. Pig pneumoniae vaccine (RespisureTM, Stellamune Mycoplas-
Vet J 30:72–76. maTM) as evaluated under both experimental and field
Bommeli, W.-R., and Nicolet, J. 1983. A method for the eval- conditions. Proc Int Congr Pig Vet Soc 13:137.
uation of enzyme linked immunoassay results for diag- Dayalu, K. I., and Ross, R. F. 1990. Evaluation of experimen-
nosing enzootic pneumonia in pig herds. Proc Int Symp tal vaccines for control of porcine pneumonia induced
World Assoc Vet Lab Diag 3(2):439–442. by Mycoplasma hyopneumoniae. Proc Int Congr Pig Vet
Bruggmann, S.; Engberg, B.; and Ehrensperger, F. 1977. Soc 11:83.
Demonstration of M. suipneumoniae in pig lungs by the DeBey, M. C., and Ross, R. F. 1994. Ciliostasis and loss of cil-
enzyme-linked immunoperoxidase technique. Vet Rec ia induced by Mycoplasma hyopneumoniae in porcine tra-
101:137. cheal organ cultures. Inf Immun 62:5312–5318.
Burch, D. G. S. 1984a. The evaluation of tiamulin by injec- Dee, S. A. 1994. Apparent prevention of Mycoplasma hyo-
tion for the treatment of enzootic pneumonia and my- pneumoniae infection in growing pigs with a low-cost
coplasmal arthritis of pigs. Proc Int Congr Pig Vet Soc modified medicated-early-weaning program. Sw Hlth
8:117. Prod 2:7–12.
———. 1984b. Tiamulin feed premix in the improvement Dial, G. D.; Wiseman, B. S.; Davies, P. R.; Marsh, W. E.;
of growth performance of pigs in herds severely affected Molitor, T. W.; Morrison, R. B.; and Thawley, D. G.
with enzootic pneumonia. Vet Rec 114:209–211. 1992. Stategies employed in the United States for im-
Burch, D. G. S.; Jones, G. T.; Heard, T. W.; and Tuck, R. E. proving the health of swine. Proc Minn Sw Conf Vet
1986. The synergistic activity of tiamulin and chlorte- 19:1–26.
tracycline: In-feed treatment of bacterially complicated Dritz, S. S.; Chengappa, M. M.; Nelssen, J. L.; Tokach, M. D.;
enzootic pneumonia in fattening pigs. Vet Rec Goodband, R. D.; Nietfeld, J. C.; and Staats, J. J. 1996.
119:108–112. Growth and microbial flora of nonmedicated, segregat-
Buttenshon, J.; Svensmark, B.; and Kyrval, J. 1995. Non-pu- ed, early weaned pigs from a commercial swine opera-
rulent arthritis in Danish slaughter pigs. I. A study of tion. J Am Vet Med Assoc 208:711–715.
field cases. J Vet Med A 42:633–641. Etheridge, J. R.; Cottew, G. S.; and Lloyd, L. C. 1979. Isola-
Caruso, J., and Ross, R. F. 1990. Effects of Mycoplasma hyo- tion of Mycoplasma hyopneumoniae from lesions in ex-
pneumoniae and Actinobacillus (Haemophilus) pleuropneu- perimentally infected pigs. Aust Vet J 55:356–359.
moniae infections on alveolar macrophage functions in Farrington, D. O. 1976. Immunization of swine against my-
swine. Am J Vet Res 51:227–231. coplasmal pneumonia. Proc Int Congr Pig Vet Soc 4:4.
Chen, J. R.; Lin, C. S.; and Weng, C. N. 1996. An adhesin of Feld, N. C.; Qvist, P.; Ahrens, P.; Friis, N. F.; and Meyling, A.
CHAPTER 36 MYCOPLASMAL DISEASES Ross 507

1992. A monoclonal blocking ELISA detecting serum induced M. hyopneumoniae pneumonia in pigs. IOM Lett
antibodies to Mycoplasma hyopneumoniae. Vet Microbiol 1(8):351–352.
30:35–46. Hannan, P. C. T., and Ripley, P. H. 1996. In vitro develop-
Friis, N. F. 1973. The pathogenicity of Mycoplasma floccu- ment of resistance in Mycoplasma hyopneumoniae to
lare. Acta Vet Scand 14:344–346. SDZ PMD 296, tiamulin and two other antimicrobial
———. 1975. Some recommendations concerning primary agents. Proc Int Congr Pig Vet Soc 14:225.
isolation of Mycoplasma suipneumoniae and Mycoplasma Hannan, P. C. T.; O’Hanlon, P. J.; and Rogers, N. H. 1989. In
flocculare. Nord Vet Med 27:337–339. vitro evaluation of various quinolone antibacterial
———. 1976. A serologic variant of Mycoplasma hyorhinis agents against veterinary mycoplasmas and porcine
recovered from the conjunctiva of swine. Acta Vet Scand respiratory bacterial pathogens. Res Vet Sci 46:202–
17:343–353. 211.
Friis, N. F.; Larsen, H.; and Ahrens, P. 1990. Propagation of Harris, D. L. 1990. The use of Isowean 3 site production to
Mycoplasma hyosynoviae in the presence of specific sup- upgrade health status. Proc Int Congr Pig Vet Soc
pressing factors against Mycoplasma hyorhinis. IOM Lett 11:374.
1:377–378. Harris, D. L.; Edgerton, S. L.; and Wilson, E. R. 1990. Large
Gois, M., and Kuksa, F. 1974. Intranasal infection of gnoto- thymus glands in Isowean pigs. Proc Int Congr Pig Vet
biotic piglets with Mycoplasma hyorhinis: Differences in Soc 11:291.
virulence of the strains and influence of age on the de- Holmgren, N. 1974. Swine enzootic pneumonia: Immuno-
velopment of infection. Zentralbl Veterinärmed (B) logic studies in infected sow herds. Res Vet Sci
21:352–361. 17:145–153.
Gois, M.; Cerny, M.; Rozkosny, V.; and Sovadina, J. 1969. Huhn, R. G. 1971. The action of certain antibiotics and
Studies on the epizootiological significance of some ether on swine enzootic pneumonia. Can J Comp Med
species of mycoplasma isolated from nasal swabs and 35:1–4.
lungs of pigs. Zentralbl Veterinärmed (B) 16:253–265. Inamoto, T.; Takahashi, H.; Yamamoto, K.; Nakai, Y.; and
Goodwin, R. F. W. 1972a. Isolation of Mycoplasma suipneu- Ogimoto, K. 1994. Antibiotic susceptibility of Mycoplas-
moniae from the nasal cavities and lungs of pigs affected ma hyopneumoniae isolated from swine. J Vet Med Sci
with enzootic pneumonia or exposed to this infection. 56:393–394.
Res Vet Sci 13:262–267. Johnson, R. J.; Maplesden, D. C.; and Szanto, J. 1978. Effica-
———. 1972b. Experiments on the transmissibility of en- cy of tiamulin in the treatment of mycoplasmal pneu-
zootic pneumonia of pigs. Res Vet Sci 13:257–261. monia of swine. Proc Int Congr Pig Vet Soc 5:M.25.
———. 1985. Apparent reinfection of enzootic pneumo- Jorsal, S. E., and Thomsen, B. L. 1988. A Cox regression
nia-free pig herds: Search for possible causes. Vet Rec analysis of risk factors related to Mycoplasma suipneu-
116:690–694. moniae reinfection in Danish SPF herds. Acta Vet Scand
Goodwin, R. F. W., and Whittlestone, P. 1967. The detection Suppl 29:436–438.
of enzootic pneumonia in pig herds. I. Eight years gen- Kobisch, M.; Labbe, A.; Morvan, P.; and Cariolet, R. 1994.
eral experience with a pilot control scheme. Vet Rec Evaluation of a Mycoplasma hyopneumoniae vaccine in
181:643–647. pigs experimentally infected with Mycoplasma hyopneu-
———. 1973. Enzootic pneumonia of pigs: Immunization moniae and Pasteurella multocida. Proc Int Congr Pig Vet
attempts inoculating Mycoplasma suipneumoniae anti- Soc 13:194.
gen by various routes and with different adjuvants. Br Koh, H. B.; Jeong, Y. U.; Kim, J. J.; and Ahn, S. H. 1996. Con-
Vet J 129:456–464. trol of Mycoplasma hyopneumoniae from infected herds
Goodwin, R. F. W.; Pomeroy, A. P.; and Whittlestone, P. by medication. Proc Int Congr Pig Vet Soc 14:240.
1965. Production of enzootic pneumonia in pigs with a Kott, B. E. 1983. Chronological Studies of Respiratory Dis-
mycoplasma. Vet Rec 77:1247–1249. ease in Baby Pigs. M.S. thesis, Iowa State Univ.
Guerrero, R. J. 1990. Respiratory disease: An important Kuwano, A.; Yamamoto, K.; Takei, M.; Kato, M.; and Tachi,
global problem in the swine industry. Proc Int Congr Pig H. 1992. Evaluation of the effect of ofloxacin on experi-
Vet Soc 11:98. mentally induced mycoplasmal pneumonia of swine.
Halbur, P. G. 1996. Changing trends in the porcine respira- Proc Int Congr Pig Vet Soc 12:320.
tory disease complex. In Proc 96 Pork Summit, Tempe, Lium, B.; Lund, A.; and Skomsoy, A. 1994. A field study on
Ariz, 2–5 May, pp. 76–80. vaccination against Mycoplasma hyopneumoniae infec-
Halbur, P. G.; Paul, P. S.; and Janke, B. H. 1993. Viral con- tion in pigs. Proc Int Congr Pig Vet Soc 13:191.
tributors to the porcine respiratory disease complex. Mare, C. J., and Switzer, W. P. 1965. New species: Mycoplas-
Proc Am Assoc Sw Pract 24:343–350. ma hyopneumoniae, a causative agent of virus pig pneu-
Hannan, P. C. T., and Goodwin, R. F. W. 1990. Efficacies of monia. Vet Med 60:841–846.
norfloxacin and its 6-chloro analogue in experimentally- ———. 1966. Virus pneumonia of pigs: Drug and ether
508 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

sensitivity of a causative agent. Am J Vet Res ———. 1993b. An epidemiologic and economic study of
27:1671–1675. respiratory diseases in two conventional Danish swine
Martineau, G.; Martineau-Doize, B.; Coignoul, F.; and De- herds. II. Associations between lesions present at
waele, A. 1980. Bilan éconmique après traitment de la slaughter and mean daily gains during specific intervals
broncho-pneumonie enzootique par la tiamuline. Ann of the growth period. Acta Vet Scand 34:331–344.
Méd Vét 124:369–377. Peterson, G.; Weiss, D.; Egan, J.; Korshus, J.; Peters, R.; and
Mattsson, J. G.; Bergstrom, K.; Wallgren, P.; and Johansson, Miron, M. 1990. Response to Mycoplasma hyopneumoni-
K.-E. 1995. Detection of Mycoplasma hyopneumoniae in ae vaccination in nursing piglets. Proc Int Congr Pig Vet
nose swabs from pigs by in vitro amplification of the 16S Soc 11:84.
rRNA gene. J Clin Micro 33:893–897. Pointon, A. M.; Byrt, D.; and Heap, P. 1985. Effect of en-
Mebus, C. A., and Underdahl, N. R. 1977. Scanning electron zootic pneumonia of pigs on growth performance. Aust
microscopy of trachea and bronchi from gnotobiotic Vet J 62:13–18.
pigs inoculated with Mycoplasma hyopneumoniae. Am J Pointon, A. M.; Hueston, W. D.; and Dial, G. D. 1990. Dis-
Vet Res 38:1249–1254. ease surveillance—Reducing the uncertainty of decision
Messier, S., and Ross, R. F. 1991. Interactions of Mycoplas- making. In Proc Allen D. Leman Swine Conf, pp. 38–57.
ma hyopneumoniae membranes with porcine lympho- Pullar, E. M. 1948. Infectious pneumonia of pigs. I. General
cytes. Am J Vet Res 52:1497–1502. description, differential diagnosis and epidemiology.
Meyling, A., and Friis, N. F. 1972. Serological identification Aust Vet J 24:320–330.
of a new porcine mycoplasma species, M. flocculare. Ac- Roberts, D. H.; Johnson, C. T.; and Tew, N. C. 1972. The iso-
ta Vet Scand 13:287–289. lation of Mycoplasma hyosynoviae from an outbreak of
Moore, C. 1996. Canadian perspective on PRDC. In Nat porcine arthritis. Vet Rec 90:307–309.
Pork Producers Council Summit, Des Moines, Iowa, pp. Rogers, D. G.; Frey, M. L.; and Hogg, A. 1991. Conjunctivitis
129–132. associated with a mycoplasma-like organism in swine. J
Morita, T.; Fukuda, H.; Awakura, T.; Shimada, A.; Umemu- Am Vet Med Assoc 198:450–452.
ra, T.; Kazama, S.; and Yagihashi, T. 1995. Demonstra- Ross, R. F. 1978. Immunization of swine against Mycoplas-
tion of Mycoplasma hyorhinis as a possible primary ma hyosynoviae. Zentralbl Bakteriol (Orig A) 241:246.
pathogen for porcine otitis media. Vet Path 32:107–111. ———. 1993. Control and elimination practices with My-
Morris, C. R.; Gardner, I. A.; Hietala, S. K.; Carpenter, T. E.; coplasma hyopneumoniae infection. In Iowa State Univ Sw
Anderson, R. J.; and Parker, K. M. 1994. Persistence of Dis Conf for Sw Pract, pp. 31–38.
passively acquired antibodies to Mycoplasma hyopneu- Ross, R. F., and Cox, D. F. 1988. Evaluation of tiamulin for
moniae in a swine herd. Prev Vet Med 21:29–41. treatment of mycoplasmal pneumonia in swine. J Am
Morrow, W. E. M.; Iglesias, G.; Stanislaw, C.; Stephenson, Vet Med Assoc 193:441–446.
A.; and Erickson, G. 1994. Effect of a mycoplasma vac- Ross, R. F., and Karmon, J. A. 1970. Heterogeneity among
cine on average daily gain in swine. Sw Hlth Prod strains of Mycoplasma granularum and identification of
2:13–18. Mycoplasma hyosynoviae, sp. n. J Bacteriol 103:707–713.
Nicolet, J.; Paroz, P.; and Bruggmann, S. 1980. Tween 20 sol- Ross, R. F., and Spear, M. L. 1973. Role of the sow as a reser-
uble proteins of Mycoplasma hyopneumoniae as antigen voir of infection for Mycoplasma hyosynoviae. Am J Vet
for an enzyme linked immunosorbent assay. Res Vet Sci Res 34:373–378.
29:305–309. Ross, R. F., and Stemke, G. W. 1996. Mycoplasma infections
Nielsen, N. C. 1988. Mycoplasma-associated enzootic bursi- of swine. In Molecular and Diagnostic Procedures in
tis in growing pigs. Proc Int Congr Pig Vet Soc 10:240. Mycoplasmology, vol 2. Ed. J. G. Tully and S. Razin. San
Nielsen, N. C.; Nielsen, E. O.; and Hagedorn-Olsen, T. 1996. Diego: Academic Press, pp. 275–281.
Field outbreaks of Mycoplasma hyosynoviae polyarthritis. Ross, R. F., and Whittlestone, P. 1983. Recovery of, identifi-
Proc Int Congr Pig Vet Soc 14:236. cation of and serological response to porcine mycoplas-
Noyes, E. P.; Feeny, D. A.; and Pijoan, C. 1990. Comparison mas. In Methods in Mycoplasmology, vol. 2. Ed. J. G.
of the effect of pneumonia detected during the lifetime Tully and S. Razin. New York: Academic Press.
with pneumonia detected at slaughter on growth in Ross, R. F.; Weiss, R.; and Kirchhoff, H. 1977. Nachweis von
swine. J Am Vet Med Assoc 197:1025–1029. M. hyorhinis und M. hyosynoviae in arthritischen Ge-
Owen, W. J. 1990. Food Animal Disease Monitoring. Ames, lenken von Schweinen. Zentralbl Veterinärmed
Iowa. Published by author. 24:741–745.
Paisley, L. G.; Vraa-Andersen, L.; Dybkjaer, L.; Moller, K.; Ross, R. F.; Zimmermann-Erickson, B. J.; and Young, T. F.
Christensen, G.; Mousing, J.; and Agger, J. F. 1993a. An 1984. Characteristics of protective activity of Mycoplas-
epidemiologic and economic study of respiratory dis- ma hyopneumoniae vaccine. Am J Vet Res 45:1899–1905.
eases in two conventional Danish swine herds. I. Preva- Ross, R. F.; Jackson, J. A.; Tanner, A. C.; Andrews, J. J.; and
lence of respiratory lesions at slaughter and their effects Magonigle, R. A. 1990. In vitro and in vivo efficacy of
on growth. Acta Vet Scand 34:319–329. danofloxacin against Mycoplasma hyopneumoniae. Proc
CHAPTER 36 MYCOPLASMAL DISEASES Ross 509

Int Congr Pig Vet Soc 11:85. response in pig after Mycoplasma hyopneumoniae infec-
Scheidt, A. B.; Mayrose, V. B.; Hill, M. A.; Clark, L. K.; Cline, tion: Effect of immunosuppression on pneumonia in pig
T. R.; Knox, K. E.; Runnels, L. J.; Frantz, S.; and Einstein, induced by M. hyopneumoniae. J Chin Soc Vet Sci
M. E. 1990a. Relationship of growth performance to 14:267–273.
pneumonia and atrophic rhinitis detected in pigs at Whittlestone, P. 1972. The role of mycoplasmas in the pro-
slaughter. J Am Vet Med Assoc 196:881–884. duction of pneumonia in the pig. In Pathogenic My-
Scheidt, A.; Froe, D.; Cline, T.; Mayrose, V.; and Einstein, M. coplasmas. Amsterdam: Associated Scientific Publish-
1990b. The use of long-acting oxytetracycline (LA200tm) ers, pp. 263–283.
in two swine herds for control of enzootic pneumonia. ———. 1979. Porcine mycoplasmas. In The Mycoplasmas.
Proc Int Congr Pig Vet Soc 11:87. Vol. 2, Human and Animal Mycoplasmas. Ed. J. G. Tully
Scheidt, A.; Clark, K.; Mayrose, V.; Cline, T.; Jones, D.; and and R. F. Whitcomb. New York: Academic Press, pp.
Frantz, S. 1990c. All-in, all-out as a means for improving 133–176.
growth in a swine herd affected by enzootic pneumonia. Windsor, H. M.; Ripley, P. H.; and Hannan, P. C. T. 1996. An
Proc Int Congr Pig Vet Soc 11:92. in vitro investigation of the sensitivity of German field
Schuller, W.; Laber, G.; and Walzl, H. 1977a. Chemothera- isolates of Mycoplasma hyopneumoniae and Mycoplasma
peutische Untersuchungen mit 81723 hfu (Tiamulin), hyosynoviae to SDZ PMD 296, a new pleuromulin deriv-
einem neuen Pleuromutilin-Derivat, an der experi- ative. Proc Int Congr Pig Vet Soc 14:226.
mentellen Mykoplasma-Pneumonie des Schweines. Yagihashi, T.; Nunoya, T.; Mitui, T.; and Tajima, M. 1984.
DTW 84:333–372. Effect of Mycoplasma hyopneumoniae infection on the de-
Schuller, W.; Neumeister, E.; and Vogl, D. 1977b. Zur velopment of Haemophilus pleuropneumoniae pneumo-
Sanierung von mit Enzootischer Pneumonie versuchten nia in pigs. Jpn J Vet Sci 46:705–713.
Schweinebestanden. Wien Tierärztl Monatsschr Yagihashi, T.; Kazama, S.; and Tajima, M. 1993. Seroepide-
64:156–160. miology of mycoplasmal pneumonia of swine in Japan
Simon, F.; Semjen, G.; Dobos-Kovacs, M.; Laczay, P.; and as surveyed by an enzyme-linked immunosorbent assay.
Cserep, T. 1990. Efficacy of enrofloxacin against enzoot- Vet Micro 34:155–166.
ic pneumonia in swine. Proc Int Congr Pig Vet Soc Young, T. F.; Ross, R. F.; and Drisko, J. 1983. Prevalence of
11:96. antibodies to Mycoplasma hyopneumoniae in Iowa swine.
Stark, K. D. C.; Keller, H.; and Eggenberger, E. 1992. Risk Am J Vet Res 44:1946–1948.
factors for the reinfection of specific pathogen-free pig Zhang, Q.; Young, T. F.; and Ross, R. F. 1995. Identification
breeding herds with enzootic pneumonia. Vet Rec and characterization of a Mycoplasma hyopneumoniae
131:532–535. adhesin. Inf Immun 63:1013–1019.
Switzer, W. P., and Ross, R. F. 1975. Mycoplasmal diseases. Zielinski, G., and Ross, R. F. 1993. Adherence of Mycoplas-
In Diseases of Swine, 4th ed. Ed. H. W. Dunne and A. D. ma hyopneumoniae to porcine ciliated respiratory tract
Leman. Ames: Iowa State Univ Press, pp. 741–764. cells. Am J Vet Res 54:1262–1269.
Thomsen, B. L.; Jorsal, S. E.; Andersen, S.; and Willeberg, P. Zielinski, G. C.; Young, T.; Ross, R. F.; and Rosenbusch, R. F.
1992. The Cox regression model applied to risk factor 1990. Adherence of Mycoplasma hyopneumoniae to cell
analysis of infections in the breeding and multiplying monolayers. Am J Vet Res 51:339–343.
herds in the Danish SPF system. Prev Vet Med Zimmerman, W. 1990. Erfarhungen mit der EP-Teil-
12:287–297. sanierung im Tilgungsprogramm des Schweizerichen
Tielen, M. 1995. Respiratory Diseases in Pigs: Prevalence Schweinegesundheitsdienstes (SGD). Tierarztl Um-
and Economical Effects. Pigs-Misset, Suppl, 4–5 June. schau 45:556–562.
Van Buren, J. W. 1983. Lincomycin and swine mycoplasmal Zimmermann, B. J., and Ross, R. F. 1982. Antibody re-
pneumonia indications and efficacy. Proc Am Assoc sponse of swine experimentally infected with Mycoplas-
Swine Pract, Cincinnati, pp. 29–40. ma hyosynoviae. Vet Microbiol 7:135–146.
Wallgren, P.; Schwan, O.; Mattsson, S.; and Bolske, G. 1996. Zimmermann, W.; Tschudi, P.; and Nicolet, J. 1986. ELISA-
Comparison of the sensitivity of two ELISA systems for serologie in Blut und Kolostralmilch: Eine Moglichkeit
detection of antibodies to Mycoplasma hyopneumoniae in zur Uberwachung der enzootischen Pneumonie (EP) in
naturally infected herds. Proc Int Congr Pig Vet Soc Schweine-Bestanden. Schweiz Arch Tierheilkd
14:217. 128:299–306.
Wannemuehler, M. W.; Minion, F. C.; and Ross, R. F. 1988. Zimmermann, W.; Odermatt, W.; and Tschudi, P. 1989. En-
Immune suppression of Mycoplasma hyopneumoniae in- zootische pneumonie (EP): Die Teilsanierung EP-Rein-
fected swine. Proc Int Org Mycoplasmol 7:72. fizierter Schweinezuchtbetriebe als alternative zur To-
Weng, C. N., and Lin, W. H. 1988. Cell-mediated immune talsanierung. Schweiz Arch Tierheilkd Bl:179–191.
Pneumonic Pasteurellosis
37 C. Pijoan

Pneumonic pasteurellosis, the result of Pasteurella multo- pneumonic pasteurellosis has been variable. In most
cida infection of the lung, is the common final stage of farms and groups of pigs, pneumonia at slaughter has
enzootic pneumonia or porcine respiratory disease com- decreased to negligible levels. However, some farms have
plex (PRDC). This syndrome is one of the most common presented with severe pneumonia during the late finish-
and costly diseases of pigs, especially when they are ing stages, around 16–18 weeks of age. This late pneu-
raised under confinement. Published data suggest that monia has been attributed mostly to infection by M. hy-
pneumonic lesions at slaughter are very common, even opneumoniae, but P. multocida is also commonly isolated.
in well-managed herds. Reports on the prevalence of pigs SEW, at least when used with weaning at 15 days, does
with pneumonic lesions at slaughter has varied from as not eliminate P. multocida from the offspring. Elimina-
low as 30% to as high as 80% in various studies through tion of M. hyopneumoniae is more variable and unpre-
the years. Recent data from the United States found a dictable, probably dependent on sow immunity, and is
prevalence of 75% of pigs with pneumonia and 13% with most probably the predisposing trigger for pneumonia
pleuritis in a sample of 6634 pigs inspected, with all in these systems.
herds studied showing some animals with lesions (Bahn-
son 1994), which highlights pneumonia as the most fre- ETIOLOGY
quent lesion seen at slaughter.
Pneumonia in pigs also appears to be a very costly P. multocida is a gram-negative coccobacillus, 0.5–1 ×
disease, although the actual cost has been difficult to cal- 1–2 µm in size. The organism is a facultative anaerobe,
culate and has varied widely in published results. Noyes growing well in most enriched media. It is oxidase posi-
et al. (1990) performed a radiographic study of pigs’ tive, nonmotile, indole positive, and urease negative. It
lungs in a commercial herd in order to evaluate lifetime does not grow well in MacConkey and is nonhemolytic
pneumonia and found a significant correlation between and does not require X and V factors. These reactions are
the extent of lifetime pneumonic lesions and the weight helpful in differentiating P. multocida from a group of
of the animals at 180 days. Bahnson (1994) compared closely related bacteria that are also involved in pul-
batches of finishing pigs sent to slaughter. The batch monary diseases of pigs, namely, P. haemolytica, Acti-
with the highest pneumonia scores had a 7.8% lower rate nobacillus suis, and A. pleuropneumoniae.
of gain than the batch with the lowest score, a difference P. multocida has five capsular serotypes, A, B, D, E,
that has considerable economic impact. and F of which A, B, and D have been reported in swine.
Pneumonic pasteurellosis occurs worldwide and in Serotype B however, is atypical in that it produces a
all climates and husbandry conditions. Specific- much more severe disease. It is also rare, confined to re-
pathogen-free (SPF) schemes, especially at the national gions of Southeast Asia, China, and India (Verma 1988).
level, do achieve a degree of control, presumably It has not been reported from natural outbreaks in pigs
through the eradication of Mycoplasma hyopneumoniae. in North America or Europe. The most common
However, P. multocida, which is a common inhabitant of serotype isolated from pneumonic lungs is A, although a
the pig’s nasal flora, is extremely difficult to eradicate variable proportion of serotype D strains is also found
and can be found in most high-health-status herds, such (Pijoan et al. 1983a, 1984; Kielstein 1986; Hoie et al.
as SPF or minimal-disease herds. Since pasteurellae can 1991; Rubies et al. 1996). P. multocida also has 16 somat-
interact directly with other agents, elimination of my- ic serotypes; strains of serotypes 3 and 5 are commonly
coplasmas does not give an absolute guarantee of con- detected in pigs, with strains A:3, A:5, D:5, and D:3 be-
trolling pneumonia. ing the most prevalent, in that order.
A more recent strategy for raising high-health-status
pigs has been to use a combination of segregation of the Virulence Factors
offspring together with early weaning. This program of The virulence factors of P. multocida are not well defined.
segregated early weaning (SEW) has been extremely suc- In particular, the importance of the dermonecrotic toxin
cessful in controlling and minimizing most of the com- (DNT) is unresolved. This toxin is central to the produc-
mon diseases that affect the pig. However, its impact on tion of atrophic rhinitis (see Chapter 27) when only tox-

511
512 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

igenic strains of P. multocida are involved in the disease. Trigo (1995) found that some strains, in particular toxi-
Toxigenic strains of P. multocida from lungs were first re- genic ones, had detectable pili on their surface, although
ported by Pijoan et al. (1984). Since then, a number of the role of these structures in adhesion is still under de-
authors have found increasing numbers of toxigenic bate. In contrast to their poor attachment to epithelial
strains (both type A and type D) in pneumonic lungs. surfaces, P. multocida strains have been shown to attach
Some reports (Kielstein 1986; Iwamatsu and Sawada readily to nasal mucus, raising questions as to where nor-
1988) show that between 25% and 45% of strains isolat- mal attachment and colonization take place.
ed from lungs are toxigenic. Kielstein (1986) found that The presence of capsule has been shown to decrease
toxigenic strains were frequently isolated from acute cas- the attachment of P. multocida to respiratory tract mu-
es but not from slaughterhouse lungs, suggesting en- cus, as well as to cultured tracheal rings (Jacques et al.
hanced virulence. However, Baekbo (1988) postulated 1993). On the other hand, the same group has reported
that toxigenicity was not important in determining the that preinfection of tracheal rings with Bordetella bron-
virulence of P. multocida in experimentally infected ani- chiseptica enhanced subsequent attachment by P. multo-
mals. cida (Dugal et al. 1992). Some other authors, however,
The role, if any, of toxigenicity in pneumonic pas- have had difficulties in confirming that P. multocida
teurellosis is still under debate. For example, Hoie et al. attached to immobilized mucus (Issacson and Trigo
(1991) found that 94% of serotype A and 90% of serotype 1995).
D isolates from pneumonic lungs were toxigenic. In con- Mucosal colonization of suckling pigs is becoming a
trast, Rubies et al. (1996) found no toxigenic strains (ei- very important issue in SEW systems. It has been postu-
ther A or D) in 218 isolates from pneumonic lungs in lated (Pijoan 1995) that pigs weaned early (at 15 days of
Spain. age or less) are not homogeneously colonized with or-
The capsule appears to be an important virulence fac- ganisms such as P. multocida and M. hyopneumoniae. As a
tor, especially in serotype A, for it may help the organism result, pigs are weaned into isolated nurseries in popula-
avoid phagocytosis by alveolar macrophages, at least in tions that have a variable prevalence of colonized ani-
vitro. Maheswaran and Thies (1979) reported that P. mals. Populations with low prevalence of colonization
multocida uptake by swine alveolar macrophages was are at risk of developing severe clinical disease, because
very low, even in the presence of opsonins. Similar re- some pigs in the population will become infected very
sults were found by Fuentes and Pijoan (1986). More re- late, at a time when no maternal immunity is available.
cent work, however, suggests that little capsule is ex- This could explain why SEW systems sometimes present
pressed when the organisms are grown under with delayed PRDC (the “18-week wall”) (Dee 1997).
iron-restricted conditions (Jacques et al. 1994). These
growth conditions mimic the scenario found in vivo. EPIDEMIOLOGY
Thus, the relevance of the capsule to virulence may have
been overestimated in the past. The epidemiology of P. multocida is not well understood.
Some strains of P. multocida are able to produce pleu- The organism is present in practically all herds and can
ritis and abscessation in experimentally infected pigs (Pi- be readily isolated from the nose and tonsils of normal,
joan and Fuentes 1987). The virulence factors that dis- healthy individuals. Transmission of the disease by
tinguish these strains from less virulent pneumonic aerosols has been postulated but is unlikely to be of im-
strains are not defined. However, Iwamatso and Sawada portance. Baekbo and Nielsen (1988) measured airborne
(1988) found that strains of serotype D or toxigenic P. multocida in herds suffering from atrophic rhinitis.
strains (of both serotypes) were associated with abscess- They were able to isolate the organism in 29 of 44 herds
es but not with pleuritis. studied. However, the low number of organisms isolated
(144 CFU/mL) led them to conclude that there was no re-
Mucosal Colonization lationship between the number of organisms recovered
The colonization of mucous surfaces by P. multocida has and the severity of the clinical problem.
received some attention lately, as it is of paramount im- Although aerosol transmission may occasionally oc-
portance in understanding the pathogenesis of this or- cur within the herd, it is probable that nose-to-nose con-
ganism. Jacques (1987) found that both serotypes A and tact is the common route of infection. Both vertical and
D adhered poorly to isolated tracheal epithelial cells, al- horizontal transmission occur, although within farms
though serotype A strains were more adherent. He later most transmission appears to be horizontal, with one
showed that serotype A strains adhered mostly to ciliat- strain predominating in pneumonic lungs (Zhao et al.
ed epithelial cells. Pijoan and Trigo (1989) also found 1993). This suggests the existence of P. multocida strains
sparse colonization by serotype A and D strains but of variable virulence, with one of the more virulent
found that serotype D strains adhered mostly to noncili- strains producing most of the disease within a popula-
ated cells. Trigo and Pijoan (1988) and later Issacson and tion. External sources of the organism include mice and
CHAPTER 37 PNEUMONIC PASTEURELLOSIS Pijoan 513

other rodents, although chickens and chicken manure Subacute Form


have also been postulated as sources. These are probably This is associated with P. multocida strains that produce
not common sources of the organism in modern swine pleuritis. In these cases, cough and abdominal breathing
farms, however. can be detected in grower or finishing pigs up to market
weight. Cough in this age pig is usually the hallmark of
PATHOGENESIS severe disease. Clinically, this form of the disease is very
similar to pleuropneumonia due to A. pleuropneumoniae
Experimental infections with P. multocida are very diffi- (see Chapter 26). The main distinguishing feature is that
cult to produce. Healthy pigs will readily tolerate large pleuritic pasteurellosis rarely results in sudden death.
doses of organisms instilled intranasally or even intra- Rather, pigs become extremely emaciated but may sur-
tracheally. Pulmonary clearance is very effective, and the vive for a long time. Recently, outbreaks of PRDC in fin-
bacteria cannot be reisolated 30 minutes after challenge. ishing pigs (about 16–18 weeks of age) on farms using
Recent work demonstrating that little capsule is present SEW methods has resulted in cough and abdominal
in vivo has clarified the apparent disparity between the breathing in pigs but usually not pleuritis (Dee 1997).
poor phagocytosis observed in vitro and the ready clear-
ance of bacteria found in healthy animals. Experimental Chronic Form
models of the disease have used serotype B organisms This is the most common form of the disease, character-
(Farrington 1986), previous infections with immunosup- ized by occasional cough, thumping, and low or nonexis-
pressive virus or mycoplasmas (Fuentes and Pijoan 1986; tent fever. Animals affected are usually in the later stages
Ciprian et al. 1988), or massive instillation of infected of the nursery or are growers (10–16 weeks of age). The
fluids into the lung (Hall et al. 1988). This has led to the signs are indistinguishable from those following M. hy-
conclusion that P. multocida is not a primary agent of opneumoniae infections, for P. multocida causes the con-
pneumonia but rather follows infections with other tinuation and exacerbation of primary mycoplasmosis.
agents. Vaccination against hog cholera virus (Pijoan and
Ochoa 1978) and infection with Aujezsky’s virus LESIONS
(Fuentes and Pijoan 1987) or M. hyopneumoniae (Ciprian
et al. 1988) have all been shown to predispose the pig to Lesions of P. multocida are confined to the thoracic cavi-
superinfections with P. multocida. In contrast, porcine re- ty and are superimposed on those of M. hyopneumoniae.
productive and respiratory syndrome (PRRS) virus could Typically, anteroventral consolidation of the lung is
not be shown to interact with P. multocida in the produc- seen, together with froth in the trachea. There is a clear
tion of pneumonia (Carvalho et al. 1997). line of demarcation between affected and healthy lung
Once established, the organism stimulates a rapid tissue. The affected portion of the lung will have discol-
suppurative reaction, characterized by neutrophil infil- oration ranging from red to grayish-green, depending on
tration. This is probably a host reaction to bacterial the course of the infection (Fig. 37.1).
lipopolysaccharide, which stimulates the release of in- Severe cases may present varying degrees of pleuritis
flammatory cytokines. Death is uncommon, probably and abscessation. Pleural adhesions to the thoracic wall
the result of endotoxic shock and respiratory fail- are common in these cases, and the pleura has a translu-
ure. cent, dry appearance (Figs. 37.2, 37.3). This is useful in
differentiating pneumonic pasteurellosis from acti-
CLINICAL SIGNS nobacillus pleuropneumonia, in which moist, yellowish
pleural adhesions with massive fibrin infiltration are
The clinical signs vary in severity depending on the strain more common (Pijoan 1989).
of P. multocida involved, together with the immune sta- Histologically, a lobular, exudative bronchopneumo-
tus of the animals. nia is found. Severe bronchopneumonia, alveolar epithe-
lial hyperplasia, and the presence of abundant neu-
Acute Form trophils are seen with mucopurulent exudate in the
This form is most commonly associated with serotype B bronchial lumen and in alveolar spaces. These lesions are
strains. It is rare and is never seen in Europe or North not specific to P. multocida infections and are similar for
America. The animals show dyspnea, labored breathing most bacterial pneumonias.
with abdominal “thumps” (sudden contractions of the Evidence has also been presented for a relationship
abdomen), prostration, and high fever (up to 42.2˚C, between pasteurella-induced bronchopneumonia and
108˚F). Mortality may be high (5–40%) in these cases; the presence of disseminated focal nephritis (Butten-
dead and moribund animals may show purplish discol- schon 1991). The author concluded that the two diseases
oration of the abdominal region, suggesting endotoxic are connected by a process of dissemination from the
shock. pulmonary lesions.
514 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

37.1. Pneumonic pasteurellosis. Lung consolidation is anteroventral with a clear demarcation


line between affected and healthy tissue.
CHAPTER 37 PNEUMONIC PASTEURELLOSIS Pijoan 515

37.2. Pleural adhesions of lung to the thoracic wall in a case of pleuritic pasteurellosis.
Note that the pleura has a translucent, dry aspect.

DIAGNOSIS in brain-heart infusion (BHI) broth, grown overnight,


and then plated (Pijoan et al. 1983b). Alternatively, se-
Since the lesions of P. multocida infection are not pathog- lective media can be used: Ackermann et al. (1994) suc-
nomonic, they cannot be used as the only criteria to es- cessfully isolated P. multocida from tonsils and turbinates
tablish a definite diagnosis. The history of the outbreak, of adult pigs using blood agar with 3.75 U/mL of baci-
histopathology, and isolation of the organism should be tracin, 5 µg/mL clindamycin, 0.75 µg/mL gentamicin,
used to confirm the original presumptive diagnosis. and 2.25 µg/mL of amphotericin B. Isolation can also be
Serology has not proven effective for diagnosis, and no enhanced by injecting the specimen intraperitoneally in-
serologic test is routinely available for P. multocida infec- to mice and then recovering the pasteurellae 24 hours
tions. later from liver and ascitic fluid.
P. multocida is a relatively easy organism to culture, Differential diagnosis must include influenza virus,
provided proper specimens are submitted to the labora- A. pleuropneumoniae, B. bronchiseptica, Salmonella choler-
tory. Specimens yielding the best isolations include aesuis, and pure M. hyopneumoniae infections. Accurate
swabs of tracheobronchial exudate and affected lung tis- clinical differentiation based on the epidemiology and le-
sue obtained from the border area between affected and sions can be readily achieved for some of these condi-
normal tissue. Nasal swabs have also been shown to be tions but may be difficult in cases of influenza, B. bron-
good samples for isolation of P. multocida (Schoss chiseptica, or M. hyopneumoniae. In these cases, histology
and Alt 1995). Swabs should be immersed in an ap- and bacterial culture will be needed. Ramirez and Pijoan
propriate transport medium, such as Stuart’s. Lung sam- (1982) and Straw (1986a) have published tables for the
ples should be obtained as aseptically as possible. All differential diagnosis of these conditions.
samples should be refrigerated (but not frozen) until cul-
tured. TREATMENT
Culture of P. multocida can be successfully achieved in
laboratories with minimal facilities. Good-quality speci- Treatment of P. multocida field infections with antibi-
mens will yield the organism on direct culture onto otics is usually difficult or unsuccessful. This is partly due
blood agar or glucose agar plates. If the samples are to widespread antibiotic resistance in P. multocida iso-
more contaminated, they can be serially diluted tenfold lates in the United States and also to difficulties in achiev-
516 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

37.3. Lung from a case of


pleuritic pasteurellosis. Note
anteroventral, well-demarcated
lesions, together with the presence
of multiple abscesses. There is
extensive interlobar adhesion.

ing adequate antibiotic concentrations in consolidated, Cote et al. (1991) found some plasmid-mediated re-
pneumonic lung. sistance to streptomycin and sulfonamides among 29
A variety of antibiotics and antibiotic combinations field isolates investigated. Gutierrez Martin and Ro-
have been commonly used (Farrington 1986). These in- driguez Ferri (1993) studied 59 Spanish isolates, finding
clude parenteral antibiotics such as oxytetracycline, 11 good activity with penicillins, aminoglycosides, tetracy-
mg/kg; long-acting oxytetracycline, 20 mg/kg; procaine clines, erythromycin, colistin, and rifampicin, with some
penicillin, 66,000 units/kg; benzathine penicillin, 32,000 resistance to tylosin, vancomycin, and tiamulin. They re-
units/kg; tiamulin, 10–12.5 mg/kg; and ampicillin, 6.6 ported that third-generation cephalosporins and fluori-
mg/kg. Many of these drugs, however, are becoming in- nated quinolones were the most effective drugs. Raem-
creasingly less efficient due to widespread antibiotic re- donck et al. (1994) found good minimum inhibitory
sistance. Treatment via feed antibiotics has also been concentrations (MIC) for danofloxacin, ceftiofur, and
suggested, although, as in the case of other pneumonias, trimethoprim-sulfonamide; they found variable to high
it is probably not very effective. resistance to erythromycin, gentamicin, lincomycin,
CHAPTER 37 PNEUMONIC PASTEURELLOSIS Pijoan 517

oxytetracyclines, and spectinomycin. Finally, Salmon et ties of spreading the organism.


al. (1995) tested isolates from several countries and Environmental changes such as increasing ventila-
found the best activity with cephalosporins and en- tion flow rate, decreasing ammonia, and minimizing
rofloxacin, with poor activity shown in vitro by ery- temperature fluctuations and dust are usually recom-
thromycin, sulfamethazine, spectinomycin, and lin- mended. Some of these recommendations are antago-
comycin. nistic; increasing airflow, especially in winter, results in a
The effectiveness of all these antimicrobials will vary decrease of both temperature and humidity, with an in-
considerably depending on strain susceptibility. Since P. crease in dust. Most of these changes have not proven
multocida readily exhibits resistance to various antimi- valuable in controlled, experimental conditions. Noyes
crobials, antibiograms should be performed before insti- et al. (1986) found that decreasing ventilation below
tuting treatment. minimal recommended levels (0.5 CFM/pig) had no ef-
As in other respiratory infections, antibiotics are fect on weaned pigs inoculated with both B. bronchisepti-
more effective when used as prophylactic, rather than ca and P. multocida. Similarly, Rafai et al. (1987) found
therapeutic, agents. Tetracyclines alone, tetracyclines that cold stress, even though it reduced immune function
combined with sulfamethazine or sulfathiazole or peni- in suckling pigs, had no effect on the course of an exper-
cillin, and tylosin combined with sulfamethazine have imental P. multocida infection. Environmental changes
been recommended for this purpose. It is probable that frequently entail extensive remodeling. They are, there-
the most effective compound in these mixtures is sul- fore, expensive to institute and maintain. It is not clear
famethazine. This antimicrobial has been the focus of that these changes are cost-effective in terms of reducing
controversy over residues. Because of this, its use has respiratory disease.
been severely limited and monitored. Tiamulin (40 ppm On the other hand, considerable improvement may
in feed) has been shown, in a number of trials, to im- be obtained by instituting management changes that re-
prove average daily gain. However, since pneumonic le- duce the spread of the organism. These include the fol-
sions are not significantly reduced by this antibiotic (Pott lowing:
and Edwards 1990), the mode of action by which these
improvements are obtained is unclear. Tiamulin has 1. Segregated early weaning: SEW has a major impact
been found to be of variable effectiveness against P. mul- on pneumonia and, when properly done, will reduce or
tocida. It is therefore probable that its main effect in eliminate pneumonia from most groups of pigs. This is
pneumonia lies in the control of M. hyopneumoniae. probably based on the control of M. hyopneumoniae,
Some new antibiotics with claims for P. multocida which in many herds is virtually eliminated. Early wean-
treatment are not available in the United States but are ing probably has less effect on P. multocida, since it has
used extensively in other countries. These include in- been shown that pigs are colonized as early as 10 days of
jectable lincomycin-spectinomycin, some cephalo- age. Occasional farms or groups of pigs will experience
sporins, and various quinolones including enrofloxacin acute outbreaks of pneumonia.
and danofloxacin. Injectable ceftiofur is available in the 2. All-in/all-out production: In farms where SEW is
United States, however, and it has been shown by a num- not possible, efforts should be made to institute an all-
ber of authors to have good activity against P. multocida. in/all-out production program. Several workers have
shown that this type of production system markedly de-
PREVENTION creases the incidence of pneumonia.
3. Closed herds: Minimizing the purchase of outside
Management Approaches pigs, especially fatteners, will result in a decrease of
Since antibiotic therapy is often unsuccessful and even pneumonia and other respiratory conditions. However,
when successful may not prove cost-effective, prevention the pressure to improve genetics in modern farms has
of pneumonia has received much attention. Prevention forced most producers to purchase their breeding ani-
is usually obtained through changes in management. mals. It has become increasingly important to evaluate
The management techniques that result in decreased the health status of the farm from which these animals
pneumonia have been reviewed by several authors (Pi- are purchased, in order to minimize the probability of
joan 1986; Straw 1986b). Caution must be taken when introducing disease.
implementing these recommendations, since they derive 4. Minimal mixing and sorting: This is a source of
mostly from retrospective epidemiological studies and stress to the pigs, while also intensifying the probabilities
not from experimental data. Also, they are intended to of disease transmission. Pigs should be mixed as few
reduce pneumonia as a whole (and other respiratory times as possible during their productive life.
problems) and do not differentiate between conditions 5. Reduction in building and pen size: Smaller rooms
of different etiology. and smaller pens both have been shown to reduce levels
Management changes can be directed either at modi- of pneumonia. Rooms should have a maximum of 250
fying the pigs’ environment or at reducing the possibili- pigs, and pens a maximum of 20–25 pigs each. This
518 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

again is difficult to achieve in modern farms. However, sions produced (and the serologic reactions) are indistin-
the recent trend of building wean-to-finish barns, where guishable from A. pleuropneumoniae, it is possible that
pigs are only mixed once, at weaning, will probably have some field outbreaks have been misdiagnosed. At
a positive effect in decreasing pneumonia. present, it is very difficult to assess the true prevalence
6. Reduction in animal density: Decreasing animal and economic impact of these strains.
density has been shown by many authors to reduce levels
of pneumonia. It is important therefore to find a reason- REFERENCES
able compromise between densities that are appropriate
for the animals’ health and those that maximize returns Ackermann, M. R.; Debey, M. C.; Register, K. C.; Larson, J.
on the building’s cost. L.; and Kinyon, J. M. 1994. Tonsil and turbinate colo-
nization by toxigenic and nontoxigenic strains of Pas-
Vaccination teurella multocida in conventionally raised swine. J Vet
Although several killed vaccines for the prevention of Diag Invest 6:375–377.
pneumonic pasteurellosis are available, their effective- Adler, B.; Chancellor, R.; Hamchampa, P.; Hunt, M.; Ruffo-
ness is questionable. Since no reliable model of experi- lo, C.; Strugnell, R.; and Wapling, D. 1996. Immunity
mental disease exists, potency testing is usually per- and vaccine development in Pasteurella multocida infec-
formed in mice. It is doubtful, therefore, that this model tions. J Biotechnol 44(1–3):139–144.
truly replicates the disease in the pig. Under field condi- Baekbo, P. 1988. Pathogenic properties of Pasteurella multo-
tions, clinicians frequently have disappointing results cida in the lungs of pigs. Proc Int Congr Pig Vet Soc
with these vaccines, and they do not appear to be cost-ef- 10:58.
fective. Baekbo, P., and Nielsen, J. P. 1988. Airborne Pasteurella mul-
Some modern approaches to vaccination against P. tocida in pig fattening units. Proc Int Congr Pig Vet Soc
multocida have been reported recently. The use of puri- 10:51.
fied lipopolysaccharide (LPS) antigen and the use of aro Bahnson, B. B. 1994. Enzootic Pneumonia of Swine: Epi-
A gene deletion mutants both resulted in protection demiology and Effect on the Rate of Gain. Ph.D. diss.
against homologous, but not heterologous, challenge Univ Minnesota.
(Adler et al. 1996). The use of a cloned, 87 kDa outer- Bisgaard, M. 1984. Comparative investigations of Pasteurel-
membrane protein had similar limitations (Ruttolo and la haemolytica sensu stricto and so-called P. haemolytica
Adler 1996). Cross-protection has been reported in isolated from different pathological lesions in pigs. Acta
chickens with the use of a protein preparation from cells Pathol Microbiol Immunol Scand (B) 92:203–207.
grown under iron-deprived conditions, which mimic the Buttenschon, J. 1991. Statistical evidence for a link between
in vivo environment (Wang and Glisson 1994). However, pleuropneumonia and disseminated focal nephritis in
none of these fraction vaccines are commercially avail- pigs. Zentralblatt für Vet Reihe A 38(4):287–299.
able or have been tested in pigs, where the disease is not Carvalho, L.; Segales, J.; and Pijoan, C. 1997. Effect of PRRS
septicemic as it is in chickens and mice. Therefore, there virus on subsequent Pasteurella multocida challenge in
is at present no successful commercial vaccine against P. pigs. Vet Microbiol (in press).
multocida in the pig. Ciprian, A.; Pijoan, C.; Cruz, T.; Camacho, J.; Tortora, J.; Col-
menares, G.; Lopez-Revilla, R.; and Garza, M. De la.
OTHER PASTEURELLA INFECTIONS 1988. Mycoplasma hyopneumoniae increases the suscepti-
IN PIGS bility of pigs to experimental Pasteurella multocida pneu-
monia. Can J Vet Res 52:434–438.
Several authors have sporadically isolated P. haemolyt- Cote, S.; Harel, J.; Higgins, R.; and Jacques, M. 1991. Resis-
ica–like organisms from cases of severe necrotizing pleu- tance to antimicrobial agents and prevalence of R plas-
ropneumonia. The lesions seen are similar to those mids in Pasteurella multocida from swine. Am J Vet Res
found in A. pleuropneumoniae infections but tend to be 52(10):1653–1657.
less severe. In this regard, they are also similar to lesions Dee, S. A. 1997. Porcine respiratory disease complex: The
reported from A. suis. A new nicotinamide adenine dinu- “18 week wall.” In Proc Am Assoc Swine Pract (Quebec),
cleotide (NAD)-independent biotype of A. pleuropneu- pp. 465–466.
moniae has now been recognized, and it is very probable Dugal, F.; Belanger, M.; and Jacques, M. 1992. Enhanced ad-
that many of these severe pneumonias are related to herence of Pasteurella multocida to porcine tracheal rings
these two organisms rather than to P. hemolytica. preinfected with Bordetella bronchiseptica. Can J Vet Res
All these organisms are very similar and differ in mi- 56(3):260–264.
nor biochemical reactions such as indole formation or V- Farrington, D. O. 1986. Pneumonic pasteurellosis. In Dis-
factor requirement. Bisgaard (1984) did a comprehen- eases of Swine, 6th ed. Ed. A. D. Leman, B. Straw, R. D.
sive study of these strains and concluded that they Glock, W. L. Mengeling, R. H. C. Penny, and E. Scholl.
differed from P. haemolytica sensu stricto. Since the le- Ames: Iowa State Univ Press, p. 436.
CHAPTER 37 PNEUMONIC PASTEURELLOSIS Pijoan 519

Fuentes, M., and Pijoan, C. 1986. Phagocytosis and killing of State Univ Press, pp. 152–162.
Pasteurella multocida by pig alveolar macrophages after ———. 1989. Pleuritis effect on growth underestimated.
infection with pseudorabies virus. Vet Immunol Im- Int Pig Lett 9:17–19.
munopathol 13:165–172. ———. 1995. Diseases of high-health pigs: Some ideas on
———. 1987. Pneumonia in pigs induced by intranasal pathogenesis. In Proc A Leman Conf (Minnesota), pp.
challenge exposure with pseudorabies virus and Pas- 16–17.
teurella multocida. Am J Vet Res 48:1446–1448. Pijoan, C., and Fuentes, M. 1987. Severe pleuritis associated
Gutierrez Martin, C. B., and Rodriguez Ferri, E. F. 1993. In with certain strains of Pasteurella multocida in swine. J
vitro susceptibility of Pasteurella multocida strains isolat- Am Vet Med Assoc 191:823–826.
ed from swine to 42 antimicrobial agents. Int J Med Mi- Pijoan, C., and Ochoa, G. 1978. Interaction between a hog
crobiol Virol Parasitol Dis 279(3):387–393. cholera vaccine strain and Pasteurella multocida in pro-
Hall, W.; Bane, D.; Kilroy, C.; and Essex-Sorlie, D. 1988. A duction of porcine pneumonia. J Comp Pathol
model for the induction of pneumonia caused by Pas- 88:167–170.
teurella multocida type A. Proc Int Congr Pig Vet Soc Pijoan, C., and Trigo, E. 1989. Bacterial adhesion to mucos-
10:59. al surfaces with special reference to Pasteurella multocida
Hoie, S.; Falk, K.; and Lium, B. M. 1991. An abbatoir survey isolates from atrophic rhinitis. Can J Vet Sci 54:516–521.
of pneumonia and pleuritis in slaughter weight swine Pijoan, C.; Morrison, R. B.; and Hilley, H. D. 1983a. Serotyp-
from 9 selected herds. IV. Bacteriological findings in ing of Pasteurella multocida isolated from swine lungs
chronic pneumonic lesions. Acta Vet Scand collected at slaughter. J Clin Microbiol 17:1074–
32(3):395–402. 1076.
Issacson, R. E., and Trigo, E. 1995. Pili of Pasteurella multo- ———. 1983b. Dilution technique for isolation of
cida of porcine origin. FEMS Microbiol Letters Haemophilus from swine lungs collected at slaughter. J
132(3):247–251. Clin Microbiol 18:143–145.
Iwamatsu, S., and Sawada, T. 1988. Relationship between Pijoan, C.; Lastra, A.; Ramirez, C.; and Leman, A. 1984. Iso-
serotypes, dermonecrotic toxin production of Pasteurel- lation of toxigenic strains of Pasteurella multocida from
la multocida isolates and pneumonic lesions of porcine lungs of pneumonic swine. J Am Vet Med Assoc
lungs. Jpn J Vet Sci 50:1200–1206. 185:522–523.
Jacques, M. 1987. Adherence of Pasteurella multocida to Pott, J., and Edwards, J. 1990. Beneficial effects of tiamutin
porcine upper respiratory tract cells. Current Microbiol administered in feed at 40 ppm to pigs with enzootic
15:115–119. pneumonia. Proc Int Congr Pig Vet Soc 11:86.
Jacques, M.; Kobisch, M.; Belanger, M.; and Dugal, F. 1993. Raemdonck, D. L.; Tanner, A. C.; Tolling, S. T.; and Michen-
Virulence of capsulated and noncapsulated Pasteurella er, S. L. 1994. Antimicrobial susceptibility of Acti-
multocida and their adherence to porcine respiratory nobacillus pleuropneumoniae, Pasteurella multocida and
tract cells and mucus. Infect Immun 61(11):4785–4792. Salmonella choleraesuis isolates from pigs. Vet Rec
Jacques, M.; Belanger, M.; Diarra, M. S.; Dargis, M.; and 134:5–7.
Malouin, F. 1994. Modulation of Pasteurella multocida Rafai, P.; Neumann, R.; Leonhardt, W.; Frenyo, L.; Rudas, P.;
capsular polysaccharide during growth under iron-re- Fodor, L.; and Boros, G. 1987. Effect of environmental
stricted conditions and in vivo. Microbiol 140 (Pt temperature on pigs infected with Pasteurella multocida
2):263–270. type A. Acta Vet Hung 35:211–223.
Kielstein, P. 1986. On the occurrence of toxin-producing Ramirez, N., and Pijoan, C. 1982. Diagnostico de las Enfer-
Pasteurella multocida strains in atrophic rhinitis and in medades del Cerdo. Ed. R. R. Necoechea and C. Pijoan.
pneumonias of swine and cattle. J Vet Med (B) Mexico City: Necoechea and Pijoan, p. 227.
33:418–424. Rubies, X.; Casal, J.; Fernandez, J.; and Pijoan, C. 1996.
Maheswaran, S., and Thies, E. 1979. Influence of encapsula- Transmission of Pasteurella multocida clones in a
tion on phagocytosis of Pasteurella multocida by bovine swine pyramid structure. Proc Int Congr Pig Vet Soc
neutrophils. Infect Immun 26:76–81. 14:243.
Noyes, E.; Pijoan, C.; and Jacobson, L. 1986. Ventilating en- Ruttolo, C. G., and Adler, B. 1996. Cloning, sequencing, ex-
vironment for the weaned pig. Proc Int Congr Pig Vet pression and protective capacity of the oma 87 gene en-
Soc 9:401. coding the Pasteurella multocida 87-kilodalton outer
Noyes, E.; Feeney, D.; and Pijoan, C. 1990. Comparison of membrane antigen. Infect Immun 64(8):316–317.
the effect of pneumonia detected during a lifetime with Salmon, S. A.; Watts, J. L.; Case, C. A.; Hoffman, L. J.; We-
pneumonia detected at slaughter on growth in swine. J gener, H. C.; and Yancey, R. J., Jr. 1995. Comparison of
Am Vet Med Assoc 197:1025–1029. MICs of Ceftiofur and other antimicrobial agents
Pijoan, C. 1986. Respiratory system. In Diseases of Swine, against bacterial pathogens of swine from the United
6th ed. Ed A. D. Leman, B. Straw, R. D. Glock, W. L. States, Canada and Denmark. J Clin Microbiol
Mengeling, R. H. C. Penny, and E. Scholl. Ames: Iowa 33(9):2435–2444.
520 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Schoss, P., and Alt, M. 1995. Are nasal swabs appropriate multocida strains associated with atrophic rhinitis. Vet
for the diagnosis of bacterial pneumonia agents? Dtsch Rec 122:19.
Tierarztl Wochen 102(11):427–430. Verma, N. D. 1988. Pasteurella multocida B:2 in haemorrhag-
Straw, B. E. 1986a. Differential diagnosis of swine diseases. ic septicaemia outbreak in pigs in India. Vet Rec 123:63.
In Diseases of Swine, 6th ed. Ed. A. D. Leman, B. Straw, Wang, C., and Glisson, J. R. 1994. Passive cross-protection
R. D. Glock, W. L. Mengeling, R. H. C. Penny, and E. provided by antisera directed against in vivo expressed
Scholl. Ames: Iowa State Univ Press, p. 214. antigens of Pasteurella multocida. Avian Dis
———. 1986b. A look at factors that contribute to the de- 38(3):506–514.
velopment of swine pneumonia. Vet Med Zhao, G.; Pijoan, C.; and Murtaugh, M. P. 1993. Epidemiol-
(Aug):747–755. ogy of Pasteurella multocida in a farrow-to-finish swine
Trigo, E., and Pijoan, C. 1988. Presence of pili in Pasteurella herd. Can J Vet Res 57(2):136–138.
Porcine Proliferative
38 Enteropathies
S. McOrist and C. J. Gebhart

Proliferative enteropathies (PE; also known as prolifera- ly Desulfovibrionaceae (McOrist et al. 1995a).
tive ileitis) are a group of acute and chronic conditions of The disease is worldwide in distribution and has been
widely differing clinical signs but with a common under- recently described in Australia, Belgium, Brazil, Canada,
lying pathological change visible at necropsy: a thicken- Denmark, France, Greece, Holland, Ireland, Japan, Mex-
ing of the mucosa of the small intestine and colon. His- ico, New Zealand, Poland, South Africa, Sweden, Tai-
tologically, the affected tissues show marked wan, Thailand, the United Kingdom, and the United
proliferation of immature epithelial cells of the intesti- States (Rowland and Lawson 1992). While there are
nal crypts, forming a hyperplastic to adenoma-like mu- some local differences, surveys conducted in several of
cosa. These proliferating cells invariably contain numer- these countries have estimated that approximately 30%
ous intracytoplasmic Lawsonia intracellularis, an obligate of pig herds are likely to be affected. It is possible that
intracellular bacterium. there has been an increase in the incidence of acute PE in
In growing pigs with uncomplicated proliferation of modern multisite production systems in the United
the mucosa, the condition is chronic proliferative en- States in the past 5 years (Winkelman 1996a).
teropathy, also known as porcine intestinal adenomato- Pathological changes closely resembling porcine PE
sis (PIA) or ileitis. In some pigs, additional changes may have been described in a number of other mammalian
be superimposed on this basic lesion, including a necrot- species, including some rodents, such as the hamster
ic enteritis, a granulomatous regional ileitis, or an acute (Frisk and Wagner 1977) and rat, (Vandenberghe et al.
hemorrhagic proliferative enteropathy (Rowland and 1985), but not the mouse. PE has been described occa-
Lawson 1975). Both the chronic and acute hemorrhagic sionally in carnivores, such as the fox (Eriksen and
forms of PE are now important enteric diseases in the Landsverk 1985) and ferret (Fox and Lawson 1988), in
modern pig industry. herbivores, such as the horse (Duhamel and Wheeldon
The lesions of PE were first described in pigs in 1982; Williams et al. 1996), rabbit (Schoeb and Fox
Ames, Iowa, by Biester and others in the 1930s (Biester 1990), and deer (Drolet et al. 1996), and also in ratite
and Schwarte 1931; Biester et al. 1939) and were subse- birds, the emu and ostrich (Cooper 1996), but not in oth-
quently found to occur in other major pig-raising areas er birds. In all these species, intracellular bacteria resem-
throughout the world, but it was not until 1973 that bling Lawsonia species have been observed within the cy-
Rowland and others investigating major outbreaks oc- toplasm of epithelial cells of proliferative portions of
curring in the United Kingdom developed a productive intestinal mucosa. Experimental transmission studies, in
research program (Rowland and Rowntree 1972; Row- situ immunostaining, and DNA analysis have suggested
land and Lawson 1974). They found that whenever these that one agent may be able to infect the intestinal cells of
proliferative changes in pigs were studied either ultra- a wide variety of host species. However, there is current-
structurally or using silver stains, intracellular bacteria ly no evidence to link these other hosts with the onset of
were consistently present within the abnormal prolifer- disease in pigs.
ating cells (Rowland and Lawson 1974). These bacteria
are curved to straight rods with either tapered or round- ETIOLOGY
ed ends and measure 1.25–1.75 µm in length by
0.25–0.43 µm in width. The bacteria lie free in the apical The primary agent that causes PE is the obligately intra-
cytoplasm of infected epithelial cells and are not mem- cellular bacterium L. intracellularis, which preferentially
brane-bound during the important stages of infection. grows within intestinal epithelial cells. It has not as yet
The identity of these bacteria and their etiologic role in been cultivated in cell-free media. Its morphology and
PE were finally resolved in 1993 with successful culture of entry processes into host epithelial cells resemble some
the intracellular organism and the reproduction of the other obligately intracellular bacteria, particularly Rick-
disease in pigs using a pure culture of this agent (Lawson ettsia species (McOrist et al. 1995b); however, these two
et al. 1993; McOrist et al. 1993). Also in 1993, its taxo- genera are not related taxonomically. The reliance on
nomic position was clarified (Gebhart et al. 1993); its de- host cells for growth, probably for preformed triphos-
finitive name is Lawsonia intracellularis, within the fami- phates or another energy source, is readily apparent

521
522 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

when the bacteria are observed in the natural lesions. as a result of artificial exposure to British or American
Unfortunately, between 1973, when the bacteria were isolates have had all the characteristics of the field dis-
first observed, and 1993, when they were first cultured ease, including the presence of mucosal proliferation
and identified, a variety of curved gram-negative bacte- and intracellular bacteria (McOrist et al. 1993; Knittel et
ria and other putative agents were cultured from the in- al. 1996).
testines of affected pigs, leading to considerable confu- Early attempts to identify the intracellular agent in
sion at the time. the tissues by immunological techniques produced con-
A variety of Campylobacter species, particularly C. flicting results (Rowland and Lawson 1974; Chang et al.
mucosalis (Lawson and Rowland 1974), C. hyointestinalis 1984), probably due to the presence of a “natural” rabbit
(Gebhart et al. 1983), C. hyoilei, a C. jejuni variant, and antibody reactive with the intracellular organism (Law-
C. coli, can be recovered from proliferative lesions. How- son et al. 1985). Preparation of specific hyperimmune
ever, neither specific proliferative lesions nor intracellu- sera and monoclonal antibodies eliminated this source
lar colonization have occurred when pigs are inoculated of confusion. These react with L. intracellularis and do
with any of these bacteria (Kashiwazaki et al. 1971; Mc- not react with cells of any of the cultivated Campylobac-
Cartney et al. 1984; Boosinger et al. 1985; Alderton et al. ter species or other bacteria (Lawson et al. 1985; McOrist
1992). This indicates that these are secondary agents tak- et al. 1987). Specific reactivity of monoclonal and poly-
ing advantage of the altered conditions in the gut for col- clonal antibody raised to the L. intracellularis organism
onization. Similar colonization by other secondary bac- mainly lies in a 25–27 kDa fragment present on its outer
teria is a feature of other enteric diseases, such as swine surface (McOrist et al. 1989a).
dysentery (Robinson et al. 1984). Much of the published
literature on PE does refer to the intracellular bacterium EPIDEMIOLOGY
being a Campylobacter-like organism; however, the desig-
nation was only based on its morphologic similarity to Documented information on the occurrence and epi-
that genus. Other agents cultured from affected in- demiological features of the PE disease complex in pig
testines are enteroviruses and intestinal Chlamydia herds of the world remains basic. The main reason for
species (Joens et al. 1987; Fox et al. 1993). However, these this deficiency is that in many instances clinical signs are
are probably present in many unaffected pigs, and inoc- not dramatic and that, until recently, specific tools for
ulation studies with these agents were unrewarding. the diagnosis of the disease in the live animal have not
Exposure of pigs to crude, or partially filtered, ho- been available. Abattoir or on-farm mortality monitor-
mogenized diseased mucosa resulted in reproduction of ing has been used to assess the presence and possible im-
specific intestinal lesions and clinical disease in some pact of the disease. The extent to which the incidence at
early challenge trials (Roberts et al. 1977; Mapother et al. slaughter may reflect the occurrence of disease on the
1987; McOrist and Lawson 1989a), first indicating the farm is largely unknown and may be a considerable un-
etiologic role of the intracellular bacteria. derestimate. As with other postweaning enteric diseases,
Experimental transmission studies using pure cul- moderate lesions may resolve by the time of slaughter.
tures of British or American strains of L. intracellularis as Various authors have noted lesions in pigs at normal
oral-challenge inocula for conventional pigs and using slaughter, but in some reports the incidence of animals
gnotobiotic pigs predosed with a minimal bacterial flora with lesions has been low, 0.7–2.0% (Emsbo 1951; Row-
of nonpathogenic enteric species have resulted in repro- land and Hutchings 1978; Kubo et al. 1984; Christensen
duction of the specific lesions of PE. Three weeks follow- and Cullinane 1990). Other reports have indicated much
ing oral inoculation of postweaned pigs with 108 L. intra- higher levels of occurrence of lesions. Studies by Pointon
cellularis bacteria, numerous L. intracellularis bacteria in Australia and in the United States found that around
were recovered from the affected proliferative intestines 30% of herds had pigs with lesions at slaughter, and the
of challenged pigs, with numerous intracytoplasmic L. incidence of lesions in particular herds reached 40%
intracellularis bacteria visible in sections of these in- (Pointon 1989). These differences may be partly due to
testines (McOrist et al. 1993, 1994). No such recovery or differences in the age and origin of animals at slaughter,
lesions occurred in control pigs inoculated with unin- making routine slaughter checks a somewhat unreliable
fected cell cultures or media. Several isolates derived guide to prevalence.
from natural lesions of acute or chronic forms of PE have Surveys of the cause of mortality on pigs farms and
proved pathogenic in these and several subsequent chal- estimates of clinical occurrence of acute hemorrhagic PE
lenge studies, indicating the ability of any single L. intra- suggest that an average of 1.0–5.0% of older growing
cellularis strain to be involved in both types of lesions. L. pigs are affected (Lawson et al. 1979; Duran 1994; Chris-
intracellularis isolates from American and Australian pigs tensen et al. 1995). Clinically important outbreaks of
have been identical to British isolates with respect to cul- this condition are now reasonably frequent, however,
tural and morphological characteristics (Knittel et al. and may affect a high proportion of animals in a herd.
1996; Collins et al. 1996). Intestinal lesions that develop Reported attack rates of 12–50% of susceptible pigs on
CHAPTER 38 PORCINE PROLIFERATIVE ENTEROPATHIES McOrist, Gebhart 523

affected farms are still representative (Love et al. 1977; ed pigs provide the likely source of new infections. Rig-
Lomax and Glock 1982; Holyoake and Cutler 1995). Par- orous removal of feces between batches of pigs in build-
ticular management situations are thought to lend them- ings capable of complete “all-in/all-out” was demon-
selves to outbreaks of acute hemorrhagic PE. Young strated to be more effective at control of PE than reliance
adults (4–12 months old) within boar and gilt perfor- on slatted floors and sunken pits for feces removal (Bane
mance testing stations, gilts within breeding programs et al. 1997; Smith 1997). It is possible that feces from in-
that involve transport to new units, and the movement fected gilts are responsible for transmission of the infec-
and mixing of boars and gilts into breeding groups are tion to their progeny of preweaned piglets. However, the
commonly associated with PE outbreaks. While this may replacement of stock with those derived from the early
partly reflect changes in use of antibiotics at these times, weaning of piglets, at 10–14 days old, has not proved an
the occurrence of major stressors is a consistent feature effective method of eradicating PE from herds (Winkel-
of herd history prior to outbreaks. White breeds, partic- man 1996a). The relevance of the occurrence of diseases
ularly the Landrace and Large White breeds, and syn- similar in etiology and pathology to PE in several species
thetic commercial breeds incorporating the white breeds other than pigs is not yet clear. Modern pig farms are
are thought to exhibit an increased breed susceptibility; generally capable of excluding other mammals, except
however, the disease has been regularly diagnosed in mice, and these are not known to suffer from naturally
Duroc and other colored pig breeds. Outbreaks have also occurring PE. Experimental inoculations of rodents oth-
been noticed in association with extreme weather condi- er than hamsters (i.e., rats and mice) with pig-derived
tions, particularly periods of extreme differentials be- isolates of L. intracellularis has not resulted in persistent
tween night and day temperatures, or hot, humid weath- infection (McOrist and Gebhart, unpublished data—
er. Lawsonia sp. may have a relatively long survival time 1996). The close adaptation of L. intracellularis to life
in the environment for an obligate intracellular bacteri- within pig epithelial cells suggests that the introduction
um. Some strains have remained viable at 5˚C for 1–2 of infection generally occurs with new pigs carrying the
weeks outside the host cells, and only quaternary ammo- infection, particularly in high-health-status herds. Sur-
nium compounds and iodine-based compounds, of 6 veys have indicated that boars and gilts within nucleus
disinfectants tested, showed complete bactericidal activ- herds are potential sources of infection that can lead to
ity (McOrist and Gebhart, unpublished data—1996). outbreaks of acute disease (Smith 1997).
Development of specific polymerase chain reaction Examination of diagnostic records in several Danish
(PCR) and immunologic detection methods has recently high-health herds indicated that introduction of PE oc-
enabled some measurement of the excretion of L. intra- curred following import of British boars of white breeds
cellularis in the feces of pigs. Where clinical disease is (McOrist and Gebhart, unpublished data—1996).
present in a group, the organism is present in the feces of
all affected, and some subclinically affected animals can PATHOGENESIS
be found to be excreting at this time (McOrist and Law-
son 1989b). Fecal excretion of L. intracellularis can per- PE can be reproduced by exposing susceptible pigs to L.
sist for at least 10 weeks, and enumeration of L. intracel- intracellularis or to diseased mucosa containing these in-
lularis in the feces of experimentally affected pigs found tracellular bacteria (Roberts et al. 1977; Mapother et al.
up to 108 organisms per gram, further indicating that the 1987; McOrist and Lawson 1989a; McOrist et al. 1993,
feces of infected pigs may contain ample infective doses 1994). Intracellular bacteria and histological changes of
for other pigs (Smith and McOrist 1997). These methods PE are first evident 8–10 days after exposure, and lesions
have also enabled surveys of the prevalence and risk fac- reach their maximum around 21 days postchallenge, in-
tors for chronic PE in growing pigs. Recent surveys in dicating a relatively long incubation period of 2–3
Spain, Denmark, United States, and the United Kingdom weeks. Experimental infection of postweaned piglets
have indicated that 20–40% of farms are infected (Lanza usually leads to moderate diarrhea in 50% of challenged
and Pozo 1996; Moller et al. 1996; Bane et al. 1997; animals 2–3 weeks after challenge. L. intracellularis
Smith 1997). These surveys have also indicated that a strains associated with one type of PE (acute or chronic)
wide age range of pigs can be infected, with the detection appear capable of initiating the range of pathological se-
of fecal excretion in boars, gilts, pre-, and postweaned quelae. The pigs exposed to diseased mucosa from the
pigs. Preliminary analysis of PE-positive farms by PCR chronic form may develop the necrotic or acute hemor-
and serologic methods has further indicated that the ma- rhagic form of the disease (Mapother et al. 1987). Also,
jority of infections occur in later postweaning areas of pigs challenged with L. intracellularis isolates derived
farms (where pigs are 8–16 weeks old). It is possible that from acute hemorrhagic PE regularly develop lesions of
these finisher pigs could act as a potent source of infec- chronic PE (McOrist et al. 1993) and occasionally
tion to other areas of affected farms, especially if fecal progress to acute hemorrhagic PE. Pigs of a wide age
contamination of boots and equipment occurs. range are susceptible to oral challenge. Pathogenic infec-
In herds with endemic chronic PE, feces from infect- tions have developed in neonatal piglets aged 7 or 14
524 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

days and in weaned pigs aged 4 or 10 weeks (McOrist In many cases of chronic PE in growing pigs, the clinical
and Lawson 1989a; McOrist et al. 1993; Jones et al. signs are slight, and little more is seen than failure to sus-
1993a). tain growth despite normal feed intake. Ileal lesions are a
PE develops as a progressive proliferation of imma- consistent feature of these pigs. In some pigs there may
ture epithelial cells populated by numerous intracellular be a degree of anorexia, characterized by curiosity about
bacteria. In order to persist and multiply within the ep- food but refusal to eat. Thus, affected animals vary from
ithelium, the L. intracellularis organisms must penetrate the clinically unremarkable to those showing marked
into the dividing crypt cells. In vivo and in vitro studies dullness and apathy. Diarrhea, when present, is general-
have elucidated the early events in bacteria-cell interac- ly moderate, with loose stools of normal color; this is
tion (McOrist et al. 1989b, 1995b; Lawson et al. 1995). probably a feature of only one-half of pigs affected with
Bacteria associate with the cell membrane and then chronic PE. Diarrhea may occur when significant large-
quickly enter the enterocyte via an entry vacuole. This intestinal lesions are present. When chronic PE is sus-
rapidly breaks down (within 3 hours) and the bacteria pected in a herd, milder cases can be relatively common
flourish and multiply free (not membrane-bound) within but difficult to detect. Therefore, such farms should be
the cytoplasm. The entry of bacteria into cells is depen- inspected for apparent wasting of well-grown animals
dent on cell, but not necessarily bacterial, viability, that with anorexia and irregular diarrhea, possibly seen as
is, a type of induced phagocytosis (Lawson et al. 1995). “slab-sided” pigs. Records should be carefully examined
The mechanism whereby the bacteria cause infected cells to detect changes in average weight gains and feed con-
to fail to mature, continue to undergo mitosis, and form version efficiency in the postweaned group (Roberts et al.
hyperplastic crypts is not yet understood. Intestinal 1979; Gogolewski et al. 1991). More severely affected cas-
glands can become enormously elongated and often es are often associated with varying degrees of inflam-
branched. Loss of body protein into the feces and the matory or necrotic change in the affected mucosa, and
blocking of nutrient absorption by the thickened intesti- those that develop necrotic enteritis or the “hose pipe”
nal mucosa are the likely causes of the reduction in ileum of regional ileitis show severe loss of condition
weight gain and altered feed conversion in pigs affected and often scour persistently. Death in regional ileitis is
with chronic uncomplicated PE lesions. not uncommon and is usually associated with perfora-
Degenerative and reparative changes may be super- tions of the hypertrophied ileal wall, leading to a gener-
imposed on the basic enterocyte proliferation. Inflam- alized terminal peritonitis.
matory changes range from a superficial fibrinous reac- Unlike chronic PE, cases of acute hemorrhagic PE oc-
tion to extensive, deep, coagulative necrosis, which is the cur more commonly in young adults 4–12 months old
lesion of necrotic enteritis. Early lesions contain very few and present a clinical picture of acute hemorrhagic ane-
infiltrating inflammatory cells, probably not above the mia. Black tarry feces are often the first visible clinical
normal for pig intestines (McOrist et al. 1992). In more sign and these may become loose. However, some ani-
developed lesions a mainly mononuclear leukocyte infil- mals die without fecal abnormality and show only
tration of the lamina propria, particularly CD8 cells, marked pallor. Probably around half of the animals clin-
may occur. In some pigs a substantial granulation tissue ically affected will die, the remainder recovering over a
reaction may occur, leading to fibrous tissue infiltration short period of time, without marked loss of body con-
and muscular hypertrophy, which is the lesion of region- dition. Pregnant animals that are clinically affected may
al ileitis (Rowland and Lawson 1992). abort, the majority within 6 days of the onset of clinical
Acute hemorrhagic PE is marked by severe bleeding signs (Beers 1984).
into the lumen of the intestine, but with underlying le- In most cases of uncomplicated chronic PE, recovery
sions of chronic PE. The hemorrhage occurs concurrent- occurs abruptly 4–10 weeks after the onset of clinical
ly with the widespread degeneration and desquamation signs with a return of appetite and growth rate to normal
of many epithelial cells and leakage from the capillary levels. However, although progress to slaughter weight
bed (Rowland and Lawson 1992). The exact trigger for can take place despite extensive lesions (Emsbo 1951;
this hemorrhagic crisis is not yet known, but it has been Rowland and Hutchings 1978), there will be a reduction
observed in pigs challenged once with L. intracellularis in average weight gain, causing a significant extension of
and also subjected to an acute stressor (Mapother et al. the time pigs take to reach market weight, with a conse-
1987; McOrist and Gebhart, unpublished data—1996), quent burden on the costs of maintaining the facilities.
indicating a direct antiprotective effect of the stressor. The increase in feed required per unit weight gain in af-
fected pigs is also a major cost in extra feed require-
CLINICAL SIGNS ments. Careful feed and weight measurements during re-
peated challenge studies have established that average
Clinical cases of chronic PE are observed most common- weight gains are reduced 6–20% in affected pigs, and the
ly in the postweaned pig between 6 and 20 weeks of age. increase in feed required per unit gain is 6–25%, com-
CHAPTER 38 PORCINE PROLIFERATIVE ENTEROPATHIES McOrist, Gebhart 525

pared to normal pigs (Gogolewski et al. 1991; McOrist et


al. 1996b, 1997). Other costs associated with reduced
weight gains in growing pigs include reduced lean-meat
gain and increased “variation” in pigs destined for mar-
ket or breeding programs.

LESIONS

Chronic PE in growing pigs occurs most commonly in


the terminal 50 cm of the small intestine and the upper
third of the proximal colon. The magnitude of the pro-
liferation varies widely, but in the developed lesions the
wall is visibly thickened and the overall diameter in-
creased. In minor lesions, the area of the terminal ileum
that is 10 cm proximal from the ileocecal valve should be
carefully examined as the most likely site of infection.
Care is needed to distinguish minor lesions from con-
tracted mucosa over the Peyer’s patches. Some subseros-
al and mesenteric edema is common, and the normal
reticulated pattern of the serosal surface is emphasized.
The mucosal surface is moist but not mucoid, sometimes
with flecks of inflammatory exudate loosely adherent.
The affected mucosa itself is thrown into deep folds, lon-
gitudinal or transverse (Fig. 38.1); similar changes in the
large intestine may result in apparent plaque or polyp 38.1. Chronic PE. Ileum showing thickened, ridged
formation. mucosa.
Histologically, the mucosa is composed of enlarged,
branching crypts lined with immature epithelial cells. ing elongated spindles. Goblet cells are absent, and their
Compared to normal crypts, which are 1 cell layer thick, reappearance in deep glands is an indication of impend-
affected crypts are often 5, 10, or more cells thick (Fig. ing resolution. In uncomplicated disease, the lamina pro-
38.2). Numerous mitotic figures occurring throughout pria is normal.
the crypt are evident. Other nuclei of affected cells may Electron microscopy reveals intracellular bacteria, of-
appear as enlarged vesicular structures or densely stain- ten in considerable numbers, lying in the apical cyto-

38.2. Chronic PE, ileum.


High-power micrograph
showing marked enlargement
of affected intestinal crypts,
5–10 epithelial cells thick,
compared with adjacent nor-
mal crypt (H&E; ×400).
526 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

plasm of the affected epithelial cells (Fig. 38.3). In recov- rin deposits and degenerative inflammatory cells. Diag-
ering lesions, the organisms become aggregated and may nosis is confirmed by the presence of remnants of the
be extruded in degenerate cells into the lumen or be con- proliferative epithelium observed in the deep layers. In
sumed by macrophages in the lamina propria. Many cas- long-standing cases, granulation tissue may become
es show little evidence of inflammatory reaction. The re- prominent.
covering lesions are notable for the resumption of
epithelial cell apoptosis with the development of a popu- Regional Ileitis
lation of mature epithelium, with goblet cells in the deep This is recognized as a smoothly contracted, almost rigid
crypts, and a rapid disappearance of the adenomatous length of lower small intestine; hence the traditional
cells from the surface (McOrist et al. 1996a). name “hose pipe gut” is appropriate (Fig. 38.4); the af-
fected portion of intestine often may be stood on end.
Necrotic Enteritis When the lumen is opened, ulceration, usually linear,
This is evident as coagulative necrosis with marked in- can be seen, with islands or strips of surviving mucosa
flammatory exudation superimposed on an established adjacent. Granulation tissue may be prominent, but the
lesion of PE. Yellow-gray cheesy masses that adhere most striking feature is hypertrophy of the outer muscle
tightly to the mucosa are present and may closely follow coats. Both necrotic enteritis and regional ileitis lesions
the original thickened mucosal architecture. Histologi- are now relatively rare occurrences in some diagnostic
cally, the coagulative necrosis is clearly defined, with fib- laboratories, for reasons that are not certain.

38.3. Chronic PE. Intestinal epithelial cell of ileum. Apical cytoplasm containing several L.
intracellularis organisms lying free in the cytoplasm and undergoing division (arrow) (uranyl
acetate and lead citrate; ×10,000).
CHAPTER 38 PORCINE PROLIFERATIVE ENTEROPATHIES McOrist, Gebhart 527

38.4. Regional ileitis. Small intestine showing marked muscular hypertrophy. Normal intestine
adjacent.

Acute Hemorrhagic PE mixed blood and digesta (Fig. 38.5). The mucosal surface
Hemorrhagic PE generally affects the terminal ileum and of the affected portion of intestine shows little gross
colon. The affected intestine is thickened and somewhat damage except for the marked hyperplastic thickening.
turgid with serosal edema. The lumen of the ileum and Bleeding points, ulcers, or erosions are not observed.
colon usually contains one or more formed blood clots, Histological examination demonstrates extensive degen-
but often with no other bloody fluids or feed contents ev- eration and hemorrhage within the proliferative epitheli-
ident. The rectum may contain black, tarry feces of um. There is marked accumulation of cellular debris

38.5. Acute hemorrhagic PE.


Small intestine showing thickened
mucosa and blood clot in lumen.
528 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

containing numerous L. intracellularis organisms above intracellularis in feces, either by a PCR assay using L. in-
the affected mucosa and in the lumina of affected in- tracellularis–specific primers or by using specific hyper-
testinal crypts (Fig. 38.6). immune rabbit serum or, preferably, a specific anti–L. in-
tracellularis monoclonal antibody (McOrist et al. 1987)
DIAGNOSIS incorporated into immunofluorescence assay tech-
niques. In both test systems, pigs with active lesions are
Because of the difficulty of culturing L. intracellularis (see usually found to be excreting the agent (McOrist et al.
below), it has been necessary to develop alternative 1987; McOrist and Lawson 1989b; Jones et al. 1993b);
methods for its detection. Confirmation of a clinical di- however, neither test is likely to prove sufficiently sensi-
agnosis of PE may be obtained by demonstration of L. tive for the diagnosis of all infections. The sensitivity of

38.6. Acute hemorrhagic PE,


ileum. Micrograph of mucosa
showing widespread degeneration
of mucosal crypts with hemor-
rhage on the surface.
CHAPTER 38 PORCINE PROLIFERATIVE ENTEROPATHIES McOrist, Gebhart 529

the PCR assay is considered to be 102–105 organisms per mospheres and cell lysis conditions, respectively (Law-
gram of feces, depending on the DNA extraction method son et al. 1993). Most cells in a monolayer are typically
and type of assay used. Collection of feces for either test infected with around 50 cytoplasmic bacteria (Fig. 38.8),
is relatively easy to perform, and organisms within feces causing no apparent cytopathic effect.
are probably robust for the test procedures. Animals
6–10 weeks old usually have the highest prevalence rates TREATMENT AND PREVENTION
for screening of farms. Older animals are usually only
sampled during outbreaks of acute PE. Feces should be Some understanding of the antimicrobial agents likely to
stored at 4˚C or below for either test. However, both tests be effective against L. intracellularis within pigs’ in-
require specialized reagents and equipment and are ex- testines has been gained. However, full correlation of in
pensive to perform. vitro data, challenge trial data, and field trial data is not
Methods described for the serologic diagnosis of PE yet finalized. Previously, speculative clinical impressions
have employed whole bacterial antigen incorporated in- of field treatments, without relevant controls, pointed
to an indirect immunofluorescence assay (Lawson et al. out many possible treatments (Connor 1991). Controlled
1988; Knittel et al. 1997) or an ELISA (Holyoake et al. field studies suggested that PE may be prevented by
1994). Those assays used bacteria extracted from affect- treatment with high doses of tylosin (100 ppm, Fleck and
ed intestines or cultured L. intracellularis. Results from Jones 1994) or oxytetracycline (400 ppm, Beers 1984).
serologic assays suggest that the serum antibody re- In vitro evaluations of the minimum inhibitory con-
sponse in pigs to L. intracellularis is specific and involves centration (MIC) of 20 antimicrobial agents and the
IgM and IgG. While detectable antibody responses relate minimum bactericidal concentration (MBC) of 10 of
well to the presence of lesions, exposure to infection may these suggested a rather broad range of antibiotic groups
not induce significant seroconversion in all cases. Al- with in vitro activity against L. intracellularis (McOrist et
though blood collection is considerably more time-con- al. 1995c; McOrist and Gebhart 1995). These included
suming than feces collection, the serotests are probably macrolides (erythromycin and tylosin), tetracyclines,
cheaper to perform and more amenable to high through- pleuromulins (tiamulin), penicillins, and fluoro-
puts. Development of sensitive methods for antibody de- quinolones. The aminoglycosides and aminocyclitols
tection may improve detection of low-level infections (neomycin, gentamicin, apramycin) were found to be in-
(Knittel et al. 1997). effective, with the bacteria being fully resistant. Follow-
At necropsy, the use of modified Ziehl-Neelsen stain ing these results, the evaluations of treatment and pre-
or the Giminez stain on mucosal smears to demonstrate vention measures in experimentally challenged pigs have
the intracellular organisms is a simple presumptive tech- indicated that macrolides, lincosamides, chlortetracy-
nique, requiring minimal time and equipment (Love et cline, and tiamulin are effective drugs in vivo (McOrist et
al. 1977). Histopathological examination of affected tis- al. 1996b, 1997; Winkelman 1996b).
sues will reveal the distinctive morphology of the prolif- Various approaches to medication are possible, de-
erative lesions. Specific identification of L. intracellularis pending on the age and type of pigs involved. Treatment
in these lesions can be achieved by immunohistochemi- of acute PE in breeding herds previously thought to be
cal staining of fixed embedded tissues (Lawson et al. free of PE requires a vigorous approach. Treatment
1985; McOrist et al. 1987). In the absence of specific im- needs to include both the clinically affected and the in-
munological reagents, silver-staining techniques will contact animals (which may be the whole herd). The pre-
clearly show the presence of intracellular bacteria (Fig. ferred treatment would be tiamulin (120 ppm), tylosin
38.7). Modifications of the Warthin-Starry silver im- (100 ppm), or chlortetracycline (400 ppm) for 14 days,
pregnation technique (Young 1969) are satisfactory for delivered orally via a water-soluble formulation or an in-
routine use. The affected crypts need to be examined feed premix or by intramuscular injection of an equiva-
carefully at high magnifications due to the small size of lent dose to affected and in-contact pigs.
L. intracellularis. Where electron microscopic facilities Animals such as replacement breeding stock which
are available, the presence of the intracellular organism are to be introduced into infected transport situations or
can be confirmed. The organisms are straight or curved into infected premises could be allowed a period of ex-
bacteria with a wavy trilaminar outer coat typical of posure followed by therapeutic levels of antimicrobial
gram-negative bacteria. agents to impede the occurrence of clinical disease. The
Cocultivation of the obligate intracellular L. intracel- preferred treatment would be tiamulin (120 ppm), ty-
lularis in the laboratory requires establishment of a suit- losin (100 ppm), lincomycin (110ppm), or chlortetracy-
able cell line, such as IEC-18 rat enterocytes or IPEC-J2 cline (300 ppm) for 14 days, delivered orally via a stabi-
pig enterocytes and the addition of purified L. intracellu- lized in-feed premix. Following the transport or on-farm
laris from pig intestines in the presence of antibiotics to period of exposure, no more than 2–3 weeks should
retard the growth of other bacteria (Lawson et al. 1993; elapse before the introduction of antibiotics (Love and
McOrist et al. 1995b). Maintenance and passage of the Love 1977). Such an approach is most suited to the man-
organism in coculture require suitable microaerobic at- agement of acute PE in young adult animals. However,
530 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

38.7. Chronic PE, ileum. Micro-


graph of enlarged mucosal crypt
showing numerous intracellular
bacteria (arrows) in the apical
cytoplasm of epithelial cells
(Warthin-Starry silver stain;
×2000).

even with this type of program, PE may occur in med- probably effective for this purpose in most cases, al-
icated animals following the end of therapy. It is possible though reduced dose rates or inadequate compliance
that treatment of pregnant gilts 1–2 weeks prior to far- would be likely to undermine proper performance of
rowing may reduce transmission of the infection to their these drugs. However, the continuous medication of
progeny. young breeding animals may result in the medicated an-
Where PE is endemic in growing and fattening pigs, imals remaining susceptible to infection, with the danger
another approach to treatment is continuous medica- of reoccurrence of PE following withdrawal of the med-
tion, to minimize the severe production losses caused by ication. These drugs should be delivered orally via a sta-
the disease. Tiamulin (50 ppm), chlortetracycline (200 bilized in-feed premix. The description of ineffective
ppm), lincomycin (110 ppm), and tylosin (100 ppm) are drugs is less certain, but recurrence or failure of preven-
CHAPTER 38 PORCINE PROLIFERATIVE ENTEROPATHIES McOrist, Gebhart 531

appropriate absolute precaution.


It remains a matter of concern that acute and chron-
ic PE continue to be serious problems in high-health-sta-
tus, minimal-disease herds, often with early weaning and
high-quality commercial breeding lines in place. It can-
not be overemphasized that in most conventional herds,
the absence of clinical PE, even over a period of years, is
no guarantee of freedom from the infection and disease.
Apparently clean animals from such herds may be re-
sponsible for the introduction of PE into a hitherto un-
contaminated environment, often followed by an explo-
sive outbreak of acute hemorrhagic PE and later by a low
level of endemic chronic PE.
38.8. Culture of L. intracellularis. Micrograph of REFERENCES
IPEC-J2 pig intestinal epithelial cells, one of which con-
tains numerous intracellular bacteria in the cytoplasm.
Alderton, M. R.; Borland, R.; and Coloe, P. J. 1992. Experi-
Immunoperoxidase stain incorporating Lawsonia-
specific monoclonal antibody. mental reproduction of porcine proliferative enteritis. J
Comp Pathol 106:159–167.
Bane, D.; Gebhart, C. J.; and Gardner, I. 1997. Epidemiology
tion of the disease has been reported in pigs receiving of porcine proliferative enteropathy: A case control
penicillin, bacitracin, neomycin, virginiamycin, salino- study. Proc AASP 27:429–431.
mycin, or olaquindox (Pointon 1989; Winkelman Beers, P. T. 1984. Studies on Porcine Adenomatosis with
1996a). Despite the apparent usage of these drugs at ap- Particular Reference to Proliferative Haemorrhagic En-
propriate levels, intestinal lesions still developed in some teropathy. Ph.D. diss, Univ Sydney.
pigs. Biester, H. E., and Schwarte, L. H. 1931. Intestinal adenoma
Severe chronic clinical disease manifest as wasting in swine. Am J Pathol 7:175–185.
pigs with or without necrotic enteritis will often appear Biester, H. E.; Schwarte, L. H.; and Eveleth, D. F. 1939. Stud-
to be moderated by the use of tylosin, lincomycin, ies on a rapidly developing intestinal adenoma in a pig.
chlortetracycline, or tiamulin. If sufficient numbers of Am J Pathol 15:385–389.
clinical cases are occurring in growing pigs, then the re- Boosinger, T. R.; Thacker, H. L.; and Armstrong, C. H. 1985.
moval of affected animals to separate accommodation, Campylobacter sputorum subsp. mucosalis and Campy-
with supportive therapy, may limit losses. Preliminary lobacter hyointestinalis infections in the intestine of gno-
field trials suggest that incorporation of in-feed antibi- tobiotic pigs. Am J Vet Res 46:2152–2156.
otics for control achieves best results at 4–8 weeks of age, Chang, K.; Kurtz, H. J.; Ward, G. E.; and Gebhart, C. J. 1984.
that is, just prior to the peak period of Lawsonia infection Immunofluorescent demonstration of Campylobacter
on most farms. These trials also suggest that medication hyointestinalis and Campylobacter spurorum subspecies
of older pigs, such as breeding stock, is not likely to elim- mucosalis in swine intestine with lesions of proliferative
inate the infection from their progeny or from other enteritis. Am J Vet Res 45:703–710.
groups. Inherent bacterial resistance to tetracycline or Christensen, G.; Vraa-Andersen, L.; and Mousing, J. 1995.
other groups of drugs has not been proven and is not Causes of mortality among sows in Danish pig herds.
common in other obligately intracellular bacteria. Vet Rec 137:395–399.
Because PE is rarely observed in apparently recovered Christensen, N. H., and Cullinane, L. C. 1990. Monitoring
pigs or in pigs over 2 years old, it is likely that some nat- the health of pigs in New Zealand abattoirs. NZ Vet J
ural immunity exists. Also, specific cell-mediated and hu- 38:136–141.
moral antibody responses to infection have been detect- Collins, A. M.; Swift, I.; and Monckton, R. P. 1996. Replica-
ed, albeit at a low level (Lawson et al. 1988; McOrist and tion of Australian porcine isolates of ileal symbiont in-
Lawson 1993). However, the common occurrence of the tracellularis in tissue culture. Vet Microbiol 49:249–255.
disease and the wide age range of susceptible pigs sug- Connor, J. F. 1991. Diagnosis, treatment and prevention of
gest that natural immunity in field situations may not porcine proliferative enteritis. Comp Contin Educ
occur regularly. 13:1172–1177.
Estimates of the financial costs of the disease suggest Cooper, D. M. 1996. Proliferative Enteritis in the Hamster,
that the costs in extra feed, extra facility time, and possi- Horse, Deer and Ostrich: Detection and Characteriza-
ble loss of carcass leanness are substantial (U.S.$5–10 tion of Lawsonia intracellularis. M.Sc. thesis, Univ Min-
per affected pig), especially considered together with the nesota.
high prevalence of the disease. Sustained isolation of the Drolet, R.; Larochelle, D.; and Gebhart, C. J. 1996. Prolifera-
herd with rigorous control of access appears the most tive enteritis associated with Lawsonia intracellularis
532 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

(ileal symbiont intracellularis) in white-tailed deer. J Vet Kashiwazaki, M.; Namioka, S.; and Yabiki, T. 1971. Gnoto-
Diagn Invest 8:250–253. biotic pigs exposed to Vibrio coli. Nat Inst Anim Health
Duhamel, G. E., and Wheeldon, E. B. 1982. Intestinal ade- Quart 11:145–150.
nomatosis in a foal. Vet Pathol 19:447–450. Knittel, J. P.; Larson, D. I.; Harris, D. L.; Roof, M. B.; and
Duran, C. O. 1994. Causes of sow mortality. Pig J McOrist, S. 1996. United States isolates of Lawsonia in-
32:107–112. tracellularis from porcine proliferative enteropathy re-
Emsbo, P. 1951. Terminal or regional ileitis in swine. Nord semble European isolates. Swine Health Prod
Vet Med 3:1–28. 4:119–122.
Eriksen, K., and Landsverk, T. 1985. Intestinal adenomato- Knittel, J. P.; Roof, M.; Schwartz, K. J.; Jordan, D. M.; Harris,
sis in the blue fox. Nord Vet Med 37:254–255. D. L.; McOrist, S. 1997. Diagnosis of porcine prolifera-
Fleck, M. K., and Jones, G. F. 1994. Porcine proliferative en- tive enteritis. Comp Contin Educ Pract Vet
teropathy outbreak and treatment using tylosin. Proc 19(suppl):S26–S29, S35.
Int Congr Pig Vet Soc 13:345. Kubo, M.; Ohya, T.; and Watase, H. 1984. Proliferative he-
Fox, J. G., and Lawson, G. H. K. 1988. Campylobacter-like morrhagic enteropathy detected at an abattoir in
omega intracellular antigen in proliferative colitis in fer- Kagoshima. Jap J Vet Sci 46:413–417.
rets. Lab Anim Sci 38:34–36. Lanza, I., and Pozo, J. 1996. Epidemiologia de la ileitis porci-
Fox, J. G.; Stills, H. F.; Paster, B. J.; Dewhirst, F. E.; Yan, L.; na. Porci Vet 32:42–49.
Palley, L.; and Pastek, K. 1993. Antigenic specificity and Lawson, G. H. K., and Rowland, A. C. 1974. Intestinal ade-
morphologic characteristics of Chlamydia trachomatis, nomatosis in the pig: A bacteriological study. Res Vet Sci
strain SFPD, isolated from hamsters with proliferative 17:331–336.
ileitis. Lab Anim Sci 43:405–410. Lawson, G. H. K.; Rowland, A. C.; Roberts, L.; Fraser G.; and
Frisk, C. S., and Wagner, J. E. 1977. Experimental hamster McCartney, E. 1979. Proliferative haemorrhagic en-
enteritis: An electron microscopic study. Am J Vet Res teropathy. Res Vet Sci 27:46–51.
38:1861–1868. Lawson, G. H. K.; Rowland, A. C.; and MacIntyre, N. 1985.
Gebhart, C. J.; Ward, G. E.; Chang, K.; and Kurtz, H. J. 1983. Demonstration of a new intracellular antigen in porcine
Campylobacter hyointestinalis (new species) isolated from intestinal adenomatosis and hamster proliferative ileitis.
swine with proliferative ileitis. Am J Vet Res 45:361–367. Vet Microbiol 10:303–313.
Gebhart, C. J.; Barns, S. M.; McOrist, S.; Lin, G.-F.; and Law- Lawson, G. H. K.; McOrist, S.; Rowland, A. C.; Roberts, L.;
son, G. H. K. 1993. Ileal symbiont intracellularis, an ob- and McCartney, E. 1988. Serological diagnosis of the
ligate intracellular bacterium of porcine intestines porcine proliferative enteropathies: Implications for ae-
showing a relationship to Desulfovibrio species. Int J Syst tiology and epidemiology. Vet Rec 122:554–557.
Bacteriol 43:533–538. Lawson, G. H. K.; McOrist, S.; Sabri, J.; and Mackie, R. A.
Gogolewski, R. P.; Cook, R. W.; and Batterham, E. S. 1991. 1993. Intracellular bacteria of porcine proliferative en-
Suboptimal growth associated with porcine intestinal teropathy: Cultivation and maintenance in vitro. J Clin
adenomatosis in pigs in nutritional studies. Aust Vet J Microbiol 31:1136–1142.
68:406–408. Lawson, G. H. K.; Mackie, R. A.; Smith, D. G. E.; and
Holyoake, P. K., and Cutler, R. S. 1995. Outbreaks of prolif- McOrist, S. 1995. Intracellular multiplication of ileal
erative haemorrhagic enteropathy on two pig farms. symbiont intracellularis depends on host cell function
Aust Vet J 72:253–256. and actin polymerisation. Vet Microbiol 45:339–350.
Holyoake, P. K.; Cutler, R. S.; Caple, I. W.; and Monckton, R. Lomax, L. G., and Glock, R. D. 1982. Naturally occurring
P. 1994. Enzyme-linked immunosorbent assay for mea- porcine proliferative enteritis: Pathologic and bacterio-
suring ileal symbiont intracellularis-specific im- logic findings. Am J Vet Res 43:1608–1614.
munoglobulin G response in sera of pigs. J Clin Micro- Love, R. J., and Love, D. N. 1977. Control of proliferative
biol 32:1980–1985. haemorrhagic enteropathy in pigs. Vet Rec 100:473.
Joens, L. A.; Finn, D. L.; and Saif, L. J. 1987. Etiology of pro- Love, R. J.; Love, D. N.; and Edwards, M. J. 1977. Prolifera-
liferative ileitis: Viral components. In Proc A. L. Leman tive haemorrhagic enteropathy in pigs. Vet Rec
Swine Herd Health Conf, pp. 137–143. 100:65–68.
Jones, G. F.; Ward, G. E.; Murtaugh, M. P.; Rose, R.; and Mapother, M. E.; Joens, L. A.; and Glock, R. D. 1987. Exper-
Gebhart, C. J. 1993a. Relationship between ileal sym- imental reproduction of porcine proliferative enteritis.
biont intracellularis and porcine proliferative enteritis. Vet Rec 121:533–536.
Infect Immun 61:5237–5244. McCartney, E.; Lawson, G. H. K.; and Rowland, A. C. 1984.
Jones, G. F.; Ward, G. E.; Murtaugh, M. P.; Lin, G.; and Geb- Behaviour of Campylobacter sputorum sub-species mucos-
hart, C. J. 1993b. Enhanced detection of intracellular or- alis in gnotobiotic pigs. Res Vet Sci 36:290–297.
ganism of swine proliferative enteritis, ileal symbiont in- McOrist, S., and Gebhart, C. J. 1995. In vitro testing of an-
tracellularis, in feces by polymerase chain reaction. J timicrobial agents for proliferative enteropathy (ileitis).
Clin Microbiol 31:2611–2615. Swine Health Prod 3:146–149.
CHAPTER 38 PORCINE PROLIFERATIVE ENTEROPATHIES McOrist, Gebhart 533

McOrist, S., and Lawson, G. H. K. 1989a. Reproduction of McOrist, S.; Morgan, J.; Veenhuizen, M. F.; Lawrence, K.;
proliferative enteritis in gnotobiotic piglets. Res Vet Sci and Kroger, H. W. 1997. Oral administration of tylosin
46:27–33. phosphate for treatment and prevention of proliferative
———. 1989b. Failure to demonstrate Campylobacter-like enteropathy in pigs. Am J Vet Res 58:136–139.
organisms of the proliferative enteropathies in pigs ex- Moller, K.; Jensen, T. K.; and Jorsal, S. E. 1996. Bacteriologi-
creting Campylobacter sp. Vet Rec 124:41. cal examination and PCR analysis of feces from growing
———. 1993. Interactions of porcine lymphocytes with pigs originating from herds with and without diarrhea.
Campylobacter-like organism membranes purified from Proc Int Congr Pig Vet Soc 14:325.
proliferative enteropathy. Vet Microbiol 34:381– Pointon, A. M. 1989. Campylobacter associated intestinal
388. pathology in pigs. Aust Vet J 66:90–91.
McOrist, S.; Boid, R.; Lawson, G. H. K.; and McConnell, I. Roberts, L.; Rowland, A. C.; and Lawson, G. H. K. 1977. Ex-
1987. Monoclonal antibodies to intracellular Campy- perimental reproduction of porcine intestinal adeno-
lobacter-like organisms of the porcine proliferative en- matosis and necrotic enteritis. Vet Rec 100:12–13.
teropathies. Vet Rec 121:421–422. Roberts, L.; Lawson, G. H. K.; Rowland, A. C.; and Laing, A.
McOrist, S.; Boid, R.; and Lawson, G. H. K. 1989a. Anti- H. 1979. Porcine intestinal adenomatosis and its detec-
genic analysis of Campylobacter species and an intracel- tion in a closed pig herd. Vet Rec 104:366–368.
lular Campylobacter-like organism associated with the Robinson, I. M.; Whipp, S. C.; Bucklin, J. A.; and Allison, M.
porcine proliferative enteropathies. Infect Immun J. 1984. Characterization of predominant bacteria from
57:957–962. the colons of normal and dysenteric pigs. Appl Environ
McOrist, S.; Lawson, G. H. K.; Rowland, A. C.; and MacIn- Microbiol 48:964–969.
tyre, N. 1989b. Early lesions of proliferative enteritis of Rowland, A. C., and Hutchings, D. A. 1978. Necrotic enteri-
pigs and hamsters. Vet Pathol 26:260–264. tis and regional ileitis in pigs at slaughter. Vet Rec
McOrist, S.; MacIntyre, N.; Stokes, C. R.; and Lawson, G. H. 103:338–339.
K. 1992. Immunocytological responses in the porcine Rowland, A. C., and Lawson, G. H. K. 1974. Intestinal ade-
proliferative enteropathies. Infect Immun nomatosis in the pig: Immunofluorescent and electron
60:4184–4191. microscopic studies. Res Vet Sci 17:323–330.
McOrist, S.; Jasni, S.; Mackie, R. A.; MacIntyre, N.; Neef, N.; ———. 1975. Porcine intestinal adenomatosis: A possible
and Lawson, G. H. K. 1993. Reproduction of porcine relationship with necrotic enteritis, regional ileitis and
proliferative enteropathy with pure cultures of ileal sym- proliferative haemorrhagic enteropathy. Vet Rec
biont intracellularis. Infect Immun 61:4286–4292. 97:178–180.
McOrist, S.; Mackie, R. A.; Neef, N.; Aitken, I.; and Lawson, ———. 1992. Porcine proliferative enteropathies. In Dis-
G. H. K. 1994. Synergism of ileal symbiont intracellu- eases of Swine, 7th ed. Ed. A. D. Leman, B. E. Straw, W.
laris and gut flora in the reproduction of porcine prolif- L. Mengeling, S. D’Allaire, and D. J. Taylor. Ames: Iowa
erative enteropathy. Vet Rec 134:331–332. State Univ Press, pp. 560–569.
McOrist, S., Gebhart, C. J.; Boid, R.; and Barns, S. M. 1995a. Rowland, A. C., and Rowntree, P. G. M. 1972. A haemor-
Characterization of Lawsonia intracellularis gen. nov., sp. rhagic bowel syndrome associated with intestinal ade-
nov., the obligately intracellular bacterium of porcine nomatosis in the pig. Vet Rec 91:235–241.
proliferative enteropathy. Int J Syst Bacteriol Schoeb, T. R., and Fox, J. G. 1990. Enterocolitis associated
45:820–825. with intraepithelial Campylobacter-like bacteria in rab-
McOrist, S.; Jasni, S.; Mackie, R. A.; Berschneider, H. M.; bits. Vet Pathol 27:73–80.
Rowland, A. C.; and Lawson, G. H. K. 1995b. Entry and Smith, S. H. 1997. Epidemiological Features of Porcine
release of the bacterium ileal symbiont intracellularis in Proliferative Enteropathies. Ph.D. diss, Univ Edin-
cultured enterocytes. Res Vet Sci 59:255–260. burgh.
McOrist, S.; Mackie, R. A.; and Lawson, G. H. K. 1995c. An- Smith, S. H., and McOrist, S. 1997. Development of persis-
timicrobial susceptibility of ileal symbiont intracellu- tent intestinal infection and excretion of Lawsonia intra-
laris isolated from pigs with porcine proliferative en- cellularis by piglets. Res Vet Sci 62:6–10.
teropathy. J Clin Microbiol 33:1314–1317. Vandenberghe, J.; Verheyen, A.; Lauwers, S.; and Geboes, K.
McOrist, S.; Roberts, L.; Jasni, S.; Rowland, A. C.; Lawson, 1985. Spontaneous adenocarcinoma of the ascending
G. H. K.; Gebhart, C. J.; and Bosworth, B. 1996a. Devel- colon in Wistar rats: The intracytoplasmic presence of a
oped and resolving lesions in porcine proliferative en- Campylobacter-like bacterium. J Comp Pathol 95:45–55.
teropathy: Possible pathogenetic mechanisms. J Comp Williams, N. M.; Harrison, L. R.; and Gebhart, C. J. 1996.
Pathol 115:35–45. Proliferative enteropathy in a foal caused by Lawsonia
McOrist, S.; Smith, S. H.; Shearn, M. F. H.; Carr, M. M.; and intracellularis–like bacterium. J Vet Diagn Invest
Miller, D. J. S. 1996b. Treatment and prevention of 8:254–256.
porcine proliferative enteropathy with oral tiamulin. Vet Winkelman, N. L. 1996a. Ileitis: An update. Comp Contin
Rec 139:615–618. Educ 18:S19–S25.
534 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

———. 1996b. Evaluation of preventive or therapeutic feed Young, B. J. 1969. A reliable method for demonstrating
medication programs with chlortetracycline in swine spirochaetes in tissue sections. J Med Lab Technol
challenged with Lawsonia intracellularis. In Roche Anim 26:248–252.
Nutr Health Vet Educ Seminar, Univ Minnesota, pp.
50–62.
Salmonellosis
39 K. J. Schwartz

Members of the genus Salmonella are notorious for their typical of the family Enterobacteriaceae, the intestinal
ability to infect a broad range of hosts, which is a major tract of warm-blooded and cold-blooded animals.
factor in their success as pathogens. Taylor and McCoy Salmonellae are hardy and ubiquitous bacteria. They
(1969) observed that salmonellae have been isolated multiply at 7–45˚C; survive freezing and desiccation
from virtually all vertebrate hosts from which they have well; and persist for weeks, months, or even years in suit-
been sought, with the possible exception of fish in un- able organic substrates. Salmonellae were reported to
polluted waters. Although many of the more than 2400 survive in meat-meal fertilizer for 8 months (Mittermey-
salmonella serotypes have a broad host range and are er and Foltz 1969) and in manure oxidation ditches for
widely distributed, several serotypes are quite adapted to 47 days (Will et al. 1973). Survival is greatly shortened
a single host species, most notably S. typhi (humans), S. below pH 5.0 (Henry et al. 1983). Numerous reports of
dublin (bovine), and S. choleraesuis (swine). Many prolonged survival in water have been cited (Williams
serotypes are not associated with overt disease and ap- 1975; Wray and Sojka 1977; Pokorny 1988). The bacteria
pear to have limited host and geographical range. are readily inactivated by heat and sunlight as well as by
Salmonella infections of swine are of concern for two common phenolic, chlorine, and iodine-based disinfec-
major reasons. The first is the clinical disease (salmonel- tants (Rubin and Weinstein 1977). Ability to survive in
losis) in swine that may result, and the second is that the environment, as well as prolonged carrier states in in-
swine can be infected with a broad range of salmonella numerable hosts, ensures the widespread distribution of
serotypes that can be a source of infection of pork prod- this genus worldwide.
ucts. Techniques for isolation of salmonellae vary widely,
Salmon and Smith (1886) first associated salmonel- depending on the nature of the suspect material and
lae with disease when they described S. choleraesuis as the sometimes with the specific serotypes sought. In sewage,
putative cause of classical swine fever (hog cholera). The feed, and polluted water, where salmonella numbers are
identification and, in many swine-producing areas, erad- likely to be low compared to other organisms, well-docu-
ication of the viral etiology of classical swine fever, rele- mented and sometimes elaborate techniques of preen-
gated S. choleraesuis to an opportunistic pathogen in richment, selective enrichment, and selective plating are
swine. The dramatic increase in salmonellosis during the commonly used (Groves et al. 1971; Edwards and Ewing
1980s in North America underscored the pathogenic po- 1972; Skovgaard et al. 1985; Vassiliadis et al. 1987). Oc-
tential of S. choleraesuis for swine. casionally these methods may be necessary for the isola-
Disease associated with host-adapted S. choleraesuis is tion of salmonellae from tissues or feces of carrier ani-
referable to septicemia, enterocolitis, or bacteremic lo- mals in which numbers are low, but in clinically affected
calization as pneumonia and hepatitis (Baskerville and animals the populations are such that direct plating of
Dow 1973) or occasionally as meningitis (Reynolds et al. internal organs on routine selective and differential en-
1967; McErlean 1968), encephalitis (Wilcock and Olan- teric media such as brilliant green agar and MacConkey
der 1977c), and abortion (Schwartz and Daniels 1987). agar are usually adequate (Committee on Salmonella
Only a handful of other serotypes are associated with 1969). The isolation of salmonellae is not sufficient for
disease in swine, usually as a cause of enterocolitis, the definitive diagnosis of salmonellosis, particularly if elab-
most notable being S. typhimurium. Reported rarely, S. orate isolation techniques are required, since subclinical
typhisuis is associated with caseous lymphadenitis infections and environmental contamination are com-
(Barnes and Bergeland 1968). mon. Isolation techniques for various types of specimens
are detailed in most standard texts on clinical microbiol-
ETIOLOGY ogy. Epidemiological investigations of zoonotic out-
breaks occasionally demand more sophisticated tech-
The genus Salmonella is a morphologically and biochem- niques of phage typing, plasmid characterization,
ically homogeneous group of gram-negative, motile, mapping of outer-membrane proteins (OMP), or DNA
non-spore-forming, facultatively anaerobic bacilli with analysis to trace a specific isolant.
peritrichous flagella. The reservoir for salmonellae is, S. choleraesuis is the type species for the genus Salmo-

535
536 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

nella as described by Salmon, although it is now more 1972; Wilcock et al. 1976; Mills and Kelly 1986;
commonly isolated as the hydrogen sulfide–producing Schwartz and Daniels 1987; Schwartz 1997a), generally
variant kunzendorf. Some authors suggest that there are manifested as septicemia.
only two actual species of salmonellae with well over S. typhimurium is the second most frequently isolated
2400 serotypes. Convention, however, is to refer to each serotype from diseased swine, usually associated with en-
of the serologically distinct serotypes, usually named by terocolitis. Disease caused by S. typhimurium occurs with
the geographic site of first isolation, as a species. greater than expected frequency in what could be con-
Serotype identification uses the Kauffmann-White sidered unusually clean herds: university research herds,
schema, based on antigenic differences in somatic (O), testing stations, closed specific-pathogen-free (SPF)
surface or capsular (Vi), phase 1 flagellar, and phase 2 fla- herds, or purebred breeding herds (Heard et al. 1965;
gellar antigens determined by agglutination serology. Gooch and Haddock 1969; Lynn et al. 1972). Presum-
Complete serotyping is laborious and is available at only ably, this is because of introduction to a previously im-
a few reference laboratories. Most laboratories use com- munologically naive population, a situation occurring
mercially available polyvalent antisera to determine the with increasing frequency in modern production units
O antigen groups of isolates for rapid and preliminary using age-segregated rearing. This organism is also fre-
identification. The serogroup designation will help pre- quently isolated as a sequel to other enteric or debilitat-
dict the serotype present or, at least, can be used to rule ing diseases.
out those serotypes found in other serogroups (Table Localized epizootics of disease caused by the bio-
39.1). chemically atypical S. typhisuis have been reported in the
In contrast to the large number of serotypes isolated American Midwest (Barnes and Bergeland 1968; An-
from carcasses and pork products, disease in swine is al- drews 1976) and at least historically in Europe (Barnes
most always caused by either the hydrogen sulfide–pro- and Sorensen 1975). This organism grows poorly in stan-
ducing variant of S. choleraesuis variety kunzendorf or by dard selective media for salmonella isolation, but the dis-
S. typhimurium. The former has been and continues to be ease produced by S. typhisuis is so characteristic that out-
the most frequent serotype causing disease in swine breaks are not likely to remain unnoticed (Barnes and
(Levine et al. 1945; Lawson and Dow 1966; Morehouse Bergeland 1968).

Table 39.1. Serogroups of selected salmonella serotypes and ranking of frequency of isolation from diseased
pigs, swine sources, and humans
Isolations from Isolations from Isolations from
Diseased Pigs Swine Sources Humans
Serogroup Serotype (Schwartz 1997a) (Ferris and Frerichs 1996) (CDC 1996)
A S. paratyphi A
B S. typhimurium 3 5 3
S. typhimurium var. copenhagen 2 3 2
S. derby 1
S. agona 4 6
S. schwarzengrund 10
S. hadar 7
S. saint paul
S. heidelberg 4 6 4
C1 S. choleraesuis 7
S. choleraesuis var. kunzendorf 1 2
S. mbandaka 9
S. typhisuis
S. montevideo 8
S. oranienburg 9
S. infantis
S. thompson 10
C2 S. newport 5
S. muenchen
D1 S. dublin
S. typhi
S. enteritidis 1
S. pullorum
E1 S. anatum 8
E2 S. newington
E4 S. senftenberg
G2 S. worthington
CHAPTER 39 SALMONELLOSIS Schwartz 537

Other serotypes are occasional causes of disease in ly, disease prevalence surveys must be carefully scruti-
swine but are usually transient and associated with pre- nized to be sure that infection is not equated with dis-
disposing factors, including other intestinal distur- ease, and that a source of infection is not inaccurately
bances or disease; circumstances which allow immuno- implicated.
logically naive pigs to be exposed to very large doses; or
debilitated and immunologically compromised pigs. A Salmonellae in Pork
variety of serotypes may be isolated from diarrheic Data collected from various countries indicate salmonel-
piglets in the immediate postweaning period, but most lae to be present in 0–48% of carcasses (Riley 1970; Not-
are usually associated with concurrent enteric tingham et al. 1972; McCaughey et al. 1973; Gustafson et
pathogens, inappropriate diets, poor hygiene and envi- al. 1976; Tacal and Lawrence 1980; Morse and Hird
ronment, or debilitation. The isolation of uncommon 1984; Jayarao et al. 1989; Carr et al. 1996) and 0–30% of
serotypes of salmonellae from diarrheic pigs generally retail pork products (Gooch and Goo 1971; Surkiewicz et
warrants further diagnostic investigation. Salmonella hei- al. 1972; Roberts et al. 1975; Banks and Board 1983; Silas
delberg has been associated with postweaning diarrhea, et al. 1984; Fukushima et al. 1987; Anon. 1994). The
with lesions more typical of enterotoxigenic diarrheal marked variation is probably due, in part, to real varia-
disease than typical salmonellosis (Reed et al. 1985). Re- tion in contamination and, in part, to differences in
ports of naturally occurring disease, such as those for S. methods of survey and methods of meat processing. The
dublin (Lawson and Dow 1966; McErlean 1968) and S. high level of infection demonstrated in some of the stud-
enteritidis (Reynolds et al. 1967), should support the iso- ies is likely the result of abattoir cross-contamination in
lation of the offending serotype with clinical and patho- crowded holding pens prior to slaughter as well as me-
logic evidence of salmonellosis. In the case of both S. chanical transfer of contamination among carcasses by
dublin and S. enteritidis, the reports were of meningitis in dehairing machines, scalding tanks, and polishers (Gal-
suckling pigs. ton et al. 1954; Hansen et al. 1964; Kampelmacher et al.
1965; Williams and Newell 1970; Michaud 1978; Mor-
EPIDEMIOLOGY gan et al. 1987). Although much of the salmonella
contamination of pork products occurs within abattoirs
The reservoir for salmonellae is the intestinal tract of during processing, infected pigs leaving the farm are con-
warm- and cold-blooded animals. Salmonellae have mas- sidered the original source of abattoir infections. The
tered virtually all of the attributes necessary to ensure stress of transport and feed deprivation increases shed-
wide distribution, including abundant reservoir hosts, ding from inapparent carriers, which then contaminate
efficient fecal shedding from carrier animals, persistence the environment of the truck and abattoir (Williams and
within the environment, and the effective use of trans- Newell 1970). The prevalence of infection within the
mission vectors (feed, fomites, vehicles, etc.). Inappar- group continues to increase with increasing length of
ent, long-term carriers that can shed salmonellae in feces stay in the pens prior to slaughter, rising by about 50%
continuously or intermittently, often in high numbers, for each 24-hour period (Craven and Hurst 1982; Mor-
are common in most host species. Shedding of the or- gan et al. 1987). It should be noted that S. choleraesuis is
ganism can be exacerbated by a long list of stressors, in- rarely associated with contamination of carcasses and
cluding commingling of pigs, transportation, concur- pork products, and although unusual as a cause of hu-
rent diseases, and food deprivation. man disease, it is particularly severe when it does occur
The epidemiology of salmonella infections in swine is (Cherubin 1980).
two relatively separate problems: salmonella infection of There is currently an explosion of investigational ac-
pork carcasses and retail products and infections that tivity related to issues of food safety, including salmonel-
cause salmonellosis in swine. Infection of swine by one la contamination of a variety of foods. Salmonellosis is
or more serotypes is common, but primary clinical dis- considered to be the most common food-borne illness in
ease caused by serotypes other than S. choleraesuis or S. humans, with increasing incidence reported worldwide.
typhimurium is uncommon. It is important to under- Reasons include improved monitoring, increased public
stand that swine can be infected with a variety of awareness of microbiological hazards of food, wide-
serotypes that do not cause disease in swine but do rep- spread distribution of virulent serotypes such as S. enter-
resent a source of infection for pork products. itidis or S. tyhimurium definitive type 104 (DT 104) in an-
Extrapolation of epidemiological data from experi- imal populations, increasing consumption of foods of
mental studies where a single serotype with predeter- animal origin, and changes in consumer eating habits.
mined dose is administered to naive healthy pigs is not Although salmonella contamination of poultry and beef
likely to represent field situations, where there are multi- products exceeds that of pork, salmonella control pro-
ple serotypes, varying doses, intermittent exposures, grams in swine will continue to be a primary focus of
variable host resistance, many management variables, food safety initiatives. Salmonella reduction programs
and various intercurrent infections and diseases. Similar- are becoming commonplace, with long-range goals to in-
538 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

clude the production and marketing of salmonellae-free S. typhimurium accounts for most of the remaining
pork products. Already, numerous, dynamic programs cases of salmonellosis in swine. This serotype has world-
are in place utilizing hazard analysis and critical control wide distribution and is not host-specific. Enterocolitis is
point (HACCP) principles. Those programs that have the primary disease referable to this serotype, most com-
been in place for a sufficient period of time, such as the monly seen in pigs with concurrent debilitating illnesses,
Danish program, have significantly reduced the rate of in conditions of poor hygiene that allow exposure to
salmonella infection in pork products (Nielsen et al. high doses of the organism, or where immunologically
1995). Fortunately, most of the methods useful for pre- naive pigs are exposed to sufficiently large doses. The lat-
harvest salmonella reduction in swine populations are ter situation appears to be more frequently encountered
related to sound management practices that also im- with modern production systems utilizing age-segregat-
prove the overall health of a swine operation. ed production.
The attribution of primary pathogenic status to oth-
Salmonellosis in Swine er serotypes should be made with caution. Most other
Most salmonellosis outbreaks occur in intensively reared serotypes are transient, sporadic causes of disease and
weaned pigs, and although disease in adults and suckling often cannot be associated with disease experimentally
pigs is infrequent, infection is not (Gooch and Haddock without unique, qualifying criteria. S. heidelberg has been
1969; Wilcock et al. 1976). The low frequency of salmo- associated with catarrhal enterocolitis in young pigs,
nellosis in suckling pigs presumably results from lacto- with enterotoxigenic properties leading to accumula-
genic immunity, since neonatal swine are susceptible to tions of large amounts of fluid in the small intestine and
oral challenge with salmonellae and develop a disease colon as a rather unusual presentation of salmonellosis
comparable to that in weaned pigs (Wilcock 1978). Dis- (Reed et al. 1985).
ease occurs worldwide but varies markedly in estimated
prevalence, morbidity, and mortality. This may be from Sources of Infection
incautious extrapolation of data gathered from microbi- The number of potential sources of salmonella infection
ological surveys applied to actual disease incidence. In for a population of swine is seemingly endless. A task
one correlative study in Indiana in 1974–75, salmonel- force study in the United States did not reach a consen-
losis accounted for 19% of 327 consecutive porcine sus as to the most important source of salmonellae for
necropsies (Wilcock et al. 1976). In contrast, Hooper and pigs (Bixler 1978), in large part due to the diversity and
Troutt (1971) reported that salmonella infection was the biology of the genus Salmonella. In general, the
considered the major disease process in only 2% of sam- source of salmonellae virulent for swine is most likely to
ples submitted in Missouri between 1967 and 1969. Dur- be other swine or environments contaminated by swine.
ing a 4-year period in Ireland, salmonellosis was the di- S. choleraesuis is the most frequent porcine isolate from
agnosis in 4.4% of 2180 swine necropsies (Lawson and clinically ill pigs, but it is a very infrequent isolate from
Dow 1966). In Taiwan, salmonellae were isolated from pig feeds or nonporcine salmonella reservoirs. The con-
about 10% of scouring pigs and 48% of fatal diarrheas or clusion seems clear that infected, shedding pigs and con-
septicemias in weaned pigs (Hsu et al. 1983). A survey of taminated environments are the major sources of new
diagnostic submissions to the Iowa State University Vet- infections of S. choleraesuis. Vertical (dam to offspring)
erinary Diagnostic Laboratory from 1994 through 1996 and horizontal transmission both occur. Feed contami-
revealed salmonellae to be present in 11% of 9109 nation and nonporcine species have not been implicated
porcine pneumonia cases, 9% of 3320 enteric cases, and as a source of S. choleraesuis infection of swine.
58% of 1612 cases of porcine bacterial septicemia The source of infection for other serotypes is less
(Schwartz 1997a). clear, since the host and vector range for salmonellae is
Host-adapted S. choleraesuis is isolated almost exclu- broad and they have amazing capability to persist in en-
sively from diseased swine, is the most common cause of vironments outside the host. For serotypes other than S.
salmonellosis in swine, and is usually manifested as sep- choleraesuis, pigs should be thought of as biological fil-
ticemia. Midwestern U.S. diagnostic laboratories and ters for the low numbers of various salmonella serotypes
veterinarians reported an increasing frequency of salmo- present in feed, water, or litter contaminated by birds,
nellosis due to S. choleraesuis from 1981 to 1990 and a de- rodents, or other animals. Evidence linking these sources
creasing frequency from 1991 to 1997 (Schwartz 1997b). of contamination to primary clinical outbreaks, without
The recent decrease in the Midwest is likely due to im- other concurrent diseases or predisposing conditions, is
provements in swine management and husbandry and generally lacking. Feed containing ingredients of animal
the advent of efficacious attenuated live vaccines. Re- origin is widely accepted as a source of salmonella infec-
gional variation in salmonellosis incidence is loosely cor- tion to herds, but it should be emphasized that ingredi-
related to pig density, husbandry practices, and, in par- ents of vegetable origin can also be a source of salmonel-
ticular, commingling of pigs of different ages and/or lae-contaminated feed. Water is not as likely a source of
origins. infection unless surface water is used for consumption or
CHAPTER 39 SALMONELLOSIS Schwartz 539

pigs have access to recycled lagoon water. Birds, insects, tablished in field situations, but disease is difficult to re-
rodents, and pets can all act as carriers, as can bedding produce experimentally at low doses. There is one report
and litter (Allred et al. 1967; Williams et al. 1969; Nape of moderate disease following oral inoculation of 106
and Murphy 1971). In a recent survey in the United cells (Dawe and Troutt 1976), but most authors report
States, salmonellae were isolated from feed or feed ingre- successful experimental disease production with doses of
dients from 14 of 30 farms and 36 of 1228 samples (Har- 108–1011 cells unless pigs are artificially stressed by injec-
ris et al. 1997). The isolation of salmonellae from feed tion of dexamethasone or in some other manner. Pigs in-
was significantly associated with the lack of bird-proof- fected with 103 organisms remained clinically normal but
ing, with on-farm feed preparation, and with the housing uninoculated pigs in the same pen did become clinically
of pigs in facilities other than total confinement, for all ill (Gray et al. 1996). It is likely that dose (and perhaps
stages of production. Interestingly, salmonella isolation virulence) is magnified when pigs are infected and se-
from complete feeds was more frequent from pelleted quential (pig-to-pig) transmission occurs in field situa-
feeds than from ground feed. No S. typhimurium was iso- tions, so that the initial infective dose in the field is con-
lated from feed samples in this study. siderably less than that required in experimental
situations. High animal density, stress of transport, and
Transmission, Shedding, and Carrier State intercurrent nutritional or infectious disease are as-
Because of the dynamic relationship between salmonel- sumed to increase the shedding by carriers as well as the
lae, host, and environment, and because infection does susceptibility of exposed pigs (Committee on Salmonel-
not equate with disease, definitive statements regarding la 1969). Pigs with nondetectable shedding of salmonel-
transmission, shedding, and carrier states are apt to be lae can become detectable within hours of an applied
misinterpreted, if not erroneous. Salmonella transmis- stress. The transmission demonstrated between feeder
sion and shedding within differing populations of ani- pigs also occurs between pigs during market transport
mals in an endless variety of environmental, feeding, and and lairage at abattoirs, with infection rates proportion-
management situations result in countless unique situa- al to time spent in transport and lairage. It is likely that
tions that cannot be experimentally reproduced. The catecholamines are released in association with stress, re-
sensitivity of most current detection methods is not ade- sulting in decreased gastric acid production and in-
quate to define these parameters in field situations. In creased intestinal motility. Increases in stomach pH in-
general, fecal-oral transmission is the most likely mode crease the likelihood that salmonellae will survive
of transmission of virulent salmonellae. This can occur passage through the stomach and will access and repli-
from pig to pig, contaminated environment to pig, or cate in the intestine and colon.
dam to offspring. Oral-pharyngeal secretions may con- Outbreaks of salmonellosis are usually characterized
tain salmonellae, which may allow nose-nose transmis- by spread from pen to pen. Situations of spread from
sion. Aerolized secretions, feces, or contaminated dust pen to distant pen are likely because of vectors or care-
particles make the potential for aerosol transmission for taker transmission. When all animals sicken simultane-
short distances quite real. ously, a common source such as feed, bedding, water, or
Salmonella infection of swine herds is much more a contaminated environment should be suspected. Sal-
common than disease. Longitudinal Dutch studies sug- monella infections tend to be more prevalent in continu-
gest that about 25% of herds are never infected, 24% are ous-flow systems than in barns managed by principles of
constantly infected, and 50% are infected most of the all-in/all-out. Prevalence is also higher in barns with
time. There appear to be infection cycles, with the en- open flush-gutters than in those with slotted floors, with
demic salmonellae having an ecological advantage over the highest rates of infection observed in outdoor finish-
newly introduced salmonella serotypes. Infection occurs ing systems (Davies et al. 1997).
in the first weeks after arrival or commingling and reach- Numerous studies in a variety of host species with a
es a maximum of 80–100% prevalence within another potpourri of serotypes have demonstrated prolonged
2–3 weeks. About 5–30% of the pigs are still excreting carrier states following infection. The pattern of shed-
salmonellae at the end of the finishing period. In 1995, ding and the duration of the carrier state after clinically
the National Animal Health Monitoring Service apparent disease have been studied only in group-
(NAHMS) reported 30–60% of U.S. herds infected with housed pigs with no barrier to repeated reinfection
at least one serotype of Salmonella, with the greatest per- (Wilcock and Olander 1978; Wood et al. 1989). After ex-
centage of postive herds found in the southeastern Unit- perimental infection, S. typhimurium was isolated from
ed States and in herds marketing greater than 10,000 feces daily during the first 10 days postinfection and fre-
head annually. quently during the next 4–5 months. When slaughtered
During acute disease, pigs will shed up to 106 S. 4–7 months after initial infection, over 90% of pigs were
choleraesuis/g feces (Smith and Jones 1967) or 107 S. ty- positive for S. typhimurium in the mesenteric lymph
phimurium (Gutzmann et al. 1976). The minimum dis- node, tonsil, cecum, or feces (Wilcock and Olander 1978;
ease-producing dose of either serotype has not been es- Wood et al. 1989; Wood and Rose 1992; Fedorka-Cray et
540 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

al. 1994). S. newport has been shown to persist in mesen- changes in normal flora or cold-induced alteration in in-
teric lymph nodes for 28 weeks. Infection of individual testinal motility may reduce the amount of replication
animals may be relatively short-lived (less than 8 weeks), required for disease or increase the ease of salmonella
but organisms may circulate within a population and be- replication (Bohnhoff et al. 1954). Infection with S.
tween pigs and the environment for extended periods of choleraesuis may not require such massive luminal prolif-
time. eration as prerequisite for disease, because it is inherent-
S. choleraesuis given by either the intranasal or the in- ly more invasive than other serotypes, can infect via the
tragastric route has been demonstrated to persist within pharyngeal tonsil, and regularly causes signs of sep-
ileocolic junction, lymph nodes, tonsils, lungs, and colon ticemia 24–72 hours before the onset of diarrhea (Smith
for at least 12 weeks (Gray et al. 1995). Infection with low and Jones 1967; Cherubin et al. 1974; Wilcock 1979;
doses of S. choleraesuis produced a shorter period of in- Reed et al. 1986).
fection than infection with moderate or high doses. S. Most pathogenic S. typhimurium organisms have type
choleraesuis has been shown to persist for at least 3 1 fimbriae and flagella, both of which appear to be in-
months in wet feces and 6 months in desiccated feces. volved in attachment and invasion. Phenotypic shifts
The influence of antibiotics on the frequency and du- may occur in salmonellae to produce adhesive pili (Isaac-
ration of shedding of salmonellae in pigs has received lit- son 1996). Flagella may also be important in allowing
tle attention. In human enteric salmonellosis, the use of survival inside macrophages. The ability to invade is a re-
antibiotics has long been recognized to prolong the car- quirement for pathogenesis and is encoded by a
rier state (Dixon 1965; Aserkoff and Bennett 1969). In serotype-specific plasmid (Helmuth et al. 1985). Re-
pigs with enterocolitis, antibiotics do not reduce the du- moval of this plasmid results in a lack of ability to invade
ration or the magnitude of fecal shedding, but neither but has no effect on ingestion or killing by murine
are they reported to prolong or intensify shedding (Fin- macrophages, LPS production, or serum resistance
layson and Barnum 1973; DeGeeter et al. 1976; Gutz- (Gulig and Curtiss 1987). During the invasion process
mann et al. 1976; Wilcock and Olander 1978; Jones et al. there is induction of synthesis of new proteins that prob-
1983; Jacks et al. 1988). In contrast, vigorous antibacter- ably enhance intracellular survival (Finlay et al. 1989).
ial therapy early in the course of septicemia caused by S. Peroxidase-antiperoxidase immunoenzymatic labeling
choleraesuis may significantly reduce the magnitude and and immunogold labeling techniques have demonstrat-
duration of fecal shedding (Jacks et al. 1981). ed that S. typhimurium has a low tendency to invade the
enteric mucosa and does not have a predilection for any
PATHOGENESIS specific intestinal location, whereas S. choleraesuis locates
preferentially in the colon on the luminal surface of ileal
The clinical and pathological features of salmonella in- M cells of Peyer’s patches (Pospischil et al. 1990). Inva-
fections are extremely variable. Severity is influenced by sion is by endocytosis by M cells of gut-associated lym-
serotype, virulence, natural and acquired host resistance, phoid tissue as well as enterocytes. Attachment of the
and route and quantity of the infective dose. Over 200 bacteria to epithelial receptors triggers microfilament-
virulence factors have been associated with salmonellae controlled uptake, vacuole formation, vacuole transport
but few have been completely characterized. Generally, through the cell cytoplasm, and entry to the lamina pro-
those that promote virulence in pathogenic salmonellae pria via exocytosis through the basement membrane
are involved in adhesion, invasion, cytotoxicity, and re- (Takeuchi 1967; Takeuchi and Sprinz 1967). Passage
sistance to intracellular killing, often working in combi- through the epithelium results in mild and transient en-
nation to promote disease. Despite distinct differences in terocyte damage. Salmonellae can synthesize over 30
clinical signs, many parallels can be drawn between S. proteins which are selectively induced during infection
choleraesuis and S. typhimurium when discussing patho- of macrophages, making them facultative intracellular
genesis. bacteria (similar to Brucella, Mycobacteria, and Listeria
Although large doses (greater than 107) are required organisms) that can survive within macrophages and
to induce disease experimentally, intraluminal replica- neutrophils in the lamina propria (Roof et al. 1992a, b).
tion may be important with small inocula from contami- Spread to mesenteric lymph nodes is rapid, occurring
nated feed or water. Disease is facilitated by factors such within 2 hours of inoculation of ligated intestinal loops
as peristaltic impairment, interference with intestinal or 24 hours after oral challenge (Reed et al. 1985, 1986).
flora, and elevation of gastric pH (Clarke and Gyles Macrophages are the most likely disseminators of infec-
1993). Replication to about 107 organisms/g of intestinal tion systemically to other sites of infection and patholo-
content is required for lesion production in pigs infected gy. Concurrent with bacillary spread is the appearance of
with S. typhimurium, a finding that probably also applies an acute, predominantly macrophagic, inflammatory re-
to other serotypes causing enterocolitis. Alterations in action and prominent microvascular damage with
normal intestinal defenses by antibiotic-induced thrombosis within the lamina propria and submucosa.
CHAPTER 39 SALMONELLOSIS Schwartz 541

When administered intranasally to esophagatomized Endotoxins have either direct effects on tissue or effects
pigs, S. choleraesuis demonstrated primary colonization via an array of cytokine mediators.
of the lung within 4 hours (Fedorka-Cray et al. 1995;
Gray et al. 1995). CLINICAL SIGNS, PATHOLOGICAL
The pathogenesis of the diarrhea typical of enteric FINDINGS, AND DIAGNOSIS
salmonellosis and of later stages of salmonella sep-
ticemia has traditionally been attributed to malabsorp- The clinical signs of porcine salmonellosis are referable
tion and net fluid leakage by a necrotic, inflamed bowel. to septicemia or to enterocolitis, and this section de-
Several studies using rabbits, monkeys, calves, or pigs scribes each syndrome separately. Pigs surviving acute
have demonstrated fluid secretion independent of mu- septicemia may develop clinical signs due to bacteremic
cosal necrosis or inflammation (Giannella et al. 1973; localization: pneumonia, hepatitis, enterocolitis, and,
Rout et al. 1974; Kinsey et al. 1976; Clarke and Gyles occasionally, meningoencephalitis. Pigs initially suffer-
1987). These studies present evidence that, at least early ing from enterocolitis may later develop chronic wasting
in the disease, the diarrhea is the result of decreased disease or, occasionally, rectal stricture.
sodium resorption and increased chloride secretion due The most available salmonella diagnostic method is
to cholera-like and shiga-like enterotoxins. Secretion bacterial isolation and identification, which, along with
stimulated by prostaglandins elaborated by endotoxin- compatible lesions, remains the basis for diagnosis. Oth-
stimulated neutrophils may also be important (Stephen er tests using more sophisticated technology, including
et al. 1985). Most of the experimental work has been polymerase chain reaction (PCR), are not required for
done using small intestine of rabbits or monkeys, species routine diagnosis. PCR currently has value as a screening
in which the lesions of salmonellosis are quite different tool but has a relatively high cost and currently lacks sen-
from those in pigs. Toxic effects of certain OMPs, as well sitivity without preenrichment. Detection of salmonel-
as lipid A associated with the LPS, are also important me- lae does not constitute diagnosis of salmonellosis.
diators of cell damage. Survival within phagocytes is an Serology is becoming increasingly available, usually
important attribute of virulent salmonellae, the mecha- in the form of an ELISA test. Most tests use surface anti-
nism of which is not clear. Salmonellae which possess gens such as OMP or LPS. These tests, some of which use
smooth LPS, O side chains, and a complete LPS core are mixed antigens containing both OMP and LPS or anti-
more resistant to phagocyte killing. gens from several serotypes, thus far appear to lack speci-
Mucosal inflammation and necrosis, as well as sep- ficity and sensitivity for individual-animal diagnosis but
ticemia, occur in concert with the diarrhea but perhaps are useful for herd screening (Baum et al. 1996). A mixed-
independently of it. Microvascular thrombosis and en- ELISA test using meat juice at slaughter to detect anti-
dothelial necrosis in the submucosa and lamina propria body to a broad range of serotypes has been useful in cat-
are consistent early lesions in porcine salmonellosis egorizing the level of salmonella infection in herds in
(Lawson and Dow 1966; Wilcock et al. 1976; Jubb et al. Denmark (Nielsen et al. 1995; Mousing et al. 1997).
1993; Reed et al. 1986), probably in response to locally
produced endotoxin. Salmonellae are not directly associ- Septicemic Salmonellosis
ated with the damaged vessels but direct the events from This form of disease, usually caused by S. choleraesuis, oc-
the protected intracellular niche of macrophages in the curs mainly in weaned pigs less than 5 months of age but
surrounding submucosa or lamina propria (Takeuchi may be seen occasionally in market swine, suckling
and Sprinz 1967). Mucosal ischemia as a result of the mi- piglets, or adult breeding stock either as a septicemia or
crovascular thrombosis is probably a major contributor as a cause of abortion.
to the mucosal necrosis so typical of salmonellosis in all
species. The second major contribution to mucosal CLINICAL SIGNS. Pigs ill with S. choleraesuis are in-
necrosis is probably from the chemical products of mu- appetent, lethargic, febrile with temperatures of
cosal inflammation. The systemic signs and lesions of 105–107˚F (40.5–41.6˚C) and may have a shallow, moist
septicemic salmonellosis, in swine almost exclusively S. cough with slight expiratory dyspnea. Icterus may be ap-
choleraesuis infection, are most commonly attributed to parent. The first evidence of disease may be finding pigs
endotoxemia from bacterial dissemination. The complex reluctant to move, huddled in the corner of a pen, or
biology of endotoxin is beyond the scope of this chapter, even dead, with cyanosis of extremities and abdomens.
and readers should consult Wolff 1973, Elin and Wolff Diarrhea is not usually a feature of septicemic salmonel-
1976, or Cybulsky et al. 1988. Briefly, endotoxin interacts losis until the third or fourth day of disease, when watery
with plasma and with leukocytes to initiate inflamma- yellow feces may be seen. In most outbreaks, the case fa-
tion and fever. Most of the effects are mediated by inter- tality rate is high; morbidity is variable but is usually less
leukin-1, a lymphokine produced by macrophages stim- than 10%. Outbreaks are frequently associated with
ulated by the endotoxin (Rubin and Weinstein 1977). stressful situations. The duration of the disease in indi-
542 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

vidual pigs, as well as the duration and severity of each phoid nodules in the liver. These are clusters of histio-
epizootic, is unpredictable but will be prolonged without cytes amid foci of acute coagulative hepatocellular necro-
successful intervention. Evaluation of therapeutic regi- sis, corresponding to the white foci seen grossly. These
mens in naturally occurring outbreaks is difficult, mak- lesions are often present (Lawson and Dow 1966) and
ing response to therapy a poor diagnostic criterion. Dis- unique for this disease, although other agents can pro-
ease spread is by ingestion of contaminated feces or duce suppurative or necrotic foci in liver. Other lesions
nasopharyngeal secretions, with an incubation period typical of salmonellosis include fibrinoid thrombi in
ranging from 2 days to at least several weeks. Surviving venules of gastric mucosa, in cyanotic skin, in glomeru-
pigs may remain carriers and fecal shedders for at least lar capillaries, and less regularly in pulmonary vessels.
12 weeks. There is hyperplasia of reticular cells of spleen and
lymph nodes as well as generalized swelling of endothe-
GROSS LESIONS. Lesions at necropsy include cyano- lial cells and histiocytes typical of gram-negative sepsis.
sis of ears, feet, tail, and ventral abdominal skin; conges- A similar diffuse histiocytic interstitial pneumonia is pre-
tion progressing to infarction of gastric fundic mucosa; sent in lung. A complete discussion of the pathology of
splenomegaly with less severe hepatomegaly; and moist, septicemic salmonellosis can be found in Lawson and
swollen gastrohepatic and mesenteric lymph nodes. Dow 1966 and Jubb et al. 1993.
Lungs are firm and resilient, diffusely congested, often
with interlobular edema and perhaps hemorrhage; cran- DIAGNOSIS. The diagnosis of septicemic salmonel-
ioventral bronchopneumonia is not uncommon. Icterus losis cannot be made on the basis of clinical signs alone,
can be severe, although not consistently present (Fig. which are similar to those of other causes of septicemia
39.1). An inconsistent, subtle lesion is miliary, random in pigs, particularly Erysipelothrix rhusiopathiae, Strep-
white foci of necrosis in the liver. In pigs surviving the tococcus suis, Actinobacillus suis, or death due to classical
first few days of disease there may be serous to necrotic swine fever or Actinobacillus pleuropneumoniae. Gross le-
enterocolitis. The features of the intestinal lesion are de- sions of splenomegaly, hepatomegaly, lymphadenopa-
scribed more fully in the section on salmonella entero- thy, interstitial pneumonia, or focal hepatic necrosis are
colitis. Petechial hemorrhages, when present, are usually very suggestive of septicemic salmonellosis, but are not
best seen in the renal cortex or on the epicardium. seen in every case. In most situations, definitive diagno-
sis requires the isolation of large numbers of salmonel-
MICROSCOPIC LESIONS. The most diagnostic le- lae from tissues of affected pigs, almost invariably S.
sion of systemic salmonellosis is the presence of paraty- choleraesuis var. kunzendorf. Samples of lung, liver, or

39.1. Splenomegaly, hepatomegaly, and swollen mesenteric lymph nodes from S. choleraesuis
infection.
CHAPTER 39 SALMONELLOSIS Schwartz 543

spleen often yield pure cultures of the organism on bril- rarely with the profuseness typical of swine dysentery or
liant green, bismuth sulfite, blood agar, or MacConkey hemorrhagic porcine proliferative enteropathy (PPE).
agar. Enrichment techniques are seldom required unless Affected pigs are febrile, have decreased feed intake, and
the organs have been contaminated by feces or careless are dehydrated, paralleling the severity and duration of
handling or have autolysis, in which case tetrathionate the diarrhea. Mortality usually is low and occurs only af-
broth at 42–43˚C is the enrichment medium of choice. ter several days of diarrhea, presumably as the result of
Selenite broth is inhibitory for S. choleraesuis and should hypokalemia and dehydration. Most pigs make complete
be avoided (Edwards and Ewing 1972). Attempts to iso- clinical recovery but a portion may remain as carriers
late salmonellae from animals that have received antimi- and intermittent shedders for at least 5 months. A few
crobial therapy are often unrewarding. Intestine or feces pigs may remain unthrifty and, occasionally, may devel-
are not reliable specimens for isolation of the organism op rectal strictures.
in pigs with acute septicemia. Differential diagnosis
must include agents associated with the particular sys- GROSS LESIONS. In pigs that have died of diarrhea,
tems affected, including those that may cause sep- the major lesion is focal or diffuse necrotic enteritis, col-
ticemia, pneumonia, hepatitis, encephalitis, or entero- itis, or typhlitis. The lesion is seen as adherent gray-yel-
colitis (Schwartz 1991). low debris on the red, roughened mucosal surface of an
edematous spiral colon, cecum, or ileum (Fig. 39.2).
Salmonella Enterocolitis Colon and cecal contents are bile stained and scant, often
Salmonellosis manifested as enterocolitis is most fre- with black or sandlike gritty material. Mesenteric lymph
quent in pigs from weaning to about 4 months of age. nodes, especially ileocecal nodes, are greatly enlarged
Disease may be acute or chronic and can usually be as- and moist. The gross lesion may extend to involve the de-
cribed to S. typhimurium (including variety copenhagen) scending colon and rectum. The necrosis may be seen as
or, less frequently, to S. choleraesuis. Although isolation sharply delineated button ulcers, particularly in resolv-
of other serotypes of salmonellae from pigs with diar- ing lesions. Necrotic ileitis has historically been attrib-
rhea occurs with some frequency, implication of uted to several agents, including Salmonella, but in con-
serotypes other than S. choleraesuis, S. typhimurium, and firmed cases of salmonellosis, ileal involvement usually
perhaps S. heidelberg as primary pathogens should be is seen as reddening and slight roughening of the mu-
with caution. cosa, suggesting mild superficial necrosis. Salmonellae
associated with necrotic enteritis may be a sequel to PPE.
CLINICAL SIGNS. The initial clinical sign is watery yel- Lesions of septicemia may be present in those cases in-
low diarrhea, initially without blood or mucus. The dis- volving S. choleraesuis. In cases of S. typhimurium entero-
ease may spread rapidly to involve most pigs in a pen colitis, the liver and spleen are not enlarged except by
within a few days. The initial diarrhea in an individual terminal congestion.
pig usually lasts 3–7 days, but it typically may recur for
second and third bouts, giving the impression of a wax- MICROSCOPIC LESIONS. The typical enteric lesion
ing and waning diarrheal disease of several weeks’ dura- is necrosis of cryptal and surface enterocytes that varies
tion. Blood may appear sporadically in the feces but from focal to diffuse. The lamina propria and submucosa

39.2. Coalescing colonic ulcers


from S. typhimurium infection.
544 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

contain numerous macrophages and moderate numbers volves marked mesenteric lymphadenopathy. The lesion
of lymphocytes; neutrophils are numerous only in the of swine dysentery is diffuse, shallow, and restricted to
very early lesions. Thrombi containing fibrin, platelets, large intestine; lymph node enlargement is absent or
and leukocytes are numerous (Fig. 39.3). The necrosis mild. In PPE ileal involvement usually overshadows the
frequently extends to involve muscularis mucosa, sub- milder colonic lesions, and the mucosa underlying the
mucosa, and lymphoid follicles. Balantidium coli is com- necrotic membrane is markedly hyperplastic (Table
monly present in necrotic debris of chronic cases. In the 39.2). Whipworms (Trichuris suis) may also cause diffuse
ileum, necrosis is usually quite superficial and is often mucohemorrhagic colitis.
seen as villous atrophy. The Peyer’s patches may be The diagnosis of salmonellosis is confirmed by mi-
necrotic in acute disease, but in pigs dying of the natu- crobiological and histological examination. The wide dis-
rally occurring disease, lymphoid hypertrophy or even tribution of environmental salmonellae makes isolation
regenerative hyperplasia is more common. The liver may alone unreliable for disease diagnosis, and a positive iso-
contain paratyphoid nodules but not with the consisten- lation should always be supported by appropriate lesions
cy or the necrosis usually seen in the septicemic disease. before a diagnosis of salmonellosis is made. A pool of
A more complete discussion of the pathology can be ileum and ileocecal lymph node should enable detection
found in Wilcock et al. 1976, Reed et al. 1986, and Jubb of virtually all active or recently recovered cases, al-
et al. 1993. though tissues such as tonsil or cecal wall will usually
yield positive cultures as well (Wilcock et al. 1976; Wood
DIAGNOSIS. The differential diagnosis of diarrhea in et al. 1989). From live animals, large (10 g) aliquots of fe-
weaned pigs must include salmonellosis, swine dysen- ces or pharyngeal tonsil scrapings are preferable to rectal
tery, and PPE due to Lawsonia intracellularis. Other viral, swabs for isolation, with tetrathionate enrichment the
bacterial, or parasitic diseases capable of causing diar- method of choice.
rhea include rotaviral and coronaviral enteritis, post-
weaning colibacillosis, trichuriasis, and coccidiosis. Sal- Other Syndromes
monellosis concurrently present with other diseases is Salmonellae are occasionally involved in disease out-
not uncommon. breaks in which the clinical signs may not suggest salmo-
Typical acute swine dysentery is distinguished from nellae as the etiologic diagnosis. Outbreaks of neurolog-
salmonellosis on the basis of the mucoid and bloody di- ic disease resembling classical swine fever or
arrhea in otherwise alert swine with dysentery, contrast- pseudorabies have been reported (Wilcock and Olander
ed with depression and profuse yellow diarrhea of sal- 1978), and brain lesions sometimes occur with sep-
monellosis. PPE may be seen as acute intestinal ticemic salmonellosis. The lesion in the brain is necrotic
hemorrhage or acute to chronic diarrhea with mucosal vasculitis and perivascular granulomatous lesions resem-
proliferation or necrosis. Differentiation among the bling typical paratyphoid nodules (Fig. 39.4). Rectal
three diseases at necropsy is primarily by recognition of strictures in growing pigs have been ascribed to defective
differences in lesion distribution rather than by differ- healing of ulcerative proctitis caused by S. typhimurium
ences in character. Salmonellosis is usually in colon and (Wilcock and Olander 1977a, b). The stricture reported-
occasionally small intestine, may be focal, and always in- ly represents fibrosis in an area of persistent ischemia,

39.3. Histological section show-


ing deep colonic ulceration and
inflammation from S. typhimuri-
um infection.
CHAPTER 39 SALMONELLOSIS Schwartz 545

Table 39.2. Differential diagnosis of enterocolitis in swine at necropsy


Extraintestinal
Condition Ileal Lesion Colonic Lesion Ileocecal Nodes Lesions
Salmonellosis Mild, usually no Focal to diffuse, Always enlarged Variable, gastric
pseudomembrane deep necrotic two to five infarction,
lesions times normal interstitial
pneumonitis,
miliary hepatic
necrosis
Swine dysentery Absent Superficial and Often normal, None except
usually diffuse slight gastric fundic
necrosis, blood enlargement infarction in
and/or mucus natural deaths
Porcine proliferative Varies from Milder than in the Variable with None
enteritis hemorrhagic to ileum, usually stage of disease
necrotic or only the
proliferative proximal spiral
colon

with the rectum predisposed because of a normally pre- TREATMENT


carious blood supply (Fig. 39.5).
Infection with the fastidious, swine-adapted serovar With either septicemic or enteric salmonellosis, the goals
S. typhisuis causes a relatively specific chronic syndrome of treatment in an outbreak of salmonellosis are to min-
of diarrhea and wasting in which caseous lymphadenitis, imize the severity of clinical disease, prevent spread of
histiocytic interstitial pneumonia, or suppurative bron- infection and disease, and prevent recurrence of the dis-
chopneumonia is added to the typical necrotic colitis ease in the herd. With salmonellosis the attainment of
(Barnes and Bergeland 1968; Andrews 1976; Fenwick these goals is particularly difficult. Both S. choleraesuis
and Olander 1987). In some pigs the intestinal lesions and S. typhimurium are often resistant in vitro to many
may have healed, leaving the lymphoid and pulmonary antibacterial agents used in swine (Barnes and Sorensen
lesions to be distinguished from tuberculosis and infec- 1975; Wilcock et al. 1976; Blackburn et al. 1984; Schultz
tion with Arcanobacterium pyogenes (Barnes and Sorensen 1989; Fales et al. 1990; Schwartz 1997a). During clinical
1975). disease, the organism inhabits a protected intracellular

39.4. Histological section show-


ing vasculitis and perivascular
granulomatous inflammation in
the brain stem from S. cholerae-
suis encephalitis.
546 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

39.5. Rectal stricture at


necropsy.

niche inaccessible to many common antibacterials. The 1991). Mass medication of the population at risk to de-
use of various antibiotics to treat enteric salmonellosis is crease severity of disease and transmission of salmonel-
widely advocated (Morehouse 1972; Barnes and lae is also widely practiced. The choice of an appropriate
Sorensen 1975; Blood et al. 1979), but much of the in- antimicrobial is aided by antibiograms and previous
formation to support this recommendation has been tak- herd experience. In the absence of either, amikacin, gen-
en from trials designed to test the prophylactic efficacy of tamicin, neomycin, apramycin, ceftiofur, and trimetho-
drugs, not their therapeutic efficacy. Thus, pigs on med- prim-sulfonamide are effective in vitro against most iso-
icated feed, when inoculated orally with salmonellae, lates (Barnes and Sorensen 1975; Wilcock et al. 1976;
have the antibiotic already present in the gastrointestinal Mills and Kelly 1986; Schultz 1989; Evelsizer 1990; Fales
tract to interact with the salmonellae, resulting in milder et al. 1990, Schwartz 1997b). Antiinflammatory agents
disease because of what amounts to a decreased inocu- are sometimes administered to critically ill animals to
lum. In the few trials designed specifically to test an- combat the effects of endotoxin (Schwartz and Daniels
tibacterial drug efficacy against clinical enteric salmonel- 1987; Schultz 1989; Evelsizer 1990).
losis, such therapy was considered to have little merit Most salmonella antimicrobial resistance is plasmid-
(Heard et al. 1968; Gutzmann et al. 1976; Olson et al. mediated, and removal of selective pressure of an an-
1977; Wilcock and Olander 1978). Although not thera- timicrobial allows for reversion to susceptibility. Of con-
peutic, oral medications may decrease efficiency of cern is the recent emergence of a S. typhimurium phage
transmission and have a prophylactic effect on pigs not type or definitive type 104 (DT 104), isolated primarily
yet affected. Antimicrobials are ordinarily administered from bovine and human populations, that has chromo-
at maximum permissible levels in feed or, preferably, wa- somally integrated multiple antimicrobial resistance
ter. Ideally, the choice of antibacterial agent should be (Low et al. 1997). This isolate has a higher morbidity and
based on in vitro susceptibility testing of isolates from mortality in humans than other S. typhimurium organ-
each outbreak. Since medication often must be initiated isms and is increasing in prevalence in human and
before such results are available, choices must be based bovine populations. Carrier swine are generally asymp-
on previous experience and results of controlled trials. tomatic. Although evidence linking emergence of such
In contrast, vigorous therapy early in the course of isolates to veterinary medical practices is generally lack-
septicemia caused by S. choleraesuis has been reported to ing, the impact will affect public health initiatives as well
significantly reduce the duration and severity of disease as the availability of therapeutic agents for food-produc-
(Jacks et al. 1981). In that report, therapy was initiated ing animals. It should be emphasized that salmonella
after inoculation but prior to the onset of clinical signs. control programs that rely strictly on antimicrobials are
Evaluation of efficacy under field conditions is difficult doomed to failure.
because of the unpredictability of the disease and be- In addition to antimicrobial therapy, the successful
cause husbandry changes often accompany the use of treatment of salmonellosis relies heavily on routine hus-
antibacterials in an outbreak. Reports and practitioner bandry procedures recommended for control of infec-
communications from the American Midwest, however, tious diseases. The diarrheic pig massively contaminates
suggest that visibly affected animals respond to aggres- its environment and is the single most important source
sive therapy with parenteral antimicrobials (Schwartz of infection for other pigs. Removal and isolation of sick
CHAPTER 39 SALMONELLOSIS Schwartz 547

animals, minimizing exposure to infective material by torically, an attenuated live S. choleraesuis vaccine was
scrupulous pen sanitation, frequent cleaning of water used widely in the United Kingdom for many years but
bowls, and restriction of animal or staff movement from was withdrawn when S. choleraesuis infection decreased
potentially contaminated to clean areas are necessary. Ef- in that country to negligible proportions. Recently, the
forts to modify management and environment to de- introduction of effective and safe modified live attenuat-
crease stress and increase pig comfort are essential ad- ed vaccines for S. choleraesuis has had a major impact on
juncts to specific therapy. the occurrence of systemic salmonellosis in North Amer-
ica. The isolates used in these vaccines are either natural-
PREVENTION ly occurring avirulent S. choleraesuis or are derived from
repeated passage through porcine neutrophils, the prod-
Prevention of infection of swine with salmonellae is not uct of which was demonstrated to have been cured of a
currently possible. Infection does not necessarily result 50 kb virulence plasmid necessary for intracellular sur-
in disease, and pigs may not sicken with disease until se- vival (Roof et al. 1992b; Kramer et al. 1987, 1992). When
verely stressed long after initial exposure. The control of given at weaning, vaccine protected pigs for at least 20
disease expression rests on efforts to minimize the expo- weeks (Roof and Doitchinoff 1995) against homologous
sure dose and to maximize pig resistance. The carrier pig serotypes, with some cross-protection suggested with
and contaminated feed or environment are the most sig- heterologous serotypes.
nificant sources of infection to pigs, and pigs are most Partial protection can be obtained with bacterins,
likely to develop disease during periods of stress or when primarily because of the nonspecific mitogenic and im-
exposed to massive numbers of salmonellae. The com- munostimulant effect of LPS (Fenwick et al. 1986). Killed
mingling and transport of weanling pigs from different vaccines for S. typhimurium are safe, but the bulk of the
sources to finishing farms enhance activation of latent evidence suggests that they have little efficacy in prevent-
carriers and ensure exposure of stressed pigs to salmo- ing disease following strong challenge because resistance
nellae (Allred 1972). The source of host-adapted S. to disease rests primarily on cell-mediated immunity
choleraesuis, which is rarely, if ever, isolated from feed or (Collins 1974; Davies and Kotlarski 1976). Extrapolation
feed ingredients, would seem to be limited to carrier pigs of information from experience in humans (Hornick et
and facilities previously contaminated with this al. 1970; Welliver and Ogra 1978) and calves (Bairey
serotype. The fact that many outbreaks occur in facilities 1978) suggests, however, that use of a potent killed vac-
with good sanitation suggests that other stresses proba- cine may increase the dose necessary to cause disease and
bly contribute to occurrence of the disease. Management may offer some protection from septicemic salmonel-
practices that allow filling of grower and finishing rooms losis, in which humoral immunity may play a role.
with single-source and single-age pigs are beneficial. Monitoring herds for salmonellae has not been com-
Minimizing the variety of stresses often involved in acute monly practiced. The detection of carrier animals is dif-
outbreaks requires constant attention to details of man- ficult because of the unpredictability of fecal shedding.
agement and husbandry, including proper animal densi- The detection of salmonellae by bacterial culture of feces
ty; dry, comfortable pens and temperatures; and ade- and tonsils of diarrheic pigs in the nursery would likely
quate ventilation. On farms with enzootic disease, be the most rewarding for identification of infected
modifications to the facility and environment and imple- herds. However, even repeated negative fecal or tonsilar
mentation of management practices that emphasize all- cultures do not guarantee that a herd or individual is not
in/all-out production should precede a prophylactic a salmonella carrier and thus a potential shedder. The
drug program. Antibiotics are probably useful as aids in use of salmonella serology can determine if an animal
preventing occurrence of disease when used prophylacti- has had previous exposure to salmonellae, but this has
cally, but their use will not prevent infection and when little relevance to the carrier status or to the probability
relied upon for prevention of disease will eventually fail. of shedding. Food safety concerns have stimulated re-
Nutritional approaches to prevent or alleviate swine newed interest in serology as a method to determine the
salmonellosis include feeding of propionic or other salmonella infection status of groups of market swine.
volatile fatty acids, mannose, lactose, probiotics, and This technology offers the possibility of sensitive and
heavy metals. Although all of these practices are based specific methods to identify infected herds, but it is not
on a sound theoretical basis and are reported to be suc- yet useful for determining the infection status of individ-
cessful experimentally or in other species, evidence for ual pigs.
their efficacy in swine is lacking. Anecdotal reports sug-
gest acidification of rations or water to be of some bene- REFERENCES
fit.
As with other facultative intracellular bacteria, live Allred, J. N. 1972. Comments on salmonellosis in swine. J
vaccines that stimulate cell-mediated immunity are the Am Vet Med Assoc 160:601–602.
most likely to be protective for salmonellae in swine. His- Allred, J. N.; Walker, J. W.; Beal, V. C.; and Germaine, F. W.
548 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

1967. A survey to determine the salmonella contamina- Clarke, R. C., and Gyles, C. L. 1987. Virulence of wild and
tion rate in livestock and poultry feeds. J Am Vet Med mutant strains of Salmonella typhimurium in ligated in-
Assoc 151:1857–1860. testinal segments of calves, pigs, and rabbits. Am J Vet
Andrews, J. J. 1976. Salmonella typhisuis infection in swine. Res 48:504–510.
In Proc North Cent Conf Vet Lab Diagn, p. 7. ———. 1993. Salmonella. In Pathogenesis of Bacterial In-
Anon. 1994. Annual Report on Zoonoses in Denmark. fections in Animals, 2d ed. Ames: Iowa State Univ Press,
Copenhagen: Ministry of Agriculture and Fisheries, pp. 133–153.
Danish Veterinary Service, Danish Zoonosis Centre. Collins, F. M. 1974. Vaccines and cell-mediated immunity.
Aserkoff, B., and Bennett, J. V. 1969. Effect of antibiotic Bacteriol Rev 38:371–402.
therapy in acute salmonellosis on the fecal excretion of Committee on Salmonella. 1969. An evaluation of the sal-
salmonellae. N Engl J Med 281:636–640. monella problem. Washington, D.C.: Nat Acad Sci.
Bairey, M. H. 1978. Immunization of calves against salmo- Craven, J. A., and Hurst, D. B. 1982. The effect of time in
nellosis. J Am Vet Med Assoc 173:610–613. lairage on the frequency of salmonella infection in
Banks, J. G., and Board, R. G. 1983. The incidence and level slaughtered pigs. J Hyg (Camb) 88:107–111.
of contamination of British fresh sausages and ingredi- Cybulsky, M. I.; Chan, M. K. W.; and Movat, H. Z. 1988. Bi-
ents with salmonellas. J Hyg (Lond) 90:213–223. ology of disease: Acute inflammation and microthrom-
Barnes, D. M., and Bergeland, M. E. 1968. Salmonella ty- bosis induced by endotoxin, interleukin-1, and tumor
phisuis infection in Minnesota swine. J Am Vet Med As- necrosis factor and their implication in gram-negative
soc 152:1766–1769. infection. Lab Invest 58:365–378.
Barnes, D. M., and Sorensen, D. K. 1975. Salmonellosis. In Davies, P. R; Morrow, W. E. M.; Jones, F. T.; Deen, J.; Fedor-
Diseases of Swine, 4th ed. Ed. H. W. Dunne and A. D. ka-Cray, P. J.; and Gray, J. T. 1997. Risk of shedding Sal-
Leman. Ames: Iowa State Univ Press, pp. 554–564. monella organisms by market-age hogs in a barn with
Baskerville, A., and Dow, C. 1973. Pathology of experimen- open-flush gutters. J Am Vet Med Assoc 210:386–389.
tal pneumonia in pigs produced by Salmonella cholerae- Davies, R., and Kotlarski, I. 1976. The role for antibody in
suis. J Comp Pathol 83:207–215. the expression of cellular immunity to Salmonella ty-
Baum, D. H.; Harris, D. L.; Nielsen, B.; Fedorka-Cray, P. J.; phimurium C5. Aust J Exp Biol Med Sci 54:207–219.
Homan, W. H.; and Rankin, R. A. 1996. Comparison of Dawe, D. L., and Troutt, H. F. 1976. Treatment of experi-
the Danish MIX-ELISA to culture for the detection of mentally induced salmonellosis in weanling pigs with
Salmonella in finishing pigs at or near slaughter. In ISU trimethoprim and sulfadiazine. Proc Int Congr Pig Vet
Swine Res Rep, p. 177. Soc 4:M4.
Bixler, W. B. 1978. FDA salmonella control activities for an- Degeeter, M. H.; Stahl, G. L.; and Geng, S. 1976. Effect of
imal feeds and feed ingredients. In Proc Nat Salmonel- lincomycin on prevalence, duration and quantity of Sal-
losis Sem, Washington, D.C. monella typhimurium excreted by swine. Am J Vet Res
Blackburn, B. O.; Schlater, L. K.; and Swanson, M. R. 1984. 37:525–529.
Antibiotic resistance of members of the genus Salmonel- Dixon, J. M. S. 1965. Effect of antibiotic treatment on dura-
la isolated from chickens, turkeys, cattle, and swine in tion of excretion of Salmonella typhimurium by children.
the United States during October 1981 through Septem- Br Med J 2:1343–1345.
ber 1982. Am J Vet Res 45:1245–1249. Edwards, P. R., and Ewing, W. H. 1972. Identification of En-
Blood, D. C.; Henderson, J. A.; and Radostits, O. M. 1979. terobacteriaceae, 3d ed. Minneapolis: Burgess, pp.
Veterinary Medicine, 4th ed. London: Baillière Tindall, 146–207.
pp. 476–486. Elin, R. J., and Wolff, S. M. 1976. Biology of endotoxin. Ann
Bohnhoff, M.; Drake, B. L.; and Miller, C. P. 1954. Effect of Rev Med 27:127–141.
streptomycin on susceptibility of intestinal tract to sal- Evelsizer, R. 1990. Salmonellosis in Grow-Finish pigs. Min-
monella infection. Proc Soc Exp Biol Med 86:133–137. nesota Swine Conf Vet, St. Paul.
Carr, M. A.; Bawcom, D. W.; Miller, M. F.; Ramsey, C. B.; Fales, W. H.; Maddox, C. W.; and Oakman, J. K. 1990. An-
and Thompson, L. D. 1996. Microbiology of Pig Car- timicrobial susceptibility of Salmonella choleraesuis from
casses. 1st Int Symp Ecology of Salmonella in Pork Prod- Missouri swine. In 33d Annu Meet AAVLD, Denver, Co-
ucts, NADC, Ames, Iowa. lo.
Centers for Disease Control and Prevention (CDC) Annual Fedorka-Cray, P. J.; Whipp, S. C.; Isaacson, R. E.; Nord, N.;
Summary, Salmonella Surveillance. 1996. and Lager, K. 1994. Transmission of Salmonella ty-
Cherubin, C. E. 1980. Epidemiologic assessment of antibi- phimurium to swine. Vet Micro 41:333–344.
otic resistance in salmonella. In CRC Handbook Series in Fedorka-Cray, P. J.; Kelley, L. C.; Stabel, T. J.; Gray, J. T.; and
Zoonosis, I Sect D. Ed. J. H. Steele, pp. 173–200. Laufer, J. A. 1995. Alternate routes of invasion may af-
Cherubin, C. E.; Neu, H. C.; Imperato, P. J.; Harvey, R. P.; fect pathogenesis of Salmonella typhimurium. Infect Im-
and Bellen, N. 1974. Septicemia with nontyphoid sal- mun 63:2658–2664.
monella. Medicine 53:365–376. Fenwick, B. S.; Cullor, J. S.; Osburn, B. I.; and Olander, H. J.
CHAPTER 39 SALMONELLOSIS Schwartz 549

1986. Mechanisms involved in protection provided by occurrence and persistence of Salmonella typhimurium in
immunization against core lipopolysaccharides of E. experimentally infected swine. Am J Vet Res
coli. J strain from lethal Haemophilus pleuropneumonia in 37:649–655.
swine. Infect Immun 53:296–302. Hansen, R.; Rogers, R.; Emge, S.; and Jacobs, N. J. 1964. In-
Fenwick, B. W., and Olander, H. J. 1987. Experimental infec- cidence of Salmonella in the hog colon as affected by
tion of weanling pigs with Salmonella typhisuis: Effect of handling practices prior to slaughter. J Am Vet Med As-
feeding low concentrations of chlortetracycline, peni- soc 145:139–140.
cillin, and sulfamethazine. Am J Vet Res 48:1568–1573. Harris, I. T.; Fedorka-Cray, P. J.; Gray, J. T.; Thomas, L. A.;
Ferris, K., and Frerichs, W. 1996. Summary of Salmonella and Ferris, K. 1997. Prevalence of Salmonella organism
Isolated from Swine. Ames, Iowa: Nat Vet Services Labo- in swine feed. J Am Vet Med Assoc 210:382–385.
ratories. Heard, T. W.; Linton, A. H.; Penny, R. H. C.; and Wilson, M.
Finlay, B. B.; Heffron, F.; and Falkow, S. 1989. Epithelial cell R. 1965. Salmonella typhimurium infection in a hysterec-
surfaces induce Salmonella proteins required for bacter- tomy-produced herd of pigs. Vet Rec 77:1276–1279.
ial adherence and invasion. Science 243:940–943. Heard, T. W.; Jennett, N. E.; and Linton, A. H. 1968. The
Finlayson, M., and Barnum, D. A. 1973. The effect of control and eradication of salmonellosis in a closed pig
chlortetracycline feed additive on experimental salmo- herd. Vet Rec 82:92–99.
nella infection of swine and antibiotic resistance trans- Helmuth, R., and Bunge, C., et al. 1985. Epidemiology of
fer. Can J Comp Med 37:139–146. virulence-associated plasmids and outer membrane pro-
Fukushima, H.; Hoshina, K.; Nakamura, R.; and Ito, Y. 1987. tein patterns within several common Salmonella
Raw beef, pork and chicken in Japan contaminated with serotypes. Infect Immun 48:175–182.
Salmonella sp., Campylobacter sp., Yersinia enterocolitica Henry, D. P.; Frost, A. J.; Samuel, J. L.; O’Boyle, D. A.; and
and Clostridium perfringens: A comparative study. Zen- Thomson, R. H. 1983. Factors affecting the survival of
tralbl Bakteriol Mikrobiol Hyg 184:60–70. Salmonella and Escherichia coli in anaerobically ferment-
Galton, M. M.; Smith, W. V.; McElrath, H. B.; and Hardy, A. ed pig waste. J Appl Bacteriol 55:89–95.
B. 1954. Salmonella in swine, cattle, and the environ- Hooper, B. E., and Troutt, H. F. 1971. Swine salmonellosis.
ment of abattoirs. J Infect Dis 95:236–245. In Proc Carbadox: Synthetic Antibacterial Agent,
Giannella, R. A.; Formal, S. B.; Dammin, G. J.; and Collins, Kansas City, pp. 83–101.
H. 1973. Pathogenesis of salmonellosis: Studies of fluid Hornick, R. B.; Griesman, S. E.; and Woodward, T. E. 1970.
secretion, mucosal invasion, and morphologic reaction Typhoid fever: Pathogenesis and immunologic control.
of the rabbit ileum. J Clin Invest 52:441–453. N Engl J Med 283:735–746.
Gooch, J. M., and Goo, V. 1971. Frequency of Salmonella iso- Hsu, F. S.; Chueck, L. L.; and Shen, Y. M. 1983. Isolation,
lation in selected foods of animal origin, Hawaii: The serotyping and drug resistance of salmonellae in scour-
red meat processor’s role in salmonella prevention. In ing pigs in Taiwan. Chung Hua Min Kuo Wei Sheng Wu
Proc Nat Salmonellosis Sem, Washington, D.C. Chi Mien I Hsueh Tsa Chih 16:283–290.
Gooch, J. M., and Haddock, R. L. 1969. Swine salmonellosis Isaacson, R. E. 1996. Persistent colonization of pigs by Sal-
in a Hawaiian piggery. J Am Vet Med Assoc monella typhimurium and effects of transportation relat-
154:1051–1054. ed stress on the shedding of Salmonella typhimurium. In
Gray, J. T.; Fedorka-Cray, P. J.; Stabel, T. J.; and Ackermann, 1st Int Symp Ecology of Salmonella in Pork Products,
M. R. 1995. Influence of inoculation route on the carrier NADC, Ames, Iowa.
state of Salmonella choleraesuis in swine. Vet Micro Jacks, T. M.; Welter, C. J.; Fitzgerald, G. R.; and Miller, B. M.
47:43–59. 1981. Cephamycin C treatment of induced swine salmo-
Gray, J. T.; Stabel, T. J.; and Fedorka-Cray, P. J. 1996. Effect nellosis. Antimicrob Agents Chemother 19:562–566.
of dose on the immune response and persistence of Sal- Jacks, T. M.; Frazier, E.; Judith, F. R.; and Olson, G. 1988. Ef-
monella choleraesuis in swine. Am J Vet Res 57:313– fect of efrotomycin in feed on the quantity, duration,
319. and prevalence of shedding and antibacterial suscepti-
Groves, B. I.; Fish, N. A.; and Mitchell, W. R. 1971. The oc- bility of Salmonella typhimurium in experimentally in-
currence of salmonella infection in market swine in On- fected swine. Am J Vet Res 49:1832–1835.
tario. Can Vet J 12:11–15. Jayarao, B. M.; Biro, G.; Kovacs, S.; Domjan, H.; and Fabian,
Gulig, P. A., and Curtiss, R. 1987. Plasmid associated viru- A. 1989. Prevalence of Salmonella serotypes in pigs and
lence of Salmonella typhimurium. Infect Immun evaluation of a rapid, presumptive test for detection of
55:2891–2901. Salmonella in pig faeces. Acta Vet Hung 37:39–44.
Gustafson, R. H.; Kobland, J. D.; and Langner, P. H. 1976. Jones, F. T.; Langlois, B. E.; Cromwell, G. L.; and Hays, V. W.
Incidence and antibiotic resistance of salmonella in 1983. Effect of feeding chlortetracycline or virgini-
market swine. Proc Int Congr Pig Vet Soc 4:M2. amycin on shedding of salmonellae from experimental-
Gutzmann, F.; Layton, H.; Simiins, K.; and Jarolmen, H. ly infected swine. J Anim Sci 57:279–285.
1976. Influence of antibiotic-supplemented feed on the Jubb, K. V. F.; Kennedy, P. C.; and Palmer, N. C. 1993.
550 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Pathology of Domestic Animals. San Diego: Academic Salmonella enterica surveillance and control in Danish
Press. slaughter swine herds. Prev Vet Med 29:247–261.
Kampelmacher, E. H.; Guinee, P. A. M.; and Van Keulen, A. Nape, W. F., and Murphy, C. 1971. Recovery of salmonellae
1965. Prevalence of salmonellosis in pigs fed decontam- in feed mills, using terminally heated and regularly
inated and normal feeds. Zentralbl Veterinärmed (B) processed animal protein. J Am Vet Med Assoc
12:258–267. 159:1569–1572.
Kinsey, M. D.; Dammin, G. J.; Formal, S. B.; and Giannella, Nielsen, B.; Baggesen, D.; Bager, F.; Haugegaard, J.; and
R. A. 1976. The role of altered intestinal permeability in Lind, P. 1995. The serological response to Salmonella
the pathogenesis of salmonella-diarrhea in the rhesus serovars typhimurium and infantis in experimentally in-
monkey. Gastroenterology 71:429–434. fected pigs: The time course followed with an indirect
Kramer, T. T.; Pardon, P.; Marly, J.; and Bernard, S. 1987. anti-LPS ELISA and bacteriological examinations. Vet
Conjunctival and intramuscular vaccination of pigs with Microbiol 47:205–218.
a live avirulent strain of Salmonella choleraesuis. Am J Vet Nottingham, P. M.; Penney, N.; and Wyborn, R. 1972. Sal-
Res 48:1072–1076. monella infection in calves and other animals. III. Fur-
Kramer, T. T.; Roof, M. B.; and Matheson, R. R. 1992. Safe- ther studies with pigs and calves. NZ J Agric Res
ty and efficacy of an attenuated strain of Salmonella 15:279–283.
choleraesuis for vaccination of swine. Am J Vet Res Olson, L. D.; Rodebaugh, D. E.; and Morehouse, L. G. 1977.
53:444–448. Comparison of furazolidone and carbadox in the feed
Lawson, G. H. K., and Dow, C. 1966. Porcine salmonellosis. for treatment of Salmonella choleraesuis in swine. Am J
J Comp Pathol 76:363–371. Vet Res 38:1471–1477.
Levine, N. D.; Peterson, E. H.; and Graham, R. 1945. Studies Pokorny, J. 1988. Survival and virulence of salmonellae in
on swine enteritis. II. Salmonella and other enteric or- water. J Hyg Epidemiol Microbiol Immunol 32:361–366.
ganisms isolated from diseased and normal swine. Am J Pospischil, A.; Wood, R. L.; and Anderson, T. D. 1990. Per-
Vet Res 6:241–246. oxidase-antiperoxidase and immunogold labeling of
Low, J. C.; Angus, M.; Hopkins, G.; Munro, D.; and Rankin, Salmonella typhimurium and Salmonella choleraesuis var
S. C. 1997. Antimicrobial resistance of Salmonella enter- kunzendorf in tissues of experimentally infected swine.
ica typhimurium DT104 isolates and investigation of Am J Vet Res 51:619–624.
strains with transferable apramycin resistance. Epid In- Reed, W. M.; Olander, H. J.; and Thacker, H. L. 1985. Stud-
fect 118:97–103. ies on the pathogenesis of Salmonella heidelberg infection
Lynn, M.; Dobson, A. W.; McClune, E. L.; and Dorn, C. R. in weanling pigs. Am J Vet Res 46:2300–2310.
1972. A study of Salmonella typhimurium infection in a ———. 1986. Studies on the pathogenesis of Salmonella ty-
swine-testing station. Vet Med Small Anim Clin phimurium and Salmonella choleraesuis var. kunzendorf in-
67:1022–1027. fection in weanling pigs. Am J Vet Res 47:75–83.
McCaughey, W. G.; McClelland, T. G.; and Roddy, R. M. Reynolds, I. M.; Miner, P. W.; and Smith, R. E. 1967. Salmo-
1973. Salmonella isolations in pigs. Vet Rec 92:191–194. nella enteritidis from porcine meningitis: A case report.
McErlean, B. A.1968. Salmonella dublin meningitis in Cornell Vet 58:180–185.
piglets. Vet Rec 82:257–258. Riley, M. G. I. 1970. The incidence of salmonellosis in nor-
Michaud, R. P. 1978. Contributory Sources of Salmonella in mal slaughtered pigs in Australia. Aust Vet J 46:40–43.
the Environment of Swine Slaughterhouses. M.S. thesis, Roberts, D.; Boag, K.; Hall, M. L. M.; and Shipp, C. R. 1975.
Univ Guelph. The isolation of salmonella from British pork sausage
Mills, K. W., and Kelly, B. L. 1986. Antibiotic susceptibilities and sausage meat. J Hyg (Camb) 75:173–184.
of swine Salmonella isolants from 1979 to 1983. Am J Vet Roof, M. B., and Doitchinoff, D. D. 1995. Safety, efficacy,
Res 47:2349–2350. and duration of immunity induced in swine by use of an
Mittermeyer, F. C., and Foltz, V. D. 1969. Salmonella survey avirulent live Salmonella choleraesuis–containing vac-
of plant foods used in and around the home. Appl Mi- cine. Am J Vet Res 56:39–44.
crobiol 18:682–683. Roof, M. B.; Kramer, T. T.; Roth, J. A.; and Minion, C. F.
Morehouse, L. G. 1972. Salmonellosis in swine and its con- 1992a. Characterization of a Salmonella choleraesuis iso-
trol. J Am Vet Med Assoc 160:594–601. late after repeated neutrophil exposure. Am J Vet Res
Morgan, I. R.; Krautil, F. L.; and Craven, J. A. 1987. Effect of 53:1328–1332.
time in lairage on caecal and carcass salmonella contam- Roof, M. B.; Kramer, T. T.; Kunesh, J. P.; and Roth, J. A.
ination of slaughter pigs. Epidemiol Infect 98:323–330. 1992b. In vivo isolation of Salmonella choleraesuis from
Morse, J. W., and Hird, D. W. 1984. Bacteria isolated from porcine neutrophils. Am J Vet Res 53:1333–1346.
lymph nodes of California slaughter swine. Am J Vet Res Rout, W. R.; Formal, S. B.; Giannella, R. A.; and Dammin, G.
45:1648–1649. J. 1974. Pathophysiology of Salmonella diarrhea in the
Mousing, J.; Thode Jensen, P.; Bager, F.; Feld, N.; Nielsen, B.; rhesus monkey: Intestinal transport, morphologic and
Nielsen, J. P.; and Bech-Nielsen, S. 1997. Nation-wide bacteriological studies. Gastroenterology 67:59–70.
CHAPTER 39 SALMONELLOSIS Schwartz 551

Rubin, R. H., and Weinstein, L. 1977. Salmonellosis. New dures based upon Rappaport-Vassiliadis medium with
York: Stratton Intercontinental Medical Book Corp., p. those using Muller-Kauffmann medium containing
25. Teepol for the isolation of Salmonella sp. Epidemiol In-
Salmon, D. E., and Smith , T. 1886. The bacterium of swine fect 99:143–147.
plague. Am Mon Microbiol J 7:204. Welliver, R. C., and Ogra, P. L. 1978. Importance of local im-
Schultz, R. A. 1989. Salmonellosis-The Problem-How Do munity in enteric infection. J Am Vet Med Assoc
We Handle It? Proc Am Assoc Swine Prod, Des Moines, 173:560–564.
Iowa. Wilcock, B. P. 1978. Experimental Klebsiella and Salmonella
Schwartz, K. J. 1991. Salmonellosis in swine. Compend Con- infection in neonatal swine. Can J Comp Med
tin Educ 13(1):139–148. 43:100–106.
———. 1997a. A summary of 1994–1996 Annual Reports ———. 1979. Serotype-associated virulence factors in
of the Iowa State University Veterinary Diagnostic Lab- porcine salmonellosis. Conf Res Workers, Anim Dis Res
oratory, Ames, Iowa. Inst, Ottawa.
———. 1997b. Salmonellosis. Proc AASP, Quebec City, Wilcock, B. P., and Olander, H. J. 1977a. The pathogenesis
Quebec. of porcine rectal stricture. I. Observations on the natu-
Schwartz, K. J., and Daniels, G. 1987. Salmonellosis. Proc rally occurring disease and its association with salmo-
Minnesota Swine Herd Health Prog Conf, Univ Min- nellosis. Vet Pathol 14:36–42.
nesota. ———. 1977b. The pathogenesis of porcine rectal stric-
Silas, J. C.; Carpenter, J. A.; and Reagan, J. O. 1984. Update: ture. II. Experimental salmonellosis and rectal stricture.
Prevalence of Salmonella in pork sausage. J Anim Sci Vet Pathol 14:43–55.
59:122–124. ———. 1977c. Neurologic disease in naturally occurring
Skovgaard, N.; Christensen, S. G.; and Culistani, A. W. Salmonella choleraesuis infection in pigs. Vet Pathol
1985. Salmonellas in Danish pigs: A comparison of 14:113–120.
three isolation methods. J Hyg (Lond) 95:69–75. ———. 1978. Influence of oral antibiotic feeding on the
Smith, H. W., and Jones, J. E. T. 1967. Observations on ex- duration and severity of clinical disease, growth perfor-
perimental oral infection with Salmonella dublin in mance and pattern of shedding in swine inoculated with
calves and Salmonella choleraesuis in pigs. J Pathol Salmonella typhimurium. J Am Vet Med Assoc
93:141–156. 172:472–477.
Stephen, J.; Wallis, T. S.; Starkey, W. G.; Candy, D. C. A.; Os- Wilcock, B. P.; Armstrong, C. H.; and Olander, H. J. 1976.
borne, M. P.; and Huddon, S. 1985. Salmonellosis: In The significance of the serotype in the clinical and
retrospect and prospect. In Microbial Toxins and Diar- pathologic features of naturally occurring porcine sal-
rhoeal Diseases. Ciba Foundation Symposium. London: monellosis. Can J Comp Med 40:80–88.
Pitman. Will, L. A.; Diesch, S. L.; and Pomeroy, B. X. 1973. Survival
Surkiewicz, B. F.; Johnston, R. W.; Elliott, E. P.; and Sim- of Salmonella typhimurium in animal manure disposed
mons, E. R. 1972. Bacteriologic survey of fresh pork in a model oxidation ditch. Am J Public Health
sausage produced at establishments under federal in- 63:322–336.
spection. Appl Microbiol 23:515–520. Williams, C. B. 1975. Environmental considerations in sal-
Tacal, J. V., and Lawrence, W. 1980. A survey for salmonella monellosis. Vet Rec 93:317–321.
among market swine in southern California, 1980. Calif Williams, L. P., and Newell, K. W. 1970. Salmonella excre-
Vet 11:15–18. tion in joy-riding pigs. Am J Public Health 50:926–929.
Takeuchi, A. 1967. Electron-microscopic studies of experi- Williams, L. P.; Vaughn, J. P.; Scott, A.; and Blanton, V.
mental salmonella infection. I. Penetration into the in- 1969. A ten-month study on salmonella contamination
testinal epithelium by Salmonella typhimurium. Am J in animal protein meals. J Am Vet Med Assoc
Pathol 50:109–136. 155:167–174.
Takeuchi, A., and Sprinz, H. 1967. Electron microscopic Wolff, S. M. 1973. Biologic effects of bacterial endotoxins in
studies of experimental salmonella infection in the pre- man. J Infect Dis 128(Suppl):159–164.
conditioned guinea pig. II. Response of the intestinal Wood, R. L., and Rose, R. 1992. Populations of Salmonella
mucosa to the invasion by Salmonella typhimurium. Am J typhimurium in internal organs of experimentally infect-
Pathol 51:137–161. ed carrier swine. Am J Vet Res 53:653–658.
Taylor, J., and McCoy, J. H. 1969. Salmonella and Arizona in- Wood, R. L.; Pospischil, A.; and Rose, R. 1989. Distribution
fections and intoxications. In Foodborne Infections and of persistent Salmonella typhimurium infection in inter-
Intoxications. New York: Academic Press, pp. 3–71. nal organs of swine. Am J Vet Res 50:1015–1021.
Vassiliadis, P.; Mavromati, C.; Trichopoulus, D.; Kalapotha- Wray, C., and Sojka, W. J. 1977. Reviews of progress in dairy
ki, V.; and Papadakis, J. 1987. Comparison of proce- science: Bovine salmonellosis. J Dairy Res 44:383–425.
Spirochetal Diarrhea/Porcine
40 Intestinal Spirochetosis
D. J. Hampson and D. J. Trott

The term “spirochetal diarrhea” has been used to de- was thought to be S. innocens but is now recognized as
scribe a colitis of growing pigs associated with infection the type strain of S. pilosicoli. The condition has since
with a weakly hemolytic intestinal spirochete distinct been experimentally reproduced in pigs on a number of
from Serpulina hyodysenteriae (the agent of swine dysen- occasions using other strains of S. pilosicoli (Andrews
tery) (Taylor 1980, 1992). The causal agent in the original and Hoffman 1982; Thompson et al. 1996; Trott et al.
study of this condition (Taylor et al. 1980) recently has 1996c). PIS is now increasingly becoming recognized as a
been shown to be a distinct species of spirochete, now of- major cause of colitis in growing pigs, particularly since
ficially named Serpulina pilosicoli (Trott et al. 1996e). S. the adoption of more intensive production strategies
pilosicoli previously has been described in the literature that have limited other major intestinal diseases such as
as “Anguillina coli” (Lee et al. 1993), “Serpulina coli” salmonellosis and swine dysentery (Duhamel 1996). It is
(Duhamel et al. 1993a), and group IV weakly hemolytic likely that at least some of the cases of “nonspecific coli-
intestinal spirochetes (Fellström and Gunnarsson tis” reported in the United Kingdom may have been
1994). caused by S. pilosicoli, although a noninfectious, diet-re-
Infection with S. pilosicoli is characterized by a mild sponsive colitis also does exist (Wood 1991). PIS has
to moderate typhlocolitis resulting in the passage of wa- been reported in most major pig-producing countries,
tery to mucoid feces and consequently loss of condition including the United Kingdom (Taylor et al. 1980), Cana-
and reduced growth rate. The most striking histological da (Girard and Higgins 1989; Girard et al. 1995; Jacques
feature of the condition is the presence of large numbers et al. 1989; Spearman et al. 1988), Australia (Hampson
of intestinal spirochetes attached by one cell end to the 1991; Hampson and Trott 1995), the United States
surface of the colonic epithelium. This characteristic at- (Duhamel et al. 1993b; Ramanathan et al. 1993), Sweden
tachment also has been observed in humans (Lee and (Fellström et al. 1996), and Denmark (Møller et al.
Hampson 1994), primates (Duhamel et al. 1996), dogs 1996). Much of the research concerning PIS has only
(Duhamel et al. 1995b), chickens (McLaren et al. 1997), been undertaken recently, and many important aspects
and ducks (Trott et al. 1996a). of the disease are still poorly understood, including
Prior to the classification of S. pilosicoli and its recog- pathogenicity, host immunity, and detailed epidemiolo-
nition as a cause of diarrhea in the pig and other host gy.
species, the attachment of intestinal spirochetes by one
end to the colonic epithelium of humans was referred to ETIOLOGY
as “intestinal spirochetosis” (Harland and Lee 1967). For
this reason, when S. pilosicoli was named, we used the Serpulina pilosicoli takes its name from the histological
term “porcine intestinal spirochetosis” (PIS) rather than appearance of PIS, where the end-on attachment of
“spirochetal diarrhea” to describe the disease it caused in spirochetes to the colonic epithelium resembles a hairy
pigs (Trott et al. 1996e). In North America the infection covering on the surface of the colon (Serpulina pilosicoli is
also has been called “porcine colonic spirochetosis” (Gi- Latin for “little serpent of the hairy colon”) (Trott et al.
rard et al. 1995), but the preferred term “porcine intesti- 1996e). S. pilosicoli has a characteristic spirochete mor-
nal spirochetosis” will be used for the condition in the re- phology and an appearance similar to other species in
mainder of this chapter. the genus Serpulina, except that it is generally shorter
PIS was first described by Taylor and coworkers (6–10 µm in length) and thinner (0.25–0.30 µm in
(1980), who challenged experimental pigs with a weakly width), has fewer periplasmic flagella (4–7 attached at
beta-hemolytic strain of intestinal spirochete each cell end), and has pointed, instead of rounded, ends
(P43/6/78), inducing a colitis associated with mucoid di- (Fig. 40.1). These differences can only be confirmed by
arrhea containing flecks of blood. This strain initially electron microscopy; however, using phase contrast mi-
croscopy, S. pilosicoli cells generally appear thinner and
shorter than cells of the other species of Serpulina. S. pi-
Recently the genus name Brachyspira has been suggested for Serpulina losicoli grows as a thin streak of weak beta-hemolysis on
hyodysenteriae, Serpulina innocens, and Serpulina pilosicoli. trypticase soy blood agar and is cultured under the same

553
554 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

conditions as those described for S. hyodysenteriae, except et al. 1977; Lee et al. 1993). However, strains of S. inter-
that it may take up to 6 days for hemolysis to appear, and media have been shown to be pathogens in commercial
the inclusion of additional antibiotics (rifampicin chickens (McLaren et al. 1997), have caused lesions when
and spiramycin) normally recommended for the isola- inoculated into isolated porcine colonic loops (Binek and
tion of S. hyodysenteriae may inhibit the growth of the Szynkiewicz 1984), and on rare occasions have been as-
more sensitive organism (Trott et al. 1996d). Slicing the sociated with diarrhea in pigs in the field (Fellström and
agar prior to primary inoculation, as reported for S. Gunnarsson 1995). Additionally, some strains of S. inno-
hyodysenteriae by Olson (1996), may help improve the re- cens isolated from pigs with diarrhea have been shown to
covery of S. pilosicoli free of other contaminating organ- cause diarrhea and lesions when inoculated into gnoto-
isms. biotic pigs (Neef et al. 1994). The occurrence of colitis
The taxonomy of the porcine intestinal spirochetes is and diarrhea in pigs colonized by S. intermedia has previ-
a rapidly developing field (Hampson and Stanton 1997), ously been termed “spirochetal colitis” (Hampson and
and in addition to S. innocens and S. pilosicoli, two new Trott 1995; Taylor and Trott 1997). Little is known about
species of weakly beta-hemolytic spirochete have been this condition; however, it appears to be much less fre-
formally proposed: Serpulina intermedia and Serpulina quently encountered than PIS and therefore will only be
murdochii (Stanton et al. 1997). All four species of weak- mentioned briefly as a differential diagnosis.
ly hemolytic intestinal spirochete have a similar appear-
ance on blood agar and can only be differentiated from EPIDEMIOLOGY
one another by biochemical and genetic typing tech-
niques. S. innocens, S. intermedia, and S. murdochii have The detailed epidemiology of PIS has not been fully de-
never been shown to cause disease in experimentally in- termined. Infection is believed to occur by the fecal/oral
fected conventional pigs and are generally considered to route, and disease in nonimmune herds may be facilitat-
be nonpathogens (Hudson and Alexander 1976; Kinyon ed by the introduction of carrier pigs. Infection has been

40.1. (A) Electron micrograph of


whole cell of P43/6/78T. Marker
bar = 0.5 µm. (B) Electron micro-
graph of magnified cell end of
Serpulina pilosicoli strain
P43/6/78T showing pointed tip,
five periplasmic flagella, and
insertion disks. Marker bar = 0.1
µm.
B
CHAPTER 40 SPIROCHETAL DIARRHEA Hampson, Trott 555

demonstrated in specific-pathogen-free (SPF) herds free PATHOGENESIS


of swine dysentery (Atyeo et al. 1996c). The organism is
believed to survive in feces for a length of time similar to The pathogenesis of PIS is not well understood but is
that previously demonstrated for S. hyodysenteriae (Chia thought to differ from swine dysentery in several impor-
and Taylor 1978). Using culture techniques, the preva- tant ways. Cells of S. pilosicoli are not attracted to porcine
lence in individual piggeries has been demonstrated to mucin in the same way as are virulent strains of S. hyo-
range from 5% (Atyeo et al. 1996c) to 37.5% (Duhamel dysenteriae (Milner and Sellwood 1994), and infection is
1996). However, these figures are often influenced by the often associated with the presence of large numbers of
concurrent administration of antibiotics, the age of the spirochetes attached by one cell end to the luminal sur-
pigs examined, the limits of detection using culture, and face of colonic and cecal epithelial cells. Additionally, the
the degree of contamination with other fecal organisms colitis associated with PIS is much less severe than swine
which may inhibit the growth of the spirochetes. In Swe- dysentery and only resembles the very early stages of
den, S. pilosicoli was isolated from 6 of 8 herds with that disease.
diarrhea but only from 1 of 11 herds with no clinical Oral infection is followed by colonization of the
signs (Fellström et al. 1996). S. pilosicoli may be isolated colonic mucosa, and the organism can be demonstrated
from pigs during any period of the growing stage, but in fecal swabs within 2–7 days postinoculation; however,
infection is most common and more severe in weaner the incubation period may range up to 20 days. In the ini-
and grower pigs. Specific serologic tests are not yet tial stages of infection, S. pilosicoli cells adhere in large
available for determining antibody titers in exposed ani- numbers to the surface of cecal and colonic epithelial
mals. cells, resulting in effacement of the microvilli and dis-
Unlike the situation with S. hyodysenteriae, where in- ruption of the terminal web cytoplasm. Attachment on-
fection throughout a piggery is usually caused by a single ly occurs to mature apical enterocytes on the villous tips,
strain of the organism, S. pilosicoli is a genetically diverse and spirochetes do not attach to immature cells within
species (Atyeo et al. 1996a; Lee and Hampson 1994), and intestinal crypts (Trott et al. 1995). Epithelial cell degen-
often more than one type of strain of the organism may eration results in an increase in the crypt cell mitotic
be isolated from a single pen of pigs (Atyeo et al. 1996c). rate, crypt elongation, and the production of an imma-
The demonstration of multiple strains within piggeries ture epithelium consisting of squamous or cuboidal
may help explain the common recurrence of PIS in con- cells. Necrosis of the epithelium is evident grossly as
valescent animals or in those that have been treated with small adherent nodules on the surface of the mucosa
antimicrobials. (Fig. 40.2).
Unlike S. hyodysenteriae, strains of S. pilosicoli colo- S. pilosicoli cells have been observed within dilated in-
nize a range of animal species (Atyeo et al. 1996a). Typi- testinal crypts (Trott et al. 1996c), invading through tight
cal clinical signs and lesions associated with PIS have junctions between epithelial cells, within goblet cells
been demonstrated in all host species, including hu- (Thompson et al. 1996), and within the lamina propria.
mans. Isolates from pigs, dogs, and humans have been The presence of S. pilosicoli within crypts and the lamina
shown to be genetically closely related (Lee and Hamp- propria is associated with neutrophilic exocytosis (crypt
son 1994), but direct evidence for zoonotic transmission abscesses) and colitis characterized by edema, with a
has only been demonstrated between dogs and humans mixed infiltrate of neutrophils and lymphocytes within
(Trott et al. 1996b). Nevertheless, human strains of S. pi- the mucosa, lamina propria, and occasionally muscularis
losicoli have been shown to cause disease when inoculat- layers. In chronic infections, neutrophils may be absent
ed into conventional pigs (Trott et al. 1996c), and the po- and the lamina propria is infiltrated with monocytes and
tential for cross-transmission between humans and pigs lymphocytes (Duhamel 1996). Invasion has been ob-
cannot be discounted. Because humans colonized with S. served both concurrent with and independent of attach-
pilosicoli tend to be either immunocompromised or live ment of spirochete cells to the epithelium. The damaged
in developing communities, healthy pig industry work- mucosa is often colonized by large numbers of Balantid-
ers are unlikely to be at risk of developing disease from ium coli cells. S. pilosicoli has been isolated from the
pigs infected with PIS. blood of humans with severe clinical disease or impaired
Other host species, particularly birds, may represent immunity (Trott et al. 1997). Although a systemic spiro-
continual sources of transmission for pigs. Although ro- chetemia has not been directly observed in pigs, its oc-
dents have been demonstrated to be carriers of S. hyo- currence cannot be excluded. Occasionally some animals
dysenteriae and may be colonized with weakly beta-he- are found dead with the only lesions being suggestive of
molytic spirochetes, all other rodent isolates examined mucohemorrhagic colitis caused by S. pilosicoli. To date,
by us to date have been shown to be members of the non- there has been no reported attempt to culture the blood
pathogenic species S. murdochii (Trott et al. 1996a). De- of such animals using media appropriate for the isola-
spite this, strain P43/6/78 has been shown to colonize tion of S. pilosicoli.
experimentally infected mice, thus demonstrating poten- Colonization of the epithelium, local invasion, and
tial for mice to act as carriers (Sacco et al. 1996). subsequent colitis combine to cause an increase in the
556 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

40.2. Colonic mucosa of a


chronic case of porcine intestinal
spirochetosis. Note the isolated
areas of necrotic material.

water content of the cecal and colonic digesta, together growers, and finishers at the same piggery. Because of
with excess mucus production and occasionally flecks of the large functional capacity of the cecum and colon, not
blood. The resulting damaged or immature epithelium all infected animals will develop diarrhea; however, sub-
may result in a reduction in surface area of the colon, re- clinical infections will still result in depressed growth
duced absorption of volatile fatty acids, and consequent- rates.
ly poor feed conversion and weight gain (Duhamel The first clinical signs that tend to be observed are
1996). hollowing of the flanks and the passage of loose, sticky
The host immune mechanisms that are directed feces that adhere to the pen floor. The consistency of the
against S. pilosicoli have not been determined. Circulat- feces then changes to that of wet cement or porridge and
ing antibodies have been demonstrated in convalescent may take on a glistening appearance. These may be the
experimentally infected pigs, but the immune response only clinical signs observed in finishers. Weaner and
has not be characterized in detail (Taylor et al. 1980). grower pigs usually develop a watery to mucoid diarrhea
Gnotobiotic pigs (Neef et al. 1994), day-old chicks which is green or brown in color and occasionally char-
(Adachi and Minato 1986; Duhamel et al. 1995a; Trott et acterized by thick tags of mucus and flecks of blood. Di-
al. 1995), and mice (Sacco et al. 1996) have successfully arrhea is usually self-limiting and lasts between 2 and 14
been used as models of experimental infection. In exper- days, although some animals may relapse and develop
imental infections of conventional pigs, only a small pro- clinical signs after convalescence or treatment.
portion (33%) of challenged animals became colonized Affected pigs appear ill-thrifty, have fecal staining of
and developed disease (Trott et al. 1996c). Adachi et al. the perineum, have a tucked-up appearance, and are
(1982) infected pigs with weakly hemolytic spirochetes sometimes febrile, but usually continue to eat. Pigs with
(most likely to be S. pilosicoli) and demonstrated that PIS may have concurrent illness such as pneumonia, and
those animals with the highest titers of complement-fix- pigs with other, more severe gastrointestinal diseases
ing antibody shed the least number of organisms in their such as swine dysentery, salmonellosis, or intestinal
feces. adenopathy may also be colonized with S. pilosicoli. Pigs
with uncomplicated PIS that develop loose feces may not
CLINICAL SIGNS show any reduction in liveweight gain, but the condition
is generally characterized by significant loss of condi-
PIS has a clinical presentation similar to other forms of tion, decreased feed conversion, and delays in reaching
colitis and to the early stages of swine dysentery. It is market weight. Mortality is generally not a feature of
commonly seen in the immediate postweaning period PIS.
and in recently mixed growers placed on a new ration but
can be observed in finishers and occasionally pregnant LESIONS
sows and recently introduced breeding stock. PIS may af-
fect groups of pigs of the same age in a unit or be present Gross Lesions
in pigs of mixed age within a house. It is not uncommon Gross lesions associated with PIS are limited to the ce-
to observe various manifestations of PIS in weaners, cum and colon and may be very subtle, particularly in
CHAPTER 40 SPIROCHETAL DIARRHEA Hampson, Trott 557

the early stages of the disease. Postmortem examina- ic or petechial hemorrhages may be apparent on the sur-
tion soon after the onset of clinical signs often reveals face. In chronic cases and in resolving lesions, the hem-
a flaccid, fluid-filled cecum and colon with an edema- orrhages are covered by small tags of adherent fibrin,
tous serosal surface and enlarged mesenteric and necrotic material, and digesta, which appear as conical
colonic lymph nodes. The contents are abundant and scales adherent on the surface of the mucosa (Fig. 40.2).
watery green or occasionally yellow and frothy. The This material may be dislodged by rinsing and can be
severity and clinical stage of the disease are reflected found as a deposit after decanting the washings.
in the appearance of the mucosal surface. There may
be few changes in the early stages of the disease, al- Microscopic Lesions
though mild congestion and hyperemia may be ap- PIS has been described as a catarrhal, multifocal, erosive
parent, with occasional ulcerations and necrotic foci. or ulcerative colitis (Andrews and Hoffman 1982; Girard
Inflammation in the later stages may result in multi- and Higgins 1989). Lesions are generally confined to the
focal ulcerative or mucohemorrhagic colitis, but it is mucosa and submucosa but may extend into the muscu-
never as severe as that observed in swine dysentery. laris. The mucosa is thickened, edematous, and occa-
The mucosa becomes thickened, and local ecchymot- sionally hyperemic and is characterized by the presence

40.3. Photomicrograph of
hematoxylin eosin–stained section
of porcine colon infected with a
Serpulina pilosicoli strain.
Neutrophilic exocytosis (crypt
abscess material) is present in
dilated intestinal crypts (arrows).
The lamina propria is diffusely
infiltrated with inflammatory
cells. Marker bar = 37 µm.

40.4. Photomicro-
graph of the colonic
epithelium. Note false
brush border of spiro-
chetes (arrow) (×400).
558 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

of dilated, elongated intestinal crypts filled with mucus, Bacteriology


cellular debris, and degenerate inflammatory cells gener- Large numbers of spirochetes can be seen in wet smears
ally referred to as crypt abscesses (Fig. 40.3). Subepithe- viewed by phase contrast microscopy or in fixed Gram-
lial capillary dilatation is a noteworthy finding. The stained smears. Swabs should be taken directly from the
crypt cell mitotic rate is increased, often with immature, colonic wall rather than collecting colonic contents.
cuboidal, or squamous epithelium present on the tips of
the villi. Where columnar epithelium is still present on DIAGNOSIS
the surface of the colon, it is often covered by a dark
fringe of spirochetes attached by one cell end (Fig. 40.4). A tentative diagnosis of PIS can be made when typical
The use of silver stains will confirm the presence of clinical signs of mucoid diarrhea without blood and no
spirochete-shaped bacteria attached to the surface of mortality are found in weaned pigs not previously re-
colonic enterocytes, within dilated intestinal crypts, and ceiving medication in the feed. The clinical signs of PIS
occasionally within the lamina propria. Balantidium coli are difficult to differentiate from those associated with
bacteria are often seen in large numbers on the surface of the proliferative enteropathies caused by Lawsonia intra-
the colon (Taylor et al. 1980; Trott et al. 1996c). cellularis. Additionally, PIS may occur concurrently with
a number of gastrointestinal diseases, including prolifer-
Electron Microscopy ative enteritis, salmonellosis, postweaning colibacillosis,
Using transmission electron microscopy, polar-attached swine dysentery, and trichuriasis. All of these should be
spirochetes with 4–7 periplasmic flagella attached to considered in the differential diagnosis of PIS, as well as
each cell end are observed invaginated into the terminal “nonspecific colitis,” an apparently dietary condition as-
web cytoplasm, effacing microvilli and disrupting micro- sociated with a form of hypersensitivity to pelleted feed
filaments without penetration of the host cell plas- (Connor 1992; Smith and Nelson 1987). Additionally,
malemma (Fig. 40.5). Scanning electron microscopy re- Clostridium perfringens type A enterotoxemia (Taylor et
veals the adherent spirochetes as a patchy fringe on the al. 1987) and Yersinia pseudotuberculosis infection (Neef
surface of colonic epithelial cells. Spirochetes also may and Lysons 1994) must be considered as causes of colitis.
be observed invading between epithelial cells in the ex- To obtain a diagnosis, postmortem examination of
trusion zone between adjacent crypt units (Duhamel several affected pigs, including routine histological and
1996). bacteriological examination, should be undertaken. Fe-

40.5. Transmission electron


micrograph of porcine colon infect-
ed with a strain of Serpulina
pilosicoli. Note attachment of
large numbers of spirochetes to
the epithelium and disruption of
terminal web microfilaments
(arrow). Marker bar = 2 µm.
CHAPTER 40 SPIROCHETAL DIARRHEA Hampson, Trott 559

cal samples for culture for Serpulina spp. and other The four recognized species of weakly hemolytic in-
pathogens also should be obtained from a cross section testinal spirochetes can be differentiated without the
of affected pigs. While waiting for confirmation of re- need for culture by digestion of PCR-amplified 16S rRNA
sults, water medication should be initiated, as it may DNA with restriction enzymes. Characteristic DNA frag-
take up to 2 weeks to obtain a definitive diagnosis from ments produced are identified after separation by elec-
culture. In histological sections from the cecum and par- trophoresis (Stanton et al. 1997). An indirect fluorescent
ticularly the colon, the presence of spirochetes attached antibody test using a monoclonal antibody (MAB) raised
to the colonic mucosa is diagnostic for PIS but will not be against a specific outer-membrane protein of S. pilosicoli
observed in every case of the disease. Other typical his- also shows considerable potential for diagnostic use on
tological lesions associated with PIS, including the feces, and with suitable modification the MAB may be
demonstration of spirochetes within intestinal crypts applied to tissue sections for immunoperoxidase stain-
and the lamina propria by silver staining or the demon- ing (Lee and Hampson 1995). There is significant sero-
stration of spirochetes in smears, should be confirmed logic cross-reactivity among the intestinal spirochetes,
by the results of culture. and to date no specific tests are available to measure
The four species of weakly beta-hemolytic spiro- serum antibody titers.
chetes can be differentiated from one another and from If weakly beta-hemolytic spirochetes distinct from S.
S. hyodysenteriae using a number of biochemical tests pilosicoli are cultured from pigs with colitis, spirochetal
outlined in Table 40.1. Unfortunately, these tests require colitis should be suspected, particularly if the organisms
the growth of pure cultures of Serpulina organisms in liq- are indole positive and have API-ZYM profiles consistent
uid medium, a process which may take several weeks to with S. intermedia. Spirochetal colitis should respond to
achieve, particularly if the initial plates are heavily con- treatment in a manner similar to PIS.
taminated with other fecal flora. Consequently, the tests
are currently performed only in several specialized labo- TREATMENT AND CONTROL
ratories. For S. pilosicoli, examination of ultrastructural
morphology under the transmission electron micro- Treatment and control of PIS are largely modeled on
scope, hippurate hydrolysis, metabolism of ribose, and procedures developed for swine dysentery. No effective
lack of beta-glucosidase activity in the API-ZYM profile vaccines have been produced to date. Affected pigs
are diagnostic (Fellström and Gunnarsson 1995; Trott et should be treated by water or feed medication at similar
al. 1996d). levels and lengths of time as recommended in Chapter
A polymerase chain reaction (PCR) test based on a 42 for the treatment of swine dysentery. Parenteral treat-
unique 16S rRNA DNA sequence is completely specific ment may sometimes be warranted for individual ani-
for S. pilosicoli (Park et al. 1995). Although not sensitive mals. A number of antimicrobials that are effective
enough for use directly on feces, the technique is more against S. hyodysenteriae, including tiamulin, carbadox,
sensitive than culture alone (Atyeo et al. 1996b). When dimetridazole, lincomycin, and tylosin, have been shown
applied to DNA harvested from 4-day-old bacteriological to have low minimum inhibitory concentration (MIC)
plates containing the organism, it can significantly re- values when tested against S. pilosicoli isolates in vitro
duce the time taken for a diagnosis, even if there is a (Trott et al. 1996d). Tetracyclines also have low MIC val-
large amount of contamination with other fecal flora ues, which suggests their effectiveness against S. pilosicoli
(Atyeo et al. 1996b). A second PCR also based on the S. (Trott et al. 1996d). Olaquindox has been shown to have
pilosicoli 16S signature sequence but producing a slightly MIC values of less than 1.0 µg/mL against S. pilosicoli
smaller product has been reported but did not signifi- (group IV spirochetes), and the organism could not be
cantly improve the sensitivity of the initial procedure isolated from herds previously receiving olaquindox in
(Fellström et al. 1997). the feed at 100 ppm (Fellström et al. 1996). However, de-

Table 40.1. Differentiation of the five recognized groups of porcine intestinal spirochetes by their hemolysis
pattern on trypticase soy blood agar, biochemical reactions, and utilization of sugars
Test S. hyodysenteriae S. intermedia S. innocens S. murdochii S. pilosicoli
Hemolysis strong weak weak weak weak
Indole + + − − −
Hippurate − − − − +
API-ZYM 1 1 2 3 4
d-ribose − − − − +
Note: + = positive reaction; − = negative reaction; 1 = alpha-glucosidase positive, alpha-galactosidase negative; 2 = alpha-glucosidase
negative, alpha-galactosidase positive; 3 = alpha-glucosidase negative, alpha-galactosidase negative; 4 = variable reactions, including positive
reactions for both enzymes, beta-glucosidase negative.
560 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

spite its preventive qualities, olaquindox is not suitable ples. Proc Int Congr Pig Vet Soc 14:291.
as a therapeutic agent (Fellström et al. 1996): olaquindox Atyeo, R. F.; Trott, D. J.; Robertson, I. D.; Buddle, J. R.; and
is insufficiently active; at the level required for effective- Hampson, D. J. 1996c. Epidemiological analysis of Ser-
ness, the pig would die. pulina pilosicoli within a high-health status pig herd.
Resistance to single antimicrobials or combinations Proc Int Congr Pig Vet Soc 14:287.
has been recorded in our laboratory. In the field, the oc- Binek, M., and Szynkiewicz, Z. 1984. Physiological proper-
currence of reinfection after medication has ceased is ties and classification of strains of Treponema sp. isolat-
common. Adjunctive therapy such as improved manage- ed from pigs in Poland. Comp Immunol Microbiol Infect
ment and pen hygiene has given inconsistent results Dis 7:141–148.
(Wilkinson and Wood 1987), whereas reducing the ener- Chia, S. P., and Taylor, D. J. 1978. Factors affecting the sur-
gy and protein content of the ration has often alleviated vival of Treponema hyodysenteriae in dysenteric pig fae-
the problem (Spearman et al. 1988; Wilkinson and ces. Vet Rec 103:68–70.
Wood 1987). It has been reported that the addition of Connor, J. F. 1992. Nonspecific colitis. In Proc Aust Assoc
zinc oxide in the feed at 3 kg/tonne helps control PIS Pig Vet, Adelaide, pp. 79–80.
(Love 1996). This method also has been effective in con- Duhamel, G. E. 1996. Porcine colonic spirochaetosis caused
trolling uncomplicated cases of “nonspecific colitis” (Ka- by Serpulina pilosicoli. Misset Pigs, May (Enteric Dis-
vanagh 1992). eases Suppl): 10–13.
The methods described for the control of swine Duhamel, G. E.; Mathiesen, M. R.; Schafer, R. W.; Ra-
dysentery in Chapter 42 may be effective at eliminating manathan, M.; and Johnston, J. L. 1993a. Description of
PIS, but the severity of PIS generally does not warrant a new species of spirochete, Serpulina coli sp. nov. asso-
such drastic procedures. Medicated early weaning, as ciated with intestinal spirochetosis of swine and human
proposed by Alexander et al. (1980), and discussed else- beings. In Annu Meet Con Res Work Anim Dis, Chicago,
where for swine dysentery, is the only recorded practice p. 14.
that has eliminated intestinal spirochetes from a group Duhamel, G. E.; Ramanathan, M.; Gardner, I.; Anderson,
of animals. Where PIS becomes endemic in a herd, regu- M. A.; Walker, R. L.; and Hampson, D. J. 1993b. Intesti-
lar periods of treatment with antimicrobials in the feed nal spirochetosis of swine associated with weakly β-he-
or water may be required to prevent sudden increases in molytic spirochetes distinct from Serpulina innocens. In
morbidity due to recent introduction of nonimmune Proc Ann Con Amer Assoc Vet Lab Diagn, Las Vegas, p.
pigs, change of diet, or other periods associated with 53.
stress. Duhamel, G. E.; Muniappa, N.; Gardner, I.; Anderson, M.
A.; Blanchard, P. C.; DeBey, B. M.; Mathiesen, M. R.;
REFERENCES and Walker, R. L. 1995a. Porcine colonic spirochaetosis:
A diarrhoeal disease associated with a newly recognized
Adachi, Y., and Minato, H. 1986. Experimental infection of species of intestinal spirochaetes. Pig J 35:101–110.
young chicks with a weak beta-hemolytic spirochete dif- Duhamel, G. E.; Muniappa, N.; Mathiesen, M. R.; Johnson,
fering from Treponema hyodysenteriae. Proc Int Congr Pig J. L.; Toth, J.; Elder, R. O.; and Doster, A. R. 1995b. Cer-
Vet Soc 9:34. tain canine weakly beta-hemolytic spirochetes are phe-
Adachi, Y.; Tanaka, T.; and Watase, H. 1982. Transition of notypically and genotypically related to spirochetes as-
antibody titres in swine infected with weak β-hemolytic sociated with human and porcine intestinal
spirochetes. Proc Int Congr Pig Vet Soc 7:34. spirochetosis. J Clin Microbiol 33:2212–2215.
Alexander, T. J. L.; Thornton, K.; Boon, G.; Lysons, R. J.; and Duhamel, G. E.; Elder, R. O.; Muniappa, N.; Mathiesen, M.
Gush, A. F. 1980. Medicated early weaning to obtain R.; Wong, V. J.; and Tarara, R. P. 1996. Colonic spiro-
pigs free from pathogens endemic in the herd of origin. chetal infections of non-human primates associated
Vet Rec 106:114–119. with Brachyspira aalborgi, Serpulina pilosicoli and unclas-
Andrews, J. J., and Hoffman, L. J. 1982. A porcine colitis sified flagellated bacteria. In Annu Meet Con Res Work
caused by a weakly beta-hemolytic treponeme (Trepone- Anim Dis, Chicago, p. Abstr 89.
ma innocens?). In Proc Annu Con Amer Assoc Vet Lab Fellström, C., and Gunnarsson, A. 1994. A biochemical re-
Diagn, Nashville, pp. 395–402. action scheme for porcine intestinal spirochetes. Proc
Atyeo, R. F.; Oxberry, S. L.; and Hampson, D. J. 1996a. Int Congr Pig Vet Soc 14:145.
Pulsed-field gel electrophoresis for sub-specific differen- ———. 1995. Phenotypic characterisation of intestinal
tiation of Serpulina pilosicoli (formerly “Anguillina coli”). spirochaetes isolated from pigs. Res Vet Sci 59:1–4.
FEMS Microbiol Lett 141:77–81. Fellström, C.; Pettersson, B.; Johansson, K.; Lundenheim,
Atyeo, R. F.; Trott, D. J.; and Hampson, D. J. 1996b. Use of N.; and Gunnarsson, A. 1996. Prevalence of Serpulina
polymerase chain reaction for the detection of swine species in relation to diarrhea and feed medication in
dysentery and intestinal spirochaetosis from faecal sam- pig-rearing herds in Sweden. Am J Vet Res 57:807–811.
CHAPTER 40 SPIROCHETAL DIARRHEA Hampson, Trott 561

Fellström, C.; Pettersson, B.; Thompson, J.; Gunnarsson, A.; spirochetes: Potential role in intestinal colonization. In-
Persson, M.; and Johansson, K. 1997. Identification of fect Immun 62:4095–4099.
Serpulina species associated with porcine colitis by bio- Møller, K.; Jensen, T. K.; and Jorsal, S. E. 1996. Bacteriologi-
chemical analysis and PCR. J Clin Microbiol cal examination and PCR analysis of feces from growing
35:462–467. pigs originating from herds with and without diarrhea.
Girard, C., and Higgins, R. 1989. Colonic spirochetosis in Proc Int Congr Pig Vet Soc 14:325.
piglets. Can Vet J 30:68. Neef, N. A., and Lysons, R. J. 1994. Pathogenicity of a strain
Girard, C.; Lemarchand, T.; and Higgins, R. 1995. Porcine of Yersinia pseudotuberculosis isolated from a pig with
colonic spirochetosis: A retrospective study of eleven porcine colitis syndrome. Vet Rec 135:58–63.
cases. Can Vet J 36:291–294. Neef, N. A.; Lysons, R. J.; Trott, D. J.; Hampson, D. J.; Jones,
Hampson, D. J. 1991. New developments in research on P. W.; and Morgan, J. M. 1994. Pathogenicity of porcine
swine dysentery and spirochaetal colitis. Pig News In- intestinal spirochetes in gnotobiotic pigs. Infect Immun
form 12:233–235. 62:2395–2403.
Hampson, D. J., and Stanton, T. B. 1997. Intestinal Olson, L. D. 1996. Enhanced isolation of Serpulina hyodysen-
Spirochaetes in Domestic Animals and Humans. teriae by using sliced agar media. J Clin Microbiol
Wallingford: CAB International. 34:2937–2941.
Hampson, D. J., and Trott, D. J. 1995. Intestinal spirochaetal Park, N. Y.; Chung, C. Y.; McLaren, A. J.; Atyeo, A. J.; and
infections of pigs: An overview and an Australian per- Hampson, D. J. 1995. Polymerase chain reaction for
spective. In Manipulating Pig Production V. Ed. D. P. identification of human and porcine spirochaetes recov-
Hennessy and P. D. Cranwell. Canberra, ACT: Aus- ered from cases of intestinal spirochaetosis. FEMS Mi-
tralasian Pig Science Association, pp. 139–169. crobiol Lett 125:225–230.
Harland, W. A., and Lee, F. D. 1967. Intestinal spirochaeto- Ramanathan, M.; Duhamel, G. E.; Mathiesen, M. R.; and
sis. Brit Med J 3:718–719. Messier, S. 1993. Identification and partial characteriza-
Hudson, M. J., and Alexander, T. J. L. 1976. Diagnosis of tion of a group of weakly β-hemolytic spirochetes of
swine dysentery: Spirochaetes which may be confused swine distinct from Serpulina innocens. Vet Microbiol
with Treponema hyodysenteriae. Vet Rec 99:498–500. 37:53–64.
Jacques, M.; Girard, C.; Higgins, R.; and Goyette, G. 1989. Sacco, R. E.; Trampel, D. W.; and Wannemueler, M. J. 1996.
Extensive colonization of the porcine colonic epithelium Experimental infection of mice with avian or porcine
by a spirochete similar to Treponema innocens. J Clin Mi- isolates of Serpulina pilosicoli. In Annu Meet Con Res
crobiol 27:1139–1141. Work Anim Dis, Chicago, p. Abstr 92.
Kavanagh, N. T. 1992. Non-specific colitis: Observations on Smith, W. J., and Nelson, E. P. 1987. Grower scour/non-spe-
methods of control and prevention at farm level. Proc cific colitis. Vet Rec 121:334.
Int Congr Pig Vet Soc 12:522. Spearman, J. G.; Nayar, G.; and Sheridan, M. 1988. Colitis
Kinyon, J. M.; Harris, D. L.; and Glock, R. D. 1977. En- associated with Treponema innocens in pigs. Can Vet J
teropathogenicity of various isolates of Treponema hyo- 29:747.
dysenteriae. Infect Immun 15:638–646. Stanton, T. B.; Fournie-Amazouz, E.; Postic, D.; Trott, D. J.;
Lee, B. J., and Hampson, D. J. 1995. A monoclonal antibody Grimont, P. A. D.; Baranton, G.; Hampson, D. J.; and
reacting with the cell envelope of spirochaetes isolated Saint Girons, I. 1997. Recognition of two new species of
from cases of intestinal spirochaetosis in pigs and hu- intestinal spirochetes: Serpulina intermedia sp. nov. and
mans. FEMS Microbiol Lett 131:179–184. Serpulina murdochii sp. nov. Submitted for publication.
Lee, J. I., and Hampson, D. J. 1994. Genetic characterisation Taylor, D. J. 1980. Spirochaetal diarrhoea. Proc Int Congr
of intestinal spirochaetes and their association with dis- Pig Vet Soc 6:235.
ease. J Med Microbiol 40:365–371. ———. 1992. Spirochetal diarrhea. In Diseases of Swine.
Lee, J. I.; Hampson, D. J.; Lymbery, A. J.; and Harders, S. J. 7th ed. Ed. A. D. Leman, B. E. Straw, W. L. Mengeling, S.
1993. The porcine intestinal spirochaetes: Identification D’Allaire, and D. J. Taylor. Ames: Iowa State Univ Press,
of new genetic groups. Vet Microbiol 34:273–285. pp. 599–616.
Love, R. 1996. Serpulina pilosicoli colitis in grower pigs. Aust Taylor, D. J., and Trott, D. J. 1997. Porcine intestinal
Assoc Pig Vet News 20:6. spirochaetosis and spirochaetal colitis. In Intestinal
McLaren, A. J.; Trott, D. J.; Swayne, D. E.; Oxberry, S. L.; and Spirochaetes in Domestic Animals and Humans. Ed. D.
Hampson, D. J. 1997. Genetic and phenotypic character- J. Hampson and T. B. Stanton. Wallingford: CAB Inter-
ization of intestinal spirochetes colonizing chickens and national, pp. 211–241.
allocation of known pathogenic isolates to three distinct Taylor, D. J.; Simmons, J. R.; and Laird, H. M. 1980. Produc-
genetic groups. J Clin Microbiol 35:412–417. tion of diarrhoea and dysentery in pigs by feeding pure
Milner, J. A., and Sellwood, R. 1994. Chemotactic response cultures of a spirochaete differing from Treponema hyo-
to mucin by Serpulina hyodysenteriae and other porcine dysenteriae. Vet Rec 106:326–332.
562 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Taylor, D. J.; Estrada Correa, A.; and Pradal Roa, P. 1987. fection of newly weaned pigs with human and porcine
Grower scour and non-specific colitis. Vet Rec strains of Serpulina pilosicoli. Infect Immun
121:479–480. 64:4648–4654.
Thompson, J. R.; Smith, W. J.; Murray, B. M.; and McOrist, Trott, D. J.; Stanton, T. A.; Jensen, N. S.; and Hampson, D. J.
S. 1996. Porcine colitis: Results of challenge studies us- 1996d. Phenotypic characteristics of Serpulina pilosicoli,
ing strains of weakly beta-haemolytic spirochaetes. Proc the agent of intestinal spirochaetosis. FEMS Microbiol
Int Congr Pig Vet Soc 14:280. Lett 142:209–214.
Trott, D. J.; McLaren, A. J.; and Hampson, D. J. 1995. Patho- Trott, D. J.; Stanton, T. B.; Jensen, N. S.; Duhamel, G. E.;
genicity of human and porcine intestinal spirochaetes in Johnson, J. L.; and Hampson, D. J. 1996e. Serpulina pi-
day-old specific pathogen free chicks: An animal model losicoli sp. nov.: The agent of porcine intestinal spiro-
of intestinal spirochetosis. Infect Immun 63:3705–3710. chetosis. Int J Syst Bacteriol 46:206–215.
Trott, D. J.; Atyeo, R. F.; Lee, J. I.; Swayne, D. A.; Stouten- Trott, D. J.; Jensen, N. S.; Saint Girons, I.; Oxberry, S. L.;
burg, J. W.; and Hampson, D. J. 1996a. Genetic related- Stanton, T. B.; Lindquist, D.; and Hampson, D. J. 1997.
ness amongst intestinal spirochaetes isolated from rats Identification and characterisation of Serpulina pilosicoli
and birds. Lett Appl Microbiol 23:431–436. isolates recovered from the blood of critically-ill pa-
Trott, D. J.; Combs, B. G.; Mikosza, A. S. J.; Alpers, M.; and tients. J Clin Microbiol 35:482–485.
Hampson, D. J. 1996b. Prevalence and epidemiological Validation list no. 64. 1998. Int J Syst Bacteriol 48:327–328.
analysis of Serpulina pilosicoli in villagers and their ani- Wilkinson, J. D., and Wood, E. N. 1987. Grower scour/non-
mals living in the Eastern Highlands of Papua New specific colitis. Vet Rec 121:406.
Guinea. Aust Microbiol 17:A86. Wood, E. N. 1991. Fashionable and future diseases. Pig Vet J
Trott, D. J.; Huxtable, C. R.; and Hampson, D. J. 1996c. In- 27:193–197.
Streptococcal Diseases
41 R. Higgins and M. Gottschalk

Several streptococcal species can be found in tonsils, in- caused by S. suis because of their emergence in the swine
testines, or feces of clinically healthy pigs, and some of industry during the last decade. Efforts were made to
them are potential pathogens. Among species consid- synthesize the more recent knowledge about the differ-
ered part of the intestinal microflora in swine, there are ent aspects of these infections, particularly pathogene-
Streptococcus intestinalis (Robinson et al. 1988), S. hyoin- sis, clinical signs, diagnosis, and prevention. Importance
testinalis (Devriese et al. 1988), S. suis, S. alactolyticus, was also given to the fact that S. suis is a zoonotic agent
and S. bovis (Devriese et al. 1994b). In tonsils, S. suis, S. with severe consequences, and that its presence in a wide
porcinus, and S. dysgalactiae are members of the mi- range of animal species and birds may influence the epi-
croflora (Devriese et al. 1994b). Members of the genus demiology and the control of the infection in swine.
Enterococcus (formerly Streptococcus) such as E. faecalis, E. Comments about infections caused by other streptococ-
faecium, E. durans, E. hirae, and E. cecorum are also pres- ci, such as S. porcinus (groups E, P, U, and V), S. dysgalac-
ent in the intestinal microflora. tiae subsp. dysgalactiae (group C, formerly S. equisimilis,
In this chapter, the different pathological conditions and group L), and S. agalactiae, have been included, as
associated with streptococci and enterococci are present- well as a discussion of diarrhea associated with entero-
ed. A particular emphasis was put on the infections cocci.

INFECTIONS CAUSED BY S. SUIS


ETIOLOGY AND PREVALENCE originated from diseased pigs except for capsular type
14, a human isolate, capsular types 17, 18, 19, and 21 iso-
New alpha-hemolytic streptococci from septicemic infec- lated from clinically healthy pigs, capsular types 20 and
tions in pigs were first biochemically and serologically 31 from diseased calves, and type 33 from a diseased
characterized by de Moor between 1956 and 1963 as new lamb (Gottschalk et al. 1989; Higgins et al. 1995). The
Lancefield groups R, S, RS, and T (de Moor 1963). In designation of S. suis as a new bacterial species was made
England, Elliott (1966) suggested that de Moor’s group S official in 1987 by Kilpper-Balz and Schleifer. This
was similar to his PM Streptococcus and that both be- species appeared genetically distinct and displayed no
longed to Lancefield’s group D; he proposed the name specific relationship with other streptococcal species ex-
Streptococcus suis capsular type 1. In 1975, Windsor and amined (Bently et al. 1991). Genetic diversity among
Elliott isolated other porcine streptococci which corre- members of the S. suis species is important (Hampson et
sponded to de Moor’s group R and named them S. suis al. 1993; Harel et al. 1994), and this should be taken into
type 2. Type 1 was associated with meningitis in baby account in diagnosis, surveillance, and control of the dis-
piglets, whereas type 2 occurred at any age. Isolates re- ease.
acting with both antisera against types 1 and 2 were des- Initial reports of S. suis infections were published by
ignated capsular type 1⁄2 (originally RS group). Isolations Jansen and Van Dorssen in the Netherlands (1951) and
of streptococci belonging to group T from tonsillar, vagi- by Field et al. (1954) in England. Since then, S. suis has
nal, and preputial swabs were reported by Clifton- been reported in all countries where the swine industry is
Hadley in 1984. The group T reference strain was desig- important, and for more than a decade, infections asso-
nated S. suis capsular type 15 in 1989 (Gottschalk et al. ciated with this microorganism have been observed in
1989). both traditional and modern intensive swine operations.
Between 1983 and 1995, a total of 32 new capsular In diseased pigs, capsular type 2 predominates in almost
types were described, out of a total number of 35 strains all countries. In Scandinavia capsular type 7 predomi-
(Perch et al. 1983; Higgins et al. 1995). Reference strains nated for several years (Perch et al. 1983; Sihvonen et al.

563
564 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

1988), but in the 1990s, capsular type 2 outnumbered Hogg et al. 1996). S. suis is also increasingly isolated from
type 7 (J. P. Nielsen, pers. comm.—1997). In Japan, cap- a wide range of animal species and birds, and this may
sular type 2 was also the most prevalent serotype (28%), affect some epidemiological aspects of this infection (see
followed by capsular type 7 (11%) (Kataoka et al. 1993). the section on infection in other animal species and
Most S. suis organisms isolated from diseased pigs be- birds). Its presence in the environment is transitory (see
long to a limited number of capsular types, often those the next section).
between 1 and 8 (Galina et al. 1992; Higgins and
Gottschalk 1996; Hogg et al. 1996; Kataoka et al. 1993; Transmission
Prieto et al. 1994; Reams et al. 1996). Transmission of the infection between herds usually oc-
Some strains belonging to less common capsular curs by the movement of healthy carrier pigs. The intro-
types have been associated with severe cases of infection. duction of healthy carriers pigs (breeding gilts, boars,
S. suis capsular type 9 was associated with outbreaks of weaners) into a noninfected herd usually results in the
septicemia, meningitis, and pneumonia in weaned pigs subsequent onset of disease in weaners and/or growing
(Orr et al. 1989; Gogolewski et al. 1990). In 1996, pigs in recipient herds. Sows presumably infect their own
MacLennan et al. reported the first isolation of serotype piglets via the respiratory route (Clifton-Hadley et al.
14 in the United Kingdom. They indicated that although 1986b), but because S. suis is found in the genital and al-
capsular type 2 still predominated (62%), 25% of isolates imentary tracts, piglets are also exposed during the birth
belonged to serotype 14, which affected piglets aged 2–4 process and while suckling (Robertson and Blackmore
weeks and caused arthritis, meningitis, and septicemia. 1989a; Robertson et al. 1991; Dee et al. 1993). It appears
The same year, also in the United Kingdom, Heath et al. that although most weaned piglets carry S. suis strains,
(1996) reported the isolation of capsular type 14 in 22 only a few of them carry strains capable of inducing the
farms with clinicopathological findings similar to those disease after weaning (Pijoan 1996).
associated with serotype 2. S. suis also appears to be easily transmitted via
Capsular type 2 can also be isolated from clinically fomites (Robertson et al. 1991; Dee and Corey 1993). S.
healthy pigs, but its prevalence may be low. British au- suis types 1 and 2 were isolated from the feed troughs of
thors reported that in four herds without any history or piglets and sows (Robertson et al. 1991). Enright et al.
clinical signs of the disease, two were negative for the (1987) demonstrated that flies can carry S. suis type 2 for
presence of type 2, one had a prevalence of 1.5%, and an- at least 5 days and can contaminate materials on which
other a prevalence of 20% (Clifton-Hadley et al. 1984). they feed for at least 4 days. Thus, flies could spread in-
This is in accordance with Canadian studies that demon- fection within farms and between farms. The importance
strated the presence of this serotype in 12% of herds of other animal species or birds as reservoirs or vectors
without clinical signs of infection and in 4% of piglets in of the infection has still to be determined. Carriage by
these herds (Brisebois et al. 1990). Also, serotype 2 was humans also seems possible (Sala et al. 1989).
found in 8 out of 19 nurseries without clinical signs of
infection; in these herds, only 1.5% of piglets were carri- Survival in the Environment
ers in their nasal cavities, whereas capsular types 19 and Studies of the survival of S. suis in the environment have
21 were found in 24% and 19% of the piglets, respective- only been carried out with capsular type 2. This organ-
ly (Monter Flores et al. 1993). ism survived in water at 4˚C for 1–2 weeks. In experi-
Hogg et al. (1996) noted a higher prevalence of mentally inoculated feces, it survived at 0˚C, 9˚C, and
serotypes 9–34 from nasal and vaginal swabs than from 22–25˚C for 104, 10, and 8 days, respectively, and in dust
tissues taken from diseased pigs. It is noteworthy that for 54, 25, and 0 days, respectively. Thus, at a summer-
several capsular types may be present in the same ani- time or weaner-accommodation temperature of
mal. In one study, 31% of pigs had only one serotype of 22–25˚C, the organism could survive about 8 days in fe-
S. suis in their nasal cavities, 38% had two or three ces but less than 24 hours in dust (Clifton-Hadley and
serotypes, and 6% had more than four serotypes (Monter Enright 1984). S. suis capsular type 2 was shown to sur-
Flores et al. 1993). Isolation of multiple serotypes also vive in pig carcasses left rotting on farms. Survival time
has to be taken into consideration in diseased animals was 6 weeks at 4˚C and 12 days at 22–25˚C, potentially
(see the section on diagnosis). providing a source of infection for indirect spread by, for
example, birds, rats, mice, or dogs (Clifton-Hadley et al.
EPIDEMIOLOGY 1986c).
With regard to the cleaning of infected pens, disin-
Natural Habitat fectants and cleansers commonly used in piggeries can
The natural habitat of S. suis is the upper respiratory kill S. suis type 2 in less than 1 minute, even at concen-
tract (particularly the tonsils and nasal cavities) and the trations in distilled water less than those recommended
genital and alimentary tracts of pigs (Clifton-Hadley et by the manufacturers (Clifton-Hadley and Enright 1984;
al. 1986b; Devriese et al. 1991; Robertson et al. 1991; Robertson et al. 1991). The presence of dirt and organic
CHAPTER 41 STREPTOCOCCAL DISEASES Higgins, Gottschalk 565

material protects some organisms from the action of dominated in North America (Reams et al. 1994, 1996;
chemical disinfectants, and hence, removal of surface Hogg et al. 1996). In the Netherlands, S. suis type 2 was
dirt from pens is an important part of the disinfection associated with pneumonia in 42% of the cases, followed
procedure. S. suis type 2 has survived for up to 2 hours at by meningitis, endocarditis, and polyserositis in 18%,
50˚C, but only 10 minutes at 60˚C. Hot water may be 18%, and 10%, respectively (Vecht et al. 1985). In Japan,
used, but water from heated pressure washers cools between 1987 and 1991, 38% of S. suis isolates were from
rapidly on pen surfaces and is usually below 50˚C. Thus, cases of meningitis and 33% from cases of pneumonia
its value is in diluting contaminating microorganisms (Kataoka et al. 1993).
and washing away surface dirt rather than in killing by S. suis isolates belonging to capsular types other than
heat (Clifton-Hadley and Enright 1984). 2 have been recovered from cases of bronchopneumonia
in Denmark (Perch et al. 1983), the Netherlands (Vecht
CLINICAL SIGNS AND LESIONS et al. 1985), Belgium (Hommez et al. 1986), Finland (Sih-
vonen et al. 1988), Australia (Gogolewski et al. 1990),
Even when the pig carrier rate is near 100%, the incidence Canada (Higgins et al. 1990a; Gottschalk et al. 1991a, b),
of the disease varies from period to period and is usually and the United States (Reams et al. 1994). In a retrospec-
less than 5%. According to Clifton-Hadley et al. (1984), tive study of 256 cases associated with S. suis serotypes
affected animals are generally between 5 and 10 weeks of 1–8 and 1⁄2, Reams et al. (1996) indicated that neither
age. In 1996, a report from the United States described clinical signs nor gross lesions were associated with spe-
cases in pigs aged between 1 and 32 weeks, but 75% of cific serotypes. This is in accordance with a report by
them were 16 weeks of age or less (Reams et al. 1996). Heath et al. (1996). Nevertheless, in an outbreak due to
The earliest sign is usually a rise in rectal temperature S. suis type 9, 100% of weaned pigs dying from the dis-
to as high as 42.5˚C. This may occur initially without any ease had arthritis, 91% had meningitis, 73% had intersti-
other obvious signs. It is accompanied by a detectable tial pneumonia, and 42% had endocarditis, and accord-
bacteremia or pronounced septicemia, which, if untreat- ing to the authors, this serotype produced a different
ed, may persist for up to 3 weeks. During this period, distribution of lesions from that reported for serotype 2
there is usually a fluctuating fever and variable degrees of (Vasconcelos et al. 1994). Some strains of certain
inappetence, depression, and shifting lameness (Clifton- serotypes seem to have particular virulence characteris-
Hadley et al. 1984). tics. This could explain cases of severe pneumonia due to
In peracute cases, pigs may be found dead with no serotype 3 in Argentina (Vena et al. 1991) or the recent
premonitory signs. Meningitis is the most striking fea- diffusion of capsular type 14 in the United Kingdom
ture and the one on which a presumptive diagnosis is (Heath et al. 1996). It is noteworthy that type 14 has al-
usually based. Early nervous signs include incoordina- ready infected humans (Gottschalk et al. 1989).
tion and adoption of unusual stances, which soon Significant microscopic lesions are usually limited to
progress to inability to stand, paddling, opisthotonus, the lung, brain, heart, and joints (Reams et al. 1994). The
convulsions, and nystagmus. The eyes are often staring, predominant lesions are suppurative bronchopneumo-
with reddening of mucous membranes (Clifton-Hadley nia, neutrophilic meningitis or encephalitis, and fibrino-
et al. 1986a). Septicemia, arthritis, and pneumonia are purulent or suppurative epicarditis (Sanford and Tilker
less remarkable manifestations of the disease, and a ten- 1982; Erickson et al. 1984; Reams et al. 1994, 1996). In-
tative diagnosis may be difficult to make. Among other terstitial pneumonia is also seen and is considered a le-
manifestations of S. suis infections, there are endocardi- sion secondary to septicemia (Reams et al. 1994). Micro-
tis, rhinitis, abortions, and vaginitis (Sanford and Tilker scopic lesions do not seem to be associated with serotype
1982; Sihvonen et al. 1988). In North America, S. suis is, (Reams et al. 1994). Rare cases of fibrinohemorrhagic
by far, the infectious agent most frequently isolated from pneumonia and septal necrosis have been seen, and it is
cases of endocarditis in pigs. Affected pigs may die sud- suggested that certain strains of S. suis may cause vascu-
denly or show various levels of dyspnea, cyanosis, and lar lesions (Reams et al. 1995). Unusual lesions of hem-
wasting. orrhagic and necrotizing myocarditis and subacute
In the United Kingdom, infections due to S. suis cap- meningoencephalitis and meningoencephalomyelitis
sular type 2 were primarily associated with septicemia have also been reported by Sanford (1987a, b).
and meningitis in weaned pigs (Windsor and Elliott
1975). In North America, early reports indicated that S. PATHOGENESIS
suis was predominantly isolated from cases of pneumo-
nia (Koehne et al. 1979; Sanford and Tilker 1982; Erick- The mechanisms that enable S. suis to disseminate
son et al. 1984). Years later, reports from the United throughout the animal are not well understood. The bac-
Kingdom mentioned septicemia, meningitis, and pol- terium could spread systemically from the nasopharynx,
yarthritis, but rarely pneumonia (MacLennan et al. 1996; occasionally resulting in septicemia and death (Windsor
Heath et al. 1996), whereas pulmonary lesions still pre- 1977). Most studies have focused on capsular type 2 and
566 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

on meningitis. Possible steps involved in the pathogene- be found in several serotypes. Since these proteins are
sis are entry of the bacterium into blood from tonsils, not present in all virulent S. suis isolates, it is now ac-
uptake of bacteria by monocytes, transport of bacteria cepted that these strains possess other virulence factors
to the cerebrospinal fluid (CSF) via the choroid plexus, (Quessy et al. 1994a; Galina et al. 1996).
and stimulation of cytokine production by mono- A hemolysin, designated suilysin, has been associated
cytes/macrophages, which leads to an inflammatory in- with virulence in some capsular type 2 strains. Antibod-
filtrate from the blood to the CSF (Williams 1990; ies against it induced protection in mice and pigs against
Chanter et al. 1993). Studies have pointed out that the in- an experimental challenge (Jacobs et al. 1996). As for
flammatory exudate itself may be detrimental in some MRP and EF, the suilysin is not produced by all virulent
cases and have recommended treatment with antiinflam- isolates. Finally, an IgG-binding protein (Serhir et al.
matory medications (Tunkel et al. 1990). 1993), a galactosyl-(alpha 1-4)-galactose-binding adhesin
In spite of the prevalence of cases of pneumonia in (Tikkanen et al. 1996), and an albumin-binding protein
which S. suis was isolated in pure culture or in associa- (Quessy et al. 1997) were described as possible virulence
tion with other microorganisms, the pathogenesis of this factors. These binding proteins may participate in the es-
condition has not been investigated. To our knowledge, tablishment of the infection, but their role is still not
there is no report of experimental induction of pneumo- considered essential in the pathogenesis of S. suis infec-
nia in swine using S. suis alone. It is likely that the infec- tions.
tion occurs via the respiratory route, then colonization It is likely that concomitant viral infections could po-
takes place along with multiplication of the bacteria. A tentiate the development of lesions in infections due to
study has demonstrated that all S. suis type 2 isolates S. suis. Iglesias et al. (1992) concluded that clinical dis-
from diseased pigs adhered to cryostat sections of new- ease associated with S. suis type 2 was enhanced by con-
born piglet lung tissue, and that isolates from cases of comitant infection with pseudorabies virus. Later, it was
pneumonia adhered in greater number than isolates suggested that the porcine reproductive and respiratory
from cases of meningitis (Gottschalk et al. 1991c). Bind- syndrome virus (PRRSV) predisposes specific-pathogen-
ing of hemagglutinating S. suis strains to frozen sections free (SPF) pigs to infection and disease caused by virulent
of pig pharyngeal tissue was also demonstrated by S. suis serotype 2 (Galina et al. 1994). Swine practitioners
Haataja et al. (1993). Another explanation is that some generally agree with this statement. However, Cooper et
pneumonic infections result from bacteremia, either by al. (1995) have obtained different results, and they con-
direct infection of the alveoli or by transport to the alve- cluded that the NEB-1 PRRSV did not potentiate com-
oli in monocytes (Alexander 1995). mon bacterial pathogens while inducing consistent clini-
The pathogenesis of an infection is influenced by the cal signs, viremia, seroconversion, and microscopic
immune status of the host, environmental factors, and lesions.
virulence attributes of the infectious agent. Researchers
have studied different possible virulence factors of S. su- DIAGNOSIS
is: fimbriae (Jacques et al. 1990), hemagglutinins
(Gottschalk et al. 1990; Haataja et al. 1993), capsular ma- Presumptive diagnosis of S. suis infections is generally
terial (Elliott and Tai 1978; Quessy et al. 1994c; Salasia et based on clinical signs, age of animals, and macroscopic
al. 1994; Charland et al. 1996; Katsumi et al. 1996), cell- lesions. Confirmation is achieved by the isolation of the
wall and extracellular proteins (Vecht et al. 1991, 1996), infectious agent and microscopic lesions in tissues.
and hemolysin (Gottschalk et al. 1995; Jacobs et al. When feasible, collection of more than one alpha-he-
1996). molytic colony from different tissues of the same animal
In several bacterial species, fimbriae and hemagglu- or from different animals in the same herd is recom-
tinins have been associated with adhesion. Haataja et al. mended, because multiple serotypes and strains of S. su-
(1993) indicated that the S. suis activity detected by is can be involved (Reams et al. 1996). Amass (1997) sug-
hemagglutination may account for binding of bacteria to gested periodic culture of CSF from pigs with meningitis
pig tissues, but the presence of other binding mecha- to ensure that the strain(s) of S. suis causing meningitis
nisms is suggested. Studies on the S. suis capsular mater- in the herd have not changed. This could be very helpful
ial have led to different conclusions, but although the when vaccination is considered. Since S. suis may be re-
capsule is one of the important virulence factors (re: covered even from healthy lung tissues (Mwaniki et al.
phagocytosis), it does not appear to be a good virulence 1994), isolates from tissues other than lungs should be
marker. preferred in septicemic cases. In respiratory case acces-
Two proteins of S. suis type 2 were identified as viru- sions, isolation of other infectious agents such as Pas-
lence markers: a muramidase-released protein (MRP) teurella multocida, Actinobacillus pleuropneumoniae, or
and an extracellular factor (EF) (Vecht et al. 1991). Vari- Arcanobacterium pyogenes is frequent, but pure cultures of
ants of these proteins were later described by Vecht et al. S. suis are also isolated (Higgins et al. 1990a; Galina et al.
(1996), who proposed a series of phenotypes which can 1992).
CHAPTER 41 STREPTOCOCCAL DISEASES Higgins, Gottschalk 567

Biochemical identification of S. suis isolates is possi- ies against S. suis have been evaluated. Recent studies
ble with a minimum of tests when serotyping is available have shown that an ELISA using purified capsular anti-
(Higgins and Gottschalk 1990). Devriese et al. (1991) gens had the best specificity (del Campo Sepulveda et al.
suggested the use of only two tests on pig isolates: amy- 1996; Kataoka et al. 1996). However, serology is more
lase positive and Voges-Proskauer (acetoin) negative. It useful in vaccination studies or as a surveillance tool in
has to be noted that isolates from the genital tract of high-health-status swine herds than as a diagnostic tool.
sows form a distinct ecovar with several unusual charac-
teristics such as CO2-dependency (Devriese et al. 1991). TREATMENT
Also, isolates from dogs have been shown to differ from
the common porcine isolates in fermenting mannitol The choice of the best antibacterial agent against S. suis
(Devriese et al. 1992). infections must be based on several criteria such as the
Serotyping is still an important part of the routine di- susceptibility of the organism, the type of infection, and
agnostic procedure. It can be carried out by different the mode of administration. Using the Kirby-Bauer
techniques, but many laboratories have adopted the co- method, susceptibility of S. suis isolates to penicillin ap-
agglutination technique. Since the majority of typeable peared to be between 80% and 95% (Sanford and Tilker
isolates belong to capsular types 1–8 and 1⁄2, it is advis- 1989; Dee et al. 1993; Tarradas et al. 1994). The determi-
able for diagnostic laboratories to only use antisera cor- nation of minimal inhibitory concentrations allowed the
responding to those serotypes and to send untypeable demonstration of a large number of isolates moderately
isolates to a reference laboratory (Higgins and susceptible to penicillin, but the sensitivity rate to amox-
Gottschalk 1996; Hogg et al. 1996). Some isolates cross- icillin and ampicillin was around 90% (Shryock et al.
react with more than one antiserum using the coaggluti- 1992; Dee et al. 1993; Turgeon et al. 1994; Tarradas et al.
nation, as well as the capsular reaction, tests. Some cross- 1994). It is now recommended that penicillin be used on-
reactions can be removed by absorption of antisera, but ly in cases where sensitivity tests have shown that S. suis
others will remain, as for type 1⁄2 (Gottschalk et al. 1989; is sensitive (Sanford 1989). Different authors have re-
Hampson et al. 1993). ported a high degree of resistance of S. suis isolates to
Genetic tools could be of valuable help in distin- some antibacterial agents such as tetracycline, clin-
guishing individual isolates of S. suis, in establishing the damycin, erythromycin, kanamycin, neomycin, and
origin of the infection in a given herd, in monitoring the streptomycin (Estoepangestie and Lämmler 1993; Dee et
kinetics of an outbreak, or in ensuring that the right al. 1993; Reams et al. 1993; Tarradas et al. 1994; Prieto et
strain is included in a vaccine. Plasmid analysis allowed al. 1994; Turgeon et al. 1994; Salmon et al. 1995). Sus-
the differentiation of some bacterial isolates belonging ceptibility to trimethoprim-sulfamethoxazole appeared
to capsular type 2 (Cantin et al. 1992). It was demon- to be variable (Sanford and Tilker 1989; Shryock et al.
strated that an important diversity existed among S. suis 1992; Turgeon et al. 1994; Tarradas et al. 1994). Other re-
isolates even for those belonging to the same serotype ports have mentioned that the most active in vitro an-
(Mogollon et al. 1990). Genomic fingerprinting has al- timicrobial agents against S. suis were enrofloxacin and
lowed the identification of outbreak isolates of S. suis ceftiofur (Dee et al. 1993; Salmon et al. 1995).
(Mogollon et al. 1991). Beaudoin et al. (1992) confirmed Prompt recognition of the early clinical signs of
the genetic diversity between isolates and explained that streptococcal meningitis followed by immediate par-
although isolates from clinically healthy animals were enteral treatment of affected pigs with an appropriate
very heterogeneous, that was not the case for most iso- antibiotic is currently the best method to maximize pig
lates from cases of septicemia. Hampson et al. (1993) in- survival (Amass 1997). Pigs in the early stages of menin-
dicated that genetic diversity was greater than anticipat- gitis may be difficult to detect, and groups of pigs should
ed on the basis of a previous DNA-DNA hybridization be checked 2–3 times daily. Affected pigs hold their ears
study (Kilpper-Balz and Schleifer 1987) and estimated back, squint their eyes, or exhibit dog-sitting (Amass
that the species, as currently defined, was diverse. The 1997). Based on data from studies on the treatment of S.
same authors did not find any tendency for isolates re- pneumoniae meningitis in children (Tuomanen 1993), ad-
covered from healthy pigs to be genetically distinct from junctive therapy with an antiinflammatory agent is rec-
those from diseased animals, nor were there consistent ommended for treatment of S. suis meningitis in pigs
differences between isolates recovered from animals with (Amass 1997). In a segregated early-weaning (SEW) pro-
different disease syndromes. They concluded that viru- gram where postweaning meningitis associated with S.
lence, as well as tissue tropism within the species, does suis was observed, excellent results were obtained when
not appear to be confined to narrow genetic groupings of piglets, in the very early stages of the disease, were in-
the bacteria. At present, there are no known common jected with both penicillin and dexamethasone (Clark
virulence markers for all S. suis serotypes (Vecht et al. 1995). In human medicine, the use of antiinflammatory
1996). agents in cases of bacterial meningitis seems nonetheless
Different serologic tests for the detection of antibod- controversial (Canadian Pediatric Society 1990).
568 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Gentamicin appeared very active against S. suis, and measurable systemic concentrations, although it is
a combination with penicillin is recommended in human known that in humans, only about one-third of an orally
cases of endocarditis due to this agent (Trottier et al. administered dose is absorbed from the intestinal tract.
1991). Higher plasma concentrations are obtained with an
equivalent dose of phenoxymethyl penicillin (McKellar
PREVENTION et al. 1987). Del Castillo et al. (1995) indicated that of all
types of oral treatments with penicillins, only penicillin
Control of Predisposing Factors V in water given to fasted piglets could reach serum con-
S. suis is an example of an emerging infection associated centrations greater than the target concentrations select-
with the intensification of the swine industry. Multiple ed for S. suis isolates. In the same conditions, penicillin G
factors are involved, and among them are the health sta- concentrations were much lower and hardly stayed
tus of the herd (such as concomitant infections and im- above the target serum level. In the presence of food, on-
munosuppression), the degree of virulence of the S. suis ly penicillin V reached the target level, but only in a few
strains involved, and the quality of the environment and piglets. Thus, penicillins should exclusively be orally ad-
of the management. ministered through drinking water to reduce the inter-
Overcrowding, poor ventilation, excessive tempera- ference in absorption due to feed (del Castillo et al.
ture fluctuations, and mixing of pigs with an age spread 1995).
of more than 2 weeks seem to be the most important Amoxicillin has advantages over natural penicillins
stress factors involved in the development of S. suis in- for mass medication. Its bioavailability is similar, but its
fection in susceptible pigs (Dee et al. 1993). Management body clearance is lower than that of penicillin V. Thus,
practices such as all-in/all-out pig flow can help reduce higher serum concentrations can be obtained (J. del
the incidence of the disease. Dividing large buildings in- Castillo, pers. comm.—1997).
to smaller rooms can help minimize temperature fluctu-
ations and the age spread between pigs. Cleaning each Immunization
room between groups of pigs reduces buildup of mi- Until now, most vaccines used to protect against S. suis
croorganisms and improves health status, average daily infections have been autogenous bacterins and results
gain, and feed conversion (Dee et al. 1993). have been inconsistent. The exact reasons for vaccine
failure are still unknown, but possible explanations are
Production Technologies degradation of protective antigens or loss of antigenicity
Medicated early weaning and SEW have been used to im- of the bacteria caused by heat or formalin processing
prove the health status of pigs and to eliminate some in- (Holt et al. 1990a), weak immunogenicity of the capsu-
fectious organisms (Alexander et al. 1980). It is now ac- lated bacteria (del Campo Sepulveda et al. 1996), pro-
cepted that although early weaning can succeed in duction of antibodies to antigens not associated with
controlling diseases such as pleuropneumonia and swine virulence factors (Holt et al. 1988), and absence of some
dysentery, its capacity to reduce or eliminate early colo- strains or serotypes involved in the pathological process.
nizers, such as S. suis, Haemophilus parasuis, and Acti- Reams et al. (1996) focused on this last argument; they
nobacillus suis, is questionable (Pijoan 1996). Control of mentioned that the presence of multiple serotypes in a
these problems requires aggressive use of new diagnostic single herd may account for poor response to vaccina-
techniques, such as serum profiling, together with some tion. Some strains of S. suis can be primary pathogens,
manipulation of sow immunity and disease transmission causing conditions such as septicemia, meningitis, or
in the nursery (Pijoan 1996). Other procedures, such as arthritis, but the majority of isolates are considered op-
nursery depopulation, are under evaluation (Dee and Joo portunistic or secondary pulmonary pathogens, which,
1997). nonetheless, should probably be taken into account in a
vaccination program (Reams et al. 1996).
Antimicrobial Preventive Medication Different types of vaccines have been or presently are
Antimicrobial preventive medication of groups of pigs under investigation. Whole-cell bacterins do not appear
via feed or water against S. suis infections must be based to be good immunogens and vaccine failures are com-
on several considerations. Bioavailability, route of ad- mon (Reams et al. 1996). None of the sera from vacci-
ministration (feed or water), competition (overcrowded nated piglets and sows were protective for mice chal-
pens), and serum concentration needed to kill S. suis lenged with a virulent strain, whereas sera from
have to be considered prior to prophylactic antimicro- hyperimmunized pigs induced a strong passive protec-
bial treatment (del Castillo et al. 1995; Amass 1997). tion (Blouin et al. 1994). This is in accordance with data
Procaine penicillin incorporated into the feed was re- from Holt et al. (1990a).
ported to significantly reduce the prevalence of strepto- Live attenuated vaccines, such as a temperature-sen-
coccal meningitis within a herd (McKellar et al. 1987). sitive mutant of S. suis capsular type 1⁄2, provided protec-
Oral administration of procaine penicillin G resulted in tion of various degrees in mice against challenge by types
CHAPTER 41 STREPTOCOCCAL DISEASES Higgins, Gottschalk 569

1, 2, or 1⁄2 (Kebede et al. 1990). Vaccination of mice with (1986b), only depopulation and restocking with clean
a streptomycin-dependent mutant of serotype 1⁄2 result- pigs will ensure eradication of the infection, and in most
ed in complete protection against challenge with herds this is not economic. Mills (1996) has recently de-
serotypes 1 and 1⁄2 and partial protection against chal- scribed the procedures that were used to establish a pure-
lenge with a capsular type 2 strain (Foster et al. 1994). Al- bred minimal-disease herd from gilts found to be carriers
though described as low, the reversion rate of these mu- of a virulent strain of S. suis type 2. Amass et al. (1996),
tants represents a risk difficult to evaluate. on the other hand, did not recommend such an ap-
Live avirulent strains have also been tested. Protec- proach. They emphasized optimization of management
tion was obtained in mice and/or pigs following the in- and environment of pigs coupled with strategic medica-
oculation of live S. suis capsular type 2 strains (Holt et al. tion of clinically ill animals for control and prevention of
1988; Quessy et al. 1994b; Kobisch et al. 1995; del Cam- mortality caused by streptococcosis.
po Sepulveda et al. 1996). Since live avirulent S. suis Cesarean section can be used to derive pigs free of S.
strains appear to induce a protection similar to that pro- suis from infected dams. Strict biosecurity measures are
duced by live virulent strains, it is suggested that the im- needed and they must include eliminating rodents and
portant immunogens may be different from S. suis viru- flies (Amass 1997).
lence factors. The importance of humoral immunity was Considering that the disease is often associated with
well established by Holt et al. (1988), who succeeded in respiratory problems and with multiple S. suis capsular
passively transferring protection against S. suis type 2. types, that some less common serotypes are more and
Nonetheless, S. suis was shown to survive a period of more involved in severe outbreaks, that reliable diagnos-
time in leukocytes, and the use of live vaccines could, tic or monitoring tools such as serology will not be avail-
along with the induction of humoral immunity, stimu- able in the short term, and, finally, that S. suis is now iso-
late an active cellular immunity. lated from a wide range of animal species and birds, it
Studies have shown that different S. suis type 2 cell- would appear reasonable to direct research efforts to-
wall proteins could induce a good protection (Holt et al. ward control measures rather than eradication.
1990b; Quessy et al. 1994a). However, some proteins
which have a good immunogenic potential, such as the INFECTION IN HUMANS
136 kDa (MRP), are not present in all virulent strains
(Quessy et al. 1994a; Galina et al. 1996). S. suis is a zoonotic agent which deserves attention. De-
Vaccines against extracellular proteins were tested in spite its high prevalence in pigs, human cases are rare,
pigs by Jacobs et al. (1996). One was made of purified but serious. Over 110 cases have been reported world-
suilysin and the other contained most of the extracellu- wide, mostly from northern Europe and Southeast Asia
lar proteins produced by S. suis, particularly the EF, a 110 (Dupas et al. 1992; Kay et al. 1995). Meningitis is the
kDa protein, described by Vecht et al. (1991) and essen- most common manifestation, followed by septicemia
tially free of suilysin. Results in pigs were comparable to and endocarditis. Clinical cases were reported as early as
those already reported with mice (Jacobs et al. 1994). 1968 in the Netherlands and Denmark (Zanen 1970;
Complete protection was obtained with the purified Perch et al. 1968). In Europe, cases have been reported
suilysin and only partial protection with the EF. Al- from the Netherlands, Denmark, Sweden, France, United
though the suilysin is produced by a large number of Kingdom, Belgium, Italy, and Germany (Colaert et al.
serotypes and field isolates from diseased pigs (Jacobs et 1985; Lütticken et al. 1986; Walsh et al. 1992; Perseghin
al. 1995), there are virulent strains in North America that et al. 1995). Cases have occurred in Hong Kong (Ho et al.
do not produce a detectable hemolysin (M. Gottschalk, 1990; Kay et al. 1995), Taiwan (Yen et al. 1994), New
unpublished data). Zealand (Robertson and Blackmore 1989b), and Canada
Some authors have suggested that capsular antigens (Trottier et al. 1991; Michaud et al. 1996). During the
are most important in the protection of mice against a past two decades, S. suis infection has become common
challenge with S. suis (Kebede et al. 1990). On the other in swine, and it is likely that the disease in humans is un-
hand, it was demonstrated that pigs experimentally or derdiagnosed. Unfortunately, because many laboratories
naturally infected with S. suis type 2 only produced low are unaware of this organism, it can easily be mistaken
levels of antibodies against the capsular polysaccharide for an enterococcus, Streptococcus bovis, pneumococcus,
(del Campo Sepulveda et al. 1996), and that a monoclon- or even Listeria species (Chattopadhyay 1979; Arends
al antibody directed against the capsule of S. suis type 2 and Zanen 1988).
was not protective for mice (Charland et al. 1996). The vast majority of human cases have been attrib-
uted to capsular type 2 of S. suis. Only one case each was
ERADICATION associated with capsular types 4 (Arends and Zanen
1988) and 14, which was designated as the reference
Attempts to eradicate the infection have focused only on strain of this serotype (Gottschalk et al. 1991a). In near-
capsular type 2. According to Clifton-Hadley et al. ly all reported cases, patients had close contact with
570 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

pigs—as farmers, butchers, abattoir workers, or veteri- and only one was related to the presence of pigs on the
narians—or handled pork products. The most frequent farm (Hommez et al. 1988). In 1990, a case of meningitis
transmission route is through skin abrasions or cuts, al- in a horse, in contact with cattle only, was reported in
though in many cases, no skin laceration can be shown Belgium; the authors indicated that S. suis appears to be
(Michaud et al. 1996). The finding that liquid soap inac- a member of the intestinal flora of cattle, sheep, goats,
tivates S. suis type 2 in less than 1 minute at a dilution in and horses (Devriese et al. 1990). In Canada, capsular
water of 1 in 500 suggests that washing with soap and type 2 was isolated in pure culture from an aborted
water would be a satisfactory way of removing skin con- bovine fetus, capsular type 16 from a lung of a 6-week-
tamination (Clifton-Hadley and Enright 1984). Since S. old calf affected with chronic purulent bronchopneumo-
suis type 2 can survive in carcasses at 4˚C for 6 weeks, nia, capsular type 31 from a calf with a cerebral edema,
chilled or frozen meat could be a hazard long after being and type 33 from a lamb affected with arthritis (Higgins
butchered. A case has been reported in a woman who en- et al. 1990b, 1995). Devriese and Haesebrouck (1992) re-
joyed eating raw meatballs (Chattopadhyay 1979). ported the isolation of S. suis from three horses, a zebra,
S. suis capsular type 2 has been isolated from the ton- and cats. The same group also confirmed the presence of
sils of two clinically healthy abattoir workers, confirm- S. suis in 20% of dog tonsils (Devriese et al. 1992). Cap-
ing a higher risk for persons in close contact with pigs sular types 1⁄2, 4, 9, 20, 22, and 26 were also isolated from
(Sala et al. 1989). However, the possible role of other an- the tonsils and the anal flora of dogs and cats by Salasia
imal species and birds as reservoirs or as sources of in- et al. (1994). In 1993, S. suis was isolated from a peri-
fection for humans has still to be determined. toneal abscess in a fallow deer (Devriese et al. 1993).
Meningitis due to capsular types 9 and 20, pneumonia
INFECTION IN OTHER ANIMAL due to types 9 and 18 in cattle, and a case of pneumonia
SPECIES AND BIRDS in a horse were reported by Japanese authors in 1993
(Kataoka et al. 1993). More recently, the group from Bel-
Formerly reported from pigs and humans only, S. suis is gium reported the isolation of S. suis from psittacine
now recovered from a variety of different animal species birds, zebra finches, bullfinches, and a duck (Devriese et
and birds. In 1983, Keymer et al. reported isolation of S. al. 1994a). Finally, capsular type 2 was isolated from the
suis type 2 from a raccoon dog fed raw pig meat in a lung of a young wild boar affected with pneumonia (Hig-
wildlife park. Isolates belonging to serotypes 2 and 5 gins et al. 1997). More studies are needed to evaluate the
have been recovered from extramammary suppurative importance of these findings for the epidemiology of S.
lesions in cattle, sheep, and goats. All cases were sporadic suis infection not only in swine but also in humans.

INFECTIONS CAUSED BY S. PORCINUS

In 1984, the name Streptococcus porcinus was proposed pharyngeal or tonsillar surfaces and are carried to the
for a species that would include streptococci of groups E, lymph nodes, primarily of the head and neck region,
P, U, and V, mostly because they shared biochemical where abscesses are formed (Wessman 1986). Losses due
properties (Collins et al. 1984a). S. porcinus has a unique to this disease in the United States were important in the
phenotypic profile in addition to serologic differences 1960s, but its incidence has since declined. The disease is
that can be used to help identify the species. By rRNA se- not recognized as an important economic entity in other
quencing, S. porcinus is closely related to the other beta- countries, where the bacterium represents only a few
hemolytic streptococci, such as groups A, B, and C (Fack- percent of the microorganisms isolated from abscesses
lam et al. 1995). S. porcinus is easily differentiated from S. in swine (Wessman 1986). A report of an outbreak from
suis by the fact that the former is beta-hemolytic and is Spain mentioned that 80% of 50 feeder pigs had
positive to cyclic AMP (cAMP), esculin, and Voges- mandibular and retropharyngeal purulent lymphadeni-
Proskauer tests (Lämmler and Bahr 1996). tis (Real et al. 1992).
S. porcinus group E was associated, particularly in the Antibiotic treatment is not usually successful in ab-
United States, with a contagious clinical entity in grow- scessed swine or in elimination of carriers. However, re-
ing pigs known as streptococcal lymphadenitis, jowl ab- duction in the prevalence of abscesses was achieved by
scesses, or cervical abscesses. Transmission is possible by feeding 138 g chlortetracycline/metric ton (125 g/ton) to
contact, drinking water, or ingestion of food contami- pigs for 4–6 weeks after weaning (Schmitz and Olson
nated by abscess discharge or infected feces. The organ- 1973). Resistance to tetracycline has been recently re-
isms enter the swine host through the mucosa of the ported (Facklam et al. 1995; Lämmler and Bahr 1996).
CHAPTER 41 STREPTOCOCCAL DISEASES Higgins, Gottschalk 571

Vaccination is possible but has not been widely used S. porcinus groups P, U, and V were isolated by Hom-
since the condition is not widespread. The vaccine is ob- mez et al. (1991) from pig lungs, genital organs, and
tained from a chemically derived mutant of the group E brains. However, no histological lesions could be associ-
streptococcus and is sprayed directly onto the tonsillar ated with their presence. More recently, S. porcinus
area, producing an immunological efficacy of more than groups P and V were associated with abortions in pigs
90% (Wessman 1986). It has to be noted that abscesses in (Plagemann 1988; Lämmler and Bahr 1996). Hommez et
pigs may also be caused by a variety of streptococcal and al. (1991) cited a report of Hinterdorfer et al. (1990) in
nonstreptococcal bacteria. which, S. porcinus group P was associated with hemor-
S. porcinus group E can be isolated from tonsils, phar- rhagic tracheitis in pigs.
ynx, and nasal cavities of clinically healthy pigs. It is also According to Wessman (1986), group E streptococci
occasionally found in the vaginal mucus of sows and in were isolated from cases of enteritis and infertility in cat-
the semen and prepuce of boars. It is considered to be tle and sheep, mastitis in cattle, pneumonia in guinea
more of a secondary invader than a primary pathogen in pigs, abscesses in rabbits, and skin and respiratory infec-
conditions such as pneumonia, enteritis, encephalitis, tions in dogs. Finally, these beta-hemolytic streptococci
and arthritis (Wessman 1986). It was isolated in Canada were found in the genitourinary tract of humans (Fack-
from the lungs of a 2-year-old pig affected with an ab- lam et al. 1995). There is, as yet, no evidence that S. porci-
scessative pneumonia, along with other bacterial agents nus has a zoonotic potential.
(A. Désilets, pers. comm.—1995).

INFECTIONS CAUSED BY STREPTOCOCCI BELONGING TO


GROUPS C AND L

ETIOLOGY AND EPIDEMIOLOGY secretions (Jones 1976). Vaginal secretions and milk
from postparturient sows are the most likely sources of
In 1984, Farrow and Collins, using DNA-DNA hybridiza- infection for the piglets (Woods and Ross 1977). Strepto-
tion, DNA base composition, and biochemical tests, in- cocci enter the bloodstream via skin wounds, the navel,
dicated that Streptococcus dysgalactiae, S. equisimilis, and and tonsils. A bacteremia or septicemia occurs, and the
streptococci of Lancefield serologic groups C, G, and L organisms then settle in one or more tissues, giving rise
were a single species. Then group C S. equi and S. zooepi- to arthritis, endocarditis, or meningitis. Insufficient con-
demicus were each classified as a subspecies of the species sumption of colostrum or milk or inadequate levels of
S. equi. In 1996, Vandamme et al. proposed that the name antibodies, especially in gilts, may predispose to disease
S. dysgalactiae subsp. dysgalactiae be used for strains of (Windsor 1978).
animal origin, and the name S. dysgalactiae subsp. equi-
similis be used for human isolates. S. dysgalactiae subsp. CLINICAL SIGNS, LESIONS,
dysgalactiae strains belong to groups C, G, and L; are al- AND DIAGNOSIS
pha-, beta-, or nonhemolytic; do not exhibit streptoki-
nase activity on human plasminogen; and are found in Infection is usually first seen in pigs between 1 and 3
the respiratory and genital tracts of various animal weeks of age. Joint swelling and lameness are the most
species. S. dysgalactiae subsp. equisimilis strains belong to obvious and persistent clinical signs. Elevated tempera-
groups C and G; are beta-hemolytic; exhibit streptoki- tures, lassitude, roughened hair coat, and inappetence
nase activity on human plasminogen; and are found in may also be noted. Early lesions consist of periarticular
the respiratory and genital tracts of humans. edema; swollen, hyperemic synovial membranes; and
In swine, the S. dysgalactiae group C “equisimilis” and turbid synovial fluid. Necrosis of articular cartilage may
the S. dysgalactiae group L serobiovars are all beta-he- be seen 15–30 days after onset and may become more se-
molytic streptococci. Although members of the normal vere. Fibrosis and multiple focal abscessation of periar-
flora, they are considered the most important beta-he- ticular tissues and hypertrophy of synovial villi also oc-
molytic streptococci involved in lesions in pigs, and cur. In lame pigs, up to 12 weeks of age, the causative
these agents were judged to be of etiological significance agents of arthritis were, in decreasing order, “S. equisim-
in autopsy reports (Jones 1976; Hommez et al. 1991). S. ilis” (26.3%), Staphylococcus hyicus subsp. hyicus (24.6%),
dysgalactiae group C streptococci are common in nasal Arcanobacterium pyogenes (13.2%), S. aureus (7.9%), and
and throat secretions, tonsils, and vaginal and preputial Haemophilus parasuis (7.9%). Most of the pigs culled for
572 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

arthritis were under 6 weeks of age (Hill et al. 1996). In TREATMENT AND CONTROL
Denmark, polyarthritis due to hemolytic streptococci
and staphylococci was shown to be common in suckling Since baby pigs are virtually assured of being exposed to
pigs, 17.8% of litters born being affected (Nielsen et al. group C and/or group L streptococci, effective preven-
1975). tive measures should be followed. Adequate intake of
Endocarditis is another condition associated with S. colostrum may ensure that the piglets receive protective
dysgalactiae infection, but it is difficult to diagnose in live antibodies. Traumatic injuries to the feet and legs should
animals. Lesions consist of yellow or white vegetations of be minimized by reducing the abrasiveness of the floor
different sizes, often covering the entire surface of the af- surface in the nursing area.
fected valve (Jones 1980). Beta-hemolytic streptococci are recognized to be sen-
Diagnosis of streptococcal septicemia, arthritis, or sitive to beta-lactam antibiotics. Long-acting antibacteri-
endocarditis is best accomplished by necropsy and bac- al agents may be beneficial, and treatment should be giv-
teriological examination of representative affected pigs. en before the inflammatory process is well advanced
Rather small numbers of organisms or no organisms (Sanford and Higgins 1992). There are no recent reports
may be isolated from affected joints, especially when the about vaccination against groups C or L streptococci. Au-
inflammatory process is advanced. Use of large inocu- togenous bacterins have been used, and a reduction in
lums and enrichment media will enhance the isolation of incidence of arthritis has been reported when sows were
the organism. Bacteria grow readily from portions of the vaccinated before farrowing. Woods and Ross (1977) re-
vegetations in cases of endocarditis (Sanford and Hig- ported that extract vaccines induced higher levels of
gins 1992). complement-fixing antibodies than did whole-cell bac-
terins.

DIARRHEA ASSOCIATED WITH ENTEROCOCCUS DURANS IN PIGLETS

Diarrhea associated with Enterococcus durans (formerly ly. The pathogenesis of enteric disease associated with
Streptococcus durans) was described in piglets between 2 adherent enterococci is unclear. Adherence occurs with
and 20 days of age (Johnson et al. 1983; Drolet et al. the help of fibrillar projections (Tzipori et al. 1984). Di-
1990; Cheon and Chae 1996). Most cases are sporadic, arrhea is not associated with enterotoxin production or
but a recently reported outbreak included 90% of the pig- substantial mucosal injury (Cheon and Chae 1996).
lets in 16 of 20 litters. Diarrhea was observed during a 2- Alone or in conjunction with other agents, they interfere
week period, but mortality was negligible. No other in- with digestion and absorption at the brush border (Tzi-
fectious agent was identified, and E. durans was con- pori et al. 1984). Because of the natural resistance of en-
sidered to be strongly associated with diarrhea in piglets terococci to some antibacterial agents, antimicrobial sus-
(Cheon and Chae 1996). Collins et al. (1984b) have pub- ceptibility testing is advised before the institution of a
lished details about the characterization and identifica- treatment. Due to the lack of knowledge about the clini-
tion of E. durans. Devriese et al. (1994b) have indicated cal and epidemiological aspects of this infection, preven-
that some strains identified as E. durans were in fact E. hi- tive measures are difficult to establish. This condition
rae. has also been reported in foals (Tzipori et al. 1984),
Enterococci are known as part of the intestinal flora, calves (Rogers et al. 1992), and puppies (Collins et al.
but it seems that some strains have the capacity to colo- 1988; Jergens et al. 1991).
nize the mucosal surface of the small intestine extensive-

OTHER STREPTOCOCCI IN SWINE


Other streptococcal species have been isolated from pigs. to these authors, the etiological significance of the S.
Hommez et al. (1991) reported the isolation of beta-he- agalactiae strains in the lesions was beyond doubt. They
molytic group B streptococci (S. agalactiae) from the ton- also suggested that the rare porcine S. agalactiae cases
sils, brain, and intestinal mucosa of pigs, and according had cattle or humans as the infection source. However, a
CHAPTER 41 STREPTOCOCCAL DISEASES Higgins, Gottschalk 573

more recent study has indicated that group B streptococ- Charland, N.; Kobisch, M.; Martineau-Doizé, B.; Jacques,
ci from pigs and nutrias differ from bovine and human M.; and Gottschalk, M. 1996. Role of capsular sialic acid
group B streptococci and seem to play no role in cross-in- in virulence and resistance to phagocytosis of Streptococ-
fections between animals (Wibawan et al. 1993). cus suis capsular type 2. Proc Int Congr Pig Vet Soc
S. bovis, S. alactolyticus, S. hyointestinalis, and S. in- 14:297.
testinalis are not considered potential pathogens in Chattopadhyay, B. 1979. Group R streptococcal infection
pigs. amongst pig meat handlers—A review. Public Health
93:140–142.
REFERENCES Cheon, D.-S., and Chae, C. 1996. Outbreak of diarrhea asso-
ciated with Enterococcus durans in piglets. J Vet Diagn In-
Alexander, T. J. L. 1995. Streptococcus suis: Pathogenesis and vest 8:123–124.
host response. In Proc Allen D. Leman Swine Conf, pp. Clark, L. K. 1995. SEW: Program, problems, performances,
49–53. Minnesota. potential profits and methods of implementation for
Alexander, T. J. L.; Thornton, K.; Boon, G.; Lysons, R. J.; and various herd sizes. In Proc 36th George A. Young Swine
Gush, A. F. 1980. Medicated early weaning to obtain Conf, pp. 1–14.
pigs free from pathogens endemic in the herd of origin. Clifton-Hadley, F. A. 1984. Studies of Streptococcus suis type
Vet Rec 106:114–119. 2 infection in pigs. Vet Res Comm 8:217–227.
Amass, S. F. 1997. Eradication, prevention, and treatment of Clifton-Hadley, F. A., and Enright, M. R. 1984. Factors af-
meningitis in weaned pigs caused by Streptococcus suis. fecting the survival of Streptococcus suis type 2. Vet Rec
In Proc Am Assoc Swine Pract, pp. 411–414. Quebec, 114:585–587.
Canada. Clifton-Hadley, F. A.; Alexander, T. J. L.; Enright, M. R.; and
Amass, S. F.; Wu, C. C.; and Clark, L. K. 1996. Evaluation of Guise, J. 1984. Monitoring herds for Streptococcus suis
antibiotics for the elimination of the tonsillar carrier type 2 by sampling tonsils of slaughter pigs. Vet Rec
state of Streptococcus suis in pigs. J Vet Diagn Invest 115:562–564.
8:64–67. Clifton-Hadley, F. A.; Alexander, T. J. L.; and Enright, M. R.
Arends, J. P., and Zanen, H. C. 1988. Meningitis caused by 1986a. Diagnosis of Streptococcus suis infection in pigs.
Streptococcus suis in humans. Rev Infect Dis 10:131–137. Proc Pig Vet Soc 14:27–34.
Beaudoin, M.; Harel, J.; Higgins, R.; Gottschalk, M.; ———. 1986b. The epidemiology, diagnosis, treatment
Frenette, M.; and MacInnes, J. I. 1992. Molecular analy- and control of Streptococcus suis type 2 infection. In Proc
sis of isolates of Streptococcus suis capsular type 2 by re- Am Assoc Swine Pract, pp. 473–491.
striction-endonuclease-digested DNA separated on Clifton-Hadley, F. A.; Enright, M. R.; and Alexander, T. J. L.
SDS-PAGE and by hybridization with an rDNA probe. J 1986c. Survival of Streptococcus suis type 2 in pig
Gen Microbiol 138:2639–2645. carcasses. Vet Rec 118:275.
Bently, R. W.; Leigh, J. A.; and Collins, M. D. 1991. Intra- Colaert, J.; Allewaert, M.; Magerman, H.; Vandeven, J.; and
generic structure of Streptococcus based on comparative Vandepitte, J. 1985. Streptococcus suis meningitis in man.
analysis of small-subunit rRNA sequences. Int J Syst Acta Clin Belgica 40:314–317.
Bacteriol 41:487–494. Collins, J. E.; Bergeland, M. E.; Lindeman, C. J.; and Duim-
Blouin, C.; Higgins, R.; Gottschalk, M.; and Simard, J. 1994. stra, J. R. 1988. Enterococcus (Streptococcus) durans ad-
Evaluation of the antibody response in pigs vaccinated herence in the small intestine of a diarrheic pup. Vet
against Streptococcus suis capsular type 2 using a double- Pathol 25:396–398.
antibody sandwich ELISA. Can J Vet Res 58:49–54. Collins, M. D.; Farrow, J. A. E.; Katic, V.; and Kandler, O.
Brisebois, L. M.; Charlebois, R.; Higgins, R.; and Nadeau, 1984a. Taxonomic studies on streptococci of serological
M. 1990. Prevalence of Streptococcus suis in four to eight groups E, P, U and V: Description of Streptococcus porci-
week old clinically healthy piglets. Can J Vet Res nus sp. nov. Syst Appl Microbiol 5:402–413.
54:174–177. Collins, M. D.; Jones, D.; Farrow, J. A. E.; Kilpper-Balz, R.;
Canadian Pediatric Society. 1990. Dexamethasone therapy and Schleifer, K. H. 1984b. Enterococcus avium nom. rev.;
for bacterial meningitis: A time for caution. Can Med comb. nov.; E. casseliflavus nom. rev.; comb. nov.; E. du-
Assoc J 142:307–310. rans nom. rev.; comb. nov.; E. gallinarum comb. nov.;
Cantin, M.; Harel, J.; Higgins, R.; and Gottschalk, M. 1992. and E. malodoratus sp. nov. Int J Syst Bacteriol
Antimicrobial resistance patterns and plasmid profiles 34:220–223.
of Streptococcus suis isolates. J Vet Diagn Invest Cooper, V. L.; Doster, A. R.; Hesse, R. A.; and Harris, N. B.
4:170–174. 1995. Porcine reproductive and respiratory syndrome-
Chanter, N.; Jones, P. W.; and Alexander, T. J. L. 1993. NEB-1 PRRSV infection did not potentiate bacterial
Meningitis in pigs caused by Streptococcus suis—A spec- pathogens. J Vet Diagn Invest 7:313–320.
ulative review. Vet Microbiol 36:39–55. Dee, S. A., and Corey, M. M. 1993. The survival of Strepto-
574 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

coccus suis on farm and veterinary equipment. Swine Elliott, S. D. 1966. Streptococcal infection in young pigs. I.
Hlth Prod 1:17–20. An immunological study of the causative agent (PM
Dee, S. A., and Joo, H. S. 1997. Elimination of PRRS virus: Is streptococcus). J Hyg Camb 64:205–212.
it possible? In Proc 28th Annu Meet Am Assoc Swine Elliott, S. D., and Tai, J. 1978. The type-specific polysaccha-
Pract, pp. 393–397. rides of Streptococcus suis. J Exp Med 148:1699–1704.
Dee, S. A.; Carlson, A. R.; Winkelman, N. L.; and Corey, M. Enright, M. R.; Alexander, T. J. L.; and Clifton-Hadley, F. A.
M. 1993. Effect of management practices on the Strepto- 1987. Role of houseflies (Musca domestica) in the epi-
coccus suis carrier rate in nursery swine. J Am Vet Med demiology of Streptococcus suis type 2. Vet Rec
Assoc 203:295–299. 121:132–133.
del Campo Sepulveda, E. M.; Altman, E.; Kobisch, M.; D’Al- Erickson, E. D.; Doster, A. R.; and Pokorny, T. S. 1984. Iso-
laire, S.; and Gottschalk, M. 1996. Detection of antibod- lation of Streptococcus suis from swine in Nebraska. J Am
ies against Streptococcus suis capsular type 2 using a puri- Med Vet Assoc 185:666–668.
fied capsular polysaccharide antigen-based indirect Estoepangestie, S., and Lämmler, C. 1993. Distribution of
ELISA. Vet Microbiol 52:113–125. capsular types 1 to 28 and further characteristics of
del Castillo, J.; Martineau, G. P.; Messier, S.; and Higgins, R. Streptococcus suis isolates from various European coun-
1995. The use of pharmacokinetics to implement peni- tries. Zentrbl Bakteriol 279:394–403.
cillin prophylaxis for streptococcal diseases. In Proc Facklam, R.; Elliott, J.; Pigott, N.; and Franklin, A. R. 1995.
Allen D. Leman Swine Conf, pp. 93–96. Minnesota. Identification of Streptococcus porcinus from human
de Moor, C. E. 1963. Septicaemic infections in pigs caused sources. J Clin Microbiol 33:385–388.
by haemolytic streptococci of new Lancefield groups Farrow, J. A. E., and Collins, M. D. 1984. Taxonomic studies
designated R, S and T. Antonie van Leeuwenhoek on streptococci of serological groups C, G and L and
29:272–280. possibly other taxa. System Appl Microbiol 5:483–493.
Devriese, L. A., and Haesebrouck, F. 1992. Streptococcus suis Field, H. I.; Buntain, D.; and Done, J. T. 1954. Studies on
in horses and cats. Vet Rec 130:380. piglet mortality. I. Streptococcal meningitis and arthri-
Devriese, L. A.; Kilpper-Balz, R.; and Schleifer, K. H. 1988. tis. Vet Rec 66:453–455.
Streptococcus hyointestinalis sp. nov. from the gut of Foster, N.; Staats, J. J.; and Chengappa, M. M. 1994. Isola-
swine. Int J Syst Bacteriol 38:440–441. tion, characterization and protection studies in mice of
Devriese, L. A.; Sustronck, B.; Maenhout, T.; and Haese- a streptomycin-dependent mutant of Streptococcus suis
brouck, F. 1990. Streptococcus suis meningitis in a horse. type 1/2. Vet Res Comm 18:155–163.
Vet Rec 127:68. Galina, L.; Collins, J. E.; Pijoan, C. 1992. Porcine Streptococ-
Devriese, L. A.; Ceyssens, K.; Hommez, J.; Kilpper-Balz, R.; cus suis in Minnesota. J Vet Diagn Invest 4:195–196.
and Schleifer, K. H. 1991. Characteristics of different Galina, L.; Pijoan, C.; Sitjar, M.; Christianson, W. T.;
Streptococcus suis ecovars and description of a simplified Rossow, K.; and Collins, J. E. 1994. Interaction between
identification method. Vet Microbiol 26:141–150. Streptococcus suis serotype 2 and porcine reproductive
Devriese, L. A.; Cruz Colque, J. I.; De Herdt, P.; and Haese- and respiratory syndrome virus in specific pathogen-
brouck, F. 1992. Identification and composition of the free piglets. Vet Rec 134:60–64.
tonsillar and anal enterococcal and streptococcal flora of Galina, L.; Vecht, U.; Wisselink, H. J.; and Pijoan, C. 1996.
dogs and cats. J Appl Bacteriol 73:421–425. Prevalence of various phenotypes of Streptococcus suis
Devriese, L. A.; Desmidt, M.; Roels, S.; Hoorens, J.; and isolated from swine in the U.S.A. based on the presence
Haesebrouck, F. 1993. Streptococcus suis infection in a fal- of muraminidase-released protein and extracellular fac-
low deer. Vet Rec 132:283. tor. Can J Vet Res 60:72–74.
Devriese, L. A.; Haesebrouck, F.; De Herdt, P.; Dom, P.; Gogolewski, R. P.; Cook, R. W.; and O’Connell, C. J. 1990.
Ducatelle, R.; Desmidt, M.; Messier, S.; and Higgins, R. Streptococcus suis serotypes associated with disease in
1994a. Streptococcus suis infections in birds. Avian Pathol weaned pigs. Austr Vet J 67:202–204.
23:721–724. Gottschalk, M.; Higgins, R.; Jacques, M.; Mittal, K. R.; and
Devriese, L. A.; Hommez, J.; Pot, B.; Haesebrouck, F. 1994b. Henrichsen, J. 1989. Description of 14 new capsular
Identification and composition of the streptococcal and types of Streptococcus suis. J Clin Microbiol
enterococcal flora of tonsils, intestines and faeces of 27:2633–2635.
pigs. J Appl Bacteriol 77:31–36. Gottschalk, M.; Lebrun, A.; Jacques, M.; and Higgins, R.
Drolet, R.; Higgins, R.; and Jacques, M. 1990. L’entéropathie 1990. Hemagglutination properties of Streptococcus suis.
associée à Enterococcus (Streptococcus) durans chez le J Clin Microbiol 28:2156–2158.
porcelet. Méd Vét Québec 20:114–115. Gottschalk, M.; Higgins, R.; Jacques, M.; Beaudoin, M.; and
Dupas, D.; Vignon, M.; and Geraut, C. 1992. Streptococcus Henrichsen, J. 1991a. Isolation and characterization of
suis meningitis: A severe noncompensated occupational Streptococcus suis capsular types 9–22. J Vet Diagn Invest
disease. J Occup Med 34:1102–1105. 3:60–65.
CHAPTER 41 STREPTOCOCCAL DISEASES Higgins, Gottschalk 575

———. 1991b. Characterization of six new capsular types Pract, pp. 79–81.
(23 through 28) of Streptococcus suis. J Clin Microbiol Holt, M. E.; Enright, M. R.; and Alexander, T. J. L. 1988. Im-
29:2590–2594. munization of pigs with live cultures of Streptococcus su-
Gottschalk, M.; Petitbois, S.; Higgins, R.; and Jacques, M. is type 2. Res Vet Sci 45:345–352.
1991c. Adherence of Streptococcus suis capsular type 2 to ———. 1990a. Immunization of pigs with killed cultures of
porcine lung sections. Can J Vet Res 55:302–304. Streptococcus suis type 2. Res Vet Sci 48:23–27.
Gottschalk, M.; Lacouture, S.; and D. J. Dubreuil. 1995. ———. 1990b. Protective effects of sera raised against dif-
Characterization of Streptococcus suis capsular type 2 ferent fractions of Streptococcus suis type 2. J Comp Path
haemolysin. Microbiology (Reading) 141:189–195. 103:85–94.
Haataja, S.; Tikkanen, K.; Liukkonen, J.; François-Gerard, Hommez, J.; Devriese, L. E.; Henrichsen, J.; and Castryck, F.
C.; and Finne, J. 1993. Characterization of a novel bacte- 1986. Identification and characterization of Streptococ-
rial adhesion specificity of Streptococcus suis recognizing cus suis. Vet Microbiol 11:349–355.
blood group P receptor oligosaccharides. J Biol Chem Hommez, J.; Wullepit, J.; Cassimon, P.; Castryck, F.;
268:4311–4317. Ceyssens, K.; and Devriese, L. A. 1988. Streptococcus suis
Hampson, D. J.; Trott, D. J.; Clarke, I. T.; Mwaniki, C. G.; and other streptococcal species as a cause of extramam-
and I. D. Robertson. 1993. Population structure of Aus- mary infection in ruminants. Vet Rec 123:626–627.
tralian isolates of Streptococcus suis. J Clin Microbiol Hommez, J.; Devriese, L. A.; Castryck, F.; and Miry, C. 1991.
31:2895–2900. Beta-hemolytic streptococci from pigs: Bacteriological
Harel, J.; Higgins, R.; Gottschalk, M.; and Bigras-Poulin, M. diagnosis. J Vet Med (B) 38:441–444.
1994. Genomic relatedness among reference strains of Iglesias, J. G.; Trujano, M.; and Xu, J. 1992. Inoculation of
different Streptococcus suis serotypes. Can J Vet Res pigs with Streptococcus suis type 2 alone or in combina-
58:259–262. tion with pseudorabies virus. Am J Vet Res 53:364–367.
Heath, P. J.; Hunt, B. W.; Duff, J. P.; and Wilkinson, J. D. Jacobs, A. A. C.; Loeffen, P. L. W.; van den Berg, A. J. G.; and
1996. Streptococcus suis serotype 14 as a cause of pig dis- Storm, P. 1994. Identification, purification, and charac-
ease in the UK. Vet Rec 139:450–451. terization of a thiol-activated hemolysin (suilysin) of
Higgins, R., and Gottschalk, M. 1990. An update on Strepto- Streptococcus suis. Infect Immun 62:1742–1748.
coccus suis identification. J Vet Diagn Invest 2:249–252. Jacobs, A. A. C.; van den Berg, A. J. G.; Baars, J. C.; Nielsen,
———. 1996. Distribution of Streptococcus suis capsular B.; and Johannsen, L. W. 1995. Production of suilysin,
types in 1995. Can Vet J 37:242. the thiol-activated haemolysin of Streptococcus suis, by
Higgins, R.; Gottschalk, M.; and Beaudoin, M. 1990a. Strep- field isolates from diseased pigs. Vet Rec 137:295–296.
tococcus suis infection in swine: A sixteen-month study. Jacobs, A. A. C.; van den Berg, A. J. G.; and Loeffen, P. L. W.
Can J Vet Res 54:170–173. 1996. Protection of experimentally infected pigs by
Higgins, R.; Gottschalk, M.; Fecteau, G.; Sauvageau, R.; De suilysin, the thiol-activated haemolysin of Streptococcus
Guise, S.; and Du Tremblay, D. 1990b. Isolation of Strep- suis. Vet Rec 139:225–228.
tococcus suis from cattle. Can Vet J 31:529. Jacques, M.; Gottschalk, M.; Foiry, B.; and Higgins, R. 1990.
Higgins, R.; Gottschalk, M.; Boudreau, M.; Lebrun, A.; and Ultrastructural study on surface components of Strepto-
Henrichsen, J. 1995. Description of six new Streptococcus coccus suis. J Bacteriol 172:2833–2838.
suis capsular types. J Vet Diagn Invest 7:405–406. Jansen, E. J.; and Van Dorssen, C. A. 1951. Meningoen-
Higgins, R.; Lagacé, A.; Messier, S.; and Julien, L. 1997. Iso- cephalitis bij varkens door streptococcen. Tijdschr Dier-
lation of Streptococcus suis from a young wild boar. Can geneeskd 76:815–832.
Vet J 38:114. Jergens, A. E.; Moore, F. M.; Prueter, J. C.; and Yankauskas,
Hill, B. D.; Corney, B. G.; and Wagner, T. M. 1996. Impor- P. J. 1991. Adherent gram-positive cocci on the intestinal
tance of Staphylococcus hyicus ssp. hyicus as a cause of villi of two dogs with gastrointestinal disease. J Am Vet
arthritis in pigs up to 12 weeks of age. Aust Vet J Med Assoc 198:1950–1952.
73:179–181. Johnson, D. D.; Duimstra, J. R.; Gates, C. A.; and
Hinterdorfer, F.; Köfer, J.; and Hahn, G. 1990. Strep- McAdaragh, J. P. 1983. Streptococcal colonization of pig
tokokkenisolate aus Untersuchungsmaterial von intestine. In Proc Vet Infect Dis Org, Saskatoon, Canada,
Schweinen in der Steiermark. Wiener Tierartzl pp. 292–298.
Monatsschr 77:153–157. Jones, J. E. T. 1976. The carriage of beta-hemolytic strepto-
Ho, A. K. C.; Woo, K. S.; Tse, K. K.; and French, G. L. 1990. cocci by healthy pigs. Br Vet J 132:276–283.
Infective endocarditis caused by Streptococcus suis ———. 1980. Bacterial endocarditis in the pig with special
serotype 2. J Infect 21:209–211. reference to streptococcal endocarditis. J Comp Pathol
Hogg, A.; Amass, S. F.; Hoffman, L. J.; Wu, C. C.; and Clark, 91:11–28.
L. K. 1996. A survey of Streptococcus suis isolations by Kataoka, Y.; Sugimoto, C.; Nakazawa, M.; Morozumi, T.;
serotype and tissue of origin. In Proc Am Assoc Swine and Kashiwazaki, M. 1993. The epidemiological studies
576 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

of Streptococcus suis infections in Japan from 1987 to Monter Flores, J. L.; Higgins, R.; D’Allaire, S.; Charette, R.;
1991. J Vet Med Sci 55:623–626. Boudreau, M.; and Gottschalk, M. 1993. Distribution of
Kataoka, Y.; Yamashita, T.; Sunaga, S.; Imada, Y.; Ishikawa, the different capsular types of Streptococcus suis in nine-
H.; Kishima, M.; and Nakazawa, M. 1996. An enzyme- teen swine nurseries. Can Vet J 34:170–171.
linked immunosorbent assay (ELISA) for the detection Mwaniki, C. G.; Robertson, I. D.; and Hampson, D. J. 1994.
of antibody against Streptococcus suis type 2 in infected The prevalence of Streptococcus suis type 2 in Western
pigs. J Vet Med Sci 58:369–372. Australian piggeries. Austr Vet J 71:385–386.
Katsumi, M.; Saito, T.; Kataoka, Y.; Itoh, T.; Kikushi, N.; and Nielsen, N. C.; Bille, N.; Larsen, J. L.; and Svendsen, J. 1975.
Hiramune, T. 1996. Comparative preparation methods Preweaning mortality in pigs. 7. Polyarthritis. Nord Vet
of sialylated capsule antigen from Streptococcus suis type Med 27:529–543.
2 with specific antigenicity. J Vet Med Sci 58:947–952. Orr, J.; Copeland, S.; and Chirino-Trejo, M. 1989. Streptococ-
Kay, R.; Cheng, A. F.; and Tse, C. Y. 1995. Streptococcus suis cus suis type 9 outbreak in swine. Can Vet J 30:680.
infection in Hong Kong. QJM 88:39–47. Perch, B.; Kristjansen, P.; and Skadhange, K. N. 1968. Strep-
Kebede, M.; Chengappa, M. M.; and Stuart, J. G. 1990. Iso- tococci pathogenic for man. Acta Pathol Microbiol
lation and characterization of temperature sensitive mu- Scand B 74:69–76.
tants of Streptococcus suis: Efficacy trial of the mutant Perch, B.; Pedersen, K. B.; and Henrichsen, J. 1983. Serology
vaccine in mice. Vet Microbiol 22:249–257. of capsulated streptococci pathogenic for pigs: Six new
Keymer, I. F.; Heath, S. E.; and Wood, J. G. P. 1983. Strepto- serotypes of Streptococcus suis. J Clin Microbiol
coccus suis type II infection in a raccoon dog (Nyctereutes 17:993–996.
procyonoide), family Canidae. Vet Rec 113:624. Perseghin, P.; Bezzi, G.; and Gallina, M. 1995. Streptococcus
Kilpper-Balz, R., and Schleifer, K. H. 1987. Streptococcus suis suis meningitis in an Italian blood donor. Lancet
sp. nov.; nom. rev. Int J Syst Bacteriol 37:160–162. 346:1305–1306.
Kobisch, M.; Gottschalk, M.; Morvan, P.; Cariolet, R.; Pijoan, C. 1996. Bacterial respiratory pathogens: What is
Bénévent, G.; and Joly, J. P. 1995. Experimental infection their impact? In Proc 4th Annu Swine Dis Conf Swine
of SPF piglets with Streptococcus suis serotype 2. J Rech Pract, pp. 45–47.
Porcine France 27:97–102. Plagemann, O. 1988. Streptococcus porcinus as a cause of
Koehne, G.; Maddux, R. L.; and Cornell, W. D. 1979. Lance- abortion in swine. Zentrbl Vet B 35:770–772.
field group R streptococci associated with pneumonia in Prieto, C.; Garcia, F. J.; Suarez, P.; Imaz, M.; and Castro, J.
swine. Am J Vet Res 40:1640–1641. M. 1994. Biochemical traits and antimicrobial suscepti-
Lämmler, C., and Bahr, K.-H. 1996. Characterization of bility of Streptococcus suis isolated from slaughtered pigs.
Streptococcus porcinus serogroup P isolated from an J Vet Med (B) 41:608–617.
aborted fetus of a pig. Med Sci Res 24:177–178. Quessy, S.; Dubreuil, D.; Caya, M.; Létourneau, R.; and Hig-
Lütticken, R.; Temme, N.; Hahn, G.; and Bartelheimer, E. W. gins, R. 1994a. Comparison of pig, rabbit and mouse
1986. Meningitis caused by Streptococcus suis: Case re- IgG response to Streptococcus suis serotype 2 proteins
port and review of the literature. Infection 14:181–185. and active immunization of mice against the infection.
MacLennan, M.; Foster, G.; Dick, K.; Smith, W. J.; and Can J Vet Res 58:220–223.
Nielsen, B. 1996. Streptococcus suis serotypes 7, 8 and 14 Quessy, S.; Dubreuil, D.; and Higgins, R. 1994b. Immuniza-
from diseased pigs in Scotland. Vet Rec 139:423–424. tion of mice against Streptococcus suis serotype 2 infec-
McKellar, Q. A.; Baxter, P.; Taylor, D.; and Bogan, J. A. 1987. tions using a live avirulent strain. Can J Vet Res
Penicillin therapy of spontaneous streptococcal menin- 58:299–301.
gitis in pigs. Vet Rec 121:347–350. Quessy, S.; Dubreuil, J. D.; Jacques, M.; Malouin, F.; and
Michaud, S.; Duperval, R.; and Higgins, R. 1996. Streptococ- Higgins, R. 1994c. Increase of capsular material thick-
cus suis meningitis: First case reported in Quebec. Can J ness following in vivo growth of virulent Streptococcus su-
Infect Dis 7:329–331. is serotype 2 strains. FEMS Microbiol Lett 115:19–26.
Mills, G. 1996. Establishing a Streptococcus suis type II-free Quessy, S.; Busque, P.; Higgins, R.; Jacques, M.; and
herd by a combination of medication and removal of Dubreuil, J. D. 1997. Description of an albumin binding
piglets at birth. Irish Vet J 49:674–677. activity for Streptococcus suis serotype 2. FEMS Microbi-
Mogollon, J. D.; Pijoan, C.; Murtaugh, M. P.; Kaplan, E. L.; ol Lett 147:245–250.
Collins, J. E.; and Cleary, P. P. 1990. Characterization of Real, F.; Ferrer, O.; and Rodriguez, J. L. 1992. Purulent strep-
prototype and clinically defined strains of Streptococcus tococcal lymphadenitis in swine. Vet Rec 131:151–152.
suis by genomic fingerprinting. J Clin Microbiol Reams, R. Y.; Glickman, L. T.; Harrington, D. D.; Bower-
28:2462–2466. sock, T. L.; and Thacker, H. L. 1993. Streptococcus suis in-
Mogollon, J. D.; Pijoan, C.; Murtaugh, M. P.; Collins, J. E.; fection in swine: A retrospective study of 256 cases. Part
and Cleary. 1991. Identification of epidemic strains of I. Epidemiologic factors and antibiotic susceptibility
Streptococcus suis by genomic fingerprinting. J Clin Mi- patterns. J Vet Diagn Invest 5:363–367.
crobiol 29:782–787. Reams, R. Y.; Glickman, L. T.; Harrington, D. D.; Thacker,
CHAPTER 41 STREPTOCOCCAL DISEASES Higgins, Gottschalk 577

H. L.; and Bowersock, T. L. 1994. Streptococcus suis infec- Schmitz, J. A., and Olson, L. D. 1973. Prevention of strepto-
tion in swine: A retrospective study of 256 cases. Part II. coccic lymphadenitis in swine: Effectiveness of medica-
Clinical signs, gross and microscopic lesions, and coex- tion with chlortetracycline and isolation from other
isting microorganisms. J Vet Diagn Invest 6:326–334. swine in an infected herd. J Am Vet Med Assoc
Reams, R. Y.; Harrington, D. D.; Glickman, L. T.; Thacker, 162:55–57.
H. L.; and Bowersock, T. L. 1995. Fibrinohemorrhagic Serhir, B.; Higgins, R.; Foiry, B.; and Jacques, M. 1993. De-
pneumonia in pigs naturally infected with Streptococcus tection of immunoglobulin-G-binding proteins in Strep-
suis. J Vet Diagn Invest 7:406–408. tococcus suis. J Gen Microbiol 139:2953–2958.
Reams, R. Y.; Harrington, D. D.; Glickman, L. T.; Thacker, Shryock, T. R.; Mortensen, J. E.; and Rhoads, S. L. 1992. An-
H. L.; and Bowersock, T. L. 1996. Multiple serotypes and tibiotic susceptibility in Streptococcus suis. Curr Therap
strains of Streptococcus suis in naturally infected swine Res 52:419–424.
herds. J Vet Diagn Invest 8:119–121. Sihvonen, L.; Kurl, D. N.; and Henrichsen, J. 1988. Strepto-
Robertson, I. D., and Blackmore, D. K. 1989a. Prevalence of coccus suis isolated from pigs in Finland. Acta Vet Scand
Streptococcus suis types I and II in domestic pigs in Aus- 29:9–13.
tralia and New Zealand. Vet Rec 124:391–394. Tarradas, M. C.; Arenas, A.; Maldonado, A.; Vicente, S.; Mi-
———. 1989b. Occupational exposure to Streptococcus suis randa, A.; and Perea, A. 1994. Susceptibility of Strepto-
type 2. Epidemiol Inf 103:157–164. coccus suis to various antimicrobial agents. J Vet Med (B)
Robertson, I. D.; Blackmore, D. K.; Hampson, D. J.; and Fu, 41:685–688.
Z. F. 1991. A longitudinal study of natural infection of Tikkanen, K.; Haataja, S.; and Finne, J. 1996. The galactosyl-
piglets with Streptococcus suis types 1 and 2. Epidemiol (alpha-1-4)-galactose-binding adhesin of Streptococcus
Infect 107:119–126. suis: Occurrence in strains of different hemagglutination
Robinson, I. M.; Stromley, J. M.; Varel, V. H.; and Cato, E. P. activities and induction of opsonic antibodies. Infect
1988. Streptococcus intestinalis: A new species from the Immun 64:3659–3665.
colons and feces of pigs. Int J Syst Bacteriol 38:245–248. Trottier, S.; Higgins, R.; Brochu, G.; and Gottschalk, M.
Rogers, D. G.; Zeman, D. H.; and Erickson, E. D. 1992. Diar- 1991. A case of human endocarditis due to Streptococcus
rhea associated with Enterococcus durans in calves. J Vet suis in North America. Rev Infect Dis 13:1251–1252.
Diagn Invest 4:471–472. Tunkel, A. R.; Wispelwey, B.; and Scheld, M. 1990. Bacterial
Sala, V.; Colombo, A.; and Gerola, L. 1989. Infection risks of meningitis: Recent advances in pathophysiology and
Streptococcus suis type 2 localizations in slaughtered treatment. Ann Intern Med 112:610–614.
swine. Arch Vet Italiano 40:180–184. Tuomanen, E. 1993. Breaching the blood-brain barrier. Sci
Salasia, S. I.; Lämmler, C.; and Devriese, L. A. 1994. Am (Feb), pp. 80–84.
Serotypes and putative virulence markers of Streptococ- Turgeon, P. L.; Higgins, R.; Gottschalk, M.; and Beaudoin,
cus suis isolates from cats and dogs. Res Vet Sci M. 1994. Antimicrobial susceptibility of Streptococcus
57:259–261. suis isolates. Br Vet J 150:263–269.
Salmon, S. A.; Watts, J. L.; Case, C. A.; Hoffman, L. J.; We- Tzipori, S.; Hayes, J.; Sims, L.; and Withers, M. 1984. Strep-
gener, H. C.; and Yancey, R. J. 1995. Comparison of tococcus durans: An unexpected enteropathogen of foals.
MICs of ceftiofur and other antimicrobial agents against J Infect Dis 150:589–593.
bacterial pathogens of swine from the United States, Vandamme, P.; Pot, B.; Falsen, E.; Kersters, K.; and Devriese,
Canada and Denmark. J Clin Microbiol 33:2435–2444. L. A. 1996. Taxonomic study of Lancefield streptococcal
Sanford, S. E. 1987a. Gross and histopathological findings groups C, G and L (Streptococcus dysgalactiae) and pro-
in unusual lesions caused by Streptococcus suis in pigs. I. posal of S. dysgalactiae subsp. equisimilis subsp. nov. Int
Cardiac lesions. Can J Vet Res 51:481–485. J Syst Bacteriol 46:774–781.
———. 1987b. Gross and histopathological findings in un- Vasconcelos, D.; Middleton, D. M.; and Chirino-Trejo, J. M.
usual lesions caused by Streptococcus suis in pigs. II. Cen- 1994. Lesions caused by natural infection with Strepto-
tral nervous system lesions. Can J Vet Res 51:486–489. coccus suis type 9 in weaned pigs. J Vet Diagn Invest
———. 1989. Streptococcus suis: A strategic update. In Proc 6:335–341.
Am Assoc Swine Pract, pp. 193–195. Vecht, U.; van Leengoed, L. A. M. G.; and Verheijen, E. R. M.
Sanford, S. E., and Higgins, R. 1992. Streptococcal diseases. 1985. Streptococcus suis infections in pigs in the Nether-
In Diseases of Swine, 7th ed. Ed. A. D. Leman, B. E. lands (Part I). Vet Quart 7:315–321.
Straw, W. L. Mengeling, S. D’Allaire, and D. J. Taylor. Vecht, U.; Wisselink, H. J.; Jellema, M. L.; and Smith, H. E.
Ames: Iowa State Univ Press, pp. 588–598. 1991. Identification of two proteins associated with vir-
Sanford, S. E., and Tilker, A. M. E. 1982. Streptococcus suis ulence of Streptococcus suis type 2. Infect Immun
type II–associated diseases in swine: Observations of a 59:3156–3162.
one-year study. J Am Med Vet Assoc 181:673–676. Vecht, U.; Wisselink, H. J.; Reek, F. H.; Stockhofe-Zur-
———. 1989. Streptococcus suis antimicrobial susceptibility. wieden, N.; and Smith, H. E. 1996. Diagnosis of several
Can Vet J 30:679. capsular serotypes of Streptococcus suis by phenotype
578 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

and PCR and the relation with virulence for pigs. Proc Windsor, R. S. 1977. Meningitis in pigs caused by Strepto-
Int Congr Pig Vet Soc 14:298. coccus suis type II. Vet Rec 101:378–379.
Vena, M. M.; Miquet, J. M.; and Isern, S. 1991. Streptococcus ———. 1978. Streptococcal infections in young pigs. Vet
suis isolated from an outbreak of pig pneumonia. Vet Annu 18:134–143.
Arg 8:316–319. Windsor, R. S., and Elliott, S. D. 1975. Streptococcal infec-
Walsh, B.; Williams, A. E.; and Satsangi, J. 1992. Streptococ- tion in young pigs. IV. An outbreak of streptococcal
cus suis type 2: Pathogenesis and clinical disease. Rev meningitis in weaned pigs. J Hyg Camb 75:69–78.
Med Microbiol 3:65–71. Woods, R. D., and Ross, R. F. 1977. Immunogenicity of ex-
Wessman, G. E. 1986. Biology of the group E streptococci: A perimental Streptococcus equisimilis vaccines in swine.
review. Vet Microbiol 12:297–328. Am J Vet Res 38:33–36.
Wibawan, I. W. T.; Lämmler, C.; and Smola, J. 1993. Prop- Yen, M. Y.; Liu, Y. C.; Wang, J. H.; Chen, Y. S.; Wang, Y. H.;
erties and type antigen patterns of group B streptococ- and Cheng, D. L. 1994. Streptococcus suis meningitis
cal isolates from pigs and nutrias. J Clin Microbiol complicated with permanent perceptive deafness: Re-
31:762–764. port of a case. J Formosan Med Assoc 93:349–351.
Williams, A. E. 1990. Relationship between intracellular Zanen, H. C. 1970. Meningitis door Lancefield-groep-R-
survival in macrophages and pathogenicity of Strepto- streptococci bij varkensslachters; een zoonose. Ned Tijd-
coccus suis type 2 isolates. Microbiol Pathogen schr Geneeskd 114:1660–1661.
8:189–196.
42 Swine Dysentery
D. L. Harris, D. J. Hampson, and R. D. Glock

Swine dysentery (SD) is a severe mucohemorrhagic diar-


rheal disease that primarily affects pigs during the grow-
ing-finishing period. This enteric disease is also referred
to as vibrionic dysentery, bloody scours, bloody dysen-
tery, black scours, or mucohemorrhagic diarrhea. SD
was originally described in 1921 and since that time has
been reported to occur in most swine-rearing areas of
the world. An anaerobic spirochete, Serpulina hyodysen-
teriae is considered to be the primary etiologic agent of
SD, and diagnostic tests are based on demonstrating the
presence of this organism. However, other indigenous
microbiota of the intestinal tract may also contribute to
lesion production.
For veterinary practitioners familiar with SD and de-
siring specific directions concerning the formulation of
advice to producers on minimizing the detrimental eco-
nomic effects of the disease, we suggest that the section
Practical Veterinary Advice to Producers (later in chap-
ter) be studied prior to referring to the other sections in
42.1. Pure culture of Serpulina (Treponema) hyo-
the chapter.
dysenteriae, carbol fuchsin stain, as viewed by light
microscopy (Kinyon 1974).
ETIOLOGY

Although SD was first described in 1921 by Whiting et by a loose outer membrane (Fig. 42.2).
al., the etiology remained unknown for 50 years. In 1971 S. hyodysenteriae produces typical signs and lesions of
Taylor and Alexander at Cambridge University reported SD when orally inoculated into conventional or specific-
the successful propagation of a pathogenic anaerobic pathogen-free (SPF) pigs (Taylor and Alexander 1971;
spirochete from an infected pig. Their work was simulta- Glock and Harris 1972; Harris et al. 1972a). Several ani-
neously confirmed at Iowa State University, and the or- mal models have been developed for the study of SD. Le-
ganism was named Treponema hyodysenteriae (Glock and sions closely resembling those of SD can be produced
Harris 1972; Harris et al. 1972a). The organism is now with pure cultures of S. hyodysenteriae in a colonic seg-
classified in the new genus Serpulina, which also includes ment of pigs prepared by surgical anastomosis (Hughes
the species S. innocens (Stanton 1992) and S. pilosicoli et al. 1975), in isolated ligated colonic loops of pigs
(Trott et al. 1996c). A number of other species in the (Whipp et al. 1978), and by oral inoculation of guinea
genus are in the process of being described and pigs (Joens et al. 1978a), mice (Joens and Glock 1979),
named. and young chicks (Adachi et al. 1985). The organism also
The organism is a gram-negative, oxygen-tolerant, has been recovered from naturally infected rheas in the
anaerobic spirochete. It is 6–8.5 µm in length, 320–380 United States, where it causes a necrotizing typhlocolitis
nm in diameter, loosely coiled (Fig. 42.1), motile, and he- (Jensen et al. 1996; Trott et al. 1996a).
molytic on blood agar. S. hyodysenteriae has 7–14 Initial studies on the pathogenicity of S. hyodysenteri-
periplasmic flagella inserted at each cell end, and these ae in gnotobiotic pigs indicated that the organism could
overlap at the middle of the protoplasmic cylinder. The not establish itself in the gut when inoculated orally in
whole cell, including the periplasmic flagella, is covered the absence of other anaerobic bacteria (reviewed by
Harris and Glock 1981). Studies in gnotobiotic mice
Recently the genus name Brachyspira has been suggested for Serpulina (Joens et al. 1982) and pigs (Whipp et al. 1982) clearly
hyodysenteriae, Serpulina innocens, and Serpulina pilosicoli. showed that S. hyodysenteriae is not dependent upon oth-

579
580 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

42.2. Electron micrographs of S.


hyodysenteriae. (A) S. hyodysen-
teriae from the intestinal mucosa of
a pig acutely affected with SD. The
organism is negatively stained with
potassium phosphotungstate. Ar-
rows indicate areas of flagellar
crossover (A. E. Ritchie and L. N.
Brown, unpublished data—1974).
(B) S. hyodysenteriae, isolate 3166
(Norway), in thin section. Trans-
verse aspect illustrating flagellar di-
ameters and apposition to the outer
envelope. The differences in flagellar
diameter are presumably due to dif-
ferent stages of morphogenetic as-
sembly (A. E. Ritchie and L. A.
Joens, unpublished data—1978).
(C) Bacteriophages ubiquitously as-
sociated with S. hyodysenteriae, in
potassium phosphotungstate nega-
tive stain illustrating uniform mor-
phology and close proximity of the
receptor sites on a fragment of the
outer envelope (Ritchie et al. 1978).

er microorganisms for the production of lesions in the ever, is also capable of causing colitis in experimentally
colon and cecum, although the disease produced in gno- infected pigs (Trott et al. 1996b). Nonpathogenic spiro-
tobiotic pigs was less severe than in conventional pigs. chetes probably exist in all pig herds (Hudson et al. 1976;
Anaerobic indigenous bacteria of the colon and cecum Kinyon et al. 1976; Joens et al. 1979b). Kinyon and Har-
are synergistic with S. hyodysenteriae by facilitating colo- ris (1979) originally proposed that nonpathogenic spiro-
nization and by augmenting lesion production. There is chetes isolated from the pig gut be named Treponema
no doubt that S. hyodysenteriae is the only transmissible (Serpulina) innocens. Other nonpathogenic porcine spiro-
agent in natural outbreaks of SD. Its ability to colonize chete species now include Serpulina murdochii and Ser-
can be inhibited by feeding diets that are highly di- pulina intermedia—although the latter has been suspect-
gestible in the small intestine, so that little fermentable ed of being pathogenic under some circumstances
substrate reaches the large intestine (Pluske et al. 1996; (Hampson and Trott 1995; Chapter 40, this volume).
Siba et al. 1996). Kinyon et al. (1977) and Kinyon and Harris (1979)
In their original work, Taylor and Alexander (1971) showed that 25 of 25 strongly beta-hemolytic isolates (S.
demonstrated a second type of anaerobic spirochete in hyodysenteriae) caused SD when orally inoculated into
the feces of normal swine, which morphologically re- SPF pigs, whereas 13 weakly beta-hemolytic isolates were
sembled pathogenic S. hyodysenteriae. It is now clear that not pathogenic.
there are a number of species of weakly beta-hemolytic It became evident that these organisms were not typ-
spirochetes which can be differentiated from pathogenic ical of the genus Treponema (Sellwood 1991). Analysis of
S. hyodysenteriae either by hemolytic pattern on blood 16S RNA sequence data from strains of T. hyodysenteriae
agar plates (Fig. 42.3) or by enteropathogenicity testing and T. innocens and data from other spirochetes and oth-
in pigs or mice. The weakly hemolytic S. pilosicoli, how- er bacteria enabled genetic relationships to be estab-
CHAPTER 42 SWINE DYSENTERY Harris, Hampson, Glock 581

ple laboratory flask apparatus in which enhanced growth


of the organism was demonstrated. Kinyon (1974), Kin-
yon and Harris (1979), and Lemcke and Burrows (1981)
compared the cultural characteristics of S. hyodysenteriae
and strains of weakly hemolytic spirochetes (S. innocens).
The organisms produced acid from a limited number of
carbohydrates, usually glucose and maltose; the end
products of fermentation were acetate, small amounts of
butyrate, H2, and CO2. The organisms degraded pyruvate
but not lactate and were negative in tests for catalase, cy-
tochrome oxidase, hydrogen sulfide, gelatin hydrolysis,
42.3. Zones of beta hemolysis (S. hyodysenteriae) meat hydrolysis, glycine tolerance, starch hydrolysis, and
and weak beta hemolysis (S. innocens) produced on urease. They were positive in tests for bile tolerance, es-
bovine blood agar. culin hydrolysis, and iodoacetic acid tolerance. S. hyo-
dysenteriae may be separated from most but not all
strains of weakly hemolytic spirochetes by differences in
lished. Taking this RNA homology, DNA-DNA reassocia- fructose fermentation and indole production; more reli-
tion, and SDS-PAGE profiles of whole-cell proteins, able is the hemolytic pattern (although in recent years
Stanton et al. (1991) proposed the new genus “Serpula” occasional strains of non–S. hyodysenteriae spirochetes
(Latin for “little serpent”) for both T. hyodysenteriae and have been reported to be strongly hemolytic: Neef et al.
T. innocens strains. These were closely related to each oth- 1994). Enteropathogenicity for conventional pigs is the
er but only distantly related to treponemes and other ultimate test. A fuller description of the metabolic activ-
bacteria. The genus name Serpula was found to duplicate ities of S. hyodysenteriae has been given by Stanton
the name of a fungal genus and has subsequently been (1997).
changed to Serpulina (Stanton 1992). Miao et al. (1978) found that S. hyodysenteriae and S.
In 1982, Lysons et al. described three isolates of S. hy- innocens have an extremely low guanine-plus-cytosine
odysenteriae, cultured from three different pig herds in content (25.8%), as does S. pilosicoli (Trott et al. 1996c).
the United Kingdom without previous histories of SD, The different Serpulina spp. have only low (<50%) de-
that were strongly beta-hemolytic on blood agar but oxyribonucleic acid (DNA) sequence homology, whereas
lacked pathogenicity for conventional experimental pigs different isolates of S. hyodysenteriae exhibit greater than
by oral inoculation. They were indistinguishable from S. 75% DNA sequence homology. A physical and genetic
hyodysenteriae in biochemical, slide agglutination, and map of the chromosome of the type strain of S. hyo-
disk growth-inhibition tests but could be differentiated dysenteriae (B78T) has been prepared (Zuerner and Stan-
from a pathogenic isolate of S. hyodysenteriae by agglu- ton 1994). The chromosome is circular and about 3.2 Mb
tinin cross-absorption tests. These isolates appear to be in size. Ritchie et al. (1978) demonstrated several differ-
avirulent strains of S. hyodysenteriae, and their existence ent types of bacteriophages on the surfaces of pure cul-
may confuse the laboratory diagnosis of SD. Lee et al. tures of S. hyodysenteriae and S. innocens. A generalized
(1993) subsequently described another avirulent isolate transducing bacteriophage from S. hyodysenteriae has
which they recovered from a pig in an Australian herd. been purified and characterized (Humphrey et al. 1997).
Both the British and Australian isolates have reduced A hemolysin produced by S. hyodysenteriae has been
motility in porcine mucus (Milner and Sellwood 1994). demonstrated in filtrates of cultures; the amount was en-
Initially, S. hyodysenteriae could not be grown in a liq- hanced when Na RNA was added (Picard et al. 1978).
uid medium, so solid agar was used. In 1974 Kinyon and Lemcke et al. (1982) used washed cells in a buffer with
Harris reported propagating the organism in a liquid various “carriers”; yeast RNA core was more efficient
medium consisting of trypticase soy broth without dex- than Na RNA, while bovine serum albumin fraction V
trose but with 10% fetal calf serum, under O2-free H2 and and Tween 80 were worse. They also determined that the
CO2. Lemcke et al. (1979) described a procedure for hemolysin of S. hyodysenteriae was oxygen-stable and re-
growth of S. hyodysenteriae in trypticase soy broth with sembled another carrier-dependent toxin, streptolysin S.
10% rabbit serum under O2-free N2 and CO2. Lemcke and It is a small peptide with a molecular weight of 19 kDa
Burrows (1980) propagated the organism in serum-free (Kent et al. 1988), which has proved difficult to separate
medium containing bovine serum albumin and choles- from the carrier. The hemolysin is cytotoxic for a number
terol. Other useful media have been described by Kunkle of tissue culture cells and also primary pig cells, lympho-
et al. (1986) and also by Stanton and Lebo (1988), who cytes being particularly sensitive (Kent and Lemcke
used 1% O2 in the culture atmosphere (S. hyodysenteriae 1984). It has also been shown to damage epithelial cells
can utilize oxygen with the aid of enzymes such as nicoti- in pig ligated intestinal loops (Lysons et al. 1991). The
namide adenine dinucleotide, reduced [NADH], oxidase role of hemolysins in virulence has been emphasized by
[NOX]; Stanton 1989). Kent et al. (1988) described a sim- the fact that hemolysin-negative mutants of S. hyodysen-
582 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

teriae have reduced virulence in pigs (Hyatt et al. 1994). printing” of individual strains, including restriction en-
Recently it has become apparent that there are at least donuclease analysis (Combs et al. 1989, 1992; Harel et al.
three different genes encoding hemolysins of S. hyo- 1994), DNA restriction fragment polymorphism analysis
dysenteriae (Ter Huurne et al. 1994), and their relative im- (Duhamel et al. 1992; Jensen et al. 1992), pulsed field gel
portance is still not clear. electrophoresis (Trott et al. 1996a), and random amplifi-
Baum and Joens (1979a) serologically characterized cation of polymorphic DNA (Dugourd et al. 1996), have
phenol extracts of S. hyodysenteriae and S. innocens. Wa- been developed and are useful tools for epidemiological
ter-phase material (lipopolysaccharide, LPS) from phe- studies.
nol extracts of S. hyodysenteriae was utilized to classify LPS from S. hyodysenteriae has been shown to resem-
isolates into four serotypes by agar gel diffusion tests. ble endotoxin by in vitro tests. Studies using LPS extract-
Mapother and Joens (1985) found three more LPS ed by phenol/water (Nuessen et al. 1982) and endotoxin
serotypes, designated 5, 6, and 7 (Table 42.1). Lemcke extracted by butanol/water (Greer and Wannemuehler
and Bew (1984) meanwhile found three new serotypes, 1989a) showed that it acts as a mitogen for murine
which have not been compared with those of Mapother splenocytes, generates a chemotoxin in fresh swine
and Joens. Having studied LPS antigens of North Amer- serum, causes an increase in uptake of red blood cells by
ican, European, and Australian isolates, Hampson et al. murine peritoneal cells via Fc and C3 receptors, and is cy-
(1989a, b, 1990) proposed that LPS serologic typing totoxic to murine peritoneal cells. The butanol/water
should be modified to a system describing serogroups preparation was also able to stimulate production of in-
(currently 11: Hampson et al. 1997), each of which may terleukin-1 (IL-1) and tumor necrosis factor (TNF) from
contain several distinct serovars. The situation in the murine peritoneal exudate cells. However, the differ-
United States appears relatively simple; most isolates be- ences in virulence between S. hyodysenteriae and S. inno-
long to serotypes 1 and 2 of Baum and Joens (1979a). In cens could not be attributed to the biological properties
Quebec “serotypes 8 and 9” predominate (Li et al. 1991), investigated. Oral inoculation of two types of conven-
whereas isolates from Europe and Australia are more tional mice with S. hyodysenteriae has shown that the LPS
serologically diverse. There is no indication that the viru- may be involved in lesion production. Lesions were pro-
lence of an isolate correlates with its serotype. duced in the colons of endotoxin-sensitive mice but not
Other typing methods suggested for S. hyodysenteriae in endotoxin-resistant mice (Nuessen et al. 1983).
have been agglutinin cross-absorption (Lemcke and Bew Other morphologic types of spirochetes are present
1984), which was likely to be very complex, and multilo- in colons of normal pigs and those affected with SD.
cus enzyme electrophoresis (Lymbery et al. 1990; Lee et These may be confused with S. hyodysenteriae when using
al. 1993). Using the latter technique, S. hyodysenteriae iso- dark-field or phase-contrast microscopy. A commonly
lates have been divided into four broad genetic groups, recognized type is the small spirochete, or pig feces spiro-
one of which contains the avirulent Australian isolate chete (Harris et al. 1972b). It has a corkscrew shape and
SA3. A number of DNA-based techniques for “finger- is approximately 220 nm in diameter with 2–4 periplas-
mic flagella. These should not be confused with the much
larger and snakelike S. hyodysenteriae and S. innocens.
Table 42.1. Serotypes of S. hyodysenteriae based on Cwyk and Canale-Parola (1979) characterized the mor-
water-soluble antigens as revised by Mapother and phology and general physiological characteristics of a
Joens (1985)
small corkscrew-shaped anaerobic spirochete that had
Serotype Origin Isolate been isolated from the colon of a pig (Harris et al. 1972b)
1 Iowa Strain B78a and named it T. succinifaciens. This organism is nonpath-
Missouri Dys 7, Den 191 ogenic for pigs by oral inoculation.
Missouri B234b
Denmark Dys 7, Den 191 EPIDEMIOLOGY
Mexico Strain G
Iowa T6
Minnesota T7 Mapother (1993) reported that 11% of herds in the Unit-
Minnesota B140 ed States were infected with S. hyodysenteriae based on an
2 Iowa B204c LPS-ELISA test. Egan et al. (1982) had previously deter-
Iowa T3, T4, T5 mined that 40% of all pig herds in Iowa, Illinois, and Mis-
Iowa B9605
3 Canada B169 souri were affected with SD by testing sera collected at
4 England A-1 slaughter plants. In comparison, Mapother (1993) indi-
5 Illinois B6933 cated that 33% of Iowa farms were positive for SD. Total
6 Missouri B8044 national losses to the U.S. swine industry have been esti-
7 Netherlands Ack 300/8
mated to be $115.2 million (Duhamel and Joens 1994). A
a
Type species: ATCC 27164; CCM 6063. serologic survey in Western Australia indicated a preva-
b
ATCC 31287. lence of 33% (Mhoma et al. 1992). Taylor (1984) con-
CHAPTER 42 SWINE DYSENTERY Harris, Hampson, Glock 583

ducted a veterinary practitioner survey that indicated ly affected herds may be mild, and disease may not be
that 27% of the feeder pig–producing herds were affect- clinically evident. This situation is due to the protection
ed with the disease. SD is the second most commonly di- afforded suckling pigs by the milk of chronically affected
agnosed pig disease (after enteric colibacillosis) in U.K. dams and the common use of drugs in nursery and grow-
Veterinary Investigation Centers, and the incidence has er rations. Under experimental conditions in which pigs
not dropped noticeably, despite some successful at- are not treated, mortality is often 50%. The severity of ex-
tempts at eradication by depopulation or cleaning/med- perimentally induced SD is related to the amount of
ication plus the availability of disease-free replacement stress on the pig, the quantity of infectious inoculum ad-
breeding stock (Lysons 1992). Roncalli and Leaning ministered, and the size of the pig.
(1976) reported that SD was present in most pig-produc- Clinical signs of SD seem to occur in a cyclic manner.
ing countries of the world. It appears to continue to be In large groups of pigs affected with the disease, symp-
prevalent but at a reduced frequency in most countries. toms may reappear at 3- to 4-week intervals. This reap-
SD is most commonly observed in 15–70 kg pigs, but pearance of symptoms often occurs only after removal of
the disease may also occur in adults, particularly in sows therapeutic levels of drugs from the water or feed. Be-
reared outdoors, and occasionally in suckling piglets. cause S. hyodysenteriae survives in the rearing environ-
Transmission of the disease occurs primarily by inges- ment, some pigs may become reinfected and show symp-
tion of fecal material either from clinically affected pigs toms of the disease. By contrast, many pigs that survive
or from clinically normal pigs that carry the disease. Ex- the acute phase of the disease do recover from SD and
perimentally, transmission has been accomplished by ex- are capable of resisting subsequent challenge. However,
posure of susceptible pigs to previously infected animals chemotherapy during the acute phase may not allow the
that have had no clinical signs for 70 days. SD has also pig to initiate an immune response.
been transmitted by animal caretakers who did not Pigs that have recovered from SD may be asympto-
change clothing or footwear between isolation units con- matic but shed S. hyodysenteriae in their feces. Songer and
taining diseased and healthy pigs. It is believed that the Harris (1978) reported that pigs that had not had clinical
use of improperly designed load-outs which allow pigs to signs of SD for 70 days would transmit the disease to sus-
return to the production unit from a contaminated trans- ceptible sentinel pigs. S. hyodysenteriae shed in the feces
port vehicle has resulted in transmission of SD. Differ- of asymptomatic sows can cause infection of suckling
ences in susceptibility due to breed have not been re- pigs. Depending on the immune status of the sow, the
ported. suckling pigs may not show clinical symptoms until re-
SD causes a tremendous financial loss due to mortal- moval from the sow. Glock et al. (1975) showed that la-
ity, decreased rate of growth, poor feed conversions, and goon water containing effluent of a herd affected with
expenses for chemotherapy. Lysons (1983) calculated SD would produce the disease when administered to sus-
that in-feed medication for SD could cost £1.50–£5.00 ceptible pigs. The organism has been isolated from the
($2.60–$8.60)/pig. Wood and Lysons (1988) demon- pit of a slotted-floor building that housed pigs affected
strated that the feed-conversion efficiency ratio in a herd with the disease. S. hyodysenteriae was isolated from a dog
deteriorated by 0.58 while it had SD, a cost increase of that frequented pens containing pigs affected with SD
£7.31 ($12.60)/pig sold, and the cost of medication was (Songer et al. 1978). Similarly, the organism has been
£1.38 ($2.40)/pig. Although there were pigs with clinical isolated by Joens and Kinyon (1982) from field mice cap-
SD in the herd, the poor feed-conversion efficiency ratio tured on three farms on which there were pigs affected
was attributed mainly to subclinical disease. Walter and with the disease. Hampson et al. (1991) isolated S. hyo-
Kinyon (1990) found that the cost of medicating an in- dysenteriae from a wild rat living on an Australian pig-
fected herd was $8.30/pig marketed and that medication gery.
costs were reduced to $.08 per pig after eradication. Experimentally, Glock et al. (1978) showed that oral
Polson et al. (1992) projected the financial impact of inoculation of dogs and birds (starlings) resulted in fecal
SD via four simulation scenarios: SD-free, endemic SD, shedding of S. hyodysenteriae for 13 days and 8 hours, re-
eradication by medication and disinfection, and total de- spectively. Flies may carry the organism for at least 4
population/repopulation. Net present value, internal hours (J. G. Songer, pers. comm.—1978). Experimental-
rate of return, and benefit/cost ratio were calculated for ly inoculated mice shed S. hyodysenteriae in feces for over
each simulation scenario over a 10-year period. The prof- 180 days (Joens 1980), while rats shed it for only 2 days
it margin per 100 kg liveweight produced was as follows: (Chia 1977). Conventional pigs exposed to the feces of
SD-free, $7.44; endemic SD, $1.67; eradication, $4.93; infected mice developed clinical symptoms of SD within
and depopulation/repopulation, $.07 (Polson et al. 11 days after the first contact with mouse feces (Joens
1992). 1980). Chia and Taylor (1978) demonstrated that S. hyo-
In field cases, morbidity of SD in weanling pigs may dysenteriae will survive in dysenteric feces diluted in wa-
approach 90% and mortality may be 30%, depending on ter for 61 days at 5˚C. The organism survives in feces for
the effectiveness of treatment. The severity in chronical- 7 days at 25˚C. I. T. Egan (pers. comm.—1980) found
584 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

that S. hyodysenteriae would survive in soil for 18 days at vitro. Bowden et al. (1989) concluded that binding ad-
4˚C. However, her attempts to transmit the disease to hesins on S. hyodysenteriae for cultured Henle intestinal
susceptible pigs by placing them on contaminated dirt epithelial (HIE 407) cells may contain sialic acid residues.
lots were unsuccessful. In pure culture, S. hyodysenteriae Cellular damage and invasion did not occur. Whether at-
survives at −80˚C for more than 10 years. The above ob- tachment is an important feature in the disease has not
servations indicate that although S. hyodysenteriae is an been clearly demonstrated. S. hyodysenteriae is an oxy-
anaerobic bacterium, it may have the potential to survive gen-tolerant anaerobe and is able to chemically reduce
under a wide range of environmental conditions. molecular oxygen to water via NOX (Stanton and Jensen
The origin of most epizootics of SD can be explained 1996). Kennedy et al. (1996) prepared nox mutant strains
by the fact that carrier pigs were introduced into a herd. of the organism that were less pathogenic for pigs and
However, outbreaks of the disease do occur in herds with appeared to have less ability to colonize the colon. There-
no history of introduction of new animals. Robertson et fore, wild-type S. hyodysenteriae may be able to associate
al. (1992) found a high odds ratio for both allowing visi- with the oxygen-rich colonic epithelium and thus exert
tors onto farms and the presence of rodents. Other fac- its pathogenicity.
tors that play a role in the induction of symptoms of SD Although the mechanism of tissue destruction has
in infected pigs are stresses, including a change in feed, not been fully elucidated, two toxins (hemolysin and
shipping, castration, overcrowding, and exposure to ex- lipooligosaccharide [LOS]) of S. hyodysenteriae have been
treme changes in environmental temperatures. Where described and characterized that may play a role in lesion
antibiotic medication is routine, any cause of loss of ap- production. The hemolysin produced by S. hyodysenteri-
petite, such as pneumonia, stops the intake of drug; the ae as extracted by Kent et al. (1988) is cytotoxic for sev-
animal can then succumb to SD. The disease seems to oc- eral types of cell cultures and is a major virulence factor
cur frequently in the late summer and fall. in the disease (Hyatt et al. 1994). By contrast, hemolysin
The incubation period of SD is variable. Specific re- prepared from the avirulent strain of S. hyodysenteriae
ports range from 2 days to 3 months, but the disease usu- VS1 was less toxic for cell cultures (Kent and Lemcke
ally occurs within 10–14 days in naturally exposed pigs. 1984). Lysons et al. (1991) demonstrated damage to ep-
Olson (1974) showed that treatment with preventive lev- ithelial cells in germfree pig ligated ileal and colonic
els of sodium arsanilate may be a factor in prolonging loops injected with hemolysin. Damage occurred after
the incubation period. 0.5–1 hour when cell organelles were disrupted; swelling
Experimental oral inoculation of starvation-stressed and shedding of cells had occurred after 3 hours. The
pigs with colonic mucosa from acutely affected donors pattern of damage was similar to that observed by Kang
has resulted in lesion formation within 24 hours. Varia- and Olander (1990) when following early changes at in-
tion in incubation time occurs with varied doses of in- tervals after injecting pig colonic loops with cultures of S.
oculum. Unfortunately, some animals may carry the in- hyodysenteriae. Definitive work on the importance of he-
fection for days with no clinical signs. Unknown molysin followed the cloning of the RNA-core hemolysin
circumstances usually described as “stress” may induce gene (tlyA) by Muir et al. (1992). Hyatt et al. (1994) pro-
the active disease and spread of infection to other ani- duced tlyA mutants and compared them to parent strains
mals. of S. hyodysenteriae by oral inoculation of pigs. The mu-
tant strains of the organism were able to colonize pigs
PATHOGENESIS but did not produce disease. The LOS described origi-
nally by Baum and Joens (1979a) has endotoxic activity
Swine dysentery is associated with the proliferation of S. that may have a direct effect on the epithelial cells of the
hyodysenteriae and perhaps other synergistic supporting large intestine (Nuessen et al. 1983) and may elicit an in-
organisms within the large intestine. Although S. hyo- flammatory response via the stimulation of production
dysenteriae can be seen within epithelial cells and the of IL-1 and TNF (Greer and Wannemuehler 1989b).
lamina propria of tissues with typical lesions, invasion However, treponemal endotoxin was less potent than Es-
may not be essential for lesion production (Glock et al. cherichia coli endotoxin at stimulating IL-1 and TNF and
1974). The organism is motile due to the presence of causing death in galactosamine-sensitized BALB/ByJ
periplasmic flagella, and motility is essential for viru- mice (Greer and Wannemuehler 1989a, b).
lence (Rosey et al. 1994, 1995). Due to its motility, S. hy- Whipp et al. (1978) reported that sterile filtrates of
odysenteriae is efficient at moving through viscous mater- broth cultures of S. hyodysenteriae caused no fluid accu-
ial such as mucus and has been shown to be attracted to mulation in ligated colonic loops of pigs or in sucking
hog gastric mucin by chemotaxis (Kennedy et al. 1988; mice. Furthermore, sterile filtrates did not produce
Milner and Sellwood 1994). It is thus able to get close to changes in Y-1 adrenal cells. Inactivated whole cells and
epithelial cells in the colon (Wilcock and Olander 1979a, sonically disrupted suspensions of S. hyodysenteriae do
b). Knoop et al. (1979) and Bowden et al. (1989) demon- not cause lesions or fluid accumulation in ligated colonic
strated attachment of S. hyodysenteriae to animal cells in segments of pigs.
CHAPTER 42 SWINE DYSENTERY Harris, Hampson, Glock 585

The causative organisms do not invade beyond the of diarrhea. This syndrome is, however, uncommon. The
lamina propria of the large intestine, and the lack of S. first evidence of the disease in most animals is soft, yel-
hyodysenteriae and significant lesions in other organs im- low to gray feces. Partial anorexia and increased rectal
plies that the entire pathogenesis of the disease can be di- temperature of 104–105˚F (40–40.5˚C) may be evident
rectly attributed to the enteric lesions (Kinyon et al. in some animals, but not uniformly so. After a few hours
1980). The primary systemic effects of typical SD are the to a few days following infection, there are large amounts
result of fluid and electrolyte imbalance induced by en- of mucus and often flecks of blood in the feces. As the di-
teritis. The pathogenesis of peracute deaths is not known arrhea progresses, watery stools containing blood, mu-
but could be attributable to endotoxin. cus, and shreds of white mucofibrinous exudate are seen
The pathophysiology of the disease has been studied with concurrent staining of the rear quarters. An arched
by Argenzio et al. (1980), Argenzio (1981), and Schmall back and occasional kicking at the abdomen suggest ab-
et al. (1983). In contrast to what would be expected from dominal pain. Prolonged diarrhea leads to dehydration
histological interpretations, the diarrhea observed is not with increased thirst, and affected animals become
the result of increased mucosal permeability and leakage gaunt, weak, incoordinated, and emaciated.
of protein and extracellular fluid from blood to lumen Most pigs follow the same general sequence of clini-
because of increased tissue hydrostatic pressures. In- cal signs, but the time involved may vary from hours to
stead, the fluid loss appears to be the result of colonic weeks, and arbitrary separation into acute, subacute,
malabsorption as a consequence of the failure of the ep- and chronic categories is often made. The feces in chron-
ithelial transport mechanisms to actively transport sodi- ic forms often contain well-mixed dark blood leading to
um and chloride ions from lumen to blood. Further- so-called black scours. A walk through the facilities hous-
more, cyclic adenosine monophosphate (cAMP) and ing an infected herd may permit the observation of soft,
cyclic guanosine monophosphate (cGMP) levels in yellow or gray feces, some mucoid feces with partially
colonic mucosa of infected pigs were normal, but their mixed blood, and uniformly dark red or brown feces of
response to a stimulus (theophylline) was markedly at- variable consistency.
tenuated. Thus, these studies strongly suggest that an en- The ultimate cause of death in most animals is asso-
terotoxin and/or prostaglandins released from the in- ciated with dehydration, acidosis, and hyperkalemia,
flamed mucosa are not involved in the production of which are discussed in greater detail below. The cause of
diarrhea. Therefore, the pathogenesis of dysentery is un- the occasional peracute deaths is not known.
like the diarrhea induced by enterotoxigenic E. coli or Suckling pigs are not commonly affected and are an
Salmonella spp. exception to the typical form of SD in that they may have
Studies of small-intestine function in infected pigs in- catarrhal enteritis without hemorrhage.
dicated that the glucose-stimulated fluid-absorptive
mechanism was intact and that no additional small-in- LESIONS
testinal secretory component was present. Therefore, the
fluid losses are exclusively the result of failure of the Gross Lesions
colon to reabsorb the animals’ own endogenous secre- The first obvious signs noted in pigs that have died from
tions. Because as much as 30–50% of the extracellular SD are emaciation and a hair coat that is often rough and
fluid volume of these animals, in the form of endoge- stained with feces. Dehydration is usually evident. A con-
nous secretions, is presented daily to the colon for ab- sistent characteristic of the disease is the presence of le-
sorption, colonic absorptive failure alone is sufficient to sions in the large intestine but not in the small intestine,
explain the progressive dehydration and death associat- often with a sharp line of demarcation at the ileocecal
ed with the disease. These studies also imply that oral junction.
glucose-electrolyte solutions would be useful as a thera- Typical changes in the acute stages of SD include hy-
peutic measure in restoring these extracellular fluid loss- peremia and edema of the walls and mesentery of the
es. large intestine. Inflammation may also induce swelling of
mesenteric lymph nodes and formation of small
CLINICAL SIGNS amounts of clear ascitic fluid. Colonic submucosal
glands are often more prominent than normal and ap-
Diarrhea is the most consistent sign of SD, but the sever- pear as white, slightly raised foci on the serosa, particu-
ity may be quite variable. The disease usually spreads larly in subacute or chronic infections. There is obvious
gradually through an infected herd, with new animals swelling of the mucosa, with loss of the typical rugose
being affected each day. The course varies not only be- appearance. The mucosa is usually covered by mucus
tween individual animals within a herd but also between and fibrin with flecks of blood, and the colonic contents
herds. are soft to watery and contain exudate.
Occasional animals are peracutely affected and die af- As the condition progresses, the amount of edema
ter a period of only a few hours with little or no evidence within the wall of the large intestine may decrease. Mu-
586 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

cosal lesions may become more severe with increased fib-


rin exudation and may form thick, mucofibrinous
pseudomembranes containing blood. As lesions become
more chronic, the mucosal surface is usually covered by a
thin, dense, fibrinous exudate, often giving the appear-
ance of marked necrosis, which is quite superficial. Le-
sions can be found in clinically healthy pigs and appear
as discrete areas of reddening of the mucosa, usually
covered with some mucus, but with colon contents of
normal consistency.
Distribution of lesions within the large intestine
varies. In some instances the entire organ may be in-
volved, while at other times only certain segments may
be affected. Lesions tend to become more diffuse in the
later stages of the disease.
Other lesions may include hepatic congestion and hy- 42.4. S. hyodysenteriae in colonic crypt and epitheli-
peremia or congestion of the gastric fundus. However, um (Warthin-Starry; ×750).
these lesions are also associated with other diseases and
are not specific for SD. The fact that the stomach may be
full is often interpreted as meaning the appetite was nor- in the acute phase (Fig. 42.4). Invasion of crypt epitheli-
mal prior to death. It is probably more reasonable to as- um is common.
sociate the full stomach with gastric stasis. Pigs with en- Chronic changes are not very specific, with less hy-
teritis also frequently ingest materials with a high fiber peremia and edema. There is often more advanced su-
content. perficial necrosis of the mucosa, which usually has a
thick, fibrinous pseudomembrane. Ulceration is uncom-
Microscopic Lesions mon unless there are secondary infections with Salmo-
The only significant microscopic lesions are found in the nella spp.
cecum, colon, and rectum. Typical acute lesions include Ultrastructural changes of the intestine during the
obvious thickening of the mucosa and submucosa due to early stages of SD have been characterized. Large num-
vascular congestion and extravasation of fluids and bers of spirochetes with the appearance of S. hyodysen-
leukocytes. There is also hyperplasia of goblet cells, and teriae are found at the luminal surface and within crypts
the epithelial cells at the base of the crypts may be elon- (Fig. 42.5). Adjacent epithelial cells have lesions, includ-
gated and hyperchromic in appearance. Increased num- ing destruction of microvilli, swelling of the mitochon-
bers of various types of leukocytes may be present in the dria and endoplasmic reticulum, loss of other or-
lamina propria, but quantitative evaluation of this lesion ganelles, and decreased density. As damage becomes
is difficult because of the relatively numerous leukocytes more pronounced, the epithelial cells often shrink and
present in normal animals. However, excessive accumu- become dark. S. hyodysenteriae invade epithelial cells,
lation of neutrophils in and around capillaries near the goblet cells, and the lamina propria and may be found in
lumen is commonly observed. large clusters within some epithelial cells, suggesting
Groups of epithelial cells on the luminal surface may that intracellular multiplication may occur (Glock 1971;
separate from the lamina propria early in the course of Taylor and Blakemore 1971; Taylor 1972; Glock et al.
the disease, resulting in exposure of capillaries. Focal ar- 1974).
eas of hemorrhage result and blood is trapped in the
overlying mucus, producing the typical blood-flecked ap- Hematology
pearance of the colonic contents in the acute stages of Hematologic changes in SD include marked alterations
the disease. in many measurable factors. Total leukocyte counts may
Later changes include accumulation of large increase, but not consistently. However, a marked left
amounts of fibrin, mucus, and cellular debris in mucos- shift usually occurs, with high numbers of immature
al crypts and on the luminal surface of the large intes- neutrophils in circulation.
tine. Superficial necrosis of the mucosa may be exten- Other changes include early transient increases in
sive, but deep ulceration is not typical. Increased erythrocyte sedimentation rates and fibrinogen levels.
numbers of neutrophils may be seen throughout the Packed-cell volumes vary but do not indicate significant
lamina propria. Large spirochetes with the appearance of blood loss, and total plasma protein may be elevated as a
S. hyodysenteriae are found in the lumen and within result of dehydration. Serum glutamic-oxaloacetic
crypts at all stages of the disease but are most numerous transaminase levels remain normal.
CHAPTER 42 SWINE DYSENTERY Harris, Hampson, Glock 587

42.5. Electron photomicrograph


of S. hyodysenteriae invading a
colonic epithelial cell (Glock and
Harris 1972).

The most significant changes occur in blood elec- diffuse enteritis limited to the large intestine. Mucofibri-
trolytes. Serum sodium, chloride, and bicarbonate levels nous exudate and free blood in the lumen are character-
decrease, and a marked metabolic acidosis develops, istic but do not eliminate some differential questions.
which may be fatal. Terminal hyperkalemia may be not- Typical microscopic lesions of mucosal edema and mi-
ed and may be a significant cause of death along with crofibrinous enteritis with superficial erosion are helpful
acidemia (Glock 1971). but not very definitive as diagnostic criteria.
Confirmation of a diagnosis of SD requires isolation
DIAGNOSIS and identification of S. hyodysenteriae from the colonic
mucosa or feces, or demonstration of S. hyodys-
The diagnosis of SD depends primarily on differentiat- enteriae–specific proteins or nucleic acid sequences. Early
ing the condition from other potential causes of diar- isolation studies involved passage of mucosal extracts
rhea. Factors that should be considered include history, through filters of decreasing pore size, but this technique
clinical signs, gross lesions, microscopic lesions, as well is no longer routinely used. Songer et al. (1976) devel-
as isolation and identification of S. hyodysenteriae. oped a selective isolation medium that utilized 400
SD may occur as a persistent problem within a herd, µg/mL spectinomycin hydrochloride incorporated in
with phases of increased or decreased severity. Diagnos- trypticase soy agar and 5% bovine or horse blood. Other
tic problems are most likely to occur in a herd in which antibiotics are often added to the medium to increase its
the disease has not been previously diagnosed. History selectivity in terms of inhibition of other fecal microflo-
may be helpful, as it is not unusual to have an outbreak ra, including colistin and vancomycin, both at 25 µg/mL
following the introduction of new animals, presumably (Jenkinson and Wingar 1981), or lower concentrations
carriers, into the herd. Other situations that disrupt the of the previous three antimicrobials with 25 µg/mL spi-
normal environment may also precipitate outbreaks in ramycin and 12.5 µg/mL rifampin also added (Kunkle
herds that have been exposed to S. hyodysenteriae but in and Kinyon 1988). Lemcke and Williams (1984) showed
which the overt disease has not been detected. that the addition of Na RNA (1%) to the selective medi-
Clinical signs such as depression, dehydration, and um containing spectinomycin increased the degree of
diarrhea with mucus and/or blood in the feces are quite hemolysis around colonies of S. hyodysenteriae and en-
suggestive but offer only presumptive evidence. Temper- hanced the viable counts of the organism. Fastidious
ature increases are too moderate and inconsistent to be anaerobe agar (Lab M) gives a more lush surface growth
of any diagnostic benefit. Hematologic changes as previ- of S. hyodysenteriae (M. R. Burrows, pers. comm.—
ously described are characteristic but not sufficiently 1988).
unique to be of any great differential value. Swabs may be used to collect samples of colonic mu-
Further evidence may be obtained by necropsy. Only cosa or feces. The samples should be stored at 4˚C and
acutely affected animals should be examined, since in a kept moist in phosphate-buffered saline or a transport
chronically affected animal various secondary infectious medium until streaked onto the selective agar medium.
agents may cause confusion. The essential finding is a The selective medium should be utilized within 3 days of
588 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

preparation if stored at room temperature. Pure cultures all isolates of S. hyodysenteriae but not with those pre-
are more readily obtained if the agar surface is kept free pared against isolates of S. innocens. Sellwood et al.
of moisture. Recently it has been recommended that S. (1989) identified a prominent 16 kDa protein band pres-
hyodysenteriae be separated from other fecal microflora ent on the outer membrane of S. hyodysenteriae, but un-
by cutting the agar in parallel cuts and streaking perpen- fortunately this lipoprotein is no longer considered to be
dicular to the cuts (Olson 1996). The spirochete grows species specific (Turner et al. 1995). A 30 kDa outer-
out along the cut lines, and if motile contaminants are membrane protein of S. hyodysenteriae, to which a mon-
present, these migrate farther along the cuts. oclonal antibody has been generated, appears to be a bet-
Whatever technique is used, after streaking, the ter species-specific target (Lee and Hampson 1996). The
plates should be placed in an anaerobic container, typi- monoclone can be used to identify S. hyodysenteriae in
cally containing a cold palladium catalyst and a mixture immunofluorescence and agglutination assays, as well as
of N2 or H2 and CO2. The anaerobic container should in Western blot analysis.
contain an indicator (methylene blue) to ensure that a re- The direct culture of S. hyodysenteriae from colonic
duced atmosphere has been obtained within 2–3 hours tissue or feces is central in confirming the diagnosis of
of closure. The plates should be incubated at 37˚C or the disease. But, as with other enteric diseases, the sensi-
preferably 42˚C for at least 24–48 hours prior to their ini- tivity of this procedure is dependent upon the number of
tial observation. On primary isolation, S. hyodysenteriae organisms present in the sample. Pigs acutely affected
produces zones of strong beta hemolysis in which with SD possess large numbers (108–109/g) of S. hyo-
colonies are hard to distinguish, but a film of growth in dysenteriae in their colonic mucosa and feces. By contrast,
the hemolytic zone is grossly visible. As a routine, plates pigs that are asymptomatic may only shed the organism
without evidence of beta hemolysis should be further in- periodically at detectable levels in their feces (Harris et
cubated anaerobically and observed at 48-hour intervals al. 1978). Furthermore, medications commonly used to
for at least 144 hours of incubation. Great care must be treat or prevent SD may reduce the number of organ-
taken to distinguish the weakly beta-hemolytic zones of isms below culturally detectable levels. Therefore, great
growth produced by S. innocens and other Serpulina spp. caution must be used in interpreting the results of nega-
from those produced by S. hyodysenteriae. To adequately tive culture results, particularly from fecal samples. In an
distinguish S. hyodysenteriae from more weakly hemolyt- attempt to increase the sensitivity of detection, nucleic
ic organisms, serial passage on trypticase soy agar with acid probes (Jensen et al. 1990; Combs and Hampson
5% blood may be required. Serial passage is readily ac- 1992; Sotiropoulos et al. 1993) and polymerase chain re-
complished by removing plugs of agar from within the action (PCR) amplification of specific sequences (Combs
hemolytic zones (free of other colony-forming bacteria) et al. 1994; Elder et al. 1994; Harel and Forget 1995;
and transferring the agar to fresh medium. The plugs of Atyeo et al. 1996) have been developed for S. hyodysente-
agar should be dispersed on fresh medium with a trans- riae. Probes have been used to detect 105 cells per gram of
fer loop to free the spirochetes from the agar. pig feces (Jensen et al. 1990), but the technique is quite
Once a strongly hemolytic spirochete is isolated, a va- technically difficult and time-consuming. The PCR is a
riety of other methods have been described that may simpler technique and can detect much fewer organisms,
help to confirm its identity. As discussed earlier, spiro- but unfortunately there often are substances in porcine
chete isolates can be tested for pathogenicity in pigs or fecal samples which inhibit the reaction. For this reason
mice, looking for pathological changes typical of SD. the PCR is usually used on growth from the primary iso-
Other, more practical methods for distinguishing be- lation plate. Even under these circumstances, PCR offers
tween S. hyodysenteriae and other spirochetes include a a more rapid and sensitive approach to diagnosis than
fluorescent antibody test with absorbed antiserum, a does the more routine isolation and biochemical identifi-
growth-inhibition test (Lemcke and Burrows 1979), en- cation of the spirochetes (Atyeo et al. 1996).
zyme analysis (Hunter and Wood 1979), rapid slide ag- The identification by Lysons et al. (1982) of avirulent
glutination (Burrows and Lemcke 1981), and biochemi- strains of S. hyodysenteriae poses a problem for the diag-
cal differential tests, such as the production of indole. nosis of SD. To date, such organisms have been identi-
None of these tests are completely specific by themselves fied only in the United Kingdom and Australia, but diag-
but are useful in conjunction with information on nostic laboratories throughout the world should be
strength of hemolysis. aware of their existence. Until we have simple laborato-
Improved identification methods include the demon- ry-based assays to detect virulence determinants, it is ul-
stration of S. hyodysenteriae–specific antigens or nucleic timately necessary to conduct in vivo tests to determine
acid sequences. Some candidate antigens have been iden- if strongly beta-hemolytic isolates from herds without
tified. Baum and Joens (1979b) described a protein iso- typical signs and lesions of SD are pathogenic S. hyo-
lated and purified from phenol-phase material of a phe- dysenteriae.
nol/water extraction of isolate B169 of S. hyodysenteriae It is common practice to conduct direct examination
by starch-block electrophoresis. In agar gel diffusion of smears prepared on slides from colonic mucosa or fe-
tests this protein reacted with antisera prepared against ces of pigs suspected of being affected with SD. Workers
CHAPTER 42 SWINE DYSENTERY Harris, Hampson, Glock 589

in the United Kingdom have utilized an absorbed anti- (1983) concluded that this ELISA could be used for de-
serum in an indirect fluorescent antibody test to detect S. termination of the prevalence of SD but not for detec-
hyodysenteriae (Hunter and Saunders 1977). Joens et al. tion of individual pigs affected with the disease. LPS-
(1978b) in the United States and Lysons and Lemcke based ELISA systems also have the disadvantage that
(1983) in the United Kingdom questioned the specificity they require a knowledge of the serotypes of organisms
of this test. However, improvements have been made to present in the herds to be tested (Mhoma et al. 1992).
the procedure in the United Kingdom by cross-absorbing Further research must be done before national con-
the serum with a number of strains of weakly hemolytic trol schemes or import-export regulations are formulat-
spirochetes to increase its specificity. The use of specific ed based on serologic tests. The specificity of such tests
monoclonal antibodies, as described by Lee and Hamp- could be improved by the identification and use of an S.
son (1996), will improve this situation further. The ex- hyodysenteriae–specific antigen that is recognized by in-
amination of smears for the presence of S. hyodysenteriae fected pigs: the 30 kDa outer-membrane protein de-
is less sensitive than isolation and identification of the scribed by Lee and Hampson (1996) is one possible can-
organism (Lysons and Lemcke 1983). Direct microscopic didate.
examination revealing large numbers of organisms re- Proliferative enteritis (porcine intestinal adenomato-
sembling S. hyodysenteriae in samples from diseased pigs sis) may clinically resemble the symptoms of SD. How-
can be used as presumptive evidence of SD, at least until ever, SD does not affect the small intestine, whereas pro-
completion of cultural examination. In such cases the liferative enteritis usually occurs primarily in the small
possibility of the condition being porcine intestinal intestine. Necrotic debris and blood that originated in
spirochetosis (PIS) caused by S. pilosicoli (see Chapter 40) the anterior intestine will often be present in the large in-
also must be considered. Mucosal or fecal smears may be testine and feces of pigs affected with proliferative en-
examined as wet mounts by phase-contrast or dark-field teritis. Weakly hemolytic spirochetes are commonly iso-
microscopy or stained with crystal violet, dilute carbol lated from cases of proliferative enteritis, although they
fuchsin, or Victoria blue 4-R stains and viewed by light are not known to play a role in the disease. A definitive
microscopy. Serpulina spp. are easily recognized by these diagnosis of proliferative enteritis depends on the lack of
techniques but cannot be differentiated. Electron micro- isolation of S. hyodysenteriae, lesions characteristic of
scopic examination of negative-stained preparations of proliferative enteritis, and the presence of organisms
intestinal or fecal fluids may detect the organisms but with the appearance of Lawsonia intracellularis in the in-
does not distinguish between the species. The organisms testinal epithelium.
can also be demonstrated by light microscopy in sections Salmonellosis, in particular Salmonella choleraesuis
of the colonic mucosa by staining with Warthin-Starry, infection, can be easily confused with SD because symp-
Victoria blue 4-R, or Goodpasture stains. toms and lesions may be quite similar. It should be kept
Campylobacter (synonym Vibrio) spp. may appear in in mind that hemorrhage or necrosis in parenchymatous
increased numbers in some affected animals, but such organs and lymph nodes may be expected with salmo-
organisms are often numerous in normal animals. There- nellosis but not with SD. Mucosal lesions may be found
fore, no diagnostic significance should be associated with in the small intestine with salmonellosis but not with un-
the observation or isolation of Campylobacter spp. complicated SD. Deep ulcerative enteric lesions are also
Several serologic tests have been reported that detect much more typical of salmonellosis. The definitive diag-
antibodies to S. hyodysenteriae in serum of experimental- nosis depends on lack of S. hyodysenteriae in the mucosa
ly affected pigs. To date, these tests have not been based of the large intestine and the isolation of Salmonella spp.
on species-specific antigens and consequently have had from the intestine or other organs such as lymph nodes
low specificity and/or sensitivity. Tests include macro- or spleen. The mere isolation of Salmonella spp. does not
scopic agglutination (Joens et al. 1978c), indirect fluores- constitute a positive diagnosis, since both normal ani-
cent antibody, passive hemolysis (Jenkins et al. 1976), mals and animals with SD may harbor Salmonella spp.
enzyme-linked immunosorbent assays (ELISAs) using Trichuriasis may be differentiated on the basis of the
various plate-coating antigens (P. Høgh, pers. comm— presence of numerous Trichuris suis in the large intestine
1979; Burrows et al. 1984), and agar gel diffusion (L. A. and the lack of S. hyodysenteriae. It should be noted that
Joens, pers. comm.—1979). Egan et al. (1983) designed a concurrent infections can occur, and possible potentia-
method for determining the accuracy of serologic testing tion of SD by T. suis has been postulated. Here again,
for SD as a means of determining the prevalence of the demonstration of the absence of S. hyodysenteriae is nec-
disease and the usefulness of serology for diagnosis. essary to eliminate the possibility of both infections be-
They compared the accuracy and sensitivity of the mi- ing present.
crotitration agglutination test and an LPS-based ELISA Gastric ulcers and other hemorrhagic conditions may
(Joens et al. 1981) by evaluating the level of antibodies in cause the presence of blood in the feces and confusion
the sera of three age groups of swine from 22 farms; 14 with SD. These conditions are easily differentiated at
of the farms had a previous history of SD and 8 had no necropsy since they generally involve the anterior diges-
history that the disease had ever occurred. Egan et al. tive tract.
590 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

“Colitis” is a diet-related disease syndrome of grow- chlortetracycline, virginiamycin, and lincomycin (Harris
ing pigs that can resemble SD both in clinical signs and and Glock 1975).
in postmortem appearance (Lysons et al. 1988). There Methods for in vitro sensitivity testing of antimicro-
has been some confusion over this condition, and some bials have been developed utilizing either liquid or solid
reported cases actually may have been spirochetal diar- media (Kitai et al. 1979; Kinyon and Harris 1980). Iso-
rhea/PIS, caused by infection with Serpulina pilosicoli lates resistant to certain antimicrobials have been re-
(Taylor et al. 1980; Hampson and Trott 1995; Chapter ported from various parts of the world. In general, iso-
40, this volume). Typically, pigs aged 7 weeks or more are lates resistant to carbadox or tiamulin are uncommon
affected. They have a watery scour, or sometimes just soft while resistance to other antimicrobials is reported more
feces, and lose body condition. Some pigs will have mu- frequently. Walter and Kinyon (1990) found that 38 iso-
cus and/or blood in the feces. Lesions are confined to the lates of S. hyodysenteriae from the United States were ful-
colon. In the early stages of the disease, the entire large ly susceptible to carbadox, dimetridazole, and tiamulin
intestine is filled with liquid contents and there is a mild while many isolates were resistant to bacitracin MD, gen-
reddening of the colon. Pigs in which the disease persists tamicin, and lincomycin. Isolates resistant to sodium ar-
can become thin, excrete mucus, and have a mucofibri- sanilate, lincomycin, and tylosin have also been reported
nous exudate on the mucosal surface of the colon. To from the United States (Kinyon and Harris 1980). Iso-
eliminate SD from the diagnosis, it is necessary to autop- lates resistant to (in order of increasing frequency of re-
sy pigs in the early stages of the disease and to carry out sistance) carbadox, dimetridazole, ronidazole, and lin-
extensive screening for the presence of S. hyodysenteriae. comycin have been reported from Canada (Messier et al.
In some laboratories samples from such cases are rou- 1990), and all isolates were found to be susceptible to tia-
tinely subjected to PCR for both S. hyodysenteriae and S. mulin. Ronne and Szancer (1990) found 25 Danish iso-
pilosicoli (Atyeo et al. 1996). Clearly, PIS represents the lates to be fully susceptible to tiamulin and most or all
most difficult differential diagnosis, since it so closely re- isolates resistant to lincomycin, dimetridazole, and ty-
sembles mild cases of SD. Although it responds to simi- losin. Buller and Hampson (1994) tested 30 Australian
lar treatment, an accurate diagnosis is important be- isolates and found 2 resistant to dimetridazole and 1 re-
cause, in general, PIS has much less economic impact in sistant to tiamulin; 90% of isolates were resistant to lin-
individual herds than SD does. Furthermore, it appears comycin and tylosin. Molnar (1996) tested 332 Hungari-
that aspects of the epidemiology of PIS may differ from an isolates of S. hyodysenteriae isolated between 1978 and
those described here for SD (Hampson 1997). 1992 and found all isolates to be susceptible to carbadox,
but isolates resistant to dimetridazole, lincomycin, and
TREATMENT tiamulin were reported. Binek et al. (1994) tested 83 Pol-
ish isolates of S. hyodysenteriae isolated from 1982 to
Pigs acutely affected with SD should be medicated via the 1993, finding all isolates remained susceptible to tia-
water. Usually, animals in the early stages of the disease mulin while complete reistance had developed for lin-
consume very low amounts of feed. This precludes plac- comycin, ronidazole, and metronidazole during the peri-
ing therapeutic levels of drugs in the feed as a method of od.
treatment. Occasionally moribund animals may require As with the treatment of any infectious disease, suc-
systemic injection of drugs, but this is usually impracti- cessful therapy is dependent on an accurate differential
cal in large-herd situations. Medication is often added to diagnosis distinguishing SD from other enteric bacterial
the feed as a method of preventing SD. or parasitic diseases. Government regulatory approval
Table 42.2 lists the dosage levels, duration of admin- (drug availability), antimicrobial resistance patterns,
istration, and withdrawal times of various drugs used for and cost typically dictate drug selection and usage. Po-
the treatment and/or prevention of SD as approved by tential effectiveness against concurrent infections, either
the U.S. Food and Drug Administration. Harris and enteric or respiratory, and the goal of treatment (con-
Glock (1975, 1981, 1986) have reviewed other drugs that trolling vs. eradicating the disease) are also important
have been used to either treat and/or prevent SD. Treat- considerations in drug selection. Ensuring adequate con-
ment of SD is usually accomplished by administering ei- sumption (dosage and duration) of properly medicated
ther chemotherapeutics or antibiotics, sometimes sup- feed and/or water is essential to achieve desired results
plemented with electrolytes. with those routes of administration.
Chemicals that have been reported to be effective for
the treatment and/or prevention of SD are sulfon- Elimination of SD by Medication
amides, nitrofurans, quinoxalines, ionophores, mutilins, and Cleaning
and the nitroimidazoles. The use of drugs on all ages of pigs on the premises and
The following antibiotics have been reported to be ef- careful cleanup and disinfection procedures have been
fective for the treatment and/or prevention of SD: strep- successfully utilized to eradicate SD from affected herds
tomycin, bacitracin, neomycin, tylosin, gentamicin, without depopulation. Songer and Harris (1978) report-
CHAPTER 42 SWINE DYSENTERY Harris, Hampson, Glock 591

Table 42.2. Dosage level, duration of administration, and withdrawal times for various drugs used for the
treatment and prevention/control of SD as approved by the U.S. Food and Drug Administration
Dosage by Dosage in Dosage in Duration Withdrawal
Drug Injection Water Feed (days) (days)
Treatment of SD
Bacitracin 1000 mg/gal, 7–14a None
264 mg/L
Gentamicin 50 mg/gal, 3 3 (oral solution)
13.2 mg/L 10 (sol. powder)
Lincomycin 3.8 mg/lb/day, 5–10a None
8.4 mg/kg/day
(250 mg/gal,
66 mg/L)
100 g/t 21a None
Tiamulin 3.5 mg/lb/day, 5a 3
7.7 mg/kg/day
(227 mg/gal,
60 mg/L)
200 g/t 14a 7
Tylosin 4 mg/lb IM ≤3 14
(8.8 mg/kg)
twice daily
250 mg/gal, 3–10 2
66 mg/kg
Virginiamycin 100/g/t 14 None

Prevention of SD
Bacitracin 250 g/t None
Carbadox 50 g/t 42

Lincomycin 40 g/ta None


Tiamulin 35 g/ta 2
Tylosin 100 g/t ≥3 weeks None
Followed by To market None
40 g/t
Virginiamycin 25–50 g/t Not over None
120 lbs
(54 kg) BW
Note: The information in this table is an abbreviated summation of product labeling. For more complete information review the prod-
uct label. BW = body weight. IM = intramuscular.
a
Not approved for use in swine over 250 lbs (109 kg) body weight.

ed that S. hyodysenteriae could be completely eliminated The former approach depended on intensive, high-level
from pigs by treatment with dimetridazole or ronidazole medication to all pigs via drinking water or oral dosing;
when pigs were housed in conditions minimizing expo- the latter concentrated on elimination of S. hyodysenteri-
sure to their feces. Rainier et al. (1980) showed that car- ae from sows as they came into the farrowing house.
badox would also eliminate the organism from pigs un- General guidelines for eradicating swine dysentery
der similar conditions. Taylor (1980) has also eliminated from a herd without depopulation are given below (Har-
SD using tiamulin. Veterinarians from several countries ris and Glock 1981):
have reported the successful elimination of SD from af-
fected herds by medication of all pigs on the premises 1. A warm season in which temperatures are higher
(Glock 1979). During the medication period the facilities than 15˚C (59˚F) is preferable.
should be cleaned and disinfected frequently to mini- 2. The number of animals in the herd should be de-
mize the environmental survival of S. hyodysenteriae. In creased to as few as possible.
addition, the rodent population on the farm should be 3. If farrowings occur in batches, the recommended
reduced and, in the pig buildings, should be eliminated. time to eradicate the disease is when no suckling pigs are
The regimes for eradication have varied greatly. The on the farm.
medication period has been as short as 5 days (Moller 4. An effective rodent control program, including
1984) or as long as 6 months (Wood and Lysons 1988). renovation of buildings, should be instituted.
592 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

5. All liquids should be removed from pits within eliminate S. hyodysenteriae from the intestinal tract to
buildings in which pigs are housed. preclude the introduction of the disease into the herd.
6. Any buildings that do not contain pigs should be It is well accepted that although SD is very prevalent,
cleaned and disinfected. the disease may be readily eliminated by surgical proce-
7. All pigs on the farm should be medicated simulta- dures used for repopulation. Once a herd is established
neously with drugs known to eliminate S. hyodysenteriae in the absence of the disease, such precautions as those
from the pig intestinal tract. Care should be taken to en- taken by the SPF or minimal-disease associations, the Pig
sure that inclusion rates of antibacterials in the diet of Health Control Association in the United Kingdom
animals, such as adult stock, on restricted feeding is ad- (Goodwin and Whittlestone 1984), and the Health Con-
justed so that the animals receive the correct dose/kg of trol Programs of certain breeding-stock companies
body weight. (Alexander 1985) can prevent recurrence of S. hyodysen-
8. After 1 week of medication, all equipment used teriae. For example, newly established surgically derived
for handling pigs, feed, and manure should be cleaned herds in some locales may eventually become infected
and disinfected. with such agents as Bordetella bronchiseptica and My-
9. During the medication period, an attempt should coplasma hyopneumoniae but often can be maintained
be made to clean and disinfect floors of buildings fre- free of S. hyodysenteriae. The introduction of S. hyodysen-
quently. Animals should not be housed in overcrowded teriae into a susceptible farm primarily occurs with carri-
conditions. er pigs. Mice are considered only as reservoir hosts be-
cause mice do not usually migrate from farm to farm.
This method of elimination of the disease without Rats are also considered reservoir hosts but do tend to
depopulation is not always successful. Muirhead (1984) migrate from farm to farm. Thus rats may be the source
reported 11 attempts to eliminate SD from six herds; that of introduction in some cases (Hampson et al. 1991;
is, the success rate for any one attempt was 54%. With Robertson et al. 1992). Such mechanical vectors of in-
careful selection of herds, including an assessment of the fected feces as boots, coveralls, vehicles, migratory ani-
ability and commitment of the workforce, and pretreat- mals, and birds are also possible modes of introduction.
ment antimicrobial susceptibility testing of the endemic Losses in affected herds can also be reduced or pre-
isolate to ensure proper drug selection, the chances of vented by various management procedures. Outbreaks
success are likely to reach 80–90% (Wood and Lysons of SD are often associated with conditions that produce
1988). To determine if the disease has been eliminated, it stress such as handling, transportation, severe weather,
is recommended that for 3–6 months following medica- or dietary changes. Minimizing stress or using preven-
tion no antimicrobials be used that are efficacious for SD. tive levels of various efficacious compounds may be use-
Clinical evidence of the disease will usually reappear if ful aids. Sanitation is also extremely important, since the
the organism is still present. The increased feed-conver- severity of the disease within an individual or a herd is
sion efficiency of SD-free pigs means that the cost of the directly related to the quantities of contaminated feces
eradication program will usually be recovered in 6–12 that are ingested. Reducing crowded conditions and pro-
months. viding a clean, dry environment can produce dramatic
results. Conversely, poor sanitation will greatly enhance
PREVENTION the distribution and severity of the disease within a herd.
An example of this may be seen in occasional herds in
The economically devastating effects of SD justify a seri- northern latitudes where severe outbreaks have followed
ous effort to keep the disease from being introduced into overfilling of waste pits under slatted-floor systems
a noninfected herd. Infectious materials may be carried when outlets became frozen.
into a herd by fomites such as workers’ boots, farm im- Depopulation is a rather drastic but frequently neces-
plements, and trucks. Isolation of a herd and rigid sani- sary measure to eliminate chronic SD. Because the
tation are essential to reduce this potential hazard. causative agent is anaerobic and susceptible to heat, oxy-
Introduction of new stock is an even greater hazard. gen, drying, and disinfectants, it is recommended that
A reliable history of the source herd is the only assurance depopulation be done during warm, dry weather if pos-
of safety. Research efforts are being directed at various sible. Thorough cleaning, disinfection and rodent exter-
methods of identification of carrier animals, and it is mination should be followed by enough time to reduce
hoped that serologic tests or other detection methods hazards of reinfection.
will soon be available for use in screening potential herd S. hyodysenteriae may survive in feces (pits and la-
additions. Quarantine of all new animals is an excellent goons) for 60 days. The organism does not survive for
procedure, especially since clinical signs often appear in long periods of time in feces-contaminated soil during
subclinically affected animals as a result of transporta- seasonal temperatures of 15˚C (59˚F) or above (Egan
tion. Furthermore, during quarantine, the newly pur- 1981). Therefore, dirt lots that have held pigs affected
chased animals could be placed on a drug(s) known to with SD should remain free of pigs for at least 30 days
CHAPTER 42 SWINE DYSENTERY Harris, Hampson, Glock 593

during a warm period of the year. Any liquid feces such pigs weaned at a younger age. Since a natural infected-
as in pits and lagoons should be considered to contain S. immune state can exist for SD involving a secretory im-
hyodysenteriae for 90 days during periods of warm weath- mune response (Joens et al. 1984; Rees et al. 1989), it is
er. Therefore, lagoon water used for recycling should not possible that a vaccine could be developed to prevent the
be utilized for several months after depopulation of the disease. Several researchers have published information
affected pigs. about experimental attempts to prevent SD by active im-
Repopulation is extremely critical because any pre- munization. Most use a parenteral administration of
ventive procedures are for naught if carrier pigs are in- killed whole cells of S. hyodysenteriae (Fernie et al. 1983;
troduced into the herd. Only surgically derived pigs or Parizek et al. 1985; Coloe and Gerraty 1988) or subunits
animals from reliable sources should be considered as re- (Joens et al. 1990; Wannemuehler et al. 1990). Cloned
placement stock. endoflagella antigens were demonstrated to confer pro-
Serious losses may be prevented even in exposed tection in the CF-1 mouse model (Boyden et al. 1989) but
herds by use of preventive levels of various therapeutic did not immunize pigs (Gabe et al. 1995). A combination
compounds. Judicious use of these compounds as de- of killed whole cells given intramuscularly followed by
scribed in the section on treatment may be very benefi- an oral vaccination with live avirulent S. hyodysenteriae
cial, but they should not be relied upon as a substitute for was a novel approach that gave better protection than in-
good management. tramuscular vaccination alone (Lysons et al. 1986).
Among veterinarians and producers, there has been Hampson et al. (1993, 1994) reported that protection
confusion regarding whether or not pigs that recover with whole-cell bacterins may be serotype specific, which
from SD are subsequently immune. Pigs with experi- may be important for bacterins marketed either region-
mentally induced SD that recover without drug treat- ally or globally.
ment can be immune to rechallenge (Joens et al. 1979a), A killed whole-cell vaccine was marketed until re-
or they may require more than one episode of disease be- cently in the United States, but with minimal sales. A
fore acquiring protective resistance (Rees et al. 1989). subunit vaccine (Wannemuehler et al. 1990) containing
Pigs that develop SD and recover without treatment both serotypes 1 and 2 of S. hyodysenteriae and a metab-
usually continue to excrete S. hyodysenteriae in their feces olizable adjuvant was licensed and marketed in the Unit-
(i.e., the immune status could be an infection immunity). ed States in mid-1997.
This immunologic state can be maintained under condi-
tions of good management. If some management stress PRACTICAL VETERINARY ADVICE
is applied, clinical symptoms of the disease can reap- TO PRODUCERS
pear. If pigs that are acutely affected with the disease are
given effective pharmaceuticals for treatment, the devel- The most important mode of transmission of SD from
opment of immunity may be impaired. Once the drug is farm to farm is the asymptomatic carrier pig. Reservoir
withdrawn, the pig may be readily reinfected by S. hyo- hosts for S. hyodysenteriae are pigs, rats, and mice. The or-
dysenteriae present in the environment and exhibit clini- ganism also survives readily in feces, particularly in
cal symptoms of SD. waste pits (Table 42.3).
Sows that have been exposed to S. hyodysenteriae pro- In herds known to be free of SD, the practitioner
tect their suckling piglets. The feces of these immune should determine that replacement breeding stock and
sows are the source of S. hyodysenteriae for the suckling purchased feeder pigs are free of the disease. In produc-
pigs. Pigs that are not weaned until 8–10 weeks of age tion facilities where the disease is endemic, reduction in
perhaps have a better chance to develop immunity than production costs can be attained by eradication of the

Tabe 42.3. Survival time of S. hyodysenteriae in animals and the environment


Temp.
Location Condition °F °C Survival Time
Pigs − − − 60 days
Mice − − − 1 year
Rats − − − 2 days
Dogs − − − 13 days
Feces
Pits Moist 45 7 60 days
Hog lots Dry 65 18 7 days
Hog lots Cold or frozen 45 7 Until temp. increases
Lagoons − − − 60 days
Source: Harris et al. 1990.
594 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

organism without depopulation; by two-site production definite financial advantage to eradication of S. hyo-
with partial depopulation; or by total depopulation, dysenteriae without depopulation versus either endemic
cleanup, disinfection, and repopulation with SD-free disease with constant medication/vaccination or total
stock (Polson et al. 1992). depopulation/repopulation. The options are as follows:

Establishing a Diagnosis of SD 1. Eradication of S. hyodysenteriae without depopula-


A diagnosis of SD based on clinical signs and/or gross tion. S. hyodysenteriae can be eliminated from endemic
and histological lesions should be confirmed by isolation herds without depopulation, but the procedure is not al-
and identification of S. hyodysenteriae. If no clinical signs ways successful. Several drugs used either alone or in
of SD have been observed in the herd, the veterinarian combination have been reported to eliminate the infec-
must be assured by the farmer that drugs and vaccines tion from endemic herds when used in conjunction with
known to be effective for the treatment and prevention of a sanitation program and rodent control (Blaha et al.
SD are not being used and thus masking the disease. 1986; Coulson 1986; Larsen 1987; Wetzel 1987; Olson
Clinical signs of SD should become apparent in 3–6 1988). Drugs that are recommended for this procedure
months, particularly in confinement facilities in the ab- are lincomycin, carbadox, and tiamulin. The principle
sence of drugs and vaccines. applied for elimination without depopulation is to med-
icate all pigs in the herd via feed, water, and/or injection
Preventing Introduction of S. hyodysenteriae for a period of several days to months. During this med-
into SD-Free Herds ication period, a sanitation program is conducted to
Herds that have been established as free of SD and are ei- eliminate the organism from the facility. In addition, the
ther closed or maintained in a closed pyramid will re- rodent population is exterminated. The reason for failure
main free of SD if situated in an isolated locale and pre- to eliminate the organism in this manner is believed to
cautions are taken to prevent contamination by feces be survival of S. hyodysenteriae in either slurry (Glock et
from carrier pigs. If breeding stock or feeder pigs are in- al. 1975) or mice (Joens and Kinyon 1982). More re-
troduced into the herd, care should be taken to limit the search is needed to determine if vaccines could be of
introductions to as few sources as possible and to pro- benefit in such programs.
cure the pigs from herds known to be free of SD. Pigs 2. Isowean two- or three-site production with partial
should be purchased from herds that do not use drugs or depopulation. Endemic SD is often not difficult or costly
vaccines known to prevent the occurrence of SD. In ad- to control in the adult population. Dams previously re-
dition, the source herd should not have made recent in- covered from SD confer resistance to their young during
troductions of pigs from questionable or SD-positive the suckling period (Harris and Glock 1986). The cost of
herds. It is not recommended that tests be conducted to medication to control SD to 18–32 kg body weight is not
ascertain the SD status, because cultural tests lack sensi- prohibitive, since certain drugs at growth-promotion lev-
tivity (Harris et al. 1978) and serologic tests often have els often control SD in this age of pig. Therefore, if pigs
false-positive reactions (Egan et al. 1983). A fecal ELISA are removed from either the farrowing rooms after re-
has been developed but it appears to be no more sensi- ceiving colostrum as in isowean (see Chapter 72) or the
tive than culture, and false-positive reactions also occur nursery accommodation up to 30 kg body weight, it is
(Taylor and Stevenson 1986). possible that the pigs will be free of S. hyodysenteriae at
If the SD status of the source herd(s) cannot be as- this time. Prior to removal from farrowing rooms or the
certained, then it is imperative that pigs be placed in nursery, medications should be administered to the
quarantine prior to entry into the SD-free herd. In quar- piglets to eliminate S. hyodysenteriae. The SD-free pigs
antine, the pigs should either be nonmedicated and ob- should be isolated from the adult herd and moved to fa-
served for signs of SD or be appropriately medicated to cilities not contaminated with S. hyodysenteriae. The use
eliminate carriers of S. hyodysenteriae. It is more practical of an SD vaccine in the sow herd may also be helpful in
to medicate, since it has been reported that unmedicated this procedure.
pigs may remain asymptomatic carriers of the disease 3. Total depopulation, cleanup, disinfection, and re-
for at least 60 days (Songer and Harris 1978). population with SD-free stock. The decision to totally de-
populate an ongoing pig operation should not be made
Herd Elimination Programs without serious deliberation and accurate financial cal-
In some situations, pig producers must continually use culations (Wood and Lysons 1988). However, in some
medications and vaccines for control of SD (Harris et al. situations, this alternative is the only method available to
1990). Quite often, however, the cost of such programs is eliminate S. hyodysenteriae from the herd/facilities. At
prohibitive due to poor profitability. If the producer is warm temperatures, the organism does not survive well
capable of maintaining a herd free of SD, then consider- in soil (less than 7 days) (Egan 1981). Therefore, the most
ation should be given to eliminating S. hyodysenteriae important reservoirs for the organism in a depopulated
from the herd/facilities. Polson et al. (1992) reported a facility are the slurry and rodents. At temperatures above
CHAPTER 42 SWINE DYSENTERY Harris, Hampson, Glock 595

60˚F (16˚C), facilities can be cleaned, disinfected, and re- dysenteriae and the epidemiology of swine dysentery.
populated very rapidly. M.V.M. thesis, Univ Glasgow, Scotland.
Chia, S. P., and Taylor, D. J. 1978. Factors affecting the sur-
REFERENCES vival of Treponema hyodysenteriae in dysenteric pig feces.
Vet Rec 103:68–70.
Adachi, Y.; Sueyoshi, M.; Miyagawa, E.; Minato, H.; and Coloe, P. J., and Gerraty, N. L. 1988. The use of vaccination
Shoya, S. 1985. Experimental infection of young broiler against swine dysentery to improve health status and
chicks with Treponema hyodysenteriae. Microbiol Im- productivity in intensively housed pigs. Proc Int Congr
munol 29:683–688. Pig Vet Soc 10:123.
Alexander, T. J. L. 1985. Methods of disease control. In Dis- Combs, B. G., and Hampson, D. J. 1992. Identification of
eases of Swine, 6th ed. Ed. A. D. Leman, B. Straw, R. D. Serpulina hyodysenteriae using a cloned DNA sequence.
Glock, W. L. Mengeling, R. H. C. Penny, and E. Scholl. Proc Int Congr Pig Vet Soc 12:275.
Ames: Iowa State Univ Press, pp. 778–800. Combs, B.; Hampson, D. J.; Mhoma, J. R. L.; and Buddle, J.
Argenzio, R. A. 1981. Glucose-stimulated fluid absorption R. 1989. Typing of Treponema hyodysenteriae by restric-
in the pig small intestine: An approach to oral fluid ther- tion endonuclease analysis. Vet Microbiol 19:351–359.
apy in swine dysentery. Am J Vet Res 41:2000–2006. Combs, B. G.; Hampson, D. J.; and Harders, S. J. 1992. Typ-
Argenzio, R. A.; Whipp, S. C.; and Glock, R. D. 1980. Patho- ing of Australian isolates of Treponema hyodysenteriae by
physiology of swine dysentery: Colonic transport and serology and by DNA restriction endonuclease analysis.
permeability studies. J Infect Dis 142:676–684. Vet Microbiol 31:273–285.
Atyeo, R. F.; Trott, D. J.; and Hampson, D. J. 1996. Poly- Combs, B. G.; Atyeo, R. F.; and Hampson, D. J. 1994. Identi-
merase chain reaction for the detection of swine dysen- fication of Serpulina hyodysenteriae by polymerase chain
tery and intestinal spirochaetosis from faecal samples. reaction. Proc Int Congr Pig Vet Soc 13:148.
Proc Int Congr Pig Vet Soc 14:291. Coulson, A. 1986. An attempt to eliminate swine dysentery
Baum, D. H., and Joens, L. A. 1979a. Serotypes of beta-he- from a 400 sow herd by the use of lincomycin in their
molytic Treponema hyodysenteriae. Infect Immun water and feed. Proc Int Congr Pig Vet Soc 9:179.
25:792–796. Cwyk, W. M., and Canale-Parola, E. 1979. Treponema suc-
———. 1979b. Partial purification of a specific antigen of cinifaciens sp. nov., an anaerobic spirochete from the
Treponema hyodysenteriae. Infect Immun 26:1211–1223. swine intestine. Arch Microbiol 122:231–239.
Binek, M.; Wojcik, U.; Szyncikwicz, Z.; and Jakubowski, T. Dugourd, D.; Jacques, M.; Bigras-Poulin, M.; and Harel, J.
1994. Dynamics of susceptibility of Serpulina hyodysen- 1996. Characterization of Serpulina hyodysenteriae iso-
teriae to different chemotherapeutics in vitro. Proc Int lates of serotype 8 and 9 by random amplification of
Congr Pig Vet Soc 13:203. polymorphic DNA analysis. Vet Microbiol 48:305–314.
Blaha, T.; Erler, W.; and Burch, D. G. S. 1986. The evaluation Duhamel, G. E., and Joens, L. A. 1994. Laboratory Proce-
of tiamulin injection for the elimination of the agent of dures for Diagnosis of Swine Dysentery. Madison, Wis.:
swine dysentery under field conditions. Proc Int Congr American Association of Veterinary Laboratory Diag-
Pig Vet Soc 9:177. nosticians.
Bowden, C. A.; Joens, L. A.; and Kelley, L. M. 1989. Charac- Duhamel, G. E.; Ramanathan, M.; Bernard, R. J.; Newman,
terization of the attachment of Treponema hyodysenteri- M. C.; and Erickson, E. D. 1992. Application of restric-
ae to Henle intestinal cells in vitro. Am J Vet Res tion fragment length polymorphism typing to epidemi-
50:1481–1485. ological tracing of Serpulina hyodysenteriae. Proc Int
Boyden, D. A.; Albert, F. G.; and Robinson, C. S. 1989. Cong Pig Vet Soc 12:276.
Cloning and characterization of Treponema hyodysenteri- Egan, I. T. 1981. The Epidemiological Aspects of Swine
ae antigens and protection in a CF-1 mouse model by im- Dysentery in the Midwestern United States. Ph.D. diss.,
munization with a cloned endoflagellar antigen. Infect Iowa State Univ.
Immun 57:3808–3815. Egan, I. T.; Harris, D. L.; and Hill, H. T. 1982. Prevalence of
Buller, N. B., and Hampson, D. J. 1994. Antimicrobial sus- swine dysentery, transmissible gastroenteritis, and
ceptibility testing of Serpulina hyodysenteriae. Aust Vet J pseudorabies in Iowa, Illinois, and Missouri swine. In
71:211–214. Proc US Anim Health Assoc, pp. 497–502.
Burrows, M. R., and Lemcke, R. M. 1981. Identification of Egan, I. T.; Harris, D. L.; and Joens, L. A. 1983. Comparison
Treponema hyodysenteriae by a rapid slide agglutination of the microtitration agglutination test and the enzyme-
test. Vet Rec 108:187–189. linked immunosorbent assay for the detection of herds
Burrows, M. R.; Lysons, R. J.; Rowlands, G. J.; and Lemcke, affected with swine dysentery. Am J Vet Res
R. M. 1984. An enzyme-linked immunosorbent assay for 44:1323–1328.
detecting serum antibody to Treponema hyodysenteriae. Elder, R. O.; Duhamel, G. E.; Schafer, R. W.; Mathieson, M.
Proc Int Congr Pig Vet Soc 8:186. R.; and Ramanathan, M. 1994. Rapid detection of Ser-
Chia, S. P. 1977. Studies of the survival of Treponema hyo- pulina hyodysenteriae in diagnostic specimens by poly-
596 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

merase chain reaction. J Clin Microbiol 32:1497–1502. 1990. Serological grouping of Treponema hyodysenteriae.
Fernie, D. S.; Ripley, P. H.; and Walker, P. D. 1983. Swine Epidemiol Infect 105:79–85.
dysentery: Protection against experimental challenge Hampson, D. J.; Combs, B. G.; Harders, S. J.; Connaughton,
following single dose parenteral immunisation with in- I. D.; and Fahy, V. A. 1991. Isolation of Treponema hyo-
activated Treponema hyodysenteriae. Res Vet Sci dysenteriae from a wild rat living on a piggery. Aust Vet J
35:217–221. 68:308.
Gabe, J. D.; Chang, R.-J.; Sloiany, R.; Andrews, W. H.; and Hampson, D. J.; Robertson, I. D.; and Mhoma, J. R. L. 1993.
McCaman, M. T. 1995. Isolation of extracytoplasmic Experiences with a vaccine being developed for the con-
proteins from Serpulina hyodysenteriae B204 and molecu- trol of swine dysentery. Aust Vet J 70:18–20.
lar cloning of the flaB1 gene encoding a 38-kilodalton Hampson, D. J.; Maltas, C. D.; Stephens, D. P.; McKechnie,
flagellar protein. Infect Immun 63:142–148. K.; and Buller, N. B. 1994. Serogroups of Australian iso-
Glock, R. D. 1971. Studies on the Etiology, Hematology and lates of Serpulina hyodysenteriae. Aust Vet J 71:347.
Pathology of Swine Dysentery. Ph.D. diss., Iowa State Hampson, D. J.; Atyeo, R. F.; and Combs, B. G. 1997. Swine
Univ. dysentery. In Intestinal Spirochaetes in Domestic Ani-
———. 1979. Swine dysentery: Is cleanup possible? In Proc mals and Humans. Ed. D. J. Hampson and T. B. Stanton.
Livest Conserv Inst, p. 130. Wallingford, England: CAB International, pp. 175–209.
Glock, R. D., and Harris, D. L. 1972. Swine dysentery. II. Harel, J., and Forget, C. 1995. DNA probe and polymerase
Characterization of lesions in pigs inoculated with Tre- chain reaction procedure for the specific detection of
ponema hyodysenteriae in pure and mixed culture. Vet Serpulina hyodysenteriae. Mol Cell Probes 9:111–119.
Med Small Anim Clin 67:65–68. Harel, J.; Belanger, M.; Forget, C.; and Jacques, M. 1994.
Glock, R. D.; Harris, D. L.; and Kluge, J. P. 1974. Localiza- Characterisation of Serpulina hyodysenteriae isolates of
tion of spirochetes with the structural characteristics of serotypes 8 and 9 from Quebec by restriction endonu-
Treponema hyodysenteriae in the lesions of swine dysen- clease fingerprinting and ribotyping. Can J Vet Res
tery. Infect Immun 9:167–178. 58:302–305.
Glock, R. D.; Vanderloo, K. J.; and Kinyon, J. M. 1975. Sur- Harris, D. L., and Glock, R. D. 1975. Swine dysentery. In Dis-
vival of certain pathogenic organisms in swine lagoon eases of Swine, 4th ed. Ed. H. W. Dunne and A. D. Le-
effluent. J Am Vet Med Assoc 166(3):277–278. man. Ames: Iowa State Univ Press, p. 541.
Glock, R. D.; Kinyon, J. M.; and Harris, D. L. 1978. Trans- ———. 1981. Swine dysentery. In Diseases of Swine, 5th
mission of Treponema hyodysenteriae by canine and avian ed. Ed. A. D. Leman, R. D. Glock, W. L. Mengeling, R. H.
vectors. Proc Int Congr Pig Vet Soc 5:KB63. C. Penny, E. Scholl, and B. Straw. Ames: Iowa State Univ
Goodwin, R. F. W., and Whittlestone, P. 1984. Monitoring Press, p. 432.
for swine dysentery: Six years’ experience with a control ———. 1986. Swine dysentery and spirochetal diseases. In
scheme. Vet Rec 115:240–241. Diseases of Swine, 6th ed. Ed. A. D. Leman, B. Straw, R.
Greer, J. M., and Wannemuehler, M. J. 1989a. Comparison D. Glock, W. L. Mengeling, R. H. C. Penny, and E. Scholl.
of the biological responses produced by lipopolysaccha- Ames: Iowa State Univ Press, p. 494.
ride and endotoxin of Treponema hyodysenteriae and Tre- Harris, D. L.; Glock, R. D.; Christensen, C. R.; and Kinyon, J.
ponema innocens. Infect Immun 57:717–723. M. 1972a. Swine dysentery. I. Inoculation of pigs with
———. 1989b. Pathogenesis of Treponema hyodysenteriae: Treponema hyodysenteriae (new species) and reproduc-
Induction of interleukin-1 and tumour necrosis factor tion of the disease. Vet Med Small Anim Clin 67:61–64.
by a treponema butanol/water extract (endotoxin). Mi- Harris, D. L.; Kinyon, J. M.; Mullin, M. T.; and Glock, R. D.
crobiol Pathol 7:279–288. 1972b. Isolation and propagation of spirochetes from
Hampson, D. J. 1997. Rapid identification of porcine Ser- the colon of swine dysentery affected pigs. Can J Comp
pulina species. Vet Rec 140:51–52. Med 36:74–76.
Hampson, D. J., and Trott, D. J. 1995. Intestinal spirochaetal Harris, D. L.; Kinyon, J. M.; Glock, R. D.; Songer, J. G.; and
infections of pigs: An overview with an Australian per- Egan, I. T. 1978. Cultural detection of swine infected
spective. In Manipulating Pig Production V. Ed. D. P. with pathogenic Treponema hyodysenteriae. Proc Int Con-
Hennessy and P. D. Cranwell. Werribee, Victoria: Aus- gr Pig Vet Soc 5:M3.
tralasian Pig Science Association, pp. 139–169. Harris, D. L.; McKean, J. D.; Joens, L. A.; Glock, R. D.; and
Hampson, D. J.; Mhoma, J. R. L.; and Combs, B. 1989a. Schultz, R. 1990. Swine dysentery–practitioner plan-
Analysis of lipopolysaccharide antigens of Treponema ning guide for herd elimination programs. Livest Con-
hyodysenteriae. Epidemiol Infect 103:275–284. serv Inst, Madison, Wis.
Hampson, D. J.; Mhoma, J. R. L.; Combs, B.; and Buddle, J. Hudson, M. J.; Alexander, T. J. L.; and Lysons, R. J. 1976. Di-
R. 1989b. Proposed revisions to the serotyping system agnosis of swine dysentery: Spirochaetes which may be
for Treponema hyodysenteriae. Epidemiol Infect confused with Treponema hyodysenteriae. Vet Rec
102:75–84. 99:498–500.
Hampson, D. J.; Mhoma, J. R. L.; Combs, B. G.; and Lee, J. I. Hughes, R.; Olander, H. J.; and Williams, C. B. 1975. Swine
CHAPTER 42 SWINE DYSENTERY Harris, Hampson, Glock 597

dysentery: Pathogenicity of Treponema hyodysenteriae. Joens, L. A.; Songer, J. G.; and Harris, D. L. 1979b. Compar-
Am J Vet Res 36:971–977. ison of selective culture and serologic agglutination of
Humphrey, S. B.; Stanton, T. B.; Jensen, N. S.; and Zuerner, Treponema hyodysenteriae for diagnosis of swine dysen-
R. L. 1997. Purification and characterization of VSH-1, a tery. Vet Rec 105:463–465.
generalized transducing bacteriophage of Serpulina hyo- Joens, L. A.; Robinson, I. M.; Glock, R. D.; and Matthews, P.
dysenteriae. J Bacteriol 179:323–329. J. 1981. Production of lesions in gnotobiotic mice by in-
Hunter, D., and Saunders, C. N. 1977. Diagnosis of swine oculation with Treponema hyodysenteriae. Infect Immun
dysentery using an absorbed fluorescent antiserum. Vet 31:504–506.
Rec 101:303–304. Joens, L. A.; Nord, N. A.; Kinyon, J. M.; and Egan, I. T. 1982.
Hunter, D. M., and Wood, T. 1979. An evaluation of the API Enzyme-linked immunosorbent assay for detection of
ZYM system as a means of classifying spirochaetes asso- antibody to Treponema hyodysenteriae antigens. J Clin
ciated with swine dysentery. Vet Rec 104:383–384. Microbiol 15:249–252.
Hyatt, D. R.; Ter Huurne, A. A. H. M.; Van Der Zeist, B. A. Joens, L. A.; DeYoung, D. W.; Cramer, J. C.; and Glock, R. D.
M.; and Joens, L. A. 1994. Reduced virulence of Serpuli- 1984. The immune response of the porcine colon to
na hyodysenteriae hemolysin-negative mutants in pigs swine dysentery. Proc Int Congr Pig Vet Soc 8:187.
and their potential to protect pigs against challenge with Joens, L. A.; Cramer, J. D.; and Mapother, M. E. 1990. Swine
a virulent strain. Infect Immun 62:2244–2248. dysentery subunit vaccine and method. Patent Coop
Jenkins, E. M.; Sinhi, P. P.; Vance, R. T.; and Reese, G. L. Treaty WO 90/02565.
1976. Passive hemolysis test for antibody to Treponema Kang, B., and Olander, H. J. 1990. Scanning electron mi-
hyodysenteriae. Infect Immun 14:1106–1107. croscopy of the colon inoculated with Treponema hyo-
Jenkinson, S. R., and Wingar, C. R. 1981. Selective medium dysenteriae in colonic loops of swine. Proc Int Congr Pig
for the isolation of Treponema hyodysenteriae. Vet Rec Vet Soc 11:117.
109:384–385. Kennedy, M. J.; Rosnick, D. K.; Ulrich, R. G.; and Yancey, R.
Jensen, N. S.; Casey, T. A.; and Stanton, T. B. 1990. Detec- J. 1988. Association of Treponema hyodysenteriae with
tion and identification of Treponema hyodysenteriae by porcine intestinal mucosa. J Gen Microbiol
using oligonucleotide probes complementary to 16S 134:1565–1567.
rRNA. J Clin Microbiol 28:2717–2721. Kennedy, M. J.; Rosey, E. L.; Frank, R. K.; Jensen, N. S.; and
———. 1992. Characterization of Serpulina (Treponema) Stanton, T. B. 1996. Generation, characterization and
hyodysenteriae and related intestinal spirochaetes by ri- virulence testing of NADH oxidase defective mutants of
bosomal RNA gene restriction patterns. FEMS Microbi- Serpulina hyodysenteriae. Abstr B105. New Orleans:
ol Lett 93:235–242. American Society for Microbiology.
Jensen, N. S.; Stanton, T. B.; and Swayne, D. E. 1996. Identi- Kent, K. A., and Lemcke, R. M. 1984. Purification and cyto-
fication of the swine pathogen Serpulina hyodysenteriae toxic activity of a haemolysin produced by Treponema
in rheas (Rhea americana). Vet Microbiol 52:259–269. hyodysenteriae. Proc Int Congr Pig Vet Soc 8:185.
Joens, L. A. 1980. Experimental transmission of Treponema Kent, K. A.; Lemcke, R. M.; and Lysons, R. J. 1988. Produc-
hyodysenteriae from mice to pigs. Am J Vet Res tion, purification and molecular weight determination
41:1225–1226. of the haemolysin of Treponema hyodysenteriae. J Med
Joens, L. A., and Glock, R. D. 1979. Experimental infection Microbiol 27:215–224.
in mice with Treponema hyodysenteriae. Infect Immun Kinyon, J. M. 1974. Characterization of Treponema hyo-
25:757–760. dysenteriae isolated from outbreaks of swine dysentery.
Joens, L. A., and Kinyon, J. M. 1982. Isolation of Treponema M.S. thesis, Iowa State Univ.
hyodysenteriae from wild rodents. Infect Immun Kinyon, J. M., and Harris, D. L. 1974. Growth of Treponema
15:994–997. hyodysenteriae in liquid medium. Vet Rec 95:219–220.
Joens, L. A.; Songer, J. G.; Harris, D. L.; and Glock, R. D. ———. 1979. Treponema innocens, a new species of intesti-
1978a. Experimental infection with Treponema hyodysen- nal bacteria and emended description of the type strain
teriae in guinea pigs. Infect Immun 22:132–135. of Treponema hyodysenteriae Harris et al. Int J Syst Bacte-
Joens, L. A.; Kinyon, J. M.; Baum, D. H.; and Harris, D. L. riol 29:102–109.
1978b. Immunofluorescent studies on Treponema hyo- ———. 1980. In vitro susceptibility of Treponema hyodysen-
dysenteriae. Proc Int Congr Pig Vet Soc 5:M2. teriae and Treponema innocens by the agar dilution
Joens, L. A.; Harris, D. L.; Kinyon, J. M.; and Kaeberle, M. L. method. In Proc Int Symp Vet Lab Diagn, Lucerne,
1978c. Microtitration agglutination for detection of Tre- Switzerland.
ponema hyodysenteriae antibody. J Clin Microbiol Kinyon, J. M.; Songer, J. G.; Janc, M.; and Harris, D. L. 1976.
8:293–298. Isolation and identification of Treponema hyodysenteriae:
Joens, L. A.; Harris, D. L.; and Baum, D. H. 1979a. Immuni- Aid to the diagnosis and treatment of swine dysentery.
ty to swine dysentery in recovered pigs. Am J Vet Res In 19th Annu Proc Am Assoc Vet Lab Diagn, pp. 65–74.
40:1352–1354. Kinyon, J. M.; Harris, D. L.; and Glock, R. D. 1977. En-
598 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

teropathogenicity of various isolates of Treponema hyo- Lysons, R. J. 1983. European perspectives on control of
dysenteriae. Infect Immun 15:638–646. swine dysentery. In Proc Am Assoc Swine Pract, Cincin-
———. 1980. Isolation of Treponema hyodysenteriae from nati, Ohio.
experimentally infected pigs at various intervals post-in- ———. 1992. Swine dysentery and other colitides. In Pig
oculation. Proc Int Congr Pig Vet Soc 6:232. Vet Soc Proc 26:69–74.
Kitai, K.; Kashiwazaki, M.; Adachi, Y.; Kume, T.; and Lysons, R. J., and Lemcke, R. M. 1983 Swine dysentery: To
Arakawa, A. 1979. In vitro activity of 39 antimicrobial isolate or to fluoresce? Vet Rec 112:203.
agents against Treponema hyodysenteriae. Antimicrob Lysons, R. J.; Lemcke, R. M.; Bew, J.; Burrows, M. R.; and
Agents Chemother 15:392–395. Alexander, T. J. L. 1982. An avirulent strain of Trepone-
Knoop, F. C.; Schrank, G. D.; and Ferraro, F. M. 1979. In vit- ma hyodysenteriae isolated from herds free of swine
ro attachment of Treponema hyodysenteriae to mam- dysentery. Proc Congr Pig Vet Soc 7:40.
malian epithelial cells. Can J Microbiol 25:399–405. Lysons, R. J.; Burrows, M. R.; Debney, T. G.; and Bew, J.
Kunkle, R. A., and Kinyon, J. M. 1988. Improved selective 1986. Vaccination against swine dysentery—An effec-
medium for the isolation of Treponema hyodysenteriae. J tive novel method. Proc Int Congr Pig Vet Soc 9:180.
Clin Microbiol 26:2357–2360. Lysons, R. J.; Lemcke, R. M.; and Duncan, A. L. 1988. “Coli-
Kunkle, R. A., Harris, D. L.; and Kinyon, J. M. 1986. Auto- tis” of pigs—An emerging disease. Proc Int Congr Pig
claved liquid medium for propagation of Treponema hy- Vet Soc 10:141.
odysenteriae. J Clin Microbiol 24:669–671. Lysons, R. J.; Kent, K. A.; Bland, A. P.; Sellwood, R.; Robin-
Larsen, L. P. 1987. Eradication of swine dysentery. In Proc son, W. P.; and Frost, A. J. 1991. A cytotoxic haemolysin
Am Assoc Swine Pract, Indianapolis, Ind., p. 565. from Treponema hyodysenteriae: A probable virulence de-
Lee, B. J., and Hampson, D. J. 1996. Production and charac- terminant in swine dysentery. J Med Microbiol
terisation of a monoclonal antibody to Serpulina hyo- 34(2):97–102.
dysenteriae. FEMS Microbiol Lett 136:193–197. Mapother, M. E. 1993. An Estimate of the Prevalence of
Lee, J. I.; Hampson, D. J.; Combs, B. G.; and Lymbery, A. J. Swine Dysentery in U.S. Swine Herds during 1989–1991.
1993. Genetic relationships between isolates of Serpuli- Survey, National Animal Health Monitoring System,
na (Treponema) hyodysenteriae, and comparison of meth- National Veterinary Services Laboratory, U.S. Dept
ods for their subspecific differentiation. Vet Microbiol Agric, Washington, D.C.
34:35–46. Mapother, M. E., and Joens, L. A. 1985. New serotypes of
Lemcke, R. M., and Bew, J. 1984. Antigenic differences Treponema hyodysenteriae. J Clin Microbiol 22:161–164.
among isolates of Treponema hyodysenteriae. Proc Int Messier, S.; Higgins, R.; and Moore, C. 1990. Minimal in-
Congr Pig Vet Soc 8:183. hibitory concentrations of five antimicrobials against
Lemcke, R. M., and Burrows, M. R. 1979. A disc growth-in- Treponema hyodysenteriae and Treponema innocens. J Vet
hibition test for differentiating Treponema hyodysenteriae Diagn Invest 2:330–333.
from other intestinal spirochetes. Vet Rec 104:548–551. Mhoma, J. R. L.; Hampson, D. J.; and Robertson, I. D. 1992.
———. 1980. Sterol requirement for the growth of Tre- A serological survey to determine the prevalence of Tre-
ponema hyodysenteriae. J Gen Microbiol 116:539–543. ponema hyodysenteriae in Western Australia. Aust Vet J
———. 1981. A comparative study of spirochaetes from 69:81–84.
the porcine alimentary tract. J Hyg Camb 86:173–182. Miao, R. M.; Feldsteel, A. H.; and Harris, D. L. 1978. Genet-
Lemcke, R. M., and Williams, L. A. 1984. Modification of ics of Treponema: Characterization of Treponema hyo-
the selective medium for Treponema hyodysenteriae. Proc dysenteriae and its relationship to Treponema pallidum.
Int Congr Pig Vet Soc 8:184. Infect Immun 22:736–739.
Lemcke, R. M.; Bew, J.; Burrows, M. R.; and Lysons, R. J. Milner, J. A., and Sellwood, R. 1994. Chemotactic response
1979. The growth of Treponema hyodysenteriae and other to mucin by Serpulina hyodysenteriae and other porcine
porcine intestinal spirochaetes in a liquid medium. Res spirochetes: Potential role in intestinal colonization. In-
Vet Sci 26:315–319. fect Immun 62:4095–4099.
Lemcke, R. M.; Burrows, M. R.; Kent, K. A.; and Lysons, R. Moller, A. F. N. 1984. Eradication of swine dysentery with a
J. 1982. Studies on a haemolysin produced by Treponema 5 day treatment of tiamulin. Dansk Vet Tidsskr 67:20.
hyodysenteriae. Proc Int Congr Pig Vet Soc 7:39. 15/10 1017–1022.
Li, Z.; Belanger, M.; and Jacques, M. 1991. Serotyping of Molnar, L. 1996. Sensitivity of strains of Serpulina hyodysen-
Canadian isolates of Treponema hyodysenteriae and de- teriae isolated in Hungary to chemotherapeutic drugs.
scription of two new serotypes. J Clin Microbiol Vet Rec 138:158–160.
29:2794–2797. Muir, A., Koopman, M., Libby, S., Joens, L., Heffron, F.; and
Lymbery, A. J.; Hampson, J. J.; Hopkins, R. M.; Combs, B.; Kusters, J. 1992. Cloning and expression of a Serpulina
and Mhoma, J. R. L. 1990. Multilocus enzyme elec- (Treponema) hyodysenteriae hemolysin gene. Infect Im-
trophoresis for identifying and typing of Treponema hyo- mun 60:529–535.
dysenteriae and related spirochaetes. Vet Microbiol Muirhead, M. R. 1984. With precise planning and imple-
22:89–99. mentation, swine dysentery can be eliminated from a
CHAPTER 42 SWINE DYSENTERY Harris, Hampson, Glock 599

herd. Int Piglett 4:1–3. geted mutagenesis of flaA1 or flaB1 flagellar genes re-
Neef, N. A.; Lyson, R. J.; Trott, D. J.; Hampson, D. J.; Jones, sults in avirulence by Serpulina hyodysenteriae in swine.
P. W.; and Morgan, J. H. 1994. Pathogenicity of porcine In Conference for Research Workers in Animal Disease,
intestinal spirochetes in gnotobiotic pigs. Infect Immun Chicago, Ill. Abstr 53.
62:2395–2403. Rosey, E. L.; Kennedy, M. J.; Petrella, D. K.; Ulrich, R. G.;
Nuessen, M. E.; Birmingham, J. R.; and Joens, L. A. 1982. Bi- and Yancey, R. J., Jr. 1995. Inactivation of Serpulina hyo-
ological activity of a lipopolysaccharide extracted from dysenteriae flaA1 and flaB1 periplasmic flagellar genes by
Treponema hyodysenteriae. Infect Immun 37:138–142. electroporation-mediated allelic exchange. J Bacteriol
Nuessen, M. E.; Joens, L. A.; and Glock, R. D. 1983. Involve- 177:5959–5970.
ment of lipopolysaccharide in the pathogenicity of Tre- Schmall, M. S.; Argenzio, R. A.; and Whipp, S. C. 1983.
ponema hyodysenteriae. J Immun 131:997–999. Pathophysiologic features of swine dysentery: Cyclic nu-
Olson, L. D. 1974. Clinical and pathological observations on cleotide-independent production of diarrhea. Am J Vet
the experimental passage of swine dysentery. Can J Res 44:1309–1316.
Comp Med 28:7–13. Sellwood, R. 1991. Antigenic characterization and genetics
———. 1996. Enhanced isolation of Serpulina hyodysenteri- of Treponema hyodysenteriae. In Genetics and Molecular
ae by using sliced agar medium. J Clin Microbiol Biology of Anaerobes. Ed. M. Sebald. New York:
34:2937–2941. Springer-Verlag.
Olson, S. 1988. Eradication of swine dysentery from a far- Sellwood, R.; Kent, K. A.; Burrows, M. R.; Lysons, R. J.; and
row to finish herd using lincomycin. Proc Am Assoc Bland, A. P. 1989. Antibodies to a common outer enve-
Swine Pract, St. Louis, Mo., p. 301. lope antigen of Treponema hyodysenteriae with antibacte-
Parizek, R.; Stewart, R.; Brown, K.; and Blevins, D. 1985. rial properties. J Gen Microbiol 135:2249–2257.
Protection against swine dysentery with an inactivated Siba, P. M.; Pethick, D. W.; and Hampson, D. J. 1996. Pigs
Treponema hyodysenteriae bacterin. Vet Med 80:80–86. experimentally infected with Serpulina hyodysenteriae
Picard, B.; Massicotte, L.; and Saheb, S. A. 1978. Effect of can be protected from developing swine dysentery by
sodium ribonucleate on the growth and the hemolytic feeding them a highly digestible diet. Epidem Infect
activity of Treponema hyodysenteriae. Experientia 116:207–216.
35:484–486. Songer, J. G., and Harris, D. L. 1978. Transmission of swine
Pluske, J. R.; Siba, P. M.; Pethick, D. W.; Durmic, Z.; Mullan, dysentery by carrier pigs. Am J Vet Res 39:913–916.
B. P.; and Hampson, D. J. 1996. The incidence of swine Songer, J. G.; Kinyon, J. M.; and Harris, D. L. 1976. Selective
dysentery in pigs can be reduced by feeding diets that medium for isolation of Treponema hyodysenteriae. J Clin
limit the amount of fermentable substrate entering the Microbiol 4:57–60.
large intestine. J Nutr 126:2920–2933. Songer, J. G.; Glock, R. D.; Schwartz, K. J.; and Harris, D. L.
Polson, D. D.; Marsh, W. E.; and Harris, D. L. 1992. Finan- 1978. Isolation of Treponema hyodysenteriae from sources
cial considerations for individual herd eradication of other than swine. J Am Vet Med Assoc 172:464–466.
swine dysentery. Proc Int Congr Pig Vet Soc 12:510. Sotiropoulos, C.; Smith, S. C.; and Coloe, P. J. 1993. Charac-
Rainier, R. H.; Harris, D. L.; Glock, R. D.; Kinyon, J. M.; and terization of two DNA probes specific for Serpulina hyo-
Brauer, M. A. 1980. Effects of carbadox and lincomycin dysenteriae. J Clin Microbiol 31:1746–1752.
for treatment and carrier state control of swine dysen- Stanton, T. B. 1989. Glucose metabolism and NADH recy-
tery. Am J Vet Res 41(9):1349–1356. cling by Treponema hyodysenteriae, the agent of swine
Rees, A. S.; Lysons, R. J.; Stokes, C. R.; and Bourne, F. J. dysentery. Appl Environ Microbiol 55:2365–2371.
1989. Antibody production in the colon during infection ———. 1992. Proposal to change the genus designation
with Treponema hyodysenteriae. Res Vet Sci 47:263–269. Serpula to Serpulina gen. nov. containing the species Ser-
Ritchie, A. E.; Robinson, I. M.; Joens, L. A.; and Kinyon, J. pulina hyodysenteriae comb. nov. and Serpulina innocens
M. 1978. A bacteriophage for Treponema hyodysenteriae. comb. nov. Int J Syst Bacteriol 42:189–190.
Vet Rec 102:34–35. ———. 1997. Physiology of ruminal and intestinal
Robertson, I. D.; Mhoma, J. R. L.; and Hampson, D. J. 1992. spirochaetes. In Intestinal Spirochaetes in Domestic An-
Risk factors associated with the occurrence of swine imals and Humans. Ed. D. J. Hampson and T. B. Stan-
dysentery in Western Australia: Results of a postal sur- ton. Wallingford, England: CAB International, pp. 7–45.
vey. Aust Vet J 69:92–93. Stanton, T. B., and Jensen, N. S. 1996. Purification and char-
Roncalli, R. A., and Leaning, W. H. D. 1976. Geographical acterization of NADH oxidase from Serpulina (Trepone-
distribution of swine dysentery. Proc Int Congr Pig Vet ma) hyodysenteriae. J Bacteriol 175:2980–2987.
Soc 4:L17. Stanton, T. B., and Lebo, D. F. 1988. Treponema hyodysenteri-
Ronne, H., and Szancer, J. 1990. In vitro susceptibility of ae growth under various culture conditions. Vet Micro-
Danish field isolates of Treponema hyodysenteriae to biol 18:177–190.
chemotherapeutics in swine dysentery (SD) therapy. Stanton, T. B.; Jensen, N. S.; Casey, T. A.; Dewhirst, F. E.;
Proc Int Congr Pig Vet Soc 11:126. and Paster, B. J. 1991. Reclassification of Treponema hyo-
Rosey, E. L.; Kennedy, M. J.; and Yancey, R. J., JR. 1994. Tar- dysenteriae and Treponema innocens in a new genus, Ser-
600 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

pula, gen. nov., as Serpula hyodysenteriae comb. nov. and 1995. Distribution of the SmpA gene from Serpulina hy-
Serpula innocens comb. nov. Int J Syst Bacteriol odysenteriae amongst intestinal spirochaetes. Microbiol
41(1):50–58. 141:2041–2046.
Taylor, D. J. 1972. Studies of Bacteria Associated with Swine Validation no. 64. 1998. Int J Syst Bacteriol 48:327–328.
Dysentery. Ph.D. diss., Univ Cambridge, England. Walter, D. H., and Kinyon, J. M. 1990. Recent MIC determi-
———. 1980. Experiments to evaluate the efficacy of par- nation of six antimicrobials for Treponema hyodysenteri-
enteral tiamulin medication in the therapy of experi- ae in the United States; use of tiamulin to eliminate
mental swine dysentery. Proc Int Congr Pig Vet Soc swine dysentery from two farrow to finish herds. Proc
6:255. Int Congr Pig Vet Soc 11:129.
———. 1984. Swine dysentery survey. Vet Rec 115:110–111. Wannemuehler, M. J.; Ostle, A. G.; Nibblelink, S. K.; Coyle,
Taylor, D. J., and Alexander, T. J. L. 1971. The production of D. C.; and Walter, C. J. 1990. Pathogenesis of swine
dysentery in swine by feeding cultures containing a dysentery: Preparation of a protective vaccine. Proc Int
spirochaete. Br Vet J 127(11):58–61. Congr Pig Vet Soc 11:124.
Taylor, D. J., and Blakemore, W. F. 1971. Spriochaetal inva- Wetzel, T. 1987. A case report: The use of carbadox to erad-
sion of the colonic epithelium in swine dysentery. Res icate swine dysentery. Proc Am Assoc Swine Pract, Indi-
Vet Sci 12:177–179. anapolis, p. 375.
Taylor, D. J., and Stevenson, R. 1986. A fecal ELISA test for Whipp, S. C.; Harris, D. L.; Kinyon, J. M.; Songer, J. G.; and
Treponema hyodysenteriae in pig feces and slurry. Proc Int Glock, R. D. 1978. Enteropathogenicity testing of Tre-
Congr Pig Vet Soc 9:175. ponema hyodysenteriae in ligated colonic loops of swine.
Taylor, D. J.; Simmons, J. R.; and Laird, H. M. 1980. Produc- Am J Vet Res 39:1293–1396.
tion of diarrhea and dysentery in pigs by feeding pure Whipp, S. C.; Pohlenz, J.; Harris, D. L.; Robinson, I. M.;
cultures of a spirochaete differing from Treponema hyo- Glock, R. D.; and Kunkle, R. 1982. Pathogenicity of Tre-
dysenteriae. Vet Rec 106:326–332. ponema hyodysenteriae in uncontaminated gnotobiotic
Ter Huurne, A. A. H. M.; Muir, S.; Van Houten, M.; Van der pigs. Proc Int Congr Pig Vet Soc 7:31.
Zeijst, B. A. M.; Gaastra, W.; and Kusters, J. G. 1994. Whiting, R. A.; Doyle, L. P.; and Spray, R. S. 1921. Swine
Characterization of three putative Serpulina hyodysen- dysentery. Purdue Univ Agric Exp Stn Bull 257:3–15.
teriae hemolysins. Microbiol Pathogen 16:269–282. Wilcock, B. D., and Olander, H. J. 1979a. Studies on the
Trott, D. J.; Atyeo, R. F.; Swayne, D. E.; Stoutenburg, J. W.; pathogenesis of swine dysentery. I. Characterization of
and Hampson, D. J. 1996a. Genetic relatedness amongst the lesions in colons and colonic segments inoculated
intestinal spirochaetes isolated from rats and birds. Lett with pure cultures or colonic content containing Tre-
Appl Microbiol 23:431–436. ponema hyodysenteriae. Vet Pathol 16:450–465.
Trott, D. J.; Huxtable, C. R.; and Hampson, D. J. 1996b. Ex- ———. 1979b. Studies on the pathogenesis of swine dysen-
perimental infection of newly weaned pigs with human tery. II. Search for a cytotoxin in spirochetal broth cul-
and porcine strains of Serpulina pilosicoli. Infect Immun tures and colon content. Vet Pathol 16:567–573.
64:4648–4654. Wood, E. N., and Lysons, R. J. 1988. The financial benefit
Trott, D. J.; Stanton, T. B.; Jensen, N. S.; Duhamel, G. E.; from the eradication of swine dysentery. Vet Rec
Johnson, J. L.; and Hampson, D. J. 1996c. Serpulina pi- 121:277–279.
losicoli sp. nov., the agent of porcine intestinal spiro- Zuerner, R. L., and Stanton, T. B. 1994. Physical and genet-
chetosis. Int J System Bacteriol 46:206–215. ic map of the Serpulina hyodysenteriae B78 chromosome.
Turner, A. K.; Atyeo, R. F.; Sellwood, R.; and Hampson, D. J. J Bacteriol 176:1087–1092.
Tuberculosis
43 C. O. Thoen

During fiscal year 1995 in the United States, 35,681,290 Table 43.1. Prevalence of tuberculosis in swine in
the United States as determined by inspection in
cattle (exclusive of tuberculin reactors) were slaughtered abattoirs under federal supervision
under federal inspection; of these, 136 carcasses
(0.0003%) were designated as being tuberculous. In con- Number Percent Percent
Year Slaughtered Tuberculosisa Condemnedb
trast, of 94,490,329 swine slaughtered during the same
period, 196,944 (0.21%) had lesions attributed to tuber- 1912 34,966,378 4.69 0.12
culosis (USDA 1996). The number of swine carcasses 1917 40,210,847 9.89 0.19
1922 34,416,439 16.38 0.20
with lesions attributed to tuberculosis is more than 1400 1927 42,650,443 13.54 0.14
times greater than that for cattle, and this is cause for 1932 45,852,422 11.38 0.08
concern. 1937 36,226,309 9.48 0.08
The processing of tuberculous swine carcasses is cost- 1942 50,133,871 7.96 0.026
1947 47,073,370 8.50 0.023
ly and results in significant economic losses. Regulations 1952 63,823,263 4.40 0.015
of the Meat and Poultry Inspection Program of the US- 1956 66,781,940 4.76 0.010
DA require that unaffected portions of swine carcasses 1962 67,109,539 2.25 0.008
with tuberculous lesions in more than one primary site, 1968 72,325,507 1.35 0.005
such as cervical and mesenteric lymph nodes, be cooked 1972 83,126,396 0.85 0.007
1978 71,805,911 0.75 0.006
at 170˚F (76.7˚C) for 30 minutes before being approved 1983 79,992,743 0.41 0.003
for human food (National Archives and Records Services 1989 82,110,688 0.67 0.002
1973). 1995 94,490,329 0.21 0.003
The public health significance of Mycobacterium avi- Sources: Data compiled from USDA 1922, 1973, 1979, 1984,
um complex infections has been recognized. Of special 1990, 1996; Feldman 1963.
a
interest are reports on the isolation of M. avium complex Includes all carcasses with evidence of tuberculosis, varying
from patients with acquired immune deficiency syn- in extent from only small foci in cervical lymph nodes to general-
ized involvement.
drome (Chin et al. 1994). b
Includes carcasses with evidence of generalized tuberculo-
There has been no direct campaign to eradicate tu- sis.
berculosis in swine. It was once believed that the cam-
paign to eradicate bovine tuberculosis, which was started
in 1917, would result in a reduction of the disease in France and Germany (Meissner et al. 1978), Hungary
swine in the United States. However, the percentage of (Szabo et al. 1975), Japan (Nishimori et al. 1995; Yugi et
swine with tuberculous lesions continued to increase for al. 1972), and South Africa (Kleeberg and Nel 1973).
a number of years (Table 43.1). These reports indicate the worldwide distribution of tu-
berculosis in swine due to M. avium. The similarity of M.
ETIOLOGY avium and so-called M. intracellulare has led to the pro-
posal that the latter be considered serovars of M. avium
Swine are susceptible to infection with Mycobacterium tu- complex (M. avium–M. intracellulare) (Wolinsky and
berculosis, M. avium, and M. bovis. The investigations of Schaefer 1973; Thoen et al. 1984); this has been done in
Van Es and Martin (1925) showed that in the United this chapter.
States most of the tuberculosis in swine was caused by A numbering scheme has been developed for report-
avian tubercle bacilli. M. avium serovars 1, 2, 4, and 8 are ing M. avium serovars (Wolinsky and Schaefer 1973).
the most common isolates from tuberculous lesions in Serovars 1, 2, and 3 occur mainly in animals but also in
swine in the United States (Mitchell et al. 1975; Thoen et humans, whereas serovars 4–8 are found in human pa-
al. 1975b). At least 15 other M. avium complex serovars tients and less commonly in animals. A micromethod
have been isolated from swine in the United States has been developed for identifying serovars of M. avium
(Thoen et al. 1975b) as well as in other countries: Aus- that saves time and materials (Thoen et al. 1975a). DNA
tralia (Tammemagi and Simmons 1971), Brazil (Pestana hybridization using probes for M. avium and M. intracel-
de Castro et al. 1978), Denmark (Jorgensen 1978), lulare has been shown to provide a rapid and reliable

601
602 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

method for identifying M. avium complex isolated from


swine (Thorensen and Saxegaard 1993).
The decrease in prevalence of tuberculosis in swine in
the United States is largely attributable to a lowering of
the incidence of tuberculosis in poultry, which in turn is
the result of the increasing practice of maintaining all-
pullet flocks of chickens. The control of tuberculosis in
swine is thus incidental to and a beneficial but secondary
effect of a changing practice of poultry husbandry. Per-
haps a more rapid decline in tuberculosis among swine
will occur if a direct and effective attack is made on tu-
43.1. Tuberculous lesions in a mesenteric lymph node
berculosis in poultry. of a pig at slaughter (Thoen 1994).
Tuberculosis has been observed in large confined
swine herds, in which the infection is caused by M. avium
serovars 4 and 8. Outbreaks usually result in large losses
involving 30–60% of slaughter swine (Pritchard et al. in Australia (Clapp 1956), Denmark (Plum 1946; Jor-
1977). The problem has been associated with the use of gensen et al. 1972), England (Cochin 1943), Finland
sawdust and peat for litter (Dalchow and Nassal 1979; (Vasenius 1965), France (LaFont and LaFont 1968), and
Songer et al. 1980). Germany (Retzlaff 1966; Dalchow and Nassal 1979). The
failure to demonstrate tubercle bacilli in lesions that ap-
EPIDEMIOLOGY pear grossly to be tuberculous may be due to inadequacy
in present-day methods for isolating tubercle bacilli, oc-
Because swine are not routinely tested with tuberculin, currence of healed processes that contain no viable tu-
the only sources of information on the prevalence and bercle bacilli, or cause of the lesions by some microor-
geographic distribution of tuberculosis in this species ganism other than tubercle bacilli, such as Rhodococcus
are the data obtained from meat inspection records. On equi or R. sputi (to be discussed later).
this basis an increase in the rate of infection occurred in
the United States until 1922 (Table 43.1). During 1922, Sources of Infection and Their Control
16.38% of all swine slaughtered under federal supervi- Swine are susceptible to infection with M. tuberculosis, M.
sion had tuberculous lesions; in 0.2%, the disease was so bovis, M. avium, and M. intracellulare. The occurrence of
extensive that the entire carcass was condemned. Since tuberculosis in swine, therefore, is related to the oppor-
1922, there has been a gradual decline; by 1995, the tunity for direct or indirect contact with tuberculous cat-
prevalence had decreased to 0.21%, with only 0.003% tle, humans, and fowl and to the prevalence of tubercu-
having evidence of generalized tuberculous disease. losis in all of these species.
Since most of the tuberculosis in swine in the United The bovine tubercle bacillus is not a frequent cause of
States is of avian origin, the disease among swine is ex- tuberculosis in swine in localities where the disease in
pected to be greater in the north central states, where tu- cattle is controlled by a campaign of eradication. In the
berculosis among chickens is greater (Thoen 1997). United States and Canada, for example, bovine tubercle
However, data from meat inspection records may be mis- bacilli are rarely found in lesions of swine (Table 43.2).
leading because in the United States swine may be In Great Britain during 1952–55, the bovine type of tu-
shipped great distances from the state of origin for berculosis in swine gradually declined concurrently with
slaughter. Data on the prevalence of tuberculosis in the eradication of the disease in cattle. The percentage of
swine from meat inspection records may also be mis- avian-type infection increased from 44% during the first
leading because the diagnoses are made on the basis of 5 years of the study to 92% for the last 5 years (Lesslie et
the macroscopic appearance of lesions (Fig. 43.1). A cer- al. 1968). However, the occasional finding of bovine tu-
tain number of tuberculous infections will escape detec- bercle bacilli in swine is a reminder that the disease in
tion because the lesions are not grossly visible. Avian tu- cattle is a constant threat. Efforts to eradicate bovine tu-
bercle bacilli have been isolated from tonsils (Feldman berculosis should not be diminished.
and Karlson 1940) and lymph nodes of apparently nor- Where tuberculosis does occur in cattle, the infection
mal swine (Langenegger and Langenegger 1981). may be transmitted to swine by the feeding of unpas-
In studies in the United States and Canada, where teurized milk and dairy by-products. Feces of tubercu-
presumably tuberculous lymph nodes of swine were col- lous cattle may contain viable tubercle bacilli; this pro-
lected at abattoirs and examined bacteriologically, a vides an obvious hazard where swine and cattle are
varying percentage failed to yield tubercle bacilli (Table maintained in a common feedlot.
43.2). Similar observations have been made by workers The practice of feeding swine the offal from abattoirs
CHAPTER 43 TUBERCULOSIS Thoen 603

Table 43.2. Summary of data compiled from reports in North America on the occurrence of tubercle bacilli in
tuberculous lymph nodes of swine
Type of Tubercle Bacillus (%)
Mammalian
Reference Datea Origin of Swine Specimens Avian only only Mixed Noneb
Van Es 1925b Nebraska 248 74.6 4.4 5.6 15.4
Van Es and Martin 1925b Michigan 14 92.9 None 7.1 None
Mitchell et al. 1934b Canada 96 38.5 None None 61.5
Feldman 1938b Southeastern 30 c 80.0 6.6 (bovine) None 13.3
Minnesota
Feldman 1939b Minnesota 75d 46.6 16.0 (human) None 37.3
Feldman and Karlson 1940b Minnesota 89 61.8 None None 38.2
Pullin 1946b Eastern Canada 232 44.8 0.9 (bovine) None 54.3
Bankier 1946b Alberta, Canada 102 88.0 1.0 (bovine) None 11.0
Karlson and Thoen 1971b Minnesota 36 72.0 None None 28.0
Thoen et al. 1975b United States 2036 76.0 <1.0 <1.0 22.0
Pritchard et al. 1977b Idaho 31 80.0 None None None
Cole et al. 1978b Georgia 112 53.6 None None 46.4
Margolis et al. 1994b Pennsylvania 125 70 None None 26
Note: Specimens obtained from abattoirs under federal supervision.
a
Several papers indicated that the work was done from 1 to 2 years before publication.
b
Tubercle bacilli not demonstrated by cultural or animal inoculation tests.
c
Selected cases of generalized tuberculosis; some of the specimens were portions of lung, liver, or spleen.
d
Garbage-fed swine.

or uncooked garbage is obviously unwise, because such cupied by tuberculous chickens for the previous 2 years.
material may contain tuberculous material from beef Viable and pathogenic avian tubercle bacilli were found
carcasses. Fichandler and Osborne (1966) described an in the soil and litter of a chicken cage after 4 years.
epizootic of tuberculosis in a herd of swine in Connecti- Schalk and coworkers concluded that soil contaminated
cut that was fed improperly cooked offal from tubercu- by feces of tuberculous fowl is the most important source
lous cattle. A serious outbreak of avian tuberculosis in a of infection for swine. No success was obtained in con-
swine-feeding establishment in Denmark was traced to trolling the disease merely by use of the tuberculin test
the improper cooking of offal from poultry plants (Bier- and elimination of reactors, because the soil remained
ing-Sorensen 1959). The human type of bacillus is occa- contaminated. They recommended that an ideal pro-
sionally isolated from tuberculous lesions in swine. No gram to control avian tuberculosis is to rear young birds
person known to have active tuberculosis should be per- on clean ground and to dispose regularly of all fowl more
mitted to have contact with swine or other animals. than 1 year old.
Uncooked garbage is a potential means of transmit- Schliesser and Weber (1973) studied the survival of
ting tuberculosis to swine. Feldman (1939) recorded that M. avium in sawdust. At 18–22˚C, the survival time of
75 (28.4%) of 264 garbage-fed swine were found to have two virulent strains was 153–160 days, and the survival
tuberculous lesions at the time of slaughter. Of these, 47 time of two avirulent strains was 169–214 days. The sur-
contained tubercle bacilli, of which 35 were avian type vival times were greatly reduced when the contaminated
and 12 were human type. It was concluded that garbage sawdust was maintained at 37˚C.
may contain the offal of tuberculous chickens and that Wild birds may be incriminated as a source of M. avi-
material from tuberculous human patients is not proper- um infections in swine. Tuberculosis was found in star-
ly disposed of. The frequent occurrence of avian tubercle lings on a farm with a high incidence of tuberculosis in
bacilli in lesions limited to the cervical and mesenteric the swine but where no poultry had been kept for 8 years
lymph nodes in naturally infected swine indicates that (Bickford et al. 1966). Tuberculosis due to M. avium has
infection usually occurs by ingestion. Janetschke (1963) been found in various wild birds, some of which fre-
found that the primary complex involved the alimentary quent feedlots (Thoen 1997).
tract in 97.3% of 1000 carcasses with tuberculous lesions; The close contact of sows and slaughter pigs in yards
a pulmonary route of infection was noted in only 2.7%, and feeding pens provides opportunity for transmission
as indicated by involvement of the bronchial lymph of tuberculosis from animal to animal (Alfredsen and
nodes. Skjerve 1993). The occurrence of intestinal lesions (Fig.
Schalk et al. (1935) found that swine contracted tu- 43.2) allows spread of tubercle bacilli in feces. Feldman
berculosis when placed on ground that had not been oc- and Karlson (1940) and Pullar and Rushford (1954)
604 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

43.2. Submucosal tuberculous lesion


due to avian tubercle bacilli in the
intestinal tract of the pig. The lesion
appears to be extending toward the
surface, where it may ulcerate and
discharge bacilli into the lumen. Diffuse
cellular proliferation with little necrosis
is typical of avian tubercle bacillus
infection in swine (H&E; ×50).

demonstrated avian tubercle bacilli in the tonsils of pigs. herds of swine in Arizona and found in at least one herd
The latter workers suggested that this may be a source of that the source was sawdust and wood shavings.
infection for other animals. Smith (1958) found avian tu-
bercle bacilli in apparently normal lymph nodes of 7% of PATHOGENESIS
swine, 5% of sheep, and 5% of cattle but was unable to
find them in adult normal chickens; he suggested, there- The development of disease in swine depends on the
fore, that domestic mammals may contract M. avium ability of the tubercle bacillus to multiply within tissues
from each other as well as from tuberculous fowl. of the host and to induce a host response. Although acid-
Pulmonary, uterine, and mammary tuberculous le- fast bacilli initially encounter granulocytes and humoral
sions in swine constitute sources of infection for other components, activated mononuclear macrophages are
animals. Jorgensen et al. (1972) described an enzootic of considered to be more important in protection of the
pulmonary tuberculosis resulting from M. avium in pigs. host against mycobacteria.
Lesslie and Birn (1967) found M. avium in the udder or The capacity of M. avium to produce progressive dis-
milk of 18 cows and concluded that such animals may be ease may be related to certain complex lipids present in
a source of M. avium in pigs. Bille and Larsen (1973) re- the cell wall, such as the glycopeptidolipids (previously
ported congenital infection in swine caused by avian tu- referred to as C-mycosides) localized in the exterior por-
bercle bacilli, suggesting that infected pregnant sows tion of the cell envelope (Rastogi and Barrow 1994).
may have a role in the transmission of this infection. Sig- However, it appears that the effect of these components
urdardottir et al. (1994) reported granulomatous enteri- alone or together on phagolysosome fusion cannot ac-
tis due to M. avium in a pig. count for virulence. Available information suggests that a
Where sawdust is used for bedding, various serovars combination of toxic lipids and factors released by viru-
of M. avium have been isolated from lesions in swine as lent tubercle bacilli may cause disruption of the phago-
well as from the sawdust. Reactions to avian and bovine some, interfere with phagolysosome formation, alter the
tuberculin have been reported in boars exposed to saw- release of hydrolytic enzymes from the attached lyso-
dust from which M. avium or other nonphotochro- somes, and/or inactivate the lysosomal enzymes released
mogenic mycobacteria were isolated (Fodstad 1977). into the cytoplasmic vacuole (Thoen and Bloom 1995).
Schliesser and Weber (1973) found that M. avium would Certain serovars of M. avium are susceptible to bacterici-
survive as long as 214 days in sawdust. In Hungary, dal mechanisms of macrophages; however, the impor-
Szabo et al. (1975) found that the incidence of tubercu- tance of reactive nitrogen intermediates and oxygen rad-
lous adenitis in swine was greater when sawdust was icals in macrophages of swine exposed to virulent
used as litter; when the use of sawdust was discontinued, tubercle bacilli remains to be elucidated (Thoen and
the occurrence of such lesions decreased significantly. Chiodini 1993). Although the mechanisms by which my-
Dalchow and Nassal (1979) recorded that the same cobacteria produce disease in swine have not been clear-
serovars of avian tubercle bacilli as found in swine could ly defined, experimental studies in piglets revealed that
be isolated from sawdust. These workers also reported nonspecific esterase activity was elevated in mononu-
that sawdust could contain infectious mycobacteria even clear macrophages of lymph nodes 7 days following in-
after 4 years of storage. Songer et al. (1980) investigated oculation of M. avium complex (M. intracellulare) serovar
CHAPTER 43 TUBERCULOSIS Thoen 605

8 (Momotani et al. 1980). Granulomas of varying stages yielded Corynebacterium equi, now reclassified as
were observed in mesenteric and mandibular lymph Rhodococcus equi (Goodfellow et al. 1982), which Clapp
nodes and intestinal mucosa at 14 days postinoculation. considered important in producing tuberculosis-like
In other investigations, sensitized lymphocytes and de- lymphadenitis in swine. In the series of 420 specimens,
tectable mycobacterial antibodies have been reported to only 5 were from swine with generalized tuberculosis,
occur at 14–28 days postexposure to M. avium or M. bo- and all of these were associated with M. bovis and M. avi-
vis (Muscoplat et al. 1975; Thoen et al. 1979a). um. Microscopically, the changes induced in swine tis-
sues are characterized by diffuse proliferation of epithe-
LESIONS lioid cells and giant cells. There may be some necrosis
and calcification, especially in older lesions, but calcifica-
Tubercle Bacilli tion is not usually prominent. Similar changes are ob-
Detailed discussions of the pathological anatomy of tu- served in sows and slaughter pigs (Thoen et al. 1976b).
berculosis in swine may be found in Pallaske (1931), Proliferation of connective-tissue elements accompanies
Feldman (1938a), Francis (1958), and Kramer (1962). As the process. Lesions caused by mammalian tubercle
seen in the abattoirs, tuberculous lesions in swine are bacilli have a tendency to become encapsulated by a well-
usually limited to lymph nodes of the cervical and the developed zone of connective tissue (Fig. 43.3). In addi-
mesenteric regions. The lesions vary in appearance from tion, there is often early caseation and marked calcifica-
small, yellowish white, caseous foci a few millimeters in tion (Karlson and Thoen 1971). However, consistent
diameter to diffuse enlargement of the entire node (Fig. histopathological differentiation between lesions caused
43.1). The disease may be localized in one group of by mammalian and avian tubercle bacilli is not possible
nodes or may involve a number of lymph nodes along (Himes et al. 1983).
the digestive tract. Generalized tuberculosis in swine is not commonly
Gross differentiation between tuberculous adenitis seen. In most instances it is from infection with M. bovis,
caused by avian tubercle bacilli and that caused by mam- but it may also result from the avian type (Feldman
malian tubercle bacilli is difficult, but in general, some 1938b; Jorgensen et al. 1972). The extent and character
features are characteristic of each. In an infection of of generalized involvement vary from the occurrence of
avian origin the lymph nodes may be enlarged and firm a few small foci in several organs to extensive nodular
with no discrete purulent foci, or there may be one or processes involving the liver, spleen, lungs, kidneys, and
more soft caseous areas with indistinct borders. Calcifi- many lymph nodes. Generalized lesions from infection
cation is seldom demonstrable. The cut surface of the le- with M. avium tend to be diffuse. The cut surface is usu-
sion has a neoplastic appearance with a few caseous foci. ally smooth, and there is no great tendency toward en-
Although there may be diffuse fibrosis, there is little ten- capsulation by fibrosis. There may be foci of caseation,
dency to encapsulation. Relatively large areas of but calcification is not pronounced. Lesions resulting
caseation may be present and occasionally will involve from infection with mammalian tubercle bacilli, howev-
the entire lymph node. The lesions due to tubercle bacil- er, are likely to be discrete, caseous, and well circum-
li of the avian type are generally not easily enucleated. In scribed by fibrosis. Calcification is prominent.
contrast, when the infection is of mammalian origin (ei-
ther bovine or human), the lesions tend to be well encap- Bacteria Other than Tubercle Bacilli
sulated and are relatively easy to separate from the sur- Various species of mycobacteria other than tubercle
rounding tissue. In addition, calcification is usually bacilli have been isolated from swine and other animals
prominent in lesions caused by infection with mam- in different countries, but reports of such are few and
malian tubercle bacilli. The individual foci appear to be usually concern only sporadic cases (Schliesser 1976).
discrete and caseous. These distinctions are by no means The significance of finding M. kansasii, M. xenopi, or M.
absolute, and there are many variations in the gross ap- fortuitum is not clear. It may be important, however, to
pearance of tuberculous lesions in lymph nodes of learn if animals and humans become infected from the
swine. same sources (Thoen and Williams 1994). Of potential
Clapp (1956) examined, by bacteriological proce- importance is the recovery of M. chelonei from swine be-
dures, 420 lymph nodes (mostly submaxillary) designat- cause this bacterium has been isolated from prosthetic
ed as tuberculous upon meat inspection. There was some heart valves that were prepared from swine (Thoen and
association between the gross appearance and the cause. Himes 1977).
Localized lesions that were not easily enucleated and In Norway, M. paratuberculosis was isolated by cul-
large, dry calcareous processes involving an entire lymph ture from lesions in the mesenteric lymph nodes of
node were usually due to M. avium. Indistinctly mottled swine as well as from normal swine that were closely as-
and streaked lesions, large encapsulated purulent ab- sociated with a herd of cattle in which Johne’s disease
scesses, and lesions that could be easily enucleated were was present (Ringdal 1963). This microorganism was
usually not caused by tubercle bacilli. Some of these isolated in the United States from a slaughter pig (Thoen
606 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

A B

43.3. Tuberculous changes in cervical lymph nodes of swine. (A) Mammalian tubercle bacillus
infection. Peripheral fibrosis, necrosis, and calcification are typical of lesions due to bovine or
human types of tubercle bacilli (H&E; ×40). (B) M. avium infection. Diffuse cellular proliferation
with little necrosis (H&E; ×95).

et al. 1975b). Jorgensen (1969) and Larsen et al. (1971) concluded that it is difficult for inspectors in abattoirs to
found that swine may be infected with M. paratuberculo- differentiate between tuberculosis and R. equi infection.
sis after oral administration of the organism. M. xenopi Barton and Hughes (1980) recorded 32 reports of R. equi
was isolated from tissues of slaughtered swine that orig- infection in swine. R. equi expressing a 20 kDa antigen
inated in the southeastern region of the United States has been observed in all pig isolates, and 2 of 5 plasmids
(Jarnagin et al. 1971). Another rare finding was the isola- from pig isolates were the same as those from human
tion of the vole bacillus, M. microti, from lymph nodes of isolates, suggesting that the source of infection for hu-
three swine (Huitema and Jaartsveld 1967). mans may be pigs or the pig environment (Takai et al.
Mention must be made of the occurrence of R. equi 1996).
in localized lesions that cannot be easily differentiated Rhodococcus sputi has been isolated from tuberculosis
from tuberculous processes either macroscopically or lesions in the mesenteric lymph nodes of swine (Tsuka-
histologically (Feldman et al. 1940). Holth and Amund- mura et al. 1988).
sen (1936) in Norway reported that of 162 tuberculous
lymph nodes from swine, only 103 yielded tubercle bacil- DIAGNOSIS
li (97 were typed, with 80 avian and 16 human strains
and 1 bovine strain). Of the other 59, 38 contained a A clinical diagnosis of tuberculosis in swine is presump-
variably acid-fast “coccobacillus.” The acid-fastness, tive at best. Generally, the tuberculous lesions are limit-
however, was not constant and was lost on subculture. ed to small foci in a few lymph nodes of the digestive
The presence of this microorganism in localized tuber- tract. It is difficult to conceive that such nonprogressive
culosis-like lesions in swine was soon confirmed by other morbid changes may elicit signs detectable by physical
Scandinavian workers. Ottosen (1945) has shown that R. examination. In extensive tuberculous infection, signs
equi occurs more frequently in the soil of hog pens than may be suggestive of an infectious disease, but the symp-
elsewhere. In Denmark, Plum (1946) studied a large toms and changes are not sufficiently characteristic to es-
number of tuberculous lymph nodes from swine and tablish a diagnosis of tuberculosis.
CHAPTER 43 TUBERCULOSIS Thoen 607

The necropsy and histopathological appearance of 25% Old Tuberculin applied into the dermis on the dor-
tuberculosis in swine has been described. Although the sal surface of the ear, slightly anterior to the base. A pos-
morbid changes are sufficiently characteristic to permit a itive reaction is indicated in 24 hours by a flat, reddish
tentative diagnosis of tuberculosis, they are not specific. swelling up to 3 cm in diameter, which in 48 hours reach-
The great similarity between localized tuberculous le- es its maximal intensity. At this time the erythema and
sions and those associated with R. equi and other bacte- swelling are more pronounced; the central area becomes
ria has already been discussed. Also, chronic granuloma- hemorrhagic, and ulceration may occur. McDiarmid
tous lesions may be difficult to differentiate grossly (1956) described a means of testing swine in which re-
because of parasitic nodules and neoplasms. straint is not necessary. While the animals are feeding
Enzyme-linked immunosorbent assay (ELISA) has from a trough, 0.1 mL tuberculin is injected at a right an-
been described for detecting antibodies in swine infected gle into the skin at the junction of the ear and neck using
with M. avium (Thoen et al. 1979a, b). Positive ELISA re- a needle only 3.5 mm long. With this short needle, most
actions were observed in pigs experimentally infected of the tuberculin is deposited in the skin. By using a sy-
and in those naturally infected. The ELISA is a rapid test ringe in each hand, avian tuberculin can be injected on
that can be automated and may be of value in testing re- one side and mammalian tuberculin on the other. Reac-
placement breeding animals. tions are recorded in 48 hours. A positive reaction varies
The mere demonstration of acid-fast bacilli in exu- from “puffy” edema to inflammation, with purple discol-
dates or in lesions may be misleading. Some workers oration and necrosis. McDiarmid used Weybridge puri-
have recorded that R. equi is acid-fast in smears of fied protein derivative (PPD), which, according to Pater-
necrotic material from lymph nodes of swine (Ottosen son (1949), has 3 mg protein/mL for mammalian
1945). Acid-fast microorganisms other than tubercle tuberculin and 0.8 mg protein/mL for avian.
bacilli have been isolated from swine (Karlson and Feld- Lanz (1955) recommended injecting the tuberculin in
man 1940; Brandes 1961). the skin of the back about 10–20 cm caudal to the shoul-
The characteristic pathological features of tuberculo- ders and slightly to the right of the midline. This was eas-
sis in swine and the presence of acid-fast microorgan- ier and less time-consuming than trying to use the ear. A
isms in such lesions provide important indications on dose of 0.1 mL PPD (as used for cattle in Switzerland) is
which to base a diagnosis of tuberculosis. However, an injected intradermally. A positive reaction reaches its
unequivocal diagnosis can be made only on the basis of peak in 72 hours and consists of a painful erythematous
bacteriological procedures designed for the isolation, swelling 22–35 mm in diameter. As determined by
identification, and typing of the tubercle bacillus. necropsy, no false-negative or atypical reactions were
found among 316 animals.
Tuberculin Test Luke (1952) described observations on the tuberculin
The tuberculin test for the diagnosis of tuberculosis in test in 100 sows, 3 years old or older, using avian and
swine appears to be a useful procedure on a herd basis. mammalian tuberculin intradermally in the ear and
Of the various techniques described for this test in recording the results in 24 hours. A positive reaction was
swine, the operator should select the method that proves ascribed when there was an increase of 2 mm in thick-
by experience to be most suitable. Separate simultaneous ness of the skin at the site of injection. Of 39 reactors,
tests with avian and mammalian types of tuberculin only 22 had visible lesions, chiefly of lymph nodes of the
must be made (Thoen and Karlson 1970). A number of digestive tract. Tubercle bacilli were demonstrable in on-
investigators have found that some tuberculous swine ly 3 of the 22 animals with lesions, and each was a mam-
may fail to react to the intradermal tuberculin test. malian strain. Lesions considered to be tuberculous were
Therefore, tests should be repeated in a herd in which an- found in 8 animals that were nonreactors, but tubercle
imals with positive reactions have been found and ex- bacilli were apparently not demonstrable in these. In
cluded. Luke’s opinion, there is a large percentage of error in the
The intradermal test, usually on the ear or vulva, may tuberculin test in swine because of nonspecific sensitivi-
be employed. Because swine are susceptible to infection ty or residual sensitivity from healed tuberculous lesions.
with avian and mammalian tubercle bacilli, avian and Negative reactions in animals with lesions may, accord-
mammalian tuberculin should be used. The tuberculins ing to Luke (1958), be ascribed to the ability of the pig to
may be diluted (1:100) to minimize nonspecific cross-re- overcome and apparently sterilize existing lesions. Luke
actions (Meyn et al. 1959). Fichandler and Osborne (1953) studied various aspects of the tuberculin test in
(1966) described an extensive outbreak of bovine tuber- swine, including the specific effect of tuberculin on the
culosis in swine in which animals reacted to mammalian leukocyte count.
tuberculin by developing erythema and swelling of the Lesslie et al. (1968), using Weybridge PPD, tested 84
ear, compared with slight reactions to the avian tuber- White pigs from a herd known to have tuberculosis. The
culin. avian tuberculin was given in injections of 0.1 mL, each
Feldman (1938a) recommended the use of 0.2 mL containing 2500 tuberculin units (TU); and the mam-
608 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

malian tuberculin was given in injections of 0.1 mL, each zootiology of tuberculosis in starlings. J Am Vet Med As-
containing 10,000 TU. The injections were made simul- soc 149:312–318.
taneously, each at the base on an ear; in 48–72 hours a Biering-Sorensen, U. 1959. Ophobning af tilfaelde af aviaer
positive reaction was recorded when the reaction con- tuberkulose I en svinebesaetning. Medd Dan Dyrlaege-
sisted of edema and erythema. Guinea pigs experimen- foren 42:550–552.
tally sensitized with M. avium serovars 3, 4, 5, 6, 8, and 9 Bille, N., and Larsen, J. L. 1973. Porcine congenital infection
each reacted similarly to tuberculin prepared from due to Mycobacterium tuberculosis typus avium: Report of
serovars 2 and 7 (Anz et al. 1970). Swine experimentally a case. Nord Vet Med 25:139–143.
infected with M. avium serovars 4 and 8 reacted well to Brandes, T. 1961. Zur makroskopischen Unterscheidung
the USDA avian Old Tuberculin and to PPD prepared zwischen tuberkulosen und tuberkuloseahnlichen Ve-
from M. avium serovar 1 (Thoen et al. 1976a; Thoen et al. randerungen in den Mesenterial-lymphoknoten des
1979b). Further studies must be made in swine to deter- Schweines. Arch Lebensmittelhyg 12:53–56.
mine whether the serovar of the infecting strain will in- Chin, D. P.; Hopwell, P. C.; Yajko, D. M.; Vittinghoff, E.;
fluence the specificity of the tuberculin test. Horsburgh, C. R.; Hadley, W. K.; Stone, E. N.; Nassos, P.
At present, intradermal injection of PPD tuberculin S.; Ostroff, S. M.; Jacobson, M. A.; Matkin, C. C.; and
in the dorsal surface of the ear is the recommended pro- Reingold, A. L. 1994. Mycobacterium avium complex in
cedure for applying the tuberculin test in swine. The in- the respiratory or gastrointestinal tract and the risk of
jection site should be observed at 48 hours. M. avium complex bacterium in patients with human
immunodeficiency virus infection. J Infect Dis
PREVENTION 169:289–295.
Clapp, K. H. 1956. Tuberculosis-like lesions in swine in
The eradication of tuberculosis in swine, as well as in South Australia. Aust Vet J 32:110–113.
other species, is dependent on the availability of an eco- Cochin, E. 1943. Tubercle bacilli in lesions of the submaxil-
nomical and specific means of detecting infected ani- lary lymph nodes of swine. J Comp Pathol 53:310–314.
mals. Additional information is needed to determine ad- Cole, J. R.; Sangster, L. T.; Thoen, C. O.; Pursell, A. R.;
equate measures for cleaning and disinfecting the Williams, D. J.; DuBois, P. R.; and McDaniel, H. T. 1978.
premises where M. avium persists in the soil, in build- Mycobacteriosis in a Georgia swine herd. In Proc 21st
ings, or on equipment. Also, we need to know how long Meet Am Assoc Vet Lab Diagn, pp. 195–208.
these organisms will remain viable in the environment. Dalchow, W., and Nassal, J. 1979. Mykobakteriose beim
Investigations also should be made to determine the Schwein durch Sagemehleinstreu. Tierärztl Umsch
sources and modes of transmission of the different 34:253–261.
serovars of M. avium. Information is needed on the sta- Feldman, W. H. 1938a. Avian Tuberculosis Infections. Balti-
bility of the serovars and the genetic relationships of the more: Williams & Wilkins.
M. avium isolated from tuberculous swine and those that ———. 1938b. Generalized tuberculosis of swine due to
cause disease in humans. This is of increased importance avian tubercle bacilli. J Am Vet Med Assoc 92:681–685.
since serovars 1, 4, and 8, which are most commonly iso- ———. 1939. Types of tubercle bacilli in lesions of
lated from patients with acquired immune deficiency garbage-fed swine. Am J Public Health 29:1231–1238.
syndrome, are also isolated from tuberculous lesions in ———. 1963. Tuberculosis. In Diseases Transmitted from
swine. Animals to Man, 5th ed. Ed. T. G. Hull. Springfield, Ill.:
Charles C. Thomas, p. 5.
REFERENCES Feldman, W. H., and Karlson, A. G. 1940. Avian tubercle
bacilli in tonsils of swine. J Am Vet Med Assoc
Alfredsen S., and Skjerve E. 1993. An abattoir-based case- 96:146–149.
control study of risk factors for mycobacteriosis in Nor- Feldman, W. H.; Moses, H. E.; and Karlson, A. G. 1940.
wegian swine. Prev Vet Med 15:253–259. Corynebacterium equi as a possible cause of tuberculosis-
Anz, W.; Lauterback, D.; Meissner, G.; and Willers, I. 1970. like lesions of swine. Cornell Vet 30:465–481.
Vergleich von Sensitin-Testen an Meerschweinchen mit Fichandler, P. D., and Osborne, A. D. 1966. Bovine tubercu-
Serotyp und Huhnervirulenz bei M. avium- and M. in- losis in swine. J Am Vet Med Assoc 148:167–169.
tracellulare-Stammen. Zentralbl Bakteriol (Orig A) Fodstad, F. H. 1977. Tuberculin reactions in bulls and boars
215:536–549. sensitized with atypical mycobacteria from sawdust. Ac-
Bankier, J. C. 1946. Tuberculous lesions of swine. II. Survey ta Vet Scand 18:374–383.
of lesions found in the prairie provinces, especially in Francis, J. 1958. Tuberculosis in Animals and Man: A Study
Alberta. Can J Comp Med 10:250–253. in Comparative Pathology. London: Cassell, p. 177.
Barton, M. D., and Hughes, K. L. 1980. Corynebacterium Goodfellow, M.; Beckham, A. R.; Feldman, W. H.; and Bar-
equi: A review. Vet Bull 50:65–80. ton, M. D. 1982. Numerical classification of Rhodococcus
Bickford, A. A.; Ellis, G. H.; and Moses, H. E. 1966. Epi- equi. J Appl Bacteriol 53:199–207.
CHAPTER 43 TUBERCULOSIS Thoen 609

Himes, E. M.; Miller, L. D.; and Thoen, C. O. 1983. Swine tu- caused by the avian type tubercle bacillus. Vet Rec
berculosis: Histologic similarities of lesions from which 80:559–564.
Mycobacterium tuberculosis, M. avium complex or M. Lesslie, I. W.; Birn, K. J.; Stuart, P.; O’Neill, P. A. F.; and
bovis was identified. In Proc 26th Meet Am Assoc Vet Smith, J. 1968. Tuberculosis in the pig and the tuber-
Lab Diagn, pp. 63–76. culin test. Vet Rec 83:647–651.
Holth, H., and Amundsen, H. 1936. Fortsattle under- Luke, D. 1952. Studies in tuberculous sensitivity in the pig.
sokelser over baciltypene ved tuberkulose hos svinet pa III. The tuberculin skin reaction in the sow. Vet Rec
Ostlandet. Nor Vet Tidsskr 48:2–17. 65:344–345.
Huitema, H., and Jaartsveld, F. H. J. 1967. Mycobacterium ———. 1953. The intradermal tuberculin test in the pig.
microti infection in a cat and some pigs. Antonie van Vet Rec 65:533–535.
Leeuwenhoek 33:209–212. ———. 1958. Tuberculosis in the horse, pig, sheep and
Janetschke, P. 1963. Über die tuberkulose beim Schwein. goat. Vet Rec 70:529–536.
Monatsschr Veterinärmed 18:860–864. Margolis, M. J.; Hutchinson, L. J.; Kephart, K. B.; Hattel, A.
Jarnagin, J. L.; Richards, W. D.; Muhm, R. L.; and Ellis, E. M. L.; Whitlock, R. H.; Payeur, J. B. 1994. Results of using
1971. The isolation of Mycobacterium xenopi from granu- histologic examination and acid-fast staining to confirm
lomatous lesions in swine. Am Rev Respir Dis a diagnosis of swine mycobacteriosis made on the basis
104:763–765. of gross examination. JAVMA 204:1571–1572.
Jorgensen, J. B. 1969. Paratuberculosis in pigs: Experimental McDiarmid, A. 1956. Tuberculin testing of pigs. Vet Rec
infection by oral administration of Mycobacterium 68:298–299.
paratuberculosis. Acta Vet Scand 10:275–287. Meissner, G.; Viallier, J.; and Coullioud, D. 1978. Identifica-
———. 1978. Serological investigation of strains of My- tion serologique de 1590 souches de Mycobacterium avi-
cobacterium avium and Mycobacterium intracellulare iso- um isolées en France et en Allemagne Federale. Ann Mi-
lated in animal and non-animal sources. Nord Vet Med crobiol (Paris) 129A:131–137.
30:155–162. Meyn, A.; Schliesser, T.; and Bederke, G. 1959. Zur Frage der
Jorgensen, J. B.; Haarbo, K.; Dam, A.; and Engbaek, H. C. Abklarung unspezifischer Tuberkulinreaktionen mit
1972. An enzootic of pulmonary tuberculosis in pigs verdunntem Rinder-Einheitstuberkulin. Rindertuberk
caused by M. avium. I. Epidemiological and pathological brucell 8:179–185.
studies. Acta Vet Scand 13:56–57. Mitchell, C. A.; Walker, R. V. L.; and Humphrey, F. A. 1934.
Karlson, A. G., and Feldman, W. H. 1940. Studies on an Types of tubercle bacilli found in swine of two accredit-
acid-fast bacterium frequently present in tonsillar tissue ed areas. In Rep Vet Dir Gen Dep Agric Can, pp. 43–44.
of swine. J Bacteriol 39:461–472. Mitchell, M. D.; Huff, I. H.; Thoen, C. O.; Himes, E. M.; and
Karlson, A. G., and Thoen, C. O. 1971. Mycobacterium avium Howder, J. W. 1975. Swine tuberculosis in South Dako-
in tuberculous adenitis of swine. Am J Vet Res ta. J Am Vet Med Assoc 167:152–153.
32:1257–1261. Momotani, E.; Yokomizo, Y.; Shoya, S.; Nakamura, K.; and
Kleeberg, H. H., and Nel, E. E. 1973. Occurrence of environ- Yugi, H. 1980. Experimental granuloma formation with
mental atypical mycobacteria in South Africa. Ann Soc Mycobacterium intracellulare in HPCD piglets. J Tokyo
Belg Med Trop 53:405–417. Vet Zootec Sci 29:25–32.
Kramer, H. 1962. Zur Beurteilung tuberkuloseahnalicher Muscoplat, C. C.; Thoen, C. O.; Chen, A. W.; Rakich, P. M.;
Veranderungen in den Gekroslymphknoten des and Johnson, D. W. 1975. Development of specific lym-
Schweines unter besonderer Beruchsichigung der bakte- phocyte immunostimulation and tuberculin skin reac-
rioskopisher Prufung. Arch Lebensmittelhyg tivity in swine infected with Mycobacterium bovis and
13:264–271. Mycobacterium avium. Am J Vet Res 36:1167–1171.
LaFont, P., and LaFont, J. 1968. Étude microbiologigue des National Archives and Records Services. 1973. Code of Fed-
adenites cervicales du porc. II. Adenites a mycobacteries eral Regulations: Animal and Animal Byproducts. Title
atypiques. Rec Méd Vét 144:611–630. 9, chap. 3, subchap. A, pt. 311. Washington, D.C.: Gen
Langenegger, C. H., and Langenegger, J. 1981. Prevalence Serv Adm.
and distribution of serotypes of mycobacteria of the Nishimori, K.; Eguchi, M.; Nakaoka, Y.; Onodera, Y.; Ito, T.;
MAIS-complex isolated from pigs in Brazil. Pesqui Vet and Tanaka, K. 1995. Distribution of IS901 in strains of
Bras 1:75–80. Mycobacterium avium complex from swine using IS901
Lanz, E. 1955. Über die Tuberkulose und die intrakutane Tu- detecting primers that discriminate between M. avium
berkulinisierung beim Schwein. Schweiz Arch Tier- and Mycobacterium intracellulare. Clin Microbiol
heilkd 97:229–245. 33:2102–2106.
Larsen, A. B.; Moon, H. W.; and Merkal, R. S. 1971. Suscep- Ottosen, H. E. 1945. Undersogelser over Corynebacterium
tibility of swine to Mycobacterium paratuberculosis. Am J Magnusson-Holth, specielt med Henblik paa dens serol-
Vet Res 32:589–595. ogiske Forhold. A/S Carl. Fr Mortensen, Copenhagen.
Lesslie, I. W., and Birn, K. J. 1967. Tuberculosis in cattle Pallaske, G. 1931. Studien zum Ablauf, zur Pathogenese und
610 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

pathologischen Anatomie der Tuberkulose des Takai, S.; Fukunaga, N.; Ochiai, S.; Imai, Y.; Sasaki, Y; Tsub-
Schweines. Beitrag zum Vergleichenden Studium der aki, S.; and Sekizaki, T. 1996. Identification of interme-
Tiertuberkulose. Z Infektioskr Parasitenkd Krankheit diately virulent Rhodococcus equi isolate from pigs. Clin
Haustiere 39:211–260. Microbiol 34:1034–1037.
Paterson, A. B. 1949. Tuberculosis in animals other than cat- Tammemagi, L., and Simmons, G. C. 1971. Pathogenicity of
tle. III. Vet Rec 61:880–881. Mycobacterium intracellulare to pigs. Aust Vet J
Pestana de Castro, A. F.; Campedelli Filho, O.; and Wais- 47:337–339.
bich, E. 1978. Opportunistic mycobacteria isolated from Thoen, C. O. 1994. Mycobacterium avium infections in ani-
the mesenteric lymph nodes of apparently healthy pigs mals. Res Microbiol 145(3):173–177.
in São Paulo, Brazil. Rev Microbiol (Brazil) 9:74–83. Thoen, C. O. 1997. Avian tuberculosis. In Diseases of Poul-
Plum, N. 1946. Om Vaerdien af den Makroskopiske Diag- try, 10th ed. Ed. B. W. Calnek, W. M. Reid, and H. W. Yo-
nose af de Holthske Processer. Maandsskr Dyrlaege- der, Jr. Ames: Iowa State Univ Press (in press).
foren 58:27–37. Thoen, C. O., and Bloom, B. R. 1995. Pathogenesis of My-
Pritchard, W. D.; Thoen, C. O.; Himes, E. M.; Muscoplat, C. cobacterium bovis. In Mycobacterium bovis Infection in
C.; and Johnson, D. W. 1977. Epidemiology of mycobac- Animals and Humans. Ed. C. O. Thoen and J. H. Steele.
terial lymphadenitis in an Idaho swine herd. Am J Epi- Ames: Iowa State Univ Press, pp. 3–14.
demiol 106:222–227. Thoen, C. O., and Chiodini, R. 1993. Mycobacterium. In
Pullar, E. M., and Rushford, B. H. 1954. The accuracy of the Pathogenesis of Bacterial Infections in Animals, 2d ed.
avian intradermal tuberculin test in pigs. Aust Vet J Ed. C. L. Gyles and C. O. Thoen. Ames: Iowa State Univ
30:221–231. Press, pp. 44–56.
Pullin, J. W. 1946. Tuberculous lesions of swine. I. Survey of Thoen, C. O., and Himes, E. M. 1977. Isolation of Mycobac-
lesions found in eastern Canada. Can J Comp Med terium chelonei from a granulomatous lesion in pig. J Clin
10:159–163. Microbiol 6:81–83.
Rastogi, N., and Barrow, W. W. 1994. Cell envelope con- Thoen, C. O., and Karlson, A. G. 1970. Epidemiologic stud-
stituents and the multifaceted nature of Mycobacterium ies on swine tuberculosis. In Proc US Anim Health As-
avium pathogenicity and drug resistance. Res Microbiol soc, pp. 459–464.
145:243–252. Thoen, C. O., and Williams, D. E. 1994. Tuberculosis, tuber-
Retzlaff, N. 1966. Histologische Untersuchungen an Lymph- culoidoses and other mycobacterial infections. In Hand-
knoten von mit Mykobakterien infizierten book of Zoonoses, 2d ed. Ed. G. W. Beran. Boca Raton,
Schlachtschwein. Arch Lebensmittelhyg 17:56–62. Fla.: CRC Press, pp. 41–60.
Ringdal, G. 1963. Johne’s disease in pigs. Nord Vet Med Thoen, C. O.; Jarnagin, J. L.; and Champion, M. L. 1975a.
15:217–238. Micromethod for serotyping strains of Mycobacterium
Schalk, A. F.; Roderick, L. M.; Foust, H. L.; and Harshfield, avium. J Clin Microbiol 1:469–471.
G. S. 1935. Avian Tuberculosis: Collected Studies. ND Thoen, C. O.; Jarnagin, J. L.; and Richards, W. D. 1975b. Iso-
Agric Exp Stn Tech Bull 279. lation and identification of mycobacteria from porcine
Schliesser, T. 1976. Vorkommen und Bedeutung von My- tissues: A three-year summary. Am J Vet Res
cobakterien bei Tieren. Zentralbl Bakteriol (Orig A) 36:1383–1386.
235:184–194. Thoen, C. O.; Johnson, D. W.; Himes, E. M.; Menke, S. B.;
Schliesser, T., and Weber, A. 1973. Untersuchungen über die and Muscoplat, C. C. 1976a. Experimentally induced
Tenazitzt von Mykobakterien der Gruppe III nach Run- Mycobacterium avium serotype 8 infection in swine. Am J
yon in Sagemehleinstreu. Zentralbl Veterinärmed (B) Vet Res 37:177–181.
20:710–714. Thoen, C. O.; Himes, E. M.; Weaver, D. E.; and Spangler, G.
Sigurdardottir, O. G.; Nordstoga, K.; Baustad, B.; and Saxe- W. 1976b. Tuberculosis in brood sows and pigs slaugh-
gaard, F. 1994. Granulomatous enteritis in a pig caused tered in Iowa. Am J Vet Res 37:775–778.
by Mycobacterium avium. Vet Pathol 31:274–276. Thoen, C. O.; Armbrust, A. L.; and Hopkins, M. P. 1979a.
Smith, H. W. 1958. The source of avian tuberculosis in the Enzyme-linked immunosorbent assay for detecting anti-
pig (letter to the editor). Vet Rec 70:586. bodies in swine infected with Mycobacterium avium. Am
Songer, J. G.; Bicknell, E. J.; and Thoen, C. O. 1980. Epi- J Vet Res 40:1096–1099.
demiological investigations of swine tuberculosis in Ari- Thoen, C. O.; Owen, W. J.; and Himes, E. M. 1979b. My-
zona. Can J Comp Med 44:115–120. cobacterium avium serotype 4 infection in swine. In Proc
Szabo, I.; Tuboly, S.; Szecky, A.; Kerekes, J.; and Udvardy, N. 83d Annu Meet US Anim Health Assoc, pp. 468–479.
1975. Swine lymphadenitis due to Mycobacterium avium Thoen, C. O.; Himes, E. M.; and Karlson, A. G. 1984. My-
and atypical mycobacteria. II. Studies on the role of lit- cobacterium avium complex. In The Mycobacteria: A
tering in mycobacterial lymphadenitis incidence in Sourcebook. Ed. G. P. Kubica and L. G. Wayne. New
large-scale pig units. Acta Vet Acad Sci Hung 25:77– York: Marcel Dekker.
83. Thorensen, O. F., and Saxegaard, F. 1993. Comparative use
CHAPTER 43 TUBERCULOSIS Thoen 611

of DNA probes for Mycobacterium avium and Mycobac- try Inspection for Calendar Year 1989. MPI-1.
terium intracellulare and serotyping for identification ———. 1996. Statistical Summary. Federal Meat and Poul-
and characterization of animal isolates of the M. avium try Inspection for Calendar Year 1995.
complex. Vet Microbiol 34:83–88. Van Es, L. 1925. Tuberculosis of Swine. Univ Nebr Agric Exp
Tsukamura, M.; Komatsuzaki, C.; Sakai, R.; Kaneda, K.; Ku- Stn Circ 25.
do, K.; and Seino, A. 1988. Mesenteric lymphadenitis of Van Es, L., and Martin, H. M. 1925. An Inquiry into the
swine caused by Rhodococcus sputi. J Clin Microbiol Cause of the Increase of Tuberculosis of Swine. Univ Ne-
26:155–157. br Agric Exp Stn Res Bull 30.
USDA. 1922. Yearbook of Agriculture. Washington, D.C. Vasenius, H. 1965. Tuberculosis-like lesions in slaughter
———. 1973. Statistical Summary. Federal Meat and Poul- swine in Finland. Nord Vet Med 17:17–21.
try Inspection for Calendar Year 1972. MPI-1. Wolinsky, E., and Schaefer, W. B. 1973. Proposed number-
———. 1979. Statistical Summary. Federal Meat and Poul- ing scheme for mycobacterial serotypes of agglutina-
try Inspection for Calendar Year 1978. MPI-1. tion. Int F Syst Bacteriol 23:182–183.
———. 1984. Statistical Summary. Federal Meat and Poul- Yugi, H.; Nemoto, H.; and Watanabe, K. 1972. Serotypes of
try Inspection for Calendar Year 1983. MPI-1. Mycobacterium intracellulare of porcine origin. Nat Inst
———. 1990. Statistical Summary. Federal Meat and Poul- Anim Health Q (Tokyo) 12:168–169.
Miscellaneous Bacterial
44 Infections
D. J. Taylor

ANTHRAX
Anthrax is relatively rare in swine compared to sheep (Norberg 1953). The colony of B. anthracis growing on
and cattle, which are highly susceptible. Swine may be- blood agar on primary isolation possesses a stickiness
come infected, however, along with other species of farm that can be readily detected by touching with the bacte-
animals and may become important as a reservoir of in- riological loop. The colonial growth tends to adhere to
fection. the loop and forms tenacious threads.
Since anthrax is a zoonosis, infections in swine are a Bacteria in these colonies do not produce capsules
threat to human health. Infected swine represent a haz- unless grown on special media or in 5% carbon dioxide
ard to the farmworker and veterinarian, to the abattoir but do produce spores. B. anthracis may be distinguished
worker, and to those preparing and eating contaminated from other members of the genus by biochemical tests.
pig products. The importance of this relatively rare dis- Those of use in differentiating the organism from related
ease in swine is increased by the public health require- bacilli are listed in the section on diagnosis below. B. an-
ment for abattoir disinfection and the disposal of car- thracis is pathogenic to laboratory animals and humans.
casses after the discovery of an infected animal at meat Culture should not be attempted unless appropriate safe-
inspection. Meat processors are becoming unwilling to ty precautions such as safety cabinets and adequate dis-
slaughter pigs from infected farms, retailers are increas- posal facilities are available. Personnel handling the or-
ingly concerned about their duty to consumers, and the ganism should be vaccinated.
safe disposal of manures can be a major problem. These
factors add a wider importance to the disease. EPIDEMIOLOGY
Anthrax is present throughout the world, and the
FAO-WHO report (1973) indicates that the disease oc- Anthrax is generally considered a soilborne infection in
curred in swine in every continent during 1972. The inci- cattle, sheep, and horses. Animal-to-animal spread does
dence remains low and sporadic, but the disease presents not commonly occur, but rather, B. anthracis is deposited
a local problem in some areas. in the soil or the environment by the infected animal at
the time of or following death. Spores are formed by
ETIOLOGY some of the organisms, and these highly resistant bodies
may remain viable for years, even under adverse condi-
Anthrax is caused by Bacillus anthracis, a large gram-pos- tions. Subsequently, the spores may be ingested by sus-
itive, aerobic, spore-forming, nonmotile rod. The indi- ceptible animals and anthrax may develop.
vidual bacilli are 1–5 µm in diameter and 3–8 µm long. Swine can presumably become infected in this man-
When observed in tissue from an infected animal, the or- ner; however, because of the small number of spores
ganisms are commonly in short chains surrounded by a likely to be picked up and the higher degree of resistance
well-developed capsule. Under suitable aerobic condi- in swine, infection probably occurs only rarely. Rather,
tions, spores highly resistant to disinfectants, heat, and anthrax in swine generally occurs following ingestion of
desiccation may be produced. feed that contains a large number of B. anthracis or vi-
B. anthracis grows very luxuriantly on most common able spores. Swine that are permitted to eat the carcass of
laboratory media. On blood agar plates, colonies can an animal dead of anthrax may consume large numbers
usually be detected within 12 hours. After 24 hours at of organisms and may therefore become infected. The
37˚C, the colonies have a “ground glass” appearance, use of bonemeal or other animal products containing
with irregular, wavy borders that give them the “medusa spores of B. anthracis in feed is the most common source
head” characteristic. No hemolysis is produced on blood of infection in swine. Davies and Harvey (1955) isolated
agar; this is useful in distinguishing the colonies from B. anthracis from 5 of 41 cargoes of bonemeal shipped to
those of certain nonpathogenic species of the genus England from the Near and Middle East. Direct cultural

613
614 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

methods were unsuccessful, but the authors isolated the sulation is related to the possession of a smaller 60 MDa
organism from guinea pigs that were first protected from plasmid (Uchido et al. 1985; Mikesell et al. 1983). The ex-
the various anaerobic species common in bonemeal by otoxin (Smith et al. 1955; Harris-Smith et al. 1958; Davis
means of clostridial antisera and antitoxins and that et al. 1973) is composed of three fractions and is pro-
were then injected with a concentrated infusion from the duced when bacteria reach 5–10 × 106 organisms per
bonemeal specimen. milliliter of blood. The toxins share the same binding
The role of feed contaminated with spores of B. an- unit—the protective antigen (PA)—and are binary tox-
thracis in the transmission of anthrax can be illustrated ins. PA binds to cell surfaces and is activated by a host
by a brief account of the 1952 outbreak that occurred in protease, allowing edema factor (EF) and lethal factor
the midwestern United States (Ferguson 1986). Anthrax (LF) to enter cells. EF is a calmodulin-dependent adeny-
was confirmed on a farm in southern Ohio in February late cyclase and affects neutrophils, preventing the respi-
1952, and further outbreaks occurred in rapid succession ratory burst and thus protecting the organisms. It is also
in widely separated areas. Within a week of the recogni- responsible for the progressive hyperglycemia and the
tion of the first case, feed was incriminated as the source severe terminal hypoglycemia seen in animals with the
of infection. A number of feed companies were involved, septicemic form. LF appears to be a zinc-dependent pro-
but all had incorporated bonemeal obtained from a com- tease affecting macrophages. All three toxins are needed
pany in Columbus, Ohio, which had processed part of a to produce typical anthrax.
shipment of 100 tons of raw bonemeal obtained from The organism appears to enter the pig through the
Belgium into a meat scrap concentrate. The companies gut or tonsil. Septicemic disease is rare, and the organ-
purchased the concentrate and included it in many hun- ism multiplies locally, resisting phagocytosis by means of
dreds of tons of swine feed sold throughout Ohio and the polyglutamic acid capsule. Edema is commonly pro-
adjoining states. The organism was isolated from the raw duced locally. Neutrophils and other phagocytes are
bonemeal and from the meat scrap concentrate but not killed by EF, and organisms multiply until LF is pro-
from any of the finished feeds. duced, resulting in the death of the animal as a result of
The organism appears to be spread in wet-feed sys- its effect on the mitochondria. Immunity against anthrax
tems but rarely affects more than 1–2 animals in an in- is associated with antibodies against the exotoxin (PA)
fected herd. This was the classic picture, but accounts of (Sargeant et al. 1960; Thorne et al. 1960). Antibody to
continuing outbreaks exist (Jackson 1967; Jackson and the cell wall may be produced but is not protective.
Taylor 1989; Edgington 1990). The outbreak referred to
by Jackson and Taylor (1989) and Edgington (1990) oc- CLINICAL SIGNS
curred in a 500-sow unit and persisted for 14 weeks, re-
sulting in at least 18 cases in sows, suckling pigs, and The first indications of an outbreak of the disease may
weaned pigs. The development of disease in weaners be an increase in mortality. Investigation of these extra
may have been delayed by maternal immunity in this deaths may indicate the presence of anthrax and the clin-
continuing outbreak. The origin of the outbreak was ical signs described below may be identified. Three forms
considered to be feed, but the disease persisted within of anthrax have been observed in swine: pharyngeal, in-
the herd in spite of antimicrobial treatment. Persistence testinal, and septicemic. The usual portal of entry is the
may have been in carrier pigs or as spores in slurry and oral cavity, and invasion occurs in the tonsils or mucosa
housing. The role of flies in persistence and transmission of the pharynx. In some cases the infection may remain
was not clear, although recent studies in the United localized in the lymph nodes of this region, and the dis-
States indicate that biting flies (Stomoxys calcitrans) and ease is classified as pharyngeal. In other cases the organ-
mosquitoes (Aedes aegyptii and A. taeniorhynchus) trans- isms may pass into the intestinal tract, where primary in-
mit the disease experimentally 4 hours after feeding vasion may also occur. When B. anthracis is not localized
(Turell and Knudson 1987). Ticks (Dermacentor margina- but gains access to the general circulation, the septicemic
tus) were shown to harbor the organism for 76 days at form of the disease develops.
4˚C and for 35 days at 22–25˚C in the former Soviet The clinical signs commonly observed in pharyngeal
Union (Akhmerov et al. 1982). anthrax are cervical edema and dyspnea. General de-
pression, inappetence, and vomiting are commonly seen.
PATHOGENESIS Fever with temperatures to 41.7˚C may occur, but it is
not consistent, and in some affected swine the tempera-
B. anthracis has two major sets of pathogenic determi- ture may be subnormal. Death follows in many of the
nants: a protective capsule composed of a polymer of swine within 24 hours after the cervical edema is noticed.
d-glutamic acid and the complex exotoxin. Molecular bi- It is not uncommon for swine to recover even in the ab-
ology studies have shown that toxin production results sence of treatment. The swelling may disappear gradual-
from the possession of a 110 MDa plasmid and that cap- ly, and complete recovery appears to occur; however,
CHAPTER 44 MISCELLANEOUS BACTERIAL INFECTIONS Taylor 615

such animals may continue to remain carriers of B. an- consistency. The tonsils are usually covered with a fibri-
thracis. nous exudate, or extensive necrotic changes may be evi-
Clinical signs of intestinal anthrax are not as obvious dent. The pharyngeal mucosa is frequently inflamed and
as those in the pharyngeal form. In severe cases an acute swollen.
digestive disturbance may be evident, with vomiting, The mandibular and suprapharyngeal lymph nodes
complete loss of appetite, and diarrhea with bloody fe- are enlarged to several times their normal size. The cut
ces. Death may follow in the most severely affected surface of the affected node may vary in color from deep
swine; however, recovery occurs in many affected with brick red to strawberry red. In more chronic cases the
the milder forms (Brennan 1953). When 50 pigs were in- color may be grayish yellow, indicative of necrotic
fected in an experimental study (Redmond et al. 1997), changes in the node. In cases of the septicemic and in-
33 developed anorexia, lethargy, dullness, shivering, testinal forms the carcass may be opened before anthrax
constipation, loose feces, blood in the feces, and ataxia at is suspected. The intestinal form is more common and
some point between 1 and 8 days after infection. Only 2 there is usually copious pinkish peritoneal fluid, which
died. Fever did not exceed 41.9˚C, peaking 48 hours after may clot on exposure to air. The small intestine is usual-
infection. ly inflamed, with fibrinous adhesions on the serous sur-
Intestinal anthrax has been reported only rarely in face. The mesenteric lymph nodes may be swollen, hem-
the United States. Many cases may be unrecognized be- orrhagic, or necrotic, and edema of the mesentery is
cause of the usual practice of avoiding a complete common. The intestinal mucosa is covered with a diph-
necropsy of animals suspected of anthrax. It is possible theritic membrane and may be hemorrhagic. The intesti-
that some of the animals dying of pharyngeal anthrax nal wall may be grossly thickened. In the septicemic form
may also have had lesions in the intestinal tract. Brennan little may be seen other than the presence of blood-
(1953) reported that intestinal anthrax was the most stained fluid in the peritoneal cavity and local petechia-
common form of the disease seen in the 1952 outbreak tion. In some cases the spleen is enlarged and there may
of anthrax in England. be marked petechiation of the kidney. Small abscesses
Septicemic anthrax is the highly acute form that re- may be present in the lymph nodes of recovered pigs
sults from the entrance of B. anthracis into the blood- (Redmond et al. 1997).
stream, followed by rapid reproduction of the organisms Microscopic lesions in the lymph nodes usually con-
throughout the body. Death frequently occurs in animals sist of hemorrhage and necrosis with encapsulated bacil-
so affected, without any period of illness being noticed li. These may also be seen in the necrotic diphtheritic le-
by the owner. In swine it is the uncommon form of the sions of the intestinal mucosa and in the capillaries of
disease. Walker et al. (1967) reported the presence of vi- any organ in septicemia.
able spores of B. anthracis in the lungs of dwarf swine for
as long as 7 days following respiratory exposure. These DIAGNOSIS
authors suggested that resistance of swine may be relat-
ed to some mechanism that inhibits germination of the Anthrax should be suspected when swine show cervical
spores. Of 30 swine examined at necropsy during the an- edema and dyspnea. However, erysipelas or malignant
thrax outbreak of 1952 in Ohio, only 3 had the enlarged, edema from Clostridium septicum may also provoke simi-
dark spleen so characteristically seen in cattle. It is possi- lar clinical signs. In malignant edema, the edema will of-
ble that young pigs develop septicemia more frequently ten be more prominent in the shoulders or axillary
than older swine (Ferguson 1986). spaces. The edematous fluid and enlarged cervical or
mesenteric lymph nodes, as seen on necropsy, are very
LESIONS suggestive of anthrax. When the carcass has been
opened, the presence of bloodstained fluid in the peri-
In the interests of controlling anthrax, complete necrop- toneum, petechiation of the kidney or serosal surfaces,
sy of animals is strongly discouraged. Pigs with anthrax enlargement of the spleen, and thickening and inflam-
may not be identified before necropsy because the dis- mation of the small intestine should lead to suspicion of
ease is relatively rare. Large pigs which have died from anthrax. A history of the type of feed products eaten by
the disease may have a bloody discharge from the nose the affected swine is always of value.
(Edgington 1990), and small ones may appear very pale The accurate diagnosis of anthrax is very important
and dehydrated. The cervical region is edematous, but and in most cases is dependent upon the isolation and
otherwise no superficial lesions are evident. Incision of identification of B. anthracis.
the region reveals an extensive infiltration of the tissues
with fluid, which is usually straw colored but may appear Microscopic Examination
pink or hemorrhagic. The tissue, containing large Impression smears and cultures should be made from
amounts of fluid, may appear to possess a gelatinous the cut surfaces of the cervical lymph nodes, spleen,
616 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

mesenteric lymph nodes, intestinal mucosa, or kidney as been described (Turnbull et al. 1986, 1992) to identify
appropriate, and peritoneal fluid should also be sampled the presence of IgG antibody to PA, and PA may be as-
when present. Smears should be fixed in Zenker’s fluid, sayed in serum in pigs which have died using the capture
which kills spores, or by low heat, which does not, and EIA (Turnbull 1990).
then stained by polychrome methylene blue for 2 min-
utes and washed with water. The bacilli of anthrax ap- CONTROL
pear as square-ended blue rods in a pinkish capsule. In
smears made from decayed carcasses, other bacilli may Control of the spread of anthrax differs significantly
be present, and where antimicrobial treatment has been from control of most of the other important animal dis-
given, the bacilli may be present only as capsules or in eases. The highly resistant spore formed by B. anthracis
aberrant forms. The failure to find anthrax bacilli imme- accounts for this difference. Some swine may become in-
diately should not rule out the disease, as up to 30 min- apparent carriers, but there is little evidence to indicate
utes’ examination may be required. Peritoneal fluid is that this forms an important source of infection to sus-
more often positive than splenic smears in septicemia. ceptible animals. Otherwise, animals that become infect-
Slides and reagents used for diagnosis should be dis- ed do show clinical signs and generally develop an acute
posed of by incineration or formaldehyde fixation. disease that terminates in death within a few days. Trans-
Spores are not observed in slides prepared from fresh mission from animal to animal rarely occurs, but soil
tissue or from freshly cut surfaces. Spore-forming anaer- contaminated by the organisms serves as a source from
obes are frequently encountered in tissues of animals which susceptible animals subsequently ingest the
that have been dead several hours prior to necropsy. Dif- spores. Because of this common form of transmission,
ferentiation is important in such cases, and the following anthrax can be controlled by preventing susceptible ani-
points are helpful. Spores are rarely seen in B. anthracis mals from contacting viable spores of B. anthracis.
in fresh-tissue preparations, whereas spores are regularly Van Ness and Stein (1956) pointed out the impor-
seen in clostridia. In the latter organism the rod is usual- tance of soil types in the survival of anthrax spores. The
ly slightly enlarged by the spore. Capsules are very rarely principal areas of enzootic anthrax are regions charac-
observed in the clostridia, and any capsules seen do not terized by soils high in nitrogen and with adequate calci-
stain purple with polychrome methylene blue. um. Where such soil types are lacking (e.g., central and
eastern United States), anthrax does not appear to per-
Cultural Studies sist.
B. anthracis grows readily on many common media and The spores can survive for years under a variety of en-
is characterized by very rapid colonial development. vironmental conditions. In the unopened carcasses of
Typical colonies can be observed after 12–18 hours of animals dead of anthrax, few spores are formed except at
incubation. This rapid growth is useful in differentiating the body openings. When the animal is opened for a
B. anthracis from other pathogens. complete necropsy or when carnivorous animals are per-
B. anthracis is readily cultured from the enlarged mitted to eat the carcass, there is usually extensive spore
lymph nodes, and it may also be demonstrated from the formation as the heavily infected blood and viscera are
surrounding connective tissue in some cases. In the occa- exposed to the oxygen of the air. For this reason, the ori-
sional septicemic case the organisms can be isolated fices and any cuts in a carcass should be covered with dis-
from the blood, spleen, or liver—in fact, from essentially infectant-soaked cotton wool to prevent sporulation and
any tissue of the body. Since B. anthracis grows more spread of infection. The most productive control mea-
rapidly than most of the saprophytic bacteria likely to be sures include the complete destruction of the carcasses
encountered, except other species of Bacillus, one should of animals dead of anthrax by incineration or deep bur-
always examine the cultures after incubation for 12–18 ial.
hours. When an animal dies in the open, it is generally rec-
Suspect colonies can be identified as B. anthracis by ommended that it be burned on the spot. If the animal
their biochemical characters using API systems or by the must be moved, the carcass must be placed on a sled or
absence of hemolysis, lack of motility, growth on chloral some other vehicle that can be thoroughly disinfected
hydrate agar, and susceptibility to anthrax phage. Final and then hauled, not dragged, to an area for disposal. If
confirmation of pathogenic B. anthracis depends on the burning is not an option, deep burial can be used. The
inoculation of culture into scratches on the footpad of a carcass should be covered with lime and at least 4 feet
guinea pig or mouse under strict containment. All cul- (1.25 m) of soil. When carefully completed, these meth-
tures and any experimental animals should be fixed in ods will minimize the chances of transmission of the in-
formaldehyde and incinerated. fection.
Disinfection can be achieved with 5% freshly pre-
Serology pared sodium hydroxide or, more controllably, with 10%
A competitive enzyme immunosorbent assay (EIA) has formaldehyde and the use of appropriate respirators.
CHAPTER 44 MISCELLANEOUS BACTERIAL INFECTIONS Taylor 617

Only disinfectants capable of inactivating anthrax ter infection in a population, and this factor must be con-
spores, such as those containing glutaraldehyde and sidered before carcasses are submitted for human con-
formaldehyde, should be used. Disinfectants should be sumption.
used prior to clearing up infected premises, and contam-
inated articles should be burned. Exposed surfaces PREVENTION
should be scrubbed or pressure washed with the disin-
fectant. Kaufmann et al. (1973) evaluated the Sterne strain an-
Edgington (1990) gives an account of the procedure thrax vaccine, an avirulent spore vaccine, in an outbreak
adopted in depopulating and disinfecting a chronically of the disease in Louisiana. The results supported the ef-
infected 500-sow unit from which purchasers would no ficacy of the vaccine in swine, but the number involved
longer take pigs. All 5000 pigs were slaughtered and was too small to provide significant data for this species.
burned, all 300,000 gallons (1,364,000 L) of slurry were Similar findings were obtained by Jackson (1967) in a
disinfected with 10% formaldehyde and disposed of in an continuing outbreak in the United Kingdom. Immuniza-
approved toxic-waste site, and the buildings were tion of swine would probably reduce incidence of infec-
formaldehyde-fumigated and cleaned—at a cost of tion when they are exposed to massive doses of B. an-
£1,000,000 (U.S.$1,700,000). Similar precautions may thracis. Immunization on a large scale has not been
have to be adopted in contaminated meat plants to safe- recommended, however, since swine possess a level of
guard public health. natural resistance adequate to prevent the disease except
Following the outbreaks of anthrax in the midwest- following heavy exposure to B. anthracis.
ern United States in 1952, which were conclusively Human infection can be prevented by the safe dis-
traced to imported bonemeal, regulations were estab- posal of all contaminated carcasses, articles, and fluids
lished that prohibit the importation of raw bonemeal in- on the farm by the methods outlined above. Persons ex-
to the United States (Stein 1953). Comparable preventive posed to the infection can be given prophylactic antimi-
legislation was adopted in Canada (Moynihan 1963). crobials such as penicillin and tetracyclines, and any cas-
Bonemeal processed by an acceptable steam treatment es can be treated with them. Where longer-term
may be imported under these regulations. In addition to exposure is likely, vaccination should be carried out.
this federal regulation, some states have laws pertaining
to the operation of rendering plants and the use of ani- REFERENCES
mal products in feed. These regulations have proved ef-
fective. Similar regulations apply in most developed Akhmerov, D. S.; Kusov, V. N.; and Chernova, A. A. 1982.
countries. Survival of Bacillus anthracis in the tick Dermacentor
marginalis. Rep Kaganskii Vet Inst, pp. 101–103.
TREATMENT Brennan, A. D. J. 1953. Anthrax, with special reference to
the recent outbreak in pigs. Vet Rec 65:255.
Treatment of animals infected with B. anthracis is possi- Davies, D. G., and Harvey, R. W. S. 1955. The isolation of
ble. Since swine may develop a chronic form of the dis- Bacillus anthracis from bones. Lancet 2:86.
ease, treatment can be successfully administered in some Davis, B. D.; Dulbecco, R.; Eisen, N. H.; Ginsberg, H. S.;
cases. In the outbreak in Ohio in 1952, penicillin in oil Wood, W. B.; and McCarty, M. 1973. Microbiology, 2d
was used at a dosage level of 10,000 units/lb (22,000 ed. New York: Harper & Row.
units/kg) body weight. According to Ferguson (1986), Edgington, A. B. 1990. An outbreak of anthrax in pigs: A
pigs that were showing clinical signs of anthrax recov- practitioner’s account. Vet Rec 127:321–324.
ered completely after this treatment, and the losses were FAO/WHO. 1973. Animal Health Yearbook for 1972. Rome:
reduced considerably when the disease was recognized FAO.
early in its course. Anthrax antiserum in doses of 20–75 Ferguson, L. C. 1986. Anthrax. In Diseases of Swine, 6th ed.
mL was also used in treatment of a limited number of Ed. A. D. Leman, B. Straw, R. D. Glock, W. L. Mengeling,
animals. The results were comparable to those following R. H. C. Penny, E. and Scholl, E. Ames: Iowa State Univ
treatment with penicillin in that the pigs in the early Press, pp. 622–627.
stages of anthrax recovered promptly. Oxytetracycline is Harris-Smith, P. W.; Smith, H.; and Keppie, J. 1958. Produc-
effective against B. anthracis and may be used parenteral- tion in vitro of the toxin of Bacillus anthracis previously
ly in daily doses of 4.4–11.0 mg/kg body weight. Edging- recognised in vivo. J Gen Microbiol 19:91.
ton (1990) reported the successful use of penicillin, Jackson, W. T. 1967. Anthrax in pigs—A series of deaths.
oxytetracycline, and chlortetracycline:sulfonamide:peni- State Vet J 22:67–71.
cillin combinations to treat or suppress infection but had Jackson, W. T., and Taylor, K. C. 1989. Anthrax in pigs—A
to withdraw treatment from animals intended for series of deaths. State Vet J 43:119–125.
slaughter. Following the study of Redmond et al. (1997), Kaufmann, A. F.; Fox, M. D.; and Kolb, R. C. 1973. Anthrax
it is clear that infection may persist for up to 21 days af- in Louisiana, 1971: An evaluation of the Sterne strain
618 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

anthrax vaccine. J Am Vet Med Assoc 163:442. aration into two components. J Bacteriol 79:450.
Mikesell, P.; Ivins, B. E.; Ristroph, J. D.; and Dreier, T. M. Turell, M. J., and Knudson, G. B. 1987. Mechanical trans-
1983. Evidence for plasmid mediated toxin production mission of Bacillus anthracis by stable flies (Stomoxys cal-
in Bacillus anthracis. Infect Immun 39:371–376. citrans) and mosquitoes (Aedes aegyptii and Aedes tae-
Moynihan, W. A. 1963. Anthrax in Canada. Can Vet J 4:283. niorhynchus). Infect Immun 55:1859–1861.
Norberg, B. K. 1953. Continued investigations of some im- Turnbull, P. C. B. 1990. Salisbury Medical Bulletin. In Proc
portant characteristics in anthrax-like microorganisms Int Workshop on Anthrax, Apr 11–13, 1989, Winches-
as viewed from a point of view of differential diagnosis. ter. 68. Special Suppl, p. 53.
Nord Vet Med 5:915. Turnbull, P. C. B.; Broster, M. G.; Carman, J. A.; Manchee, R.
Redmond, C.; Hall, G. A.; Turnbull, P. C. B.; and Gillgan, J. J.; and Melling, J. 1986. Development of antibodies to
S. 1997. Experimentally assessed public health risks as- protective antigen and lethal factor components of an-
sociated with pigs from farms experiencing anthrax. Vet thrax toxin in humans and guinea pigs and their rele-
Rec 141:244–247. vance to protective immunity. Infect Immun
Sargeant, K.; Stanley, J. L.; and Smith H. 1960. The serolog- 52:356–363.
ical relationship between purified preparations of fac- Turnbull, P. C. B.; Doganay, M.; Lindeque, P. M.; Aygen, B.;
tors I and II of the anthrax toxin produced in vivo and in and McLaughlin, J. 1992. Serology and anthrax in hu-
vitro. J Gen Microbiol 22:219. mans, livestock, and Etosha National Park wildlife. Epi-
Smith, H.; Keppie, J.; and Stanley, J. L. 1955. The chemical demiol Infect 108:299–313.
basis of the virulence of Bacillus anthracis. V. The specif- Uchido, I.; Sekizaki, T.; Hashimoto, K.; and Terakado, N.
ic toxin produced by B. anthracis in vivo. Br J Exp Pathol 1985. Association of the encapsulation of Bacillus an-
36:460. thracis with a 60 Megadalton plasmid. J Gen Microbiol
Stein, C. D. 1953. A review of anthrax in livestock during 131:363–367.
1952 with reference to outbreaks in the first eight Van Ness, G., and Stein, C. D. 1956. Soils of the United
months of 1953. In Proc US Livest Sanit Assoc, States favorable for anthrax. J Am Vet Med Assoc 128:7.
p. 101. Walker, J. S.; Klein, F.; Lincoln, R. E.; and Fernelius, A. L.
Thorne, C. B.; Molnar, D. M.; and Strange, R. E. 1960. Pro- 1967. A unique defense mechanism against anthrax
duction of toxin in vitro by Bacillus anthracis and its sep- demonstrated in dwarf swine. J Bacteriol 93:2031.

MELIOIDOSIS (BURHOLDARIA PSEUDOMALLEI INFECTION)


Melioidosis is a chronic bacterial infection of pigs in Melioidosis may be suspected on clinical grounds espe-
tropical and subtropical regions such as those of Asia cially when prolonged raised rectal temperatures and un-
and northern Australia. Pigs may become infected by steady gait are associated with subcutaneous swellings of
Burholdaria (formerly Pseudomonas) pseudomallei, which the limbs. More frequently, diagnosis is based on the
is a short, gram-negative rod, 0.8 by 1.5 µm, that pro- creamy abscesses found at slaughter or on the bacterio-
duces rough or mucoid colonies on a wide variety of lab- logical results needed to confirm the presence of B.
oratory media. It is present in water and soil in tropical pseudomallei (Ketterer et al. 1986; Veljanov et al. 1994). A
and subtropical areas and may infect pigs when water hypersensitivity test resembling a tuberculin test (the
supplies are contaminated. Infection is often clinically melioidin test) and serum-agglutination and comple-
inapparent but has been associated with clinical signs ment-fixation tests have all been described and can be
(Olds and Lewis 1955; Omar et al. 1962; Laws and Hall used to confirm a diagnosis in the live pig.
1964; Rogers and Andersen 1970; Veljanov et al. 1994). Treatment with tetracyclines has been described, and
Clinical signs include a raised rectal temperature the disease can be prevented by use of clean or chlori-
(40–42˚C, 104–108˚F) for up to 4 days, unsteady gait, nated water supplies and preventing access to contami-
lameness or weakness, slight nasal discharge, and subcu- nated soil. As the disease is of public health importance,
taneous swellings of the limbs. Deaths may occur but are infected carcasses should be disposed of safely.
rare in adults, in which abortions and uterine discharges
have been recorded. REFERENCES
Lesions are found in slaughter pigs in which clinical
signs have not been seen and in those that have died from Ketterer, P. J.; Webster, W. R.; Shield, J.; Arthur, R. J.; Black-
the disease. They consist of large abscesses in the lungs, all, P. J.; and Thomas, A. D. 1986. Melioidosis in inten-
liver, spleen, kidney, and mesenteric and subcutaneous sive piggeries in South Eastern Queensland. Aust Vet J
lymph nodes. The organism can be isolated from them. 63:146–149.
CHAPTER 44 MISCELLANEOUS BACTERIAL INFECTIONS Taylor 619

Laws, L., and Hall, W. T. K. 1964. Melioidosis in animals in Rogers, R. J., and Andersen, D. J. 1970. Intrauterine infec-
North Queensland. IV. Epidemiology. Aust Vet J tion of a pig by Pseudomonas pseudomallei. Aust Vet J
40:309–314. 46:292.
Olds, R. J., and Lewis, F. A. 1955. Melioidosis in a pig. Aust Veljanov, D.; Vesselinova, A.; Nicolova, S.; Kussovski, V.; and
Vet J 31:273–274. Najdenski, H. 1994. Experimental Pseudomonas pseudo-
Omar, A. R.; Cheah, K. K.; and Mahendranathan, T. 1962. mallei infection of pigs. Proc Int Congr Pig Vet Soc
Observations on porcine melioidosis in Malaya. Br Vet J 13:236.
118:421–429.

CHLAMYDIAE
Chlamydiae are small intracellular bacteria that cause earlier work only where it is relevant to current knowl-
disease in many mammalian species and are widespread edge.
in birds. They have been demonstrated in cases of con-
junctivitis, enteritis, pneumonia, pleurisy, pericarditis, ETIOLOGY
arthritis, orchitis, uterine infections, and abortion in
pigs. Recent work on chlamydiae in pigs has shown that Chlamydiae are gram-negative bacteria that can multiply
at least three species are involved (C. psittaci, C. tra- only inside living cells. They are unusual among bacteria
chomatis, and C. pecorum); the development of reliable in that they exist outside the cell only as an inactive,
tissue culture methods and the use of immunoperoxi- trypsin-resistant, infectious particle 0.2–0.3 µm
dase staining of tissue sections, DNA probes, and, more (200–300 nm) in diameter known as an elementary body
recently, polymerase chain reaction (PCR) methods have (Fig. 44.1). This body has an electron-dense core packed
demonstrated that the organisms are relatively common with DNA and surrounded by a trilaminar cytoplasmic
in pig populations. The literature prior to 1983 was re- membrane, outside which lies a further trilaminar enve-
viewed comprehensively by Stellmacher et al. (1983), lope and then a cell wall with projections that may be as-
and the account in the present chapter draws on sociated with attachment to cells. The outer-membrane

44.1. Electron micrograph of


elementary body (arrow) of
Chlamydia sp. in an aborted
placenta (×52,500).
620 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

proteins of the chlamydiae have been studied in consid- tions show that C. psittaci from species such as sheep can
erable detail, and sequences of the genes coding for out- be transmitted to pigs and sometimes produces lesions
er-membrane protein A (ompA) have been obtained (Harris et al. 1984; Vasquez-Cisneros et al. 1994). C.
(Kaltenbroeck and Storz 1992; Kaltenbroeck et al. 1993; psittaci occurs in many avian species and is particularly
Anderson et al. 1996). These sequences have contributed common in pigeons and doves but may occur in almost
to the current species classification and are now widely any bird. Some mammals such as sheep, cattle, and ro-
used in diagnosis. C. psittaci contains a plasmid, and dents may be infected. All these species may form a reser-
DNA sequences of it have also been identified in C. peco- voir of C. psittaci infection for the pig. The relationship
rum. between C. pecorum and C. trachomatis infections of oth-
Within 6–9 hours after an elementary body has en- er mammalian species and the pig has not yet been ex-
tered a cell, it forms a reticulate body 1 µm (1000 nm) in plored in detail. All chlamydiae can survive for consider-
diameter. This body divides by binary fission to form fur- able lengths of time as elementary bodies in the
ther reticulate bodies in an inclusion within the host cell. environment, where they are resistant to drying. The ma-
At this stage the infected host cell may divide to give in- jor routes of transmission of C. psittaci to pigs are by in-
fected daughter cells, which may account for latent infec- halation of aerosols of elementary bodies, either fresh or
tions seen in animal hosts. Within 20 hours of the first in dust, from respiratory, genital tract, or enteric infec-
division of the reticulate body, some begin to mature in- tions; by ingestion of contaminated feed; and by contact,
to elementary bodies. The chlamydial inclusions may oc- particularly venereal in the case of genital tract infec-
cupy up to three-quarters of the cell volume and contain tions. The exact methods by which C. pecorum and C. tra-
up to 10,000 elementary bodies. C. trachomatis produces chomatis are transferred are not yet clear but are proba-
glycogen inclusions in cells at this stage; C. psittaci and C. bly similar, with the fecal-oral route being of importance
pecorum do not. Infected cells may lyse to release the ele- for enteric infections and transmission by flies or dust
mentary bodies or these may be budded from persistent- being involved in C. trachomatis conjunctivitis (Rogers et
ly infected cells. al. 1993).
Chlamydiae can be grown in the laboratory by inocu- Chlamydial infection in the pig has been reported
lation of the yolk sac of 6- to 8-day-old embryonated hen from the United States (Willigan and Beamer 1955;
eggs and in neonatal mice. Most laboratories now use Pospischil and Wood 1987), Britain (Wilson and Plum-
cell cultures, usually McCoy or L929 cells, in which the mer 1966; Harris 1976), Rumania (Sorodoc et al. 1961),
chlamydiae grow readily. Some strains and species can Germany (Stellmacher et al. 1983), and more recently
be maintained in Vero cells. In some cases, isolation can from other countries. The early serologic surveys sug-
be improved by treatment of the cells, using irradiation gested that complement-fixing and microagglutinating
or cycloheximide (1 µm/mL), and by centrifugation of antibodies were present in up to 23% of slaughter pigs
the chlamydiae onto the cells. (Wilson and Plummer 1966). Most antibody titers were
The different species of chlamydiae and biotypes low (1:8–1:128), but titers as high as 1:1024 were record-
within them can be distinguished by their differential ed (Wilson and Plummer 1966). More recent surveys us-
growth in cell cultures in the presence of tissue culture ing immunoperoxidase staining of histological sections
medium supplemented with amino acids such as argi- of gut suggest that chlamydial inclusions can be found in
nine, isoleucine, and methionine (Johnson 1984) and by up to 67% of piglets (Zahn et al. 1995) and 99% of fin-
the presence of inclusions or the time taken to develop in ishing pigs (Szeredi et al. 1996). Serum antibody surveys
a particular cell line. The main methods of differentia- of the same finishing animals using an ELISA method
tion are now (1) antigenic, based on differences between confirmed antibody in 82.6%, but the complement-fixa-
the outer-membrane proteins detected using monoclon- tion test (CFT) detected antibody in only 28.6%, a figure
al antibodies in immunoperoxidase, immunofluores- similar to that of Wilson and Plummer (1966). It is clear,
cence, or ELISA methods, and (2) nucleic acid based, us- therefore, that infection with chlamydiae is widespread
ing genomic and ompA sequences for PCR (Anderson et among pigs.
al. 1996; Kaltenbroek and Storz 1992; Kaltenbroeck et al. Studies of the distribution of infection within infect-
1993). ed herds suggest that infection can occur in animals of
every age group. Samples from which chlamydiae have
EPIDEMIOLOGY been isolated include semen samples from boars; fetuses,
both live and aborted; sows; lungs, joints, and organs
The recent development of methods to differentiate the such as liver and spleen from piglets; store pigs; and pigs
three species of chlamydiae present in pigs has raised a at slaughter. Enteric infection is uncommon in piglets of
number of questions about the epidemiology of infec- less than 4 weeks of age (6.9%) and more common
tions with these species in the pig. All three species are (41.8%) in piglets aged more that 4 weeks (Zahn et al.
found in other animals or birds, and experimental infec- 1995), and conjunctival infection was recorded by
CHAPTER 44 MISCELLANEOUS BACTERIAL INFECTIONS Taylor 621

Rogers et al. (1993) as associated with clinical signs be- results in little or no further disease. This development of
tween 2 and 8 weeks of age. The presence of low levels of immunity is accompanied by development of comple-
antibody in piglets may suggest maternal antibody trans- ment-fixing antibodies to C. psittaci, which appear with-
fer. The possibility of transmission of intestinal chlamy- in 2 weeks and remain detectable for a variable period.
diae to humans from slaughter pigs was raised by Szere- Information on the time course of antibody detectable
di et al.(1996). by ELISA and indirect immunofluorescence is lacking.
In genital infections, infected semen given to sows
PATHOGENESIS has resulted in the birth of weak piglets and continued
shedding of chlamydiae for up to 20 months.
Experimental infections were carried out with “C.
psittaci” or “chlamydiae” prior to the identification of the CLINICAL SIGNS
presence of three species of chlamydiae in pigs. The ac-
count which follows uses the term “C. psittaci” unless Many chlamydial infections are inapparent, but consis-
there is evidence that the organisms were of the other tent features of respiratory tract and generalized infec-
species. tions include an incubation period of 3–11 days followed
Elementary bodies of C. psittaci enter by the respira- by inappetence and a rise in rectal temperature to
tory, oral, or genital routes and enter epithelial cells, in 39–41˚C. Dyspnea, pneumonia and conjunctivitis may
which they multiply or are taken up by macrophages and occur and may persist for 4-8 days. Evidence of pleurisy
distributed to lymph nodes. Infection may be local at the or pericarditis may be detected by auscultation, and ar-
portal of infection and remain inapparent or latent; may ticular involvement by lameness in one or more joints. In
cause local disease such as pneumonia, enteritis, or dis- slaughter pigs, polyarthritis associated with synovitis has
turbances of reproduction; or may become generalized. been reported. Other disturbances of gait include weak-
C. psittaci isolates of avian, bovine, ovine, and porcine ness in piglets and nervous signs in pigs of all age
origin have been used in experimental infections; but groups. Fatal infections are most commonly reported in
strains of porcine origin appear to be most virulent for younger animals.
the pig, provided they have not become yolk sac or tissue Diarrhea has been reported to be associated with
culture adapted. chlamydial infection (Pospischil and Wood 1987) and
There appears to be some adaptation of strains to the can be produced experimentally in gnotobiotic pigs with
method of transmission, in that strains of genital origin isolates from diarrheic animals (Rogers and Anderson
(Kielstein et al. 1983) do not appear to cause severe pneu- 1996), but retrospective analysis of cases by Nietfeld et
monia, and parenteral inoculation was found necessary al. (1997) failed to confirm that infection (demonstrated
to reproduce arthritis with an arthritis isolate. Pneumo- by immunoperoxidase staining of the intestinal epitheli-
nia has been consistently produced by intranasal or in- um) was statistically associated with diarrhea. Many re-
tratracheal inoculation with porcine strains (Kielstein et ports deal with genital tract infection and disturbances
al. 1983; Martin et al. 1983; Stellmacher et al. 1983; in reproduction. In the boar, infection is associated with
Rogers et al. 1996), but infection was found to spread orchitis, epididymitis, and urethritis; while infections in
consistently to other organs. An acute exudative or inter- gilts and sows have resulted in late abortions and the
stitial pneumonia with peribronchiolar cellular cuffing birth of dead or weak piglets. Serologic and isolation
and a lobular distribution occurs within 4–8 days of in- studies suggest that many genital tract infections are
fection. Lesions are fully developed by 8–12 days postin- clinically inapparent.
fection and are largely resolved by 4 weeks after infec-
tion, although infection may still be present in the lungs. LESIONS
Similar lesions were produced in germfree pigs by Rogers
et al. (1996) using a C. trachomatis–like isolate from a Lesions in which C. psittaci has been demonstrated often
case of pneumonia. They identified mild multifocal contain other agents, and many descriptions of the le-
rhinitis and diarrhea in addition to the pneumonic sions found in field cases may not take into account the
changes. Rogers and Anderson (1996) infected gnotobi- presence of agents such as mycoplasmas or viruses. The
otic piglets with isolates from diarrheic pigs to produce large body of work on respiratory disease suggests that
diarrhea after 4–5 days which lasted for 8 days. The api- lung lesions are distributed posteriorly in most cases, al-
cal part of the villus appeared to be colonized in the dis- though occasional patches of pneumonia may occur in
tal jejunum and ileum, with little or no infection in the the anterior lobes (Harris et al. 1984).
colon. Villous atrophy developed and this was later fol- Lesions are irregular and raised, are of firm consis-
lowed (7–10 days postinfection) by mild focal serositis. tency, extend deep into the lung tissue, are limited by
Contact infections suggest that natural infection is lobular boundaries, and are clearly demarcated from ad-
normally less severe and that reinfection after 3–4 weeks jacent grossly normal tissue. Early lesions are pale red,
622 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

becoming grayish as they age. Enlarged bronchial lymph ence of high levels of antibody (1:256) may be sufficient.
nodes may be present. The microscopic findings include As only low levels of complement-fixing antibodies may
thickening of the alveolar septae by capillaries, septal arise from infections in sites such as the respiratory, en-
edema, and neutrophils in peribronchial and subepithe- teric, and genital tracts, the absence of high levels of
lial sites. Neutrophils and macrophages are common in complement-fixing antibody does not rule out chlamydi-
the alveolar lumina, and in some areas this exudate oc- ae as a cause of disease.
cludes terminal bronchioles. Edema and massive epithe- Chlamydiae may be detected in smears of discharges
lial cell shedding have been reported in severely affected or postmortem specimens and in histological specimens
lung lobules (Martin et al. 1983). Foci of type II pneu- after staining by Giemsa’s method. The organisms are
mocyte hypertrophy and hyperplasia and vacuolated tiny (0.2–1.0 µm) and are present in large numbers in
pneumocytes and bronchial epithelium have been re- cells. A more satisfactory method is to use Koster’s stain
ported in C. trachomatis–like experimental infection in which a fixed smear is stained for 5 minutes with car-
(Rogers et al. 1996). Peribronchiolar accumulations of bol fuchsin, decolorized for 30 seconds with 0.25% acetic
plasma cells, lymphocytes, and macrophages are also acid, and counterstained for 1 minute with 1% aqueous
common. There appears to be no pleurisy in experimen- Loeffler’s methylene blue. The chlamydiae appear as
tal infections, and no gross changes in other organs were clusters of intracellular red dots against a blue back-
reported beyond enlargement of the bronchial lymph ground (Fig. 44.2). Most specific of all is the immunoflu-
nodes. Antigen can be demonstrated in bronchial and orescence test using specific fluorescein-conjugated anti-
bronchiolar epithelial cells and in pneumocytes in exper- body to C. psittaci to demonstrate infected cells.
imental studies (Rogers et al. 1996) and in field cases Immunoperoxidase tests have been described (Chasey et
(Done et al. 1992). al. 1981), and the immunoperoxidase staining of fixed
The other lesions reported to occur in field cases in- tissue sections is now standard and appears to be the
clude pericarditis, pleurisy, hemorrhages of kidney and most sensitive method of detection (Szeredi et al. 1996)
bladder, and enlargement of the spleen. There is little in tissue. Many laboratories are using PCR tests to con-
doubt that synovitis accompanies the arthritic changes firm the presence of the three species in feces and tissue
and that orchitis in boars is accompanied by interstitial specimens. Primers include genomic DNA sequences,
edema and tubular degeneration. Aborted piglets may be 16S rRNA gene sequences, ompA gene sequences, and
mummified; stillborn or weak piglets may have lung, liv- plasmid sequences.
er, or enteric lesions. The organism has been isolated Isolation can also be carried out by the inoculation of
from pseudomembranous colitis in experimental S. ty- young mice and of 6- to 8-day fertile hen eggs. More than
phimurium infections (Pospischil and Wood 1987), and one subculture may be necessary before infection can be
extensive studies of pig intestines by immunoperoxidase detected. Cell cultures using L929 or McCoy cell lines
(Zahn et al. 1995; Pospischil et al. 1996; Szeredi et al. treated with cycloheximide (1 µg/mL) may be inoculated
1996; Nietfeld et al. 1997) confirm the distribution of the by centrifugation (Farmer et al. 1982) in tissue culture
inclusions in the small-intestinal villi in piglets and in the medium at pH 7.0. Inclusions are at a maximum after 48
large-intestinal intercrypt epithelium in finisher pigs. Le- hours of incubation at 35–37˚C.
sions in experimentally infected gnotobiotic piglets in- Transport media for chlamydiae should contain
cluded watery colon contents with flakes of undigested streptomycin (50–100 mg/L) or gentamicin (10–20
curd, villous atrophy, lymphangitis, and multifocal mg/L) with vancomycin (100 µg/mL) and nystatin (25
necrosis of the apical portion of the villi (Rogers and An- mg/L). Samples can be stored at 4˚C or at −70˚C. Han-
derson 1996). dling C. psittaci is dangerous, and severe human infec-
tions and death can result. Appropriate safety precau-
DIAGNOSIS tions should be observed.

The clinical signs of chlamydial infection are not distinc- TREATMENT


tive, but it must be considered as a possible cause of
pneumonia, polyarthritis, enteritis, late abortion, still- A number of antimicrobials have some effect on C.
births, mummified piglets, and orchitis. The gross le- psittaci in vitro, but the most satisfactory compounds for
sions in the lung may be suggestive of chlamydial infec- treatment are the tetracyclines. Treatment for inade-
tion, but any firm diagnosis involves laboratory tests. quate times may result in relapse; for complete elimina-
These are serologic: complement fixation using heat-sta- tion or suppression of infection to the latent state, 21-
ble C. psittaci antigen, and ELISA using tissue culture day treatment should be given at the therapeutic level.
antigen (Szeredi et al. 1996); microscopic agglutination Tetracycline, oxytetracycline, and chlortetracycline can
(Wilson and Plummer 1966); and indirect immunofluo- all be used in drinking water or feed. Long-acting oxyte-
rescence. Complement-fixing antibodies should ideally tracycline injections are useful for treating individual in-
be found to rise in paired serum samples, but the pres- fected animals.
CHAPTER 44 MISCELLANEOUS BACTERIAL INFECTIONS Taylor 623

44.2. Photomicrograph of
elementary bodies of
Chlamydia sp. (arrow) in
aborted placenta (×1200).

PREVENTION Chlamydia psittaci isolated from the eyes of domestic


ducks (Anas platyrhynchos) with conjunctivitis and rhini-
Pigs should be prevented from coming into contact with tis. Vet Rec 110:59.
infected pigs, other mammalian species, and bird drop- Harris, J. W. 1976. Chlamydial antibodies in pigs in Scot-
pings. Infected pigs should be maintained in separate air land. Vet Rec 98:505–506.
and drainage spaces from susceptible animals. Any in- Harris, J. W.; Hunter, A. R.; and Martin, D. A. 1984. Experi-
fected breeding stock should be used only after tetracy- mental chlamydial pneumonia in pigs. Comp Immun
cline treatment or kept with other infected stock in isola- Microbiol Infect Dis 7:19–26.
tion until sufficient uninfected animals are available to Johnson, F. W. A. 1984. Isolation of C. psittaci from nasal
replace them. Disinfection with phenols and formalin fu- and conjunctival exudate of a cat. Vet Rec 114:342–344.
migation will eliminate elementary bodies from build- Kaltenbroeck, B., and Storz, J. 1992. Biological properties
ings. and genetic analysis of the ompA locus in chlamydia iso-
lated from swine. Am J Vet Res 53:1482–1487.
REFERENCES Kaltenbroeck, B.; Kousoulas, K. G.; and Storz, J. 1993. Struc-
tures of and allelic diversity and relationships among
Anderson, I. E.; Baxter, S. I. F.; Dunbar, S.; Rae, A. G.; the major outer membrane protein (omp A) genes of the
Philips, H. L.; Clarkson, M. J.; and Herring, A. J. 1996. four chlamydial species. J Bacteriol 175:487–502.
Analyses of the genomes of chlamydial isolates from ru- Kielstein, P.; Stellmacher, H.; Horsch, F.; and Martin, J.
minants and pigs support the adoption of the new 1983. Zur ChlamydienInfektion des Schweines. I. Mit-
species Chlamydia pecorum. Int J Syst Bacteriol teilung zur experimentellen Chlamydien-Pneumonie
46:245–251. des Schweines. Arch Exp Veterinärmed 37:569–586.
Chasey, D.; Davis, P.; and Dawson, M. 1981. Immunoperox- Martin, J.; Kielstein, P.; Stellmacher, P.; and Horsch, F. 1983.
idase detection of Chlamydia ovis in experimentally in- Zur Chlamydien Infektion des Schweines. II. Mitteilung:
fected cell culture. Br Vet J 137:634–638. Pathologische-histologische Besonderhect der experi-
Done, S. H.; McGill, I.; Spencer, Y.; and Simmons, J. 1992. mentellen Chlamydien Pneumonie des Schweines. Arch
Chlamydia psittaci and necrotizing interstitial pneumo- Exp Veterinärmed 37:939–949.
nia. Proc Int Congr Pig Vet Soc 12:341. Nietfeld, J. C.; Leslie-Steen, P.; Zeman, D. H.; and Nelson, D.
Farmer, H.; Chalmers, W. S. K.; and Woolcock, P. R. 1982. 1997. Prevalence of intestinal chlamydial infection in
624 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

pigs in the Midwest as determined by immunoperoxi- 1983. Zur Bedeutung der Chlamydien-Infektion des
dase staining. Am J Vet Res 58:260–264. Schweines unter besonder Berucksichtigung der Pneu-
Pospischil, A., and Wood, R. L. 1987. Intestinal Chlamydia monien. Monatsh Veterinärmed 38:601–606.
in pigs. Vet Pathol 24:568–570. Szeredi, L.; Schiller, I.; Sydler, T.; Guscetti, F.; Heinen, E.;
Pospischil, A.; Thoma, R.; Schiller, I.; Sydler, T.; and Corboz, L.; Eggenberger, E.; Jones, G. E.; and Pospischil,
Guscetti, F. 1996. Chlamydia in pigs. Pig J 37:9–13. A. 1996. Intestinal chlamydia in finishing pigs. Vet
Rogers, D. G., and Anderson, A. A. 1996. Intestinal lesions Pathol 33:369–374.
caused by two swine chlamydia isolates in gnotobiotic Vasquez-Cisneros, C.; Wilsmore, A. J.; and Bolle, E. 1994.
pigs. J Vet Diagn Invest 8:433–440. Experimental infections of pregnant sows with ovine
Rogers, D. G.; Anderson, A. A.; Hogg, A.; Nielson, D. L.; and chlamydia strains. Vet Microbiol 42:383–387.
Huebert, M. A. 1993. Conjunctivitis and keratoconjunc- Willigan, D. A., and Beamer, P. D. 1955. Isolation of trans-
tivitis associated with chlamydias in swine. J Am Vet missible agent from pericarditis of swine. J Am Vet Med
Med Assoc 203:1321–1323. Assoc 126:118–122.
Rogers, D. G.; Anderson, A. A.; and Hunsaker, B. D. 1996. Wilson, M. R., and Plummer, P. A. 1966. A survey of pig
Lung and nasal lesions caused by a swine chlamydial iso- sera for the presence of antibodies to the P.L.V. group of
late in gnotobiotic pigs. J Vet Diagn Invest 8:45–55. organisms. J Comp Pathol 76:427–433.
Sorodoc, G.; Surdan, C.; and Sarateanu, D. 1961. Cercetari Zahn, I.; Szeredi, L.; Schiller, I.; Straumann Kunz, U.; Buer-
asupra identificarii virusului pneumoniei enzootice a gi, E.; Guscetti, F.; Heinen, E.; Corboz, L.; Sydler, T.; and
porcilor. Stud Cerc Inframicrobiol 12 (Suppl):355– Pospischil, A. 1995. Immunologischer nachweis von
364. Chlamydia psittaci/pecorum und Chlamydia trachomatis
Stellmacher, H.; Kielstein, P.; Horsch, F.; and Martin, J. im Ferkel Darm. J Vet Med Series B 42:266–276.

ACTINOBACILLUS SUIS
Septicemia and death caused by Actinobacillus suis and produce catalase, oxidase, and urease; hydrolysis of es-
occasionally by A. equuli in suckling and recently weaned culin; and acid production without gas from arabinose,
pigs have been reported sporadically from several pig- lactose, salicin, and trehalose, but not from mannitol or
rearing countries (Van Dorssen and Jaartsveld 1962; Cut- sorbitol. Its biochemistry and antigenicity have recently
lip et al. 1972; Windsor 1973; Mair et al. 1974; MacDon- been studied by Bada et al. (1996). Distinction from iso-
ald et al. 1976). A. suis outbreaks resembling erysipelas lates of A. pleuropneumoniae biotype II is difficult but can
have been reported in older pigs and sows in Canada be achieved biochemically and using DNA analysis. A.
(Miniats et al. 1989), and the organism has also been the equuli differs from A. suis by being nonhemolytic; pro-
cause of disease resembling pleuropneumonia in older ducing acid from mannitol but not from arabinose, cel-
pigs in the United States (Yaeger 1996) and the United lulose, and salicin; and not splitting esculin. A. suis is
Kingdom. Most outbreaks occur as sudden death of one pathogenic for mice; A. equuli is not. A. suis and A. equuli
or several piglets in one, two, or, rarely, multiple litters in are killed within 15 minutes at 60˚C and are sensitive to
individual herds. Infection with A. suis is probably wide- most disinfectants. They die out within a few days in cul-
spread but disease is seldom reported. ture and pathologic material.

ETIOLOGY EPIDEMIOLOGY

A. suis is a gram-negative, nonmotile, nonencapsulated, A. suis can be carried in the tonsils and nostrils of
aerobic, and facultative anaerobic coccobacillus, 0.5–3 healthy pigs of any age and in the vaginas of apparently
µm long and about 0.8 µm in diameter. Filamentous healthy sows (Ross et al. 1972). Clinical disease occurs in
forms occur. Grayish, adherent, circular, translucent neonates and suckling pigs up to and just after weaning
colonies measuring 1–2 mm form on blood agar within age, and less commonly in sows and mature swine (Mini-
24 hours. On horse blood agar, colonies are surrounded ats et al. 1989; Sanford et al. 1990). With the separation
by a narrow but distinct zone of alpha hemolysis, and on of pigs and horses in modern farming systems, infection
calf and sheep blood agars, by a wide zone of beta (com- in pigs by A. equuli seems to have diminished.
plete) hemolysis. The organism grows on MacConkey Outbreaks of clinical disease associated with A. suis
agar. Biochemically, A. suis can be differentiated from infection occur more frequently in minimal-disease and
other related bacteria isolated from pigs by its ability to other high-health-status herds (Miniats et al. 1989; San-
CHAPTER 44 MISCELLANEOUS BACTERIAL INFECTIONS Taylor 625

ford et al. 1990), possibly because the lack of immunity pigs, anorexia, fever, a persistent cough, respiratory dis-
in these pigs allows virulent A. suis organisms to express tress (Yaeger 1996) and pneumonia are reported; recov-
their pathogenic potential, but the organism can be re- ered animals may remain unthrifty. In outbreaks in ma-
covered from herds in which disease is not apparent. ture animals, fever, round and rhomboid erythematous
skin lesions, inappetence, and sudden deaths are charac-
PATHOGENESIS teristic, but mortality is usually low. Metritis, meningitis,
and abortion have been reported in sows. The disease
The pathogenesis of A. suis infection has not been de- may be confused with erysipelas, especially when skin le-
fined. Infection probably occurs via the upper respirato- sions develop, and with pleuropneumonia when respira-
ry tract, and the disease has been reproduced by in- tory signs are present.
tranasal inoculation (Fenwick et al. 1996), although
invasion through abrasions in the skin and mucous LESIONS
membranes is also likely. In susceptible animals, septic
emboli then spread rapidly to multiple organs and tis- The most striking gross lesions are petechial to ecchy-
sues throughout the body and either are trapped in ves- motic hemorrhages in any of the following organs: lung,
sels or adhere to vessel walls, forming microcolonies sur- kidney, heart, liver, spleen, skin, and intestines. The le-
rounded by areas of hemorrhage and necrosis. Virulence sions are especially prominent and most frequently seen
factors of A. suis have not been specifically determined, in the lung, where lobular necrosis and serofibrinous ex-
but lipopolysaccharide, polysaccharides in the cell wall, udates also occur (Fig. 44.3). Increased serous or serofib-
outer-membrane proteins, and, in some strains, a 104 rinous exudates may occur in the thorax and the peri-
kDa hemolysin (ApxI) are all potential virulence factors cardium. Pleurisy, pericarditis, and miliary abscesses in
likely to be involved in pathogenesis. Antibodies to ApxI the lung, liver, skin, mesenteric lymph nodes, and kidney
have been demonstrated in the sera of pigs which have may be seen in older suckling or weaned pigs. The pneu-
recovered from experimental infection (Fenwick et al. monic lesions may resemble those of pleuropneumonia.
1996). Pigs may die within 15 hours of infection. Arthritis (Van Dorssen and Jaartsveld 1962; Odin 1994)
and valvular endocarditis (Jones and Simmons 1971)
CLINICAL SIGNS have been reported. In mature animals, numerous
round, rhomboid, or irregular skin lesions are com-
Sudden death of suckling piglets, 2 days to 4 weeks old, mon.
in one or more litters is often the first indication of an Histologically, bacterial thromboemboli with accom-
outbreak of actinobacillosis. Deaths in piglets are some- panying fibrinohemorrhagic necrosis in randomly scat-
times mistakenly attributed to crushing. Cyanosis, pe- tered vessels in the lung, liver, kidney, skin, spleen, heart,
techial hemorrhages, fever (up to 40˚C, 104˚F), and pericardium, meninges, and brain are characteristic (Fig.
panting, sometimes accompanied by shaking and/or 44.4). Bacterial emboli may be surrounded by radiating
paddling, may be seen prior to death in suckling pigs. eosinophilic clublike colonies. These are most obvious in
Congestion of extremities (leading to necrosis of feet, the lung, where there may be large coalescing areas of
tail, and ears) and swollen joints may occur. In weaned necrosis.

44.3. Lung of 3-day-old piglet with actinobacillosis. Note the hemorrhages.


44.4. Photomicrograph of a microabscess in the lung as shown in Fig. 44.3. Note the
microcolonies of bacteria (H&E; ×110).

DIAGNOSIS pigs, responses to treatment with ampicillin (5 mg/kg)


orally or parenterally, injectable benzathine-procaine
Sudden mortality in suckling pigs in individual litters in penicillin G (2.25–3.0 × 106 IU) intramuscularly (IM),
herds with previous A. suis outbreaks usually indicates a injectable procaine penicillin (1.8–2.4 × 106 IU) IM, or
new outbreak. The gross lesions of hemorrhages and in-feed medication with oxytetracycline hydrochloride
necrosis in the lung and/or skin and kidney and splenic (550 g/ton) and/or streptomycin for periods up to 1
enlargement are suggestive of A. suis infection. In mature week have all been excellent.
pigs, fever, inappetence, and skin lesions resembling
erysipelas, especially in herds already vaccinated against PREVENTION
erysipelas, should raise a suspicion of A. suis. Micro-
scopic lesions consisting of bacterial emboli, necrosis, Autogenous bacterins have not been critically evaluated
and inflammatory cells in the lung and other organs are but have been used in herds with repeated A. suis out-
also suggestive. Diagnosis, however, depends on isola- breaks with apparent success.
tion of A. suis or A. equuli from the lesions. A. suis infec-
tion should be considered when pleuropneumonia is sus- REFERENCES
pected in herds thought to be free from that disease. In
these herds it may cause mild or atypical lung lesions and Bada, R.; Mittal, K. R.; and Higgins, R. 1996. Biochemical
give rise to antibody to ApxI but not to the cytotoxin of and antigenic relationships between porcine and equine
A. pleuropneumoniae or to its somatic antigens (Fenwick strains of Actinobacillus suis. Vet Microbiol 51:393–396.
et al. 1996). A suis can usually be isolated from the tonsils Cutlip, R. C.; Amtower, W. C.; and Zinober, M. R. 1972. Sep-
of piglets aged 2–10 days in such herds. tic embolic actinobacillosis of swine: A case report and
laboratory reproduction of the disease. Am J Vet Res
TREATMENT 33:1621–1626.
Fenwick, B.; Rider, M.; Chengappa, M.; and Montarz, J.
A. suis is sensitive to most commonly used antibiotics. 1996. Cross protective immunity between Actinobacillus
Since outbreaks in suckling pigs are so acute and unpre- suis and A. pleuropneumoniae. Proc Int Congr Pig Vet Soc
dictable, however, treatment is usually too late. In older 14:373.
CHAPTER 44 MISCELLANEOUS BACTERIAL INFECTIONS Taylor 627

Jones, J. E. T., and Simmons, J. R. 1971. Endocarditis in the bec 24:61–65.


pig caused by Actinobacillus equuli: A field and an exper- Ross, R. F.; Hall, J. E.; Orning, A. P.; and Dale, S. E. 1972.
imental case. Br Vet J 127:25–29. Characterization of an Actinobacillus isolated from the
MacDonald, D. W.; Hewitt, M. P.; Wilton, G. S.; Rawluk, S.; sow vagina. Int J Syst Bacteriol 22:39–46.
and Childs, L. 1976. Actinobacillus suis infections in Al- Sanford, S. E.; Josephson, G. K. A.; Rehmtulla, A. J.; and
berta swine, 1973–1975: Pathology and bacteriology. Tilker, A. M. E. 1990. Actinobacillus suis infection in pigs
Can Vet J 17:251–254. in southwestern Ontario. Can Vet J 31:443–447.
Mair, N. S.; Randall, C. J.; Thomas, G. W.; Harbourne, J. F.; Van Dorssen, C. A., and Jaartsveld, F. H. J. 1962. Acti-
McCrea, C. T.; and Cowl, K. P. 1974. Actinobacillus suis nobacillus suis (novo species), a bacterium occurring in
infection in pigs: A report of four outbreaks and two swine. Tijdschr Diergeneeskd 87:450–458.
sporadic cases. J Comp Pathol 84:113–119. Windsor, R. S. 1973. Actinobacillus equuli infection in a litter
Miniats, O. P.; Spinato, M. T.; and Sanford, S. E. 1989. Acti- of pigs and a review of previous reports on similar in-
nobacillus suis septicemia in mature swine: Two out- fections. Vet Rec 92:178–180.
breaks resembling erysipelas. Can Vet J 30:943–947. Yaeger, M. J. 1996. An outbreak of Actinobacillus suis sep-
Odin, M. 1994. Les infections a Actinobacillus suis au Que- ticemia in grow/finish pigs. J Vet Diagnost Invest
bec: une étude retrospective de 22 cas. Méd Vét du Que- 8:381–383.

YEASTS
Yeasts are fungi that are normally single celled but can coccus have been recorded. Cryptococcus neoformans has
form filaments (or pseudohyphae). Some can sporulate been isolated from cryptococcosis in pigs, but the disease
to produce resistant spores. They occur in the food of the is rare in this species and occurs only where the organism
pig and in dusts. Some species are commonly found on is commonly found in other livestock.
the skin and mucous membranes. In certain situations, Candida albicans is the species of Candida most fre-
yeasts of a number of species (but principally Candida quently reported; but C. tropicalis, C. pseudotropicalis, C.
albicans) may be isolated from inflammatory lesions of brumptii, C. sloofii, C. rugosa, C. lipolytica, C. krusei, and C.
the oral cavity, gastrointestinal tract, urogenital tract, scottii have been isolated from lesions or feces of appar-
and skin. Their isolation from such lesions is often asso- ently healthy pigs. Since C. albicans is associated most
ciated with use of therapeutic antimicrobials, especially frequently with specific lesions, both it and its relation-
in piglets. Yeasts belonging to the genus Malassezia, pos- ship to these lesions will be described here.
sibly M. pachydermatis, are present in the ears and on the Candida spp. are spherical cells, 2.5–6 µm in diame-
skin of pigs, but their role in disease at these sites is not ter. They reproduce by budding (blastospores) and
known. They can reach high numbers when skin or ear chlamydospores, which bud from filaments (or pseudo-
lesions develop, but so little is currently known about hyphae) on chlamydospore agar, particularly under re-
them and their role in disease that they will not be con- duced oxygen tension at 25˚C (Carter 1979). Pseudohy-
sidered further. phae and oval yeast forms are found in lesions. Candida
Yeasts may also form a major part of the diet of the spp. grow readily on Sabouraud agar, malt agar, and of-
pig, as either yeast wastes from brewing or distilling or ten on blood agar incubated aerobically. They form 1–2
yeast grown and treated specifically as a component of mm, creamy white, opaque, circular colonies within
rations. These yeasts can provide high protein and, in 24–48 hours at 37˚C and within 2–4 days at 25˚C. C. al-
particular, high lysine. Traces of paraffin waxes have bicans produces chlamydospores and germ tubes but no
been found in the fat of pigs fed on yeasts grown on that pellicle when grown in broth and ferments glucose, mal-
substrate, and there are some reports of diarrhea and in- tose, and galactose but not sucrose or lactose. It is not
creased kidney weights in yeast-fed pigs. Most reports in- known to produce toxins, although there have been sug-
dicate that inclusion of yeasts in the ration does not ad- gestions that it can produce keratolytic enzymes in the
versely affect the health of pigs. presence of glucose.

ETIOLOGY EPIDEMIOLOGY

Yeasts identified in infections in pigs belong to a number C. albicans has been identified in the bedding, feed, and
of genera. Those of the genus Candida are most com- water supplies of pigs. It occurs on the skin and in the
monly isolated, although species of Torulopsis, Tri- oral cavity, stomach, and intestines of normal pigs in
chosporon, Rhodotorula, Pichia, Pityrosporum, and Crypto- small numbers. It can be shed in the feces and exhaled in
628 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

droplets by animals with oral infections. It may be isolat- ynx, and sometimes on the soft or hard palate. These
ed from the feces of birds, rodents, and other animal patches may coalesce to form larger areas of
species and may cause disease in those species, which pseudomembranous material that may occlude the lu-
may become sources of the organism for pigs. Organ- men of the esophagus. The lesions may extend down the
isms in the environment may multiply in moist condi- esophagus and may be seen on the gastric mucosa. There
tions in the presence of suitable substrates such as may be small hemorrhages in the cardiac area and white
spilled meal or garbage. pseudomembranous lesions in the esophageal area. De-
scriptions of the lesions distal to the stomach are rarely
PATHOGENESIS published, but in heavily infected animals they resemble
those of chronic enteritis, with villous atrophy and thick-
C. albicans appears to colonize debilitated skin surfaces ening of the mucosa. When the white pseudomembra-
and lesions on other mucous surfaces. The predisposing nous material is removed, congestion of the mucosal sur-
factors appear to include the effects of artificial rearing face may be seen, but ulceration is rare. In older pigs, C.
of piglets (Osborne et al. 1960) and chronic enteritis of- albicans may be isolated from gastric ulcers, but these do
ten associated with treatment with broad-spectrum an- not differ grossly from uninfected ones. Gross lesions
timicrobials. Gastric ulcers appear to be colonized by may be seen in cutaneous candidiasis and include a gray-
yeasts rather than initiated by them, and cutaneous can- ish surface deposit, thickening of the skin, and hair
didiasis often results from exposure to continually warm loss.
moist conditions that are accompanied by poor hygiene Microscopic lesions include the presence of numer-
and food residues (Reynolds et al. 1968). ous yeasts on the epithelial surface, with pseudohyphal
Invasion of mucous surfaces appears to follow accu- filaments visible as 1.5–2.0 µm deeply staining threads in
mulation of yeast forms on the debilitated surface and the epithelium. In lesions on the tongue, yeast cells and
develops with pseudohyphal invasion of the superficial pseudohyphae may be seen in cavities beneath the papil-
layers of the epithelium. Systemic invasion is rare and lae. They may also be present in large numbers in the pe-
the inflammatory response to infection is slight. riphery of infected gastric ulcers. Degenerative changes
are frequently present in the infected epithelium. They
CLINICAL SIGNS include desquamation of epithelial cells, capillary dila-
tion, edema of the submucosa or dermis (depending on
Yeasts have been implicated in chronic gastroenteritis in the epithelial surface attached), and presence of inflam-
piglets, gastric ulceration, and cutaneous and oropha- matory cells. These are neutrophils in the early lesions,
ryngeal infections. Piglets are often 3–5 days old (more later (in 4- to 5-day-old lesions) accompanied by
commonly, 7–14 days) before yeast infection complicates eosinophils, macrophages, plasma cells, and lympho-
the underlying problem. The clinical signs of gastroen- cytes.
teritis complicated by yeasts are not specific, but there is
often a history of dullness, inappetence, vomiting, and DIAGNOSIS
chronic diarrhea that may be grayish or blackish de-
pending on the diet and has failed to respond to the use In piglets the appearance of the white 2–5 mm lesions in
of broad-spectrum antibiotics such as tetracyclines. the oral cavity may suggest that candidiasis is present,
Piglets may die after 10–14 days of illness. In many cases but diarrhea and association of infection with gastric ul-
there are characteristic yellowish white, circular, 2–5 mm ceration may not be identified on clinical grounds. Skin
lesions on the tongue and hard palate, which resemble changes may also suggest a diagnosis of candidiasis. A
colonies of C. albicans on artificial media. When scraped history of chronic enteritis and broad-spectrum antibi-
off, no macroscopic changes are seen beneath them. Cu- otic use or housing in moist conditions is often sugges-
taneous candidiasis often presents as a moist gray exu- tive of candidiasis.
date on the surface of the skin of the abdomen with little Confirmation of diagnosis is based on demonstra-
or no effect on the hair in early lesions, but later resulting tion of the organism concerned in the lesions or, in life,
in hair loss and thickening of the skin. Affected animals isolation of large numbers from the feces. The presence
are often kept in moist conditions and exposed to food of yeasts in lesions of the intestine may be established by
residues. their demonstration in Gram-stained smears in which
oval or round, gram-positive, often budding, 2.5–6 µm
LESIONS cells may be seen (Fig. 44.5). Similar bodies may be seen
in histological sections stained by hematoxylin and eosin
Piglets with candidiasis are often in poor condition with or, more easily, stained by periodic acid-Schiff or silver
chronic diarrhea. There may be lesions in the oral cavity stains such as Grocott’s (Fig. 44.6). None of these allow
and throughout the gastrointestinal tract. These consist the complete identification of the organism.
of white specks and circular patches 2–5 mm in diameter Yeasts may be isolated using Sabouraud’s agar with or
on the dorsum of the tongue, less frequently on the phar- without chloramphenicol (Carter 1979). Some, such as
CHAPTER 44 MISCELLANEOUS BACTERIAL INFECTIONS Taylor 629

44.5. Photomicrograph of
yeast cells (arrow) from the
ileal mucosa in a 10-day-old
piglet (Gram; ×1200).

44.6. Photomicrograph of
yeast cells and pseudohyphae
adjacent to the ileal mucosa
in a 10-day-old piglet
(Grocott; ×400).
630 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

C. albicans, will grow readily on horse blood agar. Incu- include discontinuation of the use of broad-spectrum
bation at 25˚C yields colonies 1–2 mm in diameter after antibiotics and their replacement with narrow-spectrum
3–4 days, but incubation at 37˚C can allow colonies to be ones if they are a factor in yeast colonization. Animals
identified within 24–48 hours. The genera can be sepa- with cutaneous candidiasis may also be scrubbed with
rated using characters such as shape of the cells, pres- suitable detergent or with hexetidine-based shampoos.
ence or absence of pseudomycelium, presence or ab-
sence of capsule, production of chlamydospores, ability PREVENTION
to split urea, and other characters (Carter 1979). Many
yeast species may be identified in culture using commer- Pigs should be maintained in warm, dry, clean condi-
cial biochemical strips such as the API Yeast series. tions, and accumulations of moist fermenting food
Isolation of large numbers of yeasts from lesions may should be prevented. Enteric diseases in piglets should
confirm a diagnosis of candidiasis, but their isolation in be treated with appropriate antimicrobials, and lengthy
small numbers from the skin, vagina, or feces and in- treatment with broad-spectrum antibiotics should be
testines of clinically normal pigs may not be significant. avoided. Disinfection of pens and pen fittings can be car-
ried out using formaldehyde vapor or 2% formaldehyde;
TREATMENT cleaning and drying of the pens will reduce levels of
yeasts to normal background levels.
C. albicans and other yeasts are sensitive in vitro to a
number of compounds such as nystatin, miconazole, REFERENCES
and amphotericin B, but only nystatin and amphotericin
B have been used in treatment (Osborne et al. 1960). Carter, G. R. 1979. Diagnostic Procedures in Veterinary Bac-
Nystatin suppressed the clinical signs but did not elimi- teriology and Mycology, 3d ed. Springfield: Charles C.
nate infection. Amphotericin B may be effective in young Thomas, chap. 30.
piglets given at a rate of 0.5 mg/kg twice daily. In many Osborne, A. D.; McCrae, M. R.; and Manners, M. J. 1960.
instances, correction of underlying disease or husbandry Moniliasis in artificially reared pigs and its treatment
factors is sufficient. Cleaning up waste food and provid- with nystatin. Vet Rec 72:237–241.
ing a dry environment caused resolution of cutaneous Reynolds, I. M.; Miner, P.; and Smith, R. E. 1968. Cutaneous
candidiasis (Reynolds et al. 1968). Treatment should also candidiasis on swine. J Am Vet Med Assoc 152:182–186.

YERSINIAS
A number of species of Yersinia have been isolated from suis. This interference is described in Chapter 28.
pigs, and reports of association between infection and Infection in pigs is usually inapparent, but Y. pseudo-
clinical disease are increasing. Y. enterocolitica has in- tuberculosis and Y. enterocolitica have been isolated from
creasingly become recognized as a cause of human food pigs with fever, enteritis, and diarrhea.
poisoning and enteritis since the late 1960s, and the pig
is an important source. There are reports in the literature ETIOLOGY
of many surveys of pig carcasses, offal, and feces for the
presence of Y. enterocolitica (Doyle et al. 1981; Schie- Yersinias are aerobic or facultatively anaerobic, gram-
mann and Fleming 1981; Hunter et al. 1983). Results of negative coccobacilli (or short rods), 1.2 µm in length
these surveys show that infection is distributed world- and 0.5–1.0 µm in diameter. They are nonmotile at 37˚C,
wide in pigs, and serotypes considered pathogenic to hu- but some are motile at lower temperatures. Species iso-
mans are commonly present. This relationship to human lated from pigs include Y. pseudotuberculosis, Y. pseudotu-
disease has stimulated a number of reports of pathogen- berculosis subsp. pestis (the plague bacillus), Y. enterocolit-
ic determinants (Mosimbale and Gyles 1982) that have ica, Y. intermedia, Y. fredrikensii, and Y. kristensenii. Only
been demonstrated in Y. enterocolitica in both porcine two species (Y. pseudotuberculosis and Y. enterocolitica)
and human isolates, and it seems clear from the work of have yet been associated with clinical disease in pigs.
Kwaga and Iversen (1993) that the pig and human strains Yersinias may be isolated in routine media, upon
are identical. Further reports deal with the antigenic re- which they appear as grayish 1–2 mm colonies within
lationships between Yersinia spp. and Brucella spp., since 24–48 hours and as similar-sized non-lactose-fermenting
infections with certain strains of the former can cause in- colonies on MacConkey agar. The species are distin-
terference with serologic tests for both B. abortus and B. guished by biochemical tests. Y. pseudotuberculosis is
CHAPTER 44 MISCELLANEOUS BACTERIAL INFECTIONS Taylor 631

motile at 22˚C, grows on citrate media at 22˚C, splits fection of 6 pigs with the isolate obtained from the clini-
urea, and does not ferment sucrose or raffinose but does cal outbreak described above. Experimental studies by
ferment mannose. Y. pseudotuberculosis subsp. pestis is Nielsen et al. (1996) have confirmed that infection of the
negative for all these characters. Y. enterocolitica is also feces may be found between 5 and 21 days after infection
motile and splits urea but ferments sucrose and does not and that tonsillar carriage persists longer. A serum anti-
grow on citrate or ferment mannose. Individual species body response develops by 19 days after infection and
can be divided into biotypes and serotypes. Capsules, at- has disappeared by 70 days postinfection. Studies in
tachment antigens, and enterotoxins have been de- suckling mice have indicated that 10 of the 12 pig iso-
scribed in organisms of this genus. lates tested produced enterotoxin and that one isolate
Y. enterocolitica has been subdivided into at least 46 O could produce fluid in piglet gut loops. The Sereny test
groups and at least 5 biotypes. Of these, most human in- for invasiveness was negative in typical pig strains
fections are associated with biotype 2, O9 and biotype 4, (Mosimbale and Gyles 1982). Pig isolates harbor the vir-
O3. Biotype 1, O8 is also associated with some human in- ulence plasmid (Kwaga and Iversen 1993) and possess
fections. Y. enterocolitica organisms of a number of O the capsular material considered essential for patho-
groups have been recorded from pigs. The actual O genicity and detected using the congo red magnesium
groups isolated may vary from one part of a country to oxalate test. Studies by Erwerth and Natterman (1987)
another (Schiemann and Fleming 1981) but include O3, suggest that oral infection is followed by establishment
O5, O6, O7, O8, O9, O13, O18, and O46. of infection in the tonsils and the development of enteri-
tis in the ileum and large intestine. Similar colonization
EPIDEMIOLOGY was reported by Schiemann (1988) and has also been re-
produced by Shu et al. (1995a, b) and Shu et al. (1997),
Y. enterocolitica is found throughout the world and has who demonstrated that small-intestinal infection in
been recorded from pigs in many countries (Bockemuhl piglets led to microabscesses at the base of the villi and
et al. 1979; Cantoni et al. 1979; Barcellos and Castro to reductions in the levels of intestinal lactases (Shu et al.
1981; Doyle et al. 1981; Schiemann and Fleming 1981; 1997) and depressed growth (Shu et al. 1995b).
Hunter et al. 1983). Infection may not be general, since
not all herds are infected (Christensen 1980). It persists CLINICAL SIGNS
on the tonsils of infected pigs for long periods and is
shed in the feces of infected pigs for up to 30 weeks. It Clinical disease has been associated only with Y. enteroco-
has been shown to be transmitted to human food and litica and Y. pseudotuberculosis. Y. pseudotuberculosis sub-
elsewhere on farms by flies (Fukushima et al. 1979). Feed sp. pestis is clearly capable of producing serologic reac-
has been found to be infected, and studies of the dissem- tions (Clark et al. 1983), but no clinical disease has been
ination of infection in pig facilities (Fukushima et al. described.
1983) indicate that infection is transmitted from conta- Y. enterocolitica was isolated in profuse culture from
minated pens, in which infection can persist for 3 weeks. outbreaks of diarrhea in weaned pigs from which no oth-
Other studies suggest that feces can remain infected for er infectious agents could be recovered. Mild fever (to
up to 12 weeks and that, in suitable substrates, the or- 39.4˚C, 103˚F) was present, and the diarrhea contained
ganism may multiply at 20–22˚C. It appears that trans- no blood or mucus and was blackish in color. Clinical
mission from pig to pig is via fecal contamination of ac- signs resembling those described above have been seen
commodation, water, and feed. in animals receiving tylosin or lincomycin. Bloodstained
Y. pseudotuberculosis is less commonly demonstrated mucus may also be found in some diarrheic feces and on
in America than in Europe or Japan and is less common- solid feces passed by penmates. The organism has been
ly identified in pigs than Y. enterocolitica. It is commonly isolated from the rectal mucosa in cases of rectal stric-
found in rodents, which probably represent the main ture. Experimental infections in suckling piglets result in
source of infection for pigs. anorexia, vomiting, diarrhea, and reduction in weight
Y. pseudotuberculosis subsp. pestis may infect wild pigs gain (Shu et al. 1995a).
in California, presumably from infection present in ro- Y. pseudotuberculosis has been associated with clinical
dents (Clark et al. 1983). signs by Morita et al. (1968), who described an outbreak
in Japan. Affected pigs were dull and showed inappe-
PATHOGENESIS tence; bloodstained diarrhea; and edema of eyelids, low-
er face, and dependent parts of the abdomen. Diarrhea
Y. enterocolitica has been shown to infect pigs orally, to was also observed by Barcellos and Castro (1981). Neef
multiply and be found in the feces within 2–3 weeks of and Lysons (1994) were able to reproduce diarrhea in 4
infection, and to disappear from the feces within 30 of 9 pigs infected with a colitis isolate of Y. pseudotuber-
weeks (Fukushima et al. 1984). No clinical signs or le- culosis. The organism has been isolated from pigs with
sions were described and none were found following in- the rectal stricture syndrome.
632 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

LESIONS weeks, with subculture at 3 and 6 weeks onto a selective


medium (Hunter et al. 1983). The selective medium may
The lesions caused by Y. enterocolitica infection have been be MacConkey agar incubated at 30˚C or a specific
described in detail by Erwerth and Natterman (1987) medium for Y. enterocolitica. These methods may be used
and consist of catarrhal enteritis in the small and large for direct isolation. Media are described by Hunter et al.
intestines. Microcolonies of the organism can be seen in (1983) and six are reviewed by Catteau et al. (1983). Re-
the disrupted intestinal epithelium, and in pigs with rec- cent studies by food microbiologists have resulted in a
tal lesions, bacterial penetration and inflammation reach range of tests, such as the immunomagnetic separation
the muscularis mucosae. Shu et al. (1995a) confirmed and polymerase chain reaction technique of Rasmussen
this finding for the small intestine and describe the pres- et al. (1995), which can detect as few as 200 cells per
ence of microabscesses at the bases of the villi. gram of feces.
Lesions of Y. pseudotuberculosis have been described
(Morita et al. 1968). They resembled those of pseudotu- TREATMENT
berculosis in other species, with miliary gray-white spots
on the liver and spleen and swollen gray-white mesen- There is at present no general indication for the treat-
teric lymph nodes. A catarrhal and diphtheritic change ment of yersinia infections, since clinical signs are so
was described in the colon and rectum, as edema and as- rare. In vitro studies suggest that isolates are often sensi-
cites also occurred. Microscopic lesions included necrot- tive to oxytetracycline, furazolidone, neomycin, sulfon-
ic foci containing masses of bacteria surrounded by a amides, and spectinomycin. Tetracyclines have been
thin layer of granulation tissue in the lung, liver, spleen, used in feed to eliminate infection and clinical signs.
mesenteric lymph nodes, and lymphoid follicles of the
large intestine. Similar findings were made by Neef and PREVENTION
Lysons (1994), who noted the penetration of the mi-
croabscesses into the lamina propria. Y. pseudotuberculo- Since spread of Y. enterocolitica from pig to pig appears to
sis was isolated from the liver, spleen, lungs, duodenum, occur from contact with feces, hygiene coupled with
rectum, and mesenteric lymph nodes by Morita et al. housing groups of pigs in separate drainage areas will re-
(1968). Y. pseudotuberculosis can also be isolated from in- duce infection. Control of flies and rodents and disinfec-
flammatory lesions of the rectal mucosa similar to those tion of pens before restocking will reduce transmission.
described above. Morita et al. (1968) found that pseudotuberculosis could
be prevented by excluding birds and rodents. Current re-
DIAGNOSIS quirements for the control of Y. enterocolitica in pig meat
concentrate on the removal of the tonsil at slaughter, so
The clinical signs are not distinctive, but the occurrence control programs have not been developed for the live
of mild fever and blood and mucus on solid feces can in- animal. Control may be required when serologic cross-re-
dicate yersinia infection in the absence of swine dysen- action in the brucellosis test is identified in herds selling
tery. Where rectal stricture is common, the organism breeding stock. The findings of Nielsen et al. (1996) sug-
may be responsible for diarrhea in younger age groups, gest that serologic reactions peak at 33 days postinfec-
and the organism may be involved in the “colitis” syn- tion and have disappeared by 70 days postinfection, so
drome of mild diarrhea in growing pigs. Diagnosis of in- pigs may be treated or managed at the time of infection
fection with most Yersinia spp. in pigs depends upon iso- to prevent the development of antibody or held and
lation of the organism and its identification. Serology retested after antibody levels have declined.
has been used to identify Y. pseudotuberculosis subsp.
pestis (Clark et al. 1983), but most accounts of yersinia
infections suggest that although agglutinating antibody REFERENCES
may result from infection, isolation methods are ade-
quate for diagnosis. The indirect ELISA developed by Barcellos, D. E. S. N. de, and Castro, A. F. P. de. 1981. Isola-
Nielsen et al. (1996) for Y. enterocolitica O3 may be of val- tion of Yersinia pseudotuberculosis from diarrheas in pigs.
ue in the field but may not detect infections by other Br Vet J 137:95–96.
serotypes. Y. pseudotuberculosis can readily be isolated at Bockemuhl, J.; Schmitt, H.; Roth, J.; and Saupe, E. 1979. Die
37˚C on blood and MacConkey agar from lesions of the jahreszeitliche Haufigkeit der Ausscheidung von Yersinia
type described by Morita et al. (1968) and so may Y. en- enterocolitica im Kot gesunder Schlachtschweine. Zen-
terocolitica. Most isolation methods for all yersinias in- tralbl Bakteriol (Orig A) 224:494–505.
volve use of a cold enrichment technique in which tis- Cantoni, C.; D’Aubert, S.; Buogo, A.; and Guizzardi, F. 1979.
sues or samples under investigation are placed in Yersinia enterocolitica nelli feci di suino. Arch Vet Ital
phosphate-buffered saline M/15 at pH 7.6 at 4˚C for 6 30:134–136.
CHAPTER 44 MISCELLANEOUS BACTERIAL INFECTIONS Taylor 633

Catteau, M.; Krembel, C.; and Wauters, G. 1983. Isolement Mosimbale, F., and Gyles, C. L. 1982. The pathogenicity of
de Yersinia enterocolitica de langues de porc. Rec Méd Vét Yersinia enterocolitica strains isolated from various
159:89–94. sources for four test systems. Can J Comp Med
Christensen, S. G. 1980. Yersinia enterocolitica in Danish 46:70–75.
pigs. J Appl Bacteriol 48:377–382. Neef, N. A., and Lysons, R. J. 1994. Pathogenicity of a strain
Clark, R. K.; Jessup, D. A.; Hird, D. W.; Rupanner, R.; and of Yersinia pseudotuberculosis isolated from a pig with
Meyer, M. E. 1983. Serologic survey of California wild porcine colitis syndrome. Vet Rec 135:58–63.
hogs for antibody against selected zoonotic disease Nielsen, B.; Heisel, C.; and Wingstrand, A. 1996. Time
agents. J Am Vet Med Assoc 183:1248–1259. course of the serological response to Yersinia enterocolit-
Doyle, M. P.; Hugdahl, M. B.; and Taylor, S. L. 1981. Isola- ica O:3 in experimentally infected pigs. Vet Microbiol
tion of Yersinia enterocolitica from porcine tongues. Appl 48:293–303.
Environ Microbiol 41:661–666. Rasmussen, H. N.; Rasmussen, O. F.; Christensen, H.; and
Erwerth, W., and Natterman, H. 1987. Histopathologische Olsen, J. E. 1995. Detection of Yersinia enterocolitica O:3
Untersuchengen bei der experimentellen oralen Yersinia in fecal samples and tonsil swabs from pigs using im-
enterocolitica Infektion des Jung-schweines. Monatsh Vet munomagnetic separation and polymerase chain reac-
42:319–324. tion. J Appl Bacteriol 78:563–568.
Fukushima, H.; Ito, Y.; Saito, K.; Tsubokura, M.; and Otsuki, Schiemann, D. A. 1988. The pathogenicity of Yersinia ente-
K. 1979. Role of the fly in the transport of Yersinia ente- rocolitica for piglets. Can J Vet Res 52:325–330.
rocolitica. Appl Environ Microbiol 38:1009–1010. Schiemann, D. A., and Fleming, C. A. 1981. Yersinia entero-
Fukushima, H.; Nakamura, R.; Ito, Y.; Saito, K.; Tsubokura, colitica isolated from throats of swine in eastern and
M.; and Otsuki, K. 1983. Ecological studies of Y. entero- western Canada. Can J Microbiol 27:1326–1333.
colitica in pigs. I. Dissemination of Y. enterocolitica in Shu, D.; Simpson, H. V.; Xu, R. J.; Mellor, D. J.; Reynolds, G.
pigs. Vet Microbiol 8:469–483. W.; Alley, M. R.; Fenwick, S. G.; and Marshall, R. B.
Fukushima, H.; Nakamura, R.; Ito, Y.; and Saito, K. 1984. 1995a. Impact of Yersinia enterocolitica enteritis on
Ecological studies of Yersinia enterocolitica. II. Experi- disaccharidase activity and small intestinal morphology
mental infection with Y. enterocolitica in pigs. Vet Micro- in colostrum-deprived newborn piglets. New Zealand
biol 9:375–389. Vet J 45:27–36.
Hunter, D.; Hughes, S.; and Fox, E. 1983. Isolation of Shu, D.; Simpson, H. V.; Xu, R. J.; Mellor, D. J.; Reynolds, G.
Yersinia enterocolitica from pigs in the United Kingdom. W.; and Marshall, R. B. 1995b. Effects of Yersinia entero-
Vet Rec 112:322–323. colitica infection on growth of the body and internal or-
Kwaga, J., and Iversen, J. O. 1993. Plasmids and outer mem- gans in newborn colostrum-deprived piglets. Biology of
brane proteins of Yersinia enterocolitica and related the Neonate 67:360–369.
species of swine origin. Vet Microbiol 36:205–214. Shu, D.; Simpson, H. V.; Xu, R. J.; Mellor, D. J.; Reynolds, G.
Morita, M.; Nakamatsu, M.; and Goto, M. 1968. Pathologi- W.; and Marshall, R. B. 1997. Experimental infection of
cal studies on pseudotuberculosis rodentium. III. Spon- newborn piglets with Yersinia enterocolitica: An animal
taneous swine cases. Jpn J Vet Sci 30:233–239. model of enteritis. New Zealand Vet J 43:50–56.

STAPHYLOCOCCI
Staphylococci are ubiquitous. They are present on every ETIOLOGY
pig farm and involved in a wide range of lesions in pigs
of all ages. The most easily recognized are those of ex- Staphylococci are gram-positive, 0.5–1.5 µm in diameter,
udative epidermitis caused by Staphylococcus hyicus and forming grapelike clusters when grown in serum broth
described in Chapter 33. Few, if any, of the other lesions or seen in pus. They grow primarily in aerobic conditions
can be unequivocally identified as being staphylococcal but can also grow anaerobically, are oxidase-negative but
on clinical grounds; in the majority, staphylococcal in- produce catalase, and metabolize a wide variety of sug-
volvement must be confirmed by laboratory means. In ars. They produce a wide range of enzymes and some
addition to their association with pig disease, some of toxins. The species are distinguished by the presence of
the staphylococci that infect the pig, notably S. aureus, enzymes such as coagulase, DNAase, hemolysins, and
may be involved in human food poisoning if carcasses phosphatase and by the ability to utilize a variety of sug-
are contaminated or abscesses are present. ars. Major species reported from the pig are S. aureus, S.
634 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

hyicus, and S. epidermidis, although S. saprophyticus has tion of multiple abscesses. These may occur in bones to
also been described. give osteomyelitis; in joints; on the heart valves to give
S. aureus is the only species apart from S. hyicus to be vegetative endocarditis; and in the liver, kidney, or
consistently isolated from lesions in pigs. It forms yel- lymph nodes. Vegetative endocarditis may give rise to
lowish-white, opaque, circular, domed colonies, 1–2 mm septic emboli that cause abscess formation and infarc-
in diameter, on blood agar after 24 hours of incubation. tion in the kidney. Most of these systemic infections oc-
These colonies may be surrounded by a zone of complete cur in neonates or piglets and take 7–10 days to develop.
hemolysis, caused by alpha hemolysins, on horse blood They are also present in apparently normal pigs at
agar. On sheep blood agar, the ring of complete hemoly- slaughter. Abscesses contain neutrophils and micro-
sis is surrounded by a wider area of incomplete hemoly- colonies of bacteria in all stages of multiplication and
sis caused by beta hemolysins; this becomes complete on heal by fibrosis.
cooling of the plate. In addition to these hemolysins, the Mastitis, vaginitis, and metritis appear to result from
organism produces coagulase, DNAase, proteinases, ascending infection. Abortion has been associated with
hyaluronidase, and toxins that include the alpha toxin the demonstration of serum antibody to alpha he-
and the enterotoxins. Both have been demonstrated in molysin in the sow (Kohler and Wille 1980), but the im-
strains of porcine origin (Engvall and Schwan 1983); portance of this and other toxins in abortion is not
protein A (Takeuchi et al. 1995) and polysaccharide cap- known.
sules are also present. Isolates of S. aureus can be identi- In enteric infection, experimental studies have shown
fied by phage typing and plasmid profiling, and those of that staphylococcal enterotoxin A will cause emesis with-
public health significance may readily be traced. S. aureus in 90–180 minutes when given orally in doses of 40–50
is fairly resistant to drying but is readily inactivated by µg (Taylor et al. 1982). Larger doses can cause behavioral
heat. It is sensitive to a wide range of disinfectants, such changes, including inappetence, restlessness, and stag-
as phenols, hypochlorites, iodine, and iodophors. gering. The piglet is clearly susceptible, but the ability of
enteric staphylococcal infections to produce enterotoxin
EPIDEMIOLOGY in vivo is not known.

S. aureus is widely distributed in the environment and CLINICAL SIGNS


has been recovered from pig feces, food, water following
contamination of drinkers, pen floors and walls, and the The presence of S. aureus infection cannot readily be sus-
air in pig facilities. The organism can be isolated from a pected on clinical grounds. S. aureus has been isolated
wide range of hosts, including birds, rodents, dogs, cats, from a wide variety of syndromes. Most of these occur in
and humans. The extent to which isolates from lesions in individual animals, and disease rarely spreads from ani-
pigs are of nonporcine origin is not yet known. Porcine mal to animal. It can cause neonatal septicemia and is of-
strains capable of enterotoxin production have been ten identified in small, hairy, stunted piglets of 7–10
identified on carcasses and represent a source of infec- days of age with umbilical abscesses, polyarthritis, and
tion or possible food poisoning to humans (Engvall and signs of cardiac enlargement due to vegetative endo-
Schwan 1983). carditis. It is a cause of subcutaneous abscesses associat-
The pig is probably the major source of infection for ed with abrasions and foot lesions, especially in piglets.
other pigs; S. aureus can be isolated from the skin, oral These often result in arthritis of the distal phalangeal
cavity, upper respiratory tract, prepuce, vagina, and gut joints, enlargement of the hoof, and sinus formation at
of healthy pigs on a very wide scale. Transmission of the the coronary band. Creamy pus is often seen to exude
organism may be by aerosol to the upper respiratory from the abscesses.
tract, directly by skin contact, or indirectly by contact S. aureus has been isolated from enteritis in piglets
with contaminated walls or fittings. Ingestion of S. au- and in older animals and from the rectal mucosa of ani-
reus from food, contaminated water, or litter is common. mals with lesions of rectal stricture. No particular fea-
Venereal contact may be responsible for some genital in- tures of diarrhea are associated with staphylococci ex-
fections; local invasion of the mammary gland, navel, cept that the enteritis is often chronic and antibiotic
and skin lesions is common. treatment may have been given. The organism is also
present in a small percentage of cases of mastitis and has
PATHOGENESIS been isolated from pigs with metritis and agalactia. The
only reason to suspect staphylococcal involvement may
S. aureus appears to multiply on damaged mucosal sur- be the presence of a creamy white or bloodstained pus,
faces or skin and can invade to cause bacteremia. In some but this often contains other organisms. It has also been
cases, such as neonatal septicemia, animals may become isolated from aborted fetuses and placentas (Kohler and
fevered and die, but bacteremia usually leads to forma- Wille 1980).
CHAPTER 44 MISCELLANEOUS BACTERIAL INFECTIONS Taylor 635

LESIONS be confirmed before S. aureus is considered to be the sole


cause; exclusion of other agents is even more important
No gross lesions may be seen in piglet septicemia. In in diseases at mucous surfaces.
chronic infections, an inflamed mucosa may be associat-
ed with staphylococcal infection, but there is no specific TREATMENT
feature that allows their identification with staphylococ-
cal infection. Abscesses may occur in the umbilicus, liver, Individual abscesses may be opened surgically after skin
lungs, lymph nodes, spleen, kidneys, joints, and bones in cleaning and disinfection, but most treatments rely on
osteomyelitis; bone abscesses may give rise to pathologic antimicrobial treatment. Since S. aureus infection is an
fractures, especially when in the vertebrae. Body cavities individual-animal problem, there is usually no need to
(peritoneal cavity, pericardial cavity, uterine lumen) may treat the whole group. Parenteral treatment with an ap-
contain pus, especially in young animals, following um- propriate formulation and prompt treatment at any age
bilical infection. S. aureus is only one cause of such le- can prevent the development of large and potentially ex-
sions. Both gross and microscopic lesions of mastitis tensive and fatal abscesses. The use of feed medication as
may be found in the mammary glands, and in some cas- a prophylactic cannot be justified unless a severe prob-
es fibrosis may be considerable. Occasionally, a granulo- lem has been identified, since the development of an-
matous mass with fibrosis may be found in the abdomi- timicrobial resistance in staphylococci is likely to favor
nal cavity of piglets that have died after castration. In all them over other organisms after a brief period during
cases, confirmation that the lesions are staphylococcal which they are suppressed. The use of bacterins has been
depends on demonstration of the organisms. described, but they are not widely available or extensive-
ly used.
DIAGNOSIS
REFERENCES
The clinical signs resulting from multiple abscess forma-
tion in individual pigs may be suggestive, and similar Engvall, A., and Schwan, O. 1983. Isolation and partial char-
suspicions may arise from the postmortem findings. acterization of bacteria recovered from abscesses of nor-
Confirmation of involvement of staphylococci rather mally slaughtered pigs. Acta Vet Scand 24:74–83.
than Arcanobacterium pyogenes or streptococci in abscess- Kohler, B., and Wille, H. 1980. Bakteriologische Unter-
es and arthritis is obtained from Gram-stained smears of suchungen bei abortierten Schweinfeten unter Beruck-
the pus in which gram-positive cocci may be seen singly sichtigung der ätiologischen Bedeutung von Staphylococ-
or in clusters. Only the isolation of staphylococci in cul- cus aureus. Monatsch Veterinärmed 35:506–510.
ture confirms that they are involved. S. aureus can be iso- Takeuchi, S.; Matuda, K.; and Sasano, K. 1995. Protein A in
lated readily on blood and MacConkey agar, and its iden- Staphylococcus aureus isolates from pigs. J Vet Med Sci
tity can be confirmed by coagulase and DNAase tests and 57:581–582.
by its ability to ferment mannitol. Isolates may be phage Taylor, S. L.; Schlunz, L. R.; Beerey, J. T.; Cliver, D. O.; and
typed if this is considered relevant, and any toxins pro- Bergdoll, M. S. 1982. Emetic action of staphylococcal
duced may be identified. enterotoxin A on weanling pigs. Infect Immunol
In most abscesses the absence of other bacteria must 36:1263–1266.

ACTINOBACULUM (ACTINOMYCES–EUBACTERIUM–
CORYNEBACTERIUM) SUIS
Soltys and Spratling (1957) isolated, anaerobically, a in sows has been reported from Canada (Percy et al.
diphtheroid bacterium from the urine and diseased tis- 1966), Norway (Aalvik 1968), Holland (Dijkstra 1969;
sue of adult pigs in the United Kingdom affected with Frijlink et al. 1969; Narucka and Westendorp 1972),
cystitis and pyelonephritis and named it Corynebacterium Denmark (Larsen 1970, 1973), Hong Kong (Munro and
suis. Soltys (1961) described the characteristics of C. suis Wong 1972), Australia (Glazebrook et al. 1973), Switzer-
in more detail. C. suis was subsequently assigned to the land (Schallibaum et al. 1976), Finland (Kauko et al.
genus Eubacterium (Wegienek and Reddy 1982), Actino- 1977), Malaysia (Too et al. 1985), Germany (Muller et al.
myces (Ludwig et al. 1992), and finally Actinobaculum 1986; Waldmann 1987), Brazil (De Oliveira et al. 1988),
Lawson et al. 1997). and the United States (Walker and MacLachlan 1989).
A. suis infection associated with urinary tract disease Disease caused by A. suis occurs in small outbreaks or in-
636 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

dividual sows, but carriage is much more widespread. weeks of mating, suggesting that predisposing factors
The main features of the organism and the disease are are operating at this time. Wendt et al. (1994) suggest
described below. that these may include infection with other organisms,
because of a requirement for erosion of mature vesical
ETIOLOGY epithelial cells to allow attachment by A. suis. Water re-
striction and the presence of crystalluria may also pre-
A. suis is a gram-positive pleomorphic rod, 2–3 µm long dispose to infection (Wendt and Sobestiansky 1995).
and 0.3–0.5 µm wide; the organism tends to be larger in The disease may become clinically evident at any time in
tissues than in cultures. In tissues and cultures it occurs the breeding cycle of the sow (e.g., after parturition), and
in the form of so-called Chinese letters and in a palisade in such cases it is not always clear whether infection of
fashion. It is nonmotile and does not form spores. the urinary tract is recent or whether there has been a re-
A. suis grows well on blood agar under anaerobic con- crudescence of previously existing disease.
ditions. Colonies are evident at 2 days, having a diameter Studies on the adhesive properties of A. suis have
of 2–3 mm; they then begin to flatten and develop a been reported by Larsen et al. (1986). They have demon-
characteristic dry, gray, opaque surface with a crenated strated that some strains are heavily fimbriated and are
edge; some colonies attain a size of 4–5 mm in 5–6 days. able to adhere to the epithelial cells of the porcine blad-
There is no hemolysis. Growth on nutrient agar, even af- der; their findings support the hypothesis that glycocon-
ter subculture, is poor. In liquid media such as cooked jugates are specific receptor sites for the attachment of A.
meat broth and brain-heart infusion, slight turbidity is suis. Infection of the ureters and kidneys follows infec-
produced in 2–4 days; growth is more luxuriant in tryp- tion of the bladder.
ticase soy broth and is further enhanced by the addition
of urea to a final concentration of 1.2% (w/v). The addi- CLINICAL SIGNS AND LESIONS
tion of maltose to either solid or liquid media improves
growth. Although A. suis has always been described as an Hematuria is the main sign in the acute phase. As the dis-
anaerobic organism, prolonged aerobic incubation on ease progresses, there is loss of weight. Some sows may
blood agar results in the development of colonies within die suddenly, apparently from acute renal failure.
5–10 days; on subculture, aerobically, colonies are evi- Inflammatory reactions in the mucosa of the urethra,
dent in 1–3 days. bladder, and ureters may be catarrhal, fibrinopurulent,
The organism is relatively inactive when subjected to hemorrhagic, or necrotic. Affected kidneys often have ir-
conventional biochemical tests. Most strains ferment regular yellow areas of degeneration in the parenchyma
maltose and xylose and hydrolyze starch but do not at- that are visible on the surface. The renal pelvis may be di-
tack other commonly used “sugars”; all produce urease. lated and contain mucoid fluid in which flakes of necrot-
Catalase, methyl red, Voges-Proskauer, indole, and ni- ic debris and altered blood are present. The medullary
trate-reduction tests are negative. Coagulated serum and pyramids often show yellow or dark green to black foci of
egg medium are not liquefied. A slight alkalinity is pro- necrosis. The ureters are often dilated and filled with red-
duced in litmus milk. dish purulent urine. There are no related lesions else-
where in the body.
EPIDEMIOLOGY AND PATHOGENESIS
DIAGNOSIS
Most male pigs aged 6 months or more harbor A. suis in
the preputial diverticulum, which may become colonized Diagnosis is based on clinical signs and bacteriological
when pigs are only a few weeks old. Uninfected males are examination of urine. A. suis is easily seen in Gram-
readily infected when they are housed with carrier males stained films, often with other bacteria, notably strepto-
(Jones and Dagnall 1984). The organism may be found cocci. For cultural examination it is essential to incubate
on the floors of pens occupied by male pigs. Carr and the medium (e.g., blood agar), which has been inoculat-
Walton (1990) have isolated A. suis from the footwear of ed with urine or other appropriate material, anaerobical-
handlers working with boars but not from those working ly for 4 days. Results of cultural procedures should not
in the farrowing area. Only rarely is it found in the vagi- be reported as negative before that time. Rapid diagnosis
na of healthy females, but it may be that existing cultur- can be achieved by the use of immunofluorescent tech-
al techniques are insufficiently sensitive to demonstrate niques (Schallibaum et al. 1976; Kauko et al. 1977). A se-
its presence there. There are no reports of A. suis being lective medium for the isolation of A. suis has been de-
isolated from any sites in the pig other than the urogeni- scribed by Dagnall and Jones (1982). Wendt and
tal tract. Amstberg (1995) evaluated the possibility of using serol-
Cystitis and pyelonephritis caused by A. suis mainly ogy to detect infection and tested indirect immunofluo-
affect adult females. Infection of the bladder and kidneys rescence in diagnosis. They found that antibody did not
is by the ascending route. Most cases occur within 1–3 appear until 3 weeks after hemorrhagic cystitis had oc-
CHAPTER 44 MISCELLANEOUS BACTERIAL INFECTIONS Taylor 637

curred and that when a titer of ++ at 1/16 was used as an with Corynebacterium suis. Aust Vet J 49:546.
endpoint to give 100% specificity, the sensitivity was on- Jones, J. E. T., and Dagnall, G. J. R. 1984. The carriage of
ly 79%. There was no relationship between antibody lev- Corynebacterium suis in male pigs. J Hyg (Camb)
el and the degree of renal damage. 93:381–388.
Kauko, L.; Schildt, R.; and Sandholm, M. 1977.
TREATMENT AND PREVENTION Corynebacterium suis emakoiden pyelonefritin aiheut-
tajana suomessa. Suom Elainl 83:489–492.
A. suis is sensitive in vitro to several antibiotics, including Larsen, J. L. 1970. Corynebacterium suis infektioner hos
penicillin and tetracyclines. Administration of antibi- svin. Nord Vet Med 22:422–431.
otics is frequently effective, at least in the short term. ———. 1973. Et enzootisk udbrud af cystitis og
However, relapses commonly occur, and often it is best to pyelonephritis forarsaget af Corynebacterium suis.
advise early slaughter of affected animals. Prolonged Medlemsbl Dan Dyrlaegeforen 56:509–515.
treatment for 20 days with ampicillin given at 20 mg/kg Larsen, J. L.; Hogh, P.; and Hovind-Hougen, K. 1986.
may be used (Wendt and Sobestiansky 1995), and en- Haemagglutinating and hydrophobic properties of
rofloxacin given for 10 days at 10 mg/kg may also be ef- Corynebacterium (Eubacterium) suis. Acta Vet Scand
fective. In Wendt and Sobestiansky’s studies (1995), pigs 27:520–530.
with lesions confined to the bladder recovered with an- Lawson, P. A.; Falsen, E.; Akervall, E.; Vandamme, P.;
timicrobial treatment alone, but those with renal dam- and Collins, M. D. 1997. Characterization of some
age required infusion therapy for recovery. Actinomyces-like isolates from human clinical speci-
There are no proven methods of prevention. A. suis mens: reclassification of Actinomyces suis (Soltys and
may be transmitted from boars to sows at the time of Spratling) as Actinomyces suis comb nov and descrip-
mating. Culling of carrier boars has been suggested as a tion of Actinobaculum schaalii. Int J Syst Bacteriol
method of preventing infection of sows; this does not 47:899–903.
seem worthwhile, because replacement boars will almost Ludwig, W.; Kirchof, G.; Weizenegger, M.; and Weiss, N.
certainly be infected. Culling might be of value if there 1992. Phylogenetic evidence for the transfer of
are “pathogenic” and “nonpathogenic” strains of A. suis, Eubacterium suis to the genus Actinomyces as Actino-
but there is no evidence that such different strains exist. myces suis comb nov. Int J Syst Bacteriol 42:161–165.
Currently, the only means of attempting prevention of Muller, E.; Pozvari, M.; and Merkt, M. 1986. Zum
the disease is to administer antibiotics to sows immedi- Vorkommen von blutig-eitrigen Harnblasen und
ately after service or, if outbreaks of the disease are eco- Nierenentzundungen bei Zuchtsauen in Verbindung
nomically serious, to use artificial insemination. mit Corynebacterium suis. Prakt Tierärztl 67:1081–
1083.
REFERENCES Munro, R., and Wong, F. 1972. First isolation of
Corynebacterium suis in Hong Kong. Br Vet J
Aalvik, B. 1968. Corynebacterium suis isolert fra et tilfelle 128:29–32.
av pyelonefritt hos purke. Nord Vet Med 20:319–320. Narucka, U., and Westendorp, J. F. 1972. Corynebacteri-
Carr, J., and Walton, J. R. 1990. Investigation of the path- um suis in pigs. Neth J Vet Sci 4:86–92.
ogenic properties of Eubacterium (Corynebacterium) Percy, D. H.; Ruhnke, H. L.; and Soltys, M. A. 1966.
suis. Proc Int Congr Pig Vet Soc 11. A case of infectious cystitis and pyelonephritis of
Dagnall, G. J. R., and Jones, J. E. T. 1982. A selective swine caused by Corynebacterium suis. Can Vet J
medium for the isolation of Corynebacterium suis. Res 7:291–292.
Vet Sci 32:389–390. Schallibaum, M. von; Hani, H.; and Nicolet, J. 1976. In-
De Oliveira, S. J.; Barcellos, D. E. S. N.; and Borowski, S. fektion des Harntraktes beim Schwein mit Corynebac-
M. 1988. Urinary tract infections in two pig breeding terium suis: Diagnosis met Immunofluoreszenz.
herds, with emphasis on the presence of Corynebac- Schweiz Arch Tierheilkd 118:329–334.
terium suis. Proc Int Congr Pig Vet Soc 10. Soltys, M. A. 1961. Corynebacterium suis associated with
Dijkstra, R. G. 1969. Cysto-pyelonefritis bij varkens ver- specific cystitis and pyelonephritis in pigs. J Pathol
zoorzaakt door Corynebacterium suis. Tijdschr Dierge- 81:441–446.
neeskd 94:393–394. Soltys, M. A., and Spratling, F. R. 1957. Infectious cysti-
Frijlink, G. P. A.; Van Dijk, J. E.; and Goudswaard, J. tis and pyelonephritis of pigs: A preliminary commu-
1969. Een hemorragische-necrotiserende cystopy- nication. Vet Rec 69:500–504.
elonefritis bij een drachtige zeug, veroorzaakt door Too, H. L.; Chooi, K. F.; and Bahaman, A. R. 1985. Cysti-
Corynebacterium suis. Tijdschr Diergeneeskd tis in a sow due to Corynebacterium suis. Kajian Veteri-
94:389–393. nar 17:155–156.
Glazebrook, J. S.; Donaldson-Wood, C.; and Ladds, P. W. Waldmann, K. H. 1987. Die Pyelozystitis der Zuchtsau.
1973. Pyelonephritis and cystitis in sows associated Tierärztl Prax 15:263–267.
638 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Walker, R. L., and MacLachlan, J. J. 1989. Isolation of Eu- 137:129–136.


bacterium suis from sows with cystitis. J Am Vet Med Wendt, M., and Sobestiansky, J. 1995. Untersuchungen
Assoc 195:1104–1107. zur Therapie von Harnwegs-infektionen bei Sauen.
Wegienek, J., and Reddy, C. A. 1982. Taxonomic study of Dtsch Tierärztl Wochenschr 102:21–22.
Eubacterium suis (nom. rev.) Comb. nov. Int J Syst Wendt, M.; Liebhold, M.; and Drommer, W. 1994.
Bacteriol 32:218–228. Rasterelektronenmikroskopische Untersuchungen
Wendt, M., and Amstberg, G. 1995. Untersuchungen an der Harnblase von Sauen unter besonderer
zum Nachweis von Antikorpen gegen Eubacterium su- Berucksichtigung einer Eubacterium suis-Infektion. J
is beim Schwein. Schweiz Arch für Tierheilkd Vet Med Ser B 41:126–138.

RHODOCOCCUS EQUI
Rhodococcus equi is associated with granulomatous lym- idase is the major constituent (Linder and Bernheimer
phadenitis affecting the lymph nodes of the head and 1982).
neck of the pig. The lesions can be confused at slaughter R. equi possess an abundant acidic polysaccharide
with those of tuberculosis, and the organism is impor- capsule, which is the basis for several serotyping
tant for this reason rather than as a cause of clinical dis- schemes. Prescott (1981) has identified 7 serotypes, of
ease. which serotype 1 is the most frequently isolated in Cana-
da, Australia, and India. Japanese workers have identi-
ETIOLOGY fied 27 serotypes, with the most common being equiva-
lent to Prescott serotype 1 (Nakazawa et al. 1983). There
The organism was first isolated from the pneumonic is no relationship between capsular serotype and origin
lungs of a foal by Magnusson (1923) and was given the of the isolates. The capsular polysaccharides of 4
name Corynebacterium equi; it is now called Rhodococcus Prescott serotypes have been purified.
equi. This gram-positive coccobacillus is a member of the Recent studies of this species have confirmed that
nocardioform actinomycete group (Goodfellow et al. isolates from pigs possess a virulence-associated protein
1982). In common with other members of Rhodococcus, (vapA) that is encoded by a virulence plasmid. Takai et
R. equi produces pinkish colonies on solid media. The al. (1996) confirmed the presence of 79–95 kb plasmids
mycolic acids of R. equi have a chain length of 34–48 C, coding for a 20 kDa virulence protein.
and the DNA base composition is 66–72 mol% guanine
plus cytosine. Chemical properties used in defining the EPIDEMIOLOGY AND PATHOGENESIS
species were summarized by Goodfellow (1987).
Isolation of R. equi from clinical samples is easily R. equi is primarily a soil resident. Environmental distri-
achieved by aerobic culture on routine media at 37˚C, al- bution favors soils enriched with herbivore manure,
though the optimum temperature is 28–30˚C. Selective since fecal matter potentiates bacterial multiplication
media, such as that developed by Woolcock et al. (1979), (Barton and Hughes 1984). R. equi is also a transient in
are required for fecal isolation. Colonies are slow grow- the intestinal tract of many species, including pigs, cat-
ing, requiring 48 hours to reach a size of 2–4 mm. The tle, deer, horses, sheep, goats, and wild birds (Woolcock
typical colony is irregularly round, buff-pink, smooth, et al. 1979; Carman and Hodges 1987). Being an obligate
and mucoid, although colonial variation is common aerobe, R. equi is not likely to be a member of the normal
within and between strains (Mutimer and Woolcock flora. The bacterium is found in soil samples collected
1981). R. equi is biochemically unreactive. It is not prote- from arable land that has not pastured animals for many
olytic and does not ferment carbohydrates. R. equi is years, emphasizing its durability. R. equi is present in
catalase positive, usually urease positive, and oxidase dust and even in cobwebs of farm buildings in areas
negative. The API ZYM system has been found helpful in where it occurs. R. equi is relatively resistant to chemical
bacterial identification (Mutimer and Woolcock 1982). disinfectants, such as treatment with 2.5% oxalic acid
R. equi is not hemolytic but, in conjunction with the and 0.5% sodium hydroxide over periods of 15–60 min-
phospholipase D of Corynebacterium pseudotuberculosis utes (Karlson et al. 1940).
or the beta toxin of Staphylococcus aureus, produces com- Little is known of the epidemiology or pathogenesis
plete hemolysis of sheep and cattle erythrocytes of the naturally occurring disease in swine. As in horses,
(Prescott et al. 1982). Semipurification of this factor, R. equi infection is likely to be acquired from the envi-
known as “equi factor,” has indicated that cholesterol ox- ronment (Woolcock et al. 1980). Ingestion is the normal
CHAPTER 44 MISCELLANEOUS BACTERIAL INFECTIONS Taylor 639

mode of exposure in foals (Takai et al. 1986a), leading to DIAGNOSIS


the development of solid protective immunity in the ma-
jority of animals (Prescott et al. 1980; Chirino-Trejo et al. Diagnosis is at postmortem. Microbiological identifica-
1987). A similar situation probably occurs in swine tion of R. equi is necessary for it is not possible to differ-
housed on pasture or in yards contaminated with R. equi, entiate the gross lesions of R. equi–induced lymphadeni-
as the bacterium is readily isolated from the feces of such tis from those caused by Mycobacterium spp.
pigs (Barton and Hughes 1984). Several slaughterhouse
studies have also demonstrated recovery of R. equi from TREATMENT AND PREVENTION
normal cervical and submaxillary lymph nodes at rates
varying from 7% to 35% (Mutimer and Woolcock 1980; R. equi–induced disease is not sufficiently important to
Takai et al. 1986b), although the organism may now be necessitate antemortem diagnosis and treatment in
less common, as Takai et al. (1996) recovered it from on- swine. Therapy, which in foals requires long-term ad-
ly 3.1% of 1832 swine in Japan. However, no epidemio- ministration of rifampicin and erythromycin, is not fea-
logical studies have been carried out to correlate infec- sible on economic grounds. While some economic loss
tion rates, disease prevalence, and environmental may accrue from condemnation at slaughter, there are no
contamination of R. equi on pig farms, as has been done studies that indicate the extent of this loss and there ap-
for horses. There have been suggestions that isolates pears to be no incentive to institute control measures.
from pigs differ somewhat from those from horses and These would in any case be difficult; an effective vaccine
that some pig isolates resemble those from humans. is presently unavailable.
Takai et al. (1996) speculated that some human cases
may be of porcine origin. REFERENCES
The way in which R. equi causes granulomatous lym-
phadenitis of the head and neck in the pig is not clear. R. Barton, M. D., and Hughes, K. L. 1984. Ecology of Rhodococ-
equi can be recovered from normal nodes, and there are cus equi. Vet Microbiol 9:65–76.
accounts of failure to reproduce the nodal lesions exper- Carman, M. G., and Hodges, R. T. 1987. Distribution of
imentally (Karlson et al. 1940; Cotchin 1943). Mycobac- Rhodococcus equi in animals, birds and from the environ-
terium spp. may also be recovered in some cases. It is pos- ment. NZ Vet 35:114–115.
sible that the severity of the lesions reflects the Chirino-Trejo, J. M.; Prescott, J. F.; and Yager, J. A. 1987. Pro-
possession of the virulence-associated protein, which tection of foals against experimental Rhodococcus equi
may vary from strain to strain, the degree of immunity pneumonia by oral immunization. Can J Vet Res
present when infection took place, or the duration of the 51:444–447.
infection at sampling. However, R. equi typically pro- Cotchin, E. 1943. Corynebacterium equi in the submaxillary
duces a granulomatous tissue reaction in lymph nodes of lymph nodes of swine. J Comp Pathol 53:298–309.
other species, consistent with its action as a facultative Goodfellow, M. 1987. The taxonomic status of Rhodococcus
intracellular pathogen (Yager 1987). equi. Vet Microbiol 14:205–209.
R. equi has been associated with serious clinical dis- Goodfellow, M.; Beckham, A. R.; and Barton, M. D. 1982.
ease, including one outbreak of oral abscesses and one of Numerical classification of Rhodococcus equi and related
pneumonia (Thal and Rutqvist 1959; Rao et al. 1982), actinomycetes. J Appl Bacteriol 53:199–207.
but pigs are extremely resistant to experimental infec- Karlson, A. G.; Moses, H. R.; and Feldman, W. H. 1940.
tion. Following aerosolization, R. equi is cleared from the Corynebacterium equi (Magnusson, 1923) in the submax-
lungs very slowly, but clinical signs and pathological le- illary lymph nodes of swine. J Infect Dis 67:243–
sions of pneumonia are minimal despite exposure to 107 251.
organisms on 7 consecutive days (Zink and Yager 1987). Linder, R., and Bernheimer, A. W. 1982. Enzymatic oxida-
Pneumonia has, however, been induced by intratracheal tion of membrane cholesterol oxidase in relation to lysis
inoculation of fluid inocula (Thal and Rutqvist 1959). of sheep erythrocytes by corynebacterial enzymes. Arch
Biochem Biophys 213:395–404.
LESIONS Magnusson, H. 1923. Spezifische Infektiose Pneumonie
beim Fohlen: Ein neurer Eitererreger beim Pferde. Arch
Lymphadenitis causes no significant clinical signs; le- Wiss Prakt Tierheilk 50:22–38.
sions are detected only at slaughter. Affected sub- Mutimer, M. D., and Woolcock, J. B. 1980. Corynebacterium
mandibular and cervical nodes are enlarged, containing equi in cattle and pigs. Vet Q 2:25–27.
multiple yellow-tan foci, often in a subcapsular location. ———. 1981. Some problems associated with the identifi-
Caseation and calcification of these foci sometimes oc- cation of Corynebacterium equi. Vet Microbiol
cur. Histologically, the lesion is a granulomatous lym- 6:331–338.
phadenitis. Similar lesions reported in the mesenteric ———. 1982. API ZYM for identification of Corynebacteri-
lymph nodes have yielded Rhodococcus sputi on culture um equi. Zentralbl Bakteriol Hyg Abt (Orig C3):410–
(Tsukamura et al. 1988). 415.
640 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

Nakazawa, M.; Kubo, M.; Sugimoto, C.; and Isayama, Y. Takai, S.; Fukunaga, N.; Ochiai, S.; Imal, Y.; Sasaki, Y.; Tsub-
1983. Serogrouping of Rhodococcus equi. Microbiol Im- aki, S.; and Sekizaki, T. 1996. Identification of interme-
munol 27:837–846. diately virulent Rhodococcus equi isolates from pigs. J
Prescott, J. F. 1981. Capsular serotypes of Corynebacterium Clin Microbiol 34:1034–1037.
equi. Can J Comp Med 45:130–134. Thal, E., and Rutqvist, L. 1959. The pathogenicity of
Prescott, J. F.; Ogilvie, T. J. H.; and Markham, R. J. F. 1980. Corynebacterium equi for pigs and small laboratory ani-
Lymphocyte immunostimulation in the diagnosis of mals. Nord Vet Med 11:298–304.
Corynebacterium equi pneumonia of foals. Am J Vet Res Tsukamura, M.; Komatsuzaki, C.; Sakai, R.; Kane-da, K.; Ku-
41:2073–2075. do, T.; and Seino, A. 1988. Mesenteric lymphadenitis of
Prescott, J. F.; Lastra, M.; and Barksdale, L. 1982. Equi fac- swine caused by Rhodococcus sputi. J Clin Microbiol
tors in the identification of Corynebacterium equi Mag- 26:155–157.
nusson. J Clin Microbiol 16:988–990. Woolcock, J. B.; Farmer, A. M. T.; and Mutimer, M. D. 1979.
Rao, M. S.; Zaki, S.; Keshavamurthy, B. S.; and Singh, K. C. Selective medium for Corynebacterium equi isolation. J
1982. An outbreak of an acute Corynebacterium equi in- Clin Microbiol 9:640–642.
fection in piglets. Indian Vet J 59:487–488. Woolcock, J. B.; Mutimer, M. D.; and Farmer, A. M. T. 1980.
Takai, S.; Ohkura, H.; Watanabe, Y.; and Tsubaki, S. 1986a. Epidemiology of Corynebacterium equi in horses. Res Vet
Quantitative aspects of fecal Rhodococcus (Corynebacteri- Sci 28:87–90.
um) equi in foals. J Clin Microbiol 23:794–796. Yager, J. A. 1987. The pathogenesis of Rhodococcus equi
Takai, S.; Takeuchi, T.; and Tsubaki, S. 1986b. Isolation of pneumonia in foals. Vet Microbiol 14:225–232.
Rhodococcus (Corynebacterium) equi and atypical my- Zink, M. C., and Yager, J. A. 1987. Experimental infection of
cobacteria from the lymph nodes of healthy pigs. Jpn J piglets by aerosols of Rhodococcus equi. Can J Vet Res
Vet Sci 48:445–448. 51:290–296.

ARCANOBACTERIUM (ACTINOMYCES–CORYNEBACTERIUM)
PYOGENES
Arcanobacterium pyogenes, previously known as Actino- porcine strains are more biochemically active than
myces pyogenes, and even earlier as Corynebacterium bovine strains (Roberts 1968; Tainaka et al. 1983). A. pyo-
pyogenes, is a common cause of suppurative lesions in genes is proteolytic, a series of serine proteases with mol-
pigs throughout the world. Infection is opportunistic, re- ecular masses of 69, 59, and 55 kDa being produced
sulting from the invasion of skin or mucous membranes along with one of 108 kDa that is only produced by pig
by resident A. pyogenes. Clinical disease can result from strains (Takeuchi et al. 1995).
vertebral osteomyelitis, arthritis, pneumonia, endocardi- A. pyogenes had been classified into the genus Actino-
tis, mastitis, and subcutaneous and deep-tissue ab- myces by Collins and Jones (1982), chiefly on the basis of
scesses. cell wall composition. The guanine-cytosine content of
DNA is 58 mol%. Identification of Arcanobacterium
ETIOLOGY pyogenes is rapid and reliable with the API 20 Strep sys-
tem (Morrison and Tillotson 1988). Recent studies of
A. pyogenes is a small gram-positive pleomorphic rod. vaginal isolates have allowed the differentiation of
There is marked morphologic variation between and Arcanobacterium hyovaginalis (Collins et al. 1993).
within strains. Growth is enhanced by the addition of
serum or blood to media and occurs under both aerobic EPIDEMIOLOGY AND PATHOGENESIS
and anaerobic conditions. The optimal temperature for
growth is 37˚C. Colonies are translucent and small, tak- Arcanobacterium pyogenes is common on the mucous
ing 48 hours to reach a diameter of 1 mm. A. pyogenes membranes of the upper respiratory tract and genital
colonies are surrounded by a narrow zone of complete tract of several animal species, including the pig. Disease
hemolysis after 24 hours on blood agar. Strains isolated is therefore the result of endogenous infection and is
from pigs are more hemolytic than those isolated from sporadic, requiring some predisposing event, such as
cattle. A. pyogenes produces a hemolysin and an exotoxin trauma, to initiate the process. For A. pyogenes to cause
that is dermonecrotic in rabbits and guinea pigs and subcutaneous lesions, devitalized or inflamed tissue is an
lethal following intravenous injection in rabbits and apparent prerequisite, since the inoculation of A. pyo-
mice (Lovell 1944). Glucose is fermented by all strains, genes subcutaneously does not, per se, lead to abscesses.
but other carbohydrate reactions are variable. In general, Infection is often secondary. Tail biting may lead to ab-
CHAPTER 44 MISCELLANEOUS BACTERIAL INFECTIONS Taylor 641

scessation and suppurative osteomyelitis, retention of la multocida, and unidentified anaerobes (Hara 1980;
the fetal membranes may lead to endometritis and infer- Jones 1980). Diagnosis of infection within a herd has
tility, lacerations of the mammary gland may lead to been attempted serologically using an immunodiffusion
mastitis and arthritis, umbilical cord contamination may test for antibody to A. pyogenes protease (Takeuchi et al.
lead to omphalophlebitis, and iatrogenic abscesses may 1979). However, in a slaughterhouse survey, only 34.4%
result from faulty injection or castration techniques. Lo- of pigs with abscesses had an antiprotease titer (Hara
cal extension may produce pelvic lymphadenitis and 1980).
peritonitis. A. pyogenes may act as a secondary invader in
preexisting pneumonia. TREATMENT
Bacteremic spread from infective foci results in a va-
riety of lesions, including embolic pneumonia, endo- A. pyogenes is sensitive to a wide range of antimicrobial
carditis, arthritis, and vertebral osteomyelitis. Experi- agents, including penicillin, tetracycline, and ery-
mental intravenous inoculation of A. pyogenes shows thromycin. Some strains have been shown to be resistant
bacterial localization within the marrow of vertebral to sulfonamides and trimethoprim. In vivo sensitivity
body epiphyses, initiating osteolysis, abscessation, and does not necessarily reflect in vitro sensitivity, for the
the formation of osteophytes (Vladutiu et al. 1982). physicochemical properties of chronic abscesses tend to
Valvular endocarditis may result from bacteremia follow- protect the bacteria from the action of antimicrobial
ing tail-biting lesions (Van den Berg et al. 1981). A. pyo- drugs. Abscesses may be removed surgically.
genes is occasionally recovered from fetuses and fetal
membranes, but its role in abortion has not been estab- PREVENTION
lished. Some of these isolates may be A. hyovaginalis.
A. pyogenes, as denoted by its name, causes suppura- Surveys of serum antibodies to A. pyogenes protease
tive lesions. Surprisingly little, however, is known of the show that approximately one-third of pigs are positive
virulence factors important in disease causation. Both (Hara 1980). Antitoxin antibodies are also demonstrable
the hemolytic protein exotoxin and protease have been and may increase with age. However, mice vaccinated
proposed as toxic factors (Kume et al. 1983). A. pyogenes with preparations of whole cells with or without toxoid
also binds alpha-2 macroglobulin (Lammler et al. 1985), or even given live organisms are not adequately protect-
a property that could interfere with local regulation of ed against subsequent challenge (Derbyshire and
inflammation. Matthews 1963; Durner and Werner 1983). There is no
effective vaccine available for swine. Prevention requires
CLINICAL SIGNS AND LESIONS management of the environment to reduce or abolish
the various conditions that predispose the development
The clinical signs are very variable, since A. pyogenes is re- of A. pyogenes lesions. The treatment of sows with an-
sponsible for a range of pathological lesions. Some, such timicrobials such as tetracyclines in the food prior to far-
as endocarditis and adhesive peritonitis, may be fatal. rowing and throughout weaning can eliminate infection
Others, such as vertebral osteomyelitis leading to poste- and reduce vulval discharges (Taylor 1984).
rior paralysis, may necessitate euthanasia. Suppurative
osteomyelitis generally affects the vertebral bodies, lead- REFERENCES
ing to transverse pathological fractures, vertebral col-
lapse, and compression of the spinal cord. Lameness re- Collins, M. D., and Jones, D. 1982. Reclassification of
sults from polyarthritis or from cellulitis and Corynebacterium pyogenes (Glage) in the genus Actino-
periarthritis. However, many lesions, including subcuta- myces, as Actinomyces pyogenes comb. nov. J Gen Microbi-
neous and intramuscular abscesses, are clinically inap- ol 128:901–902.
parent and are discovered only at postmortem or slaugh- Collins, M. D.; Stubbs, S.; Hommez, J.; Devriese, L. A. 1993.
ter. Such abscesses vary from a few millimeters to several Molecular taxonomic studies of Actinomyces-like bacte-
centimeters in size, usually have a thick fibrous capsule, ria isolated from purulent lesions in pigs and descrip-
and contain a yellow-green pus of variable consistency. tion of Actinomyces hyovaginalis sp. nov. Int J Syst Bacte-
Mastitis may be confined to one gland or may involve riol 43:471–473.
several. Derbyshire, J. B., and Matthews, P. R. J. 1963. Immunologi-
cal studies with Corynebacterium pyogenes in mice. Res
DIAGNOSIS Vet Sci 4:537–542.
Durner, K., and Werner, B. 1983. Untersuchungen zur Im-
Diagnosis in individual cases requires the demonstration munogenitat und zu den Pathogenitatsfaktoren von
of the organism in lesional material and confirmation by Corynebacterium pyogenes. Arch Exp Vet Med Leipzig
laboratory culture and identification. Carcass abscesses 37:541–547.
typically yield mixed cultures, including clostridia, Hara, F. 1980. A study on pig pyogenic infections: Results of
Bacteroides spp., Propionibacterium granulosum, Pasteurel- clinical, pathological, bacteriological and serological ex-
642 SECTION 3 BACTERIAL DISEASES D. J. Taylor, Editor

aminations of slaughtered pigs. Bull Azabu Uni Vet Med immunodiffusion test with protease antigen. Nat Inst
1:187–202. Anim Health Q (Tokyo) 19:77–82.
Jones, J. E. T. 1980. Observations on the bacterial flora of ab- Takeuchi, S.; Azuma, R.; Nakajima, Y.; and Suto, T. 1979.
scesses in pigs. Br Vet J 136:343–348. Diagnosis of Corynebacterium pyogenes infection in pigs
Kume, T.; Tainaka, M.; Saito, M.; Hiruma, M.; Nishio, S.; by immunodiffusion test with protease antigen. Nat Inst
Kashiwazaki, M.; Mitani, K.; and Nakajima, Y. 1983. Re- Anim Health Q (Tokyo) 19:77–82.
search on experimental Corynebacterium pyogenes infec- Takeuchi, S.; Kaidoh, T.; and Azuma, R. 1995. Assay of pro-
tions in pigs. Kitasato Arch Exp Med 56:119–135. teases from Actinomyces pyogenes isolated from pigs and
Lammler, C.; Chhatwal, G. S.; and Blobel, H. 1985. Binding cows by zymography. J Vet Med Sci 57:977–979.
of a2-macroglobulin and haptoglobin to Actinomyces Taylor, D. J. 1984. Clinical and bacteriological effects of an-
pyogenes. Can J Microbiol 31:657–659. timicrobial therapy on naturally-occurring post-partum
Lovell, R. 1944. Further studies on the toxin of Corynebac- vulval discharge in sows. Proc Int Congr Pig Vet Soc
terium pyogenes. J Pathol Bacteriol 56:525–529. 8:151.
Morrison, J. R. A., and Tillotson, G. S. 1988. Identification Van den Berg, J.; Narucka, U.; Nouws, J. F. M.; Okma, B. D.;
of Actinomyces (Corynebacterium) pyogenes with the API Peelen, J. P. J.; and Soethout, A. E. E. 1981. Lesions in
20 Strep system. J Clin Microbiol 26:1865–1866. slaughtered animals. II. Inflammation of the tail and
Roberts, R. J. 1968. Biochemical reactions of Corynebacteri- embolic pneumonia in pigs. Tijdschr Diergeneeskd
um pyogenes. J Pathol Bacteriol 95:127–130. 106:407–410.
Tainaka, M.; Kume, T.; Takeuchi, S.; Nishio, S.; and Saito, Vladutiu, O.; Florescu, S.; and Murgu, I. 1982. Research in
M. 1983. Studies on the biological and serological prop- the pathogenetic mechanisms of osteophytosis in py-
erties of Corynebacterium pyogenes infection in pigs by obacillary polyarthritis. Arch Vet 16:75–95.

You might also like