You are on page 1of 208

SWINE DISEASE

MANUAL

Fifth Edition

Edited by:
E.J. Neumann, A. Ramirez, and K.J. Schwartz
ISBN 978-0-9843503-1-5

COPYRIGHT © 2020 American Association of Swine Purchase online: http://ecom.aasv.org/sdm


Veterinarians.
Order questions and bulk quantity orders
All rights reserved. No part of this manual may be American Association of Swine Veterinarians
copied, reproduced, recorded, transmitted or retrieved 830 26th Street
from an information storage and retrieval system without Perry, Iowa 50220
written permission of the American Association of Swine 515-465-5255
Veterinarians. aasv@aasv.org
DISCLAIMER: Information in this manual comes
from many sources and may contain errors. In Copyediting and proofreading
particular, information on drugs, chemicals, antibiotics, Karen Richardson
anthelmintics, parasiticides, feed additives, vaccines and Sue Schulteis
any other agents used to prevent or treat swine diseases
may contain errors and inaccuracies. Veterinarians and Text formatting and cover design
all others using those products are advised to use their Barbara Molnár-Smith
own independent, best judgment, based on the most Tina Smith
recent and authoritative information provided by those
producing the product. Also, management changes and
management practices mentioned in the manual are to be
used at the discretion of the owner(s) of the swine or the
person(s) charged with those responsibilities. Neither the
authors nor universities nor diagnostic laboratories they
represent shall be liable for damage caused by reliance on
information in this manual.

Cover photo credits


Front cover photo courtesy of
Tina Smith
Front cover histopathological
images courtesy of Iowa State
University Veterinary
Diagnostic Laboratory

Back cover photo courtesy of


Ashley Schwartz
A
B C
Histopathological image descriptions
A. Alveolar macrophages of lung have positive staining for PRRSV by immunohistochemistry (IHC). Pig.
B. Intestinal enterocytes have positive staining for PEDV by immunohistochemistry (IHC). Neonatal pig.
C. Spiral-shaped bacteria (Brachyspira hyodysenteriae) present in colon by in situ hybridization (ISH). Pig.
PREFACE

This manual was written primarily for veterinary students and those who teach swine diseases. It also
may be of interest to veterinary practitioners, veterinary technicians, and others interested in swine
diseases in production systems. This manual attempts to update, condense and simplify the great
mass of information available. Only basic material appropriate for students is included. More detailed
information is available via an array of texts, websites, and scientific publications.
Disease topics are listed as individual entities. The student should be reminded that in reality, the
occurrence and severity of disease outbreaks are strongly influenced by production practices, housing,
environment, nutrition, and genetics. Diseases may occur concurrently in modern production systems
with large populations of animals; for example, porcine respiratory disease complex (PRDC) is usually a
co-infection with Mycoplasma hyopneumoniae and/or other bacteria or viruses. Diagnostic methods for
disease complexes are usually more complicated than finding a single agent. Likewise, control of disease
frequently involves not only specific intervention, but also modification of environment, husbandry,
and production practices. For this reason, the prevention and control sections in this manual refer to
specific agents but are not always appropriate or complete in terms of control of disease complexes.
Swine practitioners expend considerable effort in diagnosis, herd investigation, and understanding the
interactions of risk factors before implementing cost-effective control strategies. These activities are
beyond the scope of this manual but our vision is that this manual will serve as a foundation on which to
build a better understanding of the complexities of disease in swine populations.
Familiarity with the organization of the manual will make it more useful. The table of contents lists the
diseases and conditions of swine covered in this manual. Most sections are based on the nature of the
etiologic agent, and each section is preceded by its own table of contents. The time-saving index at the
back of the book lists each major disease by name, often cross-referenced with a synonymous name.
Several tables are included. Some are designed to help in differential diagnosis. Others list major diseases
of a single body system and permit a rapid overview of diseases of that system. The tables may be of
special value in preparing for board exams.
Comments on the manual and how it can be made more useful are encouraged and appreciated. They
can be mailed to Dr. Alex Ramirez, 2231 Lloyd Veterinary Medical Center, 1809 S Riverside Drive,
Iowa State University, Ames, Iowa 50011 or e-mailed to ramireza@iastate.edu.
TABLE OF CONTENTS

Table of Contents
Abbreviations .....................................................................................................................................................................................1
Swine industry terminology............................................................................................................................................................3

SECTION I: Diseases caused by bacteria, mycoplasmas, and spirochetes..................................7


Actinobacillus pleuropneumoniae.....................................................................................................................................................9
Actinobacillus suis............................................................................................................................................................................ 12
Anthrax............................................................................................................................................................................................. 14
Atrophic rhinitis............................................................................................................................................................................. 16
Clostridial diarrhea......................................................................................................................................................................... 18
Colibacillosis.................................................................................................................................................................................... 21
Erysipelas.......................................................................................................................................................................................... 25
Greasy pig disease............................................................................................................................................................................ 27
Haemophilus parasuis .................................................................................................................................................................... 29
Ileitis.................................................................................................................................................................................................. 32
Leptospirosis.................................................................................................................................................................................... 35
Mycoplasma suis .............................................................................................................................................................................. 37
Mycoplasmal arthritis.................................................................................................................................................................... 39
Mycoplasmal pneumonia ............................................................................................................................................................. 41
Pneumonic pasteurellosis.............................................................................................................................................................. 44
Salmonellosis................................................................................................................................................................................... 46
Streptococcus suis.............................................................................................................................................................................. 50
Swine brucellosis............................................................................................................................................................................. 53
Swine dysentery and spirochetal colitis...................................................................................................................................... 55
Tuberculosis..................................................................................................................................................................................... 58

SECTION II: Diseases caused by viruses ........................................................................... 61


African swine fever ........................................................................................................................................................................ 63
Blue eye disease................................................................................................................................................................................ 65
Classical swine fever....................................................................................................................................................................... 67
Encephalomyocarditis virus.......................................................................................................................................................... 70
Foot-and-mouth disease ............................................................................................................................................................... 72
Hemagglutinating encephalomyelitis......................................................................................................................................... 74

i
SWINE DISEASE MANUAL

Inclusion body rhinitis................................................................................................................................................................... 76


Influenza .......................................................................................................................................................................................... 78
Japanese encephalitis...................................................................................................................................................................... 81
Parvovirus......................................................................................................................................................................................... 83
Porcine circovirus associated diseases......................................................................................................................................... 85
Porcine picornaviruses................................................................................................................................................................... 88
Porcine reproductive and respiratory syndrome...................................................................................................................... 92
Porcine respiratory coronavirus................................................................................................................................................... 96
Pseudorabies .................................................................................................................................................................................... 98
Rotavirus.........................................................................................................................................................................................101
Senecavirus A.................................................................................................................................................................................103
Swine enteric coronavirus diseases............................................................................................................................................105
Swine vesicular disease.................................................................................................................................................................108
Swinepox.........................................................................................................................................................................................110
Vesicular exanthema ....................................................................................................................................................................111
Vesicular stomatitis ......................................................................................................................................................................112

SECTION III: Emerging viruses .......................................................................................115


Filoviruses (Ebola virus and Marburg virus), Hepatitis E virus ...............................................................................................117
Paramyxoviruses (Menangle and Nipah viruses), Pestivirus F (Bungowannah virus) .........................................................118
Porcine adenovirus, Porcine astrovirus, Porcine bocavirus ..................................................................................................119
Porcine parainfluenza virus type 1, Porcine sapovirus..........................................................................................................120
Torque teno virus, West Nile virus (Kunjin virus) ................................................................................................................121

SECTION IV: Diseases caused by parasites ....................................................................123


Coccidiosis.....................................................................................................................................................................................125
Cysticercosis...................................................................................................................................................................................127
Lice...................................................................................................................................................................................................129
Lungworm......................................................................................................................................................................................130
Mange..............................................................................................................................................................................................132
Minor intestinal nematodes: Kidney worm, Nodular worm, Thorny-headed worm, Threadworm...........................134
Roundworm...................................................................................................................................................................................137
Trichinellosis..................................................................................................................................................................................139
Whipworm.....................................................................................................................................................................................141

ii
TABLE OF CONTENTS

SECTION V: Miscellaneous ...............................................................................................143


Atresia ani, Aural hematoma, Congenital tremors, Cystitis and pyelonephritis, Dermatosis vegetans......................145
Ectopic ossification, Epitheliogenesis imperfects, Gastric ulcer ........................................................................................146
Hemorrhagic bowel syndrome, Hernias, Hydronephrosis, Hypogalactia and MMA...................................................148
Hypoglycemia, Megacolon, Mulberry heart disease and related conditions...................................................................149
Osteochondrosis, Pityriasis rosea, Porcine stress syndrome................................................................................................151
Prolapses, Pustular dermatitis, Rabies, Ringworm.................................................................................................................152
Shoulder ulcers in sows, Skin necrosis of piglets, Splayleg (spraddleleg), Sunburn and photosensitization.............153
Torsion and volvulus, Vestibular syndrome, Vices (tail biting, ear biting, flank biting, navel sucking).....................154

SECTION VI: Nutritional diseases, toxicoses, and poisonings .......................................155


Mycotoxicoses................................................................................................................................................................................157
Nutritional deficiencies and excesses........................................................................................................................................159
Toxins (plants, feed additives, chemicals, and gases).............................................................................................................162

SECTION VII: Tables .........................................................................................................165


Anthelmintics and parasiticides for swine...............................................................................................................................167
Arthritis in neonatal and growing swine..................................................................................................................................168
Central nervous system (CNS) diseases in swine: comparative approach to diagnosis.................................................169
Diarrheal diseases common at various ages.............................................................................................................................171
Diarrheal diseases in swine: comparative approach to diagnosis........................................................................................172
Internal parasites...........................................................................................................................................................................175
Intestinal hemorrhage: differential diagnosis..........................................................................................................................177
Mycoplasma-related diseases......................................................................................................................................................179
Reproduction: causes of low farrowing rate............................................................................................................................180
Reproductive failure: infectious causes ...................................................................................................................................181
Respiratory disease in swine: comparative approach to diagnosis......................................................................................183
Skin lesions: differential diagnosis............................................................................................................................................186
Vesiculating viral diseases............................................................................................................................................................189
Zoonotic agents found in swine and humans.........................................................................................................................190

INDEX ........................................................................................................................................... 193

iii
iv
ABBREVIATIONS

Abbreviations
AASV American Association of Swine Veterinarians FDA US Food and Drug Administration
AAVLD American Association of Veterinary FE Feed efficiency
Laboratory Diagnosticians
FMD Foot-and-mouth disease
AD Aujeszky’s disease (pseudorabies)
FSIS Food Safety Inspection Service
ADFI Average daily feed intake
HA Hemagglutination assay
ADG Average daily gain
HACCP Hazard analysis critical control point
AI Artificial insemination
HE Hemagglutinating encephalomyelitis
AIAO All-in all-out
HEV Hepatitis E virus
AMDUCA Animal Medicinal Drug Use Clarification Act
HI Hemagglutination inhibition
APHIS Animal and Plant Health Inspection Service
HPS Haemophilus parasuis
APP Actinobacillus pleuropneumoniae
IAV-S Influenza A virus of swine
AR Atrophic rhinitis
IBR Inclusion body rhinitis
ASF African swine fever
IF Immunofluorescence
BED Blue eye disease
IFA Immunofluorescent antibody
BVD Bovine viral diarrhea
IHA Indirect hemagglutination
CF Complement fixation
IHC Immunohistochemistry
CNS Central nervous system
ISH In situ hybridization
CPE Cytopathic effect
JE Japanese encephalitis
CSF Classical swine fever
JSHAP Journal of Swine Health and Production
CVM Center for Veterinary Medicine
LPS Lipopolysaccharides
DIVA Differentiate infected from vaccinated animals
MARV Marburg virus
DON Deoxynivalenol
MEW Medicated early weaning
EBOV Ebola virus
MHP Mycoplasma hyopneumoniae
ED Edema disease
MHR Mycoplasma hyorhinis
ELISA Enzyme-linked immunosorbent assay
MHS Mycoplasma hyosynoviae
EM Electron microscopy
MLVA Multiple-locus variable number tandem
EMC Encephalomyocarditis repeat analysis
EP Enzootic pneumonia MLST Multi-locus sequence typing
EPG Eggs per gram MMA Mastitis, metritis, agalactia
ET Embryo transfer MMEW Modified medicated early weaning
FA Fluorescent antibody MPS Mycoplasmal pneumonia of swine
FAT Fluorescent antibody test MRSA Methicillin-resistant Staphylococcus aureus
FADDL Foreign animal disease diagnostic laboratory NAD Nicotinamide adenine dinucleotide
FCR Feed conversion ratio NADC National Animal Disease Center
ABBREVIATIONS 1
SWINE DISEASE MANUAL

NPB National Pork Board PSY Pigs weaned per sow per year
NPPC National Pork Producers Council PWM Preweaning mortality
NRC National Research Council RFLP Restriction fragment length polymorphism
OA Osteoarthritis SAHO State animal health official
OCD Osteochondrosis SD Swine dysentery
PAdV Porcine adenovirus SECD Swine enteric coronavirus diseases
PAR Progressive atrophic rhinitis SEW Segregated early weaning
PAstV Porcine astrovirus SIV Swine influenza virus (archaic; see IAV-S)
PBoV Porcine bocavirus SMEDI Stillbirth, mummification, embryonic death,
infertility
PCMV Porcine cytomegalovirus
SMSLV San Miguel sea lion virus
PCR Polymerase chain reaction
SN Serum neutralization
PCV Porcine circovirus
SPF Specific pathogen free
PCVAD Porcine circovirus associated disease
SVA Senecavirus A
PCVD Porcine circovirus disease
SVV Seneca Valley virus (see SVA)
PDCoV Porcine deltacoronavirus
SwPV Swinepox virus
PDNS Porcine dermatitis and nephropathy syndrome
TB Tuberculosis
PED Porcine epidemic diarrhea
TGE Transmissible gastroenteritis
PHE Porcine hemorrhagic enteropathy
USAHA United States Animal Health Association
PIA Porcine intestinal adenomatosis
USDA United States Department of Agriculture
PMWS Postweaning multisystemic wasting syndrome
VE Vesicular exanthema
PPE Porcine proliferative enteropathy
VES Vesicular exanthema of swine
PPV Porcine parvovirus
VFD Veterinary Feed Directive
PQA Pork Quality Assurance
VI Virus isolation
PRCV Porcine respiratory coronavirus
VN Virus neutralization
PRDC Porcine respiratory disease complex
VS Vesicular stomatitis
PRRS Porcine reproductive and respiratory syndrome
WNV West Nile virus
PRV Pseudorabies virus
PSS Porcine stress syndrome

2 ABBREVIATIONS
SWINE INDUSTRY TERMINOLOGY

Swine industry terminology


Following are some terms used in the swine industry. Although Carcass yield
many of the terms have never been defined precisely, the defini- Percent of the live body weight in the eviscerated carcass.
tions give some idea as to their meaning.
Commercial herd
Acclimatization Type of swine farm where the main intention is to raise pigs for
A period for new breeding stock to become immune to disease slaughter. These herds make the final cross between “terminal”
organisms on the farm. This is usually accomplished by isolating sires and “maternal” females to produce fast-growing, lean, effi-
new animals prior to their entry into the herd, while intentionally cient pigs for slaughter. Commercial herds are also referred to
exposing them to potential pathogens on the recipient farm, such as “parent” herds; they receive replacement gilts from multiplier
as by commingling with culls. The goal is for incoming breeding herds and send offspring to slaughter.
stock to become immune to and cease shedding endemic disease
agents. Acclimatization is often performed at the same time as the Conception rate
period of isolation. The number of sows detected pregnant divided by the total num-
ber of sows serviced (bred).
Average daily gain (ADG)
The average amount of weight a pig gains per day. Continuous flow
Any rearing system under which the pigs use facilities continuously
Adjusted farrowing rate or nearly so, or where a facility is not completely depopulated
The number of sows farrowed, divided by (the number of sows before more pigs are added. Often the pigs are not started, raised,
serviced minus the number of sows that were culled for non-re- or sold together, which is the opposite of the AIAO system.
productive reasons).
Creep feed
All-in all-out (AIAO) Special feed fed to piglets prior to weaning.
A production method where there are no additions to a group of
pigs following their placement into a room, barn, or location. All Cross-fostering
pigs are removed before any subsequent groups of pigs are placed. Moving pigs from one sow to another to balance the size or num-
Usually, pigs in a group are born in as brief a period as possible (1 ber of pigs per litter.
to 2 weeks), started, raised, and sold together. Each subsequent
group is separated physically and has separate air space. The Cryptorchid
facilities are totally depopulated, cleaned, and disinfected before A male pig with one testicle retained in the body cavity.
the next group of pigs enters. There are many disease control
advantages of AIAO because there is only one age group of pigs Cull rate
in a location. The number of sows removed from a herd annually, divided by
the total number of sows in the herd.
Back pressure test
Applying pressure to the back of a sow to determine if she is
exhibiting estrus. Depopulation/Repopulation
Removing all animals on the entire farm, cleaning and disinfect-
Barrow ing the facilities, and then repopulating the farm with pigs free of
Castrated male pig. specific diseases.

Boar Down time


Intact male pig. Time during which there are no animals in a barn, which facili-
tates cleaning and disinfecting.
Boar taint
The characteristic smell and taste that is often detectable in pork Farrow
derived from sexually mature boars. To give birth to piglets.

Carcass index
An index used to establish the value of a carcass based on body
weight and backfat measurement.

SWINE INDUSTRY TERMINOLOGY 3


SWINE DISEASE MANUAL

Farrow-to-finish Induced farrowing


A swine production system where all stages of production The use of chemicals to trigger parturition and enable a producer
(breeding, gestation, farrowing, nursery, and finishing) are on to attend farrowings.
the same site, often housed together or in immediately adjacent
buildings. The buildings may be subdivided or partitioned into Isolation
sections for each stage of production. Animals (eg, new breeding stock) that have a risk of introducing
a disease into a breeding herd are placed in a location that is iso-
Farrowing rate lated from the rest of the herd. There they are observed and tested
The number of sows that farrowed divided by the number of sows
for disease exposure before being allowed to enter the breeding
serviced (bred).
herd. Often, isolation and acclimatization occur simultaneously.
Feed conversion ratio (FCR) Lactogenic immunity
The amount of feed used divided by the total weight gained; also
Antibodies (especially IgA) passed from the sow to the piglet via
referred to as feed efficiency or feed to gain (F:G).
the milk.
Feed wastage Market hog
Feed that is fed but is not eaten.
Market weight pig; usually greater than 220 pounds (100 kilo-
grams) in the US.
Feeder pig
A pig moved or sold from a nursery at approximately 50 pounds
(23 kilograms).
Medicated early weaning (MEW)
A system of raising pigs under which piglets are weaned very
young, as early as 4 to 5 days of age, and are aggressively medicated
Finisher from birth until they are 10 to 20 days old. The system is used to
Stage of production after the grower phase, starting at around reduce or eliminate pathogens that might be transmitted to piglets
150 pounds (68 kilograms) and continuing to market weight of from the dams. Modified medicated early weaning (MMEW) is
250 to 290 pounds (114 to 132 kilograms). a variant of MEW where pigs are weaned later than a specified
age (eg, 17 days of age). The specific weaning age and medication
Floor feeding depend on the disease(s) in the source herd to be controlled.
Putting the pigs’ feed directly on the floor, rather than in a feeder.
Multiple mating
Genetic pyramid Mating or inseminating a sow more than once during a single
System by which animals of highest genetic value are used to pro- estrus cycle.
duce offspring that will be used to populate commercial farms. The
“nucleus” herd supplies genetically superior animals to “multiplier” Multiplier herd
herds that produce the gilts that will populate commercial farms. Herd of purebred animals that are crossed to produce replace-
ment gilts for use in commercial herds. Multiplier herds receive
Gilt gilts from nucleus herds and send their offspring to commercial
A young female pig that has not given birth. farms. Multiplier herds are also called “grandparent” farms.

Gilt pool Multi-site production


The group of gilts selected for the breeding herd but not yet bred. A system of raising pigs in well-separated facilities. In a 3-site
system, site 1 includes breeding, gestation, and farrowing, site 2
Grower is the facility where weaners are placed in a nursery, and site 3 is
Stage of production after the nursery phase, when a pig weighs a grower/finisher facility. All sites operate as AIAO. Some pro-
between 50 and 150 pounds (23 and 68 kilograms). ducers prefer to combine sites 2 and 3, and the operation is then
referred to as a 2-site system.
Hybrid vigor
The improved production due to heterosis; the combination of Not in pig (NIP)
two purebreds. A sow that is assumed to be pregnant at the end of gestation but
found not pregnant just prior to farrowing.

4 SWINE INDUSTRY TERMINOLOGY


SWINE INDUSTRY TERMINOLOGY

Nucleus herd Seasonal infertility


A herd at the top of a genetic pyramid, usually composed only Decreased production associated with breeding sows during the
of purebred stock of superior quality and housed in highly spe- summer months.
cialized, biosecure farms called “nucleus” farms. Improvement
at this level is obtained by selecting superior individuals within Segregated early weaning (SEW)
each breed. From these pure lines, individuals are selected for Weaning all piglets under a specified age, often approximately
particular characteristics such as growth rate, backfat, or litter 17 days of age, usually as part of a short-term disease control
size. The best individuals from this population will go to multi- strategy.
plier herds for crossbreeding. Nucleus farms are also referred to as
“great grandparent” farms. Service
One or more mating events that occur during a single estrus cycle
Nursery pigs constitute a service.
Pigs from weaning to 50 to 75 lbs (23 to 34 kilograms).
Specific pathogen free (SPF)
Parity A method of obtaining pigs free of specified pathogens by raising
The number of litters that a sow has farrowed. surgically derived piglets apart from their dam, usually in an
isolated facility or by a foster sow in the recipient herd. A strategy
Parity 0 used for genetic improvement, disease elimination, or starting a
A gilt that has been bred but has not yet farrowed. new, disease-free herd.

Partial depopulation Split suckle


Depopulating one area of a barn or phase of production to create Management technique where the more robust or first-farrowed
a physical “gap” in the production flow; part of some disease pigs of a large litter are removed for one to two hours to allow
control strategies. smaller, later-born pigs to suckle to ensure all piglets receive an
adequate dose of colostrum.
Pig density
The number of pigs per square area (foot or meter) of floor space. Split wean
Weaning the biggest pigs in a litter 2 to 3 days before the others
Pigs marketed per sow per year in order to increase weaning weight of the remaining piglets and
The number of pigs marketed per year divided by the number of decrease weaning-to-breeding interval.
sows in the herd.
Sow
Pigs weaned per sow per year (PSY) A female pig that has been bred and given birth.
The number of pigs weaned per year divided by the number of
sows in the herd. Starter feed
First specialized diet fed to newly weaned pigs.
Preweaning mortality (PWM)
Total number of pigs that die after birth but before weaning Weaner (weaner pigs)
(number of pigs born alive minus number of pigs weaned) Term describing pigs when they are removed from the sow and the
divided by total number of pigs born alive. early growing, ie, nursery, phase for up to 8 weeks after weaning.

Return to estrus Weaning-to-breeding interval


Sow that comes back into heat after breeding and before farrow- Number of days between weaning and breeding a sow.
ing, indicative of a failed conception or loss of pregnancy.
Wean-to-removal interval
Savaging Time between weaning and culling a sow.
When a sow or gilt is aggressive to her newborn piglets, usually
by biting or eating them. The cause of the condition is poorly Zero wean
understood. Weaning a sow the day that she farrows.

Scour
Common term for diarrhea.

SWINE INDUSTRY TERMINOLOGY 5


6
BACTERIA
Section I

Diseases caused by bacteria,


mycoplasmas, and spirochetes

Actinobacillus pleuropneumoniae ....................................................................................................................................................9


Actinobacillus suis............................................................................................................................................................................ 12
Anthrax............................................................................................................................................................................................. 14
Atrophic rhinitis............................................................................................................................................................................. 16
Clostridial diarrhea ........................................................................................................................................................................ 18
Colibacillosis ................................................................................................................................................................................... 21
Erysipelas.......................................................................................................................................................................................... 25
Greasy pig disease ........................................................................................................................................................................... 27
Haemophilus parasuis .................................................................................................................................................................... 29
Ileitis.................................................................................................................................................................................................. 32
Leptospirosis.................................................................................................................................................................................... 35
Mycoplasma suis .............................................................................................................................................................................. 37
Mycoplasmal arthritis.................................................................................................................................................................... 39
Mycoplasmal pneumonia ............................................................................................................................................................. 41
Pneumonic pasteurellosis.............................................................................................................................................................. 44
Salmonellosis................................................................................................................................................................................... 46
Streptococcus suis.............................................................................................................................................................................. 50
Swine brucellosis............................................................................................................................................................................. 53
Swine dysentery and spirochetal colitis ..................................................................................................................................... 55
Tuberculosis..................................................................................................................................................................................... 58

7
8
BACTERIA
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Actinobacillus pleuropneumoniae
Alternate names and abbreviations quantities of exotoxins produced vary among the 16 serotypes.
APP, porcine pleuropneumonia, Haemophilus pleuropneumoniae Antibodies to the specific exotoxins probably are important in
or HPP (archaic) protective immunity.
Actinobacillus suis is closely related to APP, and some strains pro-
Definition duce one or more of the RTX toxins as well. The two organisms
This organism causes severe pleuropneumonia and is a highly may cross-react on some serological tests; polymerase chain reac-
contagious disease of swine often characterized by sudden onset, tion (PCR) tests must also be interpreted with care.
short clinical course, high morbidity, and high mortality.
Epidemiology
Occurrence Actinobacillus pleuropneumoniae is usually transmitted within and
Porcine pleuropneumonia caused by Actinobacillus pleuropneumo- between herds by direct contact via nasal secretions and fomites,
niae (APP) is widely distributed throughout major swine-raising but transmission via aerosol for short distances (eg, 100 meters)
countries and is one of the most important bacterial respiratory has been reported. Fomites, including boots and coveralls, can
pathogens globally. The host range of APP is restricted to swine. spread the organism but are infectious for only a short time since
Most virulent strains can cause rapidly fatal pneumonia, with APP is fragile outside the host. APP does not persist in the envi-
abrupt and severe outbreaks not uncommon. In naïve herds, APP ronment but can survive several days to several weeks in mucus
occurs in all age groups, but it is most frequently seen in pigs 6 to exudates in moist organic matter. Routine hygienic practices with
20 weeks of age. Actinobacillus suis can cause similar disease and disinfection are effective for environmental elimination.
lesions but is not as contagious as APP.
Transmission from dam to offspring in positive herds probably
Historical information occurs late in the suckling phase (“late colonizer” of nasopharynx
In the US, APP was first reported in 1957. It was reported and tonsil), making APP elimination from populations possible
frequently in the 1960’s and 1970’s. The frequency of outbreaks by medicated early weaning techniques. Subclinical carriage of
increased with the industrialization of swine production. The APP in tonsils occurs for long (indefinite) periods in both healthy
disease and its epidemiology are well researched through exper- animals as well as in survivors of clinical outbreaks. The likelihood
imental studies and field experiences. Because of the economic of disease expression is strongly influenced by the virulence cassette
consequences of mortality and cost of control of this disease, of the particular serotype present, the status of passive or active spe-
most producers strive to eliminate the more virulent serotypes of cific immunity, and the presence of both infectious and non-infec-
APP from herds. tious risk factors. Overstocking, inadequate ventilation, coinfection
with other respiratory pathogens, or unusual stress may facilitate
Etiology transmission or precipitate outbreaks.
The APP organism is a hemolytic, Gram-negative, encapsulated,
coccobacillary rod that is highly host-specific for swine and Pathogenesis
fragile outside the host. Strains denoted by serotype vary in vir- Although carriage of organisms by asymptomatic carriers can be
ulence and pathogenicity. There are 2 biotypes, whereby biotype for indefinite periods, when finally introduced into the deep lung
I is dependent on an exogenous source of NAD (nicotinamide (alveoli), disease onset is rapid with fatal pleuropneumonia pos-
adenine dinucleotide) in culture and biotype II is not. NAD is sible within hours. This suggests that other risk factors can facili-
usually supplied by a Staphylococcus nurse streak, which allows the tate or initiate outbreaks in colonized populations of pigs. Once
“satellite” growth of APP biotype I on blood agar. Biotype 1 has introduced to the deep lung tissue, hemolysins, Apx toxins, and
14 serotypes (1-12, 15, 16) and biotype II is composed of 2 sero- other proteins produced by APP affect the function of endothe-
types (13, 14) that are usually considered to be less virulent than lial cells and pulmonary alveolar macrophages. There is variation
biotype I serotypes. However, because genetic variation, serologi- in the hemolytic and cytotoxic effects of ApxI, ApxII, and ApxII,
cal cross-reaction, and different classification schemes are utilized, which impacts virulence. Severe damage to endothelial cells,
categorical classification of isolates can sometimes be confusing if pneumocytes, and inflammatory cells from Apx cytotoxins occurs
not daunting. Serotype prevalence varies considerably across the rapidly and leads to ischemic necrosis and endotoxic shock. These
globe and by region. peracute cases typically present as cases of sudden deaths. In acute
and subacute cases, inflammation and thrombosis is followed by
The APP organisms secrete one or more of 4 exotoxins: ApxI,
infarction and sequestration of infarcts in the lungs.
ApxII, ApxIII, and ApxIV, together called RTX toxins. Actino-
bacillus pleuropneumoniae also produces abundant endotoxin. Pathogenesis involves both variation in host resistance and
The RTX toxins are cytotoxic and/or hemolytic. The types and variation in virulence among serotypes. Generally, serotypes 1

ACTINOBACILLUS PLEUROPNEUMONIAE 9
SWINE DISEASE MANUAL
BACTERIA

and 5 are considered more virulent than 7 or 3, but all can cause Accurate differentiation of APP and A suis based on gross lesions
significant disease and any of these may be present subclinically in is difficult, perhaps impossible. Because A suis infection is more
populations of pigs. often accompanied by septicemia, bacterial culture is an import-
ant tool in establishing a diagnosis. This differential is important
Other pathogens (eg, Mycoplasma hyopneumoniae, influenza A
with less virulent strains of APP where disease is sporadic and less
virus in swine, and porcine reproductive and respiratory syndrome
severe.
virus), commingling, or environmental factors that affect pulmo-
nary clearance in the lung may influence onset and course of the
disease. Colostral immunity in young pigs confers some resistance
Diagnosis
The sudden onset of an acute, rapidly spreading, respiratory dis-
to infection and permits gradual development of active immunity
ease associated with high morbidity and mortality is suggestive of
in many exposed, growing pigs. Naïve pigs suddenly exposed to
APP. Observation of dark red infarcts and pleuritis in the lungs
APP are more likely to develop fulminating clinical signs.
of acutely affected cases, sequestered nodules in lungs of chronic
cases, and a high incidence of pleuritis in the herd are also highly
Clinical signs suggestive for APP involvement. Virulent serotypes of APP exist
Signs vary considerably between peracute and chronic forms
within both biotype I (NAD dependent) and biotype II (NAD
of APP. With peracute disease, individual pigs are either found
independent). Isolation and identification of APP is commonly
dead or have high fever, lethargy, anorexia, and may vomit. Pigs
practiced in order to confirm the diagnosis. Detection by molec-
rapidly become recumbent, cyanotic, dyspneic, and die with
ular testing (PCR) is also confirmatory and may allow immediate
foamy blood-tinged discharge from the nose. With acute disease,
genotyping to predict serotype. Histopathology is useful to
more pigs are affected, with slower progression of similar signs,
confirm typical lesions as well as to elucidate whether other insult
and the outcome is not necessarily fatal. Pigs may have a shallow
types are present. Acute outbreaks of APP must be differentiated
cough, and pigs medicated at this stage may respond to antimi-
from other respiratory diseases, especially A suis infection, classi-
crobial intervention. In less severely affected pigs, chronic disease
cal swine fever, pneumonic pasteurellosis, pneumonic salmonello-
can result and is seen as intermittent cough, reduced gain, and
sis, influenza, and mycoplasmal pneumonia.
ill-thrift with signs of undifferentiated pneumonia because of the
pleural adhesion and pulmonary abscesses that are likely present. Serotyping is commonly performed on isolates confirmed as
APP to characterize expected virulence, aid in epidemiology, or
Morbidity and mortality in growing swine vary but can become
predict immune cross-protection by natural or vaccinal exposure.
high. Peak mortality usually occurs in pigs 10 to 16 weeks old.
However, cross-reactions are problematic for some isolates. Use
Mortality can reach 20% to 80% in fattening pigs. Abortions may
of monoclonal antibodies improves accuracy but some isolates
occur in acutely infected pregnant females.
still cannot be differentiated by serological tests. Advancement in
molecular techniques, particularly PCR, for predicting serotype,
Lesions is providing less uncertainty for some of the more troubling sero-
Lesions of APP are usually restricted to the lungs. Characteristic types and likely will improve over the next few years. PCR toxin
lesions include locally extensive areas of pulmonary necrosis typing does not provide serotype information but can determine
and hemorrhage that are quite firmly consolidated, dark red in which Apx toxin genes are present in a particular isolate.
color, and are nearly always accompanied by fibrinous pleuritis.
The thoracic cavity often contains blood-tinged fluid. The classic To eliminate APP from a herd, accurate detection of inapparent
lesion of APP consists of large areas of hemorrhage and necrosis carriers can be very helpful. Immunomagnetic separation and cul-
in the dorsal portion of one or both diaphragmatic lobes accom- ture of tonsil samples has been useful but is quite technical and
panied by severe fibrinous pleuritis. These lesions develop as a laborious with limited availability. PCR performed on tonsils,
result of APP gaining access to alveoli (through inhalation) and tonsil biopsies or tonsil scrapings is quite sensitive, particularly if
causing large areas of infarction overlaid with fibrinous pleuritis. performed after selective enrichment culture, but is not capable
of differentiating APP serotypes. Detection of inapparent carriers
Bloody froth often fills larger airways and there may be interlobu-
with 100% sensitivity should not be expected with either culture
lar edema along with congestion in the remainder of lung. Fibrin-
or molecular techniques.
ous pleuritis almost always overlays pneumonic areas and extends
to adjacent costal pleura. Occasionally there may be serofibrinous Serology is useful for APP surveillance and probably the most
pericarditis. Even less frequent is necrotic or ulcerative pharyngi- useful tool for detection of subclinical infections. Enzyme-linked
tis and polyarthritis. immunosorbent assay (ELISA) tests can target Apx antigens,
with a commercially available ELISA for detection of ApxIV
Chronic cases often have large, sequestered or encapsulated nod- antibodies shown to be quite sensitive for detection of subclinical
ules of necrosis in the lungs that incompletely resolve as abscesses. carriers or previously infected pigs. As with all diagnostic tests,
Lung lesions are often attached to the rib cage by fibrous adhe- communication with experts at the diagnostic laboratory is essen-
sions, leading to condemnations at slaughter. tial for sample selection, preservation, test type(s) requested, and
interpretation of results.
10 ACTINOBACILLUS PLEUROPNEUMONIAE
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Control The efficacy of various vaccines can be difficult to quantify. Gen-
Herds free of APP should be managed as closed herds since eration of antibodies or the level of serum antibodies to various
introduced swine may be carriers. Breeding stock and pigs that components in the vaccine (eg, LPS, capsule, outer membrane
must be introduced should originate from APP-free herds. They proteins, Apx toxins) may not be an accurate predictor of the
should be introduced only after testing both before and after a level of protection.
quarantine period of at least 30 days. Most commonly, vaccines are used to increase immunity in the
Introduction of naïve animals to an APP-positive herd requires sow herd, and the practice of vaccinating the sow may have the
careful planning. Usually, animals to be introduced are vacci- benefit of delaying the colonization of her piglets. However,
nated, perhaps multiple times, with a homologous bacterin-tox- routine practice of sow-herd vaccination remains controversial
oid. They are monitored closely when commingled with resident because the resulting high level of maternal antibodies that occurs
animals and, should some sicken, be treated parenterally as indi- in these piglets means that piglet vaccination must be delayed
viduals or by affected pen. until piglets are perhaps as much as 7 weeks old before the mater-
nally derived antibodies have declined to low enough levels to
Usually, control of APP in endemically infected herds requires permit effective vaccination. Vaccination tends not to eliminate a
a combination of medication, improved husbandry, and vacci- carrier state from developing.
nation. Because antibiotic therapy is only effective early in the
course of this rapidly progressing disease, outbreaks of pleuro- In some cases, particularly those associated with serotypes of high
pneumonia should be treated aggressively to decrease mortality virulence, all efforts to control the disease effectively fail and the
and mitigate lung damage in survivors. Acute outbreaks demand cost of long-term control becomes unacceptably high. At this
immediate parenteral antimicrobial treatment of entire pens con- point, eradication of APP becomes a consideration, and in fact
taining affected pigs as well as those in adjacent pens because of is achievable, though it requires careful planning, commitment,
the rapid nose-to-nose spread. Depending on the situation, anti- and effort. Complete depopulation followed by repopulation
biotics administered via the water supply may be a useful adjunct with APP-free animals is an effective but costly method of erad-
during outbreaks to suppress new cases in those pigs that are still ication. Depopulation must be followed by thorough cleaning
willing to drink. However, water medication should not be relied and disinfection of all facilities and leaving them vacant for a few
on for treatment of pigs that are sick. weeks, then restocking using APP-free swine. An alternative to
depopulation is medicated early weaning with age segregation,
Feed medication in pulse doses or continuously has been used for perhaps augmented by vaccination, medication, culling, and
prevention and on-going control in groups of pigs. This combi- repopulation. This option can result in weaning of APP-negative
nation of injectable antibiotic augmented with medicated feed piglets, even though the breeding herd remains APP positive.
or water for a few weeks can reduce mortality and decrease the
impact of chronic disease. Antimicrobials do not eliminate the Each farm and each serotype have different nuances, hence
infection from a group; outbreaks can still occur after the medica- careful review of necessary biosecurity, vaccination, colostrum
tion has been removed. management, facility limitations, interventions, procedures, and
costs are warranted before attempting elimination.
Management factors of importance in prevention include all-in,
all-out, age-segregated production with careful cleaning and
disinfection between groups, avoiding overstocking, assuring
ventilation systems are functioning properly, and eliminating or
controlling other predisposing diseases.
The use of vaccines, usually in the form of killed bacterin-toxoids
or subunit toxin-based products, is important for the prevention
of APP. Bacterins are usually serotype-specific and are sometimes
produced as autogenous products. Bacterins may induce pro-
duction of antibodies to surface antigens of APP. However, the
appearance of antibodies after vaccination may not relate to the
level of protection provided by the vaccination. Newer types of
subunit or toxin-based vaccines may produce limited cross-pro-
tection against multiple serotypes. Some of these products are
“fortified” with other components of APP such as outer mem-
brane proteins. Their use should not interfere with the serological
tests that are based on detection of antibodies to capsular or
lipopolysaccharides (LPS).

ACTINOBACILLUS PLEUROPNEUMONIAE 11
SWINE DISEASE MANUAL
BACTERIA

Actinobacillus suis
Alternate names and abbreviations Pathogenesis
None Invasion probably occurs through the tonsil with the organism
then spreading through the bloodstream. This is suggested by the
Definition presence of the organism in emboli within small blood vessels
An infectious disease caused by Actinobacillus suis characterized at many lesion sites. The agent’s pathogenic effect is probably
by septicemia, with hemorrhages and embolic lesions at multiple a result of production of hemolysin and Apx toxins, similar to
sites, more obviously in the lungs. Necrotizing bronchopneumo- those produced by APP. The organism has genes that code for
nia with pleuritis is common, often with serous or serofibrinous ApxI and ApxII, similar to APP, but appears to produce less of
exudates in the thorax, pericardium or abdomen. those toxins. Other virulence factors are suspected to be pro-
duced by A suis but are not well characterized.
Occurrence
Outbreaks occur sporadically as various manifestations of acute Clinical signs
sepsis in pigs of all ages, with serositis in young pigs, acute pneu- The first signs of an outbreak may include sudden death of young
monia in grower pigs, and acute septicemia in adults as the most pigs with lesions attributed to a bacterial septicemia. Sick piglets
common presentations. Most outbreaks are in recently weaned are febrile, breathe rapidly, and may have congested or cyanotic
pigs or in grow-finish pigs, usually with acute illness and death of extremities. Cyanosis, arthritis, enteritis, and rarely necrosis
individual pigs while the remainder of the cohort remains clini- of the feet, tail, and ears have been reported and are similar to
cally normal. other causes (streptococcal infection, erysipelas, Salmonella) of
bacterial septicemia, particularly in suckling piglets and weaners.
Historical information Occasionally, infected pigs may show central nervous system
There have been reports of the disease since 1962. In the 1980’s, signs including tremors, shaking, or paddling. Mortality within
after management techniques were introduced that minimized affected litters can be high, up to 50%.
exposure of neonates to many endemic pathogens, the incidence Older growing pigs and adults can have similar signs but may also
of A suis infections appeared to increase in the US industry. have signs of acute respiratory distress. Lethargy or depression,
Misdiagnosis of the disease as Actinobacillus pleuropneumoniae anorexia, and rarely irregular reddened skin lesions that resemble
(APP) infection or other bacterial septicemias likely contributed those of erysipelas may be observed. Pregnant sows may abort.
to under-reporting of the true prevalence of the disease.
Lesions
Etiology Lesions are similar in pigs of all age groups but vary considerably
Actinobacillus suis is a Gram-negative, nicotinamide adenine between pigs and include petechial and ecchymotic hemorrhages
dinucleotide (NAD)-dependent, non-motile coccobacillus. It can in many organs. Frequently, lesions are present in the lungs as
be aerobic or facultatively anaerobic. All isolates appear to be very disseminated foci of hemorrhagic to necrotizing pneumonia, the
similar based on biochemical characteristics, restriction fragment latter more marked in older piglets and growers and often with
length polymorphism (RFLP), and toxin analysis. Antibodies to fibrinous pleuritis. A consistent lesion in all age groups is serous
A suis and APP may cross-react on some serological tests. or serofibrinous exudate in the abdominal and thoracic cavities.
Polyarthritis may occur in piglets and grow-finish pigs.
Epidemiology
The epidemiology is not completely understood but carrier pigs Microscopic lesions occur in many major organs. These develop
probably introduce A suis into herds. The organism appears to around small colonies of A suis within the vascular emboli or
be widespread in swine populations and can be isolated from thrombi. Occasionally there are eosinophilic clubs surrounding
the nasal cavity and tonsils of many healthy pigs as well as the the colony. Both gross and microscopic lesions are typical of
reproductive tract of healthy sows. It is believed to be an “early embolic, bacterial septicemias.
colonizer” of neonates. Less than optimal environmental con-
ditions or concurrent diseases may increase the ability of A suis Diagnosis
to cause disease. Prevalence of the disease seems to be greater in History, signs and lesions are suggestive of the diagnosis, but it
newly populated herds or high health herds, presumably due to should be confirmed by culture of A suis from multiple organs.
an absence of herd immunity. PCR tests are available to detect A suis and differentiate it from
APP. Serological diagnosis is unreliable because antibodies to
A suis and APP may sometimes cross-react. Laboratory testing for
A suis is not routinely available.

12 ACTINOBACILLUS SUIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Control
Autogenous vaccines have been used but have not been evaluated
critically. Because there are multiple types of lipopolysaccharides
(LPS) in A suis, the LPS profile of the vaccine strain must match
that of the pathogen. Bacterin-toxoids containing ApxI and
ApxII are available.
Actinobacillus suis is sensitive to many antibiotics. Sick pigs
should be treated parenterally as soon as possible. The exposed
group can be treated by medicating the water.

ACTINOBACILLUS SUIS 13
SWINE DISEASE MANUAL
BACTERIA

Anthrax
Alternate names and abbreviations the animal source component (eg, bone meal, tankage, or meat
None scraps). Direct pig-to-pig transmission is rare. Biting flies and
mosquitoes have been shown to transmit anthrax.
Definition
Bacillus anthracis can cause an acute bacterial disease in most Pathogenesis
warm-blooded animals, including swine and man. During infection, the anthrax bacilli produce a complex
exotoxin. In experimental settings, a lethal factor in the exotoxin
Occurrence is responsible for the death of the animal. Lethal factor causes
In swine, anthrax is relatively rare except when large numbers depression of the central nervous system and has a harmful effect
of anthrax organisms are ingested. Although anthrax occurs on mitochondria.
throughout the world, incidence in swine in the US is low. Invasion of the anthrax bacilli is usually through defects in the
Anthrax in grazing animals other than swine tends to occur in pharyngeal mucosa or the tonsils. Invasion is often followed by
certain geographic areas and is not seasonal. Swine, however, are lymphangitis and localization of B anthracis in tonsils and/or cer-
rather resistant to anthrax. vical lymph nodes. Alternatively, there can be focal or multifocal
localization in the intestine followed by spread to one or more
Historical information mesenteric lymph nodes. Localizations tend to be necrotizing.
Although there are many historical reports of anthrax in species Frequently, both pharyngeal and intestinal lesions develop. There
other than swine, most reports on the disease in swine have seldom is a septicemic form of anthrax in swine.
occurred since 1952 to 1953 when a major epidemic occurred in
several Midwestern states. The outbreak was caused by imported, Clinical signs
contaminated bone meal used in preparing feed for swine. The Sudden, unexpected deaths may be the first signs of anthrax
feed was widely distributed before anthrax was finally diagnosed. following a highly variable incubation phase (1 to 8 days). Signs
In 1989 to 1990, an outbreak lasting over 14 weeks in a 500- depend upon the site of localization of lesions. Three forms of
sow operation was reported. Reports of this outbreak serve to anthrax, based on site of localization have been described: pha-
emphasize the great difficulties and expense in depopulating, ryngeal anthrax, intestinal anthrax, and septicemic anthrax. Swine
cleaning, and disinfecting a large contaminated confinement with pharyngeal anthrax, the most common form, show marked
facility or slaughterhouse. cervical swelling and dyspnea; they may die from suffocation.
Other signs include fever, depression, and inappetence, perhaps
vomiting. Swine with intestinal anthrax have similar signs, except
Etiology that cervical edema and dyspnea may not be apparent. Also, there
Anthrax is caused by infection with B anthracis, a large, aerobic,
may be blood in the feces. Both pharyngeal and intestinal forms
Gram-positive rod. The organism is easy to culture, and colonies
can occur in the same animal. Swine are quite resistant to anthrax
start to appear on blood agar within 12 hours. After 24 hours, the
and spontaneous recovery from cases with localized lesions is com-
colonies will be non-hemolytic and have irregular borders giving
mon. The rare septicemic cases that do occur have a short course
them a “ground glass” appearance; they are sticky when touched
and may result in sudden death.
with a bacteriologic loop. Under suitable aerobic conditions,
spores are formed that are quite resistant to heat, desiccation, and
many disinfectants. Spores can persist in the environment for over Lesions
50 years. A necropsy should not be performed on any animal suspected of
having died from anthrax; spores will be formed when the carcass is
In tissue, the organisms have a well-developed capsule and grow opened and the organism is exposed to oxygen; these spores can per-
in short chains. They have square ends when found in contact sist at the site for years. Necropsy will also expose humans to anthrax.
with others in the same chain.
In anthrax-infected cadavers, rigor mortis may be weak or absent.
The blood may fail to clot and may be discharged from nose,
Epidemiology
Decaying, anthrax-infected carcasses and their fluids contaminate mouth, or anus. Many animals have no visible external lesions
the soil with resistant spores which then serve as a source of infec- other than pharyngeal swelling. Animals with pharyngeal anthrax
tion for grazing animals for months to years. Swine, however, are usually have a gelatinous cervical edema and markedly enlarged,
rather resistant to anthrax and are seldom infected while grazing. hemorrhagic, cervical lymph nodes. Tonsils are often covered
Infection of swine usually occurs when they have fed on an with fibrinous exudates. There may be a marked pharyngitis. In
anthrax-infected carcass, or when feeds containing large numbers chronic cases, necrotic lesions in nodes or tonsils may be gray-
of spores are fed to them. The source in contaminated feeds is ish-yellow and circumscribed.

14 ANTHRAX
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Swine with intestinal anthrax usually have peritonitis with Bacillus anthracis can also be identified via polymerase chain
excessive, bloody peritoneal fluid. Fibrin may be present in the reaction or be cultured on blood agar and identified from colo-
fluid and overlay the serosa of affected segments of intestine. nies that soon develop. Caution must be used when growing B
The mesentery attached to affected intestine (ileum and spiral anthracis due to the zoonotic potential of this pathogen. Anthrax
colon) is edematous and may contain reddened streaks around must be differentiated from pharyngeal malignant edema, acute
the vasculature. There are focal or multifocal areas of severe, often erysipelas, and classical swine fever.
diphtheritic, enteritis. Regional lymph nodes draining the area All cultures, contaminated materials, disposable equipment, and
are enlarged and reddened. Animals with intestinal anthrax often infected experimental animals and their feces should be inciner-
have pharyngeal lesions. ated. For carcass disposal, deep burial (greater than 4 feet deep)
In septicemic anthrax, carcasses may appear pale and dehydrated. followed by covering with lime is recommended. Any known or
There is bloodstained peritoneal fluid and scattered petechiation. suspected areas of contamination should be disinfected. Ten per-
cent sodium hydroxide, 5% formaldehyde, and 2% glutaraldehyde
are among the better disinfectants for B anthracis.
Diagnosis
Although bacteremia and septicemia are unusual findings in
swine infected with anthrax, it is advisable to take a peripheral
Control
An attenuated live vaccine against anthrax has been used for
blood sample aseptically for smears and culture. Smears can be
prevention in some species but is seldom used for swine since they
fixed in Zenker’s solution and stained with polychrome methy-
have enough natural resistance to withstand considerable exposure.
lene blue. Anthrax bacilli will appear as long, square-ended, blue Prevention in swine is usually by avoidance of heavy exposures
rods with a pinkish capsule. Alternatively, smears can be stained that might occur through ingestion of anthrax-infected carcasses
with Gram stain. The large Gram-positive, rod-shaped organisms or contaminated feeds. Laws now exist that prevent importation
have a “boxcar” appearance that is presumptive but not definitive of raw bone meal. Some states have laws that regulate the use of
of anthrax. Sometimes it may be necessary to biopsy or excise animal-origin products in feeds.
an affected lymph node for culture using sterile technique. Any
necessary cut on a suspected cadaver should be disinfected or Treatment of infected animals can be effective if given early. Both
plugged with a mass of cotton saturated with 10% formaldehyde. penicillin and oxytetracycline have been used effectively. Infection
can persist for up to 21 days.

ANTHRAX 15
SWINE DISEASE MANUAL
BACTERIA

Atrophic rhinitis
Alternate names and abbreviations most important being a dermonecrotizing toxin. Atrophic
Progressive atrophic rhinitis, PAR, AR, rhinitis rhinitis can be reproduced experimentally by exposure of pigs to
dermonecrotizing toxin, even in the absence of the P multocida
organism.
Definition
An infectious disease characterized in its early stages by sneez- Management and husbandry factors have long been suspected
ing, snorting, and perhaps nosebleed. Advanced cases will prog- of being important predisposing or contributing factors to AR.
ress to include atrophy and distortion of the nasal turbinates of Dust, ammonia, endotoxin from bacteria, and other organic or
affected pigs. chemical irritants may increase incidence and severity of AR.

Occurrence Epidemiology
Atrophic rhinitis (AR) occurs globally and only occurs in swine, Transmission of toxigenic B bronchiseptica and P multocida is
although a similar disease has been described in dogs and goats. through direct contact or short distance aerosolization between
Signs and lesions can occur in any age group. In the US, AR has
infected (sometimes inapparent) and uninfected pigs. Carrier
decreased in prevalence in most areas due to routine use of early
sows expose their piglets to the organisms very early in lactation
weaning and age segregation, which, when combined with all-in,
all-out flow, will result in elimination of the disease from most and when these infected piglets are mingled with others, the
farms. agents can be transmitted horizontally. Immune sows do provide
their piglets with some colostral immunity, but this immunity
wanes soon after weaning, leaving young pigs susceptible to
Historical information infection. Older sows may provide more colostral immunity to
Atrophic rhinitis was described first in 1830 but not actually
reported in the US until 1944. The condition rapidly increased their piglets than do gilts and may also shed fewer organisms.
in frequency with expansion and intensification of swine pro- Management or environmental factors, particularly inadequate
duction and stimulated a great amount of research; AR has a ventilation, increase the susceptibility of pigs to AR. Other con-
complex etiology. tributing factors include high concentrations of dust or ammonia,
overstocking, substandard housing or sanitation, and continu-
Etiology ous-flow production.
Toxigenic strains of Bordetella bronchiseptica and Pasteurella mul-
tocida (usually type D), may cause AR. Both organisms are aero- Pathogenesis
bic, Gram-negative rods that readily colonize the nasal passages The initial requirement for development of AR is colonization
of swine, and non-toxigenic strains are often widely distributed of the nasal passages by toxigenic strains of B bronchiseptica or P
in the swine population. Along with its association with AR, B multocida. Bordetella bronchiseptica adheres to the nasal mucosa,
bronchiseptica can cause or contribute to bronchopneumonia in probably by attaching to the ciliated epithelial cells. The toxin
suckling pigs or weaned pigs up to 8 weeks of age. Widespread elaborated by the organisms diffuses into the turbinates and is
endemic P multocida infections are more likely to be opportun- responsible for osteopathy and hypoplastic rhinitis. Later, after
ists, contributing to bronchopneumonia in pigs of any age. Both the disease becomes more quiescent, some degree of regeneration
organisms have been isolated from many species other than swine and repair can occur. The term “nonprogressive atrophic rhinitis”
(dogs, cats, rodents, rabbits, wildlife); however, most of those is sometimes used for this form of AR. However, when toxigenic
strains are not believed to play a role in causing AR in swine. P multocida colonizes the nasal mucosa, usually after mild rhinitis
Colonization of nasal passages with toxigenic B bronchiseptica has been established by B bronchiseptica, other agents, or envi-
can cause mild turbinate atrophy in pigs 1 to 6 weeks of age. ronmental irritants, the organism produces a potent toxin that
Lesions from B bronchiseptica tend to be mild and can usually causes a rhinitis with progressive osteopathy of facial and turbinate
be resolved by the host, therefore not leading to progressive AR. bones. The term “progressive atrophic rhinitis” (PAR) is some-
However, this insult or other sources of irritation to nasal mucosa times used to indicate this form of AR.
can enhance colonization with P multocida.
Pasteurella multocida is a ubiquitous early colonizer of pigs,
Clinical signs
commonly isolated from nasal cavity, tonsils, and lungs of pigs Signs may appear in pigs as young as 1 week old but more often
without signs or lesions of AR. However, toxigenic strains of appear shortly after weaning. Sneezing, snorting, and develop-
P multocida type D associated with AR are less widespread in ment of a serous or mucopurulent nasal discharge are early signs
the swine population. Colonization with toxigenic P multocida of AR. Rhinitis obstructs the flow of tears through the infraor-
type D alone can cause AR, but disease is enhanced when bital ducts. Tears therefore travel from the medial canthus of the
the P multocida type D is associated with B bronchiseptica. eyes along the skin. The dirty and discolored hair and skin result-
Toxigenic P multocida type D produces several toxins, the ing from this is commonly known as “tear-staining.” Signs caused
16 ATROPHIC RHINITIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
by B bronchiseptica alone tend to peak after a few weeks and then Control
diminish. Signs related to toxigenic P multocida often appear After AR has been diagnosed, the source of infection confirmed,
in 4- to 8-week-old pigs and may persist for weeks or months. additional risk factors identified, and the extent of losses consid-
Signs occasionally include nosebleed, and growth retardation ered, a decision should be made to either attempt to control the
may become apparent. Occasional pigs develop secondary bron- disease or to initiate some type of elimination program.
chitis, bronchiolitis, or pneumonia as an indirect effect of AR. A
variable number of pigs develop lateral or dorsal deviation of the If the decision is made to control rather than eradicate AR,
snout. Dorsal deviation often results in shortening of the snout control measures fall into 3 major categories. The first control
or wrinkling of the skin over the snout. Marked snout deviation measure is improvement of husbandry, management, housing,
in many pigs is usually associated with toxigenic P multocida. and ventilation. The second measure is initiation of a vaccination
program for the breeding stock, pigs, or both. Vaccination of
Lesions the sows may be most cost-effective. Sows are often vaccinated
Lesions are usually restricted to turbinates, nasal septum, and twice at about 4 and 2 weeks prior to parturition. Bacterin-toxoid
nasal and facial bones, and these lesions can be most easily vaccines provide some protection and historically have been
observed by sawing transversely across the snout at the level of widely used. The third control measure involves administration of
the second premolar (first cheek tooth in pigs less than 6 months antibiotics to sows and piglets around the time of farrowing and
of age), typically located at the commissure of the lips. Lesions again at weaning in an effort to limit the extent of colonization in
vary in severity and are usually graded on a scale of 0 to 5. Minor the young pigs. The value of antibiotic therapy to control AR in
lesions must be differentiated from normal variations in anatomy growing swine is questionable.
and breed character. Mild lesions usually result in atrophy of the Eradication of toxigenic P multocida from a herd or system has
ventral scroll of the ventral turbinates; severe cases are more likely been successfully achieved. One method for elimination involves
to involve the dorsal turbinate scrolls. In extreme cases, the dorsal total herd depopulation, thorough clean up and disinfection of
and ventral turbinates may be almost completely destroyed and facilities, and restocking with swine known to be free of AR. This
there may be atrophy of ethmoid turbinates. Deviation of the method is expensive. The second method does not require depopu-
nasal septum is more common in severe cases. Nasal and facial lation and involves staged use of several control strategies. First, the
bones may be thinned or distorted. Mucoid or mucopurulent herd should be closed to gilt introductions. Second, an intensive
exudate is often present on blanched mucous membranes and period of medication and pre-farrowing vaccination of sows should
in adjacent sinuses. Microscopic study usually reveals fibrous be implemented. Finally, piglets are medicated and weaned early
replacement of the bony plates of conchae that remain. to avoid colonization. Concurrently, nasal swabs for P multocida
and toxin testing by PCR can be used to identify the remaining
Diagnosis infected animals, which can then be culled (test and removal).
History, signs, and lesions are often adequate to make a clinical
Progress in control of AR should be evaluated by repeatedly
diagnosis of AR, with confirmation to follow by isolation and
examining nasal swabs from a large number of pigs submitted for
identification of the etiologic agent(s). Bacterial cultures made
slaughter for the presence of the offending agent(s). In addition,
from nasal swabs from live or sacrificed pigs are most rewarding
routine scoring of the severity of nasal and turbinate lesions from
during the early course of infection (first few weeks post-weaning).
snouts collected at slaughter may also be a useful monitoring tool.
Toxigenic P multocida can be determined by genotyping with
polymerase chain reaction (PCR) to confirm if the isolate has the
ability to elaborate dermonecrotizing toxin. Most toxigenic strains
will be P multocida type D, whereas P multocida type A organisms
are more likely to be isolated from bronchopneumonia. Both
organisms are common residents of the nasopharynx.
Differential diagnosis should exclude other diseases or irritants
that can cause rhinitis, such as porcine reproductive and respira-
tory syndrome virus, pseudorabies virus, inclusion body rhinitis,
or excessive dust or ammonia.

ATROPHIC RHINITIS 17
SWINE DISEASE MANUAL
BACTERIA

Clostridial diarrhea
Alternate names and abbreviations Five toxigenic types of C perfringens (A, B, C, D, E) are distin-
Sometimes referred to as enterotoxemia, clostridial enteritis, guished by the major toxins they produce (alpha, beta, epsilon, or
Clostridium difficile associated disease, CDAD iota). By definition, all C perfringens produce alpha toxin. Use of
polymerase chain reaction (PCR) to detect the genes coding spe-
Definition cific toxins has simplified their identification. Type C organisms
Disease associated with Clostridium perfringens and other clos- produce beta toxin, which cause the severe necrotizing enteritis.
tridia occurs in many different animal species. Systemic disease, Clostridium perfringens type A can produce a beta2 toxin and
including myositis, cellulitis, and gangrene, is uncommon in pigs, perhaps other less characterized toxins which may contribute to a
but described with opportunists such as C chauvoei, C septicum, much milder diarrhea than C perfringens type C. Virulent strains
and C sordelli. of C difficile produce an enterotoxin (TcdA) and a cytotoxin
(TcdB), which are associated with systemic illness in piglets as
In swine, diarrhea is caused by enterotoxemia associated with well as localized typhlocolitis and mesocolonic edema.
C perfringens type C, which affects neonates and suckling pigs
and causes sudden deaths, bloody diarrhea, or necrotic enteritis Epidemiology
with high mortality. Neonatal diarrhea associated with C per- Feces from carrier sows contain pathogenic clostridial organisms
fringens type A is reported with moderate morbidity and low and, coupled with a contaminated farrowing environment, are the
mortality. Clostridium difficile causes necrotizing typhlocolitis in likely source of exposure for piglets. The bacteria and its spores
suckling piglets with variable morbidity and mortality. survive in soil and can be carried on shoes, boots, or other fomites.
Each outbreak renews the supply of organisms and spores in the
Occurrence farrowing environment to infect the next litter of susceptible pig-
Although most outbreaks of C perfringens type C are acute and lets. Most piglets are colonized with C perfringens and C difficile
occur in piglets less than 1 week old, chronic cases may occur in within hours of birth, usually with no adverse clinical outcomes.
pigs up to several weeks of age. The disease is reported globally Although other species of animals have C perfringens type C infec-
but incidence varies greatly, depending on local geography and tion, there are very few reports of interspecies transmission.
management practices. Type C clostridial enteritis also occurs
in young lambs, calves, foals, and poultry, but less often in adult The magnitude of the dose and level of active or passive immu-
sheep, cattle, and goats. Diarrhea associated with C perfringens nity play a large role in disease occurrence. Immune dams (from
type A or C difficile is sporadic, but both are recognized con- natural exposure, perhaps augmented by vaccination) likely pro-
tributors to diarrhea in young pigs on farms around the world. vide antitoxin antibodies via colostrum to offspring. In contrast
Clostridium difficile also causes antibiotic-induced typhlocolitis to C perfringens, antibodies against C difficile toxins are not pres-
in humans and other species. Evidence to implicate pigs or pork ent in colostrum. Sanitation in farrowing rooms between groups
products as sources of these human infections is generally lacking. decreases the infectious dose in the environment.

Historical information Pathogenesis


Enterotoxemia in piglets associated with C perfringens type C was Clostridium perfringens and C difficile are ubiquitous early
reported, experimentally reproduced, and researched in the colonizers of the gastrointestinal tract and normally achieve
1960’s. Many early reports on C perfringens type C enteritis are high populations (greater than 10 million organisms per gram
from species other than swine. The application of molecular tech- feces) in the intestine and colon within 24 to 48 hours of birth.
nologies allows further characterization of toxins produced by Conditions that favor proliferation include absence of acid in the
Clostridium species and facilitates study and diagnosis of C per- stomach and the presence of antitrypsin factor.
fringens type A and C difficile, with particular emphasis on identi- Pathogenic C perfringens type C can attach to epithelial cells
fying the presence of genes associated with toxin production. of the proximal small intestine, proliferate, then elaborate
necrotizing beta toxin which causes acute and severe necrosis
Etiology and desquamation of epithelial cells. This is accompanied by
Clostridium perfringens and C difficile are large, anaerobic, hemorrhage and severe suppurative inflammation. Fermentation
Gram-positive bacilli with both vegetative and sporulated forms produces gas that accumulates in the lumen and/or wall (intes-
common in the intestine, colon, feces, and soil that have been in tinal emphysema) of necrotic sections of intestine. Death results
contact with livestock. Both C perfringens and C difficile are “early from mucosal necrosis and the systemic effect of toxins on other
colonizers” of newborn piglets, achieving high numbers within 1 organs. In older suckling piglets, clostridial colonization may be
to 2 days of birth. secondary to other agents of enteritis, most notably coccidiosis,
thus contributing to necrotic enteritis.
18 CLOSTRIDIAL DIARRHEA
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Pathogenesis is less clear with C perfringens type A, as disease has necrohemorrhagic small intestine with lesions most commonly
not been successfully reproduced experimentally. Novel toxins found in the proximal half of small intestine, though lesions can
(eg, beta2 toxin, NetB, or NetF) may have a role in initiating be observed occasionally in the lower intestine, cecum, or colon.
diarrhea but presently no clear mechanisms or virulence factors Gas and bloody fluid may be present in the intestinal lumen and
for C perfringens type A have confirmed roles in neonatal pigs. wall of the intestine with fibrinous adhesion between intestinal
Diarrhea is most often associated with exotoxins produced by C loops. Piglets that live a few days may have a necrotic pseudo-
perfringens and is less commonly a result of the endotoxins which membrane lining the mucosa of affected intestine. Microscopy
may also be present. reveals acute necrosis with severe suppurative inflammation and
robust rod-shaped bacteria in close association with or end-on
Similar to C perfringens type C and C perfringens type A, C dif-
attachment to enterocytes.
ficile colonizes neonates very quickly after birth, and under
ill-defined circumstances or at high doses, toxigenic strains can Piglets with diarrhea associated with C perfringens type A rarely
produce TcdA or TcdB toxins causing lesions in the cecum and present with any gross lesions. The small intestine may be thin-
colon. Mesocolonic edema and multifocal necrosis of epithelium walled and fluid-filled or contain milky content. Colonic content
with a flux of neutrophils spewing onto the mucosal surface is usually creamy. Microscopic lesions are quite mild. Accumula-
(“volcano lesion”) is the classic microscopic lesion. Nontoxigenic tions of neutrophils at villus tips, perhaps with focal enterocyte
strains of C difficile are widespread and are commonly found in necrosis or sloughing of apical enterocytes, is described from nat-
colonic contents in the absence of any clinical signs or lesions. urally occurring cases, but experimental reproduction of specific
Although C difficile typhlocolitis is a common sequel to oral anti- microscopic lesions has not been reported. Occasionally, sporu-
biotic therapy in humans, disease in piglets does not appear to be lating rods are detected in close association with enterocytes.
associated with misuse or overuse of antimicrobials.
Lesions of C difficile are usually confined to the cecum and colon and
include moderate to severe mesocolonic edema and watery or creamy
Clinical signs colonic contents. Microscopically there is erosion of the colonic
Clinical signs produced by C perfringens type C vary depending epithelium with infiltration and exudation of neutrophils (“volcano
on infectious dose, virulence, and quantity of antitoxin provided lesions”) as a common and nearly pathognomonic finding.
via colostral immunity. Susceptible neonates can become ill in
just a few hours after exposure to virulent C perfringens type C
and may be found dead in as little as 4 to 8 hours after birth, prior
Diagnosis
Acute C perfringens type C cases manifest with an acute, rapid
to onset of diarrhea (peracute form). Sick piglets soon become clinical course with variable-to-high morbidity, high mortality,
weak, prostrate, and moribund; weakened piglets have a higher and typical gross lesions in neonatal piglets. Demonstration
risk of being crushed by the dam. Piglets able to survive infection of a large population of Gram-positive, rod-shaped bacteria in
for a few days may have bloody diarrhea. Occasionally, piglets mucosal smears is supportive of the diagnosis, which is confirmed
may live several weeks and develop a yellow or gray, mucoid diar- by typical microscopic lesions that are nearly pathognomonic for
rhea, then tend to remain unthrifty with a chronic form of the C perfringens type C. Samples should be selected from acutely
disease; fibrinonecrotic enteritis is a common lesion in these pigs. affected, non-medicated pigs early in the course of disease. Con-
Morbidity is variable, but mortality can be high, up to 100%, in firming C perfringens type C in lesions by isolation, PCR, and
susceptible litters. toxin detection is useful.
In reports of uncomplicated cases of C perfringens type C or Criteria for diagnosis of disease associated with C perfringens
C difficile, clinical signs include mild, non-watery diarrhea and type A are less clear. Diarrhea is usually mild, mortality low, and
compromised growth rate. Morbidity is variable to high, but only mild non-specific gross and microscopic lesions are typically
mortality is generally low. Diarrhea produced by C difficile may present. High populations of C perfringens can be isolated from
be indistinguishable from that of C perfringens type A or neo- intestine and colon of normal suckling piglets. Detection by
natal colibacillosis. Morbidity is variable with all three agents, isolation or PCR is not sufficient to confirm a diagnosis of C
perfringens type A.
with some litters more severely affected than others. Mortality is
expected with C perfringens type C but not C perfringens type A. Diagnosis of C difficile is often made by observing typical diarrhea
Gross lesions are expected with C perfringens type C and C diffi- at 3 to 7 days of age, the presence of gross lesions of mesocolonic
cile but are not often grossly apparent with C perfringens type A. edema, and observation of typical microscopic lesions in cecum
and colon. Detection of TcdA and TcdB in colon contents or
Lesions feces by enzyme-linked immunosorbent assay is helpful. Isolation
Necropsy of piglets with C perfringens type C infection may of organisms can be helpful for confirmation and/or additional
reveal cyanosis or darkening of the abdominal skin. The peri- characterization.
toneal and pleural cavities may contain excessive, blood-tinged Anaerobic culture and isolation of Clostridium species or detec-
fluid. Typical acute cases have a dark red, segmental, or diffuse, tion by PCR are not particularly useful for diagnosis since these

CLOSTRIDIAL DIARRHEA 19
SWINE DISEASE MANUAL
BACTERIA

agents are early colonizers of many healthy neonatal pigs. How- caused by infection with C perfringens. Assuring adequate intake
ever, isolates and molecular techniques can be useful for further of colostrum and milk is essential to prevent endemic diarrheal
characterization of the organism once a diagnosis has been made. diseases. Natural exposure via proper gilt acclimatization aug-
A number of laboratory techniques are available to detect either mented by vaccination of dams with bacterin-toxoids is generally
toxins or toxin genes in intestinal contents isolates from typical helpful in enhancing maternal immunity. Commercial vaccines are
cases. PCR techniques are generally available to detect and dif- available for C perfringens type A and C, and autogenous products
ferentiate clostridial toxins of various species. Clostridial enteritis are sometimes used for C difficile and C perfringens type A as well as
must be differentiated from other neonatal diarrheal diseases, C perfringens type C. Two doses of vaccine, given at around 6 and
particularly coccidiosis, colibacillosis, or viral insults. 2 weeks prior to farrowing is a common practice in gilts. A single
booster vaccination given to sows a few weeks prior to subsequent
Control farrowings is often recommended in problem herds.
With outbreaks of C perfringens type C, immediate passive
immunity can be provided by administering immune antiserum Hygiene and management are critical control points in the
(antitoxin) orally or parenterally within 2 hours of birth. Pigs farrowing facility. Attention to details of management, environ-
acutely affected with C perfringens type C often do not respond ment, temperature, drafts, and hygienic practices is a constant
to treatment. Treatment of milder cases with antimicrobials is requirement. Breaking cycles of infection with all-in, all-out
commonly practiced. Prophylactic antimicrobials for C perfrin- farrowing and thorough washing and disinfection of the facility
gens type C, C difficile, and C perfringens type A can be an aid between groups is often sufficient to reduce the occurrence of
in reducing severity or prevalence of the disease. Some studies diarrhea in neonates.
suggest administration of oral probiotics shortly after birth may Antimicrobials fed to sows prior to farrowing is sometimes rec-
help in preventing cases of clostridial diarrhea, but studies docu- ommended by veterinarians to decrease shedding of clostridia
menting efficacy for this treatment are lacking. in feces from sows but credible data to support this practice is
Ensuring maternal passive immune protection for piglets via generally lacking.
colostrum and milk is a preferred method to prevent disease

20 CLOSTRIDIAL DIARRHEA
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Colibacillosis
Alternate names and abbreviations (exotoxins), endotoxins, and capsules. Fimbria are hair-like
Escherichia coli (E coli) diarrhea, neonatal scours, post-weaning processes on the bacterial surface that allow attachment to genet-
diarrhea, edema disease (ED), gut edema ically determined receptors on the surface of mucosal enterocytes
(colonization). Pathogenic strains also produce one or more
enterotoxins, which are exotoxins elaborated locally in the small
Definition intestine with either local or systemic effects. These pathogenic
The focus of this chapter is the colonization of intestine and
strains are termed enterotoxigenic E coli (ETEC). There are mul-
colon by different pathotypes of E coli, resulting in 3 primary tiple schemes in use for classifying E coli:
disease manifestations: diarrhea in neonates (neonatal colibacil-
losis), diarrhea after weaning (post-weaning colibacillosis), and Serotype: Based on the formula of the surface O (somatic or
enterotoxemia (also known as edema disease or ED). The latter polysaccharide), K (capsular or microcapsular), H (flagellar),
is an acute, often fatal enterotoxemia of recently weaned pigs and F (fimbrial) antigens that are present.
characterized by tissue edema, sudden deaths, and occasionally by Pathotype: Based on virulence mechanisms, such as enterotoxi-
genic E coli (ETEC), Shiga-toxin producing E coli (STEC),
neurologic signs related to lesions in the brain.
enterohemorrhagic E coli (EHEC), attaching and effacing
Important but not discussed here are sporadic outbreaks of septice- E coli (AEEC), enteropathogenic E coli (EPEC), and extrain-
mia and polyserositis caused by E coli in young pigs, as well as coli- testinal E coli (ExPEC).
form mastitis and urinary tract infections of sows and older pigs. Pilus type: Based on presence of antigenically distinct pilus
types, including F4 (K88, archaic), F5 (K99, archaic), F41,
Occurrence F6 (987P, archaic) and F18. The first 4 pilus types mediate
Colibacillosis and ED affect pigs in all major swine-raising coun- adhesion in neonates, while F18 and F4 are much more
common in post-weaning colibacillosis.
tries. There are many different strains of E coli, each of which
Toxin type: Based on the type of enterotoxins that are elabo-
may possess several of many potential virulence factors. Neonatal
rated. Toxins elaborated by ETEC strains include labile toxin
colibacillosis occurs from 1 to 7 days of age. Post-weaning coli- (LT), heat stable toxin A (StA), heat stable toxin B (StB),
bacillosis and ED tend to occur in older pigs from 3 to 7 weeks and shiga-like toxin (referred to variably as SLT, STx2e, vero-
of age. Risk factors and strain variation are often involved in out- toxin, edema disease principal, neurotoxin, or vasotoxin).
breaks of what are usually endemic infections with potentially The first 3 toxins act locally causing hypersecretion of fluid
pathogenic E coli. Genotype, nutritional factors, environmental from the intestine while shiga-like toxins are responsible for
stress, management type, herd immune status, and the presence the systemic vascular effects of ED.
or combination of specific virulence factors of E coli account for
the wide variation in the age of pigs that are affected as well as There are many strains and types of E coli present in swine and
their environment. Enteropathogenic E coli have fimbria-me-
the significance of clinical signs that are expressed.
diated adhesion to enterocytes as well as elaborate one or more
toxins. Other forms of colibacillosis may be caused by some of the
Historical information same pilus types that cause ED but have different pathogenesis
Colibacillosis has been recognized for nearly a century as an due to differences in the enterotoxins that are elaborated. Excep-
important diarrheal disease of pigs; ED is a unique form of coli- tions and variation like this are common with E coli.
bacillosis first reported in 1938. For many years, treatment and
control were largely empirical. Recurring losses, as well as the Epidemiology
importance of E coli in other species including humans, stimulated Potentially pathogenic E coli are global in distribution, present
research on the disease. Advances in molecular biology have sim- in the intestinal tract and feces of many normal swine, and are
plified identification of different pathogenic E coli, based on the generally able to survive for long periods of time in the environ-
presence of virulence genes as well as differences in surface antigens. ment. Transmission is usually through ingestion of feces directly
Escherichia coli strain O157:H7 is an important foodborne zoo- or from fomites. Airborne dust containing E coli can transmit
nosis in humans, however these strains in people do not appear to the organism over short distances.
cause disease in swine, nor is this serotype common in swine. Neonates are most often infected by ingestion of E coli shed from
immune carrier dams or the environment; very high populations
Etiology of the organism can be established in the intestine and colon
Pathogenic strains of E coli are Gram-negative, flagellated bacilli within hours of birth. The dominant strain(s) of E coli in a pig
easily isolated from non-medicated animals. Most pathogenic can change rapidly over time and with age. Continuous farrowing
strains form smooth to mucoid colonies on agar plates, many but accompanied by poor sanitation or chilling increases the risk of
not all of which are beta-hemolytic on blood agar. Numerous colibacillosis. Piglets lacking passive immunity from colostrum
virulence factors include variations of fimbria (pili), enterotoxins and milk are likely to be the first pigs in a litter to become ill from

COLIBACILLOSIS 21
SWINE DISEASE MANUAL
BACTERIA

colibacillosis. The dose of pathogenic coliforms is magnified by factors (effective temperature, humidity, sanitation, etc). Neo-
shedding from infected pigs to littermates. Disease occurrence natal piglets have immature immune systems and limited innate
and its severity are related to the infectious dose and the occur- resistance, and therefore are largely dependent on passive immu-
rence of other risk factors and is mitigated by the presence of nity acquired from colostrum and milk. Purchased breeding stock
effective passive or active immunity. may not have immunity to specific resident E coli and may have
Similar risk factors for neonatal colibacillosis (presence of carrier increased risk for disease in newborns because their colostrum
pigs, high levels of environmental contamination, infectious and milk may not contain enough antibodies to protect their
dose, pig genotype, weaning age, and environmental stress) also piglets. Chilling of piglets and weaners impairs intestinal motility
influence the occurrence of post-weaning diarrhea and ED. In and lowers resistance to infection. Massive exposure dose can
addition, nutrition and feeding management often play a role overwhelm resistance. During the early nursery phase of recently
in post-weaning colibacillosis and ED. Increase in the rate of weaned pigs, the absence of milk antibodies, transition to a plant
enterocyte turnover can lead to increased expression of receptors protein-based diet, cool or damp environmental stresses, weaning
post-weaning which is favorable for colonization with entero- stress, commingling, and other risk factors may contribute to out-
pathogenic E coli. Other contributors favoring colonization breaks of colibacillosis or ED.
include viral infections (eg, rotavirus) or diet (ration formu-
lation, processing type, high protein content, certain dietary Clinical signs
constituents) as well as poor hygiene and environmental control. Colibacillosis is usually signaled by the appearance of watery diar-
Disease-causing E coli tends to persist on farms that do not rhea, with piglets from gilts usually more severely affected than
provide adequate attention to good management of sanitation, piglets from multiparous sows. The severity of the diarrhea within
husbandry, and other risk factors. and between litters can vary. The hypersecretory diarrhea associ-
ated with colibacillosis usually has an alkaline pH and varies from
Pathogenesis clear and watery to white or yellow; the color is influenced by
Pili of ingested pathogenic E coli allow adherence to genetically type of ingesta and duration of the disease and is neither pathog-
determined receptors on microvilli of enterocytes in the jejunum nomonic for the occurrence of colibacillosis nor an indicator of
and ileum (and sometimes in the colon) where they then colo- disease severity. Vomiting is not as prominent as with coronavirus
nize, proliferate, and elaborate various enterotoxins that cause the infections. There is progressive dehydration and the hair coat
clinical signs. becomes roughened. Body temperature is often subnormal.
Shivering is often noted unless an adequate supplementary heat
Clinical diarrhea in neonates or post-weaning pigs is the result of source is available. Signs are similar in post-weaned pigs. Death
toxin-mediated hypersecretion of water and electrolytes by crypt losses can be severe if husbandry and environmental conditions
epithelial cells. This can result in up to 40% loss of the piglet’s are poor. Diarrhea tends to persist until successful intervention is
bodyweight over a few hours to days. Death is usually the result achieved.
of dehydration and metabolic acidosis, perhaps with terminal sep-
sis and endotoxemia. Some of the enterotoxins, endotoxin, and/ Edema disease usually occurs 1 to 2 weeks post-weaning, often
or adhesins may damage the microvilli and enterocytes, which signaled by finding a few acutely dead pigs that were previously
can reduce the absorptive capacity of the intestine and contribute known to be in excellent health. Morbidity is usually low, but
to even more severe diarrhea. In some cases, the large intestine mortality is high in pigs that show signs. Signs include anorexia,
can be affected similarly to the small intestine, further compro- ataxia, stupor, and recumbency, often accompanied by paddling
mising the pig’s ability to resorb water from the gut. Damage to and running movements. When caught, affected pigs may
gut epithelial cells sometimes leads to septicemia, and death is respond with an abnormal squeal, a consequence of laryngeal
usually the result of dehydration and metabolic acidosis or from edema. Diarrhea is not usually present in pigs with typical ED
terminal septicemia. but may be present in other pigs in the same group. Swelling
of the face and eyelids may or may not be present. Sick pigs die
Pathogenesis of ED is similar to that of neonatal diarrhea caused within a few hours or days. The few that survive often have neu-
by E coli but infection with F18 pilus is much more common rologic deficits. The course of ED in a herd is usually around 2
in ED (in the US) than with the F4 typically seen in neonatal weeks. However, the disease may reappear when other batches of
diarrhea. In Canada, ED is more commonly associated with F4 pigs reach the age group at risk.
than with F18 types. The shiga-like toxin produced locally in the
intestine is absorbed and has systemic effects on the endothe- Escherichia coli is one of the most common causes of neonatal
lium of blood vessels of multiple tissues, most notably areas of septicemia and polyserositis. Often, strains associated with septi-
the brain, eyelid, stomach wall, and mesentery. cemia are not enteropathogenic.

Whether piglets develop colibacillosis or ED depends on a balance Lesions


between dose, specific virulence factors of the organism, the pig’s With colibacillosis, dehydration is severe. The small intestine
innate and acquired resistance to the disease, and environmental and colon may contain excess watery fluid or may be distended

22 COLIBACILLOSIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
and gas-filled. There may be mild reddening and congestion small intestine from a non-medicated, typically affected pig
of the stomach. Lesions are often surprisingly mild, especially will yield high populations of hemolytic colonies of E coli. It is
in very young piglets. However, in outbreaks caused by certain useful to genotype the isolate for confirmation of the presence
pathogenic strains (usually in older post-weaned pigs), there can of genes that determine pilus type and potential for production
be marked congestion in the gastrointestinal tract. Microscopy of shiga-like toxin Stx2e. Typical microscopic vascular lesions
of the mucosa of the small intestine will reveal high numbers of help confirm diagnosis. Edema disease must be differentiated
coliforms adhered to microvilli of intestinal epithelial cells. Villi from other causes of neurologic signs (eg, pseudorabies, water
and enterocytes are usually intact. With some strains of E coli, deprivation, and organic arsenical poisoning) and septicemia and
there may be necrosis of some villi and microvascular thrombosis meningoencephalitis caused by Streptococcus suis, Haemophilus
in the lamina propria. parasuis, and others.
Lesions of ED vary between individuals. In pigs found dead, Investigation of outbreaks of colibacillosis should include iden-
visceral congestion may be prominent. Usually, some will have evi- tification of any risk factors such as chilling, poor sanitation, or
dence of subcutaneous edema of the face or eyelids, gastric submu- starvation that may be contributing to the disease. These must be
cosa, or in the mesentery of the spiral colon. There may be slightly corrected if prevention or treatment is to be successful.
increased volumes of clear fluid in the peritoneal cavity, chest, or
pericardial sac. Occasionally, there are hemorrhages under the epi- Control
cardium or endocardium. In subacutely or chronically affected pigs, Escherichia coli that cause clinical colibacillosis or ED are often
microscopic degenerative changes can be found in small arteries endemic on farms, with disease expression most influenced by
and arterioles at many sites. The changes are sometimes apparent changes to the occurrence of non-infectious risk factors. For coli-
in vessels supplying the brain. Histologic sections of brain stem bacillosis, the use of all-in, all-out (AIAO) systems of farrowing
and basal ganglia may confirm suspected areas of hemorrhage and and raising piglets is recommended. Farrowing should occur in a
malacia, but these lesions are not consistently present. facility that has been thoroughly cleaned, disinfected, and dried
Escherichia coli is a common cause of septicemia in neonates, between farrowing groups, along with an on-going sanitation
characterized by fibrinous polyserositis and arthritis. program and internal biosecurity measures that prevent pathogen
build-up. The farrowing facility should be designed to provide
a dry, comfortable, draft-free environment for piglets as well as
Diagnosis accommodate dam comfort. This requires lower temperatures
The typical clinical signs, dehydration, and subtle lesions
(approximately 70°F [21°C]) for sows, areas warmed (up to
described above are useful for diagnosing colibacillosis but are
approximately 90°F [31°C]) for small piglets, and the use of hus-
not definitive. The isolation of a uniform and high population of
bandry practices that minimize stress.
smooth, mucoid, or hemolytic E coli from the small intestine is
suggestive of colibacillosis or ED in typically affected neonates It is wise to obtain breeding stock from a single source that does
and post-weaned pigs, with the hemolytic colony type more not have a historical problem with colibacillosis. Dams should
likely in post-weaned pigs. As E coli are commonly found in the be acclimatized together for 3 to 6 weeks prior to breeding so
small intestine of both normal and diseased pigs, microscopic they can develop immunity to endemic potential pathogens that
examination of sections of small intestinae is very useful in deter- exist on the farm. This will encourage production of high-quality
mining the significance of the infection. Dense colonization colostrum that contains an adequate concentration of specific
of the brush border with adherent E coli should be expected in antibodies directed at the E coli present on that farm. Enhance-
most cases of colibacillosis, with additional erosive or congestive ment of sow immunity is effective with commercially available
changes present depending on the toxins that have been elab- vaccines made from bacterial pili, or toxins, or both. Pregnant
orated. Tissue samples should be immediately collected from gilts are usually vaccinated twice at 2- to 3-week intervals prior
acutely affected, non-medicated, euthanized pigs and preserved to farrowing; sows need a single booster vaccination 2 to 3 weeks
in formalin. prior to farrowing. Immediate treatment of piglets is warranted
should signs of colibacillosis appear. Antimicrobials can be
Polymerase chain reaction (PCR) assays are available to identify
administered to neonates orally or by injection. Because the
the presence of pathogenic E coli. Genotyping by multiplex PCR
disease is contagious and sick piglets shed high numbers of organ-
is widely practiced in order to identify specific pilus and toxin
isms, it is wise to treat all pigs in the litter at the same time.
genes present in the isolate. Colibacillosis must be differentiated
from other diarrheal diseases of young pigs. Important differ- For post-weaning colibacillosis and ED, attention to details of
entials include transmissible gastroenteritis, porcine rotavirus, husbandry and management is essential. No prevention method
coccidiosis, and Strongyloides parasitism. Starvation is also a major is universally accepted or successful, because the erratic occur-
differential diagnosis. rence of colibacillosis and ED makes intervention strategies hard
With ED, the history, signs, and lesions are often sufficient for to assess. General strategies to consider include:
establishing a presumptive diagnosis. Bacterial culture of the

COLIBACILLOSIS 23
SWINE DISEASE MANUAL
BACTERIA

1. Minimizing environmental stress (eg, temperature variation, 5. Routine administration of antimicrobials in feed or water is
drafts, damp conditions), limiting commingling, and using often practiced but not desirable, as the practice is expensive
AIAO flow with scrupulous sanitation and disinfection and can contribute to development of antimicrobial resistance.
between groups. 6. Natural resistance to colibacillosis and ED is possible in pigs
2. Nutritional considerations include the use of creep feed- genetically lacking receptors for F18 and/or F4 pili. Genet-
ing, restricted feeding, multiple feedings (small quantities ically resistant pigs have been developed. In herds negative
fed 3 to 6 times per day) after weaning, increasing fiber in for ED, it is prudent to prevent introduction of the disease
the rations, adding acidifiers to feed or water, and adding by careful scrutiny of the disease history of breeding stock
zinc oxide or plasma proteins to the ration. Probiotics are that are being considered for purchase. Elimination of the
perceived by some to have benefit during the transition to clinical disease may be possible by using depopulation and
grain-based diets. Avoid abrupt dietary changes, including disinfection.
ingredient changes, and use step-wise transition diets when 7. Should diarrhea occur, weanlings may be given antibiotics in
making changes. water with the antimicrobial selection based on sensitivity
3. Passive or active immunoprophylaxis can be very helpful on data. Oral electrolyte replacement solutions are sometimes
farms known to have a colibacillosis problem. Oral products used to help treat dehydration and acidosis. Treatment of
specifically formulated to contain high levels of antibody pigs affected with ED can be frustrating since toxin pro-
(milk, plasma protein, egg powder) can help to prevent duction (and clinical signs) is generally too far advanced for
colonization and disease. Active immunity can be induced effective treatment by the time clinical signs become visible.
against specific adhesins or toxins. Oral vaccination with live During an outbreak of colibacillosis or ED, control efforts
or killed cultures that contain F18 or F4 pili (but lacking are better directed toward reducing the incidence of infec-
genes for toxin production) have been used successfully on tion in the remainder of the population at risk by adminis-
some farms. Toxoids prepared from Stx2e have also been tering antimicrobials to the unaffected pigs and including
demonstrated to be effective against ED but are not commer- acidifiers in the feed or water. In severe outbreaks, removal
cially available. While effective as a vaccination for pregnant of the existing feed with replacement by a ration containing
sows, administration of killed bacterins to newborn, nursing, less protein, a lower percentage of soybean meal, or higher
or post-weaning pigs has been much less effective. proportion of fiber may be beneficial.
4. Competitive exclusion may be an effective mechanism
whereby receptors for pathogenic E coli are occupied by
avirulent agents with products fashioned similar to avirulent
live vaccines.

24 COLIBACILLOSIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Erysipelas
Alternate names and abbreviations In addition to these inapparent carriers, affected pigs also shed
Swine erysipelas, diamond skin disease, human erysipeloid large numbers of organisms that further contaminate feed, water,
soil, and bedding. Contaminated soil probably does not remain
Definition infective for more than 35 days, but organisms can flow in surface
Erysipelas is an infectious disease that can affect swine of any age, water to adjacent premises, and a wide range of wild mammals,
though it is most common in growing or adult swine. Infection poultry, wild birds, and pets can then further disseminate the
may result in acute sepsis, chronic arthritis, endocarditis, condem- bacteria. There is empirical evidence that biting flies and ticks
nations at slaughter, or be subclinical. can sometimes transmit erysipelas between swine, though it is
unlikely they transmit the infection between farms. Erysipelas
Occurrence tends to have a seasonal occurrence with spikes during the hot
Erysipelas occurs in swine, lambs, turkeys, occasionally in summer months. Eradication of erysipelas from a herd is unlikely.
other poultry and wild birds, and sporadically in most kinds
of livestock, wild mammals, reptiles, and amphibians. Seasonal Pathogenesis
distribution is suspected, with losses greater during warm summer Variation in virulence between isolates of E rhusiopathiae is
months. Occasionally, people who work with infected animals, attributable to the presence and expression of various virulence
poultry, fish, or animal by-products can become infected through factors. In general, ingestion of contaminated feed and water
skin wounds and develop a localized skin condition referred to as allows E rhusiopathiae to gain access to the body, probably
an erysipeloid rash though an identical condition can be caused through the tonsils or other lymphoid tissue of the digestive
by other infections as well. tract. Less often, contaminated skin wounds may permit organ-
ism entry. Bacteremia and sepsis results in localization of E rhu-
In humans, the condition referred to as “erysipelas” is actually
siopathiae at various sites throughout the body.
caused by a streptococcal infection.
The precise mechanisms by which E rhusiopathiae causes disease
Historical information are speculative. The organism wall and capsule along with several
In 1885, Dr. Theobald Smith isolated an organism from swine in proteins it produces, appear to mediate the widespread vascular
the US that was similar to one Pasteur had isolated 3 years earlier damage that accompanies erysipelas and allow for persistence
from swine affected by a condition called rouget du porc; both were in tissues. Vascular damage leads to thrombosis and interference
eventually identified as Erysipelothrix rhusiopathiae, the causative with microcirculation in capillaries and venules.
agent of swine erysipelas. Some of the earliest recognized outbreaks In chronic proliferative arthritic cases, there is a vasculitis with
of erysipelas were reported from South Dakota in 1928 and led to exudation of fibrin into perivascular tissues and joints, causing
greater awareness, increased recognition of the disease, and devel- marked villous proliferation of the synovial membrane. Con-
opment of bacterins. However, erysipelas remains a constant threat nective tissue forms around joints, accompanied by perivascular
in both confinement and outdoor swine farms. fibrin and thickening of the joint capsule. Pannus may develop,
accompanied by destruction of articular cartilage and ankylosis of
Etiology adjacent bones. Persistence of inflammation around an affected
Erysipelothrix rhusiopathiae is a Gram-positive, aerobic, slightly joint may be from a few living bacteria in the joint or it may be
bent, thin bacillus. It is resistant to many environmental condi- the result of hypersensitivity to remaining bacterial antigens.
tions and survives for months in meat, cured hams, fish meal, Vegetative valvular endocarditis is another chronic lesion asso-
and decaying animal carcasses. There are at least 28 serotypes of ciated with erysipelas. Emboli of E rhusiopathiae may cause
E rhusiopathiae, of which swine are susceptible to at least 15. In inflammation of blood vessels within heart valves. During the
the US, most isolates from sick pigs are serotype 1a, followed inflammatory process, the endocardium is breached and bacterial
by 2, 5, and 1b. Non-pathogenic species (E tonsillarum) are fre- colonies are established at the site. Exuded fibrin deposits are
quently isolated from normal swine. The organism is sensitive to slowly organized to form nodular vegetations. Emboli that arise
penicillin and usually to tetracyclines. It resists many antibiotics from vegetations can cause sudden deaths. Vegetative endocarditis
and sulfonamides. It is susceptible to most disinfectants. can occur without clinical signs.

Epidemiology Susceptibility to infection is not well understood. Many swine


Worldwide in distribution, E rhusiopathiae is assumed to be spread that are exposed to organisms do not develop disease. Lactogenic
by inapparent carriers within and between herds. It has been immunity protects young piglets for several weeks. In older,
estimated that over half of healthy swine are carriers of pathogenic susceptible swine, various stresses (heat, aflatoxin, poor nutrition)
and/or non-pathogenic Erysipelothrix species and can dissemi- are suspected to play a role in precipitating outbreaks.
nate the organism through their feces and oronasal secretions.

ERYSIPELAS 25
SWINE DISEASE MANUAL
BACTERIA

Clinical signs sloughed. Vegetative valvular endocarditis of chronic cases usually


Clinical signs of erysipelas may be acute, subacute, chronic, or affects the mitral valve and may appear as irregular masses of con-
inapparent. Acute outbreaks of erysipelas are manifestations of siderable size. Such cases often have infarcts in the spleen, kidneys,
sepsis, high fever (up to 42°C/108°F), and vascular damage in or other sites as a result of embolism from valvular lesions.
various tissues. Clinically, there may be acute deaths. Affected
pigs have fever, cyanosis, stilted gait due to painful joints, and Diagnosis
often vocalize in pain when moved. These signs may be accompa- Clinical diagnosis is based on the appearance of typical clinical
nied by the classic skin lesions which are pink-to-purple, raised, signs and lesions in multiple pigs; favorable response of acute
and rhomboid or diamond-shaped. Young pigs (2 to 8 weeks of cases to high doses of penicillin and/or hyperimmune serum also
age) may have profound cyanosis of ears, snout, and extremities. helps to confirm a presumptive diagnosis of erysipelas. Acute
Pregnant sows may abort from the effects of acute sepsis or from cases usually have exceptionally high temperatures, evidence of
transplacental fetal infection. marked pain in multiple joints, and typical diamond-shaped skin
lesions. Diagnosis can be confirmed by bacterial culture or poly-
The subacute form of disease is viewed as simply a less severe
merase chain reaction of kidney, spleen, skin, lung, lymph nodes,
illness than the acute form. The classic cutaneous diamond skin
blood, or other tissues. Isolation of E rhusiopathiae is more likely
lesions may be observed on pigs that are otherwise clinically
from acute cases; failure to recover the organism should not rule
normal. Cutaneous lesions may coalesce over the rump, back, or
out a diagnosis of erysipelas. Acute erysipelas must be differenti-
shoulders, and occasionally patches of affected skin may become
ated from classical swine fever, other causes of acute septicemia
necrotic. Sloughing of the tip of the tail or tips of the ears can
(eg, salmonellosis, streptococcal infection), and other causes of
occur with erysipelas but these lesions can also have other causes.
dermatitis. Actinobacillus suis is reported to cause rhomboid cuta-
The chronic forms of erysipelas may either follow acute and sub- neous lesions that resemble those of acute erysipelas.
acute forms or develop insidiously. Chronic erysipelas is manifested
Chronic erysipelas can be challenging to diagnose. Culture of
usually as either chronic arthritis or chronic vegetative valvular
multiple joints or valvular endocarditis lesions does not often
endocarditis. With the former, there little mortality but rather a
result in isolation of E rhusiopathiae. However, in situations
gradual enlargement of leg joints, most noticeable in the hock and
where these lesions are occurring in appreciable numbers, careful
carpus, observed in an increasing number of pigs; affected pigs tend
examination of the rest of the pigs in the herd will likely result
to become unthrifty as the disease persists. With chronic vegetative
in identification of at least a few pigs periodically showing more
valvular endocarditis, pigs often present few signs. However, when
acute clinical signs and/or diamond-shaped skin lesions. This
these pigs are exerted they may show signs of acute respiratory dis-
is often accompanied by a history of change (or absence) of
tress which occasionally triggers collapse and sudden death.
vaccination for erysipelas. It is important to remember that both
arthritis and vegetative, valvular endocarditis can be caused by
Lesions many different organisms, most notably Streptococcus suis.
Pigs that die of acute erysipelas may present as sudden deaths
without the appearance of any gross lesions. Cyanosis or dia-
mond-shaped skin lesions may be apparent. Necropsy may reveal
Control
Acute outbreaks of erysipelas can usually be controlled by injec-
congested or edematous lymph nodes and perhaps subcapsular
tions of penicillin and/or erysipelas antiserum into affected pigs
hemorrhage if the animal lived a few days after onset. Petechial
with simultaneous administration of appropriate antimicrobials
hemorrhages may be present on the kidneys, epicardium, and
via the drinking water. Vaccination in the face of an outbreak has
endocardium. The liver is often swollen and lungs may be con-
merit in reducing future cases.
gested and edematous. The spleen may be enlarged, especially in
cases that survive beyond a few days; it may occasionally contain A vaccination program is advisable for farms on which outbreaks
infarcts. Microscopic lesions include vasculitis in capillaries and have occurred previously or where exposure is expected. Killed
venules at many lesion sites, including glomerular tufts, pulmo- or attenuated live vaccines are available. Most now have a dura-
nary alveolar walls, and skin. Bacteria may be apparent within tion of immunity of 26 weeks and are efficacious across a broad
microthrombi in blood vessels. spectrum of serotypes. Killed vaccines are given by injection
whereas attenuated live vaccines are given either by injection or
With chronic joint disease, there is arthritis with proliferative,
oral administration. Two doses of bacterin will usually protect
villous synovitis in one or more swollen joints. Synovial fluid
growing pigs to market weight. Boosters may be necessary in
is viscous and only modestly increased in amount. The joint
some herds, especially those with a history of repeated exposure
capsule may be visibly thickened. An organized inflammatory
to the organism. Erysipelas rhusiopathiae is a common constitu-
exudate (pannus) may be present in affected joints, sometimes
ent of pre-breeding vaccines that also include porcine parvovirus
with ulceration of the articular cartilage or ankylosis of adja-
and Leptospira. A combination of regular vaccination, good
cent bony surfaces. Chronic cutaneous lesions may appear as
sanitation, the elimination of carriers with lesions, and appropri-
one or more patches of dry, necrotic skin. Sometimes there are
ate quarantine measures for purchased stock will usually aid in
granulating, healing areas where patches of necrotic skin have
control of erysipelas.

26 ERYSIPELAS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Greasy pig disease
Alternate names and abbreviations differences in strain virulence or variations in animal immune
Exudative epidermitis status are important in the epidemiology of the disease. However,
some outbreaks do occur that are not related to introduction of
Definition new animals, suggesting that other risk factors are likely involved.
An acute, exudative dermatitis affecting pigs from a few days to The organism is hardy and may persist in facilities, particularly
about 8 weeks of age, characterized by variable to high morbid- those with poor hygiene and high humidity. Important risk
ity and variable mortality. The causative organism can contribute factors for exudative epidermitis include management practices
to skin infections in older pigs as well as sporadic cases of sepsis or environmental conditions that are associated with frequent
in pigs of any age. breaches to the skin epithelium. Sources of skin trauma and
abrasions are numerous and include frequent commingling
Occurrence events, large litter sizes (or cross-fostering techniques) that create
Exudative epidermitis occurs in most major swine-raising coun- competition for nipples, facility design or maintenance issues
tries. It is a sporadic disease that is occasionally seen by most that result in cuts or abrasions, and the use of caustic compounds
swine practitioners. Although acute exudative epidermitis usually or disinfectants that cause skin irritation. Other possible factors
occurs in pigs less than 8 weeks old, the disease also occurs in a for exudative epidermitis expression include: viruses causing
less severe form in older pigs. It seldom occurs in adult swine. transient vesicles (eg, certain strains of parvovirus, poxvirus, or
members of Picornaviridae), nutritional deficiencies (eg, zinc or
Historical information vitamins), ringworm, pityriasis rosea, parasitism (including lice
Exudative epidermitis was described over 150 years ago and infestation), lack of competing bacterial flora on the skin, and
Staphylococcus hyicus, the etiologic agent, was first identified genetic susceptibility.
1965. Despite research, the complete etiology of exudative epi-
Bacteremia and sepsis that may occur along with exudative
dermitis is unclear.
epidermitis may occasionally cause polyarthritis and serositis in
young pigs or abortion in sows.
Etiology
Staphylococcus hyicus is a hardy, Gram-positive coccus. Staphy-
lococcus hyicus may normally be present at many sites, including
Pathogenesis
Initial skin lesions are likely toxin mediated, resulting in desmo-
skin, nasal mucosa, vagina, and prepuce. At least 6 serotypes have
some lysis and focal erosions in the stratum granulosum that often
been identified. Experimentally, inoculation of pigs with cultures
form intracorneal pustules laden with bacteria. Lesions extend
of certain strains of S hyicus, as well as bacteria-free culture super-
into hair follicles as suppurative folliculitis. Sebaceous glands
natant, is able to induce exudative epidermitis in pigs. At least 6
secrete excessive amounts of sebaceous, greasy exudates that accu-
exfoliative exotoxins have been identified but not all are present
mulate over the dermatitis lesions. Severe epidermal erosion and
in every isolate; these toxins target cells of the stratum granulosum
ulceration follow with formation of surface cracks, fissures, and
in the epidermis.
ulcers that enlarge and may coalesce. Systemic effects of dehydra-
tion with loss of serum proteins and electrolytes occur.
Epidemiology
Most herds (and pigs) are endemically infected with S hyicus, but
exudative epidermitis seems to occur only as a sporadic, opportun-
Clinical signs
Listlessness and anorexia in some or all piglets in a litter, in the
ist disease in individual pigs. However, on occasion exudative epi-
absence of fever, are early warning signs of the disease. The skin in
dermitis can occur as an epidemic within certain age cohort groups.
the axilla and groin is often reddened and non-pruritic, but these
Vertical transmission of the organism occurs within 24 hours of lesions may not be noticed until brownish spots (starting as 1 to
birth, with further lateral transmission occurring from pig to pig. 2 centimeter in diameter) covered by serum and exudate appear
Maternal immunity is likely important in preventing the disease on the skin of the face or head. In some cases, the lesions will be
in pigs less than 8 weeks of age. When they occur, outbreaks of resolved without treatment but in most cases, the lesions enlarge
exudative epidermitis are usually self-limiting within 3 months as and coalesce then spread posteriorly with a generalized dermatitis
a function of herd immunity developing. becoming apparent in as few as 24 to 48 hours. Although initial
lesions usually affect haired areas, ulcers may develop on the
Under experimental conditions, only certain strains of S hyicus tongue or in the mouth.
can colonize the skin and cause disease. The epidemic form of
exudative epidermitis tends to occur in start-up herds or follow- Young pigs can die within 3 to 5 days. Overall morbidity and
ing introductions of new breeding stock to a herd, suggesting that mortality are usually high in younger pigs. Pigs older than about

GREASY PIG DISEASE 27


SWINE DISEASE MANUAL
BACTERIA

8 weeks may have only a few lesions and these may be mostly during the very early stages of the disease. As much of the pathol-
confined to the head. These older pigs often survive but tend to ogy is related to the effect of bacterial toxins rather than simply
gain weight slowly. Adult swine occasionally have a few brown bacterial colonization, antibiotic therapy often has little effect in
exudative lesions on the back and flanks but infections in this age advanced cases. Topical application of antimicrobials or solutions
group are uncommon and often unrelated to known outbreaks in (as approved in specific countries) such as 10% bleach, chlor-
younger pigs. The adult lesions can easily be confused with focal hexidine, Virkon, or dilute tamed iodine have been utilized to
areas of ringworm. some effect. Affected litters or individual pigs should be isolated
immediately and commingling of pigs should be avoided if an
Lesions outbreak has occurred. If an outbreak is well established in small
Affected pigs are usually dehydrated and malodorous, with exten- pigs before diagnosis, it may be prudent to euthanize those with
sive gross lesions on the skin. Fissured dark crusts involve most advanced lesions. Any that recover will gain weight inefficiently.
haired areas, with the head and neck most often affected, though
Prevention of exudative epidermitis starts with providing a high
feet, body, and occasionally the mouth and tongue may also be
standard of sanitation for pregnant sows, especially in farrowing;
affected. Peripheral lymph nodes draining areas of infected skin
washing of sows prior to entry into the farrowing house is a good
are swollen. Microscopically, there is a pustular dermatitis largely
practice.
in the stratum corneum and a suppurative folliculitis; suppurative
lymphadenitis may occur in enlarged peripheral lymph nodes. Control of exudative epidermitis is largely dependent on pre-
venting trauma to the skin and improving environmental hygiene
Diagnosis through good ventilation, cleaning, drying, and disinfection
Clinical signs and gross lesions are adequate for diagnosis, with between groups, all-in, all-out pig flow, low humidity, and
confirmation by isolation of S hyicus or by histopathology. Mild avoidance of high stocking densities. Vaccination with an autoge-
cases require differential testing for other skin diseases. nous bacterin of S hyicus has been reported, but there is little
information regarding the efficacy of this procedure. In addition,
Control outbreaks of exudative epidermitis often develop so rapidly that
Consistently efficacious therapies have not been reported, an autogenous bacterin cannot be prepared and administered in
although injectable antimicrobials are considered beneficial time to have any beneficial effect.

28 GREASY PIG DISEASE


DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Haemophilus parasuis
Alternate names and abbreviations is not reliable despite descriptions of at least 48 putative virulence
HPS, Glässer’s disease, Glaesserella parasuis factors. Virulence appears to be related to capsular antigens but is
not necessarily related to immunogenicity.
Definition
A common acute infectious disease of pigs, characterized by Epidemiology
various combinations of meningoencephalitis, polyserositis, and Haemophilus parasuis is widely distributed in swine populations
polyarthritis as well as a contributor to bacterial pneumonia. throughout the world; the host range is limited to swine. The
organism is considered part of the normal respiratory microbiota,
Occurrence often with multiple strains being present simultaneously in a herd
Glässer’s disease is caused by Haemophilus parasuis and occurs and in individual pigs. Some, but not all, suckling piglets are nat-
sporadically among swine, usually in pigs between 3 weeks and urally exposed from the dam and will become colonized within
4 months of age. It often occurs subsequent to stressful events days of birth.
such as weaning, environmental stressors, commingling, or as a Colostral antibodies usually protect pigs from disease during
coinfection with other disease agents. Glässer’s disease is present the period of time required for the colonized pigs to develop an
in all major swine-raising countries and is common in modern active immunity, usually by 7 to 8 weeks of age. However, piglets
age-segregated production systems. colonized later in life, perhaps during commingling events or
with different strains, may not have sufficient or specific passive
Historical information protection at this point. Therefore, some pigs in a group will tend
In 1910, Glässer observed a unique swine disease that was associ- to become infected through this horizontal transmission and
ated with a small, Gram-negative rod. In 1931, an organism was develop fulminating disease. Unusual stresses such as weaning,
isolated from pigs with influenza and was named Haemophilus prolonged transport, or the occurrence of other diseases may
influenzae suis. By 1943, it was clear that the organism was a predispose to outbreaks.
pathogen in its own right and not the cause of influenza in swine
and therefore the name was shortened to Haemophilus suis. By Haemophilus parasuis is quite labile outside of the pig and does
1976, taxonomic studies had been completed that resulted in the not persist in the environment, particularly with modern hygiene
organism’s present name, Haemophilus parasuis. The importance practices such as all-in, all-out pig flow.
of Glässer’s disease has increased with expansion of the swine
raising industry. Pathogenesis
Intranasal inoculation of susceptible pigs with virulent strains
results in colonization and suppurative rhinitis. Haemophilus
Etiology parasuis can then be picked up by macrophages and/or through
Haemophilus parasuis is a small, pleomorphic, fastidious,
lymph drainage and be distributed systemically (septicemia) to a
Gram-negative rod of the family Pasteurellaceae. It grows on variety of internal organs and epithelial surfaces. Highly virulent
blood agar as “satellite” colonies near a streak of Staphylococcus stains can persist in blood and resist clearance by macrophages,
organisms, an alternative source of nicotinamide adenine dinu- thereby enabling them to access and colonize serosal surfaces,
cleotide (NAD), or on enriched chocolate agar. Haemophilus synovium, and meninges. The resulting tissue colonization and
parasuis is a common inhabitant of the nasopharynx in healthy local tissue damage results in classic Glässer’s disease with severe
pigs. It causes disease when it becomes septic or when it infects inflammation and exudation.
lung tissue, the latter frequently as a coinfection with other dis-
ease processes. As a septicemia, H parasuis has predilection for Haemophilus parasuis has a predilection for the leptomeninges
and brain, and often localizes there. This commonly stimulates a
colonization of serosal surfaces (peritoneum, pleura, epicardium),
severe inflammatory reaction in the meninges. Damage to endo-
synovium, and/or meninges.
thelium probably facilitates formation of inflammatory exudate
At least 15 major serovars have been identified with many addi- and increased synovial fluid in joints as well.
tional serovars, including untypeable strains, also common. Herds, Haemophilus parasuis can also colonize lower respiratory tract
groups of pigs, and even individual animals often harbor several and when armed with the necessary virulence factors, can survive
different serovars of H parasuis simultaneously. Serovars 4, 5, 13, destruction by pulmonary alveolar macrophages and contribute
and 14 are the most prevalent in North America. Cross-protection substantially to suppurative bronchopneumonia that may have
may occur among serotypes but is incomplete. Molecular charac- been initiated by other pathogens.
teristics of isolates differ, depending on whether they have been
The severity of disease expressed depends on virulence factors
isolated from systemic sites or from the respiratory tract. Prediction
contained in the particular strain of H parasuis, the immune
of virulence or cross-protection based on genotyping information
status of the pig, the presence of other pathogens, and myriad risk
HAEMOPHILUS PARASUIS 29
SWINE DISEASE MANUAL
BACTERIA

factors or stressors. Haemophilus parasuis can be a primary patho- aspect of the brain. In advanced cases, the exudates may be present
gen acting alone. However, disease often occurs in conjunction over the cerebellum or cerebrum. The atlanto-occipital joint is
with other diseases such as porcine reproductive and respiratory frequently affected.
syndrome virus (PRRSV), porcine circovirus type 2 or influenza
A virus. The lesions or stress associated with concurrent diseases Less common and less specific findings of Glässer’s disease
likely enhances access of the organism to the blood or compro- include congestion of the fundus of the stomach and petechial
mises innate or acquired immune mechanisms, thus allowing hemorrhages on the kidneys. Microscopic lesions are usually
disease expression. acute, suppurative to fibrinopurulent, with suppurative infiltrates
in meninges, pleura, serosa, and synovium common findings. Vas-
Clinical signs culitis and vascular thrombi are occasionally recognized.
Onset of typical Glässer’s disease is usually sudden, often affecting
the most robust pigs in a group first. They may be found dead or Diagnosis
die after a disease course of less than 2 days. The sudden deaths A presumptive diagnosis can often be made on the basis of his-
may occur in any age group and sometimes there are signs sugges- tory, clinical signs, and the appearance of typical gross lesions.
tive of septicemia or myositis. The diagnosis should be confirmed, however, by demonstrating
the presence of H parasuis within lesions. Samples of choice for
Clinical signs depend upon where the organisms localize. Signs culture include systemic locations, while avoiding the nasophar-
of central nervous system disturbance often predominate as a ynx and lung. Samples from these sites may include fibrin or
consequence of localization of H parasuis in the brain, meninges, swabs from serous membranes, meninges, synovium, cerebro-
and spinal cord. Signs may then include tremors, incoordination, spinal fluid, or other affected tissues. Detection of the organism
posterior paresis, or lateral recumbency. In most cases, there is can be achieved by culture or polymerase chain reaction. Culture
an initial, marked rise in temperature, along with anorexia and has the advantage of being able to determine the antibiotic
depression. In some outbreaks, swollen leg joints, fever, reluctance susceptibility on the resulting isolate. In addition, the isolate is
to move, and lameness are common and may even predominate. also then available for further characterization or production of
Less frequent signs include rhinitis, dyspnea, reddening of the autogenous bacterins. Isolates must be differentiated from other
conjunctiva, cyanosis of the extremities, and edema of the eyelids Haemophilus-like organisms, especially Actinobacillus suis.
or ears. Morbidity is usually low, but mortality is high among
those pigs left untreated. Concurrent infections with Streptococcus suis or Mycoplasma
hyorhinis are not uncommon. Brain, visceral pleura and other sero-
Haemophilus parasuis infection that manifests as a respiratory sal exudates are preferred culture sites since those are most likely
disease may involve serotypes that are different from those to have virulent H parasuis present. Because it is a common inhab-
causing septicemias and classic Glässer’s disease. Compromised itant of the nasopharynx in healthy pigs, detection of H parasuis
growth or stunting may be apparent in a portion of pigs, partic- from the nasopharynx, lung parenchyma, or oral fluids has no
ularly those where fibrinous serositis resolves with fibrosis and value in confirmation of Glässer’s disease. Serology is of little value
serosal adhesions. in diagnosis of disease.

Lesions Because of the multisystemic nature of Glässer’s disease, numer-


External examination may reveal cyanosis of the skin, swollen ous other causes of sudden deaths, respiratory distress, sepsis,
joints, or conjunctivitis. Lesions will vary with each pig, but those polyserositis, lameness, arthritis, and central nervous system dis-
of classic Glässer’s disease often have copious serofibrinous or eases should be included in the list of differential diagnoses.
fibrinopurulent exudate in the peritoneal cavity, the pericardial In complicated cases of Glässer’s disease, histopathology is
sac, within meninges, and/or on pleura in the thorax. Pleuritis extremely useful to confirm the presence of typical Glässer’s
is a common lesion and may be present with or without obvious lesions and to implicate roles for other disease agents. Molecular
pneumonia. Lesions in the lungs are often red, multifocal, dis- techniques are well described for identifying H parasuis from
seminated, and are suggestive of septicemia and hematogenous clinical specimens, as well as for characterizing the organism and
spread. Haemophilus parasuis may also contribute to typical bron- for carrying out epidemiological studies.
chopneumonias caused by other agents.
One or more major leg joints are often swollen, perhaps in combi- Control
nation with lesions at other sites, and there may be fibrin within The course of the disease is often short and many of the sick ani-
mals die if left untreated. For that reason, sick animals should be
the joint capsule along with reddened and swollen synovium.
promptly treated parenterally with an appropriate antimicrobial.
Exudate in joints is often fibrinous and gray to green in color.
If numerous pigs are affected, it may be advisable to mass medi-
Occasionally, some peracutely affected pigs will contain no readily
cate. Haemophilus parasuis is sensitive to many antibiotics but the
visible gross lesions. However, in many of these cases if the brain is sensitivity pattern of isolates from individual farms may vary over
removed, a fibrinopurulent exudate may be visible on the ventral time so antimicrobial sensitivity testing is warranted.
30 HAEMOPHILUS PARASUIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Prevention of Glässer’s disease may involve changes to manage- farrowing, or piglets vaccinated when 1 to 3 weeks of age, have
ment procedures, including improvements to husbandry and resistance to disease caused by the same (homologous) serovar.
environment, control of primary diseases such as PRRSV, admin- Colostral immunity in the piglets may sometimes interfere with
istration of prophylactic antimicrobials, or vaccination. Since very early vaccination against H parasuis infection.
H parasuis colonizes relatively early in the piglet’s life (less than
The natural process of herd immunity probably involves coloni-
10 days of age), segregated rearing has not been uniformly suc-
zation of young pigs while in the presence of maternal immunity.
cessful in preventing transmission to piglets or the downstream
This prevents expression of clinical disease, yet allows induction
spread of infection and disease. Achieving herd immunity to the
of active immunity to the agent, rendering pigs resistant to
agent is helpful and is augmented by proper acclimatization and
subsequent challenge with homologous strains. One innovative
perhaps vaccination of breeding stock. Minimizing stressors asso-
approach to control of the disease is based on purposefully man-
ciated with weaning, commingling, and the nursery environment
aging controlled exposure of nursing piglets to live H parasuis
should be emphasized as well.
endemic to the farm. The objective of this strategy is to permit
Killed bacterins as well as avirulent live vaccines are available and successful infection and development of active immunity in all
can be effective for prophylaxis. Unfortunately, protection against pigs, while disease expression is suppressed by maternal immu-
one serovar of H parasuis does not ensure good protection against nity. This concept attempts to mimic the natural ecology of
all serovars; cross-protection is difficult to predict even with organism and host. However, this technique may not be appro-
molecular technologies. Limited research suggests that piglets priate on large farms with multisite production that tend to have
from dams that have been vaccinated against H parasuis prior to multiple strains of H parasuis at any given time.

HAEMOPHILUS PARASUIS 31
SWINE DISEASE MANUAL
BACTERIA

Ileitis
Alternate names and abbreviations Etiology
Porcine proliferative enteritis (PPE), porcine intestinal adeno- Early microscopic reports of lesions of ileitis identified the
matosis (PIA), porcine hemorrhagic enteropathy (PHE), regional presence of curved, rod-shaped, Gram-negative bacilli in the
ileitis, garden-hose gut apical cytoplasm of proliferative crypt enterocytes. In 1993, the
offending fastidious causal bacteria was finally cultivated in vitro
Definition in tissue culture. Isolation of L intracellularis is laborious and
Porcine proliferative enteritis, commonly known as ileitis, is a quite technical; only a few laboratories routinely attempt to iso-
bacterial enteric disease caused by Lawsonia intracellularis and late the organism from field samples. As an obligate intracellular
is characterized by hyperplasia of crypt enterocytes and mucosal bacterium, tissue culture (eg, rat enterocyte) is still required for
thickening, with inflammation and sometimes ulceration or hem- organism propagation. Experimental inoculation of susceptible
orrhage of the intestine and/or colon. Clinical signs vary con- pigs with a pure culture of L intracellularis consistently repro-
siderably between individual pigs as does gross lesion location, duces ileitis, which has greatly augmented study of the organism
extent, and duration. and disease. All L intracellularis isolates examined from various
host species or different geographical locations appear to be
Occurrence quite similar genetically. With multiple passages in tissue culture,
Lawsonia intracellularis and its associated disease is distributed L intracellularis becomes attenuated; the technique has been used
worldwide with disease expression having become more common to develop a commercially available vaccine.
as production systems have become more modern. Disease is Despite rigorous investigation, virulence factors and protective
categorized pathologically as either acute porcine hemorrhagic epitopes of L intracellularis are not well understood. Polymerase
enteropathy (most typical in older pigs and young adults) or as a chain reaction (PCR) has been useful for detection of the agent
chronic, proliferative, and/or necrotic form that is more common and continuing investigation of sequence data is being used in
and usually occurs during the grow-finish phase. attempts to characterize epidemiologically important attributes of
Disease occurs most frequently in pigs 8 to 20 weeks of age, but L intracellularis.
may also occur in pigs as young as 3 weeks of age (especially in
antibiotic free or organic farms), in market-weight pigs, replace- Epidemiology
ment gilts, or in adult swine. Disease occurrence often appears to Lawsonia intracellularis is endemic globally and is found in most
be more severe and at an increased incidence in age-segregated domestic swine herds. Carrier dams have been shown to infect
“high-health” herds. Natural infection with similar signs and their litters as early as 6 days of age, a likely reason for failure of
lesions has been reported in other mammals, including horses, segregated early weaning techniques to control disease. Passive
hamsters, ferrets, guinea pigs, foxes, lambs, rabbits, rats, dogs, immunity may prevent infection in some pigs, hence transmis-
white-tailed deer, and emus. Irrespective of the source, no sub- sion from pig to pig within groups probably continues for several
stantial differences have been detected in the genotype of the months. This is supported by the gradual seroconversion that
L intracellularis agent. Hamsters have been used in research and occurs in groups of pigs over weeks to months during the growing
are readily infected by the porcine organism. phase. It follows that clinical impact may be present for weeks
to months, if no interventions are applied. Research has demon-
Historical information strated that L intracellularis recovered from experimentally
Lesions of ileitis were first reported as adenomatosis in 1931 infected pigs spreads to naïve sentinel pigs penned with them
due to the characteristic severe crypt enterocyte hyperplasia that or penned separately in the same enclosed area. The incubation,
was associated with the disease. The name “regional ileitis” was recovery, and carrier phase of L intracellularis infection can be
applied because of the propensity for lesions to be seen in the quite long in some pigs, probably months. There undoubtedly are
ileum. Ileitis was recognized as a sporadic disease over the next carrier swine that disseminate L intracellularis to susceptible pigs
40 years. It was not until the 1970’s that rising mortality and through their feces by the fecal-oral route or via feces-contam-
performance losses from ileitis on large, modern farms stimulated inated fomites. Although other animal species may be infected
specific research efforts on the disease. Several bacteria were with L intracellularis, there is little evidence suggesting direct
erroneously implicated as causal over the next 20 years. However, interspecies transmission. However, mice have been demon-
in 1993 L intracellularis was isolated in tissue culture, and subse- strated to harbor and shed L intracellularis in feces and hence
quent experimental infection studies confirmed it as the cause of may have a role in introduction or maintenance of L intracellu-
all forms of ileitis. laris in swine herds.

32 ILEITIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Infectious dose is low and fecal excretion from clinically affected Clinical signs
pigs is high. The severity of disease tends to be proportional to There has been much confusion related to nomenclature,
the infecting dose. The incubation period for expression of the lesions, and clinical signs of ileitis, primarily due to the variation
disease is at least 2 weeks. The L intracellularis organism has been that exists in clinical signs, age of pigs affected, gross lesions,
shown to survive at least 2 weeks in organic material, a feature and clinical impact. Three clinical forms of ileitis are commonly
which also helps to assure its continuous maintenance on a recognized.
farm premises.
The acute hemorrhagic form (PHE) is characterized by the
Ileitis may occur explosively, particularly when naïve older ani- appearance of acute bloody diarrhea with blood clots and the
mals are exposed to L intracellularis through commingling with sudden death of previously healthy pigs. This form of the dis-
infected animals or when they are placed under stress in an envi- ease is most common in pigs greater than 20 weeks of age (late
ronment in which L intracellularis is present in high numbers. grow-finish or young adults). Feces in PHE-affected pigs may be
Common examples of these situations occur when naïve gilts dark or black from blood loss, and affected animals may be pale
enter a sow farm or when carrier gilts enter a naïve sow popula-
from anemia. This clinical presentation must be differentiated
tion. It is also common to have acute outbreaks of ileitis occur
from bleeding gastric ulcers and hemorrhagic bowel syndrome.
just after some, but not all pigs, are sorted from the group then
removed for transport to slaughter. The chronic clinical form is most frequent in pigs from 6 to 20
weeks of age. Affected animals lose body condition and have
Outbreaks in endemic populations have often been associated
persistent or intermittent, liquid, gray feces. Salmonellosis, swine
with a variety of stressors. Infections in these circumstances are
dysentery, whipworm infestation, and some enteric coronaviruses
often subclinical yet may have substantial impact on growth and
are common clinical differentials. Morbidity and mortality with
performance. Vaccines for L intracellularis are very effective in
both the acute and chronic presentation are quite variable.
preventing clinical signs and lesions, but do not prevent coloniza-
tion or stimulate sterilizing immunity. Some pigs may have severe proliferation of the mucosa of the
terminal small intestine yet can be clinically asymptomatic. These
Pathogenesis pigs fall into the third clinical form of ileitis referred to as the
Lawsonia intracellularis infects enterocytes within hours after subclinical form. Affected pigs are without visible or obvious
ingestion and survives intracellularly, free in cytoplasm. When clinical signs but have compromised growth performance that
enterocytes undergo mitosis, each cell retains some of the may only be measurable through production data recorded at a
bacteria. However, these infected enterocytes fail to mature. herd level. These pigs have lesions that are identifiable at a micro-
This results in the hallmark lesion of proliferative enteritis - the scopic level that are not grossly visible at necropsy. Understanding
proliferation of immature enterocytes. Whether the organism whether L intracellularis is present and its relative importance is
stimulates greater proliferation, retards maturation, or both is not critical whenever groups of pigs have intermittent diarrhea with
known. Microscopically, organisms are visible within the apex of visible variation in growth.
crypt epithelial cells. Although much is known regarding patho-
genesis, some details remain speculative, particularly the existence Lesions
and role of specific virulence factors and mechanisms. Ileitis lesions correspond to the clinical forms described above.
Regardless of form, the lesions vary between pigs based on the
Infection results in crypt epithelial hyperplasia, crypt elongation,
duration, severity, extent, and location of the infection. However,
degeneration and necrosis of enterocytes, fewer goblet cells,
lesions that include gross thickening of a portion of the mucosa
and an inflammatory response. There are often degenerative
of the small and/or large intestine virtually always occur. In the
changes, cell necrosis, inflammation, and repair simultaneously
acute form, there are often bloody intestinal contents associated
present. The variations in the reported clinical and pathological
with grossly thickened small intestine. Clotted blood in the small
syndromes may correlate with different stages in lesion develop-
intestine is highly indicative of PHE.
ment, variation in immune modulation, or different individual
pig responses to infection via variations in predisposing factors, Lesions are more variable in the chronic form. The small intestine
stresses, microbiome, or presence of other saprophytes. is commonly affected with a hose-like, palpable thickening of
the mucosa of the ileum. The serosal surface of affected loops
Seroconversion usually occurs 2 to 4 weeks after infection,
of thickened intestine has a unique wrinkled appearance due to
although detectable lesions may not develop until 3 weeks
mucosal thickening and the presence of exudates. Peyer’s patches
post-infection. The protected, intracellular environment in which
may be hyperplastic. Lesions can be found in the small or large
the organism lives probably means that a cell-mediated response
intestine, or both.
is required to eliminate infection.
In the cecum or colon, the mucosa is thrown into thick folds,
Resolution of lesions starts approximately 3 to 4 weeks after plaques, or polyps, and it may be covered by inflammatory exudate
infection, but shedding may continue for months.

ILEITIS 33
SWINE DISEASE MANUAL
BACTERIA

and vaguely resemble lesions of swine dysentery, trichuriasis, Serology has limited availability and, similar to PCR testing,
salmonellosis or even porcine circovirus type 2-associated disease. requires understanding of both the test and immunopathogen-
Healing lesions of ileitis and ileal thickening in pigs at slaughter esis of L intracellularis. Antibody specific for L intracellularis
suggest that some affected swine may show few or no signs and can be detected within 2 weeks of infection. The magnitude and
recover spontaneously. duration of antibody response is proportional to disease severity.
Histopathology is useful to confirm the nearly pathognomonic When applied in a systematic manner on a herd basis through
lesions of ileitis, which usually include easily identifiable intracellu- sequential or cross-section sampling, serology can confirm that
lar L intracellularis organisms. Multiple sections should be exam- L intracellularis is present in a herd, the age when infection likely
ined in mild cases, lesions may be microscopic and segmental. The occurs, and the prevalence.
hallmark lesion of crypt enterocyte hyperplasia should be accom-
panied by demonstration of intracellular organisms via immuno- Control
histochemistry (IHC), in situ hybridization (ISH), or silver stains, Outbreaks of ileitis require antimicrobial intervention to prevent
irrespective of the clinical or pathologic form of disease. Crypt mortality. Affected pigs are usually injected with an appropriate
hyperplasia alone is not sufficient to make the diagnosis of ileitis antimicrobial while the rest of the group may be medicated via
since some crypt regeneration (proliferation) is not uncommon in water or feed for several days. Pulse doses of antimicrobials are
subacute or chronic stages of other enteric diseases. sometimes used for ongoing control or prevention, particularly
during or after periods of stress, or during outbreaks. Because of
Diagnosis the 2- to 3-week long incubation period, pulse doses are thought
A presumptive diagnosis is best made by observation of charac- to allow colonization and initiation of immunity while prevent-
teristic gross lesions at necropsy. Clinical signs are helpful but ing the occurrence of clinical disease. Medication failures can
often not specific. Confirmation of diagnosis is by observation occur in limit-fed breeding stock where animals are not fed ad
of typical microscopic lesions and demonstrating the presence libitum. In small herds that practice continuous-flow production
of intracellular curved L intracellularis in enterocyte apical cyto- and/or late weaning, clinical disease may be mitigated by herd
plasm by IHC, ISH, fluorescent antibody test (FAT), or silver immunity. Presumably, passively acquired maternal antibody
stains; IHC is considered the gold standard test for visualizing allows infection and active immunity to develop but prevents
the organism on histopathologic examination. Lesions may be overt disease. Control of ileitis has been a major reason for use of
segmental; histopathology should therefore be performed on feed-grade antimicrobials in grow-finish swine.
multiple carefully selected and preserved sections of intestine and Vaccination for ileitis is widely practiced. Both avirulent live vac-
colon. Polymerase chain reaction (PCR) testing on samples of cines and killed vaccines are widely available and have very good
affected feces, oral fluids, or mucosa is useful to confirm presence efficacy when properly administered. Vaccination of growing pigs,
of L intracellularis, but results should be interpreted with care as well as breeding gilts during acclimatization, has been very suc-
because of the high prevalence of L intracellularis in most farms, cessful in reducing or eliminating clinical signs of ileitis. In some
regardless of whether signs of ileitis are being expressed. herds, vaccination has allowed the elimination of continuous
The detection of L intracellularis does not confirm presence of administration of feed-grade antimicrobials.
the disease. Quantitative PCR performed on samples from clin- Elimination of L intracellularis from populations of pigs is cur-
ically affected pigs may be suggestive of ileitis, but the diagnosis rently impractical because exposure of swine to L intracellularis is
is not definitive without a complete diagnostic workup. Demon- currently unavoidable. Segregated rearing has not been successful
strating L intracellularis presence by PCR may be coincidental and may even increase the likelihood of fulminating acute ileitis
with other enteric diseases such as salmonellosis, trichuriasis, occurring in “high-health” herds where normal herd immunity
swine dysentery, coronaviruses, circovirus insult, or gastric ulcers. mechanisms are often purposefully disrupted as part of control
Negative PCR tests do not rule out carrier status. programs for other diseases.

34 ILEITIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Leptospirosis
Alternate names and abbreviations swine. Serovar L hardjo is the most important serovar of bovines
Lepto and has been reported to infect pigs kept in close contact with
cattle. Leptospira bratislava is reported to be the most common
Definition strain in swine, although the role of this serotype as a cause of
Leptospirosis is a contagious disease of swine (and many animals, disease is debatable.
including humans) caused by any one of a large group of Lepto-
spira bacteria. Disease expression in swine varies with the partic- Epidemiology
ular species and serovar that is involved; swine are susceptible to Many of the pathogenic Leptospira serovars, including L pomona,
many different serovars. have a predilection for the kidneys where they tend to persist.
Both maintenance and incidental hosts pass large numbers of
Leptospira in their urine, with shedding often lasting for weeks
Occurrence to months after clinical signs have concluded. Infection is usu-
Leptospirosis occurs in swine in all parts of the world where ally introduced into a herd by infected, shedding swine (often
environmental conditions are favorable for the survival of Lep- replacement gilts or boars), by direct or indirect contact with
tospira. The occurrence of any one serovar of Leptospira in an incidental hosts (rats, mice, skunks, raccoons, foxes, opossums,
area depends on the presence of maintenance hosts in which the etc), or through Leptospira-contaminated water, soil, or effluent.
infection can persist or the presence of incidental hosts. In both Outbreaks have been reported in herds using untreated surface
instances, Leptospira can be effectively disseminated to swine. The water for drinking.
disease occurs more frequently where the hosts are numerous,
Under ideal conditions, Leptospira survives in water and damp
concentrated, or in frequent contact with swine. soil, remaining a persistent threat to a wide range of animals
including swine and humans. Incidental hosts can be infected or
Historical information merely act as passive transport hosts that carry Leptospira to other
Early reports of “swine herder’s disease” in Europe described a premises. Carnivores that consume dead pigs may be underesti-
disease among caretakers of swine that probably was leptospirosis. mated as transmitters of Leptospira.
In the US, leptospirosis was recognized as an important disease
Fetuses infected in utero may survive as infected piglets and
of swine in the 1940s and 1950’. The disease continues to be become adult carriers, eventually transmitting Leptospira via
endemic in areas where environmental conditions are favorable urine to cohorts, offspring, or other farms, or acting as a source of
for Leptospira. However, losses have been reduced through rou- contamination for vectors or the environment. Some Leptospira
tine use of confinement housing, good sanitation, management serovars, especially L bratislava, have a predilection for the genitalia
changes, vaccination, and the use of antibiotics. Exposure to Lep- of both male and female swine and can be transmitted venereally as
tospira, and consequently the occurrence of clinical leptospirosis, well as through urine.
is now relatively uncommon in modern swine production.
Pathogenesis
Etiology Leptospira penetrate mucous membranes (eyes, mouth, nose,
Leptospira are motile spirochetes, 6 to 12 microns long, and or vagina), wounds, abrasions, and perhaps water-softened skin
0.1 µm in diameter. They are usually hooked on both ends and of the host. A leptospiremia then develops and organisms may
can be stained by Giemsa stain or in tissue, by silver stains. In the affect many major organs. Many Leptospira species localize in the
laboratory, they are often studied under darkfield microscopy. kidney or in the uterus during the last half of gestation. Leptospira
In laboratory media, they are difficult to culture and grow very bratislava can localize in the oviduct and uterus of non-pregnant
slowly (12 to 26 weeks). Many pathogenic strains can survive in sows and in the genital tract of boars. The exact way in which
the environment for long periods of time under moist conditions Leptospira produce their pathogenic effect is not known but
capillary damage is a major feature. Localization in the kidney,
with a slightly alkaline pH. Most cannot withstand desiccation
first in the interstitium and later in the tubules, results in vascular
and are destroyed by common disinfectants. damage and at least temporary damage to tubules. Interstitial
Taxonomy of Leptospira remains confusing. Under the most nephritis, if extensive, can lead to kidney failure and uremia.
current classification, the family Leptospiraceae contains 8 patho- The liver may also be affected. In addition, Leptospira produce
genic species, of which three are of most importance to swine: hemolysins which result in the hemolysis of erythrocytes and
Leptospira interrogans (serovars pomona, icterohaemorrhagiae, causes hemoglobinuria and hemolytic anemia. The severe effects
canicola, and bratislava), Leptospira borgpetersenii (serovars sejroe of hemolytic anemia occur mostly in very young piglets, perhaps
and tarassovi), and Leptospira kirschneri (serovar grippotyphosa). fetuses, and may trigger abortions.
Serovars pomona and bratislava are uniquely adapted to swine;
others are maintained in other species but sometimes infect

LEPTOSPIROSIS 35
SWINE DISEASE MANUAL
BACTERIA

Clinical signs Diagnosis of outbreaks is often based on serological herd testing


The acute phase of leptospirosis largely coincides with an initial using the microscopic agglutination test. Serum samples from
stage of leptospiremia as well as virulence of the particular serovar both affected and unaffected animals should be tested. High
involved. Signs often go unnoticed in mature, non-pregnant antibody titers are usually present at the time of abortion or
swine and in growing pigs. A careful observer might notice farrowing, but paired samples may sometimes be required to
mild fever and inappetence for a few days. In very young piglets, correctly interpret titer levels. Microscopic agglutination testing
however, there may be fever, anorexia, hemolytic anemia, hemo- is serovar specific, so the use of multiple assays may be required to
globinuria, icterus, convulsions in occasional pigs, and a failure to arrive at a diagnosis. Breeding animals are commonly vaccinated
grow and gain weight. against multiple Leptospira serovars, so interpretation of serolog-
Chronic infection in dams is usually apparent only as various forms ical results must also consider vaccination history of the animal.
of reproductive failure, including poor conception rates. Pregnant, Titers from infection are usually much higher than those induced
infected animals often abort in late gestation. In many cases, fetuses by vaccination.
are carried almost full term but may be mummified, dead, or weak
at birth. Although many infected neonatal pigs die within a few Control
days, some often survive. The dams usually recover promptly. They Control is usually attempted by prevention of exposure, immu-
often conceive again and carry their litters to term. nization through vaccination, and/or the use of antibiotics. Pre-
vention of exposure is difficult to achieve because so many species
Lesions can act as carriers of Leptospira. These include infected swine,
Lesions have seldom been described in sows since most infected rodents (especially mice and rats), and many kinds of wildlife.
adults recover. Grow-finish swine may have scattered foci of Once Leptospira are introduced into a favorable wet environ-
interstitial nephritis or generalized kidney scarring that may only ment, they often persist there as a continuing source of infection.
be noticed at slaughter as white-spotted kidneys. Lesions in small Nonetheless, leptospirosis can be effectively controlled (perhaps
piglets include patchy areas of hepatic necrosis, excessive fluid even eradicated) from swine populations raised under confine-
and fibrin in the peritoneal cavity, and evidence of inflammation ment conditions using a combination of medication, vaccination,
in multiple organs. Microscopically, there is a marked focal, dis- vector control, and providing a treated or non-contaminated
seminated, interstitial nephritis; tubules and glomeruli often have drinking water source.
degeneration and inflammation. Occasionally, a non-suppurative Immunization through the use of bacterins, widely practiced in
meningitis can be observed. Placental lesions are seldom marked breeding herds, will usually reduce the prevalence of infection
or significant but may include edema and scattered hemorrhages. and abortions. The bacterin must be appropriate for the serovar
of Leptospira causing the disease on the farm. Many bacterins
Diagnosis are multivalent. The level of immunity obtained and its duration
A history of abortions in late gestation along with lesions in may not always be satisfactory, meaning sows may need to be
neonates may suggest leptospirosis, but mere observation is inad- vaccinated 2 or 3 times per year to maintain protection. Also,
equate for accurate diagnosis. Demonstration of organisms in bacterins will not usually eliminate infection in carriers.
tissues or fluids by polymerase chain reaction can confirm infec-
tion but may not identify the particular serotype. Histopathology Antibiotics used judiciously may reduce the impact of leptospi-
with immunohistochemistry or fluorescent antibody test on rosis. It is doubtful that all carriers can be eliminated through
kidney or liver is quite definitive. Silver stains of tissue smears or the use of antibiotics. Research suggests that oxytetracycline,
urine can reveal organisms with compatible morphology, as can tylosin, and erythromycin are among the most effective agents
darkfield or phase-contrast microscopy, but may not be specific or for treating leptospirosis.
sensitive enough to detect some infections. Isolation and identifi-
cation of Leptospira is both tedious and laborious.

36 LEPTOSPIROSIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Mycoplasma suis
Alternate names and abbreviations occurs after about 7 to 8 days, which is during the acute phase of
Eperythrozoon suis (previously), epy, eperythrozoonosis the disease and the time when organisms are most numerous on
the surface of erythrocytes. Infected pigs become hypoglycemic
Definition and gradually develop hemolytic anemia. Hemoglobinuria does
A sporadic disease usually observed in young growing pigs, char- not occur because hemolysis is extravascular. However, icterus
acterized by listlessness, fever, anorexia, hemolytic anemia, and can occur in some cases.
(in severe cases) icterus. Occasionally, dysgalactia has also been The number of platelets, the packed cell volume, and hemoglobin
associated with infection in sows. values decrease. Most affected animals recover in the absence of
additional stressors but some do remain as carriers. The organism
Occurrence was thought to reside in the cell membrane, but M suis is now
Mycoplasma suis is a bacterial infection that can occur in all age known to transmigrate to the interior of the red blood cells. In
groups of swine but is seldom clinically apparent except in young, most situations, M suis rarely causes substantial death loss. Rather,
growing pigs. Distribution is worldwide, with infection common it is recognized as an occasional cause of anemia or icterus in
but actual disease outbreaks rather uncommon. young pigs or as a contributor to compromised reproductive or
lactation performance in sows.
Historical information
Icteroanemia in growing swine was described in the US in 1943 Clinical signs
and was investigated in the 1950s. Recognition that there were In young growing pigs, signs may include anorexia, listlessness,
many carriers came after development of an antibody test in weakness, pale mucous membranes, and sometimes icterus. Body
1975. Research with this organism has been sporadic, but with temperature is initially raised during infection but is often sub-
the advent of polymerase chain reaction (PCR) testing for the normal by the time the pigs are examined. In chronic cases, there
organism, the disease has garnered renewed interest. is often marked icterus, and deaths may occur. Signs in breeding
stock are similar but are less obvious and often go unnoticed. In
Etiology breeding herds, M suis infection has been associated serologi-
Mycoplasma suis organisms are obligate, intracellular bacteria cally with various reproductive problems, including dysgalactia.
lacking cell walls. The organism is usually from 0.2 to 1.0 µm in However, proof that these reproductive effects are caused by the
diameter but may be larger during the acute stages of infection. It disease is still lacking.
is usually coccoid-shaped but may appear rod or ring-shaped when
adhered to an erythrocyte’s cell membrane. The organism has not Lesions
been cultured in cell-free media. Lesions at necropsy depend somewhat on the duration of illness
and the age of the animals. In younger pigs, there may be pallor
Epidemiology and watery blood compatible with anemia, and perhaps modest
Herd prevalence is quite variable, with some countries reporting enlargement of the spleen. In cold climates, there may be necro-
up to 90% of herds infected, with herd prevalence of 10% to sis of extremities. External parasites (such as lice), their ova, or
20%. There are likely numerous subclinical M suis carriers in the insect bites may be present. In older pigs, the same hematology
swine population. In utero transmission has been demonstrated findings and spleen lesions are apparent at necropsy but splenic
experimentally. Since the disease occurs more frequently in the enlargement is more noticeable. There is often icterus (more
summer and fall, it is suspected that mosquitoes and biting flies as obvious internally) that may be marked. The bone marrow is
well as the hog louse (Haematopinus suis) may play a role in trans- red, suggestive of hyperplasia. The liver often has a brownish cast
mission. The use of contaminated surgical instruments, including from hemosiderosis. There are often suffusions in the abdomen,
castration knives and needles used repeatedly during vaccination, thorax, and pericardial sac. Lymph nodes are edematous and
is also believed to transmit the organism. Ongoing infectious and swollen. The heart is often thin-walled and flabby. Careful exam-
parasitic diseases are believed to play an important role in precip- ination often reveals lesions of concurrent diseases or parasitism.
itating outbreaks.
Diagnosis
Pathogenesis Mycoplasma suis is not a major cause of disease, nor are outbreaks
Much of the information on pathogenesis is based on the study spectacular. The history, signs, and gross lesions are often subtle
of experimental infection in splenectomized pigs. In those pigs, or vague, yet are of value in diagnosis. Confirmation of the
the clinical and pathologic aspects resemble reports of natural diagnosis is made by demonstrating the presence of organisms by
infections. Once M suis is introduced into the blood, a fever microscopy, PCR, or indirectly by serology. Organisms can be
MYCOPLASMA SUIS 37
SWINE DISEASE MANUAL
BACTERIA

visualized on the surface of erythrocytes in blood smears or on Control


impression smears from spleen tissues that have been stained with Tetracyclines are used with success for treatment of M suis infec-
Wright-Giemsa or Diff-Quick stains. Blood with anticoagulant tions. Intracellular organisms may not be responsive to single tetra-
(eg, citrate, heparin, or EDTA) should be collected from pigs in cycline treatments so treatment may need to be repeated. Affected
the acute stage of disease. Another reported technique for diag- pigs can be treated individually with injectable oxytetracycline.
nosis is mixing equal parts of blood with 10% formalin, and then
staining a smear of the material with acridine orange. Diagnostic Where available, serology may be useful as a screening test prior to
laboratories now usually prefer to confirm the presence of M suis the purchase of breeding stock or to identify herd infection.
in blood or spleen by PCR. There are no vaccines for the prevention of M suis. In modern pro-
Serological tests (indirect hemagglutination [IHA] or enzyme- duction systems, clinical infections with M suis are unusual. Since
linked immunosorbent assay) may be available to confirm M suis as signs are sometimes vague, diagnostic investigations may need to be
a problem in a herd, although these tests have limited availability. expanded to assure other agents or risk factors are not contributing.
Infected pigs less than 3 months old seldom react on IHA. Differ-
ential diagnoses for M suis include systemic salmonellosis, porcine
circovirus associated disease, mycotoxicosis, hepatotoxicosis, iron
deficiency anemia, and autoimmune hemolytic anemia.

38 MYCOPLASMA SUIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Mycoplasmal arthritis
Alternate names and abbreviations Epidemiology
Mycoplasma hyosynoviae (MHS), Mycoplasma hyorhinis (MHR) Mycoplasma hyorhinis and M hyosynoviae are considered commen-
sals, colonizing the tonsils and respiratory epithelium of the nasal
Definition cavity and conducting airways. A majority of colonized pigs have
Mycoplasma hyorhinis and M hyosynoviae are considered com- no apparent clinical disease. Transmission is suspected to be pri-
mensal bacteria in swine but are also known to be important marily via nasal secretions by direct contact or close proximity with
causes of arthritis on some farms. Mycoplasma hyorhinis usually the dam (M hyorhinis) or penmates (M hyorhinis and M hyosyno-
affects younger pigs up to 10 weeks of age. In addition to arthri- viae). Prevalence of M hyorhinis is high at or shortly after weaning.
tis, it can also cause polyserositis, otitis, pneumonia, and sporadic Prevalence of M hyosynoviae is low at weaning and slowly increases,
abortions in sows. Mycoplasma hyosynoviae usually causes arthritis peaking around 10 to 16 weeks of age. The conditions or risk fac-
in older (greater than 10 weeks of age) grow-finish pigs and tors that trigger systemic spread of these tonsillar and respiratory
young adults. tract commensals are unknown. However, evidence suggests that
the dam is an important source of both organisms, leading to early
Occurrence colonization of piglets and making the likelihood of eradication of
It is suspected that both bacteria have a worldwide presence, either organism from a farm very unlikely.
but published data on regional prevalence are lacking. Even in
herds where Mycoplasma hyopneumoniae has been eradicated, Pathogenesis
M hyorhinis and M hyosynoviae are still present. In some areas, Both bacteria bind to the ciliated respiratory epithelium of the
there are perceptions that disease associated with these organ- nasal cavity and conducting airways. Systemic invasion from these
isms has increased, particularly in “high health” herds. In recent commensal sites is well documented, but mechanisms of invasion
years, diagnostic laboratories have reported increased frequency and spread are still unknown. Both bacteria can be detected in
of detection of these organisms, which may be associated with the blood of asymptomatic pigs for short periods of time. In
increased testing, improved diagnostic capabilities, or an increase some, the organisms are able to localize and colonize systemic
in the clinical impact of these agents in some herds. sites, stimulating inflammation and disease expression. With
M hyosynoviae, systemic colonization is usually limited to syno-
Historical information via, with disease and lesions referable to synovitis. On the other
Mycoplasma lack a cell wall and are the smallest and simplest hand, M hyorhinis can colonize serosal surfaces of heart, pleura,
known self-replicating organisms. Mycoplasma hyorhinis was peritoneum, or synovium, causing inflammation and fibrinous
initially described by Switzer in 1955. In 1970, Ross and Karmon exudates at any or all of those locations. In a challenge study,
first described M hyosynoviae. In recent years, M hyorhinis and M hyosynoviae was found in multiple joints at the same time,
M hyosynoviae have been increasingly recognized as swine patho- supporting the role of bacteremia as a prerequisite to disease
gens from both an economic as well as an animal welfare perspec- expression.
tive, especially in herds with higher health status. Detection of either or both organisms in lungs of healthy pigs
by polymerase chain reaction (PCR) is common and does not
Etiology appear to correlate with coughing or pneumonic lesions, nor do
The small genome of both organisms and their inability to rapidly the organisms appear to contribute to porcine respiratory disease
synthesize ribosomes results in relatively slow replication rates complex. Sporadic abortions reported with M hyorhinis are rare,
compared to most bacterial pathogens. As with most Myco- presumably a result of transplacental infection as sequel to bactere-
plasma, they attach to and replicate on epithelial cells. mia in young dams.
Both M hyorhinis and M hyosynoviae are known to infect swine
only. Both bacteria are easier to isolate and cultivate in vitro than Clinical signs
M hyopneumoniae, but each still requires unique special enriched Clinical signs of arthritis are similar for M hyorhinis and
media as well as several days to a week for isolation or propagation. M hyosynoviae, but the age of pigs affected is usually 3 to 10
weeks for the former, compared to 3 to 5 months for the latter.
The complex system of membrane lipoproteins of M hyorhinis Joint swelling can occur in single or multiple joints, manifested as
creates remarkable membrane lipoprotein diversity with variable a painful lameness. Lameness may be transient (just a few days) or
protein expression and size which is suspected to be important last for an extended time, especially when complicated by other
in pathogenesis and immune evasion. Typing methods such as health or environmental conditions. Affected pigs generally are
multi-locus sequence (MLST) and multiple-locus variable num- off feed and reluctant to move, with older pigs sometimes pre-
ber tandem repeat analysis (MLVA) have been developed recently ferring a “dog-sitting” posture. This is accompanied by reduced
to better characterize this diversity.
MYCOPLASMAL ARTHRITIS 39
SWINE DISEASE MANUAL
BACTERIA

weight gain and loss of body condition. If not rapidly resolved, Samples for confirmation of disease include fibrin or swabs col-
pigs spend more time in recumbency, are more likely to have joint lected from joints with lesions (M hyorhinis and M hyosynoviae),
calluses or chronic lameness, and are more likely to be culled. or fibrin or fluid from the pleural, pericardial, and peritoneal
cavities where lesions are present (M hyorhinis). PCR assays are
Experimental studies confirm that pigs inoculated with M hyorhinis the usual means for detection. It is important to remember that
can develop polyserositis and/or arthritis approximately 3 to 10 days as commensals, both organisms can commonly be detected in
post-inoculation. Labored breathing and dyspnea can be observed in nasal and respiratory tracts of normal pigs, hence PCR for disease
pigs with pleuritis. Studies have shown that after experimental infec- diagnosis should be performed only on suspected lesions such as
tion with M hyosynoviae, lameness can occur in up to 30% of pigs fibrinous exudates, synovium, or synovial fluid.
within 4 to 10 days after experimental inoculation.
Molecular characterization of M hyorhinis has been performed
with MLST and MLVA typing although epidemiological inter-
Lesions pretation of these results is currently being investigated.
Gross lesions associated with M hyorhinis most commonly
include polyserositis (involving pleura, pericardium and/or peri- Both M hyorhinis and M hyosynoviae can be grown in their
toneum) and/or fibrinous synovitis. Microscopic lesions include respective culture media but primary isolation attempts are easily
infiltration of lymphocytes, plasma cells, and macrophages in overgrown by other bacteria unless samples are aseptically col-
lected from lesions present in acutely affected, unmedicated pigs.
serosal connective tissues. Synovia are edematous with fibrino-
suppurative exudates and expansion of the synovial stroma by Serological enzyme-linked immunosorbent assays are not widely
infiltrates of lymphocytes, plasma cells, macrophages, and fewer available and are currently under development or investigation to
neutrophils. Co-inoculation of M hyorhinis with porcine repro- better interpret their results.
ductive and respiratory syndrome virus or porcine circovirus type There are many other causes of polyserositis and arthritis that can
2 has resulted in more severe lesions. present with similar clinical signs and gross lesions, including Strep-
Mycoplasma hyosynoviae causes lesions in one or more joints tococcus suis, Haemophilus parasuis, Actinobacillus suis, and Erysip-
without specific tropism to any one joint. Grossly, affected joints elothrix rhusiopathiae. Other potential contributors to lameness
often have increased amounts of red-tinged synovial fluid with or joint lesions include non-infectious causes and risk factors such
as trauma, commingling, nutritional diseases, and environmental
decreased viscosity and very moist, swollen synovia. Microscop-
(flooring and housing) and management factors. Osteochondrosis
ically, the synovial stroma is expanded by serofibrinous effusion
can be a differential for M hyosynoviae in older pigs.
and sparse aggregates of lymphocytes and plasma cells. As lesions
become subacute to chronic and the fibrin and neutrophilic Control
infiltrates resolve, large numbers of lymphocytes and plasma cells The widespread commensal occurrence of both of these patho-
expand the synovial stroma. The result is proliferation of synov- gens, along with the current limited knowledge on strain variabil-
iocytes and supporting connective tissue, resulting in grossly ity, strain virulence, and epidemiology, makes them difficult to
visible villous proliferation of the synovium. prevent or control. Current efforts in prevention are focused on
the use of vaccines. The effectiveness of autogenous vaccines has
Diagnosis not been documented.
Clinical signs and gross lesions are not usually sufficient for diagno- Treatment of outbreaks of arthritis with antimicrobials is often
sis of mycoplasmal arthritis or serositis. The age of the pigs affected practiced. Early detection of lameness and rapid implementation
is an important consideration to clinically differentiate the acute of specific antimicrobial therapy, often by injection of acutely
arthritis caused by these two mycoplasmas. Pigs greater than 10 affected pigs, is critical for successful treatment.
weeks of age are more likely to be afflicted with M hyosynoviae,
whereas M hyorhinis is more likely to affect pigs less than 10 weeks
of age. A rapid and favorable response of acutely affected pigs to the
injection of an appropriate antimicrobial can support a diagnosis of
M hyosynoviae involvement but is not confirmatory.

40 MYCOPLASMAL ARTHRITIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Mycoplasmal pneumonia
Alternate names and abbreviations virus (PRRSV), influenza, porcine circovirus type 2, Actinoba-
Enzootic pneumonia (EP), mycoplasmal pneumonia of swine cillus pleuropneumoniae, and Pasteurella multocida. Mycoplasma
(MPS), Mycoplasma hyopneumoniae (MHP) hyopneumoniae is quite capable of acting as a significant primary
pathogen on its own.
Definition Three other pathogenic mycoplasmas are recognized in swine.
A widespread, chronic respiratory disease of swine characterized Mycoplasma hyosynoviae is a sporadic cause of epidemic synovitis
by coughing, growth retardation, and reduced feed efficiency. in growing swine. Mycoplasma hyorhinis infection is common in
swine and can cause fibrinous polyserositis in young pigs. Both of
Occurrence these agents are of increasing concern in modern commercial pro-
Mycoplasmal pneumonia of swine is a common, widely dis- duction systems. Mycoplasma suis (previously known as Eperyth-
tributed disease that occurs throughout the year. It is restricted rozoon suis) infects red blood cells and can cause anemia in pigs.
to swine and occurs in all major swine-raising countries except
possibly Switzerland, where a national eradication program Epidemiology
appears to have been mostly successful. Chronic MPS is often Carrier swine are the most common source of infection. Since
more apparent on farms where there is continuous-flow produc- M hyopneumoniae does not survive for long in the environment,
tion and management, and where husbandry and environmental carriers are essential for its maintenance in populations. The
conditions are poor. The disease may cause acute and severe organism persists for 7 to 8 months in the lungs of infected, often
disease in immunologically naïve pigs in age-segregated systems. asymptomatic pigs, including young breeding stock. There is no
It may affect pigs early after weaning, when passive immunity has evidence to implicate infection or carriage in other species. Pigs are
waned, but more commonly occurs in grow-finish pigs from 3 to infected by M hyopneumoniae transmitted from dams, cohorts, or
6 months of age. Mycoplasmal pneumonia often interacts with or exposure to other, usually older, pigs. The organism can be detected
contributes to other respiratory diseases and is considered to have in nasal secretions, so transmission by nose-to-nose contact and
a central role in porcine respiratory disease complex (PRDC). coughing are likely. The organisms also appear to be transmitted by
Secondary bacterial infections are common. aerosol; empirical evidence suggests that aerosol spread can occur
over several miles. A mycoplasma-free herd status can be difficult to
Historical information maintain in areas with high pig density.
Chronic pneumonia in swine, part of which was probably myco-
plasmal pneumonia, has been recognized for at least a century. Pathogenesis
Mycoplasmal pneumonia was once believed to be caused by a Mycoplasma hyopneumoniae can be observed microscopically on
virus and was misnamed viral pig pneumonia. However, clear the ciliary epithelium of trachea, bronchi, and bronchioles. The
distinction of mycoplasmal pneumonia from influenza A and distribution of pulmonary lesions suggests bronchogenic distri-
recognition of it as a specific disease began in 1948. Mycoplasmal bution with settling of exudate cranioventrally because of the
pneumonia was first reproduced experimentally in the US and compromised mucociliary apparatus. Virulence factors derived
England in 1965. Subsequently, it was reported in most major from M hyopneumoniae outer membrane proteins damage several
swine-raising countries. Mycoplasmal pneumonia is recognized as respiratory defense mechanisms and facilitate infection. The cell
a very costly, widespread disease of swine due to its negative effects membrane presents a mosaic of epitopes that camouflage pro-
on growth rate and feed efficiency, its very high prevalence in tective antigens, rendering the immune response inefficient. The
infected herds, and its role in PRDC. immune response by macrophages and neutrophils also appears
to be compromised. Poor air quality (dust or noxious gases) can
Etiology irritate airways and increase susceptibility.
Mycoplasma hyopneumoniae, the etiologic agent of mycoplasmal The initial lesions are bronchitis and bronchiolitis. There is
pneumonia, is difficult to isolate and grows slowly in the labo- hyperplasia of the mucus-secreting cells in the mucosa and a loss
ratory. It is a small, filterable bacteria, survives only a short time of cilia from many epithelial cells of airways. The inflammatory
in swine housing environments, and can be destroyed by most reaction spreads into surrounding alveoli causing alveolar exu-
disinfectants. Mycoplasma hyopneumoniae is frequently found in dates, pneumonia, airway obstruction, and atelectasis. Over time,
association with other respiratory pathogens, including bacteria there is a marked hyperplasia of lymphoid tissue around airways
and viruses, the resulting combination of which is often termed and adjacent blood vessels, often described as lymphocytic
PRDC. In pigs with pneumonia, it may be difficult to determine
peribronchiolitis or hyperplasia of bronchiolar-associated lym-
which agent is the primary pathogen. Evidence suggests that
phoid tissue. Increased mucus in airways, ciliostasis, and pressure
M hyopneumoniae increases the severity of several other infec-
of surrounding lymphoid tissue interfere with the lung clearance
tions, including porcine reproductive and respiratory syndrome
MYCOPLASMAL PNEUMONIA 41
SWINE DISEASE MANUAL
BACTERIA

of mucus and exudate. Secondary bacterial infections, most of the organism but PCR alone probably should not be used to
notably with Pasteurella multocida but also others, contribute confirm a role for M hyopneumoniae in disease causation. Histo-
substantially and are the usual cause of severe bronchopneumonia pathology, ISH, IHC, and FA are preferred methods to confirm
and deaths. a role for M hyopneumoniae in the pneumonia process. Moreover,
histopathology can help rule out other contributors.
Clinical signs Serological tests including complement fixation and enzyme-
The principal clinical sign is chronic, persistent, non-productive linked immunosorbent assay can be useful on a herd basis, but
cough. In naïve animals, onset often occurs about 2 to 3 weeks interpretation must be made with care and knowledge of the
after exposure and is usually gradual in a herd. Coughing may herd, the particular age and individuals sampled, and their vac-
persist for weeks to months. Excessive dust, irritating gases, or cination status. Antibody tests on individual animals have little
concurrent infections result in more severe coughing and pneu- value because many pigs without active disease have antibodies
monia. As pneumonia develops in some pigs, dyspnea becomes to M hyopneumoniae or other cross-reacting Mycoplasma species
more marked. Growth is retarded, and feed efficiency decreases (especially M flocculare). Antibody levels develop slowly in many
in the face of near-normal appetites. Morbidity is high. Mortality infected animals. In vaccinated animals, the detection of antibod-
is low, except when the disease is complicated by other viral and ies in serum can be quite variable (ranging from 30% to 100%) as
aggressive bacterial infections or when proper treatment and can the duration of antibody detection (1 to 3 months), depend-
interventions are not instituted. ing on the vaccine used and the assay used for detection.
PCR technology can be used to determine herd status in some
Lesions situations. Sample types, in decreasing order of sensitivity, include
In affected pigs, pneumonic lesions are generally well-demarcated tracheobronchial swabs and lavages, lung tissue, laryngeal swabs,
and cranioventral in distribution. Gross lesions are not pathogno- tonsil swabs, nasal swabs, and oral fluids. The sensitivity of the
monic and usually involve apical, intermediate, and cardiac lobes different sample types is quite variable. For example, oral fluids
but may extend into diaphragmatic lobes in severe cases. Chronic have low sensitivity for detection of carriers but can detect
lesions are usually reduced in volume (atelectasis) and dark gray M hyopneumoniae quite efficiently in pigs with clinical disease.
in color. More recent lesions tend to be reddish brown or a lighter Detection of the organism by PCR confirms the presence of the
gray, with edema, mucus, and inflammatory cells apparent in organism but does not confirm its role in disease.
the airways. Raised areas adjacent to pneumonic areas are often
emphysematous and lighter pink than normal lung. On the cut The value of slaughter checks to evaluate the impact of mycoplas-
surface of pneumonic areas, mucopurulent exudate can often be mal pneumonia is questionable, since gross lesions may be healed
squeezed from airways. Secondary infection with other respira- by the time pigs reach market weight and lesions from other bac-
tory pathogens is common and may modify the appearance of terial and viral agents may mimic those of M hyopneumoniae.
mycoplasma-initiated lesions.
Control
Typical microscopic lesions, although not pathognomonic, reveal Many methods of control for mycoplasmal pneumonia are in use,
lymphohistiocytic peribronchiolar cuffing, mucocellular exu- and many herds supplying genetic stock are maintained negative
dates, and atelectasis. for M hyopneumoniae. Such herds obviously must have scrupu-
Neither gross nor microscopic lesions are pathognomonic lous isolation, acclimatization, testing, and biosecurity protocols
for M hyopneumoniae. Cranioventral, bronchogenic, clearly in place to assure the organism is not reintroduced.
demarcated consolidation of lungs is a characteristic of the bron- Most M hyopneumoniae-negative herds have been created as a
chopneumonia that is produced by M hyopneumoniae and can result of depopulation and repopulation with negative breeding
be present even when the pneumonia is complicated by other stock. In some cases, herds have become negative through one or
bacterial and viral agents. more management techniques, including strict medicated or seg-
regated early weaning, other minimal disease programs, or closing
Diagnosis the breeding herd to introductions of new animals for at least
History, clinical signs, and gross and microscopic lesions are 6 to 7 months.
suggestive of M hyopneumoniae, but laboratory assistance is
necessary to confirm a role for M hyopneumoniae in a given pig. Historically, specific pathogen free herds were established with
Isolation is slow, laborious, difficult, and generally not routinely pigs delivered via hysterectomy or cesarean section and raised in
available. Detection of M hyopneumoniae in lung samples taken isolation on colostrum-free diets. Those animals were then used
at necropsy is usually done by polymerase chain reaction (PCR), to repopulate previously depopulated farms.
in situ hybridization (ISH), immunohistochemistry (IHC), or
fluorescent antibody (FA) techniques (listed in decreasing order Maintaining commercial herds free of M hyopneumoniae infec-
of sensitivity). Since many pigs may carry M hyopneumoniae tion when located in swine-dense regions has not been successful
asymptomatically in lung tissue, PCR may detect the presence long term. A strategy referred to as “Swiss depopulation” is based

42 MYCOPLASMAL PNEUMONIA
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
on the hypothesis that most adult swine raised on an endemically for minimizing the impact of the disease. Despite reports of many
infected farm are exposed to M hyopneumoniae early in life with methods to eliminate or control M hyopneumoniae, it remains
subsequent recovery (and presumably robust immunity with a significant economic problem, particularly when PRRSV or
limited carrier status) by 10 months of age. Swiss depopulation influenza A virus is also endemically present in the herd.
involves implementation of a period of intensive vaccination of
Vaccines are generally thought to be efficacious in mitigating
all breeding animals on a farm followed by removal of all animals
losses due to M hyopneumoniae by reducing the prevalence and
less than 10 months of age. This is coupled with cessation of
severity of lung lesions and improving growth performance.
farrowing for at least 3 weeks while applying aggressive antimi-
However, vaccines do not prevent colonization or transmission
crobial therapy to eliminate any remaining M hyopneumoniae
of M hyopneumoniae from sow to piglet or between pigs. In most
that may be residing in carrier animals. Removal of all susceptible
herds, maternal antibodies do not seem to interfere with piglet
swine combined with vaccination and/or medication programs
vaccination, meaning that piglets on most farms can be success-
generally results in a significant decrease in the disease for up
fully vaccinated at or before weaning, with a booster dose given
to 2 years, with true eradication of the organism from the farm
around 3 weeks later.
reported in some instances. Modifications to these procedures
continue. Antibiotics may augment management and vaccination programs
but should not be considered as a cost-effective, long-term con-
Other measures to aid in control, but not elimination, of M hyo-
trol strategy. Antimicrobials can be strategically administered in
pneumoniae include early weaning or various forms of medicated
the feed or water, or by injection. Many antibiotics and chemo-
early weaning. Early weaning (7 to 10 days of age) may prevent
therapeutic agents have been used in feed or water for prevention
transmission of M hyopneumoniae (and other organisms) from
or treatment of mycoplasmal pneumonia, often with inconsistent
dams to piglets before they are weaned and moved to a separate,
results. The efficacy of antimicrobials in controlling M hyopneu-
clean nursery or grow-finish facility. Age-segregated rearing and
moniae losses may be related either to anti-mycoplasmal activity
the use of all-in, all-out systems of management with cleaning and
or by suppression of other bacterial infections.
disinfection of facilities between batches of pigs is widely used
and believed to be one of the more practical and useful measures

MYCOPLASMAL PNEUMONIA 43
SWINE DISEASE MANUAL
BACTERIA

Pneumonic pasteurellosis
Alternate names and abbreviations Pathogenic strains damage endothelium and microvasculature
Pasteurellosis; also see chapter on Atrophic rhinitis in the lung. This leads to the formation of fibrinous thrombi in
alveolar capillaries and fibrinous alveolitis accompanied by sup-
Definition puration. The lesion may slowly organize into fibrous connective
Pneumonic pasteurellosis is usually secondary to other infectious tissue or form abscesses as a consequence of infarction or necrosis
diseases or environmental factors that impair pulmonary func- from toxin associated with the bacteria and inflammation; only
tion. Both primary and secondary forms of disease occur. occasionally does fibrinous pleuritis or pericarditis occur. When
present, fibrinous pleuritis, serositis, and/or fibrous adhesions are
Occurrence much more likely due to prior effects of Actinobacillus or Strep-
Pneumonic pasteurellosis occurs throughout the world. Second- tococcus infections than as a direct result of P multocida. In acute
ary pasteurellosis is seen in association with several other diseases, cases, septicemia can occur as a result of the infection; deaths are
especially mycoplasmal pneumonia, Actinobacillus pleuropneu- believed to be caused by endotoxic shock and respiratory failure.
moniae, and viral lung infections, or it can accompany or follow
environmental or other stresses. Pasteurellosis occurs in many Clinical signs
other species of domestic and wild animals. Primary pasteurellosis Signs vary in severity and are similar to those seen with other
occurs occasionally as a septicemic disease in piglets, but this types of bronchopneumonia: coughing, dyspnea, fever, and pros-
rarely occurs in older or adult swine. Secondary pasteurellosis tration. Chronic cases tend to have less fever, a persistent cough,
occurs very frequently as a contributor to bronchopneumonia. and a more marked dyspnea. Swine with well-developed pneumo-
nia or extensive adhesions between the lungs and rib cage often
Historical information have a marked expiratory diaphragmatic lift or “thump.”
Louis Pasteur experimented with a disease of poultry (fowl chol-
era) caused by a bacterium now known as Pasteurella multocida. Lesions
Subsequent research implicated P multocida as both a primary Lesions are those of bronchopneumonia, with firmly consol-
and secondary pathogen in many species, including swine. idated areas affecting the cranioventral lobes of the lungs and
often extending to adjacent parts of the diaphragmatic lobes.
Etiology Pneumonic areas are remarkable for their firmness and density.
Pasteurella multocida is a Gram-negative coccobacillus easily The pleura over the lesions may be granular; scant fibrin may be
grown in the laboratory. There are both toxigenic and non-toxi- visible on the surface. Regional lymph nodes are usually enlarged.
genic strains. The organism is a common inhabitant of the Microscopic lesions include suppurative bronchopneumonia
pig’s nasal passages and can be isolated from most swine herds. with fibrin and neutrophils, with bacteria sometimes noticeable
There are 5 capsular serotypes (A, B, D, E, and F) and at least 16 in affected alveoli and airways. Other reported but rare lesions
somatic serotypes. In the US, pasteurellosis of swine is caused by include acute necrotizing pharyngitis, fibrinous polyarthritis
serotypes A and D. without joint swelling, meningitis, and/or meningoencephalitis.

Epidemiology Diagnosis
Ubiquitous in swine, P multocida is probably introduced into History, signs, and remarkable pulmonary lesions may be sugges-
most herds by carrier swine, with subsequent vertical and lateral tive of pasteurellosis, but culture of lung lesions from a non-med-
icated pig is required for confirmation. Histopathology can con-
transmission to other pigs. Transmission from dam to offspring
firm typical microscopic lesions and can implicate or rule out roles
occurs early in lactation, and lateral transmission is via nose-to-
for common primary pathogens. Pasteurellosis is often secondary
nose contact. Although many other species harbor P multocida,
to one or more of a host of primary infectious lung diseases. Since
there is little evidence of interspecies transfer of pathogenic P the agent is common, easily isolated, contributes to lesion severity,
multocida to swine. and persists in lesions, P multocida is frequently isolated from
lungs at the same time as other primary agents of pneumonia
Pathogenesis (Mycoplasma hyopneumoniae, Actinobacillus, influenza A, and
Healthy pigs are highly resistant to development of pasteurellosis porcine reproductive and respiratory syndrome virus [PRRSV]).
when experimentally inoculated with only P multocida. Many Serology and isolation of P multocida from nasal swabs are of little
normal swine carry P multocida in their nasopharynx. Chronic, diagnostic value. Non-pathogenic commensals are common and,
progressive pneumonia occurs when pulmonary immune func- in contrast to the use of polymerase chain reaction for toxins asso-
tion has been comprised, allowing the organism to colonize ciated with progressive atrophic rhinitis, tests are not available to
deeper portions of the lung. be able to predict P multocida pathogenicity in lung.

44 PNEUMONIC PASTEURELLOSIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Environmental factors such as dusty or overcrowded conditions, Control methods include all of the biosecurity measures that might
excessive ammonia gas, or poor ventilation may be important prevent introduction of a new strain of P multocida into a herd.
predisposing factors for pasteurellosis. Only when all other pos- Herd additions should be quarantined for at least a few weeks
sible causes have been eliminated should there be a diagnosis of before entry into a herd. Management practices including use of
primary pasteurellosis. Nevertheless, P multocida can cause severe all-in, all-out production with age segregation that minimizes
disease and is an important component of respiratory disease commingling will reduce the likelihood of pasteurellosis occurring.
when present with typical lesions. Mixing and sorting of pigs should be minimized. Importantly,
implementing control of other respiratory pathogens (eg, M
Control hyopneumoniae, influenza A, PRRSV, Actinobacillus pleuropneumo-
Individual acutely affected pigs require immediate antimicrobial niae) is critical to prevent outbreaks of pasteurellosis. Identification
intervention, usually by injection. Basic therapeutic caveats of and elimination of other diseases, environmental or air quality
early diagnosis, appropriate choice of antimicrobial, and appro- concerns, and other stress factors that might lower resistance are
priate dose with sufficient duration of therapy are particularly also critical components of a control program.
important for success. Many different antibiotics are available for Bacterins for P multocida are available, but their efficacy in
injection or water medication at therapeutic levels. Mass medi- preventing or minimizing the severity of respiratory disease is
cation may be warranted while identifying and controlling the questionable.
primary initiators of the pasteurellosis.

PNEUMONIC PASTEURELLOSIS 45
SWINE DISEASE MANUAL
BACTERIA

Salmonellosis
Alternate names and abbreviations Etiology
None Salmonellae are small, hardy, ubiquitous, Gram-negative bacilli
which contain endotoxin and are capable of elaborating a variety
Definition of other toxins. Isolation and propagation on a variety of culture
Salmonellosis is the disease caused by infection with any of more media have been well-described.
than 2000 Salmonella serotypes. In swine, only a few serotypes Two species of Salmonella are recognized: S enterica and Salmo-
are primary causes of disease. Disease may be manifested as nella bongori. Salmonella enterica is the type species and is further
enterocolitis or septicemia with localization of infection at divided into 6 subspecies that include over 2600 serotypes which
various tissue sites. Asymptomatic Salmonella infections are are serologically clustered into serogroups (Table). Further iden-
common. Swine may serve as a source of Salmonella infection to tification to serotype usually has to be done by reference laborato-
humans via contamination of pork products. ries though some large veterinary laboratories are equipped to do
serotyping for a limited number of strains.
Occurrence
Infection of swine and most vertebrates with a broad range of Primary pathogenic serotypes for swine are relatively few, with
Salmonella serotypes is common and occurs worldwide. Clinical most outbreaks caused by S Typhimurium, S Choleraesuis, or
disease is much less common than infection and occurs through- Salmonella ser Heidelberg. The serotypes that most commonly
out the year in all major swine-raising countries. All age groups cause disease in both people and swine include serotypes S Typh-
are susceptible, but the disease usually occurs in newly weaned imurium and its serologic variants (eg, S 1,4,[5],12:i:-), S Heidel-
or grow-finish pigs. Many other species, including humans, are berg, Salmonella ser Agona, and Salmonella ser Infantis. Salmo-
susceptible to salmonellosis caused by some of the Salmonella nella Choleraesuis and Salmonella ser Typhisuis are host-adapted
serotypes that infect swine. Contaminated pork products are not to swine and are rarely isolated from sources other than swine.
a primary source of food-borne salmonellosis outbreaks in peo- Salmonellae readily survive in many swine environments but can
ple, but efforts to reduce Salmonella in the pork food chain are a be inactivated by chlorine, iodine and phenol-based disinfectants.
high priority for the swine industry.
Epidemiology
Historical information Host-adapted S Choleraesuis is the cause of swine paratyphoid, a
In 1884, the organism now known as Salmonella enterica ser severe systemic disease of pigs; this was the major manifestation
Choleraesuis was isolated and erroneously reported to cause clas- of the disease caused by Salmonella throughout the 20th century.
sical swine fever. After the viral etiology for classical swine fever Since the 1990’s, cases of paratyphoid have decreased precipi-
was identified, salmonellae were considered only as secondary tously in modern swine industries as a result of elimination of
pathogens. However, some Salmonella serotypes frequently act the serotype from breeding herds, better hygiene, adoption of
as primary pathogens causing septicemia and/or enterocolitis in multisite, age-segregated production systems, improved biose-
swine. Species-adapted Salmonella serotypes are relatively infre- curity, and some use of effective vaccines. However, salmonel-
quent but cause severe typhoid or typhoid-like disease in host losis has not gone away in modern industries but has instead
species. Most serotypes, including Salmonella ser Typhimurium been replaced by S Typhimurium and its serological variants
and its serological variants (eg, S. 1,4,[5],12:i:-), are capable of (eg, S 1,4,[5],12:i:-); the reasons for this are unknown.
infecting a broad range of vertebrates.
Salmonellae are commonly found in intestinal tracts of most
Outbreaks of salmonellosis in people are often food-borne from warm and cold-blooded vertebrates. Fecal contamination of
products of animal origin, including pork. Both animal and premises, feed, or fomites from any species is a possible source of
human health could be improved if Salmonella could be effec- Salmonella introduction to a herd. However, it appears that most
tively controlled across animal production and food processing introductions of the disease are a result of the introduction of
systems. Salmonella reduction programs at farm level constitute asymptomatic carrier pigs. Clinically normal pigs are inapparent
a logical step in minimizing the occurrence of outbreaks in carriers of a variety of serotypes of Salmonella infections which
humans. Swine industry-led national Salmonella control pro- persist in their tonsils, intestine, lymph nodes, or gall bladder.
grams in North America and Europe are an effort to understand Individual carrier pigs may regularly or occasionally shed Sal-
what management strategies are most likely to minimize the monella in their feces and are a major source of exposure to sus-
impact of the organism on both human and animal health. ceptible cohorts. A variety of stressors can exacerbate shedding,
including weaning, abrupt feed changes, transportation, com-
mingling, and environmental stresses all of which may precipitate
outbreaks of salmonellosis. Once introduced, salmonellae are

46 SALMONELLOSIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Table: Common serotypes of Salmonella enterica within serogroups B, C1, C2, D and E isolated from US pigs.

Group B Group C1 Group C2 Group D Group E


Abortus-equi Choleraesuis Hadar Dublin Anatum
Agona Infantis Kentucky Enteritidis Meleagridis
Brandenburg Lille Litchfield Miami Muenster
Bredeney Mbandaka Manhattan Riefild New Brunswick
California Montevideo Muechen Orien
Derby Tennessee
Heidelberg Newport
Saint Paul
Schwarzengrund
Typhimurium
Thompson
Senftenberg
Uganda

spread by the fecal-oral route, in contaminated feed and water, increase shedding by inapparent carriers, contamination at swine
and sometimes by short distance aerosol. Exposed pigs may concentration points and in transportation vehicles, and com-
either develop clinical signs of salmonellosis or be colonized and mingling of pigs in holding pens at an abattoir. Efforts to decrease
remain healthy. Although individual pigs may eventually clear the the prevalence of inapparent carriers and prevent exposures from
infection, any pig exposed to Salmonella should subsequently be occurring during the marketing chain are major areas of focus in
considered as a possible carrier of the organism. food-borne Salmonella reduction programs.
Salmonella-contaminated buildings and pens can remain infectious
for weeks if not thoroughly cleaned and disinfected. Although feed Pathogenesis
is a common source of many serotypes of Salmonella, a notable Salmonella vary considerably in virulence, depending on which
exception is S Choleraesuis, which has not been reported in feed. of more than 200 known genetically or plasmid-determined vir-
Most serotypes are capable of infecting a broad range of hosts, so ulence cassettes and pathogenicity genetic elements they possess.
livestock, poultry, wild birds and mice (as well as feces-contaminated In enteric salmonellosis, mucosal damage and necrosis are the
feedstuffs) are believed to be important disseminators of Salmonella. result of mucosal ischemia caused by vasculitis and microvascular
thrombosis. Vasculitis is present in all forms of salmonellosis.
Salmonellosis often appears during or following stressful events Diarrhea is attributed to tissue fluids leaking from damaged
(eg, prolonged transport, drought, overcrowding, change of mucosa and compromise of mucosal absorption. An enterotoxin
rations, parturition, prolonged treatment with drugs or antibi- similar to that produced by enterotoxigenic E coli (causing cyclic
otics) or secondary to an outbreak of another disease in a herd. AMP-mediated hypersecretion of fluids and salts from entero-
Stressful events can lower immune resistance and/or increase the cytes) has been described for S Heidelberg.
magnitude of Salmonella shedding and therefore precipitate out-
Transient bacteremia can occur with a variety of serovars but the
breaks. A balance of virulence, dose, and resistance exists between
host-adapted S Choleraesuis is an exceptional example that causes
swine and the Salmonella they carry.
septicemia. Following ingestion or inhalation of this serotype,
Salmonella contamination of pork products as a source of food- S Choleraesuis invades the tonsil and/or intestinal mucosa and
borne salmonellosis in humans is a global concern. Market pigs spreads to regional lymph nodes. Other Salmonella serotypes usu-
exposed to new serotypes of Salmonella can be rapidly colonized, ally remain confined to the intestine and occasionally the adjacent
with high numbers of organisms detected in intestinal contents lymph nodes, producing only a transient septicemia. Host-adapted
and regional lymph nodes within hours of exposure occurring S Choleraesuis organisms, however, are phagocytized, survive
during the transport or lairage processes. The marketing chain within macrophages and neutrophils, and then are transported
provides many potential sources of Salmonella contamination, to other sites (eg, lungs, liver, meninges, joints, and lymph nodes)
including transportation and feed withdrawal stresses that where they localize and cause further severe tissue damage.
SALMONELLOSIS 47
SWINE DISEASE MANUAL
BACTERIA

Clinical signs may be only congestion with superficial fibrinous inflammation


Salmonellosis presenting as diarrhea with enterocolitis is in cecum, colon, or ileum. In long-standing cases, the intestinal
usually caused by S Typhimurium and its serological variants lesions resemble those of the enterocolitic form of salmonellosis.
(eg, S 1,4,[5],12:i:-) and less often by S Heidelberg or other sero-
types. Enteric salmonellosis mostly occurs in pigs from weaning Diagnosis
to about 5months of age. Initial signs include moderate anorexia History, clinical signs, and typical gross lesions may be adequate
and diarrhea that may be watery, yellow, and intermittent. Ini- for a presumptive diagnosis. Diagnosis should be confirmed by
tially, animals are moderately febrile, but temperatures usually laboratory culture and identification of Salmonella, especially
decrease as diarrhea continues. As the disease progresses, there if the disease is new on the premises. Sero-grouping is readily
may be mucus, fibrin, and blood in the feces, although blood is available and can aid in determining potential serotypes involved
not a prominent feature of enteric salmonellosis. Over time, mor- (Table 1). Serotyping is available in some reference laboratories
bidity is high and mortality is moderate. Surviving animals often and is sometimes useful in understanding the epidemiology of an
become emaciated or gain weight slowly. outbreak.
Historically, septicemic salmonellosis is caused by host-adapted There are several serological assays for investigating or diagnosing
S Choleraesuis. It is most commonly seen in pigs from weaning to outbreaks of Salmonella. Most are based on enzyme-linked
about 180 pounds, although the disease can occur in all ages. Lac- immunosorbent assay and typically exploit known differences in
togenic immunity is usually protective. The onset of an outbreak the lipopolysaccharides or other surface antigens to detect the
may be signaled by finding a pig dead within a group of appar- different serotypes. These assays are used on a herd basis only as
ently thrifty pigs. Within a few days, the disease spreads to other they lack sensitivity and specificity for individual animal use.
pigs in the group. Morbidity is usually low to moderate, but mor- Necropsy and sampling of typical, untreated, acutely affected pigs
tality is high among those that become ill. Clinical signs include for bacteriologic culture and histopathologic study is usually war-
inappetence, depression, huddling, weakness, temperatures up to ranted. Microscopic examination of tissues will help identify typ-
107°F (42°C), and red to purple discoloration of the skin of the ical lesions and differentiate them from those of other enteric and
extremities (cyanosis). Diarrhea is not present initially but fre- septicemic diseases that are commonly present and may be pri-
quently occurs after a few days of illness. The length of course is mary risk factors for salmonellosis expression. Common agents of
variable, both in individual animals and in the herd. Localization post-weaning diarrhea, notably coronaviruses, rotaviruses, E coli,
of S Choleraesuis in the lungs, brain, meninges, lymph nodes, dysbacteriosis, or dietary hypersensitivities often have secondary
or - less commonly - joints can lead to mild pneumonia with dys- or concurrent infections with Salmonella. Other differentials
pnea, lameness with reluctance to move, or vague central nervous include ileitis, whipworms, and swine dysentery, the latter two of
system signs. which are restricted to the large intestine. Host-adapted S Chol-
eraesuis causes rather unique paratyphoid nodules in the liver.
Lesions Septicemic salmonellosis must be differentiated carefully from
With enteric salmonellosis, the large intestine and the lower small classical swine fever, erysipelas, and other septicemias, as well as
intestine are heavy, edematous, and the wall is thickened. There other causes of interstitial pneumonia.
may be excess peritoneal fluid and strands of fibrin on the serosa.
The ileocolic and mesenteric lymph nodes are usually markedly Control
enlarged, congested, and edematous. There are usually focal, mul- Although prevention is preferred, outbreaks of clinical salmonel-
tifocal, or diffuse areas of mucosal necrosis with fibrinonecrotic losis do occur and proper treatment with appropriate antimicro-
exudates in colon, cecum, and/or small intestine. In chronic bials is warranted. The objective is to decrease mortality, reduce
cases there may be raised, circular lesions in the colon referred to the duration and severity of production losses, and to decrease
as button ulcers. Dry gangrene may occur on the ears or tail of shedding in order to minimize transmission of the organism to
survivors. other pigs or cohort groups.
With septicemic salmonellosis, gross lesions include red to Numerous antibiotics and chemotherapeutic agents have been
purplish discoloration of the extremities, marked enlargement used for prevention and/or treatment. Visibly sick pigs may be
of the spleen, moderate swelling of the liver (perhaps with slight injected with an appropriate, approved, systemic antibiotic, par-
icterus), severe pulmonary congestion and edema, and swollen ticularly if there is clinical evidence of septicemia. This treatment
and edematous lymph nodes. Frequently there will be petechial is usually augmented by concurrent mass medication through
hemorrhages in skin, larynx, lungs, heart, bladder, and kidneys. water, usually with an aminoglycoside. There is no evidence that
The liver may or may not contain small, white to yellow foci of treating infected pigs with antimicrobials increases the likelihood
necrosis (paratyphoid nodules). Less frequently observed is syno- of inducing a Salmonella carrier status as has been suggested in
vitis in joints or fibrinopurulent meningitis. Intestinal lesions vary humans. Prophylactic use of antimicrobials in the feed is widely
in severity, largely related to duration of illness. At onset there

48 SALMONELLOSIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
practiced but not preferred because of eventual antimicrobial dard of sanitation. Early weaning followed by removal of piglets
resistance. to a separate, clean site has been successful in minimizing trans-
mission of S Choleraesuis from breeding populations to their
Prevention relies on sound management that is properly and
offspring.
consistently executed. Breeding stock should be acquired only
from suppliers with no history of salmonellosis. If feasible, all There are several efficacious avirulent live vaccines for control of
breeding stock should be from a single source. Essential addi- S Choleraesuis and S Typhimurium. Killed Salmonella bacterins
tions to the herd should be made only after a quarantine period and mutant-derived endotoxin vaccines are available but thought
of 30 to 60 days. to be less effective. All vaccination programs should be coupled
with proper sanitation, husbandry, and pig flow.
Ideally, only pigs from a single source and of similar age should be
raised together in a closed, all-in, all-out system. Buildings should Successful reduction of Salmonella infections in swine herds has
be cleaned and disinfected carefully between batches. All possible been reported in Europe and the US. The programs are based
carriers, including rodents, birds, pets, and wildlife should be largely on serological testing of breeding stock, culture of sam-
eliminated or excluded from contact with pigs. Unusual stresses ples taken at slaughter, establishment of measures that reduce
should be avoided insofar as possible. Perhaps the most import- contamination of carcasses at slaughter, and controlling risks that
ant approach to control of salmonellosis involves provision of are likely to result in the introduction of Salmonella through
comfortable, dry housing, maintaining proper stocking density, feed or drinking water. The hazard analysis critical control point
managing the ventilation system, and implementing a high stan- (HACCP) programs instituted by US abattoirs have adopted
many of these measures. 

SALMONELLOSIS 49
SWINE DISEASE MANUAL
BACTERIA

Streptococcus suis
Alternate names and abbreviations Historical information
Strep, Strep encephalitis Outbreaks of streptococcal infection in swine occurred many
years before the organism S. suis was identified in 1987. Out-
Definition breaks of S suis infection now are reported frequently. Because
The predominant streptococcal disease of swine is caused by S suis colonizes piglets at birth, neither segregated early weaning
Streptococcus suis. Streptococcus suis is the most important strepto- nor medicated early weaning programs are able to prevent S suis
coccal infection of pigs and is usually seen in nursing or recently transmission to piglets.
weaned piglets. The disease is usually characterized by septicemia
There has been much research on S suis. Early research was largely
with tissue localizations, which can present as acute meningitis,
focused on the study of capsular serotypes and comparison of
polyarthritis, polyserositis, or bronchopneumonia.
signs and lesions produced by different serotypes; identification
Other less common, largely sporadic, streptococcal infections of virulence factors has been the focus of more recent research.
also occur in pigs. Sporadic infections are occasionally caused by Experimental infection models vary widely and likely contribute
Streptococcus dysgalactiae subspecies equisimilis. This beta-hemo- to the lack of agreement in findings reported in published studies.
lytic Streptococcus and a few others are endemic opportunists that
sometimes cause arthritis, septicemia, or meningitis in young pigs, Etiology
complicate other disease processes, and may contribute to cases of Streptococcus suis is Gram-positive, alpha-hemolytic, and easily
vegetative valvular endocarditis in older grow-finish pigs. These grown on blood agar. Identification is based on biochemical
organisms are indigenous on the skin (including the vagina), and reactions and capsular serotyping. There are about 35 capsular
it has been shown that most piglets become infected at the time serotypes. Their prevalence varies in different geographic regions
of birth. Cases in young pigs are a result of navel infection, breaks of the US, with types 2, 3, 4 and 7 predominating in the Midwest.
in the integument due to knee abrasions, or infection of skin Type 2 may be most prevalent worldwide and is the most likely to
wounds, including those caused by tail docking, ear notching, or express virulence. Virulence factors are not well understood, but
clipping of needle teeth. capsular polysaccharide and 2 proteins, muramidase released pro-
tein and extracellular factor, are thought to play important roles
Streptococcus porcinus is the same as Lancefield group E Strepto-
in the severity of disease caused by infection with type 2.
coccus and is associated with jowl abscess (feeder boils) in swine.
This disease was once quite prevalent but is now rare, presumably Many species of Streptococcus can be isolated from normal swine.
because of improvements in husbandry and feeder design. It is There is no single, consistent predictor of virulence, immunity,
occasionally isolated from pigs with septicemia or abscesses. or cross-protection. Environmental or other predisposing fac-
tors may influence pathogenicity. Most Streptococcus are readily
Streptococcus durans has been associated with enteritis in suck-
destroyed by common disinfectants such as phenol, quaternary
ling pigs. Additional streptococci are sporadic opportunists
ammonium, formaldehyde, hypochlorite, chlorhexidine, and 3%
isolated from the respiratory tract, mammary glands, localized
iodine.
skin lesions, subcutaneous abscesses, the reproductive tract of
aborting sows, and sows with fertility problems or agalactia.
Their role in these conditions is not clear but probably driven by Epidemiology
other risk factors. Streptococcus suis survives in dust and feces in the usual swine
environment. It can be isolated from the nasal cavity, palatine
tonsils, and genitalia of many normal pigs. It is present in the
Occurrence feces and nasal secretions of carriers. Transmission may be
Although S. suis infection occurs primarily in nursing or
through ingestion, inhalation or nose-to-nose contact. Flies and
recently weaned pigs, it can affect pigs of all age groups. The
rodents may play a role in mechanical spread. Streptococcus suis
disease is found in all major swine-raising countries. It is rel-
can survive on flies for at least 5 days.
atively common, occurs throughout the year, and is prevalent
in confinement-raised swine. Streptococcus suis infection also Sows infect piglets with the bacterium during the birthing pro-
occurs sporadically in several domestic animals (cattle, sheep, cess or shortly thereafter. Contaminated secretions, excretions,
goats, horses, dogs). It is zoonotic and occurs occasionally in and the skin of infected pigs serve to expose other pigs to the
people who work with pigs or handle meats (butchers, farmers, organism. These factors make early weaning programs, with or
and veterinarians), causing localized infections, arthritis, septi- without medication, ineffective in preventing transmission to at
cemia, and sometimes death. least some piglets that may then serve as sources of infection for
the rest of their cohort during lactation and nursery phases.

50 STREPTOCOCCUS SUIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Wounds also are a common route of entry for S suis. Once a few meningitis microscopically, seen grossly as visible clouding of
piglets are infected, they are able to disseminate the bacteria to the leptomeninges of the brain. In septicemic cases there may be
other litters or commingled pigs. Close contact favors spread. slight enlargement of the spleen and petechial hemorrhages on
The role of fomites and insects has been demonstrated but their the kidneys or other organs. With polyarthritis there is enlarge-
importance remains uncertain. ment of joints that contain turbid synovial fluid.
Multiple serotypes or genotypes may exist on the same farm. Streptococcus suis pneumonia, often secondary, has a broncho-
Control of one particular type of S suis may lead to emergence of genic cranioventral distribution pattern of firm consolidation
another serotype, frustrating long-term control strategies. in lung. Predominance of any one of these lesions varies in
outbreaks. Polyserositis is quite common. Other lesions seen in
Pathogenesis chronic cases include endocarditis (usually on the mitral valve),
Pathogenesis is somewhat speculative and may differ among myocarditis, and fibrinopurulent to fibrous pericarditis.
serotypes. Management and husbandry stresses may predispose
piglets to outbreaks. Also, preexisting injury to respiratory epi- Diagnosis
thelium, especially rhinitis, may predispose to infection. Known A presumptive diagnosis can often be made on the basis of his-
causes of rhinitis include ammonia fumes, infection with Borde- tory, signs, lesions, and the demonstration of Gram-positive cocci
tella bronchiseptica and Pasteurella multocida, and inclusion body in the lesions. Confirmation should be made through culture
rhinitis. Interaction of S suis with viruses (eg, porcine reproduc- and identification of the Streptococcus. Both pathogenic and
tive and respiratory syndrome [PRRS], pseudorabies, and influ- non-pathogenic strains can be recovered from pigs on most farms,
enza A) may trigger outbreaks. Overcrowding, poor ventilation, so demonstrating organisms from systemic locations (brain, joint,
and high humidity also favor disease expression. fibrinous exudates) with typical gross and microscopic lesions, in
addition to a typical clinical picture, is important in establishing
After oral or respiratory exposure, streptococci can be found in
the correct diagnosis. If convenient, the capsular type should be
crypts of the palatine tonsils or in nasal mucosa. From there, the
determined since that can assist in selecting an appropriate vac-
organism can become systemic, spreading through the blood
cine. The predominate capsular type may change over time within
stream to multiple organ systems. Some organisms may survive
a farm, so periodic typing of S suis organisms recovered from
within phagocytes and gain access to the cerebrospinal fluid,
affected pigs may have value as an epidemiologic tool. Outbreaks
brain, meninges, lungs, and joints where they localize and cause
are frequently associated with a variety of infectious and non-in-
meningoencephalitis, arthritis, or pneumonia. In pneumonic
fectious risk factors, so a complete diagnostic evaluation of agents
lungs, S suis can be either a primary or secondary pathogen.
as well as risk factors in the environment is usually warranted.
Clinical signs Control
Signs and lesions are similar in all S suis infections, regardless of
At present there are no highly effective measures for control of S suis.
serotype. Signs usually suggest septicemia and/or localization of
Elimination of stresses, particularly overcrowding, poor ventila-
systemic infection. High rectal temperatures (greater than 105°F
tion, high humidity, and inadequate sanitation, should be part of
[41°C]) are common early in the course of infection. Pigs with
the strategy. The all-in, all-out system with emphasis on age seg-
septicemia die after a short, acute course and are sometimes sim-
regation, multi-site rearing, and strict sanitation of facilities and
ply found dead without having ever been observed as sick. Young
instruments used in processing piglets may be beneficial. The con-
piglets with central nervous system lesions are often found in
trol of primary diseases (PRRS, influenza A, pseudorabies, Myco-
lateral recumbency and paddling. Older pigs have a wider variety
plasma  hyopneumoniae, etc) that interact with S suis is important.
of central nervous system signs that include ataxia, opisthotonus,
It may reduce incidence and improve response to treatment.
incoordination, tremors, convulsions, blindness, deafness, or
vestibular syndromes. Pigs with polyarthritis have swollen joints Affected pigs should be treated individually as early as possible
and are lame. In some outbreaks, respiratory signs associated with with injectable antibiotics. Systemically available antibiotics are
pneumonia may occur. Both morbidity and mortality vary widely sometimes pulsed strategically for a few days in feed or water to
and are affected by treatment. Streptococcus suis, much like Pasteu- groups experiencing high morbidity. Some strains of S suis are
rella multocida, is ubiquitous in swine and a frequent secondary resistant to many antibiotics (particularly tetracyclines), so sensi-
contributor to pneumonia. tivity tests are helpful in selecting a therapeutic agent. Because the
type of S suis may differ in consecutive outbreaks, sensitivity tests
Lesions often need to be repeated.
There is often reddening of the skin and modest enlargement of Vaccines and commercial antisera, when available, may be of
lymph nodes. Pigs that die suddenly often have fibrinopurulent value as management tools. The capsular type of the product
being used should be the same as the type causing the disease.

STREPTOCOCCUS SUIS 51
SWINE DISEASE MANUAL
BACTERIA

Not all vaccines have been effective, however, perhaps because


the product used was not appropriate for the serotype causing
the problem, because of a shift to another predominant serotype
within the herd, or because of a failure to address underlying risk
factors. Proper acclimatization of gilts and perhaps vaccination of
sows prior to farrowing may be helpful for control in suckling pig-
lets but is of less value for control of post-weaning streptococcosis.
Streptococcus suis can be an important zoonotic pathogen, with
spread generally thought to be a result of direct inoculation
through skin wounds. A large outbreak of S suis type 2 infection
in humans was reported from China in 2005 that included a case
fatality rate of approximately 18%.

52 STREPTOCOCCUS SUIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Swine brucellosis
Alternate names and abbreviations Cattle can be infected naturally with B suis. Milk and aborted
None fetuses can transmit the organisms to swine. Milk-borne epidem-
ics have occurred in people and swine. Infected feral swine, if in
Definition contact with domestic swine, are another source of exposure.
An infectious disease of swine characterized by reproductive The bacteremia that occurs during infection of swine often results
impairment or failure, and often by localization of Brucella suis in in infectious carcasses. Abattoir workers can be infected by
bones and joints of breeding stock. handling infectious carcasses or cuts of meat. Farmers and veteri-
narians may be exposed while assisting sows at farrowing or when
Occurrence handling infectious placentas or newborn infected piglets.
Swine brucellosis occurs primarily in domesticated and feral The prolonged survival of B suis in pastures and farrowing houses
swine; wild hares have been proposed as a reservoir of infection is probably a significant factor in the spread of infection, espe-
in some countries. The disease occurs in many countries where cially in colder climates or seasons on farms where management
domestic or feral swine are present in large numbers. and sanitation are poor. Swine moved into infectious lots or
Swine brucellosis once was common in the US, but due to rig- buildings can be infected by surviving B suis.
orous control measures the disease is now very uncommon. The
porcine type of brucellosis occurs as a serious zoonosis, primarily Pathogenesis
among abattoir workers, farmers, and veterinarians. Penetration of mucous membranes by B suis results in the coloni-
zation of regional lymph nodes, which is followed by long, con-
tinuous, or intermittent periods of bacteremia. The bacteria then
Historical information infect and persist in many other organs, including lymph nodes,
Brucellosis was first identified as a disease of swine in 1914 but
placenta, mammary gland, joint fluids, bone, and the testicles or
was believed to be a form of bovine brucellosis caused by Brucella
accessory genital organs of boars.
abortus. In 1919, the porcine organism was named as a separate
species (B suis). Swine brucellosis increased in importance as A large percentage of swine, especially females, recover after
swine production intensified and because of the severe disease several months. The recovery rate in boars is low, perhaps never
caused in people. A cooperative, 3-stage, state-federal-industry more than 50%. Enough animals remain infected in a herd to
eradication program was initiated with a goal of eradication of perpetuate the disease. Surprisingly, most piglets in infected herds
brucellosis. This goal is nearly accomplished in domestic herds, are free of infection at the time of weaning.
but feral swine remain a reservoir in the US.
Clinical signs
Etiology Most owners of B suis-infected herds notice no signs of infection
Under the present classification scheme, B suis is the organism prior to evidence of infertility in the herd. In dams infected at
breeding, disease is first suspected when numerous sows or gilts
that causes most brucellosis in swine. There are 5 biotypes of B
return to heat 30 to 45 days after breeding. Females first exposed
suis. Biotypes 1 and 3 cause generalized infections that result in during late gestation have abortions or dead and weak pigs. Early
reproductive failure. abortions often go undetected.
Brucella suis organisms are moderately resistant to environmental Sows and gilts usually recover promptly after aborting, success-
influences. Survival time of organisms in the environment decreases fully re-breed, then deliver live pigs. Infected boars present few
as temperatures rise. However, the bacteria often survive desicca- signs of infection, although their fertility may be below normal.
tion and can survive freezing and near-freezing temperatures for They may or may not show lack of sexual drive.
over 2 years. Pasteurization of milk kills B suis. Most commonly
In growing pigs, lameness may occur due to joint and bone
used disinfectants, as well as ultraviolet light, kill the organism.
infection with B suis. In mature boars, orchitis, epididymitis, and
infertility are common.
Epidemiology
Brucella suis is usually transmitted to susceptible swine through Lesions
direct contact with infected swine. The route of exposure is In dams, gross lesions are limited except in chronic cases. At the
ordinarily through the alimentary or genital tract. Pigs are often time of abortion, there may be some degree of endometritis.
infected by ingesting aborted fetuses, fetal membranes, or fluids Occasionally, some of the fetal membranes are retained. Infected
discharged at the time of abortion. The organisms are also trans- placentas may not have grossly visible lesions, or may be con-
mitted in infectious semen during natural breeding and artificial gested, edematous, and have small hemorrhages. Infected fetuses
insemination. Transmission through abraded skin, conjunctiva, may have subcutaneous edema, especially around the umbilicus,
and nasal mucous membranes is possible. and there may be effusions in body cavities.
SWINE BRUCELLOSIS 53
SWINE DISEASE MANUAL
BACTERIA

In intact males, reproductive organs with multiple granulomas Direct culture is probably the most accurate and sensitive method
are occasionally accompanied by fibrinopurulent or hemorrhagic of diagnosis of acute cases but is usually less practical than sero-
periorchitis, often resulting in enlarged testes that can become logical testing. Specimens include lymph nodes from slaughter,
atrophied with time. Calcified foci may also be in the testes and blood, semen, castrated testicles, infected joints, vaginal swabs, or
accessory sex glands. aborted materials from suspect cases. The bacteriologist should be
alerted if brucellosis is suspected since colonies are slow-growing
Chronic infections in growing pigs and breeding stock may man-
and easily missed in routine diagnostic investigations.
ifest as fibrinous or fibrinopurulent arthritis. These lesions often
affect the large joints of the legs. Osteomyelitis may be apparent
within vertebrae, especially the lumbar vertebrae, sometimes with Control
paralumbar abscesses present. Prevention is best accomplished by focusing on implementation
of those measures that are most likely to prevent exposure. No
Microscopic lesions include small granulomas in the liver during animals should be added to a herd without having originated
the bacteremic phase of acute infection. Small, focal areas of from a negative herd. Those animals should test negative initially,
inflammation or microabscesses may be found at many sites, be quarantined for at least 30 days, then again test negative before
including the bone marrow adjacent to the epiphyses of long being added to the herd. No effective vaccines are available for
bones. The microscopic lesions are suggestive of brucellosis but immunization of swine.
not specific.
Infected herds can be handled in 2 different ways. If the herd has
many reactors, as is often the case, the premises should be depop-
Diagnosis ulated and allowed to stand idle for at least 2 to 3 months before
Diagnosis is usually made on the basis of serological testing. The
restocking. This is often the least expensive way. Depopulation
buffered, acidified plate antigen test and the standard card test
with slaughter of infected herds is often used in the final efforts at
have been used extensively as presumptive tests. For confirmation,
eradication. If a herd test reveals only one or a few reactors, those
either the standard tube test or the particle concentration fluores-
can be removed, and the herd retested periodically.
cence immunoassay is used. Serological testing is very useful as a
herd test but is not infallible on individual animals. In some cases where valuable bloodlines must be saved, weaned
pigs, usually negative when tested at weaning, can be moved to
All testing for swine brucellosis has limitations. Swine may be
separate, clean premises. They must be retested at least once prior
infected for 6 to 8 weeks before developing antibodies that will
to breeding them to tested, clean boars.
result in a positive test. In uninfected herds, occasional animals that
test positive will later test negative. Infections with several other
genera of bacteria may result in positive brucellosis tests. In particu-
lar, infection of swine with Yersinia enterocolitica has led to positive
tests for brucellosis. The limitations of testing may justify the use of
a slaughter program in the later stages of an eradication effort.

54 SWINE BRUCELLOSIS
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Swine dysentery and spirochetal colitis
Alternate names and abbreviations been considered, the lack of sensitive monitoring tests precludes
Brachyspira (previously Serpulina or Treponema), swine dysen- this effort at a national or regional level. Antimicrobial resistance
tery, SD, porcine intestinal spirochetosis is common.

Definition Etiology
Swine dysentery is a severe infectious disease caused by strongly Brachyspira are short, slender, spiral-shaped bacteria with axial
beta-hemolytic Brachyspira organisms, characterized by muco- filaments that provide serpentine locomotion. The organisms are
hemorrhagic diarrhea and lesions of marked inflammation that Gram-negative, oxygen-tolerant anaerobes, and the strains that
are limited to the large intestine. Porcine intestinal spirochetosis can cause bloody diarrhea (dysentery) are strongly beta-hemolytic
causes milder mucoid colitis in young pigs and is associated with on blood agar, notably B hyodysenteriae and, more recently, B
a variety of weakly beta-hemolytic Brachyspira organisms, most hampsonii and B suanatina.
notably B pilosicoli. Hemolysin activity of Brachyspira is an important virulence
factor and pathogenicity is seemingly proportional to hemolytic
Occurrence activity, with dysentery caused only by strongly beta-hemolytic
Swine dysentery occurs only in swine and is a very important isolates. Isolation of strongly beta-hemolytic Brachyspira colonies
disease causing significant economic and production losses world- on blood agar is probably the most accurate and sensitive method
wide. Other animal species can be transiently infected and shed to confirm that the presence of organisms capable of causing
or harbor pathogenic Brachyspira. Swine dysentery can occur in mucohemorrhagic colitis typical of dysentery. Properly validated
any age pig but is most prevalent from 6 weeks of age through polymerase chain reaction (PCR) tests are useful for screening
adult. The prevalence of dysentery in the US has decreased and speciation but may lack sensitivity to detect all pathogenic
because of successful eradication programs and through wide- variants. Attempts to classify Brachyspira using a variety of other
spread adoption of swine buildings that have fully-perforated techniques have shown genotypic and phenotypic variation, but
flooring, minimizing the recycling of the causative agent between have not proven useful to predict virulence, pathogenicity, immu-
pigs and between subsequent groups. Outbreaks occur through- nogenicity, or immune cross-protection.
out the year.
Brachyspira pilosicoli and other weakly beta-hemolytic Brachyspira
With an increased interest in niche pork production in the US species, including B murdochii, B intermedia, and B innocens, are
(eg, organic, antibiotic-free, and free-range systems), there is the common in swine and can be isolated and/or detected by specific
potential for a resurgence in swine dysentery. PCR testing. These agents are common inhabitants of swine
colons and may cause or exacerbate other causes of colitis in
Spirochetal colitis, associated with B pilosicoli, also has worldwide
young and growing swine, but they do not cause swine dysentery.
distribution but is associated with a milder, non-hemorrhagic,
mucoid colitis. Several other Brachyspira species may contribute
to mild mucoid colitis as well. Epidemiology
Transmission of Brachyspira occurs by the fecal-oral route. Feces
or any materials soiled with feces are sources of infection, along
Historical information with carrier swine or other animal species capable of harboring
Swine dysentery was first reported in 1921 and was subsequently
organisms. Brachyspira persist in lagoon water and moist feces
described in many swine-raising countries throughout the world.
for at least 2 months and soil for 3 weeks. Mice can shed strongly
The etiology remained unknown until the isolation of Brachyspira
beta-hemolytic Brachyspira for at least 180 days and dogs can
hyodysenteriae in 1971, which allowed characterization and more
shed for at least 13 days. Birds, waterfowl, flies, and fomites can
focused research during the 1970s and 1980s. With the advent of
harbor and transmit organisms as well. Asymptomatic carrier
genetic sequencing, additional strongly beta-hemolytic Brachy-
swine can transmit the agent for at least 90 days. The introduc-
spira species (B hampsonii and B suanatina) have been associated
tion of inapparent carriers, including breeding stock, is often the
with swine dysentery. Spirochetal colitis associated with B pilosi-
source of initial exposure. Contaminated transport vehicles are
coli was described in the late 1980s.
frequently implicated as well. Carrier sows can intermittently
Prior to the discovery of organic arsenicals and effective antimi- transmit Brachyspira to their piglets. Infected mice on the prem-
crobials as treatments and preventatives, dysentery caused disas- ises may also be an ongoing source of infection, something to
trous losses among swine herds. Improved methods of control keep in mind when considering elimination strategies.
and elimination have markedly reduced losses in the US, but
Brachyspira survive under a wide range of environmental con-
dysentery remains a very important disease globally. Methods of
ditions but are susceptible to heat, ultraviolet light, desiccation,
elimination of dysentery from individual swine herds have been
soaps, most disinfectants, and alkaline environments.
well described. Although national eradication programs have

SWINE DYSENTERY AND SPIROCHETAL COLITIS 55


SWINE DISEASE MANUAL
BACTERIA

Pathogenesis Microscopy reveals moderate, non-suppurative colitis and typhli-


After ingestion of pathogenic Brachyspira, clinical signs of tis, mucus metaplasia, crypts dilated with mucus and organisms,
diarrhea can develop in 5 to 21 days (or more). The incubation mucosal congestion and edema, and superficial epithelial necro-
period is dependent upon dose, herd immunity, and individual sis. Spiral-shaped organisms are demonstrable using silver or
pig susceptibility, but the infective dose is quite small. Diet can immunohistochemistry staining.
influence prevailing microflora to facilitate or ameliorate colo-
Brachyspira pilosicoli appears to exert its pathogenic effect by
nization and lesion formation. The organism reaches the large
attachment to enterocytes and creating microerosions in the
intestine, colonizes, proliferates, penetrates the mucus layer,
epithelium. Lesions of spirochetal colitis are milder that those seen
becomes closely associated with epithelial cells, and causes a flux
with dysentery, with mild mucosal hyperemia and excess mucus
of mucus. This is followed by superficial epithelial erosion and
observed on the mucosa of large intestine. Microscopically, there
hemorrhage. Invasion may not be essential for lesion production.
may be abundant spiral-shaped organisms colonizing epithelium,
The exact mechanisms of tissue destruction are not known, but
accompanied by mild non-suppurative inflammation.
toxins, hemolysin, and lipopolysaccharides are probably involved.
In typical lesions, organisms are present within crypts, epithelial
cells, and occasionally in the lamina propria. The lack of lesions Diagnosis
at other sites suggests the entire pathogenesis occurs locally A field diagnosis of dysentery is based on clinical signs, necropsy
within the large intestine. Systemic effects of the disease include findings, and typical gross lesions that are limited to the large
dehydration, acidosis, and electrolyte imbalance caused by colitis. intestine. Demonstrating high numbers of spirochetes in smears
Colonic inflammation can be severe and prolonged. made from colonic scrapings or feces using crystal violet or Vic-
toria blue stains adds confidence to the field diagnosis. However,
other Brachyspira species are common flora in some herds and
Clinical signs are morphologically indistinguishable from B hyodysenteriae, B
Disease is most common in pigs from 6 weeks of age to market
hampsonii or B suanatina on a smear.
but can occur in any pig from 3 weeks of age through adults. The
appearance of gray to yellow, mucoid diarrhea is often one of the Laboratory confirmation is usually warranted, especially if the
first signs of the disease. With dysentery, diarrhea continues and outbreak is new, its impact is high, or there is doubt about the
quickly becomes mucohemorrhagic with excess mucus and fresh true cause of the clinical signs. Tissues should be collected for
blood apparent. In a small proportion of pigs, diarrhea may be culture and histopathology from acutely affected non-medicated
preceded by tail twitching or a humped, gaunt appearance. Fresh pigs. Culture of a strongly beta-hemolytic Brachyspira from
red blood in mucus-containing feces is often profuse with abun- colonic scrapings is the most accurate and sensitive way to con-
dant perineal staining. Prolonged diarrhea leads to dehydration, firm diagnosis. Biochemical differentiation of pathogenic and
electrolyte imbalance and acidosis, sunken eyes, marked weak- non-pathogenic strains is not consistent. Polymerase chain reac-
ness, hollow flanks, and weight loss. Anorexia is common, but tion techniques are available for detection of some Brachyspira
affected animals usually continue to drink. Morbidity is greater and can provide differentiation to the species level, but specificity
than 50% and mortality can approach 50% in untreated herds; limitations of molecular technologies can lead to false negative or
affected pigs may remain stunted even after successful treatment. false positive tests, especially when only feces are tested.
Spirochetal colitis is generally seen as a mild diarrhea accompa- Several diseases can mimic or occur simultaneously with dysen-
nied by excess mucus. Affected pigs may be less thrifty, but effects tery: ileitis, whipworms, salmonellosis, gastric ulcers, and intes-
on growth performance may not be overtly obvious. tinal accidents. Severe whipworm infestations can very closely
mimic the gross lesions associated with dysentery, particularly as
Lesions the worms may not be grossly visible until perhaps 3 to 4 weeks
Lesions associated with Brachyspira are limited to the large intes- after exposure to the whipworm ova. Lesions of salmonellosis
tine. With dysentery, the mesentery and serosa are edematous extend deeper into the mucosa than those seen with dysentery
and hyperemic. In acute cases, the mucosa of the affected large and tend to present as small ulcers, which may be multifocal in
intestine is diffusely reddened, edematous, and swollen. There distribution and have much less mucus and fresh blood than dys-
entery. Salmonellosis lesions are not usually confined to the large
may be only small amounts of fibrin but copious mucus. Any or
intestine. Though ileitis typically affects the small intestine, some
all parts of the large intestine may be affected. As lesions progress,
cases seem to have a predilection to cause lesions only in the large
the colon wall thickens with inflammation, congestion, and
intestine; in these cases, histopathology can easily differentiate
edema and the mucosa becomes hyperemic with excess mucus the disease from dysentery.
and flecks of blood. Further progression leads to the mucosa
becoming covered, diffusely or in patches, with layers of fibrin, Spirochetal colitis is best diagnosed by necropsy of acutely
necrotic debris, mucus, and blood. Pigs that die suddenly may be affected pigs, followed by histopathology on affected tissues,
in surprisingly good body condition but have extensive lesions in isolation of the weakly beta-hemolytic causative organism, and
the large intestine. definitive identification by biochemical tests or PCR.

56 SWINE DYSENTERY AND SPIROCHETAL COLITIS


DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
Control Any effort to eradicate dysentery requires an initial accurate
Preventing the introduction of pathogenic Brachyspira to nega- diagnosis of dysentery, a high level of commitment from the
tive herds through strict biosecurity measures is a high priority. owners and workers, and an agreement to consistently execute
Additions to the herd should be made only from herds known to the herd-specific protocol with ongoing veterinary supervision.
be historically free of dysentery and only after a quarantine period Successful implementation of a dysentery elimination project
of 30 to 60 days. requires exquisite attention to and compliance with the details
laid out in the implementation plan. The eradication effort
Elimination of dysentery from farms can be successful with should be made during the warmest time of the year. A profes-
scrupulous attention to the details of a well-designed plan. One sionally managed rodent control program is an essential part
method of elimination commonly used in the US relies on the of any eradication program. Eradication must be coordinated
use of a medicated early weaning protocol, whereby piglets are with pig flow so that thorough cleaning and disinfection can
weaned to a clean site concurrent with implementation of an be accomplished when the premises is vacant or the population
intensive off-site breeding program. While the infected sow minimal. If infected animals must be retained during the effort,
farm is depopulated and the facilities cleaned and disinfected, an a barrier system must be set up to separate clean from potentially
off-site breeding program establishes a disease-free population contaminated areas. In contrast to the diagnosis of disease in
of sows that can eventually be relocated back to the site that was overtly affected animals, detection of non-clinical carriers can be
previously depopulated. Variations of this program exist and have difficult. Repeated sampling of non-medicated pigs over time is
also been quite successful in eliminating dysentery. A second often required.
elimination method relies more heavily on use of antibacterial
medication and is generally only applicable to the breeding herd Producers trying to control endemic dysentery generally rely
population in a multi-site production system. In this method, on the use of preventive and therapeutic levels of antibiotics
the herd population is reduced as far as practical to decrease administered in the feed and/or water. Pulse-dosing or contin-
medication cost. An intensive oral medication regimen of all uous administration, coupled with good sanitation and manure
animals then follows. Debilitated animals must be culled before removal, can be effective but is costly. Development of antimicro-
the eradication program since they may not consume enough bial resistance is common, making control programs (as opposed
medicated feed or water to eliminate the infection. Few antimi- to elimination) a poor long-term strategy. Vaccination has not
crobials are effective at complete elimination of the organism, so proved to be reliably effective in control or elimination programs.
product selection should be made with care and directions for its Some dietary considerations (highly digestible, low insoluble
use should be followed carefully. Often, improvements to farm fiber) may be useful in controlling dysentery when coupled with
biosecurity are implemented at the same time as the medication consistent sanitation and manure removal strategies.
program. A third method for elimination is through complete Control of B pilosicoli should follow the same principals of sanita-
depopulation of the farm; this is most useful on farms where age tion and husbandry as described for dysentery. The disease usually
segregation, biosecurity, and sanitation are difficult to implement. responds favorably to the antimicrobials listed for swine dysentery.
Depopulation should be done during warm, dry weather. During
depopulation, all facilities and equipment should be thoroughly
cleaned, disinfected, and idled. During periods of warm, dry
weather, Brachyspira will not usually survive for more than 2
weeks in soil or grass pens. Obviously, significant investment
should be made in confirming that depopulation is followed only
by repopulation with swine known to be free of dysentery.

SWINE DYSENTERY AND SPIROCHETAL COLITIS 57


SWINE DISEASE MANUAL
BACTERIA

Tuberculosis
Alternate names and abbreviations As a genus, Mycobacteria are Gram-positive, acid-fast bacilli. They
TB are highly resistant to most environmental factors, many disin-
fectants, and an increasing number of antibiotics. Mycobacterium
Definition avium complex organisms have survived for more than 4 years in
Tuberculosis is a chronic, granulomatous disease of swine, poul- poultry lot soil, in cages, and in sawdust used as litter.
try, wild birds, many domestic and wild animals, and people.
Epidemiology
Occurrence The marked resistance of Mycobacterium to environmental factors
Tuberculosis occurs frequently in humans, domestic and wild makes contaminated premises a long-term threat to swine. The
animals, poultry, and wild birds. It occurs in almost all vertebrates tuberculosis organisms are usually spread to swine by tubercu-
and in some cold-blooded animals. The disease is present world- losis-infected poultry, wild birds, cattle or people, or by soil or
wide throughout the year. bedding materials contaminated by them. The route of exposure
is usually by ingestion, although spread by inhalation occasionally
The incidence of tuberculosis condemnations in over 100 million occurs. Infectious feeds that have spread tuberculosis bacilli to
swine slaughtered in federally inspected abattoirs of the US was swine include offal from infected poultry and cattle, uncooked
0.003% in 1995 and has remained stable at this low incidence. garbage, and unpasteurized milk and dairy products. Exposure of
swine to lots contaminated by the feces of tuberculosis-infected
Historical information poultry has often led to their infection by M avium complex.
In the US, tuberculosis was once a common disease of poultry,
cattle, swine, and people. A major effort to eradicate tuberculosis Other less common but proven routes of exposure include expo-
from cattle and in people markedly reduced incidence of the sure to serovars of avian type contained in sawdust or wood shav-
classic, severe disease caused by infection with Mycobacterium ings used as bedding, congenital exposure of fetuses of infected,
tuberculosis, M bovis, and closely related subspecies, now referred pregnant sows, and animal-to-animal exposure among swine.
to as Mycobacterium tuberculosis complex. Swine and poultry are Rarely, infected swine have tuberculosis lesions in their tonsils or
susceptible to M tuberculosis complex but are now rarely exposed intestine and shed the organisms in their feces.
to or infected with it. Infection with granulomatous lesions in
swine and poultry is usually caused by M avium and related sub- Pathogenesis
species, referred to as Mycobacterium avium complex. The complex lipid nature of the organisms may explain the diffi-
Changes in the poultry industry leading to the establishment of culty the body defenses have in destroying mycobacteria though
large, integrated production systems largely eliminated the raising phagocytosis and oxidative killing. In mononuclear phagocytes,
of poultry as a private commercial enterprise for small farmers, toxic lipids in the mycobacteria, or factors released by them,
which traditionally had been the source of exposure for swine. As a appear to interfere with phagolysosome formation or to inacti-
consequence, the incidence of tuberculosis in swine has decreased vate lysosomal enzymes. If macrophages fail to destroy ingested
substantially. Reductions were achieved without an eradication mycobacteria, they can carry them to other sites. There, the bacte-
program directed specifically at swine or poultry tuberculosis. ria are liberated upon death of the macrophages, and the released
organisms set up new foci of infection.
An increasing prevalence of tuberculosis in immunocompro-
mised people, coupled with resistance of tuberculosis to many In swine, tuberculosis bacilli appear to initially infect the tonsils
therapeutic agents, continues to be a public health concern for and intestinal mucosa and then spread to the regional lymph
much of the world. nodes, especially those of the cervical area and less often to
mesenteric nodes. Lesions in the nodes tend to develop slowly
Etiology and, in most cases, the bacilli are successfully walled off. Most M
There are at least 177 species of Mycobacterium, many of which avium complex infections are asymptomatic, with granulomatous
are soil saprophytes. Tuberculosis in bovines and humans refers lymphadenitis the predominant outcome.
specifically to disease caused by M tuberculosis complex organ-
isms. Swine and poultry can be infected and can develop lesions Clinical signs
with M tuberculosis complex but are much more likely to be In swine with localized tuberculosis infections, there are usually
infected with M avium complex. This latter group causes gran- no signs. Since most swine are slaughtered at an early age, there is
ulomatous lesions that resemble classic tuberculosis of humans little time for generalization of infection and the appearance of
and bovine and are a public health concern for immunocompro- signs. Condemnation of carcasses at slaughter is the usual presen-
mised humans. tation. In older breeding stock, tuberculosis generalization does

58
DISEASES CAUSED BY BACTERIA, MYCOPLASMAS, AND SPIROCHETES

BACTERIA
occur occasionally. Affected animals tend to waste away despite Diagnosis
adequate feed. They eventually become emaciated and may die A presumptive diagnosis can often be made on the basis
from tuberculosis.  of history, study of gross and microscopic lesions, and the
presence of acid-fast bacteria within discrete granulomas or
Lesions granulomatous tissue. A confirmed diagnosis can be made
In localized tuberculosis, grossly visible lesions are usually only after isolation, identification, and typing of the bacteria.
apparent only in cervical, submaxillary, or mesenteric lymph Polymerase chain reaction and immunohistochemistry are
nodes. Lesions vary considerably in appearance, depending on increasingly useful for confirmation.
the type of infection. Localized avian and human type infections
Antemortem diagnosis is possible but rarely done in pigs.
tend to produce enlarged, firm nodes with no discrete purulent
foci. Calcification is seldom apparent and lesions are not easily
enucleated. Nodes affected by M tuberculosis complex organisms Control
(specifically M bovis) tend to be well encapsulated, relatively easy Prevention is by avoiding exposure of swine to tuberculosis
to enucleate, and there is often marked calcification in the lesions. bacilli. Swine are quite susceptible to M tuberculosis complex
On rare occasions where disease is caused by M tuberculosis com- organisms, which are usually acquired from exposure to offal or
plex organisms, the infection is much more likely to generalize unpasteurized milk and dairy products from tuberculous cows.
than tuberculosis caused by M avium complex organisms. Lesions Fortunately, exposure is rare in modern agriculture production;
in the organs vary considerably. Initially, the lesions are miliary however, it remains a real threat where bovine tuberculosis is still
and widely distributed. Those in the lungs tend to develop along endemic and the feeding of waste food or milk is common.
pleural and septal lymphatics and resemble dewdrops. Those in Mycobacterium avium complex infections of swine occur with
the liver tend to have caseated or liquefied centers. Splenic lesions some frequency. Raising either swine or poultry in confinement
commonly occur and, when well developed, often protrude. can reduce the most common exposure of swine to infection.
Serous membranes are seldom affected. Conversely, close confinement of swine with careless exposure
to wild birds or poultry can provide an opportunity for greater
exposure. All swine, as well as all bedding, should be protected
from domestic and wild birds. Phenol-based disinfectants are
preferred for environmental disinfection.

TUBERCULOSIS 59
60
BACTERIA
Section II

Diseases caused by viruses

VIRUSES
African swine fever ........................................................................................................................................................................ 63
Blue eye disease................................................................................................................................................................................ 65
Classical swine fever....................................................................................................................................................................... 67
Encephalomyocarditis virus.......................................................................................................................................................... 70
Foot-and-mouth disease ............................................................................................................................................................... 72
Hemagglutinating encephalomyelitis......................................................................................................................................... 74
Inclusion body rhinitis................................................................................................................................................................... 76
Influenza .......................................................................................................................................................................................... 78
Japanese encephalitis...................................................................................................................................................................... 81
Parvovirus......................................................................................................................................................................................... 83
Porcine circovirus associated diseases......................................................................................................................................... 85
Porcine picornaviruses................................................................................................................................................................... 88
Porcine reproductive and respiratory syndrome...................................................................................................................... 92
Porcine respiratory coronavirus................................................................................................................................................... 96
Pseudorabies .................................................................................................................................................................................... 98
Rotavirus.........................................................................................................................................................................................101
Senecavirus A.................................................................................................................................................................................103
Swine enteric coronavirus diseases............................................................................................................................................105
Swine vesicular disease.................................................................................................................................................................108
Swinepox.........................................................................................................................................................................................110
Vesicular exanthema ....................................................................................................................................................................111
Vesicular stomatitis ......................................................................................................................................................................112

61
62
VIRUSES
DISEASES CAUSED BY VIRUSES

African swine fever


Alternate names and abbreviations in which neither species develops significant clinical disease
ASF, ASFV from the infection despite high levels of viremia during the acute
stage of infection. The second cycle describes the relationship
Definition between domestic pigs (truly domestic pigs, but also the Eurasian
A highly contagious, viral disease with signs, lesions, and other wild boar, and feral pigs or free-living pigs that are a result of

VIRUSES
features that closely resemble classical swine fever. The disease is escaped or released domestic pigs) and any of several argasid ticks
exotic to the US There is no evidence that African swine fever including but not limited to O porcinus porcinus. The third cycle
(ASF) virus infects humans. describes those situations whereby the infection can persistent
within populations of truly domestic pigs without any need for
Occurrence a tick vector (ie, direct pig-to-pig transmission). In this third
African swine fever occurs naturally in wild, feral, and domestic cycle, ASF virus is thought to be primarily transmitted via direct
swine (Suidae). At present, ASF does not occur in the western contact with excreted virus in oral and nasal secretions, feces,
hemisphere. It is endemic in several African countries, and an and perhaps short distance aerosols. The US has ticks which
outbreak of the disease has been occurring across multiple coun- could potentially act as vectors for ASF virus. The fourth cycle,
tries in Asia and Europe since 2007, with recent (2018) epidemic described as the “wild boar - habitat cycle,” is characterized both
status reported from China and Southeast Asia. Its distribution by direct transmission between infected and susceptible wild
among countries changes, depending on new infections, reinfec- boars and, importantly, by indirect transmission to wild boars
tions, and the success of eradication programs. through their contact with carcasses of wild boars that have
recently succumbed to ASF. This fourth cycle was described to
Historical information explain the persistence of the highly virulent ASF virus associated
African swine fever has been a serious problem in many African with the ongoing Eurasian wild boar population in the apparent
countries since the disease was first reported in 1921. In 1957, absence of an endemic tick vector.
the disease occurred in Portugal, the first non-African country While both sylvatic and pig-to-pig transmission models can
to be infected. Additional outbreaks have since occurred in Asia, explain the persistence of ASF virus in populations of domestic
Europe, the Caribbean, and South America. Although many of or wild pigs, it appears that the most important means of trans-
these outbreaks were eradicated at significant cost, the risk of mitting the virus over long distances is through the movement of
re-introduction remains. In 2007, ASF was introduced into the infected pigs and the feeding of pigs on swill containing infected
Caucasus region of Eurasia, where it then spread to wild boars pork products. Feeding of contaminated swill is particularly
and domesticated pigs in several member countries of the Euro- important, as it is considered to be one of the most likely routes
pean Union. In 2018, this outbreak extended into China. for introduction of the virus into ASF-free countries that do not
Prohibiting the importation of swine and pork from infected share a contiguous border with countries having populations of
countries and eliminating or regulating the practice of waste-food wild pigs known to be infected.
feeding to pigs are fundamental to most successful eradication Pigs that 1) have died acutely from ASF, 2) have died in a more
programs. The existence of a feral reservoir, widespread tick vec- chronic stage of the disease, or 3) are wild Suids living in an area
tors, and the high cost of eradication make worldwide eradication known to be endemically affected by ASF, all present a risk of
unlikely. The US has never experienced an outbreak, although virus transmission to other pigs. As omnivores, pigs that come
ASF has occurred several times in nearby Caribbean islands. across the carcass of a dead pig are likely to interact with, if not
actively feed on, the carcass. Given the propensity of the virus to
Etiology remain infectious for days to weeks in a protein-rich substrate
African swine fever virus is a DNA virus and is currently the only such as meat, tissue or blood, oral exposure to a contaminated
member of a family called Asfarviridae. More than 20 genotypes of carcass is likely to lead to transmission of the virus.
ASF virus have been identified, most from wildlife cycles in Africa.
African swine fever virus is stable across a wide range of pH and
Isolates differ greatly in virulence, ranging from highly pathogenic
temperatures, particularly when stored in the presence of pro-
viruses that kill most pigs to strains that result only in seroconversion.
tein. Virus has been shown to remain infectious for more than
400 days when stored in filtered serum at room temperature, at
Epidemiology least 6 years in a cold room, and for at least 16 days in putrefy-
In general terms, 4 transmission cycles have been described to ing blood. The virus can remain infectious for years at freezing
explain the transmission and maintenance of ASF virus in pigs. temperatures. The virus can also survive in feedstuffs. The
The first cycle describes a sylvatic relationship between sub- stability of this virus in a variety of matrices is a major threat for
Saharan warthogs and the tick Ornithodoros porcinus porcinus, spread of this disease globally.

AFRICAN SWINE FEVER 63


SWINE DISEASE MANUAL

Phenol disinfectants, in particular 1-Stroke Environ®, appear to swine fever, include hydrothorax, hydropericardium, ascites, and
be most effective in inactivating the virus from surfaces, although pulmonary edema. Hemorrhages and edema are usually more
extended contact time (greater than 1 hour) may be required. severe in ASF than with classical swine fever.
Gross lesions in chronic cases of ASF include fibrinous peri-
Pathogenesis carditis and pleuritis, spleen and lymph node enlargement, and
The primary infection usually starts in the tonsils and mandibular caseous lobular consolidation of areas in the lungs, possibly
lymph nodes. Virus then spreads in lymph and blood to secondary with mineralization.
VIRUSES

replication sites (bone marrow, lung, spleen, and kidney). A


persistent viremia begins about 1 week after infection. The main Histologic lesions of ASF include meningoencephalitis, peripor-
target cells of the virus include monocytes, macrophages, cells of tal hepatitis, extensive necrosis with karyorrhexis (cell nucleus
the reticuloendothelial system, endothelial cells, and platelets. rupture) in lymphoid tissues, and degeneration and necrosis of
Destruction of antigen processing cells in lymph nodes, spleen, endothelial cells and cells of the mononuclear-phagocytic system.
and bone marrow apparently interferes with formation of pro-
tective antibodies. Destruction of endothelial cells and vascular Diagnosis
damage is responsible for hemorrhages, edema, and transudate Diagnosis can be confirmed by detecting ASF virus, viral anti-
formation. The virus also causes a meningoencephalitis which can gen, viral genes, or antibodies to the virus. Virus isolation and
lead to ataxia in affected pigs. The virus affects multiple organs of identification using available tests (eg, hemadsorption, immuno-
fetuses and causes fetal deaths. fluorescence, and polymerase chain reaction [PCR]) confirm the
presence of ASF virus. Several techniques have been developed to
Clinical signs identify ASF virus antigen in paraffin-embedded tissue sections
Clinical signs appear 5 to 15 days after infection. Signs vary (eg, immunohistochemistry) or frozen sections (eg, indirect
greatly, depending on the virulence of the virus, and can range immunofluorescence). PCR assays on tissue or blood detect viral
from inapparent or subclinical to peracute with sudden death. genes and are in common use globally. Viral DNA can also be
Many initial outbreaks closely resemble outbreaks of acute classi- detected in tissue sections by in situ hybridization. A number
cal swine fever. of tests are used to identify ASF antibodies, including indirect
immunofluorescence, enzyme-linked immunosorbent assay, com-
In acute outbreaks, signs include anorexia, high temperatures, plement fixation, immunoblotting, and radioimmunoassay.
listlessness, incoordination, leukopenia, cutaneous hyperemia,
hemorrhages on the skin (especially on ears and flanks), and
hyperpnea. Vomiting, diarrhea, dehydration, and ocular dis-
Control
Control in most countries is limited to preventing entry of the
charge may occur. Morbidity and mortality are high and can
virus. Early diagnosis of suspected cases is important. State and
approach 100%. Death usually occurs in 4 to 7 days. In subacute
federal disease control authorities should be informed imme-
or chronic cases, signs include abnormal respiration, abortion,
diately if an ASF or classical swine fever outbreak is suspected.
and low mortality.
If ASF should gain entrance, early quarantine and restriction
of movement of animals in the area are important. Vaccines are
Lesions unsatisfactory, both for control of clinical signs and for preven-
The lesions are similar to those of classical swine fever. There tion of infection.
are petechial and ecchymotic hemorrhages at many sites (skin,
kidneys, lymph nodes, serosal and mucosal surfaces, epicardium, Massive amounts of virus are found in many tissues, especially
endocardium, larynx, bladder, gall bladder, and lung), edema, blood. Care must be taken when performing necropsies to
hydrothorax, hydropericardium, and ascites. The liver and spleen prevent substantial environmental contamination. Feeding pigs
are congested. The spleen is often enlarged, friable, and may have waste food that has not been heat treated is illegal in the US and
infarcts. There is often congestion of the meninges and brain. most other countries; good compliance with waste food feeding
Lesions that are often present with ASF, but seldom with classical regulations is a critical step in preventing an incursion of ASF
into a country.

64 AFRICAN SWINE FEVER


DISEASES CAUSED BY VIRUSES

Blue eye disease


Alternate names and abbreviations to the fetus in utero. In recovered pigs, porcine rubulavirus has
BED, Porcine rubulavirus, porcine paramyxovirus, La Piedad been isolated intermittently from semen for up to 7 weeks after
Michoacan virus experimental inoculation, and from the testes and epididymis for
as long as 20 weeks.
Definition

VIRUSES
While infectious virus is not routinely isolated from tissues in pigs
Blue eye disease is the common name for a disease caused by a after recovery from the acute illness, viral RNA has been detected
porcine rubulavirus that is characterized clinically in nursing or in numerous organs for as long as 13 months, suggesting the virus
growing pigs by the occurrence of central nervous system (CNS) may persist in at least some pigs for an extended period of time.
signs and corneal opacity. Signs in sows and boars include various
forms of reproductive failure and corneal opacity. Paramyxoviruses are readily destroyed by light, heat, and drying,
and they are normally short-lived in the environment. In general,
Occurrence members of Paramyxoviridae are susceptible to many common
Blue eye disease is only known to occur in swine. In Mexico disinfectants including sodium hypochlorite, sodium hydroxide,
(the only country that has reported the disease), blue eye disease aldehydes, iodine, chlorhexidine, detergents, oxidizing agents,
occurs throughout the year but is more common during hotter, and low pH.
drier months. Swine diseases caused by other paramyxoviruses
have occurred in several countries (Australia, Canada, Israel, Pathogenesis
Japan, and the US), but the diseases they cause are sporadic, rare, Natural infection is thought to occur by inhalation, based on
and distinctly different from blue eye disease, usually manifesting experimental exposures where intratracheal and intranasal expo-
as transient outbreaks of pneumonia, encephalitis, or reproduc- sure reproduced the disease. The initial site of viral replication is
tive disease. unknown but may be in the nasal mucosa or tonsils. Virus soon
spreads to the brain, lung and many other organs, suggesting that
Historical information viremia occurs. Reproductive problems in dams suggest that the
Blue eye disease first occurred in 1980 in a large swine-raising virus crosses the placenta and infects the fetuses.
operation in central Mexico, and within a short time it had Corneal opacities (blue eye) sometimes occur late in the course of
spread to at least half of the Mexican states. It is currently consid- the disease. The opacities disappear spontaneously, presumably as
ered to be an economically important disease in that country. The inflammatory edema resolves. The opacities are perhaps the result
first outbreaks occurred mainly in unweaned piglets less than 30 of an immunologic reaction accompanied by inflammation. Virus
days old, frequently in piglets less than 2 weeks old. After 1983, may replicate in the eyes, since inclusion bodies have been demon-
severe outbreaks tended to occur in larger pigs (33 to 99 lb [15 strated there.
to 45 kg]). Signs of reproductive failure were also observed in
mature sows and boars. Experimentally infected piglets developed clinical signs 3 to 5
days after exposure.
Etiology
The agent that causes blue eye disease is officially named porcine Clinical signs
rubulavirus and is a member of the Paramyxoviridae family of Signs vary somewhat with the age groups affected but are mainly
RNA viruses. Other members of this family include Hendra heni- related to encephalomyelitis in both young and growing pigs. In
pavirus (Hendra virus disease), Menangle rubulavirus (Menangle young piglets, onset is sudden with prostration and fever. Other
virus disease), and Nipah henipavirus (Nipah virus disease), each signs include ataxia, weakness, muscle tremors, rigidity, nystagmus,
of which are emerging as important zoonotic agents in other abnormal postures, hyperexcitability, squealing, and paddling
parts of the world. Although no wildlife reservoir has been iden- movements; occasionally there is blindness. Often there is conjunc-
tified for blue eye disease virus, pteropid fruit bats are known to tivitis with adherence of the eyelids. Corneal opacity may occur
be the reservoir for the other viruses listed above and are critically in 1% to 10% of young piglets and may be seen in the absence of
important in the epidemiology of those diseases. other signs. Twenty to 65% of litters can be affected. Morbidity
within litters is between 20% to 50%; mortality in affected pigs can
reach 90%. Piglets affected early in an outbreak often die within 48
Epidemiology hours. Those affected later in life usually die after a course of 4 to 6
Both direct contact and short distance aerosolization are import-
days. Mortality occurs over a 2- to 9-week period.
ant routes of transmission for blue eye disease virus. Direct
contact is not required for transmission. Infectious virus has also In pigs greater than 30 days old, signs are more moderate and
been found in urine and semen, and the virus can be transmitted transient. They include fever, sneezing, coughing, and anorexia.

BLUE EYE DISEASE 65


SWINE DISEASE MANUAL

Central nervous system signs include listlessness, ataxia, and cir- Diagnosis
cling. Less than 5% of the pigs are affected and mortality usually Clinical signs of encephalomyelitis as described and the presence
is low. However, in pigs weighing 35 to 45 lb (16 to 20 kg), out- of corneal opacity, particularly in suckling pigs, suggest a diagno-
breaks with 20% mortality and up to 30% corneal opacity have sis of blue eye disease. These clinical signs in sows or boars may be
occurred. Adult animals occasionally develop corneal opacities. followed by prolonged or ongoing reproductive failure, including
In pregnant swine, returns to estrus may occur and persist in the increased mummies, small litters, and infertility. Microscopic
herd for 6 to 8 months. There is also an increase in stillbirths and lesions of encephalomyelitis and the presence of intracytoplasmic
VIRUSES

mummified fetuses, and occasional abortions. In affected boars, inclusion bodies in neurons or eyes strongly suggest blue eye
there is a reduction in fertility and an increase in testicular size, disease.
usually unilateral, followed by atrophy. The hemagglutinating virus can usually be isolated from the
brain, tonsil, serum, and many other tissues using PK-15 or Vero
Lesions cells. Serological assays have been developed and include hemag-
Gross lesions are limited and nonspecific. They may include glutination inhibition, virus neutralization, and enzyme-linked
mild anteroventral pneumonia, tonsillitis, and mild ascites with immunosorbent assay. Blue eye disease must be differentiated
some fibrin. Brain congestion and an increase in the amount of from other causes of CNS disease, especially pseudorabies.
cerebrospinal fluid may be present. There is conjunctivitis and
perhaps corneal opacity in one or both eyes. Pericardial and renal Control
hemorrhages have also been reported. In affected boars there is A rigorous health control program to prevent introduction of the
orchitis, epididymitis, and testicular atrophy. virus is the best preventive measure. Maintaining a closed herd
Microscopic lesions include a nonsuppurative encephalomyelitis and use of the all-in, all-out system of production may be helpful.
affecting mainly the gray matter of the midbrain, thalamus, and Essential replacement stock should be added only after negative
cerebral cortex. Intracytoplasmic inclusions in neurons have been serological tests and a period of quarantine. Commercial vaccines
reported. Affected eyes have corneal edema and anterior uveitis, have been produced but there is little information available
perhaps with exudate in the anterior chamber. Intracytoplasmic regarding their efficacy.
inclusions may be present in epithelial cells near the drainage Blue eye disease has been eliminated from infected sites by dis-
angle of the corneoscleral junction. posing of infected animals, cleaning and disinfecting the site, and
using sentinel pigs to confirm elimination of infection. There is
no satisfactory treatment of blue eye disease: pigs that develop
CNS signs usually die. Pigs having only corneal opacities fre-
quently recover without treatment.

66 BLUE EYE DISEASE


DISEASES CAUSED BY VIRUSES

Classical swine fever


Alternate names and abbreviations passage through pigs. Strains of high virulence cause classic out-
CSF, CSFV, hog cholera, swine fever breaks with high morbidity and mortality. Strains of moderate
virulence cause subacute or chronic infections. Strains of low
Definition virulence can cause mild or inapparent infection, reproductive
Classical swine fever (CSF) is a highly contagious, viral disease of failure, or neonatal losses.

VIRUSES
swine that in its most virulent form causes morbidity and mortality New strains of CSF have emerged in Asia in the last few years.
approaching 100%. Viral strains of low to moderate virulence cause Their clinical presentation does not appear to be substantially
infections with a gradient of severity, some clinically inapparent. different than those produced by classical strains of the virus but
immunologic cross-protection (by natural exposure or vaccina-
Occurrence tion) is limited.
Classical swine fever occurs only in swine, and all age groups are
susceptible. The disease occurs in most major swine-raising coun- Classical swine fever virus can persist for days to weeks in pig
tries where eradication programs have not been successfully imple- facilities, excreta, and bedding, depending upon temperature.
mented. In the US, CSF was once a frequent occurrence but was The virus survives some curing processes as well as in frozen pork
eradicated in 1976. Classical swine fever currently exists in many for months to years, and in chilled meats for months. The virus is
countries, including areas of Central and South America, Asia, the inactivated by 2% sodium hydroxide or by lipid solvents.
Caribbean, and in localized feral pig populations in Europe. Since There is no evidence that classical swine fever virus can be trans-
1997, sporadic epidemics of CSF have been reported in many mitted to humans.
European countries, including Germany, Italy, The Netherlands,
and Spain, suggesting the disease is an ongoing threat for countries Epidemiology
with modern, commercial pork production industries. Pigs are infected with classical swine fever virus mainly by direct
contact with other infected pigs through the oral or oronasal
Historical information routes. However, the virus may also be transmitted through natu-
The historical record does not clearly indicate where CSF ral mating or artificial insemination.
originated, but a disease with clinical signs similar to CSF was
reported from the eastern US in the early 1800s. Until organized Pigs with acute infection shed large amounts of virus before they
efforts to control the disease began in the 1960s, the US routinely are visibly ill, during illness, and after recovery. The virus is shed
experienced significant regional outbreaks of CSF. The earliest in most body fluids, including saliva, urine, feces, and semen.
efforts to minimize losses from CSF were first achieved through Shedding can begin before the onset of clinical signs. Classical
the administration of CSF antiserum as either a prophylaxis swine fever virus can persist in blood and tissues after death,
or for treatment of the disease. When properly performed on explaining why an important route of transmission is related to
healthy swine, it resulted in strong immunity. However, there the feeding of uncooked waste food that contains tissues from
were many cases when serum administration triggered outbreaks infected pigs.
of other diseases (in addition to CSF), as the serum preparation Aerosol transmission has been demonstrated experimentally with
was not well regulated - meaning it could contain live viruses some strains but is probably not a significant means of moving
or other disease-causing organisms. Serum administration was virus between farms.
eventually prohibited when a national CSF eradication program
Other methods of viral spread include fomites (farm equipment,
began in 1962. Improvements in CSF vaccine, farm biosecurity,
wagons, trucks, tractors, machinery) and personnel (farmers and
and coordinated industry actions resulted in the eradication of
staff, farm visitors, veterinarians).
CSF in 1976. Eradication was a remarkable achievement, con-
sidering the highly infectious nature of virus and the extensive,
poorly regulated commerce in pigs and pork products at the time. Pathogenesis
After ingestion, the virus infects epithelial cells in the crypts of
tonsils, spreads to adjacent lymph nodes and produces viremia
Etiology within 24 hours. Tonsils are the initial site of viral replication.
Classical swine fever is a pestivirus related to the viruses that
Replication also occurs at other sites, especially in lymphoid tis-
cause bovine virus diarrhea and border disease. Classical swine
sues (spleen, Peyer’s patches, lymph nodes, and thymus), endothe-
fever virus is closely related to these ruminant pestiviruses which
lial cells, bone marrow, and circulating leukocytes. Within 3 to 4
can also occasionally infect pigs, causing serological reactions
days, the virus spreads to many epithelial-type cells and is present
that may be mistaken for CSF. Strains of CSF vary greatly in
in excretions and secretions.
antigenicity and virulence. Virulence can increase during a single

CLASSICAL SWINE FEVER 67


SWINE DISEASE MANUAL

The virus causes lymphoid depletion, making affected swine more Microscopically, there is a panencephalitis that is most apparent in
susceptible to other infections. Bone marrow damage leads to the medulla, pons, midbrain, or thalamus. Glial cell nodules often
leukopenia and thrombocytopenia. Thrombocytopenia, along cluster around destroyed capillaries. Vascular lesions are present at
with endothelial cell damage, results in petechial and ecchymotic many sites, perhaps more pronounced in splenic follicular arteries.
hemorrhages at many sites. Swine with chronic CSF infection
may develop glomerulonephritis from antigen-antibody com- Diagnosis
plexes that damage glomeruli. Typical acute CSF should be suspected on the basis of history,
VIRUSES

clinical signs, body temperature, and gross lesions. Numerous


In pregnant sows and gilts, the virus may cross the placenta and
postmortem examinations will increase the accuracy of diagnosis.
infect some or all of the fetuses. The effect depends on the stage
Leucopenia in multiple affected pigs is suggestive of CSF. The
of pregnancy and can include abortion or production of mummi-
lesions of typical acute cholera closely resemble and must be
fied fetuses, stillborn piglets, or persistently infected live piglets. carefully differentiated from those of African swine fever, acute
Fetal anomalies may result from in utero infection; a remarkable salmonellosis, and acute swine erysipelas. Lesions sometimes
anomaly is hypomyelinogenesis, a syndrome that results in shak- resemble those of other septicemic diseases, including strepto-
ing piglets (myoclonia congenita). coccosis and Glasser’s disease. Infection with mildly virulent
strains may be indistinguishable from many endemic systemic or
Clinical signs respiratory pathogens. Diagnostic testing is imperative whenever
In typical acute outbreaks, clinical signs are nonspecific and the combination of gross pathology, clinical signs, and lack of
include depression (a hunched posture with drooping head and response to antimicrobial therapy suggest that CSF should be on
a straight-hanging tail), anorexia, high fevers (106°F [41°C]), the list of possible etiologies.
conjunctivitis, and a strong desire to lie down and huddle or pile
Suspected outbreaks should be reported immediately to author-
with other affected pigs. There may be diarrhea or constipation,
ities to confirm the diagnosis. Three laboratory procedures com-
and perhaps vomiting. Signs caused by central nervous system
monly used in confirming a diagnosis include: 1) demonstration
lesions are often apparent and may include reeling when forced
of CSF viral antigen in frozen tissue sections by immunofluo-
to walk, hindquarter paresis or paralysis, and occasional tonic/
rescence with preferred tissues being tonsil, pharyngeal lymph
clonic convulsions in young growing pigs. Most affected pigs die
nodes, spleen, kidney and distal ileum; 2) isolation of virus in
within 3 weeks of onset.
cell culture and identification of viral antigen in the culture by
Outbreaks with less virulent virus or chronic cases seldom present the fluorescent antibody test; and 3) identification of antibodies
with typical signs but often have conjunctivitis, diarrhea or con- to CSF virus (CSFV) by virus neutralization tests. Regardless of
stipation, and some degree of emaciation. Mildly virulent strains the procedure used, it may still be necessary to differentiate CSF
of the virus seem to be becoming more prevalent worldwide and from bovine viral diarrhea. Virus isolation and polymerase chain
present a significant opportunity for misdiagnosis by veterinar- reaction assays for CSFV are available.
ians and producers unfamiliar with the disease. Pigs infected as
fetuses or neonates may present no signs. Control
Control of CSF is possible through prevention of exposure,
Lesions vaccination, or eradication. In most countries, a cornerstone for
In typical acute CSF, lesions include petechial and ecchymotic prevention of the disease is a ban or strict control on the impor-
hemorrhages at several common sites, including the epiglottis, tation of live pigs, fresh pork, insufficiently heated pork products,
bladder mucosa, cortex and pelvis of the kidneys, gall bladder and other possible sources of virus (imported swine semen and
mucosa, on the lungs and heart, at the ileocecal junction, and embryos, and biologicals produced with porcine serum or prod-
in the skin. Lesions sometimes considered of special value in ucts), and prohibiting the feeding of uncooked waste food. Most
diagnosis include single or multiple infarcts along the border of developed countries have strict regulations that prohibit disposal
a spleen of normal size, subcapsular hemorrhage in many lymph of garbage from international vessels (planes and ships) without
nodes, and hemorrhages on the cortices of the kidneys and on the heat treatment.
lungs. There usually is some degree of congestion in the fundus
In countries where the virus is endemic, attenuated vaccines are
of the stomach and small intestine. Small foci of necrosis may be
often used for preventing or reducing the prevalence of infection
present in the tonsils.
and minimizing the severity of clinical disease. Effective commer-
Peracute cases may have no lesions. In chronic cases, ulcers with cial vaccines are available, although some countries rely on gov-
raised edges (“button ulcers”) are often present in the cecum and/ ernment-produced vaccines that have unknown efficacy. In the
or colon. Grossly visible fetal lesions include ascites, hepatic nod- final stages of an eradication program that has relied on CSF vac-
ularity, pulmonary hypoplasia, petechiation of the skin, microen- cination, vaccine use may be stopped as ongoing use of modified
cephaly, hydrocephalus, and cerebellar hypoplasia. live vaccines will prevent true disease freedom from occurring.

68 CLASSICAL SWINE FEVER


DISEASES CAUSED BY VIRUSES

Bovine viral diarrhea (BVD) virus and


border disease (BD) virus of sheep
These two pestiviruses and CSF virus are closely related. Natu-
rally occurring infections with bovine viral diarrhea and border
disease have occurred in swine. Their major importance in swine
is that they both induce antibodies that can lead to serological
misdiagnosis of CSF. This can cause confusion in countries trying

VIRUSES
to eradicate CSF or maintain a CSF-free status. This confusion
can be avoided if specific laboratory methods of CSF diagnosis
are used.
Clinically apparent outbreaks of bovine viral diarrhea and bor-
der disease in swine are uncommon. Outbreaks that have been
suspected are manifested in breeding herds as reproductive prob-
lems, such as poor conception rates, small litters, abortions, and
an excessive number of dead and mummified fetuses. In infected
live piglets, the signs are expected to be similar to those of con-
genital bovine viral diarrhea and border disease, including deaths
in piglets less than 5 weeks old, anemia, unthriftiness, congenital
tremors, and convulsions.

CLASSICAL SWINE FEVER 69


SWINE DISEASE MANUAL

Encephalomyocarditis virus
Alternate names and abbreviations does not occur. Infected rodents excrete virus and their diseased
EMC, EMCV carcasses contain large amounts of virus. Presumably, swine are
infected by ingesting feed or water contaminated by rats, mice,
Definition or the carcasses of animals that died from encephalomyocarditis.
Encephalomyocarditis (EMC) is a disease caused by infection with Infected swine excrete virus for about 9 days, but it is not clear
VIRUSES

EMC virus. Encephalomyocarditis virus commonly infects suscep- that they play an important role in transmission to other pigs.
tible species; clinical disease is unusual in domestic animals. Virus has been isolated from certain tick and mosquito species,
but vector competency studies have not shown transmission from
Occurrence infected arthropods.
Susceptible species include humans, primates, many species of
rodents, marsupials, mongoose species, and several species of Pathogenesis
birds. In zoos, rapidly fatal infections have been reported in Following experimental oral infection of piglets, viremia occurs.
elephants, primates, and artiodactyls. Horses, cattle, swine, dogs, Lymphoid tissues contain virus and are probably the major sites
and cats are susceptible to infection, but except for swine there is of virus replication. The highest titers of virus occur in damaged
no clear evidence that encephalomyocarditis viruses are signifi- heart muscle. Most deaths are related to right-sided heart failure.
cant pathogens of domesticated species. Transplacental infections can occur in fetuses near full-term and
fetuses may develop myocardial lesions. Pigs that survive develop
Historical information antibodies that persist for long periods.
In 1940, Columbia SK virus was the first strain of encephalo-
myocarditis virus identified. At the time, it was postulated to be Clinical signs
a strain of poliovirus, but this was later shown not to be the case. Infection in swine is usually subclinical, but in rare cases it
Infection in swine was not reported until outbreaks occurred produces myocarditis and high mortality in nursing pigs and/
in Panama (1958) and Florida (1960). Subsequently, clinical or reproductive failure in dams. For unknown reasons, clinical
encephalomyocarditis was reported in Australia, Brazil, Cuba, outbreaks in swine generally occur in tropical or semi-tropical
Greece, New Zealand, and North America. climates. Clinical disease in young piglets is often characterized
Research on the encephalomyocarditis viruses in swine was by an acute course and sudden death. Less acute cases have respi-
conducted in the early 1990s because of an erroneous association ratory signs with dyspnea, fever, anorexia, depression, trembling,
with “mystery swine disease,” which was later proven to be caused staggering, and paralysis. Mortality may approach 100% in neo-
by porcine reproductive and respiratory syndrome (PRRS) virus. natal and nursing pigs. Older pigs and adults usually have subclin-
At present in the US, encephalomyocarditis may be a sporadic ical infection, but some mortality may occur, even among adults.
pathogen of dubious clinical significance as a swine pathogen. In pregnant sows and gilts, there are mid- to near-term abortions
with an increase in stillbirths and mummified fetuses.
Etiology
The encephalomyocarditis group of viruses is classified in the Lesions
genus Cardiovirus in the Picornaviridae family. They are non-en- In young piglets there are changes compatible with right-sided
veloped, single-stranded RNA viruses. These hemagglutinating heart failure, including hepatomegaly, hydropericardium,
viruses are stable in many environments and can be grown in cell hydrothorax, pulmonary edema, and ascites. The heart may be
culture. Areas contaminated with encephalomyocarditis viruses enlarged, flabby, and pale red with subepicardial, petechial hem-
can be disinfected with iodine-based disinfectants or 0.5 ppm orrhages. Lesions are most marked in the right ventricle where
residual chlorine. there may be discrete yellow or white foci, or diffuse, pale areas
of myocardial necrosis. Similar myocardial lesions may be visible
Epidemiology in the heart of aborted fetuses. Microscopic lesions include focal
The encephalomyocarditis group of viruses is present throughout nonsuppurative interstitial myocarditis and myocardial necrosis;
the world. Infection is common but clinical disease is rare in infiltration of small numbers of neutrophils may be evident.
domestic animals and humans. Rats and mice are considered to Occasionally, this finding is accompanied by a nonsuppurative
be the principal reservoirs of the virus, although the experimental meningoencephalitis.
evidence to support this is weak. Transmission of encephalomyo-
carditis virus among rodent species is difficult to achieve and a Diagnosis
chronic intestinal carrier state, a key epidemiologic feature in Disease associated with encephalomyocarditis is rare but
closely related picornaviruses (ie, polioviruses or enteroviruses), should be considered in cases where there is marked dyspnea

70 ENCEPHALOMYOCARDITIS VIRUS
DISEASES CAUSED BY VIRUSES

and sudden deaths in pre-weaning age piglets, high preweaning Control


mortality, an increase in late-term abortions, or gross and/ Until encephalomyocarditis is demonstrated to be a significant
or microscopic myocardial lesions. Myocardial lesions must be cause of disease, one should interpret any positive diagnostic test
differentiated from those of PRRS, mulberry heart disease, toxo- results with caution to ensure that other more likely agents are
plasmosis, bacterial septicemia, and cardiac infarcts. not involved.
Definitive diagnosis requires demonstration of virus associated Elimination or reduction of feral rodent populations is recom-

VIRUSES
with compatible gross and microscopic lesions. The virus can be mended in order to minimize environmental contamination with
isolated from the myocardium or spleen of acute cases in young virus. Feeds from all sources should be properly stored and kept
piglets. Polymerase chain reaction assays are available to detect free of contamination by rodents. Pigs from farms known to have
the presence of the virus. encephalomyocarditis should not be added to known negative
A number of serological assays have been developed. A serum herds. Pigs, rodents, and other animals that have died should
neutralization assay is widely available and has been shown to be be removed promptly to avoid transmission through ingestion.
specific for antibodies against encephalomyocarditis virus. Serum Basic hygienic and sanitary practices should be followed. An
neutralizing antibodies may persist for an extended period of inactivated vaccine is commercially available, but its use should be
time, so serological evidence of recent infection requires demon- clearly justified. There is no specific treatment.
strating a 4-fold increase in neutralizing antibody titers in conva-
lescent versus acute sera.

ENCEPHALOMYOCARDITIS VIRUS 71
SWINE DISEASE MANUAL

Foot-and-mouth disease
Alternate names and abbreviations culling, and production losses to farmers. The outbreaks also
FMD, FMDV had substantial effects on the national economies due to reduced
tourism, a temporary decline in meat consumption, and flow-on
Definition effects to related agribusinesses.
Foot-and-mouth disease (FMD) is a highly contagious viral
VIRUSES

Outbreaks of all vesicular diseases of swine are of concern because


disease of many wild and domestic cloven-footed mammals, and the lesions cannot be distinguished from those of FMD except
many other animals. In swine, the disease is characterized by vesi- by laboratory means. Swine are of special concern because they
cles on the feet, snout, and in the mouth. are susceptible to more vesicular diseases than other species of
livestock and they often play a major role in the spread of FMD
Occurrence by producing large, infectious aerosols of virus.
Many wild and domestic animals, especially cloven-footed mam-
mals, are susceptible to FMD virus. Important livestock hosts Etiology
include cattle, pigs, sheep, and goats, as well as water buffalo and Foot-and-mouth disease virus is a member of the genus Aphthovi-
yaks in Asian countries. Cattle can act as important maintenance rus in the family Picornaviridae. There are 7 major viral serotypes:
hosts as some individuals will harbor virus for many months after O, A, C, South African Territories type 1 (SAT1), South African
clinical disease has resolved. Some strains of FMD virus are host- Territories type 2 (SAT2), South African Territories type 3
adapted to cattle, sheep, or pigs, but one should consider all species (SAT3), and Asia 1. Serotype O is the most common serotype
to be at risk, regardless of the strain responsible for an outbreak. worldwide. Among the 7 types, one particular antigen (virus
Other susceptible species include wild and farmed cervids (deer, infection-associated antigen or “VIA”) is group reactive and use-
moose, caribou, elk, etc), llamas, and alpacas. Experiments suggest ful in serological diagnosis of FMD infection.
that Bactrian camels can develop FMD, but dromedary camels Over 60 subtypes of virus have been identified and new subtypes
appear to be resistant to infection. continue to develop. Many differ enough antigenically to require
Horses are uniformly resistant to infection with FMD virus, a preparation of subtype vaccines for their control. The antigenic
fact that can be very useful in formulating a list of differential variation of the virus and the limited cross-protection among
diagnoses in the early phase of an outbreak. The disease occurs strains has made it impossible to prepare a single vaccine that
in most countries that have large livestock populations unless the protects satisfactorily against all strains. Effective disinfectants
country has made specific efforts to eradicate it. All age groups of FMD virus include sodium hydroxide, acetic acid, sodium
are susceptible. In the absence of farmed susceptible livestock, carbonate, and Virkon™.
FMD is generally unable to sustain itself in populations of wild
susceptible animals. Epidemiology
Virus transmission occurs through respiratory aerosols and direct
Historical information or indirect contact with infected animals. Aerosol transmission
Foot-and-mouth disease is an ancient disease. A description of of FMD virus over distances as great as 30 miles is believed to
a disease observed in 1546 probably was a description of FMD, occur under certain weather conditions. For some virus subtypes,
and the disease has continued to cause major losses in livestock infected swine can act as exceptional disseminators of virus and
throughout the world. Losses have been greater from loss of pro- are capable of producing aerosols with greater virus concentration
ductivity than from mortality. than those produced by cattle or sheep.

Nine outbreaks of FMD have occurred in the US with the last one Infected animals disseminate virus in their excretions and secre-
occurring in 1929. All 9 outbreaks were followed by eradication, tions. Food products containing infectious pork can also spread
often at great expense. A new outbreak of FMD could cause great FMD virus. Virus persists for long periods of time in frozen meat
losses in the US, as it is likely that the disease would become wide- products. In several notable FMD outbreaks, the index case has
spread before it was recognized, due to the frequent long-distance been associated with pigs consuming uncooked waste food con-
transportation of livestock in the cattle and swine industries. taining infectious meat scraps. Contaminated biologics, including
vaccines, have also been responsible for outbreaks. People with
Recent outbreaks of FMD in Japan, South Korea, and other residual FMD virus in their respiratory tract can transmit the
countries with modern livestock sectors have demonstrated the virus to livestock for a short time.
devastation and frustration wrought by FMD. In addition to the
cost of the national disease response, these outbreaks resulted Some species of animals, particularly cattle, recover from FMD and
in the immediate restriction of meat and livestock exports, mass remain carriers for weeks, months, and possibly years. Occasionally,

72 FOOT-AND-MOUTH DISEASE
DISEASES CAUSED BY VIRUSES

they disseminate virus that initiates new outbreaks. Swine are not Foot lesions may involve one or more of the feet. Vesicles usually
believed to be long-term carriers of FMD virus. rupture within 24 hours and the superficial epidermis sloughs to
reveal hyperemia and hemorrhage on underlying tissue. Uncom-
The incubation period for FMD can vary with the species of
plicated lesions usually heal within 2 weeks. In a virulent form
animal, the dose of virus, the viral strain, and the route of inocu-
of FMD, young pigs and sometimes older animals may have
lation. It is reported to be 2 to 4 days in pigs (with some experi-
extensive mottled areas of myocardial necrosis on ventricles and
ments reporting clinical signs in as little as 18 to 24 hours).
in papillary muscles.

VIRUSES
Pathogenesis Diagnosis
Foot-and-mouth disease virus adheres to the mucosa of the
A diagnosis cannot be reliably made on the basis of clinical signs
respiratory tract, the usual site of virus entry. Macrophages are
and lesions, since they are similar in all of the vesicular viral dis-
believed to transport the virus to secondary sites that include eases of swine. The state animal health official (SAHO or state
epithelium, mucosa, and myocardium. In secondary sites, the veterinarian) or a USDA-APHIS Assistant District Director
virus replicates, a marked viremia develops, and then the virus (ADD, previously called the Area Veterinarian in Charge or
infects epithelium at many other sites. Within a few days, vesicles AVIC) should be contacted immediately if an outbreak is sus-
develop, usually at sites of mechanical stress. In swine, common pected. Differential diagnosis of vesicular viral diseases should
vesicle sites include the snout, mouth, tongue, and especially the only be completed in specifically designated laboratories having
feet. In cattle, FMD virus affects the mammary gland epithelium facilities to safely handle exotic disease organisms. Plans must be
and the virus is shed in milk for a prolonged period of time. made for collecting and mailing specimens. The Foreign Animal
Although unproven, similar shedding may occur in swine. Disease Diagnostic Laboratory in Plum Island, New York, often
does the diagnostic work.
The lesions of the major vesiculating viral diseases are similar. Ves-
icles develop in the epidermis and then the epithelium over the Diagnostic techniques include serological tests to identify FMD
vesicle soon sloughs. Enough of the stratum basale is preserved to virus infection-associated antigens, complement fixation and
regenerate the epidermis unless there is secondary infection of the enzyme-linked immunosorbent assay (ELISA) to detect FMD
lesions. Secondary infection occurs on the feet of some swine and virus antigen, virus isolation, virus neutralization, electron micros-
leads to chronic lameness. copy, and animal inoculation studies. Polymerase chain reaction
tests have been developed and are frequently utilized. Foot-and-
Foot-and-mouth disease can cause severe myocardial necrosis in mouth disease must be differentiated from all other vesicular viral
neonatal and young pigs which often leads to sudden deaths from diseases and from other diseases that cause erosive or ulcerative
myocardial failure. The mottled myocardial lesions are sometimes lesions in the oral cavity. Positive diagnoses usually require less time
referred to as “tiger-heart” lesions and are useful in diagnosis. than negative diagnoses. An ELISA is available that can differen-
tiate antibody titers from infected versus vaccinated animals but is
Clinical signs not yet officially recognized by many countries.
Lameness is often the first clinical sign of FMD infection. There
is an initial acute rise in temperature; slobbering and chomping Control
are common signs. Pregnant sows may abort or deliver stillborn, In the US, prevention of FMD depends on regulations that govern
infected piglets. Sudden deaths may occur in neonatal pigs, some- the importation of animals, animal products, semen, and embryos,
times before signs or lesions are apparent in the sow. The early and on regulations related to the safety of vaccines and other bio-
stages of lesions appear as blanched, small foci in the skin on the logic products. There are special regulations on both cooking and
snout, soft tissues of the feet, and perhaps the teats of lactating disposal of waste food to prevent spread of the virus. Reliance on
sows. By the time signs are readily apparent, there are usually the vigilance of veterinary practitioners and diagnostic laboratory
cutaneous vesicles or bullae. Signs develop rapidly, and morbidity personnel is important in early detection of outbreaks.
rapidly increases. Mortality is usually less than 5% but there can
be higher mortality in young pigs. In countries where FMD has not been eradicated, vaccines are
used for prevention. The recently developed subunit vaccines
have been of value in preventing some types and subtypes of
Lesions FMD infection but have not provided consistent protection
Well-developed vesicles and bullae are soon apparent. They are
frequently present on the snout, behind the rim of the snout, in for type O infection, the most prevalent form of FMD. In the
the nares, on the tongue and lips, and on the soft tissues of the event of an outbreak in the US, vaccine use will be considered.
feet including the coronary band, the bulbs of the toes, and inter- The disease will be managed through quarantine, restriction of
digital clefts. Lesions are probably more common on the feet than movement of animals in quarantined areas, slaughter followed
in the mouth. Less often, the lesions are on the vulva, the teats of by burial or incineration of infected and exposed animals, and
lactating sows, or the scrotum of boars. Extensive lesions on the disinfection of production sites. Eradication is considered to be
coronary band may lead to sloughing of the hoof and lameness. less costly than living with FMD.

FOOT-AND-MOUTH DISEASE 73
SWINE DISEASE MANUAL

Hemagglutinating encephalomyelitis
Alternate names and abbreviations of exposure to virus in the environment followed by subclinical
HE, HEV, vomiting and wasting disease infection. Clinically apparent disease develops only when young
piglets from non-immune sows are exposed.
Definition
A disease seen in pigs less than 4 weeks old, characterized by vom- Pathogenesis
VIRUSES

iting, wasting, and perhaps neurologic signs. The virus replicates in the nasal epithelial mucosa, tonsils, lungs,
and small intestine and spreads via the peripheral nervous system
Occurrence to the central nervous system. Viremia is probably of little signifi-
Hemagglutinating encephalomyelitis occurs only in swine. The cance in spread. In the central nervous system, virus localizes first
disease occurs in most major swine-raising countries but tends to in nuclei of the medulla oblongata but later moves to the brain
be quite sporadic. Infection is clinically silent except in piglets less stem, spinal cord, and sometimes to the cerebrum and cerebel-
than 4 weeks old. The disease occurs as an acute outbreak affect- lum. The virus produces lesions in a variety of nuclei, ganglia, and
ing most piglets in litters of non-immune dams. plexi (trigeminal ganglion, vagal sensory nuclei, some brain stem
nuclei, and intramural plexi of the stomach). Clinical signs vary,
Historical information depending upon which nuclei are affected by the virus. Death
In 1959 an encephalomyelitis of suckling pigs was reported occurs whenever a vital nucleus ceases to function.
in Ontario, Canada. In 1962, other investigators reported a
hemagglutinating virus producing encephalomyelitis in piglets. Clinical signs
In 1969, vomiting and wasting disease, similar in many respects Two similar clinical syndromes are caused by infection with hem-
to the encephalomyelitis reported in Canada, was reported in agglutinating encephalomyelitis virus.
England. Viruses from the outbreaks later were proven to be
similar or identical. Vomiting and wasting syndrome
Sneezing and coughing may occur at onset. Within a few days,
Much of the research on hemagglutinating encephalomyelitis vomiting of milk and retching are prominent signs. The young
infection in swine was completed in the 1970s and 1980s. Epi- pigs dehydrate rapidly and often grind their teeth. When water
demiologic studies have shown the virus to be widespread in the is supplied, piglets mouth it but drink little, presumably because
US. However, clinical outbreaks seldom occur, and the disease is of pharyngeal paralysis. After a few days they exhibit dyspnea,
of modest economic importance. cyanosis, coma, and die. Pigs in older litters survive longer but
continue to vomit occasionally. The anterior abdomen of some
Etiology of them may become distended, presumably from impaired emp-
Porcine hemagglutinating encephalomyelitis virus is a sin- tying and accumulation of gas. Most affected piglets waste away
gle-stranded, positive-sense RNA virus in the family Coronavi- over a few weeks and die. The few that survive remain unthrifty.
ridae, genus Betacoronavirus. The virus can be grown in several Morbidity and mortality can approach 100% in affected litters.
porcine cell culture systems and hemagglutinates erythrocytes
from several kinds of animals. Only one serotype of the virus is Encephalomyelitis syndrome
recognized. Differences in strain virulence, along with differences Initial signs are similar to those in the above syndrome. Vomiting
in susceptibility are responsible for 2 clinical syndromes. and retching occur less often and lead to less dehydration. In 1
to 3 days, any of a variety of signs of encephalomyelitis appear.
Epidemiology These may include stilted gait, muscle tremors, nystagmus, blind-
Only pigs are susceptible to infection with hemagglutinating ness, opisthotonus, convulsions, and progressive paresis leading
encephalomyelitis virus. While the virus can infect naïve pigs of to recumbency with paddling of the legs. Occasional pigs walk
any age, clinical signs including vomiting and wasting or enceph- backwards and assume a dog-sitting position. Affected pigs soon
alomyelitis, are generally seen only in piglets less than 4 weeks weaken, pass into coma and die. In some litters of older pigs, there
of age. Transmission of virus is presumably through actively may be only transient posterior paresis followed by recovery.
infected or carrier swine. Exposure is probably through infectious
nasal secretions or aerosol. Most large breeding herds in major Lesions
swine-raising countries are endemically infected. Lesions are similar in the two syndromes. Chronically infected
In most swine herds, immune sows provide colostral antibody pigs have cachexia and there may be distension of the anterior
that protects their piglets for 4 to 18 weeks. Passive immunity in abdomen by excessive gas in the stomach.
the growing pigs is replaced by active immunity as a consequence

74 HEMAGGLUTINATING ENCEPHALOMYELITIS
DISEASES CAUSED BY VIRUSES

Microscopic lesions include a nonsuppurative encephalitis affect- Paired early and late serum samples can sometimes be used to
ing the gray matter of the medulla and brain stem. The frequency show a rise in antibody titer against the infection. The first blood
of lesions at specific sites may vary in the two syndromes. There samples should be taken early in the course of the disease since
is inflammation of the trigeminal, paravertebral, and autonomic antibody levels rise quickly. There does not appear to be serologi-
ganglia. Also, ganglia in the stomach wall, mostly in the pyloric cal cross-reaction with other porcine coronaviruses.
area, may show degeneration.
Once the virus is introduced, it persists in the herd until all
breeding stock (including replacement females) have been

VIRUSES
Diagnosis exposed. Development of an active immunity against the virus
The history, signs, and age group affected may support a diag- allows for development of adequate passive lactogenic immunity
nosis of hemagglutinating encephalomyelitis. However, for for their piglets.
definitive diagnosis, the virus should be isolated from affected
pigs, demonstrated present within lesions using antigen-based There is no effective treatment for affected individuals.
stains, or detected by polymerase chain reaction. Virus isolation
can best be made from the brainstem of piglets showing signs for
less than 2 days.

HEMAGGLUTINATING ENCEPHALOMYELITIS 75
SWINE DISEASE MANUAL

Inclusion body rhinitis


Alternate names and abbreviations if severely stressed. These carriers then disseminate virus when
Porcine cytomegalovirus, PCM, PCMV, IBR, nonprogressive mixed with susceptible swine. By both experimental and natural
rhinitis routes, the porcine cytomegalovirus can sometimes cross the
placenta to infect developing fetuses, which may result in dead or
Definition mummified fetuses, or infected, weak piglets. Infected piglets can
VIRUSES

A common viral infection but sporadic cause of disease. Nursing then spread virus to susceptible piglets.
or newly weaned piglets may have rhinitis and conjunctivitis
accompanied by sneezing and nasal discharge. Pathogenesis
The primary site of virus replication is in the nasal mucous
Occurrence glands, and lachrymal or Harderian glands. Destruction of these
Porcine cytomegalovirus, sometimes referred to as inclusion body cells results in a marked rhinitis and conjunctivitis, sometimes
rhinitis, occurs in all age groups of previously uninfected swine, accompanied by fibrinopurulent exudates from bacterial coin-
including developing fetuses. Inclusion body rhinitis occurs only fections. Viremia follows mucosal replication. The virus localizes
in pigs and is found in all major swine-raising countries. Infection in epithelial and endothelial cells at many sites as well as within
is common but is often inapparent when herd immunity is pres- macrophages and reticuloendothelial cells, with occasional inclu-
ent or when pigs are over 6 weeks old. sions visible in affected tissues such as kidney. In fetuses and neo-
nates, capillary endothelium and sinusoids of lymphoid tissue are
Historical information infected. Congenitally infected fetuses excrete virus indefinitely.
Rhinitis accompanied by inclusion bodies was described in pigs Bone marrow damage is suspected of causing anemia in neonates.
in 1955 and was identified as a transplacental infection of fetal Widespread damage caused by the virus or secondary infections
pigs in 1961. Over the next 20 years, research reports provided can lead to death.
details on the disease and its manifestations in all ages of swine,
as well as characteristics of the virus. There are proposed rela- Clinical signs
tionships between infection with porcine cytomegalovirus and When disease is expressed, it is usually observed in 1- to 5-week-
reproductive failure in dams, neonatal losses, and as a predispos- old pigs. In suckling pigs, especially pigs less than 3 weeks old,
ing factor for other swine respiratory diseases, but evidence to signs include mucopurulent rhinitis accompanied by violent
support these associations is lacking. Losses from inclusion body sneezing, respiratory distress, conjunctivitis, shivering, and
rhinitis are small; both the disease and the infection are largely mouth-breathing. Although unusual, mortality can be high in
ignored by farmers. pigs born to naïve gilts or sows, or if the inclusion body rhinitis
is complicated by other diseases. Infected neonatal piglets appear
Etiology weak, anemic, or stunted, and there may be edema around the
Porcine cytomegalovirus is a beta-herpesvirus related to human throat and tarsal joints. Signs in older pigs or dams with viremia
herpesviruses 6 and 7. It has many of the characteristics of herpes- may include listlessness and mild anorexia but infection in these
viruses, including the ability to induce latent infection in carriers animals is often inapparent. Porcine cytomegalovirus is capable
that can later be reactivated by stress. Porcine cytomegaloviruses of causing fetal mummification, stillbirths, neonatal deaths, and
can be propagated in cell culture. In several epithelial type cells of cause failure of piglets to thrive when associated with infection
swine, the virus causes marked cell enlargement and formation of of naïve, pregnant sows but reproductive manifestations of the
large intranuclear inclusion bodies. disease appear to be rare.

Epidemiology Lesions
Serological testing has shown that most herds are antibody posi- In nursing piglets, the most commonly recognized gross lesion
is production of mucopurulent nasal exudate that can often
tive to porcine cytomegalovirus. The presence of the virus within
partially obstruct the nasal passages. Complications sometimes
a herd is often unrecognized because many sows are immune,
include sinusitis, otitis media, or pneumonia. In neonatal pigs,
show no signs of infection, and colostral antibodies protect petechial hemorrhages can be seen on the subcutis, serosal
piglets from lesions after they become infected. In endemically surfaces, and nasal turbinates, and subcutaneous edema can
infected herds, piglets 3 to 8 weeks old tend to become infected occur. Effusions in the thorax, abdomen, and pericardial sac,
and shed virus for around 30 days in nasal and ocular secretions, and edema of the lungs are often present. Microscopically, baso-
urine, cervical fluid, and the aerosol created by sneezing and philic intranuclear viral inclusions can usually be found in nasal
coughing. Transmission of virus is oronasal. Recovered ani- mucous glands, Harderian glands, and lachrymal glands of
mals may carry virus as a latent infection, becoming shedders nursing pigs. They may be present in renal glomeruli and tubu-
lar epithelium, liver, capillary endothelium, choroid plexus,
76 INCLUSION BODY RHINITIS
DISEASES CAUSED BY VIRUSES

glial cells, and in various epithelial type cells. Focal gliosis is Control
common throughout the central nervous system. Inclusions can Because the disease is seldom recognized as a cause of severe loss,
sometimes be found at several sites in fetuses. control measures are often ignored. Growing pigs are usually pro-
tected from large losses by antibodies provided by their immune
Diagnosis dams. As piglets grow, they are exposed to virus in the environ-
The disease is usually diagnosed after finding microscopic, baso- ment and respond by producing their own antibodies.
philic intranuclear inclusions in the tissues of a typical infected

VIRUSES
piglet. Where available, polymerase chain reaction can demon- Environmental conditions, if poor, should be improved. Herd
strate specific viral genetic material in affected tissues. Alterna- immunity via acclimatization of breeding stock is commonly
tively, an enzyme-linked immunosorbent assay or an immunoflu- practiced. There is no known effective treatment or medication.
orescence assay test on serum samples can be used to confirm the Antibiotics may be useful to control concurrent diseases or sec-
presence of porcine cytomegalovirus in a herd.. ondary bacterial infections.

INCLUSION BODY RHINITIS 77


SWINE DISEASE MANUAL

Influenza
Alternate names and abbreviations Etiology
Influenza A virus of swine, IAV-S, influenza A, flu, swine influ- Influenza viruses (eg, IAV-S) belong to the species Influenza A
enza (archaic), SIV (archaic), swine flu (archaic) virus, genus Influenzavirus A, and family Orthomyxoviridae.
Serological and virological evidence indicates that influenza B
Definition and C viruses from humans can occasionally infect pigs but this
VIRUSES

Influenza A in swine is a rapidly spreading viral disease character- appears to be rare and not often associated with significant clin-
ized by sudden onset of fever, oculonasal discharge, prostration, ical signs. A novel cattle-associated influenza C-like virus, with
and weakness, followed by paroxysmal coughing over a relatively the proposed designation “influenza D” has been found in some
short course of 5 to 7 days, and relatively low mortality in uncom- pigs with respiratory signs, although pigs are not thought to be
plicated outbreaks. reservoir hosts.
Several overlapping systems of nomenclature exist for influenza Influenza A viruses have glycoprotein “spikes” on their surface
viruses. With the complexity of the virus origins and the public that act as major surface antigens. There are at least 16 hemag-
health consequences of nomenclature, use of the terms swine flu, glutinin (H) and 9 neuraminidase (N) surface glycoproteins that
swine influenza, and SIV is discouraged, because these terms impli- are numbered to help identify virus strains. Influenza A viruses
cate swine as the origin of the virus. For this chapter, we will refer are very diverse and two viruses that share H and N types may
to the typical viruses that infect swine as “influenza type A viruses not be closely related. This high variability is the result of two
of swine” or IAV-S. The clinical expression of the disease caused by processes: mutation and genetic reassortment. Mutations cause
infection with IAV-S will be referred to as “influenza A.” gradual changes in the H or N proteins of the virus, a process
called antigenic drift. Genetic reassortment can cause more rapid
Occurrence changes. The influenza A genome consists of 8 individual gene
In the US, IAV-S is common and widespread. Serological surveys segments; when two different viruses infect the same cell, gene
show that nearly all herds in the US have antibodies to IAV-S. segments from both viruses may be packaged into a single, new
Influenza A occurs in swine of all ages and is present in most virus. An important aspect of reassortment is that it can generate
major swine-raising countries. viruses containing either a new H, a new N, or both. Such abrupt
changes are called antigenic shifts. There is often poor cross-pro-
Historical information tection between IAV-S subtypes because of the considerable
The first influenza virus to be recognized in pigs was an H1N1 genetic and antigenic variation among IAV-S viruses.
virus known as the classical influenza A virus. Pigs are thought
The classic subtype in swine, H1N1, has been the predominant
to have acquired this virus in 1918, at the same time as the severe
subtype in swine in the US since 1918. H3N2 viruses acquired
Spanish flu pandemic in humans. H1N1 viruses circulated in
from humans were also detected during this period. However,
both species after this time but diverged genetically in the two
they did not become established as stable, widespread lineages
host populations.
in pigs until more recently. Around 1998, “triple reassortant”
In 2009, a novel H1N1 virus emerged in human populations. It H3N2 viruses first emerged in North America and soon spread
contained genetic components from swine, avian, and human to other regions. These viruses contain H and N proteins derived
influenza viruses. This virus caused a human pandemic from from human influenza viruses, and internal proteins from
2009 to 2010, and then became established as a seasonal human the classical influenza A virus, an avian influenza virus, and a
influenza virus. It is currently the predominant H1N1 virus cir- human influenza virus. The particular combination of internal
culating in people and has been found sporadically in pigs since genes carried by these viruses is known as the triple reassortant
its emergence. internal gene (TRIG) cassette. Viruses that contain this cassette
While influenza A has always been a significant public health seem to be prone to increased antigenic drift. They also seem to
concern, the emergence of the pandemic H1N1 strain in 2009 readily acquire new H and N genes, which has resulted in new
has resulted in increased research on the disease and coordinated TRIG-containing viruses with various combinations of H1, H3,
surveillance activities among veterinary, public health, and wild- N1, or N2 from additional human influenza viruses, and/or H1
life sectors. There is clear evidence of interspecies transmission and N1 from the classical influenza A virus.
of influenza viruses among swine, chickens, ducks, turkeys, wild
birds, equines, companion animals, and people. When concur- Epidemiology
rent infection with more than one virus occurs in any of the spe- In pigs, influenza viruses are transmitted in droplets and aerosols
cies, the potential for genetic reassortment between the viruses created by coughing and sneezing, and by contact with nasal dis-
occurs which can result in development of new viral strains (ie, charges, either directly or on fomites. Viremia is not a feature and
genetic shift). there is no evidence that transplacental transmission of influenza

78 INFLUENZA
DISEASES CAUSED BY VIRUSES

A viruses occurs in pigs. Asymptomatic carrier status was not usually develops after a few days, at which time the fever has often
thought to occur, but recent evidence from persistent outbreaks started to diminish. Recovery begins about the sixth day and the
in herds not having any known ongoing risk of exposure is herd is largely recovered in 1 to 2 weeks. Morbidity is high; mor-
changing the conventional thinking on the topic. The presence of tality is low in most uncomplicated outbreaks.
antibody (maternal or active) will mitigate the severity of disease
Uncomplicated influenza A usually causes little death loss, but
and influence the rapidity of spread through a population. Thus,
losses of weight and body condition can be significant, as can
influenza A can become an endemic disease in populations that
the cost of medication and an increase in the incidence of gastric

VIRUSES
use continuous pig flows.
ulcers. Concurrent infections with other respiratory diseases can
Virus shedding can begin within 1 to 3 days after a pig becomes greatly increase mortality and culls.
infected and typically continues for 4 to 10 days. There is some
Reproductive problems occasionally associated with IAV-S are
evidence that aerosol transmission might be possible, at least
likely incidental to the systemic effects of acute illness experi-
within densely populated pig barns and possibly over longer
enced by sows. Acute illness during pregnancy may result in abor-
distances in swine-dense regions. Influenza A viruses of swine
tion and, sometimes, small, weak litters.
have been isolated from air samples inside barns and immediately
outside exhaust fans.
Lesions
Both human-to-pig and pig-to-human transmission of IAV-S The gross lesions include lobular distribution of congestion,
viruses have been documented to occur. As a result of increased firmness, atelectasis, emphysema, and perhaps pneumonia. Gross
surveillance since the 2009 pandemic, transmission events lesions are often restricted to the apical and cardiac lobes but may
between pigs and people at livestock exhibitions are detected by affect entire lungs. Areas of atelectasis are irregular, darkened, and
public health systems most years in the US. firm. Lesions often appear as distinct, scattered lobules. Airways
Influenza A virus probably does not survive more than 2 weeks may contain blood-tinged exudate. Regional lymph nodes are
outside the host and can be inactivated by many disinfectants. moderately enlarged. Microscopic lesions include degeneration and
necrosis of portions of airway epithelium and obstruction of air-
ways with exudate. There may be bronchointerstitial pneumonia.
Pathogenesis Marked edema and congestion of the entire lung may be present in
The IAV-S virus gains access by the nasopharyngeal route with
severe cases. Necrotizing bronchiolitis is highly suggestive of IAV-S.
receptor-mediated entry into respiratory epithelium, where it
causes inflammation with widespread degeneration and necro-
sis of cells lining bronchi and bronchioles. Also, virus can be Diagnosis
demonstrated in interalveolar septa, sometimes in turbinate A history of sudden onset of a severe respiratory illness in many
epithelium, and in mediastinal lymph nodes. In the lung, alveolar animals, along with typical signs and lesions, is often adequate
type 2 pneumocytes are affected and produce less surfactant. This for a field diagnosis. It is useful to confirm the diagnosis since
impairs phagocytosis of secondary microorganisms by alveolar the disease may reappear and go unrecognized in a less typical,
macrophages. Epithelial necrosis compromises the mucociliary subacute form.
escalator, compromising the removal of cellular debris and bacte- Confirmation of the diagnosis can be made in several ways, but
ria. The inflammatory exudates in the smaller airways sometimes detection of the virus or its components is most successful from
block them, leading to atelectasis, emphysema, and bronchoint- acutely affected (febrile) pigs. The virus can be identified by
erstitial pneumonia. In most cases, epithelial damage is repaired, fluorescent antibody test on fresh lung sections or by immunohis-
and the lesions resolve in 7 to 10 days if the infection is uncom- tochemistry staining on formalin-fixed lung sections from acutely
plicated. The severity of the effect of infection is mitigated by the affected, euthanized pigs. A polymerase chain reaction (PCR)
quantity of homologous circulating antibody (maternal or active). test is often used to detect virus in nasal swabs or lungs and has
Most of the mortality associated with IAV-S virus infection is the the advantage that the subtype can be immediately determined,
result of secondary bacterial infections or concurrent infections and the product can be sequenced for epidemiologic purposes.
with other primary agents of pneumonia. The IAV-S virus can be isolated from nasal swabs or lung tissue
in chick embryos. There are commercially available influenza A
Clinical signs antigen detection kits that can be used to detect IAV-S on nasal
In a typical outbreak, there is a sudden onset of a marked respira- swabs. Hemagglutination inhibition (HI) assays have been used
tory illness affecting most pigs in the herd. Depending on the herd, to evaluate titer levels but knowledge of the virus subtype respon-
clinical signs may be seen only in certain age groups, while other sible for the infection is required for accurate test results. In some
animals remain asymptomatic. The clinical signs may include fever, cases, paired serum samples taken early and late in an outbreak
lethargy, anorexia, weight loss, coughing, sneezing, nasal and ocular can be used to demonstrate a rising serotype-specific antibody
discharge, conjunctivitis, and labored breathing (“thumping”), but titer to IAV-S. Serological testing is complicated by the fact that
all of these signs do not occur in all infected animals. The cough
INFLUENZA 79
SWINE DISEASE MANUAL

several subtypes of IAV-S can be present in a group of pigs and used. Directions provided with the vaccine should be followed
most serological tests do not sufficiently cross-react to detect anti- carefully. Cross-protection between subtypes should not be
bodies of all IAV-S subtypes. expected in most situations.
Because there are multiple subtypes of IAV-S in swine popula- In modern production system units, control of influenza A in
tions, mere detection of IAV-S is not sufficient to warrant imple- recently weaned pigs usually requires stabilization of the breeding
mentation of specific control strategies. The offending IAV-S can herd by acclimatization or vaccination. Vaccination prior to far-
be subtyped by some laboratories using PCR genetic sequencing rowing to stimulate colostral protection for the young piglets is
VIRUSES

or serological methods (HI assays with multiple virus subtypes). sometimes practiced. New additions to the breeding herd should
This allows for more accurate vaccine selection and application. be vaccinated during their isolation period.
It is quite common to have other viral, bacterial, mycoplasmal, Successful control of influenza A in the grow-finish phase of
and parasitic diseases in association with IAV-S, especially in out- known IAV-S positive herds can be difficult. Nursery pigs can be
breaks with considerable mortality. vaccinated prior to moving them to the finisher. However, mater-
nal antibody interference with vaccination is a major concern in
Reproductive failure associated with influenza is due to the
timing of vaccination since antibody levels are highly variable in
effects of acute disease in sows. The virus cannot be detected in
populations and even low levels of antibody may interfere with
aborted fetuses but can be detected in acutely affected, febrile
immune response. There is little cross-protection between sub-
sows. Serology is not a reliable method of diagnosis of reproduc-
types, making vaccine selection challenging. Often, the offending
tive failure since most sow herds will be serologically positive
virus must be subtyped to aid in selection of the correct antigen.
from previous exposure to the agent.
On infected premises, all-in, all-out pig flow with cleaning and
Control disinfection of facilities between groups helps prevent virus
There are no specific therapeutics for IAV-S infection. Often, carryover, as does vaccination of the breeding herd to stabilize
antimicrobials are used to control or prevent secondary bacterial the infection to prevent virus transmission to offspring. Depopu-
infections. Aspirin or other anti-inflammatory agents may offer lation of continuous flow nurseries or finishers may be necessary
relief to severely affected pigs. Treatment should emphasize pro- to stop virus circulation. Implementing consistent and effective
viding a comfortable environment and ready access to fresh feed control strategies for other respiratory insults that may be con-
and water. currently present in the herd (eg, bacteria, viruses, parasites, air
quality, and poor ventilation) can also decrease the severity of
Prevention has depended largely on maintaining a closed herd
IAV-S outbreaks.
to avoid introducing infected pigs. Inactivated vaccines are
widely used in Europe and the US. Several types of commercial
vaccines are available and autogenous products are commonly

80 INFLUENZA
DISEASES CAUSED BY VIRUSES

Japanese encephalitis
Alternate names and abbreviations Although humans and most domesticated animals can develop
JE, JEV, Japanese B encephalitis (archaic) clinical signs after becoming infected with Japanese encephalitis
virus, they are considered to be incidental hosts as they typically
Definition produce low levels of viremia and are not thought to be signifi-
Japanese encephalitis is a mosquito-borne disease affecting many cant in further virus transmission. In contrast, birds and pigs can

VIRUSES
animals, including wild birds, horses, swine, and people. In swine, amplify Japanese encephalitis virus and are therefore important
the only evidence of disease may be reproductive failure. Japanese in the epidemiology of transmission of the virus. Domesticated
encephalitis is one of the most prevalent causes of encephalitis in pigs are considered to be particularly efficient amplifying hosts, as
humans in Asia; pigs are an important amplifying host for the virus large numbers of susceptible young swine are produced each year,
in areas where people, pigs, and the mosquito vector overlap. often in close proximity to people. In addition, boars are report-
edly able to transmit the virus in semen.
Occurrence
Japanese encephalitis occurs in most domestic animals and also Pathogenesis
in wild birds, reptiles, perhaps chickens, and people. Swine can After an incubation period of around 3 days, infected pigs remain
be infected, and all ages are susceptible. Major epidemics have viremic for several days. In porcine fetuses, the virus often causes
occurred in horses, donkeys, and people. the development of anomalies in the brain as well as encephalitis
and degenerative neuronal changes.
Japanese encephalitis is confined largely to southeastern Asia,
Indonesia, and major Pacific islands. At least 20 countries have Transplacental infection in swine sometimes occurs. The effect
reported occurrence of the disease. In many locales, epidemics of the virus depends on whether the fetuses are immunologically
correlate with the mosquito season. competent. When dams are infected between 40 to 60 days in
gestation, fetuses are often killed, and some are mummified.
Historical information There may be no effect on fetuses if dams are 85 or more days in
Japanese encephalitis was first described in 1933. Major epi- gestation. Fetal deaths are believed to be caused by destruction of
demics in people have been reported from India, Japan, Korea, vital stem cells.
and Nepal. The disease has not been reported in the western
hemisphere, although the range seems to be increasing due to Clinical signs
the expanding range of suitable mosquito vectors. Abortion Mature swine seldom show signs of infection other than those of
in women and encephalitis in children are caused by Japanese reproductive failure. Signs may occur in fetuses or piglets if their
encephalitis and the virus is recognized as an important zoonosis. dam was infected during pregnancy. Signs include stillborn and
mummified fetuses or weak pigs that may have signs of central
Etiology nervous system disease. Subcutaneous edema and hydrocephalus
Japanese encephalitis virus is an arbovirus, meaning it is transmit- may be present in stillborn pigs. Abortions seldom occur. Infected
ted to mammalian hosts through an arthropod vector. The virus boars may have orchitis, reduced libido, and disturbance of sper-
is a member of the genus Flavivirus and family Flaviviridae and is matogenesis. Young, susceptible pigs occasionally contract Japanese
related to other zoonotic arboviruses including West Nile virus, encephalitis and show signs of central nervous system lesions.
Murray Valley encephalitis virus, and St Louis encephalitis virus.
It does not persist in the environment and is easily inactivated by Lesions
many disinfectants. Gross lesions in stillborn or infected neonatal piglets include
hydrocephalus, subcutaneous edema, ascites, hydrothorax, hem-
Epidemiology orrhages on serous membranes, congestion of lymph nodes, and
Mosquitoes become infected by feeding on viremic hosts: usually necrotic foci in the liver and spleen. Other lesions may include
pigs, horses and other equids, and birds. The virus is then trans- congested meninges, hypoplastic areas in the cerebral cortex,
mitted through the mosquitoes’ saliva when they subsequently hypoplasia of the cerebellum, or spinal hypomyelinogenesis.
feed on uninfected people or animals. The virus is transmitted Histologically, in infected fetuses or piglets there is diffuse
mainly by mosquitoes of the genus Culex, although some Aedes nonsuppurative encephalitis and neuronal degeneration and
and Anopheles can also act as vectors; the virus can be transmitted necrosis in the cerebrum and cerebellum. In mature boars, there is
transovarially in some species of mosquitoes. excessive fluid in the tunica vaginalis, orchitis, epididymitis, and
degenerative changes in seminiferous epithelium.

JAPANESE ENCEPHALITIS 81
SWINE DISEASE MANUAL

Diagnosis Control
A definitive diagnosis is often based on isolation and identifica- It may be possible to break the cycle of infection by controlling
tion of the virus from fetuses or infected piglets. Virus is usually mosquito populations, but this is often impractical. The disease
isolated from brain extracts inoculated into suckling mice or cell can be controlled by vaccinating the breeding stock; vaccines are
cultures. Virus can be identified by neutralization tests in suckling common in parts of Asia. Young gilts and boars are vaccinated
mice or cell culture. Alternatively, viral antigen in tissues from twice at 2- to 3-week intervals prior to the mosquito season.
infected fetuses or stillborn pigs can be identified by fluorescent Growing pigs are also vaccinated in endemic areas.
VIRUSES

antibody or immunohistochemistry staining of formalin-fixed


tissues. In suspected outbreaks, serial serum samples from pigs
sometimes are used to show a rising titer against Japanese enceph-
alitis virus. Serological tests that reveal antibody in fetuses are also
useful in diagnosis. Polymerase chain reaction assays have been
developed to identify the virus in tissues and serum.

82 JAPANESE ENCEPHALITIS
DISEASES CAUSED BY VIRUSES

Parvovirus
Alternate names and abbreviations disseminate virus in feces and fluids, including placental fluids
PPV, parvo, SMEDI (stillbirth, mummification, embryonic and mummified fetuses. Fetuses infected during gestation can be
death, and infertility) born as infected immunotolerant piglets that can pass the virus
intermittently or continuously, but the significance or likelihood
Definition of this phenomenon is unknown. Because parvovirus is quite

VIRUSES
Porcine parvovirus can cause reproductive failure in naïve dams. It resistant to environmental influences, contaminated facilities are
is characterized by the occurrence of large numbers of mummified an important source of ongoing exposure.
fetuses, an increase in the number of returns to estrus, small litters, Oronasal transmission can occur among susceptible swine of all
failures to farrow, decreased farrowing rate, and, rarely, abortion. ages. However, serological surveys show that many herds with
Many manifestations of reproductive failure seen with porcine endemic infection contain at least some animals that are suscepti-
parvovirus accompany other reproductive diseases of swine. The ble. Transplacental infection of embryos and fetuses is the result of
many manifestations of reproductive failure should not be con- dams failing to develop an active systemic immunity prior to preg-
sidered pathognomonic for porcine parvovirus. nancy. Passive immunity persists in some gilts beyond the time they
are old enough to be bred and this maternal antibody can interfere
Occurrence with their development of an active immune response to natural
Porcine parvoviral infection is endemic in most swine herds. It is or vaccine exposure. Active immunity usually develops following
usually subclinical and is a very common infection. Prior to the exposure of gilts to parvovirus in the environment as passive immu-
emergence of porcine reproductive and respiratory syndrome nity wanes. Gilts with little or no immunity can become infected
(PRRS), porcine parvovirus was probably the most commonly during their first pregnancy. If infected during pregnancy, virus may
diagnosed infectious cause of reproductive failure in swine. Repro- cross the placenta and infect some or all of their embryos or fetuses.
ductive failure is much more likely to occur in gilts than in sows, Porcine parvovirus can be found in semen of naturally infected
since lifelong immunity is induced after active infection. Porcine boars, hence it can likely be transmitted mechanically through nat-
parvovirus occurs worldwide in major swine-raising countries. ural breeding or artificial insemination.

Historical information Pathogenesis


The earliest publication about porcine parvovirus occurred in Only porcine parvovirus 1 is known to cause reproductive failure
1965, when the agent was identified as a cell-culture contami- or disease. Pregnant, infected dams develop a viremia during the
nant. Further investigation over the next decade confirmed por-
acute phase of porcine parvovirus 1 infection. Virus then may
cine parvovirus as a major cause of reproductive failure in pigs. To
cross some or all of the separate placentas of developing embryos
this day, vaccines developed during that period remain effective at
or fetuses. Fetal infection during the first 35 days of gestation
preventing losses from porcine parvovirus.
causes death and resorption of embryos, resulting in irregular
returns to estrus or reduced litter size. Infection between 35 and
Etiology 70 days of gestation results in fetal death and mummification.
Porcine parvovirus is a single-stranded DNA virus. The strain of
porcine parvovirus typically identified in cases of reproductive Since fetuses become immunologically competent by about the
failure is the serotype designated as porcine parvovirus 1. All por- 70th day of gestation, fetuses older than that may successfully
cine parvovirus 1 isolates are similar antigenically. At least 6 other resist the effects of becoming infected and survive to full term.
parvoviruses have been detected in swine (porcine parvovirus 2, After one embryo or young fetus is infected, the virus may slowly
3, 4, 5, 6, and 7), as well as a closely related virus called porcine spread through the uterus and infect some or all of the others.
bocavirus. However, none of these appear to be pathogenic in The direct effect of porcine parvovirus 1 on the uterus may also
swine and they are therefore considered to be commensals. Por- contribute to reproductive failure.
cine parvovirus is quite resistant to environmental degradation
and many disinfectants; it can persist for at least 4 months on The death of embryos and fetuses is attributed to the direct
contaminated premises. Porcine parvovirus hemagglutinates destructive effect of porcine parvovirus on their organs and tis-
many kinds of erythrocytes, a feature useful in its laboratory iden- sues. Many kinds of cells are affected, especially mitotically active
tification. Porcine parvovirus does not cause diarrhea in swine capillary endothelium and neurons. Extensive endothelial cell
although related parvoviruses do so in several other species. damage may be reflected in damage to many organs.

Epidemiology Clinical signs


Parvoviral infection is endemic in most swine herds. Following In most swine, a transient leukopenia occurs within 10 days of
infection, virus is shed in the secretions and excretions from infection with porcine parvovirus 1. However, this and any other
the animal for approximately 2 weeks. An infected dam will

PARVOVIRUS 83
SWINE DISEASE MANUAL

indication of infection are inapparent in all swine except develop- may be difficult to interpret since cross-sectional sampling will
ing fetuses. Infection of a naïve herd of pregnant females results often reveal a wide range of titers. Demonstration of serocon-
in irregular return of bred animals to estrus, increased numbers version in many gilts and sows with paired serology may be used
of mummified fetuses, smaller litter sizes, increases in animals for a positive diagnosis but acute samples are often not available.
checked positive for pregnancy that fail to farrow, and prolonged Conversely, the absence of titers may aid in exclusion of porcine
gestation lengths. The increase in mummified fetuses after a parvovirus 1 as the cause. If mummies are not available for evalua-
normal gestation period is the hallmark of porcine parvovirus 1. tion, efforts should concentrate on gilt serology, comparing titers
VIRUSES

Abortions attributable to this virus are rare. from unmated gilts to those exhibiting signs of infertility. There
are likely other viruses that can infect unborn fetuses, particularly
Lesions in litters of naïve pregnant gilts; some of these may be present
Gross lesions are not apparent in the dams. The most important concurrently with porcine parvovirus 1 and should be considered
gross observation with porcine parvovirus 1 infection is mum- as differentials. Examples include PRRS, porcine circovirus type
mified fetuses. In experimentally infected dams, nonspecific 2, porcine circovirus type 3, members of Picornaviridae (enterovi-
microscopic and gross lesions are described but are not often of ruses, Teschovirus, Sapelovirus), and Astrovirus, among others.
diagnostic value.
Control
Diagnosis There is no effective treatment for porcine parvovirus infection.
The lack of illness in the dams, coupled with increased mummies, Probably the best way to prevent porcine parvovirus infection is
irregular return to estrus, decreased litter size, or pseudopregnancy to vaccinate all susceptible breeding stock twice at 2- to 3-week
occurring primarily in gilts is suggestive of porcine parvovirus 1 intervals with the second dose administered at least 3 weeks before
infection. A presumptive diagnosis can be confirmed by demon- breeding. Killed vaccines are available and effective. Pre-breeding
strating porcine parvovirus 1 antigen in the tissues (lungs, hearts, boosters are recommended in herds of unknown status. Vacci-
and thoracic fluids) of several truly mummified (died before 70 nation of susceptible breeding stock lacking maternal antibody
days of gestation showing severe in utero dehydration) fetuses using should stimulate an active immunity that will protect the devel-
polymerase chain reaction or immunofluorescent antibody (IFA) oping fetuses. It is important to note that virus circulation within
testing. The preferred specimen for diagnosis by IFA testing is the a population cannot be completely prevented by vaccination. The
lung tissue from several mummified fetuses less than 16 cm long. virus will continue to circulate in the face of vaccination programs;
Larger fetuses are not satisfactory since their tissues may contain hence eradication is not likely and continued vaccination should be
antibody that interferes with the procedure. an ongoing part of the herd vaccination program.
Sometimes antibody can be demonstrated in fluids or sera from Another control alternative is to expose seronegative stock to
stillborn pigs or from pre-suckle neonatal live pigs. While this may animals or environmental materials from seropositive older stock.
be an indirect indication of intrauterine infection with porcine par- In this situation, animals should be used for breeding only after
vovirus 1, there is considerable likelihood of false positive serologi- demonstrated seroconversion.
cal reaction from these specimens. Serology from breeding animals

84 PARVOVIRUS
DISEASES CAUSED BY VIRUSES

Porcine circovirus associated diseases


Alternate names and abbreviations Historical information
Porcine circovirus disease, PCVD, PCVAD, postweaning multisys- Circoviruses infect other species of mammals and avians. They
temic wasting syndrome, PMWS, porcine dermatitis and nephrop- cause psittacine beak and feather disease in chickens as well as
athy syndrome, PDNS, circovirus, circo, PCV, PCV2, PCV3 lethal infections in pigeons, canaries, and finches. Porcine circovi-

VIRUSES
rus type 2 infection of pigs was reported in North America starting
Definition around 1994. Over the next decade, similar outbreaks occurred in
Porcine circovirus is a common virus of pigs, found throughout Europe and most swine-rearing areas of the world and were consis-
the world. It was originally described as an adventitious virus of tently associated with PCV2 infection. Genomic sequencing and
porcine tissue culture cells (now known to be porcine circovirus restriction fragment length polymorphism techniques have defined
type 1). The detection and emergence of a novel porcine circo- several distinct genotypes within PCV2, but the clinical relevance
virus (porcine circovirus type 2; PCV2) in the 1990s coincided of these distinctions has not been demonstrated.
with the occurrence of new clinical syndromes in swine referred In 2016, PCV3 emerged and has been increasingly detected in
to as postweaning multisystemic wasting syndrome or porcine swine, sometimes associated with cases of PDNS, fetal infection,
circovirus associated disease. Since its appearance, PCV2 and or postnatal systemic insult.
disease associated with its presence has become widely distributed
in most developed swine industries around the world. The term
porcine circovirus associated disease (PCVAD) is now used in
Etiology
Circovirus is a non-enveloped, single-stranded DNA virus unique
North America to refer to the different disease manifestations
for its small size (approximately 1800 bp), circular genome, and
associated with PCV2. In Europe, the term porcine circovirus
hardiness within the environment. Circovirus is sensitive to most
disease (PCVD) is used preferentially to the US term PCVAD.
disinfectants, but chlorhexidine, ethanol, and iodine tend to be less
Circoviruses are also thought to be one of the causes of an
effective than other commercial products. Three types of PCV have
immune-mediated condition called porcine dermatitis and
been described in swine: porcine circovirus types 1, 2 and 3.
nephropathy syndrome (PDNS) in swine.
Multiple genotypes of PCV2 have been described (eg, PCV2a,
Recently, another type of circovirus, porcine circovirus type 3
PCV2b, PCV2c, PCV2d, and PCV2e). The prevalence of each
(PCV3), has been detected in swine herds around the world.
genotype has varied over time, but all are considered to be of the
Fetal deaths, porcine dermatitis and nephropathy syndrome, and
same type and there is little evidence to suggest significant differ-
systemic post-natal infections have recently been associated with
ences in pathogenicity, antigenicity, or cross-protection between
the presence of this virus; definitive studies of causation, patho-
these genotypes.
genesis, and immunity are in progress.
Recently characterized PCV3 appears to have distinct genetic and
Occurrence antigenic differences from PCV2, but the clinical and epidemiolog-
Infections and disease associated with PCV2 are reported ical significance of these differences is unknown at this time.
from nearly every country with a significant commercial swine
industry. Porcine circovirus type 2 is strongly associated with Epidemiology
the occurrence of postweaning multisystemic wasting syndrome, Only swine are known to be susceptible to PCV 2 or 3 infection.
porcine circovirus associated disease, porcine respiratory disease Globally, nearly all commercial swine herds are seropositive for
complex, and is occasionally associated with reproductive failure. PCV2. Seroconversion usually occurs by 2 to 4 months of age,
irrespective of whether clinical signs of porcine circovirus asso-
The number of reported cases of porcine circovirus associated
ciated disease are observed. Preliminary studies suggest PCV3 is
disease increased dramatically in the first decade of this century.
Porcine circovirus associated disease is widespread in Asia, detectable in a high percentage of herds globally as well. Oronasal
Europe, and the US, and has been reported from virtually all transmission through direct contact is the most likely and the
swine-producing areas of the world. As an apparent immune-me- most efficient mode of transmission. Porcine circovirus type 2
diated sequel to viral infection, PDNS is sporadically reported or type 3 is found in most secretions and can persist for weeks in
associated with circovirus infections. contaminated organic matter. Experimentally, infection of preg-
nant susceptible dams with PCV2 can result in transplacental
Fetal infections with PCV2 and PCV3 may be inapparent or can infection of fetuses and fetal deaths or long-term virus carriers
be associated with reproductive failure, fetal deaths, mummifica- within survivors; PCV3 can be expected to behave similarly.
tion, or abortion. Both viruses are common in swine populations,
and other viral agents or risk factors for reproductive failure are
frequently present, so disease associations with PCV2 and PCV3
are sometimes difficult to prove.
PORCINE CIRCOVIRUS ASSOCIATED DISEASES 85
SWINE DISEASE MANUAL

Pathogenesis including PCV 2 or 3. In this condition, individual pigs, usually


Pigs infected with PCV2 develop viremia of variable duration, from 8 to 18 weeks of age, develop small red-purple blotches on
with replication and persistence most extensive in macrophages the skin, sometimes slightly raised, and most obvious on the hind
and monocytes of lymphoid organs and lung, and in epithelial legs and perineum. Focal lesions can expand and coalesce, extend
cells in various organs. Characteristic microscopic lesions include over the abdomen, and eventually cover the whole body. Most
lymphoid depletion at multiple sites, chronic lymphohistiocytic pigs with PDNS eventually die.
to granulomatous inflammation, and an erosive bronchiolitis Clinical signs of transplacental infections with PCV 2 or 3 mimic
VIRUSES

with fibrosis. those of other viruses which cause in utero death of fetuses less
Reproduction of typical lesions by inoculation with PCV2 has than 70 days gestation (eg, porcine parvovirus). This is usually
been inconsistent, but there is general agreement that PCV2 is manifested as increases in mummies, small litters, sows that fail
a necessary (although perhaps not sufficient) cause of porcine to farrow with overall decreased farrowing rate or litter size, with
multisystemic wasting syndrome and porcine circovirus associated occasional abortions also reported.
disease expression. Mechanisms that allow disease expression and/
or immunity remain obscure but are related to macrophage activa- Lesions
tion with compromised antigen processing and presentation thus Gross lesions of porcine circovirus associated disease may include
thwarting an effective immune response. Studies demonstrate that pallor, marked enlargement of all lymph nodes and perhaps
PCV2 infections have immunosuppressive characteristics, which spleen, interstitial pattern of pneumonia in lungs, and occasion-
can exacerbate the impact of other pathogens in a herd. The role of ally multiple foci of pallor within swollen kidneys. Less often
PCV 2 or 3 in occurrence of PDNS, and the circumstances under present are icterus, a fluid-filled intestine, atrophy of the liver,
which that syndrome is expressed, is even less clear. The blotchy and ulceration of the pars esophagea of the stomach. Microscopy
purple skin lesions and nephropathy of this syndrome are suspected reveals depletion of germinal centers in lymphoid tissues with
to be an immune-mediated sequel to viral infection and are not replacement by histiocytes and multinucleated giant cells. Baso-
necessarily specific for either PCV 2 or 3 infections. philic intracytoplasmic inclusion bodies can sometimes be seen in
macrophages in lymph nodes, tonsils, spleen, and Peyer’s patches.
Clinical signs Lymphohistiocytic to granulomatous inflammation can be pres-
Subclinical infections are common, as many herds are infected ent in any organ, typically lungs and/or lymphoid tissues, and
with PCV 2 or 3 with no apparent clinical signs. When disease PCV2 antigen is abundantly present within characteristic lesions
occurs, multiple body systems can be affected, with considerable when examined by immunohistochemistry or in situ hybridiza-
variation in morbidity and mortality. Few infectious agents have tion assays. Erosive bronchiolitis and fibrosis of airways are also a
more variation in signs, tissues affected, or in individual and herd common finding.
impact than PCV2. With PDNS, there are small, circumscribed circular to irreg-
The effects of porcine circovirus associated disease in growing ular, deep-purple discolorations of skin that become apparent
pigs varies from severe disease with high mortality to only mild on the hindquarters, perineum, and flanks. Over the course of
clinical disease. Disease is now usually referred to simply as differ- several days, the skin discolorations either resolve or expand and
ent expressions of porcine circovirus associated disease. coalesce. Kidneys are usually swollen with foci of pallor and/
or hemorrhage in parenchyma. Microscopically, there is non-
In severe forms of the disease, pigs between 2 and 4 months of suppurative vasculitis in the affected areas of the skin. Kidneys
age sicken and rapidly waste away while cohorts seem to grow often have swollen glomerular tufts, inflammation, and protein in
normally. Morbidity varies from 2% to 30% but case fatality can tubules. Multifocal nonsuppurative interstitial nephritis can also
be high, approaching 80% in unvaccinated herds. The severity of be a feature. The lesions are thought to be due to immune-me-
clinical signs varies between individuals and includes gradual or diated (immune complex) vasculitis. Lesions of PDNS are not
rapid wasting, unthriftiness, rough hair coat, polypnea, dyspnea, specific for porcine circovirus.
pallor, diarrhea, and occasionally icterus. Many affected pigs and
clinical survivors are severely stunted. Less affected pigs continue
to grow but contribute to variation in production efficiencies and
Diagnosis
Diagnosis of porcine circovirus associated disease is based on
market weights, whereas non-clinical pigs in the same groups per-
clinical features and gross lesions and confirmed by finding typi-
form quite well. Both signs and lesions vary considerably among
cal microscopic lesions and demonstrating PCV2 within lesions.
affected herds. Because clinical signs are not specific, a diagnosis
Microscopic lesions are quite characteristic and virus inclusion
of porcine circovirus associated disease requires compatible
bodies can sometimes be identified in depleted lymph nodes,
lesions and demonstrable PCV2 within typical lesions.
spleen, tonsils, or Peyer’s patches. Polymerase chain reaction
Porcine dermatitis and nephropathy syndrome is suspected of (PCR) will confirm the presence of virus but does not confirm
being a sequel to infection by one of several different viruses, the presence of disease. However, demonstration of high virus

86 PORCINE CIRCOVIRUS ASSOCIATED DISEASES


DISEASES CAUSED BY VIRUSES

load with quantitative PCR techniques along with typical signs Control
and lesions can implicate PCV2 in the disease process. Similarly, There is no specific treatment for pigs infected with PCV type 2
isolation and identification of PCV2 is not useful for diagnosis or type 3. Anti-inflammatory agents and/or antimicrobials are
since asymptomatic infection is widespread in swine populations. sometimes used in groups of pigs experiencing outbreaks of por-
Serological testing for PCV2 has been developed to confirm cine circovirus associated disease to assist in managing secondary
infection but finding antibodies to circovirus in the herd is not infections. The use of all-in, all-out pig flow, thorough cleaning
diagnostic for disease, since most herds are positive without and disinfection between batches of pigs, and early segregation

VIRUSES
discernible disease. Porcine circovirus associated disease must be and euthanasia of severely affected pigs are measures that may
differentiated from other bacterial or viral diseases, especially por- help control the disease.
cine reproductive and respiratory syndrome (PRRS), which has Vaccines that were developed for PCV2 shortly after the virus
similarities in clinical signs and lesions. Concurrent coinfections emerged do not prevent infection but are efficacious in prevent-
and diseases are common. ing most clinical signs of PCVAD. The timing of vaccination
Diagnosis of PDNS is by observing typical clinical signs, gross and the selection of the appropriate population to be immunized
lesions, and by histopathology. Demonstration of PCV 2 or 3 (ie, breeding herds, replacement stock, neonatal pigs, or growing
antigen in the lesions is not consistent. pigs) vary between farms, depending on the characteristics of
the affected populations and the physical layout of the farm. It is
In cases where reproductive failure (evidenced by mummification, common to vaccinate gilts at the time of selection or before first
low farrowing rate, stillbirths, and abortions) is due to infection breeding. Ongoing vaccination of sow herds is not consistently
with PCV2, the virus should be demonstrable in fetal hearts as practiced. However, booster vaccination of sow herds may be war-
well as other tissues. ranted when the virus is determined to be circulating in gestating
Porcine circovirus type 2 can contribute to porcine respiratory animals or suckling piglets despite having a vaccination program
disease complex in some herds. Experimental coinfection stud- in place. Vaccination of offspring is usually performed at weaning,
ies as well as anecdotal evidence suggest that PCV2 is likely perhaps followed by booster vaccination at least 3 weeks later.
to increase the severity of pneumonia caused by Mycoplasma Although a few studies have shown that that PCV2 can be eradi-
hyopneumoniae, influenza A virus of swine, PRRS virus, and cated from an affected farm by total depopulation, the ubiquitous
others. In addition, many, but not all, field cases of PCVAD have nature of the virus makes it unlikely that commercial farms can
concurrent viral infections or some other evidence of immune be maintained free of the virus over time. Until the epidemiology,
stimulation that seems to allow replication and pathologic transmission, and virulence factors relating to PCV2 are further
effects of PCV2 in swine. Risk factors and coinfections should understood, the use of total depopulation as a control strategy is
be thoroughly investigated. Herds that are endemically infected not recommended.
with other significant pathogens, are operated on a continuous
flow basis, are in swine dense areas, or practice poor biosecurity Porcine circovirus type 2 and type 3 have been identified in the
have been shown to be more likely to experience an outbreak of semen of acutely affected boars. However, the role of semen as
porcine circovirus associated disease. a source of these infections on sow farms remains unknown.
Booster vaccination of boars prior to entry into boar studs is
At the present, PCV3 diagnosis relies on detection and identifica- probably warranted.
tion of the virus by PCR or genomic sequencing.
Currently, there is no commercial vaccine for PCV3. The extent of
cross-protection afforded by existing PCV2 vaccines is unknown.

PORCINE CIRCOVIRUS ASSOCIATED DISEASES 87


SWINE DISEASE MANUAL

Porcine picornaviruses
Alternate names and abbreviations In the 1960s, Dr. Howard Dunne coined the term “SMEDI” to
Enteroviruses, Teschen disease, Talfan disease, Sapelovirus refer to symptoms of reproductive failure (specifically stillbirths,
mummification, embryonic death, and infertility) that did not
include abortion. Eventually, picornaviruses or “enteroviruses”
Definition were implicated in several of these cases. Even though picorna-
VIRUSES

Members of the family Picornaviridae can occasionally cause viruses can cause this syndrome, they are currently considered
diverse disease syndromes in swine. At least 10 genera have been much less important than other known viral causes of reproduc-
detected in swine. Changes in the nomenclature and classification tive failure (particularly parvovirus) on most farms.
of these viruses can be a source of confusion. Foot-and-mouth
disease (genus Aphthovirus), Senecavirus A (genus Senecavirus), Picornaviruses occasionally contribute to the development of
and encephalomyocarditis (genus Cardiovirus) are discussed lesions or signs associated with other disease agents (including
in separate chapters within this manual. This chapter briefly skin vesicles, myocarditis, pericarditis, pneumonia, and diarrhea),
describes 6 additional genera, including Teschovirus, Sapelovirus, but this appears to be a relatively infrequent event that seldom
Enterovirus (with special mention of swine vesicular disease results in severe losses.
caused by Enterovirus B, formerly Coxsackie virus), Pasivirus, More recently, Sapelovirus has been demonstrated as a sporadic
Parechovirus, Kobuvirus, and an unassigned virus. cause of outbreaks of polioencephalomalacia, usually manifested
Although most infections with these agents are asymptomatic in as paresis and paralysis. It has also been implicated as a rare and
commercial swine production, the diseases associated with several minor cause of fetal wastage.
of these viruses can include polioencephalomyelitis, fetal infec- Many of the picornaviruses, particularly those currently classified
tions, fetal death seen as reproductive disorders, skin lesions or as true enteroviruses appear to be very common in the feces of
vesicles, myocarditis and pericarditis, pneumonia, or diarrhea. healthy pigs and are rarely associated with disease. Additional
enteroviruses not associated or confirmed to cause disease in
Occurrence swine are likely to continue to be detected in swine.
Porcine picornaviruses are widespread, if not ubiquitous, in con-
ventional and commercial swine herds. They occur only in swine, Etiology
and most infections are asymptomatic. Sporadic disease can occur Picornaviruses are spherical, non-enveloped, single-stranded
in susceptible herds, ranging from polioencephalomyelitis (most RNA viruses. Most can be readily grown in cell cultures of
notably teschovirus and sapelovirus) to reproductive disorders porcine origin. The current nomenclature of the most common
(formerly referred to as SMEDI: the acronym for stillbirths, porcine picornaviruses is summarized in the table on page 89.
mummification, embryonic death, infertility) to sporadic out-
breaks of mild transient skin eruptions, myocarditis, pneumonia, Picornaviruses are relatively stable and highly resistant to envi-
or diarrhea. ronmental influences and many disinfectants. Many can maintain
infectivity in processed meats. There is considerable variability in
Historical information their susceptibility to disinfectants, but all can be inactivated by
sodium hypochlorite.
Teschen disease was first reported as a cause of severe polioen-
cephalomalacia in Czechoslovakia over 75 years ago; the disease
has been limited to parts of Europe and Africa. Talfan disease, Epidemiology
described as a milder form of Teschen disease, was reported All picornaviruses are assumed to have similar epidemiology. All
nearly 60 years ago. Both diseases remain well-controlled and ages of swine are susceptible if they have not previously encoun-
geographically confined. Other members of Picornaviridae can tered a particular picornavirus species or strain. The viruses are
also occasionally cause polioencephalomyelitis. commonly and extensively shed in the feces; transmission is usu-
ally fecal-oral. Spread of the virus is usually by direct or indirect
Swine vesicular disease is an important differential for vesicular contact with infected pigs. Neonates infected in utero but born
diseases in swine caused by the species Enterovirus B. This disease alive are sometimes a source of viral exposure of other pigs. Con-
was first reported from Italy in 1966 with limited reports also from taminated fomites can easily be a mechanism to transmit picorna-
other countries in Europe and Asia; reports of disease with this viruses within and between farms as the virus family is relatively
virus are relatively infrequent. The western hemisphere is consid- stable in the environment.
ered free of the virus. Neither swine vesicular disease nor vesicular
exanthema has been reported in the US for many years, but both Less virulent strains of the virus are endemic in most herds, with
remain very important differentials for foot-and-mouth disease. disease expression mitigated by the presence of established herd

88 PORCINE PICORNAVIRUSES
DISEASES CAUSED BY VIRUSES

Table: Current nomenclature of porcine picornaviruses detected in swine.

Genus Species, virus name, and/or disease Skin CNS Fetal Other
Foot-and-mouth disease virus
Aphthovirus + + + Heart
Foot-and-mouth disease

VIRUSES
Cardiovirus A
Cardiovirus + + + Heart
Encephalomyocarditis virus (EMCV)
Cosavirus Cosavirus A No disease confirmed or implicated
Enterovirus B
Swine vesicular disease virus + +
Enterovirus (serotype is Coxsackie)
Enterovirus G
+ +
(formerly porcine enterovirus B)
Aichivirus C
Kobuvirus No disease confirmed or implicated
Porcine kobuvirus 1
Pasivirus Pasivirus A No disease confirmed or implicated
Parechovirus Parechovirus A No disease confirmed or implicated
Sapelovirus A
Sapelovirus + +
(formerly enterovirus A)
Senecavirus A
Senecavirus + Neonatal death
Seneca Valley virus
Teschovirus A
Teschovirus + + Heart, lung
Porcine teschovirus
Unassigned Porcine picornavirus Japan (unassigned) No disease confirmed or implicated
CNS = central nervous system; + = present

immunity. Alterations in production and management, including fetuses. Occasionally, mild skin eruptions, rashes, or glossitis are
segregated rearing, introduction of breeding stock or contam- associated with acute infection with some of the picornaviruses.
inated semen, failure to properly acclimate incoming breeding
Infections of the CNS vary in extent and severity and may affect
stock, commingling, as well as other risk factors may change the
different portions of that system from the olfactory bulbs to the
ecology or serve as a source for the introduction of new viruses.
lumbar cord. Most often, the lesions occur in the region between
the hypothalamus and the medulla. In prolonged cases, there may
Pathogenesis be lymphocytic meningitis, particularly in the cerebellum and
Picornaviruses are ingested and then replicate in the tonsil, intes- brain stem. Spinal cord lesions are largely confined to the gray
tinal tract, and probably in lymphoid tissue in the lamina propria. matter of the ventral horns. There is a gradient in the severity
The milder strains usually remain confined to the intestinal tract, of lesions; the outcome in an infected pig depends on location,
although transient viremia is not unusual. Epithelial cell destruc- severity, and extent of the lesions.
tion is not a feature of most picornaviral infections. With the
more virulent strains, viremia occurs, and the virus may spread to Clinical signs
the central nervous system (CNS) or other sites such as lymphoid
For more virulent disease manifestations such as Teschen, Talfan,
organs, heart, lung, or placenta. When the placenta of the preg-
or when the virus infects a naïve population, all age groups may
nant uterus becomes infected, it commonly leads to infection of
be affected. Morbidity and mortality are expected to be high.
the developing fetuses, which may then lead to sequential death of
Early signs include anorexia, listlessness, and fever that are soon
PORCINE PICORNAVIRUSES 89
SWINE DISEASE MANUAL

followed by ataxia, abnormal postures, and recumbency. There Diagnosis


may be opisthotonus, convulsions, nystagmus, and coma. Death The presence of ataxia and posterior paresis in the absence of
occurs within a few days. Occasional cases survive but may have gross lesions is not sufficiently specific to diagnose picornaviral
residual paralysis and require euthanasia. Talfan disease is less infection. There are many differential diagnoses, including
severe than Teschen disease but presents similarly. nutritional deficiencies, toxicities, and other infectious diseases.
Talfan disease, other strains of teschoviruses, sapelovirus, and Therefore, diagnosis is contingent on the observation of compati-
some enteroviruses can produce ataxia and paresis in infected ble microscopic lesions in the brain and/or spinal cord. Typically,
VIRUSES

pigs. Often the paresis does not progress to paralysis and most the lesions that are present will be suggestive of viral infection but
pigs recover with overall morbidity of 5% to 15% and mortality not pathognomonic for a particular agent.
of less than 3%. However, some outbreaks caused by strains that Detection of virus in aseptically collected neural tissue (brain,
induce CNS signs can have mortality that approaches 10%. spinal cord) or heart by polymerase chain reaction (PCR), in
Reproductive disorders caused by these viruses are usually man- situ hybridization, immunohistochemistry, fluorescent antibody
ifested by increased mummification rates, decreased farrowing staining, or by virus isolation is highly suggestive, if not confirma-
rate, variable litter size, and increased stillbirths; all have been tory, of diagnosis. Many nonpathogenic strains can be routinely
reproduced experimentally by inoculation with various members recovered from enteric tissues of both normal and clinically ill
of the Picornaviridae. Although still infrequent and sporadic, pigs. However, any virus detected within nervous tissue and
the occurrence of these SMEDI-like syndromes not explained accompanied by characteristic pathology in the tissue should be
by infection with porcine parvovirus (which therefore may be considered a significant finding. Neurotropic picornaviral dis-
caused by picornaviruses) appears to be increasing in some high eases must be differentiated from other diseases associated with
health herds. CNS disease.
Unique or new picornaviruses are occasionally detected in cases Paired serum samples taken at least 2 weeks apart may show a
of diarrhea or pneumonia, particularly as part of larger surveys, rising titer in serum neutralization or complement fixation tests
but generally do not appear to have a primary role in disease in but seroconversion to endemic viral agents may be coincidental
the surveyed farms. Efforts should be made to rule out more to disease expression.
likely causes of disease (or lesions) before attributing them to a Confirmation of reproductive disorder associated with members
“new enterovirus” that has been detected. of Picornaviridae can be challenging. Nucleic acid from a viral
infection is sometimes harbored in tissues of mummified or dead
Lesions fetuses such that detection by PCR from aseptically collected
There are no specific gross or microscopic lesions caused only samples can be highly suggestive for diagnosis. Samples should
by this group of picornaviruses. Any clinical presentation that be collected carefully to prevent fecal contamination, a common
involves skin lesions such as vesicles on lips, snout, or coronary cause of false positive PCR test results for picornaviruses.
bands should be aggressively investigated to rule out exotic vesic-
ular diseases. With Teschen, Talfan, and other sporadic picornavi- Control
ral diseases having a similar presentation related to brain or spinal Most herds develop effective immunity to endemic on-farm
cord dysfunction, there will typically be prominent nonsuppura- picornavirus infections. In herds where picornaviruses have his-
tive inflammation in the gray matter of the brain stem, cerebel- torically caused reproductive or other problems, new breeding
lum, and spinal cord (polioencephalomyelitis). In affected areas, stock should be introduced into the herd at least a month before
many neurons will be degenerated or necrotic. In prolonged cases, breeding begins and be exposed to the endemic picornaviruses
there may be lymphocytic meningitis affecting the cerebellum, prior to mating. This will provide sufficient time for recovery
brain stem, or spinal cord. Lesions tend to be more marked and and development of active immunity before pregnancy. It may
extensive with Teschen or Talfan disease than in cases caused by be preferable to expose new stock to a composite sample of feces
less virulent picornaviruses such as sapelovirus. or reproductive materials from the herd to which they are to be
With reproductive disorders, lesions are unusual. Some fetuses added.
occasionally have nonsuppurative myocarditis or encephalitis In Europe, Teschen disease and Talfan disease have been con-
seen on microscopic examination. trolled by slaughter and ring vaccination around farms or areas
Other lesions rarely observed or described include cutaneous ves- where outbreaks have occurred. Both live-attenuated and inac-
icles, nonsuppurative myocarditis, mild interstitial pneumonia, tivated vaccines have been used to control outbreaks in addition
and mild lymphadenitis. These lesions are most likely to be seen to traditional control strategies such as restricting animal move-
in young pigs. Increased edema in the mesentery or subcutis of ments out of infected areas.
neonates and fetuses is also reported yet uncommon.

90 PORCINE PICORNAVIRUSES
DISEASES CAUSED BY VIRUSES

In the US, occasional outbreaks of neurologic disease associated Outbreaks of disease (poliomyelitis, vesicular lesions) of unusual
with picornaviruses occur in nursery and grower stages. Most severity or suspected of being caused by any of the virulent types
of the outbreaks have been transient and limited to individual of picornaviruses should be reported immediately to animal
groups of pigs or production sites, but losses can be significant. As disease control authorities. Thus far, import regulations on swine
with many endemic agents, establishment of good herd immunity and pork products have kept the US free of most highly virulent
is fundamental for long-term control of the agent. However, age porcine picornaviruses including Teschen, Talfan, and foot-and-
segregation, multi-site production, and frequent commingling mouth disease virus.

VIRUSES
can make herd immunity difficult to achieve. Vaccination with
autogenous products has not been consistently effective.

PORCINE PICORNAVIRUSES 91
SWINE DISEASE MANUAL

Porcine reproductive and respiratory syndrome


Alternate names and abbreviations these type 1 and type 2 viruses, but both are able to produce
PRRS, PRRSV, mystery pig disease (archaic) similar clinical signs.
Because of the broad array of viral variants, there is considerable
Definition difficulty grouping PRRS virus (PRRSV) into species, hence it
Porcine reproductive and respiratory syndrome (PRRS) is a viral is often considered to exist in an animal (and in a population of
VIRUSES

disease characterized by two overlapping clinical presentations, animals) as a “quasispecies.” There is considerable heterogeneity
namely reproductive failure in breeding animals and/or respira- in the genome, virulence, and antigenicity of PRRSV because of
tory disease affecting pigs of all ages. Porcine reproductive and inherent errors common in the transcription of RNA viruses.
respiratory syndrome is the most economically significant infec-
tious disease in the US and most areas where it is present. Protective epitopes on the virus surface are not well known nor
can they be predicted from genomic analysis. Although much
is known about the genetic organization of PRRSV, the biolog-
Occurrence ical properties of isolates are highly variable within countries,
Porcine reproductive and respiratory syndrome occurs in all age
geographical areas, and even a single farm. This variation often
groups. Reproductive impairment or failure primarily affects sows
translates to a lack of immunologic cross-protection, posing a
or gilts, but it also can affect boar health and fertility. The respira- continuous challenge to control of the disease.
tory syndrome affects pigs of any age but is more severe in young
growing pigs. Porcine reproductive and respiratory syndrome The virus has a predilection for cells of the immune system, includ-
exists in all major swine-raising countries except Australia, New ing pulmonary intravascular macrophages and pulmonary alveolar
Zealand, Norway, Sweden, and Switzerland. macrophages, and it replicates extensively in the latter. Porcine
reproductive and respiratory syndrome virus is only moderately
Historical information resistant to environmental degradation. The virus is easily inacti-
The clinical disease was first described in 1987 and 1988 in the vated by phenol, formaldehyde, and most common disinfectants.
US, with reports of the disease occurring in multiple states by
1990. During the 1990s, PRRS spread rapidly in both Europe Epidemiology
and North America. By the end of 1992, the disease was reported Pigs (and peccaries) are the only species known to be susceptible
in Canada, Great Britain, and several European countries. Two to PRRSV infection. Pigs shed virus in oral and nasal secretions,
distinct strains of virus, one in Europe and one in the US, were urine, semen, milk, and feces. An important feature of the virus is
isolated and characterized and found to be genetically different its ability to persist for extended periods (greater than 200 days)
but clinically similar in most respects. Both strains are now in the in carrier pigs. Porcine reproductive and respiratory syndrome
US, along with a multitude of viral variants. virus can be shed in boar semen for more than 3 months.
Porcine reproductive and respiratory syndrome was first The infective dose varies by route of exposure. For example, the
described as a syndrome and was confused initially with several oronasal route requires 10,000 virions to cause infection com-
other diseases. It was referred to as swine mystery disease or swine pared to as few as 10 to 20 virions for infection by parenteral
infertility and respiratory syndrome before PRRS became the exposure or skin scarification. Infected dams experience viremia,
generally agreed upon name. Considerable research efforts over which allows transplacental transmission to fetuses, particularly
the past 30 years have revealed much about the virus, the disease, during the last trimester of pregnancy.
and its transmission. However, details related to control of the
Shedding by asymptomatic carrier pigs, contaminated transport
disease for all types of swine-raising operations are far from com-
vehicles, and fomites are probably the most common means of
plete. The consolidation of the swine industry over the past 25
virus introduction to a herd or population of pigs.
years has led to entire production systems being designed around
strategies for preventing, controlling, or eliminating this disease. Semen is a major concern as a route of viral introduction into sow
herds. Infection of sows can occur through natural breeding or
Etiology artificial insemination.
Porcine reproductive and respiratory syndrome virus is an The virus spreads readily by direct contact, and aerosol spread
enveloped RNA virus in the genus Arterivirus in the family between farms is known to occur over several kilometers depend-
Arteriviridae. The prototype US strain (porcine reproductive ing on local geography and weather conditions.
and respiratory syndrome 2, or the “North American” strain) is
related to, but distinct from, the first European isolate (porcine Despite the potential for long-term carriage, field observations
reproductive and respiratory syndrome 1, or the “European” or suggest that most, but not all, infected pigs eventually become
“Lelystad” strain) that was first identified in the Netherlands. immune to homologous virus and cease viral shedding by 60 days
There are significant genetic and antigenic differences between postinfection, although some may remain carriers for more than

92 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME


DISEASES CAUSED BY VIRUSES

200 days. Immune clearance of the virus is accompanied by a Clinical signs


steady decline in antibody titers over a period of 4 to 8 months. Where present, PRRS is probably the most important swine
In some cases, infection of a herd with PRRSV is manifested as an disease to emerge in the last half-century. Serological testing has
epidemic. In other cases, herd infection results in a more insidious revealed there are many infected herds in which significant clinical
increase in respiratory disease or reproductive failure. Although signs are not apparent. Where signs are apparent, they vary and are
debate continues as to the relative importance of vertical, hori- influenced by the virulence of the isolate, whether it is an initial
zontal, and lateral (from a source external to the farm) routes of infection of naïve animals or an on-going herd challenge, the age

VIRUSES
transmission, genetic sequencing of the offending PRRSV can be group affected, the presence of other disease-causing agents present
quite useful in helping to determine the source of infection and in the population, the herd size, and management practices.
the epidemiology of an outbreak. In breeding-age gilts, sows, and boars, clinical signs may include a
Sows infected while pregnant may deliver viremic, persistently period of anorexia, fever, lethargy, depression, and perhaps respi-
infected piglets. The infection can then cycle between young ratory distress or vomiting. Mild cyanosis of the ears, abdomen
pigs well into the nursery phase. Older infected pigs held back and vulva has been reported in some outbreaks. Reproductive
in nurseries or cross-fostered in the farrowing house are often a problems, often the most obvious signs, include a decrease in the
source of virus for younger pigs. number of dams that conceive or a decrease in farrowing rate.
There is usually an increase in premature farrowings, late term
Research suggests that the virus is unlikely to be spread to pigs abortions, stillborn or weak piglets, and mummified fetuses.
by birds or rodents. There are no known replication-competent Pre-weaning mortality is very high. Nursing pigs may have dys-
insect vectors, but mosquitoes and house flies are capable of act- pnea, fever, ill-thrift, diarrhea, or increased susceptibility to other
ing as fomites with the potential to spread the virus mechanically. bacterial infections. Reproductive signs will persist in a breeding
herd for at least 2 to 3 months until a degree of herd immunity
Pathogenesis is established; a slow improvement in reproductive performance
Once transmission of virus to the tonsil or upper respiratory sys- then begins. In larger operations, clinical signs may continue to
tem has occurred, primary replication occurs in lymphoid tissues. cycle, especially if naïve gilts or sows continue to be introduced
Viremia follows and may persist for 3 to 4 weeks ensuring virus into the herd.
access to a variety of organs, including the uterus and placenta of
pregnant sows. The virus has a predilection for lymphoid tissues Suckling piglets in farrowing rooms often propagate virus and
(spleen, thymus, tonsils, lymph nodes, and Peyer’s patches). serve as an important ongoing source of reinfection for the rest
Porcine reproductive and respiratory syndrome virus infects and of the herd, particularly when internal biosecurity measures are
compromises the function of pulmonary alveolar macrophages not practiced. Importantly, herds may be infected simultaneously
and pulmonary intravascular macrophages, resulting in intersti- with multiple, heterologous strains of PRRSV that are not com-
tial pneumonia as a primary component of respiratory disease. pletely cross-protective. This situation substantially decreases the
Besides the direct effects of interstitial pneumonia, the virus com- odds of establishing herd immunity, and clinical signs are likely to
promises pulmonary clearance mechanisms and also has other continue. In boars, clinical signs are similar to those in sows and
systemic effects that include fever, lymphadenitis, and fever. As a are usually accompanied by a transient decrease in semen quality.
consequence, infected pigs have increased susceptibility to other The primary clinical signs in young, grow-finish pigs are fever,
disease agents. Such coinfections often magnify the severity of depression, lethargy, stunting due to systemic disease, and pneu-
both PRRS and the co-infecting agent. monia. Sneezing, fever, and lethargy are followed by expiratory
Porcine reproductive and respiratory syndrome virus is known to dyspnea and stunting. The peak age for respiratory disease is 4 to
cross the placenta with great efficiency, particularly in late gestation 10 weeks, but signs can occur at any age. Postweaning mortality is
(after 70 days), and may reach a high titer in fetuses. Recently, a often markedly increased. Bacterial infections, particularly those
porcine cell receptor (CD163) present on macrophages was found capable of sepsis (eg, Streptococcus suis), are usually increased
to be an important factor, if not a prerequisite, for PRRSV infec- along with the ever-present concurrent or secondary bacterial and
tion. It is thought that the number of permissive cells containing viral infections.
the CD163 receptor is greater in older fetuses. Consequently, the
probability that transplacental infection will kill all or some of the Lesions
fetuses in later gestation is greater than when infection occurs prior Porcine reproductive and respiratory syndrome virus infection
to 70 days. The common outcome of infection of pregnant dams generally results in mild to severe lesions in lungs and lymph
is late-term abortion, the result of either the effects of acute disease nodes. The interstitial pneumonia varies from multifocal to lob-
and fever in sows, or the effects of infection and death of fetuses. ular to diffuse in distribution. Lungs appear mottled and tan, but
Recovered sows are resistant to reinfection when exposed to a lesions are highly variable in extent. Lymph nodes are generally
homologous strain of the virus but are likely to show clinical signs swollen, tan, and edematous or cystic. Microscopic lesions may
again if a heterologous strain is encountered. include nonsuppurative interstitial pneumonia, mild nonsuppu-
PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME 93
SWINE DISEASE MANUAL

rative encephalitis, myocarditis, rhinitis, and perhaps depletion of useful for diagnosis in this situation as there is a reasonable chance
germinal centers of lymph nodes. that one or more of the fetuses will contain detectable levels of
Most fetuses and stillborn pigs with uncomplicated PRRS have virus. In general, submission of serum from acutely affected dams
no discernible lesions but some may have umbilical cord arteritis and at least 3 fetuses from each of at least 3 aborted litters should
and hemorrhage, patchy distribution of slightly firm lungs (inter- be sufficient to confirm a role for PRRSV in reproductive failure.
stitial pneumonia), enlargement of lymph nodes, hemorrhages Other suitable samples to confirm a PRRS outbreak are tissues
in the skin, edema of various tissues, and dehydration with (bronchoalveolar lavage, serum, lung, lymph nodes, tonsil, and
VIRUSES

prominence of the vertebral column. Sows with acute PRRS have spleen) from weak neonates and clinically affected nursing piglets.
typical lung and systemic lesions. Endometritis, myometritis, and Techniques for differentiating viral isolates are available and
placental lesions have been reported but are generally mild. include restriction enzymes and RNA fragment analysis, and
genetic sequencing of some or all of the viral genome. Both
Diagnosis techniques can deliver epidemiologically useful information, but
Clinical signs and history are often enough to suggest a diag- neither is well suited for predicting virulence, antigenicity, or
nosis of PRRS, especially in acute outbreaks. Characteristic the likelihood of cross-protective epitopes amongst virus strains.
microscopic lesions in lungs and several other tissues also are Sequencing is helpful in documenting the various strains and/
suggestive but not pathognomonic. Any presumptive clinical or the changes that occur in the circulating virus population in
diagnosis should be confirmed by detection of the virus. Histo- endemically infected herds, with data often presented in the form
logic examination is very useful to rule out roles for other agents of a dendrogram or phylogenetic tree.
and confirm that any PRRSV that is detected is associated with
compatible lesions. A number of antigen tests including culture, Serology may be helpful in confirming the presence and perhaps
polymerase chain reaction (PCR), immunohistochemistry, and how recently PRRSV infection occurred in a non-vaccinated
in situ hybridization are widely available for detection of virus in herd. A cross-sectional sample of blood from various ages of pigs
most tissues. Serological tests are also widely available but only on a farm is useful for determining the kinetics of virus circu-
provide indirect evidence of infection; additional information is lation and therefore helps to inform control strategies. Several
required to determine if PRRSV is truly the cause of the clinical different tests for PRRS virus antibody are available, including
disease being investigated. immunofluorescence antibody, serum neutralization, and
enzyme-linked immunosorbent assay (most common); some have
To diagnose an involvement of PRRSV in respiratory disease of the ability to discriminate antibodies from European and North
postnatal pigs, growing pigs, or adults, samples should be collected American strains of virus. Maternally derived antibody may be
from acutely affected pigs with typical clinical signs. Detection of detectable up to 6 to 8 weeks of age.
the virus in serum by PCR would be highly suggestive for active
infection, but its role in disease cannot be fully elucidated and so it Control
is important to perform a complete diagnostic examination to rule Herds that are negative for PRRSV should make every effort
out other primary agents of pneumonia. The presence of PRRS to avoid becoming infected. Biosecurity protocols surrounding
viral antigen in pneumonic lesions is significant. animal introductions, transportation processes, truck disinfec-
Porcine reproductive and respiratory syndrome virus can cause tion, and staff movements need to be properly executed every
reproductive failure in two general ways. First, acute disease in day. Because airborne transmission can occur (particularly in
dams causing fever and anorexia can induce acute abortion of pig-dense areas), air is sometimes filtered to prevent airborne
fresh, non-mummified fetuses. In this case, dams will be viremic introduction of the virus. Expenses associated with extreme
at the time of abortion and the virus will be readily detectable in biosecurity measures, such as air filtering, farmer-owned truck
the dam’s serum, but the virus is not likely to be in their aborted washes, and staff biosecurity procedures, can be substantial, but
fetuses since transplacental infection will not yet have occurred. the economic impact of PRRS often warrants these measures.
Second, dams may be subacutely infected several days to weeks Should an outbreak occur, a control program should be tailored
previously but not abort their litter (or farrow early). Over time, to fit the individual farm situation.
the virus in these cases will cross the placenta and infect fetuses, Due to differences in virus strain biology, biosecurity constraints
which will result in their eventual death. Abortion may not occur for individual farms, concurrent disease challenges, incomplete
until a sufficient number of fetuses have become infected and die, knowledge of virus transmission and epidemiology, and diffi-
which typically does not happen until later in gestation (100 to culties establishing cross-protective immunity, there is no single
110 days of gestation). Weak pigs, stillbirths, and occasionally successful strategy for control of the disease
increased mummies may be present in litters that are delivered
at term. Sows in this situation are generally not sick at the time An accurate diagnosis is essential to confirm the presence of the dis-
of abortion and are usually not viremic but will usually have high ease and understand the epidemiology of the disease on a particular
antibody levels in serum. Aborted, mummified, or stillborn pigs are farm. This usually requires disease characterization (demonstration

94 PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME


DISEASES CAUSED BY VIRUSES

of the disease agents and lesions) at each stage of production or There are killed and avirulent live commercial vaccines as well as
site within the flow using a combination of serological testing and autogenous killed vaccines available for control of PRRS. Killed
virus identification and characterization. Combinations of testing vaccines are generally considered less efficacious than avirulent
strategies on statistically significant numbers of serum, oral fluid, or live vaccines; all suffer from having only a moderate ability to
environmental samples can determine status in post-weaned pigs. provide protection across the entire spectrum of genetically
Initial assessments should indicate the stage (acute or chronic) of diverse isolates.
infection in the herd as well as where and how the virus is being
Herds that have successfully stabilized will produce offspring, the

VIRUSES
spread within the herd.
vast majority of which will be virus free. Once reaching this stage,
Once the herd epidemiology has been characterized, the first it is necessary to evaluate the nursery, grower, and finisher phases
steps in development of a PRRS control program usually occur of production to determine what measures (depopulating, clean-
at the sow farm, with a strategy developed to achieve one of two ing, disinfecting, vaccinating, etc) are necessary to maximize the
objectives: 1) elimination of the virus from the breeding herd, chance for the weaned pigs to remain virus free for the duration
or 2) long-term management of the virus. For either objective, of their grow-out.
the initial goal is to manage the breeding herd in such a way that
Some producers with sow herds that consistently produce negative
batches of weaned pigs that are free of the virus are consistently
offspring may attempt to eliminate PRRSV from the sow herd. At
being weaned. Breeding herds can be considered as:
this point, all subsequent replacement seed stock should be naïve for
• Naïve (never infected) PRRSV infection and any residual infected sows in the herd should
• Actively infected (virus is detectable in sows or weaner pigs) be eliminated. The latter is accomplished by testing (serology and/or
• Previously infected but currently stable (herd is antibody PCR) and removal, or in some cases by normal attrition.
positive, but no virus is detected in the sows or weaners by
In some operations, it may be economically feasible to depopulate
routine testing)
the site, clean and disinfect the facilities, and after a few weeks
Although the naïve status is preferred, the reality is that, long-
repopulate with stock free of PRRSV and other major diseases.
term, most herds will need to be brought to and managed in the
Another means to stabilize and eliminate the virus is to close the
stable status.
herd to all new animal introductions for at least 200 days. Herd
The status of sow herds can be monitored in part by routine testing closure requires that all weaned pigs be moved off-site and that
of weaners. Naïve farms can be monitored by routine testing for anti- no internal replacement animals are allowed to be kept on-site.
body. Asymptomatic but positive sow farms are usually monitored by Most breeding stock companies today are able to provide PRRS-
PCR testing using pooled samples of serum or oral fluids at weaning free seed stock, thereby eliminating which was once a major
and/or tissue fluids collected at the time of piglet processing. limitation to producers’ control strategies.
Breeding herd stability is achieved by establishing a high level of Before embarking on a PRRS control or elimination strategy,
herd immunity, which dramatically decreases the likelihood of one should honestly assess risk factors for reinfection of the herd
transmission of virus from dams to fetuses or neonates. When as well as the level of biosecurity that can be maintained by the
coupled with segregated rearing of offspring in this situation, producer. Herds located in swine-dense areas are at great risk for
the clinical effects of infection can be minimized or eliminated. reinfection.
Breeding herd stabilization can be accomplished by vaccination,
There is no specific treatment for PRRSV infection. Broad-spec-
by intentional whole-herd infection with the resident live virus,
trum antibiotics may be useful in controlling secondary bacterial
by aggressive acclimatization of replacement breeding stock, by
infections. Anti-inflammatory products are commonly adminis-
closing the herd to additions of animals for 6 to 10 months, or by
tered during acute disease to mitigate fever and encourage appe-
a combination of these strategies.
tite. Other helpful techniques may include early weaning and
Consideration needs to be given on how one will limit the fre- isolation of piglets and various PRRS vaccination protocols.
quency of seed stock introductions to the sow herd, and to ensure
The challenge of achieving consistently effective immunologic
that the replacement gilts will be well acclimatized to the virus cur-
control of PRRSV has proven insurmountable. Recent reports that
rently resident in the sow herd prior to their entry and first mating.
pigs may be bred for genetic resistance to PRRSV infection are
Replacement gilts can be exposed to virus either by vaccination or
encouraging. Pigs lacking CD163 receptor on their cells are resis-
through exposure to field virus; in either case they must be given
tant to many strains of PRRSV infection. Commercial application
at least 60 days to recover and stop shedding virus before they are
of this finding offers some optimism for the future control of this
introduced into the sow herd. If boars are to be introduced, they
devastating disease. In the meantime, since PRRSV strains vary,
should be handled in a manner similar to the replacement gilts. If
control strategies will also vary to accommodate needs specific to
using artificial insemination, semen should be sourced only from
the farm. It is imperative that experienced practitioners, diagnos-
PRRS-negative studs that have comprehensive PRRSV monitoring
ticians, and research workers continue to objectively expand their
programs in place.
knowledge to better control and eventually eliminate this disease.

PORCINE REPRODUCTIVE AND RESPIRATORY SYNDROME 95


SWINE DISEASE MANUAL

Porcine respiratory coronavirus


Alternate names and abbreviations and additional smaller deletions in ORF 3; the deletion in the
PRCV S gene is believed to play a role in the change of tropism from
an enteric infection with TGE virus to a pneumonic infection
Definition with PRCV.
A viral disease of pigs that can cause a mild pneumonia in sus-
VIRUSES

ceptible pigs but often occurs without producing any clinical Epidemiology
signs. Porcine respiratory coronavirus (PRCV) is closely related Porcine respiratory coronavirus is spread primarily by short-dis-
to transmissible gastroenteritis (TGE) virus, and immunity gen- tance aerosols and direct contact between pigs. Most commercial
erated as a result of infection with PRCV can provide substantial herds are endemically infected with the virus, and young pigs are
cross-protection against TGE virus. assumed to be infected by their dams or horizontally from other
pigs between 4 and 10 weeks of age when maternally derived
Porcine respiratory coronavirus is not zoonotic and infects only antibody-mediated protection declines.
swine.
Porcine respiratory coronavirus is generally a subclinical infec-
Occurrence tion, although mild respiratory signs may occur in some herds;
Porcine respiratory coronavirus was first identified in Europe in severe disease has been described in experimentally infected pigs,
1986 and spread widely across the rest of the continent in the but this seems to be a very rare occurrence in field settings. In
space of a few years. In 1990, the same virus appeared on a farm either situation, mortality is rare though morbidity can often
in the US, with infections subsequently reported across the rest approach 100%.
of the country and Canada. Parts of Asia also began reporting the
appearance PRCV at about the same time. Pathogenesis
Despite its genetic similarity to TGE virus, infection with PRCV
There is no mandatory international reporting of PRCV outbreaks does not cause any pathology to the enteric system.
to the World Organization for Animal Health, so little has been
documented about the presence of the virus in specific countries. The virus replicates in the upper and lower respiratory tract (alveo-
However, the virus is assumed to be endemic in much of the world, lar cells, nasal mucosa, tracheal, bronchial, and bronchiolar epithe-
with the possible exceptions of Australia and New Zealand. lium, alveolar macrophages, and tonsils). Limited replication of the
virus in some cells of the intestine has been shown in experimen-
Historical information tally infected animals, but this finding is probably not significant
In Europe during the early and mid-1980s, veterinarians and in disease development, although it could perhaps be related to the
researchers began to report an unexplained decline in the occur- eventual cross-protection afforded against TGE virus.
rence of clinical outbreaks of both TGE and porcine epidemic Viremia may occur as early as 2 days postinfection and last for 10
diarrhea (PED). During this period in Britain, a large number of to 14 days.
herds appeared to be seroconverting to TGE virus in the absence
of any of the normally severe enteric signs typically associated Clinical signs
with that disease. In 1986, investigation of one of these herds Infection with the virus does not generally result in any signifi-
led to the identification of a viral agent that cross-reacted sero- cant clinical signs, although in naïve pigs or in some experimen-
logically with TGE virus and was designated as TLM83. During tal work, dyspnea, tachypnea, sneezing, coughing, fever, and
the late 1980s in the US, a similar phenomenon was recognized; anorexia can be seen. Severe disease or death is rare. Clinical signs
eventually a virus identical to TLM83 was identified and came to occur between around 4 and 10 days postinfection.
be known as porcine respiratory coronavirus.
Lesions
Etiology Gross lesions of infection with PRCV are not typically seen
Porcine respiratory coronavirus is a member of the Coronaviridae because the subclinical nature of the disease means that few pigs
family of enveloped, positive-sense, single-stranded RNA viruses. are actually examined by postmortem. However, experimentally,
The virus is in the sub-family Coronavirinae, genus Alphacorona- lesions including multifocal to coalescing areas of consolidation
virus along with other porcine viruses including TGE virus and and interlobular pulmonary edema may be present.
PED virus.
Microscopic lesions are similar to other viral pneumonias though
Porcine respiratory coronavirus is believed to have arisen as a less severe: thickening of interstitial septa, lymphoplasmacytic
mutant of TGE virus and is antigenically related to that virus. and histiocytic bronchiolar and alveolar exudate, hypertrophy
Compared to TGE virus, PRCV has a large deletion in the S gene

96 PORCINE RESPIRATORY CORONAVIRUS


DISEASES CAUSED BY VIRUSES

and proliferation of type II pneumocytes, airway epithelial necro- An enzyme-linked immunosorbent assay can discriminate anti-
sis, squamous metaplasia, dysplasia, and proliferation in all sizes bodies produced by infection with TGE virus from those pro-
of airways. Infections complicated by secondary bacterial patho- duced by infection with PRCV. The test is not sufficiently specific
gens can result in bronchopneumonia. enough to use on an individual-pig basis but can be used as part
of a herd-based monitoring program.
Diagnosis Other serological tests have been described for PRCV, but the
Porcine respiratory coronavirus is easily isolated from nasal user is cautioned of the need to investigate the extent to which

VIRUSES
swabs or lung tissue following inoculation onto appropriate cell each might cross-react against TGE virus. If not specifically
culture, with confirmation by virus neutralization or immuno- stated, one can assume significant cross-reaction will occur.
fluorescence. More commonly, polymerase chain reaction is used
to identify this virus from lung tissue. In situ hybridization and
immunohistochemistry tests are also available to identify the
Control
In most situations, little is done to prevent or control infection
virus in tissues.
with PRCV, since the protection that is afforded against TGE sub-
stantially outweighs the cost of disease primarily caused by PRCV.

PORCINE RESPIRATORY CORONAVIRUS 97


SWINE DISEASE MANUAL

Pseudorabies
Alternate names and abbreviations and family Herpesviridae (subfamily Alphaherpesvirinae).
Aujeszky’s disease, AD, ADV, PRV, SuHV-1, mad itch Viral variants that emerged in China in 2011 have significant
genetic differences from other pseudorabies strains that circulate
Definition worldwide. In addition, different pseudorabies strains are
Pseudorabies is a highly contagious and significant disease of pigs. maintained in feral swine, although the clinical disease caused
VIRUSES

This viral infection tends to cause central nervous system (CNS) by these strains is identical to that caused by strains found in
signs in young animals, respiratory illness in older pigs, and repro- domesticated pigs.
ductive losses in sows. Pseudorabies can result in trade restrictions Like many other herpesviruses, pseudorabies virus can persist
and economic losses in countries where it is endemic and remains in a latent state in recovered animals (usually in the trigeminal
a significant problem among domesticated pigs in countries that ganglia). The virus does not survive well outside the host, with
have not eradicated it. survival depending largely on environmental factors. Except
during cold weather, the virus probably does not survive more
Occurrence than 2 weeks outside the pig. Under exceptional conditions it
Among domestic animals, swine are the only natural host of may survive longer in infected fetuses, dried tissue, and buildings.
pseudorabies virus. All age groups not previously exposed or vac- The virus can be destroyed by many disinfectants, including
cinated are susceptible. The disease was eradicated from the US ortho-phenylphenol, quaternary ammonium, iodine compounds,
commercial pig industry in 2004 but remains in some feral swine and 5% sodium hydroxide.
populations. The disease remains common in many other major
swine-raising countries. Most domestic animals (cattle, sheep, Genetic engineering has been used to create highly effective vac-
dogs, cats, and goats, but not horses) and many wild animals (rats, cines by deleting one or more of the surface glycoproteins of the
mice, raccoons, opossums, rabbits, and several other fur-bearing virus. Antibodies to these gene-deleted vaccines differ sufficiently
mammals) are susceptible to the virus, but transmission occurs from antibodies generated by exposure to wild virus such that
only when these species are kept in close contact with acutely serological tests can differentiate infected from vaccinated ani-
infected swine; death is the usual outcome in these aberrant mals (DIVA). Protection induced by these pseudorabies vaccines
hosts. Pseudorabies occurs with some frequency in both cattle appears to be effective against all wild types of the virus.
and sheep, especially those in close contact with swine. The dis-
ease cannot be transmitted to humans. Epidemiology
Pseudorabies virus is spread and persists by several mechanisms.
Historical information Swine that recover from pseudorabies excrete large amounts of
virus in saliva and nasal secretions, and perhaps in urine and feces,
The disease is named after the Hungarian veterinarian Dr Aladar
for up to 2 weeks. Virus can persist in the tonsils of carrier swine
Aujeszky who linked the disease in cattle, dogs, and cats in 1902.
for at least several weeks. Latent virus can persist in the CNS
Pseudorabies was not identified as a viral disease in swine until 1909.
for many months. Recrudescence of the virus often occurs after
Prior to 1960, the disease in swine was important in Eastern stress. Inapparent shedders are frequently the means of introduc-
Europe but major outbreaks did not occur in the US until the tion of the virus into susceptible herds. Once introduced, the
mid-1970s. Incidence of the disease increased with the widespread virus spreads by nose-to-nose contact, through feed and water
introduction of purchased breeding stock, total confinement, and contaminated by oral secretions, and by aerosols coughed into the
continuous farrowing during this period. Active pseudorabies erad- air. The virus can be shed in the semen of acutely infected boars,
ication programs exist in many countries and have been successful, resulting in infection by venereal spread or artificial insemination.
in large part due to efficacious vaccination programs. The virus can be transmitted transplacentally and cause fetal
In 1989, the US embarked on a 5-stage federal/state/industry deaths, abortions, and viremic pigs at birth. In a farrowing house,
program for eradication of pseudorabies in swine. Eradication of aborted fetuses or live piglets born infected with or incubating
the disease soon contaminate the house. The virus may survive
pseudorabies from the commercial industry was achieved in 2004.
long enough to infect piglets of the next farrowing.
However, feral pigs remain infected in some parts of the country.
Feral swine remain a possible source of infection to domestic
Etiology swine. Other pseudorabies-infected animals (eg, rats, mice, dogs,
Pseudorabies is caused by infection with suid herpesvirus 1 cats, raccoons, and opossums) are considered dead-end hosts and
(SuHV-1); the virus is commonly known as Aujeszky’s disease will shed pseudorabies for only a short time. They may, however,
virus in most of the world and as pseudorabies virus in North visit swine facilities and either shed virus that contaminates the
America. This virus is a member of the genus Varicellovirus facility, or die and be eaten by swine. The possible role of flies or
birds in transmission is dubious.

98 PSEUDORABIES
DISEASES CAUSED BY VIRUSES

Experimental and field evidence from outbreaks in the US and In susceptible, grow-finish swine (10 weeks of age to market
Europe suggest that aerosolized virus can travel several kilometers weight), respiratory signs predominate, and morbidity is high.
between farms under ideal conditions. Signs include a febrile response, depression, anorexia, sneezing,
coughing, and nasal discharge. Central nervous system signs
The incubation period in pigs is usually 2 to 6 days.
occur in occasional pigs and vary in severity from tremors to con-
vulsions. Most pigs recover in about 7 to 10 days.
Pathogenesis
In breeding herds, clinical signs may not be apparent except

VIRUSES
Pathogenesis is variable, depending on virulence of the viral strain,
age of the pig, and the route and severity of exposure. Experimen- during the period of initial infection. These signs are primarily
tally, pigs can be infected by many routes of inoculation. Under respiratory, and most animals will recover. In pregnant sows or
field conditions the oral-nasal route of exposure is probably most gilts, reproductive failure sometimes occurs. Dams infected in the
common. Virus often localizes in the epithelium of the tonsils. first trimester may absorb their fetuses and return to estrus. Those
The primary site of replication is epithelium of tonsils, nasophar- in the second or third trimester may abort or have mummified,
ynx, and upper respiratory tract. With highly virulent strains of stillborn, or weak-born pigs. Those infected close to term may
virus there may be a short period of viremia with dissemination to deliver infected piglets that die within a few days.
many tissues. Most viral strains have a tropism for the CNS and
upper respiratory tract. Lesions
Neonates that die acutely after infection may have no gross
The virus can also spread along nerves from a primary site of
lesions. In piglets showing CNS signs, congestion of the menin-
infection to the CNS, then to the spinal cord, where it spreads
ges is often visible. In those showing respiratory signs, there may
centrifugally along nerve trunks. The virus often localizes in gan-
be a rhinitis with patchy epithelial necrosis. The rhinitis can be
glia (eg, the trigeminal nerve ganglia and paravertebral ganglia)
seen after splitting the skull and nasal bones midsagittally. The
and may become latent there. It may be reactivated later by stress.
inflammation and necrosis often extend into the fauces and
In many areas of localization, including respiratory epithelium, larynx, and occasionally into the trachea or esophagus. Other
the virus causes inflammation and necrosis of tissue. In some gross lesions may include keratoconjunctivitis and necrosis of the
organs and tissues, necrotic areas are often large enough to be tonsils. Gross and/or microscopic white foci of necrosis may be
grossly visible. Virus may cross the placenta to infect some or all seen in the liver, spleen, lungs, adrenals, and lymph nodes. Similar
fetuses. In the reproductive tract, the virus causes endometritis, foci are sometimes present in aborted fetuses.
vaginitis, and necrotic placentitis. Lesions and virus may be
Gross lesions are often subtle, absent, or difficult to find in
present in infected fetuses. Intranuclear inclusion bodies are pro-
postweaning pigs. Some pigs will have serous or fibrinonecrotic
duced in a variety of cells in swine of all ages, including fetuses.
rhinitis, pharyngitis, and tracheitis visible at necropsy. Pulmonary
edema, congestion, or consolidation is sometimes present, and
Clinical signs secondary bacterial pneumonia can result in more obvious gross
Signs vary, depending largely on the immune status of the dam
lesions. The lymph nodes may be congested and contain small
and the age of the pigs affected. In general, younger pigs are more
hemorrhages. Affected pigs may also have necrotic tonsillitis or
severely affected and become more resistant as they age. Neonatal
pharyngitis which is almost pathognomonic for the disease.
pigs born to naïve dams are highly susceptible to infection; piglets
become ill 2 to 4 days after exposure. Very young neonates may die Microscopic lesions often include a panencephalitis, ganglioneu-
without signs having even been noticed. Nursing pigs usually show ritis at multiple sites, and perhaps nonsuppurative meningitis. The
signs of neurologic involvement. Signs may include high fever, most severe lesions in the brain are usually in the cerebral cortex
depression, anorexia, tremors, incoordination, dog-sitting position, and often include neuronal degeneration and necrosis. In the
vomiting, foaming at the mouth, blindness, paddling, coma, and lungs there may be bronchitis, bronchiolitis, and alveolitis with
convulsions. Death often occurs within 1 to 3 days. Morbidity hemorrhagic foci of necrosis, but lung lesions are generally neither
and mortality may approach 100% in neonates, especially in naïve severe nor specific. Intranuclear inclusion bodies may be found at
herds. Piglets nursing recovered or vaccinated dams are well pro- many sites, often within neurons, astrocytes, oligodendroglia, and
tected from clinical signs and infection with the virus. respiratory epithelium.
Weaned pigs (3 to 9 weeks of age) have similar signs but mor- In dams that have infected fetuses or abort, the uterine wall may
tality is usually much lower. Respiratory signs often occur in be thickened by inflammatory edema. Necrotic placentitis may be
the older pigs of this age group. They include sneezing, nasal apparent in the placenta of infected fetuses and neonatal piglets.
discharge, coughing, and labored breathing. Most of the pigs
survive, provided they do not succumb to secondary infections,
especially bacterial pneumonia.

PSEUDORABIES 99
SWINE DISEASE MANUAL

Diagnosis farms can occur. Occasionally, acute central nervous disease,


Diagnosis can often be made based on knowledge of endemic fever, and rapid death in hunting dogs or animals consuming feral
circulation of pseudorabies virus in the area combined with history, pig offal will be the first indication of pseudorabies in pigs.
clinical signs, gross and microscopic lesions, and serological testing.
Control
White necrotic foci (1 to 2 mm in diameter) in the liver, tonsil,
Pseudorabies is currently considered an exotic viral disease in the
and spleen of suckling piglets is highly suggestive but should be
US since its eradication in 2004. In countries where the disease
VIRUSES

confirmed as pseudorabies by appropriate testing. Tests applicable


remains endemic, it may be possible to prevent exposure of a neg-
for all ages include fluorescent antibody testing on tissue sections,
ative herd to pseudorabies through strict testing, quarantine, and
immunohistochemistry or in situ hybridization on formalin-fixed
isolation procedures. Only seronegative animals should be added
tissues, and polymerase chain reaction or virus isolation per-
as necessary to a herd. Additions should be made only after a quar-
formed on brain, tonsils, and spleen, along with histopathology
antine period of at least one month and negative serological tests
to confirm typical microscopic lesions.
before and after quarantine. Cats, dogs, rats, mice, and all wildlife
Available serological tests include serum neutralization, latex should be prevented from coming into contact with swine.
agglutination, and enzyme-linked immunosorbent assay. Serum
Pseudorabies DIVA vaccines are essential in the control and erad-
neutralization is the standard regulatory test in the US, but the
ication of the disease. These highly efficacious vaccines increase
other test platforms are widely used as screening tests. Serological
resistance to infection, virtually eliminate clinical signs associated
testing is widely used for herd diagnosis. Accurate serological
with infection, and minimize the amount of viral shedding that
tests that can differentiate vaccine antibody from wild type (ie,
ensues if infection does occur. The incidence of latency and the
DIVA) are widely available and are commonly used as part of
frequency of recrudescence of virus in latently infected swine are
national disease control programs.
also decreased in vaccinated swine. Some sow farms have elimi-
Importantly, the clinical signs associated with acute pseudorabies nated pseudorabies through aggressive vaccination programs that
virus infection in postweaning pigs can be indistinguishable from follow a strict elimination plan that includes test and removal of
other common causes of respiratory disease. Remember that the dams seropositive to wild-type virus.
expanding US feral pig population remains infected with pseu-
dorabies virus; periodic incursions of the virus into commercial

100 PSEUDORABIES
DISEASES CAUSED BY VIRUSES

Rotavirus
Alternate names and abbreviations including Clorox®. Rotavirus infections can occur concurrently
None with or predispose pigs to other diarrheal diseases of young pigs,
particularly colibacillosis or salmonellosis. Combined infections
Definition with other agents cause more severe clinical signs and lead to
Rotaviruses cause diarrhea in nursing and post-weaned pigs and higher mortality.

VIRUSES
affect primarily the small intestine. If uncomplicated, the usual
outcome of infection is high morbidity with low mortality. Pathogenesis
The pathogenesis of rotavirus infections in pigs is similar to
Occurrence that of coronaviruses or other agents that infect enterocytes of
Rotaviruses are ubiquitous in swine populations throughout the small intestine. Transmitted mainly by the fecal-oral route,
the world. Rotavirus infections are common in most avian and rotaviruses reach the small intestine and attach, enter, replicate,
mammal species, including humans. Although all rotaviruses are and destroy enterocytes in segments of the ileum, jejunum, and
related, porcine rotaviruses infect only swine. duodenum. The pathology results in villus fusion, blunting, and
atrophy, all of which contribute to the development of a malab-
Historical information sorptive diarrhea.
In 1969, rotaviruses were identified in calves and subsequently Lesions can be subtle, segmental, or severe. Villus fusion and
detected in other species, including swine and humans. Rotavi- the destruction of villus epithelium result in impaired intestinal
ruses are important, common, and reduce productivity. function, reduced disaccharidase activity, and the accumulation
of osmotically active products in the intestine. The osmotically
Etiology active products promote the movement of water into the intestinal
There are at least 7 antigenically distinct serogroups of rotaviruses lumen. A non-structural protein, NSP4, mediates hypersecretion
(A, B, C, D, E, F, and G) within the family Reoviridae, of which which, along with malabsorption, leads to diarrhea. IgA antibody
4 (A, B, C, and E) are known to infect swine. There are also anti- effectively mitigates severity while multiple factors including
genically dissimilar subgroups within each of the major groups. malnutrition, dehydration, imbalance of electrolytes, and cardiac
Group A rotaviruses have been the most researched, but group B failure can ultimately cause death. However, death is an unusual
and C appear to be becoming more common on farms. Rotavirus outcome. The pathogenesis of all porcine rotavirus types (A, B, C
B and C and are also associated with outbreaks of diarrhea, par- and E) is likely to be similar.
ticularly in suckling piglets. Immunity is generally not cross-pro-
tective between serogroups. Clinical signs
The onset and severity of signs depend on the dose of virus that
Epidemiology has been ingested, the amount of protective antibody in the
Rotaviruses are ubiquitous; all pigs in the US are assumed to dam’s colostrum and milk, and the presence (or absence) on the
eventually become infected with one or more rotaviruses. Neonatal intestinal wall of IgA immunoglobulins secreted within the intes-
and susceptible young growing pigs are exposed to virus shed from tine by active immune processes. Outbreaks on specific premises
carriers, including sows and older pigs, or as a result of coming into often occur repeatedly when the piglets reach an age at which
contact with virus present in their environment. Herd immunity lactogenic immunity is no longer adequate to protect against the
more or less mitigates what would otherwise be a severe viral degree of exposure.
enteritis characterized by profuse diarrhea, similar to that pro-
Diarrhea, usually white to yellow in color, commences and gen-
duced by infection with swine enteric coronaviruses. Infected pigs
erally continues for a few days until the pig develops an active
may develop clinical or subclinical disease, but most pigs recover
immunity. There is usually moderate accompanying dehydration.
uneventfully, with a proportion of pigs becoming carriers. Interspe-
Vomiting occurs but is not a major clinical sign. Morbidity is
cies transmission of rotaviruses has not been documented.
variable, but mortality is usually low when good housing and
A major reason for the ubiquity of rotaviruses is that they are husbandry are present. Severity of clinical signs, morbidity, and
very stable in the environment, assuring their survival in swine mortality will be increased if there is concurrent disease, poor
populations and facilities. Rotaviruses are very resistant to husbandry, exposure to cold, or continuous exposure to high
temperature changes, many chemicals, various pH levels, and doses of virus. Sanitation is paramount in mitigating the dose.
many disinfectants, allowing them to persist in environments
for weeks or months. Recommended disinfectants for cleaned
surfaces include formaldehyde and chlorine-based disinfectants,

ROTAVIRUS 101
SWINE DISEASE MANUAL

Lesions severity of outbreaks. Antibiotics are not effective for rotaviral


Pigs that die or are euthanized are dehydrated and may have infections but may be indicated for concurrent bacterial diseases.
abundant fecal staining of the perineal area. The intestinal wall All-in, all-out pig flow with thorough cleaning and disinfection
will be thin, flaccid, and rather transparent. The small intestine between farrowing groups reduces the exposure of piglets to
and colon contain yellow or gray fluid and debris. Microscopic rotavirus. Limiting the age spread of piglets within a farrowing
lesions are largely confined to villi of the jejunum and ileum, with room minimizes the potential transmission of virus from older to
necrosis of apical enterocytes. This acute change is rapidly fol- younger pigs. If rotavirus is determined to be a consistent cause
VIRUSES

lowed by squamous and cuboidal metaplasia of epithelium, and of diarrhea in post-weaned pigs, sanitation and environmental
villus fusion and blunting, with crypt hyperplasia as enterocytes management of both the farrowing and nursery facilities should
regenerate and lesions resolve. be investigated. Decreasing the dose through good hygiene is a
priority for breaking the cycle of rotavirus disease.
Diagnosis
History, clinical signs, and gross lesions are helpful in establishing Both modified-live virus and killed rotavirus vaccines are avail-
a presumptive diagnosis but need to be confirmed by labora- able, although they are mostly focused on rotavirus type A. Dams
tory testing. Histopathology is quite important to confirm the can be vaccinated at intervals prior to farrowing to stimulate
presence of typical viral lesions, since the simple detection of them to produce higher levels of antibody in their colostrum and
rotaviruses is not unexpected, given that it is a ubiquitous endemic milk. This passive antibody protection should provide time for
agent. Typical lesions are described above but are not specific for their piglets to develop their own immunity prior to complete
rotavirus insult. With compatible lesions, the demonstration of loss of lactogenic antibody protection. Booster vaccinations to
rotavirus in the lesion by immunohistochemistry, fluorescent anti- sows are generally given several weeks before each subsequent
body, polymerase chain reaction (PCR), or electron microscopy is farrowing to maximize the concentration of immunoglobulins in
considered confirmatory. colostrum and milk. Although widely practiced, the efficacy of
various vaccination strategies has not been consistent, and it can
Not all techniques are widely available to differentiate or identify be difficult to prove their benefit on some farms.
the specific rotavirus that is present. A multiplex PCR test for
rotavirus A, B, and C coupled with histopathology is usually Acclimatization of incoming breeding stock is often practiced
sufficient for diagnosis. for rotavirus, as well as a variety of other endemic pathogens in a
herd. This is generally accomplished by intentional exposure of
Virus shedding is much greater and lesions are more definitive new breeding stock to resident stock and/or biofeedback materi-
during the early stages of the disease, therefore samples should als from the herd, prior to their first mating. In this way, the new
be obtained from acutely affected pigs. Demonstrating abundant breeding stock can develop active immunity to indigenous rota-
rotavirus by laboratory testing of feces from acutely affected viruses (and other endemic potential pathogens) that are being
pigs with an antigen-based test suggests a role for rotavirus but shed by older stock on the farm or are present in the materials
does not rule out the possibility of other diseases such as enteric used for biofeedback.
coronavirus disease, colibacillosis, salmonellosis, or coccidiosis.
Because lesions are not pathognomonic and coinfections are Some sow farms attempt to booster sow immunity through
common, it is wise to rule out coinfections with a thorough diag- intentional exposure by feeding feces-laden material from the
nostic investigation. farrowing areas or intestinal contents from acutely affected piglets
back to sows in late gestation. Although commonly practiced, the
Control efficacy of this procedure is not proven.
There are no proven specific therapies for rotavirus infection in
young piglets. Good husbandry and supportive therapy, includ-
ing electrolytes in drinking water, can be of value. A dry, warm
environment and good nutrition are important in reducing the

102 ROTAVIRUS
DISEASES CAUSED BY VIRUSES

Senecavirus A
Alternate names and abbreviations The virus is likely spread by the fecal-oral route or through
Senecavirus, SVA, Seneca Valley virus, SVV breaches in the skin, but vertical transmission is also suspected.
The duration of shedding is not known but thought to be at least
Definition 10 to 14 days; some reports suggest the virus may be detected
Senecavirus A (formerly Seneca Valley virus) can cause vesicular in pigs for at least 28 days. Senecaviruses are susceptible to most

VIRUSES
lesions in all ages of swine. The lesions are indistinguishable from modern disinfectants but because of their potential to be mis-
those of other vesiculating diseases such as foot-and-mouth dis- taken for exotic vesicular diseases, protocols effective for FMD
ease (FMD). Increased mortality in suckling piglets is reported should generally be followed.
during the acute phase of herd outbreaks with Senecavirus A.
Pathogenesis
Occurrence Experimental inoculation of susceptible pigs with Senecavirus A
Swine are considered the natural and only host for Senecavirus will cause formation of vesicles on the snout, lips, coronary bands,
infections. Senecavirus A has been reported throughout the west- and interdigital spaces within 3 to 4 days after infection. Rup-
ern hemisphere, Asia, and Europe and is one of many members of tured vesicles generally heal within a week but may persist longer,
the viral family Picornaviridae that can cause vesicular lesions and especially if complicated with secondary bacterial infections.
reproductive losses in swine. Neonates infected with Senecavirus A may have lethargy, weak-
ness, and often fail to nurse, with diarrhea and death developing
Historical information as a consequence. The morbidity may be high. Outbreaks tend to
Senecavirus was first isolated in the US as a tissue culture contam- resolve quickly, usually running their course in 5 to 10 days. Mor-
inant in a laboratory located in Gaithersburg, Massachusetts near tality in neonates can be quite high during the short, acute phase
Seneca Creek State Park. Although first thought to be nonpatho- of a herd outbreak.
genic in pigs, it is now clear from field experience and experimen-
tal inoculations that some strains of Senecavirus can cause vesicu- Viral shedding in feces and oral or nasal secretions is detectable
lar lesions indistinguishable from those of FMD, swine vesicular for up to 28 days, but viremia is relatively short, starting around
disease, vesicular stomatitis, and vesicular exanthema of swine. 3 days postinfection and resolving within a week. Immunity
appears to be rapid and long lasting, based on clinical observa-
tions and serological antibody assays.
Etiology
The genus Senecavirus and species Senecavirus A are found within
the family Picornaviridae. Senecaviruses are small, spherical, sin- Clinical signs
gle-stranded RNA viruses that have 4 structural proteins forming In acute outbreaks, clinical signs are generally of short duration,
the capsid. This virus was first isolated as a cell culture contami- lasting no more than 2 weeks. Sows are not usually systemically
nant in 2002. ill; they have mild clinical signs, the most common of which are
the presence of vesicular lesions on snouts, lips, coronary bands,
and interdigital spaces. Transient mild lameness can be associated
Epidemiology with vesicular lesions on the feet. Increased mortality in neonates
Senecaviruses have circulated in swine in the US and other parts
is often seen in acute outbreaks, but vesicles are not usually
of the world since at least 1988 with sporadic reports of vesicular
observed on piglets. Outbreaks in this group of pigs generally
disease reported since that time. Recent, increasing numbers of
have a short course (5 to 7 days), but up to 50% of piglets less
outbreaks of the disease have fostered more in-depth investi-
than a week of age can develop diarrhea, weakness, and lethargy,
gations and research on the virus. Genetic analysis suggests all
leading to death.
Senecaviruses are closely related yet have different propensity to
cause lesions. Vesicles in sows may not be noticed until investigation of
decreased piglet livability is undertaken by the farmer; vesicles
Although other species of animals have occasionally been found
can be easily overlooked without careful examination of multiple
to have antibodies against Senecaviruses, pigs are thought to be
individual animals. Along with the typical vesicular lesions, grow-
the only host; no wildlife or avian species appear to act as a reser-
ing pigs may have acute lameness that can affect up to 80% of a
voir for Senecavirus A. Morbidity associated with Senecavirus A
group; signs in this age group tend to be present for the relatively
infections can be extremely variable, but mortality is rare except
short time of 2 to 10 days.
in neonates. In growing pigs and sows, clinical signs may be
absent or exceed 50%. Morbidity in suckling piglets can also be
quite high, with mortality ranging from 5% to 60%.

SENECAVIRUS A 103
SWINE DISEASE MANUAL

Lesions Diagnosis
Grossly visible vesicles approximately 0.2 to 2 cm in diameter History, clinical signs, and the appearance of vesicles on the snout
form rapidly on snouts and coronary bands of growing or adult or feet should prompt an aggressive diagnostic investigation
animals and are apparent within 2 to 3 days of infection. The to rule out FMD and to confirm Senecavirus A as the cause of
vesicles rupture within 24 hours of emergence, form a superficial the lesions. Confirmation is usually made by polymerase chain
ulcer, and then heal over a 1- to 2-week period. In acutely affected reaction performed on vesicle fluids, lesions, or oral fluids. Immu-
neonates, gross lesions are usually absent, although increased mes- nohistochemistry, in situ hybridization, and fluorescent antibody
VIRUSES

enteric and subcutaneous edema has been reported. tests are also available for detection of the agent in lesions. The
virus can also be isolated from vesicular fluids or swabs from
In experimental challenge studies of growing or adult swine,
acute lesions using tissue culture. Serology tests such as enzyme-
characteristic histologic lesions are generally limited to skin,
linked immunosorbent assay can indirectly implicate previous
although nonspecific inflammatory changes can be seen in some
infection but should not be relied on for diagnostic confirmation.
other tissues. In neonates, microscopic changes may include mild
nonsuppurative infiltrates in lung, kidney, brain, or heart. Focal
ulcerative glossitis has also been observed. Control
Most outbreaks of Senecavirus A resolve fairly quickly as a result
of the development of herd immunity. There are no effective
treatments, immunoprophylaxis methods, or vaccines available to
prevent Senecavirus A infection. Prevention is through scrupu-
lous attention to details of external biosecurity, particularly those
involving transportation vehicles used for moving pigs to slaughter.

104 SENECAVIRUS A
DISEASES CAUSED BY VIRUSES

Swine enteric coronavirus diseases


Alternate names and abbreviations The virus family Coronaviridae is in the order Nidovirales. In the
SECD, transmissible gastroenteritis, TGE, TGEV, porcine epi- sub-family Coronavirinae, there are 4 genera: Alphacoronavirus,
demic diarrhea, PED, PEDV, porcine deltacoronavirus, PDCoV Betacoronavirus, Deltacoronavirus, and Gammacoronavirus.
Transmissible gastroenteritis virus, PED virus, and porcine respi-
Definition ratory coronavirus are members of Alphacoronavirus. Betacorona-

VIRUSES
Swine enteric coronavirus diseases (SECD) are a group of dis- virus includes porcine hemagglutinating encephalomyelitis virus
eases in swine caused by infection with one of several members and a number of human pathogens, most notably severe acute
of the Coronaviridae family of viruses including transmissible respiratory syndrome-related coronavirus and Middle Eastern
gastroenteritis (TGE) virus, porcine epidemic diarrhea (PED) respiratory coronavirus. Deltacoronavirus is relatively undescribed
virus, and porcine deltacoronavirus (PDCoV). Swine enteric and includes PDCoV.
coronavirus diseases are characterized by acute, rapidly spreading,
epidemic diarrhea in pigs of all ages. The severity of the disease Epidemiology
is generally related to the age of the pig, with younger pigs being Infected pigs excrete SECD viruses in their feces for 2 to 3 weeks
more severely affected. The World Organization for Animal postinfection with some reports of shedding for up 8 weeks. Typ-
Health lists only TGE as an internationally reportable disease. ically, the viruses can be found in intestinal tissue for much longer
than in feces, with some reports describing recovery of the virus
Occurrence from intestinal tissue for several months.
Enteric coronaviruses of pigs are found in most countries in the Contact with feces of infected pigs has been repeatedly shown to
world that have significant commercial pork industries. Exposure be the most critical risk factor for transmission of SECD viruses.
to these viruses has been detected in some feral pig populations, Direct contact between pigs and indirect contact with the virus
although prevalence appears to be at a low level and there is no through fomites is sufficient to cause infection. The role of con-
indication that feral pigs serve as a reservoir species. Viruses asso- taminated livestock trucks in transmitting PED virus between
ciated with SECD are not zoonotic. farms has recently been investigated. In this study, environmental
samples were collected from the inside floor of 575 livestock trail-
Historical information ers before and after pigs were unloaded into holding pens at 6 US
Transmissible gastroenteritis was first described in 1946 in the commercial slaughter facilities over several days. Before unload-
US, after which time the disease became endemic throughout ing, 6.6% of the trailers were PED virus positive. Of trailers that
the US and other countries. Porcine epidemic diarrhea emerged were negative at the time of arrival to the slaughter facility, 5.2%
in England in the early 1970s and spread to other parts of Asia were contaminated by the time the unloading process was com-
and Europe over the next 30 years. In 2013, an epidemic of PED pleted. Despite biosecurity recommendations to the contrary, all
occurred in the US that was associated with a highly virulent strain truck drivers were observed to have stepped into the slaughter
of the virus circulating in Asia at the same time; the epidemic facility at least once during the unloading process, leading to
eventually spread to include countries in Asia, Central and South high rates of unintentional contact between abattoir staff and the
America, and Europe, where the infection remains in an endemic driver (or trailer).
state. In 2014, an emerging PDCoV first reported in Hong Kong
in 2012 was also discovered in the US. For purposes of regulatory Several studies have implicated large herd size as a risk factor
control during the US PED epidemic, the US Department of Agri- for the occurrence of SECD. These studies suggested that larger
culture began referring to all infections associated with TGE, PED, herds tend to have continuous (weekly) farrowing, have regular
or PDCoV as SECD. The term SECD has now come into common input of replacement gilts, utilize off-site contracted grow-finish
usage in the US but less so in other countries. facilitates, are more likely to commingle postweaning pigs, and
utilize large (greater than 1000 head) grower pig facilities, all of
which increase the likelihood of bringing naïve pigs into contact
Etiology with infected pigs.
Coronaviridae is a family of enveloped, positive-sense, sin-
gle-stranded RNA viruses with a viral genome ranging from The potential for aerosol transmission of TGE and PED virus
approximately 26 to 32 kilobase (kb) pairs in length. Virions over distances of a few meters has been established under exper-
are spherical, 120 to 160 nm across and are typically covered imental conditions. However, the significance of this mode of
with large approximately 20 nm club- or petal-shaped surface transmission in the spread of SECD viruses between farms under
projections referred to as spikes, which in electron micrographs field conditions is considered to be low. Although TGE and PED
of spherical particles create an image reminiscent of the solar viruses have been identified in common house flies (Musca domes-
corona, hence the more familiar name of coronaviruses. tica), flies are not considered to act as a significant biological or
mechanical vector.
SWINE ENTERIC CORONAVIRUS DISEASES 105
SWINE DISEASE MANUAL

Porcine-origin ingredients are routinely used as a part of the animals and the death loss was usually very small …
commercial diets fed to pigs in many parts of the world. Their disease was observed affecting nearly entire herds of
risk as a possible vector for a number of pathogens has been shoats when there were no baby pigs on the farm ...
previously described and will not be reviewed here. However, the
A number of observational or experimental studies describing
potential for these ingredients to be a vector for PED virus was
similar findings have been published since that time and summa-
raised specifically as part of investigations into the introduction
rized by other authors; one should consider that TGE, PED, and
of PED virus into Canada in 2014 and the spread of PED virus
VIRUSES

PDCoV are virtually indistinguishable in clinical presentation


within the US during the same period. Studies since this time
and epidemiology.
have shown that feed contaminated with virus (as a result of the
inclusion of porcine-origin products or as a result of cross-con- When introduced into a susceptible herd, the disease usually
tamination from other sources) has the potential to introduce spreads rapidly with some degree of appetite loss and diarrhea
SECD viruses into farms or countries. in most animals, with vomiting occurring in some animals. The
occurrence of SECD in a herd where most of the pigs are naïve is
Milk from sows recently infected with TGE or PED virus has been
referred to as epidemic or epizootic SECD. In this situation, most
shown to contain infectious virus and is capable of infecting neo-
pigs within a space (barn, room, or pasture) are affected within
natal pigs that consume the milk. However, the significance of this
a 2- to 5-day period. Transmission of the infection to adjoining
finding is questionable, given the multiple sources of virus a nursing
spaces is generally inevitable through movement of clinical or
piglet would be exposed to if its dam was recently infected.
subclinical pigs, contact with contaminated inanimate vectors, or
through short-distance aerosols. The incubation period is 18 to
Pathogenesis 72 hours. In piglets under approximately 2 weeks of age, infection
Infection with an SECD virus typically occurs through oral
leads to vomiting and profuse, nonhemorrhagic diarrhea often
exposure. All parts of the small intestine are capable of replicating
associated with a characteristic foul smell. Virtually 100% of pigs
SECD viruses and their generation time is around 6 to 12 hours.
less than 1 week of age will die within 2 to 7 days of the onset of
After exposure to the virus, clinical signs of diarrhea and vomiting
clinical signs. While morbidity in piglets over 1 week of age will
usually occur within 18 to 24 hours in young pigs, although older
still approach 100%, mortality declines with age at onset, such
animals may not show clinical signs until 2 to 3 days after expo-
that most pigs infected at greater than 3 weeks of age will survive.
sure. Although these viruses have been reported from nonenteric
Older growing pigs and adults will have diarrhea, with vomiting,
tissues on occasion, the viruses replicate exclusively in enterocytes
and inappetence occurring less predictably. A sow acutely affected
of the small intestine. Virus can be shed for several days after
during lactation may produce less milk, contributing to an even
diarrhea has resolved.
more rapid decline in her affected litter.
Once infected, enterocytes rapidly cause severe damage to the
microvilli structure of the small intestine, which results in a Lesions
malabsorptive diarrhea. Younger pigs are more severely affected Gross lesions of SECD virus infection in acutely affected
than older pigs for two reasons: a larger proportion of the small neonatal piglets include dehydration, focal hemorrhages in
intestine tends to be affected in younger pigs, and the regen- the submucosa of the stomach, highly fluid intestinal contents
eration time of new enterocytes is slower in younger pigs, thus having a yellow-green color, thinning of the small intestine wall,
efforts to repair damage to the intestinal mucosa are ineffective in engorged mesenteric blood vessels, and some evidence (paleness)
controlling the clinical disease. of degeneration in the kidney. Frequently, piglets of this age will
present with excessive amounts of curdled milk in the stomach
Clinical signs and an absence of chyle in mesenteric lymphatics. Virus does not
For the purposes of this chapter, clinical signs and pathology of replicate, nor has it been identified, in peritoneal fluid, spleen,
SECD (TGE virus, PED virus, and PDCoV) will be considered kidney, pancreas, thymus, lung, or stomach. The most significant
as a single entity, based on the occurrence of essentially identical lesions are confined to the small intestine and include congestion,
syndromes among the diseases. edema, and necrosis of intestinal villi.
The clinical signs of epidemic outbreaks of SECD at both an Microscopically, lesions can vary based on the age of the pig,
individual and herd level have changed little since the first report stage of infection, and dose of virus. The key pathological feature
of TGE in 1946 when the authors described: is villus atrophy, the speed and severity of which is mostly asso-
ciated with pig age. Virus can commonly be recovered from the
... a disease characterized by diarrhea, vomiting in intestinal mucosa for 1 to 2 weeks postinfection. Desquamation
some cases, rapid loss of weight, and a high death of enterocytes accompanied by villus blunting is apparent by 12
rate in baby pigs…losses limited largely to pigs only to 18 hours postinfection. By 24 hours, sloughed cells are begin-
a few days old … recovery was prompt in older ning to be replaced by enterocytes migrating upwards from the
crypt region.
106 SWINE ENTERIC CORONAVIRUS DISEASES
DISEASES CAUSED BY VIRUSES

Diagnosis is more resistant to infection with the viruses and those entero-
The typical coronavirus morphology of SECD viruses has been cytes that do become infected can be regenerated more quickly.
described by electron microscopy. The virus is pleomorphic and This situation tends to lead to only a transient diarrhea that is
ranges in diameter from 95 to 190 nm, including the length of the self-limiting in 3 to 6 days, with little measurable effect on growth
surface projections which are approximately 15 to 20 nm in length. or mortality. Given this clinical situation, much of the research
emphasis has been on developing methods to generate high-qual-
Isolation of SEDC viruses can be challenging. Most field strains ity active immunity in breeding sows so that neonatal pigs born

VIRUSES
are difficult to grow in cell culture, generally requiring that other into a contaminated environment will be protected.
methods be used to identify the viruses.
The primary means of controlling an acute outbreak of SECD
While serological testing as part of a diagnostic plan can be useful in a breeding herd is through rapid, intentional, and controlled
in some circumstances, cross-reaction between various members exposure of all sows in the herd to live virus; the technique has
of the family has been described for at least 40 years and can been used for many diseases since the early 1950s. Prescriptive
complicate interpretation. Few commercial serological tests are step-by-step instructions for implementing this process of
available for routine diagnostic use, particularly for differentiating “auto-vaccination” or “biofeedback” are not widely available, but
exposure to the various porcine coronaviruses. the objectives of all intentional exposure programs are the same:
A number of techniques have been described for detection of to establish effective sow herd immunity to the virus as quickly as
SECD viruses in tissue samples. The fluorescent antibody tech- possible in order that high-levels of colostral and lactogenic anti-
nique is a simple method and has been an important tool in con- bodies are produced, thus protecting neonatal pigs from infec-
firming infection in field cases or experimental studies for many tion. Done correctly, this process will create an immune breeding
decades. The technique is frequently used as a rapid and accurate herd concurrent with a fully protected population of neonatal
diagnostic test on frozen intestinal tissues from clinical specimens pigs that can then be weaned into cleaned and disinfected facil-
for PED and TGE. False negative reactions can be a common ities. These two outcomes effectively eliminate the potential for
occurrence on acute or sub-acute samples if intestinal epithelial virus to come in contact with susceptible pigs, and eventual elimi-
cells have been severely denuded. Immunohistochemistry and in nation of the virus from the farm will result. Intentional exposure
situ hybridization assays have also been developed. is done by collecting feces and digesta from acutely infected
neonates and then individually exposing each breeding sow to the
Increasingly, polymerase chain reaction assays capable of detec- material via the oral route. Intentional exposure programs will
tion and differentiation of the SECD viruses are being used for typically bring a return to normal farm productivity in approxi-
diagnosis as well as for environmental monitoring. mately 4 to 6 weeks.
The following diseases should be considered in a differential In acute outbreaks of these diseases in isolated herds of growing
diagnosis of SECD based on clinical signs, lesions, or diagnostic pigs greater than 3 weeks of age, the infections are self-limiting
test cross-reaction: colibacillosis, coccidiosis, hemagglutinating and will resolve without intervention beyond supportive care.
encephalomyelitis, porcine rotavirus infection, Clostridium per- Critically, no new animals should be introduced into the popula-
fringens type A or type C infection, swine dysentery, salmonello- tion for at least 60 days after cessation of clinical signs.
sis, ileitis, classical swine fever, and African swine fever.
Vaccines are available for TGE and PED, although not for
Control PDCoV. In general, these vaccines do not generate immunity suf-
The porcine enteric coronaviruses are problematic for the ficient to stop infection from occurring, eliminate clinical signs,
immune system to control because in an acute outbreak, the or limit ongoing transmission. However, some veterinarians have
pigs that are most severely affected (neonates) are faced with a found their use helpful as part of a whole-herd program to eradi-
significant challenge dose of the virus almost immediately after cate the virus from particularly intractable infections.
birth, are born with no pre-existing humoral defense specific to There are no specific effective treatments for pigs affected by an
the virus, have the maximum number of susceptible enterocytes enteric coronavirus, although supportive care may have some
in their small intestine, and need to manage an infection caused minor benefits in neonates.
by a virus having a replication time of only 6 to 8 hours. In effect,
there is not sufficient time available for a neonatal piglet to
generate an active response to the infection before mortality is
likely to occur. Therefore, in this population of pigs, a ready and
ongoing supply of colostral and lactogenic immunoglobulin from
an immune dam is imperative in combating the infection.
In pigs older than 3 to 4 weeks, the pig is inherently less likely to
have severe clinical signs because the population of enterocytes

SWINE ENTERIC CORONAVIRUS DISEASES 107


SWINE DISEASE MANUAL

Swine vesicular disease Pigs shed virus in nasal secretions, oral fluids, feces, urine, and
serum, with shedding possible for up to 2 days prior to the
Alternate names and abbreviations appearance of clinical signs. Most pigs clear the virus within 2 to
SVD, SVDV 3 weeks, although a few may harbor the virus for longer periods
of time.
Definition The virus is quite hardy and survives over a broad pH range of
A viral disease manifested by vesicles on the feet (coronary 2.5 to 12. It survives in environment, manure, slurry, or organic
VIRUSES

bands), snout, mouth, and teats. Clinical course is mild and short substrates for months and has been shown to remain viable in
in duration with the primary concern being the need to differen- processed meats for up to 2 years; this resistance to environ-
tiate it from foot-and-mouth disease (FMD). mental extremes helps to account for the importance of indirect
methods of transmitting the virus between pigs or farms. Swine
Occurrence vesicular disease virus is fairly resistant to disinfectants. Disinfec-
The virus is reported as endemic in Italy, with sporadic reports tion protocols developed for use in foot-and-mouth elimination
of disease outbreaks in other parts of Europe and Asia. Because programs are also used for swine vesicular disease virus.
the disease is mild, it may be subclinical or not noticed. Some
countries employ active serological surveillance to monitor for Pathogenesis
infection in swine populations. Virus enters via the skin, mucus membranes, or digestive tract,
Swine are the only known natural hosts for swine vesicular disease with viremia occurring within 1 to 2 days. The period between
virus. The virus is not considered zoonotic, although there are infection and the appearance of clinical signs can vary from 2 to
rare, historic reports of transient infection of humans working 7 days, with viral shedding occurring before lesions or clinical
with the virus in a laboratory setting; however, none with recent signs are observed. The virus can be found in high titers within
circulating strains. vesicles of the skin, epithelial tissues, and occasionally myocar-
dium or brain.
Historical information
First identified in Italy in 1966, swine vesicular disease has been Clinical signs
reported throughout Europe and Asia. It has been eliminated The predominant clinical signs of infection with swine vesicular
from most swine-rearing regions but is still occasionally reported disease virus are the vesicular lesions that develop on the coronary
from Italy and Portugal with rare sporadic outbreaks in other bands or interdigital spaces of feet, and epithelium of the lips,
parts of Europe. This disease is no longer listed by the World mouth, snout, tongue, and occasionally the teats of lactating
Organization for Animal Health because improved diagnostic sows. Clinical signs can develop within 1 to 2 days and become
testing allows it to be readily detected and differentiated from apparent at about the same time lesions develop. In mild cases of
FMD and the virus does not cause significant economic losses. lameness, the lesions may easily go unnoticed.

Etiology Lesions
Swine vesicular disease virus is a member of the genus Enterovi- Any vesicular lesions of coronary bands, feet, lips, snout, mouth,
rus within the family Picornaviridae. It is closely related to and or teats should be thoroughly investigated, since the lesions of
thought to be derived from a coxsackievirus serotype of human swine vesicular disease are indistinguishable from those of FMD.
enterovirus B. Lesions of swine vesicular disease are most common around cor-
onary bands and the skin of the foot, and less prominent on the
Picornaviruses are non-enveloped, single-stranded RNA viruses snout, lips, or tongue. Lesions start as small vesicles that rupture
that tend to be quite hardy in the environment. All strains of within 24 hours, leaving a red scab. Lesions usually heal within
swine vesicular disease virus are considered as the same serotype. 7 days, with complete recovery occurring over 2 weeks. Histo-
pathology of the skin lesions is typical of other vesicular diseases
Epidemiology and is devoid of any particularly distinguishing features. Occa-
Pigs are the only natural host for swine vesicular disease virus, sionally, affected pigs may have mild nonsuppurative myocarditis
although there is a report of sheep that were held in close contact or encephalitis, but this is usually of no clinical impact.
to affected pigs seroconverting to the virus. The primary method
of transmission between pigs is through direct contact with Diagnosis
affected pigs, exudates, lesions, or feces. Other methods of trans- The investigation of swine vesicular disease often begins with
mission include orally through feeding of uncooked waste food, examination of a recumbent pig suffering from acute lameness.
contact with contaminated fomites, and through transport in Examination of peracutely and acutely affected pigs will identify
contaminated vehicles. areas of blanching on the coronary band that will be followed

108 SWINE VESICULAR DISEASE


DISEASES CAUSED BY VIRUSES

by vesicle development (and perhaps rupture) within 24 hours. Feces and serum can be used for monitoring disease occurrence
During this acute phase, ulcers may be observed in and around at a herd level in endemic regions. Serology is usually done by
the mouth as well. However, confirmation of diagnosis must be ELISA and should be used only for monitoring or surveillance
made by laboratory testing. and not relied upon for diagnosis of acute disease. Because
the goal of control is elimination, serological surveillance is an
Confirmation of acute cases is through demonstration of the
important component of control.
virus in tissues with lesions, lesion scrapings, or vesicular fluids.

VIRUSES
Polymerase chain reaction, antigen-capture enzyme-linked
immunosorbent assay (ELISA), and immuno-stains are available Control
to assist in identifying the presence of the virus in lesions. Virus There is no treatment for this disease. In most instances, the usual
isolation is possible but usually done only to assist in resolving objective after detection of an outbreak is to eradicate the virus by
other equivocal test results. Other viruses, including other strains stamping out positive herds, followed by disinfection.
or members of Picornaviridae, can cause lesions similar to swine Countries and regions that are free of the disease should strive to
vesicular disease. Care is necessary for proper sampling, process- remain so by screening the introduction of new stock, restricting
ing, and testing to ensure an accurate diagnosis. pork products from areas that have infection, and eliminating
waste food feeding.

SWINE VESICULAR DISEASE 109


SWINE DISEASE MANUAL

Swinepox
Alternate names and abbreviations Pathogenesis
Pox, congenital swine pox, congenital pox, SwPV Virus introduced into abrasions replicates in cells of the stratum
spinosum causing typical skin lesions to develop. The lesions go
Definition through the stages of macules, papules, vesicles, pustules, and
A viral disease caused by swinepox virus that is characterized by crusts. All but the vesicle stage are easily identified. As lesions
VIRUSES

the acute appearance of round to oval cutaneous lesions that heal mature and heal, crusts containing virus desquamate and leave
in 3 to 4 weeks. white macules.
A viremic stage of the disease has not been demonstrated. How-
Occurrence ever, it must occur in some animals since transplacental infection
Swinepox occurs only in swine. All age groups are susceptible, but of neonatal piglets has been reported. Antibody to swinepox
most outbreaks are encountered in young growing pigs. It occa- virus is formed as a consequence of infection and immunity is
sionally occurs congenitally and in neonates. Swinepox occurs long-lasting.
worldwide in the major swine-raising countries. It occurs more
often where the insect vectors of swinepox virus are not con- Clinical signs and lesions
trolled. Modern swine raised free of lice rarely exhibit the disease. Systemic clinical signs of disease are seldom observed, but the
skin lesions are easily seen. Initial lesions usually occur along the
Historical information ventral aspect of the abdomen, inside the legs, and in inguinal
Swinepox was first reported in Europe in 1842 and in the US in areas; all of these areas are favorite feeding sites of lice. However,
1929. The disease persisted and was relatively common, perhaps lesions have been observed at many cutaneous sites, so most of
because it caused little obvious economic loss to swine produc- the skin appears to be susceptible. Initially, there are small vesicles
ers. Outbreaks were once caused by 2 viruses: vaccinia virus (a that are largely unnoticed. The papules that result occur as 1 to 3
laboratory strain) and swinepox virus. Vaccinia virus is no longer mm circular red spots. As lesions mature, crusts form that even-
used to vaccinate people against smallpox and hence it no longer tually heal uneventfully. In severe infections, lesions may occur in
spreads to pigs. Only swinepox virus now causes the disease. upper respiratory and digestive tracts. This is particularly true of
congenitally acquired pox where piglets are born with or develop
Etiology lesions over the entire body shortly after birth. Exudative epider-
Swinepox virus, a member of the genus Suipoxvirus within the mitis and secondary bacterial dermatitis occasionally occur as a
Poxviridae family, is the etiologic agent. The virus is rather resis- sequel to swinepox. Swinepox does not appear to cause a pruritic
tant to environmental degradation; it persists in scabs for up to a response in affected pigs.
year. Microscopically, the virus is visible as large intracytoplasmic
inclusions in infected cells. Diagnosis
Swinepox is easily diagnosed by identifying the lesions. Lesions
Epidemiology are round to oval and are usually less than 1 cm in diameter. The
Persistent virus within desquamated scabs can be transmitted brown to black crusts are easily seen, but the vesicle stage can
to other pigs when introduced into skin abrasions. The virus seldom be identified grossly. If confirmation is needed, histologic
can also spread horizontally from nasal and oral secretions from diagnosis can be made by identifying typical large intracytoplas-
infected pigs. Once a few virus-containing lesions appear, the mic inclusion bodies in cutaneous lesions. Pox lesions should
virus can be transmitted mechanically by the bites of vectors that be differentiated from those of other vesiculating viral diseases,
have recently fed in pox lesion areas. Common vectors include pityriasis rosea, dermatosis vegetans, streptococcal dermatitis,
the hog louse (Haematopinus suis), mosquitoes, and biting flies. ringworm, and exudative epidermitis.
Once introduced, infected swine or virus-carrying biting insects
can initiate outbreaks in susceptible herds. There is evidence of Control
transplacental infection of neonatal pigs. Control of swine pox relies on the establishment of herd immu-
nity and prevention of transmission. Swinepox is seldom a
problem if the usual vectors of the virus, especially hog lice, are
controlled by insecticides or elimination. Congenital swinepox is
generally a sporadic, self-limiting event. There are no treatments.

110 SWINEPOX
DISEASES CAUSED BY VIRUSES

Vesicular exanthema
Alternate names and abbreviations to be the same. Caliciviruses have a cup-shaped morphology
VE, VEV, vesicular exanthema of swine virus, VES, VESV, San from which they are named. The calicivirus that causes vesicular
Miguel sea lion disease, SMSL exanthema is sensitive to many common disinfectants, includ-
ing 2% sodium hydroxide.
Definition

VIRUSES
An acute, rare, contagious disease of swine characterized by Epidemiology
vesicles on the feet, snout, mucous membranes of the mouth and The opal-eye fish (Girella nigricans) now is believed to be the
tongue, and non-haired skin. The disease in sea lions causes vesi- primary host of the calicivirus that may be passed to pinnipeds
cles on the flippers. and swine. The method of spread of the virus among sea lions and
other sea mammals is speculative. It could be through ingestion
Occurrence of infected fish or small marine life, by coastal contamination, or
Of the domestic animals, vesicular exanthema occurs only in direct contact.
swine. The disease has never been reported from any country Swine are usually exposed to the vesicular exanthema calicivirus
other than the US. Vesicular exanthema has not recurred in by eating uncooked garbage containing infectious meat scraps
domestic swine since it was eradicated in 1956. from swine or certain fish. Although unproven, some marine-ori-
San Miguel sea lion virus disease occurs in sea lions, fur seals, ele- gin feed supplements for swine may contain calicivirus. Vesicular
phant seals, opal-eye fish, and several other marine animals off the exanthema is quite contagious among swine and spreads by direct
western coast of the US. contact and fomites. Neither long-term carriers nor aerosol
spread have been demonstrated.
Historical information
The disease has only occurred in the US, and the historical Pathogenesis
record is incomplete. The disease was apparently first reported In swine, historical outbreaks of vesicular exanthema were usually
in California around 1932, presenting with lesions that closely related to feeding of uncooked food waste. Virus enters the epi-
resembled those of foot-and-mouth disease. The 1932 outbreak thelium through abrasions and multiplies in the basal layer of the
was linked to feeding of uncooked garbage and fish scraps to pigs. epidermis. Intracellular and intercellular edema and coalescence
During the 1950s, infection spread more generally across the US of disintegrating cells lead to vesicle formation. Virus spreads cell
and the principle route of transmission was thought to be a result to cell as lesions develop. A low-grade viremia occurs and leads to
of feeding untreated contaminated garbage, although direct secondary lesions at other sites. Extensive lymphocyte destruction
transmission from infected pigs was also possible. As a result of occurs in regional lymph nodes. Since much of the basal layer of
an organized and vigorous campaign, vesicular exanthema was the epidermis survives, regeneration of epithelium in swine usually
eradicated in 1959 and declared to be an exotic disease. occurs within 1 to 2 weeks. Lesions in sea lions closely resemble
those seen in swine.
Beginning in 1973, caliciviruses indistinguishable from the virus
that causes vesicular exanthema were isolated from sea lions, Experimentally, it requires much more virus by oral exposure
northern elephant seals, fur seals, and certain kinds of fish. Each to produce vesicular exanthema than is required by intradermal
of the viruses can produce lesions in swine and certain sea mam- injection. This suggests that the virus can be spread more readily
mals. Research continues with these viruses and new serotypes are through abrasions on the skin. Most swine infected with vesic-
being identified. ular exanthema will recover if allowed to do so. However, it is
anticipated that USDA-APHIS would require that infected pigs
Vesicular exanthema of swine virus and some recent serotypes of be destroyed as a precaution to prevent the surreptitious entry of
San Miguel sea lion virus have occasionally been isolated from foot-and-mouth disease.
humans with blisters. However, the virus is not considered to be a
serious public health threat. Diagnosis
Vesicular exanthema of swine virus can be grown in cell culture.
Etiology In addition, electron microscopy and polymerase chain reaction
Vesicular exanthema of swine virus is a non-enveloped RNA assays can be used to assist in identifying the virus in tissue or
virus in the family Caliciviridae. The calicivirus that causes blood. Available serological tests include complement fixation,
vesicular exanthema and San Miguel sea lion disease is believed virus neutralization, and enzyme-linked immunosorbent assay.

VESICULAR EXANTHEMA 111


SWINE DISEASE MANUAL

Vesicular stomatitis
Alternate names and abbreviations of vesicular stomatitis usually coincides with the appearance of
VS, VSV warm weather and ceases with the first frosts. This suggests that
the virus is transmitted by insects, especially to horses and cattle,
Definition which are frequently infected while on pasture. However, insect
A vesicular viral disease capable of infecting cattle, horses, swine, transmission does not appear to be essential, as contact transmis-
VIRUSES

people, and many kinds of wildlife. sion between horses has occasionally been observed.
Vesicular stomatitis virus has been isolated from both biting and
Occurrence nonbiting insects. Insect vectors are thought to introduce vesic-
Vesicular stomatitis is endemic in various regions of North, Cen- ular stomatitis virus into populations of domesticated animals.
tral, and South America. Widespread epidemics usually occur Sand flies, blackflies, and Culicoides midges can act as biolog-
about every 10 years. The virus is endemic in areas of the south- ical vectors; however, studies have not supported their role in
western US, but it has not spread to swine. Most outbreaks of vesic- long-distance transmission between farms or between geographic
ular stomatitis affect horses and cattle, less often swine. Infection regions. Once animals develop lesions, insects may become
also occurs in deer, bobcats, raccoons, feral swine, and rodents. infected by feeding on viruses in the lesions or in contaminated
secretions; these insects then pass the infection to a susceptible
Historical information animal at the next feeding.
Vesicular stomatitis was first recognized in horses during the Civil
Fomites associated with recent outbreaks have been shown to
War. In 1943, infection occurred in swine stemming from the use
play a role in the spread of vesicular stomatitis to swine.
of vesicular stomatitis virus-contaminated hog cholera serum. The
endemic nature of vesicular stomatitis in limited geographic areas
and its epidemic nature in other areas remain unexplained by Pathogenesis
epidemiologists. The close resemblance of vesicular stomatitis to Virus appears to require introduction into epithelium through
foot-and-mouth disease and other exotic vesicular diseases con- insect bites or abrasions of the skin. However, transmission may
tinues to compel interest in the disease by animal health officials. occur through the respiratory tract. After an incubation period
of 3 to 7 days, lesions appear initially as small, blanched papules.
Lesions first develop in cells of the stratum spinosum and vesicles
Etiology form. Epithelial cells above the stratum basale separate in about
Vesicular stomatitis is caused by one of four named viruses in the
30% of the cases. Loss of fluid through the stratum corneum
genus Vesiculovirus of the family Rhabdoviridae: vesicular sto-
results in inspissation (thickening by dehydration) of many
matitis New Jersey virus (VSV-NJ), vesicular stomatitis Indiana
lesions. The devitalized epithelium over vesicles soon erodes to
virus (VSV-IN), vesicular stomatitis Alagoas virus (VSVAV),
leave shallow craters that fill with serum and erythrocytes. Since
and Cocal virus. The VSV-NJ and VSV-IN appear to be most
the basal layer of epidermis usually remains intact, most lesions
common in pigs and other livestock in the US. There is little
soon heal. Gross and microscopic lesions of the vesicular diseases
immunological cross-protection after infection with one of the
of swine are similar to those of other species, and their morphol-
viruses. Vesicular stomatitis virus is not considered to be highly
ogy is not an adequate basis for differential diagnosis.
contagious among domestic animals.

Epidemiology Clinical signs and lesions


Clinical signs are similar in all of the vesicular viral diseases of
Livestock are not able to maintain vesicular stomatitis viruses
most domestic species, but there are a few differences peculiar to
long-term and the reservoir or amplifying hosts are unknown.
species and in the location of most lesions. Infection with vesic-
Deer mice (Peromyscus species) can become viremic. These and
ular stomatitis virus stimulates a sharp rise in temperature that
other small rodents have been proposed as possible reservoir
usually regresses after onset.
hosts; however, the complete epidemiology of vesicular stomatitis
is not fully understood. In swine, vesicles on the feet are common. Lameness is often the
first sign that is noticed. Vesicles that form and coalesce around
Vesicular stomatitis viruses are endemic in southern Mexico,
the coronary band may lead to loss of hooves, foot deformity,
Central America, and northern South America but VSV-NJ and
and persistent lameness. Vesicles frequently occur on the snout
VSV-IN regularly cause localized outbreaks in southern regions
and may extend into the nares. If lesions develop on the lips or
of the US. There may be unknown reservoirs and vectors of the
in the mouth, there may be excessive salivation, slobbering, and
virus, including insects. The disease remains endemic in certain
chomping. Lesions are occasionally seen on the vulva or scrotum.
river basins and coastal areas. In temperate zones, the appearance
In nursing sows, vesicles may occur on the teats. Vesicles often
develop at points of abrasion.
112 VESICULAR STOMATITIS
DISEASES CAUSED BY VIRUSES

Vesicles usually rupture within 24 hours and the overlying testing, respectively. Virus isolation and polymerase chain reac-
epithelium detaches to leave shallow erosions with reddened or tion assays are readily available for vesicular stomatitis diagnosis.
hemorrhagic surfaces. Lesions usually heal within 2 weeks unless A full set of tissues should be taken for histologic evaluation.
secondarily infected with bacteria.
Foot-and-mouth disease is of primary concern in differential
Pregnant sows may abort as a consequence of fever but none of diagnosis. If horses are affected, it is useful to know that horses
the vesicular viruses have been reported to affect fetuses. The are susceptible to vesicular stomatitis but are not susceptible to
the other exotic vesicular viral diseases.

VIRUSES
morbidity rate for vesicular stomatitis is highly variable and
ranges from 5% to more than 90%. Some reports suggest that
only 10% to 20% of the animals in a herd may become symptom- Control
atic even though nearly all may seroconvert to the virus. During outbreaks of vesicular stomatitis, uninfected livestock
should be kept away from any animals that could be infected.
Diagnosis Quarantines and animal movement restrictions can help reduce
Because signs and lesions of the vesicular viral diseases are similar, virus spread.
a variety of laboratory techniques have been developed for dif-
There should be no movement of animals from a quarantined
ferential diagnosis. In the US, the Federal Animal Disease Diag-
property for at least 21 days after all lesions are healed. Isolating
nostic Laboratory at Plum Island, New York, will usually have
symptomatic animals may also be helpful in controlling spread
responsibility for confirming a diagnosis of vesicular stomatitis. If
within a herd. Potential fomites should be cleaned and disin-
signs or lesions consistent with vesicular stomatitis are observed,
fected to prevent spread of the virus.
state animal disease control offices should be contacted and will
help make necessary arrangements for sample testing. Emergency measures for control of outbreaks of vesicular diseases
in swine require quarantine, identification of the disease, and
Epithelial and vesicular fluid samples can be tested using com-
slaughter of all infected and exposed swine. This is followed by a
plement fixation or enzyme-linked immunosorbent assay tests to
thorough cleaning and disinfection of the premises.
identify viral antigen. Heparinized and clotted blood should be
collected for virus isolation and identification, and for serological

VESICULAR STOMATITIS 113


VIRUSES

114 VESICULAR STOMATITIS


Section III

Emerging viruses

Filoviruses (Ebola virus and Marburg virus), Hepatitis E virus ...............................................................................................117


Paramyxoviruses (Menangle and Nipah viruses), Pestivirus F (Bungowannah virus) .........................................................118

EMERGING
Porcine adenovirus, Porcine astrovirus, Porcine bocavirus ..................................................................................................119
Porcine parainfluenza virus type 1, Porcine sapovirus..........................................................................................................120
Torque teno virus, West Nile virus (Kunjin virus) ................................................................................................................121

115
116
EMERGING
EMERGING VIRUSES

Filoviruses (Ebola virus and Marburg virus)


Ebola virus (EBOV) and the closely related Marburg virus A 2-part study was undertaken to determine the effect of REBOV
(MARV) belong to the family Filoviridae. They are enveloped, on pigs. For each study, 8 pathogen-free pigs were kept in 2 groups
filamentous, negative-sense RNA viruses and may be associated of 4. In study 1, REBOV genome was detected in nasopharyngeal
with severe zoonotic disease in humans. Currently four species of secretions of pigs from 2 to 8 days following exposure by the
EBOV have been reported: Zaire ebolavirus (ZEBOV), Sudan oronasal route. Neither genome nor virus was found in rectal
ebolavirus (SEBOV), Ivory Coast ebolavirus, and Reston ebola- swabs, urine, or blood of these animals at any time. Tissue samples
virus (REBOV). A fifth species, Bundibugyo ebolavirus, has been collected at euthanasia on day 28 after infection were negative by
proposed. Filoviruses occur mainly in regions of Africa; bats are polymerase chain reaction (PCR) for REBOV. In study 2, REBOV
thought to be the primary reservoir hosts. Four species of EBOV genome was found in nasopharyngeal secretions, rectal swabs,
have caused fatal disease in humans, with mortality rates of 50% blood, skeletal muscle, and most other organs and tissues that were
to 90%, depending on virus strain. In contrast, REBOV has never sampled 1 to 2 weeks after inoculation.
been identified as the cause of disease in people, though there are
In experimentally infected 3- to 6-week-old piglets, ZEBOV

EMERGING
some reports of seroconversion occurring.
caused mostly respiratory signs with no hematologic abnormali-
Reston virus has been detected in pigs that were suffering from ties. Experimentally infected pigs were able to transmit the infec-
porcine reproductive and respiratory syndrome in China and the tion to in-contact pigs and indirectly to non-human primates.
Philippines. Subsequently, several pig farmers were found to be Until recently, validated serologic tests have not been available to
seropositive for REBOV antibodies, providing a strong indica- determine the significance, if any, of wild pigs as a vector or reser-
tion that they acquired the virus from infected pigs. Experimen- voir species for EBOV in West Africa. In Sierra Leone, 400 sera
tally, infection of pigs with both REBOV and EBOV has been from village pigs were analyzed for the presence of EBOV-specific
successfully achieved. antibodies. Three samples reacted with antibody to EBOV nucle-
oprotein but had no neutralizing antibodies.

Hepatitis E virus
Hepatitis E virus (HEV) is a non-enveloped RNA virus in the The virus can be found in nearly every region in the world. Geno-
genus Orthohepevirus in the family Hepeviridae. There is a single typic niches occur, with HEV-3 and HEV-4 mainly occurring
HEV serotype, but the genus Orthohepevirus contains 4 subgen- in the US. Eighty to 100% of commercial swine farms in the US
era based on species affected. Within the subgenus Orthohepevi- show evidence of infection with HEV. Most pigs acquire HEV
rus A, there are at least 7 different genotypes. Hepatitis E virus-3 via the fecal-oral route, and HEV infections in swine are usually
and HEV-4 are zoonotic with swine serving as the reservoir host. asymptomatic. Hepatitis E virus is not cultivatable in cell culture.
Little is known about virus survival outside the host. However, Hepatitis E infection of people in both developed and developing
HEV is thought to be stable in the environment since feces are countries appears to be on the rise. A number of recent review
an important source of infection. Heating pork products to an articles are available describing important risk factors for infec-
internal temperature of 160°F (71°C) for 20 minutes is required tion with human or environmental genotypes of the virus as well
to completely inactivate HEV. as zoonotic infections arising from contact with pigs, their waste,
or pork products (usually undercooked liver or pates). Several
Hepatitis E virus can be isolated from many different species,
hundred papers are published each year on this disease in people;
including pigs, and is zoonotic. The severity of infection in
far fewer papers deal with the disease directly in pigs.
humans varies from subclinical to causing fulminant hepatitis and
occasionally reproductive disease. Pigs are the most important Hepatitis E virus infection of people caused either by environ-
animal reservoir for genotypes capable of infecting people. mental or swine-origin viruses appears to be increasing globally.

FILOVIRUSES; HEPATITIS E VIRUS 117


SWINE DISEASE MANUAL

Paramyxoviruses (Menangle and Nipah viruses)


Menangle and Nipah viruses are members of the family Menangle virus, like Nipah and Hendra viruses, circulates among
Paramyxoviridae, which also includes a number of other viruses flying foxes. It has only been identified in Australia. The virus
that have emerged following transmission to a new host species. does not cause disease in bats; however, in the single outbreak
For example, measles virus emerged in humans around 10,000 in pigs that occurred in New South Wales, Australia in 1997,
years ago from an undetermined host, which also appeared to be reproductive disease was apparent. During this outbreak, infected
a common ancestor of the ruminant viruses Rinderpest and peste sows gave birth to litters with mummified or stillborn piglets,
des petits. Also, canine distemper virus has recently emerged in some with congenital defects of the skeleton and central nervous
lions, various marine mammals, and ferrets, presumably as a result system. No clinical disease was otherwise noted in the sows or
in other pigs on the farm. There is no evidence that Menangle
of a jump from its domestic dog host. Pteropid fruit bats are the
virus continues to circulate in Australian pigs, but sporadically,
reservoir host for Menangle and Nipah viruses.
individual pigs have been found to be antibody positive to the
Nipah virus emerged in pigs in Malaysia and Singapore in virus. Menangle virus is zoonotic, as evidenced by two farm
1999. In a dramatic, multi-site outbreak, pigs were exhibiting workers who were in close contact with infected pigs on the New
EMERGING

respiratory and neurological signs. Morbidity was high in these South Wales outbreak farm becoming ill with severe but self-
outbreaks, but mortality was only moderate. Concurrently, some limiting influenza-like disease; the workers were later determined
pig-farm workers were also showing signs of severe pneumonia to be antibody positive for Menangle.
as a result of direct contact with pigs or through short-distance After experimental inoculation, 6-week-old pigs were found to
aerosols; pigs are thought to act as amplifying hosts for the shed Menangle virus in nasal and oral secretions, urine, and feces,
virus. An extensive epidemiological workup involving both and could transmit the infection to in-contact pigs. Though pigs
veterinarians and public health workers was conducted around remained asymptomatic throughout the study, virus shedding in
the outbreak. To eradicate the outbreak, approximately 1,000,000 the urine of some pigs continued for nearly 3 weeks, but there
pigs were euthanized over approximately 6 months. An estimated was no evidence that pigs become persistently infected beyond
265 people were infected with Nipah virus, of which 105 this period. No efforts have been made to reproduce the disease in
died - a case fatality rate of nearly 40%. Though this outbreak pregnant sows.
was contained, smaller outbreaks have continued to appear in
the region including Bangladesh, India, parts of South Asia, Pteropid bats have featured significantly in the last 10 to 15 years
and Oceania. These outbreaks have primarily involved people as a source of emerging viral diseases in humans and livestock. It
(including human-to-human transmission) and ruminants. is assumed that human effects (deforestation, increasingly inten-
Nipah virus is closely related to Hendra virus, which to date has sified livestock production near bat habit, climate change, etc) on
only been found in Australia. Hendra virus shares similar features the bat’s normal habitat are placing bats, livestock, and humans
to Nipah, but thus far infection has been limited to pteropid bats, in greater contact than ever before and therefore increasing the
horses, and humans. opportunity for interspecies disease transfer.

Pestivirus F (Bungowannah virus)


In June 2003, an outbreak of disease was reported on a pig farm stillborns had a distinctive subcutaneous edema over the head
in New South Wales, Australia. The outbreak initially presented and thorax. Pathological changes in affected animals consisted
as sudden death in 2- to 3-week-old piglets around the time almost exclusively of a multifocal non-suppurative myocarditis,
of weaning, but soon after the onset, deaths were observed in with myonecrosis observed in some cases. The clinical presenta-
younger pigs and there was a marked increase in the birth of tion was referred to as the porcine myocarditis syndrome. The
stillborn piglets, with a slight increase in the occurrence of mum- virus was initially named Bungowannah virus and was recently
mified piglets. Cumulative losses in some weeks exceeded 50% renamed Pestivirus F.
of piglets born, and estimates for losses to production from the
To date, the virus has only been detected in a limited number
outbreak were as high as 50,000 pigs.
of farms in Australia. Other countries, including the US, have
Gross lesions at necropsy included hydropericardium, excess undertaken limited surveillance for the agent but it has not been
pleural and pericardial fluid, and a dilated heart. Some affected detected outside Australia.

118 PARAMYXOVIRUSES; PESTIVIRUS F


EMERGING VIRUSES

Porcine adenovirus
Porcine adenovirus (PAdV) is a non-enveloped DNA virus in the Porcine adenovirus generally causes subclinical infections and
genus Mastadenovirus, family Adenoviridae. There are currently is commonly isolated from the gastrointestinal tract of normal
3 porcine species and 5 serotypes recognized: PAdV-A (serotypes swine. However, PAdV has been sporadically associated with
1-3), PAdV-B (serotype 4), and PAdV-C (serotype 5). Adeno- enteritis, encephalitis, nephritis, respiratory disease, conjunctivi-
viruses are relatively host-specific, and pigs are the only known tis, and reproductive disorders, though other concurrent diseases
hosts for PAdV. or causes of immune suppression are frequently reported in these
instances.
The geographic distribution of PAdVs is not fully known,
Porcine adenovirus has been detected on pork carcasses after
although the virus is suspected to be widespread in domestic bleeding, dehairing, pasteurization, and evisceration, and on
swine populations. Serological surveys from Canada and England some retail pork samples. Most evidence suggests adenovirus in
in the 1960s and 1970s indicated that up to 60% of swine test these locations is due to fecal or wash-water contamination rather
positive for anti-PAdV antibodies; additional countries in Europe than a primary infection of the carcass or tissue.
and South America have since reported presence of the virus and

EMERGING
Adenovirus-based products have been developed for use as vec-
it is assumed the virus is distributed globally. However, current
tors in both human and animal vaccines.
seroprevalence data is lacking.

Porcine astrovirus
Porcine astrovirus (PAstV) is a non-enveloped RNA virus in the winter, but there is little information about seasonal incidence of
family Astroviridae. There are 5 known lineages, each having sub- AstV infection in pigs.
stantial genetic variability. Astrovirus (AstV) is resistant to heat
Prevalence of PAstV in swine feces can be very high. However,
and requires very acidic pH for inactivation; it can survive for a
viral co-infection with other enteric pathogens (eg, rotavirus,
long time in the environment.
transmissible gastroenteritis virus, porcine circovirus 2, and por-
Astroviruses affect a wide variety of species and are mostly asso- cine hemagglutinating encephalitis virus) is often observed. Case
ciated with gastrointestinal disease. Porcine astrovirus was first fatality rates for PAstV-associated neurological disease on affected
isolated from the feces of pigs in 1980. Astroviruses have also US swine farms have been reported between 75% and 100%.
been linked to neurological disease in humans, mink, cattle, and
Transmission of PAstV is thought to be fecal-oral. Porcine astro-
sheep. Recently, PAstV-associated polioencephalomyelitis has
viruses are thought to cause mild, self-limiting secretory diarrhea.
been reported in pigs in Hungary and the US. Porcine astrovirus
Young pigs are most affected. Viral co-infections may contribute to
has previously been found in pigs with congenital tremors but the
observed clinical signs. Reported neurological signs associated with
significance of this is unknown.
PAstV include hind limb weakness, posterior paraplegia, quadri-
The zoonotic potential of AstV is unclear; porcine-human AstV plegia, convulsions, loss of consciousness, and flaccid paralysis of
recombinants have been documented and human-to-pig trans- muscles. Congenital tremors may be related to PAstV infection in
mission is suspected. Porcine astrovirus has been isolated from in piglets, but this is unclear.
pigs worldwide. Human AstV infections are most common in the

Porcine bocavirus
Porcine bocavirus (PBoV) is a non-enveloped, single-stranded Survival of PBoV in the environment has not been reported. In
DNA virus in the family Parvoviridae, genus Bocavirus. Although general, parvoviruses are very resistant to dry heat, requiring
bocaviruses (BoVs) have been recognized in veterinary medicine 158°F (70°C) for 10 minutes to become inactivated. In
since the early 1960s, PBoV was not isolated until 2009 in Swe- addition to infecting pigs, BoVs have been linked to respiratory
den, where the virus was found in the lymph nodes of pigs with disease, gastrointestinal disease, and reproductive loss in cattle.
post-weaning multisystemic wasting syndrome. No cases of PBoV have been reported in humans. However,
PBoVs and human strains are antigenically cross-reactive. Viral
Currently there are seven known PBoV genotypes, divided into
recombination has also been documented in both human and
three distinct groups based on the sequence of the VP1 gene:
porcine BoVs, raising the possibility of cross-species transmission.
PBoV G1, PBoV G2, and PBoV G3.

PORCINE ADENOVIRUS; PORCINE ASTROVIRUS; PORCINE BOCAVIRUS 119


SWINE DISEASE MANUAL

Porcine bocaviruses have been identified in Africa, Asia, Eastern and feces. The pathogenesis of PBoV and its ability to induce
Europe, North America, and the United Kingdom. Reported clinical signs are not well understood. Viral coinfections are com-
PBoV prevalence in sick pigs in China is around 11%, while US mon in PBoV-positive pigs. To date, PBoVs have been reported
studies have documented prevalence rates in sick pigs of approxi- in pigs with porcine circovirus associated disease (PCVAD),
mately 43% and 59%. respiratory disease, diarrhea, and in asymptomatic swine. Repro-
ductive failure may also occur. A single case of PBoV-associated
Infection occurs most commonly in weaned piglets. Transmission
encephalitis has been reported. It does not appear that any studies
routes of PBoV remain unclear. Viral DNA can be detected in a
of experimental PBoV infection have been conducted in pigs.
range of tissues, including lymph nodes, serum, lung, oral fluids,

Porcine parainfluenza virus type 1


Porcine parainfluenza virus type 1 (PPIV-1; species porcine may play a role in the porcine respiratory disease complex. How-
respirovirus 1) was first identified in 2013 from nasopharyngeal ever, the pathogenesis of PPIV-1 has not been fully elucidated
EMERGING

samples collected from deceased slaughterhouse pigs in Hong and the causal links between PPIV-1 and disease have not been
Kong, China. The PPIV-1 genome was reported at that time to established.
be a novel paramyxovirus with highest genetic similarity to Sen-
Porcine parainfluenza virus type 1 is distinctly different than
dai virus and human parainfluenza virus type 1.
parainfluenza virus 5 which has recently been isolated from
Porcine respirovirus type 1 is a member of the genus Respirovirus diarrhea-affected piglets in China. Parainfluenza virus 5 is a
in the Paramyxoviridae family and is an enveloped, single-stranded, negative-sense, non-segmented, single-stranded RNA virus that
negative sense RNA virus. Porcine respirovirus-1 is widespread in belongs to the genus Rubulavirus in the family Paramyxoviri-
the US and has been detected by PCR in all ages of swine but more dae. This virus was present in intestinal tissue samples collected
commonly in nursery and grow-finish pigs. The virus has been from diarrhea-affected piglets in 4 provinces and municipalities
detected in respiratory samples, including lung tissues, nasal swabs, of the country, but little additional information exists at this
and oral fluids. time regarding pathogenesis or its association with other enteric
pathogens.
Viral infection has been associated with respiratory disease in
pigs, with clinical signs that may include coughing, minor sneez-
ing, and serous nasal discharge. It has been suggested that PPIV-1

Porcine sapovirus
Porcine sapovirus (PSaV) is a non-enveloped RNA virus in the Canadian investigators identified porcine enteric calicivirus
family Caliciviridae. Different strains of porcine sapovirus (PSaV) (assumed to be PSaV) in 18 of 88 (20.5%) Canadian finishing
can simultaneously circulate within swine farms or herds; PSaV farms that were tested, confirming its widespread occurrence
was previously known as porcine enteric calicivirus. in that industry. Similarly, researchers in Brazil investigated the
Porcine sapovirus is stable at temperatures of 127°F (56°C) and occurrence of the virus on pig farms. Fecal samples of asymp-
at pH 3 to 8. Recombination events between human and porcine tomatic pigs aged 9 to 24 weeks old were collected from 16
sapoviruses seem likely. grow-finish herds and evaluated for porcine norovirus (assumed
to be PSaV) presence by PCR. Porcine norovirus was detected in
Porcine sapovirus has been isolated from pigs in Belgium, Brazil, 58 (51.8 %) of the 112 fecal samples and in 14 (87.5 %) of the 16
China, Denmark, Finland, Hungary, Italy, Japan, Slovenia, South herds evaluated.
Korea, Spain, the US, and Venezuela, but human sapoviruses are
found worldwide. A surveillance study among pig farms in Europe There have been no reports of PSaV-related fatalities in pigs.
found PSaV on 44.3% of farms and in 7.6% of all pigs, with the Transmission of enteric caliciviruses is thought to be fecal-oral.
highest prevalence in piglets aged 2 to 8 weeks. Porcine sapovi- Experimentally infected piglets typically develop diarrhea.
ruses were detected by PCR in 62% of fecal samples collected Vomiting and diarrhea were noted in a 2008 outbreak in pigs in
from 3 states in the US from swine of all ages. Post-weaning pigs China; both signs were reproduced experimentally in 10-day-old
had the highest prevalence, while the lowest prevalence was in piglets after feedback of feces from infected pigs. Subclinical
nursing pigs (20% to 21%). infection also seems to occur.

120 PORCINE PARAINFLUENZA VIRUS TYPE 1; PORCINE SAPOVIRUS


EMERGING VIRUSES

Torque teno virus


Torque teno viruses are small, non-enveloped viruses with a One study found that in pigs suffering from PCVAD, the levels
circular, single-stranded DNA genome belonging to the newly of TTSuV2 had higher viral DNA titers but lower anti-TTSuV2
created family Anelloviridae. In pigs, the genus Iotatorquevirus antibody levels than healthy animals. In another study of natu-
is composed of 2 species, TTSuV1a (formerly TTSuV1) and rally infected pigs from 5 weeks of age up to 24 weeks of age, the
TTSuV1b (formerly TTSuV2), and the genus Kappatorquevirus virus was present in a number of tissues (brain, lung, mediastinal
also includes 2 species, Torque teno sus virus k2a and k2b. lymph node, heart, spleen, kidney, mesenteric lymph node, and
bone marrow) with medium to high DNA loads.
The Torque teno viruses have been identified in tissue and feces
of pigs from many countries around the world. Transmission of While Anelloviruses are considered to be highly species-specific,
the viruses among pigs is horizontal and mainly via the fecal-oral one study showed porcine anelloviruses were detected in 5 out of
route. It is unknown whether they are a primary cause of disease, 21 (24%) human fecal samples from volunteers eating pork meat
but many authors seem to agree that they are most likely found as between 2 and 7 times per week, though there was no evidence
part of a co-infection with other pathogens and that in these situ- that tissue infection actually occurred in the humans.

EMERGING
ations, their presence might promote increased disease severity or
virulence. Little evidence to date supports their role as a significant
pathogen in pigs.

West Nile virus (Kunjin virus)


West Nile virus (WNV) is a mosquito-borne virus that circulates high viral amplification, significant numbers of “bridge vector”
among birds but can also affect other species, particularly humans mosquitoes (mosquitoes that feed on both birds and mammals)
and horses, and until the 1990s, tended to circulate only in parts become infected in the late summer and then are able to trans-
of Africa. Most human infections were asymptomatic. Since that mit the virus to humans, horses, and other incidental hosts. In
time, this picture has changed, with West Nile virus emerging as some birds, viremia can persist for more than 3 months, possibly
a significant human and veterinary pathogen in the Americas, contributing to the overwintering of the virus.
Europe, the Middle East, and other areas. The disease became
Among mammals, disease occurs mainly in equids (horses, don-
established in the US in 1999, and while approximately 80% of
keys, and mules), and an efficacious vaccine has been developed
the people infected with these strains were asymptomatic, 20%
for use in these species.
had flu-like signs and a small but significant percentage (<1%)
developed neurological disease. Although the introduction of an Little work has been done with West Nile virus in pigs. However,
equine vaccine has helped control the disease in horses, there is several seroprevalence studies in feral pigs have been published,
currently no vaccine for humans. Recent experiences suggest that including one from the US in which 222 samples were collected
human outbreaks may continue to occur in North America at from 2001 to 2004. The overall seroprevalence was 22.5%, with
unpredictable intervals. individual rates in Florida, Georgia, and Texas reported as 17.2%,
26.3%, and 20.5% respectively. Young adult and weaner pigs
West Nile virus is an arbovirus in the Flavivirus genus of the
were experimentally challenged with the NY99 strain of the virus
family Flaviviridae. It belongs to the Japanese encephalitis virus
through the bite of infected mosquitoes. While each of the adult
complex or serogroup. The strain that entered the US in 1999 is
pigs seroconverted to the virus, virus could only be isolated from
called NY99 and is among the most pathogenic strains. Kunjin
the serum of one pig. In the weaner pigs, however, 3 of 5 devel-
virus is a subtype of West Nile Virus found in Australia.
oped a viremia. No clinical signs attributable to WNV infection
Wild birds are the main reservoir hosts for WNV; passeri- were observed in any of these animals. The authors concluded
formes (perching birds) are important in virus amplification. that pigs are unlikely to play a significant role as amplifying hosts
In endemic regions, the virus is maintained in a cycle between of West Nile virus.
mosquitoes and birds. When environmental conditions favor

TORQUE TENO VIRUS; WEST NILE VIRUS 121


122
EMERGING
Section IV

Diseases caused by parasites

Coccidiosis.....................................................................................................................................................................................125
Cysticercosis...................................................................................................................................................................................127
Lice...................................................................................................................................................................................................129
Lungworm......................................................................................................................................................................................130
Mange..............................................................................................................................................................................................132
Minor intestinal nematodes: Kidney worm, Nodular worm, Thorny-headed worm, Threadworm...........................134
Roundworm...................................................................................................................................................................................137
Trichinellosis..................................................................................................................................................................................139

PARASITES
Whipworm.....................................................................................................................................................................................141

123
124
PARASITES
DISEASES CAUSED BY PARASITES

Coccidiosis
Alternate names and abbreviations and 36°C), which is well within the range maintained in nearly
Isosporosis (archaic), Isospora (archaic), cystoisosporosis, all confinement farrowing facilities. Fully sporulated oocysts
Cystoisospora, Eimeria of C suis have 2 sporocysts and each of these has 4 sporozoites.
Because of the rapid generation of high numbers of oocysts and
Definition the rapid onset of infectivity, Cystoisospora suis is the usual cause
Coccidiosis is the enteric disease caused by infestation with of coccidiosis in very young, confinement-raised pigs. Eimeria
variety of coccidia, usually manifested as diarrhea in suckling or species are quite similar but have a longer (18 day) life cycle and
recently weaned pigs. The disease is usually caused by Cystoisos- therefore are much less likely to cause disease in neonates. Eime-
pora (previously Isospora) suis in suckling piglets and by Eimeria ria outbreaks are more likely to occur in the nursery phase as a
species in older pigs. result of environmental contamination by oocysts from previous
groups of pigs.
Occurrence
Neonatal coccidiosis caused by C suis usually occurs in con- Epidemiology
finement-raised nursing piglets that are more than 5 days old; Sporulated coccidia are very resistant to cleaning and disin-
it is less frequent and less severe in older, recently weaned pigs. fectants, which explains their ubiquitous distribution. Disease
Coccidiosis occurs in all countries where confinement rearing results when naïve piglets or weanlings are exposed to high
and continuous farrowing are practiced, with outbreaks tending doses of infective oocysts. Coccidia are initially introduced into
to be more common in warmer months. Several coccidia of the farrowing facilities by carrier sows, but the dose in the envi-
genus Eimeria, particularly E spinosum, commonly infect 1- to ronment is then magnified over time by the accumulated fecal
3-month-old swine and can be a primary cause of enterocolitis contamination from previous litters and/or infected littermates.
when previously unexposed pigs are placed in highly contami- Sporulation is required for infectivity and this occurs in as little

PARASITES
nated environments, including unsanitary nurseries, grow-finish as 12 hours in warm environments. Parasitized piglets produce
facilities, or boar studs. very large numbers of oocysts that survive extremely well on
surfaces in the farrowing environment. Once sporulated, these
Historical information oocysts become a major source of infection for littermates and
Although first associated with enteric disease in 1934, the subsequent litters. When infected with an identical dose, piglets
coccidia found in pigs were considered as common endemic pro- infected at 1 to 3 days of age have much more severe disease than
tozoans of little consequence until the 1970s. The prevalence of those infected beyond 3 weeks of age (ie, disease is more severe
disease increased with the adoption of confinement rearing and in younger piglets). High doses of oocysts are required to cause
continuous farrowing, particularly in inadequately disinfected clinical disease, but high doses occur frequently as a result of the
farrowing crates located over solid floors in a warm environment. environmental buildup of oocysts in farrowing facilities. Inade-
These conditions facilitated rapid completion of the life cycle of quate sanitary practices between farrowing groups undoubtedly
C suis, resulting in the continuous presence of high numbers of facilitate this buildup.
infective oocysts in farrowing crates. Fortunately, prevalence of
the disease decreased with improvements in perforated flooring Pathogenesis
materials and hygiene, but the disease remains a problem for All stages of the development of coccidia occur in intestinal
some farrowing and nursery operations even when dedicated villus enterocytes. This results in death and desquamation of
efforts are made to eliminate the infection. Eimeria species are enterocytes, especially those on the distal tips of villi. The severity
also a common infection but cause disease only when naïve pigs of lesions is related to the number of oocysts ingested. Bacteria
are exposed to high environmental doses. in the intestine, including Clostridium species, may contribute
secondarily to the overall severity of the lesions. When many
Etiology coccidia are ingested, lesions are severe. In serious infections, the
Cystoisospora suis goes through repeated cycles of sporogeny, erosion of villous epithelium results in loss of fluid and failure of
excystation, and endogenous development within intestinal surviving epithelium to absorb nutrients and fluids. This leads to
villus epithelium more rapidly than most Eimeria. Development diarrhea, dehydration, and loss of electrolytes, perhaps death.
usually occurs in the small intestine, especially in the jejunum and Since regeneration of villous epithelium occurs more slowly in
ileum but less often in the duodenum, cecum, and colon. Oocysts neonates, they are more severely affected. If piglets ingest only
may be shed in as little as 5 days postinfestation with C suis. small numbers of oocysts, they survive and develop effective
Oocysts shed from piglets rapidly become infective within about immunity unless they are in a stressful environment or compro-
12 hours when held in temperatures between 68° and 97°F (20° mised by secondary infections. In a warm, moist environment,

COCCIDIOSIS 125
SWINE DISEASE MANUAL

oocysts in feces-contaminated farrowing rooms and crates soon Diagnosis


become infective. They are ingested when piglets first nurse or Diagnosis of disease states is most accurate by identifying the coc-
when they ingest feed, water, or feces in the farrowing crate. The cidia in histologic sections. The presence of paired type 1 mero-
sporozoites within the oocysts mature and are released to pene- zoites is considered diagnostic for C suis. Several areas of infected
trate enterocytes. Many enterocytes are destroyed by the develop- mucosa should be examined in order to maximize sensitivity of
ing coccidia. Released oocysts are passed in the feces. the histological examination, as the distribution of microscopic
lesions and merozoites is unpredictable.
Clinical signs
Diagnosis by fecal examinations may be possible but is thought
With C suis, diarrhea can occur as early as 5 days after birth but
to be much less sensitive than examination of affected small
is more commonly seen in piglets from 1 to 3 weeks of age. Signs
intestinal tissue. In fecal samples, either “hazy bodies” between
include yellowish, pasty to clear, watery diarrhea, followed by
the oocyst wall and the sporont, or oocysts in the 2-cell stage,
dehydration, rough hair coat, and failure to gain weight. The
are diagnostic for C suis. For Eimeria species, detection of high
clinical disease within a litter can appear in two waves. The first
numbers of oocysts in feces and absence of other pathogens are
wave (5 to 6 days of age) is the result of some piglets in the litter
indicative of infestation. Coccidiosis can occur concurrently
becoming infected shortly after birth, developing diarrhea, and
with other diseases, so it is often necessary to eliminate them as
then subsequently heavily contaminating the farrowing crate. The
additional causes of the diarrhea. These include viruses (rotavirus,
second wave occurs 4 to 8 days later, after the rest of the litter
swine enteric coronavirus diseases), colibacillosis, parasitism by
becomes infected as a result of the first wave of infection.
Strongyloides ransomi, and infection with Clostridium perfringens
Piglets usually continue to nurse when affected by coccidiosis type A or type C.
but may vomit milk. Morbidity is high, but mortality is variable,
often low or moderate. Until becoming familiar with coccidiosis, Control
producers may report the disease as an outbreak of colibacillosis For control of coccidiosis in suckling pigs, a vigorous, effective
that fails to respond to the usual antibiotic therapy. sanitation program, including careful cleaning and disinfection
PARASITES

With Eimeria, nonspecific but persistent diarrhea occurs in the of farrowing crates between farrowings, is essential. Strong bleach
nursery phase. Acute enterocolitis has also been reported in naïve or ammonium compounds can be used for disinfection after
high-health boars placed in unsanitary boar studs. Fecal blood is thorough cleaning of the crates. Steam cleaning of the entire far-
not a feature of coccidiosis from C suis or Eimeria species. rowing facility may be necessary. Sealing all surfaces with paint or
a water sealant may be an option to break the cycle of infection.
Lesions Wooden and concrete surfaces are particularly difficult to clean
Lesions are proportional to the dose of oocytes ingested and effectively. Installation of perforated metal or plastic flooring in
the age of pigs at the time of infection. The lesions are usually the crates is essential for long-term control of coccidiosis and
restricted to the intestinal tract, especially the lower jejunum other neonatal enteric diseases.
and ileum. In mild infections, the intestine tends to be turgid. On farms known to be affected by coccidiosis, routine treatment
Mild fibrinous enteritis may be visible in short segments of the of all piglets with toltrazuril in the first 1 to 2 days after birth has
lower small intestine. In severe infections, there may be extensive been used to minimize the severity of coccidiosis. There is no
fibrinonecrotic enteritis of the terminal third of small intestine proven, widely accepted anticoccidial or drug for use in dams that
and occasionally colon. Segmental necrotic enteritis in pigs 1 to is effective at controlling the disease in neonates.
4 weeks of age is fairly common with severe C suis insults. Gross
Most cases of coccidiosis associated with Eimeria species are the
lesions with Eimeria species tend to be subtle.
result of the introduction of naïve pigs into a heavily contami-
Microscopically, there is necrosis and later, atrophy and fusion of nated environment. Control is based on minimizing the dose of
some mucosal villi along with hyperplasia of cryptal epithelium, oocysts ingested. Scrupulous sanitation between groups of pigs
elongation of mucosal crypts, and nonsuppurative infiltrates in is usually sufficient to prevent disease. Amprolium is effective for
the lamina propria. Shortened villi may be covered by cuboidal or treatment of Eimeria outbreaks but not Cystoisospora.
flattened epithelial cells. Various stages of Cystoisospora or Eime-
ria are apparent in parasitized cells. If adequate time has elapsed
since initial infection, there may be oocysts in the feces.

126 COCCIDIOSIS
DISEASES CAUSED BY PARASITES

Cysticercosis The ancient Greeks were also familiar with the disease. In Aris-
tophanes’ (c 448 to 385 BC) comedic play “The Knights,” one of
Alternate names and abbreviations the supporting characters aids the local sausage-maker in berating
Taenia, taeniasis, taeniosis, pork measles the play’s protagonist by suggesting they:

set his mouth open with a wooden stick as the cooks do with
Definition pigs; we will tear out his tongue, and looking down his gaping
Cysticercosis occurs as a result of pigs consuming eggs of the
throat, will see whether his inside has any pimples,
human tapeworm, Taenia solium. The larvae of the parasite
migrate to skeletal and cardiac muscles where they form cysticerci
and remain as an important source of infection for a subsequent a clear reference to the practice of slicing the tongue of pigs
human host that consumes raw or undercooked pork. Infected during slaughter to check for the presence of cysticerci. Aristotle
pigs are generally asymptomatic. (c 384 to 322 BC) described in detail the presence of cysts in
pig muscles that were similar to hailstones. Pigs were considered
The terms taeniasis or taeniosis refer to intestinal infection of the impure in ancient Greece; it is possible that this belief induced
definitive host (humans) with the adult tapeworm. Cysticercosis Muhammad (570 to 632 AD) to prohibit the consumption of
refers to extra-intestinal infection of the intermediate host (pigs) pork when the Quran was written.
with the encysted larval stage of the parasite in muscle tissue.
Humans can also develop cysticercosis after ingesting T solium Taeniasis remains an important zoonotic disease today. An out-
eggs through contact with human sewage and thus can act both break of neurocysticercosis was identified among members of a
as a direct or intermediate host. Cysticerci in humans often occur non-pork-eating Orthodox Jewish community of New York City
in the central nervous system, causing a condition known as neu- in 1990 to 1991. Investigation of the outbreak concluded that the
rocysticercosis. source of the infection was most likely a result of incidental con-
tact between domestic staff (recent migrants from Latin America,
Taenia saginata is a related, but different, species of human tape- subsequently determined to be actively infected with the para-

PARASITES
worm that causes cysticercosis in cattle. site) and family members. The outbreak highlighted the fact that
contact with pigs is not required to explain the occurrence of
Occurrence taeniasis and neurocysticercosis in people and served to increase
Taenia solium is considered to be endemic in parts of Asia, research and awareness of the disease over the last 25 years.
South and Central America, and sub-Saharan Africa. In Europe
Neurocysticercosis is the most common parasitic infection of the
and North America, modern pig rearing systems that minimize
brain and is an endemic condition in many developing countries
contact between pigs and human sewage, in combination with
of Africa, Asia, Central America, and South America. It has
long-term commitments to meat inspection and improvements
been estimated that in these areas, 25% to 40% of patients with
to public health and sanitation, are believed to have eliminated
new-onset epilepsy have evidence of neurocysticercosis. At the
autochthonous infections. However, human cases still occur in
same time, reported cases of neurocysticercosis are increasing in
these regions related to international travel or migratory move-
the US (especially in southwestern states) and other developed
ments towards and from endemic countries.
countries, especially among immigrants from endemic areas.
Taeniasis and human cysticercosis are not notifiable in most coun-
tries; therefore it is difficult to assess the true prevalence of human Etiology
infection in various parts of the world. Porcine cysticercosis must Taenia solium belongs to the family Taeniidae, which includes
be reported to the World Organization for Animal Health, but 3 parasites affecting humans: T solium (the pork tapeworm),
there is no mandatory reporting for bovine cysticercosis. Taenia saginata (the beef tapeworm), and Taenia asiatica (a tape-
worm that is morphologically similar to T saginata but biologi-
Historical information cally analogous to T solium).
Human taeniasis and porcine and human cysticercosis are ancient
diseases and almost certainly part of the basis for certain societal and Epidemiology
religious biases against pigs and pork. In the third book of the Bible The major risk factor for cysticercosis in pigs is exposure to con-
(Leviticus, 11:7-8), it is written: taminated human sewage through a water or feed source. Pigs
can potentially become infected by consuming the carcass of an
And the pig, though it has a divided hoof, does not chew infected, deceased pig but this route of transmission does not
the cud; it is unclean for you. You must not eat their appear to feature significantly in the epidemiology of the disease.
meat or touch their carcasses; they are unclean for you. Infected pigs are generally not infectious to other pigs.

CYSTICERCOSIS 127
SWINE DISEASE MANUAL

Pathogenesis Taeniasis in humans is usually asymptomatic except for the passage


The life cycle of the human tapeworm T solium normally requires of proglottids in the stool.
the pig as an intermediate host, although humans do occasionally
develop neurocysticercosis after ingesting T solium eggs directly, Lesions
as described above. Cysticerci in pork are clear, translucent, or white; they are round
to ovoid in shape. Cysts are usually around 1 centimeter or less in
Adult tapeworms in the human consist of a scolex and numer- diameter and surrounded by a fibrous capsule. Each fluid-filled
ous body segments called proglottids. The terminal proglottid vesicle contains a single invaginated protoscolex which usually
contains thousands of eggs that are shed in carriers’ stools. Cys- appears as a single dense white body. An organ may contain one
ticercosis in pigs is a result of pigs ingesting these T solium eggs, to hundreds of cysticerci. When larvae die, the cyst degenerates
usually as a result of consuming water, food, or soil contaminated and eventually becomes filled with greenish or yellow caseous
with untreated human sewage. material. In pigs, most T solium cysticerci are found in the skeletal
When pigs ingest these eggs, oncospheres (larvae or embryos) or cardiac muscles, brain, liver, or heart.
hatch in the intestine and, after invading the intestinal wall, are
disseminated to other organs (usually muscle) where they mature Diagnosis
into cysts (cysticerci) over several weeks. Cysticerci consist of Antemortem diagnosis of cysticercosis in live pigs is difficult.
a membranous wall filled with fluid and an invaginated scolex Some serological tests have been described but are not in com-
(head). Humans who eat undercooked pork that contains cys- mon use. Cysticercosis is usually diagnosed postmortem during
ticerci can then develop a tapeworm infection (taeniasis) and meat inspection, which is why a series of sagittal slices is often
complete the life cycle of the parasite. made across the tongue of butchered pigs.
Human ingestion of cysticerci-infected pork typically leads only
to intestinal tapeworm infection and not neurocysticercosis. Neu-
Control
In theory, cysticercosis may be treated with anthelmintic drugs
rocysticercosis is generally a result of humans ingesting T solium
PARASITES

such as albendazole and praziquantel. More practically, the


eggs or larvae, with the larvae then moving to extraintestinal
disease is managed by controlling risk factors and exposure to T
sites in a manner similar to what occurs in pigs. Humans affected
solium eggs. Free-ranging pigs, particularly those with potential
by taeniasis are at very high risk of autoinfection and resultant
exposure to human sewage and untreated surface water are at
neurocysticercosis if they ingest tapeworm eggs through failures
highest risk of contracting the infection. Farm workers should
of their own personal hygiene; they are also a significant source of
use toilet facilities that ensure human feces do not come into
infection to others in their household. Humans with neurocystic-
contact with pigs; handwashing after using the toilet is a practice
ercosis are dead-end hosts and do not transmit the disease further.
that should obviously be encouraged. Those who travel to parts of
Taenia solium in humans reach the adult stage in 5 to 12 weeks the world where taeniasis is endemic should be advised to avoid
after ingestion of contaminated pork. Adult T solium are 2 to becoming infected through consumption of undercooked pork
7 meters long and may live for up to 25 years. Although up to or through contact with human sewage. Most taeniasis is asymp-
25 tapeworms have been recorded in a single person, there is tomatic; therefore, fecal examination or prophylactic treatment
usually only 1. Taenia solium eggs can survive in the environment may be advised in people known to have a high risk of exposure.
for a few weeks or months. Experimentally, pork can become
infectious to humans in as little as a few weeks after a pig ingests
T solium eggs. More typically, 2 to 3 months are required for
pork to become infectious to humans.

Clinical signs
Cysticercosis in pigs is usually asymptomatic during both the
larval migration and the establishment of cysts. Cysticerci can
induce some symptoms when the encysted larvae die or if a cyst
becomes large enough to interfere with the function of vital
organs.

128 CYSTICERCOSIS
DISEASES CAUSED BY PARASITES

Lice
Alternate names and abbreviations Pathogenesis
Haematopinus, pediculosis, hog louse Lice annoy swine when they pierce the skin to take a blood meal.
Dermatitis develops and leads to rubbing, scratching, and some
Definition loss of hair. Loss of blood is not severe in older pigs, but neonates
Infestation with the louse Haematopinus suis. and nursery pigs can exhibit anemia if heavily infested. Parasit-
ized pigs are restless. They may lose weight and utilize feed less
Occurrence efficiently.
Infestation with Haematopinus suis is common where husbandry Lice are known vectors of agents that cause swinepox, African
and management are poor or efficacious insecticides are not swine fever, Mycoplasma suis, and quite likely other blood-borne
available. These lice live only on swine, although they may tem- pathogens as well.
porarily infest people handling swine. All age groups of swine are
susceptible to infestation, with greater infestations when there Clinical signs
are many pigs in close contact. Infestation is more severe in the Persistent rubbing and scratching, patchy alopecia, and pallor
winter although it persists throughout the year. Presumably, the from anemia are clinical signs of pediculosis. Similar signs are
parasitism occurs worldwide wherever swine are raised. seen with sarcoptic mange. Pediculosis and sarcoptic mange often
occur concurrently and must be differentiated. Hog lots and
Historical information facilities with many rubbing sites also suggest that lice or mange
Prior to the development of good parasiticides, pediculosis in are present.
swine was very common. It was a common practice to use waste
oil or diesel fuel as a topical spray on swine to reduce the number Diagnosis
of lice, but the efficacy of this treatment was poor. The relatively Lesions include a rough, dull hair coat, often with patchy alo-

PARASITES
recent development of many effective parasiticides has made it pecia. A thorough examination of infested pigs will often result
possible for a producer to completely eradicate lice. Products that in seeing the lice with the naked eye. Examination of the inner
control both internal and external parasites, including lice, have aspect of the ears often reveals the lice; this area is also a preferred
made control of lice even more convenient. location to look for sarcoptic mange lesions.

Etiology It is a good practice to occasionally pick up a few small and grow-


Haematopinus suis is one of the largest species of lice, up to 6 mm ing pigs, turn them over, and examine their undersides. Lice, as
long. Haematopinus suis is a sucking louse that obtains blood well as lesions of several other diseases, are often readily apparent
meals from the host through its penetrating mouth parts. The on the posterior abdomen or on the inside surface of the legs.
louse spends its entire life on the host. It can survive off the host After the dehairing process at slaughter plants, the punctate cuta-
for only a few days. The female louse lives about 23 to 30 days, neous lesions caused by feeding lice are easily visible, especially in
lays 3 to 4 eggs (nits) per day, and attaches them to hair shafts white-skinned hogs.
near their base. Eggs are 1 to 2 mm long and light cream to gray
in color. They hatch in 12 to 20 days. Control
If they are not from a source that has been confirmed negative for
Epidemiology lice and mange, animals that are introduced into a herd should
Hog lice are host specific. They usually spread among swine when always be treated with a parasiticide while in an isolation area.
the pigs are in close contact, as when they crowd together for A minimum of 2 treatments, at 18- to 21-day intervals while in
warmth, shade, or comfort. Also, lice can spread to pigs moved into isolation prior to introduction to the herd, is required for elim-
quarters recently vacated by lousy animals. Infested animals added ination of lice and mange. Many effective parasiticides are now
to a clean herd often introduce lice. available. The repeated treatments facilitate the killing of juvenile
Lice living on a pig have certain preferred sites because they prefer lice that emerge from eggs 12 to 20 days after an initial treatment.
to feed on thin-skinned areas. Favorite sites include the neck, jowl, Several parasiticides control external parasites other than lice
flank, the inner side of the legs, and ears. In the ears, they often (flies, mites, and ticks). Injectable avermectins have been very
reside in nests. Similar nests often are found on the abdomen, effective in controlling both lice and mange; programs have been
sometimes within the circles of pityriasis rosea lesions. Location devised for eradicating both. Directions for use should be fol-
within the ear offers lice protection from parasiticides because it is lowed carefully. Some parasiticides require withholding periods
difficult to thoroughly wet the inner surface of the ear. prior to marketing.

LICE 129
SWINE DISEASE MANUAL

Lungworm
Alternate names and abbreviations Infection becomes patent in about 4 weeks. The larvated ova are
Metastrongylus, metastrongylosis coughed into the pharynx where they are swallowed and passed
out through the feces.
Definition Survival of lungworms depends largely on the survival of infective
Infection with any one or more of 3 species of metastrongyle larvae in earthworms for long periods of time as well as the ability
nematodes. of first-stage larvae to survive for long periods in swine feces.
Breeding stock and other older pigs can remain infected for long
Occurrence periods, and lungworm eggs in their feces continue to contami-
Swine can be infected by any of 3 species of metastrongyles. All nate the environment.
ages of swine are susceptible, but heavy infections occur mostly
in young pigs over 6 weeks of age. Infection is common in areas Pathogenesis
where pigs are raised outdoors with access to soil. Lungworms Third-stage larvae penetrate the small intestine mucosa and pass
likely occur in all countries where swine and the intermediate in lymph and blood to the liver and lungs. Some larvae migrate in
host (the earthworm) are present. the liver and cause gray to white areas of scarring, similar to those
caused by ascarid migration. Larvae that reach the lungs break
Historical information into alveoli where they mature, extending into terminal bronchi-
Historically, lungworm infection was a major problem in US oles. The parasites preferentially localize along the caudodorsal
swine production. With the development of confinement border of the diaphragmatic lobes. Their presence results in alve-
housing and subsequent lack of access to soil (and earthworms), olitis and bronchiolitis, which leads to coughing. Exudate that
lungworm infection has decreased in prevalence. However, where forms around lungworms and their ova partially or completely
pigs are raised in the outdoors, lungworm infection can still be a obstructs airways, resulting in alveolar emphysema and/or atelec-
PARASITES

serious problem. Parasiticides effective in controlling lungworms, tasis. Secondary bronchopneumonia is a common sequel.
as well as many other parasites, have improved control. At one
time, lungworms were erroneously believed to be important in Clinical signs
the transmission of influenza virus to swine. Common clinical signs include chronic, sometimes paroxysmal,
coughing that eventually leads to generalized unthriftiness. If
Etiology secondary pneumonia occurs, there may be dyspnea and perhaps
Lungworms that infect swine include Metastrongylus elongatus an abdominal type respiration (“thumping”). Signs are similar to
(apri), M pudendotectus, and M salmi, with the former most those caused by ascarid migration in the lungs. The two parasit-
common; infection with more than one species can occur. Lung- isms often occur concurrently where pastures are heavily contami-
worms usually have an indirect life cycle through earthworms, but nated by both ascarids and lungworms.
a direct cycle has been reported. Adult lungworms vary in length
from 14 to 66 mm. They are white, threadlike, and easily visible at Lesions
necropsy. Lungworm ova are larvated, thick-shelled, about 40 by Pigs with many lungworms are unthrifty, have a rough hair coat,
50 µm in size, and are passed in feces. and are small for their age. Pulmonary lesions often occur on the
caudodorsal border of diaphragmatic lobes. When viewed from
Epidemiology the dorsal aspect, the lesions often have a roughly triangular,
First-stage lungworm larvae hatch from eggs in swine feces and wedge-like shape and appear as recessed darker areas of atelectasis
survive in feces or moist soil for long periods of time. In some or raised, lightly colored, emphysematous areas; both types of
regions of the country, the first-stage larvae can survive over win- lesions can be present in the same pig. If the distal border of the
ter. After being ingested by earthworms, larvae are able to develop lobe is trimmed and lesion areas squeezed, adult worms can be
to the third infective stage in about 10 days. They then remain expelled. The bronchial tree should always be opened as lesions
quiescent in the earthworm for up to 18 months. Thousands of along the border are not always present. When this is done,
larvae can accumulate in a single earthworm, apparently without inflammatory exudate and lungworms may be found in terminal
harm to the earthworm. Pigs on pastures with soft or moist soils airways. Scattered 1- to 2-cm areas of gray to white scarring may
root for earthworms, ingest the parasitized earthworms, and are be visible in the liver.
then infected by the larvae within the earthworms. The lungworm
larvae penetrate the intestinal mucosa, migrate through lymphatics
and venous blood, and reach the lungs. There they localize, grow to
maturity, and produce larvated ova about 25 days after ingestion.

130 LUNGWORM
DISEASES CAUSED BY PARASITES

Diagnosis
Diagnosis is made by postmortem examination of a few typical,
affected pigs. Also, laboratory fecal examinations will usually
reveal the embryonated eggs of lungworms. Postmortem exam-
inations often require less time and have the advantage of permit-
ting examination for other parasites and diseases.

Control
Prevention of heavy infection is possible by preventing access of
pigs to soil that contains larvae or earthworms. Consequently,
most confinement-raised pigs are free of lungworms.
Pigs raised outdoors can often avoid heavy infection if they are
farrowed and raised on clean pastures. Pasture rotation is helpful
but sometimes fails to prevent infection as lungworm larvae
persist in earthworms for long periods of time. Using an effective
anthelmintic on sows prior to farrowing and then moving them
to clean pasture is helpful.

PARASITES

LUNGWORM 131
SWINE DISEASE MANUAL

Mange
Alternate names and abbreviations Pathogenesis
Sarcoptes, sarcoptic mange, acariasis, scabies Mites spread among pigs in close contact and invade the skin.
They burrow through the epidermis where eggs are laid. The
Definition mites are a strong irritant to the skin and initiate severe scratching
A skin disease caused by the sarcoptic mange mite, characterized and rubbing. In about 2 to 3 weeks, an allergic hypersensitivity
by marked pruritis and papules. develops in many pigs causing them to rub and scratch even more
vigorously, further traumatizing the skin.
Occurrence Mites tend to first colonize the skin of the inner surface of the ear.
Sarcoptic mange occurs in all age groups but is inapparent in From this site, they spread out over the body, tail, and legs. Ear
suckling pigs until signs and lesions have had time to develop. lesions first appear as small encrustations. These enlarge to form
Mange is present in all countries where pigs are raised in large plaque-like lesions. Constant rubbing and scratching irritate the
numbers. Sarcoptic mange is the most important ectoparasitic skin at many sites on the body and promote hyperkeratosis and
disease of swine. Sarcoptic mange occurs in many other species proliferation of connective tissue in the dermis. These lead to
but the mite Sarcoptes scabiei var suis is specific only for swine. thickening, wrinkling, and folding of the skin. Hyperkeratotic,
gray to white patches often form on the irritated skin. Reddened
Historical information macules and papules that develop on the skin of young growing
Sarcoptic mange, first reported about 100 years ago, was largely pigs are thought to be related to hypersensitivity.
ignored because it was widely believed that scratching was a
natural and normal behavior for pigs. Also, the economic loss Clinical signs
caused by the disease was not widely appreciated. Intensification Frequent rubbing and scratching are the usual signs and are often
of production after World War II and the economics of produc- more marked when the skin is warmed, presumably the result
PARASITES

tion focused attention on the avoidable losses caused by sarcoptic of increased mite activity. Equipment or facilities often have
mange. Development of better and more convenient acaricides polished surfaces from rubbing pigs. Decreased rate of growth,
has greatly improved control. inefficient use of feed, and low sow productivity follow. Although
morbidity is high, mortality from mange alone is unusual.
Etiology
The mite Sarcoptes scabiei var suis causes most mange in swine. Lesions
The mite is about 0.5 mm in length, gray to white, and just visible Young pigs that have developed hypersensitivity to mange mites
to the naked eye when on a black background. All stages of the display reddened macules or papules, especially over the rump,
mite (ova, larvae, nymphs, and adults) develop in the epidermis. flanks, and abdomen. Chronic mange affects relatively few older
The life cycle is completed in about 10 to l5 days. Depending on animals in the herd and is less pruritic. Lesions vary in extent,
temperature, humidity, and environment, mites and ova can sur- covering much of the body in advanced cases. In some cases, lesions
vive off the host for up to 15 days. may be confined mostly to the inner surface of the ears, where
Demodectic mange, caused by the mite Demodex phylloides, is hyperkeratotic areas often develop a brownish-red discoloration.
unrelated to sarcoptic mange and relatively unimportant in swine.
It is mentioned here because it occasionally causes dermatitis over Diagnosis
the snout, inside the thighs, and over the flanks. Occasionally, it Clinical signs and skin lesions are suggestive of mange; marked
involves the whole body and leads to excessive trimming or con- pruritis often helps differentiate mange from other diseases of
demnation at slaughter. Nodules caused by D phylloides have been the skin. Diagnosis is confirmed by identifying the mites in skin
mistaken for swine pox. scrapings or in exudates from the external auditory canal. Scrap-
ings from the inner surface of the ear usually reveal more mites
Epidemiology than scrapings from other sites. Scrapings can be wetted with
Mange in a herd usually develops after introduction of asymptom- 10% potassium hydroxide, cover-slipped, and examined under
atic swine carriers. Sarcoptes scabiei var suis is rarely if ever, trans- low-power magnification. A simple technique is to place the
mitted from other species. If swine are moved immediately into scrapings on a piece of black paper for a few minutes, carefully
quarters recently vacated by infested pigs, they are likely to become blow off the superficial debris and examine the site on the paper
infested. Spread is usually by direct body contact, often the result of for the small, light colored mites. Diseases that must be differ-
pigs crowding together for warmth or shade. Mites from the dam entiated from mange include parakeratosis, dermatomycosis,
often invade neonatal piglets within a few hours of birth. exudative epidermitis, pityriasis rosea, and photosensitization or
sunburn. Inspection of carcasses at slaughter may be useful both

132 MANGE
DISEASES CAUSED BY PARASITES

in diagnosis and in evaluating the efficacy of a mange control pro- are administered to every pig on the farm (adults, neonates, and
gram. An enzyme-linked immunosorbent assay for serological diag- growing pigs). Removal of bedding material, if present, and appli-
nosis of mange is available in some countries for herd diagnosis. cation of an effective acaricide to environmental surfaces is often
recommended at the same time injections are given. While erad-
Control ication can be an expensive task in the short term, it is reliably
Control programs should focus first on the breeding herd, which efficacious and requires only standard biosecurity procedures to
- if infected - inevitably transmits mites to the piglets. These pro- keep the mite from returning to the farm. Eradication of mange is
grams usually involve treatment of dams with external acaricide achievable in most production settings.
sprays or with an injectable avermectin a few days prior to enter- The avermectin class of parasiticides controls many external and
ing the farrowing facility. Many farms using this control strategy internal parasites. One should carefully follow the directions that
find it necessary to also treat growing pigs at 8 to 10 weeks of age. come with any product, as some require a lengthy withholding
Initiating an eradication program usually starts with culling of all period following their use.
severely infested breeding stock. Immediately following this, 2
consecutive doses of an injectable avermectin, 18 to 21 days apart,

PARASITES

MANGE 133
SWINE DISEASE MANUAL

Minor intestinal nematodes: Kidney worm, Nodular


worm,Thorny-headed worm, and Threadworm
Kidney worm necropsy by identifying ova in the urine. The first urine passed in
the morning often contains many ova.
Definition, occurrence, and historical information
The nematode Stephanurus dentatus is known as the kidney Control is possible through a breeding program that utilizes
worm, occurs only in swine, and affects all age groups, includ- only gilts and young boars. These are slaughtered after producing
ing fetuses. Pigs raised on moist, shaded pastures in tropical or one litter. This usually precludes their living long enough for
subtropical areas such as the southeastern US are most often ova of S dentatus to be passed in their urine. Eradication has
infected. Once the life cycle was determined, control became pos- been achieved on some production sites by this program and
sible via facility management and the availability of more effective by adopting management practices that preclude swine coming
parasiticides. However, kidney worms have not been eradicated into contact with the parasite on pasture. Placing feeding and
from the US and are maintained in feral pigs and anywhere there watering equipment on concrete (rather than on dirt) in a sunny,
are suitable outdoor habitats. well-drained location is a simple management practice that has
value in controlling the parasite as does the use of confinement
Etiology, pathogenesis, and epidemiology housing. Treating sows with an anthelminthic a few weeks prior
Adult kidney worms are 20 to 45 mm long and about 2 mm in to farrowing will usually prevent transplacental transmission of
diameter. They have black and white mottling related to inter- the parasite to fetuses.
nal organs and their contents. Their life cycle is lengthy, with a
prepatent period of 9 to 12 months, meaning the eggs are not Nodular worm
shed from pigs that have not yet reached breeding age (ie, adults).
Definition, occurrence, and historical information
Once a patent infection is established, ova are shed continuously
PARASITES

Oesophagostomum are frequently associated with disease in swine,


in the urine. Ova are thin walled, morulated, and measure 120
sheep, and cattle but only with host-specific species. Oesopha-
by 70 µm. In a warm, moist, shaded environment, ova can hatch
gostomum species, most commonly O dentatum and O quadrisp-
in 1 to 2 days; larvae are infective in 3 to 5 days. Under favorable
inulatum, are distributed worldwide but clinical parasitism
conditions, the larvae can survive in the environment for several
usually occurs only in pigs raised in open lots or pasture. All age
months.
groups of pigs are susceptible but pigs over 3 months of age may
Infection can occur by larval invasion prenatally across the pla- be more so.
centa, by skin penetration, by ingestion in feed, and perhaps by
ingestion of earthworms. Ingestion is probably the most common Etiology, pathogenesis, and epidemiology
route of infection. Larvae complete 5 developmental stages (L1- Adult Oesophagostomum worms are white to gray, 1 to 2 cm long,
L5) to reach maturity. Ingested or penetrating L3 larvae migrate and have slightly curved bodies. Ova are morulated, thin-shelled,
from the small or large intestine to various sites. Many larvae and about 70 by 40 µm. Ingested L3 larvae penetrate the mucosa
travel via mesenteric lymphatics or by portal venous blood to the of the cecum and colon, encyst for 6 to 20 days and molt to L4
liver where they migrate extensively for 2 to 4 months. The larvae larvae before returning to the intestinal lumen. Some L3 pene-
penetrate the liver capsule, cross the peritoneal cavity, and then trate deeply and cause marked inflammatory and fibrotic nodules
encyst in perirenal fat or near ureters. Here they establish fistu- visible on the serosa, with possible serosal adhesions and peritoni-
lous connections with the renal pelvis or ureters. When mature, tis the result. Repeated invasions and concurrent hypersensitivity
they discharge their ova into the urine after a 9- to 12-month reactions can contribute to severity.
prepatent period.
Severely infested pigs may have ill-thrift or diarrhea, but most
Most swine are marketed before patency can occur but affected infestations are asymptomatic. Adult worms reside in the intesti-
pigs will be unthrifty. Migrating larvae cause phlebitis, interstitial nal lumen and become patent as early as 3 weeks after ingestion of
hepatitis, and fibrosis in various tissues. Liver condemnations are the L3. Large numbers of ova are passed in the feces. Under favor-
frequent in infested slaughter-age swine. able conditions, the nonparasitic L1 and L2 stages can persist
on pastures for up to a year. Increased numbers of ova are passed
Diagnosis and control during the periparturient and lactation period, almost ensuring
Diagnosis is usually made at necropsy by observation of the that some of an infected sow’s piglets will be infected. Necropsy
abscesses and parasites at sites of localization. The extensively of affected pigs will reveal inflammatory nodules or necrotic
scarred, enlarged liver seen with severe infestations is rather debris present in the cecum and colon with nodules up to 20 mm
unique in swine. Diagnosis can be made in the live animal or at in diameter scattered in the mucosa and submucosa as well as
some apparent on the serosa. The lesions are easily confused with

134 MINOR INTESTINAL NEMATODES


DISEASES CAUSED BY PARASITES

dilated lymphoglandular complexes, which are common in the Diagnosis and control
colon of pigs with chronic bacterial enteritis. Oesophagostomum Diagnosis is usually made based on gross observations of worms
infestations are uncommon in most production systems. and sites of attachment at necropsy. Ova are quite characteristic
and are easily detected through laboratory fecal examinations
Diagnosis and control
Control is through avoidance of exposure to contaminated lots
Diagnosis is usually made on postmortem examination. Nodules
or pastures and to beetles, the usual intermediate hosts. Beetles
in and on the large intestine and the presence of adult parasites in
should be eradicated around feed storage areas and other sites
the lumen can easily be recognized. Lesions must be differentiated
accessible to swine. There is little data on treatment, but levami-
from abscessed lymphoglandular complexes, which tend to be very
sole has been recommended.
common in chronic colitis from various causes. Fecal examination
is also useful for diagnosis. The eggs must be differentiated from
grain mite eggs and ova of other Strongyloididae; differentiation Threadworm
from Hyostrongylus rubidus may require larval culture. Definition, occurrence, and historical information
Nodular worm infection is usually controlled by the use of Parasitism by the nematode Strongyloides ransomi occurs only in
anthelmintics. Deworming sows prior to farrowing will reduce pigs. The organism is widespread and has importance in warmer
exposure of the nursing piglets. In confinement operations, fre- climates. It can affect any age, including fetuses, but is more
quent removal of feces and good sanitation also reduce exposure common in nursing piglets. Losses in modern, indoor production
of piglets. In outdoor operations, lots and pastures that are heav- settings are relatively minor.
ily contaminated should be avoided. Pasture rotation, with tillage
Etiology, pathogenesis, and epidemiology
of land when not pastured, is helpful. Many anthelmintics are
Only asexual females of S ransomi are important in parasitism of
available for control.
swine. Free-living generations can produce progeny that are either
free-living (heterogonic cycle) or parasitic (homogonic cycle).
Thorny-headed worm

PARASITES
Free-living generations can alternate with parasitic generations.
Definition, occurrence, and historical information The asexual females are almost microscopic, about 3.3 to 4.5
Macracanthorhynchus hirudinaceus, literally the “thorny-headed mm long, and live in the mucosa of the anterior small intestine,
blood-sucking” worm, is limited to pigs and usually occurs in usually the duodenum. The ova are thin shelled, larvated, and
grow-finish pigs old enough to have had an opportunity to root are passed in the host’s feces. Infective larvae hatch in 1 to 2 days.
and forage, thus having access to the larval stages of beetles that They require warmth and moisture for survival and are usually
serve as an intermediate host for the parasite. Infection is rela- eliminated by desiccation, sunshine, and good sanitary practices.
tively common in pigs raised outdoors in southern states but can
Survival is enhanced by 3 key mechanisms: the ability of S
occur elsewhere sporadically.
ransomi to survive for some time in a nonparasitic form, the
Macracanthorhynchus hirudinaceus has an elongated, sac-like availability of 4 possible routes of transmission, and the ability to
body with no internal alimentary canal. It has a protrusible pro- achieve patency in piglets in only a few days.
boscis armed with about 6 rows of spiny hooks for attachment
Parasitic larvae may infest pigs by penetration of the skin (per-
deeply into the intestinal wall. The worms are 10 to 40 cm long,
cutaneous), ingestion in feed (oral), ingestion in colostrum
up to 9 mm wide, and are transversely wrinkled, slightly curved,
(transcolostral), or through direct invasion of developing fetuses
light pink in color and attenuated posteriorly. The brown ova are
(transplacental or perinatal). In all cases, the asexual female
large, up to 110 by 65 µm, have a 3-layered shell, and are resistant
reaches the mucosa of the small intestine, where many ova are
to environmental degradation.
produced and passed in the feces of the host.
The eggs are passed in swine feces and ingested by beetles, grubs,
Larvae sequestered in muscle or in the mammary fat of sows are
or other arthropods that act as intermediate hosts. Swine then
mobilized during the periparturient period and are transmitted
ingest these intermediate hosts. The eggs hatch in the intestinal
to piglets in the colostrum. This is the most common route of
tract of the pig and the worms attach there, usually in the ileum.
transmission to neonatal piglets. Larvae in colostrum arrive at the
The parasites develop by absorbing nutrients from the ingesta.
intestinal mucosa without migration.
Pigs with many parasites tend to become unthrifty from loss of
blood, localized inflammation at sites of attachment, the effect of Larvae that penetrate the skin of young pigs enter the blood
many localized lesions, and impaired nutrition. There are few or stream and are carried to the lungs. After breaking out into alveoli
no signs with light infections. Occasionally, a parasitized pig dies followed by tracheal migration to the pharynx, the larvae are
as a consequence of intestinal perforation followed by peritonitis. swallowed and reach the small intestine. Larvae taken into the
mouth penetrate the mucous membranes of the oral cavity and
after a similar migration, reach the small intestine.

MINOR INTESTINAL NEMATODES 135


SWINE DISEASE MANUAL

Adults that develop in the small intestine irritate the intestine, larvated ova can be identified by laboratory fecal examination
with the severity depending upon the number of parasites. The but must be differentiated from larvated ova of lungworms.
parasites cause a blunting and atrophy of intestinal villi, resulting Lesions may not be readily apparent on gross examination. On
in leakage of proteins into the gut and failure of the intestine microscopic examination of the small intestine, there is villus
to absorb nutrients and fluids effectively. These alterations lead atrophy and blunting. Adult parasites can be seen in tunnels in
to anemia, diarrhea, dehydration, and emaciation. Migration the epithelium of glandular crypts of the mucosa. In the lung,
of larvae through the lungs causes small hemorrhages as well as there is inflammation and thickening of alveolar walls, scattered
thickening and inflammation of alveolar septa that impair respira- hemorrhages, and perhaps migrating larvae. Neither hemorrhages
tion. In mild infections there are few or no signs. Most mortality nor larvae are likely to be numerous.
occurs in piglets less than 2 weeks old. Diarrhea, dehydration and
Avoidance of pastures with an environment favorable to the para-
unthriftiness can occur in pigs up to 3 months old.
site will reduce exposure of sows and piglets. Indoor housing and
confinement rearing, combined with good sanitation, will reduce
Diagnosis and control
the likelihood of threadworm infection. Avermectins given to the
Diarrhea in nursing pigs may suggest the possibility of infection
sow a week or two prior to farrowing will control transmission of
with S ransomi. However, all other common causes of diarrhea
larvae to fetuses.
must be excluded first, especially infections with rotaviruses,
coronaviruses, Escherichia coli, Clostridium perfringens, C difficile,
and coccidiosis. Strongyloides ransomi, if present, can be identi-
fied in mucosal scrapings or histologic sections. The thin-shelled,
PARASITES

136 MINOR INTESTINAL NEMATODES


DISEASES CAUSED BY PARASITES

Roundworm
Alternate names and abbreviations Epidemiology
Ascaris, worms, roundworms, large roundworms, ascarids, ascariasis Each adult ascarid can lay millions of eggs per day. The eggs are
extremely hardy and quite resistant to environmental degradation
Definition and disinfectants. Ova may be inactivated by intensive steam
Ascariasis is the infestation of swine by the roundworm, Ascaris cleaning or prolonged exposure to full sunlight. Ova can survive
suum, which can cause pneumonia, hepatitis, and ill-thrift. for years in the environment and can remain viable in swine efflu-
ent water for at least 14 months. Eggs are transported by infested
Occurrence pigs, insects, fomites, blowing dust, pig manure, and effluent.
Ascariasis occurs worldwide. All ages are affected but ascariasis is Eggs are often disseminated through the feces of purchased swine,
more severe in young, growing pigs. It is the most prevalent and including breeding stock. Once a location is infested, ascariasis
economically important internal parasite of swine. Infection and often persists in the pig population despite intensive measures
migration of ascarid larvae can occur in other animals, including to eliminate it. The extremely high number of eggs that are pro-
humans, but larvae usually do not develop to the adult stage in duced, the resistance of eggs to environmental degradation, and
the intestine of these other species. Migrating larvae occasionally the multiple means in which they can be disseminated contribute
infect cattle, calves, and lambs and can cause severe eosinophilic to their persistence on infected farms.
pneumonia.
Pathogenesis
Historical information Ascarids cause local inflammation in the intestine and compete
The ascarid of humans, A lumbricoides was once believed to be the with the host for nutrients. This adversely affects growth of pigs,
same as A suum; there is some truth to this notion as hybrids have especially those on marginal or poorly balanced diets. With heavy
been identified that can infect both humans and pigs. Knowledge infections, the most significant effect of the disease is a result of

PARASITES
of the life cycle is particularly important for this parasite, as effec- the migrating larvae causing lesions in the liver and lungs, includ-
tive economical control strategies must be tailored to each farm ing scarring of liver and interstitial pneumonia. Rarely, larvae are
site based on that knowledge. carried to ectopic sites (eg, the retina or brain), which may cause
signs of impaired vision or central nervous system lesions. Ascarid
Etiology impact also includes immunosuppression that affects the duration
Mature ascarids are 15- to 40-cm-long, thick-bodied, round and severity of other swine respiratory and systemic diseases and
worms. After a 7- to 8-week prepatent period, the prolific mature can influence the response of swine to some vaccines.
ascarids can lay almost 2 million eggs per day. Adults generally
survive in a pig for around 6 months before the gradual sponta- Clinical signs
neous expulsion of adults occurs over the next 6 months. Heavy Signs in young, growing pigs include unthriftiness, failure to gain
infections can result in hundreds of ascarids in the intestine of a weight, rough hair coat, pendulous abdomen, chronic paroxysmal
single pig. coughing, and, occasionally, abdominal expiratory dyspnea. Low
levels of ascarid exposure will stimulate development of active
Ascarid ova are thick-shelled, yellowish brown, almost spher- immunity. The most severe consequences of ascarid infection
ical, 50 to 80 µm by 40 to 60 µm, and are coated with a sticky occur when immunologically naïve pigs are placed into contam-
proteinaceous cover. Ascarid ova are ingested and hatch in the inated sites. Farrowing and nursery facilities often have a low
intestine. The larvae penetrate the mucosa and pass through the prevalence of ascarid egg contamination such that young pigs
bloodstream to the liver, lungs, and other sites. They reach the will have had little or no exposure to the parasite. Hence, these
liver 2 to 4 days after ingestion and are present in lungs within 10 animals are at risk of severe disease when subsequently placed in
days. Once arriving in the lung, they break into alveoli and bron- contaminated finishing barns. Severe, sometimes fatal, respira-
chioles, move up the mucociliary escalator, are coughed up to the tory disease may result in 7 to 14 days after naïve pigs of any age
pharynx, and are re-swallowed. These larvae reach the intestine are placed in facilities heavily contaminated with ascarid eggs.
14 to 20 days after initial ingestion. There they become adults Affected pigs are afebrile, “thump,” are gaunt, and are often mis-
over the next 6 weeks. There is a total prepatent period of approx- diagnosed as having bacterial or viral pneumonia. Other effects in
imately 8 weeks after ova are ingested before eggs are passed in heavily exposed gilts include delayed estrus, poor conception rate,
feces. Ova passed in feces require 2 to 4 weeks to mature in the pneumonia, or death. Severe liver insult can lead to stunting.
environment before they become infective.

ROUNDWORM 137
SWINE DISEASE MANUAL

Lesions Control
Scarring in the liver first appears by 7 to 14 days after exposure as A complete knowledge of the life cycle of ascarids and the infes-
scattered, gray to white “milk spots” (0.2 to 1 cm in diameter) vis- tation status of a herd or group of pigs is required to make timely
ible under the liver capsule. These lesions will eventually resolve and economical deworming decisions. All deworming programs
over 2 to 4 months; therefore, slaughter checks for liver lesions need to be designed to fit the specific situation, parasite load,
may not always be a reliable method to monitor for the presence and management practices present on each production site. One
or severity of ascarids in a herd if the infestation is occurring at a program does not fit all situations.
young age. In heavy infections, diffuse interlobular fibrosis may
Prevention of ascariasis is far better than treatment but may be
affect the entire liver and lead to its condemnation.
difficult to achieve on many production sites. Swine raised in
After their passage and molting in liver, larvae reach the lungs 7 confinement with a good ascariasis control program usually have
to 14 days after ova ingestion. There they break out of capillaries few ascarids. Some of the measures that confinement operators
into alveoli and cause small focal hemorrhages and an inflam- implement may include: deworming the sows during gestation
matory reaction that interferes with the clearance mechanism of (usually about a week before farrowing), washing the sows to
airways. Obstruction of smaller airways with atelectasis and/or remove parasite eggs prior to putting them in sanitized farrowing
emphysema (verminous pneumonia) can lead to secondary bacte- crates, early weaning of 2- to 4-week-old pigs (before ascarid eggs
rial infections that present as a suppurative bronchopneumonia. have become infective in the farrowing environment), installing
slatted flooring which minimizes contact between pigs and feces,
From the lungs, the larvae are transported or coughed into the
and using all-in, all-out production systems with thorough clean-
pharynx, swallowed, then reside in the small intestine without
ing between groups.
attachment or demonstrable lesions. Since ascarids have some
mobility, they can sometimes be found in the stomach or in the In modern swine production systems, many pigs are produced
major bile and pancreatic ducts at necropsy, and occasionally may in sanitary confinement systems that may be free of ascarid
obstruct the bile duct, leading to bile stasis and icterus. infection. When these pigs are placed in sanitized modern con-
PARASITES

finement finishers, deworming may not be warranted. However,


Diagnosis when naïve pigs are placed in contaminated grow-finish facilities,
Diagnosis of acute ascarid infestations requires postmortem they may develop mild to severe pneumonia as they ingest numer-
examination of several typically affected pigs that have been sacri- ous eggs from the environment. Feeding a continuous dewormer
ficed. Fecal flotations are not reliable during the first 6 to 8 weeks (pyrantel) for the first 30 days is a control option in this situation.
following infestation as the ova that were consumed by the pig Pigs in such conditions should be dewormed early after infesta-
have not yet become patent adults. This is an important concept tion and then at 8- to 10-week intervals to prevent additional
because most of the pathologic changes and clinical impact of egg contamination of the site. Modern confinement facilities
ascarid infestation in swine occur well before ova are passed or that recycle effluent water may increase the risk of ascarid ova
worms are grossly visible. Diagnosis of early infestations is made exposure. In these instances, routine deworming at 8- to 10-week
by observation of typical liver and lung lesions with confirmation intervals may also be warranted.
by histopathology of the liver and lung. Postmortem examination In operations that utilize pasture or open-lot housing, deworming
also permits the evaluation of concurrent diseases, including the sows before moving them to clean pasture is a good practice.
other agents of pneumonia, hepatitis and icterus, enteritis, and Pasture rotation will greatly reduce the exposure of swine to
other parasitisms. worm eggs, especially if the land is tilled when not pastured.
Diagnosis of more chronic infestations (infected greater than Anthelmintics should be selected on the basis of the entire spec-
2 months) and/or monitoring for parasitism is done by identifying trum of nematode species present on the farm, including ascarids.
eggs using fecal flotation examinations or by finding grossly visible A monitoring system should be established and followed. Some-
ascarids in the small intestine. Ascarids or their ova may not be times parasite problems change as a consequence of introduction
present in animals that have been recently dewormed, but scarring of other kinds of parasites, facility changes, or climatic changes
in the liver or hemorrhages in the lung may still be apparent. that influence the number and variety of parasites.

138 ROUNDWORM
DISEASES CAUSED BY PARASITES

Trichinellosis
Alternate names and abbreviations Larvae in the lymphatics soon gain access to the systemic circula-
Trichinella, trichina, trichinosis, pork worm tion. Many are destroyed by body defenses. Those that gain access
to muscle fibers penetrate them, usually 1 per fiber. A larva within
Definition a fiber often induces a change in the fiber that causes it to enlarge
Trichinella spiralis is a parasite that infects swine, other carniv- and act as a nurse cell. In 2 to 3 months, the larva reaches a length
orous animals, and people. The parasites are usually ingested of over 100 µm and is enclosed in a cyst-like structure in the mod-
through consumption of infected meat. ified muscle cell. Here it can survive for up to 11 years.
Larvae have a predilection for certain striated muscles, generally
Occurrence those that are normally quite active (eg, diaphragm, intercostal,
Most carnivorous, warm-blooded animals can become infected lingual, laryngeal, masseter, and ocular muscles). The larvae occa-
with this parasite. Animal sources of human trichinellosis include sionally localize in cardiac muscle fibers. Once encysted, larvae
consumption of meat that is eaten raw or has been incompletely must be ingested by another mammal for the cycle to be repeated.
cooked. In particular, meat from feral and domestic pigs, bears,
some sea mammals, and horses has been associated with significant Epidemiology
localized outbreaks in humans. Species implicated as sources of Survival of T spiralis is promoted by the wide range of animals
infection for swine include previously infected pigs, rodents, and that serve as intermediate hosts, by the long survival of larvae in
scavenged wildlife carcasses. Meat scraps from infected, slaughtered muscle fibers, and by the scavenging, cannibalistic nature of pigs.
animals that are included in uncooked swill are a significant source In swine, transmission often occurs when pigs scavenge Trichinel-
of infection for commercial swine in some countries. la-infected carcasses of swine, rats, or other animals. Transmission
The incidence of infection in swine is low in countries that also occurs when uncooked garbage containing pork or other
routinely inspect meat for T spiralis. In the US, food safety and meat scraps that harbor T spiralis are fed to pigs. Tail-biting of

PARASITES
eradication efforts have resulted in a nondetectable prevalence in infected pigs and ingestion of larvae in feces are other possible
commercial swine. sources of exposure.

Historical information Pathogenesis


Trichinellosis once caused outbreaks of severe illness in people, but Trichinella spiralis has a relatively low pathogenicity for swine but
control measures were developed and adopted by many pork-pro- can cause severe clinical signs in humans.
ducing countries in response to the problem. In many countries
including the US, T spiralis infection of swine is essentially erad- Clinical signs
icated from commercial swine production. In recent years, most Clinical signs are seldom observed in swine naturally infected
of the US human cases of trichinellosis are a result of eating game with T spiralis. Experimentally infected swine show signs of
meat (especially bear meat) and not from exposure to pork. intense muscle pain and have decreased weight gain, but most
recover and weight gain improves. In infected miniature swine,
Etiology hypergammaglobulinemia and eosinophilia have been reported.
Trichinella spiralis has 4 separately named strains, each with
minor anatomical differences; they may simply represent different Lesions
ecotypes of the same species. In swine, gross lesions are not usually visible unless there is white
streaking from calcification of affected muscle fibers. On micro-
Mature T spiralis females are 3 to 4 mm long and are found in the scopic exam, the encysted parasites are easily observed. If there
duodenal and jejunal mucosa. A fertilized female produces 500 to are degenerated or necrotic muscle fibers, an inflammatory reac-
1000 larvae over a period of 2 to 6 weeks. tion with many eosinophils may be present around them.
The parasite’s life cycle begins with ingestion of infected muscle
that contains encysted T spiralis larvae. Trichinae within muscle Diagnosis
fibers are liberated when cyst walls are exposed to gastric secre- The prevalence of trichinae in the US swine population is so low
tions after consumption. The larvae reach maturity in the small as to be reliably undetectable. In many countries, techniques used
intestine in about 4 days. They mate, and mature females pene- for surveillance include trichinoscopic examination of muscle
trate into the mucosa and liberate larvae in lymphatics. Some lar- (often diaphragm muscle) which appears to be one of the most
vae escape into the lumen of the gut and are eliminated in feces. likely sites to contain T spiralis larvae. In this test, the muscle
Recently voided feces may be infectious, but this is considered a
minor mode of transmission.

TRICHINELLOSIS 139
SWINE DISEASE MANUAL

is homogenized and enzymatically digested in order to release recommendation for cooking temperature for pork was decreased
the larvae from their cysts for efficient visualization by the tech- from 160°F (71°C) to 145°F (63°C).
nician. An enzyme-linked immunosorbent assay (ELISA) and
In addition to changes in the behavior of consumers and the
polymerase chain reaction assay are also available. Trichinella-free
pork industry, regulations have also been developed that govern
production certification in the US currently relies on an ELISA.
how ready-to-eat pork products must be processed to ensure the
destruction of T spiralis. Today, the Pork Quality Assurance pro-
Control gram promoted by the National Pork Board continues to encour-
In the US, an educational program was developed with the age good production practices that make T spiralis infection in
objective of emphasizing the need to thoroughly cook pork for swine unlikely. The program has been implemented by many
control of human trichinellosis. Concurrent with this program, producers; virtually all US pork processors require their suppliers
commercial pork production practices in the US changed, signifi- to be enrolled.
cantly reducing the opportunity for pigs to become infected with
the parasite. As a result, the incidence of human trichinellosis No effort should be made to treat animals suspected of having
decreased dramatically. In 2011, the United States Department trichinellosis, although they should be reported to state public
of Agriculture Food Safety and Inspection Service recognized health and animal health authorities.
trichinae to be of minimal risk in commercial US pork, and the
PARASITES

140 TRICHINELLOSIS
DISEASES CAUSED BY PARASITES

Whipworm
Alternate names and abbreviations Pathogenesis
Trichuris, trichuriasis After ingestion of larvated eggs, L3 larvae hatch and enter the
mucosa of the anterior small intestine. The larvae reside there
Definition temporarily, return to the intestinal lumen for a period of time,
Infestation of swine with the swine whipworm, Trichuris suis. and then deeply reinvade the mucosa or submucosa of the cecum
and colon. This penetration results in diffuse mucofibrinous to
Occurrence mucohemorrhagic typhlocolitis. Focal ulcerations may be com-
Trichuris suis infections occur in domestic and feral swine; infec- plicated by microorganisms that include Salmonella, Lawsonia,
tion has also been reported in some primates, including humans. Brachyspira, and Balantidium.
Although very uncommon in confinement-reared pigs, infection The inflammatory reaction around the parasites results in edem-
remains relatively common in swine raised on pasture or on dirt atous thickening of the wall of the gut and the formation of
lots. All age groups are susceptible, but most infestations occur inflammatory nodules around some parasites. In heavy infections,
in swine less than 12 months old. In adults, clinical signs can be a fibrinous pseudomembrane may form on the mucosa. Regional
induced when infection occurs in the face of severe stress. How- lymph nodes may be swollen.
ever, whipworms commonly infest adult pigs at a subclinical level
and serve as a major source of infection for other swine. All major Clinical signs
swine-producing countries have whipworm. Clinical effects are directly related to severity of infestation and
presence of concurrent diseases. Mild infestations are subclinical.
Historical information Signs include anorexia, mucoid or mucohemorrhagic diarrhea, dehy-
Whipworm infections were common in the past when more dration, and rarely death if severely affected. Signs are most apparent
swine were raised outdoors on pasture; the infection remains 2 to 4 weeks following exposure to contaminated facilities.

PARASITES
reasonably common today in swine reared in the same settings.
Rearing of swine in confinement housing has led to the elimina- Lesions
tion of whipworms as a significant problem for most producers. During early stages (1 to 3 weeks postinfestation), whipworms
In outdoor operations, newer anthelmintics have allowed for are not visible as they migrate within the large intestinal mucosa.
improved control over this once troublesome parasite. There is a mucoid to mucohemorrhagic typhlocolitis with
excessive mucus, fibrin, and blood on the mucosa. Lesions vary
Etiology in extent from focal to diffuse and in acute stages often resemble
Trichuris suis is the name of the whipworm in swine. Mature those of swine dysentery. The affected gut wall is thickened and
female whipworms are 6 to 8 mm long and males are about half edematous.
that length. The life span of adults is 4 to 5 months. The ante-
rior two-thirds of the parasite is quite thin and, when seen at Histopathology is required for diagnosis and will reveal the
necropsy, is buried in the mucosa of the intestine. The posterior migrating larvae. Later, mature whipworms become visible with
one-third is visibly thicker and usually seen protruding into careful gross examination. Worms often protrude from inflamma-
the intestinal lumen. However, worms are generally too small tory nodules in the gut wall, and purulent exudate may be visible
to be seen until at least 3 weeks postinfection. Microscopically, around them. Gentle traction on parasites will free them from
ova are approximately 65 by 25 µm, yellow to brown in color, the gut wall, revealing the thin anterior end buried in the mucosa.
double-operculated, and easily recognized. Ova passed in feces Dilated lymphoglandular complexes should not be mistaken as
become infective in 3 to 4 weeks. The prepatent period is 7 to 8 lesions of either whipworms or nodular worms. Inflammation of
weeks but clinical signs can be observed within 1 to 2 weeks of the cecal and colonic walls can be quite extensive and severe even
severe infestation. Ova production tends to be sporadic and there when only few adult worms are observed.
are times when there are few ova in the feces.
Diagnosis
Epidemiology Diagnosis is accomplished by necropsy of a typically affected
Although adult worms have a relatively short life span and lay ova pig. Mature parasites are easily found in the cecum or colon and
only sporadically, ova remain infective in most environments for can be identified by their size and whip-like form. Immature
up to 6 years. This long period of survival tends to perpetuate the worms are much smaller than adults but can cause severe lesions
species, even if pasture rotation is practiced. Adult swine tend to if numerous. For identification, they require careful observation,
harbor low levels of infection resulting in a continual source of perhaps with the aid of flotation over a dark background or use
ova contamination in the environment. of a magnifying lens or by histopathology. The severe lesions of
the immature worms closely resemble, and must be differentiated

WHIPWORM 141
SWINE DISEASE MANUAL

from, those of swine dysentery, salmonellosis, and proliferative Control


enteritis, any of which may be concurrently present. Control depends on avoidance of exposure to contaminated pas-
The presence of ova can be determined by laboratory fecal exam- tures or lots. Tillage of pastures or lots not being used can reduce
inations by 7 weeks after infestation, but clinical signs and lesions the number of eggs that survive. In outdoor operations, it is help-
will often be quite severe before ova are present. The double-oper- ful to treat the pregnant sows with an appropriate anthelmintic
culated eggs are distinctive, but ova production is often sporadic a week or two prior to farrowing and/or before moving them to
and there may be few or no ova in a single fecal sample. The clean pasture. This reduces exposure of the piglets to eggs. Raising
number of ova found on fecal examinations may not be a reliable swine in confinement, without access to soil, usually results in
indicator of the number of worms present. good control.
Not all anthelmintics have good efficacy against T suis, so a
careful evaluation of a product’s spectrum of activity is warranted
before incorporating it into a control program.
PARASITES

142 WHIPWORM
Section V

Miscellaneous

Atresia ani, Aural hematoma, Congenital tremors, Cystitis and pyelonephritis, Dermatosis vegetans......................145
Ectopic ossification, Epitheliogenesis imperfecta, Gastric ulcer ........................................................................................146
Hemorrhagic bowel syndrome, Hernias, Hydronephrosis, Hypogalactia and MMA...................................................148
Hypoglycemia, Megacolon, Mulberry heart disease and related conditions...................................................................149
Osteochondrosis, Pityriasis rosea, Porcine stress syndrome................................................................................................151
Prolapses, Pustular dermatitis, Rabies, Ringworm.................................................................................................................152
Shoulder ulcers in sows, Skin necrosis of piglets, Splayleg (spraddleleg), Sunburn and photosensitization.............153
Torsion and volvulus, Vestibular syndrome, Vices (tail biting, ear biting, flank biting, navel sucking).....................154

OTHER

143
144
OTHER
MISCELLANEOUS

Atresia ani
The congenital absence of a patent anus at birth is rather critical developmental stages in utero. The resultant megacolon
uncommon in swine. The cause is not known, but it may either may not be clinically apparent until several weeks of age.
have a genetic component or result from an insult to the fetus in

Aural (ear) hematoma


Aural hematomas are common lesions in pigs with large, depen- shaking is associated with mange, infestation with lice, or feed
dent ears. They are generally a result of some type of trauma to particles in the ears. Treatment or drainage of the acute (hema-
the ear followed by hemorrhage between the skin and cartilage toma) stage of the fluctuant ear lesion is not usually attempted,
(or within the cartilage). The subcutaneous clot of blood resolves nor is it likely to be successful. Segregation rather than treatment
over several weeks to months, in most cases leaving the pinna is the recommended intervention. Amputation of the ear is some-
thickened and misshapen. The inciting trauma often results from times performed in severe cases.
bites inflicted by penmates or from violent head shaking. Head

Congenital tremors
In this disease, some or all of a newborn litter of pigs have infections (eg, classical swine fever and other viruses), genetic
mild-to-violent tremors of part or all of their bodies. The trem- predispositions, or in utero exposure to certain chemicals.
ors are worse when piglets are active or stimulated and are often
Recently, experimental in utero infection of fetuses with atypical
absent while they sleep. If severe, the tremors will prevent nurs-
porcine pestivirus (a different pestivirus than classical swine fever
ing and affected piglets will starve. However, if piglets can nurse,
virus) reproduced typical clinical signs in newborn piglets. The
the condition can gradually resolve even though a mild tremor
affected piglets were born viremic and remained viremic indefi-
may persist well into finishing phase.
nitely. It appears this virus is quite widespread globally and only
This disease, recognized for over 100 years, usually occurs in gilt causes disease when it infects naïve or susceptible dams at breeding
litters, especially after introduction of a new boar. Subsequent or during gestation. Atypical porcine pestivirus can be detected
mating of the same boar and dam will result in normal, unaf- in boar semen, which is one likely mode of transmission. Once
fected piglets, suggesting that natural immunity occurs. A spe- infected, herds are generally immune and clinical cases disappear.
cific cause of congenital tremors remains elusive, but occurrence
Frequency of splayleg often increases during congenital tremor
of these “shaker pigs” has been associated with in utero viral
outbreaks.

OTHER
Cystitis and pyelonephritis
These conditions are usually the result of ascending bacterial Actinobaculum suis is present in the preputial diverticulum of
infections. Various bacteria, including Actinobaculum (formerly most boars. The organisms may be transmitted to the sow during
Eubacterium or Corynebacterium) suis and coliforms, have been copulation. Sows with cystitis or nephritis often have hematuria,
isolated from the lesions. The infections usually occur in older which is one of the earliest signs of infection, often within 3
sows and are a cause of sporadic deaths. Death often occurs weeks after breeding. This is followed by progressive loss of body
during or shortly after advanced pregnancy, a time of maximum weight.
stress on kidney function. Anything that interferes with adequate
These conditions are uncommon in boars.
water intake or the complete emptying of the bladder via urina-
tion may predispose the pig to urogenital infection.

Dermatosis vegetans
This hereditary defect originated in the Landrace breed. The lesions begin as papules and expand to form roughened plaques
defect can be expressed as foot deformities present at birth, or that soon become papillomatous. Most affected pigs die within
later as widely distributed expanding cutaneous lesions that 6 weeks. Many that survive have dyspnea related to interstitial
involve small to large areas of skin. Skin lesions are usually appar- pneumonia of giant cell type.
ent by the third week of life although some develop later. Skin

ATRESIA ANI, AURAL HEMATOMA, CONGENITAL TREMORS, CYSTITIS, DERMATOSIS VEGETANS 145
SWINE DISEASE MANUAL

Ectopic ossification of mesentery


Sows and boars over 3 years of age are occasionally found to entery is not known. Little significance is ascribed to the pres-
have ectopic ossification of the root of the intestinal mesentery. ence of ectopic ossification, but it may be involved in mechani-
The cause of these bony spicules radiating through the mes- cal injuries such as intestinal volvulus or accidents.

Epitheliogenesis imperfecta
This inherited condition is known to occur in at least 4 breeds nephrosis commonly accompany the condition. Fetuses with
and is characterized by the absence of discrete areas of skin, usu- extensive lesions may be aborted. Piglets born alive with extensive
ally over a part of the back or thigh. Lesions occasionally occur lesions usually die from bacterial invasion and septicemia. Minor
on the anterior surface of the tongue. Hydroureter and hydro- defects may heal.

Gastric ulcer
Alternate names and abbreviations • Hot weather, potentially related to decreased feed intake,
Stomach ulcers, ulceration of the pars esophagea, ulcer heat stress, or stress associated with limited access to water
• Out-of-feed events due to plugged or empty feeding systems
Definition • Chronic or acute disease (both of which may contribute to
In swine, gastric ulceration refers to the destruction of the normal anorexia)
epithelium of the pars esophagea (nonglandular region of the • Greater occurrence reported in barrows (versus gilts) and in
stomach). This ulceration can involve part or all of the entire area high-lean genotypes
of the pars esophagea. • Feeding rations with copper as a growth promoter, perhaps
without adequate zinc
Occurrence • Ad libitum feeding of whey or skimmed milk.
The lesions occur in pigs from weaning onwards. The prevalence • Diets high in wheat or cornstarch
is variable but can be quite high (5% to 90%) in some farms or • High levels of unsaturated fats in the diet, often accompa-
production systems. Clinical signs are usually noticed only in pigs nied by inadequate levels of vitamin E
8 weeks of age or older, including adults, with highest prevalence A primary infectious etiology has not been proven. Several agents
in pigs 3 to 6 months of age. Ulcers occur in all breeds, both have been associated with increased prevalence and severity of
sexes, and in most major swine-raising countries. ulcers, including Helicobacter species and Gastrospirillum spe-
OTHER

cies, but a causal role has never been established. In some severe
Historical information outbreaks of gastric ulcers in swine, porcine circovirus type 2 has
Ulceration of the pars esophagea was first reported in Illinois in been demonstrated within lesions. Gastric ulceration is a com-
1897. Subsequently, the lesion was identified in at least 30 other mon sequel to outbreaks of influenza A and porcine respiratory
countries. Although the ulcers are most often subclinical, death disease complex, particularly during the summer months.
losses can be severe.
Infectious organisms (Helicobacter pylori) and the use of non-
steroidal anti-inflammatory agents are strongly associated with
Etiology human gastric ulcers, which occur in the glandular portions of
The etiology is unknown but multiple risk factors have been iden-
the fundus and pylorus. When pigs are used as research models
tified, some of which are probably additive in their effect. Risk
for human ulcers caused by those risk factors, ulcers are induced
factors commonly mentioned include:
in the glandular mucosa. However, gastric ulcers in pigs typically
• The feeding of finely ground feed (less than 400 µm particle occur in the nonglandular pars esophagea, and the risk factors
diameter) associated with gastric ulcers in humans are not associated with
• Pelleted rations (perhaps due to the finely ground feed used typical gastric ulcers in swine.
when making pellets)
• Concentrated, nutrient dense diets that are low in fiber Epidemiology
• Stressors (eg, anxiety, fear, pain, fatigue, crowding, fasting, Both etiology and epidemiology are speculative. Epidemiology is
prolonged transportation, social stress from mixing with difficult to assess as it is likely to be multifactorial. Risk factors
unfamiliar pigs, poor air quality, etc) that appear important in one herd or production system do not
remain consistently important across others.
146 ECTOPIC OSSIFICATION, EPITHELIOGENESIS IMPERFECTA, GASTRIC ULCER
MISCELLANEOUS

Pathogenesis raised, roughened, somewhat nodular, usually bile stained, and


Irrespective of the risk factor that triggers formation of a gastric sometimes fissured with hyperkeratosis. Ulcers vary greatly in
ulcer, the pathogenesis of the lesion has been well characterized. size, shape, number, and depth. Large ulcers are often deep and
Gastric ulcers result from a disruption to the pH gradient in the have smooth, rolled, and raised borders. Sometimes the ulcerated
fluid contents of the stomach that normally exists between the area involves the entire pars esophagea and may partially obstruct
proximal end of the stomach (pars esophagea; relatively higher the esophagus. The terminal part of the esophagus may also be
pH) and the distal end (pylorus; relatively lower pH). Although hypertrophied. In animals that have recovered from deep ulcers,
causal factors are not clearly defined, when gastrin-stimulated a puckered or stellate scar often remains.
acid secretion becomes excessive, the overly fluid, very low-pH Necropsy of pigs that die suddenly will have pallor of the skin
stomach content moves to the region of the pars esophagea. The and internal organs and tissues. There may be large blood clots
stratified squamous epithelium in this region becomes hyper- in the stomach and/or dark, unclotted blood mixed with ingesta
keratotic in response to the abnormally acidic environment and, and feces throughout the intestine. The intestinal and/or colonic
if the insult persists, ulcers form. Preulcerative hyperkeratosis is contents are often very dark, black, or tar colored. The entire
extremely common in growing swine, probably because of low pars esophagea may completely ulcerate down to the level of the
fiber diets. muscular wall, giving the false impression of intact glandular
The pars esophagea undergoes hyperkeratosis and parakeratosis, mucosa. Complete destruction of the pars esophagea is easy to
creating fissures that expose the underlying lamina propria. This miss at necropsy; the color of that area may be a uniform gray and
progression from hyperkeratosis to the ulcerative process can without hemorrhage. The absence of clotted blood in the stomach
occur very quickly, often within 24 hours of the insult, with does not rule out gastric ulcer. Careful examination of the pars
significant ulcers, erosions, and hemorrhagic lesions possible in esophagea is warranted since hemorrhage may be intermittent.
5 to 7 days. If the insulting acidity persists or animals become Death caused by gastric ulcer does require a large amount of clot-
anorexic, part or all of the stratified squamous epithelium may ted blood to be found in the stomach or intestines at the time of
slough off, leaving one or more deep ulcers. Blood vessel walls of death.
various sizes within the lamina propria and submucosa may also Differential diagnoses include other causes of hemorrhagic
become eroded and hemorrhage will then occur. If blood loss is diarrhea such as porcine proliferative enteritis, swine dysentery,
slow and/or intermittent, melena, generalized pallor, and anemia salmonellosis, and whipworms.
may be observed. If a few large vessels are eroded, the animal may
die from an acute massive hemorrhage. Control
Ulcers can resolve if they are not severe, although resolution of For pale or anemic pigs, treatment is of questionable efficacy but
lesions occasionally results in stricture of the terminal esophagus may include Vitamin K, hematinics, segregation, and supportive
or pars esophagea. therapy including oral protein supplements. Access to fiber (hay
or pasture) or products that reduce gastric emptying will some-

OTHER
Clinical signs times aid in recovery.
Preulcerative hyperkeratosis of the pars esophagea is a common Prevention is based on correcting those risk factors suspected
precursor to actual ulceration. However, hyperkeratosis does not of contributing to ulcer formation. Minimizing stressors and
appear to influence feed consumption, nor is it noticeable clinically. preventing out-of-feed events are always warranted. Altering the
In fact, many swine are subclinically affected, evidenced by the high ration to minimize the effects of some of the possible risk factors
prevalence of lesions in apparently normal pigs at slaughter. With- is helpful. Changing to a more coarsely ground feed (less than
holding feed can cause some preulcerative lesions to erode in a rela- an average diameter of 700 µm) and changing from a pelleted
tively short time (as little as 2 to 3 days), which may initiate clinical diet to a meal diet is often recommended. The quality of the
signs. The clinical effects of ulcers are usually related to blood loss. feed ingredients should be improved if some of the feedstuffs
Acutely affected pigs may be found dead with carcass pallor. Others were stored for long periods, are of poor quality, or contain
may have black tarry feces (melena), anemia, and generalized pallor. mycotoxins. Efforts to decrease heat stress in warm weather are
Other signs that can sometimes be observed include anorexia, beneficial. The successful control of some of the severe swine
grinding of the teeth, or unthriftiness. diseases, especially porcine circovirus type 2, influenza A, and
Actinobacillus pleuropneumoniae has decreased the losses attrib-
Lesions utable to gastric ulcers on some swine farms.
Typical gastric ulcers occur only in the nonglandular region
(pars esophagea) of the stomach at the junction of stomach and
esophagus. Pigs that are in the early stage of developing ulcers have
an altered pars esophagea, which instead of being snowy white, is

GASTRIC ULCER 147


SWINE DISEASE MANUAL

Hemorrhagic bowel syndrome


Hemorrhagic bowel syndrome refers to a condition whereby Hemorrhagic bowel syndrome should be differentiated from
previously healthy growing or finishing pigs die acutely, with partial or complete intestinal torsion or intestinal volvulus, which
bloating and pallor at time of death. The condition was originally are common causes of sporadic death. An acute hemorrhagic gas-
described in whey-fed pigs as “whey-bloat,” but now often refers tric ulcer usually has very dark or black blood in the intestine and
to sporadic deaths of individual pigs that meet the following lacks intestinal dilation. The hemorrhagic form of ileitis may have
diagnostic criteria: blood clots and/or segmental thickening of intestinal mucosa.
Acute enterotoxemia with intestinal congestion associated with
• There is carcass pallor and the abdomen is distended by
hemolytic Escherichia coli or Salmonella is often a differential in
abdominal viscera.
nursery pigs.
• The small intestine is diffusely dilated, thin-walled, and
distended with unclotted brownish blood, watery fluid, and The actual cause of hemorrhagic bowel syndrome is unknown
gas; the large intestine may be similarly affected or relatively but anecdotal risk factors include out-of-feed events and other
normal. circumstances that foster feed engorgement (eg, commingling,
• The stomach is normal in appearance and has normal weather events, etc), liquid feeding, and feeding of whey prod-
contents. ucts. Diets higher in fiber may help ameliorate the occurrence of
The diagnosis is by default and is made only after ruling out other hemorrhagic bowel syndrome and other intestinal accidents.
causes of sudden death and intestinal hemorrhage by postmortem
examination, appropriate laboratory testing, and histopathology.

Hernias (inguinal and umbilical)


Both inguinal and umbilical hernias occur sporadically in swine. strangulation of the intestine within an inguinal hernia can occur,
Inguinal hernias, where loops of intestine can pass through an but the most significant effect of the condition is the economic
incompletely closed inguinal ring, are believed to be heritable discount applied to the pig at time of slaughter. Surgical repairs
to some extent but heritability is unlikely to be associated with a have been described that are rapid and very successful.
single gene. The overall incidence of inguinal hernias in a normal
swine population is low unless mating to a boar that is genetically Umbilical hernias occur in both sexes and tend to develop as a
predisposed to pass the risk on to his offspring. result of poor umbilical cord management in neonates, chronic
navel infections, or navel sucking by penmates. The condition is
Inguinal hernias can be bilateral or unilateral, with most not thought to be inherited. Prevention is achieved by practicing
OTHER

occurring on the left side. Protrusion of intestines through the excellent hygiene in the farrowing house along with the appro-
inguinal canal can follow castration of pigs when an inguinal priate use of antimicrobials. Surgical repair is possible but rarely
hernia is not detected prior to surgery. In pigs left uncastrated, practical.

Hydronephrosis
It is common to find variably sized, sometimes large, fluid-filled observed during postmortem examinations conducted for unre-
cysts in the kidneys of healthy swine. This incidental finding is lated conditions or at slaughter.

Hypogalactia and Mastitis, Metritis, Agalactia (MMA)


Hypogalactia and MMA usually occur within 3 days of farrowing Dams usually recover within a few days to several weeks if
and are characterized by inadequate milk production. Although the inciting cause is effectively and rapidly managed. Piglets
hypogalactia is the most consistent sign, mastitis from bacterial are at high risk of dying from starvation unless cross-fostered
infection, fever, vaginal discharge, listlessness, weakness, anorexia, or provided with supplemental milk replacer. Although the
sternal recumbency, and refusal of the dam to permit nursing by cause(s) is unknown, many risk factors have been associated
the piglets may be present in affected sows. with hypogalactia. These include lack of nursing stimulation

148 HEMORRHAGIC BOWEL SYNDROME, HERNIAS, HYDRONEPHROSIS, HYPOGALACTIA AND MMA


MISCELLANEOUS

(too few pigs, or pigs are small and weak), bacterial metritis or bacteria, especially Escherichia coli, may be the cause of many
mastitis with endotoxemia, errors in ration formulation or poor cases of mastitis. Suppression of prolactin by endotoxin from any
feeding management, confinement with little opportunity for source results in less than normal milk production.
exercise, constipation, obesity, moldy feeds (ergotism), and poor
The prevalence of MMA has decreased markedly in the past
sanitation in the farrowing environment. The condition is difficult
several decades with the use of modern sanitary practices and
to eliminate completely from farms; however, if it occurs any
the routine use of perforated, dry flooring. Most problems with
more than sporadically, a comprehensive evaluation of all aspects
hypogalactia can be solved through management with careful
of gestation and farrowing management and nutrition should be
attention to sanitation, nutritional formulation, and feeding
undertaken to help identify the root cause or main risk factors.
management.
Mastitis, often clinically undetected, is likely responsible for many
cases of hypogalactia. Endotoxin from various Gram-negative

Hypoglycemia in neonatal piglets


Hypoglycemia is a common cause of death in neonatal piglets. followed by paddling, frothing at the mouth, coma, and death
Glycogen reserves become depleted when piglets are starving, if no intervention occurs. If chilling occurs, the piglets should
chilled, competing for limited number of functional nipples, be put in a warm environment with a supplemental heat source
or when inadequate milk is available. Chilling may occur if the such as heat lamps. If the piglets are not receiving an adequate
effective temperature in the piglet sleeping area is less than 95°F amount of milk, cross-fostering or milk supplementation can
(35°C) during the first week of life. be beneficial. Porcine milk replacers, bovine colostrum, or
evaporated milk diluted equally with water can be used for
Signs of hypoglycemia include loss of condition, weak vocaliza-
supplemental nutrition until piglets are able to consume nutri-
tion, faltering gait, cold skin, and recumbency. These signs are
ent-dense solid food.

Megacolon
Enlargement and dilation of the colon with feces is usually the causing constriction of the rectum. The outcome is restricted
result of rectal stricture, atresia ani, or, less frequently, colonic fecal flow with intermittent defecation or total obstruction,
atony. Atresia ani leads to megacolon in pigs up to 4 weeks of age. causing the colon to dilate with accumulated feces while the pig

OTHER
Postnatal rectal strictures are a frequent sequel to rectal prolapse. slowly wastes away.
Damaged rectal tissues from prolapse - irrespective of whether
Colonic atony with patent rectum has been reported as a result of
the prolapse has reduced on its own, been surgically repaired,
feeding lupine-seed meal. The neural control of tone and peristal-
traumatized, or chewed off by penmates - can heal with fibrosis,
tic movement is lost, causing the colon to simply dilate with feces.

Mulberry heart disease and related conditions


Vitamin E and selenium responsive myocardial damage (uncom- noncollapsing lungs, and visible hemorrhages on the surface
pensated oxidative metabolic stress) can present with variable of the heart that tend to occur in alternating areas of necrosis
clinical signs, though 3 closely related, overlapping syndromes are and hemorrhage, creating the lesion for which the condition is
most commonly associated with the condition. named. Straw-colored fluid is often present in the pleural cavity,
and the lungs are edematous. Typically, there are hemorrhages
The most frequent presentation is mulberry heart disease,
throughout the myocardium. Microscopically, the affected
which is clinically manifested as sudden death in fast growing,
areas of the heart will show capillary hemorrhage and swollen,
previously healthy young pigs, usually a few weeks to 2 months
fragmented, and sometimes mineralized muscle cells. There
of age. However, in some cases, death due to mulberry heart
are degenerative changes in the arteriole walls at many sites.
disease may be preceded by dyspnea, recumbency, pallor, and
Although the mechanism of pathology is not clear, most cases of
leg paddling. At necropsy, it is common for these pigs to have
mulberry heart disease occur when vitamin E is low. However,
hydropericardium (often accompanied by soft fibrin clots),

HYPOGLYCEMIA, MEGACOLON, MULBERRY HEART DISEASE 149


SWINE DISEASE MANUAL

mulberry heart disease does occur in pigs with seemingly not available, severe cellular injury and death can occur. This
adequate levels of vitamin E in tissue or serum, suggesting explains the severe tissue damage that is associated with all 3 clin-
that other oxidative stress or antioxidants may be involved. ical conditions.
Supplemental vitamin E can sometimes ameliorate death loss.
Feeds high in the concentration of polyunsaturated fatty acids,
Hepatosis dietetica is a more rarely encountered presentation of copper, vitamin A, or mycotoxins can either denature vitamin
the disease, particularly since the allowable limit of selenium sup- E or make it less bioavailable. Grains from soils deficient in sele-
plementation in livestock feed was raised to 0.3 ppm. Although nium, or selenium antagonists in mixed feeds can result in feeds
sudden death is a common occurrence in hepatosis dietetica, low in selenium. Both vitamin E and selenium are important
death may be preceded by weakness, pallor, dyspnea, and ill-thrift. metabolic antioxidants. Besides the syndromes described below,
In this syndrome, irregular focal-to-large areas of necrosis and pigs deficient in vitamin E and/or selenium may be more suscep-
hemorrhage are seen in the liver, giving its surface a mottled or tible to other diseases.
speckled appearance; the liver may appear swollen, and the gall
Diagnosis of these syndromes is made on the basis of clinical pre-
bladder often is edematous. Myocardial necrosis and pulmonary
sentation, gross lesions, and microscopic lesions in heart, liver, or
edema may also be present. Microscopically, much of the liver
muscles, and analysis for vitamin E and selenium levels in the liver
parenchyma will have characteristic massive coagulative necrosis.
or serum.
Sinusoids in affected areas may be distended with blood but
occasionally the necrotic areas will be ischemic. Prudent supple- For prevention or treatment of a deficiency, pigs can be injected
mentation with selenium will usually limit further death loss due with vitamin E and/or selenium and tissue levels will rapidly
to hepatosis dietetica. Hepatosis dietetica may be present along increase. Also, prevention is possible through appropriate supple-
with mulberry heart disease or white muscle disease but can also mentation of feed or drinking water. Sows injected in late gesta-
occur in isolation. tion give birth to pigs with increased levels of both compounds.
The most infrequent manifestation of the condition in pigs is The acute manifestation of the 3 syndromes described above may
called white muscle disease, which appears to be much more com- not always be completely responsive to supplemental vitamin E
mon in calves, lambs, and chickens than in swine. Skeletal muscle and/or selenium administration. For example, mulberry heart
pallor or streaks of white, gritty mineralization are observed, disease appears to be more responsive to vitamin E compared to
particularly in the longissimus dorsi muscle or ham muscles. His- hepatosis dietetica, and white muscle disease seems to respond
tologically, there is loss of myocyte structure with vacuolation, better to supplemental selenium. However, even when mulberry
fragmentation, and mineral deposition in type I fibers. Affected heart disease occurs in the face of normal vitamin E levels, deaths
pigs present with ataxia, weakness progressing to paralysis and can usually be prevented by providing additional vitamin E, often
recumbency, and often death. Hemorrhage does not feature sig- at levels that far exceed those recommended by the National
nificantly in the skeletal muscle lesions nor is there evidence that Research Council. Supplementation with vitamin E using highly
lesions of mulberry heart disease and white muscle disease tend to bioavailable parenteral or oral forms is quite safe and effective.
OTHER

occur simultaneously in affected pigs. Selenium levels should be verified as adequate but excessive sele-
nium supplementation should be avoided because of the poten-
The underlying pathophysiological mechanisms that result in
tial for toxicity.
lesions of mulberry heart disease, white muscle disease, and
hepatosis dietetica are not well understood but it is thought they In outbreaks, feed content and quality as well as feed storage
are associated with the formation of free radicals in tissue and an conditions should be examined carefully to determine if feed
inability of the pig to scavenge them, resulting in “uncompen- quality has been compromised. Improper feed storage, high
sated oxidative metabolic stress.” Free radicals are highly reactive copper levels, high fat levels, and poor-quality feed constituents
molecules generated during normal oxidative metabolism which can result in destruction of vitamin E in a feed that has otherwise
are typically neutralized by the cell using antioxidant scavengers been adequately formulated. Pigs on pasture usually get enough
such as superoxide dismutase, and vitamins E and C. Selenium vitamin E and selenium unless the soil is deficient in selenium.
has an essential involvement in glutathione peroxidase activity, Small piglets deficient in vitamin E are quite susceptible to iron
hence its perceived role in the disease process as well. Unscav- toxicity when injected with iron dextran for anemia prevention.
enged free radicals induce damage to cellular proteins, membrane Deaths occur with myocardial lesions that closely resemble those
lipids, and nucleic acids. If adequate free radical scavengers are of mulberry heart disease.

150 MULBERRY HEART DISEASE


MISCELLANEOUS

Osteochondrosis
Osteochondrosis, also referred to as dyschondroplasia, is a gener- Despite much research, the basic cause(s) is unknown. However,
alized skeletal disease where the cartilage in physeal or epiphyseal the clinical expression of lameness is related to the presence of
growth plates fails to properly ossify. The condition often occurs one or more risk factors. Dyschondroplasia has been attributed to
in rapidly growing pigs approaching market weight or breeding rapid growth and early excessive weight-bearing pressure on carti-
age. The disease is characterized clinically by abnormal gait or lage, but is usually accompanied by additional risk factors. Genetic
lameness with characteristic pathologic lesions in cartilage and influences for rapid growth are believed to play some role. Other
bone. Lesions of the articular surfaces of joints include corru- possible risk factors for disease expression include nutritional
gations and ulcerations of cartilage, separation of cartilage from deficiencies, flooring and housing that induce trauma, concurrent
underlying bone, and development of cartilaginous cysts. Physeal joint infections, and lack of exercise. The number of pigs with
cartilaginous lesions include fractures between the diaphysis and lesions is often much higher than the incidence of lameness would
epiphysis, or islands of cartilage within epiphyseal or diaphyseal suggest. Although some clinically affected pigs recover, most
bone. Lesions are often bilateral and symmetrical. Common sites affected gilts and sows are culled because of the time required for
of lesions in joint cartilage include the medial femoral condyle, healing and the overall poor response to therapy.
humeral condyle, humeral head, glenoid of the scapula, distal
ulna, and lumbar vertebrae. Common sites of physeal lesions
include distal ulna and femur, head of the femur or humerus, and
ischial tuberosity.

Pityriasis rosea
Pityriasis rosea is a dermatitis usually seen in 4- to 12-week-old often have healed centers. Lesions heal in about 4 weeks without
pigs and is characterized by 1- to 10-cm-diameter, raised, red- intervention. Pityriasis rosea is easily diagnosed by gross exam-
dened, ring-shaped lesions on the skin. Lesions first develop on ination and does not require any treatment. The condition is not
the skin of the ventral abdomen but occasionally start in other pruritic and seems to have no apparent effect on the health or
areas. The cause is unknown. Papules first develop on the ventral growth rate of affected pigs. Nearly all affected pigs recover spon-
abdomen and inner thighs. These early lesions expand to form taneously and completely.
circular, coalescing lesions with an expanding rim. Older lesions

Porcine stress syndrome

OTHER
Porcine stress syndrome, sometimes called malignant hyper-
excessive amounts of fluid. The meat from pigs with this condi-
thermia or transport myopathy, is a complex, genetically linked
tion is referred to as pale, soft, exudative pork. In the marketplace,
myopathy usually triggered by stress or excitement. It can also be
shoppers discriminate against pork with these characteristics.
triggered by exposure to several different anesthetics, including
halothane, and by depolarizing muscle relaxants. Signs that can The basic abnormality is an inherited defect in the mechanisms
appear include tremors of the tail, back, or leg muscles, muscle for the uptake, storage, and release of calcium in muscle fibers.
rigidity, inability to walk, respiratory distress, hyperthermia, Efforts to eliminate porcine stress syndrome through genetic
blotchy dermal hyperemia, acute right heart failure, and death. selection have led to success in identification and elimination of a
stress gene by virtually all large, commercial swine genetics compa-
Postmortem lesions include early, rapid, and complete rigor mor-
nies. A polymerase chain reaction test is widely available for iden-
tis, pulmonary edema, and pale soft musculature that often con-
tifying hetero- and homozygous carriers of the halothane gene;
tains hemorrhages. When slaughtered, swine with porcine stress
both types of carrier animals should be culled when identified.
syndrome produce carcasses that are blanched, wet, and may drip

OSTEOCHONDROSIS, PITYRIASIS ROSEA, PORCINE STRESS SYNDROME 151


SWINE DISEASE MANUAL

Prolapses (rectal, vaginal, and uterine)


Prolapse of rectal or vaginal tissues (including prolapse of the situation, effort needs to be directed towards rapid identification
uterus) usually occurs sporadically in swine, but on rare occasions of the triggering factor(s). The response to individual treatment
these conditions can also appear as an outbreak or cluster of cases. of prolapses is markedly improved by timely intervention within
All age groups are susceptible to prolapses, but occurrence in a few hours of their occurrence.
suckling piglets is quite uncommon.
Prolapse of the vagina or rectum sometimes occurs as a conse-
Prolapse of the rectum is usually the result of increased abdominal quence of flaccidity and relaxation of the birth canal in sows close
pressure from piling (for warmth in cold weather or due to stress to parturition, perhaps exacerbated by trauma. Prolapse of the
during transport), severe coughing, or straining and trauma at par- vagina usually occurs first and may then be followed by prolapse
turition. Rectal prolapse has also been associated with the straining of the rectum. Genetic factors have been shown to contribute
and anal irritation that accompany some severe enteric diseases, to prolapses of the vagina and uterus in some breeds, but more
especially salmonellosis. The feeding of whey, brewer’s grains, or likely risk factors include trauma (bite wounds), severe coughing,
low-fiber diets has preceded some outbreaks. Another common or straining caused by constipation, cystitis, vaginitis, or urethral
risk factor that has been suggested is the presence of mycotoxins obstruction.
(especially zearalenone) in the feed or bedding. Estrogenic myco-
Outbreaks of uterine prolapses in periparturient sows, which have
toxins and phytoestrogens can cause vulvovaginal edema and irrita-
seemingly increased in recent years, are both a welfare concern
tion in prepuberal gilts or sows. These compounds are considered
and a substantial contributor to sow mortality. Although many
a risk factor for rectal strictures when more obvious risk factors
risk factors and contributors have been proposed, none have been
are not present. Rectal stricture is a common sequel to prolapse of
proven as causal. Factors investigated include exposure to estro-
the rectum.
genic mycotoxins, improper calcium and phosphorus status, inad-
The condition can be repaired surgically by amputation or, if the equate vitamin levels, specific pig genotypes, sow body condition,
tissue is not overly traumatized, by replacement followed by use and increased litter size. Since no specific cause has been reported
of a purse-string suture. When prolapses occur in an outbreak with convincing evidence or reproduced experimentally, efforts to
elucidate the cause and risk factors continue.

Pustular dermatitis
Pustular dermatitis is usually observed only in nursing piglets. pustules soon rupture and are replaced by dark brown to black
It is caused by a beta-hemolytic, Lancefield type C Streptococcus crusts. The streptococci are easily isolated from pustules. The
infection. Pustules appear in the inguinal area and inner surface piglets usually recover spontaneously. Recovery may be hastened
OTHER

of the rear legs but occasionally occur at other locations. The by treatment with an appropriate antibiotic.

Rabies
Rabies is a highly fatal viral disease that occurs in swine bitten 4 days of the onset of clinical signs. Furious type rabies seldom
by an infected carrier animal. The incubation period is quite occurs in swine. There are no gross lesions. Microscopic lesions
variable, reportedly 9 days to 4 months. Clinical signs are similar are typical for viral encephalitis but can be quite variable. The
to those described in other species and include sudden onset typical rabies viral inclusions are often not present. Diagnosis is
of incoordination, prostration, chewing movements, excessive usually confirmed by fluorescent antibody testing on brainstem
salivation, inability to squeal, muscular spasms and tremors, and and cerebellum sections.
perhaps twitching of the nose. Death usually occurs within 3 to

Ringworm
Ringworm (fungal infection) occurs occasionally in swine and Lesions can occur anywhere, but on older swine they are usually
may affect any age group. Although cases are often sporadic, seen on the neck or behind the ears. Ringworm lesions begin as
occasional outbreaks affect many animals, especially sows. Most brown expanding areas a few centimeters in diameter but eventu-
ringworm is caused by the fungi Microsporum nanum or Tricho- ally may enlarge to 5 to 10 cm in diameter. Mature lesions have a
phyton verrucosum. central, brown crust; hair loss is minimal or absent and lesions are
nonpruritic.
152 PROLAPSES, PUSTULAR DERMATITIS, RABIES, RINGWORM
MISCELLANEOUS

In adult swine, ringworm must be differentiated from sarcoptic lesions. Ringworm is usually self-limiting, but lesions may require
mange, especially if lesions are behind the ears. In young growing months for healing to occur. Lesions are most common during
pigs, ringworm must be differentiated from pityriasis rosea and the winter months. Ringworm can be treated but is typically left
exudative epidermitis. The diagnosis of ringworm can usually to resolve on its own. Ringworm should be considered conta-
be confirmed by microscopic examination of skin scrapings or gious to people.

Shoulder ulcers in sows


Thin sows lying on hard floors in farrowing or gestation crates are more likely to develop and are more severe during the 3 weeks
may develop ulcers over the spine of the scapula. Contributing after farrowing. The ulcers are the result of ischemia from pressure
factors include poor body condition, lack of adequate bedding or on the skin leading to skin necrosis over the spine of the scapula.
mats, prolonged recumbency while farrowing or nursing piglets, Control measures including maintaining an appropriate level of
high humidity or wetness (often from coolers used during hot body condition (fat) in sows, use of bedding or mats, and assuring
months), and reduced activity because of confinement. Lesions crates are of adequate size to allow frequent repositioning.

Skin necrosis of piglets


Young pigs on concrete or other hard floors often develop pres- with Streptococcus species, Staphylococcus species, or Trueperella
sure-related lesions on knees, coronets, or other sites over bony pyogenes, with metastasis to joints or other sites. Losses can be
prominences. Although lesions may initially appear insignificant, substantial. Prevention is achieved through good sanitation, pro-
they can become infected and lead to bacteremia, most frequently vision of good quality flooring materials, and use of floor mats.

Splayleg (spraddleleg)
This abnormality of neonatal piglets is characterized by lateral congenital tremors. Recently, this condition has also been
extension of the hind legs accompanied by an inability to adduct associated with in utero infection with atypical porcine pestivirus,
the legs; the front legs may be variably affected. The principle one of the causes of congenital tremors (myoclonia congenita).
lesion in affected muscles is myofibrillar hypoplasia, related in
If affected piglets are helped to nurse and protected from acciden-
part to delayed development and in part to degenerative change.
tal injury by the sow, many will recover in 1 to 2 weeks. Tying the
Anecdotal or speculated causes or risk factors include: genetic front legs or the back legs loosely together with sticky tape will

OTHER
predisposition, slippery or sloped floors, porcine stress syndrome increase the likelihood of survival.
in the parents, dietary deficiencies, low birth weight, and

Sunburn and photosensitization


Sunburn is injury to the skin caused by excessive exposure to the Photosensitization should be differentiated from sunburn. Pho-
ultraviolet rays of sunlight. Pigs that are unaccustomed to direct tosensitization occurs only in areas of white skin and is a result
sunlight may be sunburned if not exposed gradually to their of exposure to a photodynamic agent (via ingestion, injection,
outdoor environment. The white or light-colored breeds are more or skin contact) followed by exposure to sunlight. Sunlight
severely affected. Although suckling and weanling pigs are most includes wavelengths that activate the photosensitization process.
likely to be affected, all age groups are susceptible. The ears and Photosensitizing agents are present in some pastures (alfalfa,
back receive more direct exposure to sunlight and are often more clover, oats, rape, buckwheat, and others). Also, several common
severely sunburned. Sunburned skin is reddened, edematous, hot, pharmaceuticals (phenothiazine, tetracyclines, sulfonamides, and
and painful. Later, the affected areas appear roughened and may others) may act as photosensitizing agents. The mechanism of
peel. A common clinical presentation of sunburned pigs is a brief tissue injury probably varies with the different agents.
attack of pain manifested as squealing or dipping the back while
Lesions tend to be more severe with photosensitization than
walking. In some cases, a pig may drop to its stomach, then get up
with sunburn, often resulting in exudation of serum onto the
and walk. Presumably, this is a response to feeling pain over the
skin’s surface. Exudation is followed by drying and fissuring. The
lumbar area. There have been reports of abortions associated with
lesions tend to be mildly pruritic. Skin necrosis with sloughing of
sunburn.
SHOULDER ULCERS, SKIN NECROSIS, SPLAYLEG, SUNBURN/PHOTOSENSITIZATION 153
SWINE DISEASE MANUAL

severely affected areas (white skin areas) often follows the acute with sunburn or photosensitization should be kept out of sun-
reaction. Conjunctivitis, sometimes with corneal opacity and light. Photosensitizing agents, once identified, should be avoided.
temporary blindness, may accompany photosensitization. Pigs

Torsion and volvulus


Torsions of the stomach and/or the intestines around the dorso- competition for feeder space, use of highly fermentable ration
ventral axis of the mesentery occur sporadically in pigs. Affected ingredients that produce excessive amounts of gas in the stomach
animals die quickly without any warning and will appear pale or colon, and advanced pregnancy. Torsion of the stomach seems
and bloat very rapidly during and after death. There is no single, more prevalent in sows that become agitated during once daily or
proven cause. Factors believed to contribute to torsions include intermittent feeding protocols. Torsion of the intestine and/or
rapid ingestion of a large amount of feed or water (as with once colon must be differentiated from hemorrhagic bowel syndrome.
per day feeding), overly crowded pens with associated piling and

Vestibular syndrome
A syndrome characterized by turning of the head to the affected yngitis with extension of the infection up the Eustachian tube to
side and a tendency to circle in that direction. The condition usu- the middle and internal ear. At necropsy, exudate is often demon-
ally occurs in young, growing pigs. The syndrome is a consequence strable in the middle ear. A less likely cause is as a sequel to cerebral
of either bacterial meningitis affecting the vestibular nerve or phar- edema resulting from sodium intoxication or edema disease.

Vices (tail biting, ear biting, flank biting, navel sucking)


Tail-biting, ear-biting, flank-biting, and navel-sucking behaviors diseases. Systemic bacterial infections and toxemia are suspected
can often be attributed to management factors (comingling pigs, to be contributory but sometimes hard to prove. Agents some-
out-of-feed events, facility hazards, overcrowding, and innumer- times implicated include sepsis or toxemia with Actinobacillus,
able sources of “stress”) in groups of growing pigs. Morbidity Streptococcus, Erysipelothrix, Salmonella, Mycoplasma hyorhinis,
and mortality can be high, particularly when confounded by or Escherichia coli. Immune-mediated vascular damage from
secondary bacterial infections. Through careful observation, one infectious insult has also been speculated.
can often identify the few individuals within an affected group
OTHER

Control requires careful observation at s levels: first, to accurately


that are responsible for the abnormal behavior and are inflicting
identify the offending pigs and second, to identify the risk factors
trauma on numerous penmates.
that may be contributing to the aggressive behavior. It is important
The cause of the offending behavior is often simply ascribed to that the aggressive pigs inflicting trauma be separated from the
“stress.” A disciplined approach is required to identify the true group to prevent more cases. In addition to being removed to
source(s) of the stress, which may include: facility limitations a hospital pen environment, pigs with severe lesions will likely
(inadequate feed or water space, overstocking, etc), nutritional require systemic antibiotic therapy to prevent extension of bacterial
compromise (mineral or salt imbalances in rations, maladjust- infections locally or systemically. The administration of anti-in-
ment to solid feed immediately postweaning, etc), environmental flammatory medication may also be warranted. Concurrently, the
inadequacies (drafts, full effluent pits, temperature variation, high risk factors and potential infectious or nutritional contributors to
humidity, etc), hygiene deficiencies, and concurrent infectious stress should be systematically analyzed and eliminated.

154 TORSION AND VOLVULUS,VESTIBULAR SYNDROME,VICES


Section VI

Nutritional diseases, toxicoses, and poisonings

Mycotoxicoses................................................................................................................................................................................157
Nutritional deficiencies and excesses........................................................................................................................................159
Toxins (plants, feed additives, chemicals, and gases).............................................................................................................162

TOXICOSES

155
156
TOXICOSES
NUTRITIONAL DISEASES, TOXICOSES, AND POISONINGS

Mycotoxicoses
Mycotoxicosis is the consequence of ingestion of grains or forage get information on appropriate sample collection and storage
containing toxic metabolites produced by certain fungi. Fungi techniques.
that produce toxins often do so only under specific conditions
There are few, if any, highly effective treatments for most mycotox-
of warmth, moisture, and humidity. Growth and storage factors
icoses. A ration or ingredient suspected of containing toxin should
that adversely affect plants or their seeds (grains) often influence
be replaced with nontoxic components. If a large quantity of the
mycotoxin production. Mycotoxins can develop in field grains,
feed remains, it can sometimes be fed to less susceptible species or
damaged grains, or improperly stored feeds. Importantly, myco-
diluted with good quality feed so that the mycotoxin is no longer
toxins are heat resistant and can be concentrated in various distill-
being fed at a toxic concentration. There are several mycotoxin
ers byproducts, often at 3 times the level of the original source.
binders on the market that can be used to prevent the absorption
Of the over 200 mycotoxins identified to date, at least 7 have of some of the mycotoxins from the pig’s gut. Any investment in
been reported to cause disease in swine. Some fungi produce binders should take into account both their efficacy in controlling
more than one mycotoxin. Several different fungi can produce the particular mycotoxins that are suspected and their suitability
different mycotoxins in a single mixed feed. The toxins may be for use in the feed matrix to which they will be applied.
additive and may potentiate one another. When metabolized,
Mycotoxins have been suggested as a primary cause of abortion
they may be converted into other toxic substances. While toxico-
in swine, but none have shown that effect in an experimental
logical effects are numerous and often confusing, one should be
setting. Astute veterinarians should be reluctant to implicate
careful not to implicate mycotoxins in disease processes without
mycotoxins as a cause of ill-defined maladies without appropriate
credible evidence.
investigation and evidence.
Mycotoxins produce their toxic effects in several ways, including
impairment of metabolic, nutritional, or endocrine functions. Aflatoxicosis
Some mycotoxins damage the liver and reduce average daily feed This mycotoxicosis is caused by mycotoxins produced by Aspergil-
intake, growth, and feed efficiency. Some mycotoxins are terato- lus flavus, Aspergillus parasiticus or Penicillium puberulum. Four
genic or carcinogenic. Some are immunosuppressive and predis- major toxins (B1, B2, G1, G2) are produced, with B1 being of the
pose pigs to secondary diseases. Several mycotoxins decrease the greatest significance as a potent hepatotoxin. Fungi growing on
reproductive performance of sows. Metabolites are sometimes peanuts, corn, wheat, and several other cereal grains commonly
passed in the milk of sows to piglets in their litters. The effect produce the toxins. Maximum aflatoxin formation occurs under
of mycotoxins varies with the amount ingested (dose), the time conditions related to the specific grain, its moisture content, stor-
over which it is consumed (duration), and the age of exposed age temperature, and humidity.
swine. Young pigs are usually much more susceptible than adults.
There is a marked age-related difference in susceptibility to afla-
Within a herd, there can be great variability in response to a
toxicosis. Young nursing or weaned growing pigs are much more
mycotoxin.
susceptible than adults. When aflatoxin is ingested by a lactating
Mycotoxicoses can present with either chronic or acute onset. dam, toxic metabolites are passed in her milk and serve as a
Most exposures are probably chronic or subacute as a result of source of exposure to the nursing pigs. These toxins reduce feed
consuming small amounts of toxin over a long period of time. intake, average daily gain, and feed efficiency. Since aflatoxins are
In these instances, there may be few signs of toxicosis other than immunosuppressive, signs of toxicosis often include an increase in
decreased appetite, slow growth, and increased susceptibility to previously controlled secondary diseases.
secondary diseases. Acute outbreaks may have more obvious signs

TOXICOSES
Acute aflatoxicosis is uncommon in swine. Usually, it is a sub-
that will vary for each of the different mycotoxins. Diagnosis of
acute to chronic disease caused by daily ingestion of smaller
chronic mycotoxicosis is often difficult because clinical signs are
amounts of aflatoxin over several weeks. Lesions vary among
seldom overt, and lesions are not specific. By the time a mycotox-
pigs in the same affected group but are predominantly those of
icosis is considered, the suspected feed may already be consumed,
hepatopathy. In more severe cases there are sudden deaths, hem-
with none having been collected and stored properly for analysis.
orrhages in multiple tissues, and icterus. The liver may be swollen,
Prevention of mycotoxicosis is largely through careful selection fatty, and have areas of necrosis. There may be a prolonged
and proper storage of high-quality grains and other feed ingredi- clotting time. With subacute to chronic hepatotoxicosis, the liver
ents and the careful maintenance and cleanliness of feed prepa- may be reduced in size and fibrotic, and ascites may be present.
ration equipment. It is often worthwhile to properly dry and
Diagnosis is usually based on some combination of a history
store samples of representative batches of grain that are used on
of slow growth (often accompanied by secondary diseases that
a farm in the event they are needed for analysis later. Producers
seem unresponsive to treatment), an elevation of serum enzymes
anticipating problems should locate a competent laboratory and

MYCOTOXICOSES 157
SWINE DISEASE MANUAL

associated with hepatocellular damage, and lesions related to Trichothecene toxicoses


liver pathology. Microscopic hepatic lesions include bile duct There are numerous structurally related toxic compounds pro-
hyperplasia and enlargement of hepatocytes. In swine, chronic duced by certain Fusarium species that are classified as tricho-
aflatoxin toxicity can occur with at levels as low as 300 ppb in the thecene mycotoxins. At least 3 of these are of importance in pig
feed; acute toxicity requires concentrations beyond 1000 ppb. production. Trichothecenes are cytotoxic to many cell types and
Aflatoxin is considered to be carcinogenic in humans. are strongly immunosuppressive. Signs of trichothecene toxicity
usually include feed refusal, salivation, and sometimes vomiting.
Ergotism With chronic exposure, there may be paresis, paralysis, or sei-
Claviceps purpurea is a fungus of many grasses and several cereal zures. Lesions often include gastroenteritis, hemorrhagic diathe-
grains, especially rye, oats, and wheat. The sclerotium of the sis, skin irritation, and necrosis.
fungus is a dark, elongated body and can be seen on cereal grain
Diagnosis of trichothecene-related toxicosis can be difficult. The
heads and in processed grains. The fungus produces 3 major
presence of moldy or caked feed or a reluctance to consume feed
alkaloids that cause ergotism. The primary lesions caused by the
may suggest the presence of a trichothecene toxicosis. Improve-
alkaloids include arteriolar vasoconstriction and endothelial cell
ment following a change in feed suggests the original feed was
injury that often leads to thrombosis. When present in low levels,
contaminated.
the alkaloids can result in reduced growth rates. Larger amounts
lead to ischemic necrosis followed by a dry, gangrenous sloughing
Deoxynivalenol (DON, vomitoxin)
of parts of extremities, especially tails, ears, and hooves. Symp-
Deoxynivalenol is a commonly occurring mycotoxin in corn and
toms of ergotism are exacerbated by cold weather. In pregnant
wheat. Despite its common name of “vomitoxin,” swine rarely
sows, ergotism can inhibit mammary development, reduce litter
consume a large enough dose to produce vomiting. However,
size, reduce birth weights, and cause a profound postfarrowing
reduced feed intake is quite prominent and often the only clinical
agalactia. The agalactia is believed to be related to inhibition of
sign observed.
prolactin secretion.
Diagnosis of ergotism is based on lesions and the gross or micro- T-2 toxin
scopic identification of significant numbers of ergot sclerotia in In swine, the experimental administration of T-2 toxin, alone and
grains or the feed. Laboratory quantitative analysis can be used to with aflatoxin, has resulted in crusting and ulceration of the skin
confirm the presence of clinically significant amounts of alkaloids of the snout, lips, buccal commissures, and prepuce.
in ingredients or feedstuffs.
Zearalenone (F-2)
Fumonisin toxicosis This mycotoxin is produced by Fusarium graminearum and may be
The fumonisins include 2 principal toxins produced by Fusarium present in moldy corn, standing corn, other grains, and in pelleted
moniliforme. Signs of acute toxicity in growing and adult pigs are cereal feeds. It has an estrogenic effect that results in vulvovagi-
primarily related to the respiratory system and include dyspnea, nitis and precocious mammary development in prepuberal gilts.
cyanosis, weakness, and death within 4 to 10 days. Pulmonary Swelling and enlargement of the vulva sometimes lead to tenesmus
lesions include marked pulmonary edema and hydrothorax. with prolapse of the rectum. Similar estrogenic effects in gilts have
Pregnant sows that survive acute toxicity frequently abort in the occurred as a result of consuming estrogens from other sources,
days following their recovery. Growing pigs that survive the acute including alfalfa or other naturally occurring phytoestrogens.
syndrome suffer from clinical signs related to a hepatotoxicosis When consumed by nursing sows, some of the metabolites of
manifested as icterus, hepatic necrosis, and megalocytosis. Differ- zearalenone are passed to the offspring through the milk and can
ences in lesions and clinical presentation seem to be dose related.
TOXICOSES

lead to estrogenic effects in the piglets, namely enlargement of the


Recent research has demonstrated that fumonisins decrease the external genitalia and uterus. When the toxin is consumed by sows
ability of intravascular macrophages to clear blood-borne bacteria near the midpoint of their estrous cycles, it has a luteotropic effect
in swine, thereby potentially increasing susceptibility to respi- and can cause persistent anestrus or pseudopregnancy. Fewer pigs
ratory disease. Fumonisins are a well-known cause of leukoen- per litter are born to sows given zearalenone in the preimplantation
cephalomalacia in horses and are carcinogenic in humans at high period (7 to 10 days postmating). Zearalenone, when consumed in
concentrations. very high doses, can cause reduced libido, decreased testicular size,
and preputial enlargement in young boars.

158 MYCOTOXICOSES
NUTRITIONAL DISEASES, TOXICOSES, AND POISONINGS

Nutritional deficiencies and excesses


General comments rapid breathing; sudden deaths can occur. Anemic piglets are
Nutritional diseases are relatively uncommon in modern swine pro- quite susceptible to bacterial infection and have little resistance to
duction. When they do occur, they are usually the result of mixing environmental chilling.
errors or poor quality-control practices rather than formulation Diagnosis is often by a history that documents a lack of iron
errors. Both short- and long-term effects of deficiencies or excesses supplementation at birth concomitant with typical clinical signs
can be risk factors for expression of other diseases, so nutritional and postmortem lesions. Several laboratory procedures used to
contributors to infectious diseases should be considered whenever confirm anemia include hematocrit, red cell count, and examina-
investigating unusual disease outbreaks. Feed analysis for micro- tion of stained blood smears revealing a microcytic, hypochromic
nutrients is an inexact science exacerbated by sampling variation, anemia. Hematology and serum chemistry will reveal reduced
which can contribute to misdiagnosis or confusion. serum iron and transferrin saturation and a low hematocrit.

Vitamin and mineral deficiencies Prevention is possible by administration of supplemental iron


(100 to 200 mg) to all pigs in the first few days of life, prefera-
Iodine deficiency (goiter) bly by injection. Parenteral administration is most commonly
Hyperplastic thyroid glands (goiter) occasionally occurs in swine, practiced and has the advantage of precise dosage. Iron dextran
usually due to iodine deficiency in the pregnant sow, a genetic should be injected only into the neck muscles in order to prevent
defect in the sow for the biosynthesis of thyroid hormones, inges- any possibility of staining valuable areas of the carcass. Oral iron
tion of goitrogenic substances (certain plants, drugs, or chemi- products are available and can be effective, but one cannot be sure
cals) by the gestating sow, or iodine toxicity from dams being fed that all pigs receive an appropriate dose when using these prod-
an excess of iodine. ucts. When improperly used, both forms of administration can
In swine, goiter usually occurs in iodine-deficient regions where lead to iron toxicity, which is manifested as acute death following
iodized salt has not been included in the dam’s feed. Deficiency administration. Pigs deficient in vitamin E/selenium are highly
of iodine leads to the birth of weak or dead pigs that are largely susceptible to iron toxicity. Copper toxicity can occur at high
devoid of hair and may have a mucinous edema over enlarged doses due to mixing errors.
foreparts of the body with the skin in these areas thick and Pigs raised outdoors with access to soil do not usually have a
doughy. The tongue is often edematous and may protrude from requirement for iron supplementation, but many producers still
the oral cavity. Enlarged thyroid glands (goiter) in piglets may find it a beneficial practice even in these situations.
not be visible externally but often can be palpated or observed at
necropsy. In mature swine, iodine deficiency is not usually a sig- Zinc deficiency (parakeratosis)
nificant disease, although gestation may be prolonged by as much Parakeratosis is a zinc-responsive dermatosis usually observed in
as 7 days. Iodine deficiency is easily avoided by using iodized salt swine 2 to 4 months of age. Pigs not allowed access to soil or not
in the ration of gestating sows. supplemented with zinc are more likely to have parakeratosis. The
disease is caused by a relative deficiency of zinc. The deficiency is
Iron and/or copper deficiency (anemia) and toxicity usually caused by feeding an unbalanced diet that has one or more
Piglets are born with very little iron reserves. Colostrum and milk of the following features: excessive calcium, excessive phytic acid
from the sow provide relatively little iron, perhaps only 15% to (sometimes present in soybean meal), or a low concentration of
50% of the daily requirement. The suckling piglet’s rapid growth essential fatty acids. These features all adversely affect the avail-
and expansion of blood volume make it highly likely that iron ability of dietary zinc. In addition, enteric pathogens or changes in

TOXICOSES
deficiency and anemia will develop unless an exogenous source of intestinal flora can adversely influence zinc absorption. Parakerato-
iron is made available. This is especially true for rapidly growing sis is most often caused by consumption of excessive calcium.
piglets raised in confinement and not having access to soil or feces
that contain iron. A small amount of copper is essential for the Affected pigs show few signs of illness other than skin lesions and
utilization of iron. Copper deficiency can also lead to anemia that reduced growth rate. Initial lesions appear as reddened macules
has many of the same clinical signs and lesions as iron deficiency. and papules on the ventrolateral abdomen and medial surface
of the thighs; these lesions often go unobserved. The lesions
Signs and lesions of iron deficiency are primarily related to severe are slowly covered by thick, roughened scales and crusts. More
anemia. Piglets not supplemented with iron will be quite pale and obvious lesions soon become apparent on the lower legs and on
lose body condition and thrift after about 1 week of age. At nec- the dorsum. Lesions can sometimes be seen around the eyes, ears,
ropsy, these pigs will have pallor, watery thin blood, a thin-walled snout, and tail and may eventually become generalized. Affected
heart, and edema of the lungs, muscles, and connective tissues. areas of the skin are hyperkeratotic and there may be fissuring of
Occasionally, pigs weaned at 3 to 4 weeks of age will develop the epidermis with secondary infection of the fissures. A unique

NUTRITIONAL DEFICIENCIES AND EXCESSES 159


SWINE DISEASE MANUAL

feature occasionally seen is a focal or diffuse hyperkeratosis on Properly balanced rations that include adequate calcium, phos-
the tongue. Parakeratosis is a microscopic feature of the affected phorus present in the proper ratio, and vitamin D are essential for
epidermis and gives this dermatosis its name. the prevention of rickets and osteoporosis. Treatment tends to be
unrewarding for both rickets and osteoporosis.
Parakeratosis must be differentiated from sarcoptic mange and
exudative epidermitis. Parakeratosis is nonpruritic, whereas Vitamin D toxicity can occur with excess supplementation,
sarcoptic mange is highly pruritic. Exudative epidermitis tends usually manifested as ectopic tissue mineralization in kidney,
to occur in younger, smaller pigs. Pigs with parakeratosis will stomach, lung, and elsewhere.
recover if excessive calcium is removed from the ration and it is
properly supplemented with zinc. Most good commercial or care- Other vitamins
fully compounded rations are now supplemented with adequate Deficiencies or excesses of other fat-soluble vitamins have been
levels of zinc salts. Although once common, parakeratosis seldom reported and are usually due to ration mis-formulation, mixing
occurs today unless a feed mixing error occurs. errors, or prolonged storage in poor conditions. There are reports
of vitamin A deficiencies causing fetal malformations and ill-thrift,
Calcium, phosphorus, and vitamin D (rickets and and excessive vitamin A resulting in bone malformations and
osteoporosis) altered gait. Vitamin K deficiency is also rare but can occur under
Rickets is a disease of growing bones; consequently, it is usually similar circumstances. Exposure to coumarin-like compounds,
seen in young, growing pigs in which there is a deficiency, an either naturally occurring or as warfarin, has also caused hemor-
imbalance, or a failure of utilization of calcium, phosphorous, or rhagic episodes that will respond to vitamin K supplementation.
vitamin D. Rickets is most often caused by a dietary deficiency
Common B vitamins include thiamine (B1), riboflavin (B2),
of vitamin D or phosphorus. The basic abnormality is a failure
niacin (B3), pantothenic acid (B5), pyridoxine (B6), biotin (B7),
of mineralization of osteoid and cartilaginous matrix, especially
folic acid (B9), and cobalamin (B12). B-vitamin deficiencies are
in growth plates. This is most obvious as a thickening and irreg-
quite rare, with similar risk factors as mentioned in the previous
ularity of growth plates in long bones. In confined animals not
paragraph. Deficiency conditions that have been reported include
exposed to sunlight or properly supplemented, vitamin D may
polioencephalomalacia (B1), anemia (B12), and altered gait with
be inadequate. In pastured swine fed little or no grain or protein
myelopathy (various). Reports of B-vitamin toxicities in swine are
supplement, phosphorus may be inadequate.
extremely rare.
Signs of rickets include poor growth, short stature, enlargement
of the ends of long bones, lameness, and deformation of the Element and mineral toxicities
weight-bearing long bones. Necropsy lesions include an unusual
Arsenic
number of recent or healing fractures, ribs that bend markedly
Pigs are relatively resistant to inorganic arsenic toxicosis but can
before breaking with a weak “snap,” and widened, thickened, and
develop severe hemorrhagic gastroenteritis when exposed to
irregular growth plates. Abnormal growth plates are best seen on
older, arsenic-containing pesticides at levels greater than 100 mg/
longitudinally sawed long bones.
kg (45.4 mg/lb) of arsenic per bodyweight.
Osteoporosis is a lesion of mature bones. It follows removal of
Organic phenylarsonic forms of arsenic have toxicological prop-
much of the mineral content of the bones. It results from an
erties very different from those of inorganic arsenicals. Roxarsone
imbalance between bone formation and resorption in favor of
and arsanilic acid were traditionally used to promote growth and
resorption. In the process, there may be a softening of the bone
to treat swine dysentery and Mycoplasma suis, but they are rarely
(osteomalacia). Osteoporosis often occurs in prolific sows that
used in modern agriculture and are banned in many countries.
mobilize minerals to meet the demands of high milk production.
TOXICOSES

Poisoning is dose related. Signs of chronic poisoning with arsan-


Gilts in their first lactation are also susceptible, since their skeletal
ilic acid or roxarsone include goose-stepping, hind limb ataxia,
development may have been incomplete prior to pregnancy,
limb paresis, and blindness. A presumptive diagnosis can often be
allowing insufficient time to build a mineral reserve. Osteoporo-
based on a history of misuse of the compounds and the presence
sis often results in fractures in the latter part of a nursing period,
of characteristic clinical signs in chronically affected swine. The
immediately after weaning, or during the mating period just after
diagnosis may be assisted by toxicological assays of kidney, liver,
weaning. Inappropriate ration formulation or mixing is the most
muscle, and feed. The neurotoxic effect of poisoning is sometimes
important etiological risk factor.
reversible if the compound is removed within 2 or 3 days of the
Signs of osteoporosis include lameness, recumbency, fractures, appearance of signs. Blindness and long-standing peripheral
and paraplegia. At necropsy, fractures can often be demonstrated nerve damage may be permanent.
in the femur, humerus, or lumbar vertebrae. There may be distor-
tions or deformities of the pelvis.

160 NUTRITIONAL DEFICIENCIES AND EXCESSES


NUTRITIONAL DISEASES, TOXICOSES, AND POISONINGS

Copper, iron, selenium, and zinc Sodium ion intoxication (salt poisoning or water
Pigs tolerate fairly high doses of copper, which is sometimes deprivation)
added to feed to improve health and growth. Copper toxicity in Salt poisoning can occur in pigs as a consequence of water
pigs manifests as acute hemolytic anemia, hemoglobinuria, and deprivation or from sudden ingestion of too much salt; the latter
secondary nephropathy. Toxicity can occur when dietary copper tends to be a rare occurrence. Sodium intoxication in water-deprived
exceeds 4,000 ppm or when pigs are inadvertently exposed or pigs can occur even if the feed contains an appropriate level of salt,
overdosed via water or by injection. although the risk is increased when salt levels in the feed are excessive.
Iron toxicity can occur when pigs are inadvertently exposed to Clinical signs are precipitated or magnified by allowing the
concentrated supplements orally or when overdosed by injection. water-deprived pigs sudden, unlimited access to water. Water
Rapid respiration and cardiovascular collapse usually occur deprivation can occur for many reasons but commonly may be
within hours of exposure. Pigs deficient in vitamin E are reported the result of freezing of the water source, plugged water nipples,
to be at greater risk for iron toxicity. or inadvertently leaving a water valve closed. Operators may
not always be forthright in admitting human errors related to
Acute selenium intoxication causes posterior paresis due to acute
water deprivation. Salt poisoning has also been known to occur
poliomyelomalacia of the ventral horns of the spinal cord. Feed
following prolonged shipping without access to water, followed
formulation or mixing errors are the usual source of the toxicity for
by unlimited access to water. Salt poisoning is also reported in
weaned pigs. Overdoses of injectable selenium supplements can
swine after ingestion of salty brine from overflowing, loose-salt
cause sudden death with cardiovascular collapse. In chronic selenium
boxes provided for other livestock, or following ingestion of large
toxicosis, hair loss and separation of the hoof from the coronary band
amounts of whey. This type of poisoning is also more likely in
occurs and may precede paresis due to spinal cord lesions. Diagnosis
water-deprived pigs.
is by recognition of clinical signs, histopathology of spinal cord, and
tissue analysis for selenium. There is no treatment. Clinical signs of sodium ion toxicosis are caused by the acute
cerebral edema associated with rapid rehydration. Because the
Zinc in the oxide form is tolerated well by pigs and has been com-
condition most often occurs secondary to water deprivation (rather
monly supplemented in feed to improve health and performance.
than a primary toxic intake of salt), salt poisoning frequently occurs
Other salts of zinc may interfere with absorption of other cations
at a pen or herd level. Signs include aimless wandering, blindness,
and, at high doses, can lead to arthritis or internal hemorrhage.
deafness, and head pressing. Affected pigs sometimes dog-sit,
slowly raise their nose upward and backward, and then fall on their
Lead and mercury
side in spasms that may be followed by paddling of the legs. They
In contrast to most species, pigs are very resistant to lead poison-
may then arise and continue their wandering.
ing. Pigs fed various salts of lead have achieved very high blood
levels with no clinical effect. However, suspicions of lead exposure Diagnosis associated with water deprivation may be suggested by
may warrant blood analysis to assure adulterated pork products history, signs, and elevated sodium levels in serum or cerebrospinal
do not enter the food chain. fluid. Gross lesions may be absent or limited to mild gastritis, and
perhaps the presence of dehydrated fecal pellets in the rectum.
Swine exposure to mercury is usually limited; however, accidental
Histopathologic confirmation of the condition is by observation
exposure to pesticides or seeds treated with mercurial fungicides
of rather unique meningeal and cerebral perivascular cuffing by
does occur. Gastroenteritis, nephropathy, ataxia, central nervous
eosinophils in the brain. Later and less reliably, there may be lami-
disturbance, blindness, and death can result.
nar subcortical polioencephalomalacia or necrosis. Salt poisoning
Selenium must be differentiated from all other encephalitic diseases. In an
affected pen, a clue to the occurrence of water deprivation will be

TOXICOSES
Selenium toxicity can occur with injections, excess supplemen-
tation from mixing errors, or in geographical areas where soil the absence of any urine or wet feces on the pen floor.
selenium levels are high. Clinical signs of chronic toxicity include Water-deprived or affected pigs should be reintroduced to water
hair loss and separation of hoof from the coronary band. Acute slowly, given only small amounts of water at frequent intervals.
toxicity is manifested as paresis due to spinal cord malacia. This may reduce mortality. Pigs showing clinical signs usually die,
regardless of treatment.

NUTRITIONAL DEFICIENCIES AND EXCESSES 161


SWINE DISEASE MANUAL

Toxins (plants, feed additives, chemicals, and gases)


General comments cottonseed meal with high gossypol content can cause ill-thrift or
Historically, rooting and the inquisitive behavior of pigs coupled heart failure and death. Toxicity from gossypol is rare with prop-
with outdoor rearing practices provided pigs with access to a erly formulated swine diets fed in most commercial production
broad range of potential toxins and poisons. The list of potential systems.
toxins to which pigs are susceptible is quite long; however, modern
production practices and improvements in pharmaceuticals and Nightshade
parasiticides now limit exposure to many traditionally common Consumption of Solanum nigrum (black nightshade) occasion-
toxins and poisons. ally causes acute depression, incoordination, muscular tremors,
recumbency with kicking of feet, and death in some cases. The
Suspected cases of poisoning should be systematically investi- leaves and green berries are the most toxic parts of the plant, but
gated as appropriate for suspected toxicants. In many cases of they are quite bitter and rarely consumed unless no other forage
vague signs or sudden deaths, there is a coincidental recollec- is available.
tion of recent exposure to a “toxin.” This association can easily
be interpreted as causal for current illnesses or deaths of animals Pigweed poisoning
and should be regarded with caution. Pigs allowed access to pastures or lots containing pigweed
Several of the more common toxicants (plants, feed additives, (Amaranthus retroflexus) may be poisoned, usually in the late
organic/inorganic toxicants, chemicals, gases) for swine are summer or fall. Signs appear within 5 to 10 days after exposure
listed below. and include trembling, weakness, incoordination, knuckling, and
almost complete rear leg paralysis. Morbidity is variable, although
Plants mortality can be high (75% to 80%) in pigs that develop clinical
signs. Lesions are consistent with acute nephrosis and heart
Cocklebur poisoning failure; the hallmark gross lesion is severe perirenal edema. The
Cockleburs (Xanthium species) are widely distributed in the mid- kidneys are normal in size but may appear blanched. Often,
western US as weeds in fencerows, ditches, and low or marshy there is ascites, hydrothorax, or edema of the ventral body wall.
areas. The toxic principle, carboxyatractyloside, is present in the Long-standing cases may have renal fibrosis. Microscopic lesions
seeds and young seedlings of the plant, especially during the in the kidneys of acutely affected pigs include necrosis of both
cotyledonary (2-leaf ) stage of growth. Seedlings may be ingested proximal and distal convoluted tubules with numerous casts
by pigs on pasture, especially after spring rains when burs sprout in tubules. Diagnosis is usually made from a history of sudden
in low-lying areas, or pigs may ingest seeds present in mixed or death, typical clinical signs, severe perirenal edema at necropsy,
ground feeds. Pigs can be found dead after consuming only mod- and plant identification. The toxic principle is not known. There
est numbers of seedlings. Because the clinical course of disease is is no widely accepted treatment for affected pigs. Pigs should be
brief (several hours), signs may not be observed, and lesions may denied access to the plants. New cases may develop for as many as
be absent from dead pigs. Mortality is sporadic unless plants are 10 additional days after exposure has been terminated.
numerous. In experimentally poisoned pigs, signs include depres-
sion, hypoglycemia, occasional nausea, incoordination, convul- Feed additives
sions, and death. Lesions include serofibrinous effusions in body
cavities and subcutaneous edema. There can sometimes be edema Carbadox
of the gall bladder wall and mild gastroenteritis. Microscopic In the US, carbadox may be used to control swine dysentery and
lesions include centrilobular hepatotoxicosis with centrilobular bacterial enteritis when included in the feed at 50 ppm and, less
TOXICOSES

accentuation of the liver pattern. commonly, as a growth promotant when fed at 10 to 25 ppm.
Levels greater than 100 ppm can cause decreased feed consump-
Diagnosis is often difficult unless cocklebur seedlings can be tion, growth retardation with degeneration of the adrenal cortex,
found in the pasture or seeds can be found in the feed. Cocklebur and, at higher doses, posterior paresis.
poisoning must be differentiated from poisonings caused by
ingestion of coal tar (clay shooting pigeons), mycotoxins, gossy- Ionophores
pol (cotton seed), and others. Ionophores (eg, narasin, lasalocid, monensin) are sometimes
used in animal feeds to improve rate of gain and feed efficiency.
Gossypol Most cases of toxicity with ionophores are from mixing errors,
Gossypol is a polyphenolic binaphthalene compound found inadvertent inclusion of an ionophore in the swine feed, or most
in varying amounts in cottonseed meal. Prolonged feeding of importantly, simultaneous administration of the antibiotic tiamu-
lin in feed or water.

162 TOXINS (PLANTS, FEED ADDITIVES, CHEMICALS, AND GASES)


NUTRITIONAL DISEASES, TOXICOSES, AND POISONINGS

In the US, narasin is labeled for use in swine at 15 to 30 ppm Chemical products of historical and ongoing importance include
and is considered relatively safe when used per label instructions organophosphates, carbamates, and chlorinated hydrocarbon
and in the absence of the antibiotic tiamulin. Clinical signs of compounds administered at excessive doses. More often than
anorexia, weakness, and ataxia can occur when narasin is fed at not, toxicity is a result of their accidental addition to feed or
levels greater than 90 ppm. The greatest risk for poisoning with their presence as contaminants of feed ingredients, equipment, or
narasin (and all ionophores) is with the simultaneous adminis- swine premises.
tration of tiamulin, which effectively blocks the metabolism and
Clinical signs of organophosphate or carbamate toxicity result
excretion of narasin (and all ionophores) creating a buildup and
from stimulation of the parasympathetic nervous system, leading
toxicity. Pigs poisoned with this combination will have acute
to salivation, urination, defecation, vomiting, and death. Muscu-
ataxia with pain, necrosis of skeletal muscles, and rapid death.
lar tremors may be present, but seizures are rare. Diagnosis is by
There is no specific treatment other than immediate removal of
clinical signs, history of exposure, and finding depressed cholin-
feed and medicated water.
esterase activity in brain or whole blood. Acute cases will respond
Poisoning with monensin or lasalocid is relative rare and is usu- to atropine administration.
ally due to the inadvertent administration of very high doses in
Chlorinated hydrocarbon insecticides are stimulatory, with clin-
feed, or lower doses in feed coupled with simultaneous tiamulin
ical signs that include hyperesthesia, muscle spasms, and muscle
administration.
fasciculation leading to seizures. Diagnosis is by clinical signs,
history, and confirming the offending compound in brain, liver,
Ractopamine
or stomach contents. Treatment is symptomatic.
In the US, ractopamine is a beta-2 agonist used in the last 3 to 6
weeks of the finishing period to stimulate muscle growth. Poten- Pyrethroid compounds for fly control are relatively nontoxic
tial adverse effects that are sometimes reported include muscle to swine. Many pyrethroid pesticides are marketed for various
tremors, tachycardia, hypotension, prostration, and hypokalemia. purposes, but most are considered safe for swine unless there is
Confirmed cases of ractopamine toxicity are rare. massive exposure.
Other chemicals of historical importance include organic mer-
Sulfonamides
cury (fungicide), pentachlorophenol (wood preservative), and
Overdoses of sulfonamides in feed, particularly with restricted
chromate-copper arsenate (wood preservative), but most of these
water intake, can cause acute nephrosis. Toxicity can be exacer-
and related products are no longer available in the US.
bated by preexisting kidney diseases or simultaneous presence of
mycotoxins (eg, citrinin, rubritoxin, ochratoxin) in feed. Most modern herbicides used for weed control are rarely associ-
ated with toxicity in swine. There is very little risk from overspray,
Rodenticides with adverse reactions limited mostly to oral consumption of
Swine are susceptible to inadvertent exposures to anticoagulant concentrates, which can sometimes cause gastric irritation or
rodenticides (eg, warfarin, diphacinone, chlorophacinone, vague signs. Paraquat is an exception causing chronic pulmonary
bromadiolone, brodifacoum, or pindone) that have been placed interstitial fibrosis as a sequel to exposure.
around pig premises. Toxic doses increase clotting time, leading
to internal hemorrhages as well as epistaxis or melena. Vitamin Gases and ventilation failure
K can be a helpful treatment should accidental exposure occur, Five gases (ammonia, carbon dioxide, carbon monoxide, hydro-
although not all rodenticides are vitamin K responsive. Bro- gen sulfide, and methane) are associated with swine environ-
methalin causes depression, ataxia, and death. Cholecalciferol ments, especially confinement rearing. Ammonia (NH3) and
(vitamin D3 and analogs) are effective rodenticides causing hydrogen sulfide (H2S) are normally generated from the decom-

TOXICOSES
hypercalcemia and tissue mineralization. Careful placement and position of swine excrement.
maintenance of baiting stations is critical in helping to avoid
Carbon dioxide (CO2) can cause deaths but is perhaps a lesser
inadvertent exposures. Baits should be fixed in place so that
threat than carbon monoxide (CO), because it is heavier than air
rodents are not able to remove the bait and bring it into contact
and usually accumulates only in slurry pits below pig housing and
with pigs. Internationally, there have been incidents of rodenti-
working areas.
cide residues found in pork due exactly to this reason.
Methane (CH4) is an explosion hazard. In recent years, the num-
Chemical toxins ber of accidental methane explosions and fires has increased in
Toxicoses from chemicals, particularly insecticide and parasiticide barns with deep underfloor manure storage. This is presumably
exposures, are less frequent now due to the availability of safer due to variation in fermentation characteristics of excrement and
products, eradication programs for lice and mange, and the exten- its composition, likely from dietary inputs such as inclusion of
sive use of confinement housing, which has minimized challenges distillers waste products. All 5 gases represent potential threats
with many internal parasites.
TOXINS (PLANTS, FEED ADDITIVES, CHEMICALS, AND GASES) 163
SWINE DISEASE MANUAL

to both swine and people, but ammonia, carbon monoxide, and A “sewer gas odor/rotten egg smell” detectable by humans from
hydrogen sulfide toxicities are the most common and are dis- 0.1 to 5 ppm is sometimes offensive but is not toxic. Levels from
cussed in more detail below. 10 ppm to 100 ppm can cause eye and respiratory irritation.
Humans cannot detect the odor of H2S at levels greater than
Ammonia toxicity 150 to 250 ppm because of olfactory paralysis induced by the
Ammonia gas (NH3) is formed by the decomposition of animal gas. H2S levels greater than 300 ppm affect the nervous system.
waste and is present at some level in most animal facilities. The Immediate collapse and respiratory paralysis occur at levels
odor can be detected by most people at concentrations of around greater than 1000 ppm.
10 ppm, a level which appears to have little detrimental effect on
Hydrogen sulfide is heavier than air and accumulates in liquid
pig health. However, when the concentration reaches 50 ppm
manure-holding pits below confinement buildings. The gas usually
or more, NH3 may act as an irritant of the mucous membranes
remains dissolved in the liquid component of swine effluent and
of the eyes, nasal passages, and lungs and cause ocular and nasal
remains below the toxic level in air unless the effluent is agitated.
discharge. Concentrations of NH3 that are high enough to be
Effluent storage pits are often agitated just prior to and during
irritating to mucous membranes of pig farm workers likely have a
the emptying process at which time high levels of H2S can be
similar effect on animals continuously exposed.
released. If inhaled in high concentration, H2S can cause instant
Research on swine suggests that toxic concentrations of NH3 (over fatal systemic intoxication of exposed swine or people by directly
50 to 100 ppm) reduce growth rate, reduce bacterial clearance from suppressing the respiratory center in the brain. High levels of the
the lungs (interferes with mucociliary apparatus), exacerbate nasal gas paralyze a worker’s sense of smell and may give a false sense of
turbinate lesions in pigs infected with organisms of progressive security. Workers trying to rescue affected swine or coworkers are at
atrophic rhinitis (Bordetella bronchiseptica, Pasteurella multocida), a very high risk of asphyxiation from a single breath.
and may influence the course of other infectious diseases.
Exposure to toxic levels of H2S can be avoided by emptying and
cleaning the pit when the building is empty. Adequate ventilation
Carbon monoxide toxicity
should be provided (fans on, curtains down) whenever pits are
Carbon monoxide (CO) has a much greater affinity for hemoglo-
agitated to keep H2S at a nontoxic level.
bin than does oxygen and is therefore highly effective at displacing
oxygen from the blood. It combines with hemoglobin to form car-
Ventilation failure
boxyhemoglobin, reduces oxygen exchange, and causes mortality.
Mechanical ventilation of confinement swine facilities is com-
Carbon monoxide is produced by incomplete combustion of any
mon in temperate and cool climates. Death losses in confinement
carbonaceous fuel, but most poisonings occur because of improp-
buildings are usually due to mechanical failures or power loss
erly functioning (yellow versus blue flame) space heaters or fur-
rather than simply an accumulation of toxic gases and poisoning.
naces. Carbon monoxide is a colorless, odorless, and tasteless gas.
Importantly, ventilation failure can rapidly lead to very high
Recognition of malfunctioning heaters should alert one to the temperatures and humidity causing hyperthermia and death. The
possibility of poisoning. In late-term pregnant sows, a high level increased concentration of toxic gases can contribute or cause
of stillbirths and neonatal mortality is associated with CO poi- death by intoxication (eg, hydrogen sulfide) or asphyxiation (car-
soning. The sows themselves may show no other signs. Affected bon dioxide or carbon monoxide). Suffocation (the obstruction
piglets usually show no lesions other than a pink to bright red of air passages) as a cause of death is usually the result of piling of
color imparted to their blood and tissues by carboxyhemoglobin. pigs in cold weather.
Blood from affected piglets and fetuses with CO poisoning is
Any investigation of acute deaths suspected to be due to venti-
usually characterized as “cherry-red” in color. The concentration
lation failure should be approached with caution, putting safety
TOXICOSES

of carboxyhemoglobin in fetal thoracic fluid or blood of an


first. Human deaths from hydrogen sulfide intoxication are too
affected pig can be used to confirm CO poisoning.
common, as this gas kills quickly.
Hydrogen sulfide toxicity
Hydrogen sulfide gas (H2S) inhaled at toxic levels is dangerous
and fatal to both pigs and people. The danger of high concen-
trations (greater than 100 ppm) of H2S should be recognized,
respected, and avoided, but the usual low level of the gas in closed
confinement facilities (less than 0.2 ppm) is not toxic and is of
little consequence.

164 TOXINS (PLANTS, FEED ADDITIVES, CHEMICALS, AND GASES)


Section VII

Tables

Anthelmintics and parasiticides for swine...............................................................................................................................167


Arthritis in neonatal and growing swine..................................................................................................................................168
Central nervous system (CNS) diseases in swine: comparative approach to diagnosis.................................................169
Diarrheal diseases common at various ages.............................................................................................................................171
Diarrheal diseases in swine: comparative approach to diagnosis........................................................................................172
Internal parasites...........................................................................................................................................................................175
Intestinal hemorrhage: differential diagnosis..........................................................................................................................177
Mycoplasma-related diseases......................................................................................................................................................179
Reproduction: causes of low farrowing rate............................................................................................................................180
Reproductive failure: infectious causes ...................................................................................................................................181
Respiratory disease in swine: comparative approach to diagnosis......................................................................................183
Skin lesions: differential diagnosis............................................................................................................................................186
Vesiculating viral diseases............................................................................................................................................................189
Zoonotic agents found in swine and humans.........................................................................................................................190

TABLES

165
166
TABLES
DISEASES CAUSED BY VIRUSES

Anthelmintics and parasiticides for swine


Many effective anthelmintics are available for the treatment of internal and external parasites. Following are guidelines that show
the relative efficacy of the major categories of active ingredients against the major parasites of swine in the US. Manufacturers
generally have excellent resource materials that describe the effectiveness of their products against the various life cycle stages
of parasites, and the reader is encouraged to refer to them when making usage choices.

Agent Products
Common
Name name Piperazine Pyrantel Avermectins Levamisole Dichlorvos Fenbendazole
Ascaris suum Roundworm + + + + + +
Stomach
Ascarops strongylina - - - - + +
worm
Cystoisospora suis,
Coccidiosis - - - - - -
Eimeria species
Globocephalus
Hookworm - - + + + +
urosubulatus
Gongylonema Esophageal
- - + + - +
pulchrum worm
Haematopinus suis Lice - - + - - -
Red stomach
Hyostrongylus rubidus - - + - + +
worm
Macracanthorhynchus Thorny-
- - +/- +/- - -
hirudinaceus headed worm
Metastrongylus
Lungworm - - + + - +
species
Oesophagostomum Nodular
+/- + + + + +
species worm
Physocephalus Stomach
- - - - + +
sexalatus worm
Sarcoptes scabiei Mange mite - - + - - -
Stephanurus dentatus Kidneyworm - - + + - +
Strongyloides ransomi Threadworm - - + + +/- +/-
Trichuris suis Whipworm - - - +/- + +

TABLES

ANTHELMINTICS AND PARASITICIDES 167


SWINE DISEASE MANUAL

Arthritis in neonatal and growing swine


In young pigs, arthritis is usually caused by infection, although trauma from handling, being laid on by the sow, and entrapment
in gating or equipment must also be ruled out. Arthritis in this age group is most commonly seen in the carpus (front knee),
rear hock, and claw. Though less frequently affected, the shoulder, stifle, and atlanto-occipital joints should also be examined.
Culture is often the only way to differentiate the various etiologic agents; sterile procedures should be used as some agents are
slow-growing or difficult to culture.

Age group
Diseases and agent affected Nature of lesions Signs, other lesions, comments
Erysipelas Growers through Rear hock, carpus, and toes most May occur during, or as a sequel
(Erysipelothrix adult, rarely in often affected. Joints are swollen to, an acute occurrence of systemic
rhusiopathiae) nursery age pigs. and very painful in both acute and erysipelas (diamond skin disease).
chronic phases. May also present more insidiously
as increased prevalence of swollen
joints at the herd level, without any
acute outbreak or mortality.
GlÄsser’s disease Nursery See “streptococcal arthritis” above, Frequently accompanied by
(Haemophilus (occasionally in although fibrinous response and serofibrinous or fibrinopurulent
parasuis) growers) periarticular inflammation may be pleuritis, pericarditis, and peritonitis
more dramatic. (or in combination), and meningitis.
Urgent treatment is required, or
death is likely. Important differential
for streptococcal arthritis.
Mycoplasmal arthritis Usually pigs from Excessive serosanguinous joint fluid Stiffness and shifting lameness in
8 to 24 weeks in acute phase, with yellowing of hind legs are common. Recovery can
(Mycoplasma
of age synovial membranes and pannus be protracted, though acute cases
hyosynoviae)
developing over time. should respond well to antibiotic
injections. Occasionally, young adults
(replacement gilts) are affected.
Mycoplasmal Usually pigs from Swollen joints, serosanguinous Abdominal tenderness with sternal
polyserositis/arthritis 3 to 10 weeks or serofibrinous synovial fluid in recumbency sometimes present,
(Mycoplasma hyorhinis) of age acute phase with pannus and joint related to pleuritis. Morbidity less
erosion developing in advanced than 25%, mortality usually low.
stages. Pleuritis, pericarditis, and
polyserositis frequently present.
Septic arthritis All ages One or more hot, swollen joints Risk factors in neonates include poor
(Pyogenic Streptococcus, in acute phase, with increased technique or dirty instruments used
Staphylococcus, turbidity of synovial fluid. during piglet processing (teeth, tails,
Trueperella, and other Periarticular fibrosis and pyogenic castration). Sometimes accompanied
endemic bacteria) abscesses in advanced cases. by fever, depression, and other
evidence of septicemia.
Streptococcal arthritis Nursery Acute suppurative polyarthritis; Painful, swollen joints, and fever.
(Streptococcus suis type 2) (occasionally in concurrent meningitis and Central nervous system signs
growers) polyserositis is common. Sometimes common when meningitis is present.
accompanied by valvular Urgent treatment is required,
endocarditis. See “GlÄsser’s disease”. or death is likely. Can be major
contributor to chronic arthritis on
some farms. Important differential for
TABLES

Glasser’s disease.

168 ARTHRITIS
TABLES

Central nervous system (CNS) diseases in swine:


comparative approach to diagnosis
In addition to the diseases described in this table, other less frequent causes of CNS signs include a host of other viruses (eg,
classical swine fever (CSF), porcine reproductive and respiratory syndrome virus (PRRSV), porcine circovirus associated disease
(PCVAD), paramyxovirus, Japanese encephalitis, rabies, etc) and toxicities (organophosphate, carbamate, chlorinated hydrocar-
bons) that are not included below. Diseases that include CNS signs, particularly when they occur at a group or herd level rather
than in sporadic individuals, need to be aggressively investigated. It is important to sample multiple pigs in various stages of the
disease and to include a complete set of fresh and fixed tissues when making a laboratory submission.

Disease Agent/Cause Clinical signs Diagnostic features and methods


Bacterial Various Most often observed in postweaning Histopathology of brain will confirm
meningitis bacteria, to early growing pigs, although suppurative meningoencephalitis. Culture
including any age can be affected. Acutely of brain, meningeal swabs, or cerebrospinal
Streptococcus affected pigs show incoordination, fluid will often result in identification of the
suis, blindness, deafness, nystagmus, organism. Polyarthritis, myocarditis, and
Haemophilus tremors, opisthotonus, convulsions, pneumonia very typically accompany the
parasuis, and unconsciousness, paddling, and are central nervous signs of the disease. Most
other sporadic often simply found suddenly dead. herds are endemically infected with all of
bacteria these agents, although sporadic outbreaks
(Streptococcus, can contribute 2% to 5% to group mortality.
E coli, Stress and viral infections such as porcine
Trueperella, reproductive and respiratory syndrome can
Salmonella, etc). exacerbate the frequency and severity of the
condition.
Edema disease Shigatoxin-like Usual occurrence is within 2 weeks Edema present in the eyelids, forehead,
enterotoxin after weaning. Clinical signs include stomach wall, and mesocolon is highly
elaborated by acute onset of ataxia, incoordination, suggestive of the diagnosis. Diagnosis
E coli, usually recumbency, and occasionally sometimes difficult to confirm with certainty
of a hemolytic perhaps edema of eyelids, face, nose, though culture of hemolytic E coli (with
colony-type. or lips. Most affected pigs die within confirmed genotype for shiga-like toxin)
2 days and some die peracutely. from the ileum/colon is useful. However,
Survivors can develop head tilt, histopathology that shows angiopathy with
incoordination, and unthriftiness. polioencephalomalacia and malacia in brain
stem is pathognomonic when present.
Hemagglutinating Betacoronavirus Assumed to be endemic in the US, Viral encephalitis is observed histologically
encephalomyelitis with symptomatic infections rare. In and by PCR, ISH, or isolation.
(HEV) naïve piglets less than 4 weeks of age,
vomiting, dehydration, wasting, coma,
and eventual death occur. Tremors,
convulsions, and paddling may occur.
Case mortality can approach 100%.
FA = fluorescent antibody; IHC = immunohistochemistry; ISH = in situ hybridization; PCR = polymerase chain reaction

Table continued on next page


TABLES

CNS DISEASES: COMPARATIVE APPROACH 169


SWINE DISEASE MANUAL

Table cont’d. Central nervous system (CNS) diseases in swine: comparative approach to diagnosis

Disease Agent/Cause Clinical signs Diagnostic features and methods


Pseudorabies Suid herpesvirus Piglets less than 3 weeks of age present In a naïve herd, one is likely to see signs
(PRV) 1, also known as with incoordination, ataxia, salivation, across all age groups; the disease is exotic
pseudorabies virus opisthotonus, seizures, recumbency with in US commercial industry. Diagnosis
(PRV) or Aujeszky’s paddling, or sudden death. Older pigs confirmed by detection of virus in tissues
disease virus can show similar, but less prevalent and (tonsil, brain, spleen, lung, lymph nodes)
severe, signs. Growing pigs and adults by FA, IHC, ISH, or PCR. Highly accurate
often present with acute respiratory serologic tests are available that can
signs; pregnant sows may abort. discriminate between vaccinated and
unvaccinated pigs.
Salt poisoning Sodium ion Acute onset, usually with numerous History of water restriction or deprivation
intoxication due to affected pigs due to a pen- or barn-level followed by unrestricted access to water.
water deprivation. out-of-water event; the condition can The condition can occur regardless of salt
occur after as little as 24 hours without level in the diet; the key feature of the
water. Affected pigs wander, appear disease is almost always water deprivation.
blind, head-press, dog-sit, experience The pathognomonic histologic lesion
periodic convulsions, become prostrate, is eosinophilic meningoencephalitis,
and eventually enter a coma with death sometimes with polioencephalomalacia.
almost inevitable. Brain tissue can be assessed for sodium
levels although this is not usually required
when adequate history is available.
Spinal Miscellaneous Primary signs from compromised Careful clinical examination, review of
myelitis, viral or bacterial spinal cord function may include herd history, and knowledge of recent
myelopathy, infections persistent or sporadic occurrence of management changes on the farm will aid
and/or (including tetanus ataxia, paralysis, or posterior paresis. in arriving at a diagnosis. Necropsy should
clinical signs or botulism), Morbidity can help in the diagnosis, as be guided by the observed clinical signs in
referable to toxins (selenium, infectious causes often affect multiple order to localize the site(s) of compromise.
spinal cord arsenicals, pigs simultaneously, while traumatic Histopathology on brain and spinal cord
organophosphates, causes usually involve only individual as well as liver, muscle, and other tissues
heavy metals), animals. Toxicities or deficiencies may should be undertaken. Appropriate
deficiencies have very high morbidity. Pigs with these antigen-based tests can be selected based
(B-vitamins, conditions often remain centrally aware on the situation and test availability.
minerals), of their environment and surroundings, Chemistry or toxin screens on liver and/or
trauma (injection while encephalitic diseases tend to be feed may be warranted.
sites, fractures, associated with compromised awareness.
electrocution), or
other insults.
Teschen or Teschoviruses Historically, Teschen disease outbreaks in Histopathology shows viral myelitis or
Talfan disease or similar Europe had high morbidity and mortality encephalomyelitis and presence of virus
picornaviruses in all age groups. The disease has changed demonstrated by isolation, PCR, or ISH.
(eg, Sapelovirus, over time and now usually presents with Picornaviruses are common and definitive
Enterovirus, low morbidity but high case mortality, the genotyping is required to confirm
Astrovirus, and result of polioencephalomalacia-related diagnosis. Teschen-like strains occur
others) ataxia and paralysis. The disease is exotic sporadically in the US while most herds are
to the US. endemically infected with low virulence
strains.
TABLES

FA = fluorescent antibody; IHC = immunohistochemistry; ISH = in situ hybridization; PCR = polymerase chain reaction

170 CNS DISEASES: COMPARATIVE APPROACH


TABLES

Diarrheal diseases common at various ages


Age is an important criterion when establishing a list of differential diagnoses for diarrhea in pigs. The table below compares
the likelihood of diarrhea being produced as a primary outcome of infection by different agents. It is important to remember
that diarrhea may accompany nonenteric diseases as a result of disruptions to the pig’s appetite, systemic illness and fever, or
chronicity. Always examine the entire animal when trying to establish a diagnosis.

Unweaned Grow/finish
Disease piglets Nursery pigs pigs Adults
Enterotoxigenic E coli ++++ +++ + (early -
grower)
Porcine rotavirus ++++ +++ + (early -
grower)
Clostridium difficile ++++ - - -
Clostridium perfringens type A ++++ - - -
Clostridium perfringens type C ++++ + (rare) - -
Coccidiosis (Cystoisospora suis or Eimeria species) ++++ ++ + -
Enteric salmonellosis (Salmonella Typhimurium, + ++++ ++++ +
others)
Septicemic salmonellosis (Salmonella Choleraesuis) + +++ +++ +
Swine dysentery (Brachyspira hyodysenteriae) + (rare) ++ ++++ ++
Proliferative enteropathy (Lawsonia intracellularis) + (rare) ++ ++++ ++
Whipworm infection (Trichuris suis) - ++ ++++ +++
Swine enteric coronavirus diseases ++++ +++ ++ +++

- Agent rarely causes diarrhea in age group.


+ to ++++ Relative increases in likelihood, severity, or prevalence of diarrhea in age group.

TABLES

DIARRHEAL DISEASES AT VARIOUS AGES 171


SWINE DISEASE MANUAL

Diarrheal diseases in swine: comparative approach to


diagnosis
Many disease processes in pigs can result in diarrhea. Fortunately, the age of the affected pigs, the appearance of the feces, and
the nature of the clinical signs, when combined with appropriate diagnostic tests, can usually help one determine the primary
cause of the diarrhea.

Disease and agent Ages affected Diagnostic features and methods


Rotaviral enteritis Neonates up to 6 weeks Nonfatal diarrhea with occasional vomiting. Microscopic segmental
(Rotavirus A, B, or C) of age. Group C more atrophic enteritis. Infection confirmed by detecting agent in feces of
common in neonates while acutely affected pigs (PCR, EM, or antigen-ELISA) or by positive stain (FA
group A more common or IHC) on small intestinal sections.
in late lactation and
postweaning.
Swine enteric coronavirus All age groups susceptible Acute epidemics produce severe diarrhea, rapid dehydration, and
diseases if previously unexposed. vomiting with nearly 100% mortality in neonatal piglets. Postweaned
(Porcine epidemic Most severe in piglets less pigs have profuse diarrhea, sometimes with yellowish color due to
diarrhea virus) than 4 weeks old. maldigestion and malabsorption. Sows have less severe diarrhea and may
vomit; systemic illness may lead to lethargy and inability to effectively
nurse litter. Endemic disease occurs when only partial immunity exists
in the herd and results in milder disease, but with similar clinical signs,
though with much lower mortality. Infection confirmed by histologic
lesions of severe atrophic enteritis and positive antigen staining (FA or
IHC), or by PCR on feces from acutely affected pigs.

Swine enteric coronavirus All age groups susceptible In the absence of partial cross-protective immunity from prior infection
diseases if previously unexposed. with porcine respiratory coronavirus, signs and diagnosis are as above
(Transmissible Most severe in piglets less for porcine epidemic diarrhea. Widespread endemic porcine respiratory
gastroenteritis virus) than 4 weeks old. coronavirus has mitigated the severity of transmissible gastroenteritis virus
over the last 30 years. Partial immunity results in milder clinical signs and
less severe lesions.
Swine enteric coronavirus All age groups susceptible Acute outbreaks present with diarrhea in all age pigs but with low
diseases if previously unexposed. mortality. Once herd immunity develops, clinical signs disappear.
(Porcine delta Infection confirmed by observation of segmental atrophic enteritis and
coronavirus) PCR on feces from acutely affected pigs.
Enteric colibacillosis Neonates through 2 to 4 Watery diarrhea and possibly vomiting; clinical signs most severe in
(Escherichia coli) weeks postweaning. neonates less than 1 week old and during the first 2 weeks postweaning.
Affected pigs have few gross lesions, although occasionally fibrinous
polyserositis can be seen in neonates. Microscopically, Gram-negative
rods will be seen attached to intestinal epithelium.Infection confirmed
by isolation of uniform population of E coli from small intestine; colony
type can be smooth, mucoid, or hemolytic. PCR can be used to confirm
presence of enterotoxins and pili.
Clostridial diarrhea Neonates from few hours Moderately severe diarrhea with variable morbidity and low mortality.
(Clostridium difficile) old to 10 days of age. Mesocolonic edema may be observed and colon often filled with
creamy feces. Histology reveals multifocal suppurative and erosive colitis.
Infection confirmed by identification of alpha or beta enterotoxins using
antigen-ELISA on fresh feces.
TABLES

PCR = polymerase chain reaction; EM = electron microscopy; antigen-ELISA = enzyme-linked immunosorbent assay;
FA = fluorescent antibody; IHC = immunohistochemistry

172 DIARRHEAL DISEASES: COMPARATIVE APPROACH


TABLES

Table cont’d. Diarrheal diseases in swine: comparative approach to diagnosis

Disease and agent Ages affected Diagnostic features and methods


Clostridial diarrhea Neonates 2 to 10 days Variably severe diarrhea with variable morbidity, low mortality,
(Clostridium perfringens of age. and no obvious gross lesions. Histology may show mild multifocal
type A) suppurative enteritis associated with abundant Gram-positive
rods.
Infection confirmed by ruling out other causes, in combination
with indicative histopathology, isolation, and genotyping of an
enterotoxigenic (beta-2 toxin) C perfringens type A.
Clostridial diarrhea Neonates from 8 hours Acute presentation may appear only as an unexplained death,
(Clostridium perfringens old to 3 weeks of age. sometimes accompanied by acute diarrhea (sometimes with
type C) hemorrhage). Small intestine will be segmentally hyperemic to
hemorrhagic grossly, with transmural intestinal necrosis with
hemorrhage and emphysema microscopically. Morbidity may
be sporadic but with high case fatality. Some piglets (perhaps
those recovering from the acute infection) develop a chronic
form of the disease characterized by diarrhea and ill-thrift.
Fibrinonecrotic enteritis and colitis are evident histologically.
Infection confirmed by appropriate histopathology and isolation
of C perfringens type C determined to be beta toxin positive by
PCR.
Enteric salmonellosis All ages susceptible if Acute diarrhea with bile-stained feces and loss of condition.
(Salmonella enterica previously unexposed, Lesions of necrotizing enteritis of large (and sometimes small)
serotype Typhimurium) although most common in intestine and enlarged mesenteric lymph nodes. Occasionally
nursery and growing pigs. observe congestion in lungs or other evidence of septicemia.
Chronic cases may have “button ulcers” in large intestine.
Infection confirmed by culture and identification of bacteria from
intestine or regional lymph nodes. Salmonella Typhimurium and
Salmonella Heidelberg most commonly associated with enteritis;
other salmonellae serotypes are commonly isolated but their role
in disease is often unclear.
Septicemic salmonellosis All ages susceptible if Cases may occur as an outbreak or sporadically in individuals.
(Salmonella enterica previously unexposed, Clinical signs of sepsis with red or purple skin lesions on ears, tail,
serotype Choleraesuis ) although most common in snout, feet, and abdomen, and dyspnea; diarrhea may not appear
nursery and growing pigs. until several days after the acute stage of disease. At necropsy,
congested lungs, splenomegaly, and hepatomegaly are common,
with enteritis lesions variably present. Infection confirmed by
evidence of necrotizing enterocolitis and culture of the bacterium
from intestine, lymph nodes, and systemic sites such as liver
and lung. Paratyphoid nodules in the liver are an important
microscopic feature of the disease.
Swine dysentery Postweaning to adult. Acutely affected pigs have mucohemorrhagic diarrhea. Gross
(Strongly beta-hemolytic lesions are limited to the large intestine, which presents with
Brachyspira species mucohemorrhagic to fibrinonecrotic typhlocolitis. Microscopic
including B hyodysenteriae, lesions are restricted to mucosal layer with organisms observed
B hampsonii, and in epithelium and crypts. Infection confirmed by isolation of a
B suanatina) strongly beta-hemolytic Brachyspira from mucosa or feces. PCR
can detect some isolates but not all.
TABLES

PCR = polymerase chain reaction; EM = electron microscopy; Ag-ELISA = enzyme-linked immunosorbent assay; FA = fluorescent antibody;
IHC = immunohistochemistry

Table continued on next page


DIARRHEAL DISEASES: COMPARATIVE APPROACH 173
SWINE DISEASE MANUAL

Table cont’d. Diarrheal diseases in swine: comparative approach to diagnosis

Disease and agent Ages affected Diagnostic features and methods


Spirochetal colitis Growing and finishing pigs. Can present as a sporadic or persistent mild nonhemorrhagic
(Weakly beta-hemolytic diarrhea, generally without other overt clinical signs; gross
Brachyspira species lesions are unremarkable. Microscopically, will see attachment
including B pilosicoli, of the organism to enterocytes in the colon, microerosions in
B murdochii, B intermedia, the epithelium, and nonsuppurative inflammation. Infection
and B innocens) is confirmed by rule-out of dysenteric Brachyspira species,
characteristic histopathology, and identification of the organism,
usually by PCR.
Proliferative enteritis Postweaning to adult. In its subclinical form, disease may only be evident by poor
(Lawsonia intracellularis) performance or weight loss. Acute forms of the disease are
accompanied by hemorrhagic diarrhea, hemorrhage in the
intestine (usually clotted), and thickened intestinal mucosa of the
distal small intestine and colon. Chronic infections are typified
by fibrinonecrotic enteritis and/or colitis with mucosal thickening
due to crypt enterocyte hyperplasia and inflammation. Infection
confirmed by observation of typical gross and microscopic lesions
and presence of L intracellularis demonstrated by IHC or PCR.
Coccidiosis From 5 to 25 days of age. Sporadic diarrhea and wasting, pigs may be recruited in waves.
(Cystoisospora suis) Microscopic lesions include atrophic enteritis, sometimes with
segmental fibrinonecrotic enteritis in caudal small intestinal
sections. Infection confirmed by observation of immature stages
of Cystoisospora suis within the epithelium. The severity of
coccidiosis is highly dose- and age-dependent.
Coccidiosis Susceptible naïve pigs Enterocolitis with organisms visible in the intestinal or colonic
(Eimeria species) postweaning. epithelium by microscopy. Infection confirmed by observation
of immature stages of Eimeria within the gut epithelium. Fecal
flotations may demonstrate high numbers of oocysts, but this is
not sufficient for a diagnosis. PCR generally not available. The
severity of coccidiosis is highly dose- and age-dependent.
Whipworm infection Postweaning to adult. Mucoid or mucohemorrhagic diarrhea with loss of condition
(Trichuris suis) is suggestive of the disease. Inflammatory nodules, often with
protruding parasites, can be seen in mucosa of large intestine.
Large numbers of parasites are not necessary to create severe
disease. Clinical signs are very similar to those produced with
swine dysentery; the two diseases can occur together. Prepatent
period is around 8 weeks and eggs are shed intermittently.
Infection is confirmed by visualization of the parasite or ova and is
supported by histopathology and clinical signs.

PCR = polymerase chain reaction; EM = electron microscopy; Ag-ELISA = enzyme-linked immunosorbent assay; FA = fluorescent antibody;
IHC = immunohistochemistry
TABLES

174 DIARRHEAL DISEASES: COMPARATIVE APPROACH


TABLES

Internal parasites
Although much of modern swine production now takes place in indoor facilities and over fully perforated flooring, parasites
seem to persist on many farms. Eradication is rarely an option, so long-term control strategies, usually based on periodic treat-
ment with anthelmintics, are required. An effort should be made to understand what parasites are present on a farm and when
(and from where) the infection is occurring, so that the best value can be derived from a control program.

Disease and agent Usual location and size of adult Life cycle Mode of infection Treatment
Ascariasis Small intestine, bile duct, and Direct Orally from AVE, DCV,
(Ascaris suum) stomach contaminated FBZ, LVM, PIP,
150 to 300 mm environment. PRT
Coccidiosis Caudal half of small intestine Direct Orally from Totrazuril
(Cystoisospora suis) 20 µm diameter contaminated or related
environment. compounds
Cysticercosis Cysticerci form in skeletal and cardiac Indirect; pig is the Orally from None
(Taenia solium) muscles. intermediate host environment
Cysts range from 10 to 20 mm for this human contaminated with
tapeworm. human sewage.
Esophageal worm Sub-epithelium of esophagus Indirect; requires Ingestion of beetle AVE, FBZ, LVM
(Gongylonema and in the tongue dung beetles. with encysted larvae.
pulchrum) 30 to 60 mm
Hookworm Small intestine of feral and pastured Direct Cutaneous contact AVE, DCV,
(Globocephalus swine in southern US. with larvae. FBZ, LVM
urosubulatus) 6 to 8 mm
Kidney worm Large adults in perirenal tissues, Indirect; requires Consume earthworms AVE, FBZ, LVM
(Stephanurus kidney, and ureters. Juveniles can be earthworms. Direct; carrying larvae; some
dentatus) found in ectopic sites. some evidence evidence larvae can
20 to 50 mm exists. penetrate skin of pigs.
Liver flukes Liver of pigs on pastures used by Indirect; requires Orally from None
(Fasciola hepatica) sheep. snails. contaminated
30 mm environment.
Lungworm Edge of diaphragmatic lobes of lung Indirect; requires Consume earthworms AVE, FBZ, LVM
(Metastrongylus 20 to 50 mm earthworms. Direct; carrying larvae.
species) some evidence
exists.
Mycoplasma suis On erythrocytes Unknown Uncertain; Tetracyclines
0.8 to 1.0 µm diameter transplacental, direct
contact, insect bites,
and contaminated
instruments have
been implicated.
Red stomach worm Stomach Direct Orally from AVE, DCV, FBZ
(Hyostrongylus Less than 10 mm contaminated
rubidus) environment.

AVE = avermectin; DCV = dichlorvos; FBZ = fenbendazole; LVM = levamisole; PIP = piperazine; PRT = pyrantel.
TABLES

Table continued on next page

INTERNAL PARASITES 175


SWINE DISEASE MANUAL

Table cont’d. Internal parasites

Disease and agent Usual location and size of adult Life cycle Mode of infection Treatment
Stomach worms Stomach Indirect; requires Ingestion of beetles DCV, FBZ
(Ascarops strongylina and 10 to 20 mm dung beetles. with encysted larvae.
Physocephalus sexalatus)
Thorny-headed worm Small intestine (usually ileum) Direct Prenatal, AVE, LVM
(Macracanthorhynchus Up to 30 cm transcolostral, oral,
hirudinaceus) or cutaneous contact
with larvae.
Threadworm Small intestine of suckling pigs Direct Prenatal, AVE, DCV, FBZ,
(Strongyloides ransomi) 30 to 45 mm transcolostral, oral, LVM
or cutaneous contact
with larvae.
Trichinosis Muscle of many mammals, Many carnivorous Ingest muscle Not practical
(Trichinella spiralis) especially swine and bears. or omnivorous containing encysted
Adults 2 to 4 mm, encysted mammals. larvae (raw garbage,
larvae animal carcasses,
0.8 to 1.3 mm rats, mice, cats, or
cannibalism)
Whipworm Cecum and large intestine Direct Orally from DCV, FBZ, LVM
(Trichuris suis) 50 to 80 mm contaminated
environment.
AVE = avermectin; DCV = dichlorvos; FBZ = fenbendazole; LVM = levamisole; PIP = piperazine; PRT = pyrantel.
TABLES

176 INTERNAL PARASITES


TABLES

Intestinal hemorrhage: differential diagnosis


The listed diseases are usually seen in postweaning and growing pigs. Intestinal hemorrhage in neonatal pigs is very rare; in
the US it is usually associated only with Clostridium perfringens type C. The list of diseases and conditions that cause intestinal
hemorrhage is limited, so a logical approach to diagnosis that incorporates knowledge about the clinical signs, the location and
character of the hemorrhage, and the epidemiology and history of the outbreak, combined with the rational use of diagnostic
support, will usually identify the cause.

Enlargement
Disease Cecal and of ileocecal Diagnostic features
and agent Ileal lesions colonic lesions lymph nodes and methods
Enteric Usually mild to Severe, segmental or Mild to severe, Bile-stained feces with fibrin
salmonellosis moderate severity, with multifocal, fibrinous to often with common. Some serotypes may
(Salmonella segmental fibrinous or fibrinonecrotic colitis. edema and produce necrotic hepatic foci
enterica serotype necrotic exudates. congestion. or pneumonia.
Typhimurium)
Gastric ulcer Dark, tar-like blood Dark, tar-like blood None. Pig (or carcass) will usually
(Multiple risk (melena) in intestinal (melena) in lumen. No be pale. Ulcer may bleed
factors have been lumen. No pathology pathology present. intermittently over days to
described) present. weeks or the pig may bleed out
acutely if ulcer affects a major
vessel in the gastric wall. The
pars esophagea of the stomach
should always be examined for
the presence of an ulcer.
Hemorrhagic Transmural congestion Colon may have bloody Sometimes Cause unknown but must be
bowel syndrome of intestine contents from pathology mild edema differentiated from intestinal
(Exact cause and accompanied by that occurred in the and congestion volvulus through careful
pathogenesis unclotted blood or small intestine but no present. necropsy.
unknown) bloody fluid in the overt pathology present
lumen. in tissue.
Intestinal (or Entire intestine dilated No mucosal lesions. Sometimes Sporadic occurrence with rapid
stomach) volvulus with bloody fluid and Contents may be edema in death of healthy pigs. Typically,
or torsion gas, without mucosal normal, or bloody fluid lymph nodes. affected pigs will have feed in
lesions. With intestinal and gas may be present stomach (highly unusual for ill
volvulus, the proximal similar to small intestine. pigs). Twist may be observed
duodenum may be in the mesentery or mesenteric
normal with abrupt root, and abdominal organs
transition to congestion are often displaced. It can
at caudal flexure. be surprisingly difficult to
confirm what has occurred and
careful necropsy is required.
This condition is an important
differential for hemorrhagic
bowel syndrome.

Table continued on next page


TABLES

INTESTINAL HEMORRHAGE: DIFFERENTIAL DIAGNOSIS 177


SWINE DISEASE MANUAL

Table cont’d. Intestinal hemorrhage: differential diagnosis

Enlargement
Disease Cecal and of ileocecal Diagnostic features
and agent Ileal lesions colonic lesions lymph nodes and methods
Proliferative Mild to severe, gross and Variably present and Mild to severe Ileum may be rigid and hose-
enteritis microscopic proliferative much less common with segmental like. Clinical cases often occur
(Lawsonia changes in mucosa. than ileal lesions. When mesenteric sporadically in herd, although
intracellularis) Frank blood (often changes are present they edema small outbreaks do occur. The
clotted) can be variably can range from mild to sometimes condition frequently exists at a
present. Chronic cases severe and are restricted present. subclinical level.
often accompanied by to the proximal cecum/
fibrinonecrotic changes colon.
to mucosa.
Swine dysentery None. Severe, diffuse, Mild or absent. Mucohemorrhagic feces are
(Strongly mucohemorrhagic very common. Blood in feces is
beta-hemolytic colitis; fibrin visible in often fresh, red, and mixed with
Brachyspira chronic stages. mucus.
species including
B hyodysenteriae,
B hampsonii, and
B suanatina)
Vitamin K May contain unclotted No mucosal lesions in None. Petechial or ecchymotic
deficiency blood. No lesions cecum or colon. hemorrhage at other systemic
(warfarin toxicity) present in mucosa. locations.
TABLES

178 INTESTINAL HEMORRHAGE: DIFFERENTIAL DIAGNOSIS


TABLES

Mycoplasma-related diseases
The following table consolidates the key differentiating features of each of the 4 mycoplasmal diseases that occur on US swine
farms, so the reader can clearly understand how each needs to be approached differently with regard to prevention, diagnosis,
and control.

Disease and agent Signs and lesions Diagnostic features and methods
Mycoplasmal arthritis Age: Usually 10- to 24-week-old pigs. History, signs, and lesions are suggestive.
(Mycoplasma May respond well to lincomycin, tylosin, or
Clinical: Lameness, difficulty in arising, reluctant
hyosynoviae) tetracycline injection. Histopathology and
to walk and kneeling in some. Often in straight-
detection of organism in joints by PCR or
legged, muscular pigs with hidden joint swellings.
isolation from acutely infected pigs. Needs
Serofibrinous, blood-tinged synovial fluid with
to be differentiated from Haemophilus
synovial membranes swollen.
parasuis, Streptococcus suis, erysipelas, and
Lesions: Arthritis with serofibrinous effusion and osteochondrosis dessicans.
grossly visible villous proliferation of synovium.
Mycoplasmal Age: Three weeks old to adults, especially Often diagnosed on the basis of clinical signs
pneumonia or enzootic 2 to 4 months. and lung lesions at necropsy or slaughter.
pneumonia Confirm disease by demonstrating organisms
Clinical: Persistent dry cough, dyspnea with
(Mycoplasma by IHC, ISH or FA in lesions. Confirm presence
flare-ups of pneumonia. Good appetite but
hyopneumoniae) by PCR on lung or airway swabs. Difficult to
unthrifty.
culture. Serology (ELISA, CF) can be used
Lesions: Clearly demarcated cranioventral for herd diagnosis. Improved environment,
lobules with grey consolidation and reduced stocking density and vaccination can
mucopurulent exudate in airways. Morbidity help in control.
variable/mortality low. Histologically have
marked lymphoid hyperplasia around airways,
often with mucopurulent exudate in airways and
adjacent alveoli.
Mycoplasmal Age: Usually in 3- to 10-week-old pigs, Isolation or PCR from acute septic lesions
polyserositis and sometimes older. at systemic sites. Morbidity 20% to 35%
arthritis but mortality low. Infection may persist in
Clinical: Acutely affected pigs visibly ill and with
(Mycoplasma hyorhinis) joints, but many lame pigs slowly recover but
fever. Abdominal tenderness with preference
fibrinous adhesions and poor gains persist.
for sternal recumbency. Lameness. Sometimes
Needs to be differentiated from Haemophilus
dyspnea.
parasuis, Streptococcus suis, and Actinobacillus
Lesions: Serofibrinous pericarditis, pleuritis, and suis. Mycoplasma hyorhinis does not cause
peritonitis. Later there may be fibrous adhesions. meningitis or central nervous system signs.
Arthritis often severe in one or more joints.
Later, villous hyperplasia, possibly pannus may
occur.
Mycoplasma suis Age: Any age; usually in suckling or nursery pigs, History and clinical signs are usually not specific
or in sows. enough to be helpful in establishing a diagnosis.
Anemia may be present and the organism may
Clinical: Range from vague signs to pallor,
be detected by PCR in blood or splenic tissue.
anemia, unthriftiness, dysgalactia, skin cyanosis,
Organisms can be visualized by microscopy in
fever, and icterus.
blood smears. Serology useful on herd basis
Lesions: Anemia, icterus, and splenic but is not widely available. The historic name of
enlargement. this disease is eperythrozoonosis.
TABLES

ELISA = enzyme-linked immunosorbent assay; IHC = immunohistochemistry; CF = complement fixation; FA = fluorescent antibody;
PCR = polymerase chain reaction.

MYCOPLASMA-RELATED DISEASES 179


SWINE DISEASE MANUAL

Reproduction: causes of low farrowing rate


Low farrowing rate can be a challenging problem to solve on farms. Its occurrence can be insidious rather than sudden and
the cause is often multifactorial in nature. The table below provides a framework to assist in ensuring one looks broadly at all
potential contributors to a farrowing rate problem rather than focusing only on infectious etiologies.

Conception failure
Regular returns to estrus (19 to 24 days)
Risk factor Cause Comments
Management Number of matings per service
Mating quality and technique
Estrus detection and timing of mating
Artificial insemination Semen viability, temperature, AI technique, dose
Natural service Boar semen quality, copulation, blood/trauma
Gilt/Sow factors Infectious diseases Urogenital (ascending) infections, systemic disease
Farrowing interval Lactation length, wean-to-service interval
Parity Age and weight at first mating
Body condition Critical factor to manage during lactation
Boar factors Age, genotype, season, temperature Genotype, libido, ambient temperature too high
Infectious diseases Reduced sperm count due to systemic disease,
Trauma, blood in semen remember delayed effect due to spermatogenic cycle

Pregnancy failure
Irregular returns to estrus (greater than 25 days)
Risk factor Cause Comments
Gilt/Sow factors Infectious diseases Ascending or systemic infections
Gilt acclimatization Inadequate acclimatization of gilts (vaccine and
exposure)
Endocrine dysfunction Primary dysfunction of endocrine organ
Parity Age and weight at first mating
Feed Mycotoxins Zearalenone, phytoestrogens, others
Feed additives Carbadox
Dietary micronutrients Vitamins, trace minerals
Nutrient balance and intake Amino acids, energy, body condition
Environment Season Seasonal infertility syndrome (summer and fall matings)
Temperature Excessively cold or hot
Housing Fighting and stress due to commingling groups, trauma
Movement or commingling Embryo loss surrounding implantation (10 to 25 days
postmating postmating)
TABLES

180 REPRODUCTION: CAUSES OF LOW FARROWING RATE


TABLES

Reproductive failure: infectious causes


In addition to the most common infectious causes of reproductive failure described in the table below, many other diseases
can contribute directly to reproductive failure or indirectly through the effects of systemic disease processes. These diseases
include, but are not limited to, influenza A, classical swine fever, African swine fever, Japanese encephalitis, and virulent
enteroviral infections. Sporadic abortions may also occur from bacterial infections such as erysipelas, Streptococcus, Salmonella,
Trueperella, and others by either systemic or ascending urogenital routes (through both artificial insemination and natural
service). Any acute generalized illness that occurs during advanced pregnancy can lead to abortion.

Disease and agent Signs and lesions Diagnostic features and methods
Brucellosis Very uncommon disease in most commercial Disease is present in some feral swine in the
(Brucella suis) industries; it has been essentially eliminated US; these serve as a reservoir for the infection.
from the US indoor-housed commercial Brucellosis is a serious zoonosis and cases of
industry. When exposure does occur, infertility the disease occur in feral pig hunters every
is the most common outcome, although any year. Diagnosis of the disease in pigs is typically
manifestation of reproductive failure may occur. achieved through serology and sometimes by
Abortion can occur at any stage of pregnancy. bacterial culture.
In boars, orchitis may develop as a result of the
infection.
Leptospirosis During an acute outbreak in a naïve herd, After recovery from infection (usually as a
(Leptospira species) abortions and stillbirths can occur, particularly result of antibiotic therapy), subsequent
during late pregnancy. Piglets from infected mating and reproduction return to normal.
dams are often weak and many will die within a Leptospirosis can also cause nephritis and
few days. Adult sows often show few other signs hepatitis in nonpregnant pigs. Diagnosis in
of disease. an infected sow or litter may require use of
several testing strategies, including paired
serology, culture, PCR, or tissue staining.
Porcine circovirus A common virus in swine around the world Serology not useful for diagnosing virus
associated disease that is associated with sporadic outbreaks involvement in reproductive disease. Virus
(Porcine circovirus type of increased fetal death, mummies, or dams should be identified by PCR, ISH, or FA in
2 and type 3) confirmed pregnant but failing to farrow. fetal tissues to confirm the role of the virus in
Reproductive failure as a result of infection with disease.
these viruses is relatively uncommon compared
to their impact on performance and disease of
postweaning pigs.
Vary with gestational stage at infection. Disease Parvovirus is a major cause of embryonic and
Porcine parvovirus is most common in gilts. Infection increases fetal death, particularly in gilts. Diagnosis can
(Porcine parvovirus 1) number of mummies, returns to estrus, failure be made by PCR or FA staining of fetal lungs or
to farrow, stillbirths, and pseudopregnancy. hearts collected from mummified fetuses less
Net result is fewer pigs born alive per litter. than 16 cm in length.
Abortions or fetal anomalies are not a feature.
Affected dams remain asymptomatic.

IHC = immunohistochemistry; FA = fluorescent antibody; PCR = polymerase chain reaction; ISH = in situ hybridization.

Table continued on next page


TABLES

REPRODUCTIVE FAILURE: INFECTIOUS CAUSES 181


SWINE DISEASE MANUAL

Table cont’d. Reproductive failure: infectious causes

Disease and agent Signs and lesions Diagnostic features and methods
Porcine reproductive and Acutely infected sows may abort, or abortion In addition to reproductive disease in the
respiratory syndrome may occur from fetal infection several weeks breeding herd, respiratory signs and illness
(Porcine reproductive and after sows are infected. Decreased conception may occur in any age group. PCR testing
respiratory syndrome virus and farrowing rates during and just after of serum collected from acutely ill sows
type 1 and type 2) an outbreak are common. At a herd level, (especially if sows are ill) and tissue from
outbreaks significantly increase numbers of aborted or weak-born piglets can confirm
stillbirths, weak pigs, mummies, premature diagnosis. Depending on stage of infection,
farrowings, third trimester abortions, and both sample types (dam serum and piglet
preweaning deaths. In well-managed outbreaks, tissue) are often necessary to confirm
herd immunity can occur, with improvements diagnosis. Serology on convalescent sows is
expected in 3 to 5 months. useful if previous infection status of the herd
and/or vaccination status are known.
Pseudorabies Clinical signs depend on the gestational stage If endemic, reproductive signs less common
(Suid herpesvirus 1, also at time of infection. Dams infected in the first than central nervous signs or respiratory signs
known as pseudorabies trimester may absorb their fetuses and return in growing pigs. Diagnosis can be achieved
virus or Aujeszky’s disease to estrus; those in second or third trimester may through serology, virus isolation, IHC, PCR.
virus) abort or have increased mummified, stillborn, Pseudorabies has been eradicated from the US
or weak pigs. Dams infected near term may have commercial industry, although the disease still
congenitally infected piglets that die within a few occurs in feral pigs in parts of the country.
days; central nervous signs or sudden deaths are
common in these pigs. Recovered (or vaccinated)
sows are protected from clinical signs.

IHC = immunohistochemistry; FA = fluorescent antibody; PCR = polymerase chain reaction; ISH = in situ hybridization.
TABLES

182 REPRODUCTIVE FAILURE: INFECTIOUS CAUSES


TABLES

Respiratory disease in swine: comparative approach


to diagnosis
Respiratory disease is perhaps the most common malady faced by swine producers and veterinarians. The term porcine respi-
ratory disease complex aptly describes the multifactorial nature of most respiratory disease problems that are encountered. In
addition to management factors that may contribute to the occurrence or severity of the problem, the table below provides a
convenient comparative description of the most common infectious agents that are likely to be encountered. Not included are
Ascaris suum and lungworms, both of which can cause acute respiratory clinical signs when acute heavy exposures occur.

Disease Agent/Cause Clinical signs Diagnostic features and methods


Atrophic rhinitis Bordetella Sneezing, snorting, nasal discharge, Variable degrees of turbinate atrophy and
bronchiseptica and epistaxis (severe cases). Tear snout deviation. Secondary pneumonia
and/or toxigenic staining from the medial canthi of often present but related to other agents.
Pasteurella the eyes is very characteristic of the History, signs, and typical lesions (often
multocida disease. Coughing is not a feature of noted during slaughter check) may be
the disease. In endemically infected sufficient for a diagnosis. When assessing
herds it is usual to see deviation of risk of atrophic rhinitis in purchased stock,
the snout in at least some pigs. Most culture of nasal swabs or turbinates followed
modern production systems have by toxigenicity testing by PCR may be
eliminated the disease. necessary.
Inclusion body Porcine Sneezing, nasal discharge or Grossly, may see only moderate clear to
rhinitis cytomegalovirus occlusion, and mouth-breathing mucopurulent nasal discharge. Microscopic
in young piglets 3 to 6 weeks of evidence of rhinitis with edema, petechial
age. Morbidity can be very high hemorrhages, and mucopurulent nasal
but mortality due to the disease is exudate. Signs and lesions are nearly
negligible. In most cases, the disease pathognomonic, especially when
has only a minor and very transient characteristic intranuclear inclusions in the
effect on gain and does not appear to turbinates or Harderian glands are noted.
predispose the pig to other diseases.
Influenza A Influenza A virus of In acute outbreaks, signs include Multifocal to diffuse, red, firm areas of
swine (IAV-S) sudden onset and rapid spread pneumonia, although characteristically
of fever, serous nasal discharge, there is a lobular cranioventral distribution
and dyspnea with high morbidity. of gross lesions. Secondary bacterial
Prostration and coughing, sometimes bronchopneumonia is common. History
severe, follow. Most pigs recover in and clinical signs are very suggestive of the
7 to 10 days. In large populations, acute form though less helpful in chronic/
perhaps in combination with viral endemic situations. Morbidity is typically
evolution, endemic infections in some very high and, unless complicated by
herds are characterized by a more other diseases, mortality tends to be low.
insidious onset and clinical disease that Diagnostic tests are widely available, with
occurs only in some subpopulations most cases confirmed by PCR on lung tissue
within the herd or barn at any given or nasal swabs. Antigen staining of lung
time. Subtypes H1N1 and H3N2 tissue using IHC or FA is a common and
are most common in the US though reliable technique for diagnosis. Subtyping
ongoing evolution and interspecies (by PCR and/or sequencing) is an important
transmission is well recognized. monitoring tool. Serology by ELISA is also
available.

ELISA = enzyme-linked immunosorbent assay; IHC = immunohistochemistry; FA = fluorescent antibody; PCR = polymerase chain reaction;
TABLES

ISH = in situ hybridization; VI = virus isolation.

Table continued on next page

RESPIRATORY DISEASE: COMPARATIVE APPROACH 183


SWINE DISEASE MANUAL

Table cont’d. Respiratory disease in swine: comparative approach to diagnosis

Disease Agent/Cause Clinical signs Diagnostic features and methods


Mycoplasmal Mycoplasma Persistent, dry cough and slow Grey-to-purple pneumonia and atelectasis
pneumonia hyopneumoniae growth, typically at a high in cranioventral lobules. Marked lymphoid
prevalence but negligible mortality. hyperplasia around airways. History,
The disease is endemic in nearly signs, and lesions are very suggestive of
all herds unless specific steps have the disease. Antigen staining (FA, IHC) on
been undertaken to eliminate the affected lung or PCR on lung or airway
infection. swabs to confirm infection. Serology
has some value as an epidemiological or
monitoring tool on a herd basis.
Pneumonic Pasteurella Coughing, dyspnea, fever, and Fibrinous bronchopneumonia seen
pasteurellosis multocida prostration. cranioventrally but often extending into
diaphragmatic lobes. Pleuritis, adhesions,
and abscesses are variably present.
Pasteurella multocida can be readily isolated
from lung lesions and is often a component
of porcine respiratory disease complex.
Pleuropneumonia Actinobacillus In acute outbreaks, pigs show high Necrohemorrhagic pneumonia with
pleuropneumoniae fever, prostration, dyspnea, and fibrinous pleuritis. Chronic cases have
mouth-breathing, usually with high pleural adhesions, often accompanied by
morbidity and mortality. Dead pigs necrotic foci in the lung. Lesions tend to be
often present with blood-tinged most common in diaphragmatic lobes. Signs
foam coming from nose. Many pigs and lesions are highly suggestive of the
that recover from acute illness will diagnosis, although the agent can usually
continue to present with a chronic be readily cultured or detected by PCR.
cough and appear unthrifty. In Serotyping of the organism is important in
endemically infected herds, the developing appropriate control strategies
disease may simply contribute to and for biosecurity monitoring. Serological
poor grower-finisher performance tests are available but can be quite
(with sporadic outbreaks of the serotype specific – negative test results are
acute form) and complaints of not definitive.
high levels of pleuritis by the
abattoir. Disease severity can vary
substantially based on the strain or
serotype of A pleuropneumoniae
that is present.
ELISA = enzyme-linked immunosorbent assay; IHC = immunohistochemistry; FA = fluorescent antibody; PCR = polymerase chain reaction;
ISH = in situ hybridization; VI = virus isolation.
TABLES

184 RESPIRATORY DISEASE: COMPARATIVE APPROACH


TABLES

Table cont’d. Respiratory disease in swine: comparative approach to diagnosis

Disease Agent/Cause Clinical signs Diagnostic features and methods


Porcine Porcine Causes reproductive failure in In younger pigs, focal or diffuse interstitial
reproductive reproductive sows and respiratory disease in all pneumonia is common in any or all lobes,
and respiratory and respiratory ages. High preweaning mortality often complicated by bronchopneumonia
syndrome syndrome virus is common after outbreaks in due to secondary bacterial infection.
type 1 and type 2 a breeding herd. Respiratory Thoracic lymph nodes (and others) are
disease is accompanied by fever often enlarged. Numerous diagnostic tests
and dyspnea, which can persist for are available, although PCR on tissues or
weeks. The disease is a frequent and serum and histopathology with antigen
important contributor to porcine stains (IHC, FA, ISH) are in most common
respiratory disease complex. use. The virus is generally easy to isolate.
Genotyping of the strain involved in an
outbreak or circulating endemically on a
farm is common and epidemiologically
useful.
Pseudorabies Suid herpesvirus 1 Central nervous system signs and Characteristic white, necrotic foci in liver
death predominate in neonates. and spleen occur in suckling piglets. Rhinitis,
Respiratory disease tends to tracheitis, tonsillitis, and keratoconjunctivitis
predominate in swine greater can be seen in postweaning pigs. Often
than 3 weeks old with sneezing, there is nonsuppurative encephalitis,
coughing, nasal discharge, dyspnea, tracheitis, bronchiolitis, and alveolitis
and occasionally seizures or other that accompany the respiratory disease,
central nervous signs. The disease sometimes with grossly visible necrotic
in growing-finishing pigs can be pharyngitis, laryngitis, and tracheitis.
clinically indistinguishable from Infection can be confirmed by FA, VI, or
influenza A. PCR on tonsil, spleen or brain, and by
histopathology with antigen staining (IHC).
Serological tests that can differentiate
vaccine from wild-type virus are a critical
part of eradication programs. The US
commercial industry is free of the disease
but feral pigs in some areas are endemically
infected.

ELISA = enzyme-linked immunosorbent assay; IHC = immunohistochemistry; FA = fluorescent antibody; PCR = polymerase chain reaction;
ISH = in situ hybridization; VI = virus isolation.

TABLES

RESPIRATORY DISEASE: COMPARATIVE APPROACH 185


SWINE DISEASE MANUAL

Skin lesions: differential diagnosis


Skin lesions can be readily diagnosed in most instances if one takes into account the history, epidemiology, and clinical signs asso-
ciated with the presenting disease. In many cases, diagnostic testing may not be required. However, when a condition is occurring
at a high prevalence, diagnostic testing is warranted. Remember to examine the entire pig when faced with a disease process that
is presenting primarily as a skin problem. The skin lesion may simply be a symptom of a more significant systemic illness.

Disease and agent Signs and lesions Diagnostic features and methods
Actinobacillosis Sporadic cause of raised, red, rhomboid-shaped Signs and internal and cutaneous lesions can
(Actinobacillus suis)
lesions in growing and mature pigs. (rarely) resemble those of acute erysipelas.
Aural hematoma Hemorrhage and serous exudate between skin Condition usually a result of some trauma to the
(Trauma) and cartilage of ear pinna. Over days to weeks, ear including bites, violent head shaking,
fluid accumulation tends to resolve, resulting in a fighting from commingling, and mange (itching).
thickened and deformed ear. Lesions can be very
large and appear to be quite painful during the
acute stages.
Dermatosis vegetans Rare and semilethal hereditary defect. Coronary Defect originally detected in Landrace breed but
(Congenital defect) band lesion and hoof deformities on one or currently there is less breed predisposition due
more feet of neonate initially appear as red to evolution of composite breeds/genetics. Most
macules but expand and become papillomatous affected piglets die within a few weeks
over a few weeks. although pigs that do survive develop
interstitial pneumonia containing characteristic
multinucleate giant cells.
Ear necrosis Areas of ulceration, infection, and necrosis on Infection occasionally results in septicemia and
(Multiple causes) tips or margins of ear(s). Pathogenesis poorly death but more generally the condition appears
understood, and it is unlikely there is a single to have minimal effect on the pig.
cause; risk factors are not well identified.
Microscopic vasculitis with bacterial infection is
observed.
Epitheliogenesis Missing areas of skin at birth, often over back, Small lesions often heal by scarring, although most
imperfecta rump, or mouth. affected pigs tend to succumb to
(Congenital defect) secondary effects of the condition.
Erysipelas Acute outbreaks characterized by appearance Acute skin lesions usually resolve without issue if
(Erysipelothrix of red, raised, rhomboid-shaped lesions on skin, animal is treated promptly for the systemic disease.
rhusiopathiae) often accompanied by high fever and joint pain.
As lesions resolve, necrotic plaques or scabs peel
off. Occasionally see necrosis of ear and tail tips.
Exudative Brownish exudative lesions on skin of axilla and Triggering factors are poorly understood for
epidermitis groin but spreading to head, face, and other this toxin-mediated disease. Risk factors include
(Staphylococcus areas. Severe cases can involve almost the entire irritation or wounds to the skin which precede the
hyicus) body and develop over a few days to a week. lesion and likely provide a niche for the
Pigs tend to be systemically ill with low fever, bacterial infection to establish. Treatment is
lethargy, and poor growth. seldom satisfactory.
Lice Intense rubbing and scratching behavior. Eggs Chronic infestations will lead to unthriftiness and
(Haematopinus suis) can be seen attached to hair on jowls, flanks, anemia. Lice are easy to see and are just a bit
inner surface of legs, and behind ears. Anemia smaller than a housefly.
can develop with severe infestations.
TABLES

186 SKIN LESIONS: DIFFERENTIAL DIAGNOSIS


TABLES

Table cont’d. Skin lesions: differential diagnosis

Disease and agent Signs and lesions Diagnostic features and methods
Photosensitization White areas of skin inflamed and pruritic. Lesions Many plants and drugs are photodynamic.
(Photodynamic often become necrotic over a few days to weeks, Conjunctivitis may accompany the dermatitis.
agents) followed by peeling. Sunlight is required for the lesions to develop,
although pigs do not need to be housed
exclusively outdoors to receive adequate
exposure.
Pityriasis rosea Raised, expanding, nonexudative, circular lesions Lesions resolve within a few weeks to months
(Unknown) in 6- to 14-week-old pigs. Lesions usually on without treatment. Disease may go unnoticed
ventral abdomen and inguinal areas. Lesion unless ventrum is examined. Lesions are not
margins look quite inflamed but do not appear pruritic. Affected pigs do not seem to suffer from
to be painful. any ill effects due to pityriasis rosea.
Porcine dermatitis Red to purple, circular, 1-cm-diameter Diagnosis by recognition of typical skin lesions
and nephropathy lesions appear on perineal, rump, and ventral and histopathology on skin and kidney. Usually
syndrome regions. Lesions may enlarge and coalesce. The sporadic but morbidity can approach 10%. No
(immune-mediated) accompanying nephropathy may lead to death in treatment. Condition appears to be a sequel
days to weeks. to some systemic viral infections, notably
porcine circovirus type 2 and type 3, porcine
reproductive and respiratory syndrome virus, and
classical swine fever.
Pustular dermatitis Pustules on ventral abdomen, inguinal area, Most pigs recover in a few weeks. Risk factors
(Streptococcus inner thighs, eyes, lips, or ears. Pustules rupture for the condition are poorly understood but
species) and are then replaced by black scabs. Several the lesions are likely a result of both traumatic
bacteria have been implicated but Lancefield skin wounds and hematogenous spread from
group C Streptococcus species are most common. other bacteremias (eg, tail-docking wounds,
A specific condition called “facial pyoderma” can umbilical infections, etc). Providing clean bedding,
develop on the cheeks and jowls of neonates improving sanitation, and using good hygiene will
from wounds caused by fighting among limit occurrence. Recovery can be hastened by
littermates. antibiotic treatment.
Ringworm Approximately circular, expanding, hairless areas Condition is usually self-limiting and does not
(Microsporum nanum that develop crested scabs. Lesions typically have appear to have any significant effect on the pig.
or Trichophyton a clearly demarcated margin that may be slightly The disease is zoonotic but direct contact with
verrucosum) raised. The lesion is nonpruritic. the lesion (and abrasion on the human skin) is
probably required for transmission. Other fungi
are occasionally identified. Usually a condition of
older swine and quite common in sows.
Skin necrosis of Abrasive lesions on knees, fetlocks, coronets, Lesions often are more serious than realized. The
piglets hocks, or elbows. The condition can lead to condition is prevented by use of less abrasive
(Bacterial dermatitis) septicemia and contribute to infectious arthritis, surfaces in the farrowing environment, maintaining
valvular endocarditis, and other diseases. Left good hygiene, and providing adequate clean
unmanaged, high case mortality can result. bedding when used.
Sunburn Pigs may drop to their belly from pain associated If shade is provided, pigs will not lay in the sun.
with sunburn on their back. Reddening, Providing access to wallows will help prevent the
thickening, then peeling of affected skin is condition. Unpigmented skin more susceptible.
typical. Condition usually found on the back
and ears.
TABLES

Table continued on next page

SKIN LESIONS: DIFFERENTIAL DIAGNOSIS 187


SWINE DISEASE MANUAL

Table cont’d. Skin lesions: differential diagnosis

Disease and agent Signs and lesions Diagnostic features and methods
Swinepox Initially appear as 5 to 10 mm round to oval All age groups are susceptible but is most often
(Swinepox virus) lesions on ventrum but can extend over the seen in young, growing pigs. Infection assumed to
body. Lesions progress from papules through the be endemic in the US and the clinical condition is
vesicle, pustule, and scab stages, similar to other rarely encountered; no surveillance has occurred
pox viruses. recently to determine the true prevalence.
Congenital pox is sporadic but quite striking in
affected newborn piglets.
Vesicular diseases Round to oval vesicles on feet and coronary Lesions are similar for all of the listed vesiculating
(foot-and-mouth bands, in and around the mouth or tongue, on viral diseases. All suspicious vesicular diseases
disease, vesicular snout, in nostrils, and occasionally elsewhere. should be investigated and reported promptly, as
stomatitis, vesicular all of the agents except Senecavirus A are exotic
exanthema, swine to the US.
vesicular disease,
Senecavirus A)
Vices and Tail, ear, or flank bite wounds. Moribund swine Numerous risk factors have been studied and
cannibalism often have red, linear bite marks. Sows may show confirmed to contribute to these conditions. In
vulvar bite wounds when housed in groups. particular, overcrowding, insufficient feed or
While all tend to occur sporadically on farms, waterer space, and lack of enrichment are thought
tail-biting can occasionally appear as an outbreak to be key risk factors to manage in preventing the
within a pen or barn. conditions.
TABLES

188 SKIN LESIONS: DIFFERENTIAL DIAGNOSIS


TABLES

Vesiculating viral diseases


Foot-and-mouth disease, of primary concern, is exotic to the US. If it should be introduced to the US (or other areas currently
free of the disease), there would be significant immediate effects on trade, pig movements, and business continuity. Unfortu-
nately, vesicles produced by all of the diseases listed below, including Senecavirus A, are indistinguishable; therefore, any vesicle
occurrence should prompt urgent investigation. While most of the swine-oriented veterinary diagnostic laboratories in the US
are able to provisionally differentiate the diseases below, the obligation of producers and veterinarians is to report suspicious
vesicular lesions to the appropriate regulatory officials in their state.

Disease Family and genus of virus Other animals affected Comments


Foot-and-mouth disease Picornaviridae Most ruminants, wild US free of the disease.
Aphthovirus and domestic.
Senecavirus A Picornaviridae Swine only. Sporadic cause of vesicles that
Senecavirus A must be differentiated from
other vesiculating diseases. The
US and many other parts of the
world experienced a significant
reemergence of this disease since
about 2013.
Swine vesicular disease Picornaviridae Swine, occasionally humans. Never reported in the US, although
Enterovirus infrequent and sporadic cases occur
in Italy.
Vesicular exanthema and Caliciviridae Swine, sea lions, opal-eye Disease has not been seen since 1956,
San Miguel sea lion virus Calicivirus fish, several kinds of seals, although San Miguel sea lion virus is
and other marine animals. known to occur in marine animals off
the western US coast suggesting that a
threat of the disease remains.
Vesicular stomatitis Rhabdoviridae Cattle, horses, some wildlife, Endemic in parts of the southeastern
Vesiculovirus and humans. and southwestern US; disease
common in parts of Central and South
America. Periodic small outbreaks
occur annually in the US. Insect vector
is likely but poorly understood.

TABLES

VESICULATING VIRAL DISEASES 189


SWINE DISEASE MANUAL

Zoonotic agents found in swine and humans


Producers and veterinarians have an important role in public health. A number of agents (many of which do not cause disease in
pigs) are carried by pigs and can be transmitted to people, be they farm staff, family members, veterinarians, abattoir workers, or
the general public. The table below provides an overview of the agents known to be zoonotic that are most common on farms.

Modes of transmission to humansd,e

OIE listed disease


Included in SDMb

Exotic to the US

Direct contact

Vector-borne

Food borne
Fomites
Aerosol
(2019)c

Oral
Agenta
Bacillus anthracis X X X X X X
Balantidium coli X
Brucella suis X X X
Burkholderia pseudomallei X X X X
Campylobacter species X X
Cryptosporidium suis X
Erysipelothrix rhusiopathiae X X1
Escherichia coli X X X X X
Francisella tularensis X X X X X
Giardia species X X
Hepatitis E virus X S S S S
Influenza A virus X X2
Japanese encephalitis virus X X X X
Leptospira interrogans X X X
Listeria monocytogenes X X
Menangle virus X X X
Methicillin-resistant Staphylococcus aureus (MRSA) S S
Microsporum species X X S
Mycobacterium species X X S S
Nipah virus X X X S X
Norwalk virus X X
Pasteurella multocida X S2 S1,2 S
Salmonella species X X X X
Sarcoystis suihominis X X
Streptococcus suis X X S
TABLES

190 ZOONOTIC AGENTS


TABLES

Table cont’d. Zoonotic agents found in swine and humans

Modes of transmission to humansd,e

OIE listed disease


Included in SDMb

Exotic to the US

Direct contact

Vector-borne

Food borne
Fomites
Aerosol
(2019)c

Oral
Agenta
Taenia solium X X X
Toxoplasma gondii S S S X
Trichinella spiralis X X X
Trichophyton species X X S
Vesicular stomatitis virus X X
Yersinia enterocolitica X X X X X X

a All listed agents have the potential to infect and cause clinical disease in humans but not all are associated with clinical disease in swine.
b Agent has been described in this text, Swine Disease Manual, and is likely to be associated with clinical disease in swine.
c http://www.oie.int/animal-health-in-the-world/oie-listed-diseases-2019/. Published 2019. Accessed July 17, 2019.
d Adapted from Khan SU, Atanasova KR, Krueger WS, Ramirez A, Gray GC. Epidemiology, geographical distribution, and economic
consequences of swine zoonoses: a narrative review. Emerging Microbes & Infections, 2013 Dec; 2(12):e92.
e “X” indicates an established or primary route of transmission. “S” indicates a suspected route of transmission.
1 Requires damaged skin or mucosa
2 Droplets via close contact

TABLES

ZOONOTIC AGENTS 191


192
TABLES
INDEX

Index
A Aspergillus. See aflatoxicosis
acariasis. See mange astrovirus. See porcine astrovirus
Actinobacillus pleuropneumoniae (APP), 9-11, 12, 41, 44-45, atresia ani, 145, 149
147, 184t
atrophic rhinitis, 16-17, 44, 164, 183t
Actinobacillus suis, 9, 12-13, 26, 30, 40, 179t, 186t
Aujeszky’s disease. See pseudorabies
adenovirus. See porcine adenovirus
aural hematoma, 145, 186t
aflatoxicosis, 157
African swine fever (ASF), 63-64, 68, 107, 129, 181t B
Bacillus anthracis, 14-15, 190t
agalactia, 50, 148, 158
blue eye disease, 65-66
ammonia, 16-17, 45, 51, 163-164
bocavirus. See porcine bocavirus
anemia, 159
Bordetella bronchiseptica, 51. See also atrophic rhinitis
in association with
Brachyspira, 55-57, 141, 171t, 173-174t, 178t. See also swine
bovine viral diarrhea and border disease virus, 69
dysentery
copper toxicity, 161
Brucella suis, 53-54, 181t, 190t
gastric ulcer, 147
brucellosis. See Brucella suis
inclusion body rhinitis, 76
Bungowannah virus, 118
leptospirosis, 35-36
lice, 129, 186t C
calcium, 151, 152, 159, 160
Mycoplasma pneumonia, 41
carbadox, 162, 180t
Mycoplasma suis, 37-38, 179t,
carbon dioxide, 163, 164
porcine hemorrhagic enteropathy (PHE), 33
carbon monoxide, 163, 164
threadworm, 136
central nervous system (CNS) diseases, 169-170t
vitamin B12 deficiency, 160
chemical toxins, 163
anthelmintics, 167t, 175-176t. See also parasiticides circovirus. See porcine circovirus
anthrax, 14-15. See also Bacillus anthracis classical swine fever, 46, 67-69, 145, 169t, 181t, 187t
arsenic, 160 as a differential diagnosis, 10, 15, 26, 48, 63-64, 107
arsenicals, organic, 23, 55, 160, 170t Claviceps purpurea. See ergotism
arthritis Clostridium difficile. See diarrhea, clostridial
in neonatal and growing swine, 168t Clostridium perfringens, 107, 126, 136, 177t. See also diarrhea,
mycoplasmal, 39-40. See also Mycoplasma hyorhinis; clostridial
Mycoplasma hyosynoviae coccidiosis, 18, 125-126, 167t, 171t, 174t, 175t
ascariasis. See Ascaris suum as a differential diagnosis, 20, 23, 102, 107
Ascaris suum (roundworm), 137-138, 167t, 175t, 183t cocklebur poisoning, 162

Locators in boldface indicate main discussions. Those followed by t indicate tables.


SWINE DISEASE MANUAL 193
SWINE DISEASE MANUAL

colibacillosis, 19-20, 21-24, 101-102, 107, 126, 172t. encephalitis


See also Escherichia coli
in association with
colitis, spirochetal, 55-57, 174t
African swine fever, 64
congenital tremors, 69, 119, 145, 153
bacterial meningitis, 169t
copper, 146, 150, 159-160
blue eye disease, 65-66
cysticercosis, 127-128, 175t. See also Taenia solium
classical swine fever, 68
cystitis, 145, 152
encephalomyocarditis virus, 70
Cystoisospora. See coccidiosis
Haemophilus parasuis, 29
D hemagglutinating encephalomyelitis (HEV), 75, 169t
Demodex. See mange, demodectic pneumonic pasteurellosis, 44
deoxynivalenol, 158 porcine adenovirus, 119
dermatosis vegetans, 110, 145, 186t porcine bocavirus, 120
diamond skin disease, See erysipelas porcine picornavirus, 90
diarrhea, 171t, 172-174t porcine reproductive and respiratory syndrome (PRRS), 94
clostridial, 18-20, 171t, 172-173t pseudorabies, 99, 185t
porcine epidemic, 96, 105-107. See also swine enteric rabies, 152
coronavirus diseases
salt poisoning, 170t
post-weaning, See colibacillosis
Streptococcus suis, 50-52
rotavirus, 101-102
Japanese, 81-82, 121, 169t, 181t, 190t
transmissible gastroenteritis, 23, 96-97, 105-107, 119.
See also swine enteric coronavirus diseases encephalomyocarditis virus, 70-71, 88, 89t
differential diagnosis endometritis. See metritis
central nervous system, 169t enterotoxemia, 18, 21, 148
diarrheal diseases, 171t, 172-174t enterovirus(es), 70, 88-91, 89t, 108, 170t, 189t
intestinal hemorrhage, 177-178t Enterovirus, 88, 89t, 108
reproductive failure, 180-182t Eperythrozoon suis (eperythrozoonosis) - archaic,
See Mycoplasma suis
respiratory disease, 183-185t
epitheliogenesis imperfecta, 146
skin lesions, 186-188t
ergotism, 148, 158
dysentery. See swine dysentery
erysipelas, 25-26, 168t, 181t
E as a differential diagnosis, 12, 15, 48, 68, 179t, 186t
ear biting, 154
Erysipelothix rhusiopathiae, 25-26, 40, 190t. See also erysipelas
Ebola virus, 117
Escherichia coli (E coli), 21-24, 47-48, 136, 148, 149, 154,
ectopic ossification, 146 169t, 171t, 172t, 190t, See also colibacillosis
edema disease, 154, 169t, See also colibacillosis exudative epidermitis, 27-28, 110, 132, 153, 160, 186t
Eimeria. See coccidiosis

Locators in boldface indicate main discussions. Those followed by t indicate tables.


194 INDEX
INDEX

F I
F-2 toxin. See zearalenone ileitis, 32-34, 48, 56, 107, 148. See also Lawsonia
intracellularis
farrowing rate, low, 180t
inclusion body rhinitis, 17, 51, 76-77, 183t
feed additives, 162-163, 180t
influenza (IAV-S), 78-80, 87, 118, 130, 181t, 183t, 185t, 190t
filoviruses, 117
as a concurrent infection, 10, 29-30, 41-43, 44-45, 51
flank biting, 154
in association with gastric ulcers, 146-147
foot-and-mouth disease, 72-73, 88, 89t, 188t, 189t
insecticides, 110, 129, 163
as a differential diagnosis, 88, 103, 108, 111, 112-113,
internal parasites, 175-176t
fumonisin toxicosis, 158
intestinal hemorrhage, 177-178t
Fusarium, 158
intestinal nematodes, 134-136
G iodine, 28, 46, 50, 65, 70, 98
gastric ulcer, 33-34, 56, 79, 146-147, 148, 177t
iodine deficiency, 159
Glaesserella parasuis. See Haemophilus parasuis
ionophores, 162-163
Glasser’s disease, 29-31, 68, 168t. See also Haemophilus
iron
parasuis (HPS)
deficiency, 38, 159
goiter. See iodine deficiency
toxicity, 150, 159, 161
gossypol, 162
Isospora. See coccidiosis
greasy pig disease, 27-28. See also exudative epidermitis
J
H Japanese encephalitis, 81-82, 121, 169t, 181t, 190t
Haematopinus suis. See lice
Haemophilus parasuis (HPS), 23, 29-31, 40, 168t, 169t, 179t. K
See also Glässer’s disease kidney worm, 134, 175t
Haemophilus pleuropneumoniae. See Actinobacillus Kunjin virus (West Nile virus), 121
pleuropneumoniae
L
hemagglutinating encephalomyelitis, 74-75, 105, 107, 169t La Piedad Michoacan virus. See blue eye disease
hematoma, aural (ear), 145, 186t Lawsonia intracellularis, 32-34, 171t, 174t, 178t.
hemorrhage, intestinal, 177-178t See also ileitis
hemorrhagic bowel syndrome, 33, 146, 154, 177t lead, 161
hepatitis E virus, 117, 190t Leptospira (leptospirosis), 35-36, 181t, 190t
hernias, 148 lice, 27, 37, 110, 129, 145, 163, 167t, 186t
herpesvirus. See pseudorabies; inclusion body rhinitis lungworm, 130-131, 136, 167t, 175t, 183t
hog cholera. See classical swine fever
M
hydrogen sulfide, 163-164 Macracanthorynchus hirudinaceus. See thorny-headed worm
hydronephrosis, 148 mange, 132-133, 153, 163, 167t, 186t
hypogalactia, 148-149 as a differential diagnosis, 129, 145, 160
hypoglycemia, 149, 162 demodectic, 132
Locators in boldface indicate main discussions. Those followed by t indicate tables.
SWINE DISEASE MANUAL 195
SWINE DISEASE MANUAL

Marburg virus, 117 osteochondrosis, 40, 151, 179t


mastitis, 21, 148-149 osteoporosis, 160
megacolon, 145, 149
P
Menangle virus, 65, 118, 190t parainfluenza virus. See porcine parainfluenza virus
mercury toxicity, 161, 163 parakeratosis, 132, 147, 159-160. See also zinc
Metastrongylus. See lungworm paramyxovirus, 65, 118, 120, 169t
methane, 163 parasites, internal, 175-176t
metritis (endometritis), 53, 94, 99, 148-149 parasiticides, 129, 130, 133, 134, 162, 167t. See also
Microsporum nanum. See ringworm anthelminitics

minerals, 159-161, 170t, 180t pars esophagea, 86, 146-147, 177t

MMA (mastitis, metritis, agalactia), 148-149 parvovirus, 26, 27, 83-84, 86, 88, 90, 119, 181t

mulberry heart disease, 71, 149-150. See also selenium; Pasteurella multocida, 41-42, 44-45, 51, 164, 183t, 184t, 190t.
vitamin E See also atrophic rhinitis; pasteurellosis

Mycobacterium, 58-59, 190t pasteurellosis, 10, 44-45, 184t. See also atrophic rhinitis;
Pasteurella multocida
Mycoplasma hyopneumoniae, 10, 39, 41-43, 44-45, 51, 87,
179t, 184t pediculosis. See lice
Penicillium. See aflatoxicosis
Mycoplasma hyorhinis (MHR), 30, 39-40, 41, 154, 168t, 179t
pestivirus, 67, 69, 118, 145, 153
Mycoplasma hyosynoviae (MHS), 39-40, 41, 168t, 179t
phosphorus, 152, 160
Mycoplasma-related diseases, 179t
photosensitization, 132, 153-154, 187t
Mycoplasma suis, 37-38, 41, 129, 160, 175t, 179t
picornavirus, 70, 108, 170t. See also porcine picornavirus
mycotoxicosis, 157-158
pigweed poisoning, 162
mystery pig disease. See porcine reproductive and respiratory
syndrome (PRRS) pityriasis rosea, 27, 110, 129, 132, 151, 153, 187t
pneumonia
N
navel sucking, 148, 154 mycoplasmal (enzootic), 10, 41-43, 44, 179t, 184t.
See also Mycoplasma hyopneumoniae
nematode, 130, 138
pneumonic pasteurellosis. See pasteurellosis
nematodes, minor intestinal, 134-136
porcine adenovirus, 119
nightshade, 162
porcine astrovirus, 84, 119, 170t
Nipah virus, 65, 118, 190t
porcine bocavirus, 83, 119-120
nodular worm, 134-135, 141, 167t
porcine circovirus (PCV), 34, 85-87, 146, 147
nutritional deficiencies, excesses, 27, 90, 151, 154, 159-161
associated disease (PCVAD), 34, 38, 85-87, 120, 169t, 181t
O type 2 (PCV2), 30, 34, 40, 41, 84, 85-87, 119, 146, 147,
Oesophagostomum. See nodular worm 181t, 187t
organophosphate(s), 163, 169t, 170t type 3 (PCV3), 84, 85-87, 181t, 187t
Ornithodorus porcinus porcinus, 63 porcine cytomegalovirus. See inclusion body rhinitis

Locators in boldface indicate main discussions. Those followed by t indicate tables.


196 INDEX
INDEX

porcine deltacoronavirus. See swine enteric coronavirus ringworm, 27-28, 152-153, 187t, 190-191t
diseases
rodenticides, 163
porcine dermatitis and nephropathy syndrome (PDNS), 187t.
rotavirus, 22, 48, 101-102, 119, 126, 171t, 172t
See also porcine circovirus associated disease
as a differential diagnosis, 23, 107, 136
porcine epidemic diarrhea (PED), 96, 105-107. See also
swine enteric coronavirus diseases roundworm, 137-138, 167t. See also Ascaris suum
porcine parainfluenza virus, 120
S
porcine paramyxovirus. See blue eye disease; paramyxovirus Salmonella, 12, 46-49, 141, 148, 154, 169t, 171t, 173t, 177t,
181t, 191t. See also salmonellosis
porcine parvovirus. See parvovirus
salmonellosis, 46-49, 56, 101, 102, 142, 152, 171t, 177t. See
porcine picornavirus, 88-91, 89t
also Salmonella
porcine pleuropneumonia, See Actinobacillus
as a differential diagnosis, 10, 26, 33-34, 38, 56, 68, 107, 147
pleuropneumoniae
salt poisoning. See sodium ion intoxication
porcine reproductive and respiratory syndrome (PRRS), 70,
83, 92-95, 117, 169t, 182t, 185t, 187t San Miguel sea lion disease, 111, 189t. See also vesicular
exanthema
as a concurrent infection, 10, 30, 40, 41, 44, 51
Sapelovirus, 84, 88-91, 89t, 170t
as a differential diagnosis, 17, 87
sapovirus. See porcine sapovirus
porcine respiratory coronavirus, 96-97, 105, 172t
Sarcoptes. See mange
porcine rubulavirus. See blue eye disease
scabies. See mange
porcine sapovirus, 120
selenium, 149-150, 159, 161, 170t
porcine stress syndrome, 151, 153
Seneca Valley virus. See Senecavirus A
pork measles. See cysticercosis; Taenia solium
Senecavirus A, 88, 89t, 103-104, 188t, 189t
pork worm. See Trichinella spiralis
shoulder ulcers, 153
postweaning multisystemic wasting syndrome (PMWS).
See porcine circovirus associated disease skin lesions, 186-188t
prolapse, 149, 152, 158 skin necrosis, 153, 187t
PRRS. See porcine reproductive and respiratory syndrome SMEDI, 83-84, 88-90. See also parvovirus
pseudorabies, 17, 23, 51, 66, 98-100, 170t, 182t, 185t sodium ion intoxication, 161, 170t
pustular dermatitis, 28, 152, 187t. See also Streptococcus spirochetal colitis, See swine dysentery
pyelonephritis, 145 splayleg, 145, 153
spraddleleg. See splayleg
R
rabies, 152, 169t Staphylococcus hyicus, See exudative epidermitis; greasy pig
disease
ractopamine, 163
Stephanurus dentatus. See kidney worm
reproductive failure, 181-182t
Streptococcus, 152, 153, 168t, 169t, 187t. See also pustular
rhinitis, 29, 30, 51, 94, 99, 185. See also atrophic rhinitis;
dermatitis
inclusion body rhinitis
Streptococcus suis, 23, 26, 30, 40, 50-52, 93, 168t, 169t,
rickets, 160
179t, 191t

Locators in boldface indicate main discussions. Those followed by t indicate tables.


SWINE DISEASE MANUAL 197
SWINE DISEASE MANUAL

Strongyloides, 23, 126. See also threadworm U


suid herpesvirus 1. See pseudorabies ulcers

sulfonamides, 25, 153, 163 button, 48, 68, 173t

sunburn, 132, 153-154, 187t gastric, 33-34, 56, 79, 146-147, 148, 177t

swine dysentery, 34, 55-57, 160, 162, 171t, 173-174t, 178t shoulder, 153

as a differential diagnosis, 33-34, 48, 107, 141-142, 147 V


swine enteric coronavirus diseases, 101, 105-107, 126, 171t, ventilation failure, 163-164
172t. See also transmissible gastroenteritis (TGE); porcine vesicular exanthema, 88, 103, 111, 188t, 189t
epidemic diarrhea (PED); porcine deltacoronavirus
(PDCoV) vesicular stomatitis, 103, 112-113, 188t, 189t, 191t

swine vesicular disease, 88, 89t, 103, 108-109, 188t, 189t vesiculating viral diseases, 189

swinepox, 110, 129, 188t vestibular syndrome, 51, 154


vices, 154, 188t
T
T-2 toxin, 158 vitamin deficiencies, 160

Taenia solium, 127-128, 175t, 191t vitamin D, 160, 163

tail biting, 139, 154, 188t vitamin E, 146, 149-150, 159, 161

Talfan disease, 88-91, 170t vitamin K, 147, 160, 163, 178t

Teschen disease, 88-91, 170t vitamins, 27, 150, 160, 170t, 180t

thorny-headed worm, 135, 167t, 176t volvulus, 146, 148, 154, 177t

threadworm, 135-136, 167t, 176t. See also Strongyloides vomiting and wasting disease. See hemagglutinating
encephalomyelitis
Torque teno virus, 121
vomitoxin, 158
torsion, 148, 154, 177t
transmissible gastroenteritis (TGE), 23, 96-97, 105-107, 119. W
See also swine enteric coronavirus water deprivation. See sodium ion intoxication

tremors, congenital, 69, 119, 145, 153 West Nile virus, 81, 121

Trichinella spiralis, 139-140, 176t, 191t whipworm, 33, 48, 56, 141-142, 147, 167t, 171t, 174t, 176t

trichinellosis, 139-140. See also Trichinella spiralis Z


trichinosis. See Trichinella spiralis zearalenone, 152, 158, 180t

Trichophyton verrucosum. See ringworm zinc, 24, 27, 146, 159-161. See also parakeratosis

trichothecene, 158 zoonotic agents, 190-191t

Trichuris suis. See whipworm


Trueperella, 153, 168t, 169t, 181t,
tuberculosis, 58-59. See also Mycobacterium

Locators in boldface indicate main discussions. Those followed by t indicate tables.


198 INDEX

You might also like