You are on page 1of 27

Porcine respiratory coronavirus : molecular features and

virus-host interactions
Hubert Laude, K. van Reeth, M. Pensaert

To cite this version:


Hubert Laude, K. van Reeth, M. Pensaert. Porcine respiratory coronavirus : molecular features and
virus-host interactions. Veterinary Research, BioMed Central, 1993, 24, pp.125-150. �hal-02715975�

HAL Id: hal-02715975


https://hal.inrae.fr/hal-02715975
Submitted on 1 Jun 2020

HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est


archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents
entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non,
lished or not. The documents may come from émanant des établissements d’enseignement et de
teaching and research institutions in France or recherche français ou étrangers, des laboratoires
abroad, or from public or private research centers. publics ou privés.
Review article

Porcine respiratory coronavirus:


molecular features and virus-host interactions

H Laude K Van Reeth


2 M Pensaert
2

1
INRA, Unité de Virologie et Immunologie Moléculaires, 78650 Jouy-en-Josas, France;
2
Faculty of Veterinary Medicine, Laboratory of Virology, 9000 Ghent, Belgium

(Received 7 December 1992; accepted 11 December 1992)

Summary ― Since 1984, a previously unrecognized respiratory coronavirus, causing a mostly un-
apparent infection, has rapidly and massively spread within the swine population in Europe, and few
years later, a virus with similar characteristics has been identified in the USA. The agent, designated
PRCV, appears to be derived from the porcine enteric coronavirus TGEV. The aim of the present ar-
ticle is to review comprehensively the state of the knowledge about this new virus and its infection.
The review includes the following topics: epizootiology, molecular characterization and antigenic fea-
tures of PRCV, pathogenesis and clinical aspects, immunity and laboratory diagnosis. The authors’
views concerning the impact of the emergence of PRCV on both coronavirus research and swine
production are presented in the conclusion.

porcine respiratory coronavirus I transmissible gastroenteritis virus variant I molecular char-


acterization I epizootiology I pathogenesis I immunity I diagnosis

Résumé ― Coronavirus respiratoire porcin: aspects moléculaires et interactions virus-hôte.


À partir de 1984, un virus respiratoire inconnu jusqu’alors, responsable d’une infection essentielle-
ment inapparente, a rapidement et massivement diffusé au sein de la population porcine euro-
péenne; quelques années plus tard, un virus présentant des caractéristiques similaires a été identi-
fié aux USA. Cet agent, désigné PRCV, apparait être dérivé du coronavirus entéropathogène porcin
TGEV. L’objectif de cette revue est de faire le point de façon critique sur l’ensemble des connais-
sances accumulées sur ce nouveau virus et sur l’infection qu’il engendre. Epizootiologie, caractérisa-
tion moléculaire et antigénique du PRCV, pathogénèse et aspects cliniques, immunité et diagnostic
en laboratoire sont les principaux volets developpés. En conclusion, les auteurs donnent leur point
de vue sur les conséquences qu’a eu l’émergence du PRCV au plan des recherches en coronaviro-
logie et sur la production porcine. (92 références)

coronavirus respiratoire porcin / variant du virus de la gastro-entérite transmissible / caracté-


risation moléculaire l épizootiologie / pathogénie l immunité / diagnostic

Correspondence and reprints


INTRODUCTION groups of pigs on closed breeding-
fattening farms showed that 2 situations
The porcine respiratory coronavirus were encountered. In some farms, sows
(PRCV) is a variant of transmissible gas- were seropositive for TGEV and fattening
troenteritis virus (TGEV). It appears to pigs became negative after losing their ma-
have emerged naturally in Europe during ternal antibodies between 12 and 18 wk of
the early eighties (Pensaert et al, 1986). In age. On other farms, maternal anti-TGEV
more recent years, a similar virus has antibodies first declined but the antibody
been detected in the ,USA (Wesley et al, titer increased again around the age of 6-9
1990a). wk indicating the persistence of TGEV or a
In 1983-1984, a serological survey was TGEV-like infection. On these farms, senti-
carried out in Belgium in slaughterhouse nel pigs were placed together with farm-
sows to determine the prevalence of born pigs at the age of 4 wk and nasal
TGEV among the swine population (Pen- swabs and fecal samples were collected
saert et al, 1986). Surprisingly enough, it twice weekly. The sentinel pigs showed
was found that 68% of the sows had neu- seroconversion to TGEV without any clini-
tralizing antibodies to TGEV. This was in cal sign and, subsequently, a virus was
sharp contrast to the results of previous isolated from the nasal swabs which had
surveys, which had been carried out dur- been stored at -70°C. It was cultivable in
ing the seventies and early eighties and at cell cultures and examination with the elec-
which time the percentage of seropositive tron microscope revealed that it was a co-
sows varied between 12-24%. In France, ronavirus. The virus was neutralized by an-
similar findings were reported during a tiserum against TGEV but did not cause
study in 1985 when 73% of the farms had any disease upon inoculation of neonatal
TGE antibodies in a region in Brittany pigs. It was first called TGEV-like mutant
(Jestin et al, 1987b). because of its resemblance to TGEV and
was later named PRCV because it clearly
TGE is a porcine viral disease which is
had a tropism for the respiratory tract (and
well known all over the world. The virus
not for the enteric tract). In recent years,
was first identified in 1946 (Doyle and
PRCV has been compared to TGEV from
Hutchings, 1946) and, for decades, the the point of view of the structural and bio-
disease has been known as a severe diar-
rhea in swine of all ages, with a high (near- logical characteristics. These studies have
shown that few genetic differences in
ly 100%) mortality rate in neonatal pigs.
No epidemic of diarrhea resembling TGE PRCV have brought about large alterations
had occurred in Belgium during the in the virus-cell and virus-animal interac-
months prior to the 1983-1984 serological tions. PRCV has, therefore, appeared to
be a good model for coronavirologists to
survey and to the finding of the abnormally
high incidence of anti-TGEV antibodies. study how the modification of genes en-
Moreover, in tracing back the positive coding structural and non-structural pro-
sows to their farms of origin, it appeared
teins may modulate the development of
that a high prevalence of antibodies was the disease in the animal. Since the emer-
present in the sow population in these gence of PRCV, the clinical and economic
farms and that no history of an epidemic of importance of TGE has markedly lowered
diarrhea typical of TGE had occurred. At- in Europe.
tempts to isolate TGE virus from the feces The present article is meant to review
of pigs of different ages on such farms the state of knowledge of PRCV and its in-
failed. Longitudinal serological studies of fection.
EPIZOOTIOLOGY A sero-epizootiological study was car-
ried out in the Belgian swine population in
PRCV is widespread in Europe (Brown 1989-1990 (Pensaert et al, 1992). PRCV-
and Cartwright, 1986; Pensaert et al, 1986; induced neutralizing antibodies were
Jestin et al, 1987; Madec et al, 1987; Hen- found in 90.6% of the 160 sera from sows
ningsen et al, 1988; Yu et al, 1989; Lanza at slaughter. Fattening swine were fol-
et al, 1990). A non-diarrheic TGEV-like vi- lowed on 33 closed breeding-fattening
rus with quite similar characteristics has farms. The virus persisted on 22 farms,
also been found in the USA (Wesley et al, 11 of which were situated in a high farm
1990a), Asia (Pensaert M, unpublished re- density region (all the farms located in an
sults) and Eastern European countries area of 4 km ) and 11 of which were
2
(Deriabine, personal communication). situated in a relatively low farm density re-
In Europe, PRCV spread rapidly in all gion (1-4 farms per 12 km ). The growing
2
swine raising countries during the second pig population on the remaining 11 closed
half of the eighties. This spread to and breeding-fattening farms became tempo-
within countries was so rapid and ap- rarily free of PRCV in spring and summer.
peared to.be so unavoidable that an aero- All these farms became reinfected during
genic route of dissemination could not be the autumn. The latter pattern has been
overlooked. Rapid virus spread also oc- observed repeatedly in Belgium over
curred in countries with high hygienic stan- several successive years and in other
dards or a highly developed SPF-farm sys- countries such as France (Jestin et al,
tem. Denmark, a country which has always 1987b; Laval et al, 1990). These observa-
been free of TGEV, was a good example tions indicate that waves of infection easi-
to show that the aerogenic spread of
ly occur during the foggy and rainy
PRCV had to be the main route of dissemi- season. The infection wave is not ac-
nation. The infection was first observed in
Jutland along the German border and it
companied by manifest clinical disease
signs.
progressed towards the inside of the coun- Virus on a farm basis, oc-
try on farms where no introduction or sale persistence,
of animals had occurred and with which no curs by infection of successive litters of
other direct or indirect links could be estab- pigs. Maternal antibodies in these pigs
lished show a normal decline until the age of 5-8
(Henningsen et al, 1988).
wk and then the antibody titer rises again.
Since that time, experimental inocula-
This evolution points to a switch from pas-
tion studies have shown that aerolized vi-
sive to active immunity. In one farm, it was
rus initiates the infection very easily in the
shown, by virus isolation from tonsillar
upper and deeper respiratory pathways
and that high quantities of viruses are pro- swabs, that the infection occurs around the
duced in the lungs and excreted in nasal age of 5 wk. Maternal antibodies apparent-
fluids. Animals excrete the virus in orona- ly postpone but do not prevent the infec-
sal secretions for 8-13 d after inoculation tion of the respiratory tract.
and long term virus carriers have not been In the study earlier mentioned, antibod-
observed. There are no indications that the ies induced by TGEV as detected by a dif-
fecal-oral transmission plays a role in the ferential ELISA (Callebaut et al, 1989)
epizootiology of the natural infection and were found in 7.6 % of the 130 sow sera
persons, footwear or lorries are unimpor- and in sera from fattening pigs on 5 of the
tant in spreading the virus. Of course, in- 33 farms. This shows that TGEV is still
troduction of infected pigs serves as a present in the swine population despite the
source of infection. high prevalence of PRCV.
It is clear that seasonal circumstances, related to TGEV, had emerged and spread
swine density and distances between massively within the European swine pop-
farms influence the epizootiology. In the ulation gave rise to a number of purely
above study, there was no correlation be- conjectural hypotheses about the nature of
tween the herd size and the temporary dis- this agent. The most alarming was that
appearance of PRCV on farms. Since farm PRCV might represent a TGEV ― possibly
density and distance vary greatly from vaccine -

strain of which the tropism for


country to country and from region to re- the respiratory tract was exalted as a result
gion, the epizootiological pattern of PRCV of a systematic aerogenic immunization
may be different in different countries. (Jestin et a/ 1987a) or was a consequence
In France, a longitudinal serological of a laboratory manipulation. Other specu-
study was carried out in 10 fattening units lative views involved a spontaneous ge-
and it was observed that seroconversion nome recombination between TGEV and
occurred during the fattening period in 8 either a heterologous coronavirus such as
(Laval et al, 1991). A study on the preva- the feline infectious peritonitis virus FIPV
lence of PRCV infections in pigs shortly af- or the canine coronavirus CCV (both of
ter entering intensive fattening units was which are able to infect the porcine spe-
recently carried out in Belgium (Van Reeth cies to some extent), or a swine-specific,
et al, unpublished results). In these farms, serologically unrelated virus, such as the
an all in - all out system is the rule. Pigs porcine epidemic diarrhea virus PEDV or
originate from numerous breeding herds the hemagglutinating encephalitis virus
and are introduced for fattening at the age HEV (Sanchez et al, 1990). The possibility
of 9-12 wk. During the autumn of 1991, 10 0 that a preexisting coronavirus specific for a
groups of these pigs on 9 different farms wild animal crossed accidentally the spe-
were serologically examined 1 wk after en- cies barrier was also set forward.
try and 3 wk later. Approximately 50% of The problem of the phylogenic origin of
the pigs had PRCV-neutralizing antibody
PRCV has been largely elucidated through
titres of < 4 at the time of arrival. An infec-
the information provided by analysis of its
tion with PRCV was observed in all 10
genome. The most plausible scenario is
groups. A similar study which was per- that this agent originates from TGEV itself
formed in February-March 1992 involved 7
and that this occurred without incorpora-
groups of pigs on 7 farms. PRCV- tion of exogenous genetic material. At the
seroconversion was observed in 6 out of
same time, the studies raised stimulating
the 7 groups. These results show that the
virus is introduced in practically every questions about the genetic basis of the at-
tenuation of PRCV, or in alternative terms,
group with some animals and that an in-
of the enterotropism of TGEV.
fection occurs in those pigs which have
not yet been infected on the breeding farm
of. origin. In all these groups, the serocon- Molecular characterization
version was due to infection with PRCV
and not with TGEV, as was shown by a
differentiating ELISA (Callebaut et al, Coronavirus genome organization
1989). and expression

CAUSAL AGENT The genome of coronaviruses consists of


one single-stranded mRNA molecule of
The demonstration thata previously unrec- positive polarity. With some 4
3 x 10 nucleo-
ognized respiratory coronavirus, closely tides (nt), coronaviruses have the largest
genome among RNA viruses, yet they do has been questioned. This model derives
not encode a particularly high number of further complexity from the high rate of re-
proteins. The 5’ first 20 kilobases (kb) re- combination observed during coronavirus
gion is comprised of 2 large open reading replication.
frames (ORFs) from which a polyprotein Coronavirus proteins are translated
with polymerase motifs is translated. Thus, from the mRNA 5’ part which is absent
nearly two-thirds of the coding capacity is from the immediately smaller mRNA spe-
devoted to the RNA transcription and repli- cies. The translationally active part gener-
cation machinery, which appears to be
ally contains a single ORF; however, a few
strikingly intricated in coronaviruses. mRNAs are functionally bicistronic (eg
Most of the genes are clustered in the mRNA 3 of TGEV) or tricistronic.
3’-terminal third. They are expressed
through the synthesis of 5-7 mRNAs of
subgenomic size which are 3’ coterminal, Comparison of PRCV
thus forming a so-called nested set struc- and TGEV genomes
ture. These mRNAs are thought to be pro-
duced following a mechanism of discontin- The genomic sequence of these 2 viruses
has not been completed, contrary to the
uous, non-processive, leader-primed avian infectious bronchitis virus IBV and
transcription. The leader is a short (50- murine hepatitis virus MHV genomes. Pub-
90 nt) sequence which is homologous to
the extreme 5’ of the genome plus sense lished TGEV nucleotide data are yet limit-
RNA. A hexameric conserved sequence ed to the 3’ third region downstream the
polymerase locus, ie5 = 8. kb. This region
(CUAAAC in several coronaviruses includ-
contains 7 large ORFs (fig 1), that are ex-
ing TGEV), located upstream from each
transcription unit, is assumed to provide a pressed through 6 (in one case, 7) subge-
nomic mRNA species, numbered 2 to 7
signal to the leader-polymerase complex from 5’ to 3’.
for the specific transcription initiation of
each mRNA species (review: Lai, 1990). The 3 ORFs named S, M and N encode
Recently, the possibility that the different the major structural proteins from which all
mRNAs could be amplified independently coronaviruses are built (review: Spaan et
al, 1988). The protein S (former name )
2
E PRCV with the wild TGEV strains (Miller
is a large (220 kDa), membrane-anchored and FS772) than with the Purdue 115
glycoprotein which, by trimerization (Del- strain, which has been isolated some 20 yr
mas and Laude, 1990), forms the charac- earlier (Rasschaert et al, 1990). The leader
teristic, petal-shaped spikes protruding from sequence of 2 PRCV isolates are identical
the virion envelope. The globular and the to that of FS772 strain in length (91 nt) and
elongated domains roughly correspond to composition, whereas that of Purdue strain
the amino- and carboxy-half parts of the differs at 2 positions (Page et al, 1990;
polypeptide chain, respectively. S is the Rasschaert and Laude, unpublished data).
sole virion protein able to induce highly neu- Based on the comparison of the S gene 5’
tralizing antibodies and is considered as be- half part of 6 Eur PRCV isolates and 5
ing the attachment protein. M (formerly E )
1 TGEV strains, an evolutionary tree has
is a 29-36 kDa-glycoprotein mostly embed- been proposed in which the 2 viruses di-
ded in the virion envelope. N is a 47 kDa- verged recently from a common ancestor
phosphoprotein associated with the genom- (Sanchez et al, 1992).
ic RNA to form the nucleocapsid (review: Another important finding revealed by
Laude et al, 1990). Recently, a 10 kDa pol- the sequence data is that the deletions
ypeptide encoded by mRNA 4 has been identified in 2 USA PRCV isolates (Wesley
proposed to be an additional virion- et al, 1991; Jackwood et al, 1992) differ
associated, integral protein, designated sM from that of Eur PRCV isolates. This is
(Godet et al, 1992). The 3 other ORFs, quite unexpected in view of the fact that
numbered 3a, 3b and 7, code for assumed-
the latter have nearly identical sequences.
ly non-structural polypeptides, the function Nevertheless, as detailed below, the ge-
of which is presently unknown.
nomes of Eur and USA PRCVs show the
The global organization of PRCV ge- same alterations: i) S gene encoding a
nome is identical to that of TGEV, as de- polypeptide with a large truncation at its N-
duced from sequence data covering the terminus; ii) ORF3a converted to a pseu-
whole region downstream from the S gene dogene.
5’ end (Rasschaert et al, 1990; Britton et
al, 1991; Page et al, 1991The only strik- PRCV S gene
ing difference consists of a number of de-
letions, which essentially affect two re- The position and length of the deletions
gions of PRCV genome: the S gene and identified in the S gene 5’ region of Euro-
the ORF3a (fig 1). No additional ORF is pean and USA isolates are shown in figure
present between the polymerase locus 2. The first 4 amino acids are conserved
and the S gene. Hence, PRCV and TGEV and the next 224 amino acids are deleted
genomes differ by deletions and point mu- in the mature S polypeptide encoded by
tations, with no sequence unique to PRCV the Eur isolates, whereas the deletion in
being identified. the USA isolates involves 227 residues
The 2 viruses exhibit an overall 3% nu- from position 7 to 233 (Rasschaert et al,
cleotide and amino acid divergence, not 1990; Britton et at, 1991; Wesley et al,
significantly different from that observed 1991As a consequence, the S protein of
among 2 TGEV strains. The observed ami- Eur and USA PRCV isolates are predicted
no acid changes are not clustered but in- to be 1209 and 1206 amino acids long, re-
terspersed within the different genes. Pair- spectively, instead of 1431 or 1433 for the
wise alignment of the translated ORFs TGEV strains (Rasschaert and Laude,
also revealed a slightly higher homology of 1987; Britton and Page, 1990).
The relative mass )
r
(M of the monomer- not detected in USA PRCV-infected cells,
ic S protein synthesised in PRCV-infected consistent with the conversion of the
cells has been shown to be reduced in the CUAAAC sequence in CUAAAU, which
expected proportion, ie 170 kDa instead of should be not (or poorly) functional for
220 kDa, while the M and N proteins have transcription.
a similar M, (Rasschaert et al, 1990). As a
consequence, some antigenic sites are no PRCV ORF3-1
longer expressed by PRCV (see below).
Whether such a large truncation alters any In the Purdue-115 and FS772 TGEV
biological function of the protein is so far strains, ORF3b is assumed to be ex-
unknown.
pressed from the functionally bicistronic
mRNA3 since no functional conserved se-
PRCV ORF3a pseudogene quence is found downstream of ORFs 3a
(Rasschaert et al, 1987; Britton et al,
The ORF3a region of the Eur PRCV ge- 1990). In contrast, the Miller strain, which
nome contains 3 major deletions (Rass- has a CUAAAC motif between ORF3a and
chaert et al, 1990; Page et al, 1991):a 13 3 3b, produces an additional mRNA species
nt deletion which covers part of the con- (3.7 kb; Wesley et al, 1989), designated
sensus sequence; a 22 nt deletion which mRNA3-1 in accordance with the rules pro-
suppresses a non-coding stretch, the initia- posed by the ICTV coronavirus subgroup
tion AUG codon and 2 ORF3a codons; a (Cavanagh et al, 1990).
36 nt deletion internal to the ORF3a coding In PRCV, the ORF3b sequence is ex-
sequence (fig 3). In contrast, a unique de- pressed through the synthesis of a specific
letion 5 nt long and located within the mRNA species: in both Eur and USA iso-
ORF3a coding sequence is observed in lates a CUAAAC sequence is present up-
the USA isolate (Wesley et al, 1991How- stream (instead of CUAAAU in Purdue and
ever, transcripts of the relevant size were FS772 TGEV strains), so that transcription
can be initiated at this site (Rasschaert et could have derived from a common ances-
al, 1990; Britton et al, 1991; Wesley et al, tor having the ORF3a gene altered by mu-
1991) (fig 3). The corresponding PRCV tation of the hexameric sequence or short
transcript is thus homologous to the extra frameshift deletion in the coding sequence.
mRNA species (mRNA3-1) produced by Subsequently, additional and possibly dif-
the Miller strain. Whether the synthesis ef- ferent modifications may have occurred. It
ficiency of the ORF3b product is modified is to note that the 36 nt deletion in Eur
compared to that from mRNA3a-b has not ORF3a overlaps the 5 nt deletion in USA
been examined. ORF3a (fig 3). However, such a mecha-
nism does not apply simply to the S gene,
since the respective deletions overlap only
How related are Eur partially. Moreover, the nonoverlapping se-
and USA PRCV viruses ? quences, ie codons 5, 6 in USA PRCV and
5 to 9 in Eur PRCV are endogenous TGEV
As mentioned above, the alterations ob- sequences (fig 2). It can be inferred from
served in USA and Eur isolates, though these findings that the 2 pneumotropic var-
being identically targeted in PRCV ge- iants arose independently, a conclusion
nome, are different: i) the S gene trunca- further strengthened by the fact that they
tion differs in length and position; ii) 3 dele- do not cocirculate but spread -

up to now
tions are present in Eur PRCV ORF3a -

in geographically distinct swine popula-


gene instead of 1 short deletion for USA tions. One important implication of this no-
PRCV; in the latter, the hexameric se- tion is the possibility of a functional link be-
quence is mutated, not deleted. This rais- tween the ORF3a and S gene alterations
es the question of the phylogenic relation- (one acting in such a way that it compen-
ship of the 2 variants. Theoretically, they sates a defect induced by the other?).
How the deletions were generated into Loss of enteropathogenicity of PRCV
the PRCV genome is less clear. A unique
phenomenon associated with coronavirus- Like many spike glycoproteins, the corona-
es is the extremely high-frequency RNA re- virus S protein plays a crucial role in both
combination that can be evidenced during the attachment of virions to the target cells
a single round of infection. Recombination and the fusion between the viral and cellu-
is thought to occur through a copy-choice lar membranes. In several reports, the al-
mechanism, reflecting the tendency of the tered neurovirulence of murine hepatitis vi-
polymerase to pause then to jump to an- rus (MHV) variants has been related to the
other RNA template (review: Lai, 1990). In presence of a deletion or even of a point
addition, defective RNAs, composed of mutation in the amino-half, globular part of
several discontiguous parts of the parental the S protein, thus indicating that the latter
genomic RNA are frequently observed in bears major virulence determinant(s) (Fa-
infected cells. Therefore, deletions may zakerly et al, 1992 and references therein).
have been introduced into the PRCV ge- Also, several TGEV neutralization escape
nome by a very same mechanism, imply- mutants encoding a S protein with a single
ing the binding of pausing RNA intermedi- substitution or a short deletion within its N-
ates to a site downstream of the original terminal region were found to exhibit a
pausing sites, thus deleting various inter- markedly reduced enteropathogenicity for
nal portions of the genome. Inspection of the newborn piglet (Bernard and Laude, to
TGEV sequence did not reveal any repeat- be published). In this regard, the truncation
ed sequences flanking the different cross- of PRCV S protein appears to be highly
over sites, as frequently observed for intra- relevant.
or intergenomic homologous recombina-
Aminopeptidase N (APN), an ectoen-
tion; 2 identical heptanucleotide sequenc-
zyme abundantly expressed at the brush
es were found in the vicinity of the seg-
border membrane of the small intestinal vil-
ment unique to TGEV S gene, but not right
li, is known to act as a major receptor for
each side of the putative cross-over sites TGEV (Delmas et al, 1992a). The first hy-
(Rasschaert et al, 1990). In conclusion,
PRCV derived from TGEV by successive
pothesis coming naturally to mind is that
the modified tropism of PRCV could be
deletions which likely occurred through a
due to an impaired interaction between at-
mechanism analogous to illegitimate re-
tachment protein and cellular receptor.
combination. As an important conse-
However, recently reported observations
quence, the appearance of a revertant have indicated that PRCV may also use
PRCV is highly improbable. APN for gaining entry into cells (Delmas et
al, 1992b). First, BHK cells, which are re-
Genetic basis of the PRCV phenotype
fractory to PRCV, became susceptible
once transfected with the cDNA encoding

porcine APN. Second, anti-APN monoclo-


In this section, the possible significance of nal antibodies were able to block efficiently
the major modifications - ie deletions - the multiplication of PRCV in cell culture.
identified in the sequenced part of PRCV Yet these in vitro findings may reflect the in
genome with respect to its altered tropism vivo situation incompletely. It can be spec-
will be discussed. For the sake of clarity, 2 ulated for instance that the N-terminal do-
aspects will be considered separately, main contributes in some way to the stabili-
which does not imply that they are actually ty of the S protein. Then, exposure to the
disconnected. physicochemical environment of the diges-
tive tract could result in a decreased at- quivocally to the ORF3a defect, these find-
tachment or fusogenic capacity of PRCV S ings point to a possible involvement of
protein. This should not be an all or none ORF3a in the expression of TGEV ente-
phenomenon, however, since both isola- ropathogenicity. If confirmed, this would
tion and replication of PRCV in the neo- mean that the ORF3a product, which is
nate small intestine have been described dispensable for virus replication in cell cul-
(see Pathogenesis section). ture, plays a crucial role in modulating the
In any case, the possibility that PRCV virulence for the animal.
restriction may take place after penetration
into the enterocyte should not be disre- Respiratory tropism of PRCV
garded. Examining the susceptibility to
TGEV of clones of cell lines derived from Two kinds of cells have been shown to
various tissues or species and stably ex- support PRCV replication in the respiratory
pressing porcine APN, led to an intriguing tract: the epithelial cells, that are likely to
observation. All the transfected clones un- express APN at their membrane surface
derwent cell lysis upon infection at an ap- (Ito et al, 1980) and the alveolar macro-
propriate multiplicity and synthesized the phages; interestingly enough, APN and
viral structural polypeptides, yet only part CD13 - a marker of the monocyte-
of them yielded infectious particles (Del- macrophage lineage - are the same mole-
mas et al, 1992b). This suggests that the cule in the human species (see Delmas et
capacity to bind the receptor is a crucial al, 1992). It is thus conceivable that PRCV
but not unique determinant of the viral tro- uses APN as a receptor for gaining entry

pism. into the pulmonary cells, although the use


These observations lead focusing the of an alternative molecule cannot be for-
attention towards the second alteration of mally excluded. The recent finding that
PRCV genome, ie the conversion of HCV 229E, a human respiratory coronavi-
ORF3a to a pseudogene. ORF3a encodes rus, also uses APN as a receptor (Yeager
a 71 to 72 amino acid-long non-structural et al, 1992) brings some support to this
view.
polypeptide, which has been detected in
TGEV infected-cells (Godet, 1992). A ho- Whether PRCV possesses an increased
mologous gene is present only in the en- tropism for the respiratory tract compared
terotropic canine CCV and feline FECV/ to TGEV may be a more controversial is-
FIPV coronaviruses. Two cell-adapted sue. Indeed, replication of TGEV in the
TGEV strains, SP and Nouzilly SG188 ex- respiratory tract has been reported by a
hibit, like PRCV, a small plaque phenotype number of authors (Harada et al, 1969; Un-
in cell culture and a markedly reduced derdahl et al, 1974, 1975; Kemeny et al,
growth in the intestine. Strikingly enough, 1975, 1977, 1978; Furuuchi et al, 1979;
a deletion involving part of the ORF3a and Sprino et al, 1982; La Bonnardi6re and
part of the downstream ORF3b was identi- Laude, 1983 and unpublished data; Wes-
fied in both strains, which apparently en- ley et al, 1990a; Cubero et al, 1992). Also,
code a S protein of normal size (Wesley et the alveolar macrophages have been
al, 1990b; Britton et al, 1992). In addition, shown to support the replication of cell-
mRNA 3 of the high-passage, avirulent adapted and virulent strains in vitro or in
Miller-60 virus, could not be detected by vivo (Laude et al, 1984). Attention should
Northern blot analysis (Wesley, unpub- be drawn, however, to the nature of the
lished data). Even though the attenuation strain assayed as well as to the technique
of these viruses cannot be related une- employed for assessing virus multiplication
(eg histoimmunodetection versus infectivi- Antigenic features
ty titration), when interpreting the pub-
lished data. A tentative conclusion from
The fact that PRCV and TGEV share many
the available information is that many cell-
adapted strains (including a pathogenic antigenic determinants and cannot be dif-
ferentiated by conventional serological
strain having no more than 10 passages
in culture, such as D52) replicate in the techniques was acknowledged at the very
lungs to substantial titers. This might be beginning of the investigations. The 2
no longer true with pig-passaged wild agents exhibit a complete 2-way cross-
strains of TGEV. Growth of the Miller and neutralization when using polyclonal sera.
Purdue wild strains was found to occur At the level of individual polypeptides, im-

only at a limited extent (Wesley et al, munoblotting reveals a reciprocal cross-


1990a; Pensaert et al, unpublished re- reactivity of the S, M and N antigens
sults); there might be exceptions, howev- (Callebaut et al, 1988). The identification of
distinctive determinants could only be
er, such as the Nebraska isolate, which
has been reported to replicate to a similar achieved through the use of panels of
monoclonal antibodies (MAbs) previously
efficiency in the digestive and respiratory
tracts (Underdahl et al, 1974). Additional raised against TGEV (Jimenez et al, 1986;
studies comparing the multiplication of Laude et al, 1986; Garwes et al, 1987).
PRCV and TGEV in pulmonary tissues
may be necessary to answer that ques- S antigen
tion conclusively. In any case, it is worth
to keep in mind that, even though air- Earlier studies on the antigenic structure of
borne infection by TGEV has been report- Purdue strain S protein have defined 4,
ed to occur, the efficiency of spreading by
possibly 5 major antigenic sites, all located
aerogenic way does not appear to be in the amino-half of the polypeptide chain
comparable to that of Eur PRCV (the limit- (Delmas et al, 1986; Correa et al, 1988,
ed diffusion of USA PRCV is likely to be
1990; Delmas et al, 1990; Gebauer et al,
related to differences in the European and
US industries rather than to a distinct phe-
1991).From the analysis of the PRCV-
binding activity of anti-TGEV S MAbs
notype). (Callebaut et al, 1988; Laude et al, 1988;
Finally, one remains faced to 2 main hy- Sanchez et al, 1990), 3 main conclusions
potheses. Firstly, PRCV originates from a can be drawn (fig 4).
particular TGEV strain having originally a
Firstly, the MAbs defining the partially
respiratory tropism. Secondly, the ability of
PRCV to replicate in pulmonary cells to a overlapping sites A and B are fully reactive
towards PRCV. Both these sites are com-
high level relies on the alteration of S and/
or ORF3a genes. That a virus could ac-
prised of epitopes which are highly con-
served among TGEV strains and induce
quire such a capacity by the introduction of
deletions in its genome is puzzling and, to strongly neutralizing antibodies. The site A
has been recently reported to be immuno-
our knowledge, unprecedented. One possi-
dominant in both TGEV- and PRCV-
bility that could be explored in this respect immune sows (De Diego et al, 1992).
is whether the introduced genomic
change(s) may result in a delayed or de- Secondly, the MAbs which discriminate
creased cytopathic effect, thus allowing the between TGEV and PRCV generally exhib-
virus multiplication to take place for several it no significant neutralizing activity. By dif-
virus cyles, which is unlikely to occur in ferentiating MAbs, it is meant those which
TGEV-infected enterocytes. recognize epitopes expressed by all or
most TGEV strains, but not by any of the Britain and The Netherlands display nearly
PRCV isolates tested so far. Such epi- identical reactivity patterns. This result,
topes are grouped into 2 distinct antigenic consistent with the sequence data, con-
sites : i) the site D (probably overlapping firms the unicity of Eur PRCV. However,
the site designated B by the Madrid some antigenic variation can be evidenced

group); ii) a site not expressed on the na- by using MAbs defining the site C (D Ma-
tive virions (C Madrid), which is less con- drid), which shows some variability among
served than the former. Both of these sites TGEV strains as well. The antigenic pat-
have been mapped within the region of the tern of USA PRCV has not been studied in
polypeptide chain predicted to be removed detail, but the reactivity of the Ind89 strain
in the PRCV S molecule, thus providing a towards anti-TGEV MAbs representative of
straightfoward explanation for the lack of the sites A, B, C, and D is simiiar to that of
binding of the relevant MAbs. Few non- Eur PRCV (Wesley et al, 1990a).
neutralizing anti-S MAbs, defining epitopes Information regarding PRCV S-specific
unrelated to the major sites, have also determinants is comparatively scarce. Sev-
been found to be differentiating. Two site en PRCV MAbs, selected on the basis of
D MAbs exhibit interesting peculiarities their lack of reactivity towards the Purdue
(Laude et al, 1988): unexpectedly, the TGEV strain, were all directed against the
MAb 78.17 cross-reacts with PRCV, which S protein. Three of them, all of which were
may suggest that the cognate epitope is neutralizing, did not react with any of the 5
assembled, one component being located TGEV strains tested, whereas the other,
outside of the truncated domain; the MAb
non-neutralizing MAbs recognised com-
40.1 is the sole differentiating MAb show- mon epitopes (Deriabine and Laude, un-
ing a high neutralizing activity; strikingly, it published data). This is in contrast with the
is by use of this MAb that escape TGEV fact that nearly all differentiating anti-TGEV
mutants attenuated for the young piglet MAbs were not neutralizing. Whether the
were selected.
neutralizing PRCV MAbs define a site dis-
Thirdly, the PRCV isolates originating tinct from those previously identified on the
from Belgium, Denmark, France, Great S molecule remains to be investigated.
N and M antigens cytes similar to TGEV (Charley and Laude,
1988 and unpublished data).
Anti-N and anti-M MAbs raised against In conclusion, several epitopes absent
TGEV are not differentiating antibodies on PRCV have been identified by use of
since they recognize epitopes which are ei- anti-TGEV S monoclonal antibodies. Such
ther conserved or inconsistently expressed dissimilarities have been exploited for de-
on both the viruses (Laude et al, 1988;
veloping an ELISA test for the differential
Sanchez et al, 1990). Anti-N MAbs defin- detection of antibodies against each virus
ing the antigenic site A, located within the (see Diagnosis section).
amino-half part of the polypeptide chain,
exhibit a good cross-reactivity, whereas
those defining the site B, located within the INFECTION
carboxy half part, show an altered reactivi-
ty toward most of the PRCV isolates tested
(Alonso et al, 1992). These studies also re- Pathogenesis
vealed some antigenic heterogeneity of
PRCV N protein, not observed with TGEV,
which may be the trait of a young virus, not The pathogenesis of experimentally in-
fully adapted to its ecological niche (San- duced PRCV infections has been found to
chez et al, 1990). differ depending on the age at infection.

Concerning the M antigen, MAbs direct- Pigs < 1 wk old, either hysterectomy-
ed against epitopes previously mapped derived and colostrum-deprived (HDCD) or
within the short (some 30 residues) N- conventional but devoid of antibodies
terminal region protruding at the outer face against PRCV, have been inoculated by
of the virion envelope, were found not to aerosol (Cox et al, 1990a) or oronasally
react with any of the 3 PRCV isolates test- (O’Toole et al, 1989; Pensaert et al, 1986).
ed, whereas MAbs assumed to recognize They were euthanized at different time in-
the C-terminal, internal region of the M pro- tervals after inoculation and examined for
tein were cross-reactive (Laude et al, virus. PRCV was isolated from the nasal
1988). These findings correlate well with mucosa, tonsils, trachea and lungs. Virus
the sequence data: the 64 carboxy-last was also recovered from the stomach and
amino acids of 2 PRCV isolates arid of the small intestine and its contents
TGEV strains Purdue and FS772 are iden- (O’Toole et al, 1989; Cox et al, 1990a). In
tical, whereas 4 differences are noted with- the aerosol infected group, a number of
in the first 30 N-terminal residues, a diver- other tissues including plasma, mesenteric
gence comparable to that of the 2 TGEV lymph nodes and colon were consistently
strains (Laude et al, 1987; Rasschaert et positive, while virus was sometimes isolat-
al, 1990; Britton et al, 1991Moreover, 2 ed from other lymph nodes, spleen, liver
of these changes involve amino acids and thymus. Yet, the respiratory tract is the
shown to be crucial for epitope expression main target organ and virus titers in the ap-
(Laude et al, 1992). In essence, it would ical lung lobe are as high as 10
83 TCID
/g
sc
appear that the M protein epitopes con- tissue. These titers are significantly higher
served between TGEV and PRCV are in- than those obtained in any other organ.
ternal. Finally, despite some amino acid Using immunofluorescence (IF), the virus
changes in the N-terminal domain of M has been shown to infect the epithelial
protein, PRCV is able to induce an a- cells of nasal mucosa, tonsils, trachea and
interferon synthesis in naive swine lympho- lung tissue and the alveolar macrophages.
In the lungs, fluorescence was detected (Pospischil et al, 1990) and immunocyto-
mainly in the epithelial cells of alveoli (fig 5), chemistry (O’Toole et al, 1989). Virus isola-
but also in bronchioli and bronchi. A maxi- tion from the stomach suggests that virus
mum of infected cells is seen at 3 d post in- may reach the intestine after being ingest-
oculation (PI) in the bronchi and between 3 ed, but a spread from the respiratory tract to
and 5 d PI in alveoli and bronchioli (Cox et the gut via viremia cannot be ruled out. The
al, 1990a). Though viremia can occur as way of virus spread from the caudal to the
early as 2 d PI, as evidenced by virus isola- cranial small intestine is unclear. In piglets
tion from plasma, no virus was seen by IF inoculated with PRCV directly into the lu-
in parenchymal organs.and lymph nodes. men of the intestinal tract, more than 10 3
Viral antigen was demonstrated in a few ep- 50
TCID of virus were needed to start the in-
ithelial cells in the small intestine of neona- testinal infection (Cox et al, 1990c). These
tal pigs (Cox et al, 1990a). The intestinal in- findings, together with the very limited ex-
fection begins in the ileum and gradually tent of intestinal replication, indicate that the
moves upwards to the duodenum. Four to 5 susceptibility of the small intestine to infec-
d PI, all small intestinal segments are infect- tion with PRCV is rather low. Small intesti-
ed (Cox et al, 1990a) and a maximum of 4 nal enterocytes are very susceptible cells to
fluorescing cells are present per ileal sec- TGEV, allowing a highly productive virus
tion. The infected cells were identified as replication, but the gut is not a target organ
villous enterocytes by electron microscopy for PRCV.
In 5-wk-old HDCD pigs inoculated with Clinical signs and pathology
PRCV by aerosol, virus replication was
limited to the respiratory tract (Cox et al,
It is still a subject of debate whether or not
1990b). The pathogenesis of the res- PRCV causes clinical disease signs. At the
piratory infection was similar to that in time of the first detection of PRCV in the
neonatal piglets except for the absence of
fluorescence in tonsils. However, the Belgian swine population, the virus was
considered to be non pathogenic. First iso-
number of fluorescent cells in the lungs
lations of PRCV were made from the res-
was lower in these pigs compared to the
1-wk-old piglets and fluorescence in piratory tract of clinically normal piglets
bronchi was even rare. Viremia was not (Pensaert et al, 1986). Clinical disease is
not a feature when a PRCV infection-wave
detected. Of main importance is that no
occurs on closed breeding-fattening
intestinal replication was observed. Yet,
farms. In the longitudinal serological stud-
infectious virus was ingested since PRCV
ies mentioned above, no disease was as-
was isolated at low titres from the cranial
sociated with seroconversion of fattening
intestinal tract of some of the piglets.
When 5-wk-old pigs with maternal neutral- pigs on the closed farms.
izing antibody titer of 48 were inoculated, However, some field reports have linked
virus production in the respiratory tract a PRCV infection with respiratory disease
was not decreased compared to that in in growing, finishing and adult swine. A
pigs without PRCV-neutralizing antibod- serological,survey in pig herds with respir-
ies. atory disease in Brittany (France), re-
vealed seroconversion to PRCV and not to
In adult swine, the pathogenesis of
influenzaviruses or Aujeszky’s disease vi-
PRCV has not been studied in detail, but
rus in 2 out of 6 herds (Jestin et al,
some data are available (Van Reeth et al,

unpublished results). Conventional fatten- 1987b). Pigs became infected with PRCV
either shortly after arrival on the fattening
ing swine (80-100 kg) without PRCV- farm or towards the end of the fattening
seroneutralizing antibodies were infected
by aerosol with 7 different Belgian PRCV period. Anorexia and fever were the main
clinical signs, coughing was seen on occa-
field isolates. Virus titers in lung tissues
between 4-6 d ranged from . 1072
-
103
2 sion. Similar observations were made by
No clear-cut differences Laval et al (1991).Clinical manifestations
!g.
5c
TCID ap-
have also been reported in lactating sows
peared between the distinct isolates. with
regard to virus quantity and degree of undergoing a PRCV infection (Ulbrich et al,
fluorescence in the lungs. Fluorescence 1991They were characterized by fever
was restricted to a few alveolar epithe-
and inappetence, followed by a barking
lial cells, whereas bronchi and bronchioli cough a few days later. Such observations
were negative. The extent of virus rep-
have not been made by the authors since
lication was similar in apical, cardiac and experimental infections of sows in the field
remained completely subclinical.
diaphragmatic lung lobes. In pigs which
were clinically followed, virus shedding in Whether field infections with PRCV re-
nasal secretions started 1 to 2 d PI and sult in respiratory disease or not will re-
mean viral titres ranged from 105.5-106.5 main an open question. Multiple environ-
/g nasal secretions between 2 and
50
TCID mental factors such as climate, housing,
5 d PI. Thereafter, virus amounts gradually presence of concomitant infections and
decreased and excretion continued until many others will play a role in determining
8 d PI. the appearance of disease signs. In the
study mentioned earlier (Van Reeth et al, body temperatures did not return to normal
unpublished results) newly arrived groups until 2 wk PI. Nasal discharge and epistaxis
of feeder pigs were examined in industrial were observed occasionally.
fattening units with respiratory disease in Recently, experimental infections of con-
autumn and with minimal or negligible res- ventional fattening swine (80-100 kg) have
piratory signs in spring. In both circum- been carried out in the authors’ laboratory
stances, infections with PRCV occurred to- (Van Reeth et al, unpublished results).
gether with H1N1-and/or H3N2- Each of 7 PRCV Belgian field isolates was
influenzavirus infections. Simultaneous ex- inoculated by aerosol in 2 fattening pigs,
perimental infections with H1N1 or H3N2- free of PRCV-sero-neutralizing antibodies.
influenzavirus and PRCV did not appear to Fever and inappetence generally developed
enhance the pathogenicity of these viruses 2 d PI. Mild respiratory signs such as nasal
(Lanza et al, 1992). However, in most re- discharge and sneezing were recorded.
cent experiments where 9-wk-old piglets Two of 13 pigs, each infected with a differ-
were infected 2 or 3 d after an experimen-
ent isolate, did not show any sign of dis-
tal PRCV infection, clinical signs and lung
ease. Variation in virulence between the
lesions were more severe in dual infected
isolates tested was not detected. With one
animals than in those infected with either
of these isolates, aerosol inoculation was
of the respiratory agents alone (Van Reeth
performed on a larger group of 6 fattening
et al, unpublished results). swine. These 6 pigs showed symptoms
Clinical signs have been reported in suggesting an involvement of the deeper
some experimental inoculation studies and airways including coughing, laboured
not in others. They vary considerably in se- breathing and increased respiration.
verity and this may be influenced by the Pathological changes on field cases
age of pigs and/or the inoculation tech- have not been reported. A catarrhal lobular
nique. One-wk-old specific pathogen free bronchopneumonia is most consistently
(SPF) pigs inoculated either by aerosol found in experimentally inoculated pigs (Du-
(Cox et al, 1990a) or oronasally (O’Toole et ret et al, 1988; O’Toole et al, 1989; van
al, 1989) failed to develop clinical signs. Nieuwstadt et al, 1989; Cox et al, 1990a;
Five-wk-old piglets, either HDCD or con-
Vannier et al, 1990). Histological examina-
ventional with or without PRCV maternal
tion of the lungs of HDCD piglets revealed
antibody titers, remained asymptomatic fol- an interstitial pneumonia with some degen-
lowing aerosol inoculation (Cox et al, eration of the alveolar and bronchiolar epi-
1990b). Also, in another study, pigs at the thelium. The lumen of bronchioli and alveoli
age of 5 wk were clinically healthy after in- is filled with macrophages and cellular de-
tranasal infection with PRCV (van Nieuw-
bris. Regeneration starts at=1 wk PI and is
stadt and Pol, 1989). On the other hand, in-
characterized by hyperplasia of bronchiolar
tratracheal inoculation of SPF pigs at 90 d
of age resulted in mild clinical signs (Vanni- epithelium, alveolitis and modest peribron-
chial cuffs of lymphoblasts, macrophages
er, 1990). Dyspnea, polypnea, short lasting
and plasma cells (O’Toole et al, 1989).
fever and prostration with temporary loss of
performances were recorded in most but
not in all the pigs. More severe disease
signs have been described by Duret et al Immunity to PRCV and TGEV

(1988) following intratracheal inoculation of


SPF pigs at 25 kg. All the pigs experienced Upon aerosol inoculation of pigs with
fever, sneezing and some dyspnea and PRCV, serum antibodies are detectable 1
wk later. The antibody titre increases until nary Virology, Faculty of Veterinary Medi-
3-4 wk after inoculation. Protection of the cine, Gent; Regional Veterinary Investiga-
respiratory tract against reinfection is not tion Centre, Torhout and National Institute
long lasting since regular reinfections with for Veterinary Research, Brussels), diag-
PRCV occur in sows in the field (Callebaut nosis of TGEV has been made 68 times
et al, 1990). Also, experimental reinfection during the year 1982-1983, 61 times in
of 6 pigs was possible as soon as 6 wk af- 1985-1986, but only 7 times in 1988-
ter a primary infection using the intranasal 1989. From the beginning of 1992 till now
or intrapulmonary inoculation technique (end of November 1992) only one case
(van Nieuwstadt et al, 1992). Virus neutral- has been reported. Therefore, the question
izing antibody titers in these pigs increased has arisen if PRCV and TGEV, whose
from 794 to 10 000. In the field, attempts structural antigens are nearly identical, can
were made at experimental reinfection of induce cross-protection in vivo. Protection
fattening swine with PRCV serum antibod- of suckling piglets against an enteric TGEV
ies (VN titers 16-192). Upon challenge, vi- infection is based on the uptake of specific
rus replication did not occur in some of (lactogenic) IgA antibodies in the milk of
these pigs, as concluded from the absence TGEV-immune mothers (Bohl et al, 1972).
of seroconversion. In other pigs though, Sows immune following a natural PRCV in-
neutralizing antibody titers up to 3 072 fection do secrete TGEV-neutralizing anti-
have been detected 3 wk after challenge bodies in their milk. These antibodies
(Van Deun et al, unpublished results). though are not always of the IgA class
Because of the high prevalence of (Callebaut et al, 1990). Experiments have
PRCV, the majority of litters on a farm are been carried out in (field) sows with PRCV
born from immune sows and acquire ser- serum antibodies and thus previously in-
um antibodies through the colostrum. In fected with PRCV at an unknown time.
the absence of an infection, these mater- When these sows were reinfected with
nal antibodies decline with a mean half-life PRCV oronasally during the last weeks of
of 12.04 d and persist at the latest until the pregnancy or early in lactation, either in-
age of 16 wk (Pensaert et al, 1986). De- duction or a rise in titer of lactogenic anti-
clining maternally-derived antibody titres TGEV IgA was obtained in all of them. By
do not protect against a PRCV infection 24 wk after the experimental reinfection
because PRCV virus titres and sites of rep- however, IgA antibody titers were signifi-
lication in the respiratory tract were not dif- cantly decreased or even absent (Van
ferent when 5-wk-old pigs with or without Deun et al, 1990 and unpublished results).
maternal PRCV-antibodies were experi- Regular PRCV infection-waves on breed-
mentally inoculated (Cox et al, 1990b). Be- ing farms, therefore, most probably favor
sides, infectious virus was excreted during and maintain the presence of anti- TGEV
8-13 d by the maternally immune pigs, IgA in sow milk. The origin of IgA in sow-
compared to 6-11 d by the seronegative milk after a respiratory PRCV infection is
pigs. In the field, passive immunity fre- unclear since there is no direct evidence of
quently turns to active immunity between an immunological link between the respira-
the age of 5-10wk. tory tract and the mammary gland, as is
The wide distribution of PRCV in the known to exist between the gut and mam-
swine population has been accompanied mary gland (Bohl et al, 1972).
by a marked reduction in the number of Yet, the main point is the efficiency with
TGEV outbreaks. In the 3 diagnostic labor- which PRCV-induced lactogenic IgA pro-
atories in Belgium (Laboratory of Veteri- tects suckling piglets against an enteric
TGEV infection. In the aforementioned ex- enzootic, 90-100% death loss of neonatal
a

periment (Van Deun et al, 1990 and un- pigs during a period of 3 wk after the intro-
published results) naturally PRCV-infected duction of TGEV was a common finding. In
sows, reinfected with PRCV during preg- 1987, 3 outbreaks of enteric TGEV were fol-
nancy (and secreting lactogenic TGEV- lowed on PRCV-immune breeding farms
IgA) protected their offspring to a certain and litters born 10, 11 and 14 d after the
degree. Piglets of these sows developed start of the outbreak remained healthy (Pen-
diarrhea upon challenge with TGEV but saert et al, 1987). This can be explained by
the incubation period was delayed by 1-2 a priming effect of a PRCV infection in the
d and mortality rates were markedly low- sows. When PRCV-primed sows subse-
ered in 3 out of 6 litters (0-12.5%). Partial
quently become infected with TGEV, an an-
protection was also reported by Bernard et amnestic response will quickly develop.
al (1989) in litters of sows which had been That way, a rapid increase of IgA antibodies
naturally infected with PRCV. Five out of 7 in milk will occur and piglets born a few d af-
litters were protected against mortality af- ter the start of the outbreak already receive
ter an experimental TGEV challenge, re-
a solid lactogenic immunity.
sulting in an overall mortality rate of 44%. Another immunological aspect is the
Similar levels of protection were obtained
upon TGEV challenge of piglets nursing question of whether a respiratory PRCV in-
sows which had been infected experimen-
fection can induce an intestinal mucosal
tally with PRCV at 84 and 104 d of preg- immunity and protection against TGEV.
Van Nieuwstadt et al (1989) could not
nancy (De Diego et al, 1992). Generally,
PRCV-induced lactogenic antibodies pro- demonstrate such protection in specific
tect pigs against TGEV to a lesser degree pathogen-free pigs. TGEV antigen excre-

than TGEV-induced antibodies. This dif- tion in the faeces was of the same duration
ference is not due to IgA characteristics in PRCV immune as in non-immune con-
since the neutralizing capacity and/or the trol pigs. Recently, however, it was demon-
secretory component of both IgA’s are the strated that conventional pigs which are in-
same (Callebaut et al, unpublished re- oculated by aerosol with PRCV at the age
sults). In experiments by De
carried out of 6 wk and in the absence of maternal
Diego et al (1992) lactogenic protection in PRCV antibodies, are partially protected
piglets suckling from TGEV-immune moth- when challenged with TGEV 4 wk later
ers was higher than in piglets from PRCV- (Cox et al, 1992). Aerosol inoculation with
immune sows and this corresponded with PRCV appears to prime both the systemic
higher milk antibody titers. Cross- and the intestinal immune system. Chal-
protection between PRCV and TGEV was lenge of the PRCV-immune pigs with
not observed by Paton and Brown (1990), TGEV resulted in a rapid and drastic in-
since litters from PRCV-immune sows crease in TGEV neutralizing antibody ti-
were protected against TGEV in their
not tres-with titers ranging up to 1 280 2 wk lat-
experiments. TGEV-antibodies in sowmilk er- and infectious TGEV was excreted in
have been demonstrated in this study but feces during 0-4 d, compared to 5-6 d
no characterization with regard to immuno- when fully susceptible pigs are inoculated.
globulin class was done. The mechanism through which PRCV
In PRCV-immune breeding herds, TGEV primes the intestinal immune system is not
outbreaks are of a milder course which may known at the present time, but the PRCV-
be due to the presence of lactogenic IgA in TGEV tandem may be very suitable to
sowmilk, but they are also of a shorter dura- study the common mucosal immune sys-
tion. Prior to the situation with PRCV being tem in more detail.
LABORATORY DIAGNOSIS tions of lung tissue (O’Toole et al, 1989;
van Nieuwstadt et al, 1989), but this tech-
A clinical diagnosis of PRCV is not possi- nique is not applied for routine diagnosis.
ble. A PRCV infection can be diagnosed in By electron microscopic examination, parti-
cles with a typical coronavirus morphology
the laboratory by isolating the virus, by
were detected in bronchiolar cells, in alve-
demonstrating viral antigens in lung tissue olar macrophages and free in the alveoli
or by detecting seroconversion.
(O’Toole et al, 1989). Differentiation with
PRCV can be cultivated in a variety of TGEV is not possible with the electron mi-
cell cultures. The virus was first isolated in
croscope. Finally, the use of nucleic acid
primary pig kidney cells, but also grows in probes in dot-blot hybridisation has been
continuous cell cultures of swine kidney recently proposed as a means for selective
(PK15), swine testicle (ST) and a continu- diagnosis (Jackwood et al, 1992).
ous cell line of cat foetuses (FCWF). Be-
A serological diagnosis can be made
cause of its high susceptibility, the ST
with the classical virus neutralization test
swine testis cell line (McClurkin and Nor-
(Voets et al, 1980) or with an ELISA test
man, 1966) has become most widely ap-
(Paton et al, 1991using TGEV as an anti-
plied for virus isolation (Pensaert, 1989).
gen. One of the major shortcomings of the
Five d after seeding, fully sheeted swine
test is that it cannot differentiate between
testis cells are most sensitive. The cyto-
PRCV- and TGEV- induced antibodies. If
pathic effect in ST cells is characterized by no problems of diarrhea have been en-
syncytium formation, followed by rounding countered in a herd and if there is no evi-
up of cells. Formation of small syncytia dence of enzootic TGEV, then TGEV-
may also be observed when wild TGEV
strains are first isolated in ST cells. With
neutralizing antibodies are considered to
be PRCV-induced. However, no guarantee
PRCV, the entire monolayer may be de- can be given that TGEV is not involved. In
stroyed by 2-3 d after inoculation. Identifi- the mid eighties, the wide prevalence of
cation can be performed by immunofluo-
PRCV had created serious obstacles for
rescence, and differentiation between the
export of pigs to TGE-free countries. A
2 agents should be possible by the use of
clear need for a differential test had there-
available TGEV- or PRCV-specific anti-S fore arisen, not only for export purposes
MAbs (see above: antigenic features). but also for research. To meet this require-
Lung tissue or nasal swabs are the speci- ment, efforts have been channeled into de-
mens of choice for virus isolation.
veloping a differential competitive inhibition
The fluorescent antibody test on lung ELISA (Garwes et al, 1988; Callebaut ef al,
tissue of experimental animals is often per- 1989; Have, 1990). All these tests use a
formed. Cryostat sections are stained with non-neutralizing MAb directed against an
an anti-TGEV conjugate and examined by epitope on the S protein in TGEV with no
fluorescence microscopy. This approach is counterpart in PRCV, in order to permit the
also not PRCV-specific, since some TGEV detection of TGEV-specific antibodies with-
strains have been reported to grow in the out interference of PRCV antibodies. Two
respiratory tract (Underdahl et al, 1974; conditions are required for the test to be
Kemeny et al, 1975). Onno et al (1989) operational; first, the assay must be sensi-
have described an indirect immunofluores- tive enough to detect the low levels of anti-
cence test on smears of nasal cells. Alter- bodies that may be present early in TGEV
natively, viral antigen can be detected by infections; second, the reactivity of the
immunoperoxidase staining on frozen sec- MAb(s) used must be extended to a panel
of TGEV strains circulating in the field. The be tested first in the seroneutralization test
principle of a test which has been used at and, if positive, the differential ELISA may
a large scale and proved to be equivalent then be performed.
in sensitivity to neutralization (Callebaut et
al, 1989) is schematized in figure 6. False
positive results have not been reported but CONCLUSIONS
false negative results may occur (Calle-
baut et al, 1989; Have, 1990). It is recom- There is now strong evidence that PRCV
mended therefore, to test several serum is nothing else than a deletion variant of
samples from the same herd. The classi- TGEV. Whether a peculiar context has fa-
cal neutralization test no longer provides a voured the emergence of PRCV remains
method for the serological diagnosis of an unanswered question. In particular, the
TGE, but is still essential. It is the combi- fact that actually 2 different deletion mu-
nation of a negative result in the blocking tants have emerged independently in 2 dif-
ELISA and a positive result in the seroneu- ferent continents (Europe and the USA),
tralization test that gives evidence of a while TGEV apparently had undergone no
PRCV infection. Consequently, sera will recognizable changes since its original iso-
lation in 1946, is intriguing. Also, PRCV cused picture has emerged, and laborato-
provides the first example of a coronavirus ry tools for a selective diagnosis of each
for which spontaneous genomic deletions kind of infection have been provided. Yet,
have resulted in a considerable epizootio- much remains to uncover about the inti-
logical impact. This supports the view that mate mechanisms which led to such a re-
not only recombination but also deletion modeling of the original TGEV tropism.
events represent a driving force in corona- Two genes have been shown to be al-
virus evolution. tered in PRCV genome, but whether the
The emergence of PRCV has brought observed phenotypical changes have a
about consequences which are clearly mono- or multigenic origin remains an

beneficial to researchers and to the pig open question. However, several of the is-
sues raised in this article should be amen-
producers. The virus has become very
widespread in Europe since its first ap- able to solution, in particular by the engi-
pearance in the mid-eighties and is now neering of TGEV-PRCV chimaeras. Thus,
enzootically present. This enzootic status through this TGEV deletion mutant, mo-
will, based on the presently available in- lecular virologists can be provided with
formation, not change in the future. TGE clearer insights into the function of differ-
has clearly become a disease of minor im- ent gene products of TGEV-PRCV, infor-
portance in Europe since the establish- mation which may be useful not only for
ment of this enzootic PRCV situation. molecular pathogenesis of the respective
Widespread infections, possibly also re- infections, but also for that of other co-
peated ones, with PRCV have created an ronaviruses of different animal species.
immune status in the swine population so From a more general point of view, the
that infections with the enteropathogenic finding that the apparently enhanced con-
TGEV have acquired a mild or short last- tagiosity exhibited by PRCV relative to the
ing character. The much-feared acute out- parental virus has been gained virtually
breaks of TGE with the high pig mortali- through a loss of genetic material is worth
ties have disappeared since PRCV has reflecting upon.
become enzootic and this has given a tre- Common mucosal immune mechanisms
mendous beneficial effect on the pig pop- exist but are often difficult to study with vi-
ulation. ruses because many viruses either have a

Through the emergence of PRCV, re- tropism for several mucosae or do not
searchers have been provided with a tool have a counterpart-virus for reciprocal
to obtain new insights into molecular path- challenge and evaluation of protection at a
ogenesis and common mucosal immunity. distant mucosal site. PRCV has provided
PRCV replicates highly in the respiratory immunologists with an ideal tool. The pro-
tract and has, contrary to TGEV, no ente- tective effect of immune responses which
rotropism in clinical terms. One would ex- were initiated by a PRCV infection in the

pect that PRCV is no longer able to recog- respiratory tract can be tested in the res-
nize receptors on enterocytes, but recent piratory tract itself through challenge with
studies have shown that this may not be PRCV and can also be tested in the enter-
the case, suggesting that a later step of ic tract by challenge with the closely relat-
the virus cycle may be hampered. Even ed enterotropic TGEV and vice versa. This
more interesting is that PRCV has ac- way, immune processes interacting be-
quired a highly pronounced tropism for tween the respiratory tract and the enteric
the respiratory tract. From the molecular tract can be examined. Moreover, the
studies performed up to now, a much fo- mechanism by which a respiratory tract in-
fection with PRCV leads to IgA production ACKNOWLEDGMENTS
in the mammary gland of sows can be
studied. It has long been thought that lac- The authors express their thanks to L Van
togenic immunity based on IgA secretion Lancker and AM Spite for typing of the manu-
in the sows milk could only be obtained if script.
an infection had occurred in the enteric
tract .
Not all the effects which accompanied REFERENCES
the emergence of PRCV have been benef-
ical. The possible association of a PRCV Alonso JMM, Balbin M, Garwes DJ, Enjuanes L,
infection with the appearance of respirato- Gascon S, Parra F (1992) Antigenic structure
ry disease is still under discussion. There of transmissible gastroenteritis virus nucleo-
is no doubt that a new infectious, potential- protein. Virology 188, 168-174
ly pathogenic, respiratory virus has been Bernard S, Bottreau E, Aynaud JM, Have P,
added to the long list of now existing res- Szymansky L (1989) Natural infection with
the porcine respiratory coronavirus induces
piratory infectious agents in swine. PRCV
infections are widespread and pigs thus protective lactogenic immunity against trans-
missible gastroenteritis. Vet Microbial 21 , 1-8
have a high chance to become infected
Bohl EH, Gupta RKP, McCloskey LW, Saif L
concomitantly with other, also widespread, (1972a) Immunology of transmissible gas-
respiratory pathogens such as influenzavi- troenteritis. J Am Vet Med Assoc 160, 543-
ruses, porcine respiratory and reproduc- 549
tive disease virus, Aujeszky’s disease vi- Bohl EH, Gupta RPK, Olquin MVF, Saif LJ
rus, and some bacteria. It can be (1972b) Antibody responses in serum, colos-
reasonably accepted that PRCV in these trum and milk of swine after infection or vac-
combined infections adds to the appear- cination with transmissible gastroenteritis vi-
ance of disease particularly in pigs &dquo;at risk&dquo; rus. Infect Immun 6, 289-301

ie when maternal immunity wanes or when Britton P, Page KW (1990) Sequence of the S
transfer occurs from the breeding to the gene from a virulent British field isolate of
transmissible gastroenteritis virus. Virus Res
fattening divisions of farms. The real im-
18, 71-80
portance of PRCV pathogen for the res-
as

piratory tract is still under study both in Britton P, Otin CL, Alonso JMM, Parra F (1989)
Sequence of the coding regions from the 3.0-
field and experimental circumstances but
kb and 3.9-kb mRNA subgenomic species
the economic impact is certainly much low- from a virulent isolate of transmissible gas-
er than that of the earlier typical TGE out- troenteritis virus. Arch Virol 10, 165-178
breaks. Britton P, Mawditt ML, Page KW (1991) The clon-
In conclusion, the different features ingand sequencing of the virion protein genes
which have accompanied the emergence from a British isolate of porcine respiratory co-
of PRCV have been explained in this re- ronavirus: comparison with transmissible gas-
troenteritis virus genes. Virus Res 21, 181-198
view article. It is shown that, contrary to
other newly emerged viruses in the recent Britton P, Kottier S, Chen CM, Pocock DH, Sal-
mon H, Aynaud JM (1992) The use of PCR
history of porcine viral infections, the genome mapping for the characterisation of
emergence of PRCV has undoubtedly TGEV strains. In: Coronaviruses: Molecular
brought more favorable than unfavorable Biology and Virus-host Interactions (Laude H,
effects to the pig population itself, to the and Vautherot JF, eds) Plenum Press, New
pig producer and to researchers studying York (in press)
different aspects of viral diseases in Brown I, Cartwright S (1986) New porcine coron-
swine. avirus? Vet Rec 119, 282-283
Callebaut P, Correa I, Pensaert M, Jimenez G, piratorycoronavirus antigenically closely re-
Enjuanes L (1988) Antigenic differentiation lated to transmissible gastroenteritis virus.
between transmissible gastroenteritis virus of Vet Microbiol 23, 237-243
swine and a related porcine respiratory co- Cox E, Pensaert MB, Callebaut P (1993) Intesti-
ronavirus. J Gen Virol69, 1725-1730 nal protection against challenge with trans-
Callebaut P, Pensaert M, Hooyberghs J (1989) missible gastroenteritis virus of pigs immune
A competitive inhibition ELISA for the diffe- after infection with the porcine respiratory co-
rentiation of serum antibodies from pigs in- ronavirus. Vaccine 11, 267-272
fected with transmissible gastroenteritis virus Cubero MJ, Bernard S, Leon L, Berthon P, Con-
(TGEV) or with the TGEV-related porcine treras A (1992) Pathogenicity and antigen de-
respiratory coronavirus. Vet Microbiol 20, 9- tection of the Nouzilly strain of transmissible
199 gastroenteritis coronavirus, in 1-week-old pig-
Callebaut P, Cox E, Pensaert M, Van Deun K lets. J Comp Pathol 106, 61-73
(1990) Induction of milk IgA antibodies by De Diego M, Laviada MD, Enjuanes L, Escriba-
porcine respiratory coronavirus infection. In: no JM(1992) Epitope specificity of protective
Coronaviruses and their Diseases (Cava- lactogenic immunity against swine transmis-
nagh D, Brown TDK, eds) Plenum Press, sible gastroenteritis virus. J Virol 66, 6502-
New York, 421-428 6508
Cavanagh D, Brian DA, Enjuanes L, Holmes Delmas B, Laude H (1990) Assembly of corona-
KV, Lai MMC, Laude H, Siddell SG, Spaan virus spike protein into trimers and its role in
W, Taguchi F Talbot PJ (1990) Recommen- epitope expression. J Virol64, 5367-5375
dations of the coronavirus study group for the Delmas B, Gelfi J, Laude H (1986) Antigenic
nomenclature of the structural proteins, structure of transmissible gastroenteritis vi-
mRNAs, and genes of coronaviruses. Virolo- rus. II. Domains in the peplomer glycoprotein.
gy 176, 306-307 J Gen Virol67, 1405-1418 8
Charley B, Laude H (1988) Induction of interfer- Delmas B, Rasschaert D, Godet M, Gelfi J,
on alpha by transmissible gastroenteritis co- Laude H (1990) Four major antigenic sites of
ronavirus: role of transmembrane glycopro- the coronavirus transmissible gastroenteritis
tein E1. J Virol62, 8-111 virus are located on the amino-terminal half
Correa I, Jimenez G, Sun6 C, Bullido MJ, Enju- of spike glycoprotein S. J Gen Virol 71, 1313-
anes L (1988) Antigenic structure of the E 2 1323
glycoprotein from transmissible gastroenteri- Delmas B, Gelfi J, L’Haridon R, Vogel LH,
tis coronavirus. Virus Res 10, 77-94 Sjbstr6m H, Noren 0, Laude H (1992a) Ami-
Correa I, Gebauer F, Bullido MJ, Sune C, Baay nopeptidase N is a major receptor for the en-
MFD, Zwaagstra KA, Posthumus WPA, Len- teropathogenic coronavirus TGEV. Nature
stra JA, Enjuanes L (1990) Localization of 357, 417-420
antigenic sites of the E2 glycoprotein of Delmas B, Gelfi J, Sjbstr6m H, Noren 0, Laude
transmissible gastroenteritis coronavirus. H (1992b). Further characterization of amino-
J Gen Virol 71, 271-279 peptidase N as a receptor for coronaviruses.
Cox E, Hooyberghs J, Pensaert MB (1990a) In: Coronaviruses: Molecular Biology and Vi-
Sites of replication of a porcine respiratory rus-Host Interactions (Laude H, Vautherot
coronavirus related to transmissible gas- JF, eds) Plenum Press, New York (in press)
troenteritis virus. Res Vet Sci 48, 165-169 Doyle LP, Hutchings LM (1946) A transmissible
Cox E, Pensaert M, Hooyberghs J, Van Deun K gastroenteritis in pigs. J Am Vet Med Assoc
(1990b) Sites of replication of a porcine res- 108, 257-259
piratory coronavirus in 5-week-old pigs with Duret C, Brun A, Guilmoto H, Dauvergne M
or without maternal antibodies. In: Coronavi- (1988) Isolement, identification et pouvoir pa-
ruses and their Diseases (Cavanagh D and thogbne chez le porc d’un coronavirus appa-
Brown TDK, eds) Plenum Press, New York, rent6 au virus de la gastro-ent6rite transmis-
429-433 sible. Recl Med Vet 164, 221-226
Cox E, Pensaert M, Callebaut P, Van Deun K Fazakerley JK, Parker SE, Bloom F, Buchmeier
(1990c) Intestinal replication of a porcine res- MJ (1992) The V5A13.1 envelope glycopro-
tein deletion mutant of mouse hepatitis virus brane-bound arylamidases from small intes-
type-4 is neuroattenuated by its reduced rate tine, lung, kidney, liver, placenta and renal
of spread in the central nervous system. Vi- cell carcinoma. Clin Chim Acta 101, 139-143
rology 187, 178-188 Jackwood DJ, Bae I, Jackwood RJ, Saif LJ
Furuuchi S, Shimizu Y, Kumagai T (1979) Multi- (1992) Transmissible gastroenteritis virus
plication of low and high cell culture pas- and porcine respiratory coronavirus: molecu-
saged strains of transmissible gastroenteritis lar characterization of the S gene using
virus in organs of newborn piglets. Vet Micro- DNA probes and nucleotide sequence anal-
C
biol3, 169-178 ysis. In: Coronaviruses: Molecular Biology
Garwes DJ, Stewart F, Elleman CJ (1987) Iden- and Virus-Host Interactions (Laude H and
tification of epitopes of immunological impor- Vautherot JF, eds) Plenum Press, New York,
tance on the peplomer of porcine transmissi- (in press)
ble gastroenteritis virus. In: Coronaviruses Jimenez G, Correa I, Melgosa MP, Bullido MJ,
(Lai M, Stohlman SA, eds) Plenum Press, Enjuanes L (1986) Critical epitopes in trans-
New York missible gastroenteritis virus neutralization.
Garwes DJ, Stewart F, Cartwright SF, BrownI J Viro160, 131-139
(1988) Differentiation of porcine coronavirus Jestin A, Leforban Y, Vannier P (1987a) Les co-
from transmissible gastroenteritis virus. Vet ronavirus du porc. Recl Méd Vet 163, 583-
Rec 122, 86-87 588
Gebauer F, Posthumus WPA, Correa I, Sune C, Jestin A, Leforban Y, Vannier P, Madec F, Gour-
Smerdou C, Sanchez CM, Lenstra JA, Me- reau J (1987b) Un nouveau coronavirus por-
loen R, Enjuanes L (1991) Residues in- cin. !tudes s6ro-6pid6miologiques r6trospec-
volved in the antigenic sites of transmissible tives dans les 6levages de Bretagne. Recl
gastroenteritis coronavirus S glycoprotein. Med Vet 163, 5
571
-
7
6
Virology 183, 225-238 Kemeny LJ (1978) Isolation of transmissible
Godet M (1992) Expression en systbme baculo- gastroenteritis virus from pharyngeal swabs
virus et caracterisation de proteines structu- obtained from sows at slaughter. Am J Vet
rales et non structurales du coronavirus de la Res 39, 703-705
gastro-ent6rite transmissible du porc. Docto- Woods RD (1977) Quantitative
ral thesis, Paris Kemeny LJ,
transmissible gastroenteritis virus shedding
Godet M, L’Haridon R, Vautherot JF, Laude H
patterns in lactating sows. Am J Vet Res 38,
(1992) TGEV coronavirus ORF4 encodes a 307-3100
membrane protein that is incorporated into
virions. Virology 188, 666-675 Kemeny LJ, Wiltsey VL, Riley JL (1975) Upper
Harada K, Furuuchi
respiratory infection of lactating sows with
S, Kumagai T, Sasahara transmissible gastroenteritis virus following
(1969) Pathogenicity, immunogenicity and dis- contact exposure to infected piglets. Cornell
tribution of transmissible gastroenteritis virus
Vet65,352-362
in pigs. Nat Inst Anim Hlth Quart 9, 183-192
La Bonnardibre C, Laude H (1983) Interferon in-
Have P (1990) Infection with a new porcine res-
duction in rotavirus and coronavirus infec-
piratory coronavirus in Denmark. Serologic tions: a review of recent results. Ann Rech
differentiation from transmissible gastroenter-
itis virus using monoclonal antibodies. In: Co- et14,507-5111
V
ronaviruses and their Diseases (Cavanagh D Lai MMC (1990) Coronavirus: organization, rep-
and Brown TDK, eds) Plenum Press, New lication and expression of genome. Annu Rev
York, 435-439 Microbiol44, 303-333
Henningsen AD, Mousing J, Aalund 0 (1988) Lanza I, Rubio P, Munoz M, Callebaut P,
Porcine coronavirus (PCV) in Denmark: an C6rmenes P (1990) Epidemiology of TGEV
epidemiological study based on question- and PRCV infections in Spain. Proc llth
naire data from screening districts. Dansk Congress Int Pig Vet Soc. Lausanne, Swit-
Vet Tidsskrift7l , 1168-1177 zerland, 199
Ito T, Hiwada K, and Kokubu T Immuno-
(1980) Lanza I, Brown IH, Paton DJ (1992) Pathogenic-
logical characterization of human mem- ity of concurrent infection of pigs with porcine
respiratory coronavirus and swine influenza O’Toole D, Brown I, Bridges A, Cartwright SF
virus. Res Vet Sci 53, 309-314
4 (1989) Pathogenicity of experimental infec-
Laude H, Charley B, Gelfi J (1984) Replication tion with &dquo;pneumotropic&dquo; porcine coronavirus.
of transmissible gastroenteritis coronavirus Res Vet Sci 47, 23-29
(TGEV) in swine alveolar macrophages. Page KW, Britton P, Boursnell MEG (1990) Se-
J Gen Virol 65, 327-332 quence analysis of the leader RNA of two
Laude H, Chapsal JM, Gelfi J, Labiau, S, Gros- porcine coronaviruses: transmissible gas-
troenteritis virus and porcine respiratory co-
claude J (1986) Antigenic structure of trans-
ronavirus. Virus Genes 4, 289-301
missible gastroenteritis virus. I. Properties of
monoclonal antibodies directed against virion Page KW, Mawditt ML, Britton P (1991) Se-
proteins. J Gen Virol67, 119-130 quence comparison of the 5’ end of mRNA 3
from transmissible gastroenteritis virus and
Laude H, Rasschaert D, Huet JC (1987) Se-
porcine respiratory coronavirus. J Gen Virol
quence and N-terminal processing of the 72, 579-587
membrane protein E l of the coronavirus
transmissible gastroenteritis virus. J Gen Vir- Paton DJ, Brown IH (1990) Sows infected in
168, 1687-1693
0 pregnancy with porcine respiratory coronavi-
rus show no evidence of protecting their
Laude H, Gelfi J, Rasschaert D, Delmas B
suckling piglets against transmissible gas-
(1988) Caractérisation antigénique du coron- troenteritis. Vet Res Commun 14, 329-337
avirus respiratoire porcin 6 I’aide d’anticorps
monoclonaux dirig6s contre le virus de la Paton DJ, Brown IH, Vaz EK (1991) An ELISA
for the detection of serum antibodies to both
gastro-ent6rite transmissible. J Rech Porcine transmissible gastroenteritis virus and por-
Fr 20, 89-94
cine respiratory coronavirus. Br Vet J 147,
Laude H, Rasschaert D, Delmas B, Godet, M 370
Gelfi J, Charley B (1990) Molecular biology of
Pensaert M (1989) Transmissible gastroenteritis
transmissible gastroenteritis virus. Vet Micro-
virus (respiratory variant). In: Virus Infections
biol23, 147-154 of Porcines (Pensaert M, ed) Elsevier Sci-
Laude H, Gelfi J, Lavenant L, Charley B (1992) ence Publishers, Amsterdam, 154-158
Single amino acid changes in the viral glyco- Pensaert M, Callebaut P, Vergote J (1986) Isola-
protein M affect induction of alpha interferon tion of a porcine respiratory, non-enteric co-
by the coronavirus transmissible gastroenteri- ronavirus related to transmissible gastroen-
tis virus. J Virol66, 743-749 teritis. Vet Q 8, 257-261
Laval A, Le Foll P, Gestin G, Reynaud G (1991) Pensaert M, Callebaut P, Vergote J (1987) Un
Grippes et coronavirus respiratoire porcin : mutant naturel du virus GET chez le porc
Étude sbrologique dans dix 6levages bre- avec tropisme seulement pour I’appareil res-
tons. Recl Med Vet 167, 521-528
piratoire. In: The World Veterinary Associa-
McClurkin AW, Norman JO (1966) Studies on tion (ed). Abstracts from the XXlllth World
transmissible gastroenteritis of swine. II. Se- Veterinary Congress, Can Vet Med Assoc,
lected characteristics of a cytopathogenic vi- Montreal, 294
rus common to five isolates of transmissible Pensaert M, Cox E, Van Deun K, Callebaut P
gastroenteritis. Can J Comp Med Vet Sci 30, (1992) A seroepizootiological study of the
190-198 porcine respiratory coronavirus in the Belgian
Madec F, Kaiser C, Jestin A, Gourreau JM, Van- swine population. Vet Quart (in press)
nier Ph, Kobisch M, Paboeuf F, Aymard M Pospischil A, Cox E, Pensaert M (1990) Locali-
(1987) Les syndromes grippaux en porcherie zation of porcine respiratory coronavirus in
d’engraissement, enqu6te flash r6alis6e en the small intestine of experimentally infected
Bretagne. Point Vét 19, 654-659 piglets. In: Proc llth Congress Int Pig Vet
Onno M, Jestin A, Cariolet R, Vannier P (1989) Soc. Lausanne, Switzerland, 219
9
Rapid diagnosis of TGEV-like coronavirus in Rasschaert D, Laude H (1987) The predicted
fattened pigs by indirect immunofluorescence primary structure of the peplomer protein E2
labelling in nasal cells. J Vet Med B 36, 629- of the porcine coronavirus transmissible gas-
634 troenteritis virus. J Gen Virol68, 1883-1890
Rasschaert D, Gelfi J, Laude H (1987) Enteric Vannier P (1990) Disorders induced by the ex-
coronavirus TGEV: partial sequence of the perimental infection of pigs with the porcine
genomic RNA its organization and expres- respiratory coronavirus. J Vet Med B 37, 117-
sion. Biochimie 69, 591-600 180
Rasschaert D, Duarte M, Laude H (1990) Por- van Nieuwstadt AP, Pol JMA (1989) Isolation of
cine respiratory coronavirus differs from a TGE virus-related respiratory coronavirus
transmissible gastroenteritis virus by a few causing fatal pneumonia in pigs. Vet Rec
genomic deletions. J Gen Virol 71, 2599- 124, 43-44
2607 van Nieuwstadt AP, Boonstra J (1992) Compari-
Sanchez CM, Jimenez G, Laviada MD, Correa son of the antibody response to transmissible
I, Surie C, Bullido MJ, Gebauer F, Smerdou gastroenteritis virus and porcine respiratory
C, Callebaut P, Escribano JM, Enjuanes L coronavirus using monoclonal antibodies to
(1990) Antigenic homology among coronavi- antigenic sites A and X of the S glycoprotein.
ruses related to transmissible gastroenteritis Am J Vet Res 53, 184-190
virus. Virology 174, 410-417
7 van Nieuwstadt AP, Zetstra T, Boonstra J
Sanchez CM, Gebauer F, Suh6 C, Mendez A, (1989) Infection with porcine respiratory co-
Dopazo J, Enjuanes L (1992) Genetic evolu- ronavirus does not fully protect pigs against
tion and tropism of transmissible gastroenter- intestinal transmissible gastroenteritis virus.
itis coronaviruses. Virology 190, 92-105 Vet Rec 125, 58-60

Spaan W, Cavanagh D, Horzinek MC (1988) Voets MTh, Pensaert MB, Rondhuis PR (1980)
Coronaviruses: structure and genome ex- Vaccination of pregnant sows against trans-
pression. J Gen Virol69, 2939-2952 missible gastroenteritis virus with two attenu-
ated virus strains and different inoculation
Sprino PJ, Ristic M (1982) Intestinal, pulmo- routes. Vet Q 2, 211-2199
nary, and serum antibody responses of feed-
er pigs exposed to transmissible gastroenter- Wesley RD, Cheung AK, Michael DD, Woods
itis virus by the oral and the oral-intranasal RD (1989) Nucleotide sequence of coronavi-
routes of inoculation. Am J Vet Res 43, 255- rus TGEV genomic RNA: evidence for 2
261 mRNA species between the peplomer and
matrix protein genes. Virus Res 13, 87-100
Ulbrich F, Hendel C, Zureck J, Schneider G,
Ehrlich K, Richter M, Aschmann L, MOller C Wesley RD, Woods RD, Hill KT, Biwer JD
(1991) Auftreten klinischer Erscheinungen (1990a) Evidence for a porcine respiratory
bei laktierenden Sauen im Zusammenhang coronavirus, antigenically similar to transmis-
mit der Infektion durch Porcines respiratoris- sible gastroenteritis virus in the United
ches Coronavirus. Monatsh Veterinaer Med States. J Vet Diagn Invest 2, 312-317 7
46, 128-133 Wesley RD, Woods RD, Cheung AK (1990b)
Underdahl NR, Mebus CA, Stair EL, Rhodes Genetic basis for the pathogenesis of trans-
MB, McGill LD, Twiehaus MJ (1974) Isolation missible gastroenteritis virus. J Virol 64,
of transmissible gastroenteritis virus from 4761-4766
lungs of market-weight swine. Am J Vet Res Wesley RD, Woods RD, Cheung AK (1991) Ge-
35, 1209-1216 netic analysis of porcine respiratory coronavi-
Underdahl NR, Mebus CA, Torres-Medina A rus, an attenuated variant of transmissible
(1975) Recovery of transmissible gastroen- gastroenteritis virus. J Virol65, 3369-3373
teritis virus from chronically infected experi- Yeager CL, Ashmun RA, Williams RK, Cardelli-i-
mental pigs. Am J Vet Res 36, 1473-1476 chio CB, Shapiro LH, Look AT, Holmes KV
Van Deun K, Cox E, Callebaut P, Pensaert M (1992) Human aminopeptidase N is a recep-
tor for human coronavirus 229E. Nature 357,
(1990) Milk of sows infected with the porcine
420-422
respiratory coronavirus: induction of IgA anti-
bodies against transmissible gastroenteritis Yu E, Laviada MD, Moreno L, Castro JM, Escri-
virus and protective capacity against intesti- bano JM, Simarro (1989) Prevalencia de anti-
nal infection in piglets. In: Proc llth cuerpos frente a virus influenza y coronavirus
Congress Int Pig Vet Soc. Lausanne, Swit- respiratorio en cerdos de cebo en Espana. J
zerland, 263 Vet Med B 36, 551-556

You might also like