<<

Maedi-visna infection in : a review Michel Pépin, Christian Vitu, Pierre Russo, Jean-François Mornex, Ernst Peterhans

To cite this version:

Michel Pépin, Christian Vitu, Pierre Russo, Jean-François Mornex, Ernst Peterhans. Maedi-visna virus infection in sheep: a review. Veterinary Research, BioMed Central, 1998, 29 (3-4), pp.341-367. ￿hal-00902532￿

HAL Id: hal-00902532 https://hal.archives-ouvertes.fr/hal-00902532 Submitted on 1 Jan 1998

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

Maedi-visna virus infection in sheep: a review

Michel Pépina Christian Vitua Pierre Russoa Jean-François Mornexb Ernst Peterhans

a Unité pathologie des petits ruminants, Cneva Sophia-Antipolis, BP 111, 06902 Sophia-Antipolis cedex, France bInra- Lyon, Marcy-l’Étoile, France c Institute of Veterinary Virology, Bern, Switzerland

(Received 29 December 1997; accepted 10 March 1998)

Abstract - The maedi-visna virus (MVV) is classified as a lentivirus of the retroviridae family. The of MVV includes three : gag, which encodes for group-specific ; pol, which encodes for , , RNAse H, protease and dUTPase and env, the encoding for the surface responsible for receptor binding and entry of the virus into its host cell. In addition, analogous to other lentiviruses, the genome contains genes for regulatory proteins, i.e. vif, and . The coding regions of the genome are flanked by long ter- minal repeats (LTR) which play a crucial role in the replication of the viral genome and pro- vide binding sites for cellular transcription factors. The organs targeted by MVV are, in descend- ing order of importance, the lungs, mammary glands, joints and the brain. In these organs, the virus replicates in mature macrophages and induces slowly progressing inflammatory lesions con- taining B and T lymphocytes. The clinical signs of MVV infection, i.e. dyspnea, loss of weight, mastitis and arthritis, are related to the location of these lesions. Infection with MVV induces the formation of which can be detected by agar gel immunodiffusion, ELISA and the serum neutralization assay. As neither antiviral treatment nor vaccination is available, diagnos- tic tests are the backbone of most of the schemes implemented to prevent the spread of MVV. How- ever, since current serological assays are still lacking in sensitivity and specificity, molecular bio- logical methods are being developed permitting the detection of virus in peripheral blood, milk and tissue samples. Future research will have to focus on both the development of new diag- nostic tests and a better understanding of the pathogenesis of MV V infection. 0 Inra/Elsevier, Paris lentivirus / maedi-visna / sheep / slow infection / review

Résumé - L’infection par le virus Maedi-visna chez le mouton : une revue. Le virus du maedi-visna appartient au genre lentivirus de la famille des rétrovirus. Le génome de ce virus com-

* Correspondence and reprints Tel.: (33) 4 92 94 37 00; fax: (33) 4 92 94 37 Ol; e-mail: [email protected] prend trois gènes de structure codant pour l’enveloppe virale (env), la capside (gag) et les enzymes telles que la transcriptase réverse, l’intégrase et la dUTPase (po/), et plusieurs gènes accessoires : vif, rev et tat. Ces gènes accessoires interviennent dans la régulation de la réplication et de l’expression du virus, la modulation de son pouvoir pathogène et de son tropisme. Les organes cibles du virus maedi-visna sont par ordre d’importance le poumon, la mamelle, les articula- tions et le cerveau. Dans ces organes, le virus infecte les macrophages matures et entraîne des lésions de type inflammatoire contenant des lymphocytes B et T, évoluant lentement, il l’ori- gine des signes cliniques de la maladie : essoufflement, amaigrissement, mammite, arthrites, etc. L’infection par le virus maedi-visna conduit à une réponse humorale à l’origine des tests diagnostiques actuellement disponibles : immunodiffusion en gélose, Elisa, etc. Ces tests séro- logiques sont à la base de la plupart des plans de prophylaxie sanitaire mis en place dans de nombreux pays, en l’absence de traitement et de vaccination. Cependant de nombreuses difficultés liées il la sensibilité et à la spécificité des tests sérologiques ont conduit à s’orienter vers la mise en évidence du virus dans le sang. le lait ou les organes par des techniques d’amplification génique. Dans un futur proche, les besoins en matière de recherche sont évidents tant pour le déve- loppement de nouveaux outils diagnostiques fiables que pour une meilleure connaissance de la pathogénie du maedi-visna afin de trouver des moyens de lutte mieux adaptés. © Inra/Elsevier, Pariss lentivirus / maedi-visna / mouton / infection lente / revue 1. INTRODUCTION ruminant lentiviruses include primary interstitial pneumonia, encephalitis, lym- The lentivirus of the retroviridae phadenopathy, arthritis, mastitis and family comprises pathogens of humans, chronic weight loss (Russo et al., 1991; monkeys, horses, , sheep, goats and Phelps and Smith, 1993). Lentiviral dis- cats. The infections caused by maedi-visna eases are the cause of significant economic virus (MVV) in sheep and by caprine losses incurred by the sheep and goat arthritis-encephalitis virus (CAEV) in industries and also increasingly threaten goats share a number of features with the exports of live animals. infection caused by the human immuno- deficiency virus (HIV), such as an incu- Infections with MVV and CAEV per- bation period of several months or even sist for life in sheep and goats, respec- years and a slow development of disease tively, despite a humoral and cellular symptoms (table I). The major manifes- immune response. The infection is char- tations of the diseases induced by small acterized by progressive inflammatory et visna state of lesions in various organs (Cador6 al.,., called (’fading away - pro- 1996; Harkiss et al., 1991; Mornex et al., gressive apathy’ in Icelandic), is a 1994; Anderson et al., 1994; Cador6 et al., demyelinating leukoencephalomyelitis 1993). The major, if not the sole, host cells (Palsson, 1976; Watt et al., 1994). MVV of the virus are cells of the monocyte/ can also infect other organs or tissues, par- macrophage cell lineage (Gendelman et ticularly joints in which it causes arthri- al., 1986). In contrast to human (HIV) and tis (Watt et al., 1994) and the mammary simian (SIV) immunodeficiency , glands where it causes mastitis (Pekelder, the small-ruminant lentiviruses do not 1993; Watt et al., 1994). The properties infect CD4+ T lymphocytes. Therefore, of sheep are summarized in the diseases they cause provide a valuable table IL model for both the effects of studying In Iceland, MVV is likely to have lentivirus infection on macrophage biology appeared subsequent to the importation of and the role infected played by infected asymptomatic Karakul rams from macrophages in the absence of immuno- Germany in 1933, which resulted in deficiency. widespread dissemination of the infection This review aims to summarize the cur- to most flocks. Following its discovery in rent knowledge of the biology of MVV Iceland, MVV infections were detected in and its interaction with its host, the sheep. various countries, although with differing We also provide a short overview of the prevalences (Madewell et al., 1987; Camp- history of the discovery of MVV. The bell et al., 1994; Schaller et al., 1995; structure and organization of the MVV Pritchard et al., 1995; de la Concha- are Aus- genome and of the encoded polypeptides Bermejillo, 1997). Exceptions tralia and New where lentiviral are described, with particular emphasis on Zealand, infections have been observed in but auxiliary genes. The clinical consequences goats of infection, the epidemiology and diag- not in sheep (Greenwood et al., 1995). In nostic tests are considered and the mech- France, Russo et al. (1980) isolated the anisms of pathogenesis discussed. first French MVV strain in 1980. Complete sequences of sev- eral MVV strains have been published 2. HISTORY since 1985, i.e. those of strains K 15144 (Sonigo et al., 1985) from Iceland, SA- OMVV (Qu6rat et al., 1990) from South MVV was discovered in sheep by Sig- Africa, EV 1 from the UK et al., urdsson et al. (1952, 1957) in Iceland in (Sargan as well as clones KV 1772-kv72/67 the early 1950s, although the disease 1991) for neurovirulence (Andresson symptoms had been described prior to this [selected discovery in South Africa, the USA and et al., 1993)] and LV 1-1 KS 1 (Staskus et Iceland. The concept of ’slow viruses’ al., 1991 ).). resulting from this discovery prompted the name of the lentivirus [lentus (]at.) = 3. THE VIRUS slow] genus of which MVV is a member. Two distinct pathological situations, cor- responding to the main clinical manifes- tations of MVV infection, featured in those 3.1. Viral structure early descriptions. The first, called maedi (’dyspnea’ in Icelandic), is a progressive MVV virions are spherical and have a et structure. The size pneumonia (Palsson, 1976; Mornex al.,., unique three-layered 1994; Cador6 et al., 1996) and the second, of the virions is about 100 nm. The central part of the virus is the genome-nucleo- various auxiliary genes. Analogous to protein complex, associated with the other retroviruses, the MVV proviral DNA reverse transcriptase. This structure is is flanked by long terminal repeats (LTR) enclosed within an icosahedral sur- that provide the cis signals required for rounded by an envelope derived from the transcription, integration and polyadeny- plasma membrane of the host cell.l . lation of viral RNA (figure 1). The num- bers and role of the auxiliary genes vary depending on the lentivirus involved and, 3.2. Organization of the viral genome to a lesser degree, between the different strains of MVV. MV virus has a genetic organization that is typical of lentiviruses: its genome is 3.2.1. Structural genes and a dimer of RNA of positive strand polar- their products (table III) ity, 9.2 kb in size, which is reverse tran- scribed into DNA, some of which proviral 3.2.1.1. The gag gene will be integrated into the chromosomal DNA. It comprises three structural genes, The gag gene encodes for three glyco- i.e. gag (group-specific ), pol (poly- proteins from the precursor Pr55!a!: the merase) and env (envelope), as well as capsid (p25), the nucleocapsid (p14), and the matrix (MA) protein (p 17) which 3.2.1.2. The env gene ensures the link between the capsid and the envelope. The MA protein is respon- The env gene encodes the glycoprotein sible for the association of the gag pre- of the virus. As for other retroviruses, the cursor with the cell plasma membrane; glycoprotein is synthesized as a precursor within the virus particle, the MA protein is (gp160) and is cleaved by a host cell pro- localized between the viral membrane and tease into two subunits: the surface gly- the capsid protein of the virus. The cap- coprotein (SU; gp135) and the transmem- sid protein, the most abundant protein of brane glycoprotein (TM; gp44). The TM the virion, forms the hydrophobic core of part of the surface glyco- the virion and elicits a strong protein is anchored in the lipid bilayer. response during infection, which is valu- The SU glycoprotein is non-covalently able for diagnostic tests. Within the capsid linked to TM. The envelope protein the nucleocapsid protein coats the of lentiviruses have many important bio- viral RNA genome (Joag et al., 1996). logical functions and contain the epitopes responsible for both the induction of neu- 3.2.1.3.2.2. Integrase antibodies and the interaction of tralizing In each virion, numerous molecules of the virus with the of the host cell. receptor reverse transcriptase and integrase are To no surface for MVV date, receptors associated with the viral RNA (Marin et have been defined conclusively although a]., 1994). Following reverse transcrip- cellular proteins able to bind to the virus tion, provirus migrates towards the have been described (Dalziel et al., 1991; nucleus, and the double-stranded DNA Clements and this contrasts Payne, 1994); genome is integrated in the host cell DNA to HIV where the and the receptor (CD4) by a mechanism mediated by integrase for co-receptors (receptors chemokines) (Stormann et al., 1995). Integration have been determined et al., (Willett involves repeated inverted sequences pre- 1997). sent in the LTR (Marin et al., 1994). 3.2.1.3.2.3. Protease 3.2.1.3. The pol gene The viral protease cleaves the gag and and resem- 3.2.1.3.1. The reverse transcriptase gag-pol polyprotein precursors bles cellular aspartic acid proteases in its The key enzyme of retroviruses is the three-dimensional structure (Joag et a]., reverse transcriptase, a RNA-dependent 1996). DNA polymerase, encoded by the pol gene which permits the transcription of the viral 3.2.2. Auxiliary genes RNA into DNA; this protein is an het- erodimer which displays RNAse H activ- The MVV has three major auxiliary ity. genes: tat, vif’(; pre- viously called Q gene), and rev (regula- 3.2.1.3.2. Other enzymes tor of virion protein expression) (Clements and Zink, 1996) (figure 1).). 3.2.1.3.2.1. dUTPase 3.2.2.1. Rev In lentivirus , the gene encod- The of the rev is a ing the dUTPase enzyme is located in the product gene pro- pol reading frame between the RNAse H tein of 19 kDa (167 amino acids) derived and integrase coding regions. from an mRNA of 1.4 kb. The MVV rev consists of four exons: a leader lentiviruses lack dUTPase. This enzyme gene seg- activity has been identified only in FIV ment (exon 1), an untranslated portion (feline immunodeficiency virus), EIAV (exon 2), and two translated exons, 3 and ( virus), CAEV 4. As rev is an early gene of lentiviruses et it a and MVV (Elder et al., 1992); dUTPase (Mazarin al., 1990), plays major role in mRNA from appears to decrease the frequency of G- transporting unspliced to-A mutations. In vitro, dUTPase defi- the nucleus to the cytoplasm. The rev pro- tein contains nuclear which cient CAE viruses replicate more slowly in export signals the to the macrophages (Turelli et al., 1996). In vivo permit protein pass through nuclear membrane and to exert its they are slightly attenuated (Turelli et al., regu- role via a element 1997), albeit to a lesser degree than the latory responsive (RRE for rev located in the dUTPase-deficient EIAV (Steagall et al., responsive element) env The RRE is of 1995; Lichtenstein et al., 1997). In con- gene (figure 1). capable RNA via an RNA site. trast, dUTPase-deficient MVV appeared to binding binding be as pathogenic in vivo as the wild type The essential role of rev is illustrated virus (Petursson et al., 1998). by site-directed mutagenesis in the 4th exon (Toohey and Haase, 1994): rev-muta- be mediated by stimulation of cellular genized virus was shown to be non-infec- genes, such as cytokine genes (Philippon et tious (table lV). al., 1994). A recent paper examining the role of tat by studying tat-deleted CAE 3.2.2.2. Tat viruses has shown that this gene is not essential for virus replication (Harmache et The tat gene ( 1.7 kb mRNA) encodes al., 1995b). It may, however, contribute to for a of 10 kDa which was first protein a successful interaction between the virus described for its role in stimulating gene and its host or directed the viral by recruiting modulating expression by promoter cellular factors involved in the initiation located in the 5’ LTR. The Tat protein of the maturation pro- mediates the accumulation of viral mRNA transcription during cess of monocytes to which via the AP-1 and AP- macrophages, (activator protein-1) leads to increased viral in 4 sites in the U3 of the gene expression binding region vivo (Carruth et al., 1994). Moreover, the LTRs (Gdovin and Clements, 1992; presence or absence of AP-1 or AP-1-like Saltarelli et al., 1993) or via cellular factors sequences may at least partially explain such as c-Fos and c-Jun et (Neuveut al., the differences in tropism between vari- 1993). A leucine-rich domain in present ous MVV isolates such as EV 1 (a British Tat is to be for likely responsible target- isolate) and SA-OMVV (a South African ing the Tat at AP-1 sites in the viral protein isolate) versus K I S 14 (an Icelandic iso- LTR (Carruth et al., 1996). it has Recently, late) (Andresdottir et al., 1994; Sutton et al., been that the Tat of MVV recognized 1997). belongs to a group of Tat proteins charac- terized by a weak transactivation poten- 3.2.2.3. Vif tial, in contrast to the Tat proteins of HIV- 1 and -2, SIV or BIV, which strongly The vif gene encodes for a 29-kDa pro- transactivate their LTRs by binding to a tein (230 aa) which, in naturally infected TAR (Tat-activated region) sequence. The animals, induces a weak immune response Tat protein of MVV itself may contribute that can be detected in western blots to viral pathogenesis by inducing follicular (Audoly et al., 1992). This protein is not lymphoproliferative disorders in various homologous to cysteine protease in its organs (Hayman et al., 1993; Vellutini et entirety but contains a motif which is al., 1994). The action of the tat gene may homologous to cysteine protease and is translated during the late stages of viral may also present a problem in the diag- replication (Audoly et al., 1992; Harmache nosis of lentiviral infections and remains et al., 1995a). The importance of vif for a formidable obstacle to vaccine develop- the replication of MVV is unknown but ment. Antigenically distinct viruses have investigations using CAEV and HIV indi- been isolated from sheep persistently cate that v(fplays a crucial role in the late infected with MVV; these variants arise stages of the viral life cycle, i.e. during by point mutations in the env gene (Braun the morphogenesis of the viral nucleo- et al., 1987). protein core (Hoglund et al., 1994; Har- mache et al., 1996a; Simon et al., 1997). Genetic variation in MVV has been determined by PCR amplification of por- tions of the viral genome (Zanoni et al., 1992; Sargan et al., 1995; Rosati et al., 3.3. Variability 1995) or by analysing PCR products in denaturing gradient gel electrophoresis Genetically, lentiviruses are quite het- (Woodward et al., 1994). These studies erogeneous. This manifests itself in their have allowed the extent of variability in antigenic diversity, differences in viru- different regions of the genome (LTR, lence and growth characteristics in vitro. gag, pol, env) to be compared and the het- This genetic plasticity is believed to con- erogeneity of MVV strains to be deter- tribute to viral persistence in the host ani- mined. Moreover, these techniques per- mal by permitting evasion of the immune mitted the phylogeny of lentiviruses to be response. Moreover, antigenic diversity established (figure 2). Analysis of a 475- nt fragment in the pol gene of ovine 4.2. Vertical lentiviruses from France revealed that the French isolates form a group closely Vertical transmission (including hered- related to the Cork CAEV strain and only itary and congenital infection, and infec- distantly related to a group of ovine tion at parturition) is a key feature of the lentiviruses consisting of the K1514, EV1I epidemiology of maedi visna (Russo et and SA-OMVV strains. This analysis was al., 1991 ). In an endemically infected confirmed by studying the sequence vari- flock, virus-infected cells and free virus ability in a fragment of the env gene (Ler- are passed from ewes to their lambs via oux et al., 1995). Similarly, in North colostrum and milk (Watt et al., 1994; de America, sequence analysis in the env la Concha-Bermejillo, 1997). The perme- gene of ovine field isolates showed them ability of the gut of newborn lambs greatly to be more similar to CAEV than to the favours vertical transmission (Houwers, ovine prototype strains, which suggests 1997). The duration of infection in the that ovine and caprine lentiviruses may ewe and the extent of the contamination have descended from a common caprine of the progeny appear to be correlated ancestral genotype (Chebloune et a]., (Houwers et al., 1989). Naturally, lambing 1996). Another study suggests that CAEV is a time of high lentivirus expression originated from an ancestral ovine which facilitates the spread of infection lentivirus adapted to a caprine host (Valas (Guiguen et al., 1992). Since mastitis is et al., 1997). frequent in affected animals, vertical trans- mission may be facilitated by the recruit- ment of mononuclear infected cells to the mammary glands (Zink and Johnson, 4. VIRUS TRANSMISSION 1994).

4.2.1. In utero transmission

4.1. Horizontal transmission In utero virus transmission is a highly controversial issue. Despite strict lamb- ing controls, some unexplained cases of Free virus or virus-infected cells are seropositive lambs have been observed in horizontally transmitted by inhalation of flocks in which MVV eradication pro- respiratory secretions (Zink and Johnson, grams are carried out (Houwers et al., 1994; Watt et al., 1994). Co-infections 1983). Some authors have reported their with other viruses or bacteria may also failure to detect virus in experimentally contribute to the spread of MVV via pul- infected Texel sheep embryos (Watt et al., monary exudates (Palsson, 1976). Several 1994). Others have reported the isolation studies support the hypothesis that, under of ovine lentivirus from a 100-day-old certain circumstances, viral transmission cesarean-sectioned fetus from a naturally between adult animals may play an impor- infected ewe (de la Concha-Bermejillo, tant role in the spread of MVV (Campbell 1997). In an endemically infected flock, et al., 1994). In addition, horizontal trans- the PCR yielded positive results in periph- mission is closely associated with close eral blood mononuclear cells of about confinement in winter stabling (de la Con- 10 % of lambs tested prior to colostrum cha-Bermejillo, 1997), the duration of the ingestion (Brodie et al., 1994). Additional presence of the virus in the flock (Houw- observations suggest that co-infections, ers et al., 1989), and annual acquisition of such as Border disease, might permit an replacements (Houwers, 1997). in utero infection (Russo, pers. comm.). Embryo transfer might be a safe way of Narayan, 1986). Antiviral antibodies in ensuring virus-free flocks (Watt et al., serum of naturally infected animals are of 1994). However, in utero infection may the IgG ! subtype, with no detectable IgG2 be a rare occurrence and is not supported (Bird et al., 1995). However, this appears by epidemiological evidence (Houwers et unlikely to be related to the controversial al., 1989). role of virus neutralization in the persis- tence of virus in vivo. Although it was demonstrated that antibodies, in principle, 4.3. Venereal transmission were also capable of neutralizing virus in macrophages, the affinity of virus bind- to exceeded that of As the virus is present in all body fluids, ing macrophages its it might, theoretically, also be transmit- binding to antibody. This finding the that ted by mating. However, to date no well- prompted suggestion neutralizing antibodies be to the documented case of venereal transmission might unable prevent has been reported (de la Concha-Berme- virus from spreading between and jillo, 1997). Co-factors, such as infection macrophages (Kennedy-Stoskopf It was also that with Brucella ovis or leucocytospermia of Narayan, 1986). argued neutralization determinants of certain viral unknown origin, may increase the shed- strains not be detected antibod- ding of virus in the semen (de la Concha- might by ies and Bermejillo et al., 1996). that the emergence of antigenic variants might be yet another factor con- tributing to the antibodies’ failure to achieve control 5. PATHOGENESIS immunological (Narayan et al., 1987; Clements et at., 1988).

Maedi visna is characterized by a longg Due to the tropism for mononuclear and, typically, symp- phagocytes, lymphoid tissues are consid- toms take several months or even to years ered to be important targets for the repli- (for a review, see and develop Narayan cation of MVV. Indeed, studies Cork, 1985). Infection for life and elegant persists using lymph node cannulation infected animals are a constant reservoir of techniques have yielded important information with infection which, consequently, permits the regard to virus-host interaction. The pro- virus to persist in its host. liferative responses of efferent lymph cells In contrast to infections with HIV, SIV were shown to be depressed transiently and FIV, immunosuppression is not a fea- after experimental infection (Bird et al., ture of maedi visna (Clements and Zink, 1993, 1995). Moreover, the decrease in 1996). This explains why secondary infec- the numbers of CD4(+) and the increase in tions with opportunistic agents are infre- the numbers of CD8(+) cells caused an quent in affected flocks. Nevertheless, the inversion of the ratios of CD4(+)/CD8(+) immune response to maedi-visna virus T cell populations in bronchoalveolar exhibits certain peculiar features which lavage fluids of experimentally infected may contribute to the persistence of infec- sheep (Maslak and Schmerr, 1993). As tion. Early studies demonstrated that sheep suggested by the presence of circulating infected with maedi-visna virus develop precursors of cytotoxic T cells of the a humoral immune response that is CD8(+) phenotype, however, there also markedly slower than that directed against exists a vigorous cell-mediated immune viruses causing acute infections (Gud- response to infection (Blacklaws et al., nadottir and Kristinsdottir, 1967; Sihvo- 1994, I 995). All in all, these observations nen, 1980; Kennedy-Stoskopf and indicate that certain aspects of the immune response may lack fine tuning, even if they opmental stage of monocyte/macrophage do not suggest that the failure to eliminate host cell is a key feature of virus-cell inter- the virus may per se be due to a general action. In Maedi visna, it was demon- failure to mount an antiviral immune strated a number of years ago that viral response. For instance, as work with other gene expression increases during the lentiviruses suggests, the failure to effi- development of the monocyte to its mature ciently neutralize infection is the result of tissue form, the macrophage (Gendelman as yet undetermined mechanisms (Pan- et al., 1985). The fact that monocytes in cino et al., 1994; Bertoni et al., 1994). Par- the blood may carry the viral genome ticularly in the light of observations made without sustaining its replication was with attenuated HIV vaccines, it seems referred to as the ’Trojan horse mecha- well worth investigating whether certain nism’, which indicates that this type of immunodominant regions of Env may interaction with the host cell may permit actually serve as decoys, thus decreasing the virus to be transported to tissues with- a possible protective antibody response out being detected by the immune system (Garrity et al., 1997). Moreover, work with (Peluso et al., 1985). In this context, the the closely related CAE virus in goats sug- observation by Brodie and coworkers gests that a dominant type 2 (i.e. antibody (Brodie et al., 1995) is of interest: in con- centered) immune response may be asso- trast to entry, viral replication in ciated with disease (Perry et al., 1995).). rnacrophages may be restricted in certainn tissues. In addition, different strains of Other important factors contributing to Maedi-visna virus may, in vivo, differ in viral persistence in infected sheep are the their host cell (Clements and Zink, host-cell tropism and certain aspects of tropism lentivirus-host cell interaction. Maedi- I 996). visna virus, as well as CAE virus, were shown in vivo to have a for tropism Incidentally, the tropism for cells of the mononuclear phagocytes (Narayan et al., monocyte lineage with an attendant lack of 1982; Gendelman et al., 1985; Lairmore et viral replication in monocytes also poses Several studies that cer- al., 1987). suggest the evolu- tain other cell less interesting questions regarding types, although promi- tion of the viral in infected ani- also sustain viral in genome nent, may replication mals. The evolution of the viral infected animals. A detailed study of the genome depends on several parameters, among central nervous system of Icelandic sheep them the error rate of the viral polymerase, produced evidence of viral replication in a evolutionary pressure exerted by func- variety of cell types, inter alia, epithelial tional constraints and by the immune sys- cells and fibroblasts of the chorioid plexus tem, and the rate and extent of viral repli- et al., 1989), and viral tran- (Georgsson cation. Viral variants have been shown to scripts were detected in cells of epithelial in infected animals but there is no the and small intestines of emerge thyroid, kidneys clear evidence that these variants arise as goats infected with CAEV (Zink et a]., a result of immune et al.,., 1990). Very recently, endothelial cells pressure (Lutley 1983; Thormar et al., 1983; et were shown to the of Narayan support replication al.. 1987). Viral RNA and have MVV in vitro, with interesting differences antigen been demonstrated in bone marrow cells, on the of the tissue used depending origin but it has remained unclear whether the to prepare the endothelial cells (Craig et cells as and a]., 1997). staining macrophages pro- ducing virus were in fact monocyte pre- In all lentiviruses, the restriction of cursors or macrophages residing in the virus replication depending on the devel- marrow (Gendelman et al., 1985). A major difference between small rumi- The mechanisms responsible for main- nant lentiviruses and the immunodefi- taining and gradually increasing the ciency-causing HIV, SIV and FIV lies in mononuclear inflammation typical of the nature of the histological lesion. Even maedi visna have remained an enigma. A though the viruses causing immunodefi- lentivirus-specific interferon (IFN) may ciency may induce inflammation initially, be one of the pro-inflammatory factors. It in the later stages there is extensive cell was described as the result of an interac- depletion (Gougeon et al., 1993). By con- tion between infected macrophages and trast, both maedi-visna and CAE viruses lymphocytes (Narayan et al., 1985). The interferon was shown cause progressive inflammation. Inflam- lentivirus-specific to restrict viral slow down mation is observed in different organs, replication, and have chemo- most prominently, in the lungs and mam- macrophage maturation, tactic for and mary glands and, less frequently, in the properties lymphocytes synovial membranes of the joints and in mononuclear phagocytes (Kennedy et al., the brain. The extent of inflammation and 1985; Zink and Narayan, 1989). The over- all actions of the inter- the spectrum of affected organs may lentivirus-specific feron would tend to aggravate and per- depend on the genetics of the infected ani- petuate inflammation, providing new host mal as well as on the strain of the infecting cells for the virus and, at the same time, virus (DeMartini et al., 1991 ). Different preventing the virus from replicating at a breeds may differ in the degree of their high rate that might stimulate a more vig- susceptibility to developing ovine pro- orous antiviral immune It should the North American response. gressive pneumonia, be noted, however, that this interpretation form of maedi visna et al., 1986). (Cutlip reflects an of in vitro data In addition, visna, the classical CNS form extrapolation to a situation in vivo, which is likely to of infection originally observed in Ice- be considerably more complex. In the light land, was only rarely seen elsewhere. The of more recent data obtained relating to mechanisms of determined genetically HIV, it would be interesting to know resistance to clinical disease have not been whether part or all of the activity by this determined in but in are sheep goats they lentivirus-specific interferon may be linked to the MHC class I and class II anti- related to the recently described and gens (Ruff et al., 1993). Field isolates chemokines that were shown to have a established strains of maedi- laboratory profound effect on the extent of replica- visna virus are highly heterogeneous tion of HIV (D’Souza and Harden, 1996). (Qu6rat et al., 1984; Sargan et al., 1991; Zanoni et al., 1992; Andresdottir et al., Investigations on the role in vivo of 1994; Carey and Dalziel, 1994; Leroux et classical type 1 and type 2 interferons and differ- al., 1996). Undoubtedly, genetic of other cytokines are scarce. Maedi-visna ences are for the differences in responsible virus-infected sheep with severe lymphoid virulence between individual strains of interstitial pneumonia had significantly virus although the underlying differences elevated levels of spontaneous interferon have not yet been determined in detail. production originating from pulmonary However, work by the group of De Martini leukocytes, as compared to both infected suggests that the extent of viral replica- animals with mild or no lesions of lym- tion and degree of cytopathicity may be phoid interstitial pneumonia and non- important markers of virulence, ’rapid- infected controls. It was thought that high’ strains being more virulent than increased local production of IFN in ’slow-low’ strains (Lairmore et al., 1987, lentivirus-infected host tissues may serve 1988b). to increase the numbers of leukocytes entering the sites of viral replication. This the virus may be an important trigger for promotes cell-mediated tissue damage and the formation of the lesion. In line with also provides larger numbers of cells for this interpretation, UV-inactivated virus virus replication (Lairmore et al., 1988a). was found to stimulate the expression of Infection of alveolar macrophages resulted GM-CSF in ex vivo alveolar macrophages in increased expression of interleukin-8 (Woodall et al., 1997), indicating that virus (IL-8) mRNA. Interestingly, mRNA of replication was not required for the virus this cytokine was also demonstrated to be to have this effect. Infection with maedi increased in the lungs of sheep infected visna was also shown to alter the capacity with maedi-visna virus. Expression of IL- of macrophages for generating reactive 8 correlated with the severity of the oxygen species (Cottin et al., 1996), which lesions. This led to the suggestion that IL- indicates that the virus alters an important 8 may play an active role in shaping the effector function implicated in the defence histological changes observed in the lungs against microorganisms as well as in host of sheep suffering from maedi visna pathology. The generation of enhanced (Legastelois et al., 1996). This situation levels in response to exogenous stimula- appears to differ from that found in the tion is of particular interest because it is synovial membranes of CAE virus- indicative of ‘priming’, i.e. the cells show infected goats. Even though CAE virus a difference in function only when caused an increased expression of IL-8 in responding to exogenous stimulation. This cultured macrophages, no such effect was particular type of functional alteration may found in the synovial membranes (Lechner also occur when the cells respond to stim- et al., 1997b). Since IL-8 expression was ulation by ’physiological’ signals origi- found to be increased only in those nating from other cells in situ. We have macrophages in which virus replicated at recently observed that macrophages a very high rate, this difference between infected with CAE virus in vitro show the status in the lungs of MV-infected altered expression of some, but not all, sheep and that in the synovial membranes cytokines investigated (Woodall et al., of CAEV-infected goats may be accounted 1997; Lechner et al., 1997a). Hence, such for by a more restricted replication of priming for an altered response may con- CAEV in the synovial membranes of goats tribute to the chronic inflammation which in contrast to a higher rate of replication of is a hallmark of infection by small rumi- MVV in the lungs of sheep. An active role nant lentiviruses. of infected macrophages in promoting inflammation is further suggested by the To date, the question of how the induction of procoagulant activity both in histopathological lesions typical of maedi cultured cells and in the lungs of infected visna may be generated is unresolved. sheep (Lena et al., 1994). Recently, Clearly, the mononuclear inflammation Woodall et al. reported elevated levels of itself is the major pathogenic factor pro- mRNA for granulocyte-macrophage moting some of the more obvious alter- colony-stimulating factor (GM-CSF), ations seen in the final stages of chronic interleukin-2 receptor (IL-2R), and inter- maedi. These include, among others, lung leukins 1 f3,4 4 and 10 (IL-1 (3, IL-4, IL-10) fibrosis and its well-known manifestations (Woodall et al., 1997). Interestingly, such as a decrease in the gas exchange in expression of TNF-a was not increased, the lungs due to a decrease in the total which suggests that, in maedi, not all pro- alveolar surface and an increase in the dis- inflammatory cytokines are upregulated. tance between the luminal and vascular The severity of inflammation was corre- sides of the alveoli. Lung fibrosis is a com- lated with the viral load, indicating that plex process, to which cytokines and other immune mediators produced during the CAEV are closely related (Gogolewski et chronic inflammation contribute. In addi- al., 1985), AGID based on this antigen has tion, reactive oxygen species generated for a long time been routinely used to by macrophages and other phagocytic cells detect small ruminant lentivirus infections may shift the fragile protease/antiprotease in both sheep and goats. Precipitating anti- balance in favour of the former which, in bodies identified by AGID are anti-p25 turn, results in enzymatic tissue destruction with a crude concentrate and large periph- (Hutchison, 1987). Associated with fibro- eral wells (macroimmunodiffusion test) sis is a decrease in the compliance of the (Cutlip et al., 1977), and anti-gp135 with lung tissue which manifests as the kind of a microhexagonal well pattern, with alter- strained breathing which is called ’maedi’ nate large and small peripheral wells by the Icelanders. Similar to other forms of (microimmunodiffusion test) (Winward lung fibrosis, bacterial infections may et al., 1979; Dawson et al., 1982). Several become more frequent and more difficult studies demonstrated that an AGID assay to control efficiently. In turn, such infec- with CAEV gp 135 is more sensitive than tions have an adverse effect on the health an AGID assay with CAEV p25 (Grewal, status, as do other lung diseases such as 1986; Adams and Gorham, 1986). adenomatosis, an infectious tumor caused Radioimmunoprecipitation assays revealed by an oncovirus (Snyder et al., 1983, Daw- that goats infected with CAEV have much son et al., 1990). higher antibody titers against gp135 than against p25, emphasizing the necessity of using the appropriate antigen in AGID to 6. DIAGNOSIS attain a high sensitivity and specificity in the detection of antibody to CAEV or MVV (Knowles et al., 1994). Hence, pre- 6.1 Antibodies and nucleic acids cipitation in an agar gel requires multiple epitope-antibody interactions, whereas Due to the persistence of circulating the radioimmunoprecipitation assay antibodies, the diagnosis of MVV infec- requires only the binding to a single epi- tion is most commonly based on serolog- tope (Knowles et a]., 1994). Thus, the ori- ical tests. In recent years, as an alterna- gin and characteristics of the viral strain tive to serology, methods have been producing the precipitating antigen appear developed that allow the detection of the to be of major importance (Klein et al., viral genome by PCR. 1985). However, the interpretation of the microimmunodiffusion test with gpl35 6.1.1. The first generation assays: antigen sometimes appears difficult or AGID, whole-virus ELISA subjective and may lead to false-positive and immunoblot or false-negative results, although, theo- retically, this assay is more sensitive than The most widely used test is agar gel the anti-p25 AGID. In unclear cases, the immunodiffusion (AGID), first described results must be confirmed in ELISA or in the late 1970s (Cutlip et al., 1977; Win- immunoblot (western blot). ward et al., 1979). The antigen, a concen- trate of medium harvested from cell cul- The development of indirect ELISAs tures infected with the WLC 1 MVV strain, has improved the diagnosis of MVV and contains both major structural proteins, CAEV. The antibody response is detected i.e. the core protein p25 and the major at an earlier timepoint and yields a semi- envelope glycoprotein gp135 (Dawson et quantitative assessment of the level of cir- al., 1996). As, antigenically, MVV and culating antibodies (Vitu et al., 1982; Houwers et al., 1982; Russo et al., 1988). or monoclonal antibody conjugates These first-generation ELISAs used par- (Bosgiraud et al., 1989; Zanoni et al., tially or highly purified preparations of 1989, 1994). Generally, a sample is con- whole-virus antigens, and polyclonal sidered positive in the western blot if anti- enzyme conjugates, thus generating false- bodies to at least two gene products are positive results, and necessitating a high detected (Brodie et al., 1992; Johnson et dilution of the sera to avoid non-specific al., 1992). Nevertheless, the western blot, reactions. although valued as a confirmation test, is not suitable for large numbers of sera, and Immunoblot techniques (western blot) can still yield false-positive results. have been used to analyse the antibody responses to each of the major viral pro- 6.1.2. The second teins, i.e. the gp135 - also referred to as generation assays: gp105 (Kajikawa et al., 1990; Brodie et assays based on monoclonal recombinant al., 1992), the transmembrane (TM) pro- antibodies, proteins tein gp44 (or gp55) and the internal pro- and peptides teins p25, p17 and p14, in sera of experi- mentally or naturally infected sheep By using monoclonal antibodies anti- (Houwers and Nauta, 1989; Johnson et p25 and a double-sandwich method, al., 1992; Torfason et al., 1992) or goats improved assays with a higher specificity (Vitu et al., 1993; Barlough et al., 1994). and reliability were developed (Houwers In some cases, in the western blot, anti- and Schaake, 1987; Reyburn et a]., 1992). bodies to p25 were detected before anti- Numerous recombinant proteins have been gpl35 in experimentally infected sheep developed since 1990, following the initial (Houwers and Nauta, 1989), and localization of immunodominant regions detectable antibody responses to p14, p 177 in the Env and Gag proteins (Kwang and and even p25 may be absent in sheep with Cutlip, 1992; Kwang et al., 1996). The MVV lesions (Houwers and Nauta, 1989). gp70 (a degradation product of either the Technical problems may well explain that precursor, gp160 or of SU, gp135) and different authors recorded differing results gp40 (equivalent to TM glycoprotein, concerning a delayed anti-gp135 response gp44) were expressed in Escherichia coli, (Houwers and Nauta, 1989; Bosgiraud et and the entire gp70 was expressed in insect al., 1989; Kajikawa et al., 1990; Torfason cells by a recombinant baculovirus et al., 1992). They also explain the greater (Kwang et al., 1995). Fragments of the sensitivity of the radioimmunoprecipita- gp135 were expressed as fusion proteins tion assay in the early detection of anti- and used to analyse the antibody response bodies to viral glycoproteins in naturally to gpl 35 in MVV-infected sheep (Carey et and experimentally infected animals al., 1993). Different segments of Gag and (Gogolewski et al., 1985; Mazarin et al., of the 44 kDa TM glycoprotein were 1990; Knowles et al., 1994; Chebloune et expressed as glutathione S-transferase al., 1996). Nevertheless, as the radioim- (GST) fusion proteins (Power et al., 1995). munoprecipitation assay is expensive and These recombinant serve as time-consuming, it is rarely used in routine proteins antigens in new and promising assays diagnosis, but rather represents a ’last (Reyburn et al., 1992; Kwang and Cutlip, resort’ serological test for cases that can- Zanoni et Rosati et not be resolved in western blot. 1992; al., 1994; al., 1994; Power et al., 1995; Keen et al., The specificity of ELISA or western 1995; 1996; Boshoff et al., 1997), which blot can be improved by replacing the sec- are generally more sensitive and specific ond antibody by conjugates of protein G than whole virus tests. However, even if the results obtained with recombinant pro- for the direct detection of MVV in clinical teins turn out to be more sensitive, some specimens, either prior to culture or after non-specific reactions nevertheless remain, cocultivation with susceptible cells. The most notably due to antibodies binding to latter is more sensitive as only a few the GST fusion partner (Boshoff et al., PBMCs are infected by small ruminant 1997). In addition, insufficiently purified lentiviruses in vivo. In experimentally antigens may lead to false positive results, infected animals, PCR seems to be more making it necessary to check the speci- sensitive than serology, presumably ficity by using double-well ELISA kits because primers can be used that are per- which in turn increases the cost of serol- fectly complementary to the nucleotide ogy. sequences of the virus selected for infect- ing the animals (Vitu et al., 1997). Con- Synthetic oligopeptide assays, also results of cases of natu- called and versely, reported third-generation assays (Kwang infected animals indicated that direct Torres, 1994), have been developed, with rally PCR on when with immunodominant epitopes of the TM PBMCs, compared serological tests, might fail in a number gp44, or using a recombinant p25 com- of seropositive animals (Rimstad et al., bined with a peptide from the envelope 1993; Zanoni et al., 1996; Vitu et al., protein. In view of the cost of peptide tech- 1997). Moreover, direct PCR yields a pos- nology, the future use of these tests in itive result only in animals exhibiting a MVV eradication programmes will depend high virus load (Brodie et al., 1992). on their sensitivity and specificity. Attempts at PCR diagnosis of milk, 6.1.3. New developments: based on DNA and RNA extraction, detection of nucleic acids showed mixed results (Rimstad et a]., 1993; Barlough et al., 1994; Zanoni et al., Seroconversion may take a long time, 1996; Vitu et al., 1997; Leroux et al., and some infected animals may indeed 1997). In addition to the presence of an intermittent fail to develop a detectable antibody inhibitory contaminants, in milk is response. Moreover, as serologically pos- shedding of virus-infected cells itive animals may transiently become neg- suggested, which would limit the appli- ative, novel diagnostic methods are called cation of PCR in field conditions (Vitu et for to detect the presence of viral compo- al., 1997). nents in cells or tissues. PCR diagnosis has been improved by In recent years, the PCR, known for its an array of degenerate primer sets and by high sensitivity, has been applied to the nested, semi-nested and double-nested diagnosis of lentivirus infection in sheep procedures (Leroux et al., 1995; Barlough and goats to detect DNA and RNA in et al., 1994; Vitu et al., 1997; Leroux et peripheral blood and tissues. Preliminary al., 1997). Southern-blot hybridization reports have demonstrated proviral DNA with suitable probes is more sensitive than in cultured cells within 24 h post infec- ethidium bromide staining (Rimstad et al., tion (Zanoni et al., 1990b). These reports 1993; Russo et al., 1997), and significantly have also stressed the importance of select- increases the sensitivity and specificity, ing primers that take into account that dif- especially when samples are analysed ferent regions of the viral genome differ in without prior in vitro culture of cells. the degree of heterogeneity. In most stud- Results obtained with PCR techniques ies, conserved regions of LTR, and of gag indicated that seroconversion can be or pol genes were targeted for PCR delayed for several months following nat- (Zanoni et al., 1990a, b). PCR can be used ural infection with CAEV (Rimstad et al., 1993). However, the PCR test remains goat synovial membrane cells which also expensive and labour-intensive, and its support the replication of MVV (Johnson performance will have to be evaluated in et al., 1992). The cocultivation can also a greater number of field samples before it be performed with PBMCs or milk leu- can become the ’gold standard’ for detect- cocytes. Explant cultures of affected tissue ing MVV infection, which, theoretically, are carried out after necropsy. A cyto- should be possible because it can demon- pathic effect with formation of giant multi- strate the presence of a low number of tar- nucleated cells (syncytia) is expected after get sequences. 2-3 weeks, but some ovine lentivirus strains not be detected due to their The in situ PCR has been adapted to may failure to induce a clear effect amplify viral DNA in fixed cells, thus cytopathic or because the virus remains latent facilitating the identification of latent et al., In doubtful cases, infection of cells (Haase et al., 1990). In (Chebloune 1996). cell elec- situ hybridization, which detects MVV staining, immunocytochemistry, tron or RNA in cultured cells, has been reported to microscopy, reverse-transcriptase tests are (Mazarin et al., 1990). be as sensitive as PCR, and cocultivation performed studies using cells of latently infected seronegative sheep suggest that the infec- tion frequently remains undetected by 7. PREVENTION serological tests (Johnson et al., 1992). Combined with immunocytochemistry, in situ hybridization permits the simultaneous 7.1. Present methods detection of MVV antigens and RNA within the same cell (Roy et al., 1992). In In most countries, both MVV and situ is a hybridization complementary CAEV infections in sheep and goats are of classical but technique histopathology, currently controlled by an array of com- is more useful in experimental studies than plementary methods (Dion, 1991 ). Peri- as a tool. diagnostic odic serological tests using agar gel immunodiffusion (AGID) or ELISA rep- resent the standard method of detecting 6.2. Other diagnostic methods MVV-infected animals. For the eradica- tion of infection in the flocks, two differ- In cases of clinical disease, gross and ent strategies are adopted. Because, as microscopic post mortem pathology indi- described above, colostrum and milk are of cates the presence of histological alter- prime importance in the infection of new- ations in target organs, i.e. interstitial pneu- born lambs or kids, the lambs are removed monia with a predominant mononuclear from their infected mothers at birth and cellular infiltration (Mornex et al., 1994; raised in separate flocks. Colostrum fed Cador6 et al., 1996), presence of lymphoid to these lambs is heat-treated (56 °C for follicles in the udder parenchyma and adja- 60 min) and milk is pasteurized (Houw- cent to secretory ducts (Lujan et al., 1991) ers et al., 1983). Preferably, however, the associated with an increased number of T animals are fed colostrum and milk from lymphocytes and macrophages in milk cell certified MV-free ewes. As an alternative counts (Guiguen et al., 1992). to separating newborn lambs from their mothers, animals be Virus isolation is a delicate seropositive may technique: removed from the flock. tissue samples taken from an infected ani- mal must contain living cells for coculti- Although these methods have met with vation on sheep chorioid plexus cells or some success in several countries, these control and eradication programmes have The protective role these mutants play can a number of limitations, such as insuffi- be determined after either conventional or cient sensitivity and specificity of sero- DNA vaccination (Perk et al., 1996; Har- logical tests, relative importance of the mache et al., 1996b). Bacterial or viral other modes of transmission or resistance vectors to produce recombinant vaccines to culling seropositive animals particu- have turned out to be another promising larly in flocks raised for commercial milk approach (Cardenas and Clements, 1992; production. Bourgogne et al., 1996; Xu et al., 1997). The risk of resistant MVV strains and the cost involved have pre- emerging 7.3. Other control methods vented the development of chemotherapy for treating small ruminant lentivirus infec- The selection of with a natural tions. The only use of antiviral drugs in sheep resistance to MVV offer another maedi-visna infection is to provide a may model for in vivo testing of candidate anti- approach to controlling MVV. A few stud- ies have a natural resis- HIV drugs (Thormar et al., 1995). reported possible tance to the disease in some breeds (Perk et al., 1996). A high seroprevalence has been demonstrated in the Border 7.2. Vaccination Texel, Leicester and Finnish Landrace breeds (Vorster et al., 1996; Zink and Johnson, Not vaccines for MVV surprisingly, 1994), as opposed to the Ile-de-France are not available due to the currently breed (Houwers et al., 1989). It seems that formidable difficulties the presented by there exists a genetic susceptibility to the of the interaction between the biology disease rather than a susceptibility to the lentivirus and its host, varia- e.g. genetic infection per se (Zink and Johnson, 1994). tion of the the mechanisms virus, complex The most promising strategy would be to of and viral immunoprotection persistance learn more about the immune mechanisms et Montelaro et (Pearson al., 1989; al., involved in natural resistance to the dis- These obstacles to vaccine devel- 1989). ease at the level of the breed as a whole the of opment highlight importance pre- and at that of the individual animal. Breed- vention. ing transgenically resistant sheep, even if Despite the prospects of developing an theoretically possible (Clements et al., effective vaccine against MVV and CAEV 1994), appears, in practice, not to be an in sheep and goats remain poor (Phelps achievable goal. and Smith, 1993), a number of research groups continue to defend the feasability of a search for such a vaccine (Pearson et 8. CONCLUSION al., 1989; Cheevers et al., 1994; Perk et al., 1996; Harmache et al., 1996b), advo- In view of the impact of MVV on the cating different strategies. The use of intact farming sector of most countries, the dis- inactivated virions does not seem com- ease is classified in list B of diseases by the mendable due to inefficacy and a debat- Office International des Epizooties (Daw- able risk of such a product inducing even son et al., 1996). A continuation of more severe symptoms and lesions research is therefore called for in order to (McGuire et al., 1986; Russo et al., 1993). increase our knowledge. Inter alia, the In contrast, the use of attenuated viruses genetic variability of ovine and caprine obtained by deletion of selected genes, i.e. lentiviruses in both natural and experi- vif, tat and dUTPase, looks promising. mental infections must be analysed with a view to developing tools permitting more Andresson O.S., Elser J.E., Tobin G.J., Greenwood refined methods of lentiviral J.D., Gonda M.A., Georgsson G., Andresdottir diagnosing V., Benediktsdottir E., Carlsdottir H.M.. infections. Farmers and veterinary author- Mantyla E.O., Rafnar B., Palsson P.A., Casey ities anxious to improve their MVV erad- J.W., Petursson G., Nucleotide sequence and of ication programmes would welcome such biological properties a pathogenic proviral molecular clone of neurovirulcnt visna virus, tools. Virology 193 (1993) 89-105. In addition, the viral determinants of Audoly G., Sauze N., Harkiss G.D., Vitu C., Russo virulence and the pathogenesis of ovine P., Qu6rat G., Suzan M., Vigne R., Identifica- tion and subcellular localization of the Q gene lentivirus in its natural host must be elu- product of visna virus, Virology 189 (1992) cidated, not least to improve our under- 734-739. standing of the disease mechanisms under- Barlough J., East N.E., Rowe J.D., Vanhoosear K., lying HIV, particularly macrophage-tropic DeRock E., Bigornia L., Rimstad E., Double- nested chain-reaction for detection HIV. In this context, the determination of polymerase of caprine arthritis-encephalitis virus proviral cell receptors for MVV infection as has DNA in blood, milk, and tissues of infected recently been achieved in the case of HIV, goats, J. Virol. Methods 50 (1994) 101-1 l3. SIV and FIV (Willet et al., 1997), would Bertoni G., Zahno M.L., Zanoni R.G., Vogt H.R., constitute a major scientific advance. Peterhans E., Ruff G., Cheevers W.P., Sonigo P., Pancino G., Antibody reactivity to the Ultimately, vaccination strategies need immunodominant epitopes of the caprine arthri- virus transmembrane to be in order to extend the tis-encephalitis gp38 pro- developed tein associates with the development of arthri- armamentarium available to control maedi tis, J. Virol. 68 (1994) 7139-7147. visna in sheep. Bird P., Blacklaws B.A., Reyburn H.T., Allen D., Hopkins J., Sargan D.R., McConnell I., Early events in immune evasion by the lentivirus Maedi-Visna occurring within infected lym- ACKNOWLEDGEMENTS phoid tissue, J. Virol. 67 (1993) 5187-5197. Bird P., Reyburn H.T., Blacklaws B.A., Allen D., This review article is part of the COST Nettleton P., Yirrell D.L., Watt N.J., Sargan action no. 834 on ’Lentiviruses of sheep and D.R., McConnell I., The restricted IgGI anti- goats: pathogenesis, diagnosis and prevention’. body response to maedi-visna virus is seen fol- infection but not immuniza- This work (MP, CV, PR, JFM) was in lowing following part sup- tion with recombinant GAG Clin. from the ANRS. EP was protein, Exp. ported by grants sup- Immunol. 102 ( 1995) 274-280. ported by Swiss National Science Foundation 3100-09733.93/ and 3139-04l 859.94. Blacklaws B.A., Bird P., Allen D., McConnell L, grants T The authors wish to thank Ms R. Parham for Circulating cytotoxic lymphocyte precursors in maedi-visna virus-infected sheep, J. Gen. linguistic improvements. Virol.75 (1994) 1589-1596. Blacklaws B.A., Bird P., Allen D., Roy D.J., MacLennan I.C.M., Hopkins J., Sargan D.R., REFERENCES McConnell L, Initial lentivirus host interac- tions within lymph nodes: a study of Maedi- Visna virus infection in sheep, J. Virol. 69 Adams D.S., Gorham J.R., The gp135 of caprine (1995)1400-1407. arthritis virus affords sen- encephalitis greater Rucheton Costc Lemairc sitivity than the p28 in immunodiffusion scrot- Bosgiraud C., M., J., J.M., Nicolas J.A., Simeon de Buochberg M., ogy, Res. Vct. Sci. 40 (1986) 157-160. Beaubatie L., Detection d’anticorps et Anderson A.A., Harkiss G.D., Watt N.J., Quantita- d’antig6nes du virus dc visna-maedi par tive analysis of immunohistological changes immunotransfert, Ann. Rech. V6t. 20 ( 1989) in the synovial membrane of sheep infected 187-193. with Maedi-Visna Virus, Clin. Immunol. Boshoff C.H., Dungu B., Williams R., Vorster J., Immunopathol. 1 ( 1994) 21-29. Conradie J.D., Verwoerd D.W., York D.F., Andresdottir V., Tang X., Andresson O.S., Georgs- Detection of maedi-visna virus antibodies using son G., Sequence variation in the envelope gene a single fusion transmembrane-core p25 recom- and the LTR of maedi-visna virus, Ann. NY binant protein ELISA and a modified receiver- Acad. Sci. 724 ( 1994) 157-158. operating characteristic analysis to determine cut-off values, J. Virol. Methods 63 (1997) pressor domains, J. Virol. 68 (1994) 47-56. 6137-6146. Bourgogne A., Sanchis R., Hofnung M., P6pin M., Carruth L.M., Morse B.A., Clements J.E., The Salmonella abortu.rovi.s, souche Rv6, v6hicule leucine domain of the visna virus Tat protein vaccinal pour les ovins et caprins, 3!ll’s Ren- mediates targeting to an AP-I sitc in the viral centres des Recherches sur les Ruminants, , J. Virol. 70 (1996) Paris, France, 4-5 December, vol. 3, 1996, pp. 4338-4344. 157-I60. Chadwick B.J., Coelen R.J., Sammels L.M., Ker- Braun M.J., Clements J.E., Gonda M.A., The visna tayadnya G., Wilcox G.E., Genomic sequence virus genome: evidence for a hypervariable site analysis identifies Jemhrana disease virus as a in the env gene and sequence homology among new bovine lentivirus, J. Gen. Virol. 76 (1995) lentivirus envelope proteins, J. Virol. 61 (1987) 189-19z. 4046-4054. Chebloune Y., Karr B., Sheffer D., Leung K., Brodie S.J., Pearson L.D., Snowder G.D., DeMartini Narayan 0., Variations in lentiviral gene in J.C., Host-virus interaction as defined by ampli- expression monocyte-derived macrophages fication of viral DNA and serology in lentivirus- from naturally infected sheep, J. Gen. Virol. infected sheep, Arch. Virol. 130 (1992) 77 ( I 996) 2037-2(l51 . 413-428. Cheevers W.P., Knowles D.P., McGuire T.C., Bas- zler Brodie S.J., de la Concha-Bermejillo A., Koenig G., T.V., Hullinger G.A., Caprine arthritis- Snowder G.D., DeMartini J.C., Matemal factors encephalitis lentivirus (CAEV) challenge of immunized associated with transmission of ovine goats with recombinant vaccinia prenatal virus lentivirus, J. Infect. Dis. 169 (1994) 653-657. expressing CAEV surface and trans- membrane envelope glycoproteins, Vet. Brodie S.J., Pearson L.D., Zink M.C., Bickle H.M., Lnmunol. Immunopathol. 42 ( 1994) 237-251.1. Anderson B.C., Marcom DeMailini K.A., J.C., Clements Molecular basis of the Ovine lentivirus and disease. Virus J.E., Payne S.L., expression of lentiviruses, Virus Res. 32 but not is restricted to pathobiology Replication, entry, (1994) 97-109. macrophages of specific tissues, Am. J. Pathol. 146 (1995) 250-263. Clemcnts J.E., Zink M.C., Molecular biology and of animal lentivirus infections, Cador6 Cordier Loire pathogenesis J.L., Guiguen F., G., R., Lyon Clin. Microbiol. Rev. 9 (1996) 100-117. M., Chastang J., Greenland T., Court-Fortune I., Clements Gdovin Montelaro Revel D., Mornex J.F., Early events in the J.E., S.L., R.C., Narayan experimental interstitial lung disease induced in 0., Antigenic variation in lentiviral diseases, sheep by the visna-maedi virus, Immunol. Lett. Annu. Rev. Immunol. 60 (1988) 139-159. 1 ( 1993) 39--43. Clements J.E., Wall R.J., Narayan 0., Hauer D., Sheffer Powell Carruth Cador6 J.L., Greenland T., Cordier G., F., Schoborg R.V., D., A., Guiguen Zink Rexroad Mornex J.F., of the L.M., M.C., C.E., Development Histogenesis pulmonary that the visna virus lesions in the course of Visna Maedi Virus- of transgenic sheep express 200 370-380. induced pneumonia, Vet. Res. 27 (1996) envelope gene, Virology ( I 994) 419-426. Cottin V., Court-Fortune I., Crevon J., Mornex J.F., Oxidant-antioxidant imbalance in the Menzies Waltncr-Tocws experi- Campbell J.R., P.I., D., mental interstitial disease induced in Walton Buckrell Thorsen J., The lung sheep J.S., B.C., Visna-Maedi virus, Eur. J. 9 of maedi-visna in Ontario by Resp. (1996) seroprevalence sheep 1983-1988. flocks and its relationship to flock demo- graphics and management practices, Can. Vet. Cousens C., Minguijon E., Garcia M., Ferrer L.M., J. 35 (1994) 39-44. Dalziel R.G., Palmarini M., De Las Heras M., Sharp J.M., PCR-based detection and partial Cardenas L., Clements J.D., Oral immunization using characterization of a associated with live attenuated Salmonella as carriers of spp. contagious intranasal tumors of sheep and goats, foreign antigens, Clin. Microbiol. Rev. 5 (1992) J. Virol. 70 (1996) 7580-7583. 328-342. Craig L., Nealen M.L., Strandberg J.D., Zink C., Carey N., Dalziel R.G., Sequence variation in the Differential replication of ovine lentivirus in gp135 gene of maedi-visna virus strain EV 1, endothelial cells cultured from different tis- Virus Genes I 15-123. 8 (1994) sues, Virology 238 (1997) 316-326. Carey N., Roy D.J., Dalziel R.G., Use of recombinant Cutlip R.C., Jackson T.A., Laird G.A., Immunodif gpl35 to study epitope-specific antibody fusion test for ovine progressive pneumonia, responses to maedi-visna virus, J. Virol. Meth- Am. J. Vet. Res. 38 (1977) 1081-1084. ods 43 221-232. ( 1993) Cutlip R.C., Lehmkuhl H.D., Brogden K.A., Sacks Carruth L.M., Hardwick J.M., Morse B.A., Clements J.M., Breed suceptibility to ovine progressive J.E., Visna virus Tat protein: a potent tran- pneumonia (Maedi/Visna) virus, Vet. Micro- scription factor with both activator and sup- biol. 12 ( 1986) 283-288. Dalziel R.G., Hopkins J., Watt N.J., Dutia B.M., Stanley J., Pezeshkpour G., Tropism of sheep Clark J., McConnell I., Identification of a puta- lentiviruses for monocytes: susceptibility to tive cellular receptor for the lentivirus visna infection and virus gene expression increase virus, J. Gen. Virol. 72 (1991) 1905-1911.1 . during maturation of monocytes to 67-74. Dawson M., Biront P., Houwers D.J., Comparison of macrophages, J. Virol. 58 (1986) serological tests used in three state veterinary Georgsson G., Houwers D.J., Palsson P.A., Peturs- laboratories to identify maedi-visna virus infec- son G., Expression of viral antigens in the cen- tion, Vet. Rec. 111 (1982) 432-434. tral nervous system of visna-infected sheep: an Dawson M., Done S.H., Venables C., Jenkins C.E., histochemical study on experimental visna Maedi-visna and sheep pulmonary adeno- induced by virus strains of increased neurovir- matosis- A study of concurrent infection, Br. ulence, Acta NeuropaLhol. 77 (1989) 299-306. Vet. J. 146 (1990) 531-538. Gogolewski R.P., Adams D.S., Me Guire T.C., Banks Dawson M., Lysons R.E., Knowles D.P., Caprine K.L., Cheevers W.P., Antigenic cross-reactiv- arthritis/encephalitis & maedi-visna, in: Manuel ity between caprine arthritis-encephalitis, visna of Standards for Diagnostic Tests and Vac- and progressive pneumonia viruses involves cines, 3rd edn, Office International des Epi- all virion-associated proteins and glycopro- zooties, Paris, 1996, pp. 369-371.1. teins, J. Gen. Virol. 66 (1985) 1233-1240. de la Concha-Bermejillo A., Maedi-visna and ovine Gougeon M.L., Garcia S., Heeney J., Tschopp R., progressive pneumonia, Veterinary Clinics of Lecoeur H., Guetard D., Rame V., Dauguet C., North America: Food Animal Practice ( 1997) Montagnier L., Programmed cell death in 13-33. AIDS-related HIV and SIV infections, AIDS de la Concha-Bermejillo A., Magnus-Corral S., Res. Hum. Retroviruses 9 (1993) 553-563. Brodie S.J., DeMartini J.C., Venereal shedding Greenwood P.L., North R.N., Kirkland P.D., Preva- of ovine lentivirus in infected rams, Am. J. Vet. lence, spread and control of caprine arthritis- Res. 57 (1996) 684-688. encephalitis virus in dairy goat herds in New- DeMartini J.C., Bowen R.A., Carlson J.O., de la South-Wales, Aust. Vet. J. 72 (1995) 341-345. in: Owen Axford Concha-Bermejillo A., J.B., Grewal A.S., Comparison of two gel precipitin tests R.F.E. (Eds.), Breeding for Disease Resistance in the serodiagnosis of caprine arthritis in Farm CAB Animals, International, Walling- encephalitis virus infection in goats, Aust. Vet. ford, Oxon, 1991, pp. 293-314. J. 63 (1986) 341-342. Dion F., Mise en et evolution de la place prophy- Gudnadottir M., Kristinsdottir K., Complement-fix- laxie du visna-maedi en France, Point V6t. 23 antibodies in sera of affected with 1 . ing sheep ( 1991 ) 699-711. visna and maedi, J. Immunol. 98 (1967) D’Souza M.P., Harden V.A., Chemokines and HIV- 663-667. 1 second receptors. Confluence of two fields Lerondelle C., Ouzrout R., generates optimism in AIDS research, Nat. Guiguen F., L’expres- Med. 2 (1996) 1293-3000. sion des lentivirus au moment de I’agnelage modifie la formule leucocytaire du lait de bre- Elder J.H., Lerner D.L., Hasselkus-Light C.S., bis Ann. Rech. V6t. 23 (1992) Luciw Monte- multi pares, Fontenot D.J., Hunter E., P.A., 189-197. laro R.C., Phillips T.R., Distinct subsets of retroviruses encode dUTPase, J. Virol. 66 Haase A.T., Retzel E.F., Staskus K.A., Amplification (1992)1791-1794. and detection of lentiviral DNA inside cells, Proc. Natl. Acad. Sci. USA 87 (1990) Rimmelzwaan G., Minassian A., Tsai Garrity R.R., 4971 !975. W.P., Lin G., de Jong J.J., Goudsmit J., Nara P.L., Refocusing neutralizing antibody response Harkiss G.D., Watt N.J., King T.J., Williams J., Hop- by targeted dampening of an immunodominant kins J., Retroviral arthritis: phenotypic analysis epitope, J. Immunol. 159 (1997) 279-289. of cells in the synovial fluid of sheep with Gdovin S.L., Clements J.E., Molecular mechanisms inflammatory synovitis associated with visna virus Clin. Immune). of visna virus Tat: identification of the targets infection, Immunopathol. for transcriptional activation and evidence for 60(1991)106-117. a post-transcriptional effect, Virology 188 Harmache A., Bouyac M., Audoly G., Hi6blot C., (1992) 438-450. Peveri P., Vigne R., Suzan M., The vifgene is Gendelman H.E., Narayan 0., Molineaux S., essential for efficient replication of caprine Clements J.E., Ghotbi Z., Slow, persistent repli- arthritis encephalitis virus in goat synovial cation of lentiviruses: role of tissue membrane cells and affects the late steps of the macrophages and macrophage precursors in virus replication cycle, J. Virol. 69 (1995a) bone marrow, Proc. Natl. Acad. Sci. USA 82 3247-3257. (1985)7086-7090. Harmache A., Vitu C., Russo P., Bouyac M., Hi6blot Gendelman H.E., Narayan 0., Kennedy-Stoskopf C., Peveri P., Vigne R., Suzan M., The caprine S., Kennedy P.G.E., Ghotbi Z., Clements J.E., arthritis encephalitis virus tat gene is dispens- able for efficient viral replication in vitro and in susceptible cells, Clin. Immunol. vivo, J. Virol. 69 (1995b) 5445-5454. Immunopathol. 65 ( 1992) 254-26(l. Harmache A., Russo P., Guiguen F., Vitu C., Vignoni Kajikawa 0.. Lairmore M.D., DeMartini J.C., Anal- M., Bouyac M., Hi6blot C., P6pin M., Vigne ysis of antibody responses to phenotypically R., Suzan M., Requirement of Caprine Arthri- distinct lentiviruses, J. Clin. Microbiol. 28 tis Encephalitis Virus vif gene for in vivo repli- ( I 990) 764-77(l. cation, Virology 224 (1996a) 246-255. Keen J.E., Kwang J., Rosati S., Comparison of ovine Harmache Russo Vitu Mornex A., P., C., Guiguen F., lentivirus detection by conventional and recom- J.F., Pepin M., Vigne R., Suzan M., Replication binant serological methods, Vet. Immunol. in goats in vivo of caprine arthritis encephalitis Immunopathol. 47 (1995) 295-309. virus deleted in vif or tat genes: possible use Keen Littledike of these deletion mutants as live vaccines, AIDS J., Kwang J., E.T., Hungerford L.L., in Res. Hum. Retroviruses 12 (1996b) 409!11.] . Ovine lentivirus antibody detection serum, colostrum and milk using a recombinant trans- Hayman M., Arbuthnott G., Harkiss G., Brace H., mcmbranc protein ELISA, Vet. Immune!. Filippi P., Philippon V., Thomson D., Vigne Immunopathol. 51 ( 1996) 253-275. R., Wright A., Neurotoxicity of peptide ana- logues of the transactivating protein tat from Kennedy P.G.E., Narayan 0., Ghotbi Z., Hopkins Maedi-visna virus and human immunodefi- J., Gendelman H.E., Clements J.E., Persistent ciency virus, Neuroscience 53 ( 1993) 1-6. expression of la antigen and viral genome in visna-maedi virus-induced cells. Hecht S.J., J.M., DeMartini Retroviral inflammatory Sharp J.C., Possible role of lentivirus-induced J. of ovine carci- intetl’eron, aetiopathogenesis pulmonary Exp. Med. 162 ( 1985) 197(>-1982. noma: a critical appraisal, Br. Vet. J. 152 ( 1996) 395!09. Kcnncdy-Stoskopf S., Narayan 0., Neutralizing anti- Hoglund S., Ohagen A., Lawrence K., Gabuzda D., bodies to visna lentivirus: mechanism of action and role in virus J. Virol. Role of vi/ during packing of the core of HIV- possible persistence, 1, Virology 201 (1994) 349-355. 59 ( 1986) 37-44. Houwers D.J., Epidemiology, diagnosis and control Klein J.R., Martin J., Griffing R., Nathanson N., Pre- of SRLV-infections, 3rd European Workshop cipitating antibodies in experimental visna and natural of Res. on Ovine and Caprine Retroviruses, Jaca, Spain, progressive pneumonia sheep, 2-5 March, 1997. Vet. Sci. 38 (1985) 129-133. Houwers D.J., Schaake J., An improved ELISA for Knowles D.P., Evcrmann J.F., Shropshire C., Van- the detection of antibodies to ovine and caprine derschalie J., Bradway D.S., Gezon H.M., lentiviruses, employing monoclonal antibod- Cheevers W.P., Evaluation of agar-gel immun- ies in a one-step assay, J. Immune). Methods 98 odiffusion serology using caprine and ovine (1987)151-154. Icntiviral antigens for detection of antibody to Houwers D.J., Nauta I.M., Immunoblot analysis of caprine Arthritis-Encephalitis virus, J. Clin. Microbiol. 32 (1994) 243-245. the antibody response to ovine lentivirus infec- tions, Vet. Microbiol. 19 (1989) 127-139. Kwang J., Cutlip R.C., Detection of antibodies to Houwers D.J., Gielkens A.L.J., Schaake J., An indi- ovine lentivirus using a recombinant antigen derived from the env Biochem. rect enzyme-linked immunosorbent assay gene, Biophys. (ELISA) for the detection of antibodies to Res. Comm. 183 (1992) 104(!1046. maedi-visna virus, Vet. Microbiol. 7 (1982) Kwang J., Torres J.V., Oligopeptide-based enzyme 209-219. immunoassay for ovine lentivirus antibody Houwers D.J., Koenig G., de Boer G.F., Schaake Jr detection, J. Clin. Microbiol. 32 (1994) J., Maedi-visna control in sheep. I. Artificial 1813-1815. rearing of colostrum-deprived lambs, Vet. Kwang J., Kim H.S., Rosati S., Lehmkuhl H.D., Microbiol. 8 (1983) 179-185. Characterization of ovine lentivirus envelope Houwers D.J., Visscher A.H., Defize P.R., Impor- glycoprotein expressed in Escherichia coli cell tance of ewellamb relationship and breed in and baculovirus systems, Vet. Immunol. the epidemiology of maedi-visna virus infec- Immunopathol. 45 (1995) 185-193. tions, Res. Vet. Sci. 46 (1989) 5-8. Kwang J., Rosati S., Yang S., Justc R.A., de la Con- Hutchison D.C., The role of proteases and antipro- cha-Bennejillo A., Recognition of ovine teases in bronchial secretions, Eur. J. Respir. lentivirus gag gene products by serum from Dis. 153 (Suppl) (1987) 78-85. infected sheep, Vet. Immunol. Immunopathol. Joag S.V., Stephens E.B., Narayan 0., Lentiviruses., 55 (1996) 107-I 14. in: Fields M. (Ed.), Virology, Lippincott-Raven Lairmore M.D., Akita G.Y., Russell H.I., DeMar- Publishers, New York, 1996, 1977-1996. tini J.C., Replication and cytopathic effects of Johnson L.K., Meyer A.L., Zink M.C., Detection of ovine lentivirus strains in alveolar macrophages ovine lentivirus in seronegative sheep by in correlate with in vivo pathogenicity, J. Virol. situ hybridization, PCR, and cocultivation with 61 ()987)4038!t042. Lairmore M.D., Butera S.T., Callahan G.N., DeMar- tion of Icelandic sheep, J. Gen. Virol. 64 (1983) tini J.C., Spontaneous interferon production by 1433-1440. pulmonary leukocytes is associated with Madewell B.R., Ameghino E., Rivera H., Inope L., lentivirus-induced lymphoid interstitial pneu- DeMartini J.C., Seroreactivity of peruvian monia, J. Immunol. 140 (1988a) 779-785. sheep and goats to small ruminant lentivirus- Lairmore M.D., Poulson J.M., Adducci T.A., DeMar- ovine progressive pneumonia virus, Am. J. Vet. tini J.C., Lentivirus-induced lymphoprolifera- Res. 48 ( 1987) 372-374. tive disease. Comparative pathogenicity of phe- Marin M., Etienne-Julan M., Piechaczyk M., Noel D., notypically distinct ovine lentivirus strains, L’integration des retrovirus: faits et croyances, Am. J. Pathol. 130 (1988b) 80-90. M6decine/Sciences 10 (1994) 318-324. Lechner F., Machado J., Bertoni G., Seow H.F., Maslak D.M., Schmerr M.J., Antigenic relatedness Dobbelaere D.A.E., Peterhans E., Caprine between ovine progressive pneumonia virus Arthritis-Encephalitis virus dysregulates the (OPPV) and HIV-1, Comp. Immun. Microbiol. expression of cytokines in macrophages, J. Infect. Dis. 16 (1993) 103-111.1 . Virol. 71 (1997a)7488-7497. Mazarin V., Gourdou I., Qu6rat G., Sauze N., Audoly Lechner F., Vogt H.R., Seow H.F., Bertoni G., G., Vitu C., Russo P., Rousselot C., Filippi P., Cheevers W.P., Von Bodungen U., Zurbriggen Vigne R., Subcellular localization of rev-gene A., Peterhans E., Expression of cytokine mRNA product in visna virus-infected cells, Virology in lentivirus-induced arthritis, Am. J. Pathol. 178 (1990) 305-310. 151 (l997b) 1053-1065. McGuire T.C., Adams D.S., Johnson G.C., Klevjer- Legastelois I., Cordier G., Cozon G., Cador6 J.L., Anderson P., Barbee D.D., Gorham J.R., Acute Guiguen F., Greenland T., Mornex J.F., Visna- arthritis in caprine arthritis-encephlitis virus maedi virus-induced expression of interleukin- challenge exposure of vaccinated or persis- Am. Vet. Res. 47 8 gene in sheep alveolar cells following exper- tently infected goats, J. (1986) imental in vitro and in vivo infection, Res. Virol. 537-540. 147 (1996) 191-197. Montelaro R.C., Ball J., Rwambo P., Issel C.J., Anti- Lena P., Freyria A.M., Lyon M., Cador6 J.L., genic variation during persistent lentivirus Guiguen F., Greenland T., Belleville J., Cordier infections and its implications for vaccine G., Mornex J.F., Increased expression of tis- development, Adv. Exp. Med. Biol. 251 (1989) 251-272. sue factor mRNA and procoagulant activity in ovine lentivirus-infected alveolar macrophages, Mornex J.F., Lena P., Loire R., Cozon G., Green- Res. Virol. 145 (1994) 209-214. land T., Guiguen F., Jacquier M.F., Cordier G., Lentivirus-induced interstitial disease: Leroux C., Vuillermoz S., Momex J.F., Greenland T., lung in Genomic in the of pulmonary pathology sheep naturally heterogeneity pol region infected the visna-maedi Vet. Res. 25 ovine lentiviruses obtained from bronchoalve- by virus, 478-488. olar cells of infected sheep from France, J. Gen. (1994) Virol. 76 (1995) 1533-1537. Narayan 0., Cork L.C., Lentiviral diseases of sheep and chronic leukoen- Leroux Greenland Mornex Molecular goats: pneumonia, C., T., J.F., and arthritis, Rev. Infect. Dis. characterization of field isolates of lentiviruses cephalomyelitis 7 (1985) 89-98. of small ruminants, AIDS Res. Hum. Retro- viruses 12 (1996) 427-429. Narayan 0., Wolinsky J.S., Clements J.E., Stand- berg J.D., Griffin D.E., Cork L.C., Slow virus Leroux Lerondelle C., J., Mornex J.F., C., Chastang replication: the role of macrophages in the per- RT-PCR detection of lentiviruses in milk or sistence and expression of visna viruses of secretions of or from mammary sheep goats sheep and goats, J. Gen. Virol. 59 (1982) infected flocks, Vet. Res. 28 (1997) 115-121. 345-356. Lichtenstein D.L., Rushlow K.E., Cook R.F., Raabe Narayan 0., Sheffer D., Clements J.E., Tennekoon M.L., Swardson C.J., Kociba G.J., Issel C.J., G., Restricted replication of lentiviruses. Visna Montelaro in vitro and in vivo R.C., Replication viruses induce a unique interferon during inter- of an equine infectious anemia virus mutant action between lymphocytes and infected deficient in dUTPase activity, J. Virol. 69 macrophages, J. Exp. Med. 162 (1985) (1997) 2881-2888. 1954-1969. Lujan L., Garcia Marin J.F., De Luco D.F., Vargas Narayan 0., Clements J.E., Kennedy-Stoskopf S., A., Badiola J.J., Pathological changes in the Sheffer D., Royal W., Mechanisms of escape of lung and mammary glands of sheep and their visna lentiviruses from immunological control, relationship with maedi-visna infection, Vet. Contrib. Microbiol. Immunol. 8 (1987) 60-76. Rec. 129 (1991) 51-54. Neuveut C., Vigne R., Clements J.E., Sire J., The Lutley R., Petursson G., Palsson P.A., Georgsson visna transcriptional activator TAT: effects on G., Klein J.R., Nathanson N., Antigenic drift the viral LTR and on cellular genes, Virology in visna: virus variation during long-term infec- 197(1993) 236-244. Palmarini M., Cousens C., Dalziel R.G., Bai J., Sted- tent lentiviruses naturally present in sheep with man K., DeMartini J.C., Sharp J.M., The exoge- progressive pneumonia are genetically distinct, nous form of jaagsiekte retrovirus is specifi- J. Virol. 52 (1984) 672-679. cally associated with a contagious lung cancer Quérat G., Barban V., Sauze N., Vigne R., Payne of sheep, J. Virol. 70 (1996) 1618-1623. A., York D.F., De Villiers E.M., Verwoerd Palsson P.A., Maedi-visna in sheep, in: Kimberlin D.W., Characteristics of a novel lentivirus R.H. (Ed.), Slow Virus Diseases of Animal and derived from South African sheep with pul- Man, Amsterdam, 1976, pp. 17-43. monary adenocarcinoma (jaagsiekte), Virol- Pancino G., Ellerbrok H., Sitbon M., Sonigo P., Con- ogy 158 (1987) 158-167. served framework of envelope glycoproteins Qu6rat G., Audoly G., Sonigo P., Vigne R., among lentiviruses, Curr. Top. Microbiol. Nucleotide sequence analysis of SA-OMVV, Immunol. 188 (1994) 77-105. a visna-related ovine lentivirus: phylogenetic Pearson L.D., Poss M.L., DeMartini J.C., Animal history of lentiviruses, Virology 175 (1990) lentivirus vaccines: problems and prospects, 434!47. Vet. Immunol. Immunopathol. 20 (1989) Reyburn H.T., Roy D.J., Blacklaws B.A., Sargan 183-212. D.R., McConnell L, Expression of maedi-visna Pekelder J.J., Maedi-visna virus induced indurative virus major core protein, p25: development of lymphocytic mastitis and its effect on prewean- a sensitive p25 antigen detection assay, J. Virol. ing growth of lambs, Sheep Vet. Sci. Proc. 188 Methods 37 (1992) 305-320. (1993)53-56. Rimstad E., East N.E., Torten M., Higgins J., DeRock Peluso R., Haase A.T., Stowring L., Edwards M., E., Pedersen N.C., Delayed seroconversion fol- Ventura P., A trojan horse mechanism for the lowing naturally acquired caprine arthritis- spread of visna virus in monocytes, Virology encephalitis virus infection in goats, Am. J. 147 (1985) 231-236. Vet. Res. 54 (1993) 1858-1862. Perk K., Yaniv A., Gazit A., DeMartini J.C., Evalu- Rosati S., Kwang J., Tolari F., Keen J.E., A com- ation of vaccines for ovine lentivirus infection, parison of whole virus and recombinant trans- AIDS Res. Hum. Retroviruses 12 (1996) membrane ELISA and immunodiffusion for 425-426. detection of ovine lentivirus antibodies in Ital- ian Vet. Res. Commun. 18 L.L., Wilkerson M.J., G.A., Cheev- sheep flocks, (1994) Perry Hullinger 73-80. ers W.P., Depressed CD4+ T lymphocyte pro- liferative response and enhanced antibody Rosati S., Kwang J., Keen J.E., Genome analysis of response to viral antigen in chronic lentivirus- North American small ruminant lentiviruses induced arthritis, J. Infect. Dis. 171 (1995) by polymerase chain reaction and restriction 328-334. enzyme analysis, J. Vet. Diagn. Invest. 7 (1995) Petursson G., Turelli P., Matthiasdottir S., Georgsson 437-443. G., Andresson O.S., Torsteinsdottir S., Vigne Roy D.J., Watt N.J., Ingman T., Houwers D.J., Sar- R., Andresdottir V., Gunnarsson E., Agnars- gan D.R., McConnell I., A simplified method dottir G., Qu6rat G., Visna virus dUTPase is for the detection of maedi-visna virus RNA by dispensable for neuropathogenicity, J. Virol. in situ hybridization, J. Virol. Methods 36 72 (1998) 1657-1661.1 . (1992) 1-11. ’ Phelps S.L., Smith M.C., Caprine Arthritis- Ruff G., Regli J.G., Lazary S., Occurence of caprine Encephalitis virus infection, J. Am. Vet. Med. leucocyte class I and II antigens in Saanen goats Assoc. 203 (1993) 1663-1666. affected by caprine arthritis encephalitis (CAE), Philippon V., Vellutini C., Gambarelli D., Harkiss G., Eur. J. Immunogenet. 20 (1993) 285-288. Arbuthnott G., Metzger D., Roubin R., Filippi Russo P., Giauffret A., Lasserre M., Sarrazin C., P., The basic domain of the lentiviral tat protein Isolement et 6tude d’une souche de virus visna- is responsible for damages in mouse brain: maedi chez le mouton en France, Bull. Acad. involvement of cytokines, Virology 205 (1994) V6t. France 53 (1980) 287-293. 519-529. Russo P., Vitu C., Vigne R., Filippi P., Giauffret A., Power C., Richardson S., Briscoe M., Pasick J., Eval- Recherches exp6rimentales sur le maedi-visna, uation of two recombinant maedi-visna virus Comp. Immunol. Microbiol. Infect. Dis. 111 proteins for use in an enzyme-linked (1988) 35-41. immunosorbent assay for the detection of serum Russo P., Vitu C., Guiguen F., La maladie maedi- antibodies to ovine Clin. lentiviruses, Diagn. visna du mouton: revue et perspectives, Point Lab. Immunol. 2 (1995) 631-633. V6t. 23 (1991) 33-38. Pritchard G.C., Done S.H., Dawson M., Multiple Russo P., Vitu C., Fontaine J.J., Vignoni M., Arthrite- cases of Maedi and Visna in a flock in East- encéphalite caprine: essai d’une preparation Anglia, Vet. Rec. 137 (1995) 443. vaccinale adjuv6e. I. Etude clinique et Quérat G., Barban V., Sauze N., Filippi P., Vigne virologique, Comp. Immunol. Microbiol. Infect. R., Russo P., Vitu C., Highly lytic and persis- Dis. 16 (1993) 131-136. Russo P., Vitu C., Bourgogne A., Vignoni M.. Stormann K.D., Schlecht M.C., Pfaff E., Compara- Abadie G., David V., Pepin M., Caprine arthri- tive studies of bacterially expressed integrase tis-encephalitis virus: detection of proviral DNA proteins of caprine arthritis-encephalitis virus, in lactoserum cells, Vet. Rec. 140 (1997) maedi-visna virus and human immunodefi- 483-484. ciency virus type I, J. Gen. Virol. 76 (1995) Saltarelli M.J., Schoborg R.V., Gdovin S.L., 1651-1663. Clements J.E., The CAEV tat gene trans-acti- Sutton K.A., Lin C.T., Harkiss G.D., McConnell I., vates the viral LTR and is necessary for effi- Sargan D.R., Regulation of the long terminal cient viral replication, Virology 197 (1993) repeat in visna virus by a transcription factor 35-44. related to the AML/PEBP2/CBF superfamily, Sargan D.R., Bennct I.D., Cousens C., Roy D.J., Virology 229 (1997) 240-250. Blacklaws B.A., Dalziel R.G., Watt N.J., Thormar H., Barshatzky M.R., Arnesen K., McConnell I., Nucleotide sequence of EV I, a Kozlowski P.B., The emergence of antigenic British isolate of maedi-visna virus, J. Gen. variants is a rare event in long-term visna virus Virol. 72 (1991) 1893-1903. infection in vivo, J. Gen. Virol. 64 (1983) Sargan D.R., Sutton K.A., Bennet I.D., McConncll I., 1427-1432. Harkiss G.D., Sequence and repeat structure Thormar H., Georgsson G., Palsson P.A., Balzarini variants in the Long Terminal Repeat of maedi- J., Naesens L., Torsteinsdottir S., De Clercq visna virus EV 1, Virology 208 (1995) 343-348. E., Inhibitory effect of 9-(2-phosphonyl- Schaller P., Zanoni R.G., Vogt H.R., Strasser M., methoxyethyl) adenine on visna virus infec- Peterhans E., Infections with maedi-visna and tion in lambs: a model for iii vivo testing of Border Disease viruses in Switzerland: a cross- candidate anti-human immunodeficiency virus sectional study of seroprevalciice and associated drugs, Proc. Natl. Acad. Sci. USA 92 (1995) factors, Immunobiology of Viral Infections, 3283-3287. Proc. 3rd congress ( 1995) 365-370. Toohcy K.L., Haase A.T., The rev gene of visna virus is for Virol- Sigurdsson B., Grimsson H., Palsson P.A., Maedi, a required productive infection, chronic progressive infection of sheep lungs, ogy 200 (1994) 276-280. J. Infect. Dis. 90 (1952) 233-241.1 . Torfason E.G., Gudnadottir M., Love A., Compari- Sigurdsson B., Palsson P.A., Grimsson H., Visna, a son of immunoblots with neutralizing and com- demyelinating transmissiblc disease of sheep, J. plement fixing antibodies in experimental and Neuropathol. Exp. Neurol. 16 (1957) 389-403. natural cases of visna-maedi, Arch. V irol. 123 ( I 992) 47-58. Sihvonen L., Studies on transmission of macdi virus to lambs, Acta Vet. Scand. 21 ( 1980) 689-698. Turelli P., Petursson G., Guiguen F., Mornex J.F., of Simon J.H., Fouchier R.A., T.E., Guerra Vigne R., Qu6rat G.. Replication properties Southcrling dUTPase-deficient mutants of and ovine C.B., Grant C.K., Malim M.H., The vif and caprine lentiviruses, J. Virol. 70 (1996) 1213-1217. gag proteins of human immunodeficiency virus type I colocalize in infected human T cells, J. Turelli P., Guiguen F., Momex J.F., Vigne R., Qu6rat Virol. 71 (1997) 5259-5267. G., dUTPase-minus caprine arthritis-encephali- tis virus is attenuated for and S.P., DeMartini J.C., E., Caletti E., pathogenesis Snyder Ameghino accumulates G-to-A J. Virol. 711 Coexistence of pulmonary adenomatosis and substitutions, progressive pneumonia in sheep in the central (1997)4522-4530. sierra of Peru, Am. J. Vet. Res. 44 ( 1983) Valas S., Benoit C., Guionaud C., Perrin G., Mamoun 1334-1338. R.Z., North American and French caprine viruses from ovine Sonigo P., Alizon M., Staskus K., Klatzmann D., arthritis-encephalitis emerge Cole S., Danos 0., Retzel E., Tiollais P., Haase maedi-visna viruses, Virology 237 (1997) 307-318. A.T., Wain-Hobson S., Nucleotide sequence of the visna lentivirus: relationship to the AIDS Vellutini C., Philippon V., Gambarelli D., virus, Ccll 42 (1985) 369-382. Horschowski N., Armin-Nave K., Navarro Staskus K.A., Retzel E., Lewis E.D., Silsby J.L., J.M., Auphan M., Courcoul M.A., Filippi P., StCyr S., Rank J.M., Wictgrefe S.W., Haasc The maedi-visna virus Tat protein induces mul- A.T., Cook R., Fast D., Geiser P.T., Harty J.T., tiorgan lymphoid hyperplasia in transgenic Kong S.H., Lahti C.J., Neufeld T.P., Porter mice, J. Virol. 68 (1994) 4955!962. T.E., Shoop E., Zachow K.R., Isolation of repli- Vitu C., Russo P., Filippi P., Vigne R., Qu6rat G., cation-competent molecular clones of visna Giauffret A., Une technique Elisa pour la d6tec- virus, Virology 181 (1991) 228-240. tion des anticorps anti-virus maedi-visna. E‘tude Steagall W.K., Robek M.D., Perry S.T., Fuller F.J., comparative avec I’immunoditfusion en g6lose Payne S.L., Incorporation of uracil into viral et la fixation du complement, Comp. Immunol. DNA correlates with reduced replication of Microbiol. Infect. Dis. 5 (1982) 469-481.1. EIAV in macrophages, Virology 210 (1995) Vitu C., Russo P., Vignoni M., Arthrite-enc6phalite 302-313. caprine: essai d’une preparation vaccinale adju- vee. II. Etude de la r6ponse anticorps, Comp. and B retrovirus of sheep and goats, J. Virol. 66 Immunol. Microbiol. Infect. Dis. 16 (1993) (1992) 4930!I939. 137-144. Zanoni R., Krieg A., Peterhans E., Detection of anti- Vitu C., Russo P., Bourgogne A., Vignoni M., Suzan bodies to caprine arthritis-encephalitis virus by M., Vigne R., Qu6rat G., Harmache A., P6pin Protein G enzyme-linked immunosorbent assay M., PCR applied to diagnosis of CAEV and and immunoblotting, J. Clin. Microbiol. 27 maedi-visna: evaluation and application to goat (1989)580-582. lactoserum, 3rd European Workshop on Ovine Zanoni R.G., Pauli U., Peterhans E., Caprine arthri- and Caprine Retroviruses, Jaca, Spain, 2-5 tis-encephalitis (CAE) and Maedi-Visna viruses March, 1997. detected by polymerase chain reaction (PCR), Vorster J.H., Dungu B., Marais L.C., York D.F., Vet. Microbiol. 23 (1990a) 329-335. Williams R., Boshoff C.H., A perspective on Zanoni R.G., Pauli U., Peterhans E., Detection of Maedi-Visna in South Africa, J. South Afr. Vet. caprine arthritis-encephalitis and maedi-visna Assoc. 67 (1996) 2-7. viruses using the polymerase chain reaction, 46 316-319. Watt N.J., Scott P., Collie D.D.S., Maedi-visna virus Experientia (1990b) infection in practice, In Practice September Zanoni R.G., Nauta LM., Kuhnert P., Pauli U., Pohl (1994) 239-247. B., Peterhans E., Genomic heterogeneity of small ruminant lentiviruses detected PCR, Willett B.J., Hosie M.J., Neil J.C., Turner J.D., Hoxie by Vet. Microbiol. 33 (1992) 341-351.1. J.A., Common mechanism of infection by lentiviruses, Nature 385 (1997) 587. Zanoni R.G., Vogt H.R., Pohl B., Bottcher J., Bom- meli W., Peterhans E., An ELISA based on Winward Leendertsen Shen Microim- L.D., L., D.T., whole virus for the detection of antibodies to test for of ovine munodiffusion diagnosis pro- small ruminant lentiviruses, J. Vet. Med. B. 41 gressive pneumonia, Am. J. Vet. Res. 40 (1979) (1994)662-669. 564. Zanoni R.G., Cordano P., Nauta I.M., Peterhans E., Woodall C.J., McLaren L.J., Watt N.J., Differential PCR for the detection of small ruminant levels of mRNAs for cytokines, the interleukin- lentiviruses, Schweizer Archiv Fur Tier- 2 receptor and class II DR/DQ genes in ovine heilkunde 138 (1996) 93-98. interstitial induced maedi-visna pneumonia by inter- virus infection, Vet. Pathol. 34 (1997) 204-211. Zink M.C., Narayan 0., Lentivirus-induced feron inhibits maturation and proliferation of Woodward T.M., Carlson J.O., McClelland C., monocytes and restricts the replication of DeMartini J.C., Analysis of lentiviral genomic caprine arthritis-encephalitis virus, J. Virol. 63 variation by denaturing gradient gel elec- (1989) 2578-2584. 17 (1994) 366-371.1. trophoresis, BioTechniques Zink M.C., Johnson L.K., Pathobiology of lentivirus Xu Z.Z., Hyatt A., Boyle D.B., Both G.W., Con- infections of sheep and goats, Virus Res. 32 struction of ovine adenovirus recombinants by (1994)139-154. or of related terminal gene insertion deletion Zink M.C., J.D., of 62-71.1. Yager J.A., Myers Pathogenesis region sequences, Virology 230 (1997) caprine arthritis encephalitis virus. Cellular York D.F., Vigne R., Verwoerd D.W., Qu6rat G., localization of viral transcripts in tissues of Nucleotide sequence of the jaagsiekte retro- infected goats, Am. J. Pathol. 136 (1990) virus, an exogenous and endogenous type D 843-854.