<<

Bovine immunodeficiency : facts and questions Catherine Belloc, Bruno Polack, I. Schwartz Cornil, J. Brownlie, Danielle Levy

To cite this version:

Catherine Belloc, Bruno Polack, I. Schwartz Cornil, J. Brownlie, Danielle Levy. Bovine immunodefi- ciency virus : facts and questions. Veterinary Research, BioMed Central, 1996, 27 (4-5), pp.395-402. ￿hal-02688068￿

HAL Id: hal-02688068 https://hal.inrae.fr/hal-02688068 Submitted on 1 Jun 2020

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

Bovine immunodeficiency virus: facts and questions

C Belloc B Polack I Schwartz-Cornil J Brownlie D Lévy

1 Unité de recherche associée d’immunopathologie cellulaire et moléculaire, Inra, École nationale vétérinaire dAlfort, 7, avenue du Général-de-Gaulle, 94704 Maisons-Alfort cedex, France; 2 Royal Veterinary College, London, UK

(Received 28 November 1995; accepted 29 March 1996)

Summary ― Bovine immunodeficiency virus (BIV) is a whose serologic prevalence is worldwide. Little is known about its impact on animal health status, pathogenesis and mode of transmission. Understanding BIV biology implies isolation of new viral strains and long-term studies on experimentally-infected cows and surrogate hosts such as rabbits. bovine immunodeficiency virus / / review / lentivirus

Résumé ― Le virus de l’immunodéficience bovine : faits et interrogations. Le virus de l’immunodéficience bovine (BIV) est un lentivirus dont la séroprévalence est mondiale. Actuellement, on connaît mal ses conséquences en matière de santé animale ainsi que son pouvoir pathogène ou ses modes de transmission. Afin de répondre aux différentes questions concernant le BIV, il est nécessaire d’isoler de nouvelles souches virales et de réaliser un suivi prolongé d’animaux infectés expérimentalement. virus de l’immunodéficience bo vine l bovin / revue générale l lentivirus

.!--- * Correspondence and reprints INTRODUCTION signs suggesting bovine leukosis (Malmquist et al, 1969; Van der Maaten et al, 1972). It was first designated ’bovine visna-like virus’ The bovine immunodeficiency virus (BIV) and remained unstudied until HIV was dis- is a lentivirus and one of the three morpho- covered, which generated renewed interest biologically and genetically distinct logically, in the study of animal . BIV was syncytium-inducing infecting further characterized using molecular biology bovines. Retroviruses are a of RNA family techniques and two proviral infectious clones that are for both man pathogenic were sequenced: BIV R-29 106 and BIV R- and animals. Chronologically, BIV was the 29 127 (Gonda et al, 1987; Garvey et al, third animal lentivirus to be discovered 1990). To date, all serologic information infec- among a group of eight, after equine relating to the presence of a BIV infection tious anemia virus (Vallée and Carr6, 1904) has been obtained using the original R-29 and ovine visna-maedi virus (Sigurdss6n et isolate as the antigen. Only recently have al, 1960). However the role of BIV as a new field isolates become available in the potential pathogen as well as its actual USA (Suarez et al, 1993). in remain prevalence, especially Europe, A new bovine lentivirus, the Jembrana unknown. Our here is to totally purpose disease virus (JDV), associated with a and review the epidemiological, pathological severe acute syndrome in Bali cattle (Bos molecular knowledge concerning BIV. javanicus), has been found to be both anti- genically and genetically closely related to BIV (Kertayadnya et al, 1993; Chadwick et HISTORICAL BACKGROUND al, 1995).

BIV was discovered in 1969 during the inten- PREVALENCE OF BIV INFECTION sive search for the causative agent of enzootic bovine leukosis. A virus with the morphology of a lentivirus was isolated from Serologic evidence for BIV infection has R-29, a pregnant dairy cow with clinical been reported in many countries around the world following a non-uniform distribution. infectious agents. Nevertheless, cow R-29 The prevalence of BIV infection is 4% in the had evidence of , lym- southern and southwestern states of the phadenopathy, central nervous system USA (Black, 1989) and average frequen- lesions and emaciation (Van der Maaten et al, cies of 64% and 40% have been reported in 1972). Moreover, hematological changes, the Louisiana area within dairy and beef lymphadenopathy with follicular hyperplasia, herds respectively. In Canada and the skin lesions unresponsive to treatment and Netherlands, the prevalence is 5.5% and meningoencephalitis have been found in nat- 1.4% (McNab et al, 1994; Horzinek et ai, urally- and experimentally-infected cattle 1991 ). In France, we have found that 4% (Braun et al, 1988; Martin et al, 1991; Car- of selected cattle tested positive for BIV penter et al, 1992; Onuma et al, 1992; Flam- (Polack et al, 1996). We also observed a ing et al, 1993; Gonda et al, 1994; Rovid et al, higher prevalence in dairy than in beef herds 1995). In these animals, the virus is tran- which may be the result of herd manage- scriptionally active and can be isolated for ment practices and of the extended pro- many years after infection (Brownlie et al, ductive life of dairy relative to beef cattle. 1994; Baron et al, 1995). Jembrana disease Seropositive cattle have also been detected virus is highly pathogenic in B javanicus in Great Britain (Brownlie et al, 1994; Howie whereas it does not induce any disease in B et al, 1994), Switzerland, Australia (Forman taurus. The parameters determining this dif- et al, 1992), Costa-Rica, Venezuela (in ferential pathology remain to be discovered. Gonda et al, 1994) and Brazil (unpublished Obviously, the BIV strains, bovine breed and personal results). Since the European sera the environment are factors that may influ- react weakly with the R-29 antigen, local ence susceptibility to BIV disease. Long-term isolates may be antigenically different from studies are thus necessary to observe BIV the American ones (Horzinek et al, 1991; pathogenicity since current opinions are Polack et al, 1996). Consequently, wild-type based on a limited number of short-term isolates are also needed in order to develop experimental studies. Finally, lentiviruses specific detection assays based on anti- exhibit variable virulence according to iso- bodies or nucleic acids. lates. Isolation of new BIV variants is neces- sary in order to appreciate its pathogenesis.

CLINICAL AND PATHOLOGICAL FEATURES HOST TROPISM

Lentivi ruses are responsible for a persistent In vivo, bovines are the major naturally- lifelong infection despite inducing a strong infected animals. Antibodies against BIV immune response. The onset of the disease have been reported in sheep and goats, is preceded by a several-year incubation however, without successful isolation of the period. It affects multiple organs and is char- virus or evidence of viral DNA (Whestone acterized by progressive debilitation, which, et al, 1991; Jacobs et al, 1994). Rabbits in the case of HIV, leads to death. The sig- experimentally-infected with BIV exhibit alter- nificance of BIV infection on the health status ations in their immune response (Onuma et of field herds has not been clearly estab- al, 1990; Pifat et al, 1992; Van der Maaten lished due to the high turnover rate of pro- and Whestone, 1992; Archambault et al, duction animals. It is not known whether BIV 1993; Hirai et al, 1994). The virus can be induces a specific syndrome or whether it isolated from the peripheral blood mononu- renders animals more susceptible to other clear cells, spleen, lymph nodes and brain throughout the life time of the infected ani- VIRUS BIOLOGY mals. In transgenic mice, BIV induces a meningoencephalitis associated with an BIV infection early 50% mortality (Gonda et al, 1994). cycle Such neurological syndromes have been reported with HIV-1 transgenic mice BIV particles consist of two positive-sense (Leonard et al, 1988). single-stranded viral RNAs and have a struc- tural capsid which envelops proteins. Mice, rats and guinea pigs are not sus- ceptible to BIV infection (Gonda, 1992). BIV is an exogenous with a replication cycle similar to that of other lentiviruses. Free virus particles attach to CELL TROPISM specific surface receptors and penetrate into the cell. The viral RNA is then released and converted by a viral reverse transcriptase In vitro, BIV is able to infect a broad range of into double-stranded DNA that is integrated adherent and cells suspension originating into the cellular genome, a step that involves from different tissues and different animal a viral integrase. This integrated species (Gonda et al, 1990; Gonda, 1992). remains inactive unless appropriate cellular In fibroblast-like BIV induces a cells, cyto- signals occur. When activated, genomic and effect characterized for- pathic by syncytium subgenomic messages are translated in the mation. Through analogy with other cytoplasm. Viral particle assembly and RNA lentiviruses, the in vivo targets of BIV infec- incorporation occur near the plasma mem- tion are likely to be the immune cells of the brane. Cellular machinery and viral proteins lymphoid and monocyte/macrophage lin- contribute to each step of this process. eages. BIV provirus can be detected by polymerase chain reaction from the periph- eral blood mononuclear cells, spleen, lymph Genomic organization nodes and brain (Pifat et al, 1992; Oberste et al, 1991 However, the cellular receptor The BIV genome (proviral DNA) is 8 960 for BIV has not yet been characterized. nucleotides long and contains the obliga- tory retroviral structural genes gag, pol and envflanked on the 5’ and 3’ termini by long BIV TRANSMISSION terminal repeats (LTR) (fig 1). Each LTR is composed of three consecutive regions named U3, R and U5. The U3 region con- BIV can be transmitted experimentally by tains the the intravenous inoculation of infected transcriptional regulatory sequences. The first base of the R region material cell-free or cell-associated (blood, is the site of the viral mRNAs transcription virus). initiation. In addition, at least five non-struc- The natural transmission mode for BIV tural open reading frames (ORF) are pre- is unknown and is supposed to be analo- sent in the ’central region’ between and gous to other lentiviruses that are spread overlapping the poland envORFs (Garvey by exchange of body fluids. The virus has et al, 1990). These ORFs are designated been observed in (Nash et al, 1995a) vif (viral infectivity factor), tat (trans-activa- and bull semen (Nash et al, 1995b). At pre- tor of transcription), rev (regulator of virus sent, nothing is known about in utero trans- expression) vpw, vpy and tmx. In terms of mission or the influence of age on an ani- genomic organization, BIV is the most com- mal’s susceptibility to infection. plex non-primate lentivirus. At least five alter- nately-spliced BIV-specific mRNAs have L TR and non-structural proteins been identified in infected cells by Northern blot et al, analysis (Oberste 1991).). The U3 region of viral LTR contains tran- scription-factor binding sites (NFkB, Spl, AP1, AP4) regulating viral replication and Viral structural proteins gene expression (Liu et ai, 1992; Pallansch et al, 1992; Carpenter et al, 1993; Fong et al, The Pr53 Gag precursor is synthesized first 1995). and is responsible for virus particle budding The Tat protein is found in the nucleus (Rasmussen et al, 1990). It is subsequently of infected cells where it transactivates viral cleaved a viral into the mature by protease LTR through a trans-activating region (TAR) and proteins p16 (matrix), p26 (capsid) p7 resulting in an enhancement of virus expres- (nucleocapsid) (Battles et al, 1992). The pol sion. The Rev protein colocalizes with Tat in are a an RNA- gene products protease, infected cells and presents the same positive DNA tran- dependent polymerase (reverse effect on viral expression (Oberste et al, scriptase) and an integrase. They are 1993). derived by proteolytic cleavage from a Pr170 The role of the other BIV accessory Gag-Pol precursor. A sequence in the pol is unknown. region is associated with dUTPase activity in genes presently other lentiviruses such as virus, feline immunodeficiency virus and caprine arthritis encephalitis virus. It Genetic diversity has an unknown function in the case of BIV (Garvey et al, 1990). The two proviral molecular clones (BIV 106 The virus envelope consists of a lipid and BIV127) were both derived from the bilayer derived from the plasma membrane original BIV R-29 field isolate. Their of infected cells during the budding process. sequences exhibit a genomic variability of The gp100 (SU) and gp45 (TM) encoded 1.7% with 75% of the substitutions occur- by the env gene of BIV are inserted into this ring in the SU-coding region of the env gene bilayer (Rasmussen et al, 1992). (Garvey et al, 1990). DNA sequence anal- ysis of new field isolates (Suarez et al, 1993) have demonstrated substantial genetic vari- ation (7-8% nucleotide divergence in the conserved pol segment). This genetic diver- sity has been observed in all lentiviruses and probably plays a role in helping them to escape immune selection pressures and to new hosts. The characterization of this genetic diversity is critical for the develop- ment of diagnostic tests and the under- standing of the viral pathogenic potential.

CONCLUSION

Bovine lentiviruses have been largely over- looked due to the absence of a test for serodetection and their unknown Does BIV have variable virulence? pathogenicity. The recent availability of a test based on the R-29 diagnostic antigen Due to the current low number of character- provided evidence of seropositive animals all ized BIV isolates, this question cannot be around the world. This leads to the following answered accurately. However, a spatial and questions. temporal variation of lentiviruses in infected hosts has been well documented. After virus infection, some sort of virus-purifying selec- What is the real pathogenic potential tion occurs the initial of of BIV? during phases repli- cation in the newly-infected host, followed by rediversification into quasi-species. The The reported clinical manifestations are the progression to disease is then determined following. by complex interactions between the host, intercurrent and the BIV-R29, initially isolated from a cow factors, increasingly with a wasting condition, was able to diverse viral population. It is not certain whether the viral as induce in inoculated calves a lymphopro- burgeoning quasi-species liferative reaction associated with neural a whole or specific virulent members of the disorders. In Florida (Suarez et al, 1993), quasi-species are most critical to the pro- two wild-type isolates were recovered from gression of this disease. However, accumu- a group of cattle with the clinical manifes- lating evidence from the oncovirus FeLV- tations of ’poor doers’. More recently, in FAIDS (Mullins et al, 1991) and from the Great Britain, some seropositive animals simian lentiviruses SIV Pbjl4 and SIV-Mac were found in a herd in which a few indi- 239 (Dewhurst et al, 1990; Burns and viduals exhibited a loss of weight and bron- Desrosiers, 1991) support the hypothesis cho-alveolar manifestations. However, evi- that particular virulent variants are essential dence is lacking for a clear association for disease induction. Therefore, by analogy, between BIV infection and the reported would it be safe to evaluate the pathogenic- clinical signs. ity of BIV after studying only one BIV isolate? Lentiviruses, apart from BIV, have been to clinical char- reported produce syndromes What is the distribution of BIV? acterized by a gradual and progressive debilitation, which in the case of HIV fre- quently leads to death. The incubation An understanding of BIV pathogenesis has period varies, but in most cases, it takes been hampered in part by sparse informa- several years for the disease to develop. tion on the worldwide prevalence of BIV Moreover, the devastating consequences infection. At present, all of the serologic of EIAV (equine infectious anemia virus), information on the presence of BIV infec- MVV (maedi visna virus), CAEV (caprine tions has been obtained using the original R- arthritis encephalitis virus) and HIV (human 29 isolate as antigen. Nonetheless, emerg- immunodeficiency virus) provide evidence ing data suggest that variants of BIV, which for the pathogenesis of lentiviruses in gen- are antigenically distinct from BIV R-29 and eral (Brownlie et al, 1994). By analogy, it only demonstrate immunologic cross-reac- would be exceptional if BIV were not to have tivity with the major capsid protein, are pre- a similar clinical syndrome, however it would sent in the eastern United States and be unrealistic to expect to see BIV patho- Europe. This limited information empha- genesis before the completion of a three to sizes the need to isolate new BIV isolates in five year incubation period. order to set up more specific diagnostic tests and to obtain a more accurate estimate of active of bovine immunodeficiency-like 515-523 BIV prevalence. virus. Virology 167, Brownlie J, Collins ME, Heaton P (1994) Bovine immuno- deficiency-like virus - a potential cause of disease in cattle? Vet Rec 134, 289-291 Is BIV a risk to human health? Burns DPW, Desrosiers RC (1991) Selection of genetic variants of simian immunodeficiency virus in persis- BIV and tently infected Rhesus monkeys. J Virol 65, 1843- Considerating biology phylogeny 1854 one may ask whether BIV enbodies a dan- S, Miller LD, Alexandersen S, et al (1992) health. Several data Carpenter ger for human support Characterization of early pathogenic effects after a negative answer. First, lentiviruses are experimental infection of calves with bovine immun- highly specific in their host tropism. More- odeficiency-like virus. J Virol66, 1074-1083 over, injuries with contaminated material Carpenter S, Nadin Davis SA, Wannemuehler Y, Roth JA (1993) Identification of transactivation-response have been without inducing sero- reported sequences in the long terminal repeat of bovine conversion. Finally, no cross-reactivity immunodeficiency-like virus J Viro167, 4399-4403 between sera from HIV-infected individuals Chadwick BJ, Coelen RJ, Wilcox GE, Sammels LM, and the BIV antigen was observed (Whet- Kertayadnya G (1995) Nucleotide sequence analy- stone et al, 1992). In conclusion, there is sis of Jembrana disease virus - a bovine lentivirus associated with an acute disease syndrome. J Gen no evidence to the currently suggest pos- Viro176, 1637-1650 of cross contamination. sibility Dewhurst S, Embertson JE, Anderson DC, Mullins JI, Finally, BIV contains some peculiarities Fultz PN (1990) Sequence analysis and acute of that the need to increase coor- pathogenicity molecularly-cloned SIVSMM-PBj1 4. emphasize Nature 345, 636-640 dinated studies between practitioners and Flaming KM, Van der Maaten M, Whetstone C, Car- scientists in order to better understand the penter S, Frank D, Roth J (1993) Effect of bovine biology of this lentivirus. immunodeficiency-like virus infection on immune function in experimentally infected cattle. Vet Immunol Immunopathol36, 91-105 ACKNOWLEDGMENT Fong SE, Pallansch LA, Mikovits JA, Lackmansmith CS, Ruscetti F W, Gonda MA (1995) Cis-acting regulatory elements in the bovine immunodeficiency virus long terminal repeat. Virology 209, 604-6144 This work was partly supported by grant 92 5111 from the French Ministry of Agriculture. Forman AJ, Gibson CA, Rodwell BJ (1992) Serological evidence for the presence of bovine lentivirus infec- tion in cattle in Australia. Aust Vet J 69, 337 Oberste Elser JE, Braun MJ, Gonda REFERENCES Garvey KJ, MS, MA (1990) Nucleotide sequence and genome orga- nization of biologically active proviruses of the bovine Archambault D, Nadin Davis S, Lutze Wallace C, Bouillant immunodeficiency-like virus. Virology 175, 391-409 AMP (1993) Le virus de l’immunod6ficience bovine : Gonda MA (1992) Bovine immunodeficiency virus [edi- mise au point 1990-1992. Vet Res 24, 179-187 torial]. AIDS 6, 759-776 Baron T, Mallet F, Polack B, Betemps D, Belli P (1995) Gonda MA, Braun MJ, Carter SG et al (1987) Charac- The bovine immunodeficiency-like virus (BIV) is tran- terization and molecular cloning of a bovine lentivirus scriptionally active in experimentally infected calves. related to human immunodeficiency virus. Nature Arch Viro1140, 1461-1467 330, 388-391 Battles JK, Hu MY, Rasmussen L, Tobin GJ, Gonda MA Gonda MA, Oberste MS, Garvey KJ et al (1990) Devel- (1992) Immunological characterization of the gag opment of the bovine immunodeficiency-like virus gene products of bovine immunodeficiency virus. J as a model of lentivirus disease. Dev Biol Stand 72, Virol 66, 6868-6877 97-1100 Black JW (1989) Report of the committee on bluetongue Gonda MA, Gene Luther D, Fong SE, Tobin GJ (1994) and bovine retrovirus Proc U S Anim Health Assoc Bovine immunodeficiency virus molecular biology 93, 145-152 and virus-host interactions. Virus Res 32, 155-181 Braun MJ, Lahn S, Boyd AL, Kost TA, Nagashima K, Hirai NS, Xuan W, Ochiai K, Onuma M (1994) Alteration Gonda MA (1988) Molecular cloning of biologically of immune responses of rabbits infected with bovine immunodeficiency-like virus. Microbiot Immunol38, Onuma M, Wada M, Yasutomi Y, Yamamoto M, Okada H 943-950 M, Kawakami Y (1990) Suppression of immunological Horzinek M, Keldermans L, Stuurman T et al (1991) responses in rabbits experimentally infected with Bovine immunodeficiency virus immunochemical bovine virus. Vet Microbiol25, 131-141 characterization and serological survey. J Gen Virol Onuma M, Koomoto E, Furuyama H (1992) Infection 72, 2923-2928 and dysfunction of monocytes induced by experi- Howie NM, McKay IG, Davies AB, Brownlie J (1994) mental inoculation of calves with bovine immuno- virus. Immune Defic Problems in a Cheshire herd. Vet Rec 134, 3100 deficiency-like J Acquired Syndr 5, 1009-10155 Jacobs RM, Smith HE, Whetstone CA, Suarez DL, Jef- ferson B, Valli VEO (1994) Haematological and lym- Pallansch LA, Lackman Smith CS, Gonda MA (1992) phocyte subset analyses in sheep inoculated with Bovine immunodeficiency-like virus encodes factors bovine immunodeficiency-like virus. Vet Res Com- which transactivate the long terminal repeat. J Virol mun 18, 471-482 66,2647-2652 Kertayadnya G, Wilcox GE, Soeharsono S, et al (1993) Pifat DY, Ennis WH, Ward JM, Oberste MS, Gonda MA Characteristics of a retrovirus associated with Jem- (1992) Persistent infection of rabbits with bovine brana disease in Bali cattle. J Gen Virol74, 1765-1773 immunodeficiency-like virus. J Virol66, 4518-4524 Leonard JM, Abramczuk JW, Pezen DS, et al (1988) Polack B, Schwartz I, Berthelemy M, et al (1996) Sero- evidence for bovine virus Development of disease and virus recovery in trans- logic immunodeficiency genic mice containing HIV proviral DNA. Science infection in France. Vet Microbiol96, 165-173 242, 1665-1670 Rasmussen L, Battles JK, Ennis WH, Nagashima K, Liu ZQ, Sheridan D, Wood C (1992) Identification and Gonda MA (1990) Characterization of virus-like par- characterization of the bovine immunodeficiency- ticles produced by a recombinant baculovirus con- the of the bovine like virus tat gene. J Virol66, 5137-5140 taining gag gene immunodeficiency- like virus. Virology 178, 435-4511 Malmquist WA, Van Der Maaten MJ, Boothe AD (1969) Isolation immunodiffusion immunofluorescence and Rasmussen L, Greenwood JD, Gonda MA (1992) of bovine virus electron microscopy of a syncytial virus of lym- Expression immunodeficiency-like phosarcomatous and apparently normal cattle. Can- envelope glycoproteins by a recombinant baculovirus in insect cells. 186 551-5611 cer Res 29, 188-200 Virology Martin SJ, O’Neill TP, Bilello JA, Eiseman JL (1991) Rovid AH, Carpenter S, Roth JA (1995) Monocyte func- Lymphocyte transformation abnormalities in bovine tion in cattle experimentally infected with bovine immunodeficiency-like virus infected calves. Immunol immunodeficiency-like virus. Vet Immunol Lett27, 81-84 Immunopatho145, 31-43 McNab WB, Jacobs RM, Smith HE (1994) A serological Sigurdsson B, Thormar H, Palsson PA (1960) Cultivation of visna virus in tissue culture. Arch Gesamte survey for bovine immunodeficiency-like virus in Virus- Ontario dairy cattle and associations between test forsch 10, 368-381 results production records and management prac- Suarez DL, Van Der Maaten MJ, Wood C, Whetstone CA tices Can J Vet Res 58, 36-41 (1993) Isolation and characterization of new wild- Mullins JI, Hoover EA, Quackenbush SL, Donahue PR type isolates of bovine lentivirus. J Virol67, 5051- (1991) Disease progression and viral genome vari- 5055 ants in experimental -induced Vallee H, Carrb H (1904) Nature infectieuse de I’anbmie immunodeficiency syndrome. J Acquired Immune du cheval. CR Acad Sci 139, 331-333 Defic Syndr4, 547-557 Van Der Maaten MJ, Boothe AD, Seger CL (1972) Iso- Nash JW, Hanson LA, St Cyr Coats KS (1995a) Detec- lation of a virus from cattle with persistent lympho- tion of bovine immunodeficiency virus in blood and cytosis. J Natl Cancer Inst 49, 1649-1657 milk-derived use of chain leukocytes by polymerase Van Der Maaten MJ, Whetstone CA Infection of reaction. Am J Vet Res 56, 445-449 (1992) rabbits with bovine immunodeficiency-like virus. Vet Nash JW, Hanson LA, StCyr Coats KS (1995b) Bovine Microbio130, 125-135 virus in stud bull semen. Am J immunodeficiency Whetstone CA, Van Der Maaten MJ, Miller JM A Vet Res 56, 760-763 (1991) Western blot assay for the detection of antibodies Oberste MS, Greenwood JD, Gonda MA (1991) Analy- to bovine immunodeficiency-like virus in experi- sis of the transcription pattern and mapping of the mentally inoculated cattle sheep and goats. Arch putative rev and env splice junctions of bovine Virol116, 119-131 immunodeficiency-like virus. J Virol65, 3932-3937 Whetstone C A, Sayre K R, Dock N L, et al (1992) Exam- Oberste MS, Williamson JC, Greenwood JD, Nagashima ination of whether persistently indeterminate human K, Copeland TD, Gonda MA (1993) Characteriza- immunodeficiency virus type 1 Western immunoblot tion of bovine immunodeficiency virus rev cDNAs reactions are due to serological reactivity with bovine and identification and subcellular localization of the immunodeficiency-like virus. J Clin Microbiol 30, 764- Rev protein. J Virol67, 6395-6405 770