Diseases of Laboratory Primates PAT 707, Diseases of Laboratory Animals II Part 1: Viral Diseases Prepared by Trenton R. Schoeb Department of Comparative Medicine University of Alabama at Birmingham Winter Quarter 1989-90 Herpesvirus simiae 1. Historical: Originally reported in 1934 by Sabin and Wright, who isolated it from a man who died after being bitten by a rhesus monkey. 2. Agent: An alpha herpesvirus (same group as Herpesvirus hominis); syn. Herpes B. Antigenically similar to HSV-1, HSV-2, and SA-8. 3. Epizootiology: Macaques (especially rhesus) are natural hosts. Other species are experimentally susceptible. Transmission among monkeys by contact, fomites, aerosol; to man by bites, infected cells or tissues, aerosol. (Transmission to humans by exposure to monkey saliva via bites or scratches has not been consistently documented, however.) Probably monkeys are infected for life and a high proportion, if not 100%, of adult individuals in infected colonies harbor virus. Through 1987, 22 human cases were reported. (A few other cases are attributed to B virus infection, but lack adequate documentation.) 4. Clinical: In monkeys, lingual and labial vesicles which rupture leaving ulcers that heal in 1-2 weeks; conjunctival and cutaneous lesions in a few cases. Infection probably also occurs without lesions in many cases. In man, local erythema and vesiculation at site of inoculation, localized neurologic symptoms, fever, regional lymphadenopathy, muscular aches, CNS signs (mostly paralytic), die in coma of respiratory failure within a few days to a few weeks. Twenty of the 22 human patients developed encephalitis. Fifteen of these died. Two of the Pensacola patients had mild disease; they were treated with acyclovir but its efficacy for treating human infections has not been established. The frequency of mild or symptomatic human infections is not known. 5. Pathology: Microscopic lesions in man and monkeys are similar. "Ballooning" degeneration, multinucleation, and necrosis of epithelial and other affected cells and characteristic nuclear inclusions. In man, multifocal necrotizing encephalomyelitis and multifocal necrosis in spleen, liver, lymph nodes, and adrenals. A similar disseminated disease occurs rarely in rhesus and occasionally in cynomolgus monkeys. 6. Diagnosis: Signs, lesions, virus isolation, rabbit inoculation. Serologic tests including ELISA and serum neutralization also can be used; however, results of the usual tests can be complicated by antibodies to human herpes simplex virus and to SA-8 virus. (Competition ELISA can be used to distinguish, if necessary.) Furthermore, it is not certain that infected monkeys always have diagnostic titers. 7. Control: The CDC recommends that macaques be used for research only when necessary, and that, if possible, those used should be free of the virus and housed so as to prevent infection. Macaques should be handled only with adequate physical or chemical restraint, and handlers should wear protective clothing, gloves, and masks. The CDC does not recommend routine screening because the reliability of negative results has not been ascertained and because handling the monkeys increases the risk of human exposure. Monkeys with oral lesions suggestive of active infection should be quarantined. Handlers and investigators should be educated concerning the risks of exposure to B virus and the need for proper protective measures, immediate reporting and treatment of injuries received while handling monkeys, and obatining immediate medical attention for any condition suggestive of B virus infection. Limited experimental and clinical data indicate acyclovir to be the drug of choice for treatment. For consultation concerning diagnosis and treatment, one can contact the Viral Exanthems and Herpesvirus Branch, CDC, 404-329-1338, or the Southwest Foundation for Biomedical Research, 512-674-1410. 8. References: Balfour HH Jr. 1984. Acyclovir and other chemotherapy for herpes group viral infections. Ann Rev Med 35:27-291. Boulter EA et al. 1980. Successful treatment of B virus (Herpesvirus simiae) infection with acyclovir. Br Med J 280:681-683. Boulter EA et al. 1982. A comparison of neutralization tests for the detection of antibodies to Herpesvirus simiae (Monkey B virus) Lab Anim Sci 32:150-152. Bryan BL et al. 1975. Recovery from encephalomyelitis caused by Herpesvirus simiae. Arch Intern Med 135:868-870. Cabasso VJ et al. 1967. Correlation of B virus and herpes simplex virus antibodies in human sera. J Lab Clin Med 70:170-178. Centers for Disease Control. 1973. Herpes B encephalitis--California. Morbity and Mortality Weekly Report 22:333-334. Centers for Disease Control. 1987. B-virus infection in humans--Pensacola, Florida. Morbidity and Mortality Weekly Report 36:289-290. Centers for Disease Control. 1987. Guidelines for prevention of Herpesvirus simiae (B virus) infection in monkey handlers. Morbidity and Mortality Weekly Report 36:680- 689. DiGiacomo RF. 1988. Guidelines for prevention of herpesvirus simiae (B-virus) infection in monkey handlers [letter] Lab Anim Sci 38:558. Hartley EG. 1964. Naturally occurring B virus infection in cynomolgus monkeys. Vet Rec 76:555-557. Heberling RL, Kalter SS. 1987. A dot-immunobinding assay on nitrocellulose with psoralen inactivated Herpesvirus simiae (B virus). Lab Anim Sci 37:304-308. Hilliard JK et al. 1987. Herpesvirus simiae (B-virus): Replication of the virus and identification of viral polypeptides in infected cells. Arch Virol 93:185-198. Hilliard JK et al. 1986. Rapid identification of Herpesvirus simiae (B virus) DNA from clinical isolates in nonhuman primate colonies. J Virol Methods 13:55-62. Hull RN. 1971. B virus vaccine. Lab Anim Sci 21:1068-1071. Hummeler K et al. 1959. Encepahalomyelitis due to infection with Herpesvirus simiae (herpes B virus): a report of two fatal acquired cases. New Engl J Med 261:64-68. Juel-Jensen BE. 1982. Postexposure immunoprophylaxis against B virus infection. Brit Med J 284:113-114. Kaplan JE et al. 1987. Guidelines for prevention of Herpesvirus simiae (B-virus) infection in monkey handlers (letter). Lab Anim Sci 37:709-712. Kaplan JE. 1988. Herpesvirus simiae (B virus) infection in monkey handlers [letter] J Infect Dis 157:1090. Keeble SA. 1960. B virus infection in monkeys. Ann NY Acad Sci 85:960-969. Loomis MR et al. 1981. Fatal herpesvirus infection in patas monkeys and a black and white colobus monkey. J Am Vet Med Assoc 179:1236-1239. McClure HM et al. 1973. Disseminated herpesvirus infection in a rhesus monkey (Macaca mulatta). J Med Primatol 2:190-194. Palmer AE. 1987. B virus, Herpesvirus simiae: Historical perspective. J Med Primatol 16:99-130. Sabin AB, Wright AM. 1934. Acute ascending myelitis following a monkey bite with the isolation of a virus capable of reproducing the disease. J Exp Med 59:115-136. Van Hoosier GL Jr, Melnick JL. 1961. Neutralizing antibodies in human sera to Herpesvirus simiae. Texas Rep Biol Med 19:376-380. Vizoso AD. 1975. Recovery of Herpesvirus simiae (B virus) from both primary and latent infections in rhesus monkeys. Brit J Exp Pathol 56:485-488. Zwartouw HT et al. 1984. Transmission of B virus infection between monkeys especially in relation to breeding colonies. Lab Animal 18:125-130. Herpesvirus tamarinus 1. Agent: Syn. Herpes T, Herpesvirus saimiri type 1 2. Epizootiology: Natural host (life-long latent infection with periodic shedding) is squirrel monkey, perhaps also Cebus and Ateles spp. (serologic evidence). Severe disease in marmosets and owl monkeys. Transmission by contact, fomites, aerosol. 3. Clinical: In squirrel monkeys: usually none, but in acute disease can have lip, tongue, palate ulcers. Systemic disease and death not reported. In owl monkeys and marmosets, rapidly spreading epizootics with high morbidity and mortality (up to 100%), characterized by oral and cutaneous vesicles and ulcers, conjunctivitis, and occasionally, apparent hyperesthesia or paresthesia indicated by intensive scratching. 4. Pathology: Focal necrosis with giant cells and inclusions in skin, oral mucosa, gastrointestinal tract, lymph nodes, adrenals, and liver, and in some cases, encephalitis. (Microscopic lesions in the cases in our files do not contain giant cells.) 5. Diagnosis: Signs and lesions, virus isolation and identification. 6. Control: House reservoir and susceptible primates separately. 7. References: Desrosiers RC 1981. Herpesvirus tamarinus and its relation to Herpes simplex virus. J Gen Virol 56:119-130. Emmons RW et al. 1969. Natural fatal infection of an owl monkey with herpes-T virus. J Infect Dis 118:153-159. Hunt RD, Melendez LV. 1966. Spontaneous herpes-T infection in the owl monkey (Aotus trivirgatus). Path Vet 3:1-26. Morita M. 1981. Fatal Herpesvirus tamarinus infection in cotton-topped marmosets (Saguinus oedipus). Jap J Med Sci Biol 34:268-270. Sauer GM, Bishop W. 1963. Inclusion body hepatitis in marmosets. Lab Anim Care 13:790-792. Herpesvirus hominis 1. Agent: Herpesvirus hominis type 1 (herpes simplex) and type 2 (genital herpes). 2. Epizootiology: Man is the natural host; most people are infected. Generalized disease reported in owl monkeys, gibbons, tree shrews, and gorilla. Serologic evidence (ELISA + immunoblotting) indicates that HSV-1 infections are common in gorillas and gibbons. Marmosets and Cebus spp. are susceptible experimentally. Genital infections (HSV-2) have been reported in chimpanzees. 3. Clinical: In man, acute infection (usually in children less than 3 years old) produces vesicular gingivostomatitis. Recurrent exacerbations are characterized by vesicles and ulcers on the lips ("fever blisters"). Owl monkeys: high morbidity and mortality, conjunctivitis, nasal discharge, lethargy, course 4-7 days. Gibbons: oral ulcers; some develop CNS signs and die. Tree shrews: emaciation, conjunctivitis, some die. Generalized disease in a gorilla also has been reported. Genital herpes in chimps: localized herpetic ulcers. 4. Pathology: Morphologically indistinguishable from Herpesvirus tamarinus in owl monkeys, although encephalitis occurs in more cases. Gibbons: necrotizing meningoencephalitis, inclusions. Tree shrews: multifocal necrotizing and hemorrhagic hepatitis and adrenal adenitis with inclusions. 5. Diagnosis: Signs and lesions, virus isolation and identification. 6. Control: Persons with active herpetic infection must stay away from susceptible nonhuman primates. 7. References: Eberle R, Hilliard JK. 1989. Serological evidence for variation in the incidence of herpesvirus infections in different species of apes. J Clin Microbiol 27:1357-1366. Emmons RW, Lennette EH. 1970. Natural Herpesvirus hominis infection of a gibbon (Hylobates lar). Arch Ges Virusforsch 31:215-218. Heldstab A et al. 1981. Spontaneous generalized Herpesvirus hominis infection of a lowland gorilla (Gorilla gorilla gorilla). J Med Primatol 10:129-135. Kalter SS et al. 1972. Experimental Herpesvirus hominis type 2 infection in non-human primates. Proc Soc Exp Biol Med 139:964-968. Katzen S et al. 1971. Experimental Herpesvirus simplex infection in the owl monkey. Proc Soc Exp Biol Med 125:391-398. London WT et al. 1971. Genital Herpesvirus hominis type 2 infection of monkeys. Obstet Gynecol 35:501-509. McClure HM et al. 1972. Natural Herpesvirus hominis infection of tree shrews (Tupaia glis). Lab Anim Sci 22:517-521. McClure HM et al. 1980. Natural genital Herpesvirus hominis infection in chimpanzees (Pan troglodytes and Pan paniscus). Lab Anim Sci 30:895-901. Melendez LV et al. 1969. Natural Herpesvirus simplex infection in the owl monkey (Aotus trivirgatus). Lab Anim Care 19:38-45. Ramsey E et al. 1982. Fatal Herpesvirus hominis encephalitis in a white handed gibbon. J Am Vet Med Assoc 181:1429-1430. Smith PC et al. 1969. The gibbon (Hylobates lar): A new primate host for herpes virus hominis. I. A natural epizootic in a laboratory colony. J Infect Dis 120:292-297. Simian Varicella 1. Agent: (i) Delta herpesvirus (DHV, Syn. Patas monkey herpesvirus). First isolated in 1968 at Delta Regional Primate Research Center from patas monkeys. (ii) Liverpool vervet monkey herpesvirus (LVMHV). (iii) Medical Lake macaque herpesvirus (MLMHV). These viruses are very similar, if not identical, and are closely related to human varicella-zoster virus. 2. Epizootiology: Natural host of DHV is unknown. Most natural cases have been in patas monkeys, but disease in this species tends to feature high morbidity and mortality, which is more characteristic of herpesviral infection in aberrant or immunosuppressed hosts. African green monkeys and pigtailed macaques also are susceptible. LVMHV affects patas and African green monkeys; MLMHV, pigtailed, Japanese, and cynomolgus macaques. There has been a case of a varicella-like disease in a young chimp at the Yerkes center, but the virus was lost in culture and thus could not be identified. 3. Clinical: Cutaneous rash with vesicles, oral vesicles and ulcers, high mortality. MLMHV: lower mortality, often self-limiting exanthematous disease. 4. Pathology: Hyperplasia, "ballooning" degeneration, necrosis, blood-filled vesicles and ulceration in stratified squamous epithelium; multifocal to diffuse necrosis in most viscera, especially lung, liver, and lymphoid organs (may also be in digestive tract and aderenals); widespread necrotizing vasculitis; inclusions in epithelium, endothelium, and vascular smooth muscle of affected organs. 5. Diagnosis: Signs, lesions, virus isolation. Clinically and pathologically resembles generalized varicella in immunosuppressed human hosts. 6. Control: Uncertain--don't mix primate species. 7. References: Allen WP et al. 1974. Recent studies on the isolation and characterization of Delta herpesvirus. Lab Anim Sci 24:222-228. Ayers JP. 1971. Studies of the Delta herpesvirus isolated from the patas monkey (Erythrocebus patas). Lab Anim Sci 21:685-695. Blakely GA et al. 1973. A varicella-like disease in macaque monkeys. J Infect Dis 127:617-625. Clarkson MJ et al. 1967. A virus disease of captive vervet monkeys (Cercopithecus aethiops) caused by a new herpesvirus. Arch Ges Virusforsch 22:219-234. Felsenfeld A, Schmidt NJ. 1975. Immunological relationship between Delta herpesvirus of patas monkeys and varicella zoster virus of humans. Infect Immun 12:261-266. Eppstein DA et al. 1989. Liposomal interferon-beta: sustained release treatment of simian varicella virus infection in monkeys. J Infect Dis 159:616-620. Felsenfeld A, Schmidt WJ. 1979. Varicella zoster virus immunizes patas monkeys against varicella-like disease. J Gen Virol 42:171-178. Gard EA et al. 1983. Disseminated intravascular coagulation secondary to simian varicella in the patas monkey (Abstract). Lab Anim Sci 33:481. Gard EA. 1982. Symptomatology and pathology of delta herpesvirus infection in a patas monkey colony (Abstract). Lab Anim Sci 32:427-428. Harbour A. 1979. The serological relationship of varicella zoster virus to other primate herpesviruses. J Gen Virol 45:469-477. Heuschele WP. 1970. Varicella (chicken pox) in three young anthropoid apes. J Am Vet Med Assoc 136:256-257. Lehner NM et al. 1984. Simian varicella infection in the African green monkey (Cercopithecus aethiops). Lab Anim Sci 34:281-285. McCarthy K et al. 1968. Exanthematous disease in patas monkeys caused by herpes virus. Lancet 2:856-857. Padovan D, Cantrell CA. 1986. Varicella-like herpesvirus infections of nonhuman primates. Lab Anim Sci 36:7-13. Roberts E et al. 1984. Pathologic changes of experimental simian varicella (Delta herpesvirus) infection in African green monkeys (Cercopithecus aethiops). Am J Vet Res 45:523-530. Schmidt NJ et al. 1983. Serological investigation of an outbreak of simian varicella in Erythrocebus patas monkeys. J Clin Microbiol 18:901-904. Seibold HR, Allen WP. Varicella, Model No. 69. In Handbook of Animal Models of Human Disease. Fasc. 5, Ed TC Jones, DB Hackel and G Migaki. Registry of Comparative Pathology, AFIP, Washington DC, 1976. Soike KF, Baskin GB. Varicella, Model No. 69, Supplemental update. In Handbook: Animal Models of Human Disease, Fasc 12. Ed CC Capen, DB Hackel, TC Jones, and G Migaki. Registry of Comparative Pathology, AFIP, Washington DC, 1983. Soike KF et al. 1989. Enhancement of simian varicella virus infection in African green monkeys by recombinant human tumor necrosis factor alpha. J Infect Dis 159:331-335. Wenner HA et al. 1977. Clinical and pathogenetic studies of Medical Lake macaque virus infection in cynomolgus monkeys (simian varicella). J Infect Dis 135:611-622. White RO et al. 1972. Chicken pox in young anthropoid apes. Clinical and laboratory findings. J Am Vet Med Assoc 161-690-692. Wolf RH et al. 1974. Pathology and clinical history of Delta herpesvirus infection in patas monkeys. Lab Anim Sci 24:218-221. Other Herpesviruses 1. Oncogenic herpesviruses. (i) Herpesvirus saimiri (H. saimiri type 2): A latent virus of squirrel monkeys-- no disease. Can cause malignant lymphoma after inoculation in marmosets, owl monkeys, and spider monkeys, and in naturally infected owl monkeys. Limited serologic data and infection of some human cell lines in vitro with transformation suggest that human infections are possible, but degree of hazard is unknown. Ablashi V et al. 1971. Malignant lymphoma with lymphocytic leukemia induced in owl monkeys by Herpesvirus saimiri. J Natl Cancer Inst 47:837-855. Ablashi DV et al. 1988. Detection of antibodies to Herpesvirus saimiri late antigens in human sera. Intervirology 29:217-226. Cicmanek JL et al. 1974. Lymphoma in owl monkeys (Aotus trivirgatus) inoculated with Herpesvirus saimiri: Clinical, hematologic, and pathologic findings. J Med Primatol 3:8-17. Daniel M et al. 1970. Malignant lymphoma induced in rabbits by Herpesvirus saimiri (Abstr.) Bact Proc p. 195. Desrosiers RC et al. 1985. A region of the Herpesvirus saimiri genome required for oncogenicity. Science 228:184-187. Desrosiers RC et al. 1986. Nononcogenic deletion mutants of Herpesvirus saimiri are defective for in vitro immortalization. J Virol 57:701-705. Desrosiers RC, Falk LA. 1982. Herpesvirus saimiri strain variability. J Virol 43:352-356. Epstein MA. 1972. Burkitt's lymphoma and Herpesvirus saimiri lymphoma: Comparative aspects. J Natl Cancer Inst 49:213-216. Falk L, Wright T, Deinhardt F. 1976. Experimental infection of squirrel and marmoset monkeys with attenuated Herpesvirus saimiri. Cancer Res 36:707-710. Giddens WE. 1978. Studies of cultured lymphocytes of the owl monkey (Aotus trivirgatus) infected with Herpesvirus saimiri. Lab Anim Sci 26:1113-1126. Grassmann R, Fleckenstein B. 1989. Selectable recombinant herpesvirus saimiri is capable of persisting in a human T-cell line. J Virol 63:1818-1821 Hunt R et al. 1970. Morphology of a disease with features of malignant lymphoma in marmosets and owl monkeys inoculated with Herpesvirus saimiri. J Natl Cancer Inst 44:447-465. Hunt R et al. 1972. Herpesvirus saimiri malignant lymphoma in spider monkeys. A new susceptible host. J Med Primatol 1:114-120. Hunt R et al. 1973. Spontaneous Herpesvirus saimiri lymphoma in an owl monkey. J Infect is 127:723-725. Hunt R et al. 1976. Herpesvirus saimiri lymphoma in owl monkeys (Aotus trivirgatus). Susceptibility, latent period, hematologic picture and course. Theriogen 6:139-151. Jayo MJ et al. 1988. Maxillo-orbital lymphoma (Burkitt's-type) in an infant Macaca fascicularis. Lab Anim Sci 38:722-726. Kamine J, Bakker A, Desrosiers RC. 1984. Mapping of RNA transcribed from a region of the Herpesvirus saimiri genome required for oncogenicity. J Virol 52:532-540. Kiyotaki M, Desrosiers RC, Letvin NL. 1986. Herpesvirus saimiri strain 11 immortalizes a restricted marmoset T8 lymphocyte subpopulation in vitro. J Exp Med 164:926-931. Medveczky MM et al. 1989. Herpesvirus saimiri strains from three DNA subgroups have different oncogenic potentials in New Zealand white rabbits. J Virol 63:3601-3611. Melendez LV et al. 1968. An apparently new herpesvirus from primary kidney cultures of the squirrel monkey (Saimiri sciureus). Lab Anim Care 18:374-381. Melendez LV et al. 1969. Herpesvirus saimiri. II. Further characterization studies of a new virus from the squirrel monkey. Lab Anim Care 19:372-377. Melendez LV et al. 1969. Herpesvirus saimiri. III. Experimentally induced malignant lymphoma in primates. Lab Anim Care 19:378-386. Melendez LV et al. 1974. Prevention of lymphoma in cotton-top marmosets by Herpesvirus saimiri antiserum. J Med Primatol 3:213-220. Mohr FC, Bronson RT, Hunt RD. 1983. Failure of Herpesvirus saimiri to enhance atherogenesis in owl monkeys (Aotus trivirgatus). Atherosclerosis 46:173-179. Newbauer RH et al. 1974. Virological investigations of Herpesvirus saimiri infected owl monkeys. J Med Primatol 3:27-40. Ortaldo JR et al. 1985. Cytotoxic activity of normal and Herpesvirus saimiri-transformed nonhuman primate cells. Cell Immunol 95:349-357. Pearson GR et al. 1974. Immunological and virological investigations on owl monkeys infected with Herpesvirus saimiri. J Med Primatol 3:54-67. Rangan SRS et al. 1977. Herpesvirus saimiri-induced lymphoproliferative disease in howler monkeys. J Natl Cancer Inst 59:165-171. Wallen WC et al. 1974. In vitro immunological characteristics of lymphoid cells derived from owl monkeys infected with Herpesvirus saimiri. J Med Primatol 3:41-53. (ii) Herpesvirus ateles type 2: From spider monkey. Leukemia and malignant lymphoma in cottontop marmosets. Hunt R et al. 1972. Pathologic features of Herpesvirus ateles lymphoma in cotton- topped marmosets (Saguinus oedipus). J Natl Cancer Inst 49:1631-1639. Melendez LV et al. 1972. A new lymphoma virus of monkeys: Herpesvirus ateles. Nature 235:182-184. (iii) Epstein-Barr virus and EBV-like viruses: Experimental disease with human EBV in owl monkeys, gibbons, and cotton-top tamarins; naturally occurring viruses in chimps and baboons. Some experimental evidence indicates that survival may depend on whether or not cell-mediated response occurs. Bocker JF et al. 1980. Characterization of an EBV-like virus from African green monkey lymphoblasts. Virology 101:291-295. Deinhardt F et al. 1978. Susceptibility of marmosets to Epstein-Barr virus-like baboon herpesvirus. Prim Med 10:163-170. Epstein MA et al. 1973. Pilot experiments with EB virus in owl monkeys (Aotus trivirgatus). I. Reticuloproliferative disease in an inoculated animal. Int J Cancer 12:309-318. Epstein MA. 1976. EB virus in the owl monkey (Aotus trivirgatus). Lab Anim Sci 26:1127-1130. Gerber P et al. 1977. Biologic and antigenic characteristics of Epstein-Barr virus related herpesviruses of chimpanzees and baboons. Int J Cancer 20: 448-458. Finerty S, Scullion FT, Morgan AJ. 1988. Demonstration in vitro of cell mediated immunity to Epstein-Barr virus in cotton-top tamarins. Clin Exp Immunol 73:181-185. Heller M, Gerber P, Kieff E. 1982. DNA of Herpesvirus pan, a third member of the Epstein-Barr virus-Herpesvirus papio group. J Virol 41:931-939. Lapin BA, Timanovskaya VV, Yakovleva LA. 1985. Herpesvirus HVMA: a new representative in the group of the EBV-like B-lymphotropic herpesviruses of primates. Hamatol Bluttransfus 29:312-313. Levine PH. 1980. Infection of rhesus macaques and chimpanzees with Epstein-Barr virus. Arch Virol 66:341-351. Rangan SR et al. 1986. Epstein-Barr virus-related herpesvirus from a rhesus monkey (Macaca mulatta) with malignant lymphoma. Int J Cancer 38:425-32. Shope T et al. 1973. Malignant lymphoma in cotton-topped marmosets after inoculation with Epstein-Barr virus. Proc Natl Acad Sci 70:2487-2491. Werner J et al. 1972. Responses of gibbons to inoculation of Epstein-Barr virus. J Infect Dis 126:679-681. Werner J et al. 1975. Lymphoproliferative disease in cotton-topped marmosets after inoculation with infectious mononucleosis-derived Epstein-Barr virus. Int J Cancer 15:1000-1008. Voevodin AF et al. 1983. Increased antibody responses to Herpesvirus papio (HVP) antigens in prelymphomatous baboons (Papio hamadryas) of the Sukhumi high lymphoma stock. Int J Cancer 32:637-639. Young LS et al. 1989. Epstein-Barr virus gene expression in malignant lymphomas induced by experimental virus infection of cottontop tamarins. J Virol 63:1967-1974. 2. Cytomegaloviruses: Isolated from vervet, squirrel, owl monkeys and marmosets. Thought to occur in Cebus spp, chimp, tarsier. Generalized disease is rare, but reported in rhesus and gorilla. Confirmation of the diagnosis may be complicated by the possibility that nonhuman primate CMVs may not react with antisera against human CMV. Ablashi V et al. 1972. A cytomegalovirus isolated from an owl monkey. Lab Anim Sci 33:190-195. Nigida SM, Falk LA, Wolfe LG. 1979. Isolation of a cytomegalovirus from salivary glands of white-lipped marmosets (Saguinus fuscicollis). Lab Anim Sci 29:53-60. Swack NS, Hsiung G. 1982. Natural and experimental simian cytomegalovirus infections at a primate center. J Med Primatol 11:169-177. Vogel FS, Pinkerton H. 1955. Spontaneous salivary gland virus disease in chimpanzees. Arch Pathol 60:281-288. 3. SA-8: An alpha herpesvirus, first isolated from vervet monkey; later from baboons. Papio spp. considered the natural hosts. Persistent latent infection of nerve ganglia, transmitted venereally--possible model of human genital herpes? Usually no disease, although an epizootic was reported in which males had lesions on genitalia which began as patchy erythema and progressed to vesicles and coalescing ulcers. These were accompanied by regional lymphadenopathy. Females had similar lesions on the vulva, anus, and perineum. A few animals also had herpetic lesions on the lips and gums. In some animals, the lesions only occurred once, then healed. Others had recurrences, and a few had persistent lesions. Brack M et al. 1985. Experimental herpes neonatalis in SA8 infected baboons (Papio cynocephalus). Lab Anim 19:125-131. Eichberg J et al. 1973. Experimental herpesvirus infection of baboons (Papio cynocephalus) and African green monkeys (Cercopithecus aethiops) and recovery of virus by tissue explants. Arch Ges Virusforsch 43:304-314. Eichberg JW et al. 1976. Clinical virological, and pathological features of herpesvirus SA8 infection in conventional and gnotobiotic infant baboons (Papio cynocephalus). Arch Virol 50:225-270. Eichberg JW et al. 1980. Immunoresponsiveness of herpesvirus SA8 infected infant baboons. Dev Comp Immunol 4:137-146. Kalter SS, Weiss SA, Heberling RL et al. 1978. The isolation of herpesvirus from trigeminal ganglia of normal baboons (Papio cynocephalus). Lab Anim Sci 28:705-709. Levin JL et al. 1988. A naturally occurring epizootic of simian agent 8 in the baboon. Lab Anim Sci 38:394-397. Ochoa R et al. 1982. Herpesviral pneumonia and septicemia in two infant Gelada baboons (Theropithecus gelada). J Med Primatol 11:51-58. 4. Rhesus leukocyte-associated herpesvirus. Bissell JA et al. 1973. Rhesus leukocyte-associated herpesvirus: II. Natural and experimental infection. J Infect Dis 128:630-637. Frank AL et al. 1973. Rhesus leukocyte-associated herpesvirus. I. Isolation and characterization of a new herpesvirus recovered from rhesus monkey leukocytes. J Infect Dis 128:618-629. 5. Spider monkey herpes virus (Herpesvirus ateles type 1): Isolated in 1968 from female spider monkey that died at California zoo with erosions and ulcers on lips, nose, tongue, palate, and gums. Thought to have been fatal infection in natural host because antibodies have been detected in adult spider monkeys. Hull RN et al. 1972. Recovery and characterization of a new simian herpesvirus from a fatally infected spider monkey. J Natl Cancer Inst 49:225-230. 6. H. aotus types 1, 2 and 3: Owl monkeys. Ebeling A et al. 1983. Genome structure and virion polypeptides of the primate herpesviruses Herpesvirus aotus types 1 and 3: comparison with human cytomegalovirus. J Virol 45:715-726. Fuchs PG et al. 1985. Genome organization of Herpesvirus aotus type 2. J Virol 53:13-18. 7. Tree shrew herpesviruses: 5 types, none known to be pathogenic. Darai G. 1982. Tree shrew herpesvirus. Dev Biol Stand 52:39-51. 8. H. ateles type 3: Third herpesvirus isolated from spider monkeys. 9. H. saguinus: marmoset. Miscellaneous References-Primate Herpesviruses Ablashi V et al. 1979. Oncogenic herpesviruses of nonhuman primates. Comp Immunol Microbiol Inf Dis 2:229-241. Barahona H et al. 1974. A compendium of herpesvirus isolated from non-human primates. Intervirol 3:175-192. Barahona H et al. 1980. The owl monkey (Aotus trivirgatus) as an animal model for viral diseases and oncologic studies. Lab Anim Sci 26:1104-1112. Falk LA. 1974. Oncogenic DNA viruses of nonhuman primates: A review. Lab Anim Sci 24:182-192. Fleckenstein B. 1979. Oncogenic herpes viruses of nonhuman primates. Biochim Biophys Acta 560:301-342. Hilliard JK, Kalter SS. 1985. Development of molecular probes for simian herpesvirus detection. Dev Biol Stand. 59:79-86. Hunt R et al. Malignant lymphoma, Model No. 60. In Handbook Animal Models of Human Disease, fasc. 5. Ed TC Jones, DB Hackel and G Migaki. Registry of Comparative Pathology, AFIP, Washington, DC, 1976. Hutt R. 1981. Detection of antibodies to Herpesvirus simiae and Herpesvirus hominis in nonhuman primates. Lab Anim Sci 31:184-189. Kalter SS, Hilliard JK, Heberling RL. 1982. The differential diagnosis of herpesvirus infections in man and animals. Dev Biol Stand 52:101-113. McCarthy K et al. 1975. A review of primate herpesviruses. Proc Royal Soc Med 68:145- 150. Melendez LV et al. 1971. New herpesviruses from South American monkeys. Preliminary report. Lab Anim Sci 21:1050-1054. Melendez LV 1972. Two new herpesviruses from spider monkeys (Ateles geoffroyi). J Natl Cancer Inst 49:233-237. Melendez LV et al. Lymphoma virus of monkeys: Herpesvirus saimiri and Herpesvirus ateles, the first oncogenic herpesviruses of primates. A review. In Oncogenesis and Herpesviruses, International Agency for Research on Cancer, Lyon, pp 451-461, 1972. Mou SW et al. 1986. Comparison of the primate alphaherpesviruses. I. Characterization of two herpesviruses from spider monkeys and squirrel monkeys and viral polypeptides synthesized in infected cells. Arch Virol 91:117-133. Rangan SRS. 1979. Tumors and viruses in nonhuman primates. Adv Virus Res 24:1-123. Rasheed S. 1979. Herpesvirus of orangutan (H. pongo) and search for retrovirus in nonhuman primates. Comp Immunol Microbiol Inf Dis 2:265-274. Measles (Rubeola) 1. Agent: A paramyxovirus, genus Morbillivirus. 2. Epizootiology: Macaques (especially rhesus), baboons, African green, marmosets, tamarins, squirrel monkeys, chimps, and Presbytis cristatus are susceptible to human measles. Can be fatal if complicated by bacteria. Not a disease of wild monkeys, but most wild- caught monkeys seroconvert within a few months of capture. Virus is shed in most secretions and in urine. 3. Clinical: Can be asymptomatic or rapidly fatal. Marmosets are said to be especially susceptible. Course of clinical disease usually is less than 10 days, and is characterized by maculopapular skin rash, serous to mucopurulent nasal discharge, ocular discharge, maybe Koplik's spots, diarrhea, and rarely, abortion. Immuno- suppression during measles virus infection is clearly documented. 4. Pathology: Interstitial pneumonia with syncytial giant cells in alveoli and bronchioles, with nuclear and cytoplasmic inclusions. This commonly is complicated by secondary bacterial invaders, such as Streptococcus pneumoniae, resulting in suppurative bronchopneumonia. In such cases, the lesions due to the viral infection may be obscured. Skin lesions include focal acanthosis, hyperkeratosis, and parakeratosis; hair follicle necrosis; epidermal and follicular giant cells; no inclusions. Lymphoid tissues can have syncytial giant cells (Warthin-Finkeldey cells). Similar cells can occur in the liver and transitional epithelium of the urinary tract, although these are rare, and on occasion can be seen in other epithelia throughout the body and in the choroid plexus. It is more usual for transitional epithelium to have inclusions without much cellular change. Such inclusions must be differentiated from the nonspecific inclusions in transitional epithelium of macaques, especially cynomolgus. Enterocolitis with inclusions in some cases. Rarely, endometritis. SSPE-like disease reported in baboon. 5. Diagnosis: Signs and lesions, virus isolation and identification, various serologic techniques such as hemagglutination inhibition. Remember that macaques, especially cynomolgus, often have nonspecific inclusions in the urinary bladder epithelium that resemble measles virus inclusions. 6. Control: Keep known exposed personnel and children away from primates. Newly imported primates will have been exposed before reaching lab. Zoonotic (monkeys can transmit back to people). Chick embryo measles vaccine can be used. Not necessary if already exposed, but primates raised in closed colonies could be susceptible; vaccine could be helpful prophylaxis. 7. References: Albrecht P et al. 1977. SSPE: Experimental infection in nonhuman primates. Science 195(4273):64-66. Albrecht P et al. 1980. Fatal measles infection in marmosets. Pathogenesis and prophylaxis. Infect Immun 27:969-978. Frazer CEO et al. 1978. A paramyxovirus causing fatal gastroenterocolitis in marmoset monkeys. Prim Med 10:261-270. Fujimoto K et al. 1983. Immunoglobulins and measles antibody in sera of newborn cynomolgus monkeys and in milk of their mothers. Jpn J Med Sci Biol 36:209-14. Hall WC et al. 1971. Pathology of measles in rhesus monkeys. Vet Path 8:307-319. Kamahora J. 1965. Pathological and immunological studies of monkeys infected with measles virus. Arch ges Virusforsch 16:161-167. Kim LS et al. 1970. Subacute sererosing panencephalitis-like syndrome in the adult baboon (Papio sp.). J Am Vet Med Assoc 157:730-735. Klutch MJ et al. 1979. Fatal measles infection in marmosets. Proc Am Soc Microbiol. p. 308. Levy BM, Mirkovic RR. 1971. An epizootic of measles in a marmoset colony. Lab Anim Sci 21:33-39. Lorenz D, Albrecht P. 1980. Susceptibility of tamarins to measles virus. Lab Anim Sci 30:661-665. MacArthur JA et al. 1982. The effect of measles vaccination in rhesus monkeys (Macaca mulatta). Lab Anim 16:267-270. Manning PJ et al. 1968. Naturally occurring giant cell pneumonia in the rhesus monkey (Macaca mulatta). J Am Vet Med Assoc 153:899-904. McChesney MB et al. 1989. Virus-induced immunosuppression: infection of peripheral blood mononuclear cells and suppression of immunoglobulin synthesis during natural measles virus infection of rhesus monkeys. J Infect Dis 159:757-760. Montrey R et al. 1980. An epizootic of measles in captive silvered leaf monkeys (Presbytis cristatus) in Malaysia. Lab Anim Sci 30:694-697. Potkay S et al. 1966. An epizootic of measles in a colony of rhesus monkeys (Macaca mulatta). Am J Vet Res 27:331-334. Remfry J. 1976. A measles epizootic with five deaths in newly imported rhesus monkeys (Macaca mulatta). Lab Anim 10:49-57. Renne RA. 1973. Measles virus-associated endometritis, cervicitis, and abortion in a rhesus monkey. J Am Vet Med Assoc 163:639-641. Steele M et al. 1982. Spontaneous paramyxoviral encephalitis in nonhuman primates (Macaca mulatta and M. nemestrina). Vet Pathol 19:132-139. Welshman MD. 1989. Measles in the cynomolgus monkey (Macaca fascicularis). Vet Rec 124:184-186. Simian Adenoviruses 1. Agents: Many (> 20) serotypes isolated, some only from cell cultures. 2. Epizootiology: Wide host range, some perhaps including man. Common in gastrointestinal and respiratory tracts, not usually pathogenic. 3. Clinical: Mostly respiratory, if any. Various serotypes associated with mild upper respiratory signs and conjunctivitis or "pneumoenteritis." Diarrhea reported in experimental disease. Usually subclinical in adults. Fatalities reported mostly in neonatal macaques (rhesus, bonnet, pigtailed, cynomolgus); African green and baboons also susceptible. 4. Pathology: Necrotizing bronchiolitis, multifocal necrotizing pneumonitis, and interstitial pneumonia. Typical nuclear inclusions in epithelial cells. Necrotizing pancreatitis due to adenoviral infection has been reported. Several serotypes are oncogenic in newborn hamsters (lymphomas, sarcomas). 5. Diagnosis: Morphology, virus isolation and identification. 6. Control: No vaccine available. 7. References: Boyce JT, Giddens WE Jr, Valerio M. 1978. Simian adenoviral pneumonia. Am J Pathol 91:259-276. Chandler FW, McClure HM. 1982. Adenoviral pancreatitis in rhesus monkeys: Current knowledge. Vet Pathol 19(Suppl 7):171-180. Darai G et al. 1980. An adenovirus from Tupaia (tree shrew). Growth of the virus, characterization of viral DNA, and transforming ability. Virol 104:122-138. Eugster AK et al. 1969. Isolation of adenoviruses from baboons (Papio sp.) with respiratory and enteric infections. Arch ges Virusforsch 26:260. Landon JC, Bennett G. 1969. Viral induced simian conjunctivitis. Nature 222:683-684. Li QG, Wadell G. 1989. The degree of genetic variability among adenovirus type 4 strains isolated from man and chimpanzee. Arch Virol 101:65-77. Merkow LP, Slifikin M. 1973. Simian adenoviruses. Progr Exp Tumor Res 18: 67. Moe JB et al. 1979. Experimental adenovirus (SV-20) pneumonia in fetal rhesus monkeys. Lab Invest 41:211-219. Moe JB et al. Fetal pneumonia, Model No. 210. In Handbook: Animal Models of Human Disease, Fasc 9, ed. CC Capen, DB Hackel, TC Jones, and G Migaki. Registry of Comparative Pathology, AFIP, Washington, DC, 1980. Moe JB, Osburn BI. 1983. Fetal and maternal immunologic manifestations of intrauterine Adenovirus SV-20 infection. Vet Immunol Immunopathol 4:319-32. Ohtaki S, Kato K. 1989. Simian adenovirus type 7 (SA-7) induces tumours of nerve-supporting or paraneural cell origin in newborn hamsters. Br J Exp Pathol 70:415-424. Schoeb TR, DaRif CA. 1984. Adenoviral infection in infant tree shrews. J Am Vet Med Assoc 185:1363-1366. Shortridge KF et al. 1976. Serological evidence of human adenovirus infections in animals. J Comp Pathol 86:629-635. Shroyer EL et al. 1979. Three serologic types of adenovirus infection of owl monkeys. Am J Vet Res 110:532-536. Wigand R, Mauss M, Adrian T. 1989. Chimpanzee adenoviruses are related to four subgenera of human adenoviruses. Intervirology 30:1-9. Miscellaneous Respiratory Viruses 1. Agents: (i) Influenza (orthomyxovirus). Gibbons. (ii) Parainfluenza type 1 (paramyxovirus). Marmosets. (iii) Parainfluenza type 3 (paramyxovirus). Patas monkeys and gibbons. (iv) Respiratory syncytial virus (pneumovirus). Chimp. (v) Reoviruses (Reoviridae). Apes. (vi) Rhinoviruses (Picornaviridae). Apes. (vii) SV5, SV41 (paramyxoviruses). 2. Disease: None (SV5, SV41) to mild upper respiratory disease (most) to moderately severe respiratory disease with bronchitis (influenza). Infection seldom significant in itself but may predispose to bacterial pneumonia, a major cause of morbidity and mortality among captive nonhuman primates. 3. References: Belshe RB et al. 1977. Experimental respiratory syncytial virus infection of four species of primates. J Med Virol 1:157-162. Churchill AE. 1963. The isolation of parainfluenza-3 virus from fatal cases of pneumonia in Erythrocebus patas monkeys. Br J Exp Pathol 44:529-557. Flecknell PA et al. 1983. Respiratory disease associated with parainfluenza type 1 (Sendai) virus in a colony of marmosets (Callithrix jacchus). Lab Anim 17:111-113. Fraser CEO et al. 1978. A paramyxovirus causing fatal gastroenterocolitis in marmoset monkeys. Prim Med 10:261-270. Johnson DO et al. 1971. An epizootic of A2/Hong Kong/68 influenza in gibbons. J Infect Dis 123:365-370. Jones EE et al. 1984. Predisposition to invasive pneumococcal illness following parainfluenza type 3 virus infection in chimpanzees. J Am Vet Med Assoc 185:1351- 1353. Martin P, Kaye HS. 1983. Epizootic of parainfluenza-3 virus infection in gibbons. J Am Vet Med Assoc 183:1185-1187. Murphy BR et al. 1983. Evaluation of the A/Seal/Mass/1/80 virus in squirrel monkeys. Infect Immun 42:424-6. Richardson LS et al. 1978. Experimental respiratory syncytial virus pneumonia in cebus monkeys. J Med Virol 2:45-59. Richardson-Wyatt LS 1981. Respiratory syncytial virus antibodies in nonhuman primates and domestic animals. Lab Anim Sci 31:413-415. Sutherland SD et al. 1986. Rapid diagnosis and management of parainfluenza 1 virus infection in common marmosets (Callithrix jaccus). Lab Anim 20:121-126. Monkeypox 1. Agent: A poxvirus antigenically related to smallpox and vaccinia viruses. 2. Epizootiology: Reported in rhesus and cynomolgus macaques, owl-faced monkeys (Cercopithecus hamlyn), gorillas, gibbons, squirrel monkeys, marmosets, chimpanzees, and humans. Occurs naturally in West and Central Africa. A high prevalence (24.7%) of monkeypox-specific antibodies was found in two species of squirrels (Funisciurus anerythrus and Heliosciurus rufobrachium), suggesting that these animals may serve as reservoirs or vectors. (The animals commonly are trapped by natives.) Three of 39 primates were seropositive for monkeypox, but as yet it is not known whether primates maintain the virus or, like humans, are merely occasional hosts. About 10 or so outbreaks in captive primates have been reported. Apes appear to be quite susceptible. 3. Clinical: Fever, "pocks" over trunk, tail, face, and limbs, including palms and soles. Wide range of susceptibility: in some, general health unaffected but others become ill and some die. 4. Pathology: Pathogenesis is classical for systemic poxviral disease, with local multiplication, primary viremia, secondary internal multiplication, secondary viremia, disseminated pox. Skin lesions begin as firm elevations resulting from epithelial proliferation. As "ballooning" degeneration then necrosis proceed from the center outward, lesions develop into umbilicated pustules ("pocks"). Crusts then form over healing lesions. Other lesions include multifocal necrotizing pneumonitis, orchitis, and lymphadenitis with lymphoid hyperplasia. 5. Diagnosis: Microscopic exam of lesion scrapings, morphology, EM, virus isolation. 6. Control: An uncommon disease. Common sense and good colony management should suffice. 7. References: Arita I et al. 1972. Outbreaks of monkey pox and serological surveys in nonhuman primates. Bull WHO 46:625-631. Arita I et al. 1985. Human monkeypox: A newly emerged orthopoxvirus zoonosis in the tropical rain forests of Africa. Am J Trop Med Hyg 34:781-789. Bourke ATC, Dumbell KR. 1972. An unusual pox virus from Nigeria. Bull WHO 46:621- 623. Cho CT, Wenner WA. 1973. Monkey pox virus. Bacteriol Rev 37:1-18. Foster SO et al. 1972. Human monkey pox. Bull WHO 46:569-576. Heberling RL, Lalter SS. 1971. Induction, course, and transmissibility of monkey pox in the baboon (Papio cynocephalus). J Infect is 124:33-38. Janseghers L et al. 1984. Fatal monkeypox in a child in Kikwit, Zaire. Ann Soc Belg Med Trop 64:295-298. Khodakevich L et al. 1987. Monkeypox virus in relation to the ecological features surrounding human settlements in Bumba zone, Zaire. Trop Geogr Med 39:56-63. Khodakevich L et al. 1987. The role of squirrels in sustaining monkeypox virus transmission. Trop Geogr Med 39:115-122. Lourie G et al. 1972. Human infection with monkey pox virus. Laboratory investigation of six cases in West Africa. Bull WHO 46:633-639. Marennikova SS et al. 1972. Isolation and properties of the causal agent of a new variola-like disease (monkey pox) in man. Bull WHO 46:599-611. Mutombo M. 1983. Human monkey pox transmitted by a chimpanzee in a tropical rain forest area of Zaire. Lancet 1(8237):735-737. Stagles MJ et al. 1985. The histopathology and electron microscopy of a human monkeypox lesion. Trans R Soc Trop Med Hyg 79:192-202. _____. 1984. Human monkeypox: The past five years. WHO Chron 38:227-229. Benign Epidermal Monkeypox (BEMP, Tana Pox) 1. Agent: A pox virus antigenically related to Yaba virus but not to vaccinia and smallpox. Syn. OrTeCa virus. 2. Epizootiology: Disease of man and monkeys in Kenya in late 1950s and early 1960s. Appeared simultaneously in macaques and handlers in labs in Oregon, Texas, and California (hence "OrTeCa") in mid 1960s, and slightly later in Maryland. In the first 3 labs the animals were from a single importer, where the disease was also seen. May not be indigenous monkey disease; if not, natural host unknown. 3. Clinical: In rhesus monkeys, typical pocks on face, arms and perineum, without general illness. Most affected people have only localized lesions, e.g., at sites of scratches on hands and arms, but some had regional lymphadenopathy, fever, and debility, and a few had generalized pocks. 4. Pathology: Similar to monkey pox. Sequential epidermal changes of proliferation (acanthosis), necrosis, sloughing, and healing. However, proliferation tends to extend into hair follicles. Cytoplasmic inclusions are numerous, nuclear vacuoles occur which reportedly help differentiate from monkey pox. No visceral lesions reported. 5. Diagnosis: As for monkeypox. 6. Control: ?--Apparently has not occurred since original outbreak. 7. References: Casey HW et al. 1967. Electron microscopy of a benign epidermal pox disease of rhesus monkeys. Am J Pathol 51:443-446. Downie AW, Espana C 1972. Comparison of tanapox and Yaba-like viruses causing epidemic disease in monkeys. J Hyg 70:23-32. Hall AS, McNulty WP. 1967. A contagious pox disease in monkeys. J Am Vet Med Assoc 151:833-838. Kupper JC. 1970. Experimental Yaba and benign epidermal monkey pox in rhesus monkeys. Lab Anim Sci 20:979-988. McNulty WP et al. 1968. A pox disease in monkeys transmitted to man. Clinical and histological features. Arch Dermatol 97:386-293. Yaba Virus 1. Agent: A poxvirus serologically unrelated to the vaccinia group but related to OrTeCa virus. 2. Epizootiology: Not understood; may be a natural indigenous infection of wild African primates. Subcutaneous tumors in rhesus monkeys and a baboon in Yaba, Nigeria in the late 1950s were shown to be caused by a pox virus. Mode of transmission obscure, but experimentally can be done by aerosol, direct inoculation. Man, cynomolgus, rhesus, stumptailed, and pigtailed macaques, African green monkeys, sooty mangabeys and patas monkeys are susceptible. Apparently common. 3. Clinical: Rapidly growing subcutaneous masses which spontaneously regress in a few weeks. 4. Pathology: Proliferation of large "histiocytes" in dermis or other connective tissue (internal tumors occurred after IV inoculation). Cytoplasmic inclusions are usually visible. Multinucleate giant cells appear with regression, which is accompanied by lymphocytic and plasmacytic reaction. 5. Diagnosis: Signs and lesions, EM, virus isolation. 6. References: Behbehani AM et al. 1968. Yaba tumor virus. I. Studies on pathogenesis and immunity. Proc Soc Exp Biol Med 129:556-561. Bruestle ME et al. 1981. Naturally occurring Yaba tumor in a baboon (Papio papio). Lab Anim Sci 31:392-394. Grace JT, Mirand EA. 1963. Human susceptibility to a simian tumor virus. Ann NY Acad Sci 108:1123-1128. Kupper JL. 1970. Experimental Yaba and benign epidermal monkey pox in rhesus monkeys. Lab Anim Sci 20:979-988. Spencer AJ. Diagnostic exercise. 1985. Subcutaneous nodules in rhesus monkeys. Lab Anim Sci 35:79-80. Tsuchiya y, Tagaya I. 1971. Sero-epidemiological survey on Yaba and 1211 virus infections among several species of monkeys. J Hyg 69:445-451. Whittaker D, Glaister JR. 1985. A Yaba-like condition in a young baboon. Lab Animal 19:177-179. Wolfe LG et al. 1968. Experimental aerosol transmission of Yaba virus in monkeys. J Natl Cancer Inst 41:1175-1195. Molluscum Contagiosum 1. Agent: Poxvirus of man, unrelated to smallpox or vaccinia, never isolated but has poxvirus ultrastructure. 2. Epizootiology: Mildly contagious disease, mostly of children. One report of disease in chimps. 3. Clinical: Proliferative lesions mostly around the eyes in chimps. 4. Pathology: Very characteristic. Verrucous or plaque-like lesions characterized by extreme epidermal proliferation. Very large inclusions. 5. Diagnosis: Morphology, EM. 6. Reference: Douglas J et al. 1967. Molluscum contagiosum in chimpanzees. J Am Vet Med Assoc 151:901-904. Simian Hemorrhagic Fever 1. Agent: Complete classification not yet assigned. Tentatively, genus Pestivirus (Togaviridae). 2. Epizootiology: Asymptomatic infections in patas monkeys, vervet monkeys, and baboons. Patas monkeys thought to be natural host; about 50% of wild patas monkeys have antibodies, much less prevalent in vervets and baboons. Some patas monkeys, maybe about 10%, are asymptomatic carriers. In macaques (M. mulatta, M. fascicularis, M. arctoides, M. assamensis, M. nemestrina) epizootics of acute severe disease with high mortality. (Mortality can ÷100%.) Seen in Washington DC, California, Russia, and England. Puzzling: epizootics have occurred in when macaques introduced into macaque colonies, without obvious exposure to patas monkeys--source unclear. 3. Clinical: Rapid onset, fever, facial edema, cyanosis, anorexia, melena; cutaneous, subcutaneous, and retrobulbar hemorrhage; thrombocytopenia. Course 10-15 days. 4. Pathology: Hemorrhages in any tissue, especially skin, nasal mucosa, lung, gastrointestinal tract, renal capsule and perirenal tissue. Hemorrhagic necrosis of the duodenal mucosa, distinctly demarcated at the pylorus, usually present. There is also splenomegaly with hemorrhagic necrosis of the lymphoid areas; necrosis of the lymph nodes, Peyer's patches, and tonsils; and many thrombi in small veins and capillaries. Disseminated intravascular coagulation is an important component of the pathogenesis of the disease. 5. Diagnosis: Signs and lesions, virus isolation, serologic test in survivors. 6. Control: Keep primate species separate; proper hygeine. Serologic tests cannot distinguish carriers from previously infected, and virus isolation is unreliable for diagnosis. 7. References: Abilgaard C et al. 1975. Simian hemorrhagic fever: Studies of coagulation and pathology. Am J Trop Med Hyg 24:537-544. Allen AM et al. 1972. Simian hemorrhagic fever. II. Studies in pathology. Am J Trop Med Hyg 17:413-421. Giddens WE et al. 1975. The pathogenesis of simian hemorrhagic fever: Hematologic and histopathologic studies. Lab Invest 32:424-___. Gravell M et al. 1980. Simian hemorrhagic fever (SHF): New virus isolate from a chronically infected patas monkey. J Gen Virol 51:99-106. Gravell M et al. 1980. Methods to detect asymptomatic carriers of simian hemorrhagic fever virus. Lab Anim Sci 30:988-991. Gravell M et al. 1980. Studies on simian hemorrhagic fever virus infection of patas monkeys. I. Serology. In The Comparative Pathology of Zoo Animals, ed. Montali RJ, Migaki G, Smithsonian Institution Press, Washington, DC, pp. 167-170. Gravell M et al. 1982. Method to eliminate persistent simian hemorrhagic fever virus infection of patas monkeys (Abstract). Lab Anim Sci 32-427. London WT. 1977. Epizootiology, transmission and approach to prevention of fatal simian hemorrhagic fever in rhesus monkeys. Nature 268:344-345. London WT et al. 1982. Problems associated with treatment and prophylaxis of simian hemorrhagic fever (SHF) in macaques (Abstract). Lab Anim Sci 32:427- . Madden L et al. 1968. Antigenic relationship of two strains of simian hemorrhagic fever virus. Lab Anim Sci 28:422-427. Palmer AE et al. 1968. Simian hemorrhagic fever. Am J Trop Med Hyg 17:404-431. Tauraso WM et al. 1968. Simian hemorrhagic fever. III. Isolation and characterization of a viral agent. Am J Trop Med Hyg 17:423-431. Trousdale M et al. 1975. Simian hemorrhagic fever virus: A new togavirus (3911). Proc Soc Exp Biol Med 150:707-711. Yellow Fever 1. Agent: Genus Flavivirus, Togaviridae. 2. Epizootiology: A disease of Central and South America and Africa. In urban areas passed from person to person by mosquito (Aedes spp.). In the wild many mammals are natural hosts, including the primate genera Alouatta, Ateles, Callicebus, Cebus, Marikina, and Saimiri. Man is only an accidental host. Transmitted among day-sleeping monkeys by day-feeding tree-living mosquitos. Alouatta spp. are especially susceptible. Disease less severe in African primates. Rhesus monkeys susceptible but no natural disease in Asia. 3. Clinical: In man, fever, muscular aches, headaches, jaundice, nausea and vomiting (sometimes with blood)-- "black vomit"), hemorrhage, leukopenia, albuminuria. 4. Pathology: Icterus, hemorrhage, severe midzonal hepatocellular necrosis with "Councilman bodies." 5. Diagnosis: Lesions, demonstration of virus. 6. Control: Mosquito control. Monkeys from areas where enzootic must be shipped in mosquito-proof cages and examined for evidence of yellow fever if dead within 10 days. 7. References: Bearcroft WCG 1957. The histopathology of the liver of yellow fever-infected rhesus monkeys. J Pathol Bacteriol 74:295-303. Bearcroft WCG 1962. Electron microscope studies on the livers of yellow fever infected African monkeys. J Pathol Bacteriol 83:59-64. Pinheird FB. 1981. An epidemic of yellow fever in central Brazil 1972-1973 II. Ecological studies. Am J Trop Med Hyg 30:204-211. Smetana HF. 1962. The histopathology of experimental yellow fever. Virch Arch Pathol Anat 335:411-427. Marburg Virus 1. Historical: Marburg disease was first recognized in 1967. Severe disease in lab workers in Germany and Yugoslavia working with tissue cultures from African green monkeys from Uganda; 7/30 affected died. Monkeys not ill in German lab but there were deaths among new monkeys in Yugoslav lab. Since then, sporadic in Africa. 2. Agents: Previously thought to be rhabdoviruses, these agents are morphologically quite different from Vesiculovirus spp. (vesicular stomatitis group) and Lyssavirus spp. (rabies group). Some authors propose that Marburg virus and the similar Ebola virus be placed in a new, as yet not official, family, the Filoviridae. 3. Epizootiology: Transmission by aerosol, handling infected tissue, saliva, urine. Monkeys are susceptible to even small doses of virus; thus are probably not natural or reservoir host. If this be true, the natural host is unknown. Antibodies to these viruses are found at low prevalence rates in wild-caught primates in Kenya, but the significance of this is not known. 4. Clinical: (i). Man: 5-8 day incubation. Fever, headache, impaired consciousness, vomiting, diarrhea, rash, conjunctivitis, lymphadenopathy, hemorrhagic diathesis. (ii). Experimental disease in African green, rhesus, and squirrel monkeys: Febrile until about 24 hours before death, then temperature dropped to subnormal; diarrhea, rash, dyspnea, course 6-13 days, uniformly fatal. 5. Pathology: (i). Man: Cerebral edema, multifocal necrosis of many tissues especially liver, lymphoid tissue, kidney, pancreas, adrenal, and skin; lymphocytic encephalitis. (ii). Monkeys: Similar to man. Multifocal to diffuse hemorrhagic necrosis of liver, spleen, lungs; lymphoid necrosis, multifocal interstitial pneumonia, pulmonary vasculitis. 6. Diagnosis: Signs and lesions, virus isolation and identification. Marburg and Ebola diseases are indistinguishable. 7. Control: Effective measures unknown--source unknown. Use care when working with African green monkeys and cells derived from them. 8. References: Anon. 1980. Marburg virus in Kenya. Lab Prim Newsletter 19:10-11. Bowen ETW et al. 1978. Ebola hemorrhagic fever: Experimental infection of monkeys. Trans Royal Soc Trop Med Hyg 72:188-191. Conrad JL et al. 1978. Epidemiologic investigation of Marburg virus disease, Southern Africa, 1975. Am J Trop Med Hyg 27:1210-1215. Ellis S et al. 1979. Ebola virus: A comparison at ultrastructural level of the behavior of the Sudan and Zaire strains in monkeys. Br J Exp Pathol 59:584-593. Gear JSS et al. 1975. Outbreak of Marburg virus disease in Johannesburg. Brit Med J 4:489-493. Gordon-Smith CE et al. 1967. Fatal human disease from vervet monkeys. Lancet 2:1119- 1121. Johnson BK et al. 1982. Marburg, Ebola and Rift Valley Fever virus antibodies in East African primates. Trans R Soc Trop Med Hyg 76:307-310. Johnson BK. 1981. Marburg and Ebola virus antibodies in Kenyan primates. Lancet 1 (8235):1420-1421. Johnson KM et al. 1977. Isolation and partial characterization of a new virus causing acute hemorrhagic fever in Zaire. Lancet 1:569-572. Kiley MP et al. 1988. Physicochemical properties of Marburg virus: evidence for three distinct virus strains and their relationship to Ebola virus. J Gen Virol 69:1957-1967. Kissling RE et al. 1970. Marburg virus. Ann NY Acad Sci 174:932-945. Morbidity and Mortality Weekly Report 29:145-146. Simpson IH. 1969. Marburg agent disease: In monkeys. Trans Royal Soc Trop Med Hyg 63:303-309. Smith H et al. 1982. Marburg-virus disease in Kenya. Lancet 1(8276):816-820. Zlotnik I. 1969. Marburg agent disease: Pathology. Trans Royal Soc Trop Med Hyg 63:310-323. Miscellaneous Hemorrhagic Fevers 1. Dengue Fever (i). Agent: Genus Flavivirus (Togaviridae). (ii). Epizootiology: Southeast Asia, Mediterranean area, South Africa, Central America, and Hawaii. Monkeys are reservoir. Spread to man by Aedes aegypti. (iii). Disease: In man, mild (fever, lymphadenopathy, rash) to severe (hemorrhagic fever). In monkeys, no disease to forms similar to those in man but usually without rash. (iv). Control: Mosquito control; vaccination. (v). References: Halstead SB et al. 1973. Studies on the pathogenesis of Dengue infection in monkeys. I. Clinical laboratory responses to primary infection. II. Clinical laboratory responses to heterologous infection. III. Sequential distribution of virus in primary and heterologous infections. J Infect Dis 128:7-30. Marchette NN, Halstead SB. 1974. Immunopathogenesis of Dengue infection in the rhesus monkey. Transplantation Proc. 6:197-201. Sabin AB. 1959. Dengue. In Viral and Rickettsial Infections of Man, ed. TM Rivers. JB Lippincott, Philadelphia, pp 361-372. Yuwono J et al. 1984. Seroepidemiological survey on dengue and Japanese encephalitis virus infections in Asian monkeys. Southeast Asian J Trop Med Public Health 15:194- 200. 2. Kyasanur Forest Disease (i). Agent: a Flavivirus. (ii). Epizootiology: Langurs (Presbytis entellus), bonnet macaques, and man in India. (iii). Disease: Similar to Dengue fever. (iv). References: Shah KV. 1965. Experimental infection of lactating monkeys with Kyasanur Forest disease virus. Acta Virol (Prague) 9:72-75. Singh KR et al. 1963. Experimental transovarial transmission of Kyasanur Forest disease virus in Haemaphysalis spinigera. Nature 199:513. Webb HE, Burton J. 1966. Clinical and pathological observations with special reference to the nervous system in Macaca radiata infected with Kyasanur Forest disease virus. Trans Royal Soc Trop Med Hyg 60:325-331. Work TH. Kyasanur Forest disease. Progr Med Virol 1:248, 1958. Work TH et al. 1959. Virological epidemiology of the 1958 epidemic of Kyasanur Forest disease. Am J Publ Hlth 49:869-874. 3. Other Gonzalez PH et al. 1983. Junin virus infection of Callithrix jacchus: pathologic features. Am J Trop Med Hyg 32:417-423. Johnson BK et al. 1982. Marburg, Ebola and Rift Valley Fever virus antibodies in East African primates. Trans R Soc Trop Med Hyg 76:307-310. Gear JH. 1989. Clinical aspects of African viral hemorrhagic fevers. Rev Infect Dis 11(Suppl 4):S777-S782. Viral Hepatitis 1. Agents: Hepatitis A virus is a picornavirus; hepatitis B virus, a hepadnavirus; the delta or hepatitis D agent is an unclassified, defective RNA virus; and non-A-non-B agents are not identified with certainty. 2. Epizootiology: (i) Hepatitis A ("infectious hepatitis") in man is transmitted by fecal-oral route. Monkeys and apes get from man. Serologic evidence of infection in chimps, marmosets, baboons, and patas, African green, and Cebus monkeys. Chimps and marmosets susceptible to overt disease (marmosets used as model), in others inapparent. Zoonotic. Human hepatitis A has a short icubation period and is generally mild with very low mortality. (ii) Hepatitis B ("serum hepatitis") in man is usually transmitted in blood or serum but can be transmitted by contact or indirectly since virus is in most secretions. Chimps and other apes and monkeys (including rhesus) are readily infected (experimental in chimps and gibbons; evidence for natural infection in chimp, baboon, squirrel monkey). Zoonotic. Human hepatitis B has a long incubation period, and there can be carriers with or without hepatitis. The disease may be manifested as acute hepatitis which infrequently is fulminant. Chronic hepatitis occurs in many cases; it can be self-limiting or progressive, and can be fatal. The HBs Ag is the viral coat antigen; many of the particles are not infective, having no core, but there are lots of them in the blood. Chronic hepatitis B can be associated with hepatic carcinoma. (iii) The delta or hepatitis D agent, which is a defective virus that replicates only in the presence of HBV, typically causes a more virulent disease than uncomplicated hepatitis B, more frequently resulting in progressive cirrhosis and eventual liver failure. (iv) Non-A-non-B hepatitis in man is transmitted primarily via blood or serum and is the major cause of post-transfusion hepatitis. NANB hepatitis tends to be chronic. There are probably two (possibly more) forms of NANBH: hepatitis C, which is transmitted parenterally, and an epidemic form, which is transmitted enterically. Agent(s) can persist for up to 6 years. NANBH has been experimentally transmitted to chimps. Zoonotic? 3. Clinical: Usually inapparent but can have fever, jaundice, even death. 4. Pathology: Resembles that in man: Hepatocellular degeneration and necrosis which is multifocal or irregular but tends to greater severity in the centrilobular zone, accompanied by Kupffer cell hyperplasia and mononuclear inflammatory response. Inflammatory infiltrates tend to be mostly portal, but there can be some around the central veins also, and in sinusoids depending on severity. Chronic active NANB hepatitis reported in chimps. 5. Diagnosis: Serologic, morphologic. 6. Control: Hygiene, training of personnel, exclusion of any known infected persons, prophylactic immune globulin for handlers. 7. References: Abe K, Shikata T. 1982. Fulminant type A viral hepatitis in a chimpanzee. Acta Pathol Jpn 32:143-148. April M. Non-A, Non-B Hepatitis, Model No. 221. In Handbook: Animal Models of Human Disease, fasc. 10. Ed. by CC Capen, DB Hackel, TC Jones, and G Migaki. Registry of Comparative Pathology, AFIP, Washington, DC, 1981. Barker LF et al. 1973. Transmission of type B viral hepatitis to chimpanzees. J Infect Dis 127:648-662. Berthelot P et al. 1984. Hepatitis B vaccine safety monitoring in the chimpanzee: Interpretation of results. J Med Primatol 13:119-134. Bradley DW et al. 1987. Enterically transmitted non-A, non-B hepatitis: Serial passage of disease in cynomolgus macaques and tamarins and recovery of disease-associated 27- to 34-nm viruslike particles. Proc Natl Acad Sci USA 84:6277-6281. Burke DS et al. 1981. Hepatitis A virus in primates outside captivity. Lancet 2:928. Cote PJ et al. 1986. Protection of chimpanzees from type B hepatitis by immunization with woodchuck hepatitis virus surface antigen. J Virol 60:895-901. Coulepis AG. 1982. Biophysical and biochemical characterization of hepatitis A virus. Intervirol 18:107-127. Deinhardt F. et al. 1967. Studies on the transmission of human viral hepatitis to marmoset monkeys. I. Transmission of disease, serial passages, and description of liver lesions. J Exp Med 125:673-688. Dienstag JL. 1976. Pathology of hepatitis A and B in chimpanzees. Am J Pathol 85:131. Eichberg JW, Kalter SS. 1980. Hepatitis A and B: Serologic survey of human and nonhuman primate sera. Lab Anim Sci 30:541-543. Holmes AW et al. 1973. Specific neutralization of human hepatitis type A in marmoset monkeys. Nature (Lond) 243:419-420. Jensen DM et al. 1984. Antibodies directed against human liver specific membrane lipoprotein (LSP) in marmosets experimentally infected with the hepatitis A virus. Clin Exp Immunol 55:535-540. Karayiannis P et al. 1989. Studies of GB hepatitis agent in tamarins. Hepatology 9:186-192. Keenan CM et al. 1984. Pathology of hepatitis A infection in the owl monkey (Aotus trivirgatus). Am J Pathol 115:1-8. Keenan CM et al. 1987. Animal models of human disease: Hepatitis A infection in the owl monkey. Comp Pathol Bull 19:3-5. Kessler H. et al. 1982. Hepatitis A and B at the London Zoo. J Infect 4:63-67. Korba BE et al. 1986. Hepadnavirus infection of peripheral blood lymphocytes in vivo: Woodchuck and chimpanzee models of viral hepatitis. J Virol 58:1-8. Kornegay RW et al. 1985. Subacute nonsuppurative hepatitis associated with hepatitis B virus infection in two cynomolgus monkeys. Lab Anim Sci 35:400-404. Kos A et al. 1987. Biological and molecular characterization of the hepatitis delta virus. Prog Clin Biol Res 234:83-87. Krawczynski K et al. 1979. Immunopathologic aspects of the HBsAG carrier state in chimpanzees. J Med Primatol 8:222-232. Krawczynski KK et al. 1981. Pathogenetic aspects of hepatitis A virus infection in enterally inoculated marmosets. Am J Clin Pathol 76:698-706. Lankas GR, Jensen RD. 1987. Evidence of hepatitis A infection in immature rhesus monkeys. Vet Pathol 24:340-344. Lapin BA, Shevtsova ZV. 1989. Sensitivity of the Old World monkeys to hepatitis A virus (spontaneous and experimental infection) [letter] Exp Pathol 36:63-64. LeDuc JW, Escajadillo A, Lemon SM. 1981. Hepatitis A virus among captive Panamanian owl monkeys (letter). Lancet 2(8260-61):1427-1428. LeDuc JW et al. 1983. Experimental infection of the New World owl monkey (Aotus trivirgatus) with hepatitis A virus. Infect Immun 40:766-772. Lemon SM, Binn LN. 1983. Serum neutralizing antibody response to hepatitis A virus. J Infect Dis 148:1033-1039. Lemon SM et al. 1982. Transmission of hepatitis A virus among recently captured Panamanian owl monkeys. J Med Virol 10:25-36. Lemon SM. 1985. Type A hepatitis: New developments in an old disease. N Engl J Med 313:1059-1067. Lemon SM. 1987. Genomic heterogeneity among human and nonhuman strains of hepatitis A virus. J Virol 61:735-742. Lucke VM, Bennett AM. 1982. An outbreak of hepatitis in marmosets in a zoological collection. Lab Anim 16:73-77. Maeda T et al. 1989. Production of antibodies directed against microtubular aggregates in hepatocytes of chimpanzees with non-A, non-B hepatitis. J Gen Virol 70:1401-1407. Mao JS et al. 1981. Susceptibility of monkeys to human hepatitis A virus. J Infect Dis 144:55-60. Mascoli CC et al. 1973. Recovery of hepatitis agents in the marmoset from human cases occurring in Costa Rica. Proc Soc Exp Biol Med 142:276-282. Maynard JE. 1972. Experimental infection of chimpanzees with the virus of hepatitis B. Nature (Lond) 237:514-515. Negro F et al. 1988. Chronic hepatitis D virus (HDV) infection in hepatitis B virus carrier chimpanzees experimentally superinfected with HDV. J Infect Dis 158:151-159. Pisi E et al. 1987. Autoantibodies in chronic hepatitis delta virus infection. Prog Clin Biol Res 234:249-256. Polesky HF, Hanson MR. 1989. Transfusion-associated hepatitis C virus (non-A, non-B) infection. Arch Pathol Lab Med 113:232-235. [Published erratum appears in Arch Pathol Lab Med 1989; 113(4):368] Ponzetto A et al. 1987. Hepatitis delta virus infection in Pekin ducks chronically infected by the duck hepatitis B virus. Prog Clin Biol Res 234:47-49. Provost PJ et al. 1973. Etiologic relationship of marmoset-propagated CR326 hepatitis A virus to hepatitis in man. Proc Soc Exp Biol Med 142:1257-1267. Purcell R H et al. 1987. Experimental hepatitis delta virus infection in the chimpanzee. Prog Clin Biol Res 234:27-36. Rizzetto M, Verme G. 1985. Delta hepatitis--present status. J Hepatol 1:187-93. Rizzetto M. 1989. Hepatitis delta virus (HDV) infection and disease. Ric Clin Lab 19:11-26. Robbins SL, Cotran RS, Kumar V. Pathologic Basis of Disease, 3rd ed.; WB Saunders Co., Philadelphia, 1984; pp. 900-910. Robinson WS. 1979. Viruses of human hepatitis A and B. Comp Virol 14:471-526. Scalise G et al. 1980. Transmission of hepatitis B to rhesus monkeys. J Med Primatol 7:114-118. Scheuer PJ. 1989. Non-A, non-B hepatitis. Virchows Arch [A] 415:301-303. Schulman AN et al. 1976. Hepatitis A antigen particles in liver, bile and stool of chimpanzees. J Infect Dis 134:80. Shao-Nan Huang L et al. 1979. Electron and immunoelectron microscopic study on liver tissues of marmosets infected with hepatitis A virus. Lab Invest 41:63-71. Shevtsova ZV et al. 1988. Spontaneous and experimental hepatitis A in Old World monkeys. J Med Primatol 17:177-194. Shouval D et al. 1980. Chronic hepatitis in chimpanzee carriers of hepatitis virus: Morphologic, immunologic, and viral DNA studies. Proc Natl Acad Sci USA 77:6147-6151. Slaughter RL. 1985. Viral infections of the liver. Ala J Med Sci 22:164-168. Smith MS et al. 1980. Hepatitis A in nonhuman primates in nature (letter). Lancet 1(8206):1241-1242. Tabor E et al. 1983. Primate animal models and titered inocula for the study of human hepatitis A, hepatitis B, and non-A, non-B hepatitis. Med Primatol 12:305-318. Trahan CJ et al. 1987. Induced oral infection of the owl monkey (Aotus trivirgatus) with hepatitis A virus. Lab Anim Sci 37:45-50. Tsiquaye KN et al. 1984. Acute hepatitis A infection in hepatitis B chimpanzee carriers. Hepatology 4:504-509. Valenza FP, Muchmore E. 1985. The clinical chemistry of chimpanzees. II. Gamma glutamyl transferase levels in hepatitis studies. J Med Primatol 14:305-315. Zuckerman AJ et al. 1978. Hepatitis B outbreak among chimpanzees at the London Zoo. Lancet 2:652-654. Zuckerman AJ. 1978. The three types of human viral hepatitis. Bull WHO 56:1-20. Rift Valley Fever A hepatitis of man and other animals enzootic in many areas of Africa. Nonhuman primates may constitute reservoirs for human infection. Agent is a Bunyavirus (Bunyaviridae). Cosgriff TM et al. 1989. Hemostatic derangement produced by Rift Valley fever virus in rhesus monkeys. Rev Infect Dis 11:(Suppl 4):S807-S814. Johnson BK et al. 1982. Marburg, Ebola and Rift Valley Fever virus antibodies in East African primates. Trans R Soc Trop Med Hyg 76:307-310. Poliomyelitis 1. Agent: Human poliomyelitis viruses type 1, 2 and 3 - genus Enterovirus, family Picornaviridae. 2. Epizootiology: Macaques, African green monkeys, and Cebus spp. are susceptible to infection but not to natural paralytic disease. Naturally occurring paralytic disease, due to infections probably acquired via human contact, has been reported in chimpanzees, orangutans, gorillas, and colobus monkeys. 3. Clinical: Fever, partial or complete flaccid paralysis of one or more limbs. 4. Pathology: Classic lesion is poliomyelitis with degeneration and necrosis of anterior horn cells. Lesions can occur in other parts of spinal gray matter, brain stem, cerebellum, and meninges. 5. Diagnosis: Signs and lesions, virus isolation, serology. 6. Control: Apes can be vaccinated with human polio vaccine. 7. References: Allmond BW et al. 1967. Paralytic poliomyelitis in large laboratory primates. Virologic investigation and report on the use of oral poliomyelitis virus (OPV) vaccine. Am J Epidemiol 85:229-239. Bodian D. 1972. Poliomyelitis. In Pathology of the Nervous System, vol 3. Ed. Minckler J, McGraw-Hill, New York, pp. 2323-2344. Craig E et al. 1958. Contact transmission of poliomyelitis among monkeys. Proc Soc Exp Biol Med 99:325-329. Douglas J et al. 1970. The incidence of polio virus in chimpanzees (Pan troglodytes). Lab Anim Care 20:265-268. Guilloud NB et al. 1966. Paralytic poliomyelitis in the gorilla and orangutan. J Am Phys Ther A 46:516-518. Guilloud NB et al. 1969. Paralytic poliomyelitis in laboratory primates. J Am Vet Med Assoc 155:1190-1193. Howe HA, Bodian D. 1948. Poliomyelitis in the cynomolgus monkey following oral inoculation. Am J Hyg 48:99-106. Howe HA, Bodian D. 1944. Poliomyelitis by accidental contagion in the chimpanzee. J Exp Med 80:383-390. Howe HA, Bodian D. 1941. Poliomyelitis in the chimpanzee: A clinical-pathological study. Bull Johns Hopkins Hosp 69:149-181. Paul JR. 1971. A History of Poliomyelitis. Yale University Press, New Haven. Suleman MA et al. 1984. An outbreak of poliomyelitis caused by poliovirus type I in captive black and white colobus monkeys (Colobus abyssinicus kikuyuensis) in Kenya. Trans R Soc Trop Med Hyg 78:665-669. Papovaviridae 1. Papillomatosis. Hollander CF, Van Noord MJ. 1972. Focal epithelial hyperplasia: A virus-induced oral mucosal lesion in the chimpanzee. Oral Surg Oral Med Oral Pathol 33:220-26. Lucke B et al. 1950. Transmissible papilloma in monkeys (brown Cebus). Fed Proc 9:337. Rangan SRS et al. 1980. Virus associated papillomas in colobus monkeys (Colobus guereza). Lab Anim Sci 30:885-889. Tate CL et al. 1973. Focal epithelial hyperplasia in the oral mucosa of a chimpanzee. J Am Vet Med Assoc 163:619-621. 2. SV40 Usually inapparent in rhesus monkeys, the natural hosts; others are susceptible. Sarcomas and ependymomas in neonatal hamsters and mastomys. Once a contaminant of human polio vaccine. Gerber P. 1967. Patterns of antibodies to SV40 in children following the last booster with inactivated poliomyelitis vaccines. Proc Soc Exp Biol NY 121:303-307. Holmberg CA et al. 1977. Isolation of simian virus 40 from rhesus monkeys with spontaneous progressive multifocal leukoencephalopathy. J Infect Dis 136:593-596. London WT et al. 1983. Hydrocephalus, seizures, and incoordination in rhesus monkeys infected in utero with SV-40 virus (Abstract). Lab Anim Sci 33:482-483. Sheffield W. 1980. Simian virus 40 associated fatal interstial pneumonia and renal tubular necrosis in a rhesus monkey. J Infect Dis 142:618-622. 3. Progressive multifocal leukoencephalopathy (PML) A progressive neurologic disease originally described in human patients; also thought to be caused by papovavirus in humans. Gribble DH et al. 1975. Spontaneous progressive multifocal leukoencephalopathy (PML) in macaques. Nature 254:602-604. Holmberg CA et al. 1977. Isolation of simian virus 40 from rhesus monkeys with spontaneous progressive multifocal leukoencephalopathy. J Infect Dis 136:593-596. 4. Miscellaneous Takemoto KK, Segawa K. 1983. A new monkey lymphotropic papovavirus: Characterization of the virus and evidence of a related virus in humans. Prog Clin Biol Res 10:587-596. Takemoto KK et al. 1982. Biological and biochemical studies of African green monkey lymphotropic papovavirus. J Virol 42:502-509. Rotavirus Reoviridae. Many infections described, but disease rare; expect mostly in neonates. References: Ashley CR et al. 1978. Rotavirus infections of apes. Lancet 2:477. Hrdy B. 1982. Rotavirus antibodies in hanaman langurs (Presbytis entellus). J Med Primatol 11:35-38. Kalter SS et al. 1983. Infection of baboons (Papio cynocephalus) with rotavirus (SV- 11). Dev Biol Stand 53:257-261. Kalter SS. 1979. Electron microscopic examination of primate feces for rotaviruses. Lab Anim Sci 29:516-518. Kalter SS et al. 1982. Rotavirus (SA 11) antibody in nonhuman primates. Lab Anim Sci 32:291-293. Majer M et al. 1978. Diarrhea in newborn cynomologus monkeys infected with human rotavirus. Infection 6:71-72. Malherbe HH, Strickland-Cholmley M. 1967. Simian virus SA 11 and the related 0 agent. Arch ges Virusforsch 22:235-245. Mitchell J et al. 1977. Transmission of rotavirus gastroenteritis from children to a monkey. Gut 18:156-160. Soike KF et al. 1980. Susceptibility of nonhuman primate species to infection by simian rotavirus SV-11. AmJ Vet Res 41:1098-1103. Stuker G et al. 1979. Virus detection in monkeys with diarrhea: The association of adenoviruses with diarrhea and the possible role of rotaviruses. Lab Anim Sci 29:610- 616. Wyatt RG et al. 1976. Induction of diarrhea in colostrum-deprived newborn rhesus monkeys with the human reovirus-like agent of infantile gastroenteritis. Arch Virol 50:17-27. Wyatt RG et al. 1978. Experimental infection of chimpanzees with the Norwalk agent of epidemic viral gastroenteritis. J Med Virol 2:89-96. Encephalomyocarditis A cardiovirus (Picornaviridae). Encephalomyocarditis virus can cause necrotizing and interstitial myocarditis in gibbons and owl monkeys. Rhesus sometimes have encephalomyelitis and paralysis; may recover. Worldwide distribution, rats probably natural host. Simian disease in North and South America and Africa. Lesions: Cardiac dilation, pericardial effusion, pulmonary edema, pleural effusion. Gainer JH 1967. Encephalomyocarditis virus infection in Florida. J Am Vet Med Assoc 151:421-425. Helwig FC, Schmidt ECH. 1945. A filter-passing agent producing interstitial myocarditis in anthropoid apes and small animals. Science 102:31-33. Roca-Garcia M, Sanmartin-Barberi C. 1957. The isolation of encephalomyocarditis virus from Aotus monkeys. Am J Trop Med 6:840-852. Simian Acquired Immunodeficiency Syndrome (SAIDS) 1. Historical: Human AIDS was first reported by CDC in 1981. Simian AIDS-like conditions have been recognized in New England, California, Washington, Oregon, and Delta Regional Primate Research Centers. 2. Cause: Retroviral, probably several different ones. The family Retroviridae is divided into three subfamilies. The Oncovirinae include three unnamed genera comprising the types B, C, and D viruses. The Spumavirinae (foamy viruses) include the single genus Spumavirus, and the Lentivirinae (slow viruses) include the single genus Lentivirus. Morphology and gene organization are major criteria for classification. Typical oncoviruses have a more nearly round (eicosahedral) core than lentiviruses, which have a cylindrical or bar-shaped core, although some type D viruses have an eccentric elongated core intermediate between that of a typical oncovirus and that of a lentivirus. The basic distinction between types C and D viruses is that the core of type D particles forms in the cytoplasm first, then migrates to the cell membrane where the viral envelope forms and budding occurs. The core of type C particles, on the other hand, forms as the envelope assembles and budding occurs. Size, growth characteristics, antigenic relationships, nucleic acid homology, and host range also are used in various classification schemes. Human AIDS is caused by HIV-1 (human immunodeficiency virus-1), a lentivirus. Synonyms include LAV (lymphadenopathy virus), and HTLV-III (human T- lymphotropic virus-III). HIV-2 (HTLV-IV), another human lentivirus, is found only in west Africa. It appears to be less pathogenic than HIV-1; most people from whom it has been recovered were apparently healthy. SAIDS also is associated with retroviral infections, but information concerning the specific agent or agents responsible is somewhat confused. Lentiviruses have been isolated from macaques with SAIDS at NERPRC, CRPRC, and WRPRC. These agents are closely related genetically and are now generally considered to cause SAIDS in macaques. They currently are designated SIVmac (simian immunodeficiency virus of macaques). In terms of sequence homology in gag and pol genes and in gene organization, SIVmac viruses are more closely related to HIV-2 than to HIV-1. Another simian lentivirus, SIVsmm (STLV-III/Delta), is closely related to SIVmac and apparently is prevalent in captive sooty mangabeys (Cercocebus atys). No evidence of immunodeficiency disease has been found in the mangabeys; however, macaques inoculated with lepromatous material from sooty mangabeys developed combinations of lymphoma, CMV disease, candidiasis, cryptosporidiosis, wasting, lymphoid atrophy, marrow hyperplasia, amyloidosis, gastritis, hepatitis, and syncytia in the meninges, lung, liver, lymph nodes, kidney, and gastrointestinal tract. This appears to have been due to SIVsmm infection in the macaques. Still another nonhuman primate lentivirus, SIVagm (STLV-IIIagm), has been isolated from apparently healthy wild-caught African green monkeys, and serologic evidence indicates that infection is common. It has not been associated with disease. It cross-reacts serologically with HIV-1 and HIV-2, but based on sequence homologies SIVagm viruses form a group distinct from HIV-1 viruses and from the HIV- 2/SIVmac/SIVsmm group. An additional SIV has been found in mandrills (Papio sphinx), but its prevalence and pathogenicity are unknown. Knowledge of the cause(s) of SAIDS is clouded by the isolation of type D retroviruses from affected monkeys at NERPRC, CRPRC, WRPRC, and ORPRC. The relationships among these agents have not been clarified, and it is unclear whether or not any of them can cause or contribute to SAIDS. The New England type D agent did not cause disease after experimental inoculation. The California virus, which is easily transmitted and which is reported to cause SAIDS-like disease experimentally, is referred to as SRV-1 (simian retrovirus-1). SRV-1 has antigenic similarities to Mason-Pfizer monkey virus. The Washington type D virus, which is antigenically similar to SRV-1, is called SRV-2; it appears to cause a fibroblastic proliferative disease affecting mostly the abdominal viscera and called retroperitoneal fibromatosis. The Oregon virus is reported to be serologically similar to, but nonetheless distinct from, the California virus; its relationship to SRV-1 has not been established. Thus, there are at least 2, and possibly more, type D retroviruses that have been associated with SAIDS in addition to the aforementioned lentiviruses. 3. Epizootiology: SAIDS is most common in rhesus and other macaques. Natural reservoir, mode of transmission, incubation period, etc., are not fully understood. It is possible that lentiviruses that cause immunodeficiency diseases are derived from lentiviruses that are not pathogenic in the natural host species. However, up to 50% of the African green monkeys in their native habitat (sub-Saharan Africa) have antibodies to SIVagm, up to 80% of captive sooty mangabeys have titers to SIVsmm. The widespread occurrence of SIVs in these species without apparent adverse effects suggests a long evolutionary relationship. Macaques appear not to have SIV infections in the wild; SIVmac is closely related to SIVsmm. Simian immunodeficiency viruses replicate well in cultured human T cells, and the area of Africa in which HIV-2 (closely related to SIVsmm) is endemic corresponds to the range of sooty mangabeys. At CRPRC the California type D virus has been found in saliva and urine of a healthy carrier, and infection was transmitted by inoculation of infected saliva. 4. Clinical signs and laboratory results: Human AIDS is characterized by lymphoid depletion, depressed cellular and humoral immunity, opportunistic infections, and neoplasms such as Kaposi's sarcoma (endothelial origin) and lymphoma. There usually is lymphopenia and decreased ratio of T4+ (helper-inducer) lymphocytes to T8+ (suppressor-cytotoxic) lymphocytes. Human patients usually have normal or increased immunoglobulins. Some patients have CNS signs. Signs of SAIDS are not specific and usually are related to secondary infectious conditions. M. cyclopis at NERPRC in 1980-81 had anemia, neutropenia, bizarre circulating mononuclear cells, increased hepatic enzymes, decreased serum albumin, hypogammaglobulinemia, wasting, diarrhea, lymphoma, and diseases such pneumocystosis or noma. Responses of their lymphocytes to mitogens and mixed lymphocyte culture were decreased, but their lymphocyte counts and T4/T8 ratios were normal. Rhesus monkeys from which SIV was isolated did, however, have decreased numbers of T4+ lymphocytes. Naturally infected monkeys had lymphoma, opportunistic infections, or both. Monkeys inoculated with lymphoma tissue or plasma died rapidly with conditions such as candidiasis, generalized cytomegalovirus disease, cryptosporidiosis, intestinal trichomoniasis, and encephalitis. At the California RPRC, 42 rhesus monkeys were found to have lymphoma and concurrent progressive multifocal leukoencephalopathy or disease due to M. avium- intracellulare or Herpesvirus simiae from 1965-1975. A second outbreak began in 1976 in stumptailed macaques, and featured high mortality with encephalitis, oral candidiasis, and enteric mycobacteriosis (M. avium). In 1976-1981, a third outbreak affected rhesus monkeys and was characterized by generalized lymphadenopathy, severe anemia and lymphopenia, secondary bacterial infections, and high mortality. A fourth outbreak began in 1981 and continues at present. At the Washington center, SAIDS has affected primarily pigtail macaques but also other species of macaques. These animals had persistent diarrhea, progressive weight loss, anemia, lymphopenia, opportunistic infections, and a high incidence of retroperitoneal fibromatosis, a neoplasm-like condition whose cells bear some similarities to those of Kaposi's sarcoma. The ORPRC retrospectively recognized a SAIDS-like condition seen as early as 1978 in Celebes black macaques and rhesus monkeys. These animals had weight loss, diarrhea, anemia, lymphopenia, lymphadenopathy, splenomegaly, hypoproteinemia, and retroperitoneal fibromatosis. 5. Pathology: Lesions related specifically to SAIDS include lymphoid hyperplasia followed by progressive depletion. Some monkeys have had lymphoproliferative lesions such as infiltrates in liver, kidney, and bone marrow, or outright lymphosarcoma. Encephalitis similar to that occurring in human AIDS (perivascular granulomatous inflammation with multinucleated giant cells) occurs in some cases. Other lesions result from infections with CMV, SV40, Pneumocystis carinii, mycobacteria, Cryptosporidium, Candida, and other agents. The pathogenesis of the disease still is unclear; however, the California group has reported that monkeys dying of fulminant SAIDS after inoculation with SRV-1 had much circulating virus and did not have detectable antibody responses to the virus. Monkeys that developed a milder form of the disease had chronic mild viremia, and those that did not develop signs of SAIDS had no or transient viremia and high concentrations of antibody against the virus. Chakrabarti et al (1987) report that SIV causes disease more similar to human AIDS in that it also is tropic for lymphocytes of the T helper phenotype. 6. Diagnosis: serologic techniques, such as indirect IFA and ELISA; immunoblotting; virus isolation & identification 7. Control: Isolation. May be difficult because of long incubation period, and best measures not really known because transmission not fully understood. 8. References: _____. 1988. Animal models for HIV infection and AIDS: memorandum from a WHO meeting. Bull World Health Organ 66:561-574. _____. 1988. Guidelines to prevent simian immunodeficiency virus infection in laboratory workers and animal handlers. MMWR 37:693-694, 699-704. _____. 1989. Animal models for HIV infection and AIDS. Bull Pan Am Health Organ 23:215-234. _____. 1989. Nonhuman primate models for AIDS. San Antonio, Texas, November 16-18, 1988. Proceedings. J Med Primatol 18:165-369. Adams SW. 1987. Simian T-lymphotropic viruses. Lab Animal 16:33-38. Arthur LO et al. 1986. Simian acquired immunodeficiency syndrome. Prog Allergy 37:332-352. Arya SK et al. 1987. New human and simian HIV-related retroviruses possess functional transactivator (tat) gene. Nature 328:548-550. Baskin GB et al. 1986. Transmissible lymphoma and simian acquired immunodeficiency syndrome in rhesus monkeys. JNCI 77:127-139. Baskin GB et al. 1988. Necropsy findings in rhesus monkeys experimentally infected with cultured simian immunodeficiency virus (SIV)/delta. Vet Pathol 25:456-467. Baskin GB, Soike KF. 1989. Adenovirus enteritis in SIV-infected rhesus monkeys [letter] J Infect Dis 160:905-907. Benveniste RE et al. 1986. Isolation of a lentivirus from a macaque with lymphoma: Comparison with HTLV-III/LAV and other lentiviruses. J Virol 60:483-490. Benveniste RE et al. 1988. Inoculation of baboons and macaques with simian immunodeficiency virus/Mne, a primate lentivirus closely related to human immunodeficiency virus type 2. J Virol 62:2091-2101. Benveniste RE et al. 1989. Molecular characterization and comparison of simian immunodeficiency virus isolates from macaques, mangabeys, and African green monkeys. J Med Primatol 18:287-303. Blanchard JL et al. 1987. Disseminated cryptosporidiosis in simian immunodeficiency virus/delta-infected rhesus monkeys. Vet Pathol 24:454-456. Budzko DB et al. 1985. Immunologic alterations in monkeys with simian acquired immunodeficiency syndrome (SAIDS). Proc Soc Exp Biol Med 179:227-231. Castro BA et al. 1989. Human immunodeficiency virus (HIV) from experimentally infected chimpanzees: isolation and characterization. J Med Primatol 18:337-342. Chakrabarti L et al. 1987. Sequence of simian immunodeficiency virus from macaques and its relationship to other human and simian retroviruses. Nature 328:543-547. Chalifoux LV et al. 1984. Morphologic changes in lymph nodes of macaaques with an immunodeficiency syndrome. Lab Invest 51:22-26. Chalifoux LV et al. 1986. Lymphoproliferative syndrome in an immunodeficient rhesus monkey naturally infected with an HTLV-III-like virus (STLV-III). Lab Invest 55:43- 50. Chalifoux LV et al. 1987. Lymphadenopathy in macaques experimentally infected with the simian immunodeficiency virus (SIV). Am J Pathol 128:104-110. Dalakas MC et al. 1987. Morphological changes of an inflammatory myopathy in rhesus monkeys with simian acquired immunodeficiency syndrome. Proc Soc Exp Biol Med 185:368-376. Daniel MD et al. 1984. A new type D retrovirus from macaques with an immunodeficiency syndrome. Science 223:602-605. Daniel MD et al. 1985. Isolation of T-cell tropic HTLV-III-like retrovirus from macaques. Science 228:1201-1204. Daniel MD et al. 1987. Long-term persistent infection of macaque monkeys with the simian immunodeficiency virus. J Gen Virol 68:3183-3189. Daniel MD et al. 1988. Prevalence of antibodies to 3 retroviruses in a captive colony of macaque monkeys. Int J Cancer 41:601-608. Daniel MD et al. 1988. Simian immunodeficiency virus from African green monkeys. J Virol 62:4123-4128. Desrosiers RC, Daniel MD, Li Y. 1989. Minireview: HIV-related lentiviruses of nonhuman primates. AIDS Res Hum Retroviruses 5:465-473. Desrosiers RC et al. 1985. Retrovirus D/New England and its relation to Mason-Pfizer monkey virus. J Virol 54:552-560. Desrosiers RC et al. 1989. Vaccine protection against simian immunodeficiency virus infection. Proc Natl Acad Sci U S A 86:6353-6357. Desrosiers RC. 1988. Simian immunodeficiency viruses. Annu Rev Microbiol 42:607-625. Doolittle RF. 1989. Immunodeficiency viruses: the simian-human connection [news] Nature 339:338-339. Dormont D et al. 1989. HIV-2 in rhesus monkeys: serological, virological and clinical results. Intervirology 30(Suppl 1):59-65. Eichberg JW et al. 1985. Longitudinal study of HTLV-III-infected chimpanzees by lymphocyte subpopulation analysis. J Med Primatol 14:317-326. Eichberg JW et al. 1988. In utero infection of an infant chimpanzee with HIV [letter] N Engl J Med 319:722-723. Essex M, Kanki PJ. 1988. The origins of the AIDS virus. Sci Am 259:64-71. Fauci AS. 1982. The syndrome of Kaposi's sarcoma and opportunistic infections: An epidemiologically restricted disorder of immunoregulation. Ann Intern Med 96:777-779. Fincham JE et al. 1989. Increased peripheral lymphocytes, lymphoid hepatitis and anaemia in African vervet monkeys seropositive to retroviruses. J Comp Pathol 101:53-68. Franchini G, Bosch ML. 1989. Genetic relatedness of the human immunodeficiency viruses type 1 and 2 (HIV-1, HIV-2) and the simian immunodeficiency virus (SIV). Ann N Y Acad Sci 554:81-87. Franchini G et al. 1987. Sequence of simian immunodeficiency virus and its relationship to the human immunodeficiency viruses. Nature 328:539-543. Franchini G. 1989. Human and non-human primate immunodeficiency viruses: genetic relatedness and phylogeny. Curr Opin Immunol 1:513-515. Fukasawa M et al. 1988. Sequence of simian immunodeficiency virus from African green monkeys, a new member of the HIV/SIV group. Nature 333:457-461. Fultz PN et al. 1986. Isolation of a T-lymphotropic retrovirus from naturally infected sooty mangabey monkeys (Cercocebus atys). Proc Natl Acad Sci USA 83:5286-5291. Fultz PN et al. 1986. Persistent infection of chimpanzees with human T-lymphotropic virus type III/lymphadenopathy-associated virus: A potential model for acquired immunodeficiency syndrome. J Virol 58:116-124. Fultz PN et al. 1986. Vaginal transmission of human immunodeficiency virus (HIV) to a chimpanzee. J Infect Dis 154:896-899. Fultz PN et al. 1989. HIV infection of chimpanzees as a model for testing chemotherapeutics. Intervirology 30 Suppl 1:51-58. Fultz PN et al. 1989. Identification and biologic characterization of an acutely lethal variant of simian immunodeficiency virus from sooty mangabeys (SIV/SMM). AIDS Res Hum Retroviruses 5:397-409. Fultz PN. 1989. Nonhuman primates and the acquired immunodeficiency syndrome: a union of necessity. J Med Primatol 18:73-83. Gajdusek DC. 1988. Retrovirus encephalomyelitides. Ann Neurol 23(Suppl):S207. Gallatin WM et al. 1989. Selective replication of simian immunodeficiency virus in a subset of CD4+ lymphocytes. Proc Natl Acad Sci U S A 86:3301-3305. Gallo RC, Montagnier L. 1988. AIDS in 1988. Sci Am 259:41-48. Gallo RC. 1987. The AIDS virus. Sci Am 256:46-73. Gardner M et al. 1984. Simian AIDS--evidence for a retroviral etiology. Hematol Oncol 2:259-268. Gardner M, Luciw P. 1988. Simian immunodeficiency viruses and their relationship to the human immunodeficiency viruses. AIDS 2:S3-S10. Gardner MB et al. 1988. Nonhuman primate retrovirus isolates and AIDS. Adv Vet Sci Comp Med 32:171-226. Gardner MB et al. 1989. Postexposure immunotherapy of simian immunodeficiency virus (SIV) infected rhesus with an SIV immunogen. J Med Primatol 18:321-328. Gardner MB, Luciw PA. 1988. Simian immunodeficiency viruses and their relationship to the human immunodeficiency viruses. Aids 2(Suppl 1):S3-S10. Giddens WE Jr et al. 1983. Enzootic retroperitoneal fibromatosis in Macaca sp. In Viral and Immunologic Diseases in Nonhuman Primates; Alan R Liss, Inc, New York. Giddens WE Jr et al. 1985. Retroperitoneal fibromatosis and acquired immunodeficiency syndrome in macaques. Pathologic observations and transmission studies. Am J Pathol 119:253-263. Gilden RV et al. 1986. HTLV-III antibody in a breeding chimpanzee not experimentally exposed to the virus (letter). Lancet Mar. 22, 1986:679. Gormus BJ et al. 1989. Interactions between simian immunodeficiency virus and Mycobacterium leprae in experimentally inoculated rhesus monkeys. J Infect Dis 160:405-413. Goudsmit J et al. 1987. IgG response to human immunodeficiency virus in experimentally infected chimpanzees mimics the IgG response in humans. J Infect Dis 155:327-330. Gravell M et al. 1984. Transmission of simian acquired immunodeficiency syndrome (SAIDS) with type D retrovirus isolated from saliva or urine. Proc Soc Exp Biol Med 177:491-494. Gravell M et al. 1984. Transmission of simian acquired immunodeficiency syndrome (SAIDS) with blood or filtered plasma. Science 223:74-76. Gravell M et al. 1989. Infection of macaque monkeys with simian immunodeficiency virus from African green monkeys: virulence and activation of latent infection. J Med Primatol 18:247-254. Gravell M. 1988. Animal models of AIDS. Ann Neurol 23 Suppl:S213-S214. Heidecker G et al. 1987. Induction of simian acquired immune deficiency syndrome (SAIDS) with a molecular clone of a type D SAIDS retrovirus. J Virol 61:3066-3071. Hendry RM et al. 1986. Antibodies to simian immunodeficiency virus in African green monkeys in Africa in 1957-62 [letter]. Lancet 2(8504):455. Henrickson RV et al. 1983. Epidemic of acquired immunodeficiency in rhesus monkeys. Lancet Feb 19, 1983:388-390. Henrickson RV et al. 1984. Clinical features of simian acquired immunodeficiency syndrome (SAIDS) in rhesus monkeys. Lab Anim Sci 34:140-145. Herchenroder O et al. 1989. Experimental infection of rhesus monkeys with SIV isolated from African green monkeys. Intervirology 30(Suppl 1):66-72. Hirsch V et al. 1986. Cross-reactivity to human T-lymphotropic virus type III/lymphadenopathy-associated virus and molecular cloning of simian T-cell lymphotropic virus type IV from African green monkeys. Proc Natl Acad Sci USA 83:9754-9758. Hirsch VM et al. 1987. The genome organization of STLV-3 is similar to that of the AIDS virus except for a truncated transmembrane protein. Cell 49:307-319. Hirsch VM et al. 1989. An African primate lentivirus (SIVsm) closely related to HIV-2. Nature 339:389-392. Hirsch VM et al. 1989. Molecular cloning of SIV from sooty mangabey monkeys. J Med Primatol 18:279-285. Hirsch VM et al. 1989. SIV adaptation to human cells [letter] Nature 341:573-574. Holmberg CA et al. 1985. Immunologic abnormality in a group of Macaca arctoides with high mortality due to atypical mycobacterial and other disease processes. Am J Vet Res 46:1192-1196. Holmberg CA et al. 1985. Malignant lymphoma in a colony of Macaca arctoides. Vet Pathol 22:42-45. Homma T et al. 1984. Lymphoma in macaques: Association with virus of human T lymphotropic family. Science 225:716-718. Hu SL et al. 1989. Protection of macaques against simian AIDS by immunization with a recombinant vaccinia virus expressing the envelope glycoproteins of simian type D retrovirus. Proc Natl Acad Sci U S A 86:7213-7217. Hunt R et al. 1983. Transmission of naturally occurring lymphoma in macaque monkeys. Proc Natl Acad Sci USA 80:5085-5089. Jacobs JL, Murray HW. 1986. Immunologic developments in AIDS--1985. In: Year in Immunology, vol. 2. Ed. Cruise JM, Lewis RE; S Karger (Basel), pp 323-328. Jacobs JL. 1989. Immunologic developments in AIDS--1987. Year Immunol 4:276-285. Johnson PR et al. 1989. Genetic diversity among simian immunodeficiency virus isolates from African green monkeys. J Med Primatol 18:271-277. Kanki PJ et al. 1985. Antibodies to simian T-lymphotropic retrovirus type III in African green monkeys and recognition of STLV-III viral proteins by AIDS and related sera. Lancet 1:1330-1332. Kanki PJ et al. 1985. Isolation of T-lymphotropic retrovirus related to HTLV-III/LAV from wild-caught African green monkeys. Science 230:951-954. Kanki PJ et al. 1985. Serologic identification and characterization of a macaque T- lymphotropic retrovirus closely related to HTLV-III. Science 228:1199-1201. Kanki PJ et al. 1986. New human T-lymphotropic retrovirus related simian T- lymphotropic virus type III (STLV-IIIagm). Science 232:238-243. Kanki PJ et al. 1987. Relationship of simian T-lymphotropic virus type III to human retroviruses in Africa. Antibiot Chemother 38:21-27. Kannagi M et al. 1986. Humoral immune response to T cell tropic retrovirus simian T lymphotropic virus type III in monkeys with experimentally induced acquired immune deficiency-like syndrome. J Clin Invest 78:1229-1236. Kannagi M et al. 1987. Simian immunodeficiency virus induces expression of class II major histocompatibility complex structures on infected target cells in vitro. J Virol 61:1421-1426. Kawamura M et al. 1989. Immunological reactivities of Ghanaian sera with HIV-1, HIV-2, and simian immunodeficiency virus SIVagm. Aids 3:609-611. Kestler HW III et al. 1988. Comparison of simian immunodeficiency virus isolates. Nature 331:619-622. Kestler HW III et al. 1989. Use of infectious molecular clones of simian immunodeficiency virus for pathogenesis studies. J Med Primatol 18:305-309. King NW et al. 1983. Histopathologic changes in macaques with an acquired immunodeficiency syndrome (AIDS). Am J Pathol 113:382-388. King NW. 1986. Simian models of acquired immunodeficiency syndrome (AIDS): A review. Vet Pathol 23:345-353. Kodama T et al. 1989. Prevalence of antibodies to SIV in baboons in their native habitat. AIDS Res Hum Retroviruses 5:337-343. Koenig S et al. 1989. Selective infection of human CD4+ cells by simian immunodeficiency virus: productive infection associated with envelope glycoprotein-induced fusion. Proc Natl Acad Sci U S A 86:2443-7. Kornfeld H et al. 1987. Cloning of HTLV-4 and its relation to simian and human immunodeficiency viruses. Nature 326:610-613. Kraus G et al. 1989. Isolation of human immunodeficiency virus-related simian immunodeficiency viruses from African green monkeys. Proc Natl Acad Sci U S A 86:2892-2896. Krause H, Wunderlich V, Uckert W. 1989. Molecular cloning of a type D retrovirus from human cells (PMFV) and its homology to simian acquired immunodeficiency type D retroviruses. Virology 173:214-222. Krugner-Higby L et al. 1990. Serological survey for two simian retroviruses in macaques and African green monkeys. Lab Anim Sci 40:24-28. Kurth R et al. 1988. AIDS: animal retrovirus models and vaccines. J Acquir Immune Defic Syndr 1:284-294. Kwang HS et al. 1987. Viremia, antigenemia, and serum antibodies in rhesus macaques infected with simian retrovirus type 1 and their relationship to disease course. Lab Invest 56:591-597. Lackner AA et al. 1989. Asymptomatic infection of the central nervous system by the macaque immunosuppressive type D retrovirus, SRV-1. J Gen Virol 70:1641-1651. Lackner AA et al. 1989. Mucosal epithelial cells and Langerhans cells are targets for infection by the immunosuppressive type D retrovirus simian AIDS retrovirus serotype 1. J Med Primatol 18:195-207. Lafrado LJ et al. 1989. Polymorphonuclear leukocyte function in HIV-1-infected chimpanzees. Vet Immunol Immunopathol 21:3-12. Lairmore MD et al. 1989. Guidelines for the prevention of simian immunodeficiency virus infection in laboratory workers and animal handlers. J Med Primatol 18:167-174. Lane HC et al. 1983. Abnormalities of B-cell activation and immunoregulation in patients with the acquired immunodeficiency syndrome. N Engl J Med 309:453:458. Lecatsas G et al. 1984. Morphology of the retroviruses associated with AIDS and SAIDS. Proc Soc Exp Biol Med 177:495-498. Lee MR et al. 1987. Functional interaction and partial homology between human immunodeficiency virus and neuroleukin. Science 237:1047-1051. Legrand EK et al. 1985. Monocyte function in rhesus monkeys with simian acquired immune deficiency syndrome. Vet Immunol Immunopathol 10:131-146. Lerche N et al. 1984. Epidemiologic aspects of an outbreak of acquired immunodeficiency syndrome in rhesus monkeys (Macaca mulatta). Lab Anim Sci 34:146- 150. Lerche NW et al. 1986. Inapparent carriers of simian acquired immune deficiency syndrome type D retrovirus and disease transmission with saliva. JNCI 77:489-496. Lerche NW et al. 1987. Natural history of endemic type D retrovirus infection and acquired immune deficiency syndrome in group-housed rhesus monkeys. JNCI 79:847- 854. Letvin NL et al. 1983. Acquired immunodeficiency syndrome in a colony of macaque monkeys. Proc Natl Acad Sci 80:2718-2722. Letvin NL et al. 1983. Experimental transmission of macaque AIDS by means of inoculation of macaque lymphoma tissue. Lancet 2:599-602. Letvin NL et al. 1984. Experimental infection of rhesus monkeys with type D retrovirus. J Virol 52:683-686. Letvin NL et al. 1985. Induction of AIDS-like disease in macaque monkeys with T-cell tropic retrovirus STLV-III. Science 230:71-73. Letvin NL, Hunt RD. 1984. An acquired immunodeficiency syndrome of macaque monkeys. Ann NY Acad Sci 437:121-130. Letvin NL, King NW. 1984. Clinical and pathologic features of acquired immunodeficiency syndrome (AIDS) in macaque monkeys. Adv Vet Sci Comp Med 28:237-265. Li Y et al. 1989. Extensive genetic variability of simian immunodeficiency virus from African green monkeys. J Virol 63:1800-1802. Li Y et al. 1989. Genetic diversity of simian immunodeficiency virus. J Med Primatol 18:261-269. Ljunggren K et al. 1989. Antibody-dependent cellular cytotoxicity detects type- and strain-specific antigens among human immunodeficiency virus types 1 and 2 and simian immunodeficiency virus SIVmac isolates. J Virol 63:3376-3381. London WT et al. 1983. Experimental transmission of simian acquired immunodeficiency syndrome (SAIS) and Kaposi-like skin lesions. Lancet 2(8355):869-873. Lowenstine LJ et al. 1986. Seroepidemiologic survey of captive old world primates for antibodies to human and simian retroviruses, and isolation of a lentivirus from sooty mangabeys (Cercocebus atys). Int J Cancer 38:563-574. Lusso P et al. 1988. Cell-mediated immune response toward viral envelope and core antigens in gibbon apes (Hylobates lar) chronically infected with human immunodeficiency virus-1. J Immunol 141:2467-2473. Marthas ML et al. 1989. Rhesus macaques inoculated with molecularly cloned simian immunodeficiency virus. J Med Primatol 18:311-319. Marx PA et al. 1984. Simian AIDS: Isolation of a type D retrovirus and transmission of the disease. Science 223:1083-1086. Marx PA et al. 1985. Isolation of a new serotype of simian acquired immunne deficiency syndrome type D retrovirus from Celebes black macaques (Macaca nigra) with immune deficiency and retroperitoneal fibromatosis. J Virol 56:571-578. Marx PA et al. 1986. Prevention of simian acquired immune deficiency syndrome with a formalin-inactivated type D retrovirus vaccine. J Virol 60:431-435. Maul DH et al. 1985. Immune defects in simian acquired immunodeficiency syndrome. Vet Immunol Immunopathol 8:201-214. Maul DH et al. 1988. Simian retrovirus D serogroup 1 has a broad cellular tropism for lymphoid and nonlymphoid cells. J Virol 62:1768-1773. McCarthy TJ et al. 1990. Spontaneous malignant lymphoma and leukemia in a simian T-lymphotropic virus type I (STLV-I) antibody positive olive baboon. Lab Anim Sci 40:79-81. McClure HM et al. 1989. Spectrum of disease in macaque monkeys chronically infected with SIV/SMM. Vet Immunol Immunopathol 21:13-24. McClure MO et al. 1989. Inoculation of New World primates with the human immunodeficiency virus. J Med Primatol 18:329-335. Miller CJ et al. 1989. Genital mucosal transmission of simian immunodeficiency virus: animal model for heterosexual transmission of human immunodeficiency virus. J Virol 63:4277-4284. Miura T et al. 1989. Genetic analysis and infection of SIVAGM and SIVMND. J Med Primatol 18:255-259. Montagnier L. 1988. Origin and evolution of HIVs and their role in AIDS pathogenesis. J Acquir Immune Defic Syndr 1:517-520. Morrow WJ et al. 1989. Long-term observation of baboons, rhesus monkeys, and chimpanzees inoculated with HIV and given periodic immunosuppressive treatment. AIDS Res Hum Retroviruses 5:233-245. Morton WR et al. 1989. Transmission of the simian immunodeficiency virus SIVmne in macaques and baboons. J Med Primatol 18:237-245. Munn RJ et al. 1985. Ultrastructural comparison of the retroviruses associated with human and simian acquired immunodeficiency syndromes. Lab Invest 53:194-199. Murphey-Corb M et al. 1986. Isolation of an HTLV-III related retrovirus from macaques with simian AIDS and its possible origin in asymptomatic mangabeys. Nature 321:425-437. Naidu YM et al. 1988. Characterization of infectious molecular clones of simian immunodeficiency virus (SIVmac) and human immunodeficiency virus type 2: persistent infection of rhesus monkeys with molecularly cloned SIVmac. J Virol 62:4691-4696. Nara P et al. 1989. The biology of human immunodeficiency virus-1 IIIB infection in the chimpanzee: in vivo and in vitro correlations. J Med Primatol 18:343-355. Narayan O, Clements JE. 1989. Biology and pathogenesis of lentiviruses. J Gen Virol 70:1617-1639. Nicol I et al. 1989. Persistent HIV-2 infection of rhesus macaque, baboon, and mangabeys. Intervirology 30:258-267. Nicol I et al. 1989. Use of Old World monkeys for acquired immunodeficiency syndrome research. J Med Primatol 18:227-236. Ohta Y et al. 1988. Isolation of simian immunodeficiency virus from African green monkeys and seroepidemiologic survey of the virus in various nonhuman primates. Int J Cancer 41:115-122. Osborn KG et al. 1984. The pathology of an epizootic of acquired immunodeficiency in rhesus macaques. Am J Pathol 114:94-103. Powell JD et al. 1989. Phenotypic and functional differences in NK and LAK cells in the peripheral blood of sooty mangabeys and rhesus macaques. Cell Immunol 124:107-118. Prince AM et al. 1988. Chimpanzees and AIDS research. Nature 333:513. Prince AM et al. 1988. Failure of a human immunodeficiency virus (HIV) immune globulin to protect chimpanzees against experimental challenge with HIV. Proc Natl Acad Sci U S A 85:6944-6948. Putkonen P et al. 1989. Experimental infection of cynomolgus monkeys (Macaca fascicularis) with HIV-2. J Acquir Immune Defic Syndr 2:366-373. Putkonen P et al. 1989. Experimental infection of cynomolgus monkeys (Macaca fascicularis) with simian immunodeficiency virus (SIVsm). J Acquir Immune Defic Syndr 2:359-365. Ringler DJ et al. 1987. Immunophenotypic characterization of the cutaneous exanthem of SIV-infected rhesus monkeys: Apposition of degenerative Langerhans cells and cytotoxic lymphocytes during the development of acquired immunodeficiency syndrome. Am J Pathol 126:199-208. Ringler DJ et al. 1988. Simian immunodeficiency virus-induced meningoencephalitis: natural history and retrospective study. Ann Neurol 23(Suppl):S101-S107. Ringler DJ et al. 1989. Cellular localization of simian immunodeficiency virus in lymphoid tissues. I. Immunohistochemistry and electron microscopy. Am J Pathol 134:373-383. Ringler DJ et al. 1989. The productive infection of alveolar macrophages by simian immunodeficiency virus. J Med Primatol 18:217-226. Ringler DJ, Murphy GF, King NW Jr. 1986. An erythematous maculopapular eruption in macaques infected with an HTLV-III-like virus (STLV-III). J Invest Dermatol 87:674-677. Robinson WE Jr et al. 1989. Antibody-dependent enhancement of human immunodeficiency virus type 1 (HIV-1) infection in vitro by serum from HIV-1-infected and passively immunized chimpanzees. Proc Natl Acad Sci U S A 86:4710-4714. Schidt M et al. 1988. Oral lesions in rhesus monkeys associated with infection by simian AIDS retrovirus, serotype-I (SRV-1). Oral Surg Oral Med Oral Pathol 65:50-55. Schultz KT et al. 1989. ELISA for detection of antibodies to a type D retrovirus, SRV-W. Viral Immunol 2:47-55. Shafferman A et al. 1989. Antibody recognition of SIVmac envelope peptides in plasma from macaques experimentally infected with SIV/Mne. AIDS Res Hum Retroviruses 5:327-336. Sharer LR et al. 1988. Comparison of simian immunodeficiency virus and human immunodeficiency virus encephalitides in the immature host. Ann Neurol 23 Suppl:S108-S112. Shiigi S et al. 1989. Serologic and virologic analysis of type D simian retrovirus infection in a colony of Celebes black macaques (Macaca nigra). J Med Primatol 18:185-193. Shiigi S, Howard C Jr. 1989. Serologic and virologic analysis of type D simian retrovirus infection in a colony of Celebes black macaques [letter] J Med Primatol 18:421-422. Shiigi SM et al. 1985. Virus-associated deficiencies in the mitogen reactivity in Celebes black macaques (Macaca nigra). Clin Immonol Immunopathol 35:200-210. Shiigi SM et al. 1986. Association of SAIDS/RF-related signs with current or past SAIDS type 2 retrovirus infection in a colony of Celebes black macaques. Lab Anim Sci 36:20-23. Shiigi SM et al. 1986. Neutralizing antibody in Celebes black macaques recovering from infection with simian acquired immunodeficiency syndrome retrovirus type 2. Clin Immunol Immunopathol 40:283-290. Spinner A, Sutjipto S. 1988. Simian AIDS ELISA: sensitivity, specificity and predictive values based on a comparison with Western blot technique. Lab Anim Sci 38:568-572. Stromberg K et al. 1984. Characterization of exogenous type retrovirus from a fibroma of a macaque with simian AIDS and fibromatosis. Science 224:289-292. Terrell TG et al. 1975. Spontaneous malignant lymphoma in nonhuman primates: A morphologic study of 43 cases. Lab Invest 32:437. Terrell TG et al. 1980. Malignant lymphoma in macaques: A clinicopathologic study of 45 cases. JNCI 64:561-568. Thayer RM et al. 1987. Sequence relationships of type D retroviruses which cause simian acquired immunodeficiency syndrome. Virology 157:317-329. Tsai CC et al. 1984. Retroperitoneal fibromatosis (Kaposi's sarcoma-like tumor) with lymph node lesions in pigtail macaques. Lab Invest 50:61. Tsai CC et al. 1985. Retroperitoneal fibromatosis and acquired immunodeficiency syndrome in macaques: Epidemiologic studies. Lab Anim Sci 35:460-464. Tsai CC et al. 1986. Retroperitoneal fibromatosis and acquired immunodeficiency syndrome in macaques: Clinical and immunologic studies. Lab Anim Sci 36:119-125. Tsujimoto H et al. 1988. Isolation and characterization of simian immunodeficiency virus from mandrills in Africa and its relationship to other human and simian immunodeficiency viruses. J Virol 62:4044-4050. Tsujimoto H et al. 1989. Sequence of a novel simian immunodeficiency virus from a wild-caught African mandrill. Nature 341:539-541. van der Graaf M, Diepersloot RJA. 1986. Transmission of human immunodeficiency virus (HIV/HTLV-III): A review. Infection 14:203-211. Ward JM et al. 1987. Immunohistochemical localization of human and simian immunodeficiency viral antigens in fixed tissue sections. Am J Pathol 127:199-205. Watanabe M et al. 1989. Effect of recombinant soluble CD4 in rhesus monkeys infected with simian immunodeficiency virus of macaques. Nature 337:267-270. Weiss R et al (eds). RNA Tumor Viruses: Molcular Biology of Turor Viruses, 2nd ed. Cold Spring Harbor, Cold Spring Harbor NY, 1985. Wilson BJ et al. 1986. Relationship of mitogen reactivity to type D retrovirus infection in Celebes black macaques (Macaca nigra). Lab Anim Sci 36:237-239. Wilson BJ et al. 1986. Transmission of simian acquired immunodeficiency syndrome with a type D retrovirus: Immunological aspects. Clin Immunol Immunopathol 41:453- 460. Wyand MS et al. 1989. Cellular localization of simian immunodeficiency virus in lymphoid tissues. II. In situ hybridization. Am J Pathol 134:385-393. Wyand MS et al. 1989. Detection of simian immunodeficiency virus in macaque lymph nodes with a SIVmac envelope probe. J Med Primatol 18:209-215. Yokoyama S. 1988. Molecular evolution of the human and simian immunodeficiency viruses. Mol Biol Evol 5:645-659. Yoshimura N et al. 1988. Killer cell systems of cynomolgus monkeys experimentally infected with HTLV-1. J Immunol 141:1970-1974. Zarling JM. 1988. Primate models for evaluation of AIDS vaccines. Aids 2 Suppl 1:S113-S117. Zhang JY et al. 1988. Simian immunodeficiency virus/delta-induced immunodeficiency disease in rhesus monkeys: relation of antibody response and antigenemia. J Infect Dis 158:1277-1286. Other Retroviruses 1. Type C oncornaviruses (i) Endogenous. Integral part of genome, transmitted genetically. Low virus production, low infectivity, usually no disease in natural host species but may cause natural or experimental disease in other species. Examples: (a) Baboon endogenous virus (b) Macaca arctoides C type virus (c) Owl monkey endogenous C type virus (d) Colobus polykomos C type virus (e) Macaca mulatta C type virus (f) "Fetoplacental type C particles" seen in Old World primates (baboon, rhesus, chimp, patas, cynomolgus), New World primates (marmoset, capuchin), and prosimians (tree shrews) (g)"New World monkey type C particles" seen in howler (Alouatta sp.) and capuchin. (ii) Exogenous. Not genetically transmitted, but infectious and horizontally transmitted; potential cause of disease. Examples: (i) Simian sarcoma virus (helper) and simian sarcoma-associated virus (defective), isolated from a woolly monkey (Lagothrix sp.) with fibrosarcomas. Fibrosarcomas and gliomas in marmosets. (ii) Gibbon ape leukemia-lymphosarcoma virus, isolated from gibbons with hematopoietic neoplasms. Experimentally reproducible, but unknown whether natural disease in wild. 2. Type D oncornaviruses (i) Endogenous (a) Squirrel monkey retrovirus (squirrel monkey endogenous virus). Broad host range including man, rhesus, nonprimate mammals. No transformation in vitro. (b) Langur endogenous virus. (ii) Exogenous (i) Mason-Pfizer monkey virus. Isolated from mammary cancer in rhesus. Little or no tumorigenic activity demonstrable, but is pathogenic for infant rhesus. Three patterns observed: thymic atrophy and immuno- suppression with runting, increased susceptibility to pneumonia and enteritis, death; erythrocytic basophilia with or without anemia, death; and neutropenia with or without anemia, most survived. 3. References Ahmed M et al. 1974. Occurrence of Mason-Pfizer monkey virus in healthy rhesus monkeys. Cancer Res 34:3504-3508. Benveniste RE et al. 1974. Detection of baboon type C viral sequences in various primate tissues by molecular hybridization. J Virol 14:56-67. Bryant ML, Sherr CJ, Sen A et al. 1978. Molecular diversity among five different endogenous primate retroviruses. J Virol 28:300-313. Chopra HC. 1976. An oncorna-type virus from monkey breast tumor. Prog Med Virol 22:104-122. Chopra HC, Mason MM. 1970. A new virus in a spontaneous mammary tumor of a rhesus monkey. Cancer Res 30:2081-2086. Colcher D, Termato YA, Schlom J. 1977. Interspecies radioimmunoassay for the major structural proteins of primate type D retroviruses. Proc Natl Acad Sci 74:5739-5743. Deinhart F. 1980. Biology of primate retroviruses. In Viral Oncology, ed. Klein, Raven Press, NY. DePaoli A. 1973. Granulocytic leukemia in whitehanded gibbons. J Am Vet Med Assoc 163:624-628. Donehower L et al. 1977. Divergence of baboon endogenous type C virogene in primates: Genomic viral RNA in molecular hybridization experiments. J Virol 21:932- 941. Feldman D. 1975. An electron microscopic study of virus particles in rhesus monkey placenta. Proc Natl Acad Sci USA 72:118-121. Fine L et al. 1975. Responses of infant rhesus monkeys to inoculation with Mason- Pfizer monkey virus materials. J Natl Cancer Inst 54:651-658. Fine L, Schochetman G. 1978. Type D primate retroviruses: A review. Cancer Res 38:3123-3139. Flugel RM et al. 1978. Activation of an endogenous retrovirus from Tupaia (tree shrew). Nature 271:543-545. Gilden RV et al. 1974. Similarity between gibbon ape and woolly monkey type C virus internal antigens by quantitative micro-complement with MPMV. Arch Virol 2:360-365. Gonda MA et al. 1978. Squirrel monkey retrovirus: Electron microscopy of a virus from New World monkeys and comparison with MPMV. Arch Virol 56:297-307. Heberling RL et al. 1977. Oncornavirus: Isolation from a squirrel monkey lung culture. Science 195:289-292. Heberling RL et al. 1976. Isolation and biological properties of endogenous baboon (Papio cynocephalus) type C viruses. Bibl Hematol 43:158-160. Hefti E et al. 1983. Isolation of a unique retrovirus, MNV-1, from Macaca nemestrina. Virol 127:309-319. Hehlmann R, Brack-Werner R, Leib-Mosch C. 1988. Human endogenous retroviruses. Leukemia 2:167S-177S. Hino S, Tronick SR, Heberling RL et al. 1977. Endogenous New World primate retrovirus: Interspecies antigenic determinants shared with the major structural protein of type D RNA viruses of Old World monkeys. Proc Natl Acad Sci 74:5734-????. Hirsch MS et al. 1978. Immunity to antigens associated with primate C-type oncoviruses in pregnant women. Science 199:1337-1340. Johnson PB. 1974. Studies on simian foamy viruses and syncytium forming viruses of lower animals. Lab Anim Sci 24:159-166. Kalter SS et al. 1975. A comparative study on the presence of C-type viral particles in placentas from primates and other animals. Comparative leukemia research 1973- leukomogenesis. Bibl Haematology 40:391-401. Kalter SS et al. 1975. Biohazards and simian viruses. Bibl Haematology 40:759-769. Kalter SS et al. 1975. Brief communication: C-type viruses in chimpanzee placentas. J Natl Cancer Inst 55:735-736. Kalter SS et al. 1975. C-type particles in baboon placenta. Proc Royal Soc Med 68:135- 140. Kawakami TG et al. 1972. C-type virus associated with gibbon lymphosarcoma. Nature New Biol 235:170-171. Miyoshi I et al. 1982. Detection of type C virus particles in Japanese monkeys seropositive to adult T-cell leukemia-associated antigens. Gann 73:848-849. Nemo GJ. 1978. Antigenic relationship of human foamy virus to the simian foamy viruses. Infect Immun 20:69-72. Neubauer RH et al. 1974. Attempts to demonstrate type C virus in normal and neoplastic tissues of nonhuman primate origin. Lab Anim Sci 24:235-240. Nooter K. 1980. Primate type-C oncoviruses. Biochem Biophys Acta 605:461-487. Parks WP et al. 1973. Radioimmunoassay of mammalian type C polypeptides. In Characterization of woolly monkey and gibbon viral antigens. Int J Cancer 12:129-137. Prowten AW et al. 1985. T-cell lymphoma associated with immunologic evidence of retrovirus infection in a lowland gorilla. J Am Vet Med Assoc 187:1281- . Rangan SRS. 1974. C type oncogenic viruses of nonhuman primates. Lab Anim Sci 24:193-203. Rangan SRS, Gallather RE. 1979. Tumors and viruses in nonhuman primates. Adv Virus Res 24:1-45. Schochetman G, Fine L. 1978. Natural distribution of squirrel monkey retrovirus proviral sequences in primate DNAs. J Gen Virol 40:257-261. Scolnick EM, Parks WP. 1973. Isolation and characterization of a primate sarcoma virus: Mechanism of rescue. Int J Cancer 12:138-147. Schidlovsky G, Ahmed M. 1973. C-type virus particles in placenta and fetal tissues of rhesus monkeys. J Nat Cancer Inst 51:225-233. Seman G et al. 1975. Type C virus particles in placenta of the cottontop marmoset. J Natl Cancer Inst 54:251-252. Sherr CJ, Todara GJ. 1974. Radioimmunoassay of the major group specific protein of endogenous baboon type C viruses: Relation to the R-114/ccc group and detection of antigen in normal baboon tissue. Virol 61:168-181. Sherwin SA, Todara GJ. 1979. A new endogenous primate type C virus isolated from the Old World monkey Colobus polykomos. Proc Natl Acad Sci 76:5041-5045. Smith GC, Heberling RL, Kalter SS. 1978. Comparison of Mason-Pfizer monkey virus and squirrel monkey (Saimiri sciureus) retrovirus by immunoelectron microscopy. J Natl Cancer Inst 61:411-413. Stephenson JR. 1977. Endogenous C-type viral expression in primates. Nature 266:469-472. Todaro GJ et al. 1975. Infectious primate type C viruses: Three isolates belonging to a new subgroup from the brains of normal gibbons. Virol 67:335-343. Todaro GJ et al. 1978. Endogenous New World primate type C viruses isolated from owl monkey (Aotus trivirgatus) kidney cell line. Proc Natl Acad Sci USA 75:1004-1008. Todaro GJ et al. 1978. MAC-1, a new genetically transmitted type C virus of primates: "Low frequency" activation from stumptail monkey cell cultures. Cell 13:775-782. Todaro GJ et al. 1975. Endogenous primate and feline type C viruses. Cold Spring Harbor Symposium on Quantitative Biology 39,pt.2:1159-1168. Tronick SR et al. 1975. Antigenic characterization of type C RNA virus isolates of gibbon apes. J Virol 15:115-120. Weislow OS et al. 1976. Autogenous humoral immunity to baboon xenotropic endogenous type C virus. J Natl Cancer Inst 57:561-565. Yetz JM, Letvin NL. 1987. Macaque monkey type D retrovirus replicates in vitro in a distinct subpopulation of lymphocytes-B. J Gen Virol 68:573-576. Miscellaneous & General References--Viral Diseases Anon. Rabies in a monkey in New York state. CC Vet Publ Hlth Notes, 2 Aug 1963. Boulger LR. 1966. Natural rabies in a laboratory monkey. Lancet 1:941-943. Caul EO, Egglestone SI. 1979. Coronavirus-like particles in simian feces. Vet Rec 104:168-169. Eichhorn W, Czerny CP. 1988. Enteric coronaviruses in primates. Zentralbl Veterinarmed [B] 35:709-712. Espana C. 1971. Review of some outbreaks of viral disease in captive nonhuman primates. Lab Anim Sci 21:1023-1031. Fields BN et al (eds). Fundamental Virology. Raven Press, New York, 1986. Heberling RL. 1971. Viral diseases of nonhuman primates in the wild. Lab Anim Sci 21:1019-1022. Kalter SS, Heberling RL. 1990. Viral battery testing in nonhuman primate colony management. Lab Anim Sci 40:21-23. Kalter SS (ed). Viral and immunologic diseases in nonhuman primates. New York, Alan R Liss, 1983. Kalter SS et al. 1967. A survey of primate sera for antibodies to viruses of human and simian origin. Am J Epidemiol 86:552-568. Kalter SS et al. 1969. Antibodies to human and simian viruses in the gorilla (Gorilla gorilla). Lab Anim Care 19:63-66. Kalter SS. 1982. Enteric viruses of nonhuman primates. Vet Pathol 19(Suppl 7):33-43. Kaschula VR et al. 1978. Some infectious diseases of wild vervet monkeys (Cercopithecus aethiops pyserythrus) in South Afrida. J South Afr Vet Assoc 49:223- 227. Kessler MJ et al. 1989. Serological survey for viral diseases in the Cayo Santiago rhesus macaque population. P R Health Sci J 8:95-97. Matthews REF. 1982. Classification and nomenclature of viruses. Fourth report of the international committee on taxonomy of viruses. Intervirol 17(1-3). Mei Z et al. 1990. Virological survey of rhesus monkeys in China. Lab Anim Sci 40:29- 32. Prince AM et al. 1989. Appropriate conditions for maintenance of chimpanzees in studies with blood-borne viruses: an epidemiologic and psychosocial perspective. J Med Primatol 18:27-42. Richardson JH et al. 1971. Rabies in imported nonhuman primates. Lab Anim Sci 21:1083-1084. Sasagawa A et al. 1981. Absence of antibodies against SV5, measles and herpes simplex in indoor colony-bred cynomolgus monkeys. Jpn J Med Sci Biol 34:381-384. Shah KV, Southwick H. 1965. Prevalence of antibodies to certain viruses in sera of free-living rhesus and of captive monkeys. Indian J Med Res 53:488-500. Soave O. 1981. Viral infections common to humans and nonhuman primates. J Am Vet Med Assoc 179:1385-1388. Soike KF et al. 1984. Viral disease models in primates. Adv Vet Sci Comp Med. 1984 28:151-199. Stuker G et al. 1979. Virus detection in monkeys with diarrhea: The association of adenoviruses with diarrhea and the possible role of rotaviruses. Lab Anim Sci 29:610- ???. Suzuki M et al. 1981. Serological survey for SV5, measles, and herpes simplex infections in newly imported cynomolgus monkeys. Jap J Med Sci Biol 34:69-80. Suzuki M. 1981. Natural virus infections in cynomolgus monkeys. Jap J Med Sci Biol 34:261-263. Suzuki M et al. 1981. Serological survey for SV5, measles and herpes simplex infections in newly-imported cynomolgus monkeys. Jpn J Med Sci Biol 34:69-80. Tauraso NM. 1973. Review of recent epizootics in nonhuman primate colonies and their relation to man. Lab Anim Sci 23:201-210. Valerio A. 1971. Colony management as applied to disease control with mention of some viral diseases. Lab Anim Sci 21:1011-1014.