1
|
Henning TR, Hanson D, Vishwanathan SA, Butler K, Dobard C, Garcia-Lerma G, Radzio J, Smith J, McNicholl JM, Kersh EN. Short communication: Viremic control is independent of repeated low-dose SHIVSF162p3 exposures. AIDS Res Hum Retroviruses 2014; 30:1125-9. [PMID: 25313448 DOI: 10.1089/aid.2014.0238] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The repeat low-dose virus challenge model is commonly used in nonhuman primate studies of HIV transmission and biomedical preventions. For some viruses or challenge routes, it is uncertain whether the repeated exposure design might induce virus-directed innate or adaptive immunity that could affect infection or viremic outcomes. Retrospective cohorts of male Indian rhesus (n=40) and female pigtail (n=46) macaques enrolled in repeat low-dose rectal or vaginal SHIV(SF162p3) challenge studies, respectively, were studied to compare the relationship between the number of previous exposures and peak plasma SHIV RNA levels or viral load area under the curve (AUC), surrogate markers of viral control. Repeated mucosal exposures of 10 or 50 TCID50 of virus for rectal and vaginal exposures, respectively, were performed. Virus levels were measured by quantitative reverse-transcriptase real-time PCR. The cumulative number of SHIV(SF162p3) exposures did not correlate with observed peak virus levels or with AUC in rectally challenged rhesus macaques [peak: rho (ρ)=0.04, p=0.8; AUC: ρ=0.33, p=0.06] or vaginally challenged pigtail macaques (peak: ρ=-0.09, p=0.7; AUC: ρ=0.11, p=0.6). Infections in these models occur independently of exposure history and provide assurance that neither inoculation route nor number of exposures required for infection correlates with postinfection viremia. These data also indicate that both the vaginal and rectal repeated low-dose virus exposure models using SHIV(SF162p3) provide a reliable system for nonhuman primate studies.
Collapse
Affiliation(s)
- Tara R. Henning
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Debra Hanson
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | - Katherine Butler
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Charles Dobard
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Gerardo Garcia-Lerma
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jessica Radzio
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - James Smith
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Janet M. McNicholl
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Ellen N. Kersh
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
2
|
Abstract
Most infants born to human immunodeficiency virus (HIV)-infected women escape HIV infection. Infants evade infection despite an immature immune system and, in the case of breastfeeding, prolonged repetitive exposure. If infants become infected, the course of their infection and response to treatment differs dramatically depending upon the timing (in utero, intrapartum, or during breastfeeding) and potentially the route of their infection. Perinatally acquired HIV infection occurs during a critical window of immune development. HIV's perturbation of this dynamic process may account for the striking age-dependent differences in HIV disease progression. HIV infection also profoundly disrupts the maternal immune system upon which infants rely for protection and immune instruction. Therefore, it is not surprising that infants who escape HIV infection still suffer adverse effects. In this review, we highlight the unique aspects of pediatric HIV transmission and pathogenesis with a focus on mechanisms by which HIV infection during immune ontogeny may allow discovery of key elements for protection and control from HIV.
Collapse
|
3
|
Delwart E, Bernardin F, Lee TH, Winkelman V, Liu C, Sheppard H, Liu A, Greenblatt R, Anastos K, DeHovitz J, Nowicki M, Cohen M, Golub ET, Barbour J, Buchbinder S, Busch MP. Absence of reproducibly detectable low-level HIV viremia in highly exposed seronegative men and women. AIDS 2011; 25:619-23. [PMID: 21297421 DOI: 10.1097/qad.0b013e3283440269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Transient HIV infections have been invoked to account for the cellular immune responses detected in highly virus-exposed individuals who have remained HIV-seronegative. We tested for very low levels of HIV RNA in 524 seronegative plasma samples from 311 highly exposed women and men from three longitudinal HIV cohorts. DESIGN Two thousand and seventy-three transcription-mediated amplification (TMA) HIV RNA tests were performed for an average of 3.95 TMA assays per plasma sample. Quadruplicate TMA assays, analyzing a total of 2 ml of plasma, provided an estimated sensitivity of 3.5 HIV RNA copies/ml. RESULTS Four samples from individuals who did not seroconvert within the following 6 months were positive for HIV RNA. For one sample, human polymorphism DNA analysis indicated a sample mix-up. Borderline HIV RNA detection signals were detected for the other three positive samples but further replicate TMA testing yielded no positive results. Nested PCR assays (n = 254) for HIV proviral DNA in peripheral blood mononuclear cells (PBMCs) from these three individuals were negative. CONCLUSION Transient viremia was not reproducibly detected in highly HIV-exposed seronegative men and women. If transient infections do occur, plasma HIV RNA levels may remain below the detection limits of the sensitive assay used here, be of very short duration, or viral replication may be restricted to mucosal surfaces or their draining lymphoid tissues.
Collapse
|
4
|
Biesinger T, White R, Yu Kimata MT, Wilson BK, Allan JS, Kimata JT. Relative replication capacity of phenotypic SIV variants during primary infections differs with route of inoculation. Retrovirology 2010; 7:88. [PMID: 20942954 PMCID: PMC2964591 DOI: 10.1186/1742-4690-7-88] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 10/13/2010] [Indexed: 01/08/2023] Open
Abstract
Background Previous studies of human and simian immunodeficiency virus (HIV and SIV) have demonstrated that adaptive mutations selected during the course of infection alter viral replicative fitness, persistence, and pathogenicity. What is unclear from those studies is the impact of transmission on the replication and pathogenicity of the founding virus population. Using the SIV-macaque model, we examined whether the route of infection would affect the establishment and replication of two SIVmne variants of distinct in vitro and in vivo biological characteristics. For these studies, we performed dual-virus inoculations of pig-tailed macaques via intrarectal or intravenous routes with SIVmneCl8, a miminally pathogenic virus, and SIVmne027, a highly pathogenic variant that replicates more robustly in CD4+ T cells. Results The data demonstrate that SIVmne027 is the dominant virus regardless of the route of infection, indicating that the capacity to replicate efficiently in CD4+ T cells is important for fitness. Interestingly, in comparison to intravenous co-infection, intrarectal inoculation enabled greater relative replication of the less pathogenic virus, SIVmneCl8. Moreover, a higher level of SIVmneCl8 replication during primary infection of the intrarectally inoculated macaques was associated with lower overall plasma viral load and slower decline in CD4+ T cells, even though SIVmne027 eventually became the dominant virus. Conclusions These results suggest that the capacity to replicate in CD4+ T cells is a significant determinant of SIV fitness and pathogenicity. Furthermore, the data also suggest that mucosal transmission may support early replication of phenotypically diverse variants, while slowing the rate of CD4+ T cell decline during the initial stages of infection.
Collapse
Affiliation(s)
- Tasha Biesinger
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
5
|
Abstract
Measures to prevent sexual mucosal transmission of human immunodeficiency virus (HIV)-1 are urgently needed to curb the growth of the acquired immunodeficiency syndrome (AIDS) pandemic and ultimately bring it to an end. Studies in animal models and acute HIV-1 infection reviewed here reveal potential viral vulnerabilities at the mucosal portal of entry in the earliest stages of infection that might be most effectively targeted by vaccines and microbicides, thereby preventing acquisition and averting systemic infection, CD4 T-cell depletion and pathologies that otherwise rapidly ensue.
Collapse
Affiliation(s)
- Ashley T Haase
- Department of Microbiology, University of Minnesota, Minnesota 55455, USA.
| |
Collapse
|
6
|
Kersh EN, Luo W, Adams DR, Srinivasan P, Smith JM, Promadej-Lanier N, Ellenberger D, Garcia-Lerma JG, Butera S, Otten R. Repeated rectal SHIVSF162P3 exposures do not consistently induce sustained T cell responses prior to systemic infection in the repeat-low dose preclinical macaque model. AIDS Res Hum Retroviruses 2009; 25:905-17. [PMID: 19689194 DOI: 10.1089/aid.2008.0287] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The macaque model of repeated SHIV exposures is increasingly used as a preclinical tool to evaluate biomedical HIV intervention strategies. It is unclear whether multiple virus exposures induce immune responses in macaques, as documented in uninfected individuals repeatedly exposed to HIV. We here address whether repeated, rectal SHIV(SF162P3) exposures lead to systemic T cell activation in 12 rhesus macaques, and whether this is associated with increased infection resistance. Eight macaques became systemically infected after 2-7 exposures, three macaques were less susceptible (infection after 10-12 exposures), and one macaque remained uninfected after 14 exposures. PBMCs were retrospectively monitored for increases in T cell activation by analyzing the proportion of CD8(+) T cells, recently activated or proliferated T cells (markers CD38, Ki67), a marker for cytotoxicity (granzyme B), or T cell-produced plasma cytokines (IFN-gamma, RANTES, IL-2). Repeated virus exposures did not induce sustained, potent, or diverse T cell responses prior to systemic infection. Some changes occurred in the analyzed parameters during repeated virus exposures, but similar T cell activities were also observed in five SHIV-unexposed control macaques. Thus, we found no evidence that delayed infection or resistance to infection was associated with systemic, long-lasting, protective T cell responses to repeated rectal virus exposures. Our results provide further insights into the repeat exposure macaque model. We find that this model can be used for testing biomedical prevention strategies without concern of eliciting a systemic vaccination effect.
Collapse
Affiliation(s)
- Ellen N. Kersh
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Wei Luo
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Debra R. Adams
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Priya Srinivasan
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - James M. Smith
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Nattawan Promadej-Lanier
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Dennis Ellenberger
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - J. Gerardo Garcia-Lerma
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Salvatore Butera
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| | - Ron Otten
- Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333
| |
Collapse
|
7
|
Attenuated disease in SIV-infected macaques treated with a monoclonal antibody against FasL. Clin Dev Immunol 2008; 2007:93462. [PMID: 18317535 PMCID: PMC2248700 DOI: 10.1155/2007/93462] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Accepted: 10/31/2007] [Indexed: 11/18/2022]
Abstract
Acute SIVmac infection in macaques is accompanied by high levels of plasma viremia that decline with the appearance of viral immunity and is a model for acute HIV disease in man. Despite specific immune responses, the virus establishes a chronic, persistent infection. The destruction of CD4+
and CD4- lymphocyte subsets in macaques
contributes to viral persistence and suggests the
importance of mechanisms for depleting both infected
and uninfected (bystander) cells. Bystander cell killing
can occur when FasL binds the Fas receptor on activated lymphocytes,
which include T and B cell subpopulations that are responding to the
infection. Destruction of specific immune cells could be an important
mechanism for blunting viral immunity and establishing persistent infection
with chronic disease. We inhibited the Fas pathway in vivo with a monoclonal
antibody against FasL (RNOK203). Here we show that treatment with anti-FasL
reduced cell death in circulating T and B cells, increased CTL and antibody
responses to viral proteins, and lowered the setpoint viremia. By blocking
FasL during only the first few weeks after infection, we attenuated SIVmac
disease and increased the life span for infected and treated macaques.
Collapse
|
8
|
Subbarao S, Ramos A, Kim C, Adams D, Monsour M, Butera S, Folks T, Otten RA. Direct stringency comparison of two macaque models (single-high vs. repeat-low) for mucosal HIV transmission using an identical anti-HIV chemoprophylaxis intervention. J Med Primatol 2007; 36:238-43. [PMID: 17669212 DOI: 10.1111/j.1600-0684.2007.00241.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND In our previous work, oral chemoprophylaxis with tenofovir disoproxil fumarate (TDF) provided partial protection in rhesus macaques against repeated low-dose (RL) intrarectal SHIV162p3 exposure. METHODS Here, we make a direct comparison of these previous findings with data generated using a single high (SH)-dose challenge strategy. RESULTS All 5 (100%) control macaques were infected after a SH challenge and only three of five (60%) TDF-treated macaques became infected. The remaining two TDF-treated macaques remained virus-negative and were susceptible to virus infection upon re-challenge in the absence of oral TDF. Thus, two of five (40%) TDF-treated macaques were protected by the pre-exposure chemoprophylaxis regimen. By comparison with the RL challenge system, only one of four (25%) of TDF-treated macaques were protected from infection, whereas four of four (100%) control macaques became infected using RL challenges. CONCLUSION Taken together, these findings indicate that the stringency of the RL challenge model for testing antiretroviral interventions is not lower and possibly greater than that of the SH challenge model.
Collapse
Affiliation(s)
- Shambavi Subbarao
- Laboratory Branch, Division of HIV/AIDS Prevention, NCHHSTP, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Tasca S, Tsai L, Trunova N, Gettie A, Saifuddin M, Bohm R, Chakrabarti L, Cheng-Mayer C. Induction of potent local cellular immunity with low dose X4 SHIV(SF33A) vaginal exposure. Virology 2007; 367:196-211. [PMID: 17574643 PMCID: PMC2756750 DOI: 10.1016/j.virol.2007.05.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2007] [Revised: 04/25/2007] [Accepted: 05/15/2007] [Indexed: 11/15/2022]
Abstract
Intravaginal inoculation of rhesus macaques with varying doses of the CXCR4 (X4)-tropic SHIV(SF33A) isolate revealed a threshold inoculum for establishment of systemic virus infection and a dose dependency in overall viral burden and CD4+ T cell depletion. While exposure to inoculum size of 1000 or greater 50% tissue infectious dose (TCID(50)) resulted in high viremia and precipitous CD4+ T cell loss, occult infection was observed in seven of eight macaques exposed to 500 TCID(50) of the same virus. The latter was characterized by intermittent detection of low level virus with no evidence of seroconversion or CD4+ T cell decline, but with signs of an ongoing antiviral T cell immune response. Upon vaginal re-challenge with the same limiting dose 11-12 weeks after the first, classic pathogenic X4 SHIV(SF33A) infection was established in four of the seven previously exposed seronegative macaques, implying enhanced susceptibility to systemic infection with prior exposure. Pre-existing peripheral SIV gag-specific CD4+ T cells were more readily demonstrable in macaques that became systemically infected following re-exposure than those that were not. In contrast, early presence of circulating polyfunctional cytokine secreting CD8+ T cells or strong virus-specific proliferative responses in draining lymph nodes and in the gut associated lymphoid tissue (GALT) following the first exposure was associated with protection from systemic re-infection. These studies identify the gut and lymphoid tissues proximal to the genital tract as sites of robust CD8 T lymphocyte responses that contribute to containment of virus spread following vaginal transmission.
Collapse
Affiliation(s)
- Silvana Tasca
- Aaron Diamond AIDS Research Center, The Rockefeller University, 455 First Ave, 7 Floor, New York, NY 10016
| | - Lily Tsai
- Aaron Diamond AIDS Research Center, The Rockefeller University, 455 First Ave, 7 Floor, New York, NY 10016
| | - Nataliya Trunova
- Aaron Diamond AIDS Research Center, The Rockefeller University, 455 First Ave, 7 Floor, New York, NY 10016
| | - Agegnehu Gettie
- Aaron Diamond AIDS Research Center, The Rockefeller University, 455 First Ave, 7 Floor, New York, NY 10016
| | - Mohammed Saifuddin
- CONRAD, Eastern Virginia Medical School, 1611 North Kent Street Suite 806, Arlington, VA 22209
| | - Rudolf Bohm
- Tulane National Primate Research Center, Tulane University Medical Center, 18702 Three Rivers Road, Covington, LA 70433
| | - Lisa Chakrabarti
- Institut Pasteur, Unite d'Immunologie Virale, 28 rue du Dr roux, 75724 Paris Cedex 15, France
| | - Cecilia Cheng-Mayer
- Aaron Diamond AIDS Research Center, The Rockefeller University, 455 First Ave, 7 Floor, New York, NY 10016
| |
Collapse
|
10
|
Brewer DD, Potterat JJ, Roberts JM, Brody S. Male and female circumcision associated with prevalent HIV infection in virgins and adolescents in Kenya, Lesotho, and Tanzania. Ann Epidemiol 2007; 17:217-26. [PMID: 17320788 DOI: 10.1016/j.annepidem.2006.10.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2006] [Revised: 10/11/2006] [Accepted: 10/17/2006] [Indexed: 11/22/2022]
Abstract
PURPOSE Remarkable proportions of self-reported virgins and adolescents in eastern and southern Africa are infected with HIV, yet non-sexual routes of transmission have not been systematically investigated in such persons. Many observers in this region have recognized the potential for HIV transmission through unhygienic circumcision procedures. We assessed the relation between male and female circumcision (genital cutting) and prevalent HIV infection in Kenyan, Lesothoan, and Tanzanian virgins and adolescents. METHODS We analyzed data from recent cross-sectional national probability sample surveys of adolescents and adults in households, focusing on populations in which circumcision was common and usually occurred in puberty or later. RESULTS Circumcised male and female virgins were substantially more likely to be HIV infected than uncircumcised virgins (Kenyan females: 3.2% vs. 1.4%, odds ratio [OR] = 2.38; Kenyan males: 1.8% vs. 0%, OR undefined; Lesothoan males: 6.1% vs. 1.9%, OR 3.36; Tanzanian males: 2.9% vs. 1.0%, OR 2.99; weighted mean phi correlation = 0.07, 95% confidence interval, 0.03 to 0.11). Among adolescents, regardless of sexual experience, circumcision was just as strongly associated with prevalent HIV infection. However, uncircumcised adults were more likely to be HIV positive than circumcised adults. Self-reported sexual experience was independently related to HIV infection in adolescent Kenyan females, but was unrelated to HIV infection in adolescent Kenyan, Lesothoan, and Tanzanian males. CONCLUSIONS HIV transmission may occur through circumcision-related blood exposures in eastern and southern Africa.
Collapse
Affiliation(s)
- Devon D Brewer
- Interdisciplinary Scientific Research, Seattle, WA 98115, USA. via www.interscientific.net/contact.html
| | | | | | | |
Collapse
|
11
|
Jayaraman P, Zhu T, Misher L, Mohan D, Kuller L, Polacino P, Richardson BA, Bielefeldt-Ohmann H, Anderson D, Hu SL, Haigwood NL. Evidence for persistent, occult infection in neonatal macaques following perinatal transmission of simian-human immunodeficiency virus SF162P3. J Virol 2006; 81:822-34. [PMID: 17079310 PMCID: PMC1797486 DOI: 10.1128/jvi.01759-06] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To model human immunodeficiency virus (HIV) perinatal transmission, we studied infection of simian-human immunodeficiency virus (SHIV) SF162P3 in 10 pregnant Macaca nemestrina females and their offspring. Four of nine infants born to and suckled by these dams had evidence of infection, a transmission rate of 44.4% (95% confidence interval, 13.7% to 78.8%). We quantified transplacentally acquired and de novo Env-specific immunoglobulin G (IgG), IgM, and neutralizing antibodies in newborns. Transmission of escape variants was confirmed. In utero infection (n = 1) resulted in high viremia, depletion of peripheral CD4+ T cells, and rapid evolution of env in blood and tissues. Peripartum or postpartum SHIV infection (n = 3) resulted in postacute viral control that was undetectable by very sensitive multiplex PCR, despite increasing antibodies. Seropositive infants with highly controlled viremia had homogeneous peripheral blood env sequences, and their tissues had <3 copies per million cells. A high incidence of seropositive virus-low or -negative SHIV infection in infant macaques has implications for HIV type 1 perinatal transmission and detection.
Collapse
Affiliation(s)
- Pushpa Jayaraman
- Departments of Pathobiology, National Primate Research Center, University of Washington, Seattle, Washington 98195, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Waterman PM, Kitabwalla M, Hatfield GS, Evans PS, Lu Y, Tikhonov I, Bryant JL, Pauza CD. Effects of virus burden and chemokine expression on immunity to SHIV in nonhuman primates. Viral Immunol 2005; 17:545-57. [PMID: 15671751 DOI: 10.1089/vim.2004.17.545] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
HIV-1 vaccine candidates are designed to elicit Type 1 immune responses, including cytotoxic T cells and neutralizing antibodies. The type of immune response is influenced by many factors, including the levels of antigen expression and production of cytokines or chemokines; we designed a nonhuman primate study to evaluate the influence of these factors on protective immunity. Recombinant SHIV were engineered to express macrophage inflammatory protein-1 alpha (MIP-1alpha), regulated upon activation, normal T-cell expressed and secreted (RANTES), or Lymphotactin (Ltn) in place of nef in SHIV(89.6) (SHIV(89.6-MIP-1), SHIV(89.6-RANTES), SHIV(89.6-Ltn)). The parental virus SHIV(89.6) was included because it replicates to higher titer while still not causing disease. Control groups included animals that received a recombinant SHIV with a truncated chemokine construct (SHIV(89.6-dLtn)) and unvaccinated macaques. After pathogenic challenge with SHIV(89.6pd), animals from groups that received recombinant (nef-deleted) viruses had peak viremia levels three orders of magnitude lower than unvaccinated controls and increased survival times. Animals that received the original SHIV(89.6) (nef+) were highly resistant to both intrarectal and intravenous challenge with SHIV(89.6PD), and showed no signs of disease. There were no differences in survival times comparing unvaccinated and SHIV(89.6-dLtn) (control) groups, indicating that nef deleted viruses did not provide durable protection in this model. Strongest protection was seen in animals with the highest replicating virus (SHIV(89.6)), and the lower effect on survival after SHIV(89.6) nef-deleted vaccination, likely reflects differences in replication capacity. The protective effect of nef-deleted virus was partly restored by expressing Type 1 chemokines to augment viral immunity.
Collapse
Affiliation(s)
- Paul M Waterman
- Institute of Human Virology, University of Maryland Biotechnology Institute, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Zhu T, Hu SL, Feng F, Polacino P, Liu H, Hwangbo Y, Learn GH, Mullins JI, Corey L. Persistence of low levels of simian immunodeficiency virus in macaques that were transiently viremic by conventional testing. Virology 2004; 323:208-19. [PMID: 15193917 DOI: 10.1016/j.virol.2004.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2003] [Revised: 12/16/2003] [Accepted: 03/08/2004] [Indexed: 11/16/2022]
Abstract
Transient SIV viremia after experimental SIV challenge has been documented. Whether SIV persists in these transiently viremic macaques remains unclear. In the present study, we applied a sensitive PCR and found persistent low levels of SIVmne infection (LLSI) (range: 0.1-5.3 SIV DNA copies/10(6) PBMC) in seven macaques that were transiently positive by conventional assays, which was 10(2)- to 10(6)-fold less than those of SIVmne infected monkeys with typical disease progression. SIV envelope V1 sequences remained homogeneous in these macaques for the 6-year study period, with a mean evolution rate of 0.005% per site per year, which was not different from zero (P = 0.612) and significantly lower than that (0.56-1.18%) in macaques with progressive infection of SIVmne. LLSI macaques have remained free from SIV-associated illness, and are still alive 10 years after virus inoculation. Understanding the mechanisms underlying this outcome may provide valuable insight into therapy and vaccine development.
Collapse
Affiliation(s)
- Tuofu Zhu
- Department of Laboratory Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Bertley FMN, Kozlowski PA, Wang SW, Chappelle J, Patel J, Sonuyi O, Mazzara G, Montefiori D, Carville A, Mansfield KG, Aldovini A. Control of simian/human immunodeficiency virus viremia and disease progression after IL-2-augmented DNA-modified vaccinia virus Ankara nasal vaccination in nonhuman primates. THE JOURNAL OF IMMUNOLOGY 2004; 172:3745-57. [PMID: 15004179 DOI: 10.4049/jimmunol.172.6.3745] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A successful HIV vaccine may need to stimulate antiviral immunity in mucosal and systemic immune compartments, because HIV transmission occurs predominantly at mucosal sites. We report here the results of a combined DNA-modified vaccinia virus Ankara (MVA) vaccine approach that stimulated simian/human immunodeficiency virus (SHIV)-specific immune responses by vaccination at the nasal mucosa. Fifteen male rhesus macaques, divided into three groups, received three nasal vaccinations on day 1, wk 9, and wk 25 with a SHIV DNA plasmid producing noninfectious viral particles (group 1), or SHIV DNA plus IL-2/Ig DNA (group 2), or SHIV DNA plus IL-12 DNA (group 3). On wk 33, all macaques were boosted with rMVA expressing SIV Gag-Pol and HIV Env 89.6P, administered nasally. Humoral responses were evaluated by measuring SHIV-specific IgG and neutralizing Abs in plasma, and SHIV-specific IgA in rectal secretions. Cellular responses were monitored by evaluating blood-derived virus-specific IFN-gamma-secreting cells and TNF-alpha-expressing CD8+ T cells, and blood- and rectally derived p11C tetramer-positive T cells. Many of the vaccinated animals developed both mucosal and systemic humoral and cell-mediated anti-SHIV immune responses, although the responses were not homogenous among animals in the different groups. After rectal challenge of vaccinated and naive animals with SHIV89.6P, all animals became infected. However a subset, including all group 2 animals, were protected from CD4+ T cell loss and AIDS development. Taken together, these data indicate that nasal vaccination with SHIV-DNA plus IL-2/Ig DNA and rMVA can provide significant protection from disease progression.
Collapse
Affiliation(s)
- Frederic M N Bertley
- Department of Medicine, Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Negri DRM, Baroncelli S, Catone S, Comini A, Michelini Z, Maggiorella MT, Sernicola L, Crostarosa F, Belli R, Mancini MG, Farcomeni S, Fagrouch Z, Ciccozzi M, Boros S, Liljestrom P, Norley S, Heeney J, Titti F. Protective efficacy of a multicomponent vector vaccine in cynomolgus monkeys after intrarectal simian immunodeficiency virus challenge. J Gen Virol 2004; 85:1191-1201. [PMID: 15105535 DOI: 10.1099/vir.0.79794-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
We investigated the protective efficacy of a systemic triple vector (DNA/rSFV/rMVA)-based vaccine against mucosal challenge with pathogenic simian immunodeficiency virus (SIV) in cynomolgus monkeys. Animals were immunized at week 0 with DNA (intradermally), at weeks 8 and 16 with recombinant Semliki Forest virus (rSFV, subcutaneously) and finally, at week 24, with recombinant modified vaccinia virus Ankara strain (rMVA, intramuscularly). Both DNA and recombinant viral vectors expressed a wide range of SIV proteins (Gag, Pol, Tat, Rev, Env and Nef). This immunization strategy elicited cell-mediated rather than humoral responses that were especially increased following the last boost. Upon intrarectal challenge with pathogenic SIVmac251, three of the four vaccinated monkeys dramatically abrogated virus load to undetectable levels up to 41 weeks after challenge. A major contribution to this vaccine effect appeared to be the T-cell-mediated immune response to vaccine antigens (Gag, Rev, Tat, Nef) seen in the early phase of infection in three of the four vaccinated monkeys. Indeed, the frequency of T-cells producing antigen-induced IFN-γ mirrored virus clearance in the vaccinated and protected monkeys. These results, reminiscent of the efficacy of live attenuated virus vaccines, suggest that vaccination with a combination of many viral antigens can induce a robust and stable vaccine-induced immunity able to abrogate virus replication.
Collapse
Affiliation(s)
- Donatella R M Negri
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Silvia Baroncelli
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Stefania Catone
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Antonella Comini
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Zuleika Michelini
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Maria T Maggiorella
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Leonardo Sernicola
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Federica Crostarosa
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Roberto Belli
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Maria G Mancini
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Stefania Farcomeni
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Zahra Fagrouch
- Department of Virology, Biomedical Primate Research Center, PO Box 3306, 2280 GH Rijswijk, The Netherlands
| | - Massimo Ciccozzi
- Laboratory of Epidemiology and Biostatistics, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Stefano Boros
- Laboratory of Epidemiology and Biostatistics, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| | - Peter Liljestrom
- Microbiology and Tumor-Biology Center, Karolinska Institute, Box 280, S-17177 Stockholm, Sweden
| | - Stephen Norley
- AIDS Immunopathogenesis and Vaccine Development, Robert Koch Institute, 13353 Berlin, Germany
| | - Jonathan Heeney
- Department of Virology, Biomedical Primate Research Center, PO Box 3306, 2280 GH Rijswijk, The Netherlands
| | - Fausto Titti
- Laboratory of Virology, Istituto Superiore di Sanità, Viale R. Elena 299, 00161 Rome, Italy
| |
Collapse
|
16
|
Rybarczyk BJ, Montefiori D, Johnson PR, West A, Johnston RE, Swanstrom R. Correlation between env V1/V2 region diversification and neutralizing antibodies during primary infection by simian immunodeficiency virus sm in rhesus macaques. J Virol 2004; 78:3561-71. [PMID: 15016879 PMCID: PMC371075 DOI: 10.1128/jvi.78.7.3561-3571.2004] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Evolution of the domain encoding the V1/V2 variable region of the simian immunodeficiency virus sm (SIVsm) envelope (env) gene was analyzed in relation to route of virus challenge, virus load, and neutralizing antibody (NAb) titers during primary infection of rhesus macaques with the pathogenic SIVsmE660 isolate. In this model system animals are initially infected with multiple viruses as evidenced by the presence of multiple V1/V2 genotypic variants that could be resolved by using a heteroduplex tracking assay (HTA). Overlapping subsets of the multiple variants were established in each animal. There was no selection for the establishment of specific variants in comparing intravenous- and intrarectal-challenged macaques at week 2 postinfection, suggesting that no genotypic selection occurred at the mucosal surface. There was an initial period of significant stability of the V1/V2 variants. Macaques challenged intravenously displayed subsequent V1/V2 diversification significantly earlier than macaques challenged intrarectally and well past the initial resolution of viremia. The time when SIVsmE660-specific NAbs reached a threshold titer of 100 was significantly correlated with the timing of V1/V2 diversification, even though antibodies to the Env protein could be detected much earlier. The time when NAbs reached a titer of 400 was significantly correlated with virus load late in infection. These results show that the route of infection affects the timing of V1/V2 diversification and that this diversification is correlated with the maturation of a specific NAb response. However, prior immunization capable of priming an anamnestic Env antibody response did not accelerate V1/V2 diversification. This result suggests that diversification of the SIV env V1/V2 region is the result of a type-specific antibody response.
Collapse
Affiliation(s)
- Brian J Rybarczyk
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, USA
| | | | | | | | | | | |
Collapse
|
17
|
Avery PR, Hoover EA. Gamma interferon/interleukin 10 balance in tissue lymphocytes correlates with down modulation of mucosal feline immunodeficiency virus infection. J Virol 2004; 78:4011-9. [PMID: 15047817 PMCID: PMC374259 DOI: 10.1128/jvi.78.8.4011-4019.2004] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2003] [Accepted: 01/06/2004] [Indexed: 11/20/2022] Open
Abstract
Understanding the early cytokine response to lentiviral infections may be critical to the design of prevention and treatment strategies. By using the feline immunodeficiency virus (FIV) model, we have documented an interleukin 10 (IL10)-dominated response in lymphoid tissue CD4(+) and CD8(+) T lymphocytes within the first 4 weeks after mucosal FIV infection. This profile coincided with the period of high tissue viral replication. By 10 weeks postinfection, tissue viral levels decreased significantly, and gamma interferon (IFN gamma) production in CD8(+) T cells had increased to restore the IL10/IFN gamma ratio to control levels. Concurrently, increased production of IL6 and viral RNA was detected in macrophages. These temporal associations of viral replication with cytokine balance in tissues suggest roles for IL10 in the permissive stage of infection and IFN gamma in the subsequent down modulation of lentiviral infection.
Collapse
Affiliation(s)
- Paul R Avery
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, USA.
| | | |
Collapse
|
18
|
Rodas JD, Lukashevich IS, Zapata JC, Cairo C, Tikhonov I, Djavani M, Pauza CD, Salvato MS. Mucosal arenavirus infection of primates can protect them from lethal hemorrhagic fever. J Med Virol 2004; 72:424-35. [PMID: 14748066 PMCID: PMC2562566 DOI: 10.1002/jmv.20000] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Arenaviruses are transmitted from rodents to human beings by blood or mucosal exposure. The most devastating arenavirus in terms of human disease is Lassa fever virus, causing up to 300,000 annual infections in West Africa. We used a model for Lassa fever in which Rhesus macaques were infected with a related virus, lymphocytic choriomeningitis virus (LCMV). Our goals were to determine the outcome of infection after mucosal inoculation and later lethal challenge, to characterize protective immune responses, and to test cross-protection between a virulent (LCMV-WE) and an avirulent (LCMV-ARM) strain of virus. Although intravenous infections in the monkey model were uniformly lethal, intragastric infections recapitulated the spectrum of clinical outcomes seen in human exposure to Lassa fever virus: death, recovery from disease, and most often, subclinical infection. Plaque neutralization, ELISA, lymphocyte proliferation, and chromium-release assays were used to monitor humoral and cellular immune responses. Cross protection between the two strains was observed. The three out of seven monkeys that experienced protection were also the three with the strongest cell-mediated immunity.
Collapse
Affiliation(s)
- Juan D. Rodas
- Institute of Human Virology, University of Maryland Biotechnology Institute, Baltimore, Maryland
| | - Igor S. Lukashevich
- Institute of Human Virology, University of Maryland Biotechnology Institute, Baltimore, Maryland
| | - Juan C. Zapata
- Institute of Human Virology, University of Maryland Biotechnology Institute, Baltimore, Maryland
| | - Cristiana Cairo
- Institute of Human Virology, University of Maryland Biotechnology Institute, Baltimore, Maryland
| | - Ilia Tikhonov
- Institute of Human Virology, University of Maryland Biotechnology Institute, Baltimore, Maryland
| | - Mahmoud Djavani
- Institute of Human Virology, University of Maryland Biotechnology Institute, Baltimore, Maryland
| | - C. David Pauza
- Institute of Human Virology, University of Maryland Biotechnology Institute, Baltimore, Maryland
| | - Maria S. Salvato
- Institute of Human Virology, University of Maryland Biotechnology Institute, Baltimore, Maryland
| |
Collapse
|
19
|
Couëdel-Courteille A, Prétet JL, Barget N, Jacques S, Petitprez K, Tulliez M, Guillet JG, Venet A, Butor C. Delayed viral replication and CD4+ T cell depletion in the rectosigmoid mucosa of macaques during primary rectal SIV infection. Virology 2003; 316:290-301. [PMID: 14644611 DOI: 10.1016/j.virol.2003.08.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Rectal infection of macaques by SIV is a model for rectal HIV transmission. We focus here on the digestive tract during days 7-14 of primary rectal infection by SIV in 15 rhesus macaques. Surprisingly, we did not detect productively infected cells in the rectosigmoid colon at early stages of viral dissemination. This strongly suggests that there is no massive viral amplification in the rectosigmoid colon prior to viral dissemination. As dissemination proceeds, productively infected T cells are observed in the rectosigmoid colon and small intestine, with rectosigmoid colon showing the heaviest viral load. Lymphoid follicles are infected prior to lamina propria at both sites. When viral dissemination is widespread, inflammatory infiltrates are visible in the rectosigmoid colon, but not in the small intestine. An important decrease in CD4(+) T cells is then observed in the lamina propria of the rectosigmoid colon only.
Collapse
Affiliation(s)
- Anne Couëdel-Courteille
- Viral Transmission and Dissemination Group, Immunology Department, Institut Cochin, INSERM U567, CNRS UMR8104, Université Paris 5-René Descartes, 22 rue Méchain, 75014 Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Zhu T, Corey L, Hwangbo Y, Lee JM, Learn GH, Mullins JI, McElrath MJ. Persistence of extraordinarily low levels of genetically homogeneous human immunodeficiency virus type 1 in exposed seronegative individuals. J Virol 2003; 77:6108-16. [PMID: 12743268 PMCID: PMC154986 DOI: 10.1128/jvi.77.11.6108-6116.2003] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Some individuals remain inexplicably seronegative and lack evidence for human immunodeficiency virus type 1 (HIV-1) infection by conventional serologic or virologic testing despite repeated high-risk virus exposures. Here, we examined 10 exposed seronegative (ES) individuals exhibiting HIV-1-specific cytotoxicity for the presence of HIV-1. We discovered HIV-1 DNA in resting CD4(+) T cells (mean, 0.05 +/- 0.01 copies per million cells) at multiple visits spanning 69 to 130 weeks in two ES individuals at levels that were on average 10(4)- to 10(6)-fold lower than those of other HIV-1-infected populations reported. Sequences of HIV-1 envelope and gag genes remained markedly homogeneous, indicating little to undetectable virus replication. These results provide the evidence for HIV-1 infection in ES individuals below the detection limit of standard assays, suggesting that extraordinary control of infection can occur. The two HIV-infected ES individuals remained healthy and were not superinfected with other HIV-1 strains despite continued high-risk sexual exposures to multiple HIV-infected partners. Understanding the mechanisms that confer diminished replicative capacity of HIV-1 in these hosts is paramount to developing strategies for protection against and control of HIV-1 infection.
Collapse
Affiliation(s)
- Tuofu Zhu
- Department of Laboratory Medicine, University of Washington School of Medicine, Seattle 98195, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
Hu DJ, Subbarao S, Vanichseni S, Mock PA, van Griensven F, Nelson R, Nguyen L, Kitayaporn D, Young NL, Des Jarlais D, Byers R, Choopanya K, Mastro TD. Higher viral loads and other risk factors associated with HIV-1 seroconversion during a period of high incidence among injection drug users in Bangkok. J Acquir Immune Defic Syndr 2002; 30:240-7. [PMID: 12045687 DOI: 10.1097/00042560-200206010-00013] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We analyzed data from a prospective cohort study of injection drug users (IDUs) attending methadone treatment clinics in Bangkok, Thailand, during 1995-1998 to characterize factors associated with a period of high incidence (PHI) from July 1996 through January 1997 compared with periods of lower incidence. Sociobehavioral characteristics were similar for all participants during and outside the PHI except for the following: there was more reported drug injection while IDUs were incarcerated during the PHI (odds ratio, 1.67; p =.02) and significantly higher proportions of persons reported heroin injection (91% vs. 75%, respectively; p =.02) and higher frequencies of daily injection and sharing of injection equipment (40% vs. 25%, respectively; p =.05) during the PHI than outside the PHI. Through most of the first year after seroconversion, plasma HIV-1 loads were significantly higher in persons who seroconverted during the PHI than in those who seroconverted outside the PHI. Higher viral loads may potentially contribute to faster disease progression and increased infectiousness or transmissibility to subsequent contacts. Our findings suggest that prevention efforts to reduce the effective size and turnover within IDU sharing networks may have a significant impact on the epidemic by disrupting the rapid transmission of HIV-1 from recently infected, highly infectious individuals.
Collapse
Affiliation(s)
- Dale J Hu
- HIV Vaccine Section Epidemiology Branch/DHAP, Centers for Disease Control and Prevention, Atlanta, Georgia 30333, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
O'Connor DH, Allen TM, Vogel TU, Jing P, DeSouza IP, Dodds E, Dunphy EJ, Melsaether C, Mothé B, Yamamoto H, Horton H, Wilson N, Hughes AL, Watkins DI. Acute phase cytotoxic T lymphocyte escape is a hallmark of simian immunodeficiency virus infection. Nat Med 2002; 8:493-9. [PMID: 11984594 DOI: 10.1038/nm0502-493] [Citation(s) in RCA: 319] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Cytotoxic T-lymphocyte (CTL) responses peak coincident with the decline in acute HIV viremia. Despite two reports of CTL-resistant HIV variants emerging during acute infection, the contribution of acute CTL escape to HIV pathogenesis remains unclear. Difficulties inherent in studying acute HIV infection can be overcome by modeling virus-host interactions in SIV-infected rhesus macaques. We sequenced 21 complete simian immunodeficiency virus (SIV)mac239 genomes at four weeks post-infection to determine the extent of acute CTL escape. Here we show that viruses from 19 of 21 macaques escaped from CTLs during acute infection and that these escape-selecting CTLs were responsive to lower concentrations of peptide than other SIV-specific CTLs. Interestingly, CTLs that require low peptide concentrations for stimulation (high 'functional avidity') are particularly effective at controlling other viral infections. Our results suggest that acute viral escape from CTLs is a hallmark of SIV infection and that CTLs with high functional avidity can rapidly select for escape variants.
Collapse
Affiliation(s)
- David H O'Connor
- Wisconsin Regional Primate Research Center and Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Hu DJ, Vanichseni S, Mastro TD, Raktham S, Young NL, Mock PA, Subbarao S, Parekh BS, Srisuwanvilai L, Sutthent R, Wasi C, Heneine W, Choopanya K. Viral load differences in early infection with two HIV-1 subtypes. AIDS 2001; 15:683-91. [PMID: 11371682 DOI: 10.1097/00002030-200104130-00003] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Information on early HIV-1 infection has come primarily from studies of persons infected with subtype B in North America and Europe; much less is known about other subtypes. The purpose of the present study was to compare the virologic and immunologic parameters following seroconversion among recently-infected persons infected with either of two different HIV-1 subtypes. METHOD A prospective cohort study was carried out at methadone treatment clinics administered by the Bangkok Metropolitan Administration, Thailand. A total of 130 HIV-1-infected seroconverters (103 with HIV-1 subtype E and 27 with subtype B) were included in the study. The main outcome measures were serial HIV-1 RNA viral load, natural killer cell percentage, CD4 and CD8 lymphocyte counts since seroconversion. RESULTS The demographic and behavioral characteristics of persons with either subtype were similar. Median RNA viral levels at the earliest time within 3 months of seroconversion were more than three times higher for persons infected with subtype E than subtype B (63 100 versus 18 050 copies/ml, P = 0.001). However, this difference decreased over time such that viral loads were similar at 12, 18, and 24 months following seroconversion. The CD4 and CD8 lymphocyte counts were similar in infections with either subtype during the entire period up to 24 months post-seroconversion. CONCLUSIONS Higher viral loads associated with subtype E may result from inter-subtype biological differences; however, the epidemiological dynamics of transmission in Bangkok may have also contributed to this phenomenon.
Collapse
Affiliation(s)
- D J Hu
- Division of HIV/AIDS Prevention--Surveillance and Epidemiology, National Center for HIV, STD, and TB Prevention, Centers for Disease Control and Prevention (CDC), Atlanta, Georgia 30333, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Polyanskaya N, Bergmeier LA, Sharpe SA, Cook N, Leech S, Hall G, Dennis M, ten Haaft P, Heeney J, Manca F, Lehner T, Cranage MP. Mucosal exposure to subinfectious doses of SIV primes gut-associated antibody-secreting cells and T cells: lack of enhancement by nonneutralizing antibody. Virology 2001; 279:527-38. [PMID: 11162808 DOI: 10.1006/viro.2000.0704] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It has been suggested that the presence of immunoglobulin and complement receptors on rectal epithelium may facilitate the entry of HIV complexed to nonneutralizing antibody. We tested this hypothesis using simian immunodeficiency virus (SIV) infection of rhesus macaques. First, in a pilot study, a nonneutralizing IgG fraction of macaque anti-SIV gp120 was shown to enhance the immunogenicity of SIV envelope following rectal immunization. The same antibody was then mixed with a subinfectious dose of SIV and the occurrence of rectal infection was compared with virus alone. Animals were not infected overtly and were rechallenged with a 10-fold higher dose of virus with and without addition of antibody. There was no evidence of antibody-mediated infection, since equal numbers of macaques became infected, regardless of the presence of antibody. In addition, the application of immune complexes did not alter significantly the subsequent virus load or the immune responses generated. In seronegative animals, in which virus and proviral DNA were undetectable in PBMC and tissues, SIV-specific T-cell responses and antibody-secreting cells were found in systemic and gut-associated sites. Our results show that nonneutralizing antibody neither facilitated nor enhanced rectal infection with SIV, in the small number of animals used, despite the consistent trend for this antibody to enhance antibody responses to gp120 following rectal immunization with immune-complexed antigen. However, mucosal exposure to subinfectious doses of virus primed both systemic and local immunity, regardless of addition of nonneutralizing antibody.
Collapse
Affiliation(s)
- N Polyanskaya
- Centre for Applied Microbiology and Research (CAMR), Salisbury, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Otten RA, Smith DK, Adams DR, Pullium JK, Jackson E, Kim CN, Jaffe H, Janssen R, Butera S, Folks TM. Efficacy of postexposure prophylaxis after intravaginal exposure of pig-tailed macaques to a human-derived retrovirus (human immunodeficiency virus type 2). J Virol 2000; 74:9771-5. [PMID: 11000253 PMCID: PMC112413 DOI: 10.1128/jvi.74.20.9771-9775.2000] [Citation(s) in RCA: 160] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Postexposure prophylaxis (PEP) after intravaginal exposure to human immunodeficiency virus (HIV) was investigated using the HIV type 2 (HIV-2)/pig-tailed macaque transmission model. PEP for 28 days with the reverse transcriptase inhibitor (R)-9-(2-phosphonylmethoxypropyl)adenine (PMPA; tenofovir) was initiated 12 to 72 h following HIV-2 exposure. Systemic infection was not evident in the 12- and 36-h groups, as defined by plasma viremia, cell-associated provirus, antibody responses, and lymph node virus. Breakthrough infection in the 72-h group was detected at week 16 post-virus exposure. These results demonstrate for the first time using a vaginal transmission model that early intervention after high-risk sexual exposures may prevent infection.
Collapse
Affiliation(s)
- R A Otten
- Division of AIDS, STD, and TB Laboratory Research, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia 30333, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Luabeya MK, Dallasta LM, Achim CL, Pauza CD, Hamilton RL. Blood-brain barrier disruption in simian immunodeficiency virus encephalitis. Neuropathol Appl Neurobiol 2000; 26:454-62. [PMID: 11054186 DOI: 10.1046/j.1365-2990.2000.00275.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Infected monocyte-derived macrophages (MDM) are thought by some investigators to play a central role in the neuropathogenesis of human immunodeficiency virus encephalitis (HIVE). It was recently proposed that these cells gain access to the central nervous system (CNS) through disruptions in blood-brain barrier (BBB) tight junctions, which occur in HIVE in association with accumulation of activated, HIV-1-infected, perivascular macrophages and serum protein extravasation (Am J Pathol 1999, 155: 1915-27). The present study tested this hypothesis in basal ganglia tissue from simian immunodeficiency virus (SIV)-infected macaques with encephalitis by examining vessels for immunohistochemical alterations in the tight junction-associated proteins, occludin and zonula occludens-1 (ZO-1). Compared to non-infected macaques and SIV-infected macaques without encephalitis, cerebral vessels from macaques with SIVE showed fragmentation and decreased immunoreactivity for both tight junction proteins. These alterations were associated with accumulation of perivascular macrophages and aberrant occludin and ZO-1 immunoreactivity within these cells. In addition, perivascular extravasation of fibrinogen, a plasma protein, and a change from a strong linear staining pattern to a more irregular pattern of glucose transporter isoform-1 (GLUT-1), a metabolic BBB marker, were observed in regions with vascular tight junction protein alterations. These findings demonstrate that tight junction disruption occurs in SIVE in association with perivascular macrophage accumulation. While it cannot be ascertained from these studies whether such changes precede macrophage infiltration, or are secondary to the chronic presence of macrophages around cerebral vessels, disruptions in BBB integrity could serve as portals for additional accumulation of perivascular macrophages in SIVE.
Collapse
Affiliation(s)
- M K Luabeya
- Department of Pathology (Division of Neuropathology), University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | |
Collapse
|
27
|
Mäkitalo B, Böttiger1 P, Biberfeld G, Thorstensson R. Cell-mediated immunity to low doses of SIVsm in cynomolgus macaques did not confer protection against mucosal rechallenge. Vaccine 2000; 19:298-307. [PMID: 10930685 DOI: 10.1016/s0264-410x(00)00164-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Simian immunodeficiency virus (SIV) infection of macaques is a useful model for studies of the roles of different immune responses against viruses that cause (AIDS). In this study, six cynomolgus macaques were inoculated intrarectally with subinfectious or infectious doses of SIVsm to assess the SIV specific immunity, in particular protective immunity against subsequent challenge with a higher dose of SIVsm. Following the first inoculation with SIVsm, the two monkeys given the highest doses of cell-free SIVsm stock and one monkey given the intermediate dose became infected. In the three remaining animals, one animal inoculated with an intermediate dose and two animals given low doses of SIVsm, no overt infection occurred. Nevertheless, SIV specific cytotoxic T-cells against Gag/Pol and Nef proteins and T-cell proliferative responses against HIV-2 whole viral lysate, native HIV-2 gp125, recombinant SIV gp140 and SIV Env synthetic peptides were detected. After intrarectal rechallenge of the uninfected macaques with a higher dose of SIVsm all the animals became infected. These results demonstrate that cell mediated immunity can occur in the absence of detectable infection in monkeys inoculated with a low dose of SIVsm. Despite the presence of cellular immune responses, the animals were not protected when challenged with a higher dose of virus later.
Collapse
Affiliation(s)
- B Mäkitalo
- Swedish Institute for Infectious Disease Control and Microbiology Tumour Biology Center, Karolinska Institute, SE-171 82, Solna, Sweden.
| | | | | | | |
Collapse
|
28
|
Wallace M, Pyzalski R, Horejsh D, Brown C, Djavani M, Lu Y, Hanson JM, Mitchen JL, Perlman SB, Pauza CD. Whole body positron emission tomography imaging of activated lymphoid tissues during acute simian-human immunodeficiency virus 89.6PD infection in rhesus macaques. Virology 2000; 274:255-61. [PMID: 10964769 DOI: 10.1006/viro.2000.0479] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mechanisms of acute retroviral pathogenesis have been examined during primary infection of rhesus macaques with simian-human immunodeficiency virus 89.6PD (SHIV(89.6PD)). During acute infection, between initial exposure and establishment of antigen-specific immune responses that stabilize the virus burden, rapid immune system changes influence the viral set-point and dictate subsequent steps in disease progression. In a previous study, we described specific patterns of lymphocyte activation during acute SHIV(89.6PD) infection. We now extend these studies to describe lymphoid tissue activation, using whole body positron emission tomography (PET) and the radioactive tracer 2-[(18)F]fluorodeoxyglucose (FDG). Within a few days after primary infection by intravenous, intrarectal, or intravaginal routes, PET-FDG imaging revealed a distinct pattern of lymphoid tissue activation centered on axillary, cervical, and mediastinum lymph nodes. Increased tissue FDG uptake preceded fulminant virus replication at these sites, suggesting that a diffusible factor of host or viral origin was responsible for lymphoid tissue changes. These data show that activation of lymphoid tissues in the upper body is an early response to virus infection and that diffusible mediators of activation might be important targets for vaccine or therapeutic intervention strategies.
Collapse
Affiliation(s)
- M Wallace
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Wisconsin 53705-1532, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Obert LA, Hoover EA. Feline immunodeficiency virus clade C mucosal transmission and disease courses. AIDS Res Hum Retroviruses 2000; 16:677-88. [PMID: 10791878 DOI: 10.1089/088922200308909] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The transmissibility and pathogenicity of a clade C feline immunodeficiency virus (FIV-C) was examined via the oral-nasal, vaginal, or rectal mucosa. FIV-C was transmissible by all three mucosal routes. Vaginal transmission was most efficient (100%), oral exposure resulted in a 80% infection rate, and rectal transmission was least effective (44%). In contrast to previous intravenous passage studies, a broader range of host-virus relationships was observed after mucosal exposure. Three categories of FIV-C infection were defined: (1) rapidly progressive infection marked by high virus burdens and rapid CD4+ cell depletion (43% of vaginally exposed animals); (2) conventional (typical) infection featuring slowly progressive CD4+ cell decline (61% of all exposed animals); and (3) regressive (transient) infection marked by low and then barely detectable virus burdens and no CD4+ cell alterations (22% of rectally inoculated cats). These disease courses appear to have parallels in mucosal HIV and SIV infections, emphasizing the importance of the virus-mucosa interface in lentiviral pathogenesis.
Collapse
Affiliation(s)
- L A Obert
- Department of Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins 80523, USA
| | | |
Collapse
|
30
|
Wallace M, Waterman PM, Mitchen JL, Djavani M, Brown C, Trivedi P, Horejsh D, Dykhuizen M, Kitabwalla M, Pauza CD. Lymphocyte activation during acute simian/human immunodeficiency virus SHIV(89.6PD) infection in macaques. J Virol 1999; 73:10236-44. [PMID: 10559340 PMCID: PMC113077 DOI: 10.1128/jvi.73.12.10236-10244.1999] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Host-virus interactions control disease progression in human immunodeficiency virus-infected human beings and in nonhuman primates infected with simian or simian/human immunodeficiency viruses (SHIV). These interactions evolve rapidly during acute infection and are key to the mechanisms of viral persistence and AIDS. SHIV(89.6PD) infection in rhesus macaques can deplete CD4(+) T cells from the peripheral blood, spleen, and lymph nodes within 2 weeks after exposure and is a model for virulent, acute infection. Lymphocytes isolated from blood and tissues during the interval of acute SHIV(89.6PD) infection have lost the capacity to proliferate in response to phytohemagglutinin (PHA). T-cell unresponsiveness to mitogen occurred within 1 week after mucosal inoculation yet prior to massive CD4(+) T-cell depletion and extensive virus dissemination. The lack of mitogen response was due to apoptosis in vitro, and increased activation marker expression on circulating T cells in vivo coincided with the appearance of PHA-induced apoptosis in vitro. Inappropriately high immune stimulation associated with rapid loss of mature CD4(+) T cells suggested that activation-induced cell death is a mechanism for helper T-cell depletion in the brief period before widespread virus dissemination. Elevated levels of lymphocyte activation likely enhance SHIV(89.6PD) replication, thus increasing the loss of CD4(+) T cells and diminishing the levels of virus-specific immunity that remain after acute infection. The level of surviving immunity may dictate the capacity to control virus replication and disease progression. We describe this level of immune competence as the host set point to show its pivotal role in AIDS pathogenesis.
Collapse
Affiliation(s)
- M Wallace
- Department of Pathology, University of Wisconsin, Madison, Wisconsin 53705-1532, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Luciw PA, Mandell CP, Himathongkham S, Li J, Low TA, Schmidt KA, Shaw KE, Cheng-Mayer C. Fatal immunopathogenesis by SIV/HIV-1 (SHIV) containing a variant form of the HIV-1SF33 env gene in juvenile and newborn rhesus macaques. Virology 1999; 263:112-27. [PMID: 10544087 DOI: 10.1006/viro.1999.9908] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
SIV/HIV-1 (SHIV) chimeric clones, constructed by substituting portions of the pathogenic molecular clone SIVmac239 with counterpart portions from HIV-1 clones, provide a means to analyze functions of selected HIV-1 genes in vivo in nonhuman primates. Our studies focused on SHIVSF33, which contains the vpu, tat, rev, and env genes of the cytopathic, T-cell line tropic clone HIV-1sf33 (subtype-B); this clone has a premature stop codon in the vpu gene. In three juvenile macaques inoculated intravenously with SHIVSF33, low-level persistent infection was established; no disease was observed for a period of >2 years. However, at approximately 16 months p.i., one of four SHIVSF33-infected juvenile macaques exhibited an increase in virus load, depletion of CD4(+) T cells in peripheral blood and lymph nodes, and other symptoms of simian AIDS (SAIDS). Virus recovered from this animal in the symptomatic stage was designated SHIVSF33a (A, adapted); this virus displayed multiple amino acid sequence changes throughout the HIV-1 env gene compared with the input SHIVSF33 clone. Additionally, a mutation in all clones from SHIVSF33a restored the open reading frame for the vpu gene. In vitro evaluations in tissue-culture systems revealed that SHIVSF33a replicated to higher levels and exhibited greater cytopathicity than SHIVSF33. Furthermore cloned env genes for SHIVSF33a were more fusogenic in a cell-fusion assay compared with the env gene of the SHIVSF33. Intravenous inoculation of SHIVsf33a into juvenile and newborn macaques resulted in a rapid decline in CD4(+) T cells to very low levels and development of a fatal AIDS-like disease. A cell-free preparation of this pathogenic chimeric virus also established persistent infection when applied to oral mucosal membranes of juvenile macaques and produced a fatal AIDS-like disease. These studies on pathogenic SHIVSF33a establish the basis for further investigations on the role of the HIV-1 env gene in virus adaptation and in mechanism(s) of immunodeficiency in primates; moreover, the chimeric virus SHIVSF33a can play a role in elucidating mucosal membrane transmission and development of antiviral vaccines in newborns as well as juvenile and adult macaques.
Collapse
Affiliation(s)
- P A Luciw
- Department of Medical Pathology, University of California, Davis, California 95616, USA.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Couëdel-Courteille A, Butor C, Juillard V, Guillet JG, Venet A. Dissemination of SIV after rectal infection preferentially involves paracolic germinal centers. Virology 1999; 260:277-94. [PMID: 10417263 DOI: 10.1006/viro.1999.9809] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Homosexual transmission remains a major mode of contamination in developed countries. Early virological and immunological events in lymphoid tissues are known to be important for the outcome of HIV infections. Little data are available, however, on viral dissemination during primary rectal infection. We therefore studied this aspect of rectal infection in rhesus macaques inoculated with the biological isolate SIVmac251. We show that infection is established initially in lymph nodes draining the rectum. Infected cells and virions are localized mainly in germinal centers at that stage. With increasing viral burden, infected cells are found throughout the lymph node parenchyma. In addition the difference in viral load between lymph nodes draining the rectum and other lymph nodes is attenuated or abolished. We discuss this pattern of viral dissemination with respect to the physiology of the mucosal immune system. The pattern and kinetics of viral dissemination after rectal infection have important implications for the development of efficient mucosal vaccines.
Collapse
Affiliation(s)
- A Couëdel-Courteille
- Laboratoire d'Immunologie des Pathologies Infectieuses et Tumorales, Institut National de la Santé et de la Recherche Médicale U445, Institut Cochin de Génétique Moléculaire, 22 rue Méchain, Paris, 75014, France.
| | | | | | | | | |
Collapse
|
33
|
Staprans SI, Dailey PJ, Rosenthal A, Horton C, Grant RM, Lerche N, Feinberg MB. Simian immunodeficiency virus disease course is predicted by the extent of virus replication during primary infection. J Virol 1999; 73:4829-39. [PMID: 10233944 PMCID: PMC112526 DOI: 10.1128/jvi.73.6.4829-4839.1999] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To elucidate the relationship between early viral infection events and immunodeficiency virus disease progression, quantitative-competitive and branched-DNA methods of simian immunodeficiency virus (SIV) RNA quantitation were cross-validated and used to measure viremia following infection of rhesus macaques with the pathogenic SIVmac251 virus isolate. Excellent correlation between the methods suggests that both accurately approximate SIV copy number. Plasma viremia was evident 4 days postinfection, and rapid viral expansion led to peak viremia levels of 10(7) to 10(9) SIV RNA copies/ml by days 8 to 17. Limited resolution of primary viremia was accompanied by relatively short, though variable, times to the development of AIDS (81 to 630 days). The persistent high-level viremia observed following intravenous inoculation of SIVmac251 explains the aggressive disease course in this model. Survival analyses demonstrated that the disease course is established 8 to 17 days postinfection, when peak viremia is observed. The most significant predictor of disease progression was the extent of viral decline following peak viremia; larger decrements in viremia were associated with both lower steady-state viremia (P = 0.0005) and a reduced hazard of AIDS (P = 0.004). The data also unexpectedly suggested that following SIVmac251 infection, animals with the highest peak viremia were better able to control virus replication rather than more rapidly developing disease. Analysis of early viral replication dynamics should help define host responses that protect from disease progression and should provide quantitative measures to assess the extent to which protective responses may be induced by prophylactic vaccination.
Collapse
Affiliation(s)
- S I Staprans
- Department of Medicine-AIDS Program, University of California, San Francisco, San Francisco, California 94110, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Yin C, Wu MS, Pauza CD, Salvato MS. High major histocompatibility complex-unrestricted lysis of simian immunodeficiency virus envelope-expressing cells predisposes macaques to rapid AIDS progression. J Virol 1999; 73:3692-701. [PMID: 10196261 PMCID: PMC104144 DOI: 10.1128/jvi.73.5.3692-3701.1999] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/1998] [Accepted: 01/13/1999] [Indexed: 11/20/2022] Open
Abstract
Before the development of virus-specific immune responses, peripheral blood mononuclear cells (PBMC) from uninfected rhesus monkeys and human beings have the capacity to lyse target cells expressing simian immunodeficiency virus (SIV) or human immunodeficiency virus-1 (HIV) envelope (gp130 and gp120) antigens. Lysis by naive effector cells does not require major histocompatibility complex (MHC)-restricted antigen presentation, is equally effective for allogeneic and xenogeneic targets, and is designated MHC-unrestricted (UR) lysis. UR lysis is not sensitive to EGTA and does not require de novo RNA or protein synthesis. Several kinds of envelope-expressing targets, including cells that poorly express MHC class I antigens, can be lysed. CD4(+) effectors are responsible for most of the lytic activity. High lysis is correlated with high expression of HIV or SIV envelope, specifically, the central one-third of the gp130 molecule, and lysis is completely inhibited by a monoclonal antibody against envelope. Our work extends observations of human lymphocytes expressing HIV gp120 to the SIV/rhesus monkey model for AIDS. Additionally, we address the relevance of UR lysis in vivo. A survey of PBMC from 56 uninfected rhesus monkeys indicates that 59% of the individuals had peak UR lytic activity above 15% specific lysis. Eleven of these monkeys were subsequently infected with SIV. Animals with UR lytic activity above 15% specific lysis were predisposed to more rapid disease progression than animals with low UR lytic activity, suggesting a strong correlation between this form of innate immunity and disease progression to AIDS.
Collapse
Affiliation(s)
- C Yin
- Department of Pathology and Laboratory Medicine, University of Wisconsin Medical School, Madison, Wisconsin 53706, USA
| | | | | | | |
Collapse
|
35
|
Fultz PN, Wei Q, Yue L. Rectal transmission of human immunodeficiency virus type 1 to chimpanzees. J Infect Dis 1999; 179 Suppl 3:S418-21. [PMID: 10099110 DOI: 10.1086/314796] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Inoculation of chimpanzees with human immunodeficiency virus type 1 (HIV-1) has been used as a model system to define mechanisms of pathogenesis and to test protective efficacy of candidate HIV-1 vaccines. In most of these studies, the animals were inoculated intravenously. However, because HIV-1 is transmitted primarily across mucosal surfaces, future evaluations of vaccines should employ mucosal routes for administering infectious virus to immunized animals. To develop a model of rectal transmission of HIV-1, chimpanzees were exposed without trauma to 4 different HIV-1 strains at doses ranging from 200 to 10,000 TCIDs. Infection, characterized by seroconversion and repeated isolation of virus from lymphocytes, was established in 1 of 5 animals. This animal was sequentially inoculated with a subtype B and then an E strain and was infected with both strains. The results show that rectal exposure of adult chimpanzees to cell-free HIV-1 was not an efficient mode of transmission in this cohort.
Collapse
Affiliation(s)
- P N Fultz
- Department of Microbiology, University of Alabama School of Medicine, Birmingham, AL 35294, USA.
| | | | | |
Collapse
|
36
|
Harouse JM, Gettie A, Tan RC, Blanchard J, Cheng-Mayer C. Distinct pathogenic sequela in rhesus macaques infected with CCR5 or CXCR4 utilizing SHIVs. Science 1999; 284:816-9. [PMID: 10221916 DOI: 10.1126/science.284.5415.816] [Citation(s) in RCA: 267] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Infection of macaques with chimeric simian-human immunodeficiency virus (SHIV) provides an excellent in vivo model for examining the influence of envelope on HIV-1 pathogenesis. Infection with a pathogenic CCR5 (R5)-specific enveloped virus, SHIVSF162P, was compared with infection with the CXCR4 (X4)-specific SHIVSF33A.2. Despite comparable levels of viral replication, animals infected with the R5 and X4 SHIV had distinct pathogenic outcomes. SHIVSF162P caused a dramatic loss of CD4+ intestinal T cells followed by a gradual depletion in peripheral CD4+ T cells, whereas infection with SHIVSF33A.2 caused a profound loss in peripheral T cells that was not paralleled in the intestine. These results suggest a critical role of co-receptor utilization in viral pathogenesis and provide a reliable in vivo model for preclinical examination of HIV-1 vaccines and therapeutic agents in the context of the HIV-1 envelope protein.
Collapse
Affiliation(s)
- J M Harouse
- Aaron Diamond AIDS Research Center, The Rockefeller University, 455 First Avenue, 7th Floor, New York, NY 10016, USA
| | | | | | | | | |
Collapse
|
37
|
Polacino P, Stallard V, Montefiori DC, Brown CR, Richardson BA, Morton WR, Benveniste RE, Hu SL. Protection of macaques against intrarectal infection by a combination immunization regimen with recombinant simian immunodeficiency virus SIVmne gp160 vaccines. J Virol 1999; 73:3134-46. [PMID: 10074165 PMCID: PMC104075 DOI: 10.1128/jvi.73.4.3134-3146.1999] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/1998] [Accepted: 01/03/1999] [Indexed: 11/20/2022] Open
Abstract
We previously reported that immunization with recombinant simian immunodeficiency virus SIVmne envelope (gp160) vaccines protected macaques against intravenous challenge by the cloned homologous virus E11S but that this protection was only partially effective against the uncloned virus, SIVmne. In the present study, we examine the protective efficacy of this immunization regimen against infection by a mucosal route. We found that the same gp160-based vaccines were highly effective against intrarectal infection not only with the E11S clone but also with the uncloned SIVmne. Protection against mucosal infection is therefore achievable by parenteral immunization with recombinant envelope vaccines. Protection appears to correlate with high levels of SIV-specific antibodies and, in animals protected against the uncloned virus, the presence of serum-neutralizing activities. To understand the basis for the differential efficacies against the uncloned virus by the intravenous versus the intrarectal routes, we examined viral sequences recovered from the peripheral blood mononuclear cells of animals early after infection by both routes. We previously showed that the majority (85%) of the uncloned SIVmne challenge stock contained V1 sequences homologous to the molecular clone from which the vaccines were made (E11S type), with the remainder (15%) containing multiple conserved changes (the variant types). In contrast to intravenously infected animals, from which either E11S-type or the variant type V1 sequences could be recovered in significant proportions, animals infected intrarectally had predominantly E11S-type sequences. Preferential transmission or amplification of the E11S-type viruses may therefore account in part for the enhanced efficacy of the recombinant gp160 vaccines against the uncloned virus challenge by the intrarectal route compared with the intravenous route.
Collapse
Affiliation(s)
- P Polacino
- Regional Primate Research Center, University of Washington, Seattle, Washington, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Murphey-Corb M, Wilson LA, Trichel AM, Roberts DE, Xu K, Ohkawa S, Woodson B, Bohm R, Blanchard J. Selective Induction of Protective MHC Class I-Restricted CTL in the Intestinal Lamina Propria of Rhesus Monkeys by Transient SIV Infection of the Colonic Mucosa. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.1.540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
The identification of mucosal immune responses required for protection against sexual transmission of HIV is essential for the development of an efficacious vaccine. To gain a better understanding of these responses, we have characterized the immune responses in the lamina propria (LP) and epithelium of the jejunum, the mesenteric lymph nodes, and peripheral blood (PBMC) of 11 rhesus monkeys following colonic exposure to two molecular clones of SIV. Two monkeys had no signs of infection. Three monkeys became persistently infected. Transient infections, characterized by the sporadic detection of virus in the periphery and/or detection of SIV-specific immune responses in either the gut-associated tissues or PBMC, were induced in six of the monkeys. One persistently infected and three transiently infected monkeys had high levels of SIV env-specific MHC class I restricted CTL in the jejunal LP. Another transiently infected monkey had SIV-specific IgA secreting B cells in the LP. Three or six months postexposure, these animals and four naive controls were challenged intracolonically with the heterologous primary isolate, SIV/DeltaB670. All four monkeys with strong SIV env-specific MHC-restricted CTL in the LP were protected, whereas none of the naive controls or the remaining seven monkeys with little or no CTL in the LP were protected. These experiments provide the first direct evidence that transient mucosal infection can induce SIV-specific immunity that remains localized to the gut-associated tissues. Furthermore, a strong correlation between SIV env-specific MHC-restricted CTL in the LP and protection against colonic mucosal challenge was observed.
Collapse
Affiliation(s)
| | | | - Anita M. Trichel
- *Tulane Regional Primate Research Center, Covington, LA 70433; and
| | | | - Keyu Xu
- *Tulane Regional Primate Research Center, Covington, LA 70433; and
| | - Susumu Ohkawa
- *Tulane Regional Primate Research Center, Covington, LA 70433; and
| | - Bruce Woodson
- †Department of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Rudolf Bohm
- *Tulane Regional Primate Research Center, Covington, LA 70433; and
| | - James Blanchard
- *Tulane Regional Primate Research Center, Covington, LA 70433; and
| |
Collapse
|
39
|
Enose Y, Ibuki K, Shimada T, Ui M, Hayami M. Genomic analysis of the viral population in genital secretions early after infection of simian immunodeficiency viruses in macaque monkeys. Microbiol Immunol 1998; 42:715-22. [PMID: 9858467 DOI: 10.1111/j.1348-0421.1998.tb02344.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
To clarify the change in the viral population during passage from the vaginal cavity to blood circulation and vice versa, we examined the viral clones detected in cells in vaginal washes (VWCs) early after inoculation and after systemic infection with polyclonal SIV. In two intravaginally inoculated monkeys, the viral clones found in VWCs at 18 days p.i. were shown to be some of those contained in the inoculum, whereas the viral population in the peripheral blood mononuclear cells (PBMCs) was a monotype. This gradual decrease of viral clones suggested the possible existence of two barriers, one at the genital tract and the other between the genital tract and the blood. Later, at one month p.i., the viral clones in VWCs became rather restricted, whereas those in PBMCs diverged from a single clone to several clones. This suggested that different mechanisms affect the viral populations in PBMCs and VWCs. In order to examine how the viral population was affected by passage from the blood to the vaginal cavity, a monkey was intravenously inoculated and the viral clones in VWCs were analyzed at 14 days p.i., at a time of the heterogeneous population in PBMCs. The viral population in VWCs was found to be a single clone and this clone was a minor type in PBMCs, suggesting that the major clone in PBMCs was not always secreted to the vaginal cavity.
Collapse
Affiliation(s)
- Y Enose
- Institute for Virus Research, Kyoto University, Kyoto, Japan
| | | | | | | | | |
Collapse
|
40
|
McChesney MB, Collins JR, Lu D, Lu X, Torten J, Ashley RL, Cloyd MW, Miller CJ. Occult systemic infection and persistent simian immunodeficiency virus (SIV)-specific CD4(+)-T-cell proliferative responses in rhesus macaques that were transiently viremic after intravaginal inoculation of SIV. J Virol 1998; 72:10029-35. [PMID: 9811741 PMCID: PMC110525 DOI: 10.1128/jvi.72.12.10029-10035.1998] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The intact cervicovaginal mucosa is a relative barrier to the sexual transmission of human immunodeficiency virus type 1 (HIV-1). In the simian immunodeficiency virus (SIV) macaque model of HIV infection, seronegative transient viremia (STV; virus isolation positive followed by repeated negative cultures) occurs after intravaginal inoculation of a low dose of pathogenic SIVmac251 (C. J. Miller, M. Marthas, J. Torten, N. Alexander, J. Moore, G. Doncel, and A. Hendrickx, J. Virol. 68:6391-6400, 1994). Thirty-one adult female macaques that had been inoculated intravaginally with pathogenic SIVmac251 became transiently viremic. One monkey that had been culture negative for a year after SIV inoculation became persistently viremic and developed simian AIDS. No other STV monkey developed persistent viremia or disease. Results of very sensitive assays showed that 6 of 31 monkeys had weak SIV-specific antibody responses. SIV-specific antibodies were not detected in the cervicovaginal secretions of 10 STV monkeys examined. Twenty of 26 monkeys had lymphocyte proliferative responses to p55(gag) and/or gp130(env) antigens; 3 of 6 animals, including the monkey that became persistently viremic, had detectable cytotoxic T-lymphocyte (CTL) responses to SIV. At necropsy, lymphoid tissues and vaginal mucosa were virus culture negative, but in 10 of 10 animals, SIV provirus was detected by PCR using gag-specific primer pairs. Fifty percent of the PCR-positive tissue samples were also positive for SIV gag RNA by reverse transcriptase PCR. Thus, transient viremia following intravaginal inoculation of pathogenic SIV is associated with persistent, systemic infection, either latent or very low level productive. Atypical immune responses, characterized by lymphocyte proliferation and some CTL responses in the absence of conventionally detectable antibodies, develop in transiently viremic monkeys.
Collapse
Affiliation(s)
- M B McChesney
- California Regional Primate Research Center, University of California-Davis, Davis, California 95616, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Girard M, Mahoney J, Wei Q, van der Ryst E, Muchmore E, Barré-Sinoussi F, Fultz PN. Genital infection of female chimpanzees with human immunodeficiency virus type 1. AIDS Res Hum Retroviruses 1998; 14:1357-67. [PMID: 9788677 DOI: 10.1089/aid.1998.14.1357] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
To develop an animal model for mucosal HIV-1 infection, adult chimpanzees were inoculated without trauma by depositing the virus inoculum at the entrance to the cervical canal with a rigid catheter to which flexible tubing was attached. By this procedure, persistent infections were established in some chimpanzees with various infectious doses of either cell-associated HIV-1LAI(IIIB) (peripheral blood mononuclear cells from an infected chimpanzee) or with cell-free HIV-1 strains representing subtypes B and E, but not with a subtype A strain. Although some animals did not become infected until after the second or third cervicovaginal exposure, one chimpanzee was clearly infected after one exposure by several criteria, including virus isolation, but this animal did not seroconvert. A second chimpanzee appeared to be resistant to infection despite repeated mucosal exposures at irregular intervals. However, lymphocytes from both of these animals exhibited low-level proliferative responses to HIV-1 but not SIV antigens. Despite these apparently abortive or latent infections, after exposure to HIV-1 by the intravenous route, both animals developed systemic infections and seroconverted. Overall, 8 of 10 chimpanzees were infected systemically after one to three cervicovaginal exposures to HIV-1LAI(IIIB). The results indicate that (1) HIV-1 productive infection of female chimpanzees by the cervicovaginal route generally requires more than one exposure, just as with humans; (2) low level infections without seroconversion can be established after mucosal exposure to HIV; and (3) vaccine efficacy studies involving a single virus challenge of immunized chimpanzees by the cervicovaginal route probably will not be possible.
Collapse
|
42
|
Harouse JM, Tan RC, Gettie A, Dailey P, Marx PA, Luciw PA, Cheng-Mayer C. Mucosal transmission of pathogenic CXCR4-utilizing SHIVSF33A variants in rhesus macaques. Virology 1998; 248:95-107. [PMID: 9705259 DOI: 10.1006/viro.1998.9236] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Infection of macaques with chimeric simian/human immunodeficiency virus (SHIV) expressing the envelope protein of HIV-1 provides a model system for studying HIV-1 infection in humans. To this end, four rhesus macaques (Macaca mulatta) were given a single intravaginal (IVAG) inoculation of cell-free SHIVSF33A and longitudinal samples of peripheral blood and lymph nodes were analyzed for viremia, antigenemia, and various T-cell populations. Rhesus macaques infected IVAG with SHIVSF33A demonstrated a dramatic decrease in the CD4(+) PBMC subset in the initial weeks after viral exposure, a time that corresponded to peak in plasma viremia and antigenemia. Within 4 months of SHIVSF33A inoculation, partial to complete rebound of the CD4(+) PBMC was seen in these animals. Notably, the regeneration of the CD4(+) subset was associated with regeneration of the naive T-cell population and was concordant with clearance of plasma viremia. DNA heteroduplex tracking assays revealed transmission of minor variants within the SHIVSF33A inoculum to the IVAG-inoculated animals. The cell-free SHIVSF33A inoculum as well as virus isolated from animals early after transmission used the chemokine molecule CXCR4 as the primary cellular coreceptor, demonstrating that viruses expressing envelope glycoproteins of the syncytia inducing (SI) phenotype can be transported across the vaginal mucosa. Although none of the animals has yet to develop clinical symptoms of simian AIDS (SAIDS), infectious virus and viral nucleic acids could be persistently isolated from each animal. Furthermore, animals transfused with blood from IVAG-infected macaques drawn 2 weeks after inoculation suffered a more profound and sustained CD4(+) T-cell loss, persistent plasma viremia, and the development of SAIDS in one animal, indicating that IVAG-passaged SHIVSF33A was pathogenic. Taken together, these results establish that a pathogenic CXCR4-utilizing SHIVSF33A species crossed the cervicovaginal mucosa. Different courses of infection in the IVAG versus transfusion animals suggest that host-mediated responses elicited upon transmission across mucosal barriers may serve to limit viral replication and delay disease progression in the IVAG-infected animals.
Collapse
Affiliation(s)
- J M Harouse
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, New York, 10016, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Steger KK, Dykhuizen M, Mitchen JL, Hinds PW, Preuninger BL, Wallace M, Thomson J, Montefiori DC, Lu Y, Pauza CD. CD4+-T-cell and CD20+-B-cell changes predict rapid disease progression after simian-human immunodeficiency virus infection in macaques. J Virol 1998; 72:1600-5. [PMID: 9445063 PMCID: PMC124641 DOI: 10.1128/jvi.72.2.1600-1605.1998] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Simian-human immunodeficiency virus 89.6PD (SHIV89.6PD) was pathogenic after intrarectal inoculation of rhesus macaques. Infection was achieved with a minimum of 2,500 tissue culture infectious doses of cell-free virus stock, and there was no evidence for transient viremia in animals receiving subinfectious doses by the intrarectal route. Some animals experienced rapid progression of disease characterized by loss of greater than 90% of circulating CD4+ T cells, sustained decreases in CD20+ B cells, failure to elicit virus-binding antibodies in plasma, and high levels of antigenemia. Slower-progressing animals had moderate but varying losses of CD4+ T cells; showed increases in circulating CD20+ B cells; mounted vigorous responses to antibodies in plasma, including neutralizing antibodies; and had low or undetectable levels of antigenemia. Rapid progression led to death within 30 weeks after intrarectal inoculation. Plasma antigenemia at 2 weeks after inoculation (P < or = 0.002), B- and T-cell losses (P < or = 0.013), and failure to seroconvert (P < or = 0.005) were correlated statistically with rapid progression. Correlations were evident by 2 to 4 weeks after intrarectal SHIV inoculation, indicating that early events in the host-pathogen interaction determined the clinical outcome.
Collapse
Affiliation(s)
- K K Steger
- Department of Pathology and Laboratory Medicine, University of Wisconsin Medical School, Madison 53706, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Sodora DL, Lee F, Dailey PJ, Marx PA. A genetic and viral load analysis of the simian immunodeficiency virus during the acute phase in macaques inoculated by the vaginal route. AIDS Res Hum Retroviruses 1998; 14:171-81. [PMID: 9462928 DOI: 10.1089/aid.1998.14.171] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A comparative genetic analysis of SIV-infected female macaques during the first 120 days postinfection was undertaken. The same dose of a macaque-passaged SIVmac239(nef open) was administered to three macaques intravenously (i.v.) and to three macaques intravaginally (i.VAG). Clinical outcomes observed ranged from rapid to nonprogression, while two of the i.v.-infected macaques developed an uncommon hindleg paresis. Analysis of viral load (bDNA assay) determined that both i.v.- and i.VAG-infected macaques had comparable high viral loads at the observed viral peak of 14 days postinfection. A study of viral quasispecies diversity by the heteroduplex mobility assay indicated that (1) the i.v.-infected macaques had a highly heterogeneous quasispecies population similar to the infecting viral stock; and (2) in two of three i.VAG-infected macaques multiple viral genotypes (minimum, three or four) were observed in blood and lymph tissues at early times postinfection, which indicated that limited numbers of viral variants crossed the vaginal mucosa and established infection. Therefore, the route of infection can clearly influence early viral selection and diversity. In addition, a third i.VAG-infected macaque, which was a rapid progressor, did not seroconvert and progressed to AIDS in 120 days. This macaque exhibited a high viral load and heterogeneous quasispecies. These data demonstrate differences in the quasispecies complexity associated with route of infection and rate of disease progression.
Collapse
Affiliation(s)
- D L Sodora
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, New York 10016, USA.
| | | | | | | |
Collapse
|
45
|
Bosch ML, Schmidt A, Agy MB, Kimball LE, Morton WR. Infection of Macaca nemestrina neonates with HIV-1 via different routes of inoculation. AIDS 1997; 11:1555-63. [PMID: 9365759 DOI: 10.1097/00002030-199713000-00003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Receptive anal intercourse but not orogenital sex has been identified as a major risk factor for transmission of HIV-1. Recent studies using simian immunodeficiency virus (SIV) in rhesus macaques have demonstrated relatively efficient infection following oral administration, indicating that modes of transmission may vary between HIV-1 and SIV. Here, we investigate whether HIV-1 infection of macaques via the oral route is more efficient than via the rectal route. DESIGN Eleven Macaca nemestrina neonates were exposed to HIV-1 via different routes (four oral, two intravenous, and five rectal). One animal was orally inoculated with a sham inoculum and two control animals were not exposed. METHODS All animals were followed for virological signs of infection, and for pathogenesis associated with HIV-1 infection by general physical examinations, complete blood cell counts and lymphocyte subset analysis, and full necropsies. RESULTS Three out of five rectally exposed macaques and both of the intravenously inoculated animals became infected with HIV-1, whereas none of the orally exposed animals showed evidence of HIV-1 infection. Clinical observations following exposure included failure to thrive in the orally inoculated animals and low CD4/CD8 ratios in the rectally exposed macaques. CONCLUSIONS The finding that, contrary to what has been reported for SIV, transmission of HIV-1 via the oral route is not more efficient than via the rectal route, indicates important biological differences between HIV-1 and SIV, with direct implications for the spread of HIV and associated AIDS, and for development of anti-HIV-1 vaccines.
Collapse
Affiliation(s)
- M L Bosch
- Department of Pathobiology, University of Washington, Seattle 98195, USA
| | | | | | | | | |
Collapse
|
46
|
Trichel AM, Roberts ED, Wilson LA, Martin LN, Ruprecht RM, Murphey-Corb M. SIV/DeltaB670 transmission across oral, colonic, and vaginal mucosae in the macaque. J Med Primatol 1997; 26:3-10. [PMID: 9271183 DOI: 10.1111/j.1600-0684.1997.tb00313.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The primary mode of human immunodeficiency virus (HIV) transmission worldwide is by exposure to the virus at vaginal, rectal, and oral mucosal surfaces. To understand HIV/simian immunodeficiency virus (SIV) transmission events at mucosal portals of entry, we used the SIV-macaque model to determine if mucosal surfaces function as barriers and select for particular viral genotypes. Rhesus macaques were inoculated intravaginally, intracolonically, intrarectally, or orally with the complex primary viral isolate SIV/DeltaB670. Peripheral blood mononuclear cells, collected within the first two weeks postinoculation, were cloned and sequenced from all infected macaques. In the majority of the animals analyzed, multiple genotypes were identified, independent of the route of infection. These findings suggest that the mucosal barrier may play a minor role in the genotypic selection observed during sexual transmission of HIV and emphasize the need to evaluate the viral diversity present within the mucosal secretions of chronically infected individuals.
Collapse
Affiliation(s)
- A M Trichel
- Tulane Regional Primate Research Center, Covington, LA 70433, USA
| | | | | | | | | | | |
Collapse
|
47
|
Warren JT, Levinson MA. Preclinical AIDS vaccine development: formal survey of global HIV, SIV, and SHIV in vivo challenge studies in vaccinated nonhuman primates. J Med Primatol 1997; 26:63-81. [PMID: 9271191 DOI: 10.1111/j.1600-0684.1997.tb00321.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- J T Warren
- The EMMES Corporation, Potomac, Maryland 20854, USA
| | | |
Collapse
|