Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
J Med Primatol ; 46(4): 158-161, 2017 08.
Article in English | MEDLINE | ID: mdl-28748668

ABSTRACT

Simian betaretroviruses include the well-known exogenous simian retroviruses (SRV-1 through SRV-8), and some closely related simian endogenous retroviruses (SERV). Here, we characterized two new viral genomes, which appear to represent novel SERVs but have characteristics of both SRV and SERV highlighting the need to develop new assays providing molecular and serologic differentiation of SERV and SRV to avoid false positives.


Subject(s)
Endogenous Retroviruses/isolation & purification , Genome, Viral , Macaca nemestrina/virology , Retroviridae Infections/diagnosis , Retroviruses, Simian/isolation & purification , Tumor Virus Infections/diagnosis , Animals , Endogenous Retroviruses/genetics , Indonesia , Retroviruses, Simian/genetics
2.
J Med Primatol ; 46(4): 149-153, 2017 08.
Article in English | MEDLINE | ID: mdl-28748661

ABSTRACT

To better understand Simian betaretrovirus (SRV) seropositivity in virus-negative macaques, we transfused blood from SRV-infected or suspect donors into immunosuppressed naive recipients. Our results do not support typical SRV1-5 infection as the cause, but provide evidence for several possibilities including serological artifact, new/different SRV, or an endogenous virus.


Subject(s)
Betaretrovirus/physiology , Macaca , Monkey Diseases/diagnosis , Retroviridae Infections/diagnosis , Animals , Monkey Diseases/virology , Retroviridae Infections/virology
3.
Front Virol ; 32023.
Article in English | MEDLINE | ID: mdl-37383986

ABSTRACT

Zika virus (ZIKV) is a mosquito-borne flavivirus that causes an acute febrile illness. ZIKV can be transmitted between sexual partners and from mother to fetus. Infection is strongly associated with neurologic complications in adults, including Guillain-Barré syndrome and myelitis, and congenital ZIKV infection can result in fetal injury and congenital Zika syndrome (CZS). Development of an effective vaccine is imperative to protect against ZIKV vertical transmission and CZS. Recombinant Vesicular Stomatitis virus (rVSV) is a highly effective and safe vector for the delivery of foreign immunogens for vaccine purposes. Here, we evaluate an rVSV vaccine expressing the full length pre-membrane (prM) and ZIKV envelope (E) proteins (VSV-ZprME), shown to be immunogenic in murine models of ZIKV infection, for its capacity to induce immune responses in nonhuman primates. Moreover, we assess the efficacy of the rVSVΔM-ZprME vaccine in the protection of pigtail macaques against ZIKV infection. Administration of the rVSVΔM-ZprME vaccine was safe, but it did not induce robust anti-ZIKV T-cell responses, IgM or IgG antibodies, or neutralizing antibodies in most animals. Post ZIKV challenge, animals that received the rVSVΔM control vaccine lacking ZIKV antigen had higher levels of plasma viremia compared to animals that received the rVSVΔM-ZprME vaccine. Anti-ZIKV neutralizing Ab titers were detected in a single animal that received the rVSVΔM-ZprME vaccine that was associated with reduced plasma viremia. The overall suboptimal ZIKV-specific cellular and humoral responses post-immunization indicates the rVSVΔM-ZprME vaccine did not elicit an immune response in this pilot study. However, recall antibody response to the rVSVΔM-ZprME vaccine indicates it may be immunogenic and further developments to the vaccine construct could enhance its potential as a vaccine candidate in a nonhuman primate pre-clinical model.

4.
J Immunol ; 182(6): 3718-27, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19265150

ABSTRACT

Previously, chronic-phase protection against SHIV(89.6P) challenge was significantly greater in macaques primed with replicating adenovirus type 5 host range mutant (Ad5hr) recombinants encoding HIVtat and env and boosted with Tat and Env protein compared with macaques primed with multigenic adenovirus recombinants (HIVtat, HIVenv, SIVgag, SIVnef) and boosted with Tat, Env, and Nef proteins. The greater protection was correlated with Tat- and Env-binding Abs. Because the macaques lacked SHIV(89.6P)-neutralizing activity prechallenge, we investigated whether Ab-dependent cellular cytotoxicity (ADCC) and Ab-dependent cell-mediated viral inhibition (ADCVI) might exert a protective effect. We clearly show that Tat can serve as an ADCC target, although the Tat-specific activity elicited did not correlate with better protection. However, Env-specific ADCC activity was consistently higher in the Tat/Env group, with sustained cell killing postchallenge exhibited at higher levels (p < 0.00001) for a longer duration (p = 0.0002) compared with the multigenic group. ADCVI was similarly higher in the Tat/Env group and significantly correlated with reduced acute-phase viremia at wk 2 and 4 postchallenge (p = 0.046 and 0.011, respectively). Viral-specific IgG and IgA Abs in mucosal secretions were elicited but did not influence the outcome of the i.v. SHIV(89.6P) challenge. The higher ADCC and ADCVI activities seen in the Tat/Env group provide a plausible mechanism responsible for the greater chronic-phase protection. Because Tat is known to enhance cell-mediated immunity to coadministered Ags, further studies should explore its impact on Ab induction so that it may be optimally incorporated into HIV vaccine regimens.


Subject(s)
Antibodies, Viral/biosynthesis , SAIDS Vaccines/administration & dosage , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , env Gene Products, Human Immunodeficiency Virus/administration & dosage , env Gene Products, Human Immunodeficiency Virus/immunology , tat Gene Products, Human Immunodeficiency Virus/administration & dosage , tat Gene Products, Human Immunodeficiency Virus/immunology , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Antibodies, Viral/physiology , Antibody-Dependent Cell Cytotoxicity/genetics , Antibody-Dependent Cell Cytotoxicity/immunology , Antiviral Agents/administration & dosage , Antiviral Agents/immunology , Binding Sites, Antibody/genetics , Cell Line, Transformed , Cells, Cultured , Female , Humans , Macaca mulatta , Male , Neutralization Tests , SAIDS Vaccines/genetics , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccinia virus/genetics , Vaccinia virus/immunology , env Gene Products, Human Immunodeficiency Virus/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics
5.
PLoS One ; 16(4): e0240495, 2021.
Article in English | MEDLINE | ID: mdl-33914754

ABSTRACT

RATIONALE/STUDY DESIGN: A major challenge in the development of HIV vaccines is finding immunogens that elicit protection against a broad range of viral strains. Immunity to a narrow range of viral strains may protect infants of HIV-infected women or partners discordant for HIV. We hypothesized that immunization to the relevant viral variants could be achieved by exposure to infectious virus during prophylaxis with antiretroviral drugs. To explore this approach in an animal model, macaques were exposed to live virus (SIVmne or HIV-2287) during prophylaxis with parenteral tenofovir and humoral and cellular immune responses were quantified. Subsequently, experimental animals were challenged with homologous virus to evaluate protection from infection, and if infection occurred, the course of disease was compared to control animals. Experimental animals uninfected with SIVmne were challenged with heterologous HIV-2287 to assess resistance to retroviral infection. METHODOLOGY/PRINCIPAL FINDINGS: Juvenile female Macaca nemestrina (N = 8) were given ten weekly intravaginal exposures with either moderately (SIVmne) or highly (HIV-2287) pathogenic virus during tenofovir prophylaxis. Tenofovir protected all 8 experimental animals from infection, while all untreated control animals became infected. Specific non-neutralizing antibodies were elicited in blood and vaginal secretions of experimental animals, but no ELISPOT responses were detected. Six weeks following the cessation of tenofovir, intravaginal challenge with homologous virus infected 2/4 (50%) of the SIVmne-immunized animals and 4/4 (100%) of the HIV-2287-immunized animals. The two SIVmne-infected and 3 (75%) HIV-2287-infected had attenuated disease, suggesting partial protection. CONCLUSIONS/SIGNIFICANCE: Repeated exposure to SIVmne or HIV-2287, during antiretroviral prophylaxis that blocked infection, induced binding antibodies in the blood and mucosa, but not neutralizing antibodies or specific cellular immune responses. Studies to determine whether antibodies are similarly induced in breastfeeding infants and sexual partners discordant for HIV infection and receiving pre-exposure antiretroviral prophylaxis are warranted, including whether these antibodies appear to confer partial or complete protection from infection.


Subject(s)
Anti-Retroviral Agents/therapeutic use , HIV Infections/prevention & control , HIV-2/drug effects , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/drug effects , Tenofovir/therapeutic use , Animals , Disease Models, Animal , Female , HIV Infections/immunology , HIV-2/immunology , Humans , Immunization/methods , Macaca nemestrina , Pre-Exposure Prophylaxis/methods , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology
6.
Comp Med ; 70(1): 75-82, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31747991

ABSTRACT

Despite the lack of confirmed reports of an exogenous Simian betaretrovirus (SRV) isolated from baboons (Papio sp.), reports of simian endogenous gammaretrovirus (SERV) in baboons with complete genomes suggest that such viruses may be potentially infectious. In addition, serologic tests have repeatedly demonstrated antibody reactivity to SRV in baboons from multiple colonies. These findings complicate the management and use of such animals for research. To provide further insight into this situation, we performed in vitro and in vivo studies to determine if baboons are or can be infected with SRV. In our initial experiment, we were not able to isolate SRV from 6 seropositive or sero-indeterminate baboons by coculturing their peripheral blood mononuclear cells (PBMC) with macaque PBMC or permissive cell lines. In a subsequent experiment, we found that baboon PBMC infected in vitro with high dose SRV were permissive to virus replication. To test in vivo infectibil- ity, groups of naive baboons were infused intravenously with either (i) the same SRV tissue culture virus stocks used for the in vitro studies, (ii) SRV antibody positive and PCR positive macaque blood, (iii) SRV antibody positive or indeterminate, but PCR negative baboon blood, or (iv) SRV antibody and PCR negative baboon blood. Sustained SRV infection, as defined by reproducible PCR detection and/or antibody seroconversion, was confirmed in 2 of 3 baboons receiving tissue culture virus but not in any recipients of transfused blood from seropositive macaques or baboons. In conclusion, the data indicate that even though baboon cells can be infected experimentally with high doses of tissue culture grown SRV, baboons that are repeatedly SRV antibody positive and PCR negative are unlikely to be infected with exogenous SRV and thus are unlikely to transmit a virus that would threaten the SPF status of captive baboon colonies.


Subject(s)
Monkey Diseases/transmission , Papio , Retroviridae Infections/transmission , Animals , Betaretrovirus/isolation & purification , Female , Leukocytes, Mononuclear/virology , Male , Monkey Diseases/blood , Monkey Diseases/virology , Retroviridae Infections/blood , Retroviridae Infections/virology , Virus Replication
7.
Emerg Infect Dis ; 14(8): 1200-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18680642

ABSTRACT

In Asia, contact between persons and nonhuman primates is widespread in multiple occupational and nonoccupational contexts. Simian foamy viruses (SFVs) are retroviruses that are prevalent in all species of nonhuman primates. To determine SFV prevalence in humans, we tested 305 persons who lived or worked around nonhuman primates in several South and Southeast Asian countries; 8 (2.6%) were confirmed SFV positive by Western blot and, for some, by PCR. The interspecies interactions that likely resulted in virus transmission were diverse; 5 macaque taxa were implicated as a potential source of infection. Phylogenetic analysis showed that SFV from 3 infected persons was similar to that from the nonhuman primate populations with which the infected persons reported contact. Thus, SFV infections are likely to be prevalent among persons who live or work near nonhuman primates in Asia.


Subject(s)
Retroviridae Infections/transmission , Simian foamy virus , Zoonoses/transmission , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Ape Diseases/transmission , Ape Diseases/virology , Asia/epidemiology , DNA, Viral/genetics , Female , Humans , Male , Middle Aged , Occupational Exposure , Phylogeny , Retroviridae Infections/epidemiology
8.
Nat Med ; 24(3): 368-374, 2018 03.
Article in English | MEDLINE | ID: mdl-29400709

ABSTRACT

Zika virus (ZIKV) is a flavivirus with teratogenic effects on fetal brain, but the spectrum of ZIKV-induced brain injury is unknown, particularly when ultrasound imaging is normal. In a pregnant pigtail macaque (Macaca nemestrina) model of ZIKV infection, we demonstrate that ZIKV-induced injury to fetal brain is substantial, even in the absence of microcephaly, and may be challenging to detect in a clinical setting. A common and subtle injury pattern was identified, including (i) periventricular T2-hyperintense foci and loss of fetal noncortical brain volume, (ii) injury to the ependymal epithelium with underlying gliosis and (iii) loss of late fetal neuronal progenitor cells in the subventricular zone (temporal cortex) and subgranular zone (dentate gyrus, hippocampus) with dysmorphic granule neuron patterning. Attenuation of fetal neurogenic output demonstrates potentially considerable teratogenic effects of congenital ZIKV infection even without microcephaly. Our findings suggest that all children exposed to ZIKV in utero should receive long-term monitoring for neurocognitive deficits, regardless of head size at birth.


Subject(s)
Fetus/virology , Pregnancy Complications, Infectious/physiopathology , Zika Virus Infection/virology , Zika Virus/pathogenicity , Animals , Disease Models, Animal , Female , Fetus/physiopathology , Humans , Macaca nemestrina/virology , Microcephaly/diagnostic imaging , Microcephaly/physiopathology , Microcephaly/virology , Neurogenesis/genetics , Pregnancy , Pregnancy Complications, Infectious/diagnostic imaging , Pregnancy Complications, Infectious/virology , Zika Virus/genetics , Zika Virus Infection/genetics , Zika Virus Infection/physiopathology
9.
AIDS Res Hum Retroviruses ; 22(10): 1022-30, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17067273

ABSTRACT

In the present study, macaques were coimmunized with VEErep/SINenv chimeric alphavirus replicon particles expressing SIVp55Gag and HIVDeltaV2gp140Env or only with replicon particles expressing HIVDeltaV2gp140Env. All animals were subsequently immunized with recombinant trimeric HIVDeltaV2gp140Env protein. During alphavirus immunization, anti-SIVGag and anti-HIVEnv-specific interferon (IFN)-gamma responses, as well as high titers of anti-HIVEnv binding (gp120 but not gp41 specific) and anti-HIV neutralizing antibodies, were generated. The subsequent immunization with recombinant HIVDeltaV2gp140 enhanced the neutralizing antibody titers and Env-specific IFN-gamma responses. Following intravenous challenge with the R5- tropic SHIV(SF162P4) virus, significantly lower primary plasma viremia levels were recorded in the immunized animals, as compared to control animals immunized with replicon particles expressing influenza virus HA. Our results show that this method of immunization elicits both strong cellular immunity and neutralizing antibodies in primates and, thus, merits further investigation.


Subject(s)
AIDS Vaccines/immunology , Antibodies, Viral/biosynthesis , Gene Products, env/immunology , Gene Products, gag/immunology , HIV Antibodies/biosynthesis , Replicon , Animals , Encephalitis Virus, Venezuelan Equine/genetics , Genetic Vectors , Macaca mulatta , Recombinant Proteins/immunology , Sindbis Virus , Vaccines, Synthetic/immunology , env Gene Products, Human Immunodeficiency Virus
10.
Nat Med ; 22(11): 1256-1259, 2016 11.
Article in English | MEDLINE | ID: mdl-27618651

ABSTRACT

We describe the development of fetal brain lesions after Zika virus (ZIKV) inoculation in a pregnant pigtail macaque. Periventricular lesions developed within 10 d and evolved asymmetrically in the occipital-parietal lobes. Fetal autopsy revealed ZIKV in the brain and significant cerebral white matter hypoplasia, periventricular white matter gliosis, and axonal and ependymal injury. Our observation of ZIKV-associated fetal brain lesions in a nonhuman primate provides a model for therapeutic evaluation.


Subject(s)
Brain/diagnostic imaging , Fetus/diagnostic imaging , Pregnancy Complications, Infectious/diagnostic imaging , Zika Virus Infection/diagnostic imaging , Animals , Aspartic Acid/analogs & derivatives , Aspartic Acid/metabolism , Brain/metabolism , Brain/pathology , Brain/virology , Choline/metabolism , Creatine/metabolism , Echoencephalography , Female , Fetus/metabolism , Fetus/pathology , Fetus/virology , Glutamic Acid/metabolism , Glutamine/metabolism , Inositol/metabolism , Macaca nemestrina , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Pregnancy , Pregnancy Complications, Infectious/metabolism , Pregnancy Complications, Infectious/pathology , RNA, Viral/metabolism , Ultrasonography, Prenatal , Zika Virus/genetics , Zika Virus Infection/metabolism , Zika Virus Infection/pathology
11.
Diagn Microbiol Infect Dis ; 53(3): 185-93, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16243475

ABSTRACT

Our goal was to determine if a multiplex technique using a fluorescent bead-based flow cytometric assay could yield results comparable to traditional enzyme-linked immunosorbent assay (ELISA) in terms of sensitivity, specificity, cross-reactivity, and throughput. We applied both techniques to serologic screening of specific pathogen-free macaques, for type D simian retrovirus, simian T-lymphotropic virus, Cercopithicine herpesvirus 1, and simian immunodeficiency virus, and found a high correlation between the bead-based multiplex assay and ELISA. The multiplex assay demonstrated greater sensitivity with no loss in specificity when compared to the ELISA. A lower false-positive rate with the multiplex assay decreased the number of confirmatory Western blots required. Using the multiplex assay, we were able to screen samples for 4 viruses simultaneously in the time it took to perform a single-virus ELISA, resulting in a faster turnaround time and higher throughput. The multiplexed assay provided greater sensitivity, increased stability, and better performance than ELISA.


Subject(s)
Antibodies, Viral/blood , Flow Cytometry/methods , Virus Diseases/diagnosis , Virus Diseases/immunology , Viruses/immunology , Animals , Enzyme-Linked Immunosorbent Assay , Flow Cytometry/instrumentation , Herpesvirus 1, Cercopithecine/immunology , Immunoglobulin G/blood , Microspheres , Retroviruses, Simian/immunology , Sensitivity and Specificity , Simian Immunodeficiency Virus/immunology , Simian T-lymphotropic virus 1/immunology , Specific Pathogen-Free Organisms , Virus Diseases/virology
12.
AIDS Res Hum Retroviruses ; 20(4): 425-34, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15157361

ABSTRACT

We utilized SIV(mne) infection of Macaca fascicularis to assess the efficacy of DNA vaccination alone, and as a priming agent in combination with subunit protein boosts. All SIV(mne) structural and regulatory genes were expressed using the human cytomegalovirus Immediate Early-1 promoter in plasmids that directed the formation of virus-like particles in vitro. Macaques (n = 4) were immunized intradermally and intramuscularly four times over 36 weeks with 3 mg plasmid DNA. A second group (n = 4) received two DNA priming inoculations followed by two intramuscular boosts consisting of 250 microg recombinant Env gp160 and 250 microg recombinant Gag-Pol particles in MF-59 adjuvant. These regimens elicited modest cellular immunity prior to challenge. Humoral immune responses to Env gp160 were elicited and sustained by both vaccine protocols, and as expected antibody titers were higher in the protein subunit-boosted animals. Neutralizing antibodies prior to challenge were measurable in two of four subunit-boosted macaques. The two vaccine regimens elicited comparable helper T cell responses at the time of challenge. Vaccinees and mock-immunized controls (n = 4) were challenged intrarectally at week 38 with uncloned SIV(mne). Following challenge all macaques became infected, but both vaccine regimens resulted in reduced peak virus loads (p = 0.07) and significantly improved maintenance of peripheral CD4(+) T cell counts postchallenge (p = 0.007, DNA alone and p = 0.01, all vaccinees). There was no significant difference between the two vaccine groups in levels of plasma viremia or maintenance of CD4(+) T cell counts postchallenge.


Subject(s)
SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Viral/blood , Antigens, Viral/immunology , CD4 Lymphocyte Count , Fusion Proteins, gag-pol/genetics , Fusion Proteins, gag-pol/immunology , Gene Products, env/immunology , HIV Envelope Protein gp160/genetics , HIV Envelope Protein gp160/immunology , Immunity, Cellular , Macaca fascicularis , Neutralization Tests , Plasmids , Proviruses/genetics , Proviruses/isolation & purification , RNA, Viral/blood , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Acquired Immunodeficiency Syndrome/virology , Vaccines, DNA/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Load
13.
Comp Med ; 64(1): 63-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24512963

ABSTRACT

A 2.25-y-old male pigtailed macaque (Macaca nemestrina) was experimentally irradiated and received a bone marrow transplant. After transplantation and engraftment, the macaque had unexpected recurring pancytopenia and dependent edema of the prepuce, scrotum, and legs. The diagnostic work-up included a blood smear, which revealed a trypomastigote consistent with Trypanosoma cruzi, the causative agent of Chagas disease (CD). We initially hypothesized that the macaque had acquired the infection when it lived in Georgia. However, because the animal had received multiple blood transfusions, all blood donors were screened for CD. One male pigtailed macaque blood donor, which was previously housed in Louisiana, was positive for T. cruzi antibodies via serology. Due to the low prevalence of infection in Georgia, the blood transfusion was hypothesized to be the source of T. cruzi infection. The transfusion was confirmed as the mechanism of transmission when screening of archived serum revealed seroconversion after blood transfusion from the seropositive blood donor. The macaque made a full clinical recovery, and further follow-up including thoracic radiography, echocardiography, and gross necropsy did not show any abnormalities associated with CD. Other animals that received blood transfusions from the positive blood donor were tested, and one additional pigtailed macaque on the same research protocol was positive for T. cruzi. Although CD has been reported to occur in many nonhuman primate species, especially pigtailed macaques, the transmission of CD via blood transfusion in nonhuman primates has not been reported previously.


Subject(s)
Blood Transfusion/veterinary , Chagas Disease/veterinary , Immunocompromised Host , Macaca nemestrina/parasitology , Monkey Diseases/parasitology , Trypanosoma cruzi/isolation & purification , Animals , Antibodies, Protozoan/blood , Biomarkers/blood , Chagas Disease/blood , Chagas Disease/immunology , Chagas Disease/transmission , Dose Fractionation, Radiation , Genetic Therapy , Macaca nemestrina/blood , Macaca nemestrina/immunology , Male , Models, Animal , Monkey Diseases/blood , Monkey Diseases/immunology , Stem Cell Transplantation , Transfusion Reaction , Trypanosoma cruzi/immunology
14.
Nat Med ; 16(10): 1117-9, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20890292

ABSTRACT

Maternal HIV-1-specific antibodies are efficiently transferred to newborns, but their role in disease control is unknown. We administered neutralizing IgG, including the human neutralizing monoclonal IgG1b12, at levels insufficient to block infection, to six newborn macaques before oral challenge with simian-HIV strain SF162P3 (SHIV(SF162P3)). All of the macaques rapidly developed neutralizing antibodies and had significantly reduced plasma viremia for six months. These studies support the use of neutralizing antibodies in enhancing B cell responses and viral control in perinatal settings.


Subject(s)
Antibodies, Neutralizing/immunology , B-Lymphocytes/immunology , Immunization, Passive , Simian Acquired Immunodeficiency Syndrome/prevention & control , Viremia/prevention & control , Animals , CD4 Lymphocyte Count , Immunoglobulin G/immunology , Macaca , Simian Acquired Immunodeficiency Syndrome/immunology , Viremia/immunology
15.
J Virol ; 81(2): 822-34, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17079310

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.


Subject(s)
Infectious Disease Transmission, Vertical , Simian Acquired Immunodeficiency Syndrome/physiopathology , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/pathogenicity , Viremia/physiopathology , Animals , Animals, Newborn , Antibody Specificity , Disease Models, Animal , Female , Gene Products, env/immunology , Immunoglobulin G/blood , Immunoglobulin M/blood , Macaca nemestrina , Molecular Sequence Data , Neutralization Tests , Pregnancy , Pregnancy Complications, Infectious/virology , Sequence Analysis, DNA , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/isolation & purification , Viremia/immunology , Viremia/virology
16.
Virology ; 349(2): 276-89, 2006 Jun 05.
Article in English | MEDLINE | ID: mdl-16527321

ABSTRACT

Immunization by the SF162gp140 or the DeltaV2gp140 HIV-1 envelope proteins results in the generation of strong homologous neutralizing antibodies (NAbs) that offer similar degree of protection from disease-development to macaques challenged with homologous virus. These two immunogens elicit weak cross-reactive NAbs and their effectiveness against heterologous challenge is currently unknown. To examine this issue, we immunized macaques with SIVGag p55 and either the SF162gp140 or the DeltaV2gp140 and challenged them intravenously with SHIV-89.6P. All animals became infected but previous immunization with SF162gp140 accelerated the development of anti-SHIV89.6P neutralizing antibody responses following infection. DeltaV2gp140 is derived from SF162gp140 following the deletion of 30 amino acids and one N-linked glycosylation site from the V2 loop. Our results suggest that even small differences in HIV Envelope immunogen structure can affect the neutralizing antibody responses generated following infection.


Subject(s)
AIDS Vaccines/immunology , Gene Products, env/immunology , HIV Infections/prevention & control , HIV-1/immunology , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , AIDS Vaccines/administration & dosage , Animals , CD4 Lymphocyte Count , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Gene Products, env/genetics , HIV Antibodies/blood , HIV Infections/immunology , HIV Infections/virology , HIV-1/physiology , Interferon-gamma/biosynthesis , Leukocytes, Mononuclear/immunology , Macaca mulatta , Neutralization Tests , RNA, Viral/blood , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Load , env Gene Products, Human Immunodeficiency Virus
17.
J Med Primatol ; 33(5-6): 243-50, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15525325

ABSTRACT

We developed a SHIV/macaque model of transmission from infected dams to their infants. Ten pregnant dams were infected intravenously with 100 MID(50) of macaque-titered SHIV-SF162P3 during the second trimester. Nine infants were born; the seven surviving beyond day of birth suckled for 6 months. Four of nine infants were infected (transmission rate = 44.4%), with one infection in utero, and three intrapartum and/or immediately post-birth via suckling. Varying levels of binding and neutralizing antibodies were transplacentally transferred to infants. Passive antibodies were detected in plasma on the day of birth and persisted for 5 weeks. Infants infected at or after birth controlled acute and post-acute viremia. Exposure to maternal SHIV-SF162P3 during birth and suckling in the presence of autologous maternal neutralizing antibodies may have affected transmission or pathogenesis in the infants. This transmission model can allow investigation of key parameters involved in perinatal transmission of HIV.


Subject(s)
Infectious Disease Transmission, Vertical , Macaca nemestrina , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/physiology , Animals , Animals, Suckling , Antibodies, Viral/blood , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Lymphocyte Count , Polymerase Chain Reaction , Pregnancy , RNA, Viral/blood , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Viral Load
18.
Virology ; 308(1): 178-90, 2003 Mar 30.
Article in English | MEDLINE | ID: mdl-12706101

ABSTRACT

Although most HIV-1 infections worldwide result from heterosexual transmission, most vaccine candidates have focused on induction of systemic immunity and protection. We hypothesized that combining systemic priming with mucosal boosting would induce mucosal immunity that would protect from intravaginal challenge. Macaques were primed systemically with recombinant vaccinia viruses and boosted mucosally using inactivated SHIV(89.6) plus adjuvant. Other animals received protein boosts with adjuvant alone. Priming and boosting induced antiviral IgG and IgA antibodies. Such antibodies were induced to a lesser degree in animals receiving boosts alone. Anti-SHIV T cell responses were induced only in the prime-boost animals. Immunized animals and controls were challenged intravaginally with SHIV(89.6) and significant reductions in proviral and viral RNA loads were observed in the prime-boost animals. The boost-only animals did not have significant viral load reductions. These data suggest that cellular immunity was required for protection from intravaginal challenge. This immunization regimen provides a promising lead for vaccine development.


Subject(s)
AIDS Vaccines/administration & dosage , HIV Infections/immunology , Lentiviruses, Primate/immunology , Virus Replication/immunology , Administration, Cutaneous , Administration, Intranasal , Animals , Antibodies, Viral/blood , DNA, Viral/analysis , Disease Models, Animal , Female , HIV Infections/blood , HIV Infections/prevention & control , Immunization/methods , Immunoglobulin A/blood , Immunoglobulin G/blood , Macaca nemestrina , Proviruses/genetics , RNA, Viral/blood , T-Lymphocytes/immunology , Vaccines, Synthetic/administration & dosage , Viral Load
19.
J Med Primatol ; 32(4-5): 218-28, 2003 Aug.
Article in English | MEDLINE | ID: mdl-14498982

ABSTRACT

We assessed four prime-boost vaccine regimens with a Gene Gun component for SHIV89.6P in Macaca nemestrina. A dosing experiment using beta-galactosidase plasmid showed that 30 or 45 shots per dose elicited higher titer antibody than smaller doses. For SHIV89.6P, we administered a six-plasmid vaccine capable of producing non-infectious virions in vivo in combination with either vaccinia recombinants or inactivated virus. DNA prime/vaccinia boost, or the reverse, elicited strong immune responses. The SHIV89.6P challenge virus was grown in M. nemestrina peripheral blood mononuclear cells and titered in vivo intrarectally. As has been observed for SHIV89.6P in M. mulatta, the infected M. nemestrina experienced rapid and severe loss of circulating CD4+ T cells. Vaccinated macaques were challenged three weeks after the last boost. DNA prime/vaccina boost or vaccina prime/DNA boost protected 11/12 animals from acute CD4+ T cell depletion and disease, while other regimens were not effective.


Subject(s)
AIDS Vaccines/immunology , CD4-Positive T-Lymphocytes/immunology , HIV/immunology , Immunization, Secondary , SAIDS Vaccines/immunology , Simian Immunodeficiency Virus/immunology , Animals , Biolistics , DNA Primers , Dose-Response Relationship, Immunologic , Enzyme-Linked Immunosorbent Assay , Macaca nemestrina/immunology , Plasmids , Vaccines, DNA/immunology , Vaccinia virus
SELECTION OF CITATIONS
SEARCH DETAIL