Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Front Cell Infect Microbiol ; 14: 1408245, 2024.
Article in English | MEDLINE | ID: mdl-39006742

ABSTRACT

While simian immunodeficiency virus (SIV) infection is non-pathogenic in naturally infected African nonhuman primate hosts, experimental or accidental infection in rhesus macaques often leads to AIDS. Baboons, widely distributed throughout Africa, do not naturally harbor SIV, and experimental infection of baboons with SIVmac results in transient low-level viral replication. Elucidation of mechanisms of natural immunity in baboons could uncover new targets of antiviral intervention. We tested the hypothesis that an SIVmac adapted to replicate in baboon primary cells will gain the capacity to establish chronic infections in vivo. Here, we generated SIVmac variants in baboon cells through serial passage in PBMC from different donors (SIVbn-PBMC s1), in PBMC from the same donors (SIVbn-PBMC s2), or in isolated CD4 cells from the same donors used for series 2 (SIVbn-CD4). While SIVbn-PBMC s1 and SIVbn-CD4 demonstrated increased replication capacity, SIVbn-PBMC s2 did not. Pharmacological blockade of CCR5 revealed SIVbn-PBMC s1 could more efficiently use available CCR5 than SIVmac, a trait we hypothesize arose to circumvent receptor occupation by chemokines. Sequencing analysis showed that all three viruses accumulated different types of mutations, and that more non-synonymous mutations became fixed in SIVbn-PBMC s1 than SIVbn-PBMC s2 and SIVbn-CD4, supporting the notion of stronger fitness pressure in PBMC from different genetic backgrounds. Testing the individual contribution of several newly fixed SIV mutations suggested that is the additive effect of these mutations in SIVbn-PBMC s1 that contributed to its enhanced fitness, as recombinant single mutant viruses showed no difference in replication capacity over the parental SIVmac239 strain. The replicative capacity of SIVbn-PBMC passage 4 (P4) s1 was tested in vivo by infecting baboons intravenously with SIVbn-PBMC P4 s1 or SIVmac251. While animals infected with SIVmac251 showed the known pattern of transient low-level viremia, animals infected with SIVbn-PBMC P4 s1 had undetectable viremia or viral DNA in lymphoid tissue. These studies suggest that adaptation of SIV to grow in baboon primary cells results in mutations that confer increased replicative capacity in the artificial environment of cell culture but make the virus unable to avoid the restrictive factors generated by a complex multicellular organism.


Subject(s)
Papio , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Virus Replication , Animals , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/physiology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Acquired Immunodeficiency Syndrome/immunology , Leukocytes, Mononuclear/virology , Leukocytes, Mononuclear/immunology , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , CD4-Positive T-Lymphocytes/virology , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Serial Passage
2.
Front Cell Infect Microbiol ; 12: 880860, 2022.
Article in English | MEDLINE | ID: mdl-35493734

ABSTRACT

Nonhuman primates (NHP) are particularly important for modeling infections with viruses that do not naturally replicate in rodent cells. Zika virus (ZIKV) has been responsible for sporadic epidemics, but in 2015 a disseminated outbreak of ZIKV resulted in the World Health Organization declaring it a global health emergency. Since the advent of this last epidemic, several NHP species, including the baboon, have been utilized for modeling and understanding the complications of ZIKV infection in humans; several health issues related to the outcome of infection have not been resolved yet and require further investigation. This study was designed to validate, in baboons, the molecular signatures that have previously been identified in ZIKV-infected humans and macaque models. We performed a comprehensive molecular analysis of baboons during acute ZIKV infection, including flow cytometry, cytokine, immunological, and transcriptomic analyses. We show here that, similar to most human cases, ZIKV infection of male baboons tends to be subclinical, but is associated with a rapid and transient antiviral interferon-based response signature that induces a detectable humoral and cell-mediated immune response. This immunity against the virus protects animals from challenge with a divergent ZIKV strain, as evidenced by undetectable viremia but clear anamnestic responses. These results provide additional support for the use of baboons as an alternative animal model to macaques and validate omic techniques that could help identify the molecular basis of complications associated with ZIKV infections in humans.


Subject(s)
Zika Virus Infection , Zika Virus , Animals , Immunity, Cellular , Male , Papio , Viremia
3.
J Virol ; 95(23): e0088221, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34549979

ABSTRACT

Human and simian immunodeficiency virus (HIV and SIV) infections establish lifelong reservoirs of cells harboring an integrated proviral genome. Genome editing CRISPR-associated Cas9 nucleases, combined with SIV-specific guiding RNA (gRNA) molecules, inactivate integrated provirus DNA in vitro and in animal models. We generated RNA-guided Cas9 nucleases (RGNu) and nickases (RGNi) targeting conserved SIV regions with no homology in the human or rhesus macaque genome. Assays in cells cotransfected with SIV provirus and plasmids coding for RGNus identified SIV long terminal repeat (LTR), trans-activation response (TAR) element, and ribosome slip site (RSS) regions as the most effective at virus suppression; RGNi targeting these regions inhibited virus production significantly. Multiplex plasmids that coexpressed these three RGNu (Nu3), or six (three pairs) RGNi (Ni6), were more efficient at virus suppression than any combination of individual RGNu and RGNi plasmids. Both Nu3 and Ni6 plasmids were tested in lymphoid cells chronically infected with SIVmac239, and whole-genome sequencing was used to determine on- and off-target mutations. Treatment with these all-in-one plasmids resulted in similar levels of mutations of viral sequences from the cellular genome; Nu3 induced indels at the 3 SIV-specific sites, whereas for Ni6 indels were present at the LTR and TAR sites. Levels of off-target effects detected by two different algorithms were indistinguishable from background mutations. In summary, we demonstrate that Cas9 nickase in association with gRNA pairs can specifically eliminate parts of the integrated provirus DNA; also, we show that careful design of an all-in-one plasmid coding for 3 gRNAs and Cas9 nuclease inhibits SIV production with undetectable off-target mutations, making these tools a desirable prospect for moving into animal studies. IMPORTANCE Our approach to HIV cure, utilizing the translatable SIV/rhesus macaque model system, aims at provirus inactivation and its removal with the least possible off-target side effects. We developed single molecules that delivered either three truncated SIV-specific gRNAs along with Cas9 nuclease or three pairs of SIV-specific gRNAs (six individual gRNAs) along with Cas9 nickase to enhance efficacy of on-target mutagenesis. Whole-genome sequencing demonstrated effective SIV sequence mutation and inactivation and the absence of demonstrable off-target mutations. These results open the possibility to employ Cas9 variants that introduce single-strand DNA breaks to eliminate integrated proviral DNA.


Subject(s)
DNA , Deoxyribonuclease I/genetics , Deoxyribonuclease I/metabolism , Proviruses/genetics , RNA, Guide, Kinetoplastida/genetics , Simian Immunodeficiency Virus/genetics , Animals , CRISPR-Cas Systems , Endonucleases/genetics , Gene Editing , HEK293 Cells , Humans , Macaca mulatta/metabolism , Mutagenesis , Plasmids
4.
Immun Ageing ; 18(1): 16, 2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33827617

ABSTRACT

Individuals over the age of 65 are highly susceptible to infectious diseases, which account for one-third of deaths in this age group. Vaccines are a primary tool to combat infection, yet they are less effective in the elderly population. While many groups have aimed to address this problem by studying vaccine-induced peripheral blood responses in the elderly, work from our lab and others demonstrate that immune responses to vaccination and infectious challenge may differ between tissue sites and the periphery. In this pilot study, we established an in vivo delayed-type hypersensitivity model of Mycobacterium bovis BCG vaccination and tuberculin skin test in two adult and two aged baboons. Vaccination generates BCG-specific immune cells that are recruited to the skin upon tuberculin challenge. We tested short term recall responses (8 weeks post-vaccination) and long term recall responses (25 weeks post-vaccination) by performing skin punch biopsies around the site of tuberculin injection. In short term recall responses, we found increased oxidation and decreased production of immune proteins in aged baboon skin at the site of TST challenge, in comparison to adult skin. Differences between adult and aged animals normalized in the long term response to tuberculin. In vitro, aged peripheral blood mononuclear cells had increased migration and functional responses to antigen-specific stimulation, suggesting that age-related changes in the tissue in vivo impairs aged immune recall responses to antigenic challenge. These findings highlight the impact of age-associated changes in the local tissue environment in memory recall responses, which may be more broadly applied to the study of other tissues. Moreover, these findings should be considered in future studies aimed at understanding and improving aging immune responses to vaccination and tissue challenge.

6.
Nat Microbiol ; 6(1): 73-86, 2021 01.
Article in English | MEDLINE | ID: mdl-33340034

ABSTRACT

Non-human primate models will expedite therapeutics and vaccines for coronavirus disease 2019 (COVID-19) to clinical trials. Here, we compare acute severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in young and old rhesus macaques, baboons and old marmosets. Macaques had clinical signs of viral infection, mild to moderate pneumonitis and extra-pulmonary pathologies, and both age groups recovered in two weeks. Baboons had prolonged viral RNA shedding and substantially more lung inflammation compared with macaques. Inflammation in bronchoalveolar lavage was increased in old versus young baboons. Using techniques including computed tomography imaging, immunophenotyping, and alveolar/peripheral cytokine response and immunohistochemical analyses, we delineated cellular immune responses to SARS-CoV-2 infection in macaque and baboon lungs, including innate and adaptive immune cells and a prominent type-I interferon response. Macaques developed T-cell memory phenotypes/responses and bystander cytokine production. Old macaques had lower titres of SARS-CoV-2-specific IgG antibody levels compared with young macaques. Acute respiratory distress in macaques and baboons recapitulates the progression of COVID-19 in humans, making them suitable as models to test vaccines and therapies.


Subject(s)
COVID-19/veterinary , Callithrix/immunology , Lung/immunology , Macaca mulatta/immunology , Monkey Diseases/virology , Papio/immunology , SARS-CoV-2/immunology , Adaptive Immunity , Animals , Antibodies, Viral/immunology , Bronchoalveolar Lavage , Bronchoalveolar Lavage Fluid , COVID-19/diagnostic imaging , COVID-19/immunology , COVID-19/pathology , Female , Humans , Immunity, Cellular/immunology , Immunoglobulin G/immunology , Inflammation/pathology , Lung/virology , Male , Monkey Diseases/immunology , Myeloid Cells/immunology , Viral Load , Virus Shedding
7.
J Med Primatol ; 49(5): 269-279, 2020 10.
Article in English | MEDLINE | ID: mdl-32905624

ABSTRACT

BACKGROUND: One approach for a functional HIV cure is to prevent transcription from integrated proviral DNA. A critical step in HIV transcription is the Tat protein interaction with the TAR element viral RNA. We tested the strategy of blocking this Tat-TAR interaction in the SIVmac model. METHODS: We designed five CRISPR short guiding RNAs (sgRNAs) targeting the SIVmac TAR element, along with inactive versions of Cas9 (dCas9). These sgRNA constructs were delivered as ribonucleoproteins or plasmid DNA, along with SIV DNA. The constructs were also tested in integrated viral DNA in a cell line chronically infected by SIV. RESULTS: The sgRNAs targeting the coding strand of the TAR element inhibited SIV RNA transcription in association with dCas9-KRAB, but not with dCas9. CONCLUSIONS: Induction of epigenetic modifications may be more effective in inactivating provirus than transcriptional interference and thus may be a better strategy to achieve a functional cure in vivo.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , DNA, Viral/genetics , Gene Silencing , HIV Long Terminal Repeat/genetics , Proviruses/genetics , Simian Immunodeficiency Virus/genetics , HEK293 Cells , Humans
8.
PLoS Negl Trop Dis ; 14(5): e0008285, 2020 05.
Article in English | MEDLINE | ID: mdl-32463814

ABSTRACT

Little is known about the contribution of virus-specific and cross-reacting antibodies (Abs) or the cellular immune response generated by a primary dengue (DENV) infection on the course of a secondary zika (ZIKV) infection in vivo. Here we show that the length of time between DENV/ZIKV infections has a qualitative impact on controlling early ZIKV replication. Depletion of DENV2-specific Abs in sera confirmed that those type-specific Abs do not contribute to ZIKV control. We show that the magnitude and durability of the neutralizing antibodies (nAbs) induced by a secondary ZIKV infection is modest compared to the response induced after a secondary heterologous DENV infection. Our in vivo results are showing a complex interplay between the cellular and innate immune responses characterized by a high frequency of plasmacytoid dendritic cells (pDC) correlating with an increase in the frequency of DENV antigen specific T cells and a significant control of ZIKV replication which is time dependent. Taken together, our results suggest that early after ZIKV infection other mechanisms such as the innate and cellular immune responses may play a predominant role in controlling ZIKV replication. Regardless of the time elapsed between infections there was no evidence of in vivo antibody-dependent enhancement (ADE) of ZIKV by DENV immunity. These findings have pivotal implications while interpreting ZIKV pathogenesis in flavivirus-experimented populations, diagnostic results interpretation and vaccine designs and schedules among others.


Subject(s)
Dengue/immunology , Immunity, Cellular , Immunity, Humoral , Immunity, Innate , Zika Virus Infection/immunology , Zika Virus Infection/prevention & control , Zika Virus/immunology , Animals , Dendritic Cells/immunology , Immunologic Factors , Macaca mulatta , Male , T-Lymphocytes/immunology , Time Factors
9.
Nat Commun ; 10(1): 4316, 2019 09 20.
Article in English | MEDLINE | ID: mdl-31541110

ABSTRACT

Zika virus (ZIKV) and dengue virus (DENV) are co-endemic in many parts of the world, but the impact of ZIKV infection on subsequent DENV infection is not well understood. Here we show in rhesus macaques that the time elapsed after ZIKV infection affects the immune response to DENV infection. We show that previous ZIKV exposure increases the magnitude of the antibody and T cell responses against DENV. The time interval between ZIKV and subsequent DENV infection further affects the immune response. A mid-convalescent period of 10 months after ZIKV infection results in higher and more durable antibody and T cell responses to DENV infection than a short period of 2 months. In contrast, previous ZIKV infection does not affect DENV viremia or pro-inflammatory status. Collectively, we find no evidence of a detrimental effect of ZIKV immunity in a subsequent DENV infection. This supports the implementation of ZIKV vaccines that could also boost immunity against future DENV epidemics.


Subject(s)
Dengue/immunology , Host-Pathogen Interactions/immunology , T-Lymphocytes/immunology , Zika Virus Infection/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cell Line , Cross Reactions/immunology , Cytokines/metabolism , Dengue Virus/immunology , Humans , Immunity , Immunity, Cellular , Macaca mulatta/immunology , Male , Time Factors , Viremia , Zika Virus/immunology
10.
Am J Primatol ; 81(2): e22952, 2019 02.
Article in English | MEDLINE | ID: mdl-30664265

ABSTRACT

The development of the marmoset as a translational model for healthspan and lifespan studies relies on the characterization of health parameters in young and geriatric marmosets. This cross-sectional study examined health phenotypes in marmosets for five domains of interest for human health and aging: mobility, cognition, metabolism, homeostasis, and immune function. Geriatric marmosets were found to have significant executive function impairment when compared to young animals. While geriatric animals did not show gross abnormalities in mobility and measures of locomotion, their types of movement were altered from young animals. Geriatric marmosets had alterations in cardiac function, with significantly increased mean arterial pressures; metabolism, with significantly lower VO2 ; and suppressed immune function. Further, this study sought to characterize and describe histopathology for both young and geriatric healthy marmosets. Overall this study provides a characterization of health parameters for young and geriatric marmosets which will greatly enhance future aging and interventional testing in marmosets.


Subject(s)
Aging , Callithrix/physiology , Health Status , Animals , Callithrix/anatomy & histology , Callithrix/immunology , Callithrix/metabolism , Cognition , Cross-Sectional Studies , Female , Homeostasis , Male , Mobility Limitation , Models, Animal , Phenotype
12.
Cytokine ; 111: 408-419, 2018 11.
Article in English | MEDLINE | ID: mdl-29807688

ABSTRACT

Simian immunodeficiency virus (SIV) infection in rhesus macaques is often characterized by high viremia and CD4 T cell depletion. By contrast, SIV infection in African nonhuman primate natural hosts is typically nonpathogenic despite active viral replication. Baboons are abundant in Africa and have a geographical distribution that overlaps with natural hosts, but they do not harbor SIVs. Previous work has demonstrated baboons are resistant to chronic SIV infection and/or disease in vivo but the underlying mechanisms remain unknown. Using in vitro SIVmac infections, we sought to identify SIV restriction factors in baboons by comparing observations to the pathogenic rhesus macaque model. SIVmac replicated in baboon PBMC but had delayed kinetics compared to rhesus PBMC. However, SIVmac replication in baboon and rhesus isolated CD4 cells were similar to the kinetics seen for rhesus PBMC, demonstrating intracellular restriction factors do not play a strong role in baboon inhibition of SIVmac replication. Here, we show CD8 T cells contribute to the innate SIV-suppressive activity seen in naïve baboon PBMC. As one mechanism of restriction, we identified higher production of MIP-1α, MIP-1ß, and RANTES by baboon PBMC. Contact between CD4 and CD8 T cells resulted in maximum production of these chemokines and suppression of viral replication, whereas neutralization of CCR5-binding chemokines in baboon PBMC increased viral loads. Our studies indicate baboon natural restriction of SIVmac replication is largely dependent on CD4-extrinsinc mechanisms mediated, in part, by CD8 T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chemokine CCL3/immunology , Chemokine CCL4/immunology , Chemokine CCL5/immunology , Papio/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Animals , Coculture Techniques/methods , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Macaca mulatta/immunology , Macaca mulatta/virology , Papio/virology , Receptors, CCR5/immunology , Simian Immunodeficiency Virus/immunology , Viral Load/immunology , Virus Replication/immunology
13.
Sci Rep ; 8(1): 6851, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29717225

ABSTRACT

During its most recent outbreak across the Americas, Zika virus (ZIKV) was surprisingly shown to cause fetal loss and congenital malformations in acutely and chronically infected pregnant women. However, understanding the underlying pathogenesis of ZIKV congenital disease has been hampered by a lack of relevant in vivo experimental models. Here we present a candidate New World monkey model of ZIKV infection in pregnant marmosets that faithfully recapitulates human disease. ZIKV inoculation at the human-equivalent of early gestation caused an asymptomatic seroconversion, induction of type I/II interferon-associated genes and proinflammatory cytokines, and persistent viremia and viruria. Spontaneous pregnancy loss was observed 16-18 days post-infection, with extensive active placental viral replication and fetal neurocellular disorganization similar to that seen in humans. These findings underscore the key role of the placenta as a conduit for fetal infection, and demonstrate the utility of marmosets as a highly relevant model for studying congenital ZIKV disease and pregnancy loss.


Subject(s)
Abortion, Spontaneous/virology , Embryo Loss/virology , Fetus/abnormalities , Nervous System Malformations/virology , Placenta/virology , Pregnancy Complications, Infectious/virology , Zika Virus Infection/complications , Zika Virus , Animals , Callithrix , Cytokines/immunology , Disease Models, Animal , Female , Gestational Age , Humans , Interferon Type I/immunology , Interferon-gamma/immunology , Pregnancy , Pregnancy Complications, Infectious/immunology , Viremia , Virus Replication
14.
Sci Rep ; 7(1): 17126, 2017 12 07.
Article in English | MEDLINE | ID: mdl-29215081

ABSTRACT

A monkey model of Zika virus (ZIKV) infection is urgently needed to better understand transmission and pathogenesis, given its proven association with fetal brain defects in pregnant women and acute neurological illness. Here we experimentally infected 4 male marmosets with ZIKV (prototype 1947 African strain) and monitored them clinically with sampling of various body fluids and tissues for nearly 3 months. We show that the course of acute infection with ZIKV in these New World monkeys resembles the human illness in many respects, including (1) lack of apparent clinical symptoms in most cases, (2) persistence of the virus in body fluids such as semen and saliva for longer periods of time than in serum, and (3) generation of neutralizing antibodies as well as an antiviral immunological host response. Importantly, ZIKV-infected saliva samples (in addition to serum) were found to be infectious, suggesting potential capacity for viral transmission by the oral route. Re-challenge of a previously infected marmoset with a contemporary outbreak strain SPH2015 from Brazil resulted in continued protection against infection, no viral shedding, and boosting of the immune response. Given the key similarities to human infection, a marmoset model of ZIKV infection may be useful for testing of new drugs and vaccines.


Subject(s)
Zika Virus Infection/pathology , Animals , Callithrix , Chlorocebus aethiops , Disease Models, Animal , Male , Saliva/virology , Vero Cells , Zika Virus/pathogenicity , Zika Virus Infection/transmission , Zika Virus Infection/virology
15.
Nat Commun ; 8: 15674, 2017 06 23.
Article in English | MEDLINE | ID: mdl-28643775

ABSTRACT

Zika virus (ZIKV) is a re-emerging virus that has recently spread into dengue virus (DENV) endemic regions and cross-reactive antibodies (Abs) could potentially affect ZIKV pathogenesis. Using DENV-immune serum, it has been shown in vitro that antibody-dependent enhancement (ADE) of ZIKV infection can occur. Here we study the effects of pre-existing DENV immunity on ZIKV infection in vivo. We infect two cohorts of rhesus macaques with ZIKV; one cohort has been exposed to DENV 2.8 years earlier and a second control cohort is naïve to flaviviral infection. Our results, while confirming ADE in vitro, suggest that pre-existing DENV immunity does not result in more severe ZIKV disease. Rather our results show a reduction in the number of days of ZIKV viremia compared to naïve macaques and that the previous exposure to DENV may result in modulation of the immune response without resulting in enhancement of ZIKV pathogenesis.


Subject(s)
Antibody-Dependent Enhancement , Dengue/immunology , Zika Virus Infection/immunology , Zika Virus/pathogenicity , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cross Reactions/immunology , Cytokines/immunology , Dengue Virus , Humans , Immune Sera , K562 Cells , Macaca mulatta , Male , Models, Animal , Viral Envelope Proteins/immunology
16.
AIDS Res Hum Retroviruses ; 32(10-11): 1125-1134, 2016.
Article in English | MEDLINE | ID: mdl-26986800

ABSTRACT

Vaccines based on live attenuated viruses are highly effective immunogens in the simian immunodeficiency virus (SIV)/rhesus macaque animal model and offer the possibility of studying correlates of protection against infection with virulent virus. We utilized a tether system for studying, in naive macaques and animals vaccinated with a live-attenuated vaccine, the acute events after challenge with pathogenic SIV. This approach allowed for the frequent sampling of small blood volumes without sedation or restraining of the animals, thus reducing the confounding effect of sampling stress. Before challenge, vaccinated animals presented significantly higher levels of proliferating and activated B cells than naive macaques, which were manifested by high expression of CD8 on B cells. After SIV challenge, the only changes observed in protected vaccinated macaques were significant increases in expression of the NK marker NKG2C on CD4 and CD8 T cells. We also identified that infection of naive macaques with SIV resulted in a transient peak of expression of CD20 on CD8 T cells and a constant rise in the number of B cells expressing CD8. Finally, analysis of a larger cohort of vaccinated animals identified that, even when circulating levels of vaccine virus are below the limit of detection, live attenuated vaccines induce systemic increases of IP-10 and perforin. These studies indicate that components of both the innate and adaptive immune systems of animals inoculated with a live-attenuated SIV vaccine respond to and control infection with virulent virus. Persistence of the vaccine virus in tissues may explain the elevated cytokine and B-cell activation levels. In addition, our report underpins the utility of the tether system for the intensive study of acute immune responses to viral infections.


Subject(s)
B-Lymphocytes/immunology , CD8 Antigens/analysis , Gene Expression , NK Cell Lectin-Like Receptor Subfamily C/biosynthesis , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Animals , B-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/immunology , Macaca mulatta , SAIDS Vaccines/administration & dosage , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
17.
PLoS Pathog ; 11(9): e1005146, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26360709

ABSTRACT

The virus-host relationship in simian immunodeficiency virus (SIV) infected chimpanzees is thought to be different from that found in other SIV infected African primates. However, studies of captive SIVcpz infected chimpanzees are limited. Previously, the natural SIVcpz infection of one chimpanzee, and the experimental infection of six chimpanzees was reported, with limited follow-up. Here, we present a long-term study of these seven animals, with a retrospective re-examination of the early stages of infection. The only clinical signs consistent with AIDS or AIDS associated disease was thrombocytopenia in two cases, associated with the development of anti-platelet antibodies. However, compared to uninfected and HIV-1 infected animals, SIVcpz infected animals had significantly lower levels of peripheral blood CD4+ T-cells. Despite this, levels of T-cell activation in chronic infection were not significantly elevated. In addition, while plasma levels of ß2 microglobulin, neopterin and soluble TNF-related apoptosis inducing ligand (sTRAIL) were elevated in acute infection, these markers returned to near-normal levels in chronic infection, reminiscent of immune activation patterns in 'natural host' species. Furthermore, plasma soluble CD14 was not elevated in chronic infection. However, examination of the secondary lymphoid environment revealed persistent changes to the lymphoid structure, including follicular hyperplasia in SIVcpz infected animals. In addition, both SIV and HIV-1 infected chimpanzees showed increased levels of deposition of collagen and increased levels of Mx1 expression in the T-cell zones of the lymph node. The outcome of SIVcpz infection of captive chimpanzees therefore shares features of both non-pathogenic and pathogenic lentivirus infections.


Subject(s)
Ape Diseases/virology , HIV-1/physiology , Lentivirus Infections/veterinary , Lentiviruses, Primate/physiology , Pan troglodytes , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/physiology , Animals , Ape Diseases/immunology , Ape Diseases/pathology , Ape Diseases/physiopathology , Autoimmune Diseases/etiology , Autoimmune Diseases/veterinary , Biomarkers/blood , CD4 Lymphocyte Count , Female , HIV-1/immunology , HIV-1/isolation & purification , Hyperplasia , Lentivirus Infections/immunology , Lentivirus Infections/physiopathology , Lentivirus Infections/virology , Lentiviruses, Primate/immunology , Lentiviruses, Primate/isolation & purification , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymph Nodes/virology , Male , Myxovirus Resistance Proteins/metabolism , Neopterin/blood , Peptide Fragments/blood , Peptide Fragments/chemistry , Receptors, TNF-Related Apoptosis-Inducing Ligand/blood , Receptors, TNF-Related Apoptosis-Inducing Ligand/chemistry , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/physiopathology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/isolation & purification , Thrombocytopenia/etiology , Thrombocytopenia/veterinary , Viral Load , beta 2-Microglobulin/blood
18.
Am J Cardiovasc Dis ; 4(3): 123-32, 2014.
Article in English | MEDLINE | ID: mdl-25360390

ABSTRACT

Studies have shown that high-fat diets cause blood vessel damage, however, assessing pathological effects accurately and efficiently is difficult. In this study, we measured particle levels of static endothelium (CD31+ and CD105+) and activated endothelium (CD62E+, CD54+ and CD106+) in plasma. We determined individual responses to two dietary regimens in two groups of baboons. One group (n = 10), was fed a diet high in simple carbohydrates and saturated fats (the HSF diet) and the other (n = 8) received a diet high in simple carbohydrates and unsaturated fats (the HUF diet). Plasma samples were collected at 0, 3, and 7 weeks. The percentages of CD31+ and CD62E+ particles were elevated at 3 weeks in animals fed either diet, but these elevations were statistically significant only in animals fed the HUF diet. Surprisingly, both percentages and counts of CD31+ particles were significantly lower at week 7 compared to week 0 and 3 in the HSF group. The median absolute counts of CD105+ particles were progressively elevated over time in the HSF group with a significant increase from week 0 to 7; the pattern was somewhat different for the HUF group with significant increase from week 3 to 7. The counts of CD54+ particles exhibited wide variation in both groups during the dietary challenge, while the median counts of CD106+ particles were significantly lower at week 3 than at week 0 and week 7. Endothelial particles exhibited time-dependent changes, suggesting they were behaving as quantifiable surrogates for the early detection of vascular damage caused by dietary factors.

19.
J Med Primatol ; 43(4): 258-71, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24660852

ABSTRACT

BACKGROUND: γδT cells are effector cells that eliminate cancer and virus-infected cells. Chimpanzees are an endangered species that can naturally and experimentally be infected with SIV and HIV, respectively, but no information about the functionality of γδT cells during chronic lentiviral infection is currently available. METHODS: Healthy and HIV-infected chimpanzee γδT cells were characterized by flow cytometry. γδT subsets were studied after stimulation with T-cell activators, and the release of cytokines was analyzed by Luminex assay. RESULTS: γδT-cell subsets, Vδ1 and Vδ2Vγ9, showed different patterns in the expression of CD4, CD195, CD159a, and CD159c. Stimulation of γδT cells resulted in increased levels of CD4 and HLA-DR, which is more pronounced in Vδ1 T cells. Distinct cytokine patterns were found between healthy and HIV-infected chimpanzees. CONCLUSIONS: Analyses of major chimpanzee γδT subsets show similarities to human γδT cells and suggest different functionality and roles in their immune response against HIV infection.


Subject(s)
HIV Infections/immunology , Pan troglodytes/immunology , T-Lymphocytes/physiology , Animals , Cells, Cultured , Cytokines/metabolism , Immunophenotyping , Receptors, HIV/metabolism , Viral Load
20.
Cell Immunol ; 288(1-2): 24-30, 2014.
Article in English | MEDLINE | ID: mdl-24565973

ABSTRACT

Interleukin-15 (IL-15) contributes to natural killer cell development and immune regulation. However, IL-15 and interferon-gamma (IFN-γ) production are significantly reduced during progression to AIDS. We have previously reported that HIV infected chimpanzees (Pan troglodytes) express CD3-CD8+ IFN-γ+ natural killer (NK) cells with an inverse correlation to plasma HIV viral load. To expand on our initial study, we examined a larger population of HIV infected chimpanzees (n=10). Whole blood flow cytometry analyses showed that recombinant gp120 (rgp120) or recombinant IL-15 induces specific CD3-CD8+ IFN-γ+ NK cells at higher levels than CD3+CD8+ IFN-γ+ T cells in HIV infected specimens. Interestingly, peripheral blood T cells exhibited 0.5-3% IL-15 surface Tcell/NKT cell phenotypes, and rIL-15 stimulation significantly (P<0.007) up-regulated CD4+CD25+ T cell expression. Importantly, these data demonstrate novel T cell interleukin-15 expression and indicate a plausible regulatory mechanism for this cell-type during viral infection.


Subject(s)
Gene Expression/immunology , HIV Infections/veterinary , HIV-1/immunology , Interleukin-15/genetics , Killer Cells, Natural/virology , Pan troglodytes/virology , T-Lymphocytes/virology , Animals , CD3 Complex/genetics , CD3 Complex/immunology , CD4 Antigens/genetics , CD4 Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/pharmacology , HIV Infections/immunology , HIV Infections/virology , Humans , Immunophenotyping , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-15/immunology , Interleukin-15/pharmacology , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Killer Cells, Natural/immunology , Pan troglodytes/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , T-Lymphocytes/immunology , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL