Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Mucosal Immunol ; 17(1): 41-53, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37866719

ABSTRACT

Despite their importance for immunity against sexually transmitted infections, the composition of female reproductive tract (FRT) memory T-cell populations in response to changes within the local tissue environment under the regulation of the menstrual cycle remains poorly defined. Here, we show that in humans and pig-tailed macaques, the cycle determines distinct clusters of differentiation 4 T-cell surveillance behaviors by subsets corresponding to migratory memory (TMM) and resident memory T cells. TMM displays tissue-itinerant trafficking characteristics, restricted distribution within the FRT microenvironment, and distinct effector responses to infection. Gene pathway analysis by RNA sequencing identified TMM-specific enrichment of genes involved in hormonal regulation and inflammatory responses. FRT T-cell subset fluctuations were discovered that synchronized to cycle-driven CCR5 signaling. Notably, oral administration of a CCR5 antagonist drug blocked TMM trafficking. Taken together, this study provides novel insights into the dynamic nature of FRT memory CD4 T cells and identifies the menstrual cycle as a key regulator of immune surveillance at the site of STI pathogen exposure.


Subject(s)
CD4-Positive T-Lymphocytes , Genitalia, Female , Menstrual Cycle , Receptors, CCR5 , Signal Transduction , Female , Humans , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Genitalia, Female/immunology , Genitalia, Female/metabolism , Menstrual Cycle/immunology , Menstrual Cycle/physiology , Receptors, CCR5/genetics , Receptors, CCR5/metabolism , T-Lymphocyte Subsets/immunology , Macaca nemestrina/immunology , Immunologic Memory , Cellular Microenvironment/immunology , Cellular Microenvironment/physiology , CCR5 Receptor Antagonists/pharmacology
2.
Front Immunol ; 12: 749891, 2021.
Article in English | MEDLINE | ID: mdl-34867979

ABSTRACT

Broadly neutralising antibodies (bNAbs) may play an important role in future strategies for HIV control. The development of anti-drug antibody (ADA) responses can reduce the efficacy of passively transferred bNAbs but the impact of ADA is imperfectly understood. We previously showed that therapeutic administration of the anti-HIV bNAb PGT121 (either WT or LALA version) controlled viraemia in pigtailed macaques with ongoing SHIV infection. We now report on 23 macaques that had multiple treatments with PGT121. We found that an increasing number of intravenous doses of PGT121 or human IgG1 isotype control antibodies (2-4 doses) results in anti-PGT121 ADA induction and low plasma concentrations of PGT121. ADA was associated with poor or absent suppression of SHIV viremia. Notably, ADA within macaque plasma recognised another human bNAb 10E8 but did not bind to the variable domains of PGT121, suggesting that ADA were primarily directed against the constant regions of the human antibodies. These findings have implications for the development of preclinical studies examining multiple infusions of human bNAbs.


Subject(s)
Broadly Neutralizing Antibodies/administration & dosage , HIV Antibodies/administration & dosage , Immunoglobulin G/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , Viremia/prevention & control , Animals , Broadly Neutralizing Antibodies/blood , Broadly Neutralizing Antibodies/immunology , HIV Antibodies/blood , HIV Antibodies/immunology , Macaca nemestrina/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Viremia/immunology
3.
J Neurovirol ; 26(4): 511-519, 2020 08.
Article in English | MEDLINE | ID: mdl-32488843

ABSTRACT

HIV-associated neuroinflammation is primarily driven by CNS macrophages including microglia. Regulation of these immune responses, however, remains to be characterized in detail. Using the SIV/macaque model of HIV, we evaluated CNS expression of triggering receptor expressed on myeloid cells 2 (TREM2) which is constitutively expressed by microglia and contributes to cell survival, proliferation, and differentiation. Loss-of-function mutations in TREM2 are recognized risk factors for neurodegenerative diseases including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Nasu-Hakola disease (NHD); recent reports have also indicated a role for TREM2 in HIV-associated neuroinflammation. Using in situ hybridization (ISH) and qRT-PCR, TREM2 mRNA levels were found to be significantly elevated in frontal cortex of macaques with SIV encephalitis compared with uninfected controls (P = 0.02). TREM2 protein levels were also elevated as measured by ELISA of frontal cortex tissue homogenates in these animals. Previously, we characterized the expression of CSF1R (colony-stimulating factor 1 receptor) in this model; the TREM2 and CSF1R promoters both contain a PU.1 binding site. While TREM2 and CSF1R mRNA levels in the frontal cortex were highly correlated (Spearman R = 0.79, P < 0.001), protein levels were not well correlated. In SIV-infected macaques released from ART to study viral rebound, neither TREM2 nor CSF1R mRNA increased with rebound viremia. However, CSF1R protein levels remained significantly elevated unlike TREM2 (P = 0.02). This differential expression suggests that TREM2 and CSF1R play unique, distinct roles in the pathogenesis of HIV CNS disease.


Subject(s)
Encephalitis, Viral/genetics , Macaca nemestrina/immunology , Macrophages/immunology , Membrane Glycoproteins/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Immunodeficiency Virus/immunology , Animals , Antiretroviral Therapy, Highly Active/methods , Antiviral Agents/pharmacology , Drug Administration Schedule , Encephalitis, Viral/drug therapy , Encephalitis, Viral/immunology , Encephalitis, Viral/virology , Frontal Lobe/drug effects , Frontal Lobe/immunology , Frontal Lobe/virology , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Macaca nemestrina/genetics , Macaca nemestrina/virology , Macrophages/drug effects , Macrophages/virology , Male , Membrane Glycoproteins/immunology , Microglia/drug effects , Microglia/immunology , Microglia/virology , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/growth & development , Trans-Activators/genetics , Trans-Activators/immunology
4.
PLoS Pathog ; 15(7): e1007925, 2019 07.
Article in English | MEDLINE | ID: mdl-31260493

ABSTRACT

HIV-1 does not persistently infect macaques due in part to restriction by several macaque host factors. This has been partially circumvented by generating chimeric SIV/HIV-1 viruses (SHIVs) that encode SIV antagonist of known restriction factors. However, most SHIVs replicate poorly in macaques unless they are further adapted in culture and/or macaques (adapted SHIVs). Therefore, development of SHIVs encoding HIV-1 sequences derived directly from infected humans without adaptation (unadapted SHIVs) has been challenging. In contrast to the adapted SHIVs, the unadapted SHIVs have lower replication kinetics in macaque lymphocytes and are sensitive to type-1 interferon (IFN). The HIV-1 Envelope (Env) in the chimeric virus determines both the reduced replication and the IFN-sensitivity differences. There is limited information on macaque restriction factors that specifically limit replication of the more biologically relevant, unadapted SHIV variants. In order to identify the IFN-induced host factor(s) that could contribute to the inhibition of SHIVs in macaque lymphocytes, we measured IFN-induced gene expression in immortalized pig-tailed macaque (Ptm) lymphocytes using RNA-Seq. We found 147 genes that were significantly upregulated upon IFN treatment in Ptm lymphocytes and 31/147 were identified as genes that encode transmembrane helices and thus are likely present in membranes where interaction with viral Env is plausible. Within this group of upregulated genes with putative membrane-localized proteins, we identified several interferon-induced transmembrane protein (IFITM) genes, including several previously uncharacterized Ptm IFITM3-related genes. An evolutionary genomic analysis of these genes suggests the genes are IFITM3 duplications not found in humans that are both within the IFITM locus and also dispersed elsewhere in the Ptm genome. We observed that Ptm IFITMs are generally packaged at higher levels in unadapted SHIVs when compared to adapted SHIVs. CRISPR/Cas9-mediated knockout of Ptm IFITMs showed that depletion of IFITMs partially rescues the IFN sensitivity of unadapted SHIV. Moreover, we found that the depletion of IFITMs also increased replication of unadapted SHIV in the absence of IFN treatment, suggesting that Ptm IFITMs are likely important host factors that limit replication of unadapted SHIVs. In conclusion, this study shows that Ptm IFITMs selectively restrict replication of unadapted SHIVs. These findings suggest that restriction factors including IFITMs vary in their potency against different SHIV variants and may play a role in selecting for viruses that adapt to species-specific restriction factors.


Subject(s)
HIV-1/physiology , HIV-1/pathogenicity , Simian Immunodeficiency Virus/physiology , Simian Immunodeficiency Virus/pathogenicity , env Gene Products, Human Immunodeficiency Virus/physiology , Adaptation, Physiological , Animals , Genes, env , HIV-1/genetics , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Host Specificity , Humans , Interferon-alpha/metabolism , Macaca nemestrina/genetics , Macaca nemestrina/immunology , Macaca nemestrina/virology , Protein Processing, Post-Translational , Reassortant Viruses/genetics , Reassortant Viruses/pathogenicity , Reassortant Viruses/physiology , Simian Immunodeficiency Virus/genetics , Virus Replication
5.
Sci Rep ; 9(1): 5661, 2019 04 05.
Article in English | MEDLINE | ID: mdl-30952887

ABSTRACT

A HIV vaccine that provides mucosal immunity is urgently needed. We evaluated an intranasal recombinant Fowlpox virus (rFPV) priming vaccine followed by intramuscular Modified Vaccinia Ankara (rMVA) booster vaccine, both expressing SIV antigens. The vaccination generated mucosal and systemic SIV-specific CD4+ T cell mediated immunity and was associated with partial protection against high-dose intrarectal SIVmac251 challenge in outbred pigtail macaques. Three of 12 vaccinees were completely protected and these animals elicited sustained Gag-specific poly-functional, cytotoxic mucosal CD4+ T cells, complemented by systemic poly-functional CD4+ and CD8+ T cell immunity. Humoral immune responses, albeit absent in completely protected macaques, were associated with partial control of viremia in animals with relatively weaker mucosal/systemic T cell responses. Co-expression of an IL-4R antagonist by the rFPV vaccine further enhanced the breadth and cytotoxicity/poly-functionality of mucosal vaccine-specific CD4+ T cells. Moreover, a single FPV-gag/pol/env prime was able to induce rapid anamnestic gp140 antibody response upon SIV encounter. Collectively, our data indicated that nasal vaccination was effective at inducing robust cervico-vaginal and rectal immunity, although cytotoxic CD4+ T cell mediated mucosal and systemic immunity correlated strongly with 'complete protection', the different degrees of protection observed was multi-factorial.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Fowlpox virus/immunology , Macaca nemestrina/immunology , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Vaccines, Synthetic/immunology , AIDS Vaccines/immunology , Administration, Intranasal/methods , Animals , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Immunity, Mucosal/immunology , Immunization, Secondary/methods , Immunologic Memory/immunology , Injections, Intramuscular/methods , Macaca nemestrina/virology , Simian Acquired Immunodeficiency Syndrome/virology , Vaccination/methods , Vaccinia/immunology , Vaccinia virus/immunology
6.
Nat Commun ; 9(1): 3371, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30135445

ABSTRACT

The immunological and virological events that contribute to the establishment of Zika virus (ZIKV) infection in humans are unclear. Here, we show that robust cellular innate immune responses arising early in the blood and tissues in response to ZIKV infection are significantly stronger in males and correlate with increased viral persistence. In particular, early peripheral blood recruitment of plasmacytoid dendritic cells and higher production of monocyte chemoattractant protein (MCP-1) correspond with greater viral persistence and tissue dissemination. We also identify non-classical monocytes as primary in vivo targets of ZIKV infection in the blood and peripheral lymph node. These results demonstrate the potential differences in ZIKV pathogenesis between males and females and a key role for early cellular innate immune responses in the blood in viral dissemination and ZIKV pathogenesis.


Subject(s)
Immunity, Innate/physiology , Macaca nemestrina/immunology , Macaca nemestrina/virology , Zika Virus/immunology , Animals , Chemokine CCL2/metabolism , Macaca nemestrina/metabolism , Zika Virus Infection/immunology , Zika Virus Infection/metabolism
7.
Immunogenetics ; 70(6): 381-399, 2018 06.
Article in English | MEDLINE | ID: mdl-29134258

ABSTRACT

Pig-tailed macaques (Macaca nemestrina, Mane) are important models for human immunodeficiency virus (HIV) studies. Their infectability with minimally modified HIV makes them a uniquely valuable animal model to mimic human infection with HIV and progression to acquired immunodeficiency syndrome (AIDS). However, variation in the pig-tailed macaque major histocompatibility complex (MHC) and the impact of individual transcripts on the pathogenesis of HIV and other infectious diseases is understudied compared to that of rhesus and cynomolgus macaques. In this study, we used Pacific Biosciences single-molecule real-time circular consensus sequencing to describe full-length MHC class I (MHC-I) transcripts for 194 pig-tailed macaques from three breeding centers. We then used the full-length sequences to infer Mane-A and Mane-B haplotypes containing groups of MHC-I transcripts that co-segregate due to physical linkage. In total, we characterized full-length open reading frames (ORFs) for 313 Mane-A, Mane-B, and Mane-I sequences that defined 86 Mane-A and 106 Mane-B MHC-I haplotypes. Pacific Biosciences technology allows us to resolve these Mane-A and Mane-B haplotypes to the level of synonymous allelic variants. The newly defined haplotypes and transcript sequences containing full-length ORFs provide an important resource for infectious disease researchers as certain MHC haplotypes have been shown to provide exceptional control of simian immunodeficiency virus (SIV) replication and prevention of AIDS-like disease in nonhuman primates. The increased allelic resolution provided by Pacific Biosciences sequencing also benefits transplant research by allowing researchers to more specifically match haplotypes between donors and recipients to the level of nonsynonymous allelic variation, thus reducing the risk of graft-versus-host disease.


Subject(s)
Genes, MHC Class I/genetics , Macaca nemestrina/genetics , Macaca nemestrina/immunology , Alleles , Amino Acid Sequence , Animals , Cloning, Molecular/methods , HIV , Haplotypes/immunology , Histocompatibility Antigens/immunology , Histocompatibility Antigens Class I/genetics , Major Histocompatibility Complex/immunology , Simian Immunodeficiency Virus
8.
J Neurovirol ; 22(4): 498-507, 2016 08.
Article in English | MEDLINE | ID: mdl-26727909

ABSTRACT

In the fourth decade of the HIV epidemic, the relationship between host immunity and HIV central nervous system (CNS) disease remains incompletely understood. Using a simian immunodeficiency virus (SIV)/macaque model, we examined CNS outcomes in pigtailed macaques expressing the MHC class I allele Mane-A1*084:01 which confers resistance to SIV-induced CNS disease and induces the prototypic viral escape mutation Gag K165R. Insertion of gag K165R into the neurovirulent clone SIV/17E-Fr reduced viral replication in vitro compared to SIV/17E-Fr. We also found lower cerebrospinal fluid (CSF), but not plasma, viral loads in macaques inoculated with SIV/17E-Fr K165R versus those inoculated with wildtype. Although escape mutation K165R was genotypically stable in plasma, it rapidly reverted to wildtype Gag KP9 in both CSF and in microglia cultures. We induced robust Gag KP9-specific CTL tetramer responses by vaccinating Mane-A*084:01-positive pigtailed macaques with a Gag KP9 virus-like particle (VLP) vaccine. Upon SIV/17E-Fr challenge, vaccinated animals had lower SIV RNA in CSF compared to unvaccinated controls, but showed no difference in plasma viral loads. These data clearly demonstrate that viral fitness in the CNS is distinct from the periphery and underscores the necessity of understanding the consequences of viral escape in CNS disease with the advent of new therapeutic vaccination strategies.


Subject(s)
Gene Products, gag/immunology , Histocompatibility Antigens Class I/immunology , RNA, Viral/cerebrospinal fluid , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/prevention & control , Vaccines, Virus-Like Particle/administration & dosage , Alleles , Animals , Central Nervous System/immunology , Central Nervous System/virology , Gene Expression , Gene Products, gag/genetics , Histocompatibility Antigens Class I/genetics , Immune Evasion/genetics , Macaca nemestrina/immunology , Macaca nemestrina/virology , Male , Microglia/immunology , Microglia/virology , Mutation , Primary Cell Culture , RNA, Viral/blood , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/pathogenicity , Vaccination , Viral Load/immunology , Virus Replication/immunology
9.
Comp Med ; 65(5): 448-54, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26473350

ABSTRACT

Measles is a highly contagious viral disease in NHP. The infection can range from asymptomatic to rapidly fatal, resulting in significant morbidity and mortality in captive populations. In addition to appropriate quarantine practices, restricted access, the immunization of all personnel in contact with NHP, and the wearing of protective clothing including face masks, measles immunization further reduces the infection risk. Commercially available measles vaccines are effective for use in NHP, but interruptions in their availability have prevented the implementation of ongoing, consistent vaccination programs. This need for a readily available vaccine led us to perform a broad, multicenter safety and immunogenicity study of another candidate vaccine, MVac (Serum Institute of India), a monovalent measles vaccine derived from live Edmonston-Zagreb strain virus that had been attenuated after 22 passages on human diploid cells.


Subject(s)
Macaca mulatta/virology , Macaca nemestrina/virology , Measles Vaccine/administration & dosage , Measles virus/immunology , Measles/veterinary , Monkey Diseases/prevention & control , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Female , Immunization Schedule , Macaca mulatta/immunology , Macaca nemestrina/immunology , Male , Measles/blood , Measles/immunology , Measles/prevention & control , Measles/virology , Measles Vaccine/adverse effects , Measles Vaccine/immunology , Measles virus/pathogenicity , Monkey Diseases/blood , Monkey Diseases/immunology , Monkey Diseases/virology , Serologic Tests/veterinary , Time Factors , United States , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/immunology
10.
Braz. j. phys. ther. (Impr.) ; 19(3): 211-217, May-Jun/2015. tab, graf
Article in English | LILACS | ID: lil-751376

ABSTRACT

Background: Hippotherapy uses horseback riding movements for therapeutic purposes. In addition to the horse's movement, the choice of equipment and types of floor are also useful in the intervention. The quantification of dynamic parameters that define the interaction of the surface of contact between horse and rider provides insight into how the type of floor surface variations act upon the subject's postural control. Objective: To test whether different types of surfaces promote changes in the amplitude (ACOP) and velocity (VCOP) of the center of pressure (COP) displacement during the rider's contact with the saddle on the horse's back. Method: Twenty two healthy adult male subjects with experience in riding were evaluated. The penetration resistances of asphalt, sand and grass surfaces were measured. The COP data were collected on the three surfaces using a pressure measurement mat. Results: ACOP values were higher in sand, followed by grass and asphalt, with significant differences between sand and asphalt (anteroposterior, p=0.042; mediolateral, p=0.019). The ACOP and VCOP values were higher in the anteroposterior than in the mediolateral direction on all surfaces (ACOP, p=0.001; VCOP, p=0.006). The VCOP did not differ between the surfaces. Conclusion: Postural control, measured by the COP displacement, undergoes variations in its amplitude as a result of the type of floor surface. Therefore, these results reinforce the importance of the choice of floor surface when defining the strategy to be used during hippotherapy intervention. .


Subject(s)
Animals , Male , Blood Transfusion/veterinary , Chagas Disease/veterinary , Immunocompromised Host , Macaca nemestrina/parasitology , Monkey Diseases/parasitology , Trypanosoma cruzi/isolation & purification , Antibodies, Protozoan/blood , Biomarkers/blood , Blood Transfusion/adverse effects , Chagas Disease/blood , Chagas Disease/immunology , Chagas Disease/transmission , Dose Fractionation, Radiation , Genetic Therapy , Models, Animal , Macaca nemestrina/blood , Macaca nemestrina/immunology , Monkey Diseases/blood , Monkey Diseases/immunology , Stem Cell Transplantation , Trypanosoma cruzi/immunology
11.
J Neurovirol ; 21(2): 148-58, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25672885

ABSTRACT

Immune pressure exerted by MHC class I-restricted cytotoxic T cells drives the development of viral escape mutations, thereby regulating HIV disease progression. Nonetheless, the relationship between host immunity and HIV central nervous system (CNS) disease remains poorly understood. The simian immunodeficiency virus (SIV) macaque model recapitulates key features of HIV infection including development of AIDS and CNS disease. To investigate cell-mediated immunity regulating SIV CNS disease progression, we compared the incidence of SIV encephalitis and the influence of MHC class I allele expression on the development of CNS disease in rhesus macaques (Macaca mulatta) versus pigtailed macaques (Macaca nemestrina). After inoculation with the immunosuppressive swarm SIV/DeltaB670 and the neurovirulent molecular clone SIV/17E-Fr, pigtailed macaques progressed more rapidly to AIDS, had higher plasma and cerebrospinal fluid (CSF) viral loads, and were more likely to progress to SIV-associated encephalitis (SIVE) compared to rhesus macaques. In addition, MHC class I alleles were neuroprotective in both species (Mamu-A*001 in rhesus macaques and Mane-A1*084:01:01 in pigtailed macaques); animals expressing these alleles were less likely to develop SIV encephalitis and correspondingly had lower viral replication in the brain. Species-specific differences in susceptibility to SIV disease demonstrated that cell mediated immune responses are critical to SIV CNS disease progression.


Subject(s)
Disease Susceptibility/immunology , Macaca mulatta/virology , Macaca nemestrina/virology , Simian Acquired Immunodeficiency Syndrome/immunology , AIDS Dementia Complex/immunology , Animals , Disease Progression , Histocompatibility Antigens Class I/immunology , Macaca mulatta/immunology , Macaca nemestrina/immunology , Simian Immunodeficiency Virus
12.
J Leukoc Biol ; 97(3): 599-609, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25605872

ABSTRACT

The testis is a site of immune privilege in rodents, and there is evidence that T cell responses are also suppressed in the primate testis. Local immunosuppression is a potential mechanism for HIV persistence in tissue reservoirs that few studies have examined. The response of the pig-tailed macaque testis to SIVmac239 infection was characterized to test this possibility. Testes were surgically removed during early-chronic (10 wk) and late-chronic (24-30 wk) SIV infection in 4 animals and compared with those from 7 uninfected animals. SIV infection caused only minor disruption to the seminiferous epithelium without marked evidence of inflammation or consistent changes in total intratesticular leukocyte numbers. Infection also led to an increase in the relative proportion of testicular effector memory CD8(+) T cell numbers and a corresponding reduction in central memory CD4(+) T cells. A decrease in the relative proportion of resident-type CD163(+) macrophages and DCs was also observed. SIV-specific CD8(+) T cells were detectable in the testis, 10-11 wk after infection by staining with SIV Gag-specific or Tat-specific MHC-I tetramers. However, testicular CD8(+) T cells from the infected animals had suppressed cytokine responses to mitogen activation. These results support the possibility that local immunosuppression in the testis may be restricting the ability of T cells to respond to SIV or HIV infection. Local immunosuppression in the testis may be an underexplored mechanism allowing HIV persistence.


Subject(s)
Immunity , Macaca nemestrina/immunology , Macaca nemestrina/virology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Testis/immunology , Animals , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/immunology , Cytokines/metabolism , Dendritic Cells/pathology , Granulocytes/pathology , HEK293 Cells , Humans , Killer Cells, Natural/pathology , Leukocyte Common Antigens/metabolism , Macrophages/pathology , Male , Phenotype , Seminiferous Tubules/immunology , Seminiferous Tubules/pathology , Seminiferous Tubules/virology , Simian Acquired Immunodeficiency Syndrome/blood , Testis/pathology , Testis/virology
13.
Science ; 344(6190): 1401-5, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24948736

ABSTRACT

Primate lentiviruses exhibit narrow host tropism, reducing the occurrence of zoonoses but also impairing the development of optimal animal models of AIDS. To delineate the factors limiting cross-species HIV-1 transmission, we passaged a modified HIV-1 in pigtailed macaques that were transiently depleted of CD8(+) cells during acute infection. During adaptation over four passages in macaques, HIV-1 acquired the ability to antagonize the macaque restriction factor tetherin, replicated at progressively higher levels, and ultimately caused marked CD4(+) T cell depletion and AIDS-defining conditions. Transient treatment with an antibody to CD8 during acute HIV-1 infection caused rapid progression to AIDS, whereas untreated animals exhibited an elite controller phenotype. Thus, an adapted HIV-1 can cause AIDS in macaques, and stark differences in outcome can be determined by immunological perturbations during early infection.


Subject(s)
Acquired Immunodeficiency Syndrome/virology , Disease Models, Animal , HIV-1/physiology , Host-Pathogen Interactions/immunology , Macaca nemestrina/virology , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/transmission , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/immunology , CD8 Antigens/immunology , HIV-1/genetics , Human Immunodeficiency Virus Proteins/chemistry , Human Immunodeficiency Virus Proteins/genetics , Human Immunodeficiency Virus Proteins/metabolism , Lymphocyte Depletion , Macaca nemestrina/immunology , Molecular Sequence Data , Protein Structure, Tertiary , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication
14.
Immunogenetics ; 66(6): 361-77, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24811270

ABSTRACT

Macaques are the most widely used experimental nonhuman primate (NHP) species. Rhesus (Macaca mulatta, Macmul), cynomolgus (Macaca fascicularis, Macfas), and pigtail (Macaca nemestrina, Macnem) macaques continue to be popular models for vaccine and infectious diseases research, especially HIV infection and AIDS, and for the development of antibody-based therapeutic strategies. Increased understanding of the immune system of these species is necessary for their optimal use as models of human infections and intervention. In the past few years, the antibody/Fc receptor system has been characterized in a stepwise manner in these species. We have continued this characterization by identifying the four IG heavy gamma (IGHG) genes of Macfas and Macnem in this study. Our results show that these genes share a high degree of similarity with those from other NHP species, while presenting consistent differences when compared to human IGHG genes. Furthermore, comparison of Macfas IGHG genes with those described in other studies suggests the existence of polymorphism. Using sequence- and structure-based computational tools, we performed in silico analysis on multiple polymorphic Macfas IgG and their interactions with human IgG Fc receptors (FcγR), thus predicting that Macfas IGHG polymorphisms influence IgG protein stability and/or binding affinity towards FcγR. The presence of macaque IGHG polymorphisms and macaque/human amino acid changes at locations potentially involved in antibody functional properties indicate the need for cautious design and data interpretation of studies in these models, possibly requiring the characterization of antibody/Fc receptor interactions at the individual level.


Subject(s)
Immunoglobulin Gm Allotypes/genetics , Macaca fascicularis/genetics , Macaca nemestrina/genetics , Models, Immunological , Receptors, IgG/genetics , Amino Acid Sequence , Animals , Antibody Affinity , Binding Sites, Antibody , Computer Simulation , Humans , Macaca fascicularis/immunology , Macaca nemestrina/immunology , Molecular Sequence Data , Protein Binding , Receptors, IgG/immunology , Sequence Alignment , Sequence Homology, Amino Acid
15.
PLoS One ; 9(4): e94375, 2014.
Article in English | MEDLINE | ID: mdl-24732038

ABSTRACT

HIV immune pathogenesis is postulated to involve two major mechanisms: 1) chronic innate immune responses that drive T cell activation and apoptosis and 2) induction of immune regulators that suppress T cell function and proliferation. Both arms are elevated chronically in lymphoid tissues of non-natural hosts, which ultimately develop AIDS. However, these mechanisms are not elevated chronically in natural hosts of SIV infection that avert immune pathogenesis despite similarly high viral loads. In this study we investigated whether minocycline could modulate these pathogenic antiviral responses in non-natural hosts of HIV and SIV. We found that minocycline attenuated in vitro induction of type I interferon (IFN) and the IFN-stimulated genes indoleamine 2,3-dioxygenase (IDO1) and TNF-related apoptosis inducing ligand (TRAIL) in human plasmacytoid dendritic cells and PBMCs exposed to aldrithiol-2 inactivated HIV or infectious influenza virus. Activation-induced TRAIL and expression of cytotoxic T-lymphocyte antigen 4 (CTLA-4) in isolated CD4+ T cells were also reduced by minocycline. Translation of these in vitro findings to in vivo effects, however, were mixed as minocycline significantly reduced markers of activation and activation-induced cell death (CD25, Fas, caspase-3) but did not affect expression of IFNß or the IFN-stimulated genes IDO1, FasL, or Mx in the spleens of chronically SIV-infected pigtailed macaques. TRAIL expression, reflecting the mixed effects of minocycline on activation and type I IFN stimuli, was reduced by half, but this change was not significant. These results show that minocycline administered after infection may protect against aspects of activation-induced cell death during HIV/SIV immune disease, but that in vitro effects of minocycline on type I IFN responses are not recapitulated in a rapid progressor model in vivo.


Subject(s)
HIV Infections/immunology , HIV-1/immunology , Immunity , Minocycline/therapeutic use , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/immunology , Tetracycline/chemistry , Animals , Anti-Retroviral Agents/pharmacology , Anti-Retroviral Agents/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/metabolism , Cell Separation , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drug Therapy, Combination , Flow Cytometry , Gene Expression Regulation/drug effects , HIV Infections/drug therapy , HIV Infections/genetics , HIV Infections/virology , HIV-1/drug effects , Humans , Immunity/drug effects , Immunity/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Influenza, Human/immunology , Influenza, Human/virology , Interferon Type I/metabolism , Lymphocyte Activation/drug effects , Macaca nemestrina/immunology , Macaca nemestrina/virology , Minocycline/pharmacology , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tetracycline/pharmacology
16.
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
17.
J Med Primatol ; 43(5): 349-59, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24372425

ABSTRACT

BACKGROUND: Long-acting, hormonal contraception may increase HIV risk. Copper intrauterine devices (IUDs) could serve as non-hormonal alternatives. We pilot a pigtail macaque model for evaluating HIV susceptibility factors during copper IUD use. METHODS: Frameless and flexible GyneFix(®) copper IUDs were surgically implanted into three SHIVSF 162p3 -positive macaques via hysterotomy and monitored for up to 4 months. Four macaques served as non-IUD controls. RESULTS: All animals retained the devices without complications. No consistent change in vaginal viral RNA or inflammatory cytokines was seen. Two animals had altered menstrual cycles and experienced marked thinning of vaginal epithelium after IUD insertion. Histological examination of uterine tissue at necropsy revealed endometrial ulceration and lymphocytic inflammation with glandular loss at sites of direct IUD contact. CONCLUSIONS: Although the need for insertion surgery could limit its usefulness, this model will allow studies on copper IUDs and SHIV shedding, disease progression, and HIV susceptibility factors.


Subject(s)
HIV Infections/prevention & control , Intrauterine Devices, Copper/adverse effects , Macaca nemestrina , Simian Acquired Immunodeficiency Syndrome/prevention & control , Animals , Contraception , Disease Models, Animal , Disease Susceptibility/immunology , Disease Susceptibility/physiopathology , Disease Susceptibility/virology , Female , HIV/physiology , HIV Infections/immunology , HIV Infections/physiopathology , HIV Infections/virology , Humans , Macaca nemestrina/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/physiopathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/isolation & purification , Uterus/immunology , Virus Shedding
18.
J Virol ; 87(12): 6678-92, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23552412

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) antagonizes innate restriction factors in order to infect and persistently replicate in a host. In a previous study, we demonstrated that HIV-1 NL4-3 with a simian immunodeficiency virus mne (SIVmne) vif gene substitution (HSIV-vif-NL4-3) could infect and replicate in pig-tailed macaques (PTM), indicating that APOBEC3 proteins are primary barriers to transmission. Because viral replication was persistent but low, we hypothesized that HSIV-vif-NL4-3 may be suppressed by type I interferons (IFN-I), which are known to upregulate the expression of innate restriction factors. Here, we demonstrate that IFN-α more potently suppresses HSIV-vif-NL4-3 in PTM CD4(+) T cells than it does pathogenic SIVmne027. Importantly, we identify a variant (HSIV-vif-Yu2) that is resistant to IFN-α, indicating that the IFN-α-induced barrier can be overcome by HSIV-vif chimeras in PTM CD4(+) T cells. Interestingly, HSIV-vif-Yu2 and HSIV-vif-NL4-3 are similarly restricted by PTM BST2/Tetherin, and neither virus downregulates it from the surface of infected PTM CD4(+) T cells. Resistance to IFN-α-induced restriction appears to be conferred by a determinant in HSIV-vif-Yu2 that includes env su. Finally, we show that the Yu-2 env su allele may overcome an IFN-α-induced barrier to entry. Together, our data demonstrate that the prototype macaque-tropic HIV-1 clones based on NL4-3 may not sufficiently antagonize innate restriction in PTM cells. However, variants with resistance to IFN-α-induced restriction factors in PTM CD4(+) T cells may enhance viral replication by overcoming a barrier early in the viral replication cycle.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV-1/genetics , HIV-1/physiology , Host-Pathogen Interactions/immunology , Interferon-alpha/immunology , Macaca nemestrina/virology , Animals , CD4 Lymphocyte Count , Drug Resistance, Viral/immunology , HEK293 Cells , HeLa Cells , Humans , Interferon-alpha/pharmacology , Macaca nemestrina/immunology , Virus Replication
19.
PLoS One ; 8(2): e57343, 2013.
Article in English | MEDLINE | ID: mdl-23460840

ABSTRACT

SerpinB2, also known as plasminogen activator inhibitor type 2, is a major product of activated monocytes/macrophages and is often strongly induced during infection and inflammation; however, its physiological function remains somewhat elusive. Herein we show that SerpinB2 is induced in peripheral blood mononuclear cells following infection of pigtail macaques with CCR5-utilizing (macrophage-tropic) SIVmac239, but not the rapidly pathogenic CXCR4-utilizing (T cell-tropic) SHIVmn229. To investigate the role of SerpinB2 in lentiviral infections, SerpinB2(-/-) mice were infected with EcoHIV, a chimeric HIV in which HIV gp120 has been replaced with gp80 from ecotropic murine leukemia virus. EcoHIV infected SerpinB2(-/-) mice produced significantly lower anti-gag IgG1 antibody titres than infected SerpinB2(+/+) mice, and showed slightly delayed clearance of EcoHIV. Analyses of published microarray studies showed significantly higher levels of SerpinB2 mRNA in monocytes from HIV-1 infected patients when compared with uninfected controls, as well as a significant negative correlation between SerpinB2 and T-bet mRNA levels in peripheral blood mononuclear cells. These data illustrate that SerpinB2 can be induced by lentiviral infection in vivo and support the emerging notion that a physiological role of SerpinB2 is modulation of Th1/Th2 responses.


Subject(s)
Lentivirus Infections/immunology , Plasminogen Activator Inhibitor 2/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Cell Movement , Gene Expression Regulation , HIV Infections/genetics , HIV Infections/immunology , HIV Infections/virology , HIV-1/physiology , Humans , Immunoglobulin G/immunology , Lentivirus Infections/genetics , Lentivirus Infections/pathology , Lentivirus Infections/virology , Macaca nemestrina/immunology , Macaca nemestrina/virology , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Plasminogen Activator Inhibitor 2/deficiency , Plasminogen Activator Inhibitor 2/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Simian Immunodeficiency Virus/physiology , Virus Replication/physiology
20.
J Clin Invest ; 123(2): 594-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23281401

ABSTRACT

Long-lived memory T cells are able to persist in the host in the absence of antigen; however, the mechanism by which they are maintained is not well understood. Recently, a subset of human T cells, stem cell memory T cells (TSCM cells), was shown to be self-renewing and multipotent, thereby providing a potential reservoir for T cell memory throughout life. However, their in vivo dynamics and homeostasis still remain to be defined due to the lack of suitable animal models. We identified T cells with a TSCM phenotype and stem cell-like properties in nonhuman primates. These cells were the least-differentiated memory subset, were functionally distinct from conventional memory cells, and served as precursors of central memory. Antigen-specific TSCM cells preferentially localized to LNs and were virtually absent from mucosal surfaces. They were generated in the acute phase of viral infection, preferentially survived in comparison with all other memory cells following elimination of antigen, and stably persisted for the long term. Thus, one mechanism for maintenance of long-term T cell memory derives from the unique homeostatic properties of TSCM cells. Vaccination strategies designed to elicit durable cellular immunity should target the generation of TSCM cells.


Subject(s)
Immunologic Memory , Multipotent Stem Cells/immunology , T-Lymphocyte Subsets/immunology , Animals , Antigens, Viral , Cytokines/biosynthesis , Humans , Immunophenotyping , Lymphocyte Activation , Macaca mulatta/immunology , Macaca nemestrina/immunology , Simian Immunodeficiency Virus/immunology , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...