Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 101
Filtrer
1.
Front Immunol ; 15: 1445209, 2024.
Article de Anglais | MEDLINE | ID: mdl-39346922

RÉSUMÉ

Introduction: Epstein-Barr virus (EBV) is an oncogenic human herpesvirus associated with ~350,000 cases of lymphoid and epithelial malignancies every year, and is etiologically linked to infectious mononucleosis and multiple sclerosis. Despite four decades of research, no EBV vaccine candidate has yet reached licensure. Most previous vaccine attempts focused on a single viral entry glycoprotein, gp350, but recent data from clinical and pre-clinical studies, and the elucidation of viral entry mechanisms, support the inclusion of multiple entry glycoproteins in EBV vaccine design. Methods: Here we generated a modified vaccinia Ankara (MVA)-vectored EBV vaccine, MVA-EBV5-2, that targets five EBV entry glycoproteins, gp350, gB, and the gp42gHgL complex. We characterized the genetic and translational stability of the vaccine, followed by immunogenicity assessment in BALB/c mice and rhesus lymphocryptovirus-negative rhesus macaques as compared to a gp350-based MVA vaccine. Finally, we assessed the efficacy of MVA-EBV5-2-immune rhesus serum at preventing EBV infection in human CD34+ hematopoietic stem cell-reconstituted NSG mice, under two EBV challenge doses. Results: The MVA-EBV5-2 vaccine was genetically and translationally stable over 10 viral passages as shown by genetic and protein expression analysis, and when administered to female and male BALB/c mice, elicited serum EBV-specific IgG of both IgG1 and IgG2a subtypes with neutralizing activity in vitro. In Raji B cells, this neutralizing activity outperformed that of serum from mice immunized with a monovalent MVA-vectored gp350 vaccine. Similarly, MVA-EBV5-2 elicited EBV-specific IgG in rhesus macaques that were detected in both serum and saliva of immunized animals, with serum antibodies demonstrating neutralizing activity in vitro that outperformed serum from MVA-gp350-immunized macaques. Finally, pre-treatment with serum from MVA-EBV5-2-immunized macaques resulted in fewer EBV-infected mice in the two challenge experiments than pretreatment with serum from pre-immune macaques or macaques immunized with the monovalent gp350-based vaccine. Discussion: These results support the inclusion of multiple entry glycoproteins in EBV vaccine design and position our vaccine as a strong candidate for clinical translation.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Infections à virus Epstein-Barr , Herpèsvirus humain de type 4 , Macaca mulatta , Animaux , Humains , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Infections à virus Epstein-Barr/immunologie , Infections à virus Epstein-Barr/prévention et contrôle , Souris , Herpèsvirus humain de type 4/immunologie , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Souris de lignée BALB C , Vaccins à ADN/immunologie , Femelle , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Vecteurs génétiques/génétique , Virus de la vaccine/immunologie , Virus de la vaccine/génétique
2.
PLoS Pathog ; 20(8): e1012496, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39173097

RÉSUMÉ

Persistence of the rebound-competent viral reservoir (RCVR) within the CD4+ T cell compartment of people living with HIV remains a major barrier to HIV cure. Here, we determined the effects of the pan-lymphocyte-depleting monoclonal antibody (mAb) alemtuzumab on the RCVR in SIVmac239-infected rhesus macaques (RM) receiving antiretroviral therapy (ART). Alemtuzumab administered during chronic ART or at the time of ART initiation induced >95% depletion of circulating CD4+ T cells in peripheral blood and substantial CD4+ T cell depletion in lymph nodes. However, treatment was followed by proliferation and reconstitution of CD4+ T cells in blood, and despite ongoing ART, levels of cell-associated SIV DNA in blood and lymphoid tissues were not substantially different between alemtuzumab-treated and control RM after immune cell reconstitution, irrespective of the time of alemtuzumab treatment. Upon ART cessation, 19 of 22 alemtuzumab-treated RM and 13 of 13 controls rebounded with no difference in the time to rebound between treatment groups. Time to rebound and reactivation rate was associated with plasma viral loads (pVLs) at time of ART initiation, suggesting lymphocyte depletion had no durable impact on the RCVR. However, 3 alemtuzumab-treated RM that had lowest levels of pre-ART viremia, failed to rebound after ART withdrawal, in contrast to controls with similar levels of SIV replication. These observations suggest that alemtuzumab therapy has little to no ability to reduce well-established RCVRs but may facilitate RCVR destabilization when pre-ART virus levels are particularly low.


Sujet(s)
Alemtuzumab , Déplétion lymphocytaire , Macaca mulatta , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Charge virale , Animaux , Virus de l'immunodéficience simienne/effets des médicaments et des substances chimiques , Virus de l'immunodéficience simienne/immunologie , Syndrome d'immunodéficience acquise du singe/traitement médicamenteux , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/virologie , Alemtuzumab/pharmacologie , Déplétion lymphocytaire/méthodes , Charge virale/effets des médicaments et des substances chimiques , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/virologie , Lymphocytes T CD4+/effets des médicaments et des substances chimiques
3.
Cell Rep Med ; 5(7): 101655, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39019010

RÉSUMÉ

Yellow fever virus (YFV) is endemic in >40 countries and causes viscerotropic disease with up to 20%-60% mortality. Successful live-attenuated yellow fever (YF) vaccines were developed in the mid-1930s, but their use is restricted or formally contraindicated in vulnerable populations including infants, the elderly, and people with compromised immune systems. In these studies, we describe the development of a next-generation hydrogen peroxide-inactivated YF vaccine and determine immune correlates of protection based on log neutralizing index (LNI) and neutralizing titer-50% (NT50) studies. In addition, we compare neutralizing antibody responses and protective efficacy of hydrogen peroxide-inactivated YF vaccine candidates to live-attenuated YFV-17D (YF-VAX) in a rhesus macaque model of viscerotropic YF. Our results indicate that an optimized, inactivated YF vaccine elicits protective antibody responses that prevent viral dissemination and lethal infection in rhesus macaques and may be a suitable alternative for vaccinating vulnerable populations who are not eligible to receive replicating live-attenuated YF vaccines.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Modèles animaux de maladie humaine , Peroxyde d'hydrogène , Macaca mulatta , Vaccins inactivés , Vaccin antiamaril , Fièvre jaune , Virus de la fièvre jaune , Animaux , Vaccins inactivés/immunologie , Vaccin antiamaril/immunologie , Fièvre jaune/prévention et contrôle , Fièvre jaune/immunologie , Virus de la fièvre jaune/immunologie , Anticorps neutralisants/immunologie , Anticorps antiviraux/immunologie , Vaccins atténués/immunologie , Chlorocebus aethiops , Cellules Vero , Humains
4.
Sci Adv ; 10(19): eadm7515, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38728394

RÉSUMÉ

The nonpolymorphic major histocompatibility complex E (MHC-E) molecule is up-regulated on many cancer cells, thus contributing to immune evasion by engaging inhibitory NKG2A/CD94 receptors on NK cells and tumor-infiltrating T cells. To investigate whether MHC-E expression by cancer cells can be targeted for MHC-E-restricted T cell control, we immunized rhesus macaques (RM) with rhesus cytomegalovirus (RhCMV) vectors genetically programmed to elicit MHC-E-restricted CD8+ T cells and to express established tumor-associated antigens (TAAs) including prostatic acidic phosphatase (PAP), Wilms tumor-1 protein, or Mesothelin. T cell responses to all three tumor antigens were comparable to viral antigen-specific responses with respect to frequency, duration, phenotype, epitope density, and MHC restriction. Thus, CMV-vectored cancer vaccines can bypass central tolerance by eliciting T cells to noncanonical epitopes. We further demonstrate that PAP-specific, MHC-E-restricted CD8+ T cells from RhCMV/PAP-immunized RM respond to PAP-expressing HLA-E+ prostate cancer cells, suggesting that the HLA-E/NKG2A immune checkpoint can be exploited for CD8+ T cell-based immunotherapies.


Sujet(s)
Antigènes néoplasiques , Lymphocytes T CD8+ , HLA-E Antigens , Animaux , Humains , Mâle , Acid phosphatase , Présentation d'antigène/immunologie , Antigènes néoplasiques/immunologie , Vaccins anticancéreux/immunologie , Lymphocytes T CD8+/immunologie , Lignée cellulaire tumorale , Cytomegalovirus/immunologie , Antigènes d'histocompatibilité de classe I/immunologie , Antigènes d'histocompatibilité de classe I/métabolisme , Macaca mulatta , Mésothéline
5.
J Clin Invest ; 134(7)2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38557496

RÉSUMÉ

Programmed cell death protein 1 (PD-1) is an immune checkpoint marker commonly expressed on memory T cells and enriched in latently HIV-infected CD4+ T cells. We engineered an anti-PD-1 chimeric antigen receptor (CAR) to assess the impact of PD-1 depletion on viral reservoirs and rebound dynamics in SIVmac239-infected rhesus macaques (RMs). Adoptive transfer of anti-PD-1 CAR T cells was done in 2 SIV-naive and 4 SIV-infected RMs on antiretroviral therapy (ART). In 3 of 6 RMs, anti-PD-1 CAR T cells expanded and persisted for up to 100 days concomitant with the depletion of PD-1+ memory T cells in blood and tissues, including lymph node CD4+ follicular helper T (TFH) cells. Loss of TFH cells was associated with depletion of detectable SIV RNA from the germinal center (GC). However, following CAR T infusion and ART interruption, there was a marked increase in SIV replication in extrafollicular portions of lymph nodes, a 2-log higher plasma viremia relative to controls, and accelerated disease progression associated with the depletion of CD8+ memory T cells. These data indicate anti-PD-1 CAR T cells depleted PD-1+ T cells, including GC TFH cells, and eradicated SIV from this immunological sanctuary.


Sujet(s)
Lymphocytes T CD4+ , Récepteurs chimériques pour l'antigène , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Lymphocytes T CD4+/immunologie , Centre germinatif/immunologie , Infections à VIH/thérapie , Macaca mulatta/métabolisme , Récepteur-1 de mort cellulaire programmée , Récepteurs chimériques pour l'antigène/génétique , Syndrome d'immunodéficience acquise du singe/thérapie
6.
PLoS Negl Trop Dis ; 17(11): e0011742, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37983245

RÉSUMÉ

Mayaro virus (MAYV) is a mosquito-transmitted alphavirus that causes debilitating and persistent arthritogenic disease. While MAYV was previously reported to infect non-human primates (NHP), characterization of MAYV pathogenesis is currently lacking. Therefore, in this study we characterized MAYV infection and immunity in rhesus macaques. To inform the selection of a viral strain for NHP experiments, we evaluated five MAYV strains in C57BL/6 mice and showed that MAYV strain BeAr505411 induced robust tissue dissemination and disease. Three male rhesus macaques were subcutaneously challenged with 105 plaque-forming units of this strain into the arms. Peak plasma viremia occurred at 2 days post-infection (dpi). NHPs were taken to necropsy at 10 dpi to assess viral dissemination, which included the muscles and joints, lymphoid tissues, major organs, male reproductive tissues, as well as peripheral and central nervous system tissues. Histological examination demonstrated that MAYV infection was associated with appendicular joint and muscle inflammation as well as presence of perivascular inflammation in a wide variety of tissues. One animal developed a maculopapular rash and two NHP had viral RNA detected in upper torso skin samples, which was associated with the presence of perivascular and perifollicular lymphocytic aggregation. Analysis of longitudinal peripheral blood samples indicated a robust innate and adaptive immune activation, including the presence of anti-MAYV neutralizing antibodies with activity against related Una virus and chikungunya virus. Inflammatory cytokines and monocyte activation also peaked coincident with viremia, which was well supported by our transcriptomic analysis highlighting enrichment of interferon signaling and other antiviral processes at 2 days post MAYV infection. The rhesus macaque model of MAYV infection recapitulates many of the aspects of human infection and is poised to facilitate the evaluation of novel therapies and vaccines targeting this re-emerging virus.


Sujet(s)
Infections à alphavirus , Alphavirus , Virus du chikungunya , Animaux , Souris , Mâle , Macaca mulatta , Virémie , Souris de lignée C57BL , Anticorps antiviraux
7.
Immunity ; 56(7): 1649-1663.e5, 2023 07 11.
Article de Anglais | MEDLINE | ID: mdl-37236188

RÉSUMÉ

Allogeneic hematopoietic stem cell transplantation (alloHSCT) from donors lacking C-C chemokine receptor 5 (CCR5Δ32/Δ32) can cure HIV, yet mechanisms remain speculative. To define how alloHSCT mediates HIV cure, we performed MHC-matched alloHSCT in SIV+, anti-retroviral therapy (ART)-suppressed Mauritian cynomolgus macaques (MCMs) and demonstrated that allogeneic immunity was the major driver of reservoir clearance, occurring first in peripheral blood, then peripheral lymph nodes, and finally in mesenteric lymph nodes draining the gastrointestinal tract. While allogeneic immunity could extirpate the latent viral reservoir and did so in two alloHSCT-recipient MCMs that remained aviremic >2.5 years after stopping ART, in other cases, it was insufficient without protection of engrafting cells afforded by CCR5-deficiency, as CCR5-tropic virus spread to donor CD4+ T cells despite full ART suppression. These data demonstrate the individual contributions of allogeneic immunity and CCR5 deficiency to HIV cure and support defining targets of alloimmunity for curative strategies independent of HSCT.


Sujet(s)
Infections à VIH , Transplantation de cellules souches hématopoïétiques , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Macaca fascicularis , Charge virale
9.
Cell Host Microbe ; 30(9): 1207-1218.e7, 2022 09 14.
Article de Anglais | MEDLINE | ID: mdl-35981532

RÉSUMÉ

Strain 68-1 rhesus cytomegalovirus expressing simian immunodeficiency virus (SIV) antigens (RhCMV/SIV) primes MHC-E-restricted CD8+ T cells that control SIV replication in 50%-60% of the vaccinated rhesus macaques. Whether this unconventional SIV-specific immunity and protection is unique to rhesus macaques or RhCMV or is intrinsic to CMV remains unknown. Here, using cynomolgus CMV vectors expressing SIV antigens (CyCMV/SIV) and Mauritian cynomolgus macaques, we demonstrate that the induction of MHC-E-restricted CD8+ T cells requires matching CMV to its host species. RhCMV does not elicit MHC-E-restricted CD8+ T cells in cynomolgus macaques. However, cynomolgus macaques vaccinated with species-matched 68-1-like CyCMV/SIV mounted MHC-E-restricted CD8+ T cells, and half of the vaccinees stringently controlled SIV post-challenge. Protected animals manifested a vaccine-induced IL-15 transcriptomic signature that is associated with efficacy in rhesus macaques. These findings demonstrate that the ability of species-matched CMV vectors to elicit MHC-E-restricted CD8+ T cells that are required for anti-SIV efficacy is conserved in nonhuman primates, and these data support the development of HCMV/HIV for a prophylactic HIV vaccine.


Sujet(s)
Vaccins contre le SIDA , Infections à cytomégalovirus , Vaccins contre le cytomégalovirus , Vaccins contre le SIDA simien , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Lymphocytes T CD8+ , Cytomegalovirus/génétique , Interleukine-15 , Macaca fascicularis , Macaca mulatta
10.
Sci Immunol ; 7(72): eabn9301, 2022 06 24.
Article de Anglais | MEDLINE | ID: mdl-35714200

RÉSUMÉ

The strain 68-1 rhesus cytomegalovirus (RhCMV)-based vaccine for simian immunodeficiency virus (SIV) can stringently protect rhesus macaques (RMs) from SIV challenge by arresting viral replication early in primary infection. This vaccine elicits unconventional SIV-specific CD8+ T cells that recognize epitopes presented by major histocompatibility complex (MHC)-II and MHC-E instead of MHC-Ia. Although RhCMV/SIV vaccines based on strains that only elicit MHC-II- and/or MHC-Ia-restricted CD8+ T cells do not protect against SIV, it remains unclear whether MHC-E-restricted T cells are directly responsible for protection and whether these responses can be separated from the MHC-II-restricted component. Using host microRNA (miR)-mediated vector tropism restriction, we show that the priming of MHC-II and MHC-E epitope-targeted responses depended on vector infection of different nonoverlapping cell types in RMs. Selective inhibition of RhCMV infection in myeloid cells with miR-142-mediated tropism restriction eliminated MHC-E epitope-targeted CD8+ T cell priming, yielding an exclusively MHC-II epitope-targeted response. Inhibition with the endothelial cell-selective miR-126 eliminated MHC-II epitope-targeted CD8+ T cell priming, yielding an exclusively MHC-E epitope-targeted response. Dual miR-142 + miR-126-mediated tropism restriction reverted CD8+ T cell responses back to conventional MHC-Ia epitope targeting. Although the magnitude and differentiation state of these CD8+ T cell responses were generally similar, only the vectors programmed to elicit MHC-E-restricted CD8+ T cell responses provided protection against SIV challenge, directly demonstrating the essential role of these responses in RhCMV/SIV vaccine efficacy.


Sujet(s)
Vaccins contre le cytomégalovirus , microARN , Vaccins contre le SIDA simien , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Lymphocytes T CD8+ , Cytomegalovirus/génétique , Épitopes , Macaca mulatta , Complexe majeur d'histocompatibilité , Cellules myéloïdes , Syndrome d'immunodéficience acquise du singe/génétique , Virus de l'immunodéficience simienne/génétique , Tropisme , 59641
11.
PLoS One ; 17(4): e0266616, 2022.
Article de Anglais | MEDLINE | ID: mdl-35442982

RÉSUMÉ

Surgical antimicrobial prophylaxis is indicated when performing contaminated surgeries, when specific surgical implants are placed, and for prolonged surgical procedures. Unnecessary prophylactic antibiotics are often utilized for macaque surgeries, despite medical and veterinary guidelines. In this study we compared complication rates in macaques receiving peripheral lymph node (PLN) and laparoscopic biopsies, with and without antimicrobial prophylaxis. A majority of animals were SIV or SHIV infected at the time of surgery, so we also compared post-operative complication rates based on infection status. We found no significant difference in PLN biopsy complication rates for animals that received antimicrobial prophylaxis versus those that did not. Animals who underwent laparoscopic procedures and received prophylactic antibiotics had a higher complication rate than those who did not receive them. Complication rates did not differ significantly for SIV/SHIV infected versus uninfected animals for both laparoscopic biopsy procedures and PLN biopsy procedures. SIV/SHIV infected animals that underwent PLN biopsies had no significant difference in complication rates with and without antimicrobial prophylaxis, and SIV/SHIV infected animals receiving prophylactic antibiotics for laparoscopic biopsies had a higher complication rate than those that did not. This study suggests that perioperative prophylactic antibiotics have no role in the management of SIV/SHIV-infected and uninfected macaques undergoing clean, minimally invasive surgeries. Additionally, we recommend eliminating unnecessary antibiotic use in study animals due to their potential confounding impacts on research models and their potential to promote antimicrobial resistance.


Sujet(s)
Anti-infectieux , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Antibactériens/usage thérapeutique , Anti-infectieux/usage thérapeutique , Macaca fascicularis , Macaca mulatta , Études rétrospectives , Syndrome d'immunodéficience acquise du singe/traitement médicamenteux , Résultat thérapeutique
12.
J Clin Invest ; 132(10)2022 05 16.
Article de Anglais | MEDLINE | ID: mdl-35316218

RÉSUMÉ

Proliferation of latently infected CD4+ T cells with replication-competent proviruses is an important mechanism contributing to HIV persistence during antiretroviral therapy (ART). One approach to targeting this latent cell expansion is to inhibit mTOR, a regulatory kinase involved with cell growth, metabolism, and proliferation. Here, we determined the effects of chronic mTOR inhibition with rapamycin with or without T cell activation in SIV-infected rhesus macaques (RMs) on ART. Rapamycin perturbed the expression of multiple genes and signaling pathways important for cellular proliferation and substantially decreased the frequency of proliferating CD4+ memory T cells (TM cells) in blood and tissues. However, levels of cell-associated SIV DNA and SIV RNA were not markedly different between rapamycin-treated RMs and controls during ART. T cell activation with an anti-CD3LALA antibody induced increases in SIV RNA in plasma of RMs on rapamycin, consistent with SIV production. However, upon ART cessation, both rapamycin and CD3LALA-treated and control-treated RMs rebounded in less than 12 days, with no difference in the time to viral rebound or post-ART viral load set points. These results indicate that, while rapamycin can decrease the proliferation of CD4+ TM cells, chronic mTOR inhibition alone or in combination with T cell activation was not sufficient to disrupt the stability of the SIV reservoir.


Sujet(s)
Infections à VIH , Syndrome d'immunodéficience acquise du singe , Virus de l'immunodéficience simienne , Animaux , Antirétroviraux/pharmacologie , Antirétroviraux/usage thérapeutique , Lymphocytes T CD4+ , Prolifération cellulaire , Infections à VIH/traitement médicamenteux , Macaca mulatta/génétique , ARN , Sirolimus/pharmacologie , Sérine-thréonine kinases TOR/génétique , Sérine-thréonine kinases TOR/pharmacologie , Charge virale , Réplication virale
13.
PLoS Pathog ; 17(7): e1009278, 2021 07.
Article de Anglais | MEDLINE | ID: mdl-34228762

RÉSUMÉ

Simian immunodeficiency virus (SIV) challenge of rhesus macaques (RMs) vaccinated with strain 68-1 Rhesus Cytomegalovirus (RhCMV) vectors expressing SIV proteins (RhCMV/SIV) results in a binary outcome: stringent control and subsequent clearance of highly pathogenic SIV in ~55% of vaccinated RMs with no protection in the remaining 45%. Although previous work indicates that unconventionally restricted, SIV-specific, effector-memory (EM)-biased CD8+ T cell responses are necessary for efficacy, the magnitude of these responses does not predict efficacy, and the basis of protection vs. non-protection in 68-1 RhCMV/SIV vector-vaccinated RMs has not been elucidated. Here, we report that 68-1 RhCMV/SIV vector administration strikingly alters the whole blood transcriptome of vaccinated RMs, with the sustained induction of specific immune-related pathways, including immune cell, toll-like receptor (TLR), inflammasome/cell death, and interleukin-15 (IL-15) signaling, significantly correlating with subsequent vaccine efficacy. Treatment of a separate RM cohort with IL-15 confirmed the central involvement of this cytokine in the protection signature, linking the major innate and adaptive immune gene expression networks that correlate with RhCMV/SIV vaccine efficacy. This change-from-baseline IL-15 response signature was also demonstrated to significantly correlate with vaccine efficacy in an independent validation cohort of vaccinated and challenged RMs. The differential IL-15 gene set response to vaccination strongly correlated with the pre-vaccination activity of this pathway, with reduced baseline expression of IL-15 response genes significantly correlating with higher vaccine-induced induction of IL-15 signaling and subsequent vaccine protection, suggesting that a robust de novo vaccine-induced IL-15 signaling response is needed to program vaccine efficacy. Thus, the RhCMV/SIV vaccine imparts a coordinated and persistent induction of innate and adaptive immune pathways featuring IL-15, a known regulator of CD8+ T cell function, that support the ability of vaccine-elicited unconventionally restricted CD8+ T cells to mediate protection against SIV challenge.


Sujet(s)
Lymphocytes T CD8+/immunologie , Interleukine-15/immunologie , Vaccins contre le SIDA simien/immunologie , Virus de l'immunodéficience simienne/immunologie , Animaux , Cytomegalovirus , Femelle , Vecteurs génétiques , Macaca mulatta , Mâle , Syndrome d'immunodéficience acquise du singe/prévention et contrôle
14.
PLoS Pathog ; 17(5): e1009565, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-33970966

RÉSUMÉ

Here, we assessed the efficacy of a short-course multimodal therapy (enrofloxacin, azithromycin, fenbendazole, and paromomycin) to eliminate common macaque endemic pathogens (EPs) and evaluated its impact on gastrointestinal (GI) microbiota, mucosal integrity, and local and systemic inflammation in sixteen clinically healthy macaques. Treatment combined with expanded practices resulted in successful maintenance of rhesus macaques (RM) free of common EPs, with no evidence of overt microbiota diversity loss or dysbiosis and instead resulted in a more defined luminal microbiota across study subjects. Creation of a GI pathogen free (GPF) status resulted in improved colonic mucosal barrier function (histologically, reduced colonic MPO+, and reduced pan-bacterial 16s rRNA in the MLN), reduced local and systemic innate and adaptive inflammation with reduction of colonic Mx1 and pSTAT1, decreased intermediate (CD14+CD16+) and non-classical monocytes (CD14-CD16+), reduced populations of peripheral dendritic cells, Ki-67+ and CD38+ CD4+ T cells, Ki-67+IgG+, and Ki-67+IgD+ B cells indicating lower levels of background inflammation in the distal descending colon, draining mesenteric lymph nodes, and systemically in peripheral blood, spleen, and axillary lymph nodes. A more controlled rate of viral acquisition resulted when untreated and treated macaques were challenged by low dose intrarectal SIVmac239x, with an ~100 fold increase in dose required to infect 50% (AID50) of the animals receiving treatment compared to untreated controls. Reduction in and increased consistency of number of transmitted founder variants resulting from challenge seen in the proof of concept study directly correlated with post-treatment GPF animal's improved barrier function and reduction of key target cell populations (Ki-67+ CD4+T cells) at the site of viral acquisition in the follow up study. These data demonstrate that a therapeutic and operational strategy can successfully eliminate varying background levels of EPs and their associated aberrant immunomodulatory effects within a captive macaque cohort, leading to a more consistent, better defined and reproducible research model.


Sujet(s)
Inflammation/thérapie , Microbiote/effets des médicaments et des substances chimiques , Syndrome d'immunodéficience acquise du singe/thérapie , Virus de l'immunodéficience simienne/immunologie , Immunité acquise , Animaux , Lymphocytes B , Lymphocytes T CD4+ , Prolifération cellulaire , Association thérapeutique , Tube digestif/immunologie , Tube digestif/microbiologie , Humains , Immunité innée , Muqueuse intestinale , Noeuds lymphatiques , Macaca mulatta , Mâle , Monocytes , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/virologie
15.
Sci Immunol ; 6(57)2021 03 25.
Article de Anglais | MEDLINE | ID: mdl-33766849

RÉSUMÉ

Simian immunodeficiency virus (SIV) insert-expressing, 68-1 rhesus cytomegalovirus (RhCMV/SIV) vectors elicit major histocompatibility complex E (MHC-E)- and MHC-II-restricted, SIV-specific CD8+ T cell responses, but the basis of these unconventional responses and their contribution to demonstrated vaccine efficacy against SIV challenge in the rhesus monkeys (RMs) have not been characterized. We show that these unconventional responses resulted from a chance genetic rearrangement in 68-1 RhCMV that abrogated the function of eight distinct immunomodulatory gene products encoded in two RhCMV genomic regions (Rh157.5/Rh157.4 and Rh158-161), revealing three patterns of unconventional response inhibition. Differential repair of these genes with either RhCMV-derived or orthologous human CMV (HCMV)-derived sequences (UL128/UL130; UL146/UL147) leads to either of two distinct CD8+ T cell response types-MHC-Ia-restricted only or a mix of MHC-II- and MHC-Ia-restricted CD8+ T cells. Response magnitude and functional differentiation are similar to RhCMV 68-1, but neither alternative response type mediated protection against SIV challenge. These findings implicate MHC-E-restricted CD8+ T cell responses as mediators of anti-SIV efficacy and indicate that translation of RhCMV/SIV vector efficacy to humans will likely require deletion of all genes that inhibit these responses from the HCMV/HIV vector.


Sujet(s)
Lymphocytes T CD8+/immunologie , Reprogrammation cellulaire/immunologie , Infections à cytomégalovirus/médecine vétérinaire , Cytomegalovirus/immunologie , Maladies des singes/prévention et contrôle , Syndrome d'immunodéficience acquise du singe/immunologie , Vaccins antiviraux/immunologie , Animaux , Antigènes viraux/génétique , Antigènes viraux/immunologie , Lymphocytes T CD8+/métabolisme , Reprogrammation cellulaire/génétique , Génie génétique/méthodes , Vecteurs génétiques/génétique , Immunogénicité des vaccins , Mémoire immunologique , Macaca mulatta , Maladies des singes/immunologie , Maladies des singes/virologie , Cadres ouverts de lecture/génétique , Cadres ouverts de lecture/immunologie , Syndrome d'immunodéficience acquise du singe/prévention et contrôle , Virus de l'immunodéficience simienne/immunologie , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , 59641
16.
Science ; 372(6541)2021 04 30.
Article de Anglais | MEDLINE | ID: mdl-33766941

RÉSUMÉ

Strain 68-1 rhesus cytomegalovirus (RhCMV) vectors expressing simian immunodeficiency virus (SIV) antigens elicit CD8+ T cells recognizing epitopes presented by major histocompatibility complex II (MHC-II) and MHC-E but not MHC-Ia. These immune responses mediate replication arrest of SIV in 50 to 60% of monkeys. We show that the peptide VMAPRTLLL (VL9) embedded within the RhCMV protein Rh67 promotes intracellular MHC-E transport and recognition of RhCMV-infected fibroblasts by MHC-E-restricted CD8+ T cells. Deletion or mutation of viral VL9 abrogated MHC-E-restricted CD8+ T cell priming, resulting in CD8+ T cell responses exclusively targeting MHC-II-restricted epitopes. These responses were comparable in magnitude and differentiation to responses elicited by 68-1 vectors but did not protect against SIV. Thus, Rh67-enabled direct priming of MHC-E-restricted T cells is crucial for RhCMV/SIV vaccine efficacy.


Sujet(s)
Lymphocytes T CD8+/immunologie , Cytomegalovirus/métabolisme , Vecteurs génétiques/métabolisme , Antigènes d'histocompatibilité de classe I/métabolisme , Fragments peptidiques/métabolisme , Vaccins contre le SIDA simien/immunologie , Animaux , Lignée cellulaire , Cytomegalovirus/génétique , Déterminants antigéniques des lymphocytes T/immunologie , Fibroblastes/métabolisme , Vecteurs génétiques/génétique , Antigènes d'histocompatibilité de classe I/génétique , Ligands , Macaca mulatta , Fragments peptidiques/génétique , Transport des protéines , Virus de l'immunodéficience simienne , HLA-E Antigens
17.
Cell Host Microbe ; 29(4): 594-606.e6, 2021 04 14.
Article de Anglais | MEDLINE | ID: mdl-33711270

RÉSUMÉ

CD4 T cell effector function is required for optimal containment of Mycobacterium tuberculosis (Mtb) infection. IFNÉ£ produced by CD4 T cells is a key cytokine that contributes to protection. However, lung-infiltrating CD4 T cells have a limited ability to produce IFNÉ£, and IFNÉ£ plays a lesser protective role within the lung than at sites of Mtb dissemination. In a murine infection model, we observed that IFNÉ£ production by Mtb-specific CD4 T cells is rapidly extinguished within the granuloma but not within unaffected lung regions, suggesting localized immunosuppression. We identified a signature of TGFß signaling within granuloma-infiltrating T cells in both mice and rhesus macaques. Selective blockade of TGFß signaling in T cells resulted in an accumulation of terminally differentiated effector CD4 T cells, improved IFNÉ£ production within granulomas, and reduced bacterial burdens. These findings uncover a spatially localized immunosuppressive mechanism associated with Mtb infection and provide potential targets for host-directed therapy.


Sujet(s)
Granulome/immunologie , Lymphocytes T/immunologie , Facteur de croissance transformant bêta/métabolisme , Tuberculose/immunologie , Immunité acquise , Animaux , Lymphocytes T CD4+ , Mort cellulaire , Cytokines , Modèles animaux de maladie humaine , Femelle , Granulome/microbiologie , Inflammation , Interféron gamma , Poumon/microbiologie , Macaca mulatta , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Mycobacterium tuberculosis , Lymphocytes auxiliaires Th1
18.
J Clin Invest ; 131(8)2021 04 15.
Article de Anglais | MEDLINE | ID: mdl-33630764

RÉSUMÉ

To define the contribution of CD8+ T cell responses to control of SIV reactivation during and following antiretroviral therapy (ART), we determined the effect of long-term CD8+ T cell depletion using a rhesusized anti-CD8ß monoclonal antibody on barcoded SIVmac239 dynamics on stable ART and after ART cessation in rhesus macaques (RMs). Among the RMs with full CD8+ T cell depletion in both blood and tissue, there were no significant differences in the frequency of viral blips in plasma, the number of SIV RNA+ cells and the average number of RNA copies/infected cell in tissue, and levels of cell-associated SIV RNA and DNA in blood and tissue relative to control-treated RMs during ART. Upon ART cessation, both CD8+ T cell-depleted and control RMs rebounded in fewer than 12 days, with no difference in the time to viral rebound or in either the number or growth rate of rebounding SIVmac239M barcode clonotypes. However, effectively CD8+ T cell-depleted RMs showed a stable, approximately 2-log increase in post-ART plasma viremia relative to controls. These results indicate that while potent antiviral CD8+ T cell responses can develop during ART-suppressed SIV infection, these responses effectively intercept post-ART SIV rebound only after systemic viral replication, too late to limit reactivation frequency or the early spread of reactivating SIV reservoirs.


Sujet(s)
Antirétroviraux/pharmacologie , Lymphocytes T CD8+/immunologie , Déplétion lymphocytaire , Syndrome d'immunodéficience acquise du singe/immunologie , Virus de l'immunodéficience simienne/physiologie , Activation virale/immunologie , Animaux , Lymphocytes T CD8+/anatomopathologie , Femelle , Macaca mulatta , Mâle , Syndrome d'immunodéficience acquise du singe/traitement médicamenteux , Syndrome d'immunodéficience acquise du singe/anatomopathologie , Activation virale/effets des médicaments et des substances chimiques
19.
Ann Clin Transl Neurol ; 8(2): 456-470, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33440071

RÉSUMÉ

OBJECTIVE: To determine whether animals with Japanese macaque encephalomyelitis (JME), a spontaneous demyelinating disease similar to multiple sclerosis (MS), harbor myelin-specific T cells in their central nervous system (CNS) and periphery. METHODS: Mononuclear cells (MNCs) from CNS lesions, cervical lymph nodes (LNs) and peripheral blood of Japanese macaques (JMs) with JME, and cervical LN and blood MNCs from healthy controls or animals with non-JME conditions were analyzed for the presence of myelin-specific T cells and changes in interleukin 17 (IL-17) and interferon gamma (IFNγ) expression. RESULTS: Demyelinating JME lesions contained CD4+ T cells and CD8+ T cells specific to myelin oligodendrocyte glycoprotein (MOG), myelin basic protein (MBP), and/or proteolipid protein (PLP). CD8+ T-cell responses were absent in JME peripheral blood, and in age- and sex-matched controls. However, CD4+ Th1 and Th17 responses were detected in JME peripheral blood versus controls. Cervical LN MNCs from eight of nine JME animals had CD3+ T cells specific for MOG, MBP, and PLP that were not detected in controls. Mapping myelin epitopes revealed a heterogeneity in responses among JME animals. Comparison of myelin antigen sequences with those of JM rhadinovirus (JMRV), which is found in JME lesions, identified six viral open reading frames (ORFs) with similarities to myelin antigen sequences. Overlapping peptides to these JMRV ORFs did not induce IFNγ responses. INTERPRETATIONS: JME possesses an immune-mediated component that involves both CD4+ and CD8+ T cells specific for myelin antigens. JME may shed new light on inflammatory demyelinating disease pathogenesis linked to gamma-herpesvirus infection.


Sujet(s)
Maladies démyélinisantes/imagerie diagnostique , Maladies démyélinisantes/anatomopathologie , Encéphalomyélite/imagerie diagnostique , Encéphalomyélite/anatomopathologie , Gaine de myéline/immunologie , Lymphocytes T/immunologie , Animaux , Maladies auto-immunes/immunologie , Maladies démyélinisantes/virologie , Encéphalomyélite/virologie , Test ELISA , Cartographie épitopique , Épitopes/génétique , Épitopes/immunologie , Femelle , Infections à Herpesviridae/immunologie , Interféron gamma/analyse , Interleukine-17/analyse , Macaca fuscata , Mâle , Maladies des singes , Protéine basique de la myéline/génétique , Protéine basique de la myéline/immunologie , Protéine protéolipidique myéline/génétique , Protéine protéolipidique myéline/immunologie , Gaine de myéline/anatomopathologie , Glycoprotéine MOG/génétique , Glycoprotéine MOG/immunologie , Rhadinovirus/génétique , Rhadinovirus/immunologie
20.
J Clin Apher ; 36(1): 67-77, 2021 Feb.
Article de Anglais | MEDLINE | ID: mdl-32941672

RÉSUMÉ

Macaques are physiologically relevant animal models of human immunology and infectious disease that have provided key insights and advanced clinical treatment in transplantation, vaccinology, and HIV/AIDS. However, the small size of macaques is a stumbling block for studies requiring large numbers of cells, such as hematopoietic stem cells (HSCs) for transplantation, antigen-specific lymphocytes for in-depth immunological analysis, and latently-infected CD4+ T-cells for HIV cure studies. Here, we provide a detailed protocol for collection of large numbers of HSCs and T-cells from cynomolgus macaques as small as 3 kg using the Terumo Spectra Optia apheresis system, yielding an average of 5.0 × 109 total nucleated cells from mobilized animals and 1.2 × 109 total nucleated cells from nonmobilized animals per procedure. This report provides sufficient detail to adapt this apheresis technique at other institutions, which will facilitate more efficient and detailed analysis of HSCs and their progeny blood cells.


Sujet(s)
Aphérèse/méthodes , Cellules souches hématopoïétiques/cytologie , Lymphocytes T/cytologie , Animaux , Benzylamines/pharmacologie , Créatinine/sang , Cyclames/pharmacologie , Femelle , Facteur de stimulation des colonies de granulocytes/pharmacologie , Mobilisation de cellules souches hématopoïétiques/méthodes , Macaca fascicularis , Mâle
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE