Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 22
Filtrer
1.
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
2.
J Virol ; 96(16): e0072822, 2022 08 24.
Article de Anglais | MEDLINE | ID: mdl-35924920

RÉSUMÉ

The 1918 H1N1 influenza pandemic was among the most severe in history, taking the lives of approximately 50 million people worldwide, and novel prophylactic vaccines are urgently needed to prevent another pandemic. Given that macaques are physiologically relevant preclinical models of human immunology that have advanced the clinical treatment of infectious diseases, a lethal pandemic influenza challenge model would provide a stringent platform for testing new influenza vaccine concepts. To this end, we infected rhesus macaques and Mauritian cynomolgus macaques with highly pathogenic 1918 H1N1 influenza virus and assessed pathogenesis and disease severity. Despite infection with a high dose of 1918 influenza delivered via multiple routes, rhesus macaques demonstrated minimal signs of disease, with only intermittent viral shedding. Cynomolgus macaques infected via intrabronchial instillation demonstrated mild symptoms, with disease severity depending on the infection dose. Cynomolgus macaques infected with a high dose of 1918 influenza delivered via multiple routes experienced moderate disease characterized by consistent viral shedding, pulmonary infiltrates, and elevated inflammatory cytokine levels. However, 1918 influenza was uniformly nonlethal in these two species, demonstrating that this isolate is insufficiently pathogenic in rhesus and Mauritian cynomolgus macaques to support testing novel prophylactic influenza approaches where protection from severe disease combined with a lethal outcome is desired as a highly stringent indication of vaccine efficacy. IMPORTANCE The world remains at risk of an influenza pandemic, and the development of new therapeutic and preventative modalities is critically important for minimizing human death and suffering during the next influenza pandemic. Animal models are central to the development of new therapies and vaccine approaches. In particular, nonhuman primates like rhesus and cynomolgus macaques are highly relevant preclinical models given their physiological and immunological similarities to humans. Unfortunately, there remains a scarcity of macaque models of pandemic influenza with which to test novel antiviral modalities. Here, we demonstrate that even at the highest doses tested, 1918 influenza was not lethal in these two macaque species, suggesting that they are not ideal for the development and testing of novel pandemic influenza-specific vaccines and therapies. Therefore, other physiologically relevant nonhuman primate models of pandemic influenza are needed.


Sujet(s)
Sous-type H1N1 du virus de la grippe A , Vaccins antigrippaux , Grippe humaine , Animaux , Humains , Macaca fascicularis , Macaca mulatta
3.
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
4.
PLoS Pathog ; 18(3): e1010396, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35358290

RÉSUMÉ

The CCR5-specific antibody Leronlimab is being investigated as a novel immunotherapy that can suppress HIV replication with minimal side effects. Here we studied the virological and immunological consequences of Leronlimab in chronically CCR5-tropic HIV-1 infected humans (n = 5) on suppressive antiretroviral therapy (ART) and in ART-naïve acutely CCR5-tropic SHIV infected rhesus macaques (n = 4). All five human participants transitioned from daily combination ART to self-administered weekly subcutaneous (SC) injections of 350 mg or 700 mg Leronlimab and to date all participants have sustained virologic suppression for over seven years. In all participants, Leronlimab fully occupied CCR5 receptors on peripheral blood CD4+ T cells and monocytes. In ART-naïve rhesus macaques acutely infected with CCR5-tropic SHIV, weekly SC injections of 50 mg/kg Leronlimab fully suppressed plasma viremia in half of the macaques. CCR5 receptor occupancy by Leronlimab occurred concomitant with rebound of CD4+ CCR5+ T-cells in peripheral blood, and full CCR5 receptor occupancy was found in multiple anatomical compartments. Our results demonstrate that weekly, self-administered Leronlimab was safe, well-tolerated, and efficacious for long-term virologic suppression and should be included in the arsenal of safe, easily administered, longer-acting antiretroviral treatments for people living with HIV-1. Trial Registration: ClinicalTrials.gov Identifiers: NCT02175680 and NCT02355184.


Sujet(s)
Virus de l'immunodéficience simienne , Animaux , Anticorps monoclonaux humanisés/pharmacologie , Anticorps anti-VIH , Humains , Macaca mulatta , Récepteurs CCR5
5.
Front Immunol ; 12: 794638, 2021.
Article de Anglais | MEDLINE | ID: mdl-34868084

RÉSUMÉ

CCR5 plays a central role in infectious disease, host defense, and cancer progression, thereby making it an ideal target for therapeutic development. Notably, CCR5 is the major HIV entry co-receptor, where its surface density correlates with HIV plasma viremia. The level of CCR5 receptor occupancy (RO) achieved by a CCR5-targeting therapeutic is therefore a critical predictor of its efficacy. However, current methods to measure CCR5 RO lack sensitivity, resulting in high background and overcalculation. Here, we report on two independent, flow cytometric methods of calculating CCR5 RO using the anti-CCR5 antibody, Leronlimab. We show that both methods led to comparable CCR5 RO values, with low background on untreated CCR5+CD4+ T cells and sensitive measurements of occupancy on both blood and tissue-resident CD4+ T cells that correlated longitudinally with plasma concentrations in Leronlimab-treated macaques. Using these assays, we found that Leronlimab stabilized cell surface CCR5, leading to an increase in the levels of circulating and tissue-resident CCR5+CD4+ T cells in vivo in Leronlimab-treated macaques. Weekly Leronlimab treatment in a chronically SIV-infected macaque led to increased CCR5+CD4+ T cells levels and fully suppressed plasma viremia, both concomitant with full CCR5 RO on peripheral blood CD4+ T cells, demonstrating that CCR5+CD4+ T cells were protected from viral replication by Leronlimab binding. Finally, we extended these results to Leronlimab-treated humans and found that weekly 700 mg Leronlimab led to complete CCR5 RO on peripheral blood CD4+ T cells and a statistically significant increase in CCR5+CD4+ T cells in peripheral blood. Collectively, these results establish two RO calculation methods for longitudinal monitoring of anti-CCR5 therapeutic antibody blockade efficacy in both macaques and humans, demonstrate that CCR5+CD4+ T cell levels temporarily increase with Leronlimab treatment, and facilitate future detailed investigations into the immunological impacts of CCR5 inhibition in multiple pathophysiological processes.


Sujet(s)
Anticorps monoclonaux humanisés , Lymphocytes T CD4+ , Anticorps anti-VIH , Récepteurs CCR5 , Syndrome d'immunodéficience acquise du singe , Animaux , Femelle , Humains , Anticorps monoclonaux humanisés/usage thérapeutique , Numération des lymphocytes CD4 , Lymphocytes T CD4+/immunologie , Traitements médicamenteux de la COVID-19 , Cytométrie en flux , Anticorps anti-VIH/usage thérapeutique , Infections à VIH/traitement médicamenteux , Primates , Liaison aux protéines , Récepteurs CCR5/immunologie , Récepteurs CCR5/métabolisme , Syndrome d'immunodéficience acquise du singe/traitement médicamenteux , Résultat thérapeutique
6.
J Immunol ; 207(12): 2913-2921, 2021 12 15.
Article de Anglais | MEDLINE | ID: mdl-34810222

RÉSUMÉ

CD8+ T cells are key mediators of antiviral and antitumor immunity. The isolation and study of Ag-specific CD8+ T cells, as well as mapping of their MHC restriction, has practical importance to the study of disease and the development of therapeutics. Unfortunately, most experimental approaches are cumbersome, owing to the highly variable and donor-specific nature of MHC-bound peptide/TCR interactions. Here we present a novel system for rapid identification and characterization of Ag-specific CD8+ T cells, particularly well suited for samples with limited primary cells. Cells are stimulated ex vivo with Ag of interest, followed by live cell sorting based on surface-trapped TNF-α. We take advantage of major advances in single-cell sequencing to generate full-length sequence data from the paired TCR α- and ß-chains from these Ag-specific cells. The paired TCR chains are cloned into retroviral vectors and used to transduce donor CD8+ T cells. These TCR transductants provide a virtually unlimited experimental reagent, which can be used for further characterization, such as minimal epitope mapping or identification of MHC restriction, without depleting primary cells. We validated this system using CMV-specific CD8+ T cells from rhesus macaques, characterizing an immunodominant Mamu-A1*002:01-restricted epitope. We further demonstrated the utility of this system by mapping a novel HLA-A*68:02-restricted HIV Gag epitope from an HIV-infected donor. Collectively, these data validate a new strategy to rapidly identify novel Ags and characterize Ag-specific CD8+ T cells, with applications ranging from the study of infectious disease to immunotherapeutics and precision medicine.


Sujet(s)
Lymphocytes T CD8+ , Infections à VIH , Animaux , Épitopes , Déterminants antigéniques des lymphocytes T , Macaca mulatta , Récepteurs aux antigènes des cellules T , Facteur de nécrose tumorale alpha
7.
Nat Commun ; 12(1): 3343, 2021 06 07.
Article de Anglais | MEDLINE | ID: mdl-34099693

RÉSUMÉ

In the absence of a prophylactic vaccine, the use of antiretroviral therapy (ART) as pre-exposure prophylaxis (PrEP) to prevent HIV acquisition by uninfected individuals is a promising approach to slowing the epidemic, but its efficacy is hampered by incomplete patient adherence and ART-resistant variants. Here, we report that competitive inhibition of HIV Env-CCR5 binding via the CCR5-specific antibody Leronlimab protects rhesus macaques against infection following repeated intrarectal challenges of CCR5-tropic SHIVSF162P3. Injection of Leronlimab weekly at 10 mg/kg provides significant but partial protection, while biweekly 50 mg/kg provides complete protection from SHIV acquisition. Tissue biopsies from protected macaques post challenge show complete CCR5 receptor occupancy and an absence of viral nucleic acids. After Leronlimab washout, protected macaques remain aviremic, and adoptive transfer of hematologic cells into naïve macaques does not transmit viral infection. These data identify CCR5 blockade with Leronlimab as a promising approach to HIV prophylaxis and support initiation of clinical trials.


Sujet(s)
Récepteurs CCR5/métabolisme , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/transmission , Virus de l'immunodéficience simienne/immunologie , Animaux , Anticorps monoclonaux humanisés/pharmacologie , Lymphocytes T CD4+ , Lymphocytes T CD8+ , Femelle , Anticorps anti-VIH/pharmacologie , Infections à VIH , Humains , Macaca mulatta , Mâle , Muqueuse , Prophylaxie pré-exposition , Charge virale
8.
J Transl Autoimmun ; 4: 100083, 2021.
Article de Anglais | MEDLINE | ID: mdl-33521616

RÉSUMÉ

Coronavirus disease 2019 (COVID-19) is associated with considerable morbidity and mortality. The number of confirmed cases of infection with SARS-CoV-2, the virus causing COVID-19 continues to escalate with over 70 million confirmed cases and over 1.6 million confirmed deaths. Severe-to-critical COVID-19 is associated with a dysregulated host immune response to the virus, which is thought to lead to pathogenic immune dysregulation and end-organ damage. Presently few effective treatment options are available to treat COVID-19. Leronlimab is a humanized IgG4, kappa monoclonal antibody that blocks C-C chemokine receptor type 5 (CCR5). It has been shown that in patients with severe COVID-19 treatment with leronlimab reduces elevated plasma IL-6 and chemokine ligand 5 (CCL5), and normalized CD4/CD8 ratios. We administered leronlimab to 4 critically ill COVID-19 patients in intensive care. All 4 of these patients improved clinically as measured by vasopressor support, and discontinuation of hemodialysis and mechanical ventilation. Following administration of leronlimab there was a statistically significant decrease in IL-6 observed in patient A (p=0.034) from day 0-7 and patient D (p=0.027) from day 0-14. This corresponds to restoration of the immune function as measured by CD4+/CD8+ T cell ratio. Although two of the patients went on to survive the other two subsequently died of surgical complications after an initial recovery from SARS-CoV-2 infection.

9.
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
10.
Int J Infect Dis ; 103: 25-32, 2021 Feb.
Article de Anglais | MEDLINE | ID: mdl-33186704

RÉSUMÉ

OBJECTIVE: Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is now a global pandemic. Emerging results indicate a dysregulated immune response. Given the role of CCR5 in immune cell migration and inflammation, we investigated the impact of CCR5 blockade via the CCR5-specific antibody leronlimab on clinical, immunological, and virological parameters in severe COVID-19 patients. METHODS: In March 2020, 10 terminally ill, critical COVID-19 patients received two doses of leronlimab via individual emergency use indication. We analyzed changes in clinical presentation, immune cell populations, inflammation, as well as SARS-CoV-2 plasma viremia before and 14 days after treatment. RESULTS: Over the 14-day study period, six patients survived, two were extubated, and one discharged. We observed complete CCR5 receptor occupancy in all donors by day 7. Compared with the baseline, we observed a concomitant statistically significant reduction in plasma IL-6, restoration of the CD4/CD8 ratio, and resolution of SARS-CoV2 plasma viremia (pVL). Furthermore, the increase in the CD8 percentage was inversely correlated with the reduction in pVL (r = -0.77, p = 0.0013). CONCLUSIONS: Our study design precludes clinical efficacy inferences but the results implicate CCR5 as a therapeutic target for COVID-19 and they form the basis for ongoing randomized clinical trials.


Sujet(s)
Antagonistes des récepteurs CCR5/usage thérapeutique , Lymphocytes T CD8+/immunologie , Traitements médicamenteux de la COVID-19 , Cytokines/sang , ARN viral/sang , SARS-CoV-2 , Adulte , Sujet âgé , COVID-19/immunologie , COVID-19/virologie , Femelle , Humains , Mâle , Adulte d'âge moyen , Facteurs temps
11.
medRxiv ; 2020 May 05.
Article de Anglais | MEDLINE | ID: mdl-32511656

RÉSUMÉ

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), is now pandemic with nearly three million cases reported to date. Although the majority of COVID-19 patients experience only mild or moderate symptoms, a subset will progress to severe disease with pneumonia and acute respiratory distress syndrome (ARDS) requiring mechanical ventilation. Emerging results indicate a dysregulated immune response characterized by runaway inflammation, including cytokine release syndrome (CRS), as the major driver of pathology in severe COVID-19. With no treatments currently approved for COVID-19, therapeutics to prevent or treat the excessive inflammation in severe disease caused by SARS-CoV-2 infection are urgently needed. Here, in 10 terminally-ill, critical COVID-19 patients we report profound elevation of plasma IL-6 and CCL5 (RANTES), decreased CD8+ T cell levels, and SARS-CoV-2 plasma viremia. Following compassionate care treatment with the CCR5 blocking antibody leronlimab, we observed complete CCR5 receptor occupancy on macrophage and T cells, rapid reduction of plasma IL-6, restoration of the CD4/CD8 ratio, and a significant decrease in SARS-CoV-2 plasma viremia. Consistent with reduction of plasma IL-6, single-cell RNA-sequencing revealed declines in transcriptomic myeloid cell clusters expressing IL-6 and interferon-related genes. These results demonstrate a novel approach to resolving unchecked inflammation, restoring immunologic deficiencies, and reducing SARS-CoV-2 plasma viral load via disruption of the CCL5-CCR5 axis, and support randomized clinical trials to assess clinical efficacy of leronlimab-mediated inhibition of CCR5 for COVID-19.

12.
J Immunol ; 204(8): 2169-2176, 2020 04 15.
Article de Anglais | MEDLINE | ID: mdl-32161099

RÉSUMÉ

Currently 247 million people are living with chronic hepatitis B virus infection (CHB), and the development of novel curative treatments is urgently needed. Immunotherapy is an attractive approach to treat CHB, yet therapeutic approaches to augment the endogenous hepatitis B virus (HBV)-specific T cell response in CHB patients have demonstrated little success. In this study, we show that strain 68-1 rhesus macaque (RM) CMV vaccine vectors expressing HBV Ags engender HBV-specific CD8+ T cells unconventionally restricted by MHC class II and the nonclassical MHC-E molecule in RM. Surface staining of human donor and RM primary hepatocytes (PH) ex vivo revealed the majority of PH expressed MHC-E but not MHC class II. HBV-specific, MHC-E-restricted CD8+ T cells from RM vaccinated with RM CMV vaccine vectors expressing HBV Ags recognized HBV-infected PH from both human donor and RM. These results provide proof-of-concept that MHC-E-restricted CD8+ T cells could be harnessed for the treatment of CHB, either through therapeutic vaccination or adoptive immunotherapy.


Sujet(s)
Lymphocytes T CD8+/immunologie , Virus de l'hépatite B/immunologie , Hépatite B chronique/immunologie , Hépatocytes/immunologie , Antigènes d'histocompatibilité de classe I/immunologie , Animaux , Hépatite B chronique/virologie , Hépatocytes/virologie , Macaca mulatta
13.
PLoS Pathog ; 16(3): e1008339, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-32163523

RÉSUMÉ

Despite the success of antiretroviral therapy (ART) to halt viral replication and slow disease progression, this treatment is not curative and there remains an urgent need to develop approaches to clear the latent HIV reservoir. The human IL-15 superagonist N-803 (formerly ALT-803) is a promising anti-cancer biologic with potent immunostimulatory properties that has been extended into the field of HIV as a potential "shock and kill" therapeutic for HIV cure. However, the ability of N-803 to reactivate latent virus and modulate anti-viral immunity in vivo under the cover of ART remains undefined. Here, we show that in ART-suppressed, simian-human immunodeficiency virus (SHIV)SF162P3-infected rhesus macaques, subcutaneous administration of N-803 activates and mobilizes both NK cells and SHIV-specific CD8+ T cells from the peripheral blood to lymph node B cell follicles, a sanctuary site for latent virus that normally excludes such effector cells. We observed minimal activation of memory CD4+ T cells and no increase in viral RNA content in lymph node resident CD4+ T cells post N-803 administration. Accordingly, we found no difference in the number or magnitude of plasma viremia timepoints between treated and untreated animals during the N-803 administration period, and no difference in the size of the viral DNA cell-associated reservoir post N-803 treatment. These results substantiate N-803 as a potent immunotherapeutic candidate capable of activating and directing effector CD8+ T and NK cells to the B cell follicle during full ART suppression, and suggest N-803 must be paired with a bona fide latency reversing agent in vivo to facilitate immune-mediated modulation of the latent viral reservoir.


Sujet(s)
Antirétroviraux/administration et posologie , Lymphocytes B/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Infections à VIH/traitement médicamenteux , Interleukine-15/antagonistes et inhibiteurs , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Protéines/administration et posologie , Animaux , Lymphocytes B/cytologie , Lymphocytes B/immunologie , Lymphocytes T CD8+/cytologie , Lymphocytes T CD8+/immunologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Infections à VIH/génétique , Infections à VIH/immunologie , Infections à VIH/physiopathologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/physiologie , Humains , Interleukine-15/génétique , Interleukine-15/immunologie , Cellules tueuses naturelles/cytologie , Cellules tueuses naturelles/immunologie , Noeuds lymphatiques/effets des médicaments et des substances chimiques , Noeuds lymphatiques/immunologie , Macaca mulatta , Protéines de fusion recombinantes , Syndrome d'immunodéficience acquise du singe/traitement médicamenteux , Syndrome d'immunodéficience acquise du singe/immunologie , Syndrome d'immunodéficience acquise du singe/physiopathologie , Syndrome d'immunodéficience acquise du singe/virologie , Virus de l'immunodéficience simienne/effets des médicaments et des substances chimiques , Virus de l'immunodéficience simienne/physiologie , Latence virale/effets des médicaments et des substances chimiques
14.
Xenotransplantation ; 27(4): e12578, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-31930750

RÉSUMÉ

Allogeneic hematopoietic stem cell transplantation (HSCT) and xenotransplantation are accompanied by viral reactivations and virus-associated complications resulting from immune deficiency. Here, in a Mauritian cynomolgus macaque model of fully MHC-matched allogeneic HSCT, we report reactivations of cynomolgus polyomavirus, lymphocryptovirus, and cytomegalovirus, macaque viruses analogous to HSCT-associated human counterparts BK virus, Epstein-Barr virus, and human cytomegalovirus. Viral replication in recipient macaques resulted in characteristic disease manifestations observed in HSCT patients, such as polyomavirus-associated hemorrhagic cystitis and tubulointerstitial nephritis or lymphocryptovirus-associated post-transplant lymphoproliferative disorder. However, in most cases, the reconstituted immune system, alone or in combination with short-term pharmacological intervention, exerted control over viral replication, suggesting engraftment of functional donor-derived immunity. Indeed, the donor-derived reconstituted immune systems of two long-term engrafted HSCT recipient macaques responded to live attenuated yellow fever 17D vaccine (YFV 17D) indistinguishably from untransplanted controls, mounting 17D-targeted neutralizing antibody responses and clearing YFV 17D within 14 days. Together, these data demonstrate that this macaque model of allogeneic HSCT recapitulates clinical situations of opportunistic viral infections in transplant patients and provides a pre-clinical model to test novel prophylactic and therapeutic modalities.


Sujet(s)
Modèles animaux de maladie humaine , Transplantation de cellules souches hématopoïétiques , Infections opportunistes , Maladies virales , Allogreffes , Animaux , Transplantation de cellules souches hématopoïétiques/effets indésirables , Humains , Macaca fascicularis , Infections opportunistes/virologie
15.
J Immunol ; 200(1): 49-60, 2018 01 01.
Article de Anglais | MEDLINE | ID: mdl-29150562

RÉSUMÉ

MHC-E is a highly conserved nonclassical MHC class Ib molecule that predominantly binds and presents MHC class Ia leader sequence-derived peptides for NK cell regulation. However, MHC-E also binds pathogen-derived peptide Ags for presentation to CD8+ T cells. Given this role in adaptive immunity and its highly monomorphic nature in the human population, HLA-E is an attractive target for novel vaccine and immunotherapeutic modalities. Development of HLA-E-targeted therapies will require a physiologically relevant animal model that recapitulates HLA-E-restricted T cell biology. In this study, we investigated MHC-E immunobiology in two common nonhuman primate species, Indian-origin rhesus macaques (RM) and Mauritian-origin cynomolgus macaques (MCM). Compared to humans and MCM, RM expressed a greater number of MHC-E alleles at both the population and individual level. Despite this difference, human, RM, and MCM MHC-E molecules were expressed at similar levels across immune cell subsets, equivalently upregulated by viral pathogens, and bound and presented identical peptides to CD8+ T cells. Indeed, SIV-specific, Mamu-E-restricted CD8+ T cells from RM recognized antigenic peptides presented by all MHC-E molecules tested, including cross-species recognition of human and MCM SIV-infected CD4+ T cells. Thus, MHC-E is functionally conserved among humans, RM, and MCM, and both RM and MCM represent physiologically relevant animal models of HLA-E-restricted T cell immunobiology.


Sujet(s)
Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Antigènes d'histocompatibilité de classe I/métabolisme , Antigènes d'histocompatibilité/métabolisme , Cellules tueuses naturelles/immunologie , Syndrome d'immunodéficience acquise du singe/immunologie , Virus de l'immunodéficience simienne/immunologie , Animaux , Présentation d'antigène , Antigènes viraux/immunologie , Antigènes viraux/métabolisme , Cellules cultivées , Séquence conservée/génétique , Antigènes d'histocompatibilité/génétique , Antigènes d'histocompatibilité/immunologie , Antigènes d'histocompatibilité de classe I/génétique , Antigènes d'histocompatibilité de classe I/immunologie , Humains , Macaca fascicularis , Macaca mulatta , Modèles animaux , Peptides/immunologie , Peptides/métabolisme ,
16.
Nat Commun ; 8(1): 2146, 2017 12 15.
Article de Anglais | MEDLINE | ID: mdl-29247188

RÉSUMÉ

Hepatitis B virus (HBV) is a major global health concern, and the development of curative therapeutics is urgently needed. Such efforts are impeded by the lack of a physiologically relevant, pre-clinical animal model of HBV infection. Here, we report that expression of the HBV entry receptor, human sodium-taurocholate cotransporting polypeptide (hNTCP), on macaque primary hepatocytes facilitates HBV infection in vitro, where all replicative intermediates including covalently closed circular DNA (cccDNA) are present. Furthermore, viral vector-mediated expression of hNTCP on hepatocytes in vivo renders rhesus macaques permissive to HBV infection. These in vivo macaque HBV infections are characterized by longitudinal HBV DNA in serum, and detection of HBV DNA, RNA, and HBV core antigen (HBcAg) in hepatocytes. Together, these results show that expressing hNTCP on macaque hepatocytes renders them susceptible to HBV infection, thereby establishing a physiologically relevant model of HBV infection to study immune clearance and test therapeutic and curative approaches.


Sujet(s)
Virus de l'hépatite B/physiologie , Hépatocytes/métabolisme , Hépatocytes/virologie , Transporteurs d'anions organiques sodium-dépendants/métabolisme , Symporteurs/métabolisme , Animaux , Cellules cultivées , ADN viral/métabolisme , Hépatite B/génétique , Hépatite B/métabolisme , Hépatite B/virologie , Antigènes de la nucléocapside du virus de l'hépatite virale B/métabolisme , Virus de l'hépatite B/génétique , Virus de l'hépatite B/métabolisme , Hépatocytes/cytologie , Interactions hôte-pathogène , Humains , Macaca mulatta , Transporteurs d'anions organiques sodium-dépendants/génétique , ARN viral/métabolisme , Symporteurs/génétique
17.
Nat Commun ; 8(1): 1418, 2017 11 10.
Article de Anglais | MEDLINE | ID: mdl-29127275

RÉSUMÉ

Allogeneic hematopoietic stem cell transplantation (HSCT) is a critically important therapy for hematological malignancies, inborn errors of metabolism, and immunodeficiency disorders, yet complications such as graft-vs.-host disease (GvHD) limit survival. Development of anti-GvHD therapies that do not adversely affect susceptibility to infection or graft-vs.-tumor immunity are hampered by the lack of a physiologically relevant, preclinical model of allogeneic HSCT. Here we show a spectrum of diverse clinical HSCT outcomes including primary and secondary graft failure, lethal GvHD, and stable, disease-free full donor engraftment using reduced intensity conditioning and mobilized peripheral blood HSCT in unrelated, fully MHC-matched Mauritian-origin cynomolgus macaques. Anti-GvHD prophylaxis of tacrolimus, post-transplant cyclophosphamide, and CD28 blockade induces multi-lineage, full donor chimerism and recipient-specific tolerance while maintaining pathogen-specific immunity. These results establish a new preclinical allogeneic HSCT model for evaluation of GvHD prophylaxis and next-generation HSCT-mediated therapies for solid organ tolerance, cure of non-malignant hematological disease, and HIV reservoir clearance.


Sujet(s)
Transplantation de cellules souches hématopoïétiques/méthodes , Macaca fascicularis/immunologie , Complexe majeur d'histocompatibilité , Animaux , Femelle , Maladie du greffon contre l'hôte/prévention et contrôle , Test d'histocompatibilité , Humains , Macaca fascicularis/génétique , Mâle , Modèles animaux , Spécificité d'espèce , Chimère obtenue par transplantation/génétique , Chimère obtenue par transplantation/immunologie , Tolérance à la transplantation/génétique , Tolérance à la transplantation/immunologie , Transplantation homologue , Résultat thérapeutique
18.
Retrovirology ; 13: 6, 2016 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-26767784

RÉSUMÉ

BACKGROUND: Endogenous retroviruses (ERVs) are remnants of ancient retroviral infections that have invaded the germ line of both humans and non-human primates. Most ERVs are functionally crippled by deletions, mutations, and hypermethylation, leading to the view that they are inert genomic fossils. However, some ERVs can produce mRNA transcripts, functional viral proteins, and even non-infectious virus particles during certain developmental and pathological processes. While there have been reports of ERV-specific immunity associated with ERV activity in humans, adaptive immune responses to ERV-encoded gene products remain poorly defined and have not been investigated in the physiologically relevant non-human primate model of human disease. FINDINGS: Here, we identified the rhesus macaque equivalent of the biologically active human ERV-K (HML-2), simian ERV-K (SERV-K1), which retains intact open reading frames for both Gag and Env on chromosome 12 in the macaque genome. From macaque cells we isolated a spliced mRNA product encoding SERV-K1 Env, which possesses all the structural features of a canonical, functional retroviral Envelope protein. Furthermore, we identified rare, but robust T cell responses as well as frequent antibody responses targeting SERV-K1 Env in rhesus macaques. CONCLUSIONS: These data demonstrate that SERV-K1 retains biological activity sufficient to induce cellular and humoral immune responses in rhesus macaques. As ERV-K is the youngest and most active ERV family in the human genome, the identification and characterization of the simian orthologue in rhesus macaques provides a highly relevant animal model in which to study the role of ERV-K in developmental and disease states.


Sujet(s)
Anticorps antiviraux/sang , Rétrovirus endogènes/immunologie , Produits du gène env/immunologie , Lymphocytes T/immunologie , Animaux , Rétrovirus endogènes/génétique , Femelle , Produits du gène env/génétique , Macaca mulatta , Mâle , Données de séquences moléculaires , Analyse de séquence d'ADN
19.
PLoS Pathog ; 12(1): e1005349, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-26741490

RÉSUMÉ

HIV infection induces phenotypic and functional changes to CD8+ T cells defined by the coordinated upregulation of a series of negative checkpoint receptors that eventually result in T cell exhaustion and failure to control viral replication. We report that effector CD8+ T cells during HIV infection in blood and SIV infection in lymphoid tissue exhibit higher levels of the negative checkpoint receptor TIGIT. Increased frequencies of TIGIT+ and TIGIT+ PD-1+ CD8+ T cells correlated with parameters of HIV and SIV disease progression. TIGIT remained elevated despite viral suppression in those with either pharmacological antiretroviral control or immunologically in elite controllers. HIV and SIV-specific CD8+ T cells were dysfunctional and expressed high levels of TIGIT and PD-1. Ex-vivo single or combinational antibody blockade of TIGIT and/or PD-L1 restored viral-specific CD8+ T cell effector responses. The frequency of TIGIT+ CD4+ T cells correlated with the CD4+ T cell total HIV DNA. These findings identify TIGIT as a novel marker of dysfunctional HIV-specific T cells and suggest TIGIT along with other checkpoint receptors may be novel curative HIV targets to reverse T cell exhaustion.


Sujet(s)
Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Infections à VIH/immunologie , Récepteurs immunologiques/immunologie , Animaux , Antigène CD274/immunologie , Séparation cellulaire , ADN viral/analyse , Évolution de la maladie , Cytométrie en flux , Humains , Activation des lymphocytes/immunologie , Macaca mulatta , ARN viral/analyse , Syndrome d'immunodéficience acquise du singe/immunologie
20.
Science ; 351(6274): 714-20, 2016 Feb 12.
Article de Anglais | MEDLINE | ID: mdl-26797147

RÉSUMÉ

Major histocompatibility complex E (MHC-E) is a highly conserved, ubiquitously expressed, nonclassical MHC class Ib molecule with limited polymorphism that is primarily involved in the regulation of natural killer (NK) cells. We found that vaccinating rhesus macaques with rhesus cytomegalovirus vectors in which genes Rh157.5 and Rh157.4 are deleted results in MHC-E-restricted presentation of highly varied peptide epitopes to CD8αß(+) T cells, at ~4 distinct epitopes per 100 amino acids in all tested antigens. Computational structural analysis revealed that MHC-E provides heterogeneous chemical environments for diverse side-chain interactions within a stable, open binding groove. Because MHC-E is up-regulated to evade NK cell activity in cells infected with HIV, simian immunodeficiency virus, and other persistent viruses, MHC-E-restricted CD8(+) T cell responses have the potential to exploit pathogen immune-evasion adaptations, a capability that might endow these unconventional responses with superior efficacy.


Sujet(s)
Lymphocytes T CD8+/immunologie , Cytomegalovirus/immunologie , Déterminants antigéniques des lymphocytes T/immunologie , Antigènes d'histocompatibilité de classe I/immunologie , Virus de l'immunodéficience simienne/immunologie , Animaux , Présentation d'antigène , Variation des antigènes , Cytomegalovirus/génétique , Déterminants antigéniques des lymphocytes T/composition chimique , Vecteurs génétiques/génétique , Vecteurs génétiques/immunologie , Antigènes d'histocompatibilité de classe I/composition chimique , Interactions hôte-pathogène/immunologie , Échappement immunitaire , Cellules tueuses naturelles/immunologie , Macaca mulatta , Structure secondaire des protéines , Vaccination
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE