Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 24
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Sci Transl Med ; 14(670): eabn7336, 2022 11 09.
Article de Anglais | MEDLINE | ID: mdl-36350986

RÉSUMÉ

Chimeric antigen receptor (CAR) T cells have not induced meaningful clinical responses in solid tumors. Loss of T cell stemness, poor expansion capacity, and exhaustion during prolonged tumor antigen exposure are major causes of CAR T cell therapeutic resistance. Single-cell RNA-sequencing analysis of CAR T cells from a first-in-human trial in metastatic prostate cancer identified two independently validated cell states associated with antitumor potency or lack of efficacy. Low expression of PRDM1, encoding the BLIMP1 transcription factor, defined highly potent TCF7 [encoding T cell factor 1 (TCF1)]-expressing CD8+ CAR T cells, whereas enrichment of HAVCR2 [encoding T cell immunoglobulin and mucin-domain containing-3 (TIM-3)]-expressing CD8+ T cells with elevated PRDM1 was associated with poor outcomes. PRDM1 knockout promoted TCF7-dependent CAR T cell stemness and proliferation, resulting in marginally enhanced leukemia control in mice. However, in the setting of PRDM1 deficiency, a negative epigenetic feedback program of nuclear factor of activated T cells (NFAT)-driven T cell dysfunction was identified. This program was characterized by compensatory up-regulation of NR4A3 and other genes encoding exhaustion-related transcription factors that hampered T cell effector function in solid tumors. Dual knockout of PRDM1 and NR4A3 skewed CAR T cell phenotypes away from TIM-3+CD8+ and toward TCF1+CD8+ to counter exhaustion of tumor-infiltrating CAR T cells and improve antitumor responses, effects that were not achieved with PRDM1 and NR4A3 single knockout alone. These data underscore dual targeting of PRDM1 and NR4A3 as a promising approach to advance adoptive cell immuno-oncotherapy.


Sujet(s)
Tumeurs , Récepteurs aux stéroïdes , Mâle , Humains , Souris , Animaux , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Lymphocytes T CD8+ , Immunothérapie adoptive/méthodes , Récepteur cellulaire-2 du virus de l'hépatite A/génétique , Récepteur cellulaire-2 du virus de l'hépatite A/métabolisme , Tumeurs/anatomopathologie , Lignée cellulaire tumorale , Protéines de liaison à l'ADN/métabolisme , Récepteurs aux stéroïdes/métabolisme , Récepteurs des hormones thyroïdiennes/métabolisme , Protéines de tissu nerveux/métabolisme
2.
Mol Ther Oncolytics ; 20: 387-398, 2021 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-33614919

RÉSUMÉ

Metastatic medullary thyroid cancer (MTC) is a rare but often aggressive thyroid malignancy with a 5-year survival rate of less than 40% and few effective therapeutic options. Adoptive T cell immunotherapy using chimeric antigen receptor (CAR)-modified T cells (CAR Ts) is showing encouraging results in the treatment of cancer, but development is challenged by the availability of suitable target antigens. We identified glial-derived neurotrophic factor (GDNF) family receptor alpha 4 (GFRα4) as a putative antigen target for CAR-based therapy of MTC. We show that GFRα4 is highly expressed in MTC, in parafollicular cells within the thyroid from which MTC originates, and in normal thymus. We isolated two single-chain variable fragments (scFvs) targeting GFRα4 isoforms a and b by antibody phage display. CARs bearing the CD3ζ and the CD137 costimulatory domains were constructed using these GFRα4-specific scFvs. GFRα4-specific CAR Ts trigger antigen-dependent cytotoxicity and cytokine production in vitro, and they are able to eliminate tumors derived from the MTC TT cell line in an immunodeficient mouse xenograft model of MTC. These data demonstrate the feasibility of targeting GFRα4 by CAR T and support this antigen as a promising target for adoptive T cell immunotherapy and other antibody-based therapies for MTC.

4.
Mol Ther ; 28(2): 548-560, 2020 02 05.
Article de Anglais | MEDLINE | ID: mdl-31870622

RÉSUMÉ

The prognosis of patients diagnosed with advanced ovarian or endometrial cancer remains poor, and effective therapeutic strategies are limited. The Müllerian inhibiting substance type 2 receptor (MISIIR) is a transforming growth factor ß (TGF-ß) receptor family member, overexpressed by most ovarian and endometrial cancers while absent in most normal tissues. Restricted tissue expression, coupled with an understanding that MISIIR ligation transmits apoptotic signals to cancer cells, makes MISIIR an attractive target for tumor-directed therapeutics. However, the development of clinical MISIIR-targeted agents has been challenging. Prompted by the responses achieved in patients with blood malignancies using chimeric antigen receptor (CAR) T cell therapy, we hypothesized that MISIIR targeting may be achieved using a CAR T cell approach. Herein, we describe the development and evaluation of a CAR that targets MISIIR. T cells expressing the MISIIR-specific CAR demonstrated antigen-specific reactivity in vitro and eliminated MISIIR-overexpressing tumors in vivo. MISIIR CAR T cells also recognized a panel of human ovarian and endometrial cancer cell lines, and they lysed a battery of patient-derived tumor specimens in vitro, without mediating cytotoxicity of a panel of normal primary human cells. In conclusion, these results indicate that MISIIR targeting for the treatment of ovarian cancer and other gynecologic malignancies is achievable using CAR technology.


Sujet(s)
Tumeurs de l'appareil génital féminin/immunologie , Immunothérapie adoptive , Tumeurs de l'ovaire/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs peptidiques/immunologie , Récepteurs TGF-bêta/immunologie , Lymphocytes T/immunologie , Animaux , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Épitopes/génétique , Épitopes/immunologie , Femelle , Tumeurs de l'appareil génital féminin/thérapie , Humains , Souris , Tumeurs de l'ovaire/thérapie , Récepteurs chimériques pour l'antigène/métabolisme , Anticorps à chaîne unique/génétique , Anticorps à chaîne unique/immunologie , Lymphocytes T/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
5.
Front Immunol ; 9: 2740, 2018.
Article de Anglais | MEDLINE | ID: mdl-30559740

RÉSUMÉ

Chimeric antigen receptor (CAR)-engineered T cells represent a breakthrough in personalized medicine. In this strategy, a patient's own T lymphocytes are genetically reprogrammed to encode a synthetic receptor that binds a tumor antigen, allowing T cells to recognize and kill antigen-expressing cancer cells. As a result of complete and durable responses in individuals who are refractory to standard of care therapy, CAR T cells directed against the CD19 protein have been granted United States Food and Drug Administration (FDA) approval as a therapy for treatment of pediatric and young adult acute lymphoblastic leukemia and diffuse large B cell lymphoma. Human trials of CAR T cells targeting CD19 or B cell maturation antigen in multiple myeloma have also reported early successes. However, a clear and consistently reproducible demonstration of the clinical efficacy of CAR T cells in the setting of solid tumors has not been reported to date. Here, we review the history and status of CAR T cell therapy for solid tumors, potential T cell-intrinsic determinants of response and resistance as well as extrinsic obstacles to the success of this approach for much more prevalent non-hematopoietic malignancies. In addition, we summarize recent strategies and innovations that aim to augment the potency of CAR T cells in the face of multiple immunosuppressive barriers operative within the solid tumor microenvironment. Advances in the field of CAR T cell biology over the coming years in the areas of safety, reliability and efficacy against non-hematopoietic cancers will ultimately determine how transformative adoptive T cell therapy will be in the broader battle against cancer.


Sujet(s)
Tumeurs hématologiques/immunologie , Tumeurs hématologiques/thérapie , Récepteurs aux antigènes des cellules T/immunologie , Lymphocytes T/immunologie , Animaux , Antigènes CD19/immunologie , Thérapie cellulaire et tissulaire/méthodes , Humains , Microenvironnement tumoral/immunologie
6.
Mol Ther Oncolytics ; 11: 20-38, 2018 Dec 21.
Article de Anglais | MEDLINE | ID: mdl-30306125

RÉSUMÉ

We generated two humanized interleukin-13 receptor α2 (IL-13Rα2) chimeric antigen receptors (CARs), Hu07BBz and Hu08BBz, that recognized human IL-13Rα2, but not IL-13Rα1. Hu08BBz also recognized canine IL-13Rα2. Both of these CAR T cell constructs demonstrated superior tumor inhibitory effects in a subcutaneous xenograft model of human glioma compared with a humanized EGFRvIII CAR T construct used in a recent phase 1 clinical trial (ClinicalTrials.gov: NCT02209376). The Hu08BBz demonstrated a 75% reduction in orthotopic tumor growth using low-dose CAR T cell infusion. Using combination therapy with immune checkpoint blockade, humanized IL-13Rα2 CAR T cells performed significantly better when combined with CTLA-4 blockade, and humanized EGFRvIII CAR T cells' efficacy was improved by PD-1 and TIM-3 blockade in the same mouse model, which was correlated with the levels of checkpoint molecule expression in co-cultures with the same tumor in vitro. Humanized IL-13Rα2 CAR T cells also demonstrated benefit from a self-secreted anti-CTLA-4 minibody in the same mouse model. In addition to a canine glioma cell line (J3T), canine osteosarcoma lung cancer and leukemia cell lines also express IL-13Rα2 and were recognized by Hu08BBz. Canine IL-13Rα2 CAR T cell was also generated and tested in vitro by co-culture with canine tumor cells and in vivo in an orthotopic model of canine glioma. Based on these results, we are designing a pre-clinical trial to evaluate the safety of canine IL-13Rα2 CAR T cells in dog with spontaneous IL-13Rα2-positive glioma, which will help to inform a human clinical trial design for glioblastoma using humanized scFv-based IL-13Rα2 targeting CAR T cells.

7.
J Virol ; 92(21)2018 11 01.
Article de Anglais | MEDLINE | ID: mdl-30111569

RÉSUMÉ

Influenza virus outbreaks remain a serious threat to public health. A greater understanding of how cells targeted by the virus respond to the infection can provide insight into the pathogenesis of disease. Here we examined the transcriptional profile of in vivo-infected and uninfected type 2 alveolar epithelial cells (AEC) in the lungs of influenza virus-infected mice. We show for the first time the unique gene expression profiles induced by the in vivo infection of AEC as well as the transcriptional response of uninfected bystander cells. This work allows us to distinguish the direct and indirect effects of infection at the cellular level. Transcriptome analysis revealed that although directly infected and bystander AEC from infected animals shared many transcriptome changes compared to AEC from uninfected animals, directly infected cells produce more interferon and express lower levels of Wnt signaling-associated transcripts, while concurrently expressing more transcripts associated with cell death pathways, than bystander uninfected AEC. The Wnt signaling pathway was downregulated in both in vivo-infected AEC and in vitro-infected human lung epithelial A549 cells. Wnt signaling did not affect type I and III interferon production by infected A549 cells. Our results reveal unique transcriptional changes that occur within infected AEC and show that influenza virus downregulates Wnt signaling. In light of recent findings that Wnt signaling is essential for lung epithelial stem cells, our findings reveal a mechanism by which influenza virus may affect host lung repair.IMPORTANCE Influenza virus infection remains a major public health problem. Utilizing a recombinant green fluorescent protein-expressing influenza virus, we compared the in vivo transcriptomes of directly infected and uninfected bystander cells from infected mouse lungs and discovered many pathways uniquely regulated in each population. The Wnt signaling pathway was downregulated in directly infected cells and was shown to affect virus but not interferon production. Our study is the first to discern the in vivo transcriptome changes induced by direct viral infection compared to mere exposure to the lung inflammatory milieu and highlight the downregulation of Wnt signaling. This downregulation has important implications for understanding influenza virus pathogenesis, as Wnt signaling is critical for lung epithelial stem cells and lung epithelial cell differentiation. Our findings reveal a mechanism by which influenza virus may affect host lung repair and suggest interventions that prevent damage or accelerate recovery of the lung.


Sujet(s)
Pneumocytes/immunologie , Sous-type H1N1 du virus de la grippe A/immunologie , Infections à Orthomyxoviridae/immunologie , Muqueuse respiratoire/immunologie , Voie de signalisation Wnt/immunologie , Cellules A549 , Pneumocytes/virologie , Animaux , Lignée cellulaire , Chiens , Femelle , Analyse de profil d'expression de gènes , Humains , Interféron de type I/immunologie , Interférons/immunologie , Cellules rénales canines Madin-Darby , Souris , Souris de lignée C57BL , Infections à Orthomyxoviridae/virologie , Muqueuse respiratoire/cytologie , Muqueuse respiratoire/virologie , Transcriptome/génétique , Voie de signalisation Wnt/génétique , Interféron lambda
8.
Nat Med ; 24(5): 563-571, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-29713085

RÉSUMÉ

Tolerance to self-antigens prevents the elimination of cancer by the immune system1,2. We used synthetic chimeric antigen receptors (CARs) to overcome immunological tolerance and mediate tumor rejection in patients with chronic lymphocytic leukemia (CLL). Remission was induced in a subset of subjects, but most did not respond. Comprehensive assessment of patient-derived CAR T cells to identify mechanisms of therapeutic success and failure has not been explored. We performed genomic, phenotypic and functional evaluations to identify determinants of response. Transcriptomic profiling revealed that CAR T cells from complete-responding patients with CLL were enriched in memory-related genes, including IL-6/STAT3 signatures, whereas T cells from nonresponders upregulated programs involved in effector differentiation, glycolysis, exhaustion and apoptosis. Sustained remission was associated with an elevated frequency of CD27+CD45RO-CD8+ T cells before CAR T cell generation, and these lymphocytes possessed memory-like characteristics. Highly functional CAR T cells from patients produced STAT3-related cytokines, and serum IL-6 correlated with CAR T cell expansion. IL-6/STAT3 blockade diminished CAR T cell proliferation. Furthermore, a mechanistically relevant population of CD27+PD-1-CD8+ CAR T cells expressing high levels of the IL-6 receptor predicts therapeutic response and is responsible for tumor control. These findings uncover new features of CAR T cell biology and underscore the potential of using pretreatment biomarkers of response to advance immunotherapies.


Sujet(s)
Antigènes CD19/métabolisme , Immunothérapie adoptive , Leucémie chronique lymphocytaire à cellules B/immunologie , Leucémie chronique lymphocytaire à cellules B/thérapie , Récepteurs chimériques pour l'antigène/métabolisme , Animaux , Femelle , Interleukine-6/métabolisme , Mâle , Souris , Facteur de transcription STAT-3/métabolisme , Transcription génétique , Résultat thérapeutique
9.
Immunity ; 44(6): 1444-54, 2016 06 21.
Article de Anglais | MEDLINE | ID: mdl-27332733

RÉSUMÉ

Genetically modified T cells expressing chimeric antigen receptors (CARs) demonstrate robust responses against lineage restricted, non-essential targets in hematologic cancers. However, in solid tumors, the full potential of CAR T cell therapy is limited by the availability of cell surface antigens with sufficient cancer-specific expression. The majority of CAR targets have been normal self-antigens on dispensable hematopoietic tissues or overexpressed shared antigens. Here, we established that abnormal self-antigens can serve as targets for tumor rejection. We developed a CAR that recognized cancer-associated Tn glycoform of MUC1, a neoantigen expressed in a variety of cancers. Anti-Tn-MUC1 CAR T cells demonstrated target-specific cytotoxicity and successfully controlled tumor growth in xenograft models of T cell leukemia and pancreatic cancer. These findings demonstrate the therapeutic efficacy of CAR T cells directed against Tn-MUC1 and present aberrantly glycosylated antigens as a novel class of targets for tumor therapy with engineered T cells.


Sujet(s)
Adénocarcinome/thérapie , Déterminants antigéniques des lymphocytes T/immunologie , Immunothérapie/méthodes , Mucine-1/immunologie , Lymphocytes T/physiologie , Adénocarcinome/immunologie , Animaux , Lignée cellulaire tumorale , Cytotoxicité immunologique , Génie génétique , Glycosylation , Humains , Cellules Jurkat , Souris , Lignées consanguines de souris , Mucine-1/composition chimique , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/métabolisme , Protéines de fusion recombinantes/génétique , Protéines de fusion recombinantes/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
10.
J Immunol ; 196(6): 2602-13, 2016 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-26873987

RÉSUMÉ

Currently, there is little consensus regarding the most appropriate animal model to study acute infection and the virus-specific CD8(+) T cell (CTL) responses in neonates. TCRß high-throughput sequencing in naive CTL of differently aged neonatal mice was performed, which demonstrated differential Vß family gene usage. Using an acute influenza infection model, we examined the TCR repertoire of the CTL response in neonatal and adult mice infected with influenza type A virus. Three-day-old mice mounted a greatly reduced primary NP(366-374)-specific CTL response when compared with 7-d-old and adult mice, whereas secondary CTL responses were normal. Analysis of NP(366-374)-specific CTL TCR repertoire revealed different Vß gene usage and greatly reduced public clonotypes in 3-d-old neonates. This could underlie the impaired CTL response in these neonates. To directly test this, we examined whether controlling the TCR would restore neonatal CTL responses. We performed adoptive transfers of both nontransgenic and TCR-transgenic OVA(257-264)-specific (OT-I) CD8(+) T cells into influenza-infected hosts, which revealed that naive neonatal and adult OT-I cells expand equally well in neonatal and adult hosts. In contrast, nontransgenic neonatal CD8(+) T cells when transferred into adults failed to expand. We further demonstrate that differences in TCR avidity may contribute to decreased expansion of the endogenous neonatal CTL. These studies highlight the rapid evolution of the neonatal TCR repertoire during the first week of life and show that impaired neonatal CTL immunity results from an immature TCR repertoire, rather than intrinsic signaling defects or a suppressive environment.


Sujet(s)
Animaux nouveau-nés/immunologie , Lymphocytes T CD8+/immunologie , Lymphopoïèse/immunologie , Infections à Orthomyxoviridae/immunologie , Récepteur lymphocytaire T antigène, alpha-bêta/immunologie , Transfert adoptif , Animaux , Séparation cellulaire , Modèles animaux de maladie humaine , Cytométrie en flux , Séquençage nucléotidique à haut débit , Virus de la grippe A , Souris , Souris de lignée C57BL , Souris transgéniques
11.
J Immunol ; 196(3): 1186-98, 2016 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-26740110

RÉSUMÉ

The p110δ isoform of PI3K is known to play an important role in immunity, yet its contribution to CTL responses has not been fully elucidated. Using murine p110δ-deficient CD8(+) T cells, we demonstrated a critical role for the p110δ subunit in the generation of optimal primary and memory CD8(+) T cell responses. This was demonstrated in both acute viral and intracellular bacterial infections in mice. We show that p110δ signaling is required for CD8(+) T cell activation, proliferation and effector cytokine production. We provide evidence that the effects of p110δ signaling are mediated via Akt activation and through the regulation of TCR-activated oxidative phosphorylation and aerobic glycolysis. In light of recent clinical trials that employ drugs targeting p110δ in certain cancers and other diseases, our study suggests caution in using these drugs in patients, as they could potentially increase susceptibility to infectious diseases. These studies therefore reveal a novel and direct role for p110δ signaling in in vivo CD8(+) T cell immunity to microbial pathogens.


Sujet(s)
Infections bactériennes/enzymologie , Lymphocytes T CD8+/enzymologie , Activation des lymphocytes/immunologie , Phosphatidylinositol 3-kinases/immunologie , Maladies virales/enzymologie , Transfert adoptif , Animaux , Infections bactériennes/immunologie , Lymphocytes T CD8+/immunologie , Femelle , Cytométrie en flux , Mémoire immunologique/immunologie , Isoenzymes/immunologie , Souris , Souris knockout , Transduction du signal/immunologie , Maladies virales/immunologie
12.
Sci Transl Med ; 7(275): 275ra22, 2015 Feb 18.
Article de Anglais | MEDLINE | ID: mdl-25696001

RÉSUMÉ

Chimeric antigen receptors (CARs) are synthetic molecules designed to redirect T cells to specific antigens. CAR-modified T cells can mediate long-term durable remissions in B cell malignancies, but expanding this platform to solid tumors requires the discovery of surface targets with limited expression in normal tissues. The variant III mutation of the epidermal growth factor receptor (EGFRvIII) results from an in-frame deletion of a portion of the extracellular domain, creating a neoepitope. We chose a vector backbone encoding a second-generation CAR based on efficacy of a murine scFv-based CAR in a xenograft model of glioblastoma. Next, we generated a panel of humanized scFvs and tested their specificity and function as soluble proteins and in the form of CAR-transduced T cells; a low-affinity scFv was selected on the basis of its specificity for EGFRvIII over wild-type EGFR. The lead candidate scFv was tested in vitro for its ability to direct CAR-transduced T cells to specifically lyse, proliferate, and secrete cytokines in response to antigen-bearing targets. We further evaluated the specificity of the lead CAR candidate in vitro against EGFR-expressing keratinocytes and in vivo in a model of mice grafted with normal human skin. EGFRvIII-directed CAR T cells were also able to control tumor growth in xenogeneic subcutaneous and orthotopic models of human EGFRvIII(+) glioblastoma. On the basis of these results, we have designed a phase 1 clinical study of CAR T cells transduced with humanized scFv directed to EGFRvIII in patients with either residual or recurrent glioblastoma (NCT02209376).


Sujet(s)
Tumeurs du cerveau/thérapie , Récepteurs ErbB/immunologie , Glioblastome/thérapie , Immunothérapie , Récepteurs aux antigènes des cellules T/immunologie , Animaux , Modèles animaux de maladie humaine , Hétérogreffes , Humains , Souris
13.
Antimicrob Agents Chemother ; 58(12): 7056-71, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25224013

RÉSUMÉ

In the absence of universally available antiretroviral (ARV) drugs or a vaccine against HIV-1, microbicides may offer the most immediate hope for controlling the AIDS pandemic. The most advanced and clinically effective microbicides are based on ARV agents that interfere with the earliest stages of HIV-1 replication. Our objective was to identify and characterize novel ARV-like inhibitors, as well as demonstrate their efficacy at blocking HIV-1 transmission. Abasic phosphorothioate 2' deoxyribose backbone (PDB) oligomers were evaluated in a variety of mechanistic assays and for their ability to inhibit HIV-1 infection and virus transmission through primary human cervical mucosa. Cellular and biochemical assays were used to elucidate the antiviral mechanisms of action of PDB oligomers against both lab-adapted and primary CCR5- and CXCR4-utilizing HIV-1 strains, including a multidrug-resistant isolate. A polarized cervical organ culture was used to test the ability of PDB compounds to block HIV-1 transmission to primary immune cell populations across ectocervical tissue. The antiviral activity and mechanisms of action of PDB-based compounds were dependent on oligomer size, with smaller molecules preventing reverse transcription and larger oligomers blocking viral entry. Importantly, irrespective of molecular size, PDBs potently inhibited virus infection and transmission within genital tissue samples. Furthermore, the PDB inhibitors exhibited excellent toxicity and stability profiles and were found to be safe for vaginal application in vivo. These results, coupled with the previously reported intrinsic anti-inflammatory properties of PDBs, support further investigations in the development of PDB-based topical microbicides for preventing the global spread of HIV-1.


Sujet(s)
Col de l'utérus/effets des médicaments et des substances chimiques , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Oligonucléotides phosphorothioates/pharmacologie , Inhibiteurs de la transcriptase inverse/pharmacologie , Transcription inverse/effets des médicaments et des substances chimiques , Pénétration virale/effets des médicaments et des substances chimiques , Animaux , Col de l'utérus/virologie , Désoxyribose/composition chimique , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/virologie , Femelle , Expression des gènes , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/enzymologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/génétique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/croissance et développement , Humains , Mâle , Souris , Souris de lignée C57BL , Muqueuse/effets des médicaments et des substances chimiques , Muqueuse/virologie , Techniques de culture d'organes , Oligonucléotides phosphorothioates/synthèse chimique , Récepteurs CCR5/génétique , Récepteurs CCR5/métabolisme , Récepteurs CXCR4/antagonistes et inhibiteurs , Inhibiteurs de la transcriptase inverse/synthèse chimique , Mobilité des spermatozoïdes/effets des médicaments et des substances chimiques , Relation structure-activité , Vagin/effets des médicaments et des substances chimiques , Vagin/virologie
14.
PLoS Pathog ; 9(10): e1003658, 2013.
Article de Anglais | MEDLINE | ID: mdl-24130482

RÉSUMÉ

The role of Type I interferon (IFN) during pathogenic HIV and SIV infections remains unclear, with conflicting observations suggesting protective versus immunopathological effects. We therefore examined the effect of IFNα/ß on T cell death and viremia in HIV infection. Ex vivo analysis of eight pro- and anti-apoptotic molecules in chronic HIV-1 infection revealed that pro-apoptotic Bak was increased in CD4+ T cells and correlated directly with sensitivity to CD95/Fas-mediated apoptosis and inversely with CD4+ T cell counts. Apoptosis sensitivity and Bak expression were primarily increased in effector memory T cells. Knockdown of Bak by RNA interference inhibited CD95/Fas-induced death of T cells from HIV-1-infected individuals. In HIV-1-infected patients, IFNα-stimulated gene expression correlated positively with ex vivo T cell Bak levels, CD95/Fas-mediated apoptosis and viremia and negatively with CD4+ T cell counts. In vitro IFNα/ß stimulation enhanced Bak expression, CD95/Fas expression and CD95/Fas-mediated apoptosis in healthy donor T cells and induced death of HIV-specific CD8+ T cells from HIV-1-infected patients. HIV-1 in vitro sensitized T cells to CD95/Fas-induced apoptosis and this was Toll-like receptor (TLR)7/9- and Type I IFN-dependent. This sensitization by HIV-1 was due to an indirect effect on T cells, as it occurred in peripheral blood mononuclear cell cultures but not purified CD4+ T cells. Finally, peak IFNα levels and viral loads correlated negatively during acute SIV infection suggesting a potential antiviral effect, but positively during chronic SIV infection indicating that either the virus drives IFNα production or IFNα may facilitate loss of viral control. The above findings indicate stage-specific opposing effects of Type I IFNs during HIV-1 infection and suggest a novel mechanism by which these cytokines contribute to T cell depletion, dysregulation of cellular immunity and disease progression.


Sujet(s)
Apoptose/immunologie , Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Infections à VIH/immunologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/immunologie , Interféron alpha/immunologie , Interféron bêta/immunologie , Régulation positive/immunologie , Protéine Bak/immunologie , Adolescent , Adulte , Animaux , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD4+/anatomopathologie , Lymphocytes T CD8+/métabolisme , Lymphocytes T CD8+/anatomopathologie , Enfant , Enfant d'âge préscolaire , Femelle , Infections à VIH/métabolisme , Infections à VIH/anatomopathologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/métabolisme , Humains , Immunité cellulaire , Nourrisson , Interféron alpha/métabolisme , Interféron bêta/métabolisme , Macaca mulatta , Mâle , Récepteur de type Toll-7/immunologie , Récepteur de type Toll-7/métabolisme , Récepteur-9 de type Toll-like/immunologie , Récepteur-9 de type Toll-like/métabolisme , Charge virale/immunologie , Virémie/immunologie , Virémie/métabolisme , Protéine Bak/biosynthèse , Antigènes CD95/immunologie , Antigènes CD95/métabolisme
15.
Nat Immunol ; 14(6): 593-602, 2013 Jun.
Article de Anglais | MEDLINE | ID: mdl-23603793

RÉSUMÉ

We found upregulation of expression of the microRNA miR-155 in primary effector and effector memory CD8(+) T cells, but low miR-155 expression in naive and central memory cells. Antiviral CD8(+) T cell responses and viral clearance were impaired in miR-155-deficient mice, and this defect was intrinsic to CD8(+) T cells, as miR-155-deficient CD8(+) T cells mounted greatly diminished primary and memory responses. Conversely, miR-155 overexpression augmented antiviral CD8(+) T cell responses in vivo. Gene-expression profiling showed that miR-155-deficient CD8(+) T cells had enhanced type I interferon signaling and were more susceptible to interferon's antiproliferative effect. Inhibition of the type I interferon-associated transcription factors STAT1 or IRF7 resulted in enhanced responses of miR-155-deficient CD8(+) T cells in vivo. We have thus identified a previously unknown role for miR-155 in regulating responsiveness to interferon and CD8(+) T cell responses to pathogens in vivo.


Sujet(s)
Lymphocytes T CD8+/immunologie , Interférons/immunologie , microARN/immunologie , Transduction du signal/immunologie , Animaux , Lymphocytes T CD8+/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cytométrie en flux , Analyse de profil d'expression de gènes , Réseaux de régulation génique/génétique , Réseaux de régulation génique/immunologie , Immunotransfert , Mémoire immunologique/génétique , Mémoire immunologique/immunologie , Facteur-7 de régulation d'interféron/génétique , Facteur-7 de régulation d'interféron/immunologie , Facteur-7 de régulation d'interféron/métabolisme , Interférons/métabolisme , Interférons/pharmacologie , Souris , Souris de lignée C57BL , Souris knockout , microARN/génétique , microARN/métabolisme , Séquençage par oligonucléotides en batterie , Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/génétique , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Interférence par ARN , RT-PCR , Facteur de transcription STAT-1/génétique , Facteur de transcription STAT-1/immunologie , Facteur de transcription STAT-1/métabolisme , Transduction du signal/génétique , Protéine-1 suppressive de la signalisation des cytokines , Protéines SOCS/génétique , Protéines SOCS/immunologie , Protéines SOCS/métabolisme
16.
Arthritis Rheum ; 63(11): 3563-74, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-21792841

RÉSUMÉ

OBJECTIVE: Systemic sclerosis (SSc) is a chronic idiopathic disease of unknown etiology that is characterized by fibrosis of the skin and visceral organs mediated by activated myofibroblasts. The recently identified inflammasomes are cytosolic receptors that tightly regulate the activity of caspase 1 and downstream signaling molecules such as interleukin-1ß (IL-1ß) and IL-18. The nucleotide-binding oligomerization domain (NOD)-like receptor 3 (NLRP3) inflammasome has been implicated in the development of idiopathic pulmonary fibrosis. This study was undertaken to assess the role of the inflammasome in SSc-related dermal or pulmonary fibrosis. METHODS: Inflammasome gene transcripts were assayed in fibroblasts obtained from patients with SSc. Caspase 1 activation in SSc primary dermal and lung fibroblasts was inhibited, and the levels of hydroxyproline, COL1A1, COL3A1, IL-1ß, IL-18, and α-smooth muscle actin (α-SMA) were measured. The role of the inflammasome in dermal fibrosis was investigated in NLRP3(-/-) and ASC(-/-) mice. RESULTS: We identified increased expression of 40 genes associated with the inflammasome or downstream signaling molecules in SSc fibroblasts. Inhibition of caspase 1 in SSc dermal and lung fibroblasts abrogated the secretion of collagens, IL-1ß, and IL-18. In addition, we observed decreased expression of the myofibroblast protein α-SMA in SSc dermal fibroblasts treated with a caspase 1 inhibitor. Furthermore, NLRP3(-/-) mice and ASC(-/-) mice were resistant to bleomycin-induced skin fibrosis, which suggests a key role for the inflammasome in in vivo fibrosis. CONCLUSION: Innate immune signaling contributes to SSc fibrosis via activation of the inflammasome and caspase 1. These results suggest that inflammasome activation may play an important role in the pathogenesis of SSc.


Sujet(s)
Caspase-1/métabolisme , Inflammasomes/métabolisme , Myofibroblastes/métabolisme , Sclérodermie systémique/immunologie , Peau/immunologie , Animaux , Caspase-1/génétique , Différenciation cellulaire , Fibrose , Humains , Immunité innée , Inflammasomes/génétique , Souris , Souris knockout , Myofibroblastes/anatomopathologie , Sclérodermie systémique/anatomopathologie , Transduction du signal/physiologie , Peau/anatomopathologie
17.
J Immunol ; 186(8): 4599-608, 2011 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-21389258

RÉSUMÉ

Although much is known about the initiation of immune responses, much less is known about what controls the effector phase. CD8(+) T cell responses are believed to be programmed in lymph nodes during priming without any further contribution by dendritic cells (DCs) and Ag. In this study, we report the requirement for DCs, Ag, and CD28 costimulation during the effector phase of the CD8(+) T cell response. Depleting DCs or blocking CD28 after day 6 of primary influenza A virus infection decreases the virus-specific CD8(+) T cell response by inducing apoptosis, and this results in decreased viral clearance. Furthermore, effector CD8(+) T cells adoptively transferred during the effector phase fail to expand without DC, CD28 costimulation, and cognate Ag. The absence of costimulation also leads to reduced survival of virus-specific effector cells as they undergo apoptosis mediated by the proapoptotic molecule Bim. Finally, IL-2 treatment restored the effector response in the absence of CD28 costimulation. Thus, in contrast to naive CD8(+) T cells, which undergo an initial Ag-independent proliferation, effector CD8(+) T cells expanding in the lungs during the effector phase require Ag, CD28 costimulation, and DCs for survival and expansion. These requirements would greatly impair effector responses against viruses and tumors that are known to inhibit DC maturation and in chronic infections and aging where CD28(-/-) CD8(+) T cells accumulate.


Sujet(s)
Antigène CD28/immunologie , Lymphocytes T CD8+/immunologie , Cellules dendritiques/immunologie , Virus de la grippe A/immunologie , Infections à Orthomyxoviridae/immunologie , Animaux , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Apoptose/immunologie , Antigène CD80/immunologie , Antigène CD80/métabolisme , Antigène CD86/immunologie , Antigène CD86/métabolisme , Lymphocytes T CD8+/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/immunologie , Cellules dendritiques/métabolisme , Cytométrie en flux , Poumon/immunologie , Poumon/métabolisme , Poumon/virologie , Souris , Souris de souche-129 , Souris de lignée C57BL , Infections à Orthomyxoviridae/virologie , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/immunologie , Facteurs temps
18.
Vaccine ; 29(2): 314-22, 2010 Dec 16.
Article de Anglais | MEDLINE | ID: mdl-21034826

RÉSUMÉ

Current influenza virus vaccines primarily elicit antibodies and can be rendered ineffective by antigenic drift and shift. Vaccines that elicit CD8+ T cell responses targeting less variable proteins may function as universal vaccines that have broad reactivity against different influenza virus strains. To generate such a universal vaccine, we encapsulated live influenza virus in a biopolymer and delivered it to mice subcutaneously. This vaccine was safe, induced potent CD8+ T cell immunity and protected mice against heterosubtypic lethal challenge. Safety of subcutaneous (SQ) vaccination was tested in Rag-/-γc-/- double knockout mice which we show cannot control intranasal infection. Biopolymer encapsulation of live influenza virus could be used to develop universal CD8+ T cell vaccines against heterosubtypic and pandemic strains.


Sujet(s)
Biopolymères/administration et posologie , Vecteurs de médicaments/administration et posologie , Vaccins antigrippaux/immunologie , Orthomyxoviridae/immunologie , Lymphocytes T/immunologie , Animaux , Modèles animaux de maladie humaine , Femelle , Protéines foetales , Vaccins antigrippaux/administration et posologie , Vaccins antigrippaux/effets indésirables , Injections sous-cutanées , Souris , Souris de lignée C57BL , Souris knockout , Infections à Orthomyxoviridae/prévention et contrôle , Protéines à domaine boîte-T , Vaccins
19.
Antimicrob Agents Chemother ; 54(10): 4064-73, 2010 Oct.
Article de Anglais | MEDLINE | ID: mdl-20625151

RÉSUMÉ

Topical microbicides may prove to be an important strategy for preventing human immunodeficiency virus type 1 (HIV-1) transmission. We examined the safety and efficacy of sequence-nonspecific phosphorothioate 2' deoxyribose oligomers as potential novel microbicides. A short, 13-mer poly(T) phosphorothioate oligodeoxynucleotide (OPB-T) significantly inhibited infection of primary peripheral blood mononuclear cells (PBMC) by high-titer HIV-1(Ba-L) and simian immunodeficiency virus mac251 (SIV(mac251)). Continuous exposure of human vaginal and foreskin tissue explants to OPB-T showed no toxicity. An abasic 14-mer phosphorothioate 2' deoxyribose backbone (PDB) demonstrated enhanced anti-HIV-1 activity relative to OPB-T and other homo-oligodeoxynucleotide analogs. When PDB was used to pretreat HIV-1, PDB was effective against R5 and X4 isolates at a half-maximal inhibitory concentration (IC(50)) of <1 µM in both PBMC and P4-R5 MAGI cell infections. PDB also reduced HIV-1 infectivity following the binding of virus to target cells. This novel topical microbicide candidate exhibited an excellent in vitro safety profile in human PBMC and endocervical epithelial cells. PDB also retained activity in hydroxyethylcellulose gel at pH 4.4 and after transition to a neutral pH and was stable in this formulation for 30 days at room temperature. Furthermore, the compound displayed potent antiviral activity following incubation with a Lactobacillus strain derived from normal vaginal flora. Most importantly, PDB can inhibit HIV-1-induced alpha interferon production. Phosphorothioate 2' deoxyribose oligomers may therefore be promising microbicide candidates that inhibit HIV-1 infection and also dampen the inflammation which is critical for the initial spread of the virus.


Sujet(s)
Anti-infectieux/pharmacologie , Infections à VIH/prévention et contrôle , Oligonucléotides/pharmacologie , Récepteur de type Toll-7/métabolisme , Récepteur-9 de type Toll-like/métabolisme , Anti-infectieux/effets indésirables , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Humains , Techniques in vitro , Interféron alpha/métabolisme , Oligonucléotides/effets indésirables
20.
J Immunol ; 182(11): 6697-708, 2009 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-19454664

RÉSUMÉ

The failure of CD8(+) T cells to respond to chronic infection has been termed "exhaustion" and describes the condition in which CD8(+) T cells exhibit reduced differentiation, proliferation, and effector function. CD8(+) T cell exhaustion has been extensively studied in the murine model of chronic infection, lymphocytic choriomeningitis virus (LCMV). Although LCMV-based studies have yielded many interesting findings, they have not allowed for discrimination between the roles of cytokine- and Ag-driven exhaustion. We have created a system of chronic Ag stimulation using murine influenza A virus that leads to exhaustion and functional disability of virus-specific CD8(+) T cells, in the absence of high viral titers, sustained proinflammatory cytokine production and lymphocyte infection. Our findings show that Ag alone is sufficient to drive CD8(+) T cell impairment, that Ag-driven loss of virus-specific CD8(+) T cells is TRAIL mediated, and that removal of Ag reverses exhaustion. Although programmed death 1 was up-regulated on chronic Ag-stimulated CD8(+) T cells, it played no role in the exhaustion. These findings provide a novel insight into the mechanisms that control functional exhaustion of CD8(+) T cells in chronic infection.


Sujet(s)
Lymphocytes T CD8+/anatomopathologie , Infections/immunologie , Animaux , Antigènes viraux/immunologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/virologie , Maladie chronique , Virus de la grippe A/immunologie , Souris , Souris de lignée C57BL , Souris transgéniques , Ligand TRAIL
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...