Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cell Rep ; 43(3): 113879, 2024 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-38416647

RÉSUMÉ

Maintenance of CD4 T cells during chronic infections is vital for limiting pathogen burden and disease recrudescence. Although inhibitory receptor expression by CD4 T cells is commonly associated with immune suppression and exhaustion, such cell-intrinsic mechanisms that control activation are also associated with cell survival. Using a mouse model of visceral leishmaniasis (VL), we discovered a subset of lymphocyte activation gene 3 (LAG-3)-expressing CD4 T cells that co-express CXCR5. Although LAG3+CXCR5+ CD4 T cells are present in naive mice, they expand during VL. These cells express gene signatures associated with self-renewal capacity, suggesting progenitor-like properties. When transferred into Rag1-/- mice, these LAG3+CXCR5+ CD4 T cells differentiated into multiple effector types upon Leishmania donovani infection. The transcriptional repressor B cell lymphoma-6 was partially required for their maintenance. Altogether, we propose that the LAG3+CXCR5+ CD4 T cell subset could play a role in maintaining CD4 T cell responses during persistent infections.


Sujet(s)
Lymphocytes T CD4+ , Leishmaniose viscérale , Humains , Sous-populations de lymphocytes T , Facteurs de transcription , Récepteurs CXCR5
2.
Antimicrob Agents Chemother ; 67(2): e0133122, 2023 02 16.
Article de Anglais | MEDLINE | ID: mdl-36700643

RÉSUMÉ

Dengue virus (DENV) is a Flavivirus that causes the most prevalent arthropod-borne viral disease. Clinical manifestation of DENV infection ranges from asymptomatic to severe symptoms that can lead to death. Unfortunately, no antiviral treatments against DENV are currently available. In order to identify novel DENV inhibitors, we screened a library of 1,604 chemically diversified fragment-based compounds using DENV reporter viruses that allowed quantification of viral replication in infected cells. Following a validation screening, the two best inhibitor candidates were N-phenylpyridine-3-carboxamide (NPP3C) and 6-acetyl-1H-indazole (6A1HI). The half maximal effective concentration of NPP3C and 6A1H1 against DENV were 7.1 µM and 6.5 µM, respectively. 6A1H1 decreased infectious DENV particle production up to 1,000-fold without any cytotoxicity at the used concentrations. While 6A1HI was DENV-specific, NPP3C also inhibited the replication of other flaviviruses such as West Nile virus and Zika virus. Structure-activity relationship (SAR) studies with 151 analogues revealed key structural elements of NPP3C and 6A1HI required for their antiviral activity. Time-of-drug-addition experiments identified a postentry step as a target of these compounds. Consistently, using a DENV subgenomic replicon, we demonstrated that these compounds specifically impede the viral RNA replication step and exhibit a high genetic barrier-to-resistance. In contrast, viral RNA translation and the de novo biogenesis of DENV replication organelles were not affected. Overall, our data unveil NPP3C and 6A1H1 as novel DENV inhibitors. The information revealed by our SAR studies will help chemically optimize NPP3C and 6A1H1 in order to improve their anti-flaviviral potency and to challenge them in in vivo models.


Sujet(s)
Virus de la dengue , Dengue , Flavivirus , Infection par le virus Zika , Virus Zika , Animaux , Humains , Antiviraux/pharmacologie , Antiviraux/usage thérapeutique , Dengue/traitement médicamenteux , Virus de la dengue/génétique , Étapes du cycle de vie , Réplication de l'ARN , ARN viral/génétique , Réplication virale , Virus Zika/génétique , ARN subgénomique/génétique
3.
Virology ; 567: 77-86, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-35032866

RÉSUMÉ

Type-I interferon (IFN-I) signals exert a critical role in disease progression during viral infections. However, the immunomodulatory mechanisms by which IFN-I dictates disease outcomes remain to be fully defined. Here we report that IFN-I signals mediate thymic atrophy in viral infections, with more severe and prolonged loss of thymic output and unique kinetics and subtypes of IFN-α/ß expression in chronic infection compared to acute infection. Loss of thymic output was linked to inhibition of early stages of thymopoiesis (DN1-DN2 transition, and DN3 proliferation) and pronounced apoptosis during the late DP stage. Notably, infection-associated thymic defects were largely abrogated upon ablation of IFNαßR and partially mitigated in the absence of CD8 T cells, thus implicating direct as well as indirect effects of IFN-I on thymocytes. These findings provide mechanistic underpinnings for immunotherapeutic strategies targeting IFN-1 signals to manipulate disease outcomes during chronic infections and cancers.


Sujet(s)
Atrophie/virologie , Interféron alpha/immunologie , Interféron bêta/immunologie , Chorioméningite lymphocytaire/virologie , Virus de la chorioméningite lymphocytaire/immunologie , Thymocytes/virologie , Thymus (glande)/virologie , Animaux , Atrophie/génétique , Atrophie/immunologie , Atrophie/anatomopathologie , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/virologie , Maladie chronique , Femelle , Régulation de l'expression des gènes , Humains , Mémoire immunologique , Interféron alpha/génétique , Interféron bêta/génétique , Noeuds lymphatiques/immunologie , Noeuds lymphatiques/anatomopathologie , Noeuds lymphatiques/virologie , Déplétion lymphocytaire , Chorioméningite lymphocytaire/génétique , Chorioméningite lymphocytaire/immunologie , Chorioméningite lymphocytaire/anatomopathologie , Virus de la chorioméningite lymphocytaire/pathogénicité , Souris , Souris de lignée C57BL , Souris knockout , Récepteur à l'interféron alpha-bêta/déficit , Récepteur à l'interféron alpha-bêta/génétique , Récepteur à l'interféron alpha-bêta/immunologie , Transduction du signal/immunologie , Analyse sur cellule unique , Thymocytes/immunologie , Thymocytes/anatomopathologie , Thymus (glande)/immunologie , Thymus (glande)/anatomopathologie
4.
J Immunol ; 200(12): 4102-4116, 2018 06 15.
Article de Anglais | MEDLINE | ID: mdl-29712774

RÉSUMÉ

Macrophages represent one of the first lines of defense during infections and are essential for resolution of inflammation following pathogen clearance. Rapid activation or suppression of protein synthesis via changes in translational efficiency allows cells of the immune system, including macrophages, to quickly respond to external triggers or cues without de novo mRNA synthesis. The translational repressors eIF4E-binding proteins 4E-BP1 and 4E-BP2 (4E-BP1/2) are central regulators of proinflammatory cytokine synthesis during viral and parasitic infections. However, it remains to be established whether 4E-BP1/2 play a role in translational control of anti-inflammatory responses. By comparing translational efficiencies of immune-related transcripts in macrophages from wild-type and 4E-BP1/2 double-knockout mice, we found that translation of mRNAs encoding two major regulators of inflammation, IL-10 and PG-endoperoxide synthase 2/cyclooxygenase-2, is controlled by 4E-BP1/2. Genetic deletion of 4E-BP1/2 in macrophages increased endogenous IL-10 and PGE2 protein synthesis in response to TLR4 stimulation and reduced their bactericidal capacity. The molecular mechanism involves enhanced anti-inflammatory gene expression (sIl1ra, Nfil3, Arg1, Serpinb2) owing to upregulation of IL-10-STAT3 and PGE2-C/EBPß signaling. These data provide evidence that 4E-BP1/2 limit anti-inflammatory responses in macrophages and suggest that dysregulated activity of 4E-BP1/2 might be involved in reprogramming of the translational and downstream transcriptional landscape of macrophages during pathological conditions, such as infections and cancer.


Sujet(s)
Protéines de transport/métabolisme , Cyclooxygenase 2/métabolisme , Facteurs d'initiation eucaryotes/métabolisme , Inflammation/métabolisme , Interleukine-10/métabolisme , Macrophages/métabolisme , Phosphoprotéines/métabolisme , Protéines adaptatrices de la transduction du signal , Animaux , Protéines du cycle cellulaire , Dinoprostone/métabolisme , Expression des gènes/physiologie , Souris , Souris de lignée C57BL , Souris knockout , Liaison aux protéines/physiologie , Biosynthèse des protéines/physiologie , ARN messager/métabolisme , Protéines de répression/métabolisme , Transduction du signal/physiologie , Transcription génétique/physiologie , Régulation positive/physiologie
5.
Trends Parasitol ; 34(2): 155-171, 2018 02.
Article de Anglais | MEDLINE | ID: mdl-29089182

RÉSUMÉ

B cells are notorious actors for the host's protection against several infectious diseases. So much so that early vaccinology seated its principles upon their long-term protective antibody secretion capabilities. Indeed, there are many examples of acute infectious diseases that are combated by functional humoral responses. However, some chronic infectious diseases actively induce immune deregulations that often lead to defective, if not deleterious, humoral immune responses. In this review we summarize how Leishmania and Trypanosoma spp. directly manipulate B cell responses to induce polyclonal B cell activation, hypergammaglobulinemia, low-specificity antibodies, limited B cell survival, and regulatory B cells, contributing therefore to immunopathology and the establishment of persistent infections.


Sujet(s)
Lymphocytes B/immunologie , Lymphocytes B/parasitologie , Interactions hôte-parasite/immunologie , Leishmania/immunologie , Leishmaniose/immunologie , Trypanosoma/immunologie , Trypanosomiase/immunologie , Maladie chronique , Humains , Leishmaniose/parasitologie , Trypanosomiase/parasitologie
6.
PLoS Pathog ; 13(9): e1006616, 2017 Sep.
Article de Anglais | MEDLINE | ID: mdl-28892492

RÉSUMÉ

Leishmania donovani is known to induce myelopoiesis and to dramatically increase extramedullary myelopoiesis. This results in splenomegaly, which is then accompanied by disruption of the splenic microarchitecture, a chronic inflammatory environment, and immunosuppression. Chronically inflamed tissues are typically hypoxic. The role of hypoxia on myeloid cell functions during visceral leishmaniasis has not yet been studied. Here we show that L. donovani promotes the output from the bone marrow of monocytes with a regulatory phenotype that function as safe targets for the parasite. We also demonstrate that splenic myeloid cells acquire MDSC-like function in a HIF-1α-dependent manner. HIF-1α is also involved in driving the polarization towards M2-like macrophages and rendering intermediate stage monocytes more susceptible to L. donovani infection. Our results suggest that HIF-1α is a major player in the establishment of chronic Leishmania infection and is crucial for enhancing immunosuppressive functions and lowering leishmanicidal capacity of myeloid cells.


Sujet(s)
Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Leishmaniose viscérale , Macrophages/métabolisme , Cellules myéloïdes/métabolisme , Animaux , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Tolérance immunitaire/effets des médicaments et des substances chimiques , Interféron gamma/pharmacologie , Macrophages/parasitologie , Souris , Monocytes/métabolisme , Cellules myéloïdes/parasitologie , Rate/parasitologie
7.
Nat Commun ; 8: 13834, 2017 02 07.
Article de Anglais | MEDLINE | ID: mdl-28169274

RÉSUMÉ

Armadillo repeat containing 5 (ARMC5) is a cytosolic protein with no enzymatic activities. Little is known about its function and mechanisms of action, except that gene mutations are associated with risks of primary macronodular adrenal gland hyperplasia. Here we map Armc5 expression by in situ hybridization, and generate Armc5 knockout mice, which are small in body size. Armc5 knockout mice have compromised T-cell proliferation and differentiation into Th1 and Th17 cells, increased T-cell apoptosis, reduced severity of experimental autoimmune encephalitis, and defective immune responses to lymphocytic choriomeningitis virus infection. These mice also develop adrenal gland hyperplasia in old age. Yeast 2-hybrid assays identify 16 ARMC5-binding partners. Together these data indicate that ARMC5 is crucial in fetal development, T-cell function and adrenal gland growth homeostasis, and that the functions of ARMC5 probably depend on interaction with multiple signalling pathways.


Sujet(s)
Glandes surrénales/anatomopathologie , Protéines à domaine armadillo/génétique , Encéphalomyélite auto-immune expérimentale/immunologie , Développement foetal/physiologie , Immunité cellulaire/physiologie , Lymphocytes T/physiologie , Glandes surrénales/croissance et développement , Animaux , Différenciation cellulaire/génétique , Lignée cellulaire , Prolifération cellulaire/génétique , Encéphalomyélite auto-immune expérimentale/diagnostic , Femelle , Mutation germinale , Humains , Hyperplasie/génétique , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Mutation , Glycoprotéine MOG/immunologie , Fragments peptidiques/immunologie , Délétion de séquence , Indice de gravité de la maladie , Chimère obtenue par transplantation/immunologie
8.
J Immunol ; 197(9): 3618-3627, 2016 11 01.
Article de Anglais | MEDLINE | ID: mdl-27664281

RÉSUMÉ

Diverse signals received by CD8+ T cells are integrated to achieve the required magnitude of cell expansion and the appropriate balance of effector/memory CD8+ T cell generation. Notably, the strength and nature of TCR signaling influence the differentiation and functional capacity of effector and memory CD8+ T cells. Dok-1 and Dok-2, the two members of the Dok family expressed in T cells, negatively regulate TCR signaling in vitro. However, the role of Dok proteins in modulating T cell function in vivo has not yet studied. We studied the function of Dok-1 and Dok-2 proteins in the regulation of the CD8+ T cell response to vaccinia virus infection. Comparison of responses to vaccinia virus expressing OVA peptide SIINFEKL by wild-type and Dok-1/2-/- CD8+ OT-I cells showed that the absence of Dok-1 and Dok-2 slightly reduced the magnitude of virus-specific effector CD8+ T cell expansion. This was not due to reduced proliferation or enhanced apoptosis of effector CD8+ T cells. Dok-1/2-deficient effector CD8+ T cells showed increased cell surface TCR expression following virus infection in vivo and increased expression of granzyme B and TNF upon stimulation with peptide Ag ex vivo. Finally, Dok-1/2-deficient effector CD8+ T had a severe defect in survival that resulted in impaired generation of memory CD8+ T cells. These results reveal the critical involvement of Dok-1 and Dok-2 in a negative-feedback loop that prevents overactivation of CD8+ T cells and promotes memory formation.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Lymphocytes T CD8+/immunologie , Protéines de liaison à l'ADN/métabolisme , Mémoire immunologique , Phosphoprotéines/métabolisme , Protéines de liaison à l'ARN/métabolisme , Vaccine/immunologie , Maladies virales/immunologie , Protéines adaptatrices de la transduction du signal/génétique , Animaux , Lymphocytes T CD8+/virologie , Différenciation cellulaire , Survie cellulaire , Cellules cultivées , Protéines de liaison à l'ADN/génétique , Activation des lymphocytes/immunologie , Souris , Souris de lignée C57BL , Souris knockout , Phosphoprotéines/génétique , Protéines de liaison à l'ARN/génétique , Récepteur lymphocytaire T antigène, alpha-bêta/métabolisme , Transduction du signal
9.
Proc Natl Acad Sci U S A ; 113(37): E5444-53, 2016 09 13.
Article de Anglais | MEDLINE | ID: mdl-27573835

RÉSUMÉ

Exhaustion of CD8(+) T cells severely impedes the adaptive immune response to chronic viral infections. Despite major advances in our understanding of the molecular regulation of exhaustion, the cytokines that directly control this process during chronicity remain unknown. We demonstrate a direct impact of IL-2 and IL-15, two common gamma-chain-dependent cytokines, on CD8(+) T-cell exhaustion. Common to both cytokine receptors, the IL-2 receptor ß (IL2Rß) chain is selectively maintained on CD8(+) T cells during chronic lymphocytic choriomeningitis virus and hepatitis C virus infections. Its expression correlates with exhaustion severity and identifies terminally exhausted CD8(+) T cells both in mice and humans. Genetic ablation of the IL2Rß chain on CD8(+) T cells restrains inhibitory receptor induction, in particular 2B4 and Tim-3; precludes terminal differentiation of highly defective PD-1(hi) effectors; and rescues memory T-cell development and responsiveness to IL-7-dependent signals. Together, we ascribe a previously unexpected role to IL-2 and IL-15 as instigators of CD8(+) T-cell exhaustion during chronic viral infection.


Sujet(s)
Interleukine-15/génétique , Sous-unité bêta du récepteur à l'interleukine-2/génétique , Interleukine-2/génétique , Chorioméningite lymphocytaire/immunologie , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lymphocytes T CD8+/anatomopathologie , Hepacivirus/immunologie , Hepacivirus/pathogénicité , Récepteur cellulaire-2 du virus de l'hépatite A/génétique , Récepteur cellulaire-2 du virus de l'hépatite A/métabolisme , Humains , Mémoire immunologique , Interleukine-15/métabolisme , Interleukine-2/métabolisme , Sous-unité bêta du récepteur à l'interleukine-2/immunologie , Interleukine-7/génétique , Interleukine-7/immunologie , Chorioméningite lymphocytaire/génétique , Chorioméningite lymphocytaire/anatomopathologie , Chorioméningite lymphocytaire/virologie , Virus de la chorioméningite lymphocytaire/immunologie , Virus de la chorioméningite lymphocytaire/pathogénicité , Souris , Famille des molécules de signalisation de l'activation des lymphocytes/génétique , Famille des molécules de signalisation de l'activation des lymphocytes/métabolisme
10.
Cell Immunol ; 309: 42-49, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27372381

RÉSUMÉ

Organisms typically react to foreign pathogens by initiating an inflammatory response. However, in order to limit inflammatory tissue injury, it is essential for the organism to maintain the balance between inflammatory and anti-inflammatory responses. Dysregulation of this process can result in the strong inhibition of protective pro-inflammatory responses, and ultimately in pathogen persistence. Chronic infections are often associated with inflammation and tissue disruption. Inflamed tissues are characterized by low levels of oxygen and glucose, a microenvironment that triggers the stabilization of the hypoxia-inducible transcription factor HIF-1α. HIF-1α is the master regulator of the response to hypoxia. Here, we review the role of HIF-1α in the immune response to various pathogens and we highlight how certain microorganisms, including Leishmania parasites, have evolved to hijack the HIF-1 pathway to their advantage.


Sujet(s)
Interactions hôte-pathogène , Sous-unité alpha du facteur-1 induit par l'hypoxie/immunologie , Immunité , Inflammation , Leishmania/immunologie , Animaux , Humains , Hypoxie , Échappement immunitaire , Transduction du signal
11.
Eur J Immunol ; 45(12): 3324-38, 2015 Dec.
Article de Anglais | MEDLINE | ID: mdl-26426795

RÉSUMÉ

The ability to mount effective secondary responses is a cardinal feature of memory CD8(+) T cells. An understanding of the factors that regulate the generation and recall capacities of memory T cells remains to be ascertained. Several cues indicate that two highly related cytokines, IL-2 and IL-15, share redundant functions in this process. To establish their combined roles in memory CD8(+) T-cell development, maintenance, and secondary responses, we compared the outcome of adoptively transferred IL2Rß(+/-) or IL2Rß(-/-) CD8(+) T cells after an acute viral infection in mice. Our results demonstrate that both IL-2 and IL-15 signals condition the differentiation of primary and secondary short-lived effector cells by altering the transcriptional network governing lineage choices. These two cytokines also regulate the homeostasis of the memory T-cell pool, with effector memory CD8(+) T cells being the most sensitive to these two interleukins. Noticeably, the inability to respond to both cytokines limits the proliferation and survival of primary and secondary effectors cells, whereas it does not preclude potent cytotoxic functions and viral control either initially or upon rechallenge. Globally, these results indicate that lack of IL-2 and IL-15 signaling modulates the CD8(+) T-cell differentiation program but does not impede adequate effector functions.


Sujet(s)
Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Mémoire immunologique/effets des médicaments et des substances chimiques , Interleukine-15/pharmacologie , Interleukine-2/pharmacologie , Animaux , Lymphocytes T CD8+/cytologie , Sous-unité bêta du récepteur à l'interleukine-2/physiologie , Souris , Souris de lignée C57BL
12.
PLoS Pathog ; 11(6): e1004938, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-26046638

RÉSUMÉ

Inflammation is known to be necessary for promoting, sustaining, and tuning CD8+ T cell responses. Following experimental Leishmania donovani infection, the inflammatory response is mainly induced by the transcription factor IRF-5. IRF-5 is responsible for the activation of several genes encoding key pro-inflammatory cytokines, such as IL-6 and TNF. Here, we investigate the role of IRF-5-mediated inflammation in regulating antigen-specific CD8+ T cell responses during L. donovani infection. Our data demonstrate that the inflammatory response induced by IRF-5 limits CD8+ T cell expansion and induces HIF-1α in dendritic cells. Ablation of HIF-1α in CD11c+ cells resulted into a higher frequency of short-lived effector cells (SLEC), enhanced CD8+ T cell expansion, and increased IL-12 expression by splenic DCs. Moreover, mice with a targeted depletion of HIF-1α in CD11c+ cells had a significantly lower splenic parasite burden, suggesting that induction of HIF-1α may represent an immune evasive mechanism adopted by Leishmania parasites to establish persistent infections.


Sujet(s)
Lymphocytes T CD8+/immunologie , Cellules dendritiques/immunologie , Sous-unité alpha du facteur-1 induit par l'hypoxie/biosynthèse , Échappement immunitaire/immunologie , Facteurs de régulation d'interféron/immunologie , Leishmaniose viscérale/immunologie , Transfert adoptif , Animaux , Technique de Western , Modèles animaux de maladie humaine , Cytométrie en flux , Sous-unité alpha du facteur-1 induit par l'hypoxie/immunologie , Inflammation/immunologie , Leishmania donovani , Souris , Souris de lignée C57BL , Souches mutantes de souris , Réaction de polymérisation en chaine en temps réel
13.
Cell Mol Gastroenterol Hepatol ; 1(3): 325-341.e1, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-28210682

RÉSUMÉ

BACKGROUND & AIMS: The constant exposure of the liver to food and bacterial antigens through the mesenteric circulation requires it to maintain tolerance while preserving the ability to mount an effective immune response against pathogens. We investigated the contribution of the liver's tolerogenic nature on the establishment of chronic viral infections. METHODS: TTR-NP mice, which express the nucleoprotein (NP) of lymphocytic choriomeningitis virus (LCMV) specifically in hepatocytes under control of a modified transthyretin (TTR) promoter, were infected with the Armstrong (Arm) or WE acute strains of LCMV. RESULTS: The infection persisted for at least 147 days in TTR-NP mice. Expression of NP by the liver induced a strong peripheral tolerance against NP that was mediated by interleukin-10-secreting CD4+ regulatory T cells, leading to high PD-1 (programmed death-1) expression and reduced effector function of virus-specific T cells. Despite an active immune response against LCMV, peripheral tolerance against a single viral protein was sufficient to induce T-cell exhaustion and chronic LCMV Armstrong (Arm) or WE infection by limiting the antiviral T-cell response in an otherwise immunocompetent host. Regulatory T-cell depletion of chronically infected TTR-NP mice led to functional restoration of LCMV-specific CD4+ and CD8+ T cell responses and viral clearance. CONCLUSIONS: Expression of a viral antigen by hepatocytes can induce a state of peripheral tolerance mediated by regulatory T cells that can lead to the establishment of a chronic viral infection. Strategies targeting regulatory T cells in patients chronically infected with hepatotropic viruses could represent a promising approach to restore functional antiviral immunity and clear infection.

14.
Mol Ther ; 22(6): 1198-1210, 2014 Jun.
Article de Anglais | MEDLINE | ID: mdl-24590047

RÉSUMÉ

Cancer therapy using oncolytic viruses has gained interest in the last decade. Vesicular stomatitis virus is an attractive candidate for this alternative treatment approach. The importance of the immune response against tumor antigens in virotherapy efficacy is now well recognized, however, its relative contribution versus the intrinsic oncolytic capacity of viruses has been difficult to evaluate. To start addressing this question, we compared glycoprotein and matrix mutants of vesicular stomatitis virus (VSV), showing different oncolytic potentials for B16/B16gp33 melanoma tumor cells in vitro, with the wild-type virus in their ability to induce tumor-specific CD8(+) T cell responses and control tumor progression in vivo. Despite the fact that wild-type and G mutants induced a stronger gp33-specific immune response compared to the MM51R mutant, all VSV strains showed a similar capacity to slow down tumor progression. The effectiveness of the matrix mutant treatment proved to be CD8(+) dependent and directed against tumor antigens other than gp33 since adoptive transfer of isolated CD8(+) T lymphocytes from treated B16gp33-bearing mice resulted in significant protection of naive mice against challenge with the parental tumor. Remarkably, the VSV matrix mutant induced the upregulation of major histocompatibility class-I antigen at the tumor cell surface thus favoring recognition by CD8(+) T cells. These results demonstrate that VSV mutants induce an antitumor immune response using several mechanisms. A better understanding of these mechanisms will prove useful for the rational design of viruses with improved therapeutic efficacy.


Sujet(s)
Lymphocytes T CD8+/métabolisme , Mélanome expérimental/immunologie , Mélanome expérimental/thérapie , Glycoprotéines membranaires/génétique , Virus oncolytiques/génétique , Vesiculovirus/immunologie , Protéines de l'enveloppe virale/génétique , Protéines de la matrice virale/génétique , Animaux , Lignée cellulaire , Chlorocebus aethiops , Gènes MHC de classe I , Cellules HepG2 , Humains , Immunothérapie adoptive , Mélanome expérimental/virologie , Antigènes spécifiques du mélanome/métabolisme , Glycoprotéines membranaires/immunologie , Souris , Souris de lignée C57BL , Thérapie virale de cancers , Virus oncolytiques/immunologie , Cellules Vero , Vesiculovirus/métabolisme , Protéines de l'enveloppe virale/immunologie , Protéines de la matrice virale/immunologie
15.
J Immunol ; 191(11): 5420-9, 2013 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-24140642

RÉSUMÉ

TNF-like ligand 1A (TL1A), also known as TNFSF15, is a member of the TNF superfamily. Its known receptor is death receptor 3 (DR3). In humans, TL1A also binds to a secreted TNF family member called decoy receptor 3, which interferes with the interaction between TL1A and DR3. TL1A/DR3 signal has been implicated in several autoimmune diseases in animal models as well as in clinical conditions. We generated TL1A gene knockout (KO) mice to assess its role in collagen-induced arthritis (CIA), a mouse model of human rheumatoid arthritis. The KO mice were fertile and had no visible anomalies. Their lymphoid organ size and cellularity, T and B cell subpopulations, Th cell and regulatory T cell development in vivo and in vitro, and antiviral immune responses were comparable to those of wild-type mice. However, the KO mice presented ameliorated CIA in terms of clinical scores, disease incidence, and pathological scores. The KO mice had reduced titers of pathogenic anti-collagen Abs in the sera. No apparent defect was found in the function of follicular Th cells. We revealed that plasma cells but not B cells expressed high levels of DR3 and were direct targets of TL1A. In the presence of TL1A, they survived better and produced more pathogenic Ab. This study presented novel knowledge about the role of TL1A in humoral immune responses and its mechanism of action in CIA pathogenesis.


Sujet(s)
Arthrite expérimentale/immunologie , Polyarthrite rhumatoïde/immunologie , Plasmocytes/immunologie , Membre-25 de la superfamille des récepteurs au TNF/métabolisme , Membre-15 de la superfamille du facteur de nécrose tumorale/métabolisme , Animaux , Production d'anticorps/génétique , Arthrite expérimentale/génétique , Polyarthrite rhumatoïde/génétique , Survie cellulaire/génétique , Cellules cultivées , Collagène/immunologie , Humains , Immunité humorale , Souris , Souris de lignée C57BL , Souris knockout , Membre-25 de la superfamille des récepteurs au TNF/génétique , Membre-15 de la superfamille du facteur de nécrose tumorale/génétique , Régulation positive
16.
PLoS One ; 8(12): e82808, 2013.
Article de Anglais | MEDLINE | ID: mdl-24391722

RÉSUMÉ

Our earlier study revealed that STRA6 (stimulated by retinoic acid gene 6) was up-regulated within 3 h of TCR stimulation. STRA6 is the high-affinity receptor for plasma retinol-binding protein (RBP) and mediates cellular vitamin A uptake. We generated STRA6 knockout (KO) mice to assess whether such up-regulation was critical for T-cell activation, differentiation and function. STRA6 KO mice under vitamin A sufficient conditions were fertile without apparent anomalies upon visual inspection. The size, cellularity and lymphocyte subpopulations of STRA6 KO thymus and spleen were comparable to those of their wild type (WT) controls. KO and WT T cells were similar in terms of TCR-stimulated proliferation in vitro and homeostatic expansion in vivo. Naive KO CD4 cells differentiated in vitro into Th1, Th2, Th17 as well as regulatory T cells in an analogous manner as their WT counterparts. In vivo experiments revealed that anti-viral immune responses to lymphocytic choriomeningitis virus in KO mice were comparable to those of WT controls. We also demonstrated that STRA6 KO and WT mice had similar glucose tolerance. Total vitamin A levels are dramatically lower in the eyes of KO mice as compared to those of WT mice, but the levels in other organs were not significantly affected after STRA6 deletion under vitamin A sufficient conditions, indicating that the eye is the mouse organ most sensitive to the loss of STRA6. Our results demonstrate that 1) in vitamin A sufficiency, the deletion of STRA6 in T cells does no affect the T-cell immune responses so-far tested, including those depend on STAT5 signaling; 2) STRA6-independent vitamin A uptake compensated the lack of STRA6 in lymphoid organs under vitamin A sufficient conditions in mice; 3) STRA6 is critical for vitamin A uptake in the eyes even in vitamin A sufficiency.


Sujet(s)
Protéines membranaires/déficit , Protéines membranaires/génétique , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Animaux , Différenciation cellulaire , Prolifération cellulaire , Oeil/immunologie , Oeil/métabolisme , Femelle , Immunité cellulaire , Activation des lymphocytes , Virus de la chorioméningite lymphocytaire/immunologie , Tissu lymphoïde/immunologie , Tissu lymphoïde/métabolisme , Mâle , Protéines membranaires/métabolisme , Souris , Souris de lignée C57BL , Souris knockout , ARN messager/génétique , ARN messager/métabolisme , Récepteurs aux antigènes des cellules T/métabolisme , Transduction du signal , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Lymphocytes T/cytologie , Régulation positive , Rétinol/métabolisme
17.
J Biol Chem ; 286(48): 41135-41152, 2011 Dec 02.
Article de Anglais | MEDLINE | ID: mdl-21976681

RÉSUMÉ

Erythropoietin-producing hepatocellular kinases (Eph kinases) constitute the largest family of cell membrane receptor tyrosine kinases, and their ligand ephrins are also cell surface molecules. Because of promiscuous interaction between Ephs and ephrins, there is considerable redundancy in this system, reflecting the essential roles of these molecules in the biological system through evolution. In this study, both Efnb1 and Efnb2 were null-mutated in the T cell compartment of mice through loxP-mediated gene deletion. Mice with this double conditional mutation (double KO mice) showed reduced thymus and spleen size and cellularity. There was a significant decrease in the DN4, double positive, and single positive thymocyte subpopulations and mature CD4 and CD8 cells in the periphery. dKO thymocytes and peripheral T cells failed to compete with their WT counterparts in irradiated recipients, and the T cells showed compromised ability of homeostatic expansion. dKO naive T cells were inferior in differentiating into Th1 and Th17 effectors in vitro. The dKO mice showed diminished immune response against LCMV infection. Mechanistic studies revealed that IL-6 signaling in dKO T cells was compromised, in terms of abated induction of STAT3 phosphorylation upon IL-6 stimulation. This defect likely contributed to the observed in vitro and in vivo phenotype in dKO mice. This study revealed novel roles of Efnb1 and Efnb2 in T cell development and function.


Sujet(s)
Différenciation cellulaire/immunologie , Éphrine B1/immunologie , Immunité innée , Interleukine-6/immunologie , Chorioméningite lymphocytaire/immunologie , Virus de la chorioméningite lymphocytaire/immunologie , Transduction du signal/immunologie , Lymphocytes auxiliaires Th1/immunologie , Cellules Th17/immunologie , Thymocytes/immunologie , Animaux , Lymphocytes T CD8+ , Différenciation cellulaire/génétique , Éphrine B1/génétique , Interleukine-6/génétique , Chorioméningite lymphocytaire/génétique , Souris , Souris knockout , Taille d'organe/génétique , Taille d'organe/immunologie , Phosphorylation/génétique , Phosphorylation/immunologie , Facteur de transcription STAT-3/génétique , Facteur de transcription STAT-3/immunologie , Transduction du signal/génétique , Rate/immunologie , Rate/anatomopathologie , Rate/virologie , Lymphocytes auxiliaires Th1/anatomopathologie , Cellules Th17/anatomopathologie , Thymocytes/anatomopathologie , Thymocytes/virologie , Thymus (glande)/immunologie , Thymus (glande)/anatomopathologie , Thymus (glande)/virologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE