Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Antimicrob Agents Chemother ; 67(2): e0133122, 2023 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-36700643

RESUMO

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.


Assuntos
Vírus da Dengue , Dengue , Flavivirus , Infecção por Zika virus , Zika virus , Animais , Humanos , Antivirais/farmacologia , Antivirais/uso terapêutico , Dengue/tratamento farmacológico , Vírus da Dengue/genética , Estágios do Ciclo de Vida , Replicação do RNA , RNA Viral/genética , Replicação Viral , Zika virus/genética , RNA Subgenômico/genética
2.
J Immunol ; 198(1): 292-299, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27864474

RESUMO

The increasing use of plant viruses for the development of new vaccines and immunotherapy approaches poses questions regarding the mechanism by which the mammalian immune system recognizes these viruses. For example, although natural Abs (NA) and complement are key components of the innate immune system involved in the opsonization, phagocytosis, and destruction of microorganisms infecting mammals, their implication in plant virus recognition and immunogenicity is not well defined. In this study, we address the involvement of NA and the complement system in the activation of innate immunity through engagement of TLR7 with papaya mosaic virus (PapMV)-like nanoparticles. We demonstrate that NA, although binding to PapMV, are not involved in its recognition by the immune system. On the other hand, C3 strongly binds to PapMV nanoparticles and its depletion significantly reduces PapMV's interaction with immune cells. Unexpectedly, however, we observed increased immune cell activation following administration of PapMV to complement-depleted mice. TLR7 activation by PapMV in the absence of C3 induced higher IFN-α production, resulting in superior immune cell activation and increased immunotherapeutic properties. In conclusion, in this study we established the involvement of the complement system in the recognition and the phagocytosis of PapMV nanoparticles and identified an unsuspected role for C3 in regulating the production of IFN-α following TLR7 activation.


Assuntos
Complemento C3/imunologia , Células Dendríticas/imunologia , Interferon gama/biossíntese , Glicoproteínas de Membrana/imunologia , Vírus do Mosaico/imunologia , Receptor 7 Toll-Like/imunologia , Animais , Células Dendríticas/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas , Fagocitose/imunologia , Reação em Cadeia da Polimerase , Receptor 7 Toll-Like/metabolismo
3.
Proc Natl Acad Sci U S A ; 113(37): E5444-53, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27573835

RESUMO

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.


Assuntos
Interleucina-15/genética , Subunidade beta de Receptor de Interleucina-2/genética , Interleucina-2/genética , Coriomeningite Linfocítica/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Hepacivirus/imunologia , Hepacivirus/patogenicidade , Receptor Celular 2 do Vírus da Hepatite A/genética , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Humanos , Memória Imunológica , Interleucina-15/metabolismo , Interleucina-2/metabolismo , Subunidade beta de Receptor de Interleucina-2/imunologia , Interleucina-7/genética , Interleucina-7/imunologia , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/patologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Camundongos , Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo
4.
J Immunol ; 197(9): 3618-3627, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27664281

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Linfócitos T CD8-Positivos/imunologia , Proteínas de Ligação a DNA/metabolismo , Memória Imunológica , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Vacínia/imunologia , Viroses/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Linfócitos T CD8-Positivos/virologia , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Proteínas de Ligação a DNA/genética , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfoproteínas/genética , Proteínas de Ligação a RNA/genética , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Transdução de Sinais
5.
Nano Lett ; 16(3): 1826-32, 2016 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-26891174

RESUMO

The recent development of novel immunotherapies is revolutionizing cancer treatment. These include, for example, immune checkpoint blockade, immunomodulation, or therapeutic vaccination. Although effective on their own, combining multiple approaches will most likely be required in order to achieve the maximal therapeutic benefit. In this regard, the papaya mosaic virus nanoparticle (PapMV) has shown tremendous potential as (i) an immunostimulatory molecule, (ii) an adjuvant, and (iii) a vaccine platform through its intrinsic capacity to activate the innate immune response in an IFN-α-dependent manner. Here, we demonstrate that intratumor administration of PapMV significantly slows down melanoma progression and prolongs survival. This correlates with enhanced chemokine and pro-inflammatory-cytokine production in the tumor and increased immune-cell infiltration. Proportions of total and tumor-specific CD8(+) T cells dramatically increase following PapMV treatment whereas those of myeloid-derived suppressor cells (MDSC) concomitantly decrease. Moreover, systemic PapMV administration prevents metastatic tumor-implantation in the lungs. Importantly, PapMV also synergistically improves the therapeutic benefit of dendritic cell (DC)-based vaccination and PD-1 blockade by potentiating antitumor immune responses. This study illustrates the immunostimulatory potential of a plant virus-derived nanoparticle for cancer therapy either alone or in conjunction with other promising immunotherapies in clinical development.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Melanoma/prevenção & controle , Vírus do Mosaico/imunologia , Nanopartículas , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Carica/virologia , Linhagem Celular Tumoral , Citocinas/imunologia , Feminino , Imunoterapia , Melanoma/imunologia , Melanoma/patologia , Camundongos Endogâmicos C57BL , Vírus do Mosaico/química , Nanopartículas/química
7.
Eur J Immunol ; 45(12): 3324-38, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26426795

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Memória Imunológica/efeitos dos fármacos , Interleucina-15/farmacologia , Interleucina-2/farmacologia , Animais , Linfócitos T CD8-Positivos/citologia , Subunidade beta de Receptor de Interleucina-2/fisiologia , Camundongos , Camundongos Endogâmicos C57BL
8.
J Immunol ; 192(3): 1071-8, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24376264

RESUMO

Developing new adjuvants and vaccination strategies is of paramount importance to successfully fight against many life-threatening infectious diseases and cancer. Very few adjuvants are currently authorized for human use, and these mainly stimulate a humoral response. However, specific Abs are not sufficient to confer protection against persisting infections or cancer. Therefore, development of adjuvants and immunomodulators able to enhance cell-mediated immune responses represents a major medical need. We recently showed that papaya mosaic virus nanoparticles (PapMV), self-assembled from the coat protein of a plant virus and a noncoding ssRNA molecule, are highly immunogenic in mice. PapMV can be used either as a vaccine delivery platform, through fusion of various epitopes to the coat protein or as adjuvant to enhance humoral immune responses against coadministered Ags or vaccines. However, the mechanisms that confer these immunomodulatory properties to PapMV and its ability to enhance T cell vaccines remain unknown. Using immunization studies in mice, we demonstrate in this paper that PapMV represents a novel TLR7 agonist with strong immunostimulatory properties. More importantly, pretreatment with PapMV significantly improves effector and memory CD8(+) T cell responses generated through dendritic cell vaccination increasing protection against a Listeria monocytogenes challenge.


Assuntos
Adjuvantes Imunológicos , Linfócitos T CD8-Positivos/imunologia , Listeria monocytogenes/imunologia , Listeriose/prevenção & controle , Glicoproteínas de Membrana/agonistas , Subpopulações de Linfócitos T/imunologia , Receptor 7 Toll-Like/agonistas , Tymovirus/imunologia , Vacinação , Imunidade Adaptativa , Animais , Células Dendríticas/imunologia , Avaliação Pré-Clínica de Medicamentos , Feminino , Imunoglobulina G/biossíntese , Memória Imunológica , Interferon Tipo I/imunologia , Listeriose/imunologia , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/imunologia , Nanopartículas , Ovalbumina/imunologia , RNA Viral/imunologia , Receptor de Interferon alfa e beta/deficiência , Receptor 7 Toll-Like/deficiência , Receptor 7 Toll-Like/imunologia , Tymovirus/genética
9.
PLoS Pathog ; 9(4): e1003298, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23633948

RESUMO

The RIG-I like receptor pathway is stimulated during RNA virus infection by interaction between cytosolic RIG-I and viral RNA structures that contain short hairpin dsRNA and 5' triphosphate (5'ppp) terminal structure. In the present study, an RNA agonist of RIG-I was synthesized in vitro and shown to stimulate RIG-I-dependent antiviral responses at concentrations in the picomolar range. In human lung epithelial A549 cells, 5'pppRNA specifically stimulated multiple parameters of the innate antiviral response, including IRF3, IRF7 and STAT1 activation, and induction of inflammatory and interferon stimulated genes - hallmarks of a fully functional antiviral response. Evaluation of the magnitude and duration of gene expression by transcriptional profiling identified a robust, sustained and diversified antiviral and inflammatory response characterized by enhanced pathogen recognition and interferon (IFN) signaling. Bioinformatics analysis further identified a transcriptional signature uniquely induced by 5'pppRNA, and not by IFNα-2b, that included a constellation of IRF7 and NF-kB target genes capable of mobilizing multiple arms of the innate and adaptive immune response. Treatment of primary PBMCs or lung epithelial A549 cells with 5'pppRNA provided significant protection against a spectrum of RNA and DNA viruses. In C57Bl/6 mice, intravenous administration of 5'pppRNA protected animals from a lethal challenge with H1N1 Influenza, reduced virus titers in mouse lungs and protected animals from virus-induced pneumonia. Strikingly, the RIG-I-specific transcriptional response afforded partial protection from influenza challenge, even in the absence of type I interferon signaling. This systems approach provides transcriptional, biochemical, and in vivo analysis of the antiviral efficacy of 5'pppRNA and highlights the therapeutic potential associated with the use of RIG-I agonists as broad spectrum antiviral agents.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A Subtipo H1N1/imunologia , Infecções por Orthomyxoviridae/tratamento farmacológico , RNA Viral/farmacologia , Receptores do Ácido Retinoico/agonistas , Receptores do Ácido Retinoico/metabolismo , Animais , Antivirais/uso terapêutico , Linhagem Celular , Ativação Enzimática , Humanos , Imunidade Inata , Inflamação , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Interferência de RNA , RNA Viral/genética , RNA Viral/metabolismo , RNA Viral/uso terapêutico , Receptores do Ácido Retinoico/genética , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais
10.
J Immunol ; 191(11): 5420-9, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24140642

RESUMO

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.


Assuntos
Artrite Experimental/imunologia , Artrite Reumatoide/imunologia , Plasmócitos/imunologia , Membro 25 de Receptores de Fatores de Necrose Tumoral/metabolismo , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Animais , Formação de Anticorpos/genética , Artrite Experimental/genética , Artrite Reumatoide/genética , Sobrevivência Celular/genética , Células Cultivadas , Colágeno/imunologia , Humanos , Imunidade Humoral , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Regulação para Cima
11.
J Immunol ; 190(12): 5949-60, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23667108

RESUMO

Activation-induced deaminase converts deoxycytidine to deoxyuridine at the Ig loci. Complementary pathways, initiated by the uracil-DNA glycosylase (UNG) or the mismatch repair factor MSH2/MSH6, must process the deoxyuridine to initiate class-switch recombination (CSR) and somatic hypermutation. UNG deficiency most severely reduces CSR efficiency and only modestly affects the somatic hypermutation spectrum in vitro. This would predict isotype-switching deficiency but normal affinity maturation in Ung(-/-) mice in vivo, but this has not been tested. Moreover, puzzling differences in the amount of circulating Ig between UNG-deficient humans and mice make it unclear to what extent MSH2/MSH6 can complement for UNG in vivo. We find that Ab affinity maturation is indeed unaffected in Ung(-/-) mice, even allowing IgM responses with higher than normal affinity. Ung(-/-) mice display normal to only moderately reduced basal levels of most circulating Ig subclasses and gut-associated IgA, which are elicited in response to chronically available environmental Ag. In contrast, their ability to produce switched Ig in response to immunization or vesicular stomatitis virus infection is strongly impaired. Our results uncover a specific need for UNG in CSR for timely and efficient acute Ab responses in vivo. Furthermore, Ung(-/-) mice provide a novel model for separating isotype switching and affinity maturation during acute (but not chronic) Ab responses, which could be useful for dissecting their relative contribution to some infections. Interestingly, Ung(-/-) mice present with circulating autoantibodies, suggesting that UNG may impinge on tolerance.


Assuntos
Afinidade de Anticorpos/imunologia , Linfócitos B/imunologia , Switching de Imunoglobulina/imunologia , Ativação Linfocitária/imunologia , Uracila-DNA Glicosidase/imunologia , Animais , Autoanticorpos/sangue , Autoanticorpos/imunologia , Linfócitos B/metabolismo , Diferenciação Celular/imunologia , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Uracila-DNA Glicosidase/deficiência
12.
Mol Ther ; 22(6): 1198-1210, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24590047

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Glicoproteínas de Membrana/genética , Vírus Oncolíticos/genética , Vesiculovirus/imunologia , Proteínas do Envelope Viral/genética , Proteínas da Matriz Viral/genética , Animais , Linhagem Celular , Chlorocebus aethiops , Genes MHC Classe I , Células Hep G2 , Humanos , Imunoterapia Adotiva , Melanoma Experimental/virologia , Antígenos Específicos de Melanoma/metabolismo , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Terapia Viral Oncolítica , Vírus Oncolíticos/imunologia , Células Vero , Vesiculovirus/metabolismo , Proteínas do Envelope Viral/imunologia , Proteínas da Matriz Viral/imunologia
13.
Cell Rep ; 43(3): 113879, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38416647

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos , Leishmaniose Visceral , Humanos , Subpopulações de Linfócitos T , Fatores de Transcrição , Receptores CXCR5
14.
J Autoimmun ; 42: 19-28, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23137675

RESUMO

CD8(+) T-cell immune response to liver antigens is often functionally diminished or absent. This may occur via deletion of these autoaggressive T-cells, through the acquisition of an anergic phenotype, or via active suppression mediated by other cell populations. We generated a double transgenic model in which mice express CD8(+) T-cells specific for the lymphocytic choriomeningitis virus nucleoprotein (LCMV-NP) and LCMV-NP as a hepatic neo-autoantigen, to study the immunological response of potentially liver antigen autoaggressive CD8(+) T-cells. Autoreactive transgenic CD8(+) T-cells were analyzed for functionality and cytotoxic effector status. Despite severe peripheral deletion of liver-specific CD8(+) T-cells, a fraction of autoreactive NP-specific CD8(+) T-cells accumulate in liver, resulting in hepatocyte injury and production of auto-antibodies in both male and female mice. NP-specific intrahepatic T-cells showed capacity to proliferate, produce cytokines and up-regulate activation markers. These data provide in vivo evidence that autoreactive CD8(+) T-cells are activated in the liver and developed an inflammatory process, but require additional factors to cause severe autoimmune destruction of hepatocytes. Our new model will provide a valuable tool for further exploration of the immunological response involved in inflammatory liver diseases, including autoimmune hepatitis.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Hepatócitos/imunologia , Fígado/imunologia , Nucleoproteínas/metabolismo , Animais , Antígenos Virais/genética , Antígenos Virais/imunologia , Apoptose/genética , Apoptose/imunologia , Células Cultivadas , Citotoxicidade Imunológica/genética , Modelos Animais de Doenças , Feminino , Hepatite Autoimune/imunologia , Hepatócitos/patologia , Vírus da Coriomeningite Linfocítica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Nucleoproteínas/genética , Nucleoproteínas/imunologia , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Transgenes/genética
15.
Med Sci (Paris) ; 29(2): 175-82, 2013 Feb.
Artigo em Francês | MEDLINE | ID: mdl-23452604

RESUMO

Cancer is a complex disease that affects more and more people around the world. Unfortunately, existing treatments are only partially efficient and often induce major side effects. Thus, the use of viruses to selectively kill cancer cells is a new promising therapeutic approach. Recently, VSV has been used in oncolytic virotherapy because of its capacity to preferentially infect most human tumor cells. However, despite the availability of good oncolytic VSV mutants, the large variability of tumor cell types and the multiple ways in which they can evade viral infection suggests that therapeutic combinations of various viruses will be necessary to efficiently treat most cancers. A better understanding of the infection mechanisms and immune system recruitment by oncolytic viruses will be of great value for the development of safe and efficient strategies for cancer treatment.


Assuntos
Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vesiculovirus/fisiologia , Humanos , Sistema Imunitário/fisiologia , Modelos Biológicos , Neoplasias/imunologia , Terapia Viral Oncolítica/tendências , Vírus Oncolíticos/genética , Estomatite Vesicular/virologia , Vesiculovirus/genética , Vesiculovirus/imunologia
16.
J Biol Chem ; 286(48): 41135-41152, 2011 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-21976681

RESUMO

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.


Assuntos
Diferenciação Celular/imunologia , Efrina-B1/imunologia , Imunidade Inata , Interleucina-6/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Transdução de Sinais/imunologia , Células Th1/imunologia , Células Th17/imunologia , Timócitos/imunologia , Animais , Linfócitos T CD8-Positivos , Diferenciação Celular/genética , Efrina-B1/genética , Interleucina-6/genética , Coriomeningite Linfocítica/genética , Camundongos , Camundongos Knockout , Tamanho do Órgão/genética , Tamanho do Órgão/imunologia , Fosforilação/genética , Fosforilação/imunologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/imunologia , Transdução de Sinais/genética , Baço/imunologia , Baço/patologia , Baço/virologia , Células Th1/patologia , Células Th17/patologia , Timócitos/patologia , Timócitos/virologia , Timo/imunologia , Timo/patologia , Timo/virologia
17.
J Virol ; 85(13): 6513-20, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21561919

RESUMO

Vesicular stomatitis virus (VSV) has been widely used to characterize cellular processes, viral resistance, and cytopathogenicity. Recently, VSV has also been used for oncolytic virotherapy due to its capacity to selectively lyse tumor cells. Mutants of the matrix (M) protein of VSV have generally been preferred to the wild-type virus for oncolysis because of their ability to induce type I interferon (IFN) despite causing weaker cytopathic effects. However, due to the large variability of tumor types, it is quite clear that various approaches and combinations of multiple oncolytic viruses will be needed to effectively treat most cancers. With this in mind, our work focused on characterizing the cytopathogenic profiles of four replicative envelope glycoprotein (G) VSV mutants. In contrast to the prototypic M mutant, VSV G mutants are as efficient as wild-type virus at inhibiting cellular transcription and host protein translation. Despite being highly cytopathic, the mutant G(6R) triggers type I interferon secretion as efficiently as the M mutant. Importantly, most VSV G mutants are more effective at killing B16 and MC57 tumor cells in vitro than the M mutant or wild-type virus through apoptosis induction. Taken together, our results demonstrate that VSV G mutants retain the high cytopathogenicity of wild-type VSV, with G(6R) inducing type I IFN secretion at levels similar to that of the M mutant. VSV G protein mutants could therefore prove to be highly valuable for the development of novel oncolytic virotherapy strategies that are both safe and efficient for the treatment of various types of cancer.


Assuntos
Glicoproteínas de Membrana/genética , Mutação , Vírus Oncolíticos/patogenicidade , Vírus da Estomatite Vesicular Indiana/patogenicidade , Proteínas do Envelope Viral/genética , Animais , Linhagem Celular Tumoral , Chlorocebus aethiops , Efeito Citopatogênico Viral , Fibroblastos/virologia , Humanos , Células L , Camundongos , Dados de Sequência Molecular , Terapia Viral Oncolítica , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Análise de Sequência de DNA , Células Vero , Vírus da Estomatite Vesicular Indiana/genética , Vírus da Estomatite Vesicular Indiana/metabolismo
18.
Virology ; 567: 77-86, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35032866

RESUMO

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.


Assuntos
Atrofia/virologia , Interferon-alfa/imunologia , Interferon beta/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Timócitos/virologia , Timo/virologia , Animais , Atrofia/genética , Atrofia/imunologia , Atrofia/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Doença Crônica , Feminino , Regulação da Expressão Gênica , Humanos , Memória Imunológica , Interferon-alfa/genética , Interferon beta/genética , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/virologia , Depleção Linfocítica , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/patologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais/imunologia , Análise de Célula Única , Timócitos/imunologia , Timócitos/patologia , Timo/imunologia , Timo/patologia
19.
Eur J Immunol ; 40(8): 2190-9, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20518030

RESUMO

The initiation of CD8(+) T cell (CTL) immune responses can occur via cross-priming. Recent data suggested a relationship between cross-presentation and immunodominance of epitope-specific T cells. To test this association, we evaluated the efficacy of cross-presentation for several virus epitopes in vitro and examined if this can be extrapolated in vivo. Employing lymphocytic choriomeningitis virus (LCMV), we demonstrate that the cross-presentation and cross-priming of LCMV antigens were dominated by NP396, but not NP205 when analyzing the LCMV-NP. Although with LCMV-GP, cross-presentation was dominated by GP276, and cross-priming was dominated by GP33. Importantly, although NP396 was significantly more efficient than GP33 in cross-presentation, cross-priming of their specific CTL was comparable. In a subsequent virus challenge after cross-priming, GP33-specific CTL dominated the response. Accordingly, based on our data, the ability of viral epitopes to be cross-presented in vitro does not entirely reflect what would occur in cross-priming. Thus, weak cross-presenting antigens may still cross-prime an efficient CTL response depending on other in vivo elements such as the naïve T-cell precursor frequencies.


Assuntos
Infecções por Arenaviridae/imunologia , Apresentação Cruzada , Células Dendríticas/metabolismo , Vírus da Coriomeningite Linfocítica/imunologia , Macrófagos/metabolismo , Linfócitos T Citotóxicos/metabolismo , Animais , Antígenos Virais/imunologia , Antígenos Virais/metabolismo , Linhagem Celular , Proliferação de Células , Células Dendríticas/imunologia , Células Dendríticas/virologia , Humanos , Epitopos Imunodominantes/imunologia , Epitopos Imunodominantes/metabolismo , Ativação Linfocitária , Vírus da Coriomeningite Linfocítica/patogenicidade , Macrófagos/imunologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/patologia
20.
Immunohorizons ; 4(4): 217-230, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-32332052

RESUMO

CD8+ T cell-mediated immunity is critical for host defense against viruses and requires mitochondria-mediated type I IFN (IFN-I) signaling for optimal protection. Cyclophilin D (CypD) is a mitochondrial matrix protein that modulates the mitochondrial permeability transition pore, but its role in IFN-I signaling and CD8+ T cell responses to viral infection has not been previously explored. In this study, we demonstrate that CypD plays a critical extrinsic role in the survival of Ag-specific CD8+ T cell following acute viral infection with lymphocytic choriomeningitis virus in mice. CypD deficiency resulted in reduced IFN-I and increased CD8+ T cell death, resulting in a reduced antiviral CD8+ T cell response. In addition, CypD deficiency was associated with an increase in pathogen burden at an early time-point following infection. Furthermore, our data demonstrate that transfer of wild-type macrophages (expressing CypD) to CypD-deficient mice can partially restore CD8+ T cell responses. These results establish that CypD plays an extrinsic role in regulating optimal effector CD8+ T cell responses to viral infection. Furthermore, this suggests that, under certain circumstances, inhibition of CypD function may have a detrimental impact on the host's ability to respond to viral infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Sobrevivência Celular/genética , Interações entre Hospedeiro e Microrganismos/genética , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Peptidil-Prolil Isomerase F/metabolismo , Transferência Adotiva/métodos , Animais , Antígenos CD8/metabolismo , Peptidil-Prolil Isomerase F/genética , Feminino , Interações entre Hospedeiro e Microrganismos/imunologia , Coriomeningite Linfocítica/terapia , Coriomeningite Linfocítica/virologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/imunologia , Mitocôndrias/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA