Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Immunol ; 210(9): 1292-1304, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36961447

RESUMO

It is generally accepted that influenza A virus (IAV) infection promotes a Th1-like CD4 T cell response and that this effector program underlies its protective impact. Canonical Th1 polarization requires cytokine-mediated activation of the transcription factors STAT1 and STAT4 that synergize to maximize the induction of the "master regulator" Th1 transcription factor, T-bet. Here, we determine the individual requirements for these transcription factors in directing the Th1 imprint primed by influenza infection in mice by tracking virus-specific wild-type or T-bet-deficient CD4 T cells in which STAT1 or STAT4 is knocked out. We find that STAT1 is required to protect influenza-primed CD4 T cells from NK cell-mediated deletion and for their expression of hallmark Th1 attributes. STAT1 is also required to prevent type I IFN signals from inhibiting the induction of the Th17 master regulator, Rorγt, in Th17-prone T-bet-/- cells responding to IAV. In contrast, STAT4 expression does not appreciably impact the phenotypic or functional attributes of wild-type or T-bet-/- CD4 T cell responses. However, cytokine-mediated STAT4 activation in virus-specific CD4 T cells enhances their Th1 identity in a T-bet-dependent manner, indicating that influenza infection does not promote maximal Th1 induction. Finally, we show that the T-bet-dependent protective capacity of CD4 T cell effectors against IAV is optimized by engaging both STAT1 and STAT4 during Th1 priming, with important implications for vaccine strategies aiming to generate T cell immunity.


Assuntos
Linfócitos T CD4-Positivos , Influenza Humana , Camundongos , Animais , Humanos , Antivirais/metabolismo , Proteínas com Domínio T/metabolismo , Interferon gama/metabolismo , Fatores de Transcrição/metabolismo , Células Th1 , Fator de Transcrição STAT4/metabolismo , Diferenciação Celular , Fator de Transcrição STAT1/metabolismo
2.
J Immunol ; 210(5): 628-639, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36645384

RESUMO

Overcoming interfering impacts of pre-existing immunity to generate universally protective influenza A virus (IAV)-specific T cell immunity through vaccination is a high priority. In this study, we passively transfer varied amounts of H1N1-IAV-specific immune serum before H1N1-IAV infection to determine how different levels of pre-existing Ab influence the generation and protective potential of heterosubtypic T cell responses in a murine model. Surprisingly, IAV nucleoprotein-specific CD4 and CD8 T cell responses are readily detected in infected recipients of IAV-specific immune serum regardless of the amount transferred. When compared with responses in control groups and recipients of low and intermediate levels of convalescent serum, nucleoprotein-specific T cell responses in recipients of high levels of IAV-specific serum, which prevent overt weight loss and reduce peak viral titers in the lungs, are, however, markedly reduced. Although detectable at priming, this response recalls poorly and is unable to mediate protection against a lethal heterotypic (H3N2) virus challenge at later memory time points. A similar failure to generate protective heterosubtypic T cell immunity during IAV priming is seen in offspring of IAV-primed mothers that naturally receive high titers of IAV-specific Ab through maternal transfer. Our findings support that priming of protective heterosubtypic T cell responses can occur in the presence of intermediate levels of pre-existing Ab. These results have high relevance to vaccine approaches aiming to incorporate and evaluate cellular and humoral immunity towards IAV and other viral pathogens against which T cells can protect against variants escaping Ab-mediated protection.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vírus da Influenza A , Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Animais , Camundongos , Humanos , Vírus da Influenza A Subtipo H3N2 , Linfócitos T CD8-Positivos , Anticorpos Antivirais , Soros Imunes
3.
J Immunol ; 204(12): 3307-3314, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32376651

RESUMO

IL-2 is a pleotropic cytokine with potent pro- and anti-inflammatory effects. These divergent impacts can be directed in vivo by forming complexes of IL-2 and anti-IL-2 mAbs (IL-2C) to target IL-2 to distinct subsets of cells based on their expression of subunits of the IL-2R. In this study, we show that treatment of mice with a prototypical anti-inflammatory IL-2C, JES6-1-IL-2C, best known to induce CD25+ regulatory CD4 T cell expansion, surprisingly causes robust induction of a suite of inflammatory factors. However, treating mice infected with influenza A virus with this IL-2C reduces lung immunopathology. We compare the spectrum of inflammatory proteins upregulated by pro- and anti-inflammatory IL-2C treatment and uncover a pattern of expression that reveals potentially beneficial versus detrimental aspects of the influenza-associated cytokine storm. Moreover, we show that anti-inflammatory IL-2C can deliver survival signals to CD4 T cells responding to influenza A virus that improve their memory fitness, indicating a novel application of IL-2 to boost pathogen-specific T cell memory while simultaneously reducing immunopathology.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Memória Imunológica/imunologia , Vírus da Influenza A/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Interleucina-2/imunologia , Infecções por Orthomyxoviridae/imunologia , Animais , Anticorpos Monoclonais/imunologia , Citocinas/imunologia , Feminino , Inflamação/imunologia , Inflamação/virologia , Pulmão/imunologia , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Reguladores/imunologia , Regulação para Cima/imunologia
4.
PLoS Pathog ; 15(8): e1007989, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31412088

RESUMO

Defining the most penetrating correlates of protective memory T cells is key for designing improved vaccines and T cell therapies. Here, we evaluate how interleukin (IL-2) production by memory CD4 T cells, a widely held indicator of their protective potential, impacts immune responses against murine influenza A virus (IAV). Unexpectedly, we show that IL-2-deficient memory CD4 T cells are more effective on a per cell basis at combating IAV than wild-type memory cells that produce IL-2. Improved outcomes orchestrated by IL-2-deficient cells include reduced weight loss and improved respiratory function that correlate with reduced levels of a broad array of inflammatory factors in the infected lung. Blocking CD70-CD27 signals to reduce CD4 T cell IL-2 production tempers the inflammation induced by wild-type memory CD4 T cells and improves the outcome of IAV infection in vaccinated mice. Finally, we show that IL-2 administration drives rapid and extremely potent lung inflammation involving NK cells, which can synergize with sublethal IAV infection to promote acute death. These results suggest that IL-2 production is not necessarily an indicator of protective CD4 T cells, and that the lung environment is particularly sensitive to IL-2-induced inflammation during viral infection.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Memória Imunológica/imunologia , Vírus da Influenza A/imunologia , Interleucina-2/metabolismo , Infecções por Orthomyxoviridae/imunologia , Pneumonia/imunologia , Animais , Feminino , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Pneumonia/metabolismo , Pneumonia/virologia
5.
Cell Immunol ; 331: 121-129, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29935764

RESUMO

Although cigarette smoke is known to alter immune responses, whether and how CD4 T cells are affected is not well-described. We aimed to characterize how exposure to cigarette smoke extract impacts CD4 T cell effector generation in vitro under Th1-polarizing conditions. Our results demonstrate that cigarette smoke directly acts on CD4 T cells to impair effector expansion by decreasing division and increasing apoptosis. Furthermore, cigarette smoke enhances Th1-associated cytokine production and increases expression of the transcription factor T-bet, the master regulator of Th1 differentiation. Finally, we show that exposure to cigarette smoke extract during priming impairs the ability of effectors to form memory cells. Our findings thus demonstrate that cigarette smoke simultaneously enhances effector functions but promotes terminal differentiation of CD4 T cell effectors. This study may be relevant to understanding how smoking can both aggravate autoimmune symptoms and reduce vaccine efficacy.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Memória Imunológica/imunologia , Ativação Linfocitária/imunologia , Nicotiana/química , Fumaça , Células Th1/imunologia , Animais , Apoptose/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/imunologia , Divisão Celular/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Humanos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Células Th1/metabolismo
6.
J Immunol ; 197(10): 3936-3949, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27798159

RESUMO

Although memory CD4 T cells are critical for effective immunity to pathogens, the mechanisms underlying their generation are still poorly defined. We find that following murine influenza infection, most effector CD4 T cells undergo apoptosis unless they encounter cognate Ag at a defined stage near the peak of effector generation. Ag recognition at this memory checkpoint blocks default apoptosis and programs their transition to long-lived memory. Strikingly, we find that viral infection is not required, because memory formation can be restored by the addition of short-lived, Ag-pulsed APC at this checkpoint. The resulting memory CD4 T cells express an enhanced memory phenotype, have increased cytokine production, and provide protection against lethal influenza infection. Finally, we find that memory CD4 T cell formation following cold-adapted influenza vaccination is boosted when Ag is administered during this checkpoint. These findings imply that persistence of viral Ag presentation into the effector phase is the key factor that determines the efficiency of memory generation. We also suggest that administering Ag at this checkpoint may improve vaccine efficacy.


Assuntos
Apresentação de Antígeno/imunologia , Antígenos Virais/imunologia , Linfócitos T CD4-Positivos/imunologia , Memória Imunológica , Orthomyxoviridae/imunologia , Animais , Apoptose , Citocinas/biossíntese , Citocinas/imunologia , Genes cdc , Humanos , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Interferon gama/biossíntese , Interferon gama/imunologia , Camundongos , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia
7.
Immunol Rev ; 255(1): 149-64, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23947353

RESUMO

Over the last decade, the known spectrum of CD4(+) T-cell effector subsets has become much broader, and it has become clear that there are multiple dimensions by which subsets with a particular cytokine commitment can be further defined, including their stage of differentiation, their location, and, most importantly, their ability to carry out discrete functions. Here, we focus on our studies that highlight the synergy among discrete subsets, especially those defined by helper and cytotoxic function, in mediating viral protection, and on distinctions between CD4(+) T-cell effectors located in spleen, draining lymph node, and in tissue sites of infection. What emerges is a surprising multiplicity of CD4(+) T-cell functions that indicate a large arsenal of mechanisms by which CD4(+) T cells act to combat viruses.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Memória Imunológica , Infecções Respiratórias/imunologia , Infecções Respiratórias/virologia , Viroses/imunologia , Vírus/imunologia , Animais , Citocinas/imunologia , Citocinas/metabolismo , Regulação da Expressão Gênica , Humanos , Vírus da Influenza A/imunologia , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções Respiratórias/genética , Infecções Respiratórias/metabolismo , Fatores de Transcrição/metabolismo , Viroses/genética , Viroses/metabolismo
8.
J Immunol ; 192(11): 5140-50, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24752446

RESUMO

Our previous in vivo studies show that both the amount of Ag and the number of available naive CD4 T cells affect the Th1/Th2 phenotype of the effector CD4 T cells generated. We examined how the number of OVA-specific CD4 TCR transgenic T cells affects the Th1/Th2 phenotype of anti-SRBC CD4 T cells generated in vivo upon immunization with different amounts of OVA-SRBC. Our observations show that a greater number of Ag-dependent CD4 T cell interactions are required to generate Th2 than Th1 cells. We established an in vitro system that recapitulates our main in vivo findings to more readily analyze the underlying mechanism. The in vitro generation of Th2 cells depends, as in vivo, upon both the number of responding CD4 T cells and the amount of Ag. We demonstrate, using agonostic/antagonistic Abs to various costimulatory molecules or their receptors, that the greater number of CD4 T cell interactions, required to generate Th2 over Th1 cells, does not involve CD40, OX40, or ICOS costimulation, but does involve B7/CD28 interactions. A comparison of the level of expression of B7 molecules by APC and CD4 T cells, under different conditions resulting in the substantial generation of Th1 and Th2 cells, leads us to propose that the critical CD28/B7 interactions, required to generate Th2 cells, may directly occur between CD4 T cells engaged with the same B cell acting as an APC.


Assuntos
Antígenos/imunologia , Antígeno B7-1/imunologia , Antígenos CD28/imunologia , Células Th1/imunologia , Células Th2/imunologia , Animais , Células Apresentadoras de Antígenos/citologia , Células Apresentadoras de Antígenos/imunologia , Antígenos/genética , Linfócitos B/citologia , Linfócitos B/imunologia , Antígeno B7-1/genética , Antígenos CD28/genética , Antígenos CD40/genética , Antígenos CD40/imunologia , Relação Dose-Resposta Imunológica , Proteína Coestimuladora de Linfócitos T Induzíveis/genética , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Camundongos Endogâmicos BALB C , Receptores OX40/genética , Receptores OX40/metabolismo , Células Th1/citologia , Células Th2/citologia
9.
J Immunol ; 190(1): 296-306, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23197262

RESUMO

We have previously shown that mice challenged with a lethal dose of A/Puerto Rico/8/34-OVA(I) are protected by injection of 4-8 × 10(6) in vitro-generated Tc1 or Tc17 CD8(+) effectors. Viral load, lung damage, and loss of lung function are all reduced after transfer. Weight loss is reduced and survival increased. We sought in this study to define the mechanism of this protection. CD8(+) effectors exhibit multiple effector activities, perforin-, Fas ligand-, and TRAIL-mediated cytotoxicity, and secretion of multiple cytokines (IL-2, IL-4, IL-5, IL-9, IL-10, IL-17, IL-21, IL-22, IFN-γ, and TNF) and chemokines (CCL3, CCL4, CCL5, CXCL9, and CXCL10). Transfer of CD8(+) effectors into recipients, before infection, elicits enhanced recruitment of host neutrophils, NK cells, macrophages, and B cells. All of these events have the potential to protect against viral infections. Removal of any one, however, of these potential mechanisms was without effect on protection. Even the simultaneous removal of host T cells, host B cells, and host neutrophils combined with the elimination of perforin-mediated lytic mechanisms in the donor cells failed to reduce their ability to protect. We conclude that CD8(+) effector T cells can protect against the lethal effects of viral infection by means of a large number of redundant mechanisms.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Vírus da Influenza A/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Animais , Linfócitos T CD8-Positivos/transplante , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Cultivadas , Testes Imunológicos de Citotoxicidade/métodos , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Infecções por Orthomyxoviridae/virologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/transplante , Linfócitos T Citotóxicos/virologia
10.
Proc Natl Acad Sci U S A ; 109(38): E2551-60, 2012 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-22927425

RESUMO

Whether differences between naive cell-derived primary (1°) and memory cell-derived secondary (2°) CD4(+) T-cell effectors contribute to protective recall responses is unclear. Here, we compare these effectors directly after influenza A virus infection. Both develop with similar kinetics, but 2° effectors accumulate in greater number in the infected lung and are the critical component of memory CD4(+) T-cell-mediated protection against influenza A virus, independent of earlier-acting memory-cell helper functions. Phenotypic, functional, and transcriptome analyses indicate that 2° effectors share organ-specific expression patterns with 1° effectors but are more multifunctional, with more multicytokine (IFN-γ(+)/IL-2(+)/TNF(+))-producing cells and contain follicular helper T-cell populations not only in the spleen and draining lymph nodes but also in the lung. In addition, they express more CD127 and NKG2A but less ICOS and Lag-3 than 1° effectors and express higher levels of several genes associated with survival and migration. Targeting two differentially expressed molecules, NKG2A and Lag-3, reveals differential regulation of 1° and 2° effector functions during pathogen challenge.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Memória Imunológica/imunologia , Animais , Antígenos CD/biossíntese , Galinhas , Citocinas/metabolismo , Perfilação da Expressão Gênica , Proteína Coestimuladora de Linfócitos T Induzíveis/biossíntese , Vírus da Influenza A/metabolismo , Interferon gama/metabolismo , Interleucina-2/metabolismo , Subunidade alfa de Receptor de Interleucina-7/biossíntese , Cinética , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Subfamília C de Receptores Semelhantes a Lectina de Células NK/biossíntese , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Baço/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteína do Gene 3 de Ativação de Linfócitos
11.
Immunol Rev ; 236: 110-24, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20636812

RESUMO

Signals orchestrating productive CD4+ T-cell responses are well documented; however, the regulation of contraction of CD4+ T-cell effector populations following the resolution of primary immune responses is not well understood. While distinct mechanisms of T-cell death have been defined, the relative importance of discrete death pathways during the termination of immune responses in vivo remains unclear. Here, we review the current understanding of cell-intrinsic and -extrinsic variables that regulate contraction of CD4+ T-cell effector populations through multiple pathways that operate both initially during T-cell priming and later during the effector phase. We discuss the relative importance of antigen-dependent and -independent mechanisms of CD4+ T-cell contraction during in vivo responses, with a special emphasis on influenza virus infection. In this model, we highlight the roles of greater differentiation and presence in the lung of CD4+ effector T cells, as well as their polarization to particular T-helper subsets, in maximizing contraction. We also discuss the role of autocrine interleukin-2 in limiting the extent of contraction, and we point out that these same factors regulate contraction during secondary CD4+ T-cell responses.


Assuntos
Apoptose/imunologia , Linfócitos T CD4-Positivos/imunologia , Influenza Humana/imunologia , Ativação Linfocitária/imunologia , Linfócitos T CD4-Positivos/metabolismo , Humanos , Influenza Humana/virologia , Interleucina-2/metabolismo , Modelos Imunológicos , Transdução de Sinais/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
12.
Cells ; 13(7)2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38607077

RESUMO

Current Influenza A virus (IAV) vaccines, which primarily aim to generate neutralizing antibodies against the major surface proteins of specific IAV strains predicted to circulate during the annual 'flu' season, are suboptimal and are characterized by relatively low annual vaccine efficacy. One approach to improve protection is for vaccines to also target the priming of virus-specific T cells that can protect against IAV even in the absence of preexisting neutralizing antibodies. CD4 T cells represent a particularly attractive target as they help to promote responses by other innate and adaptive lymphocyte populations and can also directly mediate potent effector functions. Studies in murine models of IAV infection have been instrumental in moving this goal forward. Here, we will review these findings, focusing on distinct subsets of CD4 T cell effectors that have been shown to impact outcomes. This body of work suggests that a major challenge for next-generation vaccines will be to prime a CD4 T cell population with the same spectrum of functional diversity generated by IAV infection. This goal is encapsulated well by the motto 'ex pluribus unum': that an optimal CD4 T cell response comprises many individual specialized subsets responding together.


Assuntos
Vírus da Influenza A , Vacinas contra Influenza , Influenza Humana , Animais , Humanos , Camundongos , Anticorpos Neutralizantes , Linfócitos T CD4-Positivos , Vacinas , Vacinas contra Influenza/imunologia
13.
J Exp Med ; 204(9): 2199-211, 2007 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-17724126

RESUMO

The majority of highly activated CD4 T cell effectors die after antigen clearance, but a small number revert to a resting state, becoming memory cells with unique functional attributes. It is currently unclear when after antigen clearance effectors return to rest and acquire important memory properties. We follow well-defined cohorts of CD4 T cells through the effector-to-memory transition by analyzing phenotype, important functional properties, and gene expression profiles. We find that the transition from effector to memory is rapid in that effectors rested for only 3 d closely resemble canonical memory cells rested for 60 d or longer in the absence of antigen. This is true for both Th1 and Th2 lineages, and occurs whether CD4 T cell effectors rest in vivo or in vitro, suggesting a default pathway. We find that the effector-memory transition at the level of gene expression occurs in two stages: a rapid loss of expression of a myriad of effector-associated genes, and a more gradual gain of expression of a cohort of genes uniquely associated with memory cells rested for extended periods.


Assuntos
Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Memória Imunológica/imunologia , Animais , Biomarcadores/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Citocinas/biossíntese , Citotoxicidade Imunológica , Fluorescência , Perfilação da Expressão Gênica , Selectina L/metabolismo , Camundongos , Fenótipo , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Células Th1/citologia , Células Th1/imunologia , Células Th2/citologia , Células Th2/imunologia
14.
Biomolecules ; 12(11)2022 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-36358898

RESUMO

Central to the impacts of CD4 T cells, both positive in settings of infectious disease and cancer and negative in the settings of autoimmunity and allergy, is their ability to differentiate into distinct effector subsets with specialized functions. The programming required to support such responses is largely dictated by lineage-specifying transcription factors, often called 'master regulators'. However, it is increasingly clear that many aspects of CD4 T cell immunobiology that can determine the outcomes of disease states involve a broader transcriptional network. Eomesodermin (Eomes) is emerging as an important member of this class of transcription factors. While best studied in CD8 T cells and NK cells, an increasing body of work has focused on impacts of Eomes expression in CD4 T cell responses in an array of different settings. Here, we focus on the varied impacts reported in these studies that, together, indicate the potential of targeting Eomes expression in CD4 T cells as a strategy to improve a variety of clinical outcomes.


Assuntos
Linfócitos T CD4-Positivos , Fatores de Transcrição , Fatores de Transcrição/metabolismo , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Diferenciação Celular , Linfócitos T CD8-Positivos
15.
J Immunol ; 182(12): 7353-63, 2009 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-19494257

RESUMO

We examined the expression and influence of IL-10 during influenza infection. We found that IL-10 does not impact sublethal infection, heterosubtypic immunity, or the maintenance of long-lived influenza Ag depots. However, IL-10-deficient mice display dramatically increased survival compared with wild-type mice when challenged with lethal doses of virus, correlating with increased expression of several Th17-associated cytokines in the lungs of IL-10-deficient mice during the peak of infection, but not with unchecked inflammation or with increased cellular responses. Foxp3(-) CD4 T cell effectors at the site of infection represent the most abundant source of IL-10 in wild-type mice during high-dose influenza infection, and the majority of these cells coproduce IFN-gamma. Finally, compared with predominant Th1 responses in wild-type mice, virus-specific T cell responses in the absence of IL-10 display a strong Th17 component in addition to a strong Th1 response and we show that Th17-polarized CD4 T cell effectors can protect naive mice against an otherwise lethal influenza challenge and utilize unique mechanisms to do so. Our results show that IL-10 expression inhibits development of Th17 responses during influenza infection and that this is correlated with compromised protection during high-dose primary, but not secondary, challenge.


Assuntos
Vírus da Influenza A Subtipo H1N1/imunologia , Interleucina-10/deficiência , Interleucina-10/imunologia , Interleucina-17/imunologia , Infecções por Orthomyxoviridae/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Antígenos Virais/imunologia , Fatores de Transcrição Forkhead/imunologia , Interleucina-10/genética , Interleucina-10/metabolismo , Camundongos , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/metabolismo , Taxa de Sobrevida
16.
Adv Exp Med Biol ; 780: 57-68, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21842365

RESUMO

How memory CD4 T cells contribute to protection upon pathogen -challenge is not fully understood. Beyond traditional helper functions for CD8 T cell and B cell responses, memory CD4 T cells can have a potent impact on the character and a magnitude of inflammatory responses. Here we discuss how memory CD4 T cell control of innate immunity at early time points after pathogen encounters can influence protective responses. We also discuss important aspects of the mechanism whereby memory CD4 T cells directly and indirectly impact the activation status of antigen-presenting cells and production of inflammatory cytokines and chemokines from multiple cell types. We suggest that control of innate immune responses by the adaptive immune system is a powerful protective mechanism associated with the memory state and represents an important fail-safe in the face of pathogens that fail to trigger robust inflammatory responses through conserved pattern recognition receptors.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD4-Positivos , Imunidade Inata , Memória Imunológica , Vírus da Influenza A/fisiologia , Ativação Linfocitária/imunologia , Infecções por Orthomyxoviridae/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Citocinas/biossíntese , Citocinas/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Camundongos , Infecções por Orthomyxoviridae/virologia , Receptores de Reconhecimento de Padrão/imunologia , Receptores de Reconhecimento de Padrão/metabolismo
17.
Biosci Rep ; 41(7)2021 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-34240739

RESUMO

The serine protease prostasin is a negative regulator of lipopolysaccharide-induced inflammation and has a role in the regulation of cellular immunity. Prostasin expression in cancer cells inhibits migration and metastasis, and reduces epithelial-mesenchymal transition. Programmed death-ligand 1 (PD-L1) is a negative regulator of the immune response and its expression in cancer cells interferes with immune surveillance. The aim of the present study was to investigate if prostasin regulates PD-L1 expression. We established sublines overexpressing various forms of prostasin as well as a subline deficient for the prostasin gene from the Calu-3 human lung cancer cells. We report here that PD-L1 expression induced by interferon-γ (IFNγ) is further enhanced in cells overexpressing the wildtype membrane-anchored prostasin. The PD-L1 protein was localized on the cell surface and released into the culture medium in extracellular vesicles (EVs) with the protease-active prostasin. The epidermal growth factor-epidermal growth factor receptor (EGF-EGFR), protein kinase C (PKC), and mitogen-activated protein kinase (MAPK) participated in the prostasin-mediated up-regulation of PD-L1 expression. A Gene Set Enrichment Analysis (GSEA) of patient lung tumors in The Cancer Genome Atlas (TCGA) database revealed that prostasin and PD-L1 regulate common signaling pathways during tumorigenesis and tumor progression.


Assuntos
Adenocarcinoma de Pulmão/enzimologia , Antígeno B7-H1/metabolismo , Vesículas Extracelulares/enzimologia , Neoplasias Pulmonares/enzimologia , Serina Endopeptidases/metabolismo , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/imunologia , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Fator de Crescimento Epidérmico/farmacologia , Receptores ErbB/genética , Receptores ErbB/metabolismo , Vesículas Extracelulares/efeitos dos fármacos , Vesículas Extracelulares/genética , Vesículas Extracelulares/imunologia , Regulação Neoplásica da Expressão Gênica , Humanos , Interferon gama/farmacologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Quinase C/metabolismo , Serina Endopeptidases/genética , Transdução de Sinais , Regulação para Cima
18.
Biosci Rep ; 2021 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-34195807

RESUMO

The serine protease prostasin is a negative regulator of lipopolysaccharide-induced inflammation and has a role in the regulation of cellular immunity.  Prostasin expression in cancer cells inhibits migration and metastasis, and reduces epithelial-mesenchymal transition.  Programmed death-ligand 1 (PD-L1) is a negative regulator of the immune response and its expression in cancer cells interferes with immune surveillance.  The aim of this study was to investigate if prostasin regulates PD-L1 expression.  We established sublines over-expressing various forms of prostasin as well as a subline deficient for the prostasin gene from the Calu-3 human lung cancer cells.  We report here that PD-L1 expression induced by interferon-gamma (IFNg) is further enhanced in cells over-expressing the wild-type membrane-anchored prostasin.  The PD-L1 protein was localized on the cell surface and released into the culture medium in extracellular vesicles (EVs) with the protease-active prostasin.  The epidermal growth factor-epidermal growth factor receptor (EGF-EGFR), protein kinase C (PKC), and mitogen-activated protein kinase (MAPK) participated in the prostasin-mediated up-regulation of PD-L1 expression.  A Gene Set Enrichment Analysis (GSEA) of patient lung tumors in The Cancer Genome Atlas (TCGA) database revealed that prostasin and PD-L1 regulate common signaling pathways during tumorigenesis and tumor progression.

19.
Artigo em Inglês | MEDLINE | ID: mdl-33903157

RESUMO

We have discovered that the determination of CD4 effector and memory fates after infection is regulated not only by initial signals from antigen and pathogen recognition, but also by a second round of such signals at a checkpoint during the effector response. Signals to effectors determine their subsequent fate, inducing further progression to tissue-restricted follicular helpers, cytotoxic CD4 effectors, and long-lived memory cells. The follicular helpers help the germinal center B-cell responses that give rise to high-affinity long-lived antibody responses and memory B cells that synergize with T-cell memory to provide robust long-lived protection. We postulate that inactivated vaccines do not provide extended signals from antigen and pathogen beyond a few days, and thus elicit ineffective CD4 T- and B-cell effector responses and memory. Defining the mechanisms that underlie effective responses should provide insights necessary to develop vaccine strategies that induce more effective and durable immunity.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Memória Imunológica , Infecções/imunologia , Animais , Apresentação de Antígeno , Humanos , Vacinas contra Influenza/imunologia , Moléculas com Motivos Associados a Patógenos
20.
ACS Omega ; 6(8): 5591-5606, 2021 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-33681599

RESUMO

Nanoparticle-mediated cancer drug delivery remains an inefficient process. The protein corona formed on nanoparticles (NPs) controls their biological identity and, if optimized, could enhance cancer cell uptake. In this study, a hyperbranched polyester polymer (HBPE) was synthesized from diethyl malonate and used to generate NPs that were subsequently coated with normal sera (NS) collected from mice. Cellular uptake of NS-treated HBPE-NPs was compared to PEGylated HBPE-NPs and was assessed using MDA-MB-231 triple-negative breast cancer (TNBC) cells as well as endothelial and monocytic cell lines. NS-treated HBPE-NPs were taken up by TNBC cells more efficiently than PEGylated HBPE-NPs, while evasion of monocyte uptake was comparable. NS coatings facilitated cancer cell uptake of HBPE-NPs, even after prior interaction of the particles with an endothelial layer. NS-treated HBPE-NPs were not inherently toxic, did not induce the migration of endothelial cells that could lead to angiogenesis, and could efficiently deliver cytotoxic doses of paclitaxel (taxol) to TNBC cells. These findings suggest that HBPE-NPs may adsorb select sera proteins that improve uptake by cancer cells, and such NPs could be used to advance the discovery of novel factors that improve the bioavailability and tissue distribution of drug-loaded polymeric NPs.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA