Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nat Immunol ; 22(12): 1524-1537, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34795443

RESUMO

Inhibiting PD-1:PD-L1 signaling has transformed therapeutic immune restoration. CD4+ T cells sustain immunity in chronic infections and cancer, yet little is known about how PD-1 signaling modulates CD4+ helper T (TH) cell responses or the ability to restore CD4+ TH-mediated immunity by checkpoint blockade. We demonstrate that PD-1:PD-L1 specifically suppressed CD4+ TH1 cell amplification, prevents CD4+ TH1 cytokine production and abolishes CD4+ cytotoxic killing capacity during chronic infection in mice. Inhibiting PD-L1 rapidly restored these functions, while simultaneously amplifying and activating TH1-like T regulatory cells, demonstrating a system-wide CD4-TH1 recalibration. This effect coincided with decreased T cell antigen receptor signaling, and re-directed type I interferon (IFN) signaling networks towards dominant IFN-γ-mediated responses. Mechanistically, PD-L1 blockade specifically targeted defined populations with pre-established, but actively suppressed proliferative potential, with limited impact on minimally cycling TCF-1+ follicular helper T cells, despite high PD-1 expression. Thus, CD4+ T cells require unique differentiation and functional states to be targets of PD-L1-directed suppression and therapeutic restoration.


Assuntos
Antígeno B7-H1/antagonistas & inibidores , Inibidores de Checkpoint Imunológico/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Coriomeningite Linfocítica/tratamento farmacológico , Vírus da Coriomeningite Linfocítica/imunologia , Células Th1/efeitos dos fármacos , Transferência Adotiva , Animais , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Proliferação de Células/efeitos dos fármacos , Doença Crônica , Citocinas/metabolismo , Citotoxicidade Imunológica/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Redes Reguladoras de Genes , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/metabolismo , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Camundongos Endogâmicos C57BL , Fenótipo , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/virologia , Transcriptoma
2.
Immunity ; 55(7): 1143-1145, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35830820

RESUMO

How CD4+ T cell responses are maintained during chronic infection is unknown. In this issue of Immunity, Xia et al. (2022) identify a progenitor T cell subset that gives rise to effector and follicular helper T cells to sustain antiviral responses.


Assuntos
Infecção Persistente , Subpopulações de Linfócitos T , Linfócitos T CD4-Positivos , Humanos , Células-Tronco , Linfócitos T Auxiliares-Indutores
3.
Immunity ; 55(12): 2369-2385.e10, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36370712

RESUMO

Type I and II interferons (IFNs) stimulate pro-inflammatory programs that are critical for immune activation, but also induce immune-suppressive feedback circuits that impede control of cancer growth. Here, we sought to determine how these opposing programs are differentially induced. We demonstrated that the transcription factor interferon regulatory factor 2 (IRF2) was expressed by many immune cells in the tumor in response to sustained IFN signaling. CD8+ T cell-specific deletion of IRF2 prevented acquisition of the T cell exhaustion program within the tumor and instead enabled sustained effector functions that promoted long-term tumor control and increased responsiveness to immune checkpoint and adoptive cell therapies. The long-term tumor control by IRF2-deficient CD8+ T cells required continuous integration of both IFN-I and IFN-II signals. Thus, IRF2 is a foundational feedback molecule that redirects IFN signals to suppress T cell responses and represents a potential target to enhance cancer control.


Assuntos
Interferon Tipo I , Neoplasias , Humanos , Fator Regulador 2 de Interferon/genética , Linfócitos T CD8-Positivos , Fatores de Transcrição , Exaustão das Células T , Neoplasias/patologia
4.
Immunity ; 54(3): 526-541.e7, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33515487

RESUMO

Chronic viral infections increase severity of Mycobacterium tuberculosis (Mtb) coinfection. Here, we examined how chronic viral infections alter the pulmonary microenvironment to foster coinfection and worsen disease severity. We developed a coordinated system of chronic virus and Mtb infection that induced central clinical manifestations of coinfection, including increased Mtb burden, extra-pulmonary dissemination, and heightened mortality. These disease states were not due to chronic virus-induced immunosuppression or exhaustion; rather, increased amounts of the cytokine TNFα initially arrested pulmonary Mtb growth, impeding dendritic cell mediated antigen transportation to the lymph node and subverting immune-surveillance, allowing bacterial sanctuary. The cryptic Mtb replication delayed CD4 T cell priming, redirecting T helper (Th) 1 toward Th17 differentiation and increasing pulmonary neutrophilia, which diminished long-term survival. Temporally restoring CD4 T cell induction overcame these diverse disease sequelae to enhance Mtb control. Thus, Mtb co-opts TNFα from the chronic inflammatory environment to subvert immune-surveillance, avert early immune function, and foster long-term coinfection.


Assuntos
Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/fisiologia , Mycobacterium tuberculosis/fisiologia , Neutrófilos/imunologia , Células Th1/imunologia , Células Th17/imunologia , Tuberculose/imunologia , Imunidade Adaptativa , Animais , Doença Crônica , Coinfecção , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fagocitose , Índice de Gravidade de Doença , Fatores de Tempo
5.
Immunity ; 49(4): 678-694.e5, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30314757

RESUMO

CD8+ T cell exhaustion impedes control of chronic viral infection; yet how new T cell responses are mounted during chronic infection is unclear. Unlike T cells primed at the onset of infection that rapidly differentiate into effectors and exhaust, we demonstrate that virus-specific CD8+ T cells primed after establishment of chronic LCMV infection preferentially generate memory-like transcription factor TCF1+ cells that were transcriptionally and proteomically distinct, less exhausted, and more responsive to immunotherapy. Mechanistically, adaptations of antigen-presenting cells and diminished T cell signaling intensity promoted differentiation of the memory-like subset at the expense of rapid effector cell differentiation, which was now highly dependent on IL-21-mediated CD4+ T cell help for its functional generation. Chronic viral infection similarly redirected de novo differentiation of tumor-specific CD8+ T cells, ultimately preventing cancer control. Thus, targeting these T cell stimulatory pathways could enable strategies to control chronic infection, tumors, and enhance immunotherapeutic efficacy.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Imunidade/imunologia , Memória Imunológica/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Células Apresentadoras de Antígenos/virologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Diferenciação Celular/genética , Doença Crônica , Perfilação da Expressão Gênica/métodos , Imunidade/genética , Memória Imunológica/genética , Imunoterapia , Coriomeningite Linfocítica/terapia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/fisiologia , Camundongos Endogâmicos C57BL , Proteômica/métodos , Fator 1 de Transcrição de Linfócitos T/genética , Fator 1 de Transcrição de Linfócitos T/imunologia , Fator 1 de Transcrição de Linfócitos T/metabolismo
6.
Proc Natl Acad Sci U S A ; 117(10): 5420-5429, 2020 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-32094187

RESUMO

Chronic infection provokes alterations in inflammatory and suppressive pathways that potentially affect the function and integrity of multiple tissues, impacting both ongoing immune control and restorative immune therapies. Here we demonstrate that chronic lymphocytic choriomeningitis virus infection rapidly triggers severe thymic depletion, mediated by CD8 T cell-intrinsic type I interferon (IFN) and signal transducer and activator of transcription 2 (Stat2) signaling. Occurring temporal to T cell exhaustion, thymic cellularity reconstituted despite ongoing viral replication, with a rapid secondary thymic depletion following immune restoration by anti-programmed death-ligand 1 (PDL1) blockade. Therapeutic hematopoietic stem cell transplant (HSCT) during chronic infection generated new antiviral CD8 T cells, despite sustained virus replication in the thymus, indicating an impairment in negative selection. Consequently, low amounts of high-affinity self-reactive T cells also escaped the thymus following HSCT during chronic infection. Thus, by altering the stringency and partially impairing negative selection, the host generates new virus-specific T cells to replenish the fight against the chronic infection, but also has the potentially dangerous effect of enabling the escape of self-reactive T cells.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Interferon Tipo I/metabolismo , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/patologia , Vírus da Coriomeningite Linfocítica , Timo/patologia , Timo/virologia , Animais , Atrofia/virologia , Antígeno B7-H1/antagonistas & inibidores , Doença Crônica , Transplante de Células-Tronco Hematopoéticas , Interferon Tipo I/genética , Coriomeningite Linfocítica/terapia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fator de Transcrição STAT2/metabolismo , Transdução de Sinais , Replicação Viral
7.
Can J Surg ; 66(2): E132-E138, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36931653

RESUMO

BACKGROUND: The number of surgical residents experiencing childbearing during residency training is increasing, and there is an absence of clarity with respect to parental-leave, lactation and return-to-work policies in support of residents. The aim of this review was to examine parental-leave policies during residency training in surgery and the perceptions of these policies by residents, program directors and coresidents, as described in the literature. METHODS: We performed a scoping review of the literature based on the following themes: maternity or parental-leave policies; antepartum work-restriction policies and obstetric complications; accommodations for training absences; support for, and perceptions of, maternity or parental leave during residency training; and challenges upon return to work, namely resident performance and breastfeeding. RESULTS: Parental-leave policies during surgical residency training have historically lacked clarity and enforcement. Although recommendations for parental leave are now in place, this may have historically contributed to a lack of perceived support for surgical residents and may result in variable leave permitted to residents. Unclear policies may also contribute to career dissatisfaction among resident parents, which may deter qualified individuals from selecting surgical subspecialties. CONCLUSION: A call for a cultural shift is required to inform policies that would better support residents across all surgical specialties to pursue success in their dual roles as parents and surgeons. With increased awareness, progress in policy and guideline development is under way.


Assuntos
Internato e Residência , Humanos , Feminino , Gravidez , Inquéritos e Questionários , Licença Parental , Políticas , América do Norte , Pais
8.
J Reconstr Microsurg ; 39(4): 254-263, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-35477116

RESUMO

BACKGROUND: Breast reconstruction (BR) using autologous free flaps has been shown to have numerous psychosocial and quality-of-life benefits. Unfortunately, the microsurgical learning curve is quite steep due to some unique operative challenges. Currently, there is no realistic simulation model that captures real-life respiratory excursion and the depth of internal mammary vessels within the compact recipient site. The purpose of this study was to delineate intraoperative measurements of depth and motion, describe the resulting simulation model, and conduct a pilot study evaluating the simulator as an educational resource. METHODS: This is a single-center, ethics-approved study. For the intraoperative measurements, all consecutive patients undergoing free flap BR using internal mammary vessels as recipients were recruited. Patient and intraoperative factors as well as intraoperative measurements were recorded. A dynamic model was developed based on intraoperative parameters. For the pilot study, plastic and reconstructive surgery trainees were recruited to complete a hand-sewn internal mammary artery (IMA) anastomosis using the new simulator and completed objective questionnaires pre- and postsimulation. Subjective feedback was recorded and themes determined. RESULTS: Fifteen operative sites were analyzed. Flap pocket was found to be between 4 and 5 cm in depth with vertical excursion of 3.7 ± 1.0mm and a respiratory rate of 9 to 14 breaths/minute. Previous radiation, rib space, body mass index (BMI), blood pressure, heart rate, tidal volume, and respiratory rate showed no correlation to vessel depth/excursion. Laterality, rib space, BMI, radiation, vitals, and tidal volume had no correlation with vessel movement. Twenty-two trainees were included in the pilot. An increase in confidence and mixed results for anxiety was reported. CONCLUSION: This study reports a novel microsurgical simulation model that provides a realistic deep inferior epigastric perforator free flap BR IMA anastomosis experience. It replicates movement of vessels in situ with real-time respiratory excursion and similar physical structures of the internal mammary system. This model shows promising results for increased use in microsurgical education.


Assuntos
Retalhos de Tecido Biológico , Mamoplastia , Artéria Torácica Interna , Retalho Perfurante , Humanos , Artéria Torácica Interna/cirurgia , Projetos Piloto , Microcirurgia/métodos , Mamoplastia/métodos , Retalhos de Tecido Biológico/irrigação sanguínea , Artérias Epigástricas/cirurgia , Respiração , Retalho Perfurante/irrigação sanguínea , Estudos Retrospectivos
9.
Trends Immunol ; 38(8): 542-557, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28579323

RESUMO

Type I interferons (IFN-Is) are emerging as key drivers of inflammation and immunosuppression in chronic infection. Control of these infections requires IFN-I signaling; however, prolonged IFN-I signaling can lead to immune dysfunction. IFN-Is are also emerging as double-edged swords in cancer, providing necessary inflammatory signals, while initiating feedback suppression in both immune and cancer cells. Here, we review the proinflammatory and suppressive mechanisms potentiated by IFN-Is during chronic virus infections and discuss the similar, newly emerging dichotomy in cancer. We then discuss how this understanding is leading to new therapeutic concepts and immunotherapy combinations. We propose that, by modulating the immune response at its foundation, it may be possible to widely reshape immunity to control these chronic diseases.


Assuntos
Interferon Tipo I/imunologia , Neoplasias/imunologia , Transdução de Sinais/fisiologia , Viroses/imunologia , Animais , Antineoplásicos/uso terapêutico , Antivirais/uso terapêutico , Doença Crônica , Humanos , Tolerância Imunológica/imunologia , Inflamação/etiologia , Inflamação/imunologia , Interferon Tipo I/uso terapêutico , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Viroses/complicações , Viroses/tratamento farmacológico
10.
PLoS Pathog ; 12(1): e1005356, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26808628

RESUMO

Persistent viral infections are simultaneously associated with chronic inflammation and highly potent immunosuppressive programs mediated by IL-10 and PDL1 that attenuate antiviral T cell responses. Inhibiting these suppressive signals enhances T cell function to control persistent infection; yet, the underlying signals and mechanisms that program immunosuppressive cell fates and functions are not well understood. Herein, we use lymphocytic choriomeningitis virus infection (LCMV) to demonstrate that the induction and functional programming of immunosuppressive dendritic cells (DCs) during viral persistence are separable mechanisms programmed by factors primarily considered pro-inflammatory. IFNγ first induces the de novo development of naive monocytes into DCs with immunosuppressive potential. Type I interferon (IFN-I) then directly targets these newly generated DCs to program their potent T cell immunosuppressive functions while simultaneously inhibiting conventional DCs with T cell stimulating capacity. These mechanisms of monocyte conversion are constant throughout persistent infection, establishing a system to continuously interpret and shape the immunologic environment. MyD88 signaling was required for the differentiation of suppressive DCs, whereas inhibition of stimulatory DCs was dependent on MAVS signaling, demonstrating a bifurcation in the pathogen recognition pathways that promote distinct elements of IFN-I mediated immunosuppression. Further, a similar suppressive DC origin and differentiation was also observed in Mycobacterium tuberculosis infection, HIV infection and cancer. Ultimately, targeting the underlying mechanisms that induce immunosuppression could simultaneously prevent multiple suppressive signals to further restore T cell function and control persistent infections.


Assuntos
Células Dendríticas/imunologia , Tolerância Imunológica/imunologia , Interferons/imunologia , Viroses/imunologia , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , HIV , Infecções por HIV/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/imunologia , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real , Linfócitos T/imunologia , Tuberculose/imunologia
11.
Proc Natl Acad Sci U S A ; 111(20): 7409-14, 2014 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-24799699

RESUMO

CD4 T cells are central to orchestrate, sustain, and potentially regenerate antiviral immunity throughout persistent viral infections. Although the evolving immune environment during persistent infection reshapes established CD4 T-cell responses, the fate of naïve CD4 T cells primed in the midst of persistent infection is unclear. We demonstrate that, in marked contrast to the onset of infection, virus-specific CD4 T cells primed during an established persistent infection have diminished ability to develop Th1 responses, to efficiently accumulate in peripheral tissues, and almost exclusively differentiate into T follicular helper cells. Consistent with suppressed Th1 and heightened Tfh differentiation, virus-specific CD4 T cells primed during the established persistent infection provide help to B cells, but only limited help to CD8 T cells. The suppression of de novo Th1 generation and tissue distribution was mediated by chronic type I IFN (IFN-I) production and was effectively restored by blocking IFN-I signaling during CD4 T-cell priming. Thus, we establish a suppressive function of chronic IFN-I signaling and mechanism of immunoregulation during an established persistent virus infection.


Assuntos
Infecções por Arenaviridae/imunologia , Linfócitos T CD4-Positivos/virologia , Regulação da Expressão Gênica , Interferon Tipo I/metabolismo , Células Th1/virologia , Animais , Linfócitos B/imunologia , Linfócitos B/virologia , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular , Terapia de Imunossupressão , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores de Interferon/metabolismo , Transdução de Sinais , Células Th1/imunologia , Distribuição Tecidual
12.
Environ Manage ; 60(3): 383-395, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28577051

RESUMO

Riparian meadows occupy a small proportion of the public lands in the western United States but they provide numerous ecosystem services, including the production of high-quality forage for livestock grazing. Modern conservation management strategies (e.g., reductions in livestock stocking rates and adoption of new riparian grazing standards) have been implemented to better balance riparian conservation and livestock production objectives on publicly managed lands. We examined potential relationships between long-term changes in plant community, livestock grazing pressure and environmental conditions at two spatial scales in meadows grazed under conservation management strategies. Changes in plant community were not associated with either livestock stocking rate or precipitation at the grazing allotment (i.e., administrative) scale. Alternatively, both grazing pressure and precipitation had significant, albeit modest, associations with changes in plant community at the meadow (i.e., ecological site) scale. These results suggest that reductions in stocking rate have improved the balance between riparian conservation and livestock production goals. However, associations between elevation, site wetness, precipitation, and changes in plant community suggest that changing climate conditions (e.g., reduced snowpack and changes in timing of snowmelt) could trigger shifts in plant communities, potentially impacting both conservation and agricultural services (e.g., livestock and forage production). Therefore, adaptive, site-specific management strategies are required to meet grazing pressure limits and safeguard ecosystem services within individual meadows, especially under more variable climate conditions.


Assuntos
Criação de Animais Domésticos/métodos , Conservação dos Recursos Naturais/métodos , Pradaria , Gado/crescimento & desenvolvimento , Animais , Mudança Climática , Ecossistema , Herbivoria , Estados Unidos
13.
J Reconstr Microsurg ; 33(6): 381-388, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28399607

RESUMO

Background This study examined the need for improved training in the identification and management of free flap (FF) compromise and assessed a potential role for simulated scenario training. Methods Online needs assessment surveys were completed by plastic surgeons and a subsample with expertise in microsurgery education participated in focus groups. Data were analyzed using descriptive statistics and mixed qualitative methods. Results In this study, 77 surgeons completed surveys and 11 experts participated in one of two focus groups. Forty-nine (64%) participants were educators, 65 and 45% of which reported having an insufficient volume of FF cases to adequately teach the management and identification of compromise, respectively. Forty-three percent of educators felt that graduating residents are not adequately prepared to manage FF compromise independently. Exposure to normal and abnormal FF cases was felt to be critical for effective training by focus group participants. Experts identified low failure rates, communication issues, and challenging teaching conditions as current barriers to training. Most educators (74%) felt that simulated scenario training would be "very useful" or "extremely useful" to current residents. Focus groups highlighted the need for a widely accepted algorithm for re-exploration and salvage on which to base the development of a training adjunct consisting of simulated scenarios. Conclusion Trainee exposure to FF compromise is inadequate in existing plastic surgery programs. Early exposure, high case volume, and a standardized algorithmic approach to management with a focus on decision making may improve training. Simulated scenario training may be valuable in addressing current barriers.


Assuntos
Competência Clínica/normas , Educação Médica Continuada/normas , Retalhos de Tecido Biológico , Rejeição de Enxerto/prevenção & controle , Microcirurgia/educação , Avaliação das Necessidades , Cirurgiões , Cirurgia Plástica/educação , Algoritmos , Atitude do Pessoal de Saúde , Canadá , Simulação por Computador , Retalhos de Tecido Biológico/transplante , Humanos , Microcirurgia/métodos , Projetos Piloto , Estudos Prospectivos , Cirurgiões/normas , Cirurgia Plástica/normas
14.
J Immunol ; 190(9): 4627-39, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23536631

RESUMO

The TNFR family member 4-1BB plays a key role in the survival of activated and memory CD8 T cells. However, the mechanisms that regulate 4-1BB re-expression on memory CD8 T cells after Ag clearance are unknown. In unimmunized mice, ∼10% of CD8 CD44(hi) memory T cells in the bone marrow (BM) and liver express 4-1BB, with minimal 4-1BB expression in spleen and lymph node. IL-2, IL-15, and IL-7 are collectively dispensable for 4-1BB expression on the memory CD8 T cells. Rather, T cell-intrinsic glucocorticoid-induced TNFR-related protein (GITR) contributes to 4-1BB expression on CD8 T cells upon their entry into the BM or liver. Consistent with its role in regulation of 4-1BB, GITR is required on memory CD8 T cells for their persistence in vivo. These findings reveal site-specific effects of the BM and liver microenvironment on CD8 memory T cells. Previous work has demonstrated that 4-1BB agonists given to unimmunized mice induce splenomegaly, hepatitis, and other immune system anomalies. Moreover, severe liver pathology has been observed in a subset of anti-4-1BB-treated melanoma patients. Remarkably, the absence of GITR in mice almost completely abrogates cellular expansions, splenomegaly, and liver inflammation associated with anti-4-1BB agonist treatment of unimmunized mice. In contrast, lack of CD8 T cells selectively improves liver pathology, but not splenomegaly in the mice. Thus, the regulation of 4-1BB expression by GITR on CD8 T cells, as well as on other cells, contributes to the pathological effects of anti-4-1BB in unimmunized mice.


Assuntos
Ligante 4-1BB/imunologia , Ligante 4-1BB/metabolismo , Linfócitos T CD8-Positivos/imunologia , Proteína Relacionada a TNFR Induzida por Glucocorticoide/imunologia , Proteína Relacionada a TNFR Induzida por Glucocorticoide/metabolismo , Memória Imunológica/imunologia , Animais , Medula Óssea/imunologia , Medula Óssea/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Hepatite/imunologia , Hepatite/metabolismo , Humanos , Receptores de Hialuronatos/imunologia , Receptores de Hialuronatos/metabolismo , Interleucinas/imunologia , Interleucinas/metabolismo , Fígado/imunologia , Fígado/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Melanoma/imunologia , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Baço/imunologia , Baço/metabolismo , Esplenomegalia/imunologia , Esplenomegalia/metabolismo
15.
Immunol Rev ; 244(1): 197-217, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22017440

RESUMO

GITR [glucocorticoid inducible tumor necrosis factor receptor (TNFR)-related protein] and 4-1BB are costimulatory TNFR family members that are expressed on regulatory and effector T cells as well as on other cells of the immune system. Here we discuss the role of GITR and 4-1BB on T cells during viral infections and in cancer immunotherapy. Systemic treatment with agonistic anti-4-1BB antibody leads to a number of immune system abnormalities, and clinical trials of anti-4-1BB have been terminated. However, other modes of 4-1BB ligation may be less toxic. To date, similar toxicities have not been reported for anti-GITR treatment of mice, although anti-GITR antibodies can exacerbate mouse autoimmune models. Intrinsic effects of GITR and 4-1BB on effector T cells appear to predominate over their effects on other cell types in some models. Despite their similarities in enhancing T-cell survival, 4-1BB and GITR are clearly not redundant, and both pathways are required for maximal CD8(+) T-cell responses and mouse survival following severe respiratory influenza infection. GITR uses TNFR-associated factor (TRAF) 2 and TRAF5, whereas 4-1BB recruits TRAF1 and TRAF2 to mediate survival signaling in T cells. The differential use of signaling adapters combined with their differential expression may explain the non-redundant roles of GITR and 4-1BB in the immune system.


Assuntos
Anticorpos/imunologia , Proteína Relacionada a TNFR Induzida por Glucocorticoide/imunologia , Imunidade Inata , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/imunologia , Animais , Anticorpos/administração & dosagem , Autoimunidade/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Expressão Gênica/imunologia , Proteína Relacionada a TNFR Induzida por Glucocorticoide/antagonistas & inibidores , Proteína Relacionada a TNFR Induzida por Glucocorticoide/genética , Proteína Relacionada a TNFR Induzida por Glucocorticoide/metabolismo , Humanos , Imunidade Inata/efeitos dos fármacos , Imunoterapia/métodos , Vírus da Influenza A/imunologia , Camundongos , Camundongos Knockout , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/metabolismo , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/genética , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo
16.
J Immunol ; 188(12): 5915-23, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22581858

RESUMO

CD8 memory T cells are enriched in the bone marrow, a site where these cells are thought to receive homeostatic signals. However, the primary site where CD8 memory T cells receive their cytokine-induced homeostatic signals has recently come under debate. In this study, we demonstrate that the bone marrow contains a fraction of CD8 memory phenotype T cells with elevated expression of glucocorticoid-induced TNFR-related protein (GITR). In contrast, splenic and lymph node memory phenotype T cells have GITR levels similar to those on naive T cells. The bone marrow GITR(hi) memory T cells have a phenotype indicative of cytokine activation, with higher CD122 and lower CD127 than do the GITR(basal) memory T cells. Remarkably, these bone marrow-specific GITR(hi) cells are almost completely ablated in the absence of IL-15, whereas TNFR2 and 4-1BB expression on the CD8 memory T cells are IL-15 independent. Furthermore, adoptively transferred splenic CD8 memory phenotype T cells show IL-15-dependent GITR upregulation upon entry into the bone marrow. This result implies that the selective appearance of GITR(hi) memory phenotype T cells in the bone marrow reflects the local microenvironment rather than a different subset of memory T cells. GITR(-/-) mice have a lower frequency of CD8 memory phenotype cells in the bone marrow, yet the GITR(-/-) cells hyperproliferate compared with those in wild-type mice. Taken together, these data suggest that GITR plays a role in the survival of CD8 memory phenotype T cells and that GITR upregulation represents a precise marker of cells that have responded to IL-15.


Assuntos
Medula Óssea/imunologia , Linfócitos T CD8-Positivos/imunologia , Proteína Relacionada a TNFR Induzida por Glucocorticoide/imunologia , Memória Imunológica/imunologia , Interleucina-15/imunologia , Transferência Adotiva , Animais , Medula Óssea/metabolismo , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/metabolismo , Citometria de Fluxo , Proteína Relacionada a TNFR Induzida por Glucocorticoide/metabolismo , Homeostase/imunologia , Interleucina-15/metabolismo , Linfonodos/citologia , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Baço/citologia , Baço/imunologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Regulação para Cima
17.
J Biol Chem ; 287(27): 23010-9, 2012 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-22570473

RESUMO

T cells lacking TRAF1 hyperproliferate in response to T cell receptor signaling but have impaired signaling downstream of specific TNFR family members such as 4-1BB. Here we resolve this paradox by showing that while TRAF1 is required for maximal activation of the classical NF-κB pathway downstream of 4-1BB in primary T cells, TRAF1 also restricts the constitutive activation of NIK in anti-CD3-activated T cells. Activation of the alternative NF-κB pathway is restricted in unstimulated cells by a cIAP1/2:TRAF2:TRAF3:NIK complex. Using knockdown of NIK by siRNA we show that in activated CD8 T cells TRAF1 is also involved in this process and that constitutive activation of the alternative NF-κB pathway is responsible for costimulation independent hyperproliferation and excess cytokine production in TRAF1-deficient CD8 T cells compared with WT CD8 T cells. The T cell costimulatory molecule 4-1BB critically regulates the survival of activated and memory CD8 T cells. We demonstrate that stimulation through 4-1BB induces cIAP1-dependent TRAF3 degradation and activation of the alternative NF-κB pathway. We also show that while both TRAF1 and cIAP1 have non-redundant roles in suppressing the alternative NF-κB pathway in T cells activated in the absence of costimulation, activation of the classical NF-κB pathway downstream of 4-1BB requires TRAF1, whereas cIAP1 plays a redundant role with cIAP2. Collectively these results demonstrate that TRAF1 plays a critical role in regulating T cell activation both through restricting the costimulation independent activation of NIK in activated T cells and by promoting the 4-1BB-induced classical NF-κB pathway.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/imunologia , Fator 1 Associado a Receptor de TNF/imunologia , Fator 1 Associado a Receptor de TNF/metabolismo , Animais , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/imunologia , Divisão Celular/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Fator 1 Associado a Receptor de TNF/genética , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo , Quinase Induzida por NF-kappaB
18.
Eur J Immunol ; 42(11): 2861-74, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22886791

RESUMO

The persistence of memory lymphocytes is a critical feature of adaptive immunity. The TNF family ligand 4-1BBL supports the antigen-independent survival of CD8⁺ memory T cells. Here, we show that mice lacking 4-1BB only on αß T cells show a similar defect in CD8⁺ T-cell recall responses, as previously shown in 4-1BBL-deficient mice. We show that 4-1BB is selectively expressed on BM CD8⁺ but not CD4⁺ memory T cells of unimmunized mice. Its ligand, 4-1BBL, is found on VCAM-1⁺ stromal cells, CD11c⁺ cells, and a Gr1(lo) myeloid population in unimmunized mice. Adoptive transfer of in vitro generated memory T cells into mice lacking 4-1BBL only on radioresistant cells recapitulates the defect in CD8⁺ T-cell survival seen in the complete knockout mice, with smaller effects of 4-1BBL on hematopoietic cells. In BM, adoptively transferred DsRed CD8⁺ memory T cells are most often found in proximity to VCAM-1⁺ cells or Gr1⁺ cells, followed by B220⁺ cells and to a much lesser extent near CD11c⁺ cells. Thus, a VCAM-1⁺CD45(-) stromal cell is a plausible candidate for the radioresistant cell that provides 4-1BBL to CD8⁺ memory T cells in the BM.


Assuntos
Ligante 4-1BB/imunologia , Células da Medula Óssea/imunologia , Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Molécula 1 de Adesão de Célula Vascular/imunologia , Ligante 4-1BB/genética , Transferência Adotiva , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos da radiação , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos da radiação , Feminino , Citometria de Fluxo , Imuno-Histoquímica , Memória Imunológica/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , RNA Viral/química , RNA Viral/genética , Tolerância a Radiação/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Organismos Livres de Patógenos Específicos , Quimeras de Transplante
19.
bioRxiv ; 2023 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-37961130

RESUMO

Biological sex plays an integral role in the immune response to various pathogens. The underlying basis for these sex differences is still not well defined. Here, we show that Coxsackievirus B3 (CVB3) induces a viral-specific CD4 + T cell response that can protect female mice from mortality. We found that CVB3 can induce expansion of CD62L lo CD4 + T cells in the mesenteric lymph node and spleen of female but not male mice as early as 5 days post-inoculation, indicative of activation. Using a recombinant CVB3 virus expressing a model CD4 + T cell epitope, we found that this response is due to viral antigen and not bystander activation. Finally, the depletion of CD4 + T cells before infection increased mortality in female mice, indicating that CD4 + T cells play a protective role against CVB3 in our model. Overall, these data demonstrated that CVB3 can induce an early CD4 response in female but not male mice and further emphasize how sex differences in immune responses to pathogens affect disease outcomes.

20.
Front Immunol ; 14: 1327384, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38274806

RESUMO

Background: Biological sex plays an integral role in the immune response to various pathogens. The underlying basis for these sex differences is still not well defined. Here, we show that Coxsackievirus B3 (CVB3) induces a viral-specific CD4+ T cell response that can protect female mice from mortality. Methods: We inoculated C57BL/6 Ifnar-/- mice with CVB3. We investigated the T cell response in the spleen and mesenteric lymph nodes in male and female mice following infection. Results: We found that CVB3 can induce expansion of CD62Llo CD4+ T cells in the mesenteric lymph node and spleen of female but not male mice as early as 5 days post-inoculation, indicative of activation. Using a recombinant CVB3 virus expressing a model CD4+ T cell epitope, we found that this response is due to viral antigen and not bystander activation. Finally, the depletion of CD4+ T cells before infection increased mortality in female mice, indicating that CD4+ T cells play a protective role against CVB3 in our model. Conclusions: Overall, these data demonstrated that CVB3 can induce an early CD4 response in female but not male mice and further emphasize how sex differences in immune responses to pathogens affect disease.


Assuntos
Infecções por Coxsackievirus , Enterovirus Humano B , Feminino , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T CD4-Positivos , Antígenos Virais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA