Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 188
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Immunol ; 34: 65-92, 2016 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-26666651

RESUMO

T cell responses display two key characteristics. First, a small population of epitope-specific naive T cells expands by several orders of magnitude. Second, the T cells within this proliferating population take on diverse functional and phenotypic properties that determine their ability to exert effector functions and contribute to T cell memory. Recent technological advances in lineage tracing allow us for the first time to study these processes in vivo at single-cell resolution. Here, we summarize resulting data demonstrating that although epitope-specific T cell responses are reproducibly similar at the population level, expansion potential and diversification patterns of the offspring derived from individual T cells are highly variable during both primary and recall immune responses. In spite of this stochastic response variation, individual memory T cells can serve as adult stem cells that provide robust regeneration of an epitope-specific tissue through population averaging. We discuss the relevance of these findings for T cell memory formation and clinical immunotherapy.


Assuntos
Células-Tronco Adultas/imunologia , Diferenciação Celular , Imunoterapia/métodos , Análise de Célula Única/métodos , Linfócitos T/imunologia , Animais , Biodiversidade , Linhagem da Célula , Proliferação de Células , Diversidade Cultural , Epitopos de Linfócito T/imunologia , Epitopos de Linfócito T/metabolismo , Humanos , Memória Imunológica , Ativação Linfocitária
2.
Cell ; 184(5): 1245-1261.e21, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33636132

RESUMO

How early events in effector T cell (TEFF) subsets tune memory T cell (TMEM) responses remains incompletely understood. Here, we systematically investigated metabolic factors in fate determination of TEFF and TMEM cells using in vivo pooled CRISPR screening, focusing on negative regulators of TMEM responses. We found that amino acid transporters Slc7a1 and Slc38a2 dampened the magnitude of TMEM differentiation, in part through modulating mTORC1 signaling. By integrating genetic and systems approaches, we identified cellular and metabolic heterogeneity among TEFF cells, with terminal effector differentiation associated with establishment of metabolic quiescence and exit from the cell cycle. Importantly, Pofut1 (protein-O-fucosyltransferase-1) linked GDP-fucose availability to downstream Notch-Rbpj signaling, and perturbation of this nutrient signaling axis blocked terminal effector differentiation but drove context-dependent TEFF proliferation and TMEM development. Our study establishes that nutrient uptake and signaling are key determinants of T cell fate and shape the quantity and quality of TMEM responses.


Assuntos
Aminoácidos/metabolismo , Linfócitos T CD8-Positivos/citologia , Memória Imunológica , Transdução de Sinais , Sistemas de Transporte de Aminoácidos/metabolismo , Animais , Linfócitos T CD8-Positivos/imunologia , Sistemas CRISPR-Cas , Ciclo Celular , Diferenciação Celular , Modelos Animais de Doenças , Feminino , Técnicas de Introdução de Genes , Coriomeningite Linfocítica/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Células Precursoras de Linfócitos T/citologia
3.
Immunity ; 56(7): 1664-1680.e9, 2023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-37392736

RESUMO

Memory CD8+ T cells can be broadly divided into circulating (TCIRCM) and tissue-resident memory T (TRM) populations. Despite well-defined migratory and transcriptional differences, the phenotypic and functional delineation of TCIRCM and TRM cells, particularly across tissues, remains elusive. Here, we utilized an antibody screening platform and machine learning prediction pipeline (InfinityFlow) to profile >200 proteins in TCIRCM and TRM cells in solid organs and barrier locations. High-dimensional analyses revealed unappreciated heterogeneity within TCIRCM and TRM cell lineages across nine different organs after either local or systemic murine infection models. Additionally, we demonstrated the relative effectiveness of strategies allowing for the selective ablation of TCIRCM or TRM populations across organs and identified CD55, KLRG1, CXCR6, and CD38 as stable markers for characterizing memory T cell function during inflammation. Together, these data and analytical framework provide an in-depth resource for memory T cell classification in both steady-state and inflammatory conditions.


Assuntos
Linfócitos T CD8-Positivos , Células T de Memória , Camundongos , Animais , Linhagem da Célula , Memória Imunológica
4.
Immunity ; 56(6): 1320-1340.e10, 2023 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-37315535

RESUMO

CD8+ T cell exhaustion (Tex) limits disease control during chronic viral infections and cancer. Here, we investigated the epigenetic factors mediating major chromatin-remodeling events in Tex-cell development. A protein-domain-focused in vivo CRISPR screen identified distinct functions for two versions of the SWI/SNF chromatin-remodeling complex in Tex-cell differentiation. Depletion of the canonical SWI/SNF form, BAF, impaired initial CD8+ T cell responses in acute and chronic infection. In contrast, disruption of PBAF enhanced Tex-cell proliferation and survival. Mechanistically, PBAF regulated the epigenetic and transcriptional transition from TCF-1+ progenitor Tex cells to more differentiated TCF-1- Tex subsets. Whereas PBAF acted to preserve Tex progenitor biology, BAF was required to generate effector-like Tex cells, suggesting that the balance of these factors coordinates Tex-cell subset differentiation. Targeting PBAF improved tumor control both alone and in combination with anti-PD-L1 immunotherapy. Thus, PBAF may present a therapeutic target in cancer immunotherapy.


Assuntos
Linfócitos T CD8-Positivos , Montagem e Desmontagem da Cromatina , Cromatina , Diferenciação Celular , Epigênese Genética
5.
Cell ; 171(2): 398-413.e21, 2017 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-28942919

RESUMO

A fundamental challenge in immunology is to decipher the principles governing immune responses at the whole-organism scale. Here, using a comparative infection model, we observe immune signal propagation within and between organs to obtain a dynamic map of immune processes at the organism level. We uncover two inter-organ mechanisms of protective immunity mediated by soluble and cellular factors. First, analyzing ligand-receptor connectivity across tissues reveals that type I IFNs trigger a whole-body antiviral state, protecting the host within hours after skin vaccination. Second, combining parabiosis, single-cell analyses, and gene knockouts, we uncover a multi-organ web of tissue-resident memory T cells that functionally adapt to their environment to stop viral spread across the organism. These results have implications for manipulating tissue-resident memory T cells through vaccination and open up new lines of inquiry for the analysis of immune responses at the organism level.


Assuntos
Memória Imunológica , Interferon Tipo I/imunologia , Vaccinia virus/fisiologia , Vacínia/imunologia , Vacínia/prevenção & controle , Vacinas Virais/imunologia , Administração Cutânea , Animais , Feminino , Perfilação da Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Organismos Livres de Patógenos Específicos , Linfócitos T/imunologia , Vacinas Virais/administração & dosagem
6.
Immunity ; 50(5): 1249-1261.e5, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-30952606

RESUMO

Regulated activation of the cytokine TGF-ß by integrins αvß6 and αvß8 expressed on keratinocytes is required for residence of epidermal-resident memory T cells, but whether skin-derived signals also affect recirculating memory cells in the skin remains unclear. Here, we show that after resolution of skin vaccinia virus (VV) infection, antigen-specific circulating memory CD8+ T cells migrated into skin. In mice lacking αvß6 and αvß8 integrins (Itgb6-/-Itgb8fl/fl-K14-cre), the absence of epidermal-activated TGF-ß resulted in a gradual loss of E- or P-selectin-binding central and peripheral memory populations, which were rescued when skin entry was inhibited. Skin recirculating memory cells were required for optimal host defense against skin VV infection. These data demonstrate that skin migration can persist after resolution of local skin infection and that the cytokine environment within this nonlymphoid tissue shapes the differentiation state and persistence of the central and peripheral memory-T-cell pool.


Assuntos
Antígenos de Neoplasias/metabolismo , Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/imunologia , Integrinas/metabolismo , Queratinócitos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Vaccinia virus/imunologia , Animais , Antígenos de Neoplasias/genética , Linfócitos T CD8-Positivos/enzimologia , Diferenciação Celular/imunologia , Citocinas/imunologia , Ativação Enzimática , Feminino , Integrinas/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pele/citologia , Pele/imunologia
7.
Immunity ; 50(1): 91-105.e4, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30638736

RESUMO

Memory CD4+ T cells mediate long-term immunity, and their generation is a key objective of vaccination strategies. However, the transcriptional circuitry controlling the emergence of memory cells from early CD4+ antigen-responders remains poorly understood. Here, using single-cell RNA-seq to study the transcriptome of virus-specific CD4+ T cells, we identified a gene signature that distinguishes potential memory precursors from effector cells. We found that both that signature and the emergence of memory CD4+ T cells required the transcription factor Thpok. We further demonstrated that Thpok cell-intrinsically protected memory cells from a dysfunctional, effector-like transcriptional program, similar to but distinct from the exhaustion pattern of cells responding to chronic infection. Mechanistically, Thpok- bound genes encoding the transcription factors Blimp1 and Runx3 and acted by antagonizing their expression. Thus, a Thpok-dependent circuitry promotes both memory CD4+ T cells' differentiation and functional fitness, two previously unconnected critical attributes of adaptive immunity.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Subpopulações de Linfócitos T/fisiologia , Fatores de Transcrição/metabolismo , Animais , Antígenos Virais/imunologia , Diferenciação Celular , Células Cultivadas , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Humanos , Memória Imunológica/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 1 de Ligação ao Domínio I Regulador Positivo/metabolismo , Ligação Proteica , Análise de Sequência de RNA , Análise de Célula Única , Fatores de Transcrição/genética , Transcriptoma
8.
Semin Immunol ; 69: 101800, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37494738

RESUMO

The defense against infectious diseases, either through natural immunity or after vaccinations, relies on the generation and maintenance of protective T cell memory. Naïve T cells are at the center of memory T cell generation during primary responses. Upon activation, they undergo a complex, highly regulated differentiation process towards different functional states. Naïve T cells maintained into older age have undergone epigenetic adaptations that influence their fate decisions during differentiation. We review age-sensitive, molecular pathways and gene regulatory networks that bias naïve T cell differentiation towards effector cell generation at the expense of memory and Tfh cells. As a result, T cell differentiation in older adults is associated with release of bioactive waste products into the microenvironment, higher stress sensitivity as well as skewing towards pro-inflammatory signatures and shorter life spans. These maladaptations not only contribute to poor vaccine responses in older adults but also fuel a more inflammatory state.


Assuntos
Memória Imunológica , Linfócitos T , Humanos , Idoso , Diferenciação Celular , Envelhecimento , Ativação Linfocitária , Linfócitos T CD8-Positivos
9.
Proc Natl Acad Sci U S A ; 121(13): e2306763121, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38498711

RESUMO

Lactate-proton symporter monocarboxylate transporter 1 (MCT1) facilitates lactic acid export from T cells. Here, we report that MCT1 is mandatory for the development of virus-specific CD8+ T cell memory. MCT1-deficient T cells were exposed to acute pneumovirus (pneumonia virus of mice, PVM) or persistent γ-herpesvirus (Murid herpesvirus 4, MuHV-4) infection. MCT1 was required for the expansion of virus-specific CD8+ T cells and the control of virus replication in the acute phase of infection. This situation prevented the subsequent development of virus-specific T cell memory, a necessary step in containing virus reactivation during γ-herpesvirus latency. Instead, persistent active infection drove virus-specific CD8+ T cells toward functional exhaustion, a phenotype typically seen in chronic viral infections. Mechanistically, MCT1 deficiency sequentially impaired lactic acid efflux from activated CD8+ T cells, caused an intracellular acidification inhibiting glycolysis, disrupted nucleotide synthesis in the upstream pentose phosphate pathway, and halted cell proliferation which, ultimately, promoted functional CD8+ T cell exhaustion instead of memory development. Taken together, our data demonstrate that MCT1 expression is mandatory for inducing T cell memory and controlling viral infection by CD8+ T cells.


Assuntos
Linfócitos T CD8-Positivos , Transportadores de Ácidos Monocarboxílicos , Simportadores , Animais , Camundongos , Transporte Biológico , Linfócitos T CD8-Positivos/metabolismo , Ácido Láctico/metabolismo , Simportadores/genética , Simportadores/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo
10.
Immunol Rev ; 316(1): 136-159, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37038909

RESUMO

Upon resolution of an acute viral infection, during latent-reactivating infection and during chronic active infections virus-specific T-cells differentiate into distinct subsets that differ in phenotype, longevity, transcriptional, metabolic, and epigenetic profiles, and effector functions. With recent advances in single-cell profiling, this substantial heterogeneity has become apparent and new subsets of virus-specific T cells, either of stable or transitory nature, are being identified. A unifying principle of T cells emerging in these different conditions is their precursor-progeny relationship. For acute and resolved viral infections, this relationship becomes apparent during re-challenge, whereas a constant differentiation of progenitor T cells into more differentiated cells occurs during latent-reactivating and active chronic viral infections. In this review, we summarize and discuss current knowledge about T-cell heterogeneity and progenitor-progeny relationships in the setting of persistent viral infections.


Assuntos
Linfócitos T CD8-Positivos , Viroses , Humanos , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular , Fenótipo , Memória Imunológica
11.
Trends Immunol ; 44(9): 663-664, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37591711

RESUMO

Human tissue-resident memory T (TRM) cells seeded early in life undergo an age-associated functional maturation and residency acquisition throughout childhood.


Assuntos
Envelhecimento , Células T de Memória , Criança , Humanos , Especificidade de Órgãos
12.
Proc Natl Acad Sci U S A ; 120(3): e2205049120, 2023 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-36634134

RESUMO

Stimulator of interferon genes (STING) signaling has been extensively studied in inflammatory diseases and cancer, while its role in T cell responses to infection is unclear. Using Listeria monocytogenes strains engineered to induce different levels of c-di-AMP, we found that high STING signals impaired T cell memory upon infection via increased Bim levels and apoptosis. Unexpectedly, reduction of TCR signal strength or T cell-STING expression decreased Bim expression, T cell apoptosis, and recovered T cell memory. We found that TCR signal intensity coupled STING signal strength to the unfolded protein response (UPR) and T cell survival. Under strong STING signaling, Indoleamine-pyrrole 2,3-dioxygenase (IDO) inhibition also reduced apoptosis and led to a recovery of T cell memory in STING sufficient CD8 T cells. Thus, STING signaling regulates CD8 T cell memory fitness through both cell-intrinsic and extrinsic mechanisms. These studies provide insight into how IDO and STING therapies could improve long-term T cell protective immunity.


Assuntos
Receptores de Antígenos de Linfócitos T , Transdução de Sinais , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T CD8-Positivos , Células T de Memória , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo
13.
Proc Natl Acad Sci U S A ; 120(1): e2210490120, 2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36574651

RESUMO

γδ T cells are involved in the control of Staphylococcus aureus infection, but their importance in protection compared to other T cells is unclear. We used a mouse model of systemic S. aureus infection associated with high bacterial load and persistence in the kidney. Infection caused fulminant accumulation of γδ T cells in the kidney. Renal γδ T cells acquired tissue residency and were maintained in high numbers during chronic infection. At day 7, up to 50% of renal γδ T cells produced IL-17A in situ and a large fraction of renal γδ T cells remained IL-17A+ during chronic infection. Controlled depletion revealed that γδ T cells restricted renal S. aureus replication in the acute infection and provided protection during chronic renal infection and upon reinfection. Our results demonstrate that kidney-resident γδ T cells are nonredundant in limiting local S. aureus growth during chronic infection and provide enhanced protection against reinfection.


Assuntos
Interleucina-17 , Infecções Estafilocócicas , Camundongos , Animais , Staphylococcus aureus , Receptores de Antígenos de Linfócitos T gama-delta , Infecção Persistente , Reinfecção , Rim , Camundongos Endogâmicos C57BL
14.
J Virol ; 98(4): e0191223, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38501661

RESUMO

The corona virus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome corona-virus 2 (SARS-CoV-2) spurred a worldwide race for the development of an efficient vaccine. Various strategies were pursued; however, the first vaccines to be licensed presented the SARS-CoV-2 spike protein either in the context of a non-replicating adenoviral vector or as an mRNA construct. While short-term efficacies have extensively been characterized, the duration of protection, the need for repeated boosting, and reasonable vaccination intervals have yet to be defined. We here describe the adaptive immune response resulting from homologous and heterologous vaccination regimen at 18 months after primary vaccination. To that extent, we monitored 176 healthcare workers, the majority of whom had recovered from previous SARS-CoV-2 infection. In summary, we find that differences depending on primary immunization continue to exist 18 months after the first vaccination and these findings hold true irrespective of previous infection with the virus. Homologous primary immunization with BNT162b2 was repeatedly shown to produce higher antibody levels and slower antibody decline, leading to more effective in vitro neutralization capacities. Likewise, cellular responses resulting from in vitro re-stimulation were more pronounced after primary immunization involving BNT162b2. In contrast, IL-2 producing memory T helper and cytotoxic T cells appeared independent from the primary vaccination regimen. Despite these differences, comparable infection rates among all vaccination groups suggest comparable real-life protection.IMPORTANCEVaccination against the severe acute respiratory syndrome corona-virus 2 (SARS-CoV-2) was shown to avert severe courses of corona virus disease 2019 (COVID-19) and to mitigate spreading of the virus. However, the duration of protection and need for repeated boosting have yet to be defined. Monitoring and comparing the immune responses resulting from various vaccine strategies are therefore important to fill knowledge gaps and prepare for future pandemics.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Humanos , Anticorpos Neutralizantes , Anticorpos Antivirais , Vacina BNT162 , COVID-19/imunologia , COVID-19/prevenção & controle , Vacinas contra COVID-19/administração & dosagem , Vacinas contra COVID-19/imunologia , Imunidade Celular , Imunidade Humoral , RNA , SARS-CoV-2/fisiologia , Glicoproteína da Espícula de Coronavírus
15.
Brief Bioinform ; 24(6)2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37864296

RESUMO

Advances in single-cell sequencing and data analysis have made it possible to infer biological trajectories spanning heterogeneous cell populations based on transcriptome variation. These trajectories yield a wealth of novel insights into dynamic processes such as development and differentiation. However, trajectory analysis relies on an assumption of trajectory continuity, and experimental limitations preclude some real-world scenarios from meeting this condition. The current lack of assessment metrics makes it difficult to ascertain if/when a given trajectory deviates from continuity, and what impact such a divergence would have on inference accuracy is unclear. By analyzing simulated breaks introduced into in silico and real single-cell data, we found that discontinuity caused precipitous drops in the accuracy of trajectory inference. We then generate a simple scoring algorithm for assessing trajectory continuity, and found that continuity assessments in real-world cases of intestinal stem cell development and CD8 + T cells differentiation efficiently identifies trajectories consistent with empirical knowledge. This assessment approach can also be used in cases where a priori knowledge is lacking to screen a pool of inferred lineages for their adherence to presumed continuity, and serve as a means for weighing higher likelihood trajectories for validation via empirical studies, as exemplified by our case studies in psoriatic arthritis and acute kidney injury. This tool is freely available through github at qingshanni/scEGRET.


Assuntos
Algoritmos , Transcriptoma , Diferenciação Celular , Análise de Célula Única
16.
Immunity ; 45(2): 415-27, 2016 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-27533016

RESUMO

Chronic infections promote the terminal differentiation (or "exhaustion") of T cells and are thought to preclude the formation of memory T cells. In contrast, we discovered a small subpopulation of virus-specific CD8(+) T cells that sustained the T cell response during chronic infections. These cells were defined by, and depended on, the expression of the transcription factor Tcf1. Transcriptome analysis revealed that this population shared key characteristics of central memory cells but lacked an effector signature. Unlike conventional memory cells, Tcf1-expressing T cells displayed hallmarks of an "exhausted" phenotype, including the expression of inhibitory receptors such as PD-1 and Lag-3. This population was crucial for the T cell expansion that occurred in response to inhibitory receptor blockade during chronic infection. These findings identify a memory-like T cell population that sustains T cell responses and is a prime target for therapeutic interventions to improve the immune response in chronic infections.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Hepacivirus/imunologia , Hepatite C Crônica/imunologia , Imunoterapia/métodos , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Fator 1 de Transcrição de Linfócitos T/metabolismo , Adulto , Animais , Antígenos CD/metabolismo , Linfócitos T CD8-Positivos/virologia , Proliferação de Células , Células Cultivadas , Senescência Celular , Doença Crônica , Feminino , Humanos , Memória Imunológica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pessoa de Meia-Idade , Receptor de Morte Celular Programada 1/metabolismo , Fator 1 de Transcrição de Linfócitos T/genética , Transcriptoma , Proteína do Gene 3 de Ativação de Linfócitos
17.
Immunity ; 45(5): 988-998, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27851926

RESUMO

T cells rapidly undergo contraction upon viral clearance, but how T cell function and fate are determined during this phase is unclear. During the contraction phase of an acute infection with lymphocytic choriomeningitis virus, we found that virus-specific CD8+ T cells within the splenic red pulp (RP) had higher two-dimensional (2D) effective affinity than those within the white pulp (WP). This increased antigen recognition of RP-derived CD8+ T cells correlated with more efficient target cell killing and improved control of viremia. FoxP3+ regulatory T cells and cytokine TGF-ß limited the 2D-affinity in the WP during the contraction phase. Anatomical location drove gene expression patterns in CD8+ T cells that led to preferential differentiation of memory precursor WP T cells into long-term memory cells. These results highlight that intricate regulation of T cell function and fate is determined by anatomic compartmentalization during the early immune contraction phase.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Memória Imunológica/imunologia , Coriomeningite Linfocítica/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Baço/imunologia , Animais , Separação Celular , Citocinas/imunologia , Modelos Animais de Doenças , Citometria de Fluxo , Perfilação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transcriptoma
18.
Proc Natl Acad Sci U S A ; 119(43): e2209021119, 2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36260745

RESUMO

Interleukin-15 (IL-15) is often considered a central regulator of memory CD8+ T cells, based primarily on studies of recirculating subsets. However, recent work identified IL-15-independent CD8+ T cell memory populations, including tissue-resident memory CD8+ T cells (TRM) in some nonlymphoid tissues (NLTs). Whether this reflects the existence of IL-15-insensitive memory CD8+ T cells is unclear. We report that IL-15 complexes (IL-15c) stimulate rapid proliferation and expansion of both tissue-resident and circulating memory CD8+ T cell subsets across lymphoid and nonlymphoid tissues with varying magnitude by tissue and memory subset, in some sites correlating with differing levels of the IL-2Rß. This was conserved for memory CD8+ T cells recognizing distinct antigens and elicited by different pathogens. Following IL-15c-induced expansion, divided cells contracted to baseline numbers and only slowly returned to basal proliferation, suggesting a mechanism to transiently amplify memory populations. Through parabiosis, we showed that IL-15c drive local proliferation of TRM, with a degree of recruitment of circulating cells to some NLTs. Hence, irrespective of homeostatic IL-15 dependence, IL-15 sensitivity is a defining feature of memory CD8+ T cell populations, with therapeutic potential for expansion of TRM and other memory subsets in an antigen-agnostic and temporally controlled fashion.


Assuntos
Linfócitos T CD8-Positivos , Interleucina-15 , Memória Imunológica , Subpopulações de Linfócitos T
19.
Immunol Rev ; 300(1): 100-124, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33682165

RESUMO

Adaptive immunity to intracellular pathogens and tumors is mediated by antigen-experienced CD8 T cells. Individual naive CD8 T cells have the potential to differentiate into a diverse array of antigen-experienced subsets that exhibit distinct effector functions, life spans, anatomic positioning, and potential for regenerating an entirely new immune response during iterative pathogenic exposures. The developmental process by which activated naive cells undergo diversification involves regulation of chromatin structure and transcription but is not entirely understood. This review examines how alterations in chromatin structure, transcription factor binding, extracellular signals, and single-cell gene expression explain the differential development of distinct effector (TEFF ) and memory (TMEM ) CD8 T cell subsets. Special emphasis is placed on how Runx proteins function with additional transcription factors to pioneer changes in chromatin accessibility and drive transcriptional programs that establish the core attributes of cytotoxic T lymphocytes, subdivide circulating and non-circulating TMEM cell subsets, and govern terminal differentiation. The discussion integrates the roles of specific cytokine signals, transcriptional circuits and how regulation of individual nucleosomes and RNA polymerase II activity can contribute to the process of differentiation. A model that integrates many of these features is discussed to conceptualize how activated CD8 T cells arrive at their fates.


Assuntos
Linfócitos T CD8-Positivos , Subunidades alfa de Fatores de Ligação ao Core , Diferenciação Celular , Cromatina , Subunidades alfa de Fatores de Ligação ao Core/genética , Memória Imunológica , Subpopulações de Linfócitos T
20.
Eur J Immunol ; 53(6): e2250246, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37015057

RESUMO

The proprotein convertase subtilisin/kexins (PCSKs) regulate biological actions by cleaving immature substrate proteins. The archetype PCSK, FURIN, promotes the pathogenicity of viruses by proteolytically processing viral proteins. FURIN has also important regulatory functions in both innate and adaptive immune responses but its role in the CD8+ CTLs remains enigmatic. We used a T-cell-specific FURIN deletion in vivo to demonstrate that FURIN promotes host response against the CTL-dependent lymphocytic choriomeningitis virus by virtue of restricting viral burden and augmenting interferon gamma (IFNG) production. We also characterized Furin KO CD8+ T cells ex vivo, including after their activation with FURIN regulating cytokines IL12 or TGFB1. Furin KO CD8+ T cells show an inherently activated phenotype characterized by the upregulation of effector genes and increased frequencies of CD44+ , TNF+ , and IFNG+ cells. In the activated CTLs, FURIN regulates the productions of IL2, TNF, and GZMB and the genes associated with the TGFBR-signaling pathway. FURIN also controls the expression of Eomes, Foxo1, and Bcl6 and the levels of ITGAE and CD62L, which implies a role in the development of CTL memory. Collectively, our data suggest that the T-cell expressed FURIN is important for host responses in viral infections, CTL homeostasis/activation, and memory development.


Assuntos
Coriomeningite Linfocítica , Linfócitos T Citotóxicos , Camundongos , Animais , Linfócitos T CD8-Positivos , Furina/genética , Camundongos Endogâmicos C57BL , Vírus da Coriomeningite Linfocítica , Memória Imunológica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA