Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 584
Filtrar
2.
Nature ; 613(7944): 565-574, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36410718

RESUMO

Programming T cells to distinguish self from non-self is a vital, multi-step process that occurs in the thymus1-4. Signalling through the pre-T cell receptor (preTCR), a CD3-associated heterodimer comprising an invariant pTα chain and a clone-specific ß chain, is a critical early checkpoint in thymocyte development within the αß T cell lineage5,6. PreTCRs arrayed on CD4-CD8- double-negative thymocytes ligate peptides bound to major histocompatibility complex molecules (pMHC) on thymic stroma, similar to αß T cell receptors that appear on CD4+CD8+ double-positive thymocytes, but via a different molecular docking strategy7-10. Here we show the consequences of these distinct interactions for thymocyte progression using synchronized fetal thymic progenitor cultures that differ in the presence or absence of pMHC on support stroma, and single-cell transcriptomes at key thymocyte developmental transitions. Although major histocompatibility complex (MHC)-negative stroma fosters αß T cell differentiation, the absence of preTCR-pMHC interactions leads to deviant thymocyte transcriptional programming associated with dedifferentiation. Highly proliferative double-negative and double-positive thymocyte subsets emerge, with antecedent characteristics of T cell lymphoblastic and myeloid malignancies. Compensatory upregulation of diverse MHC class Ib proteins in B2m/H2-Ab1 MHC-knockout mice partially safeguards in vivo thymocyte progression, although disseminated double-positive thymic tumours may develop with ageing. Thus, as well as promoting ß chain repertoire broadening for subsequent αß T cell receptor utilization, preTCR-pMHC interactions limit cellular plasticity to facilitate normal thymocyte differentiation and proliferation that, if absent, introduce developmental vulnerabilities.


Assuntos
Desdiferenciação Celular , Antígenos de Histocompatibilidade Classe I , Receptores de Antígenos de Linfócitos T , Timócitos , Animais , Camundongos , Camundongos Knockout , Simulação de Acoplamento Molecular , Peptídeos/imunologia , Peptídeos/metabolismo , Timócitos/citologia , Timócitos/imunologia , Timo/citologia , Timo/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo
3.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-35055071

RESUMO

Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by dysregulated T and B lymphocytes. Type I interferons (IFN-I) have been shown to play important pathogenic roles in both SLE patients and mouse models of lupus. Recent studies have shown that B cell intrinsic responses to IFN-I are enough to drive B cell differentiation into autoantibody-secreting memory B cells and plasma cells, although lower levels of residual auto-reactive cells remain present. We speculated that IFN-I stimulation of T cells would similarly drive specific T-cell associated lupus phenotypes including the upregulation of T follicular helper cells and Th17, thereby affecting autoantibody production and the development of glomerulonephritis. Using the B6.Nba2 mouse model of lupus, we evaluated disease parameters in T cell specific IFN-I receptor (IFNAR)-deficient mice (cKO). Surprisingly, all measured CD4+ T cell abnormalities and associated intra-splenic cytokine levels (IFNγ, IL-6, IL-10, IL-17, IL-21) were unchanged and thus independent of IFN-I. In contrast B6.Nba2 cKO mice displayed reduced levels of effector CD8+ T cells and increased levels of Foxp3+ CD8+ regulatory T cells, suggesting that IFN-I induced signaling specifically affecting CD8+ T cells. These data suggest a role for both pathogenic and immunosuppressive CD8+ T cells in Nba2-driven autoimmunity, providing a model to further evaluate the role of these cell subsets during lupus-like disease development in vivo.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/imunologia , Ativação Linfocitária/imunologia , Receptor de Interferon alfa e beta/metabolismo , Animais , Anticorpos Antinucleares/imunologia , Autoimunidade , Biomarcadores , Linfócitos T CD4-Positivos/citologia , Diferenciação Celular/genética , Citocinas , Modelos Animais de Doenças , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Imuno-Histoquímica , Imunofenotipagem , Lúpus Eritematoso Sistêmico/etiologia , Lúpus Eritematoso Sistêmico/metabolismo , Lúpus Eritematoso Sistêmico/patologia , Ativação Linfocitária/genética , Camundongos , Camundongos Knockout , Especificidade de Órgãos/imunologia , Fenótipo , Receptor de Interferon alfa e beta/genética , Esplenomegalia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Timócitos/citologia , Timócitos/imunologia , Timócitos/metabolismo
4.
J Immunol ; 208(4): 910-928, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35082160

RESUMO

Enhancers activate transcription through long-distance interactions with their cognate promoters within a particular subtopologically associated domain (sub-TAD). The TCRα enhancer (Eα) is located at the sub-TAD boundary between the TCRα and DAD1 genes and regulates transcription toward both sides in an ∼1-Mb region. Analysis of Eα activity in transcribing the unrearranged TCRα gene at the 5'-sub-TAD has defined Eα as inactive in CD4-CD8- thymocytes, active in CD4+CD8+ thymocytes, and strongly downregulated in CD4+ and CD8+ thymocytes and αß T lymphocytes. Despite its strongly reduced activity, Eα is still required for high TCRα transcription and expression of TCRαß in mouse and human T lymphocytes, requiring collaboration with distant sequences for such functions. Because VαJα rearrangements in T lymphocytes do not induce novel long-range interactions between Eα and other genomic regions that remain in cis after recombination, strong Eα connectivity with the 3'-sub-TAD might prevent reduced transcription of the rearranged TCRα gene. Our analyses of transcriptional enhancer dependence during T cell development and non-T lineage tissues at the 3'-sub-TAD revealed that Eα can activate the transcription of specific genes, even when it is inactive to transcribe the TCRα gene at the 5'-sub-TAD. Hence distinct requirements for Eα function are necessary at specific genes at both sub-TADs, implying that enhancers do not merely function as chromatin loop anchors that nucleate the formation of factor condensates to increase gene transcription initiated at their cognate promoters. The observed different regulated Eα activity for activating specific genes at its flanking sub-TADs may be a general feature for enhancers located at sub-TAD boundaries.


Assuntos
Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Animais , Diferenciação Celular/genética , Mapeamento Cromossômico , Rearranjo Gênico da Cadeia alfa dos Receptores de Antígenos dos Linfócitos T , Loci Gênicos , Humanos , Células Jurkat , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Timócitos/imunologia , Timócitos/metabolismo
5.
Virology ; 567: 77-86, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35032866

RESUMO

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


Assuntos
Atrofia/virologia , Interferon-alfa/imunologia , Interferon beta/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Timócitos/virologia , Timo/virologia , Animais , Atrofia/genética , Atrofia/imunologia , Atrofia/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Doença Crônica , Feminino , Regulação da Expressão Gênica , Humanos , Memória Imunológica , Interferon-alfa/genética , Interferon beta/genética , Linfonodos/imunologia , Linfonodos/patologia , Linfonodos/virologia , Depleção Linfocítica , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/patologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais/imunologia , Análise de Célula Única , Timócitos/imunologia , Timócitos/patologia , Timo/imunologia , Timo/patologia
6.
J Exp Med ; 219(2)2022 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-34910107

RESUMO

The Tcra repertoire is generated by multiple rounds of Vα-Jα rearrangement. However, Tcrd recombination precedes Tcra recombination within the complex Tcra-Tcrd locus. Here, by ablating Tcrd recombination, we report that Tcrd rearrangement broadens primary Vα use to diversify the Tcra repertoire in mice. We reveal that use of Trav15-dv6 family V gene segments in Tcrd recombination imparts diversity in the Tcra repertoire by instigating use of central and distal Vα segments. Moreover, disruption of the regions containing these genes and their cis-regulatory elements identifies the Trav15-dv6 family as being responsible for driving central and distal Vα recombinations beyond their roles as substrates for Tcrd recombination. Our study demonstrates an indispensable role for Tcrd recombination in general, and the Trav15-dv6 family in particular, in the generation of a combinatorially diverse Tcra repertoire.


Assuntos
Rearranjo Gênico da Cadeia alfa dos Receptores de Antígenos dos Linfócitos T , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T gama-delta/genética , Animais , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Camundongos Knockout , Linfócitos T/imunologia , Linfócitos T/metabolismo , Timócitos/imunologia , Timócitos/metabolismo , Recombinação V(D)J
7.
Transl Res ; 239: 103-123, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34461306

RESUMO

CD160 is a member of the immunoglobulin superfamily with a pattern of expression mainly restricted to cytotoxic cells. To assess the functional relevance of the HVEM/CD160 signaling pathway in allogeneic cytotoxic responses, exon 2 of the CD160 gene was targeted by CRISPR/Cas9 to generate CD160 deficient mice. Next, we evaluated the impact of CD160 deficiency in the course of an alloreactive response. To that aim, parental donor WT (wild-type) or CD160 KO (knock-out) T cells were adoptively transferred into non-irradiated semiallogeneic F1 recipients, in which donor alloreactive CD160 KO CD4 T cells and CD8 T cells clonally expanded less vigorously than in WT T cell counterparts. This differential proliferative response rate at the early phase of T cell expansion influenced the course of CD8 T cell differentiation and the composition of the effector T cell pool that led to a significant decreased of the memory precursor effector cells (MPECs) / short-lived effector cells (SLECs) ratio in CD160 KO CD8 T cells compared to WT CD8 T cells. Despite these differences in T cell proliferation and differentiation, allogeneic MHC class I mismatched (bm1) skin allograft survival in CD160 KO recipients was comparable to that of WT recipients. However, the administration of CTLA-4.Ig showed an enhanced survival trend of bm1 skin allografts in CD160 KO with respect to WT recipients. Finally, CD160 deficient NK cells were as proficient as CD160 WT NK cells in rejecting allogeneic cellular allografts or MHC class I deficient tumor cells. CD160 may represent a CD28 alternative costimulatory molecule for the modulation of allogeneic CD8 T cell responses either in combination with costimulation blockade or by direct targeting of alloreactive CD8 T cells that upregulate CD160 expression in response to alloantigen stimulation.


Assuntos
Antígenos CD/imunologia , Linfócitos T CD8-Positivos/imunologia , Rejeição de Enxerto/etiologia , Receptores Imunológicos/imunologia , Ligante 4-1BB/metabolismo , Aloenxertos , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Sistemas CRISPR-Cas , Diferenciação Celular , Feminino , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Proteínas Ligadas por GPI/metabolismo , Regulação da Expressão Gênica , Genes MHC Classe I , Rejeição de Enxerto/imunologia , Células Matadoras Naturais/imunologia , Lectinas Tipo C/metabolismo , Camundongos Endogâmicos , Camundongos Knockout , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Membro 14 de Receptores do Fator de Necrose Tumoral/metabolismo , Transplante de Pele , Timócitos/imunologia
8.
Scand J Immunol ; 94(4): e13094, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34780092

RESUMO

The thymus produces self-limiting and self-tolerant T cells through the interaction between thymocytes and thymus epithelial cells (TECs), thereby generating central immune tolerance. The TECs are composed of cortical and medullary thymic epithelial cells, which regulate the positive and negative selection of T cells, respectively. During the process of negative selection, thymocytes with self-reactive ability are deleted or differentiated into regulatory T cells (Tregs). Tregs are a subset of suppressor T cells that are important for maintaining immune homeostasis. The differentiation and development of Tregs depend on the development of TECs and other underlying molecular mechanisms. Tregs regulated by thymic epithelial cells are closely related to human health and are significant in autoimmune diseases, thymoma and pregnancy. In this review, we summarize the current molecular and transcriptional regulatory mechanisms by which TECs affect the development and function of thymic Tregs. We also review the pathophysiological models of thymic epithelial cells regulating thymic Tregs in human diseases and specific physiological conditions.


Assuntos
Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Imunidade Adaptativa , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Diferenciação Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Epiteliais/classificação , Células Epiteliais/citologia , Células Epiteliais/imunologia , Feminino , Homeostase , Humanos , Masculino , Modelos Imunológicos , Poliendocrinopatias Autoimunes/genética , Poliendocrinopatias Autoimunes/imunologia , Gravidez , Transdução de Sinais/imunologia , Linfócitos T Reguladores/classificação , Timócitos/classificação , Timócitos/citologia , Timócitos/imunologia , Timoma/imunologia , Timo/citologia , Timo/imunologia , Neoplasias do Timo/imunologia
9.
Nat Commun ; 12(1): 6230, 2021 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-34711828

RESUMO

T cells undergo rigorous selection in the thymus to ensure self-tolerance and prevent autoimmunity, with this process requiring innocuous self-antigens (Ags) to be presented to thymocytes. Self-Ags are either expressed by thymic stroma cells or transported to the thymus from the periphery by migratory dendritic cells (DCs); meanwhile, small blood-borne peptides can access the thymic parenchyma by diffusing across the vascular lining. Here we describe an additional pathway of thymic Ag acquisition that enables circulating antigenic macromolecules to access both murine and human thymi. This pathway depends on a subset of thymus-resident DCs, distinct from both parenchymal and circulating migratory DCs, that are positioned in immediate proximity to thymic microvessels where they extend cellular processes across the endothelial barrier into the blood stream. Transendothelial positioning of DCs depends on DC-expressed CX3CR1 and its endothelial ligand, CX3CL1, and disrupting this chemokine pathway prevents thymic acquisition of circulating proteins and compromises negative selection of Ag-reactive thymocytes. Thus, transendothelial DCs represent a mechanism by which the thymus can actively acquire blood-borne Ags to induce and maintain central tolerance.


Assuntos
Sangue/imunologia , Células Dendríticas/imunologia , Células Endoteliais/imunologia , Timócitos/imunologia , Timo/imunologia , Animais , Autoantígenos/imunologia , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/imunologia , Diferenciação Celular , Movimento Celular , Quimiocina CX3CL1/genética , Quimiocina CX3CL1/imunologia , Células Dendríticas/citologia , Células Endoteliais/citologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Tolerância a Antígenos Próprios , Timócitos/citologia , Timo/citologia
10.
Iran J Allergy Asthma Immunol ; 20(5): 623-634, 2021 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-34664821

RESUMO

The rates of invariant natural killer T (iNKT) cells in vivo are very low, and the amounts of cells obtained directly from the body are hard enough to fulfill their potential in clinical application. To overcome this problem, we subcutaneously injected alpha-galactosylceramide (α-GalCer) into DBA/1 mice and thymic single cells were isolated and cultured in vitro. Fluorescence-activated cell sorting was used to detect the iNKT cells and their subsets in the thymus after the injection of α-GalCer by different methods. In addition, in vitro changes of single-cell suspensions and their cytokines in culture supernatants were assessed. Compared with the α-GalCer multiple subcutaneous injection group, the rates of iNKT cells in the α-GalCer single subcutaneous injection group were markedly higher at each time point, while the highest levels of iNKT1 and iNKT2 cells were observed on day 4 and  8, respectively. In α-GalCer single subcutaneous injection for 8 days and thymic mononuclear cell cultured for 14 days group, the expansion rate of iNKT cells was significantly faster than the other groups, while it reached a peak for iNKT1 cells. Interferon-gamma was consistent with the development of iNKT1 cells, however no difference was found between the cultured iNKT cells in vitro and the natural iNKT cells in vivo in terms of cytokine production. Herein, we introduced a method in which antigenic stimulation in vivo and directed induction in vitro yielded high levels of iNKT cells with specific functions.


Assuntos
Diferenciação Celular/imunologia , Ativação Linfocitária/imunologia , Células T Matadoras Naturais/imunologia , Células T Matadoras Naturais/metabolismo , Timócitos/imunologia , Timócitos/metabolismo , Animais , Antígenos/metabolismo , Biomarcadores , Células Cultivadas , Citocinas/metabolismo , Camundongos , Células T Matadoras Naturais/citologia , Timócitos/citologia
11.
Front Immunol ; 12: 716661, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34394122

RESUMO

Although metabolic pathways have been shown to control differentiation and activation in peripheral T cells, metabolic studies on thymic T cell development are still lacking, especially in human tissue. In this study, we use transcriptomics and extracellular flux analyses to investigate the metabolic profiles of primary thymic and in vitro-derived mouse and human thymocytes. Core metabolic pathways, specifically glycolysis and oxidative phosphorylation, undergo dramatic changes between the double-negative (DN), double-positive (DP), and mature single-positive (SP) stages in murine and human thymus. Remarkably, despite the absence of the complex multicellular thymic microenvironment, in vitro murine and human T cell development recapitulated the coordinated decrease in glycolytic and oxidative phosphorylation activity between the DN and DP stages seen in primary thymus. Moreover, by inducing in vitro T cell differentiation from Rag1-/- mouse bone marrow, we show that reduced metabolic activity at the DP stage is independent of TCR rearrangement. Thus, our findings suggest that highly conserved metabolic transitions are critical for thymic T cell development.


Assuntos
Diferenciação Celular , Metabolismo Energético , Linfócitos T/citologia , Linfócitos T/metabolismo , Timócitos/citologia , Timócitos/metabolismo , Animais , Evolução Biológica , Biomarcadores , Linhagem Celular , Biologia Computacional/métodos , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Linfopoese , Metaboloma , Metabolômica/métodos , Camundongos , Organoides , Timócitos/imunologia , Técnicas de Cultura de Tecidos
12.
Front Immunol ; 12: 668528, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34220815

RESUMO

The microenvironments formed by cortical (c) and medullary (m) thymic epithelial cells (TECs) play a non-redundant role in the generation of functionally diverse and self-tolerant T cells. The role of TECs during the first weeks of the murine postnatal life is particularly challenging due to the significant augment in T cell production. Here, we critically review recent studies centered on the timely coordination between the expansion and maturation of TECs during this period and their specialized role in T cell development and selection. We further discuss how aging impacts on the pool of TEC progenitors and maintenance of functionally thymic epithelial microenvironments, and the implications of these chances in the capacity of the thymus to sustain regular thymopoiesis throughout life.


Assuntos
Diferenciação Celular , Proliferação de Células , Células Epiteliais/fisiologia , Células-Tronco/fisiologia , Timo/fisiologia , Fatores Etários , Animais , Microambiente Celular , Células Epiteliais/imunologia , Humanos , Tolerância a Antígenos Próprios , Células-Tronco/imunologia , Linfócitos T/imunologia , Timócitos/imunologia , Timo/imunologia
13.
Front Immunol ; 12: 669893, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34140950

RESUMO

Down syndrome (DS) patients prematurely show clinical manifestations usually associated with aging. Their immune system declines earlier than healthy individuals, leading to increased susceptibility to infections and higher incidence of autoimmune phenomena. Clinical features of accelerated aging indicate that trisomy 21 increases the biological age of tissues. Based on previous studies suggesting immune senescence in DS, we hypothesized that induction of cellular senescence may contribute to early thymic involution and immune dysregulation. Immunohistochemical analysis of thymic tissue showed signs of accelerated thymic aging in DS patients, normally seen in older healthy subjects. Moreover, our whole transcriptomic analysis on human Epcam-enriched thymic epithelial cells (hTEC), isolated from three DS children, which revealed disease-specific transcriptomic alterations. Gene set enrichment analysis (GSEA) of DS TEC revealed an enrichment in genes involved in cellular response to stress, epigenetic histone DNA modifications and senescence. Analysis of senescent markers and oxidative stress in hTEC and thymocytes confirmed these findings. We detected senescence features in DS TEC, thymocytes and peripheral T cells, such as increased ß-galactosidase activity, increased levels of the cell cycle inhibitor p16, telomere length and integrity markers and increased levels of reactive oxygen species (ROS), all factors contributing to cellular damage. In conclusion, our findings support the key role of cellular senescence in the pathogenesis of immune defect in DS while adding new players, such as epigenetic regulation and increased oxidative stress, to the pathogenesis of immune dysregulation.


Assuntos
Proliferação de Células , Senescência Celular , Síndrome de Down/metabolismo , Células Epiteliais/metabolismo , Imunossenescência , Estresse Oxidativo , Timócitos/metabolismo , Timo/metabolismo , Fatores Etários , Estudos de Casos e Controles , Proliferação de Células/genética , Senescência Celular/genética , Criança , Pré-Escolar , Síndrome de Down/genética , Síndrome de Down/imunologia , Síndrome de Down/patologia , Epigênese Genética , Células Epiteliais/imunologia , Células Epiteliais/patologia , Feminino , Perfilação da Expressão Gênica , Humanos , Imunossenescência/genética , Lactente , Masculino , Estresse Oxidativo/genética , Timócitos/imunologia , Timócitos/patologia , Timo/imunologia , Timo/patologia , Transcriptoma
14.
Commun Biol ; 4(1): 681, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-34083746

RESUMO

T cells rely for their development and function on the correct folding and turnover of proteins generated in response to a broad range of molecular cues. In the absence of the eukaryotic type II chaperonin complex, CCT, T cell activation induced changes in the proteome are compromised including the formation of nuclear actin filaments and the formation of a normal cell stress response. Consequently, thymocyte maturation and selection, and T cell homeostatic maintenance and receptor-mediated activation are severely impaired. In the absence of CCT-controlled protein folding, Th2 polarization diverges from normal differentiation with paradoxical continued IFN-γ expression. As a result, CCT-deficient T cells fail to generate an efficient immune protection against helminths as they are unable to sustain a coordinated recruitment of the innate and adaptive immune systems. These findings thus demonstrate that normal T cell biology is critically dependent on CCT-controlled proteostasis and that its absence is incompatible with protective immunity.


Assuntos
Chaperonina com TCP-1/imunologia , Proteostase/imunologia , Linfócitos T/imunologia , Timócitos/imunologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Cultivadas , Chaperonina com TCP-1/genética , Chaperonina com TCP-1/metabolismo , Humanos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteoma/imunologia , Proteoma/metabolismo , Proteostase/genética , Linfócitos T/citologia , Linfócitos T/metabolismo , Timócitos/citologia , Timócitos/metabolismo , Transcriptoma/genética , Transcriptoma/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
15.
Eur J Immunol ; 51(7): 1698-1714, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33949677

RESUMO

CD4+ CD8+ double-positive thymocytes give rise to both conventional TCRαß+ T cells and invariant natural killer T cells (iNKT cells), but these two kinds of cells display different characteristics. The molecular mechanism underlying iNKT cell lineage development and function acquisition remain to be elucidated. We show that the loss of chromatin assembly factor 1B (CHAF1b) maintains the normal development of conventional TCRαß+ T cells but severely impairs early development of iNKT cells. This dysregulation is accompanied by the impairment in chromatin activation and gene transcription at Vα14-Jα18 locus. Notably, ectopic expression of a Vα14-Jα18 TCR rescues Chaf1b-deficient iNKT cell developmental defects. Moreover, cytokine secretion and antitumor activity are substantially maintained in Vα14-Jα18 TCR transgene-rescued Chaf1b-deficient iNKT cells. Our study identifies CHAF1b as a critical factor that controls the early development but not function acquisition of iNKT cells via lineage- and stage-specific regulation.


Assuntos
Fator 1 de Modelagem da Cromatina/imunologia , Células T Matadoras Naturais/imunologia , Animais , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Linhagem da Célula/imunologia , Montagem e Desmontagem da Cromatina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Timócitos/imunologia
16.
Front Immunol ; 12: 681110, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34025680

RESUMO

The discovery of innate lymphoid cells (ILCs) has revolutionized our understanding of innate immunity and immune cell interactions at epithelial barrier sites. Their presence and maintenance are critical for modulating immune homeostasis, responding to injury or infection, and repairing damaged tissues. To date, ILCs have been defined by a set of transcription factors, surface antigens and cytokines, and their functions resemble those of three major classes of helper T cell subsets, Th1, Th2 and Th17. Despite this, the lack of antigen-specific surface receptors and the notion that ILCs can develop in the absence of the thymic niche have clearly set them apart from the T-cell lineage and promulgated a dogma that ILCs develop directly from progenitors in the bone marrow. Interestingly however, emerging studies have challenged the BM-centric view of adult ILC development and suggest that ILCs could arise neonatally from developing T cell progenitors. In this review, we discuss ILC development in parallel to T-cell development and summarize key findings that support a T-cell-centric view of ILC ontogeny.


Assuntos
Imunidade Inata , Linfócitos/imunologia , Linfócitos/metabolismo , Linfopoese , Timo/fisiologia , Fatores Etários , Animais , Biomarcadores , Medula Óssea/imunologia , Medula Óssea/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Transdução de Sinais , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Timócitos/citologia , Timócitos/imunologia , Timócitos/metabolismo
17.
Int J Mol Sci ; 22(8)2021 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-33916948

RESUMO

In Hashimoto's thyroiditis (HT), oxidative stress (OS) is driven by Th1 cytokines' response interfering with the normal function of thyrocytes. OS results from an imbalance between an excessive production of reactive oxygen species (ROS) and a lowering of antioxidant production. Moreover, OS has been shown to inhibit Sirtuin 1 (SIRT1), which is able to prevent hypoxia-inducible factor (HIF)-1α stabilization. The aims of this study were to determine the involvement of NADPH-oxidases (NOX), SIRT1, and HIF-1α in HT pathophysiology as well as the status of antioxidant proteins such as peroxiredoxin 1 (PRDX1), catalase, and superoxide dismutase 1 (SOD1). The protein expressions of NOX2, NOX4, antioxidant enzymes, SIRT1, and HIF-1α, as well as glucose transporter-1 (GLUT-1) and vascular endothelial growth factor A (VEGF-A), were analyzed by Western blot in primary cultures of human thyrocytes that were or were not incubated with Th1 cytokines. The same proteins were also analyzed by immunohistochemistry in thyroid samples from control and HT patients. In human thyrocytes incubated with Th1 cytokines, NOX4 expression was increased whereas antioxidants, such as PRDX1, catalase, and SOD1, were reduced. Th1 cytokines also induced a significant decrease of SIRT1 protein expression associated with an upregulation of HIF-1α, GLUT-1, and VEGF-A proteins. With the exception of PRDX1 and SOD1, similar results were obtained in HT thyroids. OS due to an increase of ROS produced by NOX4 and a loss of antioxidant defenses (PRDX1, catalase, SOD1) correlates to a reduction of SIRT1 and an upregulation of HIF 1α, GLUT-1, and VEGF-A. Our study placed SIRT1 as a key regulator of OS and we, therefore, believe it could be considered as a potential therapeutic target in HT.


Assuntos
Doença de Hashimoto/etiologia , Doença de Hashimoto/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Estresse Oxidativo , Sirtuína 1/genética , Células Th1/imunologia , Células Th1/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Adulto , Doenças Autoimunes/etiologia , Doenças Autoimunes/metabolismo , Autoimunidade/genética , Biomarcadores , Citocinas/genética , Citocinas/metabolismo , Feminino , Imunofluorescência , Regulação da Expressão Gênica , Doença de Hashimoto/diagnóstico , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Modelos Biológicos , NADPH Oxidase 2/genética , NADPH Oxidase 2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sirtuína 1/metabolismo , Superóxido Dismutase-1/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Timócitos/imunologia , Timócitos/metabolismo , Testes de Função Tireóidea , Glândula Tireoide/imunologia , Glândula Tireoide/metabolismo , Glândula Tireoide/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem
18.
Cell Rep ; 35(2): 108967, 2021 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-33852867

RESUMO

T lymphocyte differentiation in the steady state is characterized by high cellular turnover whereby thymocytes do not self-renew. However, if deprived of competent progenitors, the thymus can temporarily maintain thymopoiesis autonomously. This bears a heavy cost, because prolongation of thymus autonomy causes leukemia. Here, we show that, at an early stage, thymus autonomy relies on double-negative 3 early (DN3e) thymocytes that acquire stem-cell-like properties. Following competent progenitor deprivation, DN3e thymocytes become long lived, are required for thymus autonomy, differentiate in vivo, and include DNA-label-retaining cells. At the single-cell level, the transcriptional programs of thymopoiesis in autonomy and the steady state are similar. However, a new cell population emerges in autonomy that expresses an aberrant Notch target gene signature and bypasses the ß-selection checkpoint. In summary, DN3e thymocytes have the potential to self-renew and differentiate in vivo if cell competition is impaired, but this generates atypical cells, probably the precursors of leukemia.


Assuntos
Hematopoese/genética , Leucemia/genética , Receptores Notch/genética , Timócitos/imunologia , Timo/imunologia , Fatores de Transcrição/genética , Animais , Diferenciação Celular , Proliferação de Células , Família de Proteínas EGF/genética , Família de Proteínas EGF/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Hematopoese/imunologia , Humanos , Imunofenotipagem , Rim , Leucemia/imunologia , Leucemia/patologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptores Notch/imunologia , Transdução de Sinais , Análise de Célula Única , Timócitos/classificação , Timócitos/patologia , Timo/patologia , Timo/transplante , Fatores de Transcrição/imunologia , Transplante Heterotópico , Transplante Homólogo
19.
Elife ; 102021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33884954

RESUMO

Functional tuning of T cells based on their degree of self-reactivity is established during positive selection in the thymus, although how positive selection differs for thymocytes with relatively low versus high self-reactivity is unclear. In addition, preselection thymocytes are highly sensitive to low-affinity ligands, but the mechanism underlying their enhanced T cell receptor (TCR) sensitivity is not fully understood. Here we show that murine thymocytes with low self-reactivity experience briefer TCR signals and complete positive selection more slowly than those with high self-reactivity. Additionally, we provide evidence that cells with low self-reactivity retain a preselection gene expression signature as they mature, including genes previously implicated in modulating TCR sensitivity and a novel group of ion channel genes. Our results imply that thymocytes with low self-reactivity downregulate TCR sensitivity more slowly during positive selection, and associate membrane ion channel expression with thymocyte self-reactivity and progress through positive selection.


Assuntos
Diferenciação Celular , Antígenos de Histocompatibilidade Classe I/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Tolerância a Antígenos Próprios , Timócitos/imunologia , Timo/imunologia , Animais , Linhagem da Célula , Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo , Cinética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Timócitos/metabolismo , Timo/crescimento & desenvolvimento , Timo/metabolismo , Transcriptoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...