Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 20(11): 1456-1468, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31636466

RESUMEN

T cell specification and commitment require Notch signaling. Although the requirement for Notch signaling during intrathymic T cell development is known, it is still unclear whether the onset of T cell priming can occur in a prethymic niche and whether RBPJ-dependent Notch signaling has a role during this event. Here, we established an Rbpj-inducible system that allowed temporal and tissue-specific control of the responsiveness to Notch in all hematopoietic cells. Using this system, we found that Notch signaling was required before the early T cell progenitor stage in the thymus. Lymphoid-primed multipotent progenitors in the bone marrow underwent Notch signaling with Rbpj induction, which inhibited development towards the myeloid lineage in thymus-seeding progenitors. Thus, our results indicated that the onset of T cell differentiation occurred in a prethymic setting, and that Notch played an important role during this event.


Asunto(s)
Diferenciación Celular/inmunología , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Células Precursoras de Linfocitos T/fisiología , Receptores Notch/metabolismo , Subgrupos de Linfocitos T/inmunología , Animales , Linaje de la Célula/inmunología , Separación Celular , Femenino , Citometría de Flujo , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/genética , Masculino , Ratones , Ratones Transgénicos , Cultivo Primario de Células , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/metabolismo , Timo/citología , Timo/inmunología
2.
Nat Immunol ; 20(10): 1381-1392, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31451788

RESUMEN

Proliferation is tightly regulated during T cell development, and is limited to immature CD4-CD8- thymocytes. The major proliferative event is initiated at the 'ß-selection' stage following successful rearrangement of Tcrß, and is triggered by and dependent on concurrent signaling by Notch and the pre-T cell receptor (TCR); however, it is unclear how these signals cooperate to promote cell proliferation. Here, we found that ß-selection-associated proliferation required the combined activity of two Skp-cullin-F-box (SCF) ubiquitin ligase complexes that included as substrate recognition subunits the F-box proteins Fbxl1 or Fbxl12. Both SCF complexes targeted the cyclin-dependent kinase inhibitor Cdkn1b for polyubiquitination and proteasomal degradation. We found that Notch signals induced the transcription of Fbxl1, whereas pre-TCR signals induced the transcription of Fbxl12. Thus, concurrent Notch and pre-TCR signaling induced the expression of two genes, Fbxl1 and Fbxl12, whose products functioned identically but additively to promote degradation of Cdkn1b, cell cycle progression, and proliferation of ß-selected thymocytes.


Asunto(s)
Proteínas F-Box/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores Notch/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Linfocitos T/fisiología , Timocitos/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Selección Clonal Mediada por Antígenos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas F-Box/genética , Regulación de la Expresión Génica , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T , Ratones , Ratones Endogámicos C57BL , Receptor Cross-Talk , Transducción de Señal
3.
Semin Immunol ; 70: 101837, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37659170

RESUMEN

Thymus is a primary lymphoid organ essential for the development of T lymphocytes. Age-related thymic involution is a prominent feature of immune senescence. The thymus undergoes rapid growth during fetal and neonatal development, peaks in size before puberty and then begins to undergo a decrease in cellularity with age. Dramatic changes occur with age-associated thymic involution. The most prominent features of thymic involution include: (i) epithelial structure disruption, (ii) adipogenesis, and (iii) thymocyte development arrest. There is a sex disparity in thymus aging. It is a multifactorial process controlled and regulated by a series of molecules, including the transcription factor FOXN1, fibroblast and keratinocyte growth factors (FGF and KGF, respectively), sex steroids, Notch signaling, WNT signaling, and microRNAs. Nevertheless, there is still no satisfactory evolutionary or physiological explanation for age-associated thymic involution, and understanding the precise mechanism(s) for thymus aging remains challenging. Sustained thymic regeneration has yet to be achieved by sex steroid ablation. Recent preclinical studies indicate that long-term thymic reconstitution can be achieved via adoptive transfer of in vitro-generated progenitor T (proT) cells, and improvements in the methods for the generation of human proT cells make this an attractive approach. Future clinical applications may rely on new applications integrating proT cells, cytokine support and sex-steroid inhibition treatments.


Asunto(s)
Reconstitución Inmune , Recién Nacido , Humanos , Envejecimiento , Timo/fisiología , Linfocitos T , Hormonas Esteroides Gonadales , Esteroides
4.
Nat Immunol ; 14(10): 1037-44, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23974957

RESUMEN

The transcription factor GATA-3 is expressed and required for differentiation and function throughout the T lymphocyte lineage. Despite evidence it may also be expressed in multipotent hematopoietic stem cells (HSCs), any role for GATA-3 in these cells has remained unclear. Here we found GATA-3 was in the cytoplasm in quiescent long-term stem cells from steady-state bone marrow but relocated to the nucleus when HSCs cycled. Relocation depended on signaling via the mitogen-activated protein kinase p38 and was associated with a diminished capacity for long-term reconstitution after transfer into irradiated mice. Deletion of Gata3 enhanced the repopulating capacity and augmented the self-renewal of long-term HSCs in cell-autonomous fashion without affecting the cell cycle. Our observations position GATA-3 as a regulator of the balance between self-renewal and differentiation in HSCs that acts downstream of the p38 signaling pathway.


Asunto(s)
Factor de Transcripción GATA3/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Factor de Transcripción GATA3/genética , Eliminación de Gen , Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/efectos de los fármacos , Ligandos , Ratones , Ratones Noqueados , Poli I-C/farmacología , Transporte de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Receptores Toll-Like/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
J Immunol ; 211(1): 81-90, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37154711

RESUMEN

Recent thymic emigrant (RTE) cells are nascent T cells that continue their post-thymic maturation in the periphery and dominate T cell immune responses in early life and in adults having undergone lymphodepletion regimens. However, the events that govern their maturation and their functionality as they transition to mature naive T cells have not been clearly defined. Using RBPJind mice, we were able to identify different stages of RTE maturation and interrogate their immune function using a T cell transfer model of colitis. As CD45RBlo RTE cells mature, they transition through a CD45RBint immature naive T (INT) cell population that is more immunocompetent but shows a bias toward IL-17 production at the expense of IFN-γ. Additionally, the levels of IFN-γ and IL-17 produced in INT cells are highly dependent on whether Notch signals are received during INT cell maturation or during their effector function. IL-17 production by INT cells showed a total requirement for Notch signaling. Loss of Notch signaling at any stage of INT cells resulted in an impaired colitogenic effect of INT cells. RNA sequencing of INT cells that had matured in the absence of Notch signals showed a reduced inflammatory profile compared with Notch-responsive INT cells. Overall, we have elucidated a previously unknown INT cell stage, revealed its intrinsic bias toward IL-17 production, and demonstrated a role for Notch signaling in INT cell peripheral maturation and effector function in the context of a T cell transfer model of colitis.


Asunto(s)
Colitis , Linfocitos T , Ratones , Animales , Timo , Interleucina-17 , Transducción de Señal
6.
J Immunol ; 209(1): 77-92, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35705252

RESUMEN

The zinc-finger transcription factor GATA-3 plays a crucial role during early T cell development and also dictates later T cell differentiation outcomes. However, its role and collaboration with the Notch signaling pathway in the induction of T lineage specification and commitment have not been fully elucidated. We show that GATA-3 deficiency in mouse hematopoietic progenitors results in an early block in T cell development despite the presence of Notch signals, with a failure to upregulate Bcl11b expression, leading to a diversion along a myeloid, but not a B cell, lineage fate. GATA-3 deficiency in the presence of Notch signaling results in the apoptosis of early T lineage cells, as seen with inhibition of CDK4/6 (cyclin-dependent kinases 4 and 6) function, and dysregulated cyclin-dependent kinase inhibitor 2b (Cdkn2b) expression. We also show that GATA-3 induces Bcl11b, and together with Bcl11b represses Cdkn2b expression; however, loss of Cdkn2b failed to rescue the developmental block of GATA-3-deficient T cell progenitor. Our findings provide a signaling and transcriptional network by which the T lineage program in response to Notch signals is realized.


Asunto(s)
Factor de Transcripción GATA3/metabolismo , Transducción de Señal , Linfocitos T , Animales , Diferenciación Celular , Linaje de la Célula , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina , Redes Reguladoras de Genes , Ratones , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Linfocitos T/metabolismo , Proteínas Supresoras de Tumor/metabolismo
7.
Development ; 147(23)2020 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-33144398

RESUMEN

E protein transcription factors are crucial for many cell fate decisions. However, the roles of E proteins in the germ-layer specification of human embryonic stem cells (hESCs) are poorly understood. We disrupted the TCF3 gene locus to delete the E protein E2A in hESCs. E2A knockout (KO) hESCs retained key features of pluripotency, but displayed decreased neural ectoderm coupled with enhanced mesoendoderm outcomes. Genome-wide analyses showed that E2A directly regulates neural ectoderm and Nodal pathway genes. Accordingly, inhibition of Nodal or E2A overexpression partially rescued the neural ectoderm defect in E2A KO hESCs. Loss of E2A had little impact on the epigenetic landscape of hESCs, whereas E2A KO neural precursors displayed increased accessibility of the gene locus encoding the Nodal agonist CRIPTO. Double-deletion of both E2A and HEB (TCF12) resulted in a more severe neural ectoderm defect. Therefore, this study reveals critical context-dependent functions for E2A in human neural ectoderm fate specification.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas Ligadas a GPI/genética , Células Madre Embrionarias Humanas/citología , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de Neoplasias/genética , Proteína Nodal/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Diferenciación Celular/genética , Linaje de la Célula/genética , Ectodermo/crecimiento & desarrollo , Ectodermo/metabolismo , Epigénesis Genética/genética , Regulación del Desarrollo de la Expresión Génica/genética , Genoma Humano/genética , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células-Madre Neurales/citología , Proteína Nodal/antagonistas & inhibidores , Transducción de Señal/genética
8.
Development ; 147(12)2020 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-32467240

RESUMEN

The cortical and medullary thymic epithelial cell (cTEC and mTEC) lineages are essential for inducing T cell lineage commitment, T cell positive selection and the establishment of self-tolerance, but the mechanisms controlling their fetal specification and differentiation are poorly understood. Here, we show that notch signaling is required to specify and expand the mTEC lineage. Notch1 is expressed by and active in TEC progenitors. Deletion of Notch1 in TECs resulted in depletion of mTEC progenitors and dramatic reductions in mTECs during fetal stages, consistent with defects in mTEC specification and progenitor expansion. Conversely, forced notch signaling in all TECs resulted in widespread expression of mTEC progenitor markers and profound defects in TEC differentiation. In addition, lineage-tracing analysis indicated that all mTECs have a history of receiving a notch signal, consistent with notch signaling occurring in mTEC progenitors. These data provide strong evidence for a requirement for notch signaling in specification of the mTEC lineage.


Asunto(s)
Desarrollo Fetal/genética , Receptor Notch1/metabolismo , Timo/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Células Epiteliales/citología , Células Epiteliales/metabolismo , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organogénesis , Receptor Notch1/deficiencia , Receptor Notch1/genética , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Timo/citología , Timo/crecimiento & desarrollo
9.
Clin Exp Immunol ; 213(1): 13-22, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37085947

RESUMEN

The Forkhead Box P3 (FOXP3) protein is an essential transcription factor for the development and function of regulatory T cells (Tregs), involved in the maintenance of immunological tolerance. Although extensive research over the last decade has investigated the critical role of FOXP3+ cells in preserving immune homeostasis, our understanding of their specific functions remains limited. Therefore, unveiling the molecular mechanisms underpinning the up- and downstream transcriptional regulation of and by FOXP3 is crucial for developing Treg-targeted therapeutics. Dysfunctions in FOXP3+ Tregs have also been found to be inherent drivers of autoimmune disorders and have been shown to exhibit multifaceted functions in the context of cancer. Recent research suggests that these cells may also be involved in tissue-specific repair and regeneration. Herein, we summarize current understanding of the thymic-transcriptional regulatory landscape of FOXP3+ Tregs, their epigenetic modulators, and associated signaling pathways. Finally, we highlight the contributions of FOXP3 on the functional development of Tregs and reflect on the clinical implications in the context of pathological and physiological immune responses.


Asunto(s)
Enfermedades Autoinmunes , Linfocitos T Reguladores , Humanos , Enfermedades Autoinmunes/metabolismo , Tolerancia Inmunológica , Inmunidad , Factores de Transcripción Forkhead
10.
Immunity ; 41(5): 675-6, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25517607

RESUMEN

Knowledge about the cells giving rise to and maintaining the thymic structure remains limited. In this issue of Immunity, Sekai et al. (2014) identify a postnatal self-renewing unipotential progenitor population capable of generating thymic medullary cells and lay the foundation for research into thymic regeneration.


Asunto(s)
Organogénesis/inmunología , Autotolerancia/inmunología , Células Madre/inmunología , Linfocitos T/inmunología , Timo/citología , Animales
11.
Immunity ; 41(6): 934-46, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25526308

RESUMEN

Gradations in extracellular regulated kinase (ERK) signaling have been implicated in essentially every developmental checkpoint or differentiation process encountered by lymphocytes. Yet, despite intensive effort, the molecular basis by which differences in ERK activation specify alternative cell fates remains poorly understood. We report here that differential ERK signaling controls lymphoid-fate specification through an alternative mode of action. While ERK phosphorylates most substrates, such as RSK, by targeting them through its D-domain, this well-studied mode of ERK action was dispensable for development of γδ T cells. Instead, development of γδ T cells was dependent upon an alternative mode of action mediated by the DEF-binding pocket (DBP) of ERK. This domain enabled ERK to bind a distinct and select set of proteins required for specification of the γδ fate. These data provide the first in vivo demonstration for the role of DBP-mediated interactions in orchestrating alternate ERK-dependent developmental outcomes.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos T/inmunología , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Activación Enzimática/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Ratones , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Estabilidad Proteica , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Transducción de Señal/genética , Especificidad por Sustrato/genética
12.
J Immunol ; 206(2): 249-256, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33397738

RESUMEN

The thymus is an intricate primary lymphoid organ, wherein bone marrow-derived lymphoid progenitor cells are induced to develop into functionally competent T cells that express a diverse TCR repertoire, which is selected to allow for the recognition of foreign Ags while avoiding self-reactivity or autoimmunity. Thymus stromal cells, which can include all non-T lineage cells, such as thymic epithelial cells, endothelial cells, mesenchymal/fibroblast cells, dendritic cells, and B cells, provide signals that are essential for thymocyte development as well as for the homeostasis of the thymic stroma itself. In this brief review, we focus on the key roles played by thymic stromal cells during early stages of T cell development, such as promoting the homing of thymic-seeding progenitors, inducing T lineage differentiation, and supporting thymocyte survival and proliferation. We also discuss recent advances on the transcriptional regulation that govern thymic epithelial cell function as well as the cellular and molecular changes that are associated with thymic involution and regeneration.


Asunto(s)
Células del Estroma/inmunología , Linfocitos T/inmunología , Timo/citología , Animales , Diferenciación Celular , Linaje de la Célula , Humanos , Activación de Linfocitos
13.
J Immunol ; 206(10): 2271-2276, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33941655

RESUMEN

T cell development is predicated on the successful rearrangement of the TCR gene loci, which encode for Ag-specific receptors. Recombination-activating gene (RAG) 2 is required for TCR gene rearrangements, which occur during specific stages of T cell development. In this study, we differentiated human pluripotent stem cells with a CRISPR/Cas9-directed deletion of the RAG2 gene (RAG2-KO) to elucidate the requirement for the TCR ß-chain in mediating ß-selection during human T cell development. In stark contrast to mice, human RAG2-KO T lineage progenitors progressed to the CD4+CD8+ double-positive (DP) stage in the absence of TCRß rearrangements. Nonetheless, RAG2-KO DPs retrovirally transduced to express a rearranged TCR ß-chain showed increased survival and proliferation as compared with control-transduced RAG2-KO DPs. Furthermore, transcriptomic analysis showed that TCRß- and control-transduced RAG2-KO DPs differed in gene pathways related to survival and proliferation. Our results provide important insights as to the distinct requirement for the TCR ß-chain during human T cell development.


Asunto(s)
Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Diferenciación Celular/genética , Células Madre Embrionarias Humanas/citología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Técnicas de Inactivación de Genes , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T/genética , Hematopoyesis/genética , Humanos , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Transducción Genética
14.
Proc Natl Acad Sci U S A ; 117(10): 5420-5429, 2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32094187

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Interferón Tipo I/metabolismo , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica , Timo/patología , Timo/virología , Animales , Atrofia/virología , Antígeno B7-H1/antagonistas & inhibidores , Enfermedad Crónica , Trasplante de Células Madre Hematopoyéticas , Interferón Tipo I/genética , Coriomeningitis Linfocítica/terapia , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Transcripción STAT2/metabolismo , Transducción de Señal , Replicación Viral
15.
Immunity ; 36(1): 105-19, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22284418

RESUMEN

Epithelial cells of mucosal tissues provide a barrier against environmental stress, and keratinocytes are key decision makers for immune cell function in the skin. Currently, epithelial signaling networks that instruct barrier immunity remain uncharacterized. Here we have shown that keratinocyte-specific deletion of a disintegrin and metalloproteinase 17 (Adam17) triggers T helper 2 and/or T helper 17 (Th2 and/or Th17) cell-driven atopic dermatitis and myeloproliferative disease. In vivo and in vitro deficiency of ADAM17 dampened Notch signaling, increasing production of the Th2 cell-polarizing cytokine TSLP and myeloid growth factor G-CSF. Ligand-independent Notch activation was identified as a regulator of AP-1 transcriptional activity, with Notch antagonizing c-Fos recruitment to the promoters of Tslp and Csf3 (G-CSF). Further, skin inflammation was rescued and myeloproliferation ameliorated by delivery of active Notch to Adam17(-)(/-) epidermis. Our findings uncover an essential role of ADAM17 in the adult epidermis, demonstrating a gatekeeper function of the ADAM17-Notch-c-Fos triad in barrier immunity.


Asunto(s)
Proteínas ADAM/metabolismo , Citocinas/metabolismo , Epidermis/enzimología , Epidermis/inmunología , Células Precursoras de Granulocitos/citología , Receptores Notch/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/inmunología , Proteína ADAM17 , Animales , Proliferación Celular , Células Epidérmicas , Eliminación de Gen , Humanos , Inflamación , Queratinocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Receptores Notch/inmunología , Transducción de Señal
16.
Proc Natl Acad Sci U S A ; 115(8): 1889-1894, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29432160

RESUMEN

Unlike αß-T lineage cells, where the role of ligand in intrathymic selection is well established, the role of ligand in the development of γδ-T cells remains controversial. Here we provide evidence for the role of a bona fide selecting ligand in shaping the γδ-T cell-receptor (TCR) repertoire. Reactivity of the γδ-TCR with the major histocompatibility complex (MHC) Class Ib ligands, H2-T10/22, is critically dependent upon the EGYEL motif in the complementarity determining region 3 (CDR3) of TCRδ. In the absence of H2-T10/22 ligand, the commitment of H2-T10/22 reactive γδ-T cells to the γδ fate is diminished, and the specification of those γδ committed cells to the IFN-γ or interleukin-17 effector fate is altered. Furthermore, those cells that do adopt the γδ fate and mature exhibit a profound alteration in the γδTCR repertoire, including depletion of the EGYEL motif and reductions in both CDR3δ length and charge. Taken together, these data suggest that ligand plays an important role in shaping the TCR repertoire of γδ-T cells.


Asunto(s)
Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Subgrupos de Linfocitos T/fisiología , Animales , Linaje de la Célula , Ligandos , Ratones , Unión Proteica , Receptores de Antígenos de Linfocitos T gamma-delta/genética
17.
Nat Methods ; 14(5): 531-538, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28394335

RESUMEN

The molecular and cellular signals that guide T-cell development from hematopoietic stem and progenitor cells (HSPCs) remain poorly understood. The thymic microenvironment integrates multiple niche molecules to potentiate T-cell development in vivo. Recapitulating these signals in vitro in a stromal cell-free system has been challenging and limits T-cell generation technologies. Here, we describe a fully defined engineered in vitro niche capable of guiding T-lineage development from HSPCs. Synergistic interactions between Notch ligand Delta-like 4 and vascular cell adhesion molecule 1 (VCAM-1) were leveraged to enhance Notch signaling and progenitor T-cell differentiation rates. The engineered thymus-like niche enables in vitro production of mouse Sca-1+cKit+ and human CD34+ HSPC-derived CD7+ progenitor T-cells capable of in vivo thymus colonization and maturation into cytokine-producing CD3+ T-cells. This engineered thymic-like niche provides a platform for in vitro analysis of human T-cell development as well as clinical-scale cell production for future development of immunotherapeutic applications.


Asunto(s)
Diferenciación Celular , Células Madre Hematopoyéticas/citología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Linfocitos T/citología , Molécula 1 de Adhesión Celular Vascular/metabolismo , Biotecnología/métodos , Complejo CD3/inmunología , Células Madre Hematopoyéticas/inmunología , Humanos , Transducción de Señal , Linfocitos T/inmunología , Timo/citología , Timo/inmunología
18.
J Immunol ; 201(9): 2664-2682, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30257885

RESUMEN

During T cell development, progenitor thymocytes undergo a large proliferative burst immediately following successful TCRß rearrangement, and defects in genes that regulate this proliferation have a profound effect on thymus cellularity and output. Although the signaling pathways that initiate cell cycling and nutrient uptake after TCRß selection are understood, less is known about the transcriptional programs that regulate the metabolic machinery to promote biomass accumulation during this process. In this article, we report that mice with whole body deficiency in the nuclear receptor peroxisome proliferator-activated receptor-δ (PPARδmut) exhibit a reduction in spleen and thymus cellularity, with a decrease in thymocyte cell number starting at the double-negative 4 stage of thymocyte development. Although in vivo DNA synthesis was normal in PPARδmut thymocytes, studies in the OP9-delta-like 4 in vitro system of differentiation revealed that PPARδmut double-negative 3 cells underwent fewer cell divisions. Naive CD4+ T cells from PPARδmut mice also exhibited reduced proliferation upon TCR and CD28 stimulation in vitro. Growth defects in PPAR-δ-deficient thymocytes and peripheral CD4+ T cells correlated with decreases in extracellular acidification rate, mitochondrial reserve, and expression of a host of genes involved in glycolysis, oxidative phosphorylation, and lipogenesis. By contrast, mice with T cell-restricted deficiency of Ppard starting at the double-positive stage of thymocyte development, although exhibiting defective CD4+ T cell growth, possessed a normal T cell compartment, pointing to developmental defects as a cause of peripheral T cell lymphopenia in PPARδmut mice. These findings implicate PPAR-δ as a regulator of the metabolic program during thymocyte and T cell growth.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/inmunología , Receptores Citoplasmáticos y Nucleares/metabolismo , Timocitos/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Proliferación Celular/fisiología , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Timocitos/inmunología
19.
Immunol Cell Biol ; 97(4): 427-434, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30633397

RESUMEN

The differentiation of human hematopoietic stem cells into CD8 T cells can be achieved in vitro with the OP9-DL4 system. We considered that in the absence of medullary thymic epithelial cells, which serve to restrict the breath of the T-cell receptor (TCR) repertoire by expressing tissue-restricted antigens, a distinct repertoire would be generated in vitro. To test this notion, we compared the TCR-Vα/Vß (TRAV/TRBV) gene usage of major histocompatibility complex-restricted antigen (MART-1)-specific T cells generated in vitro to that from ex vivo naïve T cells and tumor-infiltrating lymphocytes (TILs) using high-throughput DNA sequencing. In contrast to naïve T cells and TILs, which showed the expected narrow TRAV repertoire, in vitro-generated MART-1-specific T cells used almost all TRAV gene families and displayed unique CDR3 lengths. Our work demonstrates that the OP9-DL4 system supports the creation of a broad antigen-specific TCR repertoire, suggesting that T cells generated in vitro may undergo a different set of selection events that otherwise constrains the TCR repertoire of thymus-derived T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Antígeno MART-1/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Línea Celular , Regiones Determinantes de Complementariedad/genética , Células Madre Hematopoyéticas/metabolismo , Humanos
20.
Int Immunol ; 30(12): 541-550, 2018 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-30102361

RESUMEN

T lymphocytes are critical mediators of the adaptive immune system and they can be harnessed as therapeutic agents against pathogens and in cancer immunotherapy. T cells can be isolated and expanded from patients and potentially generated in vitro using clinically relevant systems. An ultimate goal for T-cell immunotherapy is to establish a safe, universal effector cell type capable of transcending allogeneic and histocompatibility barriers. To this end, human pluripotent stem cells offer an advantage in generating a boundless supply of T cells that can be readily genetically engineered. Here, we review emerging T-cell therapeutics, including tumor-infiltrating lymphocytes, chimeric antigen receptors and progenitor T cells (proT cells) as well as feeder cell-free in vitro systems for their generation. Furthermore, we explore their potential for adoption in the clinic and highlight the challenges that must be addressed to increase the therapeutic success of a universal immunotherapy.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T/inmunología , Inmunidad Adaptativa/inmunología , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA