Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
Nat Immunol ; 24(12): 2121-2134, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37945821

RESUMEN

The T cell antigen receptor (TCR) contains ten immunoreceptor tyrosine-based activation motif (ITAM) signaling sequences distributed within six CD3 subunits; however, the reason for such structural complexity and multiplicity is unclear. Here we evaluated the effect of inactivating the three CD3ζ chain ITAMs on TCR signaling and T cell effector responses using a conditional 'switch' mouse model. Unexpectedly, we found that T cells expressing TCRs containing inactivated (non-signaling) CD3ζ ITAMs (6F-CD3ζ) exhibited reduced ability to discriminate between low- and high-affinity ligands, resulting in enhanced signaling and cytokine responses to low-affinity ligands because of a previously undetected inhibitory function of CD3ζ ITAMs. Also, 6F-CD3ζ TCRs were refractory to antagonism, as predicted by a new in silico adaptive kinetic proofreading model that revises the role of ITAM multiplicity in TCR signaling. Finally, T cells expressing 6F-CD3ζ displayed enhanced cytolytic activity against solid tumors expressing low-affinity ligands, identifying a new counterintuitive approach to TCR-mediated cancer immunotherapy.


Asunto(s)
Motivo de Activación del Inmunorreceptor Basado en Tirosina , Receptores de Antígenos de Linfocitos T , Animales , Ratones , Complejo CD3 , Ligandos , Péptidos , Linfocitos T
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
4.
Nat Immunol ; 18(4): 433-441, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28250424

RESUMEN

THEMIS, a T cell-specific protein with high expression in CD4+CD8+ thymocytes, has a crucial role in positive selection and T cell development. THEMIS lacks defined catalytic domains but contains two tandem repeats of a distinctive module of unknown function (CABIT). Here we found that THEMIS directly regulated the catalytic activity of the tyrosine phosphatase SHP-1. This action was mediated by the CABIT modules, which bound to the phosphatase domain of SHP-1 and promoted or stabilized oxidation of SHP-1's catalytic cysteine residue, which inhibited the tyrosine-phosphatase activity of SHP-1. Deletion of SHP-1 alleviated the developmental block in Themis-/- thymocytes. Thus, THEMIS facilitates thymocyte positive selection by enhancing the T cell antigen receptor signaling response to low-affinity ligands.


Asunto(s)
Selección Clonal Mediada por Antígenos/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular , Eliminación de Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Oxidación-Reducción , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteína Tirosina Fosfatasa no Receptora Tipo 6/antagonistas & inhibidores , Proteína Tirosina Fosfatasa no Receptora Tipo 6/química , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Especies Reactivas de Oxígeno/metabolismo , Linfocitos T/citología , Timocitos/citología , Timocitos/inmunología , Timocitos/metabolismo
5.
Nat Immunol ; 18(2): 205-213, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27992403

RESUMEN

The positive and negative selection of lymphocytes by antigen is central to adaptive immunity and self-tolerance, yet how this is determined by different antigens is not completely understood. We found that thymocyte-selection-associated family member 2 (Themis2) increased the positive selection of B1 cells and germinal center B cells by self and foreign antigens. Themis2 lowered the threshold for B-cell activation by low-avidity, but not high-avidity, antigens. Themis2 constitutively bound the adaptor protein Grb2, src-kinase Lyn and signal transducer phospholipase γ2 (PLC-γ2), and increased activation of PLC-γ2 and its downstream pathways following B cell receptor stimulation. Our findings identify a unique function for Themis2 in differential signaling and provide insight into how B cells discriminate between antigens of different quantity and quality.


Asunto(s)
Linfocitos B/fisiología , Selección Clonal Mediada por Antígenos , Centro Germinal/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Activación de Linfocitos , Inmunidad Adaptativa , Animales , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Proteína Adaptadora GRB2/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fosfolipasa C gamma/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Autotolerancia , Familia-src Quinasas/metabolismo
6.
Nat Immunol ; 16(9): 980-90, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26214741

RESUMEN

Follicular helper T cells (T(FH) cells) are specialized effector CD4(+) T cells that help B cells develop germinal centers (GCs) and memory. However, the transcription factors that regulate the differentiation of T(FH) cells remain incompletely understood. Here we report that selective loss of Lef1 or Tcf7 (which encode the transcription factor LEF-1 or TCF-1, respectively) resulted in T(FH) cell defects, while deletion of both Lef1 and Tcf7 severely impaired the differentiation of T(FH) cells and the formation of GCs. Forced expression of LEF-1 enhanced T(FH) differentiation. LEF-1 and TCF-1 coordinated such differentiation by two general mechanisms. First, they established the responsiveness of naive CD4(+) T cells to T(FH) cell signals. Second, they promoted early T(FH) differentiation via the multipronged approach of sustaining expression of the cytokine receptors IL-6Rα and gp130, enhancing expression of the costimulatory receptor ICOS and promoting expression of the transcriptional repressor Bcl6.


Asunto(s)
Diferenciación Celular/inmunología , Receptor gp130 de Citocinas/inmunología , Proteínas de Unión al ADN/inmunología , Centro Germinal/inmunología , Factor Nuclear 1-alfa del Hepatocito/inmunología , Factor de Unión 1 al Potenciador Linfoide/inmunología , Receptores de Interleucina-6/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Linfocitos B/inmunología , Diferenciación Celular/genética , Receptor gp130 de Citocinas/genética , Proteínas de Unión al ADN/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Centro Germinal/metabolismo , Factor Nuclear 1-alfa del Hepatocito/genética , Factor de Unión 1 al Potenciador Linfoide/genética , Ratones , Proteínas Proto-Oncogénicas c-bcl-6 , Receptores de Interleucina-6/genética , Linfocitos T Colaboradores-Inductores/metabolismo
7.
Nat Immunol ; 15(10): 947-56, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25129370

RESUMEN

The transcription factor ThPOK promotes CD4(+) T cell differentiation in the thymus. Here, using a mouse strain that allows post-thymic gene deletion, we show that ThPOK maintains CD4(+) T lineage integrity and couples effector differentiation to environmental cues after antigenic stimulation. ThPOK preserved the integrity and amplitude of effector responses and was required for proper differentiation of types 1 and 2 helper T cells in vivo by restraining the expression and function of Runx3, a nuclear factor crucial for cytotoxic T cell differentiation. The transcription factor LRF acts redundantly with ThPOK to prevent the transdifferentiation of mature CD4(+) T cells into CD8(+) T cells. As such, the ThPOK-LRF transcriptional module was essential for CD4(+) T cell integrity and responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Proteínas de Unión al ADN/inmunología , Timo/inmunología , Factores de Transcripción/inmunología , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Células TH1/inmunología , Células TH1/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Timo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma/inmunología
9.
Trends Immunol ; 42(8): 735-750, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34261578

RESUMEN

T cell receptor (TCR) ß-selection (herein referred to as ß-selection) is a pivotal checkpoint in mammalian T cell development when immature CD4-CD8- T-cells (thymocytes) express pre-TCR following successful Tcrb gene rearrangement. At this stage, αß T cell lineage commitment and allelic exclusion to restrict one ß-chain per cell take place and thymocytes undergo a proliferative burst. ß-selection is known to be crucially dependent upon synchronized Notch and pre-TCR signaling; however, other necessary inputs have been identified over the past decade, expanding our knowledge and understanding of the ß-selection process. In this review, we discuss recent mechanistic findings that have enabled a more detailed decoding of the molecular dynamics of the ß-selection checkpoint and have helped to elucidate its role in early T cell development.


Asunto(s)
Receptores de Antígenos de Linfocitos T alfa-beta , Transducción de Señal , Animales , Diferenciación Celular , Activación de Linfocitos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Timo
10.
Immunity ; 42(3): 395-6, 2015 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-25786168

RESUMEN

The role of co-receptor molecules in the generation of inducible regulatory T cells (iTregs) remains incompletely defined. In this issue of Immunity, Henderson et al. (2015) show that CD5 regulates iTreg cell induction by rendering emerging iTreg cells refractory to signals mediated by effector-differentiating cytokines.


Asunto(s)
Autoantígenos/inmunología , Antígenos CD5/inmunología , Linfocitos T Reguladores/metabolismo , Animales
11.
Nat Immunol ; 12(2): 129-36, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21186366

RESUMEN

The nuclear adaptor Ldb1 functions as a core component of multiprotein transcription complexes that regulate differentiation in diverse cell types. In the hematopoietic lineage, Ldb1 forms a complex with the non-DNA-binding adaptor Lmo2 and the transcription factors E2A, Scl and GATA-1 (or GATA-2). Here we demonstrate a critical and continuous requirement for Ldb1 in the maintenance of both fetal and adult mouse hematopoietic stem cells (HSCs). Deletion of Ldb1 in hematopoietic progenitors resulted in the downregulation of many transcripts required for HSC maintenance. Genome-wide profiling by chromatin immunoprecipitation followed by sequencing (ChIP-Seq) identified Ldb1 complex-binding sites at highly conserved regions in the promoters of genes involved in HSC maintenance. Our results identify a central role for Ldb1 in regulating the transcriptional program responsible for the maintenance of HSCs.


Asunto(s)
Células Madre Adultas/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Desarrollo Fetal , Células Madre Hematopoyéticas/metabolismo , Traslado Adoptivo , Células Madre Adultas/citología , Células Madre Adultas/inmunología , Células Madre Adultas/trasplante , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Células Madre Embrionarias/citología , Células Madre Embrionarias/inmunología , Células Madre Embrionarias/trasplante , Femenino , Desarrollo Fetal/genética , Desarrollo Fetal/inmunología , Regulación del Desarrollo de la Expresión Génica/inmunología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Proteínas con Dominio LIM , Ratones , Ratones Noqueados , Ratones Transgénicos , Embarazo , Unión Proteica , Elementos Reguladores de la Transcripción/genética , Elementos Reguladores de la Transcripción/inmunología , Activación Transcripcional/genética , Activación Transcripcional/inmunología
12.
Proc Natl Acad Sci U S A ; 117(25): 14342-14353, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32513716

RESUMEN

Immature T cells undergo a process of positive selection in the thymus when their new T cell receptor (TCR) engages and signals in response to self-peptides. As the T cell matures, a slew of negative regulatory molecules, including the inhibitory surface glycoprotein CD5, are up-regulated in proportion to the strength of the self-peptide signal. Together these regulators dampen TCR-proximal signaling and help avoid any subsequent peripheral activation of T cells by self-peptides. Paradoxically, antigen-specific T cells initially expressing more CD5 (CD5hi) have been found to better persist as effector/memory cells after a peripheral challenge. The molecular mechanisms underlying such a duality in CD5 function is not clear. We found that CD5 alters the basal activity of the NF-κB signaling in resting peripheral T cells. When CD5 was conditionally ablated, T cells were unable to maintain higher expression of the cytoplasmic NF-κB inhibitor IκBα. Consistent with this, resting CD5hi T cells expressed more of the NF-κB p65 protein than CD5lo cells, without significant increases in transcript levels, in the absence of TCR signals. This posttranslationally stabilized cellular NF-κB depot potentially confers a survival advantage to CD5hi T cells over CD5lo ones. Taken together, these data suggest a two-step model whereby the strength of self-peptide-induced TCR signal lead to the up-regulation of CD5, which subsequently maintains a proportional reserve of NF-κB in peripheral T cells poised for responding to agonistic antigen-driven T cell activation.


Asunto(s)
Antígenos CD5/metabolismo , Regulación del Desarrollo de la Expresión Génica/inmunología , Inhibidor NF-kappaB alfa/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Linfocitos T/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno/inmunología , Antígenos CD5/genética , Línea Celular Tumoral , Separación Celular , Supervivencia Celular/inmunología , Femenino , Citometría de Flujo , Lipopolisacáridos/inmunología , Activación de Linfocitos , Ratones , Ratones Noqueados , Modelos Animales , Cultivo Primario de Células , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Transducción de Señal/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante , Timo/citología , Timo/crecimiento & desarrollo , Timo/inmunología , Factor de Transcripción ReIA/metabolismo , Regulación hacia Arriba
13.
Proc Natl Acad Sci U S A ; 117(23): 12969-12979, 2020 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-32434911

RESUMEN

CD5 is characterized as an inhibitory coreceptor with an important regulatory role during T cell development. The molecular mechanism by which CD5 operates has been puzzling and its function in mature T cells suggests promoting rather than repressing effects on immune responses. Here, we combined quantitative mass spectrometry and genetic studies to analyze the components and the activity of the CD5 signaling machinery in primary T cells. We found that T cell receptor (TCR) engagement induces the selective phosphorylation of CD5 tyrosine 429, which serves as a docking site for proteins with adaptor functions (c-Cbl, CIN85, CRKL), connecting CD5 to positive (PI3K) and negative (UBASH3A, SHIP1) regulators of TCR signaling. c-CBL acts as a coordinator in this complex enabling CD5 to synchronize positive and negative feedbacks on TCR signaling through the other components. Disruption of CD5 signalosome in mutant mice reveals that it modulates TCR signal outputs to selectively repress the transactivation of Foxp3 and limit the inopportune induction of peripherally induced regulatory T cells during immune responses against foreign antigen. Our findings bring insights into the paradigm of coreceptor signaling, suggesting that, in addition to providing dualistic enhancing or dampening inputs, coreceptors can engage concomitant stimulatory and inhibitory signaling events, which act together to promote specific functional outcomes.


Asunto(s)
Antígenos/inmunología , Antígenos CD5/metabolismo , Diferenciación Celular/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T Reguladores/fisiología , Animales , Antígenos CD5/genética , Diferenciación Celular/genética , Regulación de la Expresión Génica/inmunología , Activación de Linfocitos/genética , Espectrometría de Masas , Ratones , Ratones Transgénicos , Cultivo Primario de Células , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Transducción de Señal/genética , Transducción de Señal/inmunología
14.
Blood ; 135(25): 2252-2265, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32181817

RESUMEN

Prolonged or enhanced expression of the proto-oncogene Lmo2 is associated with a severe form of T-cell acute lymphoblastic leukemia (T-ALL), designated early T-cell precursor ALL, which is characterized by the aberrant self-renewal and subsequent oncogenic transformation of immature thymocytes. It has been suggested that Lmo2 exerts these effects by functioning as component of a multi-subunit transcription complex that includes the ubiquitous adapter Ldb1 along with b-HLH and/or GATA family transcription factors; however, direct experimental evidence for this mechanism is lacking. In this study, we investigated the importance of Ldb1 for Lmo2-induced T-ALL by conditional deletion of Ldb1 in thymocytes in an Lmo2 transgenic mouse model of T-ALL. Our results identify a critical requirement for Ldb1 in Lmo2-induced thymocyte self-renewal and thymocyte radiation resistance and for the transition of preleukemic thymocytes to overt T-ALL. Moreover, Ldb1 was also required for acquisition of the aberrant preleukemic ETP gene expression signature in immature Lmo2 transgenic thymocytes. Co-binding of Ldb1 and Lmo2 was detected at the promoters of key upregulated T-ALL driver genes (Hhex, Lyl1, and Nfe2) in preleukemic Lmo2 transgenic thymocytes, and binding of both Ldb1 and Lmo2 at these sites was reduced following Cre-mediated deletion of Ldb1. Together, these results identify a key role for Ldb1, a nonproto-oncogene, in T-ALL and support a model in which Lmo2-induced T-ALL results from failure to downregulate Ldb1/Lmo2-nucleated transcription complexes which normally function to enforce self-renewal in bone marrow hematopoietic progenitors.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Autorrenovación de las Células , Proteínas de Unión al ADN/fisiología , Proteínas con Dominio LIM/fisiología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Timocitos/citología , Proteínas Adaptadoras Transductoras de Señales/genética , Traslado Adoptivo , Animales , Antígenos CD/biosíntesis , Transformación Celular Neoplásica , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Eliminación de Gen , Técnicas de Sustitución del Gen , Proteínas con Dominio LIM/deficiencia , Proteínas con Dominio LIM/genética , Linfopoyesis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proto-Oncogenes Mas , RNA-Seq , Quimera por Radiación , Tolerancia a Radiación , Timocitos/metabolismo , Timocitos/efectos de la radiación , Timocitos/trasplante
15.
Eur J Immunol ; 50(7): 986-999, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32144749

RESUMEN

SOCS3 is a cytosolic inhibitor of cytokine signaling that suppresses the activation of cytokine receptor-associated JAK kinases. Mechanistically, SOCS3 is recruited to a site in the cytokine receptors known as the SOCS3-interaction motif, and then binds JAK molecules to inhibit their kinase activity. The SOCS3-interaction motif is found in receptors of the gp130 cytokine family but mostly absent from other cytokine receptors, including γc. Thus, SOCS3 has been considered a selective suppressor of gp130 family cytokines, but not γc cytokines. Considering that γc signaling induces SOCS3 expression in T cells, here we revisited the role of SOCS3 on γc signaling. Using SOCS3 transgenic mice, we found that increased abundance of SOCS3 not only suppressed signaling of the gp130 family cytokine IL-6, but also signaling of the γc family cytokine IL-7. Consequently, SOCS3 transgenic mice were impaired in IL-7-dependent T cell development in the thymus and the homeostasis of mature T cells in peripheral tissues. Moreover, enforced SOCS3 expression interfered with the generation of Foxp3+ regulatory T cells that requires signaling by the γc family cytokine IL-2. Collectively, we report an underappreciated role for SOCS3 in suppressing γc cytokine signaling, effectively expanding its scope of target cytokines in T cell immunity.


Asunto(s)
Citocinas/inmunología , Inmunidad Celular , Transducción de Señal/inmunología , Proteína 3 Supresora de la Señalización de Citocinas/inmunología , Linfocitos T Reguladores/inmunología , Animales , Femenino , Factores de Transcripción Forkhead/inmunología , Masculino , Ratones , Linfocitos T Reguladores/citología
16.
Nat Immunol ; 10(8): 840-7, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19597498

RESUMEN

During positive selection, thymocytes transition through a stage during which T cell antigen receptor (TCR) signaling controls CD4-versus-CD8 lineage 'choice' and subsequent maturation. Here we describe a previously unknown T cell-specific protein, Themis, that serves a distinct function during this stage. In Themis(-/-) mice, thymocyte selection was impaired and the number of transitional CD4(+)CD8(int) thymocytes as well as CD4(+) or CD8(+) single-positive thymocytes was lower. Notably, although we detected no overt TCR-proximal signaling deficiencies, Themis(-/-) CD4(+)CD8(int) thymocytes showed developmental defects consistent with attenuated signaling that were reversible by TCR stimulation. Our results identify Themis as a critical component of the T cell developmental program and suggest that Themis functions to sustain and/or integrate signals required for proper lineage commitment and maturation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Linaje de la Célula/fisiología , Proteínas/fisiología , Receptores de Antígenos de Linfocitos T/fisiología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Células Madre Embrionarias/metabolismo , Femenino , Citometría de Flujo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas/genética , Proteínas/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal
17.
Trends Immunol ; 38(9): 622-632, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28697966

RESUMEN

THEMIS, a recently identified T-lineage-restricted protein, is the founding member of a large metazoan protein family. Gene inactivation studies have revealed a critical requirement for THEMIS during thymocyte positive selection, implicating THEMIS in signaling downstream of the T cell antigen receptor (TCR), but the mechanistic underpinnings of THEMIS function have remained elusive. A previous model posited that THEMIS prevents thymocytes from inappropriately crossing the positive/negative selection threshold by dampening TCR signaling. However, new data suggest an alternative model where THEMIS enhances TCR signaling enabling thymocytes to reach the threshold for positive selection, avoiding death by neglect. We review the data supporting each model and conclude that the preponderance of evidence favors an enhancing function for THEMIS in TCR signaling.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Inmunológicos , Linfocitos T/fisiología , Timocitos/fisiología , Animales , Diferenciación Celular , Cisteína/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Activación de Linfocitos , Dominios Proteicos/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal
18.
Proc Natl Acad Sci U S A ; 113(8): E1016-25, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26862168

RESUMEN

Primary T-cell acute lymphoblastic leukemia (T-ALL) cells require stromal-derived signals to survive. Although many studies have identified cell-intrinsic alterations in signaling pathways that promote T-ALL growth, the identity of endogenous stromal cells and their associated signals in the tumor microenvironment that support T-ALL remains unknown. By examining the thymic tumor microenvironments in multiple murine T-ALL models and primary patient samples, we discovered the emergence of prominent epithelial-free regions, enriched for proliferating tumor cells and dendritic cells (DCs). Systematic evaluation of the functional capacity of tumor-associated stromal cells revealed that myeloid cells, primarily DCs, are necessary and sufficient to support T-ALL survival ex vivo. DCs support T-ALL growth both in primary thymic tumors and at secondary tumor sites. To identify a molecular mechanism by which DCs support T-ALL growth, we first performed gene expression profiling, which revealed up-regulation of platelet-derived growth factor receptor beta (Pdgfrb) and insulin-like growth factor I receptor (Igf1r) on T-ALL cells, with concomitant expression of their ligands by tumor-associated DCs. Both Pdgfrb and Igf1r were activated in ex vivo T-ALL cells, and coculture with tumor-associated, but not normal thymic DCs, sustained IGF1R activation. Furthermore, IGF1R signaling was necessary for DC-mediated T-ALL survival. Collectively, these studies provide the first evidence that endogenous tumor-associated DCs supply signals driving T-ALL growth, and implicate tumor-associated DCs and their mitogenic signals as auspicious therapeutic targets.


Asunto(s)
Células Dendríticas/inmunología , Proteínas de Neoplasias/inmunología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/inmunología , Receptores de Somatomedina/inmunología , Transducción de Señal/inmunología , Microambiente Tumoral/inmunología , Animales , Línea Celular Tumoral , Supervivencia Celular , Células Dendríticas/patología , Femenino , Humanos , Masculino , Ratones , Proteínas de Neoplasias/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Receptor IGF Tipo 1 , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/inmunología , Receptores de Somatomedina/genética , Transducción de Señal/genética , Microambiente Tumoral/genética
19.
Trends Genet ; 30(1): 1-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24290192

RESUMEN

Elucidation of the genetic pathways that control red blood cell development has been a central goal of erythropoiesis research over the past decade. Notably, data from several recent studies have provided new insights into the regulation of erythroid gene transcription. Transcription profiling demonstrates that erythropoiesis is mainly controlled by a small group of lineage-restricted transcription factors [Gata binding protein 1 (Gata1), T cell acute lymphocytic leukemia 1 protein (Tal1), and Erythroid Kruppel-like factor (EKLF; henceforth referred to as Klf1)]. Binding-site mapping using ChIP-Seq indicates that most DNA-bound Gata1 and Tal1 proteins are contained within higher order complexes (Ldb1 complexes) that include the nuclear adapters Ldb1 and Lmo2. Ldb1 complexes regulate Klf1, and Ldb1 complex-binding sites frequently colocalize with Klf1 at erythroid genes and cis-regulatory elements, indicating strong functional synergy between Gata1, Tal1, and Klf1. Together with new data demonstrating that Ldb1 can mediate long-range promoter-enhancer interactions, these findings provide a foundation for the first comprehensive models of the global regulation of erythroid gene transcription.


Asunto(s)
Eritropoyesis/genética , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Sitios de Unión/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Represión Epigenética , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Regulación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Ratones , Modelos Animales , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Análisis de Secuencia de ADN , Proteína 1 de la Leucemia Linfocítica T Aguda , Factores de Transcripción/genética
20.
Immunol Cell Biol ; 95(5): 473-477, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28244489

RESUMEN

V-(D)-J recombination generates the antigen receptor diversity necessary for immune cell function, while allelic exclusion ensures that each cell expresses a single antigen receptor. V-(D)-J recombination of the Ig, Tcrb, Tcrg and Tcrd antigen receptor genes is ordered and sequential so that only one allele generates a productive rearrangement. The mechanism controlling sequential rearrangement of antigen receptor genes, in particular how only one allele is selected to initiate recombination while at least temporarily leaving the other intact, remains unresolved. Genomic imprinting, a widespread phenomenon wherein maternal or paternal allele inheritance determines allele activity, could represent a regulatory mechanism for controlling sequential V-(D)-J rearrangement. We used strain-specific single-nucleotide polymorphisms within antigen receptor genes to determine if maternal vs paternal inheritance could underlie chromosomal choice for the initiation of recombination. We found no parental chromosomal bias in the initiation of V-(D)-J recombination in T or B cells, eliminating genomic imprinting as a potential regulator for this tightly regulated process.


Asunto(s)
Cromosomas de los Mamíferos/genética , Impresión Genómica , Recombinación V(D)J/genética , Animales , Femenino , Masculino , Ratones , Mapeo Restrictivo , Análisis de Secuencia de ADN
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda