Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Methods Mol Biol ; 2580: 151-163, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36374455

RESUMEN

Many analytical or cell culture procedures require homogeneous starting cell populations that cannot be obtained directly from organ dissection. Here, we describe two enrichment procedures to achieve this goal and discuss their respective advantages in specific experimental contexts.


Asunto(s)
Subgrupos de Linfocitos T , Timocitos , Citometría de Flujo/métodos
2.
Nat Immunol ; 23(4): 594-604, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35354951

RESUMEN

While T cell receptor (TCR) αß+CD8α+CD8ß- intraepithelial lymphocytes (CD8αα+ IELs) differentiate from thymic IEL precursors (IELps) and contribute to gut homeostasis, the transcriptional control of their development remains poorly understood. In the present study we showed that mouse thymocytes deficient for the transcription factor leukemia/lymphoma-related factor (LRF) failed to generate TCRαß+CD8αα+ IELs and their CD8ß-expressing counterparts, despite giving rise to thymus and spleen CD8αß+ T cells. LRF-deficient IELps failed to migrate to the intestine and to protect against T cell-induced colitis, and had impaired expression of the gut-homing integrin α4ß7. Single-cell RNA-sequencing found that LRF was necessary for the expression of genes characteristic of the most mature IELps, including Itgb7, encoding the ß7 subunit of α4ß7. Chromatin immunoprecipitation and gene-regulatory network analyses both defined Itgb7 as an LRF target. Our study identifies LRF as an essential transcriptional regulator of IELp maturation in the thymus and subsequent migration to the intestinal epithelium.


Asunto(s)
Linfocitos Intraepiteliales , Leucemia , Linfoma , Animales , Antígenos CD8/genética , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/metabolismo , Cadenas beta de Integrinas , Mucosa Intestinal/metabolismo , Linfocitos Intraepiteliales/metabolismo , Leucemia/metabolismo , Linfoma/metabolismo , Ratones , Ratones Noqueados , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Factores de Transcripción/metabolismo
3.
J Immunol ; 199(5): 1716-1728, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28754678

RESUMEN

The CD4+ lineage-specific transcription factor Thpok is required for intrathymic CD4+ T cell differentiation and, together with its homolog LRF, supports CD4+ T cell helper effector responses. However, it is not known whether these factors are needed for the regulatory T cell (Treg) arm of MHC class II responses. In this study, by inactivating in mice the genes encoding both factors in differentiated Tregs, we show that Thpok and LRF are redundantly required to maintain the size and functions of the postthymic Treg pool. They support IL-2-mediated gene expression and the functions of the Treg-specific factor Foxp3. Accordingly, Treg-specific disruption of Thpok and Lrf causes a lethal inflammatory syndrome similar to that resulting from Treg deficiency. Unlike in conventional T cells, Thpok and LRF functions in Tregs are not mediated by their repression of the transcription factor Runx3. Additionally, we found that Thpok is needed for the differentiation of thymic Treg precursors, an observation in line with the fact that Foxp3+ Tregs are CD4+ cells. Thus, a common Thpok-LRF node supports both helper and regulatory arms of MHC class II responses.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Linfocitos T Reguladores/inmunología , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Proteínas de Unión al ADN/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Interleucina-2/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/microbiología , Factores de Transcripción/genética
4.
JCI Insight ; 2(12)2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28614804

RESUMEN

Hematopoietic stem cell transplantation (HSCT) offers a cure for cancers that are refractory to chemotherapy and radiation. Most HSCT recipients develop chronic graft-versus-host disease (cGVHD), a systemic alloimmune attack on host organs. Diagnosis is based on clinical signs and symptoms, as biopsies are risky. T cells are central to the biology of cGVHD. We found that a low Treg/CD4+ T effector memory (Tem) ratio in circulation, lymphoid, and target organs identified early and established mouse cGVHD. Using deuterated water labeling to measure multicompartment in vivo kinetics of these subsets, we show robust Tem and Treg proliferation in lymphoid and target organs, while Tregs undergo apoptosis in target organs. Since deuterium enrichment into DNA serves as a proxy for cell proliferation, we developed a whole-body clinically relevant deuterium MRI approach to nonradioactively detect cGVHD and potentially allow imaging of other diseases characterized by rapidly proliferating cells.

5.
Methods Mol Biol ; 1323: 87-97, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26294400

RESUMEN

Many analytical or cell culture procedures require homogeneous starting cell populations that cannot be obtained directly from organ dissection. Here, we describe two enrichment procedures to achieve this goal and discuss their respective advantages in specific experimental contexts. Notes in this chapter include some tips on how to determine the appropriate level of purity (see Note 1 ).


Asunto(s)
Separación Inmunomagnética/métodos , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo , Timocitos/citología , Timocitos/metabolismo , Citometría de Flujo/métodos , Inmunofenotipificación
6.
Methods Mol Biol ; 1323: 117-28, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26294403

RESUMEN

In vitro culture is an important complement, or substitute, to in vivo approaches in order to study T cell effector differentiation. Here, we describe culture conditions that generate specific effector cell types by exposing naïve T cells to appropriate cytokine signals.


Asunto(s)
Diferenciación Celular , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/metabolismo , Animales , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/metabolismo , Separación Celular , Células Dendríticas/citología , Células Dendríticas/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Inmunofenotipificación , Técnicas In Vitro , Ratones , Bazo/citología
7.
J Immunol ; 193(8): 4032-42, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-25217159

RESUMEN

Differentiation of CD4(+) helper and CD8(+) cytotoxic αß T cells from CD4(+)CD8(+) thymocytes involves upregulation of lineage-specifying transcription factors and transcriptional silencing of CD8 or CD4 coreceptors, respectively, in MHC class II or I (MHCII or I)-restricted thymocytes. In this study, we demonstrate that inactivation of the Dicer RNA endonuclease in murine thymocytes impairs initiation of Cd4 and Cd8 silencing, leading to development of positively selected MHCI- and MHCII-restricted mature CD4(+)CD8(+) thymocytes. Expression of the antiapoptotic BCL2 protein or inactivation of the p53 proapoptotic protein rescues these thymocytes from apoptosis, increasing their frequency and permitting accumulation of CD4(+)CD8(+) αß T cells in the periphery. Dicer-deficient MHCI-restricted αß T cells fail to normally silence Cd4 and display impaired induction of the CD8 lineage-specifying transcription factor Runx3, whereas Dicer-deficient MHCII-restricted αß T cells show impaired Cd8 silencing and impaired induction of the CD4 lineage-specifying transcription factor Thpok. Finally, we show that the Drosha RNA endonuclease, which functions upstream of Dicer in microRNA biogenesis, also regulates Cd4 and Cd8 silencing. Our data demonstrate a previously dismissed function for the microRNA biogenesis machinery in regulating expression of lineage-specifying transcription factors and silencing of Cd4 and Cd8 during αß T cell differentiation.


Asunto(s)
ARN Helicasas DEAD-box/genética , MicroARNs/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Ribonucleasa III/genética , Linfocitos T Citotóxicos/citología , Linfocitos T Colaboradores-Inductores/citología , Animales , Apoptosis/inmunología , Antígenos CD4/genética , Antígenos CD4/inmunología , Antígenos CD8/genética , Antígenos CD8/inmunología , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Subunidad alfa 3 del Factor de Unión al Sitio Principal/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Ratones , Ratones Noqueados , MicroARNs/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Factores de Transcripción/inmunología , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba
8.
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.
Immunity ; 37(4): 622-33, 2012 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-23041065

RESUMEN

T helper (Th) cells are critical for defenses against infection and recognize peptides bound to class II major histocompatibility complex (MHC II) molecules. Although transcription factors have been identified that direct Th cells into specific effector fates, whether a "master" regulator controls the developmental program common to all Th cells remains unclear. Here, we showed that the two transcription factors Thpok and LRF share this function. Although disruption of both factors did not prevent the generation of MHC II-specific T cells, these cells failed to express Th cell genes or undergo Th cell differentiation in vivo. In contrast, T cells lacking Thpok, which only displayed LRF-dependent functions, contributed to multiple effector responses, both in vitro and in vivo, with the notable exception of Th2 cell responses that control extracellular parasites. These findings identify the Thpok-LRF pair as a core node of Th cell differentiation and function.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Factores de Transcripción/inmunología , Animales , Células Cultivadas , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Colaboradores-Inductores/citología , Factores de Transcripción/deficiencia
10.
Proc Natl Acad Sci U S A ; 108(45): 18330-5, 2011 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-22025728

RESUMEN

Cd8a and Cd8b1 coreceptor gene (Cd8) expression is tightly controlled during T-cell development by the activity of five Cd8 enhancers (E8(I)-E8(V)). Here we demonstrate a unique transcriptional program regulating CD8 expression during CD8(+) effector T-cell differentiation. The Cd8 enhancer E8(I) and Runx/core-binding factor-ß (CBFß) complexes were required for the establishment of this regulatory circuit, because E8(I)-, Runx3-, or CBFß-deficient CD8(+) T cells down-regulated CD8α expression during activation. This finding correlated with enhanced repressive histone marks at the Cd8a promoter in the absence of E8(I), and the down-regulation of CD8α expression could be blocked by treating E8(I)-, Runx3-, or CBFß-deficient CD8(+) T cells with the histone deacetylase inhibitor trichostatin A. Moreover, Runx/CBFß complexes bound the Cd8ab gene cluster in activated CD8(+) T cells, suggesting direct control of the Cd8a locus. However, CD8(+) effector T cells maintained high levels of CD8α when CBFß was conditionally deleted after activation. Thus, our data suggest an E8(I)- and Runx3/CBFß-dependent epigenetic programming of the Cd8a locus during T-cell activation, leading to Runx/CBFß complex-independent maintenance of CD8α expression in effector T cells.


Asunto(s)
Antígenos CD8/fisiología , Linfocitos T CD8-positivos/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/fisiología , Animales , Antígenos CD8/genética , Inmunoprecipitación de Cromatina , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Expresión Génica , Histonas/metabolismo , Activación de Linfocitos , Ratones , Regiones Promotoras Genéticas
11.
Nat Immunol ; 12(4): 312-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21297643

RESUMEN

The molecular mechanisms that direct the development of TCRαß+CD8αα+ intestinal intraepithelial lymphocytes (IELs) are not thoroughly understood. Here we show that transforming growth factor-ß (TGF-ß) controls the development of TCRαß+CD8αα+ IELs. Mice with either a null mutation in the gene encoding TGF-ß1 or T cell-specific deletion of TGF-ß receptor I lacked TCRαß+CD8αα+ IELs, whereas mice with transgenic overexpression of TGF-ß1 had a larger population of TCRαß+CD8αα+ IELs. We observed defective development of the TCRαß+CD8αα+ IEL thymic precursors (CD4⁻CD8⁻TCRαß+CD5+) in the absence of TGF-ß. In addition, we found that TGF-ß signaling induced CD8α expression in TCRαß+CD8αα+ IEL thymic precursors and induced and maintained CD8α expression in peripheral populations of T cells. Our data demonstrate a previously unrecognized role for TGF-ß in the development of TCRαß+CD8αα+ IELs and the expression of CD8α in T cells.


Asunto(s)
Antígenos CD8/metabolismo , Linfocitos/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Antígenos CD8/genética , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/metabolismo , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Recuento de Linfocitos , Linfocitos/citología , Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína smad3/genética , Proteína smad3/metabolismo , Timo/citología , Timo/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/farmacología
12.
Nat Immunol ; 11(8): 666-73, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20644572

RESUMEN

The development of T cells in the thymus involves several differentiation and proliferation events, during which hematopoietic precursors give rise to T cells ready to respond to antigen stimulation and undergo effector differentiation. This review addresses signaling and transcriptional checkpoints that control the intrathymic journey of T cell precursors. We focus on the divergence of alphabeta and gammadelta lineage cells and the elaboration of the alphabeta T cell repertoire, with special emphasis on the emergence of transcriptional programs that direct lineage decisions.


Asunto(s)
Diferenciación Celular/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Timo/inmunología , Animales , Diferenciación Celular/genética , Linaje de la Célula , Regulación de la Expresión Génica/inmunología , Humanos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/citología , Timo/citología , Transcripción Genética/genética , Transcripción Genética/inmunología
13.
J Immunol ; 185(2): 1055-62, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20562258

RESUMEN

Feedback inhibition of V(D)J recombination enforces Ag receptor allelic exclusion in mammalian lymphocytes. Yet, in-frame VbetaDJbeta exons can assemble on both alleles in human and mouse alphabeta T lineage cells. To elucidate mechanisms that enforce TCRbeta allelic exclusion in such cells, we analyzed Vbeta expression and rearrangement in mice containing a functional Vbeta14DJbeta1.5Cbeta1 gene (Vbeta14(NT)) and/or Vbeta8.2DJbeta1.1Cbeta1 transgene (Vbeta8(Tg)). The majority of Vbeta14(NT) and Vbeta8(Tg) alphabeta T lineage cells expressed only Vbeta14(+) or Vbeta8(+) TCRbeta-chains, respectively, and lacked Vbeta rearrangements on wild-type TCRbeta loci. However, endogenous Vbeta rearrangements and alphabeta T lineage cells expressing endogenous Vbetas from wild-type alleles alone or with the prerearranged Vbeta in cell surface TCRbeta-chains were observed in Vbeta14(NT) and Vbeta8(Tg) mice. Although nearly all Vbeta8(Tg):Vbeta14(NT) thymocytes and splenic alphabeta T cells expressed Vbeta8(+) TCRbeta-chains, only half of these lymphocytes expressed Vbeta14(+) TCRbeta-chains, even though similar steady-state levels of Vbeta14(NT) mRNA were expressed in Vbeta8(+)Vbeta14(+) and Vbeta8(+)Vbeta14(-) populations. Our data demonstrated that posttranscriptional silencing of functionally assembled endogenous VbetaDJbetaCbeta genes can enforce TCRbeta allelic exclusion and reveal another mechanism that contributes to the development of lymphocytes with monospecific Ag receptors.


Asunto(s)
Interferencia de ARN , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Linfocitos T/metabolismo , Alelos , Animales , Citometría de Flujo , Reordenamiento Génico de Linfocito T , Hibridomas/metabolismo , Ratones , Ratones Endogámicos , Ratones Noqueados , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/citología
14.
J Immunol ; 184(3): 1369-78, 2010 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-20042591

RESUMEN

Ag receptor allelic exclusion is thought to occur through monoallelic initiation and subsequent feedback inhibition of recombinational accessibility. However, our previous analysis of mice containing a V(D)J recombination reporter inserted into Vbeta14 (Vbeta14(Rep)) indicated that Vbeta14 chromatin accessibility is biallelic. To determine whether Vbeta14 recombinational accessibility is subject to feedback inhibition, we analyzed TCRbeta rearrangements in Vbeta14(Rep) mice containing a preassembled in-frame transgenic Vbeta8.2Dbeta1Jbeta1.1 or an endogenous Vbeta14Dbeta1Jbeta1.4 rearrangement on the homologous chromosome. Expression of either preassembled VbetaDJbetaC beta-chain accelerated thymocyte development because of enhanced cellular selection, demonstrating that the rate-limiting step in early alphabeta T cell development is the assembly of an in-frame VbetaDJbeta rearrangement. Expression of these preassembled VbetaDJbeta rearrangements inhibited endogenous Vbeta14-to-DJbeta rearrangements as expected. However, in contrast to results predicted by the accepted model of TCRbeta feedback inhibition, we found that expression of these preassembled TCR beta-chains did not downregulate recombinational accessibility of Vbeta14 chromatin. Our findings suggest that TCRbeta-mediated feedback inhibition of Vbeta14 rearrangements depends on inherent properties of Vbeta14, Dbeta, and Jbeta recombination signal sequences.


Asunto(s)
Diversidad de Anticuerpos , Cromatina/fisiología , Retroalimentación Fisiológica/fisiología , Reordenamiento Génico de Linfocito T/inmunología , Región de Unión de la Inmunoglobulina , Receptores de Antígenos de Linfocitos T alfa-beta/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Transducción de Señal/inmunología , Animales , Diversidad de Anticuerpos/genética , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Regulación del Desarrollo de la Expresión Génica/inmunología , Genes Reporteros/inmunología , Mutación de Línea Germinal/inmunología , Región de Unión de la Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Pérdida de Heterocigocidad/inmunología , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T alfa-beta/biosíntesis , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Transducción de Señal/genética , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
15.
Mol Cell ; 34(3): 298-310, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19450528

RESUMEN

A hallmark of the cellular response to DNA double-strand breaks (DSBs) is histone H2AX phosphorylation in chromatin to generate gamma-H2AX. Here, we demonstrate that gamma-H2AX densities increase transiently along DNA strands as they are broken and repaired in G1 phase cells. The region across which gamma-H2AX forms does not spread as DSBs persist; rather, gamma-H2AX densities equilibrate at distinct levels within a fixed distance from DNA ends. Although both ATM and DNA-PKcs generate gamma-H2AX, only ATM promotes gamma-H2AX formation to maximal distance and maintains gamma-H2AX densities. MDC1 is essential for gamma-H2AX formation at high densities near DSBs, but not for generation of gamma-H2AX over distal sequences. Reduced H2AX levels in chromatin impair the density, but not the distance, of gamma-H2AX formed. Our data suggest that H2AX fuels a gamma-H2AX self-reinforcing mechanism that retains MDC1 and activated ATM in chromatin near DSBs and promotes continued local phosphorylation of H2AX.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos B/citología , Linfocitos B/fisiología , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Endonucleasas , Fase G1/fisiología , Genes Codificadores de la Cadena alfa de los Receptores de Linfocito T/genética , Histonas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Recombinación Genética , Timo/citología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética
16.
J Immunol ; 182(9): 5586-95, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19380806

RESUMEN

TCRbeta chain repertoire of peripheral alphabeta T cells is generated through the stepwise assembly and subsequent selection of TCRbeta V region exons during thymocyte development. To evaluate the influence of a two-step recombination process on Vbeta rearrangement and selection, we generated mice with a preassembled Dbeta1Jbeta1.1 complex on the Jbeta1(omega) allele, an endogenous TCRbeta allele that lacks the Dbeta2-Jbeta2 cluster, creating the Jbeta1(DJbeta) allele. As compared with Jbeta1(omega/omega) mice, both Jbeta1(DJbeta/omega) and Jbeta1(DJbeta/DJbeta) mice exhibited grossly normal thymocyte development and TCRbeta allelic exclusion. In addition, Vbeta rearrangements on Jbeta1(DJbeta) and Jbeta1(omega) alleles were similarly regulated by TCRbeta-mediated feedback regulation. However, in-frame VbetaDJbeta rearrangements were present at a higher level on the Jbeta1(DJbeta) alleles of Jbeta1(DJbeta/omega) alphabeta T cell hybridomas, as compared with on the Jbeta1(omega) alleles. This bias was most likely due to both an increased frequency of Vbeta-to-DJbeta rearrangements on Jbeta1(DJbeta) alleles and a preferential selection of cells with in-frame VbetaDJbeta exons assembled on Jbeta1(DJbeta) alleles during the development of Jbeta1(DJbeta/omega) alphabeta T cells. Consistent with the differential selection of in-frame VbetaDJbeta rearrangements on Jbeta1(DJbeta) alleles, the Vbeta repertoire of alphabeta T cells was significantly altered during alphabeta TCR selection in Jbeta1(DJbeta/omega) and Jbeta1(DJbeta/DJbeta) mice, as compared with in Jbeta1(omega/omega) mice. Our data indicate that the diversity of DJbeta complexes assembled during thymocyte development influences TCRbeta chain selection and peripheral Vbeta repertoire.


Asunto(s)
Diversidad de Anticuerpos/genética , Reordenamiento Génico de Linfocito T/inmunología , Región de Unión de la Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Alelos , Animales , Diversidad de Anticuerpos/inmunología , Secuencia de Bases , Línea Celular Tumoral , Células Cultivadas , Marcación de Gen , Región de Unión de la Inmunoglobulina/biosíntesis , Región Variable de Inmunoglobulina/biosíntesis , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Receptores de Antígenos de Linfocitos T alfa-beta/biosíntesis , Recombinación Genética , Subgrupos de Linfocitos T/citología
17.
J Immunol ; 180(4): 2339-46, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18250443

RESUMEN

To elucidate mechanisms that regulate Vbeta rearrangement, we generated and analyzed mice with a V(D)J recombination reporter cassette of germline Dbeta-Jbeta segments inserted into the endogenous Vbeta14 locus (Vbeta14(Rep)). As a control, we first generated and analyzed mice with the same Dbeta-Jbeta cassette targeted into the generally expressed c-myc locus (c-myc(Rep)). Substantial c-myc(Rep) recombination occurred in both T and B cells and initiated concurrently with endogenous Dbeta to Jbeta rearrangements in thymocytes. In contrast, Vbeta14(Rep) recombination was restricted to T cells and initiated after endogenous Dbeta to Jbeta rearrangements, but concurrently with endogenous Vbeta14 rearrangements. Thus, the local chromatin environment imparts lineage and developmental stage-specific accessibility upon the inserted reporter. Although Vbeta14 rearrangements occur on only 5% of endogenous TCRbeta alleles, the Vbeta14(Rep) cassette underwent rearrangement on 80-90% of alleles, supporting the suggestion that productive coupling of accessible Vbeta14 segments and DJbeta complexes influence the frequency of Vbeta14 rearrangements. Strikingly, Vbeta14(Rep) recombination also occurs on TCRbeta alleles lacking endogenous Vbeta to DJbeta rearrangements, indicating that Vbeta14 accessibility per se is not subject to allelic exclusion.


Asunto(s)
Reordenamiento Génico de Linfocito B , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T/genética , Región Variable de Inmunoglobulina/genética , Región Variable de Inmunoglobulina/metabolismo , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Animales , Células Cultivadas , Cromosomas/genética , Regiones Determinantes de Complementariedad/genética , Células Madre Embrionarias/metabolismo , Genes Reporteros , Región de Unión de la Inmunoglobulina/genética , Ratones , Mutagénesis Insercional , Recombinación Genética , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
19.
Blood ; 106(12): 3898-906, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16118316

RESUMEN

Although Notch receptor expression on malignant B cells is widespread, the effect of Notch signaling in these cells is poorly understood. To investigate Notch signaling in B-cell malignancy, we assayed the effect of Notch activation in multiple murine and human B-cell tumors, representing both immature and mature subtypes. Expression of constitutively active, truncated forms of the 4 mammalian Notch receptors (ICN1-4) inhibited growth and induced apoptosis in both murine and human B-cell lines but not T-cell lines. Similar results were obtained in human precursor B-cell acute lymphoblastic leukemia lines when Notch activation was achieved by coculture with fibroblasts expressing the Notch ligands Jagged1 or Jagged2. All 4 truncated Notch receptors, as well as the Jagged ligands, induced Hes1 transcription. Retroviral expression of Hairy/Enhancer of Split-1 (Hes1) recapitulated the Notch effects, suggesting that Hes1 is an important mediator of Notch-induced growth arrest and apoptosis in B cells. Among the B-cell malignancies that were susceptible to Notch-mediated growth inhibition/apoptosis were mature B-cell and therapy-resistant B-cell malignancies, including Hodgkin, myeloma, and mixed-lineage leukemia (MLL)-translocated cell lines. These results suggest that therapies capable of activating Notch/Hes1 signaling may have therapeutic potential in a wide range of human B-cell malignancies.


Asunto(s)
Apoptosis/fisiología , Linfocitos B/metabolismo , Trastornos Linfoproliferativos/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Animales , Linfocitos B/patología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular , Proliferación Celular , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/metabolismo
20.
Mol Cell Biol ; 24(21): 9265-73, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15485896

RESUMEN

Notch proteins are transmembrane receptors that participate in a highly conserved signaling pathway that regulates morphogenesis in metazoans. Newly synthesized Notch receptors are proteolytically cleaved during transit to the cell surface, creating heterodimeric mature receptors comprising noncovalently associated extracellular (N(EC)) and transmembrane (N) subunits. Ligand binding activates Notch by inducing two successive proteolytic cleavages, catalyzed by metalloproteases and gamma-secretase, respectively, that permit the intracellular portion of N to translocate to the nucleus and activate transcription of target genes. Prior work has shown that the presence of N(EC) prevents ligand-independent activation of N, but the mechanisms involved are poorly understood. Here, we define the roles of two regions at the C-terminal end of N(EC) that participate in maintaining the integrity of resting Notch receptors through distinct mechanisms. The first region, a hydrophobic, previously uncharacterized portion of N(EC), is sufficient to form stable complexes with the extracellular portion of N. The second region, consisting of the three Lin12/Notch repeats, is not needed for heterodimerization but acts to protect N from ligand-independent cleavage by metalloproteases. Together, these two contiguous regions of N(EC) impose crucial restraints that prevent premature Notch receptor activation.


Asunto(s)
Proteínas de la Membrana/metabolismo , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Secuencias Repetitivas de Aminoácido , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Línea Celular , Dimerización , Regulación de la Expresión Génica , Humanos , Ligandos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Subunidades de Proteína/genética , Receptores Notch , Alineación de Secuencia , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...