Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 5152, 2023 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-37620389

RESUMEN

Intestinal intraepithelial lymphocytes (IELs) reside in the gut epithelial layer, where they help in maintaining intestinal homeostasis. Peripheral CD4+ T cells can develop into CD4+CD8αα+ IELs upon arrival at the gut epithelium via the lamina propria (LP). Although this specific differentiation of T cells is well established, the mechanisms preventing it from occurring in the LP remain unclear. Here, we show that chemokine receptor 9 (CCR9) expression is low in epithelial CD4+CD8αα+ IELs, but CCR9 deficiency results in CD4+CD8αα+ over-differentiation in both the epithelium and the LP. Single-cell RNA sequencing shows an enriched precursor cell cluster for CD4+CD8αα+ IELs in Ccr9-/- mice. CD4+ T cells isolated from the epithelium of Ccr9-/- mice also display increased expression of Cbfß2, and the genomic occupancy modification of Cbfß2 expression reveals its important function in CD4+CD8αα+ differentiation. These results implicate a link between CCR9 downregulation and Cbfb2 splicing upregulation to enhance CD4+CD8αα+ IEL differentiation.


Asunto(s)
Linfocitos Intraepiteliales , Receptores CCR , Animales , Ratones , Diferenciación Celular , Regulación hacia Abajo , Epitelio , Regulación hacia Arriba , Receptores CCR/metabolismo
2.
Nat Commun ; 11(1): 1562, 2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32218434

RESUMEN

CCL5 is a unique chemokine with distinct stage and cell-type specificities for regulating inflammation, but how these specificities are achieved and how CCL5 modulates immune responses is not well understood. Here we identify two stage-specific enhancers: the proximal enhancer mediates the constitutive CCL5 expression during the steady state, while the distal enhancer located 1.35 Mb from the promoter induces CCL5 expression in activated cells. Both enhancers are antagonized by RUNX/CBFß complexes, and SATB1 further mediates the long-distance interaction of the distal enhancer with the promoter. Deletion of the proximal enhancer decreases CCL5 expression and augments the cytotoxic activity of tissue-resident T and NK cells, which coincides with reduced melanoma metastasis in mouse models. By contrast, increased CCL5 expression resulting from RUNX3 mutation is associated with more tumor metastasis in the lung. Collectively, our results suggest that RUNX3-mediated CCL5 repression is critical for modulating anti-tumor immunity.


Asunto(s)
Quimiocina CCL5/genética , Subunidades alfa del Factor de Unión al Sitio Principal/metabolismo , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica , Inmunidad , Animales , Antígenos CD/metabolismo , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Homeostasis/genética , Inmunidad/genética , Activación de Linfocitos/inmunología , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Ratones Transgénicos
3.
Life Sci Alliance ; 3(2)2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31980555

RESUMEN

Temporal down-regulation of the CD8 co-receptor after receiving positive-selection signals has been proposed to serve as an important determinant to segregate helper versus cytotoxic lineages by generating differences in the duration of TCR signaling between MHC-I and MHC-II selected thymocytes. By contrast, little is known about whether CD8 also modulates TCR signaling engaged by the non-classical MHC-I-like molecule, CD1d, during development of invariant natural killer T (iNKT) cells. Here, we show that constitutive transgenic CD8 expression resulted in enhanced differentiation of innate memory-like CD8+ thymocytes in both a cell-intrinsic and cell-extrinsic manner, the latter being accomplished by an increase in the IL-4-producing iNKT2 subset. Skewed iNKT2 differentiation requires cysteine residues in the intracellular domain of CD8α that are essential for transmitting cellular signaling. Collectively, these findings shed a new light on the relevance of CD8 down-regulation in shaping the balance of iNKT-cell subsets by modulating TCR signaling.


Asunto(s)
Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/genética , Inmunidad Innata , Células T Asesinas Naturales/inmunología , Animales , Antígenos CD8/genética , Diferenciación Celular/inmunología , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/genética , Timocitos/inmunología , Transfección
4.
Nat Commun ; 10(1): 1075, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30824704

RESUMEN

The original version of this Article contained an error in the author affiliations. Affiliation 5 incorrectly read 'Laboratory for Prediction of Cell Systems Dynamics, RIKEN Center for Biosystems Dynamics Research (BDR), Suite, Hyogo 565-0874, Japan.' This has now been corrected in both the PDF and HTML versions of the Article.

5.
Nat Commun ; 10(1): 447, 2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30683858

RESUMEN

Group 2 innate lymphoid cells (ILC2s) have tissue-resident competence and contribute to the pathogenesis of allergic diseases. However, the mechanisms regulating prolonged ILC2-mediated TH2 cytokine production under chronic inflammatory conditions are unclear. Here we show that, at homeostasis, Runx deficiency induces excessive ILC2 activation due to overly active GATA-3 functions. By contrast, during allergic inflammation, the absence of Runx impairs the ability of ILC2s to proliferate and produce effector TH2 cytokines and chemokines. Instead, functional deletion of Runx induces the expression of exhaustion markers, such as IL-10 and TIGIT, on ILC2s. Finally, these 'exhausted-like' ILC2s are unable to induce type 2 immune responses to repeated allergen exposures. Thus, Runx confers competence for sustained ILC2 activity at the mucosa, and contributes to allergic pathogenesis.


Asunto(s)
Asma/inmunología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/inmunología , Subunidad beta del Factor de Unión al Sitio Principal/inmunología , Inmunidad Innata , Pulmón/inmunología , Linfocitos/inmunología , Animales , Asma/inducido químicamente , Asma/genética , Asma/patología , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , Proliferación Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/deficiencia , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/deficiencia , Subunidad beta del Factor de Unión al Sitio Principal/genética , Modelos Animales de Enfermedad , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/inmunología , Regulación de la Expresión Génica/inmunología , Interleucina-10/genética , Interleucina-10/inmunología , Intestino Delgado/efectos de los fármacos , Intestino Delgado/inmunología , Intestino Delgado/patología , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/patología , Pulmón/efectos de los fármacos , Pulmón/patología , Activación de Linfocitos , Linfocitos/clasificación , Linfocitos/efectos de los fármacos , Linfocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Papaína/administración & dosificación , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Transducción de Señal , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/patología
6.
Nat Commun ; 9(1): 3593, 2018 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-30185787

RESUMEN

An intronic silencer, S4, in the Cd4 gene has been shown to be responsible for the helper-lineage-specific expression of CD4; S4 requires Runx complex binding to exert its silencer function against the enhancer-mediated Cd4 activation by modulating the epigenetic state of the Cd4 gene. Here we identify a late-acting maturation enhancer. Bcl11b plays essential roles for activation of both the early-acting proximal enhancer and maturation enhancer of Cd4. Notably, Runx complexes suppress these enhancers by distinct mechanisms. Whereas repression of the proximal enhancer depends on the S4 silencer, the maturation enhancer is repressed by Runx in the absence of S4. Moreover, ThPOK, known to antagonize S4-mediated Cd4 repression, assists Runx complexes to restrain maturation enhancer activation. Distinct modes of S4 silencer action upon distinct enhancers thus unravel a pathway that restricts CD4 expression to helper-lineage cells by silencer-independent and Runx-dependent repression of maturation enhancer activity in cytotoxic-lineage cells.


Asunto(s)
Antígenos CD4/genética , Elementos de Facilitación Genéticos/fisiología , Regulación de la Expresión Génica/fisiología , Proteínas Represoras/metabolismo , Linfocitos T Colaboradores-Inductores/fisiología , Proteínas Supresoras de Tumor/metabolismo , Animales , Antígenos CD4/metabolismo , Células Cultivadas , Quimera , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Intrones/genética , Ratones , Ratones Transgénicos , Proteínas Represoras/genética , Elementos Silenciadores Transcripcionales/fisiología , Linfocitos T Citotóxicos/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética
7.
J Exp Med ; 215(2): 595-610, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29343500

RESUMEN

Multipotent hematopoietic progenitors must acquire thymus-homing capacity to initiate T lymphocyte development. Despite its importance, the transcriptional program underlying this process remains elusive. Cbfß forms transcription factor complexes with Runx proteins, and here we show that Cbfß2, encoded by an RNA splice variant of the Cbfb gene, is essential for extrathymic differentiation of T cell progenitors. Furthermore, Cbfß2 endows extrathymic progenitors with thymus-homing capacity by inducing expression of the principal thymus-homing receptor, Ccr9. This occurs via direct binding of Cbfß2 to cell type-specific enhancers, as is observed in Rorγt induction during differentiation of lymphoid tissue inducer cells by activation of an intronic enhancer. As in mice, an alternative splicing event in zebrafish generates a Cbfß2-specific mRNA, important for ccr9 expression. Thus, despite phylogenetically and ontogenetically variable sites of origin of T cell progenitors, their robust thymus-homing capacity is ensured by an evolutionarily conserved mechanism emerging from functional diversification of Runx transcription factor complexes by acquisition of a novel splice variant.


Asunto(s)
Subunidad beta del Factor de Unión al Sitio Principal/genética , Subunidad beta del Factor de Unión al Sitio Principal/inmunología , Células Precursoras de Linfocitos T/citología , Células Precursoras de Linfocitos T/inmunología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/inmunología , Empalme Alternativo , Animales , Diferenciación Celular , Linaje de la Célula , Subunidades alfa del Factor de Unión al Sitio Principal/metabolismo , Subunidad beta del Factor de Unión al Sitio Principal/deficiencia , Elementos de Facilitación Genéticos , Evolución Molecular , Técnicas de Silenciamiento del Gen , Ratones , Ratones Noqueados , Ratones Mutantes , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , ARN Mensajero/genética , Receptores CCR/genética , Receptores CCR/inmunología , Especificidad de la Especie , Timo/citología , Timo/embriología , Timo/inmunología , Pez Cebra , Proteínas de Pez Cebra/deficiencia
8.
Nat Commun ; 8(1): 702, 2017 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-28951542

RESUMEN

T-lineage committed precursor thymocytes are screened by a fate-determination process mediated via T cell receptor (TCR) signals for differentiation into distinct lineages. However, it remains unclear whether any antecedent event is required to couple TCR signals with the transcriptional program governing lineage decisions. Here we show that Bcl11b, known as a T-lineage commitment factor, is essential for proper expression of ThPOK and Runx3, central regulators for the CD4-helper/CD8-cytotoxic lineage choice. Loss of Bcl11b results in random expression of these factors and, thereby, lineage scrambling that is disconnected from TCR restriction by MHC. Initial Thpok repression by Bcl11b prior to the pre-selection stage is independent of a known silencer for Thpok, and requires the last zinc-finger motif in Bcl11b protein, which by contrast is dispensable for T-lineage commitment. Collectively, our findings shed new light on the function of Bcl11b in priming lineage-specifying genes to integrate TCR signals into subsequent transcriptional regulatory mechanisms.CD4 and CD8 T cells develop in the thymus with their transcription programs controlled by ThPOK and Runx3, respectively. Here the authors show that a pre-commitment event modulated by the transcription factor, Bcl11b, is required for the proper expression of ThPOK and Runx3 and correct CD4/CD8 lineage commitment.


Asunto(s)
Diferenciación Celular/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Proteínas Represoras/genética , Linfocitos T Citotóxicos/citología , Linfocitos T Colaboradores-Inductores/citología , Timocitos/citología , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Animales , Linaje de la Célula , Regulación de la Expresión Génica , Ratones , Receptores de Antígenos de Linfocitos T/genética
9.
Cell Rep ; 19(6): 1176-1188, 2017 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-28494867

RESUMEN

T cell receptor (TCR) signaling by MHC class I and II induces thymocytes to acquire cytotoxic and helper fates via the induction of Runx3 and ThPOK transcription factors, respectively. The mechanisms by which TCR signaling is translated into transcriptional programs for each cell fate remain elusive. Here, we show that, in post-selection thymocytes, a genome organizer, SATB1, activates genes for lineage-specifying factors, including ThPOK, Runx3, CD4, CD8, and Treg factor Foxp3, via regulating enhancers in these genes in a locus-specific manner. Indeed, SATB1-deficient thymocytes are partially re-directed into inappropriate T lineages after both MHC class I- and II-mediated selection, and they fail to generate NKT and Treg subsets. Despite its essential role in activating enhancers for the gene encoding ThPOK in TCR-signaled thymocytes, SATB1 becomes dispensable for maintaining ThPOK in CD4+ T cells. Collectively, our findings demonstrate that SATB1 shapes the primary T cell pool by directing lineage-specific transcriptional programs in the thymus.


Asunto(s)
Linfopoyesis , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Subgrupos de Linfocitos T/citología , Animales , Antígenos CD4/genética , Antígenos CD4/metabolismo , Antígenos CD8/genética , Antígenos CD8/metabolismo , Linaje de la Célula , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Elementos de Facilitación Genéticos , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Ratones , Subgrupos de Linfocitos T/metabolismo , Timo/citología , Timo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional
10.
Nat Immunol ; 18(3): 274-282, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28135253

RESUMEN

Although invariant Vα14+ natural killer T cells (NKT cells) are thought to be generated from CD4+CD8+ double-positive (DP) thymocytes, the developmental origin of CD4-CD8- double-negative (DN) NKT cells still remains unresolved. Here we provide definitive genetic evidence obtained, through studies of mice with DP-stage-specific ablation of expression of the gene encoding the recombinase component RAG-2 (Rag2) and by a fate-mapping approach, that supports the proposal of the existence of an alternative developmental pathway through which a fraction of DN NKT cells with strong T-helper-type-1 (TH1)-biased and cytotoxic characteristics develop from late DN-stage thymocytes, bypassing the DP stage. These findings provide new insight into understanding of the development of NKT cells and propose a role for timing of expression of the invariant T cell antigen receptor in determining the functional properties of NKT cells.


Asunto(s)
Células T Asesinas Naturales/fisiología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Timocitos/fisiología , Animales , Antígenos CD4/metabolismo , Antígenos CD8/metabolismo , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Citocinas/metabolismo , Citotoxicidad Inmunológica/genética , Proteínas de Unión al ADN/genética , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células TH1/inmunología
11.
Proc Natl Acad Sci U S A ; 113(24): E3394-402, 2016 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-27226296

RESUMEN

Invariant natural killer T (iNKT) cells are a subset of innate-like T cells that act as important mediators of immune responses. In particular, iNKT cells have the ability to immediately produce large amounts of IFN-γ upon activation and thus initiate immune responses in various pathological conditions. However, molecular mechanisms that control IFN-γ production in iNKT cells are not fully understood. Here, we report that basic helix-loop-helix transcription factor family, member e40 (Bhlhe40), is an important regulator for IFN-γ production in iNKT cells. Bhlhe40 is highly expressed in stage 3 thymic iNKT cells and iNKT1 subsets, and the level of Bhlhe40 mRNA expression is correlated with Ifng mRNA expression in the resting state. Although Bhlhe40-deficient mice show normal iNKT cell development, Bhlhe40-deficient iNKT cells show significant impairment of IFN-γ production and antitumor effects. Bhlhe40 alone shows no significant effects on Ifng promoter activities but contributes to enhance T-box transcription factor Tbx21 (T-bet)-mediated Ifng promoter activation. Chromatin immunoprecipitation analysis revealed that Bhlhe40 accumulates in the T-box region of the Ifng locus and contributes to histone H3-lysine 9 acetylation of the Ifng locus, which is impaired without T-bet conditions. These results indicate that Bhlhe40 works as a cofactor of T-bet for enhancing IFN-γ production in iNKT cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Regulación de la Expresión Génica/inmunología , Proteínas de Homeodominio/inmunología , Interferón gamma/inmunología , Células T Asesinas Naturales/inmunología , Regiones Promotoras Genéticas/inmunología , Proteínas de Dominio T Box/inmunología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Regulación de la Expresión Génica/genética , Proteínas de Homeodominio/genética , Inmunidad Celular/genética , Interferón gamma/genética , Ratones , Ratones Noqueados , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Proteínas de Dominio T Box/genética
12.
PLoS One ; 11(4): e0153347, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27064277

RESUMEN

Invariant Vα14 natural killer T (NKT) cells, characterized by the expression of a single invariant T cell receptor (TCR) α chain encoded by rearranged Trav11 (Vα14)-Traj18 (Jα18) gene segments in mice, and TRAV10 (Vα24)-TRAJ18 (Jα18) in humans, mediate adjuvant effects to activate various effector cell types in both innate and adaptive immune systems that facilitates the potent antitumor effects. It was recently reported that the Jα18-deficient mouse described by our group in 1997 harbors perturbed TCRα repertoire, which raised concerns regarding the validity of some of the experimental conclusions that have been made using this mouse line. To resolve this concern, we generated a novel Traj18-deficient mouse line by specifically targeting the Traj18 gene segment using Cre-Lox approach. Here we showed the newly generated Traj18-deficient mouse has, apart from the absence of Traj18, an undisturbed TCRα chain repertoire by using next generation sequencing and by detecting normal generation of Vα19Jα33 expressing mucosal associated invariant T cells, whose development was abrogated in the originally described Jα18-KO mice. We also demonstrated here the definitive requirement for NKT cells in the protection against tumors and their potent adjuvant effects on antigen-specific CD8 T cells.


Asunto(s)
Reordenamiento Génico de la Cadena alfa de los Receptores de Antígenos de los Linfocitos T/inmunología , Genes Codificadores de la Cadena alfa de los Receptores de Linfocito T/fisiología , Células Asesinas Naturales/inmunología , Neoplasias Hepáticas/inmunología , Melanoma Experimental/inmunología , Animales , Citometría de Flujo , Reordenamiento Génico de la Cadena alfa de los Receptores de Antígenos de los Linfocitos T/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Subgrupos de Linfocitos T/inmunología
13.
Proc Jpn Acad Ser B Phys Biol Sci ; 91(7): 292-304, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26194854

RESUMEN

Natural Killer T (NKT) cells are unique lymphocytes characterized by their expression of a single invariant antigen receptor encoded by Vα14Jα18 in mice and Vα24Jα18 in humans, which recognizes glycolipid antigens in association with the monomorphic CD1d molecule. NKT cells mediate adjuvant activity to activate both CD8T cells to kill MHC-positive tumor cells and NK cells to eliminate MHC-negative tumor at the same time in patients, resulting in the complete eradication of tumors without relapse. Therefore, the NKT cell-targeted therapy can be applied to any type of tumor and also to anyone individual, regardless of HLA type.Phase IIa clinical trials on advanced lung cancers and head and neck tumors have been completed and showed significantly prolonged median survival times with only the primary treatment. Another potential treatment option for the future is to use induced pluripotent stem cell (iPS)-derived NKT cells, which induced adjuvant effects on anti-tumor responses, inhibiting in vivo tumor growth in a mouse model.


Asunto(s)
Inmunoterapia/métodos , Células T Asesinas Naturales/citología , Neoplasias/inmunología , Neoplasias/terapia , Secuencia de Aminoácidos , Animales , Ensayos Clínicos como Asunto , Evolución Molecular , Humanos , Células Madre Pluripotentes Inducidas/citología , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Neoplasias/genética
14.
PLoS One ; 9(10): e111826, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25356669

RESUMEN

Recent progress in regenerative medicine has enabled the utilization of pluripotent stem cells (PSCs) such as embryonic stem cells (ESCs) as a donor resource for transplantation. However, immune suppression is still needed when the donor-recipient combination is allogeneic. Protection of ESCs-derived grafts from host immune response might be achieved thought the utilization of immunosuppressive cells generated from ESCs. In the present study, we show that a certain fraction of immunosuppressive cells can be generated from ESCs and help to suppress immune response against allogeneic grafts. ESCs-derived suppressor cells (ES-SCs) resembled macrophages in terms of cell surface molecule and gene expressions. Furthermore, gene expression analysis including microarray showed that ES-SCs have M1/M2 hybrid phenotype with high expression of genes correlated to immunosuppression of T cell response. Indeed, ES-SCs were effective to block allogeneic T cell proliferation in a nitric oxide-dependent manner, and prolonged the survival of ESCs-derived embryoid bodies or cardiomyocytes grafts transplanted into mouse kidney capsule. Thus, we consider the potential use of these ESCs-derived macrophage-like immunosuppressive cells as cellular therapies to promote long-term graft survival in future therapies.


Asunto(s)
Supervivencia de Injerto/inmunología , Terapia de Inmunosupresión , Macrófagos/citología , Células Madre Embrionarias de Ratones/citología , Miocitos Cardíacos/trasplante , Animales , Proliferación Celular , Células Dendríticas/citología , Femenino , Isoantígenos/inmunología , Ratones , Ratones Endogámicos C3H , Miocitos Cardíacos/citología , Fenotipo , Linfocitos T/citología , Trasplante Homólogo
15.
Histol Histopathol ; 28(8): 965-9, 2013 08.
Artículo en Inglés | MEDLINE | ID: mdl-23467909

RESUMEN

Infantile neuroaxonal dystrophy (INAD) is a severe neurodegenerative disease characterized by its early onset. PLA2G6, which encodes a phospholipase A2, iPLA2ß, has been identified as a causative gene of INAD. iPLA2ß has been shown to be involved in various physiological and pathological processes, including immunity, cell death, and cell membrane homeostasis. Gene targeted mice with a null mutation of Pla2g6 develop the INAD phenotype as late as approximately 1 to 2 years after birth. Recently, another INAD mouse model, Pla2g6-INAD mice line, has been established. The Pla2g6-INAD mice bear a point mutation in the ankyrin repeat domain of Pla2g6 generated by N-ethyl-N-nitrosourea mutagenesis. These mutant mice develop severe motor dysfunction and hematopoietic abnormality in a manner following Mendelian law. The mice showed the abnormal gait and poor performance as early as 7 to 8 weeks of age, detected by hanging grip test. Neuropathological examination revealed widespread formation of spheroids containing tubulovesicular membranes similar to human INAD. Molecular and biochemical analysis revealed that the mutant mice expressed Pla2g6 mRNA and protein, but the mutated Pla2g6 protein had no glycerophospholipid-catalyzing enzyme activity. When analyzed the offspring which bear Pla2g6 knockout allele and Pla2g6-INAD allele, abnormal gait appeared slightly later than Pla2g6-INAD homozygotes but with earlier onset than the Pla2g6 knockout homozygotes. This result suggests that mutant Pla2g6 protein contributes to early onset of INAD symptoms in the absence of intact Pla2g6 protein. The analysis of various INAD mouse models may help to understand the pathogenesis of neurodegenerative diseases, including INAD.


Asunto(s)
Fosfolipasas A2 Grupo VI/genética , Distrofias Neuroaxonales/genética , Alelos , Animales , Modelos Animales de Enfermedad , Fosfolipasas A2 Grupo VI/deficiencia , Hematopoyesis/genética , Humanos , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Mutación , Distrofias Neuroaxonales/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Fosfolipasas A2/metabolismo , Mutación Puntual
16.
Hum Cell ; 24(1): 9-12, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21547690

RESUMEN

Recently, accumulating evidence has suggested that tumors, including ovarian cancer, are composed of a heterogeneous cell population with a small subset of cancer stem cells (CSCs) that sustain tumor formation and growth. The emergence of drug resistance is one of the most difficult problems in the treatment of ovarian cancer, which has been explained recently by the potential of CSCs to have superior resistance against anti-cancer drugs than conventional cancer cells. In this study, we expanded this line of study to examine whether this phenomenon is also observed in clinical specimens of ovarian cancer cells. In total we could analyze 28 samples out of 60 obtained from ovarian cancer patients. The clinical samples were subjected to testing of the expression of side population (SP) as a CSC marker, and according to the presence of SP (SP+) or absence of SP (SP-), clinicopathological significances were analyzed. Although there was no statistical significance, there were more SP+s in recurrent cases as well as in ascitic and peritoneal dissemination than in primary tumor of the ovary. There was no correlation between SP status and FIGO staging. In 19 cases of those who could be followed more than 6 months from initial therapy, there were 8 cases of recurrence or death from disease, and all of these were SP+. On the other hand, in 11 cases of disease-free survivors, 6 were SP+. There was a significant difference in prognosis between SP+ and SP- (p = 0.017). Although this study was limited, it revealed that SP could be contained more in recurrent or metastatic tumors than in primary tumors, and also that the presence of SP could be a risk factor of recurrence in ovarian cancer. Therefore, a novel therapeutic strategy targeting SP could improve the prognosis of ovarian cancer.


Asunto(s)
Células Madre Neoplásicas/patología , Neoplasias Ováricas/patología , Células de Población Lateral/patología , Adulto , Anciano , Femenino , Citometría de Flujo , Humanos , Persona de Mediana Edad , Metástasis de la Neoplasia , Recurrencia Local de Neoplasia , Estadificación de Neoplasias/métodos , Neoplasias Ováricas/mortalidad , Pronóstico
17.
Gynecol Oncol ; 121(2): 390-4, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21272926

RESUMEN

OBJECTIVES: In recent years, cancer stem cells (CSCs) have been reported to be correlated with chemoresistance and may also be enriched in side populations (SPs). In this study, the relationship between resistance to paclitaxel (PTX) and cisplatin (CDDP) and side populations was examined in three parental PTX- and CDDP-sensitive ovarian cancer cell lines (2008, KF28, and TU-OM-1) and several other cell lines derived from these as well as the additional effects of interferon-alpha (INF-α). METHODS: SP of three different parental cell lines and PTX- and/or CDDP-resistant cell lines derived from these was analyzed with flow cytometry. The expression of ABCB1 and ABCG2 in KF28 and its derived cell lines was examined. Additional cell-death effect of INF-α with PTX was also examined. RESULTS: In the three parental cell lines and the PTX-sensitive cell lines derived from these lines, SP was very low. Conversely, in PTX-resistant cell lines, regardless of CDDP resistance, SP increased. ABCB1 was strongly expressed in the PTX-resistant cells, but not in their parental lines, which are sensitive to PTX. While INF-α showed only slight enhancement of the cell-death effect of PTX in PTX-sensitive cells, INF-α itself strongly induced apoptosis in PTX-resistant cells regardless of PTX concentration. CONCLUSIONS: The SP could be correlated with resistance to PTX. SP could be a target of INF-α, and resistance to PTX might be overcome by INF-α.


Asunto(s)
Cisplatino/farmacología , Células Madre Neoplásicas/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Paclitaxel/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/biosíntesis , Transportadoras de Casetes de Unión a ATP/genética , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cistadenocarcinoma Seroso/tratamiento farmacológico , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patología , Resistencia a Antineoplásicos , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Interferón-alfa/farmacología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo
18.
Int Immunol ; 23(1): 65-74, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21135032

RESUMEN

Forced expression of certain transcription factors in somatic cells results in generation of induced pluripotent stem (iPS) cells, which differentiate into various cell types. We investigated T-cell and B-cell lineage differentiation from iPS cells in vitro. To evaluate the impact of iPS cell source, murine splenic B-cell-derived iPS (B-iPS) cells were generated after retroviral transduction of four transcription factors (Oct4, Sox2, Klf4 and c-Myc). B-iPS cells were identical to embryonic stem (ES) cells and mouse embryonic fibroblast (MEF)-derived iPS cells in morphology, ES cell marker expression as well as teratoma and chimera mouse formation. Both B-iPS and MEF-derived iPS cells differentiated into lymphocytes in OP9 co-culture systems. Both efficiently differentiated into T-cell lineage that produced IFN-γ on T-cell receptor stimulation. However, iPS cells including B-iPS cells were relatively resistant to B-cell lineage differentiation. One of the reasons of the failure of B-cell lineage differentiation seemed due to a defect of Pax5 expression in the differentiated cells. Therefore, current in vitro differentiation systems using iPS cells are sufficient for inducing T-cell but not B-cell lineage.


Asunto(s)
Linfocitos B/citología , Diferenciación Celular/inmunología , Células Madre Pluripotentes Inducidas/citología , Linfocitos T/citología , Animales , Linfocitos B/inmunología , Línea Celular , Linaje de la Célula/genética , Técnicas de Cocultivo , Islas de CpG , Metilación de ADN , Expresión Génica , Células Madre Pluripotentes Inducidas/inmunología , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones SCID , Factor de Transcripción PAX5/genética , Linfocitos T/inmunología
19.
Am J Pathol ; 175(6): 2257-63, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19893029

RESUMEN

Calcium-independent group VIA phospholipase A(2) (iPLA(2)beta), encoded by PLA2G6, has been shown to be involved in various physiological and pathological processes, including immunity, cell death, and cell membrane homeostasis. Mutations in the PLA2G6 gene have been recently identified in patients with infantile neuroaxonal dystrophy (INAD). Subsequently, it was reported that similar neurological impairment occurs in gene-targeted mice with a null mutation of iPLA(2)beta, whose disease onset became apparent approximately 1 to 2 years after birth. Here, we report the establishment of an improved mouse model for INAD that bears a point mutation in the ankyrin repeat domain of Pla2g6 generated by N-ethyl-N-nitrosourea mutagenesis. These mutant mice developed severe motor dysfunction, including abnormal gait and poor performance in the hanging grip test, as early as 7 to 8 weeks of age, in a manner following Mendelian law. Neuropathological examination revealed widespread formation of spheroids containing tubulovesicular membranes similar to human INAD. Molecular and biochemical analysis revealed that the mutant mice expressed Pla2g6 mRNA and protein, but the mutated Pla2g6 protein had no glycerophospholipid-catalyzing enzyme activity. Because of the significantly early onset of the disease, this mouse mutant (Pla2g6-inad) could be highly useful for further studies of pathogenesis and experimental interventions in INAD and neurodegeneration.


Asunto(s)
Modelos Animales de Enfermedad , Fosfolipasas A2 Grupo VI/genética , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Edad de Inicio , Animales , Secuencia de Bases , Western Blotting , Ratones , Ratones Mutantes , Microscopía Electrónica de Transmisión , Mutación Puntual , Reacción en Cadena de la Polimerasa
20.
Proc Natl Acad Sci U S A ; 106(42): 17847-51, 2009 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-19815501

RESUMEN

Repeated injection of alpha-galactosylceramide, an agonistic ligand for natural killer T (NKT) cells, results in long-term unresponsiveness or anergy, which severely limits its clinical application. However, the molecular mechanisms leading to NKT anergy induction remain unclear. We show here that the decreased IFN-gamma production and failed tumor rejection observed in anergized NKT cells are rescued by Cbl-b deficiency. Cbl-b E3 ligase activity is critical for the anergy induction, as revealed by the similarity between Cbl-b(-/-) and its RING finger mutant NKT cells. Cbl-b binds and promotes monoubiquitination to CARMA1, a critical signaling molecule in NFkappaB activation. Ubiquitin conjugation to CARMA1 disrupts its complex formation with Bcl10 without affecting its protein stability. In addition, CARMA1(-/-) NKT cells are defective in IFN-gamma production. The study identifies an important signaling pathway linking Cbl-b-induced monoubiquitination to NFkappaB activation in NKT cell anergy induction, which may help design approaches for human cancer therapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Anergia Clonal , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas Adaptadoras de Señalización CARD/deficiencia , Proteínas Adaptadoras de Señalización CARD/genética , Galactosilceramidas/inmunología , Humanos , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/secundario , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-cbl/deficiencia , Proteínas Proto-Oncogénicas c-cbl/genética , Transducción de Señal/inmunología , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA