Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Banco de datos
Tipo del documento
Asunto de la revista
País de afiliación
Intervalo de año de publicación
1.
Blood ; 139(16): 2483-2498, 2022 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-35020836

RESUMEN

NOTCH1 is a well-established lineage specifier for T cells and among the most frequently mutated genes throughout all subclasses of T cell acute lymphoblastic leukemia (T-ALL). How oncogenic NOTCH1 signaling launches a leukemia-prone chromatin landscape during T-ALL initiation is unknown. Here we demonstrate an essential role for the high-mobility-group transcription factor Tcf1 in orchestrating chromatin accessibility and topology, allowing aberrant Notch1 signaling to convey its oncogenic function. Although essential, Tcf1 is not sufficient to initiate leukemia. The formation of a leukemia-prone epigenetic landscape at the distal Notch1-regulated Myc enhancer, which is fundamental to this disease, is Tcf1-dependent and occurs within the earliest progenitor stage even before cells adopt a T lymphocyte or leukemic fate. Moreover, we discovered a unique evolutionarily conserved Tcf1-regulated enhancer element in the distal Myc-enhancer, which is important for the transition of preleukemic cells to full-blown disease.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células T Precursoras , Carcinogénesis/genética , Línea Celular Tumoral , Cromatina/genética , Humanos , Oncogenes , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Receptor Notch1/genética
2.
Stem Cells ; 32(8): 2188-200, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24578291

RESUMEN

Mesenchymal stromal cells (MSCs) are radioresistant bone marrow progenitors that support hematopoiesis and its reconstitution following total body irradiation. MSCs reside in hypoxic niches within the bone marrow and tumor microenvironments. The DNA damage response (DDR) represents a network of signaling pathways that enable cells to activate biological responses to DNA damaging agents. Hypoxia-mediated alterations in the DDR contribute to the increased radioresistance of hypoxic cancer cells, limiting therapeutic efficacy. The DDR is important in mediating mouse MSC radioresistance. However, the effects of hypoxia on MSC radioresistance are currently unknown. In this report, hypoxia was found to (a) increase MSC proliferation rate and colony size; (b) increase long-term survival post-irradiation (IR), and (c) improve MSC recovery from IR-induced cell cycle arrest. DNA double-strand break (DSB) repair in MSCs was upregulated in hypoxia, accelerating the resolution of highly genotoxic IR-induced DNA DSBs. In addition, HIF-1α was found to contribute to this enhanced DSB repair by regulating (a) the expression of DNA ligase IV and DNA-PKcs and (b) Rad51 foci formation in response to DNA DSBs in hypoxic MSCs. We have demonstrated, for the first time, that hypoxia enhances mouse MSC radioresistance in vitro. These findings have important implications for our understanding of MSC functions in supporting allogeneic bone marrow transplantation and in tumorigenesis.


Asunto(s)
Reparación del ADN/fisiología , Células Madre Mesenquimatosas/fisiología , Células Madre Mesenquimatosas/efectos de la radiación , Tolerancia a Radiación/fisiología , Animales , Western Blotting , Hipoxia de la Célula , Daño del ADN/fisiología , Daño del ADN/efectos de la radiación , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Interferente Pequeño , Microambiente Tumoral/fisiología , Microambiente Tumoral/efectos de la radiación
3.
Stem Cells ; 31(1): 137-45, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22961695

RESUMEN

The regeneration of the hematopoietic system following total body irradiation is supported by host-derived mesenchymal stromal cells (MSCs) within the bone marrow. The mechanisms used by MSCs to survive radiation doses that are lethal to the hematopoietic system are poorly understood. The DNA damage response (DDR) represents a cohort of signaling pathways that enable cells to execute biological responses to genotoxic stress. Here, we examine the role of the DDR in mediating the resistance of MSCs to ionizing radiation (IR) treatment using two authentic clonal mouse MSC lines, MS5 and ST2, and primary bulk mouse MSCs. We show that multiple DDR mechanisms contribute to the radio-resistance of MSCs: robust DDR activation via rapid γ-H2AX formation, activation of effective S and G(2)/M DNA damage checkpoints, and efficient repair of IR-induced DNA double-strand breaks. We show that MSCs are intrinsically programmed to maximize survival following IR treatment by expressing high levels of key DDR proteins including ATM, Chk2, and DNA Ligase IV; high levels of the anti-apoptotic, Bcl-2 and Bcl-(XL); and low levels of the pro-apoptotic, Bim and Puma. As a result, we demonstrate that irradiated mouse MSCs withstand IR-induced genotoxic stress, continue to proliferate, and retain their capacity to differentiate long-term along mesenchymal-derived lineages. We have shown, for the first time, that the DDR plays key roles in mediating the radioresistance of mouse MSCs which may have important implications for the study and application of MSCs in allogeneic bone marrow transplantation, graft-versus-host disease, and cancer treatment.


Asunto(s)
Daño del ADN/efectos de la radiación , Reparación del ADN/efectos de la radiación , Células Madre Mesenquimatosas/efectos de la radiación , Tolerancia a Radiación/fisiología , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos T CD4-Positivos/efectos de la radiación , Linfocitos T CD8-positivos/efectos de la radiación , Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/biosíntesis , Diferenciación Celular/efectos de la radiación , Línea Celular , Proliferación Celular/efectos de la radiación , Quinasa de Punto de Control 2 , Roturas del ADN de Doble Cadena/efectos de la radiación , ADN Ligasa (ATP) , ADN Ligasas/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Histonas/biosíntesis , Histonas/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Radiación Ionizante , Transducción de Señal , Proteínas Supresoras de Tumor/biosíntesis , Proteína bcl-X/biosíntesis
4.
Immunol Cell Biol ; 91(1): 5-11, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23165606

RESUMEN

Mesenchymal stromal cells (MSCs) are multi-potent adult stem cells located in various tissues, including the bone marrow. MSCs are key components of the haematopoietic stem cell (HSC) niche within the bone marrow where they function to maintain haematopoietic homoeostasis by regulating HSC self-renewal and function. Bone marrow exposure to ionising radiation causes rapid depletion of radio-sensitive HSCs and their progenitors, leading to haematopoietic failure. However, host-/patient-derived MSCs can survive radiation doses lethal to the haematopoietic system. The mechanisms underlying MSC radio-resistance are currently under intense investigation. Here, we review the current knowledge of MSC radio-biology. The DNA damage response (DDR) represents an orchestrated network of signalling pathways that enable cells to respond to genotoxic damage. We discuss in detail the emerging importance of the DDR in mediating MSC radio-resistance and examine the DDR of MSCs in the context of other stem cell types. Finally, we examine future advances in understanding MSC radio-resistance and discuss the potential impact of the radio-resistance of these stem cells for the clinic.


Asunto(s)
Células Madre Adultas/inmunología , Células de la Médula Ósea/inmunología , Homeostasis/inmunología , Células Madre Mesenquimatosas/inmunología , Tolerancia a Radiación/inmunología , Nicho de Células Madre/inmunología , Células Madre Adultas/citología , Animales , Células de la Médula Ósea/citología , Daño del ADN/inmunología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Humanos , Células Madre Mesenquimatosas/citología
5.
Front Immunol ; 9: 1312, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29942310

RESUMEN

For successful bone marrow transplantation (BMT), a preconditioning regime involving chemo and radiotherapy is used that results in DNA damage to both hematopoietic and stromal elements. Following radiation exposure, it is well recognized that a single wave of host-derived thymocytes reconstitutes the irradiated thymus, with donor-derived thymocytes appearing about 7 days post BMT. Our previous studies have demonstrated that, in the presence of donor hematopoietic cells lacking T lineage potential, these host-derived thymocytes are able to generate a polyclonal cohort of functionally mature peripheral T cells numerically comprising ~25% of the peripheral T cell pool of euthymic mice. Importantly, we demonstrated that radioresistant CD44+ CD25+ CD117+ DN2 progenitors were responsible for this thymic auto-reconstitution. Until recently, the mechanisms underlying the radioresistance of DN2 progenitors were unknown. Herein, we have used the in vitro "Plastic Thymus" culture system to perform a detailed investigation of the mechanisms responsible for the high radioresistance of DN2 cells compared with radiosensitive hematopoietic stem cells. Our results indicate that several aspects of DN2 biology, such as (i) rapid DNA damage response (DDR) activation in response to ionizing radiation-induced DNA damage, (ii) efficient repair of DNA double-strand breaks, and (iii) induction of a protective G1/S checkpoint contribute to promoting DN2 cell survival post-irradiation. We have previously shown that hypoxia increases the radioresistance of bone marrow stromal cells in vitro, at least in part by enhancing their DNA double-strand break (DNA DSB) repair capacity. Since the thymus is also a hypoxic environment, we investigated the potential effects of hypoxia on the DDR of DN2 thymocytes. Finally, we demonstrate for the first time that de novo DN2 thymocytes are able to rapidly repair DNA DSBs following thymic irradiation in vivo.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA