Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cell Death Discov ; 3: 17018, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28417018

RESUMEN

PP2C serine-threonine phosphatase, Wip1, is an important regulator of stress response. Wip1 controls a number of critical cellular functions: proliferation, cell cycle arrest, senescence and programmed cell death, apoptosis or autophagy. Ppm1d, the gene encoding Wip1 phosphatase, is expressed in hematopoietic progenitors, stem cells, neutrophils, macrophages B and T lymphocytes in bone marrow and peripheral blood. The Wip1-/- mice display immunodeficiency, abnormal lymphoid histopathology in thymus and spleen, defects in B- and T-cell differentiation, as well as susceptibility to viral infection. At the same time, Wip1 knockout mice exhibit pro-inflammatory phenotype in skin and intestine in the model of inflammatory bowel disease (IBD) with elevated levels of inflammation-promoting cytokines TNF-α, IL-6, IL-12, IL-17. Several Wip1 downstream targets can mediate Wip1 effects on hematopoietic system including, p53, ATM, p38MAPK kinase, NFkB, mTOR. Here, we summarized the current knowledge on the role of Wip1 in the differentiation of various hematopoietic lineages and how Wip1 deficiency affects the functions of immune cells.

2.
Oncogene ; 36(16): 2328-2336, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-27819670

RESUMEN

Heat shock protein 110 (HSP110) is induced by different stresses and, through its anti-apoptotic and chaperoning properties, helps cells survive these adverse situations. In colon cancers, HSP110 is abnormally abundant. We have recently shown that colorectal cancer patients with microsatellite instability (MSI) had an improved response to chemotherapy because they harbor an HSP110-inactivating mutation (HSP110DE9). In this work, we used patient biopsies, human colorectal cancer cells grown in vitro and in vivo (xenografts), and intestinal crypts to demonstrate that HSP110 is also involved in colon cancer growth. We showed that HSP110 induces colon cancer cell proliferation and that this effect is associated with STAT3 activation, specifically an increase in STAT3 phosphorylation, nuclear translocation and transcription factor activity. STAT3 inhibition blocks the proliferative effect of HSP110. From a molecular standpoint, we demonstrated that HSP110 directly binds to STAT3, thereby facilitating its phosphorylation by JAK2. Finally, we showed a correlation between HSP110 expression and STAT3 phosphorylation in colon cancer patient samples. Thus, the expression of HSP110 in colon cancer contributes to STAT3-dependent tumor growth and the frequent inactivating mutation of this chaperone is probably an important event underlying the improved prognosis in colon cancer displaying MSI.


Asunto(s)
Neoplasias Colorrectales/patología , Proteínas del Choque Térmico HSP110/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Biopsia , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Femenino , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fosforilación , Unión Proteica
3.
Cell Death Dis ; 7: e2195, 2016 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-27077811

RESUMEN

Inactivation of p53 found in more than half of human cancers is often associated with increased tumor resistance to anti-cancer therapy. We have previously shown that overexpression of the phosphatase Wip1 in p53-negative tumors sensitizes them to chemotherapeutic agents, while protecting normal tissues from the side effects of anti-cancer treatment. In this study, we decided to search for kinases that prevent Wip1-mediated sensitization of cancer cells, thereby interfering with efficacy of genotoxic anti-cancer drugs. To this end, we performed a flow cytometry-based screening in order to identify kinases that regulated the levels of γH2AX, which were used as readout. Another criterion of the screen was increased sensitivity of p53-negative tumor cells to cisplatin (CDDP) in a Wip1-dependent manner. We have found that a treatment with a low dose (75 nM) of MK-1775, a recently described specific chemical inhibitor of Wee1, decreases CDDP-induced H2AX phosphorylation in p53-negative cells and enhances the Wip1-sensitization of p53-negative tumors. We were able to reduce CDDP effective concentration by 40% with a combination of Wip1 overexpression and Wee1 kinase inhibition. We have observed that Wee1 inhibition potentiates Wip1-dependent tumor sensitization effect by reducing levels of Hipk2 kinase, a negative regulator of Wip1 pathway. In addition, during CDDP treatment, the combination of Wee1 inhibition and Wip1 overexpression has a mild but significant protective effect in normal cells and tissues. Our results indicate that inhibition of the negative regulators of Wip1 pathway, Wee1 and Hipk2, in p53-negative tumors could potentiate efficiency of chemotherapeutic agents without concomitant increase of cytotoxicity in normal tissues. The development and clinical use of Wee1 and Hipk1 kinase chemical inhibitors might be a promising strategy to improve anti-cancer therapy.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Nucleares/metabolismo , Proteína Fosfatasa 2C/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteína p53 Supresora de Tumor/genética , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 3/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Cisplatino/farmacología , Cisplatino/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Daño del ADN/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Histonas/metabolismo , Humanos , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2C/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/genética , Interferencia de ARN , Tasa de Supervivencia , Proteína p53 Supresora de Tumor/deficiencia
4.
Oncogene ; 35(22): 2842-51, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-26364610

RESUMEN

Graft versus host disease (GvHD), which is the primary complication of allogeneic bone marrow transplantation, can alter the intestinal barrier targeted by activated donor T-cells. Chemical inhibition of the stress protein HSP90 was demonstrated in vitro to inhibit T-cell activation and to modulate endoplasmic reticulum (ER) stress to which intestinal cells are highly susceptible. Since the HSP90 inhibitor 17-allylamino-demethoxygeldanamycin (17AAG) is developed in clinics, we explored here its ability to control intestinal acute GvHD in vivo in two mouse GvHD models (C57BL/6BALB/c and FVB/NLgr5-eGFP), ex vivo in intestine organoids and in vitro in intestinal epithelial cultures. We show that 17AAG decreases GvHD-associated mortality without impairing graft versus leukemia effect. While 17AAG effect in T-cell activation is just moderate at the dose used in vivo, we observe a striking intestinal integrity protection. At the intestine level, the drug promotes the splicing of the transcription factor X-box binding protein 1 (XBP1), which is a key component of the ER stress. This effect is associated with a decrease in intestinal damage and an increase in Lgr5(+) stem cells, Paneth cells and defensins production. The importance of XBP1 splicing control is further confirmed in cultured cells and organoids of primary intestinal epithelium where XBP1 is either shRNA depleted or inhibited with toyocamycin. In conclusion, 17AAG has a protective effect on the epithelial intestinal barrier in mouse models of acute GvHD. This compound deserves to be tested in the therapeutic control of acute GvHD.


Asunto(s)
Benzoquinonas/farmacología , Citoprotección/efectos de los fármacos , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Intestinos/patología , Lactamas Macrocíclicas/farmacología , Nicho de Células Madre/efectos de los fármacos , Animales , Benzoquinonas/uso terapéutico , Femenino , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/patología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Intestinos/efectos de los fármacos , Lactamas Macrocíclicas/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Empalme del ARN/efectos de los fármacos , Proteína 1 de Unión a la X-Box/genética
5.
Tsitologiia ; 46(5): 431-6, 2004.
Artículo en Ruso | MEDLINE | ID: mdl-15344888

RESUMEN

Alteration in osmotic conditions leads to activation of some defensive mechanisms resulting in blocking the cell cycle progression. One of stress kinases, activated after osmotic stress, is p38 MAPK. In the present study the role of p38 both in intra-S-phase and G2/M checkpoint is shown. Besides, p38-dependent degradation of Cdc25A phosphatase after osmotic stress is demonstrated. Expression of stable form of Cdc25A results in a partial abrogation of G2/M block of the cell cycle, but has no effect on DNA synthesis arrest.


Asunto(s)
Ciclo Celular/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , División Celular/fisiología , ADN/biosíntesis , Fase G2/fisiología , Células HeLa , Humanos , Presión Osmótica , Fase S/fisiología , Fosfatasas cdc25/metabolismo
6.
Tsitologiia ; 46(5): 423-30, 2004.
Artículo en Ruso | MEDLINE | ID: mdl-15344887

RESUMEN

Cdc25A phosphatase regulates cell cycle progression by removing the inhibitory phosphates from cyclin-dependent kinases. Activity of Cdc25A depends on its phosphorylation status. During normal cell cycle progression and after DNA damage phosphorylation by Chk1 (or Chk2) triggers Cdc25A degradation via ubiquitin-proteasome pathway. In this study we investigate the role of various phosphorylation sites (Ser123, Ser75, Ser17 and Ser115) in the regulation of Cdc25A stability. We have shown that only S75A mutation abrogates Cdc25A degradation both in normal and stress conditions. We also studied the influence of stable form of Cdc25A on checkpoint progression after DNA damage. We have found out that delay in DNA synthesis after UV and IR does not depend on Cdc25A activity. However, the presence of stable Cdc25A increases the number of mitotic cells after these stresses.


Asunto(s)
Fosfatasas cdc25/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , ADN/biosíntesis , Estabilidad de Enzimas , Células HeLa , Humanos , Mutagénesis Sitio-Dirigida , Mutación , Fosforilación , Proteínas Quinasas/metabolismo , Serina/química , Serina/genética , Rayos Ultravioleta , Fosfatasas cdc25/química , Fosfatasas cdc25/genética
7.
Cell Death Differ ; 19(11): 1761-8, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22555459

RESUMEN

Conversion of intestinal stem cells into tumor-initiating cells is an early step in Apc(Min)-induced polyposis. Wild-type p53-induced phosphatase 1 (Wip1)-dependent activation of a DNA damage response and p53 has a permanent role in suppression of stem cell conversion, and deletion of Wip1 lowers the tumor burden in Apc(Min) mice. Here we show that cyclin-dependent kinase inhibitor 2a, checkpoint kinase 2, and growth arrest and DNA damage gene 45a (Gadd45a) exert critical functions in the tumor-resistant phenotype of Wip1-deficient mice. We further identified Gadd45a as a haploinsufficient gene in the regulation of Wip1-dependent tumor resistance in mice. Gadd45a appears to function through its ability to activate the Jnk-dependent signaling pathway that in turn is a necessary mediator of the proapoptotic functions of p53 that respond to activation of the ß-catenin signaling pathway. We propose that silencing of Gadd45a is sufficient to override p53 activation in the presence of active ß-catenin under conditions of an enhanced DNA damage response.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Animales , Apoptosis , Transformación Celular Neoplásica , Quinasa de Punto de Control 2 , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Reparación del ADN , Genes APC , Poliposis Intestinal/metabolismo , Poliposis Intestinal/patología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Noqueados , Fosfoproteínas Fosfatasas/deficiencia , Fosfoproteínas Fosfatasas/genética , Proteína Fosfatasa 2C , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Células Madre/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda