Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Death Discov ; 7(1): 335, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34741025

RESUMEN

Endogenous clocks generate rhythms in gene expression, which facilitates the organisms to cope through periodic environmental variations in accordance with 24-h light/dark time. A core question that needs to be elucidated is how such rhythms proliferate throughout the cells and regulate the dynamic physiology. In this study, we demonstrate the role of REGγ as a new regulator of circadian clock in mice, primary MEF, and SY5Y cells. Assessment of circadian conduct reveals a difference in circadian period, wheel mode, and the ability to acclimate the external light stimulus between WT and KO littermates. Compared to WT mice, REGγ KO mice attain the phase delay behavior upon light shock at early night. During the variation of 12/12 h light/dark (LD) exposure, levels of Per1, Per2, Cry1, Clock, Bmal1, and Rorα circadian genes in suprachiasmatic nucleus are significantly higher in REGγ KO than in WT mice, concomitant with remarkable changes in BMAL1 and PER2 proteins. In cultured cells depleted of REGγ, serum shock induces early response of the circadian genes Per1 and Per2 with the cyclic rhythm maintained. Mechanistic study indicates that REGγ directly degrades BMAL1 by the non-canonical proteasome pathway independent of ATP and ubiquitin. Silencing BMAL1 abrogates the changes in circadian genes in REGγ-deficient cells. However, inhibition of GSK-3ß, a known promoter for degradation of BMAL1, exacerbates the action of REGγ depletion. In conclusion, our findings define REGγ as a new factor, which functions as a rheostat of circadian rhythms to mitigate the levels of Per1 and Per2 via proteasome-dependent degradation of BMAL1.

3.
Nat Commun ; 11(1): 3904, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32764536

RESUMEN

A major challenge in chemotherapy is chemotherapy resistance in cells lacking p53. Here we demonstrate that NIP30, an inhibitor of the oncogenic REGγ-proteasome, attenuates cancer cell growth and sensitizes p53-compromised cells to chemotherapeutic agents. NIP30 acts by binding to REGγ via an evolutionarily-conserved serine-rich domain with 4-serine phosphorylation. We find the cyclin-dependent phosphatase CDC25A is a key regulator for NIP30 phosphorylation and modulation of REGγ activity during the cell cycle or after DNA damage. We validate CDC25A-NIP30-REGγ mediated regulation of the REGγ target protein p21 in vivo using p53-/- and p53/REGγ double-deficient mice. Moreover, Phosphor-NIP30 mimetics significantly increase the growth inhibitory effect of chemotherapeutic agents in vitro and in vivo. Given that NIP30 is frequently mutated in the TCGA cancer database, our results provide insight into the regulatory pathway controlling the REGγ-proteasome in carcinogenesis and offer a novel approach to drug-resistant cancer therapy.


Asunto(s)
Autoantígenos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Animales , Autoantígenos/genética , Ciclo Celular , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Resistencia a Antineoplásicos , Células HEK293 , Xenoinjertos , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Fosforilación , Complejo de la Endopetidasa Proteasomal/deficiencia , Complejo de la Endopetidasa Proteasomal/genética , Proteína p53 Supresora de Tumor/genética , Fosfatasas cdc25/metabolismo
4.
Cell Death Differ ; 27(6): 1795-1806, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31767934

RESUMEN

Lung cancer is one of the cancers with highest morbidity and mortality rates and the metastasis of lung cancer is a leading cause of death. Mechanisms of lung cancer metastasis are yet to be fully understood. Herein, we demonstrate that mice deficient for REGγ, a proteasome activator, exhibited a significant reduction in tumor size, numbers, and metastatic rate with prolonged survival in a conditional Kras/p53 mutant lung cancer model. REGγ enhanced the TGFß-Smad signaling pathway by ubiquitin-ATP-independent degradation of Smad7, an inhibitor of the TGFß pathway. Activated TGFß signaling in REGγ-positive lung cancer cells led to diminished expression of E-cadherin, a biomarker of epithelial-mesenchymal transitions (EMT), and elevated mesenchymal markers compared with REGγ-deficient lung cancer cells. REGγ overexpression was found in lung cancer patients with metastasis, correlating with the reduction of E-Cadherin/Smad7 and a poor prognosis. Overall, our study indicates that REGγ promotes lung cancer metastasis by activating TGF-ß signaling via degradation of Smad7. Thus, REGγ may serve as a novel therapeutic target for lung cancers with poor prognosis.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas Asociadas a Pancreatitis/metabolismo , Proteína smad7/metabolismo , Células A549 , Animales , Transición Epitelial-Mesenquimal , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
5.
Cell Signal ; 64: 109412, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31491459

RESUMEN

Thyroid cancer is the most common endocrine cancer with an increasing incidence and mortality. Epithelial-mesenchymal transition (EMT) is a biological process contributing to tumor progression, metastasis, and the acquisition of chemotherapy resistance. The impact of the REGγ proteasome activator on EMT in human thyroid cancer cells and the molecular mechanism is still unclear. Here, we found silencing REGγ in thyroid cancer cells inhibited cell migration and invasion, with concurrent upregulation of E-cadherin and Smurf2 expression. Mechanistically, REGγ dependent regulation of Smurf2, an E3 ligase for Smad3, contributed to alteration of Zeb1/2, Snail, Slug, and Twist. Consistently, TGF-ß mediated suppression of E-cadherin was attenuated in REGγ deficient cells, coupled with changes in cell morphology, migration and invasion. Furthermore, xenograft metastasis mouse model showed a reduced E-cadherin expression at both mRNA and protein levels, and decreased cell migration. Taken together, our findings provided an important evidence for the role of REGγ in tumor suppression, thereby implicating REGγ as a potential anti-cancer strategy in thyroid cancer therapy.


Asunto(s)
Antígenos CD/metabolismo , Autoantígenos/metabolismo , Cadherinas/metabolismo , Transición Epitelial-Mesenquimal , Complejo de la Endopetidasa Proteasomal/metabolismo , Neoplasias de la Tiroides/patología , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos
6.
Exp Dermatol ; 26(11): 1118-1124, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28605165

RESUMEN

It has been reported that the proteasome activator REGγ is associated with multiple oncogenic pathways in human cancers. However, the role of REGγ in the development of melanoma and the underlying mechanisms remain unclear. In this study, we attempted to investigate the effects of REGγ on human melanoma cell proliferation in vitro and in vivo. We demonstrated that knockdown of REGγ inhibited melanoma cell growth and arrested melanoma cell at G1 phase. Furthermore, depletion of REGγ also inhibited the xenograft growth of human melanoma. Mechanistically, REGγ activates Wnt/ß-catenin signal pathway by degrading GSK-3ß in melanoma cell lines and mouse models. Transient knockdown of ß-catenin effectively blocked cell proliferation in REGγ wild-type melanoma cells. In human melanoma samples, REGγ was overexpressed and positively correlated with ß-catenin levels. This study demonstrates that REGγ is a central molecule in the development of melanoma by regulating Wnt/ß-catenin pathway. This suggests that targeting REGγ could be an alternative therapeutic approach for melanoma.


Asunto(s)
Autoantígenos/genética , Proliferación Celular/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Melanoma/genética , Complejo de la Endopetidasa Proteasomal/genética , Vía de Señalización Wnt/genética , beta Catenina/metabolismo , Animales , Autoantígenos/metabolismo , Línea Celular Tumoral , Femenino , Puntos de Control de la Fase G1 del Ciclo Celular , Técnicas de Silenciamiento del Gen , Glucógeno Sintasa Quinasa 3 beta/genética , Humanos , Melanoma/metabolismo , Ratones , Trasplante de Neoplasias , Complejo de la Endopetidasa Proteasomal/metabolismo , ARN Mensajero/metabolismo , ARN Interferente Pequeño , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...