Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
J Biol Chem ; 300(6): 107309, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38657867

RESUMEN

Novel components in the noncanonical Hippo pathway that mediate the growth, metastasis, and drug resistance of breast cancer (BC) cells need to be identified. Here, we showed that expression of SAM and SH3 domain-containing protein 1 (SASH1) is negatively correlated with expression of mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4) in a subpopulation of patients with luminal-subtype BC. Downregulated SASH1 and upregulated MAP4K4 synergistically regulated the proliferation, migration, and invasion of luminal-subtype BC cells. The expression of LATS2, SASH1, and YAP1 and the phosphorylation of YAP1 were negatively regulated by MAP4K4, and LATS2 then phosphorylated SASH1 to form a novel MAP4K4-LATS2-SASH1-YAP1 cascade. Dephosphorylation of Yes1 associated transcriptional regulator (YAP1), YAP1/TAZ nuclear translocation, and downstream transcriptional regulation of YAP1 were promoted by the combined effects of ectopic MAP4K4 expression and SASH1 silencing. Targeted inhibition of MAP4K4 blocked proliferation, cell migration, and ER signaling both in vitro and in vivo. Our findings reveal a novel MAP4K4-LATS2-SASH1-YAP1 phosphorylation cascade, a noncanonical Hippo pathway that mediates ER signaling, tumorigenesis, and metastasis in breast cancer. Targeted intervention with this noncanonical Hippo pathway may constitute a novel alternative therapeutic approach for endocrine-resistant BC.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Neoplasias de la Mama , Péptidos y Proteínas de Señalización Intracelular , Proteínas Serina-Treonina Quinasas , Factores de Transcripción , Proteínas Supresoras de Tumor , Proteínas Señalizadoras YAP , Humanos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Femenino , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Señalizadoras YAP/metabolismo , Proteínas Señalizadoras YAP/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Animales , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Ratones , Transducción de Señal , Metástasis de la Neoplasia , Movimiento Celular , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/genética , Fosforilación , Ratones Desnudos , Carcinogénesis/genética , Carcinogénesis/metabolismo
2.
J Exp Clin Cancer Res ; 42(1): 219, 2023 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-37620897

RESUMEN

BACKGROUND: The Makorin ring finger protein 1 (MKRN1) gene, also called RNF61, is located on the long arm of chromosome 7 and is a member of the RING finger protein family. The E3 ubiquitin ligase MKRN1 is closely linked to tumour development, but the exact mechanism needs to be elucidated. In this study, we aimed to investigate the specific mechanism and role of MKRN1 in colorectal cancer (CRC) development. METHODS: MKRN1 expression in CRC was analysed using the Cancer Cell Line Encyclopaedia and the Cancer Genome Atlas (TCGA) databases. Rectal tumour tissues were frozen to explore the MKRN1 expression in CRC and its clinical significance. The impact of MKRN1 on CRC cell proliferation and migration was observed using CCK8, colony formation, wound healing, and transwell assays. A combination of MKRN1 quantitative proteomics, ubiquitination modification omics analysis, and a string of in vitro and in vivo experiments revealed the potential mechanisms by which MKRN1 regulates CRC metastasis. RESULTS: MKRN1 expression was significantly elevated in CRC tissues compared to paracancerous tissues and was positively linked with prognosis (P < 0.01). MKRN1 downregulation inhibits CRC cell proliferation, migration, and invasion. Conversely, MKRN1 overexpression promotes the proliferation, migration, and invasion of CRC cells. Mechanistically, MKRN1 induces epithelial-mesenchymal transition (EMT) in CRC cells via ubiquitination and degradation of Smad nuclear-interacting protein 1 (SNIP1). Furthermore, SNIP1 inhibits transforming growth factor-ß (TGF-ß) signalling, and MKRN1 promotes TGF-ß signalling by degrading SNIP1 to induce EMT in CRC cells. Finally, using conditional knockout mice, intestinal lesions and metastatic liver microlesions were greatly reduced in the intestinal knockout MKRN1 group compared to that in the control group. CONCLUSIONS: High MKRN1 levels promote TGF-ß signalling through ubiquitination and degradation of SNIP1, thereby facilitating CRC metastasis, and supporting MKRN1 as a CRC pro-cancer factor. The MKRN1/SNIP1/TGF-ß axis may be a potential therapeutic target in CRC.


Asunto(s)
Neoplasias Colorrectales , Proteínas de Unión al ARN , Ribonucleoproteínas , Animales , Ratones , Línea Celular , Proliferación Celular , Neoplasias Colorrectales/genética , Proteolisis , Humanos , Ribonucleoproteínas/metabolismo , Proteínas de Unión al ARN/genética , Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal
3.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 38(3): 237-243, 2022 Mar.
Artículo en Zh | MEDLINE | ID: mdl-35365989

RESUMEN

Objective To investigate the possible mechanism of doxorubicin hydrochloride (DOX) inhibiting the proliferation of HT29 and HCT15 colon cancer cells. Methods The gradient concentrations of (0, 0.08, 0.16, 0.32, 0.64, 1.28) µmol/L DOX were used to treat HT29 and HCT15 cells for 24, 48 and 72 hours, and the cell proliferation activity was detected by CCK-8 assay to determine the optimal DOX concentration and treatment time. According to different treatments, HT29 and HCT15 cells were divided into 2 groups: control group (only DMSO treatment) and (0.16, 0.32, 0.64, 1.28) µmol/L DOX group. Western blot was used to detect the effect of inhibiting autophagy on apoptosis, with 3-methyladenine (3-MA) group and 3-MA combined with DOX group supplemented. The colony formation of colon cancer cells was detected by colony formation assay, and the expression of cell B-cell lymphoma 2 (Bcl2), Bcl2-associated X protein (BAX), beclin 1, and LC3 protein were detected by Western blot. Results DOX inhibited the proliferation and colony formation of colon cancer cells, and promoted cell apoptosis in a concentration-dependent manner; DOX promoted autophagy in cells, and the expression of beclin 1 and LC3 II increased in a concentration-dependent manner; DOX promoted apoptosis of colon cancer cells, which was improved by inhibiting autophagy. Conclusion DOX inhibits the proliferation of colon cancer cells and promotes their apoptosis, and inhibition of autophagy in colon cancer cells can increase the sensitivity of apoptosis induced by DOX.


Asunto(s)
Neoplasias del Colon , Doxorrubicina , Apoptosis , Autofagia , Línea Celular Tumoral , Doxorrubicina/farmacología , Humanos
4.
Int J Mol Med ; 46(3): 1118-1134, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32582980

RESUMEN

The SAM and SH3 domain­containing 1 (SASH1) genes have been identified as the causal genes of dyschromatosis universalis hereditaria (DUH); these genes cause the pathological phenotypes of DUH, and SASH1 variants have been shown to regulate the abnormal pigmentation phenotype in human skin in various genodermatoses. However, investigations into the mutated SASH1 gene have been limited to in vitro studies. In the present study, to recapitulate the molecular pathological phenotypes of individuals with DUH induced by SASH1 mutations, a heterozygous BALB/c mouse model, in which the human SASH1 c.1654 T>G (p. Tyr 551Asp, Y551D) mutation was knocked in was first generated. The in vivo functional experiments on Y551D SASH1 indicated that the increased expression of microphthalmia­associated transcription factor (Mitf) was uniformly induced in the tails of heterozygous BALB/c mice, and an increased quantity of Mitf­positive epithelial cells was also detected. An increased expression of Mitf­ and Mitf­positive cells was also demonstrated in the epithelial tissues of Y551D­SASH1 affected individuals. In the present study, Mitf expression was also found to be increased by Y551D SASH1 in vitro. Taken together, these findings indicate that the upregulation of Mitf is the bona fide effector of the Y551D SASH1­mediated melanogenesis signaling pathway in vivo. SASH1 may function as a scaffold molecule for the assembly of a SASH1­Mitf molecular complex to regulate Mitf expression in the cell nucleus and thus to promote the hyperpigmented phenotype in the pathogenesis of DUH and other genodermatoses related to pigment abnormalities.


Asunto(s)
Factor de Transcripción Asociado a Microftalmía/metabolismo , Trastornos de la Pigmentación/congénito , Enfermedades Cutáneas Genéticas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Núcleo Celular/metabolismo , Femenino , Heterocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Factor de Transcripción Asociado a Microftalmía/genética , Mutación/genética , Linaje , Trastornos de la Pigmentación/genética , Trastornos de la Pigmentación/metabolismo , Unión Proteica , ARN Mensajero/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Enfermedades Cutáneas Genéticas/genética , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA