Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Metab ; 35(12): 2216-2230.e8, 2023 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-37979583

RESUMEN

Mammalian target of rapamycin complex 1 (mTORC1) monitors cellular amino acid changes for function, but the molecular mediators of this process remain to be fully defined. Here, we report that depletion of cellular amino acids, either alone or in combination, leads to the ubiquitination of mTOR, which inhibits mTORC1 kinase activity by preventing substrate recruitment. Mechanistically, amino acid depletion causes accumulation of uncharged tRNAs, thereby stimulating GCN2 to phosphorylate FBXO22, which in turn accrues in the cytoplasm and ubiquitinates mTOR at Lys2066 in a K27-linked manner. Accordingly, mutation of mTOR Lys2066 abolished mTOR ubiquitination in response to amino acid depletion, rendering mTOR insensitive to amino acid starvation both in vitro and in vivo. Collectively, these data reveal a novel mechanism of amino acid sensing by mTORC1 via a previously unknown GCN2-FBXO22-mTOR pathway that is uniquely controlled by uncharged tRNAs.


Asunto(s)
Proteínas Serina-Treonina Quinasas , Serina-Treonina Quinasas TOR , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Aminoácidos/metabolismo , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo
2.
Cell Death Dis ; 13(6): 532, 2022 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-35668069

RESUMEN

PTENα and PTENß (PTENα/ß), two long translational variants of phosphatase and tensin homolog on chromosome 10 (PTEN), exert distinct roles from canonical PTEN, including promoting carcinogenesis and accelerating immune-resistant cancer progression. However, their roles in carcinogenesis remain greatly unknown. Herein, we report that, after secreting into the extracellular space, PTENα/ß proteins are efficiently cleaved into a short N-terminal and a long C-terminal fragment by the proprotein convertase Furin at a polyarginine stretch in their N-terminal extensions. Although secreted PTENα/ß and their cleaved fragment cannot enter cells, treatment of the purified C-terminal fragment but not cleavage-resistant mutants of PTENα exerts a tumor-suppressive role in vivo. As a result, overexpression of cleavage-resistant PTENα mutants manifest a tumor-promoting role more profound than that of wild-type PTENα. In line with these, the C-terminal fragment is significantly downregulated in liver cancer tissues compared to paired normal tissues, which is consistent with the downregulated expression of Furin. Collectively, we show that extracellular PTENα/ß present opposite effects on carcinogenesis from intracellular PTENα/ß, and propose that the tumor-suppressive C-terminal fragment of PTENα/ß might be used as exogenous agent to treat cancer.


Asunto(s)
Furina , Neoplasias Hepáticas , Carcinogénesis , Furina/genética , Humanos , Proproteína Convertasas
3.
Cell Death Differ ; 29(8): 1569-1581, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35140358

RESUMEN

Linker histone H1 proteins contain many variants in mammalian and can stabilize the condensed state of chromatin by binding to nucleosomes and promoting a more inaccessible structure of DNA. However, it is poorly understood how the binding of histone H1s to chromatin DNA is regulated. Screened as one of a collection of epithelial cells-enriched long non-coding RNAs (lncRNAs), here we found that small nucleolar RNA host gene 8 (SNHG8) is a chromatin-localized lncRNA and presents strong interaction and phase separation with histone H1 variants. Moreover, SNHG8 presents stronger ability to bind H1s than linker DNA, and outcompetes linker DNA for H1 binding. Consequently, loss of SNHG8 increases the amount of H1s that bind to chromatin, promotes chromatin condensation, and induces an epithelial differentiation-associated gene expression pattern. Collectively, our results propose that the highly abundant SNHG8 in epithelial cells keeps histone H1 variants out of nucleosome and its loss contributes to epithelial cell differentiation.


Asunto(s)
Histonas , ARN Largo no Codificante , Animales , Cromatina , ADN/metabolismo , Células Epiteliales/metabolismo , Histonas/genética , Histonas/metabolismo , Mamíferos/metabolismo , Nucleosomas , ARN Largo no Codificante/genética
4.
Blood ; 138(24): 2485-2498, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34359074

RESUMEN

Proper regulation of p53 signaling is critical for the maintenance of hematopoietic stem cells (HSCs) and leukemic stem cells (LSCs). The hematopoietic cell-specific mechanisms regulating p53 activity remain largely unknown. Here, we demonstrate that conditional deletion of acidic leucine-rich nuclear phosphoprotein 32B (ANP32B) in hematopoietic cells impairs repopulation capacity and postinjury regeneration of HSCs. Mechanistically, ANP32B forms a repressive complex with p53 and thus inhibits the transcriptional activity of p53 in hematopoietic cells, and p53 deletion rescues the functional defect in Anp32b-deficient HSCs. Of great interest, ANP32B is highly expressed in leukemic cells from patients with chronic myelogenous leukemia (CML). Anp32b deletion enhances p53 transcriptional activity to impair LSC function in a murine CML model and exhibits synergistic therapeutic effects with tyrosine kinase inhibitors in inhibiting CML propagation. In summary, our findings provide a novel strategy to enhance p53 activity in LSCs by inhibiting ANP32B and identify ANP32B as a potential therapeutic target in treating CML.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Células Cultivadas , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Proteína p53 Supresora de Tumor/genética
5.
Nat Commun ; 11(1): 1720, 2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-32249768

RESUMEN

Nuclear localization of PTEN is essential for its tumor suppressive role, and loss of nuclear PTEN is more prominent than cytoplasmic PTEN in many kinds of cancers. However, nuclear PTEN-specific regulatory mechanisms were rarely reported. Based on the finding that nuclear PTEN is more unstable than cytoplasmic PTEN, here we identify that F-box only protein 22 (FBXO22) induces ubiquitylation of nuclear but not cytoplasmic PTEN at lysine 221, which is responsible for the degradation of nuclear PTEN. FBXO22 plays a tumor-promoting role by ubiquitylating and degrading nuclear PTEN. In accordance, FBXO22 is overexpressed in various cancer types, and contributes to nuclear PTEN downregulation in colorectal cancer tissues. Cumulatively, our study reports the mechanism to specifically regulate the stability of nuclear PTEN, which would provide the opportunity for developing therapeutic strategies aiming to achieve complete reactivation of PTEN as a tumor suppressor.


Asunto(s)
Carcinogénesis/genética , Núcleo Celular/metabolismo , Transformación Celular Neoplásica/genética , Neoplasias Colorrectales/metabolismo , Proteínas F-Box/metabolismo , Fosfohidrolasa PTEN/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Línea Celular Tumoral , Cromatografía Liquida , Neoplasias Colorrectales/genética , Citoplasma/metabolismo , Proteínas F-Box/genética , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Fosfohidrolasa PTEN/química , Fosfohidrolasa PTEN/genética , ARN Interferente Pequeño , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal/genética , Espectrometría de Masas en Tándem , Análisis de Matrices Tisulares , Trasplante Heterólogo , Ubiquitinación
6.
Nat Cell Biol ; 22(1): 135, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31730051

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

7.
Nat Cell Biol ; 21(11): 1436-1448, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31685992

RESUMEN

PTENα and PTENß are two longer translational variants of phosphatase and tensin homolog (PTEN) messenger RNA. Their expressional regulations and functions in carcinogenesis remain largely unknown. Here, we demonstrate that, in contrast with the well-established tumour-suppressive role of canonical PTEN, PTENα and PTENß promote tumourigenesis by directly interacting with the histone H3 lysine 4 (H3K4) presenter WDR5 to promote H3K4 trimethylation and maintain a tumour-promoting signature. We also show that USP9X and FBXW11 bind to the amino-terminal extensions of PTENα/ß, and respectively deubiquitinate and ubiquitinate lysines 235 and 239 in PTENα to regulate PTENα/ß stability. In accordance, USP9X promotes tumourigenesis and FBXW11 suppresses tumourigenesis through PTENα/ß. Taken together, our results indicate that the Pten gene is a double-edged sword for carcinogenesis, and reinterpretation of the importance of the Pten gene in carcinogenesis is warranted.


Asunto(s)
Carcinogénesis/genética , Carcinoma Hepatocelular/genética , Regulación Neoplásica de la Expresión Génica , Histonas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Hepáticas/genética , Fosfohidrolasa PTEN/genética , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Histonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Desnudos , Fosfohidrolasa PTEN/metabolismo , Proteolisis , Transducción de Señal , Análisis de Supervivencia , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas con Repetición de beta-Transducina/genética , Proteínas con Repetición de beta-Transducina/metabolismo
8.
Nat Commun ; 9(1): 2392, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29921876

RESUMEN

Dysregulation of pre-mRNA alternative splicing (AS) is closely associated with cancers. However, the relationships between the AS and classic oncogenes/tumor suppressors are largely unknown. Here we show that the deletion of tumor suppressor PTEN alters pre-mRNA splicing in a phosphatase-independent manner, and identify 262 PTEN-regulated AS events in 293T cells by RNA sequencing, which are associated with significant worse outcome of cancer patients. Based on these findings, we report that nuclear PTEN interacts with the splicing machinery, spliceosome, to regulate its assembly and pre-mRNA splicing. We also identify a new exon 2b in GOLGA2 transcript and the exon exclusion contributes to PTEN knockdown-induced tumorigenesis by promoting dramatic Golgi extension and secretion, and PTEN depletion significantly sensitizes cancer cells to secretion inhibitors brefeldin A and golgicide A. Our results suggest that Golgi secretion inhibitors alone or in combination with PI3K/Akt kinase inhibitors may be therapeutically useful for PTEN-deficient cancers.


Asunto(s)
Empalme Alternativo , Genes Supresores de Tumor , Aparato de Golgi/metabolismo , Fosfohidrolasa PTEN/metabolismo , Precursores del ARN/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Línea Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Células Cultivadas , Células HEK293 , Células HeLa , Humanos , Ratones Noqueados , Fosfohidrolasa PTEN/genética , Precursores del ARN/genética , Transducción de Señal , Empalmosomas/genética , Empalmosomas/metabolismo
9.
Carcinogenesis ; 37(11): 1079-1088, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27543779

RESUMEN

Recently, we have reported that apoptosis-inducing factor (AIF) regulates the epithelial-mesenchymal transition (EMT) process of cancers, but the mechanisms underlying the regulation of AIF expression in cancers remain greatly unknown. Here, we report that hypoxia inversely correlates with the expression of AIF in tumor tissues from a cohort of colon cancer patients and inhibits AIF expression in multiple colon cancer cell lines. This inhibition is mediated by hypoxia-inducible factor-1 (HIF-1), which transcriptionally represses AIF through direct binding to the hypoxia-response element in AIF promoter as revealed by luciferase reporter and chromatin immunoprecipitation assays. We also show that downregulation of AIF contributes to hypoxia-induced EMT as overexpression or silencing of AIF partially reverses or potentiates the EMT program initiated by hypoxic treatment. Mechanistic study reveals that downregulation of AIF by hypoxia causes oxidative inactivation of the lipid phosphatase activity of phosphatase and tensin homolog on chromosome 10 (PTEN), with ensuing activation of Akt kinase, phosphorylation of the Akt substrate GSK-3ß and activation of WNT/ß-catenin signaling in colon cancer cells. These results identify AIF as a novel target gene of HIF-1 and reveal the role of AIF downregulation in hypoxia-induced EMT.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Neoplasias del Colon/metabolismo , Transición Epitelial-Mesenquimal , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Animales , Factor Inductor de la Apoptosis/metabolismo , Secuencia de Bases , Sitios de Unión , Hipoxia de la Célula , Línea Celular Tumoral , Neoplasias del Colon/patología , Regulación hacia Abajo , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Oxidación-Reducción , Fosfohidrolasa PTEN/metabolismo , Regiones Promotoras Genéticas , Vía de Señalización Wnt
10.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 24(2): 519-25, 2016 Apr.
Artículo en Chino | MEDLINE | ID: mdl-27151022

RESUMEN

OBJECTIVE: To investigate the effect and mechanism of Factor Xa on the differentiation of Meg-01 cells into platelet-like particles. METHODS: The Meg-01 cells were used as experimental object, Factor Xa was used as agonist. Cell proliferation was detected by CCK-8 assay. The viability of platelet-like particles was analyzed by AlamaBlue kit. MAPK/ERK pathway and PI3K/AKT pathway were assayed by Western blot. The expression of CD41b was analyzed by Western blot and flow cytometry. Cell cycle and apoptosis were detected by flow cytometry. RESULTS: The Factor Xa (1 µg/ml) inhibited cell viability, induced apoptosis. Factor Xa triggered cell arrest at the G(2)/M stage and down-regulated the expression of SKP2. After Meg-01 cells were stimulated by Factor Xa, the expression of CD41b was up-regulated and the MAPK/ERK pathway and PI3K/AKT pathway were activated. The platelets-like particles stimulated by FXa activation were viable. CONCLUSION: The Factor Xa maybe display some effect on the differentiation of megakaryocytes into platelets.


Asunto(s)
Plaquetas/citología , Diferenciación Celular/efectos de los fármacos , Factor Xa/farmacología , Megacariocitos/citología , Apoptosis , Plaquetas/efectos de los fármacos , Puntos de Control del Ciclo Celular , Línea Celular , Proliferación Celular , Supervivencia Celular , Humanos , Sistema de Señalización de MAP Quinasas , Megacariocitos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
11.
Biomed Res Int ; 2016: 9313269, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27064425

RESUMEN

OBJECTIVES: After development and differentiation, megakaryocytes (MKs) can produce platelets. As is well known, thrombopoietin (TPO) can induce MKs to differentiate. The effect of thrombin on MKs differentiation is not clear. In this study, we used a human megakaryoblastic leukemia cell line (Meg-01) to assess the effect of thrombin on MKs differentiation. METHODS: In order to interrogate the role of thrombin in Meg-01 cells differentiation, the changes of morphology, cellular function, and expression of diverse factors were analyzed. RESULTS: The results show that thrombin suppresses Meg-01 cells proliferation and induces apoptosis and cell cycle arrest. Thrombin upregulates the expression of CD41b, which is one of the most important MK markers. Globin transcription factor 1 (GATA-1), an important transcriptional regulator, controls MK development and maturation. The expression of GATA-1 is also upregulated by thrombin in Meg-01 cells. The expression of B-cell lymphoma 2 (Bcl-2), an apoptosis-inhibitory protein, is downregulated by thrombin. Phosphorylated protein kinase B (p-AKT) and phosphorylated extracellular signal-regulated kinase (p-ERK) were upregulated by thrombin in Meg-01 cells. All the results are consistent with Meg-01 cells treated with TPO. DISCUSSION AND CONCLUSION: In conclusion, all these data indicate that thrombin maybe plays an important role in MK differentiation into platelets. However, whether the platelet-like particles are certainly platelets remains unknown.


Asunto(s)
Plaquetas , Proliferación Celular/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Megacariocitos , Trombina , Plaquetas/citología , Plaquetas/efectos de los fármacos , Ciclo Celular , Línea Celular , Tamaño de la Célula/efectos de los fármacos , Humanos , Integrina beta3 , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Trombina/metabolismo , Trombina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...