Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nature ; 597(7874): 132-137, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34408321

RESUMEN

Protein quality control systems are crucial for cellular function and organismal health. At present, most known protein quality control systems are multicomponent machineries that operate via ATP-regulated interactions with non-native proteins to prevent aggregation and promote folding1, and few systems that can broadly enable protein folding by a different mechanism have been identified. Moreover, proteins that contain the extensively charged poly-Asp/Glu (polyD/E) region are common in eukaryotic proteomes2, but their biochemical activities remain undefined. Here we show that DAXX, a polyD/E protein that has been implicated in diverse cellular processes3-10, possesses several protein-folding activities. DAXX prevents aggregation, solubilizes pre-existing aggregates and unfolds misfolded species of model substrates and neurodegeneration-associated proteins. Notably, DAXX effectively prevents and reverses aggregation of its in vivo-validated client proteins, the tumour suppressor p53 and its principal antagonist MDM2. DAXX can also restore native conformation and function to tumour-associated, aggregation-prone p53 mutants, reducing their oncogenic properties. These DAXX activities are ATP-independent and instead rely on the polyD/E region. Other polyD/E proteins, including ANP32A and SET, can also function as stand-alone, ATP-independent molecular chaperones, disaggregases and unfoldases. Thus, polyD/E proteins probably constitute a multifunctional protein quality control system that operates via a distinctive mechanism.


Asunto(s)
Proteínas Co-Represoras/metabolismo , Chaperonas Moleculares/metabolismo , Pliegue de Proteína , Animales , Línea Celular , Células/metabolismo , Evolución Molecular , Humanos , Modelos Moleculares , Mutación , Agregado de Proteínas , Agregación Patológica de Proteínas/prevención & control , Conformación Proteica , Dominios Proteicos , Desplegamiento Proteico , Deficiencias en la Proteostasis/prevención & control , Proteínas Proto-Oncogénicas c-mdm2/química , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
2.
Cell Rep ; 33(9): 108418, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33264628

RESUMEN

Neurodegenerative diseases are characterized by the formation and propagation of protein aggregates, especially amyloid fibrils. However, what normally suppresses protein misfolding and aggregation in metazoan cells remains incompletely understood. Here, we show that TRIM11, a member of the metazoan tripartite motif (TRIM) family, both prevents the formation of protein aggregates and dissolves pre-existing protein deposits, including amyloid fibrils. These molecular chaperone and disaggregase activities are ATP independent. They enhance folding and solubility of normal proteins and cooperate with TRIM11 SUMO ligase activity to degrade aberrant proteins. TRIM11 abrogates α-synuclein fibrillization and restores viability in cell models of Parkinson's disease (PD). Intracranial adeno-associated viral delivery of TRIM11 mitigates α-synuclein-mediated pathology, neurodegeneration, and motor impairments in a PD mouse model. Other TRIMs can also function as ATP-independent molecular chaperones and disaggregases. Thus, we define TRIMs as a potent and multifunctional protein quality-control system in metazoa, which might be applied to treat neurodegenerative diseases.


Asunto(s)
Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Enfermedad de Parkinson/patología , Agregado de Proteínas
3.
Onco Targets Ther ; 11: 2097-2109, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29713182

RESUMEN

OBJECTIVE: Lung cancer is the leading cause of cancer-related death worldwide. Aerobic glycolysis is considered the seventh hallmark of cancer. The M2 isoform of pyruvate kinase (PKM2) is an important rate-limiting enzyme in glycolytic pathway, and is strongly expressed in several types of cancer. Thus, understanding the underlying mechanisms of regulation of PKM2 is of great value for targeted therapy for lung cancer. PATIENTS AND METHODS: Seventy-three lung adenocarcinoma patients were analyzed in our study. The expression levels of PKM2 were analyzed by immunohistochemistry on tissues. The effect of small ubiquitin-like modifier 1 (SUMO1) on PKM2 expression was investigated using Western blot assay and quantitative polymerase chain reaction. PKM2 SUMO1 modification was determined by in vitro and in vivo SUMOylation assays. 18F-deoxyglucose uptake and lactate production measurements were conducted to research the levels of glycolysis. The level of oxidative phosphorylation in cells was determined by cellular oxygen consumption rate measurements. Cell proliferation assays were carried out to confirm the growth ability of tumor cells. RESULTS: PKM2 was overexpressed in lung adenocarcinoma patients based on immunohistochemical staining. Patients with high PKM2 expression had reduced overall survival rate (P=0.017) and disease-free survival rate (P=0.027) compared with those with low PKM2 expression. SUMO1 promoted PKM2-dependent glycolysis. Western blotting analysis showed that SUMO1 knockdown in A549 cells led to a significant decrease in PKM2 protein expression. PKM2 could be covalently modified by SUMO1 at K336 (Lys336) site. SUMO1 modification of PKM2 at Lys-336 site increased glycolysis and promoted its cofactor functions. Moreover, PKM2 SUMO1 modification promoted the proliferation of A549 cells in vitro. CONCLUSION: This information is important in elucidating a new mechanism of regulation of PKM2, and suggested that SUMO1 modification of PKM2 could be a potential therapeutic target in lung cancer.

4.
Biochem Biophys Res Commun ; 473(4): 953-958, 2016 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-27045080

RESUMEN

Pyruvate kinase M2 (PKM2) is a key enzyme of glycolysis which is highly expressed in many tumor cells, and plays an important role in the Warburg effect. In previous study, we found PIM2 phosphorylates PKM2 at Thr454 residue (Yu, etl 2013). However, the functions of PKM2 Thr454 modification in cancer cells still remain unclear. Here we find PKM2 translocates into the nucleus after Thr454 phosphorylation. Replacement of wild type PKM2 with a mutant (T454A) enhances mitochondrial respiration, decreases pentose phosphate pathway, and enhances chemosensitivity in A549 cells. In addition, the mutant (T454A) PKM2 reduces xenograft tumor growth in nude mice. These findings demonstrate that PKM2 T454 phosphorylation is a potential therapeutic target in lung cancer.


Asunto(s)
Proteínas Portadoras/metabolismo , Núcleo Celular/metabolismo , Neoplasias Pulmonares/enzimología , Proteínas de la Membrana/metabolismo , Piruvato Quinasa/metabolismo , Hormonas Tiroideas/metabolismo , Células A549 , Transporte Activo de Núcleo Celular , Animales , Proteínas Portadoras/química , Proliferación Celular , Resistencia a Antineoplásicos , Neoplasias Pulmonares/patología , Proteínas de la Membrana/química , Ratones Desnudos , Mitocondrias/metabolismo , Vía de Pentosa Fosfato , Fosforilación , Piruvato Quinasa/química , Treonina/metabolismo , Hormonas Tiroideas/química , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas de Unión a Hormona Tiroide
5.
Sci Rep ; 6: 22449, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26926077

RESUMEN

Pyruvate kinase M2 (PKM2), which is predominantly expressed in most cancers, plays a key role in the Warburg effect. However, how PKM2 functions as a tumor supportive protein has not been fully elucidated. Here, we identified tristetraprolin (TTP), an AU-rich, element-binding protein that regulates mRNA stability, as a new binding partner of PKM2. Our data reveal that PKM2 suppresses TTP protein levels by promoting its phosphorylation, ubiquitination, and proteasome degradation, reducing its mRNA turnover ability and ultimately impairing cell viability in breast cancer cells. The p38/mitogen-activated protein kinase (MAPK) pathway might be involved in PKM2-mediated TTP degradation, while treatment with the p38 inhibitor or siRNA abolished PKM2-induced TTP protein degradation. These findings demonstrate that PKM2-TTP association is crucial for regulating breast cancer cell proliferation and is therefore a potential therapeutic target in cancer.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas Portadoras/metabolismo , Proteínas de la Membrana/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Piruvato Quinasa/metabolismo , Hormonas Tiroideas/metabolismo , Tristetraprolina/metabolismo , Empalme Alternativo/genética , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular/genética , Femenino , Células HEK293 , Humanos , Células MCF-7 , Fosforilación , Unión Proteica , Interferencia de ARN , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas de Unión a Hormona Tiroide
6.
Cell Signal ; 26(7): 1560-6, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24686083

RESUMEN

HIF-1α is implicated in hepatocellular carcinoma (HCC) pathologies. Here, we screened a human liver cDNA library for HIF-1α-interacting partners using a yeast two-hybrid system. We identified 53 genes, including formiminotransferase cyclodeaminase (FTCD), which was confirmed by co-immunoprecipitation. Moreover, our data indicated that HIF-1α mediated the effects of hypoxia on FTCD induction via binding to the hypoxia-responsive elements of the FTCD promoter. Knockdown of FTCD reduced the effects of HIF-1α in hypoxia and enhanced chemosensitivity in HepG2 cells. Our findings suggested crosstalk between FTCD and HIF signaling and promoted HCC progression, thus implicating FTCD as a therapeutic target for HCC.


Asunto(s)
Amoníaco-Liasas/metabolismo , Carcinoma Hepatocelular/patología , Glutamato Formimidoiltransferasa/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Hepáticas/patología , Técnicas del Sistema de Dos Híbridos , Amoníaco-Liasas/biosíntesis , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Proliferación Celular , Glutamato Formimidoiltransferasa/biosíntesis , Células Hep G2 , Humanos , Enzimas Multifuncionales , Invasividad Neoplásica/patología , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño , Elementos de Respuesta/genética , Transducción de Señal
7.
PLoS One ; 9(3): e92949, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24658033

RESUMEN

Pyruvate kinase M2 (PKM2) is predominantly expressed in cancers, which is considered as a key regulator of the Warburg effect. In this study, HSP40 was identified as a novel binding partner of PKM2. HSP40-PKM2 association destabilized PKM2 protein through HSC70. In the presence of HSP40, PKM2 protein level and PKM2-mediated PDK1 expression were down-regulated. Moreover, HSP40 was involved in regulating glucose metabolism on PKM2 dependent way and at the mean time had an effect on mitochondrial oxygen respiration. In line with inhibition effect of HSP40 on glycolysis, the growth of cancer cells was inhibited by HSP40.Our data provided a new regulation mechanism of PKM2, which suggested a new therapeutic target for cancer therapy.


Asunto(s)
Proteínas del Choque Térmico HSP40/metabolismo , Neoplasias/metabolismo , Piruvato Quinasa/metabolismo , Proteínas Portadoras/metabolismo , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Glucosa/metabolismo , Glucólisis , Humanos , Unión Proteica , Mapeo de Interacción de Proteínas , Estabilidad Proteica
8.
PLoS One ; 9(2): e88301, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24505470

RESUMEN

To survive under hypoxic conditions, cancer cells remodel glucose metabolism to support tumor progression. HIF transcription factor is essential for cellular response to hypoxia. The underlying mechanism how HIF is constitutively activated in cancer cells remains elusive. In the present study, we characterized a regulatory feedback loop between HIF-1α and PIM2 in HepG2 cells. Serine/threonine kinase proto-oncogene PIM2 level was induced upon hypoxia in a HIF-1α-mediated manner in cancer cells. HIF-1α induced PIM2 expression via binding to the hypoxia-responsive elements (HREs) of the PIM2 promoter. In turn, PIM2 interacted with HIF-1α, especially a transactivation domain of HIF-1α. PIM2 as a co-factor but not an upstream kinase of HIF-1α, enhanced HIF-1α effect in response to hypoxia. The positive feedback loop between PIM2 and HIF-1α was correlated with glucose metabolism as well as cell survival in HepG2 cells. Such a regulatory mode may be important for the adaptive responses of cancer cells in antagonizing hypoxia during cancer progression.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Hipoxia de la Célula , Regulación Neoplásica de la Expresión Génica , Glucosa/metabolismo , Células Hep G2 , Humanos , Neoplasias/genética , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/genética , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/genética
9.
J Biol Chem ; 288(49): 35406-16, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24142698

RESUMEN

Pyruvate kinase M2 (PKM2) is a key player in the Warburg effect of cancer cells. However, the mechanisms of regulating PKM2 are not fully elucidated. Here, we identified the protein-serine/threonine kinase PIM2, a known oncogene, as a novel binding partner of PKM2. The interaction between PIM2 and PKM2 was confirmed by multiple biochemical approaches in vitro and in cultured cells. Importantly, we found that PIM2 could directly phosphorylate PKM2 on the Thr-454 residue, resulting in an increase of PKM2 protein levels. Compared with wild type, PKM2 with the phosphorylation-defective mutation displayed a reduced effect on glycolysis, co-activating HIF-1α and ß-catenin, and cell proliferation, while enhancing mitochondrial respiration of cancer cells. These findings demonstrate that PIM2-dependent phosphorylation of PKM2 is critical for regulating the Warburg effect in cancer, highlighting PIM2 as a potential therapeutic target.


Asunto(s)
Neoplasias/genética , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Piruvato Quinasa/metabolismo , Integración Viral/genética , Línea Celular Tumoral , Glucólisis , Células HEK293 , Células Hep G2 , Humanos , Oncogenes , Fosforilación , Unión Proteica , Piruvato Quinasa/química , Piruvato Quinasa/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Treonina/química , Técnicas del Sistema de Dos Híbridos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...