Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Am J Hum Genet ; 107(6): 1149-1156, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33186543

RESUMEN

The Congenital Dyserythropoietic Anemia (CDA) Registry was established with the goal to facilitate investigations of natural history, biology, and molecular pathogenetic mechanisms of CDA. Three unrelated individuals enrolled in the registry had a syndrome characterized by CDA and severe neurodevelopmental delay. They were found to have missense mutations in VPS4A, a gene coding for an ATPase that regulates the ESCRT-III machinery in a variety of cellular processes including cell division, endosomal vesicle trafficking, and viral budding. Bone marrow studies showed binucleated erythroblasts and erythroblasts with cytoplasmic bridges indicating abnormal cytokinesis and abscission. Circulating red blood cells were found to retain transferrin receptor (CD71) in their membrane, demonstrating that VPS4A is critical for normal reticulocyte maturation. Using proband-derived induced pluripotent stem cells (iPSCs), we have successfully modeled the hematologic aspects of this syndrome in vitro, recapitulating their dyserythropoietic phenotype. Our findings demonstrate that VPS4A mutations cause cytokinesis and trafficking defects leading to a human disease with detrimental effects to erythropoiesis and neurodevelopment.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/genética , Anemia Diseritropoyética Congénita/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , ATPasas de Translocación de Protón Vacuolares/genética , Adenosina Trifosfatasas/metabolismo , Anemia Diseritropoyética Congénita/patología , Médula Ósea/patología , Células de la Médula Ósea/metabolismo , Niño , Preescolar , Citocinesis , Endosomas/metabolismo , Eritroblastos/metabolismo , Eritrocitos/citología , Eritropoyesis , Femenino , Humanos , Células Madre Pluripotentes Inducidas/citología , Masculino , Trastornos del Neurodesarrollo/metabolismo , Fenotipo , Transporte de Proteínas , Reticulocitos/citología
2.
J Virol ; 96(6): e0181121, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35044214

RESUMEN

We previously reported that hepatitis C virus (HCV) infection activates the reactive oxygen species (ROS)/c-Jun N-terminal kinase (JNK) signaling pathway. However, the roles of ROS/JNK activation in the HCV life cycle remain unclear. We sought to identify a novel role of the ROS/JNK signaling pathway in the HCV life cycle. Immunoblot analysis revealed that HCV-induced ROS/JNK activation promoted phosphorylation of Itch, a HECT-type E3 ubiquitin ligase, leading to activation of Itch. The small interfering RNA (siRNA) knockdown of Itch significantly reduced the extracellular HCV infectivity titers, HCV RNA, and HCV core protein without affecting intracellular HCV infectivity titers, HCV RNA, and HCV proteins, suggesting that Itch is involved in the release of HCV particles. HCV-mediated JNK/Itch activation specifically promoted polyubiquitylation of an AAA-type ATPase, VPS4A, but not VPS4B, required to form multivesicular bodies. Site-directed mutagenesis revealed that two lysine residues (K23 and K121) on VPS4A were important for VPS4A polyubiquitylation. The siRNA knockdown of VPS4A, but not VPS4B, significantly reduced extracellular HCV infectivity titers. Coimmunoprecipitation analysis revealed that HCV infection specifically enhanced the interaction between CHMP1B, a subunit of endosomal sorting complexes required for transport (ESCRT)-III complex, and VPS4A, but not VPS4B, whereas VPS4A K23R/K121R greatly reduced the interaction with CHMP1B. HCV infection significantly increased ATPase activity of VPS4A, but not VPS4A K23R/K121R or VPS4B, suggesting that HCV-mediated polyubiquitylation of VPS4A contributes to activation of VPS4A. Taken together, we propose that the HCV-induced ROS/JNK/Itch signaling pathway promotes VPS4A polyubiquitylation, leading to enhanced VPS4A-CHMP1B interaction and promotion of VPS4A ATPase activity, thereby promoting the release of HCV particles. IMPORTANCE The ROS/JNK signaling pathway contributes to liver diseases, including steatosis, metabolic disorders, and hepatocellular carcinoma. We previously reported that HCV activates the ROS/JNK signaling pathway, leading to the enhancement of hepatic gluconeogenesis and apoptosis induction. This study further demonstrates that the HCV-induced ROS/JNK signaling pathway activates the E3 ubiquitin ligase Itch to promote release of HCV particles via polyubiquitylation of VPS4A. We provide evidence suggesting that HCV infection promotes the ROS/JNK/Itch signaling pathway and ESCRT/VPS4A machinery to release infectious HCV particles. Our results may lead to a better understanding of the mechanistic details of HCV particle release.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas , Complejos de Clasificación Endosomal Requeridos para el Transporte , Hepacivirus , Hepatitis C , Proteínas Represoras , Ubiquitina-Proteína Ligasas , ATPasas de Translocación de Protón Vacuolares , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Adenosina Trifosfatasas/metabolismo , Línea Celular , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Técnicas de Silenciamiento del Gen , Hepacivirus/fisiología , Hepatitis C/fisiopatología , Hepatitis C/virología , Humanos , Sistema de Señalización de MAP Quinasas , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Virión
3.
Int J Mol Sci ; 24(13)2023 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-37445978

RESUMEN

Autophagy has stabilizing functions for cardiomyocytes. Recent studies indicate that an impairment in the autophagy pathway can seriously affect morphology and function, potentially leading to heart failure. However, the role and the underlying mechanism of the endosomal sorting complex required for transport (ESCRT) family protein, in particular the AAA-ATPase vacuolar protein sorting 4a (Vps4a), in regulating myocardial autophagy remains unclear. In the present study, cardiomyocyte-specific Vps4a knockout mice were generated by crossing Vps4aflox/flox (Vps4afl/fl) with Myh6-cre transgenic mice. As a result, we observed a partially dilated left ventricular (LV) chamber, a significant increase in heart weight to body weight ratio (HW/BW), and heart weight to tibial length ratio (HW/TL), hypertrophic cardiomyopathy and early lethality starting at 3 months of age. Hematoxylin-eosin (HE), immunofluorescence assay (IFA), and Western blot (WB) revealed autophagosome accumulation in cardiomyocytes. A transcriptome-based analysis and autophagic flux tracking by AAV-RFP-GFP-LC3 showed that the autophagic flux was blocked in Vps4a knockout cardiomyocytes. In addition, we provided in vitro evidence demonstrating that Vps4a and LC3 were partially co-localized in cardiomyocytes, and the knockdown of Vps4a led to the accumulation of autophagosomes in cardiomyocytes. Similarly, the transfection of cardiomyocytes with adenovirus (Adv) mCherry-GFP-LC3 further indicated that the autophagic flux was blocked in cells with deficient levels of Vps4a. Finally, an electron microscope (EM) showed that the compromised sealing of autophagosome blocked the autophagic flux in Vps4a-depleted cardiomyocytes. These findings revealed that Vps4a contributed to the sealing of autophagosomes in cardiomyocytes. Therefore, we demonstrated that Vps4a deletion could block the autophagic flux, leading to the accumulation of degradation substances and compromised cardiac function. Overall, this study provides insights into a new theoretical basis for which autophagy may represent a therapeutic target for cardiovascular diseases.


Asunto(s)
Autofagia , Cardiomiopatía Hipertrófica , Ratones , Animales , Autofagia/genética , Miocardio/metabolismo , Autofagosomas/metabolismo , Miocitos Cardíacos/metabolismo , Ratones Transgénicos , Cardiomiopatía Hipertrófica/metabolismo , Ratones Noqueados , Transporte de Proteínas
4.
Proc Natl Acad Sci U S A ; 113(14): 3879-84, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-27001827

RESUMEN

Dysfunctional reward processing is implicated in various mental disorders, including attention deficit hyperactivity disorder (ADHD) and addictions. Such impairments might involve different components of the reward process, including brain activity during reward anticipation. We examined brain nodes engaged by reward anticipation in 1,544 adolescents and identified a network containing a core striatal node and cortical nodes facilitating outcome prediction and response preparation. Distinct nodes and functional connections were preferentially associated with either adolescent hyperactivity or alcohol consumption, thus conveying specificity of reward processing to clinically relevant behavior. We observed associations between the striatal node, hyperactivity, and the vacuolar protein sorting-associated protein 4A (VPS4A) gene in humans, and the causal role of Vps4 for hyperactivity was validated in Drosophila Our data provide a neurobehavioral model explaining the heterogeneity of reward-related behaviors and generate a hypothesis accounting for their enduring nature.


Asunto(s)
Anticipación Psicológica/fisiología , Trastorno por Déficit de Atención con Hiperactividad/genética , Trastorno por Déficit de Atención con Hiperactividad/fisiopatología , Mapeo Encefálico , Cuerpo Estriado/fisiopatología , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Recompensa , ATPasas de Translocación de Protón Vacuolares/genética , ATPasas Asociadas con Actividades Celulares Diversas , Adolescente , Consumo de Bebidas Alcohólicas/psicología , Animales , Niño , Drosophila , Femenino , Predicción , Estudio de Asociación del Genoma Completo , Haplotipos/genética , Humanos , Masculino , Motivación , Pruebas Neuropsicológicas
5.
Metab Brain Dis ; 32(2): 565-575, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28064406

RESUMEN

Vps4, vacuolar protein sorting 4, belongs to ATPases Associated with diverse cellular Activities (AAA) protein family which is made up of Vps4A and Vps4B. Previous studies demonstrated that Vps4A plays vital roles in diverse aspects such as virus budding, the efficient transport of H-Ras to the PM (plasma membrane) and the involvement in the MVB (multivesiculate bodies) pathway. Interestingly, Vps4A is also expressed in the brain. However, the distribution and function of Vps4A in ICH diseases remain unclear. In this study, we show that Vps4A may be involved in neuronal apoptosis during pathophysiological processes of intracerebral hemorrhage (ICH). Based on the results of Western blot and immunohistochemistry, we found a remarkable up-regulation of Vps4A expression surrounding the hematoma after ICH. Double labeled immunofluorescence showed that Vps4A was co-expressed with NeuN but rarely with astrocytes and microglia. Morever, we detected that neuronal apoptosis marker active caspase-3 had co-localizations with Vps4A. Additionaly, Vps4A knockdown in vitro specifically leads to decreasing neuronal apoptosis coupled with increased Akt phosphorylation. All datas suggested that Vps4A was involved in promoting neuronal apoptosis via inhibiting Akt phosphorylation after ICH.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/biosíntesis , Apoptosis/efectos de los fármacos , Hemorragia Cerebral/metabolismo , ATPasas de Translocación de Protón Vacuolares/biosíntesis , Animales , Antígenos Nucleares/metabolismo , Conducta Animal/efectos de los fármacos , Caspasa 3/metabolismo , Hemorragia Cerebral/patología , Hemorragia Cerebral/psicología , Femenino , Técnicas de Silenciamiento del Gen , Masculino , Proteínas del Tejido Nervioso/metabolismo , Proteína Oncogénica v-akt/metabolismo , Fosforilación , Embarazo , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos
6.
Biochim Biophys Acta Gen Subj ; 1868(1): 130497, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37931722

RESUMEN

BACKGROUND: In advanced and recurrent endometrial carcinoma (EC), the current state of immuno- or targeted therapy remains in the clinical research phase. Our study aimed to explore the role of the ESCRT machinery in maintaining cell membrane integrity and reversing pyroptotic cell death. METHODS: Immunohistochemistry, western blotting, and co-immunoprecipitation were performed to determine the expression and relationship between GSDMD, CHMP4B, and VPS4A. We employed techniques such as FITC Annexin V/propidium iodide staining, Ca2+ fluorescence intensity, IL-1ß enzyme-linked immunosorbent assay, and lactate dehydrogenase release assay to detect pyroptosis in endometrial cancer cells. Plasma membrane perforations and CHMP4B/VPS4A puncta were observed through electron and fluorescence confocal microscopy. RESULTS: We showed that GSDMD, CHMP4B, and VPS4A were differentially expressed in the pyroptotic EC xenograft mouse model group, as well as high, moderate, and mild expression in EC cells treated with LPS and nigericin compared to endometrial epithelial cells. Co-IP confirmed the interaction between GSDMD, CHMP4B, and VPS4A. We found that GSDMD knockdown reduced PI-positive cells, Ca2+ efflux, IL-1ß, and LDH release, while CHMP4B and VPS4A depletion enhanced these indicators in HEC1A and AN3CA cells. Electron microscopy showed membrane perforations correspondingly decreased with inactivated GSDMD and increased or decreased after CHMP4B and VPS4A depletion or overexpression in EC cells. Fluorescence confocal microscopy detected CHMP4B protein puncta associated with VPS4A at the injured plasma membrane in GSDMDNT cells. CONCLUSIONS: We preliminary evidenced that CHMP4B and VPS4A reverses GSDMD-mediated pyroptosis by facilitating cell membrane remodeling in endometrial carcinoma. Targeting CHMP4B related proteins may promote pyroptosis in endometrial tumors.


Asunto(s)
Neoplasias Endometriales , ATPasas de Translocación de Protón Vacuolares , Femenino , Humanos , Ratones , Animales , Piroptosis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Membrana Celular/metabolismo , Modelos Animales de Enfermedad , Neoplasias Endometriales/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Gasderminas , Proteínas de Unión a Fosfato/metabolismo
7.
Adv Sci (Weinh) ; 11(31): e2308307, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39166458

RESUMEN

Aloperine (ALO), a quinolizidine-type alkaloid isolated from a natural Chinese herb, has shown promising antitumor effects. Nevertheless, its common mechanism of action and specific target remain elusive. Here, it is demonstrated that ALO inhibits the proliferation and migration of non-small cell lung cancer cell lines in vitro and the tumor development in several mouse tumor models in vivo. Mechanistically, ALO inhibits the fusion of autophagosomes with lysosomes and the autophagic flux, leading to the accumulation of sequestosome-1 (SQSTM1) and production of reactive oxygen species (ROS), thereby inducing tumor cell apoptosis and preventing tumor growth. Knockdown of SQSTM1 in cells inhibits ROS production and reverses ALO-induced cell apoptosis. Furthermore, VPS4A is identified as a direct target of ALO, and the amino acids F153 and D263 of VPS4A are confirmed as the binding sites for ALO. Knockout of VPS4A in H1299 cells demonstrates a similar biological effect as ALO treatment. Additionally, ALO enhances the efficacy of the anti-PD-L1/TGF-ß bispecific antibody in inhibiting LLC-derived subcutaneous tumor models. Thus, ALO is first identified as a novel late-stage autophagy inhibitor that triggers tumor cell death by targeting VPS4A.


Asunto(s)
Autofagosomas , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Lisosomas , Quinolizidinas , Animales , Ratones , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Humanos , Autofagosomas/metabolismo , Autofagosomas/efectos de los fármacos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Lisosomas/metabolismo , Lisosomas/efectos de los fármacos , Línea Celular Tumoral , Quinolizidinas/farmacología , Modelos Animales de Enfermedad , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/genética , Progresión de la Enfermedad , Proliferación Celular/efectos de los fármacos , Autofagia/efectos de los fármacos , Apoptosis/efectos de los fármacos
8.
Dev Cell ; 58(23): 2746-2760.e5, 2023 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-37683632

RESUMEN

The sequence of morphological intermediates that leads to mammalian autophagosome formation and closure is a crucial yet poorly understood issue. Previous studies have shown that yeast autophagosomes evolve from cup-shaped phagophores with only one closure point, and mammalian studies have inferred that mammalian phagophores also have single openings. Our superresolution microscopy studies in different human cell lines in conditions of basal and nutrient-deprivation-induced autophagy identified autophagosome precursors with multifocal origins that evolved into unexpected finger-like phagophores with multiple openings before becoming more spherical structures. Compatible phagophore structures were observed with whole-mount and conventional electron microscopy. This sequence of events was visualized using advanced SIM2 superresolution live microscopy. The finger-shaped phagophore apertures remained open when ESCRT function was compromised. The efficient closure of autophagic structures is important for their release from the recycling endosome. This has important implications for understanding how autophagosomes form and capture various cargoes.


Asunto(s)
Autofagosomas , Autofagia , Animales , Humanos , Endosomas/metabolismo , Línea Celular , Fagocitosis , Mamíferos
9.
Am J Chin Med ; 50(6): 1599-1615, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35786171

RESUMEN

Improving autophagy-lysosome fusion has been considered a key method in the treatment of Alzheimer's disease (AD). Cornel iridoid glycoside (CIG) is extracted from Cornus officinalis and has been shown to promote the clearance of tau oligomers via the autophagy pathway. However, the mechanisms of CIG on autophagy deficits are not understood. Here, we found autophagy deficit and tau aggregation in the brains of P301S tau transgenic mice and MAPT cells edited using CRISPR-Cas9 technology. CIG decreased tau aggregation and alleviated autophagic markers involving the JNK/Beclin-1 signaling pathway which demonstrated CIG that might enhance lysosome formation by upregulating ATPase Vps4A expression. Knocking down VPS4A increased autophagosome accumulation and attenuated the effect of CIG on p62. In addition, CIG had no effect on tau oligomers but still inhibited the level of tau monomer in VPS4A knockout cells. The effective component (Sweroside, SWE) of CIG attenuated tau oligomers accumulation and increased Vps4A level but not CHMP2B. SWE could not change the level of tau oligomers in VPS4A knockout cells. In conclusion, CIG suppressed autophagosome accumulation by regulating the ATPase Vps4A/JNK. SWE is a core of active factors of CIG in Vps4A regulation. These findings suggest CIG may be a potential drug in AD treatment.


Asunto(s)
Enfermedad de Alzheimer , Autofagosomas , Adenosina Trifosfatasas , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Autofagosomas/metabolismo , Autofagia/genética , Glicósidos Iridoides/farmacología , Iridoides/farmacología , Ratones
10.
Cell Rep ; 33(11): 108493, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33326793

RESUMEN

Few therapies target the loss of tumor suppressor genes in cancer. We examine CRISPR-SpCas9 and RNA-interference loss-of-function screens to identify new therapeutic targets associated with genomic loss of tumor suppressor genes. The endosomal sorting complexes required for transport (ESCRT) ATPases VPS4A and VPS4B score as strong synthetic lethal dependencies. VPS4A is essential in cancers harboring loss of VPS4B adjacent to SMAD4 on chromosome 18q and VPS4B is required in tumors with co-deletion of VPS4A and CDH1 (E-cadherin) on chromosome 16q. We demonstrate that more than 30% of cancers selectively require VPS4A or VPS4B. VPS4A suppression in VPS4B-deficient cells selectively leads to ESCRT-III filament accumulation, cytokinesis defects, nuclear deformation, G2/M arrest, apoptosis, and potent tumor regression. CRISPR-SpCas9 screening and integrative genomic analysis reveal other ESCRT members, regulators of abscission, and interferon signaling as modifiers of VPS4A dependency. We describe a compendium of synthetic lethal vulnerabilities and nominate VPS4A and VPS4B as high-priority therapeutic targets for cancers with 18q or 16q loss.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Neoplasias/genética , ATPasas de Translocación de Protón Vacuolares/metabolismo , Línea Celular Tumoral , Humanos
11.
Brain Res ; 1706: 224-236, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30414727

RESUMEN

Charged multivesicular body protein 2b (CHMP2B) is a subunit of the endosomal sorting complex required for transport (ESCRT)-III that mediates scission of budded membranes. Neurons with CHMP2B-positive granulovacuolar inclusions in the cytoplasm are much more frequent in hippocampi of cases with Alzheimer's disease when compared with controls. We analyzed immunolabeled brain sections from tau-transgenic mice, APP-transgenic mice, non-transgenic mice, and human hippocampi to investigate the relation between CHMP2B and tau and plaque pathology that are major histopathological features of Alzheimer's disease. Neurons undergoing granulovacuolar degeneration (GVD) were found in human hippocampi and old tau-trangenic mice but not in the APP-transgenic strains. 57% of neurons with GVD displayed GVD-granules double-labeled for CHMP2B and the GVD-marker casein kinase 1δ in 24 months-old tau-transgenic mice and 5.7% of neurons with tau hyper-phosphorylated at Thr212 and Ser214 (immunoreactive with antibody AT100) displayed CHMP2B-positive GVD-granules, in human hippocampi it was 100% and 46% respectively. The number of neurons with GVD-inclusions increased in tau-transgenic mice with the number of AT100-positive neurons, suggesting a link between tau-pathology and GVD. GVD-granules in human hippocampi also displayed immunoreactivity for Vps4a, another protein component of ESCRT-III. In cases with aging-related tau astrogliopathy (ARTAG), astrocytes containing hyper-phosphorylated tau immunoreactive with antibody AT8 displayed strong CHMP2B immunoreactivity. The results suggest dysregulation of CHMP2B together with tau-pathology and possibly a disturbance of the regulation of vesicular compartments. The absence of combined Aß- and tau-associated pathology in the transgenic mice may account for the difference in CHMP2B-immunoreactivity between the transgenic mice and human hippocampus.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Cuerpos Multivesiculares/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas tau/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Biomarcadores/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Cuerpos de Inclusión/metabolismo , Cuerpos de Inclusión/patología , Masculino , Ratones , Ratones Transgénicos , Cuerpos Multivesiculares/metabolismo , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Neuronas/metabolismo , Neuronas/patología , Fosforilación
12.
Methods Enzymol ; 535: 25-38, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24377915

RESUMEN

While Ras GTPases are best known for mediating growth factor signaling on the plasma membrane, these proteins also have surprisingly complex activities in the endosome. Assisted by a method called bimolecular fluorescent complementation (BiFC), which can detect weak and transient protein-protein interactions and reveal where the binding takes place in live cells, we have identified three effectors, Cdc42, CHMP6, and VPS4A that interact with Ras proteins in endosomes. These effectors are all necessary for Ras-induced transformation, suggesting that for Ras proteins to efficiently induce tumor formation, they must also activate effectors in cytoplasm, such as those in endosomes. Here, we describe how BiFC can be used to detect and screen for Ras effectors and for readily revealing where in the cell the binding occurs.


Asunto(s)
Endosomas/metabolismo , Mapeo de Interacción de Proteínas/métodos , Transducción de Señal , Proteínas ras/metabolismo , Animales , Proteínas Bacterianas/biosíntesis , Línea Celular Tumoral , Biblioteca de Genes , Células HEK293 , Humanos , Proteínas Luminiscentes/biosíntesis , Ratones , Microscopía Fluorescente , Células 3T3 NIH , Unión Proteica , Transporte de Proteínas , Proteínas Recombinantes de Fusión/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA