Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 528
Filtrar
1.
Protein Cell ; 13(1): 6-25, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33306168

RESUMEN

The serine/threonine p21-activated kinases (PAKs), as main effectors of the Rho GTPases Cdc42 and Rac, represent a group of important molecular switches linking the complex cytoskeletal networks to broad neural activity. PAKs show wide expression in the brain, but they differ in specific cell types, brain regions, and developmental stages. PAKs play an essential and differential role in controlling neural cytoskeletal remodeling and are related to the development and fate of neurons as well as the structural and functional plasticity of dendritic spines. PAK-mediated actin signaling and interacting functional networks represent a common pathway frequently affected in multiple neurodevelopmental and neurodegenerative disorders. Considering specific small-molecule agonists and inhibitors for PAKs have been developed in cancer treatment, comprehensive knowledge about the role of PAKs in neural cytoskeletal remodeling will promote our understanding of the complex mechanisms underlying neurological diseases, which may also represent potential therapeutic targets of these diseases.


Asunto(s)
Citoesqueleto/enzimología , Enfermedades del Sistema Nervioso/enzimología , Neuronas/enzimología , Transducción de Señal , Quinasas p21 Activadas/metabolismo , Animales , Citoesqueleto/genética , Humanos , Enfermedades del Sistema Nervioso/genética , Quinasas p21 Activadas/genética
2.
Small GTPases ; 12(5-6): 416-428, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33985411

RESUMEN

Epithelial folding is a common means to execute morphogenetic movements. The gastrulating Drosophila embryo offers many examples of epithelial folding events, including the ventral, cephalic, and dorsal furrows. Each of these folding events is associated with changes in intracellular contractility and/or cytoskeleton structures that autonomously promote epithelial folding. Here, we review accumulating evidence that suggests the progression and final form of ventral, cephalic, and dorsal furrows are also influenced by the behaviour of cells neighbouring these folds. We further discuss the prevalence and importance of junctional rearrangements during epithelial folding events, suggesting adherens junction components are prime candidates to modulate the transmission of the intercellular forces that influence folding events. Finally, we discuss how recently developed methods that enable precise spatial and/or temporal control of protein activity allow direct testing of molecular models of morphogenesis in vivo.


Asunto(s)
Citoesqueleto/fisiología , Proteínas de Drosophila/metabolismo , Drosophila/fisiología , Embrión no Mamífero/fisiología , Células Epiteliales/fisiología , Proteínas de Unión al GTP Monoméricas/metabolismo , Morfogénesis , Animales , Citoesqueleto/enzimología , Drosophila/enzimología , Embrión no Mamífero/citología , Embrión no Mamífero/enzimología , Células Epiteliales/enzimología , Microtúbulos/enzimología , Microtúbulos/fisiología
4.
Small GTPases ; 12(5-6): 399-415, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33570449

RESUMEN

Megakaryocytes (MKs) are the bone marrow (BM) cells that generate blood platelets by a process that requires: i) polyploidization responsible for the increased MK size and ii) cytoplasmic organization leading to extension of long pseudopods, called proplatelets, through the endothelial barrier to allow platelet release into blood. Low level of localized RHOA activation prevents actomyosin accumulation at the cleavage furrow and participates in MK polyploidization. In the platelet production, RHOA and CDC42 play opposite, but complementary roles. RHOA inhibits both proplatelet formation and MK exit from BM, whereas CDC42 drives the development of the demarcation membranes and MK migration in BM. Moreover, the RhoA or Cdc42 MK specific knock-out in mice and the genetic alterations in their down-stream effectors in human induce a thrombocytopenia demonstrating their key roles in platelet production. A better knowledge of Rho-GTPase signalling is thus necessary to develop therapies for diseases associated with platelet production defects.Abbreviations: AKT: Protein Kinase BARHGEF2: Rho/Rac Guanine Nucleotide Exchange Factor 2ARP2/3: Actin related protein 2/3BM: Bone marrowCDC42: Cell division control protein 42 homologCFU-MK: Colony-forming-unit megakaryocyteCIP4: Cdc42-interacting protein 4mDIA: DiaphanousDIAPH1; Protein diaphanous homolog 1ECT2: Epithelial Cell Transforming Sequence 2FLNA: Filamin AGAP: GTPase-activating proteins or GTPase-accelerating proteinsGDI: GDP Dissociation InhibitorGEF: Guanine nucleotide exchange factorHDAC: Histone deacetylaseLIMK: LIM KinaseMAL: Megakaryoblastic leukaemiaMARCKS: Myristoylated alanine-rich C-kinase substrateMKL: Megakaryoblastic leukaemiaMLC: Myosin light chainMRTF: Myocardin Related Transcription FactorOTT: One-Twenty Two ProteinPACSIN2: Protein Kinase C And Casein Kinase Substrate In Neurons 2PAK: P21-Activated KinasePDK: Pyruvate Dehydrogenase kinasePI3K: Phosphoinositide 3-kinasePKC: Protein kinase CPTPRJ: Protein tyrosine phosphatase receptor type JRAC: Ras-related C3 botulinum toxin substrate 1RBM15: RNA Binding Motif Protein 15RHO: Ras homologousROCK: Rho-associated protein kinaseSCAR: Suppressor of cAMP receptorSRF: Serum response factorSRC: SarcTAZ: Transcriptional coactivator with PDZ motifTUBB1: Tubulin ß1VEGF: Vascular endothelial growth factorWAS: Wiskott Aldrich syndromeWASP: Wiskott Aldrich syndrome proteinWAVE: WASP-family verprolin-homologous proteinWIP: WASP-interacting proteinYAP: Yes-associated protein.


Asunto(s)
Plaquetas/fisiología , Citoesqueleto/fisiología , Megacariocitos/fisiología , Proteínas de Unión al GTP rho/metabolismo , Animales , Plaquetas/enzimología , Citoesqueleto/enzimología , Humanos , Megacariocitos/enzimología , Transducción de Señal
5.
Adv Biol Regul ; 80: 100769, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33307285

RESUMEN

Protein kinase C α (PKCα) is a ubiquitously expressed member of the PKC family of serine/threonine kinases with diverse functions in normal and neoplastic cells. Early studies identified anti-proliferative and differentiation-inducing functions for PKCα in some normal tissues (e.g., regenerating epithelia) and pro-proliferative effects in others (e.g., cells of the hematopoietic system, smooth muscle cells). Additional well documented roles of PKCα signaling in normal cells include regulation of the cytoskeleton, cell adhesion, and cell migration, and PKCα can function as a survival factor in many contexts. While a majority of tumors lose expression of PKCα, others display aberrant overexpression of the enzyme. Cancer-related mutations in PKCα are uncommon, but rare examples of driver mutations have been detected in certain cancer types (e. g., choroid gliomas). Here we review the role of PKCα in various cancers, describe mechanisms by which PKCα affects cancer-related cell functions, and discuss how the diverse functions of PKCα contribute to tumor suppressive and tumor promoting activities of the enzyme. We end the discussion by addressing mutations and expression of PKCα in tumors and the clinical relevance of these findings.


Asunto(s)
Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Proteína Quinasa C-alfa/genética , Transducción de Señal/genética , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Citoesqueleto/enzimología , Citoesqueleto/patología , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Mutación , Metástasis de la Neoplasia , Neoplasias/clasificación , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Oligonucleótidos Fosforotioatos/uso terapéutico , Proteína Quinasa C-alfa/metabolismo
6.
J Genet Genomics ; 47(6): 321-331, 2020 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-32912804

RESUMEN

In 2010, cytidine 5'-triphosphate synthase (CTPS) was reported to form the filamentous or serpentine structure in Drosophila, which we termed the cytoophidium. In the last decade, CTPS filaments/cytoophidia have been found in bacteria, budding yeast, human cells, mice, fission yeast, plants, and archaea, indicating that this mechanism is highly conserved in evolution. In addition to CTPS, other metabolic enzymes have been identified to have the characteristics of forming cytoophidia or similar advanced structures, demonstrating that this is a basic strategy of cells. Nevertheless, our understanding of the physiological function of the cytoophidium remains incomplete and elusive. Here, we took the larva of Drosophila melanogaster as a model to systematically describe the localization and distribution of cytoophidia in different tissues during larval development. We found that the distribution pattern of CTPS cytoophidia is dynamic and heterogenic in larval tissues. Our study provides a road map for further understanding of the function and regulatory mechanism of cytoophidia.


Asunto(s)
Ligasas de Carbono-Nitrógeno/genética , Citoesqueleto/genética , Drosophila melanogaster/genética , Animales , Citidina Trifosfato/genética , Citoesqueleto/enzimología , Drosophila melanogaster/enzimología , Humanos , Larva/enzimología , Larva/genética , Linfa/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 40(10): 2391-2403, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32787521

RESUMEN

OBJECTIVE: Reelin, a secreted glycoprotein, was originally identified in the central nervous system, where it plays an important role in brain development and maintenance. In the cardiovascular system, reelin plays a role in atherosclerosis by enhancing vascular inflammation and in arterial thrombosis by promoting platelet adhesion, activation, and thrombus formation via APP (amyloid precursor protein) and GP (glycoprotein) Ib. However, the role of reelin in hemostasis and arterial thrombosis is not fully understood to date. Approach and Results: In the present study, we analyzed the importance of reelin for cytoskeletal reorganization of platelets and thrombus formation in more detail. Platelets release reelin to amplify alphaIIb beta3 integrin outside-in signaling by promoting platelet adhesion, cytoskeletal reorganization, and clot retraction via activation of Rho GTPases RAC1 (Ras-related C3 botulinum toxin substrate) and RhoA (Ras homolog family member A). Reelin interacts with the collagen receptor GP (glycoprotein) VI with subnanomolar affinity, induces tyrosine phosphorylation in a GPVI-dependent manner, and supports platelet binding to collagen and GPVI-dependent RAC1 activation, PLC gamma 2 (1-phosphatidylinositol-4,5-bisphosphate phosphodiesterase gamma-2) phosphorylation, platelet activation, and aggregation. When GPVI was deleted from the platelet surface by antibody treatment in reelin-deficient mice, thrombus formation was completely abolished after injury of the carotid artery while being only reduced in either GPVI-depleted or reelin-deficient mice. CONCLUSIONS: Our study identified a novel signaling pathway that involves reelin-induced GPVI activation and alphaIIb beta3 integrin outside-in signaling in platelets. Loss of both, GPVI and reelin, completely prevents stable arterial thrombus formation in vivo suggesting that inhibiting reelin-platelet-interaction might represent a novel strategy to avoid arterial thrombosis in cardiovascular disease.


Asunto(s)
Plaquetas/enzimología , Traumatismos de las Arterias Carótidas/enzimología , Moléculas de Adhesión Celular Neuronal/sangre , Proteínas de la Matriz Extracelular/sangre , Proteínas del Tejido Nervioso/sangre , Neuropéptidos/sangre , Fosfolipasa C gamma/sangre , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Serina Endopeptidasas/sangre , Trombosis/enzimología , Proteína de Unión al GTP rac1/sangre , Proteína de Unión al GTP rhoA/sangre , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Coagulación Sanguínea , Traumatismos de las Arterias Carótidas/sangre , Traumatismos de las Arterias Carótidas/etiología , Moléculas de Adhesión Celular Neuronal/deficiencia , Moléculas de Adhesión Celular Neuronal/genética , Retracción del Coagulo , Citoesqueleto/enzimología , Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/deficiencia , Proteínas de la Matriz Extracelular/genética , Ratones de la Cepa 129 , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Activación Plaquetaria , Proteína Reelina , Serina Endopeptidasas/deficiencia , Serina Endopeptidasas/genética , Transducción de Señal , Trombosis/sangre , Trombosis/etiología
8.
J Genet Genomics ; 47(4): 213-223, 2020 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-32507415

RESUMEN

CTP synthase (CTPS) is an important metabolic enzyme that catalyzes the rate-limiting reaction of nucleotide CTP de novo synthesis. Since 2010, a series of studies have demonstrated that CTPS can form filamentous structures in bacteria and eukaryotes, which are termed cytoophidia. However, it is unknown whether cytoophidia exist in the third domain of life, archaea. Using Haloarcula hispanica as a model system, here we demonstrate that CTPS forms distinct intracellular compartments in archaea. Under stimulated emission depletion microscopy, we find that the structures of H. hispanica CTPS are elongated, similar to cytoophidia in bacteria and eukaryotes. When Haloarcula cells are cultured in low-salt medium, the occurrence of cytoophidia increases dramatically. In addition, treatment of H. hispanica with a glutamine analog or overexpression of CTPS can promote cytoophidium assembly. Our study reveals that CTPS can form cytoophidia in all three domains of life, suggesting that forming cytoophidia is an ancient property of CTPS.


Asunto(s)
Ligasas de Carbono-Nitrógeno/genética , Citoesqueleto/enzimología , Haloarcula/enzimología , Archaea/enzimología , Archaea/metabolismo , Ligasas de Carbono-Nitrógeno/metabolismo , Citoesqueleto/genética , Citoesqueleto/metabolismo , Regulación de la Expresión Génica Arqueal/efectos de los fármacos , Glutamina/metabolismo , Glutamina/farmacología , Haloarcula/genética
9.
Pharmacol Res ; 159: 104995, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32534100

RESUMEN

The principle of mechanopharmacology of airway smooth muscle (ASM) is based on the premise that physical agitation, such as pressure oscillation applied to an airway, is able to induce bronchodilation by reducing contractility and softening the cytoskeleton of ASM. Although the underlying mechanism is not entirely clear, there is evidence to suggest that large-amplitude stretches are able to disrupt the actomyosin interaction in the crossbridge cycle and weaken the cytoskeleton in ASM cells. Rho-kinase is known to enhance force generation and strengthen structural integrity of the cytoskeleton during smooth muscle activation and plays a key role in the maintenance of force during prolonged muscle contractions. Synergy in relaxation has been observed when the muscle is subject to oscillatory length change while Rho-kinase is pharmacologically inhibited. In this review, inhibition of Rho-kinase coupled to therapeutic pressure oscillation applied to the airways is explored as a combination treatment for asthma.


Asunto(s)
Antiasmáticos/uso terapéutico , Asma/tratamiento farmacológico , Broncoconstricción/efectos de los fármacos , Broncodilatadores/uso terapéutico , Pulmón/efectos de los fármacos , Mecanotransducción Celular/efectos de los fármacos , Músculo Liso/efectos de los fármacos , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinasas Asociadas a rho/antagonistas & inhibidores , Animales , Asma/enzimología , Asma/fisiopatología , Citoesqueleto/efectos de los fármacos , Citoesqueleto/enzimología , Humanos , Pulmón/enzimología , Pulmón/fisiopatología , Terapia Molecular Dirigida , Músculo Liso/enzimología , Músculo Liso/fisiopatología , Quinasas Asociadas a rho/metabolismo
10.
J Cardiovasc Pharmacol ; 76(1): 86-93, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32324654

RESUMEN

Aortic dissection (AD) is a serious condition and a health issue on a global scale. ß-Aminopropionitrile-induced AD in mice is similar to the pathogenesis of AD in humans. Resveratrol (RSV) is a natural polyphenolic substance that provides anti-inflammatory and cardiovascular effects, but the role of RSV in AD is unclear. In this study, we investigated the effects and mechanisms of RSV on ß-aminopropionitrile-induced AD in mice. Our results indicate that RSV can prevent the occurrence of AD. More meaningfully, we found that the protective effect comprises an increase in sirtuin 1 (SIRT1) expression in endothelial cells for the reconstruction of their structure, reducing the recruitment of inflammatory cells by endothelial cells and inhibiting the inflammation response, thereby suppressing the occurrence of AD.


Asunto(s)
Antiinflamatorios/farmacología , Aorta/efectos de los fármacos , Aneurisma de la Aorta/prevención & control , Disección Aórtica/prevención & control , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Resveratrol/farmacología , Sirtuina 1/metabolismo , Aminopropionitrilo , Disección Aórtica/inducido químicamente , Disección Aórtica/enzimología , Disección Aórtica/patología , Animales , Aorta/enzimología , Aorta/patología , Aneurisma de la Aorta/inducido químicamente , Aneurisma de la Aorta/enzimología , Aneurisma de la Aorta/patología , Adhesión Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Citoesqueleto/enzimología , Citoesqueleto/patología , Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Masculino , Ratones Endogámicos C57BL , Transducción de Señal , Células THP-1
11.
Fungal Biol ; 124(1): 15-23, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31892373

RESUMEN

Metabolons are dynamic associations of enzymes catalyzing consecutive reactions within a given pathway. Association results in enzyme stabilization and increased metabolic efficiency. Metabolons may use cytoskeletal elements, membranes and membrane proteins as scaffolds. The effects of glucose withdrawal on a putative glycolytic metabolon/F-actin system were evaluated in three Saccharomyces cerevisiae strains: a WT and two different obligate fermentative (OxPhos-deficient) strains, which obtained most ATP from glycolysis. Carbon source withdrawal led to inhibition of fermentation, decrease in ATP concentration and dissociation of glycolytic enzymes from F-actin. Depending on the strain, inactivation/reactivation transitions of fermentation took place in seconds. In addition, when ATP was very low, green fluorescent protein-labeled F-actin reorganized from highly dynamic patches to large, non-motile actin bodies containing proteins and enzymes. Glucose addition restored fermentation and cytoskeleton dynamics, suggesting that in addition to ATP concentration, at least in one of the tested strains, metabolon assembly/disassembly is a factor in the control of the rate of fermentation.


Asunto(s)
Citoesqueleto de Actina/ultraestructura , Actinas/metabolismo , Citoesqueleto/enzimología , Glucólisis , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Citoesqueleto/ultraestructura , Fermentación , Glucosa/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Proteínas de Microfilamentos/metabolismo , Fosforilación Oxidativa , Fosfoglicerato Quinasa/metabolismo , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/ultraestructura
12.
Mol Cell Biochem ; 456(1-2): 167-178, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30739223

RESUMEN

Exchange protein directly activated by cAMP (Epac) and protein kinase A are effectors for cAMP with distinct actions and regulatory mechanisms. Epac is a Rap guanine nucleotide exchange factor that activates Rap1; protein kinase C (PKC) is a major downstream target of Epac-Rap1 signaling that has been implicated in a variety of pathophysiological processes, including cardiac hypertrophy, cancer, and nociceptor sensitization leading to chronic pain. Despite the implication of both Epac and PKC in these processes, few downstream targets of Epac-PKC signaling have been identified. This study characterized the regulation of PKC activity downstream of Epac activation. Using an antibody that recognizes phospho-serine residues within the consensus sequence phosphorylated by PKC, we analyzed the 1-dimensional banding profile of PKC substrate protein phosphorylation from the Neuro2A mouse neuroblastoma cell line. Activation of Epac either indirectly by prostaglandin PGE2, or directly by 8-pCPT-2-O-Me-cAMP-AM (8pCpt), produced distinct PKC phospho-substrate protein bands that were suppressed by co-administration of the Epac inhibitor ESI09. Different PKC isoforms contributed to the induction of individual phospho-substrate bands, as determined using isoform-selective PKC inhibitors. Moreover, the banding profile after Epac activation was altered by disruption of the cytoskeleton, suggesting that the orchestration of Epac-dependent PKC signaling is regulated in part by interactions with the cytoskeleton. The approach described here provides an effective means to characterize Epac-dependent PKC activity.


Asunto(s)
Citoesqueleto/enzimología , Dinoprostona/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteína Quinasa C/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rap1/metabolismo , Animales , Línea Celular Tumoral , Citoesqueleto/genética , Factores de Intercambio de Guanina Nucleótido/genética , Ratones , Fosforilación , Proteína Quinasa C/genética , Proteínas de Unión al GTP rap1/genética
13.
Vascul Pharmacol ; 111: 62-70, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30287213

RESUMEN

INTRODUCTION: Platelets activation/aggregation with subsequent thrombus formation is the main event in the pathophysiology of acute coronary syndrome. Once activated, platelets show an extensive cytoskeleton rearrangement that leads to recruitment of additional platelets to finally cause haemostatic plug formation. Thus, the cytoskeleton plays a pivotal role in this phenomenon. Colchicine (COLC) is an anti-inflammatory drug proven to reduce major cardiovascular events in patients with coronary artery disease. The molecular mechanisms by which COLC exerts these protective effects remain partially still unknown. Since COLC causes disruption of tubulin, a component of cell cytoskeleton, we investigated whether this drug might interfere with platelet aggregation by acting on cytoskeleton rearrangement. METHODS AND RESULTS: Platelets isolated from healthy volunteers were activated with Adenosine Diphosphate (ADP, 20 µM) Collagen (COLL, 60 µg/ml) and Thrombin Activating Receptor Peptide (TRAP 25 µM) with/without COLC 10 µM pretreatment. After stimulus, aggregation was measured by light aggregometry overtime. Microtubules structure was assessed by immunohistochemistry and key proteins involved in regulation of actin-filament assembly and contractility such as Myosin Phosphatase Targeting subunit (MYPT), LIM domain kinase 1(LIMK1) and cofilin were evaluated by Western Blot analysis. Colchicine pretreatment significantly blunted ADP/COLL/TRAP-induced platelet aggregation (up to 40%). COLC effects appeared mediated by microtubules depolymerization and cytoskeleton disarrangement associated to inactivation of MYPT and LIMK1 that finally interfered with cofilin activity. CONCLUSIONS: Our data indicate that colchicine exerts anti-platelet effects in vitro via inhibition of key proteins involved in cytoskeleton rearrangement, suggesting that its beneficial cardiovascular properties may be due, at least in part, to an inhibitory effect of platelet activity.


Asunto(s)
Plaquetas/efectos de los fármacos , Colchicina/farmacología , Citoesqueleto/efectos de los fármacos , Quinasas Lim/antagonistas & inhibidores , Inhibidores de Agregación Plaquetaria/farmacología , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Plaquetas/enzimología , Cofilina 1/metabolismo , Citoesqueleto/enzimología , Humanos , Quinasas Lim/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación , Transducción de Señal/efectos de los fármacos
14.
Essays Biochem ; 62(4): 501-514, 2018 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-30315098

RESUMEN

An unprecedented opportunity to integrate ~100 years of meticulous in vitro biomolecular research is currently provided in the light of recent advances in methods to visualize closer-to-native architectures of biomolecular machines, and metabolic enzymes in particular. Traditional views of enzymes, namely biomolecular machines, only partially explain their role, organization and kinetics in the cellular milieu. Enzymes self- or hetero-associate, form fibers, may bind to membranes or cytoskeletal elements, have regulatory roles, associate into higher order assemblies (metabolons) or even actively participate in phase-separated membraneless organelles, and all the above in a transient, temporal and spatial manner in response to environmental changes or structural/functional changes of their assemblies. Here, we focus on traditional and emerging concepts in cellular biochemistry and discuss new opportunities in bridging structural, molecular and cellular analyses for metabolic pathways, accumulated over the years, highlighting functional aspects of enzymatic complexes discussed across different levels of spatial resolution.


Asunto(s)
Enzimas/metabolismo , Biología de Sistemas , Fenómenos Bioquímicos , Dominio Catalítico , Citoesqueleto/enzimología , Cinética , Redes y Vías Metabólicas , Polimerizacion , Especificidad por Sustrato
15.
Methods Mol Biol ; 1821: 235-246, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30062416

RESUMEN

B-cell migration and adhesion are critical to form a germinal center response, the site for B-cell production of high-affinity antibodies. Here, we describe two assays that can be used to examine B-cell cytoskeletal responses needed during the germinal center response: B-cell spreading and homotypic adhesion. Spreading of B cells is dependent on Cdc42, while Rac1 and Rac2 are necessary for homotypic adhesion. These in vitro assays can be used to examine functional responses of B cells mediated by the cell cytoskeleton, for example when comparing B cells from different gene knockout animals.


Asunto(s)
Linfocitos B/enzimología , Citoesqueleto/enzimología , Centro Germinal/enzimología , Neuropéptidos/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Linfocitos B/citología , Adhesión Celular/fisiología , Citoesqueleto/genética , Centro Germinal/citología , Ratones , Ratones Noqueados , Neuropéptidos/genética , Proteína de Unión al GTP cdc42/genética , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1/genética , Proteína RCA2 de Unión a GTP
16.
Cell Rep ; 24(8): 2029-2041, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30134165

RESUMEN

Synaptic cytoskeleton dysfunction represents a common pathogenesis in neurodevelopmental disorders, such as autism spectrum disorder (ASD). The serine/threonine kinase PAK2 is a critical regulator of cytoskeleton dynamics. However, its function within the central nervous system and its role in ASD pathogenesis remain undefined. Here, we found that Pak2 haploinsufficiency resulted in markedly decreased synapse densities, defective long-term potentiation, and autism-related behaviors in mice. Phosphorylation levels of key actin regulators LIMK1 and cofilin, together with their mediated actin polymerization, were reduced in Pak2+/-mice. We identified one de novo PAK2 nonsense mutation that impaired PAK2 function in vitro and in vivo and four de novo copy-number deletions containing PAK2 in large cohorts of patients with ASD. PAK2 deficiency extensively perturbed functional networks associated with ASD by regulating actin cytoskeleton dynamics. Our genetic and functional results demonstrate a critical role of PAK2 in brain development and autism pathogenesis.


Asunto(s)
Trastorno del Espectro Autista/genética , Emparejamiento Cromosómico/genética , Quinasas p21 Activadas/genética , Actinas/genética , Actinas/metabolismo , Animales , Trastorno del Espectro Autista/enzimología , Citoesqueleto/enzimología , Citoesqueleto/genética , Citoesqueleto/patología , Células HEK293 , Haploinsuficiencia , Humanos , Potenciación a Largo Plazo , Masculino , Ratones , Mutación Missense , Conducta Social , Conducta Estereotipada , Quinasas p21 Activadas/metabolismo
17.
Cell Rep ; 24(4): 895-908.e6, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-30044986

RESUMEN

The role of primary cilia in mechanosensation is essential in endothelial cell (EC) shear responsiveness. Here, we find that venous, capillary, and progenitor ECs respond to shear stress in vitro in a cilia-dependent manner. We then demonstrate that primary cilia assembly in human induced pluripotent stem cell (hiPSC)-derived ECs varies between different cell lines with marginal influence of differentiation protocol. hiPSC-derived ECs lacking cilia do not align to shear stress, lack stress fiber assembly, have uncoordinated migration during wound closure in vitro, and have aberrant calcium influx upon shear exposure. Transcriptional analysis reveals variation in regulatory genes involved in ciliogenesis among different hiPSC-derived ECs. Moreover, inhibition of histone deacetylase 6 (HDAC6) activity in hiPSC-ECs lacking cilia rescues cilia formation and restores mechanical sensing. Taken together, these results show the importance of primary cilia in hiPSC-EC mechano-responsiveness and its modulation through HDAC6 activity varies among hiPSC-ECs.


Asunto(s)
Cilios/enzimología , Células Endoteliales/enzimología , Histona Desacetilasa 6/metabolismo , Células Madre Pluripotentes/enzimología , Calcio/metabolismo , Movimiento Celular/fisiología , Citoesqueleto/enzimología , Células Endoteliales/citología , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Mecanotransducción Celular , Técnicas Analíticas Microfluídicas , Células Madre Pluripotentes/citología , Arterias Umbilicales/citología , Arterias Umbilicales/enzimología
18.
Clin Exp Med ; 18(4): 523-533, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29855744

RESUMEN

Myosin light chain kinase (MYLK) is found to catalyze the phosphorylation of myosin light chains (MLC) and regulate invasion and metastasis in some malignancies. However, there is little knowledge on the role of MYLK in hepatocellular carcinoma (HCC), and no studies have been conducted to investigate the mechanisms underlying MYLK-mediated promotion of HCC invasion and metastasis until now. In this study, we investigated the expression of MYLK in 50 pairs of human HCC and adjacent liver specimens. High MYLK expression was significantly correlated with aggressive clinicopathological features including tumor encapsulation, microvascular invasion and metastasis. In vitro assays showed that shRNA-induced MYLK knockdown significantly inhibited the wound-healing ability of HCC cells and the ability to migrate and invade through Matrigel. We next uncovered that MYLK knockdown resulted in a reduction in the number of F-actin stress fibers, disorganization of F-actin architectures and morphological alterations of HCC cells. Phosphorylated MLC, rather than total MLC, was found to be markedly reduced in response to downregulation of MYLK expression, and MYLK-regulated actin cytoskeleton through phosphorylating MLC in HCC cells. In addition, Western blotting assay revealed downregulation of the epithelial marker E-cadherin and upregulation of mesenchymal markers Vimentin, N-cadherin and Snail. Taken together, our findings indicate that MYLK promotes HCC progression by altering cytoskeleton to enhance epithelial-mesenchymal transition (EMT).


Asunto(s)
Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Transición Epitelial-Mesenquimal , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Quinasa de Cadena Ligera de Miosina/biosíntesis , Citoesqueleto/enzimología , Citoesqueleto/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Quinasa de Cadena Ligera de Miosina/genética , Metástasis de la Neoplasia
19.
Arterioscler Thromb Vasc Biol ; 37(12): 2301-2310, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29051140

RESUMEN

OBJECTIVE: VEGF (vascular endothelial growth factor-A) signaling to the endothelial cell (EC) through VEGFR2 (VEGF receptor-2) is the principal cue driving new blood vessel formation. FGD5 (faciogenital dysplasia-5)-a Rho-family guanine nucleotide exchange factor-is selectively expressed in EC. Deficiency of FGD5 is embryonically lethal in mice and perturbs angiogenesis and VEGF signal transduction. However, the mechanism of FGD5 regulation of VEGF signaling is poorly understood. APPROACH AND RESULTS: Angiogenic sprouting and EC cytoskeletal remodeling were evaluated in a 3-dimensional in vitro model. We examined the subcellular localization of FGD5 and VEGFR2 in EC by immunofluorescent staining and studied the association by immunoprecipitation. FGD5 deficiency reduced the number of angiogenic sprouts and tip cell filopodia by ≈80% and ≈70%, respectively. These defects were accompanied by downregulation of the expression of tip cell-specific markers. FGD5 inactivation led to a decrease in EC migration and early protrusion (lamellipodia) formation. In resting and VEGF-stimulated EC, FGD5 forms a complex with VEGFR2 and was enriched at the leading edge of the cell and among endosomes. FGD5 loss reduced mTORC2 (mammalian target of rapamycin complex-2)/Akt-dependent cortactin activation downstream of VEGFR2 but did not alter VEGFR2 plasma membrane expression, Y1175 phosphorylation, or endocytosis. However, FGD5 loss decreased endosomal VEGFR2 coupling to phosphoinositide-3 kinase and diverted VEGFR2 to lysosomal degradation. CONCLUSIONS: FGD5 regulates VEGFR2 retention in recycling endosomes and coupling to PI3 (phosphoinositide-3) kinase/mTORC2-dependent cytoskeletal remodeling.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Células Endoteliales de la Vena Umbilical Humana/enzimología , Neovascularización Fisiológica , Fosfatidilinositol 3-Quinasa/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Cortactina/metabolismo , Citoesqueleto/enzimología , Endosomas/enzimología , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Lisosomas/enzimología , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Unión Proteica , Proteolisis , Proteínas Proto-Oncogénicas c-akt/metabolismo , Seudópodos/enzimología , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transfección , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
20.
Toxicology ; 389: 74-84, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28743511

RESUMEN

Both RhoA/ROCK and Raf-1/CK2 pathway play essential roles in cell proliferation, apoptosis, differentiation, and multiple other common cellular functions. We previously reported that vimentin is responsible for TNF-α-induced cell apoptosis. Herein, we investigated the regulation of RhoA/ROCK and Raf-1/CK2 signaling on vimentin filaments and endothelial apoptosis mediated by TNF-α. Treatment with TNF-α significantly induced the activation of RhoA and ROCK, and the expression of ROCK1. RhoA deficiency could obviously inhibit ROCK activation and ROCK1 expression induced by TNF-α. Both RhoA deficiency and ROCK activity inhibition (Y-27632) greatly inhibited endothelial apoptosis and preserved cell viability in TNF-α-induced human umbilical vein endothelial cells (HUVECs). Also vimentin phosphorylation and the remodeling of vimentin or phospho-vimentin induced by TNF-α were obviously attenuated by RhoA suppression and ROCK inhibition. TNF-α-mediated vimentin cleavage was significantly inhibited by RhoA suppression and ROCK inhibition through decreasing the activation of caspase3 and 8. Furthermore, TNF-α treatment greatly enhanced the activation of Raf-1. Suppression of Raf-1 or CK2 by its inhibitor (GW5074 or TBB) blocked vimentin phosphorylation, remodeling and endothelial apoptosis, and preserved cell viability in TNF-α-induced HUVECs. However, Raf-1 inhibition showed no significant effect on TNF-α-induced ROCK expression and activation, suggesting that the regulation of Raf-1/CK2 signaling on vimentin was independent of ROCK. Taken together, these results indicate that both RhoA/ROCK and Raf-1/CK2 pathway are responsible for TNF-α-mediated endothelial cytotoxicity via regulating vimentin cytoskeleton.


Asunto(s)
Apoptosis/efectos de los fármacos , Quinasa de la Caseína II/metabolismo , Citoesqueleto/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/metabolismo , Factor de Necrosis Tumoral alfa/toxicidad , Vimentina/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Quinasa de la Caseína II/antagonistas & inhibidores , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citoesqueleto/enzimología , Citoesqueleto/patología , Células Endoteliales de la Vena Umbilical Humana/enzimología , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Transfección , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Quinasas Asociadas a rho/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA