Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34810254

RESUMEN

Enzymes that produce second messengers are highly regulated. Revealing the mechanisms underlying such regulation is critical to understanding both how cells achieve specific signaling outcomes and return to homeostasis following a particular stimulus. Pooled genome-wide CRISPR screens are powerful unbiased approaches to elucidate regulatory networks, their principal limitation being the choice of phenotype selection. Here, we merge advances in bioorthogonal fluorescent labeling and CRISPR screening technologies to discover regulators of phospholipase D (PLD) signaling, which generates the potent lipid second messenger phosphatidic acid. Our results reveal glycogen synthase kinase 3 as a positive regulator of protein kinase C and PLD signaling. More generally, this work demonstrates how bioorthogonal, activity-based fluorescent tagging can expand the power of CRISPR screening to uncover mechanisms regulating specific enzyme-driven signaling pathways in mammalian cells.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Fosfolipasa D/metabolismo , Proteína Quinasa C-alfa/metabolismo , Fenómenos Biológicos , Sistemas CRISPR-Cas/genética , Química Clic/métodos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Glucógeno Sintasa Quinasa 3/fisiología , Células HEK293 , Humanos , Células K562 , Ácidos Fosfatidicos/metabolismo , Fosfolipasa D/fisiología , Proteína Quinasa C-alfa/fisiología , Sistemas de Mensajero Secundario , Transducción de Señal
2.
Biochem J ; 478(4): 839-854, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33528492

RESUMEN

Aberrant differentiation of keratinocytes disrupts the skin barrier and causes a series of skin diseases. However, the molecular basis of keratinocyte differentiation is still poorly understood. In the present study, we examined the expression of C7ORF41 using tissue microarrays by immunohistochemistry and found that C7ORF41 is specifically expressed in the basal layers of skin epithelium and its expression is gradually decreased during keratinocytes differentiation. Importantly, we corroborated the pivotal role of C7ORF41 during keratinocyte differentiation by C7ORF41 knockdown or overexpression in TPA-induced Hacat keratinocytes. Mechanismly, we first demonstrated that C7ORF41 inhibited keratinocyte differentiation mainly through formatting a complex with IKKα in the cytoplasm, which thus blocked the nuclear translocation of IKKα. Furthermore, we also demonstrated that inhibiting the PKCα/ERK signaling pathway reversed the reduction in C7ORF41 in TPA-induced keratinocytes, indicating that C7ORF41 expression could be regulated by upstream PKCα/ERK signaling pathway during keratinocyte differentiation. Collectively, our study uncovers a novel regulatory network PKCα/ERK/C7ORF41/IKKα during keratinocyte differentiation, which provides potential therapeutic targets for skin diseases.


Asunto(s)
Epidermis/metabolismo , Quinasa I-kappa B/metabolismo , Queratinocitos/citología , Transducción de Señal/fisiología , Transporte Activo de Núcleo Celular , Diferenciación Celular , Línea Celular Transformada , Regulación hacia Abajo , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Queratinocitos/metabolismo , Proteína Quinasa C-alfa/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología
3.
J Cancer Res Clin Oncol ; 147(2): 393-401, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33104883

RESUMEN

BACKGROUND: Ewing's sarcoma (ES) is a kind of malignant tumor, which often occurs in the long bone, pelvis, and other bone tissues, as well as some soft tissues. It often occurs in children and adolescents, second only to osteosarcoma and rhabdomyosarcoma. In the past 30 years, little progress has been made on the genomic mechanism of ES metastasis. METHODS: The gene expression sequence of ES metastasis samples was compared with that of primary tumor samples to obtain differentially expressed genes (DEGs). Subsequently, we annotated the gene functions and enriched pathways of DEGs. Additionally, the protein and protein interaction network were constructed to screen key genes that can lead to the metastasis in ES. Then, cell and molecular biology experiments were conducted to verify the results obtained from the bioinformatics analysis. Finally, we assessed the correlation of expression between the key genes EWSR and FLI1, and conducted a survival analysis of ICAM1. RESULTS: Our study revealed 153 DEGs. Of these, 82 (53.59%) were upregulated and the remaining 71 (46.41%) were downregulated. The bioinformatics analysis showed that ICAM1 was the key gene leading to the invasion and metastasis of ES. Through cell biology and molecular biology experiments, inactivation of ICAM1 inhibited the metastasis of ES cells. The survival and correlation analyses showed that ICAM1 was a risk factor in patients with ES, and that ICAM1 expression was correlated with EWSR and FLI1 expression. CONCLUSION: Our study shows that inactivation of ICAM1 inhibits metastasis and improves the prognosis of ES. Additionally, our findings provide a better understanding of the underlying mechanisms of metastatic ES, a basis for an accurate diagnosis, and therapeutic targets for ES patients.


Asunto(s)
Neoplasias Óseas/patología , Molécula 1 de Adhesión Intercelular/fisiología , Sarcoma de Ewing/patología , Neoplasias Óseas/mortalidad , Línea Celular Tumoral , Biología Computacional , Humanos , Molécula 1 de Adhesión Intercelular/genética , Invasividad Neoplásica , Metástasis de la Neoplasia/prevención & control , Proteínas de Fusión Oncogénica/genética , Pronóstico , Proteína Quinasa C-alfa/fisiología , Sarcoma de Ewing/mortalidad , Sarcoma de Ewing/secundario
4.
Cell Signal ; 64: 109418, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31525436

RESUMEN

G Protein-activated K+ channels (GIRK) channels are inhibited by depletion of PtdIns(4,5)P2(PIP2), and/or channel phosphorylation by proteinkinase C (PKC). By using FRET-based biosensors, expressed in HEK293 cells or in atrial myocytes, we quantified receptor-specific Gq-coupled receptor (GqPCR) signalling on the level of phospholipase C (PLC) activation by monitoring PIP2-depletion and diacylglycerol (DAG) formation. Simultaneous voltage-clamp experiments on GIRK channel activity were performed as a functional readout for Gq-coupled α1B- and ET-receptor-induced signalling. GqPCR-induced fast inhibition of GIRK channel activity is mediated by depletion of PIP2, whereas phosphorylation of GIRK channels results in delayed, but effective GIRK current inhibition. We demonstrate a receptor-induced inhibitory component on GIRK activity that is independent of PIP2-depletion, but attributed to the activation of Ca2+-dependent PKC isoforms. As a novel finding, we demonstrate receptor-dependent differences in GIRK inhibition according to receptor-specific activation of the Ca2+-dependent PKC isoforms PKCα and PKCß. Pharmacological inhibition of PKCα, but not of PKCß, abolishes GIRK inhibition induced by stimulation of α1B-receptors. In contrast, ET-R-induced reduction of GIRK activity is sensitive to pharmacological block of PKCß, but not of PKCα. Coexpression of α1B-receptors (or ETB-R) and PKCα (or PKCß) in HEK 293 cells increased homologous receptor desensitization as indicated by a rapid decline of the CKAR FRET signal monitoring receptor activity. These data suggest that receptor-species dependent differences in PKC isoform activation regulate both GIRK channel activity and the strength of the receptor signal via a negative feedback mechanism.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Miocitos Cardíacos/metabolismo , Proteína Quinasa C beta/fisiología , Proteína Quinasa C-alfa/fisiología , Animales , Transferencia Resonante de Energía de Fluorescencia/métodos , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HEK293 , Atrios Cardíacos , Humanos , Ratas , Receptores Adrenérgicos alfa 1/metabolismo
5.
Nat Neurosci ; 21(8): 1027-1037, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30013171

RESUMEN

The protein kinase C (PKC) enzymes have long been established as critical for synaptic plasticity. However, it is unknown whether Ca2+-dependent PKC isozymes are activated in dendritic spines during plasticity and, if so, how this synaptic activity is encoded by PKC. Here, using newly developed, isozyme-specific sensors, we demonstrate that classical isozymes are activated to varying degrees and with distinct kinetics. PKCα is activated robustly and rapidly in stimulated spines and is the only isozyme required for structural plasticity. This specificity depends on a PDZ-binding motif present only in PKCα. The activation of PKCα during plasticity requires both NMDA receptor Ca2+ flux and autocrine brain-derived neurotrophic factor (BDNF)-TrkB signaling, two pathways that differ vastly in their spatiotemporal scales of signaling. Our results suggest that, by integrating these signals, PKCα combines a measure of recent, nearby synaptic plasticity with local synaptic input, enabling complex cellular computations such as heterosynaptic facilitation of plasticity necessary for efficient hippocampus-dependent learning.


Asunto(s)
Comunicación Autocrina/fisiología , Factor Neurotrófico Derivado del Encéfalo/fisiología , Señalización del Calcio/fisiología , Plasticidad Neuronal/fisiología , Proteína Quinasa C-alfa/fisiología , Animales , Comunicación Autocrina/genética , Factor Neurotrófico Derivado del Encéfalo/genética , Señalización del Calcio/genética , Espinas Dendríticas , Activación Enzimática , Hipocampo/fisiología , Isoenzimas , Cinética , Aprendizaje/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa C-alfa/genética , Receptores de N-Metil-D-Aspartato/metabolismo
6.
Biomed Pharmacother ; 103: 838-843, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29710499

RESUMEN

This study was designed to investigate the effects of astragalosides on cardiac diastolic function, and an emphasis was placed on the variation of the upstream molecular regulators of phospholamban. Chronic heart failure (CHF) rats were induced by ligaturing the left anterior coronary artery, and rats in the therapeutic groups were treated with either a 50 mg/kg dose of captopril, 10 mg/kg dose of astragalosides or 20 mg/kg dose of astragalosides. Four weeks after treatment, the ratio of the early and atrial peak filling velocities (E/A) and maximal slope diastolic pressure decrement (-dp/dt) both decreased in CHF rats (by 30.3% and 25.5%, respectively) and significantly increased in 20 mg/kg astragalosides and captopril-treated rats. The protein phosphatase-1 activity was lower in the 20 mg/kg astragalosides group than in the CHF group (0.22 vs 0.44, P < 0.01), and the inhibitor-1 levels in the astragalosides and captopril-treated groups were increased. Chronic heart failure increased expression of protein kinase C-α and calcium-sensing receptor, and these changes were attenuated by astragalosides therapy. Astragalosides restored the diastolic dysfunction of chronic heart failure rats, possibly by downregulation of calcium-sensing receptor and protein kinase C-α, which in turn augmented inhibitor-1 expression, reduced protein phosphatase-1 activity and increased phospholamban phosphorylation.


Asunto(s)
Presión Sanguínea/fisiología , Insuficiencia Cardíaca/tratamiento farmacológico , Proteína Quinasa C-alfa/fisiología , Proteína Fosfatasa 1/fisiología , Receptores Sensibles al Calcio/fisiología , Saponinas/farmacología , Triterpenos/farmacología , Animales , Animales Recién Nacidos , Presión Sanguínea/efectos de los fármacos , Células Cultivadas , Diástole , Relación Dosis-Respuesta a Droga , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Insuficiencia Cardíaca/fisiopatología , Masculino , Proteína Quinasa C-alfa/antagonistas & inhibidores , Proteína Fosfatasa 1/antagonistas & inhibidores , Distribución Aleatoria , Ratas , Ratas Wistar , Receptores Sensibles al Calcio/antagonistas & inhibidores , Saponinas/uso terapéutico , Triterpenos/uso terapéutico
7.
Biomol Concepts ; 8(3-4): 143-153, 2017 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-28841566

RESUMEN

Protein kinase C (PKC) comprises a family of lipid-sensitive enzymes that have been involved in a broad range of cellular functions. PKC-α is a member of classical PKC with ubiquitous expression and different cellular localization. This unique PKC isoform is activated by various signals which evoke lipid hydrolysis, after activation it interacts with various adapter proteins and is localized to specific cellular compartments where it is devised to work. The universal expression and activation by various stimuli make it a perfect player in uncountable cellular functions including differentiation, proliferation, apoptosis, cellular transformation, motility, adhesion and so on. However, these functions are not intrinsic properties of PKC-α, but depend on cell types and conditions. The activities of PKC-α are managed by the various pharmacological activators/inhibitors and antisense oligonucleotides. The aim of this review is to elaborate the structural feature, and provide an insight into the mechanism of PKC-α activation and regulation of its key biological functions in different cellular compartments to develop an effective pharmacological approach to regulate the PKC-α signal array.


Asunto(s)
Proteína Quinasa C-alfa/fisiología , Apoptosis , Adhesión Celular , Movimiento Celular , Proliferación Celular , Hidrólisis , Metabolismo de los Lípidos , Modelos Moleculares , Proteína Quinasa C-alfa/química , Proteína Quinasa C-alfa/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas , Transducción de Señal
8.
Biochim Biophys Acta Gen Subj ; 1861(8): 2039-2047, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28435021

RESUMEN

BACKGROUND: Compensation of the pancreatic ß cell functional mass in response to metabolic stress is key to the pathogenesis of Type 2 Diabetes. The mTORC2 pathway governs fuel metabolism and ß cell functional mass. It is unknown whether mTORC2 is required for regulating metabolic stress-induced ß cell compensation. METHODS: We challenged four-week-old ß-cell-specific Rictor (a key component of mTORC2)-knockout mice with a high fat diet (HFD) for 4weeks and measured metabolic and pancreatic morphological parameters. We performed ex vivo experiments to analyse ß cell insulin secretion and electrophysiology characteristics. Adenoviral-mediated overexpression and lentiviral-ShRNA-mediated knocking down proteins were applied in Min6 cells and cultured primary mouse islets. RESULTS: ßRicKO mice showed a significant glucose intolerance and a reduced plasma insulin level and an unchanged level ß cell mass versus the control mice under HFD. A HFD or palmitate treatment enhanced both glucose-induced insulin secretion (GIIS) and the PMA (phorbol 12-myristate 13-acetate)-induced insulin secretion in the control islets but not in the ßRicKO islets. The KO ß cells showed similar glucose-induced Ca2+ influx but lower membrane capacitance increments versus the control cells. The enhanced mTORC2/PKC proteins levels in the control HFD group were ablated by Rictor deletion. Replenishing PKCα by overexpression of PKCα-T638D restored the defective GIIS in ßRicKO islets. CONCLUSIONS: The mTORC2/Rictor pathway modulates ß cell compensatory GIIS under nutrient overload mediated by its phosphorylation of PKCα. GENERAL SIGNIFICANCE: This study suggests that the mTORC2/PKC pathway in ß cells is involved in the pathogenesis of T2D.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Complejos Multiproteicos/fisiología , Proteína Quinasa C-alfa/fisiología , Transducción de Señal/fisiología , Estrés Fisiológico/fisiología , Serina-Treonina Quinasas TOR/fisiología , Animales , Diabetes Mellitus Tipo 2/etiología , Dieta Alta en Grasa , Secreción de Insulina , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Acetato de Tetradecanoilforbol/farmacología
9.
J Exp Med ; 214(1): 209-226, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28028151

RESUMEN

Directional autoreactive CD4+ T cell migration into the central nervous system plays a critical role in multiple sclerosis. Recently, DOCK8 was identified as a guanine-nucleotide exchange factor (GEF) for Cdc42 activation and has been associated with human mental retardation. Little is known about whether DOCK8 is related to multiple sclerosis (MS) and how to restrict its GEF activity. Using two screening systems, we found that LRCH1 competes with Cdc42 for interaction with DOCK8 and restrains T cell migration. In response to chemokine stimulation, PKCα phosphorylates DOCK8 at its three serine sites, promoting DOCK8 separation from LRCH1 and translocation to the leading edge to guide T cell migration. Point mutations at the DOCK8 serine sites block chemokine- and PKCα-induced T cell migration. Importantly, Dock8 mutant mice or Lrch1 transgenic mice were protected from MOG (35-55) peptide-induced experimental autoimmune encephalomyelitis (EAE), whereas Lrch1-deficient mice displayed a more severe phenotype. Notably, DOCK8 expression was markedly increased in PBMCs from the acute phase of MS patients. Together, our study demonstrates LRCH1 as a novel effector to restrain PKCα-DOCK8-Cdc42 module-induced T cell migration and ameliorate EAE.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Factores de Intercambio de Guanina Nucleótido/fisiología , Proteínas de Microfilamentos/fisiología , Linfocitos T/fisiología , Proteína de Unión al GTP cdc42/fisiología , Animales , Movimiento Celular , Encefalomielitis Autoinmune Experimental/etiología , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/etiología , Fosforilación , Proteína Quinasa C-alfa/fisiología
10.
Mol Biol Cell ; 27(9): 1511-23, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26941335

RESUMEN

Tiam1 is one of the most extensively analyzed activators of the small GTPase Rac. However, fundamental aspects of its regulation are poorly understood. Here we demonstrate that Tiam1 is functionally suppressed by internal interactions and that the PAR complex participates in its full activation. The N-terminal region of Tiam1 binds to the protein-binding and catalytic domains to inhibit its localization and activation. Atypical PKCs phosphorylate Tiam1 to relieve its intramolecular interactions, and the subsequent stabilization of its interaction with PAR3 allows it to exert localized activity. By analyzing Tiam1 regulation by PAR3-aPKC within the context of PDGF signaling, we also show that PAR3 directly binds PDGF receptor ß. Thus we provide the first evidence for the negative regulation of Tiam1 by internal interactions, elucidate the nature of Tiam1 regulation by the PAR complex, and reveal a novel role for the PAR complex in PDGF signaling.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Factores de Intercambio de Guanina Nucleótido/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/fisiología , Técnicas de Cultivo de Célula , Proteínas de Ciclo Celular , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Ratones , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Unión Proteica , Dominios Proteicos , Proteína Quinasa C-alfa/genética , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-alfa/fisiología , Ratas , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Proteínas de Unión al GTP rac/metabolismo
11.
J Cell Biochem ; 117(3): 647-58, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26284819

RESUMEN

Hypoxia-inducible factor-1α (HIF-1α) is one of the key transcription factors that mediate adaptation to hypoxia. Despite increasing evidence implicating the PKC family as potential modulators of HIF-1α, the molecular mechanisms of PKC isoform-dependent HIF-1α activity under hypoxic conditions have not been systematically elucidated in cancer cell lines. Here, we collectively investigated how each isoform of the PKC family contributes to HIF-1α accumulation in the human cervical cancer cell line HeLa. Among the abundant PKC isoforms, blockade of either PKCα or PKCδ was found to substantially reduce HIF-1α accumulation and transcriptional activity in hypoxic cells. Knockdown of PKCδ resulted in a reduction of HIF-1α mRNA levels, whereas the HIF-1α mRNA level was unchanged regardless of PKCα knockdown. Upon searching for the downstream effectors of these kinases, we found that PKCα controls HIF-1α translation via AKT-mTOR under hypoxic conditions. On the other hand, one of the well-known transcriptional regulation pathways of HIF-1α, nuclear factor-κB (NF-κB) is identified as a downstream effector of PKCδ. Taken together, our findings provide insights into the roles of PKC isoforms as additional, discrete modulators of hypoxia-stimulated HIF-1α accumulation through different signaling pathways.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteína Quinasa C-alfa/fisiología , Proteína Quinasa C-delta/fisiología , Hipoxia de la Célula , Células HeLa , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Isoenzimas/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Biosíntesis de Proteínas , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
12.
Br J Pharmacol ; 173(5): 870-87, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26660275

RESUMEN

BACKGROUND AND PURPOSE: We investigated the hypothesis that elevated glucose increases contractile responses in vascular smooth muscle and that this enhanced constriction occurs due to the glucose-induced PKC-dependent inhibition of voltage-gated potassium channels. EXPERIMENTAL APPROACH: Patch-clamp electrophysiology in rat isolated mesenteric arterial myocytes was performed to investigate the glucose-induced inhibition of voltage-gated potassium (Kv ) current. To determine the effects of glucose in whole vessel, wire myography was performed in rat mesenteric, porcine coronary and human internal mammary arteries. KEY RESULTS: Glucose-induced inhibition of Kv was PKC-dependent and could be pharmacologically dissected using PKC isoenzyme-specific inhibitors to reveal a PKCß-dependent component of Kv inhibition dominating between 0 and 10 mM glucose with an additional PKCα-dependent component becoming evident at concentrations greater than 10 mM. These findings were supported using wire myography in all artery types used, where contractile responses to vessel depolarization and vasoconstrictors were enhanced by increasing bathing glucose concentration, again with evidence for distinct and complementary PKCα/PKCß-mediated components. CONCLUSIONS AND IMPLICATIONS: Our results provide compelling evidence that glucose-induced PKCα/PKCß-mediated inhibition of Kv current in vascular smooth muscle causes an enhanced constrictor response. Inhibition of Kv current causes a significant depolarization of vascular myocytes leading to marked vasoconstriction. The PKC dependence of this enhanced constrictor response may present a potential therapeutic target for improving microvascular perfusion following percutaneous coronary intervention after myocardial infarction in hyperglycaemic patients.


Asunto(s)
Vasos Coronarios/efectos de los fármacos , Glucosa/farmacología , Arterias Mamarias/efectos de los fármacos , Arterias Mesentéricas/efectos de los fármacos , Proteína Quinasa C beta/fisiología , Proteína Quinasa C-alfa/fisiología , Animales , Vasos Coronarios/fisiología , Humanos , Técnicas In Vitro , Isoenzimas/antagonistas & inhibidores , Isoenzimas/fisiología , Masculino , Arterias Mamarias/fisiología , Arterias Mesentéricas/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , Canales de Potasio con Entrada de Voltaje/fisiología , Proteína Quinasa C beta/antagonistas & inhibidores , Proteína Quinasa C-alfa/antagonistas & inhibidores , Ratas Wistar , Porcinos , Vasoconstricción/efectos de los fármacos
13.
Zhongguo Dang Dai Er Ke Za Zhi ; 17(12): 1354-9, 2015 Dec.
Artículo en Chino | MEDLINE | ID: mdl-26695679

RESUMEN

OBJECTIVE: To study the roles of PKCα on the proliferation, apoptosis, differentiation, cytokine production and inducible regulatory T cell (iTreg) induction of T cells. METHODS: T cells from WT (PKCα⁺/⁺) or PKCα knockout (PKCα⁻/⁻) mice were isolated and cultured in vitro. T cell proliferation and apoptosis were determined using ³H thymidine incorporation and CSFE/Annexin V staining. Cytokines production (IL-2, IL-4, IFN-γ and IL-17) was detected using ELISA. CD4⁺T cells were isolated and cultured in vitro via Th17 or iTreg biased condition. Flow cytometry was used to detect the cell differentiation. RESULTS: The production of IL-2 upon TCR stimulation increased, while the contents of IL-4 and IL-17 decreased in the PKCα⁻/⁻ group compared with the PKCα⁺/⁺ group. The differentiation rate of Th17 cells decreased, while the iTreg production increased in the PKCα⁻/⁻ group compared with the PKCα⁺/⁺ group. CONCLUSIONS: PKC-α is proinflammatory.


Asunto(s)
Proteína Quinasa C-alfa/fisiología , Linfocitos T/fisiología , Animales , Diferenciación Celular , Citocinas/biosíntesis , Activación de Linfocitos , Ratones , Receptores de Antígenos de Linfocitos T/fisiología , Células Th17/inmunología
14.
Mol Cell ; 58(5): 711-2, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-26046643

RESUMEN

In this issue, Lee et al. (2015) show that PKC directly phosphorylates the glucose transporter Glut1, in order to promote glucose uptake in response to growth factor signaling.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos/genética , Transportador de Glucosa de Tipo 1/metabolismo , Proteínas de Transporte de Monosacáridos/deficiencia , Proteína Quinasa C-alfa/fisiología , Animales , Humanos
15.
Mol Cell ; 58(5): 845-53, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25982116

RESUMEN

Protein kinase C has been implicated in the phosphorylation of the erythrocyte/brain glucose transporter, GLUT1, without a clear understanding of the site(s) of phosphorylation and the possible effects on glucose transport. Through in vitro kinase assays, mass spectrometry, and phosphospecific antibodies, we identify serine 226 in GLUT1 as a PKC phosphorylation site. Phosphorylation of S226 is required for the rapid increase in glucose uptake and enhanced cell surface localization of GLUT1 induced by the phorbol ester 12-O-tetradecanoyl-phorbol-13-acetate (TPA). Endogenous GLUT1 is phosphorylated on S226 in primary endothelial cells in response to TPA or VEGF. Several naturally occurring, pathogenic mutations that cause GLUT1 deficiency syndrome disrupt this PKC phosphomotif, impair the phosphorylation of S226 in vitro, and block TPA-mediated increases in glucose uptake. We demonstrate that the phosphorylation of GLUT1 on S226 regulates glucose transport and propose that this modification is important in the physiological regulation of glucose transport.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos/genética , Transportador de Glucosa de Tipo 1/metabolismo , Proteínas de Transporte de Monosacáridos/deficiencia , Proteína Quinasa C-alfa/fisiología , Secuencia de Aminoácidos , Animales , Transporte Biológico , Errores Innatos del Metabolismo de los Carbohidratos/enzimología , Línea Celular , Células Endoteliales/metabolismo , Eritrocitos/metabolismo , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1/genética , Células HeLa , Humanos , Datos de Secuencia Molecular , Proteínas de Transporte de Monosacáridos/genética , Mutación Missense , Fosforilación , Procesamiento Proteico-Postraduccional , Ratas , Xenopus laevis
16.
Neurochem Int ; 78: 76-85, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25217805

RESUMEN

INTRODUCTION: The natural product tetramethylpyrazine (TMP) has a variety of biologic activities, including neuroprotection. Nitrones are powerful free radical scavengers. We have designed and synthesized a TMP derivative, TN-2, which is armed with two nitrone moieties. AIMS: In this study, we investigated the neuroprotective effect of TN-2 against 6-hydroxydopamine (6-OHDA)-induced neurotoxicity in vitro and in zebrafish. METHODS: PC12 cells, zebrafish and rats were exposed to 6-OHDA challenge. MTT assay, LDH release, Hoechst staining, DAF-FM staining, luciferase reporter construct transfection, and western blotting were applied to detect cell viability, apoptosis, intracellular nitric oxide (NO), NF-κB transcriptional activity and proteins expression. In zebrafish, whole-mount staining and real-time PCR were performed to quantify dopaminergic neurons and mRNA expression. Hematoxylin and eosin staining and immunohistochemistry for glial fibrillary acidic protein were used to detect the astrocyte activation in the unilateral 6-OHDA rat model. RESULTS: TN-2 but not TMP exhibited potent neuroprotective effect against 6-OHDA-induced apoptosis in PC12 cells. Moreover, TN-2 prevented dopaminergic neuron loss and suppressed mRNA expression of pro-inflammatory genes, including IL-1ß, TNF-α and COX-2, in 6-OHDA-treated zebrafish. TN-2 remarkably attenuated microglial/astrocyte activation in the unilateral 6-OHDA rat model. The mechanistic study demonstrated that TN-2 inhibited over-production of intracellular NO and protein expression of inducible nitric oxide synthase through down-regulating NF-κB activity. Additionally, the PKCα/PI3-K/Akt pathway was also involved in the neuroprotection of TN-2. CONCLUSION: These results suggest that TN-2 protected against 6-OHDA-induced neurotoxicity via modulating the NF-κB-medicated neuroinflammation and PKCα/PI3-K/Akt pathways.


Asunto(s)
FN-kappa B/fisiología , Oxidopamina/toxicidad , Fosfatidilinositol 3-Quinasas/fisiología , Proteína Quinasa C-alfa/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Pirazinas/farmacología , Animales , Masculino , Fármacos Neuroprotectores/farmacología , Células PC12 , Pirazinas/química , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Pez Cebra
17.
Am J Physiol Lung Cell Mol Physiol ; 307(5): L374-85, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25015976

RESUMEN

We used a PKC-α knockout model to investigate the regulation of alveolar epithelial Na(+) channels (ENaC) by PKC. Primary alveolar type II (ATII) cells were subjected to cell-attached patch clamp. In the absence of PKC-α, the open probability (Po) of ENaC was decreased by half compared with wild-type mice. The channel density (N) was also reduced in the knockout mice. Using in vivo biotinylation, membrane localization of all three ENaC subunits (α, ß, and γ) was decreased in the PKC-α knockout lung, compared with the wild-type. Confocal microscopy of lung slices showed elevated levels of reactive oxygen species (ROS) in the lungs of the PKC-α knockout mice vs. the wild-type. High levels of ROS in the knockout lung can be explained by a decrease in both cytosolic and mitochondrial superoxide dismutase activity. Elevated levels of ROS in the knockout lung activates PKC-δ and leads to reduced dephosphorylation of ERK1/2 by MAP kinase phosphatase, which in turn causes increased internalization of ENaC via ubiquitination by the ubiquitin-ligase Nedd4-2. In addition, in the knockout lung, PKC-δ activates ERK, causing a decrease in ENaC density at the apical alveolar membrane. PKC-δ also phosphorylates MARCKS, leading to a decrease in ENaC Po. The effects of ROS and PKC-δ were confirmed with patch-clamp experiments on isolated ATII cells in which the ROS scavenger, Tempol, or a PKC-δ-specific inhibitor added to patches reversed the observed decrease in ENaC apical channel density and Po. These results explain the decrease in ENaC activity in PKC-α knockout lung.


Asunto(s)
Células Epiteliales/metabolismo , Canales Epiteliales de Sodio/metabolismo , Pulmón/metabolismo , Proteína Quinasa C-alfa/fisiología , Alveolos Pulmonares/metabolismo , Animales , Células Epiteliales/citología , Femenino , Immunoblotting , Masculino , Ratones , Ratones Noqueados , Alveolos Pulmonares/citología , Especies Reactivas de Oxígeno/metabolismo
18.
J Am Heart Assoc ; 3(3): e000716, 2014 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-24895160

RESUMEN

BACKGROUND: Right ventricular (RV) diastolic function is impaired in patients with pulmonary arterial hypertension (PAH). Our previous study showed that elevated cardiomyocyte stiffness and myofilament Ca(2+) sensitivity underlie diastolic dysfunction in PAH. This study investigates protein modifications contributing to cellular diastolic dysfunction in PAH. METHODS AND RESULTS: RV samples from PAH patients undergoing heart-lung transplantation were compared to non-failing donors (Don). Titin stiffness contribution to RV diastolic dysfunction was determined by Western-blot analyses using antibodies to protein-kinase-A (PKA), Cα (PKCα) and Ca(2+)/calmoduling-dependent-kinase (CamKIIδ) titin and phospholamban (PLN) phosphorylation sites: N2B (Ser469), PEVK (Ser170 and Ser26), and PLN (Thr17), respectively. PKA and PKCα sites were significantly less phosphorylated in PAH compared with donors (P<0.0001). To test the functional relevance of PKA-, PKCα-, and CamKIIδ-mediated titin phosphorylation, we measured the stiffness of single RV cardiomyocytes before and after kinase incubation. PKA significantly decreased PAH RV cardiomyocyte diastolic stiffness, PKCα further increased stiffness while CamKIIδ had no major effect. CamKIIδ activation was determined indirectly by measuring PLN Thr17phosphorylation level. No significant changes were found between the groups. Myofilament Ca(2+) sensitivity is mediated by sarcomeric troponin I (cTnI) phosphorylation. We observed increased unphosphorylated cTnI in PAH compared with donors (P<0.05) and reduced PKA-mediated cTnI phosphorylation (Ser22/23) (P<0.001). Finally, alterations in Ca(2+)-handling proteins contribute to RV diastolic dysfunction due to insufficient diastolic Ca(2+) clearance. PAH SERCA2a levels and PLN phosphorylation were significantly reduced compared with donors (P<0.05). CONCLUSIONS: Increased titin stiffness, reduced cTnI phosphorylation, and altered levels of phosphorylation of Ca(2+) handling proteins contribute to RV diastolic dysfunction in PAH.


Asunto(s)
Hipertensión Pulmonar/fisiopatología , Miocitos Cardíacos/química , Disfunción Ventricular Derecha/fisiopatología , Adulto , Western Blotting , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/análisis , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Estudios de Casos y Controles , Conectina/análisis , Conectina/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/análisis , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Femenino , Ventrículos Cardíacos/química , Ventrículos Cardíacos/fisiopatología , Humanos , Masculino , Miocitos Cardíacos/fisiología , Fosforilación , Proteína Quinasa C-alfa/análisis , Proteína Quinasa C-alfa/fisiología , Troponina I/fisiología
19.
PLoS One ; 9(5): e98018, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24842701

RESUMEN

Everyday functioning relies on episodic memory, the conscious retrieval of past experiences, but this crucial cognitive ability declines severely with aging and disease. Vulnerability to memory decline varies across individuals however, producing differences in the time course and severity of memory problems that complicate attempts at diagnosis and treatment. Here we identify a key source of variability, by examining gene dependent changes in the neural basis of episodic remembering in healthy adults, targeting seven polymorphisms previously linked to memory. Scalp recorded Event-Related Potentials (ERPs) were measured while participants remembered words, using an item recognition task that requires discrimination between studied and unstudied stimuli. Significant differences were found as a consequence of a Single Nucleotide Polymorphism (SNP) in just one of the tested genes, PRKCA (rs8074995). Participants with the common G/G variant exhibited left parietal old/new effects, which are typically seen in word recognition studies, reflecting recollection-based remembering. During the same stage of memory retrieval participants carrying a rarer A variant exhibited an atypical pattern of brain activity, a topographically dissociable frontally-distributed old/new effect, even though behavioural performance did not differ between groups. Results replicated in a second independent sample of participants. These findings demonstrate that the PRKCA genotype is important in determining how episodic memories are retrieved, opening a new route towards understanding individual differences in memory.


Asunto(s)
Memoria Episódica , Recuerdo Mental/fisiología , Polimorfismo de Nucleótido Simple/genética , Proteína Quinasa C-alfa/genética , Proteína Quinasa C-alfa/fisiología , Análisis de Varianza , Discriminación en Psicología/fisiología , Potenciales Evocados/fisiología , Humanos
20.
BMC Cancer ; 14: 93, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24528886

RESUMEN

BACKGROUND: Netrin-1 and its receptor UNC5B play important roles in angiogenesis, embryonic development, cancer and inflammation. However, their expression patttern and biological roles in bladder cancer have not been well characterized. The present study aims to investigating the clinical significance of PKC α, netrin-1 and UNC5B in bladder cancer as well as their association with malignant biological behavior of cancer cells. METHODS: Netrin-1 and UNC5B expression was examined in 120 bladder cancer specimens using immunohistochemistry and in 40 fresh cancer tissues by western blot. Immunofluorescence was performed in cancer cell lines. PKC α agonist PMA and PKC siRNA was employed in bladder cancer cells. CCK-8, wound healing assays and flow cytometry analysis were used to examine cell proliferation, migration and cell cycle, respectively. RESULTS: Netrin-1 expression was positively correlated with histological grade, T stage, metastasis and poor prognosis in bladder cancer tissues. Immunofluorescence showed elevated netrin-1 and decreased UNC5B expression in bladder cancer cells compared with normal bladder cell line. Furthermore, cell proliferation, migration and cell cycle progression were promoted with PMA treatment while inhibited by calphostin C. In addition, PMA treatment could induce while calphostin C reduce netrin-1 expression in bladder cancer cells. CONCLUSIONS: The present study identified netrin-1/UNC5B, which could be regulated by PKC signaling, was important mediators of bladder cancer progression.


Asunto(s)
Factores de Crecimiento Nervioso/biosíntesis , Proteína Quinasa C-alfa/fisiología , Receptores de Superficie Celular/biosíntesis , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/biosíntesis , Neoplasias de la Vejiga Urinaria/metabolismo , Anciano , Línea Celular Tumoral , Supervivencia Celular/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Receptores de Netrina , Netrina-1 , Neoplasias de la Vejiga Urinaria/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA