Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Commun Signal ; 18(1): 26, 2020 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-32059739

RESUMEN

BACKGROUND: The limited options to treat obesity and its complications result from an incomplete understanding of the underlying molecular mechanisms regulating white adipose tissue development, including adipocyte hypertrophy (increase in size) and hyperplasia (increase in number through adipogenesis). We recently demonstrated that lack of the adaptor protein Nck1 in mice is associated with reduced adiposity and impaired adipocyte differentiation. In agreement, Nck1 depletion in 3 T3-L1 cells also attenuates adipocyte differentiation by enhancing PDGFRα activation and signaling. This is accompanied by higher expression of PDGF-A, a specific PDGFRα ligand, that may contribute to enhanced activation of PDGFRα signaling in the absence of Nck1 in white adipose tissue. However, whether Nck1 deficiency also impairs adipogenic differentiation in bone marrow still remains to be determined. METHODS: To address this point, Nck1-deficient derived bone marrow mesenchymal stem/stromal cells (BM-MSCs) and C3H10T1/2 mesenchymal stem cells were differentiated into adipocytes in vitro. Genes and proteins expression in these cellular models were determined using qPCR and western blotting respectively. Pharmacological approaches were used to assess a role for Nrf2 in mediating Nck1 deficiency effect on mesenchymal stem cells adipocyte differentiation. RESULTS: Nck1 deficiency in both BM-MSCs and C3H10T1/2 results in impaired adipocyte differentiation, accompanied by increased activation of the transcription factor Nrf2, as shown by increased mRNA levels of Nrf2 target genes, including PDGF-A. Using pharmacological activator and inhibitor of Nrf2, we further provide evidence that Nrf2 is an important player in PDGFRα signaling that mediates expression of PDGF-A and impaired adipogenesis in Nck1-deficient BM-MSCs and C3H10T1/2 cells. CONCLUSION: This study demonstrates that Nck1 deficiency in mesenchymal stem cells impairs adipogenesis through activation of the PDGFRα-Nrf2 anti-adipogenic signaling pathway. Video Abstract.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Células Madre Mesenquimatosas , Factor 2 Relacionado con NF-E2/metabolismo , Obesidad , Proteínas Oncogénicas/fisiología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Adipogénesis , Animales , Células Cultivadas , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones Noqueados , Obesidad/metabolismo , Obesidad/patología , Proteínas Oncogénicas/genética
2.
Adipocyte ; 8(1): 98-104, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30848691

RESUMEN

Obesity and associated metabolic complications, including diabetes, cardiovascular and hepatic diseases, and certain types of cancers, create a major socioeconomic burden. Obesity is characterized by excessive expansion of white adipose tissue resulting from increased adipocyte size, and enhanced adipocyte precursor cells proliferation and differentiation into mature adipocytes, a process well-defined as adipogenesis. Efforts to develop therapeutically potent strategies to circumvent obesity are impacted by our limited understanding of molecular mechanisms regulating adipogenesis. In this review, we discuss recently discovered molecular mechanisms restraining adipogenesis. In this perspective, the discoveries of white adipose tissue endogenous adipogenesis-regulatory cells (Aregs) that negatively regulate adipocyte differentiation, platelet-derived growth factor receptor isoform α (PDGFRα) activation and downstream signaling that hinder adipocyte precursors differentiation, and a group of obesity-associated non-coding RNAs (ncRNAs) that regulate adipogenesis open up promising therapeutic avenues to prevent and/or treat obesity.


Asunto(s)
Adipogénesis/fisiología , Obesidad/metabolismo , Obesidad/prevención & control , Adipocitos Blancos/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Diferenciación Celular/fisiología , Humanos , MicroARNs/genética , ARN Largo no Codificante/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal
3.
iScience ; 6: 22-37, 2018 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-30240612

RESUMEN

Obesity results from an excessive expansion of white adipose tissue (WAT), which is still poorly understood from an etiologic-mechanistic perspective. Here, we report that Nck1, a Src homology domain-containing adaptor, is upregulated during WAT expansion and in vitro adipogenesis. In agreement, Nck1 mRNA correlates positively with peroxisome proliferator-activated receptor (PPAR) γ and adiponectin mRNAs in the WAT of obese humans, whereas Nck1-deficient mice display smaller WAT depots with reduced number of adipocyte precursors and accumulation of extracellular matrix. Furthermore, silencing Nck1 in 3T3-L1 preadipocytes increases the proliferation and expression of genes encoding collagen, whereas it decreases the expression of adipogenic markers and impairs adipogenesis. Silencing Nck1 in 3T3-L1 preadipocytes also promotes the expression of platelet-derived growth factor (PDGF)-A and platelet-derived growth factor receptor (PDGFR) α activation and signaling. Preventing PDGFRα activation using imatinib, or through PDGF-A or PDGFRα deficiency, inhibits collagen expression in Nck1-deficient preadipocytes. Finally, imatinib rescues differentiation of Nck1-deficient preadipocytes. Altogether, our findings reveal that Nck1 modulates WAT development through PDGFRα-dependent remodeling of preadipocytes.

4.
J Biol Chem ; 293(32): 12516-12524, 2018 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-29941454

RESUMEN

One feature of diabetes is the failure of pancreatic ß cells to produce insulin, but the molecular mechanisms leading to this failure remain unclear. Increasing evidence supports a role for protein kinase R-like endoplasmic reticulum kinase (PERK) in the development and function of healthy pancreatic ß cells. Previously, our group identified the adaptor protein Nck1 as a negative regulator of PERK. Indeed, we demonstrated that Nck1, by directly binding PERK autophosphorylated on Tyr561, limits PERK activation and signaling. Accordingly, we found that stable depletion of Nck1 in ß cells promotes PERK activation and signaling, increases insulin biosynthesis, and improves cell viability in response to diabetes-related stresses. Herein, we explored the therapeutic potential of abrogating the interaction between Nck and PERK to improve ß-cell function and survival. To do so, we designed and used a peptide containing the minimal PERK sequence involved in binding Nck1 conjugated to the cell-permeable protein transduction domain from the HIV protein TAT. In the current study, we confirm that the synthetic TAT-Tyr(P)561 phosphopeptide specifically binds the SH2 domain of Nck and prevents Nck interaction with PERK, thereby promoting basal PERK activation. Moreover, we report that treatment of ß cells with TAT-Tyr(P)561 inhibits glucolipotoxicity-induced apoptosis, whereas it enhances insulin production and secretion. Taken together, our results support the potential of sequestering Nck using a synthetic peptide to enhance basal PERK activation and create more robust ß cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Diabetes Mellitus/fisiopatología , Células Secretoras de Insulina/efectos de los fármacos , Insulina/biosíntesis , Insulinoma/prevención & control , Proteínas Oncogénicas/metabolismo , Fragmentos de Péptidos/farmacología , Sustancias Protectoras/farmacología , eIF-2 Quinasa/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis , Células Cultivadas , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/prevención & control , Células Secretoras de Insulina/patología , Insulinoma/genética , Insulinoma/metabolismo , Ratones , Proteínas Oncogénicas/genética , Fosforilación , Transducción de Señal , Estrés Fisiológico
5.
Biol Cell ; 110(2): 27-32, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29168198

RESUMEN

In humans, the pathogenesis of diabetes is characterised by two major pancreatic ß cell defects: a reduction in ß cell mass and the failure of ß cells to produce enough insulin. Over the past two decades, multiple studies involving cell cultures, animal models and human subjects have established the importance of the protein kinase RNA-like endoplasmic reticulum kinase (PERK) in the adaptive functional capacity of pancreatic ß cells during embryonic development and into adulthood. In this review, we will highlight major findings identifying PERK as a crucial player in ß cell physiology and in diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Epífisis/anomalías , Factor 2 Eucariótico de Iniciación/genética , Homeostasis/genética , Células Secretoras de Insulina/enzimología , Osteocondrodisplasias/genética , eIF-2 Quinasa/genética , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/inmunología , Adulto , Animales , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/enzimología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/patología , Epífisis/enzimología , Epífisis/inmunología , Epífisis/patología , Factor 2 Eucariótico de Iniciación/inmunología , Regulación del Desarrollo de la Expresión Génica , Homeostasis/inmunología , Humanos , Recién Nacido , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/patología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/inmunología , Osteocondrodisplasias/enzimología , Osteocondrodisplasias/inmunología , Osteocondrodisplasias/patología , Transducción de Señal , eIF-2 Quinasa/deficiencia , eIF-2 Quinasa/inmunología
6.
Cell Signal ; 36: 79-90, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28455143

RESUMEN

PTP1B, a prototype of the non-receptor subfamily of the protein tyrosine phosphatase superfamily, plays a key role in regulating intracellular signaling from various receptor and non-receptor protein tyrosine kinases. Previously, we reported that silencing Nck1 in human hepatocellular carcinoma HepG2 cells enhances basal and growth factor-induced activation of the PI3K-Akt pathway through attenuating PTP1B expression. However, the underlying mechanism by which Nck1 depletion represses PTP1B expression remains unclear. In this study, we found that silencing Nck1 attenuates PTP1B expression in HepG2 cells through down-regulation of IRE1α. Indeed, we show that silencing Nck1 in HepG2 cells leads to decreased IRE1α expression and signaling. Accordingly, IRE1α depletion using siRNA in HepG2 cells enhances PI3K-dependent basal and growth factor-induced Akt activation, reproducing the effects of silencing Nck1 on activation of this pathway. In addition, depletion of IRE1α also leads to reduced PTP1B expression, which was rescued by ectopic expression of IRE1α in Nck1-depleted cells. Mechanistically, we found that silencing either Nck1 or IRE1α in HepG2 cells decreases PTP1B mRNA levels and stability. However, despite miR-122 levels, a miRNA targeting PTP1B 3' UTR and inducing PTP1B mRNA degradation in HepG2 cells, are increased in both Nck1- and IRE1α-depleted HepG2 cells, a miR-122 antagomir did not rescue PTP1B expression in these cells. Overall, this study highlights an important role for Nck1 in fine-tuning IRE1α expression and signaling that regulate PTP1B expression and subsequent activation of the PI3K-Akt pathway in HepG2 cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Endorribonucleasas/metabolismo , Proteínas Oncogénicas/deficiencia , Proteínas Oncogénicas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Animales , Activación Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Silenciador del Gen/efectos de los fármacos , Células HeLa , Células Hep G2 , Humanos , Ratones , MicroARNs/metabolismo , Proteínas Oncogénicas/química , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Tapsigargina/farmacología
7.
Mol Biol Cell ; 28(12): 1636-1651, 2017 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-28428258

RESUMEN

Inositol-requiring enzyme-1α (IRE1α) is an endoplasmic reticulum (ER)-transmembrane endoribonuclease kinase that plays an essential function in extraembryonic tissues during normal development and is activated during ER stress. To address the functional role of IRE1α in glomerular podocytes, we produced podocyte-specific IRE1α-deletion mice. In male mice, deletion of IRE1α in podocytes resulted in albuminuria beginning at 5 mo of age and worsening with time. Electron microscopy revealed focal podocyte foot-process effacement in 9-mo-old male IRE1α-deletion mice, as well as microvillous transformation of podocyte plasma membranes. Compared with control, glomerular cross-sectional and capillary lumenal areas were greater in deletion mice, and there was relative podocyte depletion. Levels of microtubule-associated protein 1A/1B-light chain 3 (LC3)-II expression and c-Jun N-terminal kinase-1 phosphorylation were decreased in IRE1α-deletion glomeruli, in keeping with reduced autophagy. Deletion of IRE1α exacerbated glomerular injury in anti-glomerular basement membrane nephritis. In cell culture, IRE1α dominant-negative mutants reduced the physiological (basal) accumulation of LC3B-II and the size of autophagic vacuoles but did not affect ER-associated degradation. Thus IRE1α is essential for maintaining podocyte and glomerular integrity as mice age and in glomerulonephritis. The mechanism is related, at least in part, to the maintenance of autophagy in podocytes.


Asunto(s)
Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Podocitos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Autofagia/fisiología , Células COS , Capilares , Chlorocebus aethiops , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Degradación Asociada con el Retículo Endoplásmico , Células Epiteliales/metabolismo , Inositol/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Glomérulos Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo
8.
Biochim Biophys Acta Mol Cell Res ; 1864(3): 475-486, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27965113

RESUMEN

Aging affects numerous aspects of cell biology, but the senescence-associated changes in the stress response are only beginning to emerge. To obtain mechanistic insights into these events, we examined the formation of canonical and non-canonical stress granules (SGs) in the cytoplasm. SG generation is a key event after exposure to physiological or environmental stressors. It requires the SG-nucleating proteins G3BP1 and TIA-1/TIAR and stress-related signaling events. To analyze SG formation, we used two independent models of somatic cell aging. In both model systems, cellular senescence impaired the assembly of two SG classes: (i) it compromised the formation of canonical SGs, and (ii) skewed the production of non-canonical SGs. We dissected the mechanisms underlying these senescence-dependent changes in granule biogenesis and identified several specific targets that were modulated by aging. Thus, we demonstrate a depletion of G3BP1 and TIA-1/TIAR in senescent cells and show that the loss of G3BP1 contributed to impaired SG formation. We further reveal that aging reduced Sp1 levels; this transcription factor regulated G3BP1 and TIA-1/TIAR abundance. The assembly of canonical SGs relies on the phosphorylation of translation initiation factor eIF2α. We show that senescence can cause eIF2α hyperphosphorylation. CReP is a subunit of protein phosphatase 1 and critical to reverse the stress-dependent phosphorylation of eIF2α. We demonstrate that the loss of CReP correlated with the aging-related hyperphosphorylation of eIF2α. Together, we have identified significant changes in the stress response of aging cells and provide mechanistic insights. Based on our work, we propose that the decline in SG formation can provide a new biomarker to evaluate cellular aging.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Endorribonucleasas/genética , Células Epiteliales/metabolismo , Factor 2 Eucariótico de Iniciación/genética , Proteínas de Unión a Poli(A)/genética , Proteínas Represoras/genética , Animales , Línea Celular , Senescencia Celular , Gránulos Citoplasmáticos/ultraestructura , Endorribonucleasas/metabolismo , Células Epiteliales/ultraestructura , Factor 2 Eucariótico de Iniciación/metabolismo , Regulación de la Expresión Génica , Riñón/citología , Riñón/metabolismo , Fosforilación , Proteínas de Unión a Poli(A)/metabolismo , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Porcinos
9.
Diabetes ; 65(9): 2652-66, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27325288

RESUMEN

Obesity results from an excessive expansion of white adipose tissue (WAT) from hypertrophy of preexisting adipocytes and enhancement of precursor differentiation into mature adipocytes. We report that Nck2-deficient mice display progressive increased adiposity associated with adipocyte hypertrophy. A negative relationship between the expression of Nck2 and WAT expansion was recapitulated in humans such that reduced Nck2 protein and mRNA levels in human visceral WAT significantly correlate with the degree of obesity. Accordingly, Nck2 deficiency promotes an adipogenic program that not only enhances adipocyte differentiation and lipid droplet formation but also results in dysfunctional elevated lipogenesis and lipolysis activities in mouse WAT as well as in stromal vascular fraction and 3T3-L1 preadipocytes. We provide strong evidence to support that through a mechanism involving primed PERK activation and signaling, Nck2 deficiency in adipocyte precursors is associated with enhanced adipogenesis in vitro and adiposity in vivo. Finally, in agreement with elevated circulating lipids, Nck2-deficient mice develop glucose intolerance, insulin resistance, and hepatic steatosis. Taken together, these findings reveal that Nck2 is a novel regulator of adiposity and suggest that Nck2 is important in limiting WAT expansion and dysfunction in mice and humans.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Adiposidad/fisiología , Hipertrofia/metabolismo , Proteínas Oncogénicas/metabolismo , Células 3T3-L1 , Proteínas Adaptadoras Transductoras de Señales/genética , Adipogénesis/genética , Adipogénesis/fisiología , Adiposidad/genética , Animales , Composición Corporal/genética , Composición Corporal/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Células Cultivadas , Citometría de Flujo , Humanos , Hipertrofia/genética , Grasa Intraabdominal/citología , Grasa Intraabdominal/metabolismo , Masculino , Ratones , Ratones Mutantes , Proteínas Oncogénicas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
Nat Commun ; 7: 11127, 2016 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-27040916

RESUMEN

Ternary complex (TC) and eIF4F complex assembly are the two major rate-limiting steps in translation initiation regulated by eIF2α phosphorylation and the mTOR/4E-BP pathway, respectively. How TC and eIF4F assembly are coordinated, however, remains largely unknown. We show that mTOR suppresses translation of mRNAs activated under short-term stress wherein TC recycling is attenuated by eIF2α phosphorylation. During acute nutrient or growth factor stimulation, mTORC1 induces eIF2ß phosphorylation and recruitment of NCK1 to eIF2, decreases eIF2α phosphorylation and bolsters TC recycling. Accordingly, eIF2ß mediates the effect of mTORC1 on protein synthesis and proliferation. In addition, we demonstrate a formerly undocumented role for CK2 in regulation of translation initiation, whereby CK2 stimulates phosphorylation of eIF2ß and simultaneously bolsters eIF4F complex assembly via the mTORC1/4E-BP pathway. These findings imply a previously unrecognized mode of translation regulation, whereby mTORC1 and CK2 coordinate TC and eIF4F complex assembly to stimulate cell proliferation.


Asunto(s)
Quinasa de la Caseína II/fisiología , Factor 4F Eucariótico de Iniciación/metabolismo , Complejos Multiproteicos/fisiología , Serina-Treonina Quinasas TOR/fisiología , Factores Complejos Ternarios/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Quinasa de la Caseína II/genética , Quinasa de la Caseína II/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Factor 2 Eucariótico de Iniciación/fisiología , Regulación de la Expresión Génica , Células HEK293 , Humanos , Células MCF-7 , Diana Mecanicista del Complejo 1 de la Rapamicina , Modelos Genéticos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Proteínas Oncogénicas/metabolismo , Iniciación de la Cadena Peptídica Traduccional , Fosforilación , Transducción de Señal , Estrés Fisiológico , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
11.
Cell Signal ; 27(12): 2555-67, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26434994

RESUMEN

Increasing evidence strongly supports a critical role for PERK in regulating pancreatic ß cell function. In agreement, we previously reported that enhancing PERK basal activity, by silencing the SH domain-containing adaptor protein Nck1 in pancreatic ß cells, increased insulin content in a PERK-dependent manner. Here we report that Nck1-deficient MIN6 cells display normal overall morphology while as expected increased number of secretory granules. Furthermore, we demonstrate that cell survival to diabetes-relevant stresses is increased, while cell viability in response to chemical endoplasmic reticulum (ER) stress inducers is not changed. In agreement, PERK activation in Nck1-depleted MIN6 cells exposed to palmitate was significantly reduced while it remained strongly induced by the ER stress inducer thapsigargin. Interestingly, silencing Nck1 in MIN6 cells results in increased PERK basal activity and expression of the PERK downstream target sestrin2, which promotes autophagy by attenuating mTORC1 activation through AMPK-dependent and -independent mechanisms. Accordingly, activated AMPK was increased, mTORC1 signaling decreased, and autophagy markers increased in Nck1-silenced MIN6 cells. Increased autophagy was recapitulated in Nck1(-/-) mice pancreatic ß cells. In addition, basal levels of the PERK substrate Nrf2 and its antioxidant gene targets (HO-1 and Nqo1) were upregulated in Nck1-silenced MIN6 cells, revealing an active PERK-Nrf2 signaling in these cells. Finally, Akt activation was increased in Nck1-silenced MIN6 cells. Altogether, this study demonstrates that Nck1 silencing in pancreatic ß cells promotes PERK activation and signaling to protect ß cells against pathological stresses. These findings further provide new perspectives to advance our understanding of molecular mechanisms and signaling systems regulating pancreatic ß cell fates.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Diabetes Mellitus/enzimología , Células Secretoras de Insulina/enzimología , Proteínas Oncogénicas/genética , eIF-2 Quinasa/metabolismo , Factor de Transcripción Activador 4/metabolismo , Transporte Activo de Núcleo Celular , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autofagia , Supervivencia Celular , Células Cultivadas , Activación Enzimática , Técnicas de Silenciamiento del Gen , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/patología , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Oncogénicas/metabolismo , Transducción de Señal , Estrés Fisiológico , Factor de Transcripción CHOP/metabolismo , Activación Transcripcional
12.
Mol Biol Cell ; 26(5): 913-23, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25568336

RESUMEN

The USP19 deubiquitinating enzyme modulates the expression of myogenin and myofibrillar proteins in L6 muscle cells. This raised the possibility that USP19 might regulate muscle cell differentiation. We therefore tested the effects of adenoviral-mediated overexpression or small interfering RNA (siRNA)-mediated silencing of either the cytoplasmic or endoplasmic reticulum (ER)-localized isoforms of USP19. Only the ER-localized isoform of USP19 (USP19-ER) modulated myoblast fusion as well as the expression of myogenin and myofibrillar proteins, and these effects were also dependent on USP19 catalytic activity. USP19-ER inhibited muscle cell differentiation and the induction of CHOP, a transcription factor in the unfolded-protein response (UPR) that is activated during differentiation. Inducing the UPR by creating mild ER stress with thapsigargin was able to reverse the defect in myoblast fusion caused by the overexpression of USP19-ER, suggesting strongly that USP19 exerts its effects on fusion through its effects on UPR signaling. USP19 also functions similarly in vivo, as USP19(-/-) mice display improved muscle regeneration concomitant with enhanced expression of CHOP. Collectively these results implicate a deubiquitinating enzyme as a regulator of the UPR. They also suggest that inhibition of USP19 may be a therapeutic approach for the enhancement of muscle growth following injury.


Asunto(s)
Diferenciación Celular/fisiología , Endopeptidasas/metabolismo , Retículo Endoplásmico/metabolismo , Desarrollo de Músculos/fisiología , Transducción de Señal , Respuesta de Proteína Desplegada , Animales , Endopeptidasas/genética , Ratones , Ratones Noqueados , ARN Interferente Pequeño , Ratas , Factor de Transcripción CHOP/metabolismo
13.
Cell Commun Signal ; 12: 71, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25398386

RESUMEN

BACKGROUND: Activation of the PI3K/Akt pathway mediates crucial cellular functions regulated by receptor tyrosine kinases, such as cell growth, proliferation, survival and metabolism. Previously, we reported that the whole-body knockout of the Src homology domain-containing adaptor protein Nck1 improves overall glucose homeostasis and insulin-induced activation of the PI3K/Akt pathway in liver of obese mice. The aim of the current study is to elucidate the mechanism by which Nck1 depletion regulates hepatic insulin signaling. RESULTS: Here, we demonstrate that Nck1 regulates the activation of the PI3K/Akt pathway in a protein tyrosine phosphatase 1B (PTP1B)-dependent mechanism. Indeed, depletion of Nck1 by siRNA in HepG2 cells enhances PI3K-dependent basal and growth factor-induced Akt activation. In accordance, primary hepatocytes isolated from Nck1 (-/-) mice also display enhanced Akt activation in response to insulin. Activation of the PI3K/Akt pathway in Nck1-depleted HepG2 cells relies on higher levels of tyrosine-phosphorylated proteins and correlates with decreased PTP1B levels. Interestingly, Nck1 and PTP1B in cells are found in a common molecular complex and their interaction is dependent on the SH3 domains of Nck1. Finally, Nck1 depletion in HepG2 cells neither affects PTP1B gene transcription nor PTP1B protein stability, suggesting that Nck1 modulates PTP1B expression at the translational level. CONCLUSION: Our study provides strong evidence supporting that the adaptor protein Nck1 interacts with PTP1B and also regulates PTP1B expression. In this manner, Nck1 plays a role in regulating the PI3K/Akt pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Oncogénicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células Cultivadas , Fibroblastos , Glucosa/metabolismo , Células HEK293 , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Masculino , Ratones Noqueados , Proteínas Oncogénicas/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 1/genética , ARN Interferente Pequeño/genética
14.
Mol Biol Cell ; 25(5): 702-11, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24371088

RESUMEN

PERK, the PKR-like endoplasmic reticulum (ER) kinase, is an ER transmembrane serine/threonine protein kinase activated during ER stress. In this study, we provide evidence that the Src-homology domain-containing adaptor Nck1 negatively regulates PERK. We show that Nck directly binds to phosphorylated Y(561) in the PERK juxtamembrane domain through its SH2 domain. We demonstrate that mutation of Y(561) to a nonphosphorylatable residue (Y561F) promotes PERK activity, suggesting that PERK phosphorylation at Y(561) (pY(561)PERK) negatively regulates PERK. In agreement, we show that pY(561)PERK delays PERK activation and signaling during ER stress. Compatible with a role for PERK in pancreatic ß-cells, we provide strong evidence that Nck1 contributes to PERK regulation of pancreatic ß-cell proteostasis. In fact, we demonstrated that down-regulation of Nck1 in mouse insulinoma MIN6 cells results in faster dephosphorylation of pY(561)PERK, which correlates with enhanced PERK activation, increased insulin biosynthesis, and PERK-dependent increase in proinsulin content. Furthermore, we report that pancreatic islets in whole-body Nck1-knockout mice contain more insulin than control littermates. Together our data strongly suggest that Nck1 negatively regulates PERK by interacting with PERK and protecting PERK from being dephosphorylated at its inhibitory site pY(561) and in this way affects pancreatic ß-cell proinsulin biogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Células Secretoras de Insulina/metabolismo , Proteínas Oncogénicas/fisiología , Proinsulina/biosíntesis , eIF-2 Quinasa/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Fosforilación , Proinsulina/metabolismo , eIF-2 Quinasa/metabolismo
15.
Int J Cell Biol ; 2012: 684684, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22496693

RESUMEN

Actin cytoskeleton remodeling is under the regulation of multiple proteins with various activities. Here, we demonstrate that the γ2 isoform of Casein Kinase I (CKIγ2) is part of a novel molecular path regulating the formation of actin stress fibers. We show that overexpression of CKIγ2 in fibroblasts alters cell morphology by impairing actin stress fibers formation. We demonstrate that this is concomitant with increased phosphorylation of the CDK inhibitor p27(Kip) and lower levels of activated RhoA, and is dependent on CKIγ2 catalytic activity. Moreover, we report that roscovitine, a potent inhibitor of cyclin-dependent kinases, including Cdk5, decreases p27(Kip) protein levels and restores actin stress fibers formation in CKIγ2 overexpressing cells, suggesting the existence of a CKIγ2-Cdk5-p27(Kip)-RhoA pathway in regulating actin remodeling. On the other hand, we also show that in a manner independent of its catalytic activity, CKIγ2 delays cell cycle progression through G1. Collectively our findings reveal that CKIγ2 is a novel player in the control of actin cytoskeleton dynamics and cell proliferation.

16.
BMC Cancer ; 11: 443, 2011 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-21992144

RESUMEN

BACKGROUND: Nck1 and Nck2 adaptor proteins are involved in signaling pathways mediating proliferation, cytoskeleton organization and integrated stress response. Overexpression of Nck1 in fibroblasts has been shown to be oncogenic. Through the years this concept has been challenged and the consensus is now that overexpression of either Nck cooperates with strong oncogenes to transform cells. Therefore, variations in Nck expression levels in transformed cells could endorse cancer progression. METHODS: Expression of Nck1 and Nck2 proteins in various cancer cell lines at different stages of progression were analyzed by western blots. We created human primary melanoma cell lines overexpressing GFP-Nck2 and investigated their ability to proliferate along with metastatic characteristics such as migration and invasion. By western blot analysis, we compared levels of proteins phosphorylated on tyrosine as well as cadherins and integrins in human melanoma cells overexpressing or not Nck2. Finally, in mice we assessed tumor growth rate of human melanoma cells expressing increasing levels of Nck2. RESULTS: We found that expression of Nck2 is consistently increased in various metastatic cancer cell lines compared with primary counterparts. Particularly, we observed significant higher levels of Nck2 protein and mRNA, as opposed to no change in Nck1, in human metastatic melanoma cell lines compared with non-metastatic melanoma and normal melanocytes. We demonstrated the involvement of Nck2 in proliferation, migration and invasion in human melanoma cells. Moreover, we discovered that Nck2 overexpression in human primary melanoma cells correlates with higher levels of proteins phosphorylated on tyrosine residues, assembly of Nck2-dependent pY-proteins-containing molecular complexes and downregulation of cadherins and integrins. Importantly, we uncovered that injection of Nck2-overexpressing human primary melanoma cells into mice increases melanoma-derived tumor growth rate. CONCLUSIONS: Collectively, our data indicate that Nck2 effectively influences human melanoma phenotype progression. At the molecular level, we propose that Nck2 in human primary melanoma promotes the formation of molecular complexes regulating proliferation and actin cytoskeleton dynamics by modulating kinases or phosphatases activities that results in increased levels of proteins phosphorylated on tyrosine residues. This study provides new insights regarding cancer progression that could impact on the therapeutic strategies targeting cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Movimiento Celular , Melanoma/metabolismo , Melanoma/patología , Invasividad Neoplásica , Proteínas Oncogénicas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Línea Celular Transformada , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Melanoma/genética , Ratones , Ratones Desnudos , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Proteínas Oncogénicas/genética , Fosforilación , ARN Mensajero , Transducción de Señal , Carga Tumoral/genética , Tirosina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Med Sci (Paris) ; 27(8-9): 746-52, 2011.
Artículo en Francés | MEDLINE | ID: mdl-21880263

RESUMEN

Nck is an adaptor protein composed of three N-terminal Src Homology (SH) 3 domains followed by a unique C­terminal SH2 domain. Like other SH2/SH3 domains-containing adaptor proteins, Nck mediates signal transduction from activated cell surface receptors by directing the flow of information to elicit properly orchestrated cell responses. In this way, Nck appears to be unique in its contribution to a wide variety of cellular processes. Moreover, in addition to the typical signal/pY-SH2/SH3-effectors mode of signaling, Nck also transduces signals through an inverse mode of -signaling (signal-SH3/SH2-pY/effectors) and from various cell compartments. Since Nck contributes to important morphogenic and mitogenic processes, deregulated expression of Nck could be detrimental to cellular homeostasis. In agreement, Nck expression has been found upregulated in numerous types of cancer. In this paper we delineate the main molecular -signaling -complexes associated with Nck, focusing on those involved in cancer progression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Proteínas Oncogénicas/fisiología , Actinas/metabolismo , Adaptación Fisiológica , Proteínas Adaptadoras Transductoras de Señales/química , Uniones Adherentes/fisiología , Animales , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Transformación Celular Neoplásica/genética , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Retículo Endoplásmico/metabolismo , Humanos , Melanoma/metabolismo , Ratones , Modelos Biológicos , Proteínas de Neoplasias/fisiología , Proteínas Oncogénicas/química , Conformación Proteica , Mapeo de Interacción de Proteínas , Transducción de Señal , Transducción Genética , Dominios Homologos src
18.
Am J Physiol Endocrinol Metab ; 300(3): E423-34, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20587749

RESUMEN

Obesity has been shown to create stress in the endoplasmic reticulum (ER), and that initiates the activation of the unfolded protein response (UPR). This has been reported to cause insulin resistance in selective tissues through activation of the inositol-requiring enzyme 1α (IRE1α)-c-Jun NH(2)-terminal kinase (JNK) pathway, which results in the phosphorylation of the insulin receptor substrate-1 (IRS-1) at an inhibitory site and blocks insulin receptor signaling. In this study, we report that the Src homology domain-containing adaptor protein Nck1, previously shown to modulate the UPR, is of functional importance in obesity-induced ER stress signaling and inhibition of insulin actions. We have examined obese Nck1(-/-) and Nck1(+/+) mice for glucose tolerance, insulin sensitivity, and signaling as well as for ER stress markers and IRS-1 phosphorylation at Ser(307). Our findings show that obese Nck1-deficient mice display improved glucose disposal accompanied by enhanced insulin signaling in liver. This correlates with attenuated IRE1α and JNK activation and IRS-1 phosphorylation at Ser(307) compared with obese wild-type mice. Consistent with our in vivo data, we report that downregulation of Nck1 using siRNA in HepG2 cells results in decreased thapsigargin-induced IRE1α activation and signaling and IRS-1 phosphorylation at Ser(307), whereas it markedly enhances insulin signaling. Overall, in liver and in cultured cells, we show that depletion of Nck1 attenuates the UPR signal and its inhibitory action on insulin signaling. Taken all together, our findings implicate Nck1 in regulating the UPR, which secondary to obesity impairs glucose homeostasis and insulin actions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Retículo Endoplásmico/fisiología , Intolerancia a la Glucosa/genética , Intolerancia a la Glucosa/fisiopatología , Insulina/fisiología , Hígado/fisiología , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Animales , Glucemia/metabolismo , Western Blotting , Glucógeno/biosíntesis , Células HEK293 , Homeostasis/fisiología , Humanos , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Ratones Obesos , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Respuesta de Proteína Desplegada/genética , Respuesta de Proteína Desplegada/fisiología
19.
J Cell Sci ; 123(Pt 8): 1306-19, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20332103

RESUMEN

The polarised distribution of signals downstream from receptor tyrosine kinases (RTKs) regulates fundamental cellular processes that control cell migration, growth and morphogenesis. It is poorly understood how RTKs are involved in the localised signalling and actin remodelling required for these processes. Here, we show that the Gab1 scaffold is essential for the formation of a class of polarised actin microdomain, namely dorsal ruffles, downstream from the Met, EGF and PDGF RTKs. Gab1 associates constitutively with the actin-nucleating factor N-WASP. Following RTK activation, Gab1 recruits Nck, an activator of N-WASP, into a signalling complex localised to dorsal ruffles. Formation of dorsal ruffles requires interaction between Gab1 and Nck, and also requires functional N-WASP. Epithelial cells expressing Gab1DeltaNck (Y407F) exhibit decreased Met-dependent Rac activation, fail to induce dorsal ruffles, and have impaired cell migration and epithelial remodelling. These data show that a Gab1-Nck signalling complex interacts with several RTKs to promote polarised actin remodelling and downstream biological responses.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Extensiones de la Superficie Celular/enzimología , Proteínas Oncogénicas/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Extensiones de la Superficie Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Receptores ErbB/metabolismo , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas Oncogénicas/química , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-crk/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Tirosina/metabolismo , Proteína Neuronal del Síndrome de Wiskott-Aldrich/metabolismo , Proteínas de Unión al GTP rac/metabolismo
20.
Biochemistry ; 48(27): 6369-78, 2009 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-19505147

RESUMEN

The Eph family of tyrosine kinase receptors and their ligands, the ephrins, participates in the regulation of a wide variety of biological functions under normal and pathological conditions. During embryonic development, interactions between the ligands and receptors define tissue boundaries, guide migrating axons, and regulate angiogenesis, as well as bone morphogenesis. These molecules have also been shown to modify neural activity in the adult nervous system and influence tumor progression. However, the molecular mechanisms underlying these diverse functions are not completely understood. In this study, we conducted a yeast two-hybrid screen to identify molecules that physically interact with Eph receptors using the cytoplasmic domain of EphA3 as "bait". This study identified Nck1 as a strong binding partner of EphA3 as assayed using both GST fusion protein pull down and co-immunoprecipitation techniques. The interaction is mediated through binding of the Nck1 SH2 domain to the phosphotyrosine residue at position 602 (Y602) of the EphA3 receptor. The removal of the SH2 domain or the mutation of the Y602 residue abolishes the interaction. We further demonstrated that EphA3 activation inhibits cell migration and process outgrowth, and these inhibiting effects are partially alleviated by dominant-negative Nck1 mutants that lack functional SH2 or SH3 domains, but not by the wild-type Nck1 gene. These results suggest that Nck1 interacts with EphA3 to regulate cell migration and process retraction.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Movimiento Celular/fisiología , Proteínas Oncogénicas/fisiología , Receptor EphA3/fisiología , Proteínas Adaptadoras Transductoras de Señales/química , Inmunoprecipitación , Proteínas Oncogénicas/química , Transducción de Señal , Técnicas del Sistema de Dos Híbridos , Dominios Homologos src
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA