Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Hepatology ; 72(4): 1204-1218, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31950520

RESUMO

BACKGROUND AND AIMS: Older patients with obesity/type II diabetes mellitus frequently present with advanced NASH. Whether this is due to specific molecular pathways that accelerate fibrosis during aging is unknown. Activation of the Src homology 2 domain-containing collagen-related (Shc) proteins and redox stress have been recognized in aging; however, their link to NASH has not been explored. APPROACH AND RESULTS: Shc expression increased in livers of older patients with NASH, as assessed by real time quantitative PCR (RT-qPCR) or western blots. Fibrosis, Shc expression, markers of senescence, and nicotinamide adenine dinucleotide phosphate, reduced form oxidases (NOXs) were studied in young/old mice on fast food diet (FFD). To inhibit Shc in old mice, lentiviral (LV)-short hairpin Shc versus control-LV were used during FFD. For hepatocyte-specific effects, floxed (fl/fl) Shc mice on FFD were injected with adeno-associated virus 8-thyroxine-binding globulin-Cre-recombinase versus control. Fibrosis was accelerated in older mice on FFD, and Shc inhibition by LV in older mice or hepatocyte-specific deletion resulted in significantly improved inflammation, reduction in senescence markers in older mice, lipid peroxidation, and fibrosis. To study NOX2 activation, the interaction of p47phox (NOX2 regulatory subunit) and p52Shc was evaluated by proximity ligation and coimmunoprecipitations. Palmitate-induced p52Shc binding to p47phox , activating the NOX2 complex, more so at an older age. Kinetics of binding were assessed in Src homology 2 domain (SH2) or phosphotyrosine-binding (PTB) domain deletion mutants by biolayer interferometry, revealing the role of SH2 and the PTB domains. Lastly, an in silico model of p52Shc/p47phox interaction using RosettaDock was generated. CONCLUSIONS: Accelerated fibrosis in the aged is modulated by p52Shc/NOX2. We show a pathway for direct activation of the phagocytic NOX2 in hepatocytes by p52Shc binding and activating the p47phox subunit that results in redox stress and accelerated fibrosis in the aged.


Assuntos
Envelhecimento/metabolismo , NADPH Oxidase 2/fisiologia , Hepatopatia Gordurosa não Alcoólica/etiologia , Animais , Hepatócitos/metabolismo , Humanos , Cirrose Hepática/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Adaptadoras da Sinalização Shc/antagonistas & inibidores , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Domínios de Homologia de src
2.
Subcell Biochem ; 81: 201-230, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27830506

RESUMO

For the past century, vitamin A has been considered to serve as a precursor for retinoids that facilitate vision or as a precursor for retinoic acid (RA), a signaling molecule that modulates gene expression. However, vitamin A circulates in plasma at levels that far exceed the amount needed for vision or the synthesis of nanomolar levels of RA, and this suggests that vitamin A alcohol (i.e. retinol) may possess additional biological activity. We have pursued this question for the last 20 years, and in this chapter, we unfold the story of our quest and the data that support a novel and distinct role for vitamin A (alcohol) action. Our current model supports direct binding of vitamin A to the activation domains of serine/threonine kinases, such as protein kinase C (PKC) and Raf isoforms, where it is involved in redox activation of these proteins. Redox activation of PKCs was first described by the founders of the PKC field, but several hurdles needed to be overcome before a detailed understanding of the biochemistry could be provided. Two discoveries moved the field forward. First, was the discovery that the PKCδ isoform was activated by cytochrome c, a protein with oxidoreduction activity in mitochondria. Second, was the revelation that both PKCδ and cytochrome c are tethered to p66Shc, an adapter protein that brings the PKC zinc-finger substrate into close proximity with its oxidizing partner. Detailed characterization of the PKCδ signalosome complex was made possible by the work of many investigators. Our contribution was determining that vitamin A is a vital co-factor required to support an unprecedented redox-activation mechanism. This unique function of vitamin A is the first example of a general system that connects the one-electron redox chemistry of a heme protein (cytochrome c) with the two-electron chemistry of a classical phosphoprotein (PKCδ). Furthermore, contributions to the regulation of mitochondrial energetics attest to biological significance of vitamin A alcohol action.


Assuntos
Mitocôndrias/metabolismo , Proteína Quinase C-delta/metabolismo , Vitamina A/fisiologia , Animais , Citocromos c/metabolismo , Metabolismo Energético , Ativação Enzimática/efeitos dos fármacos , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/fisiologia , Previsões , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/fisiologia , Humanos , Mitocôndrias/enzimologia , Oxirredução , Estresse Oxidativo , Ligação Proteica , Isoformas de Proteínas/metabolismo , Proteína Quinase C-épsilon/fisiologia , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Vitamina A/análogos & derivados , Dedos de Zinco , Quinases raf/metabolismo
3.
Eur Heart J ; 36(48): 3392-403, 2015 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-26543043

RESUMO

Western societies are aging due to an increasing life span, decreased birth rates, and improving social and health conditions. On the other hand, the prevalence of cardiovascular (CV) and cerebrovascular (CBV) diseases rises with age. Thus, in view of the ongoing aging pandemic, it is appropriate to better understand the molecular pathways of aging as well as age-associated CV and CBV diseases. Oxidative stress contributes to aging of organs and the whole body by an accumulation of reactive oxygen species promoting oxidative damage. Indeed, increased oxidative stress produced in the mitochondria and cytosol of heart and brain is a common denominator to almost all CV and CBV diseases. The mitochondrial adaptor protein p66(Shc) and the family of deacetylase enzymes, the sirtuins, regulate the aging process, determine lifespan of many species and are involved in CV diseases. GDF11, a member of TGFß superfamily with homology to myostatin also retards the aging process via yet unknown mechanisms. Recent evidence points towards a promising role of this novel 'rejuvenation' factor in reducing age-related heart disease. Finally, telomere length is also involved in aging and the development of age-related CV dysfunction. This review focuses on the latest scientific advances in understanding age-related changes of the CV and CBV system, as well as delineating potential novel therapeutic targets derived from aging research for CV and CBV diseases.


Assuntos
Envelhecimento/fisiologia , Doenças Cardiovasculares/etiologia , Envelhecimento/genética , Animais , Proteínas Morfogenéticas Ósseas/fisiologia , Restrição Calórica , Radicais Livres/metabolismo , Fatores de Diferenciação de Crescimento/fisiologia , Humanos , Longevidade/fisiologia , Mitocôndrias/fisiologia , Modelos Biológicos , Estresse Oxidativo/fisiologia , Proteínas Proto-Oncogênicas c-jun/fisiologia , Comportamento de Redução do Risco , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Sirtuínas/fisiologia , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Encurtamento do Telômero/fisiologia , Rigidez Vascular/fisiologia
4.
Proc Natl Acad Sci U S A ; 112(44): E6068-77, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26487680

RESUMO

Despite extensive efforts to understand the monogenic contributions to perturbed glucose homeostasis, the complexity of genetic events that fractionally contribute to the spectrum of this pathology remain poorly understood. Proper maintenance of glucose homeostasis is the central feature of a constellation of comorbidities that define the metabolic syndrome. The ability of the liver to balance carbohydrate uptake and release during the feeding-to-fasting transition is essential to the regulation of peripheral glucose availability. The liver coordinates the expression of gene programs that control glucose absorption, storage, and secretion. Herein, we demonstrate that Steroid Receptor Coactivator 2 (SRC-2) orchestrates a hierarchy of nutritionally responsive transcriptional complexes to precisely modulate plasma glucose availability. Using DNA pull-down technology coupled with mass spectrometry, we have identified SRC-2 as an indispensable integrator of transcriptional complexes that control the rate-limiting steps of hepatic glucose release and accretion. Collectively, these findings position SRC-2 as a major regulator of polygenic inputs to metabolic gene regulation and perhaps identify a previously unappreciated model that helps to explain the clinical spectrum of glucose dysregulation.


Assuntos
Glucose/metabolismo , Homeostase/fisiologia , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Animais , Glucoquinase/genética , Glucoquinase/metabolismo , Camundongos , Camundongos Knockout , Transcrição Gênica
5.
Biochem Biophys Res Commun ; 463(4): 673-8, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26043699

RESUMO

8-Chloro-cyclic AMP (8-Cl-cAMP) is a cyclic AMP analog that induces growth inhibition and apoptosis in a broad spectrum of cancer cells. Previously, we found that 8-Cl-cAMP-induced growth inhibition is mediated by AMP-activated protein kinase (AMPK) as well as p38 mitogen-activated protein kinase (p38 MAPK). To identify downstream mediators of the 8-Cl-cAMP signaling, we performed co-immunoprecipitation combined with mass spectrometry using the anti-AMPK or p38 MAPK antibodies. Through this approach, SHC1 was identified as one of the binding partners of p38 MAPK. SHC1 phosphorylation was suppressed by 8-Cl-cAMP in HeLa and MCF7 cancer cells, which was mediated by its metabolites, 8-Cl-adenosine and 8-Cl-ATP; however, 8-Cl-cAMP showed no effect on SHC1 phosphorylation in normal human fibroblasts. SHC1 siRNA induced AMPK and p38 MAPK phosphorylation and growth inhibition in cancer cells, and SHC1 overexpression re-sensitized human foreskin fibroblasts to the 8-Cl-cAMP treatment. SHC1 phosphorylation was unaffected by Compound C (an AMPK inhibitor) and SB203580 (a p38 MAPK inhibitor), which suggests that SHC1 is upstream of AMPK and p38 MAPK in the 8-Cl-cAMP-stimulated signaling cascade. On the basis of these findings, we conclude that SHC1 functions as a sensor during the 8-Cl-cAMP-induced growth inhibition in SHC1-overexpressing cancer cells.


Assuntos
8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , Proteínas Adaptadoras da Sinalização Shc/fisiologia , 8-Bromo Monofosfato de Adenosina Cíclica/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Adenilato Quinase/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Espectrometria de Massas , Neoplasias/enzimologia , Neoplasias/patologia , Neoplasias/fisiopatologia , Fosforilação , RNA Interferente Pequeno/genética , Proteínas Adaptadoras da Sinalização Shc/genética , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Eur Heart J ; 36(25): 1590-600, 2015 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-25904764

RESUMO

AIM: Constitutive genetic deletion of the adaptor protein p66(Shc) was shown to protect from ischaemia/reperfusion injury. Here, we aimed at understanding the molecular mechanisms underlying this effect in stroke and studied p66(Shc) gene regulation in human ischaemic stroke. METHODS AND RESULTS: Ischaemia/reperfusion brain injury was induced by performing a transient middle cerebral artery occlusion surgery on wild-type mice. After the ischaemic episode and upon reperfusion, small interfering RNA targeting p66(Shc) was injected intravenously. We observed that post-ischaemic p66(Shc) knockdown preserved blood-brain barrier integrity that resulted in improved stroke outcome, as identified by smaller lesion volumes, decreased neurological deficits, and increased survival. Experiments on primary human brain microvascular endothelial cells demonstrated that silencing of the adaptor protein p66(Shc) preserves claudin-5 protein levels during hypoxia/reoxygenation by reducing nicotinamide adenine dinucleotide phosphate oxidase activity and reactive oxygen species production. Further, we found that in peripheral blood monocytes of acute ischaemic stroke patients p66(Shc) gene expression is transiently increased and that this increase correlates with short-term neurological outcome. CONCLUSION: Post-ischaemic silencing of p66(Shc) upon reperfusion improves stroke outcome in mice while the expression of p66(Shc) gene correlates with short-term outcome in patients with ischaemic stroke.


Assuntos
Lesões Encefálicas/prevenção & controle , Inativação Gênica/fisiologia , Traumatismo por Reperfusão/prevenção & controle , Proteínas Adaptadoras da Sinalização Shc/genética , Acidente Vascular Cerebral/prevenção & controle , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Animais , Barreira Hematoencefálica/fisiologia , Estudos de Casos e Controles , Células Cultivadas , Claudina-5/efeitos dos fármacos , Células Endoteliais/fisiologia , Feminino , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Infarto da Artéria Cerebral Média , Pós-Condicionamento Isquêmico/métodos , Masculino , Camundongos Endogâmicos C57BL , Microcirculação/fisiologia , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Espécies Reativas de Oxigênio/farmacologia , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Resultado do Tratamento
7.
Zhongguo Dang Dai Er Ke Za Zhi ; 17(3): 275-80, 2015 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-25815500

RESUMO

OBJECTIVE: To explore the roles of PKCß/P66Shc oxidative stress signal pathway in mediating hyperoxia-induced reactive oxgen species (ROS) production in alveolar epithelial cells (A549) and the protective effects of PKCß inhibitor on hyperoxia-induced injuries of alveolar epithelial cells. METHODS: A549 cells were cultured in vitro and randomly divided into three groups: control, hyperoxia and PKCß inhibitor LY333531 treatment. The hyperoxia group was exposed to a mixture of O2 (950 mL/L) and CO2 (50 mL/L) for 10 minutes and then cultured in a closed environment. The LY333531 group was treated with PKCß inhibitor LY333531 of 10 µmol/L for 24 hours before hyperoxia induction. Cells were collected 24 hours after culture and the levels of PKCß, Pin1, P66Shc and P66Shc-Ser36 were detected by Western blot. The intracellular translocation of P66Shc, the production of ROS and cellular mitochondria membrane potential were measured using the confocal microscopy. RESULTS: Compared with the control group, the levels of PKCß, Pin1, P66Shc and P-P66Shc-Ser36 in A549 cells 24 hours after culture increased significantly in the hyperoxia group. These changes in the hyperoxia group were accompanied with an increased translocation rate of P66Shc from cytoplasm into mitochondria, an increased production of mitochondrial ROS, and a reduced mitochondrial membrane potential. Compared with the hyperoxia group, the levels of Pin1, P66Shc and P66Shc-Ser36 in A549 cells, the translocation rate of P66Shc from cytoplasm into mitochondria and the production of mitochondrial ROS decreased significantly, while the mitochondrial membrane potential increased significantly in the LY333531 treatment group. However, there were significant differences in the above mentioned measurements between the LY333531 treatment and control groups. CONCLUSIONS: Hyperoxia can increase the expression of PKCß in alveolar epithelial cells and production of mitochondrial ROS and decrease mitochondrial membrane potential. PKCß inhibitor LY333531 can partially disrupt these changes and thus alleviate the hyperoxia-induced alveolar epithelial cell injury.


Assuntos
Hipóxia Celular , Estresse Oxidativo , Proteína Quinase C beta/fisiologia , Alvéolos Pulmonares/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Transdução de Sinais/fisiologia , Células Cultivadas , Células Epiteliais/metabolismo , Humanos , Indóis/farmacologia , Maleimidas/farmacologia , Alvéolos Pulmonares/citologia , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
8.
Cell Death Differ ; 22(7): 1192-202, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25613377

RESUMO

Cancer stem cells (CSCs) have been implicated in the initiation and maintenance of tumour growth as well as metastasis. Recent reports link stemness to epithelial-mesenchymal transition (EMT) in cancer. However, there is still little knowledge about the molecular markers of those events. In silico analysis of RNA profiles of 36 pancreatic ductal adenocarcinomas (PDAC) reveals an association of the expression of CD95 with EMT and stemness that was validated in CSCs isolated from PDAC surgical specimens. CD95 expression was also higher in metastatic pancreatic cells than in primary PDAC. Pharmacological inhibition of CD95 activity reduced PDAC growth and metastasis in CSC-derived xenografts and in a murine syngeneic model. On the mechanistic level, Sck was identified as a novel molecule indispensable for CD95's induction of cell cycle progression. This study uncovers CD95 as a marker of EMT and stemness in PDAC. It also addresses the molecular mechanism by which CD95 drives tumour growth and opens tantalizing therapeutic possibilities in PDAC.


Assuntos
Carcinoma Ductal Pancreático/fisiopatologia , Metástase Neoplásica , Neoplasias Pancreáticas/fisiopatologia , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Receptor fas/fisiologia , Animais , Biomarcadores Tumorais , Carcinoma Ductal Pancreático/metabolismo , Transição Epitelial-Mesenquimal , Feminino , Humanos , Camundongos , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais , Proteína 2 de Transformação que Contém Domínio 2 de Homologia de Src , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Eur J Clin Invest ; 45 Suppl 1: 25-31, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25524583

RESUMO

The adaptor protein p66Shc links membrane receptors to intracellular signalling pathways and has the potential to respond to energy status changes and regulate mitogenic signalling. Initially reported to mediate growth signals in normal and cancer cells, p66Shc has also been recognized as a pro-apoptotic protein involved in the cellular response to oxidative stress. Moreover, it is a key element in processes such as cancer cell proliferation, tumor progression, metastasis and metabolic reprogramming. Recent findings on the role of p66Shc in the above-mentioned processes have been obtained through the use of various tumor cell types, including prostate, breast, ovarian, lung, colon, skin and thyroid cancer cells. Interestingly, the impact of p66Shc on the proliferation rate was mainly observed in prostate tumors, while its impact on metastasis was mainly found in breast cancers. In this review, we summarize the current knowledge about the possible roles of p66Shc in different cancers.


Assuntos
Apoptose/fisiologia , Neoplasias/metabolismo , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Proliferação de Células/fisiologia , Humanos , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Transdução de Sinais , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
10.
Ageing Res Rev ; 19: 34-42, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25462193

RESUMO

To date, most studies of Shc family of signaling adaptor proteins have been focused on the near-ubiquitously expressed ShcA, indicating its relevance to age-related diseases and longevity. Although the role of the neuronal ShcC protein is much less investigated, accumulated evidence suggests its importance for neuroprotection against such aging-associated conditions as brain ischemia and oxidative stress. Here, we summarize more than decade of studies on the ShcC expression and function in normal brain, age-related brain pathologies and immune disorders with a focus on the interactions of ShcC with signaling proteins/pathways, and the possible implications of these interactions for changes associated with aging.


Assuntos
Envelhecimento/fisiologia , Encéfalo/crescimento & desenvolvimento , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Animais , Neoplasias Encefálicas/fisiopatologia , Humanos
11.
Oncogene ; 34(13): 1729-35, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-24837366

RESUMO

The ShcA adaptor protein is engaged by numerous receptor tyrosine kinases (RTKs) in breast cancer cells. Once activated, RTKs phosphorylate three key tyrosine phosphorylation sites (Y239, Y240 and Y317) within ShcA that creates a docking site for Grb2/SOS and Grb2/Gab-containing complexes to activate the MAPK and AKT signaling pathways, respectively. We previously demonstrated that a tyrosine to phenylalanine substitution of the ShcA tyrosine phosphorylation sites (Shc3F-Y239/240/313F) significantly impairs breast tumor growth and angiogenesis in transgenic mouse models, in part, through the regulation of vascular endothelial growth factor (VEGF) production. Despite this fact, the underlying molecular mechanisms by which ShcA transduces pro-tumorigenic signals in breast cancer cells remain poorly defined. In this study, we demonstrate that ShcA-dependent activation of AKT, but not the RAS/MAPK pathway, induces VEGF production by bolstering VEGF mRNA translation. Accordingly, ShcA drives breast tumor growth and angiogenesis in vivo in a 4E-BP-dependent manner. These findings establish ShcA as a biological bridge that links AKT activation downstream of RTKs to cap-dependent VEGF mRNA translation in order to promote mammary tumorigenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Neoplasias da Mama/irrigação sanguínea , Neovascularização Patológica/etiologia , Fosfoproteínas/fisiologia , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas c-akt/fisiologia , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Proteínas de Ciclo Celular , Feminino , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/fisiologia , RNA Mensageiro/genética , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Proteína 3 de Transformação que Contém Domínio 2 de Homologia de Src , Fator A de Crescimento do Endotélio Vascular/biossíntese
12.
Atherosclerosis ; 236(2): 426-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25150941

RESUMO

OBJECTIVE: Obesity-induced insulin resistance (IR) precipitates cardiovascular disease (CVD). Impairment of insulin signalling in the endothelium is emerging as a trigger of IR but the underlying mechanisms remain elusive. The mitochondrial adaptor p66(Shc) drives endothelial dysfunction via reactive oxygen species (ROS) generation. This study investigates p66(Shc) role in obesity-induced impairment of endothelial insulin signalling. METHODS: All experiments were performed in leptin-deficient (Lep(Ob/Ob)) and wild-type (WT) mice. RESULTS: Endothelium-dependent relaxations to insulin were blunted in Lep(Ob/Ob) as compared to WT. Interestingly, in vivo gene silencing of p66(Shc) restored insulin response via IRS-1/Akt/eNOS pathway. Furthermore, p66(Shc) knockdown in endothelial cells isolated from Lep(Ob/Ob) mice attenuated ROS production, free fatty acids (FFA) oxidation and prevented dysregulation of redox-sensitive pathways such as nuclear factor-kappa-B (NF-kB), AGE precursor methylglyoxal and PGI2 synthase. CONCLUSIONS: Targeting endothelial p66(Shc) may represent a promising strategy to prevent IR and CVD in obese individuals.


Assuntos
Endotélio Vascular/fisiopatologia , Resistência à Insulina/fisiologia , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Animais , Sistema Enzimático do Citocromo P-450/metabolismo , Ácidos Graxos não Esterificados/metabolismo , Insulina/farmacologia , Oxirredutases Intramoleculares/metabolismo , Leptina/deficiência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Obesos , Terapia de Alvo Molecular , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Obesidade/genética , Obesidade/metabolismo , Oxirredução , Fenilefrina/farmacologia , Aldeído Pirúvico/metabolismo , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Espécies Reativas de Oxigênio , Proteínas Adaptadoras da Sinalização Shc/antagonistas & inibidores , Proteínas Adaptadoras da Sinalização Shc/deficiência , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Superóxidos/metabolismo , Vasodilatação/efeitos dos fármacos
13.
Int J Cardiol ; 175(3): 446-50, 2014 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-25012499

RESUMO

BACKGROUND: Aging is an independent risk factor for cardiovascular and cerebrovascular disease. To date, little is known about the mechanisms of aging of cerebral arteries and whether the aging gene p66(Shc) is implicated in it. The present study was designed to assess age-induced vascular dysfunction in cerebral and systemic arteries of wild type (wt) and p66(Shc-/-) mice. METHODS: Basilar arteries and size matched second order femoral arteries of 3-month (3M), 6-month (6M) and 2-year old (2Y) mice were studied in wt and p66(Shc-/-) mice. To assess vascular function, arterial rings mounted in a myograph for isometric tension recordings were exposed to increasing concentrations of acetylcholine and sodium nitroprusside. Reactive oxygen species (ROS) generation was assessed in femoral and basilar arteries using the spin trap 1-hydroxy-3-methoxycarbonyl-2,2,5,5-tetramethyl-pyrrolidine. RESULTS: In wt mice, endothelial function of the femoral artery was not affected by age unlike in the basilar artery where an age-dependent dysfunction was observed. In p66(Shc-/-) a similar response was observed in the femoral artery; however, age-dependent endothelial dysfunction of the basilar artery was blunted as compared to wt. Levels of ROS were comparable in the femoral arteries of 3M and 2Y of wt and p66(Shc-/-) mice. Differently, ROS levels in the basilar artery of wt mice were strongly increased by age unlike in p66(Shc-/-) mice where they remained comparable irrespective of age. CONCLUSIONS: Endothelial function in cerebral arteries, but not in size-matched systemic ones, is heavily impaired by aging. This process is paralleled by an increased ROS production and is mediated by the p66(Shc) gene.


Assuntos
Envelhecimento/metabolismo , Transtornos Cerebrovasculares/metabolismo , Endotélio Vascular/metabolismo , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Envelhecimento/patologia , Animais , Artéria Basilar/metabolismo , Artéria Basilar/patologia , Transtornos Cerebrovasculares/patologia , Endotélio Vascular/patologia , Artéria Femoral/metabolismo , Artéria Femoral/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Cultura de Órgãos , Proteínas Adaptadoras da Sinalização Shc/biossíntese , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
14.
Mol Biol Cell ; 25(13): 2116-27, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24807908

RESUMO

The mammalian Shc family, composed of p46, p52, and p66 isoforms, serves as an adaptor protein in cell growth and stress response. p66Shc was shown to be a negative lifespan regulator by acting as a prooxidant protein in mitochondria; however, the regulatory mechanisms of p66Shc expression and function are incompletely understood. This study provides evidence for new features of p66Shc serving as an antioxidant and critical protein in cell differentiation. Unique among the Shc family, transcription of p66Shc is activated through the antioxidant response element (ARE)-nuclear factor erythroid 2-related factor 2 (Nrf2) pathway in K562 human erythroleukemia and other cell types after treatment with hemin, an iron-containing porphyrin. Phosphorylated p66Shc at Ser-36, previously reported to be prone to mitochondrial localization, is increased by hemin treatment, but p66Shc remains exclusively in the cytoplasm. p66Shc knockdown inhibits hemin-induced erythroid differentiation, in which reactive oxygen species production and apoptosis are significantly enhanced in conjunction with suppression of other ARE-dependent antioxidant genes. Conversely, p66Shc overexpression is sufficient for inducing erythroid differentiation. Collectively these results demonstrate the isoform-specific regulation of the Shc gene by the Nrf2-ARE pathway and a new antioxidant role of p66Shc in the cytoplasm. Thus p66Shc is a bifunctional protein involved in cellular oxidative stress response and differentiation.


Assuntos
Antioxidantes/fisiologia , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Ativação Transcricional , Apoptose , Diferenciação Celular , Células Eritroides/metabolismo , Hemina/farmacologia , Humanos , Células K562 , Mitocôndrias/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Fosforilação , Isoformas de Proteínas/fisiologia , Processamento de Proteína Pós-Traducional , Transporte Proteico , Espécies Reativas de Oxigênio/metabolismo , Elementos de Resposta , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Transcrição Gênica
15.
ScientificWorldJournal ; 2014: 387640, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24757420

RESUMO

Intestinal ischemia-reperfusion (I/R) injury is a serious clinical pathophysiological process that may result in acute local intestine and remote liver injury. Protocatechuic acid (PCA), which has been widely studied as a polyphenolic compound, induces expression of antioxidative genes that combat oxidative stress and cell apoptosis. In this study, we investigated the effect of PCA pretreatment for protecting intestinal I/R-induced local intestine and remote liver injury in mice. Intestinal I/R was established by superior mesenteric artery occlusion for 45 min followed by reperfusion for 90 min. After the reperfusion period, PCA pretreatment markedly alleviated intestine and liver injury induced by intestinal I/R as indicated by histological alterations, decreases in serological damage parameters and nuclear factor-kappa B and phospho-foxo3a protein expression levels, and increases in glutathione, glutathione peroxidase, manganese superoxide dismutase protein expression, and Bcl-xL protein expression in the intestine and liver. These parameters were accompanied by PCA-induced adaptor protein p66shc suppression. These results suggest that PCA has a significant protective effect in the intestine and liver following injury induced by intestinal I/R. The protective effect of PCA may be attributed to the suppression of p66shc and the regulation of p66shc-related antioxidative and antiapoptotic factors.


Assuntos
Hidroxibenzoatos/uso terapêutico , Mucosa Intestinal/metabolismo , Hepatopatias/metabolismo , Traumatismo por Reperfusão/metabolismo , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Transdução de Sinais/fisiologia , Animais , Hidroxibenzoatos/farmacologia , Intestinos/irrigação sanguínea , Intestinos/efeitos dos fármacos , Hepatopatias/tratamento farmacológico , Masculino , Camundongos , Camundongos Endogâmicos ICR , Substâncias Protetoras/farmacologia , Substâncias Protetoras/uso terapêutico , Distribuição Aleatória , Traumatismo por Reperfusão/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
16.
J Gerontol A Biol Sci Med Sci ; 69(10): 1177-85, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24336818

RESUMO

The signaling molecule p66Shc is often described as a longevity protein. This conclusion is based on a single life span study that used a small number of mice. The purpose of the present studies was to measure life span in a sufficient number of mice to determine if longevity is altered in mice with decreased Shc levels (ShcKO). Studies were completed at UC Davis and the European Institute of Oncology (EIO). At UC Davis, male C57BL/6J WT and ShcKO mice were fed 5% or 40% calorie-restricted (CR) diets. In the 5% CR group, there was no difference in survival curves between genotypes. There was also no difference between genotypes in prevalence of neoplasms or other measures of end-of-life pathology. At 40% calorie restriction group, 70th percentile survival was increased in ShcKO, while there were no differences between genotypes in median or subsequent life span measures. At EIO, there was no increase in life span in ShcKO male or female mice on C57BL/6J, 129Sv, or hybrid C57BL/6J-129Sv backgrounds. These studies indicate that p66Shc is not a longevity protein. However, additional studies are needed to determine the extent to which Shc proteins may influence the onset and severity of specific age-related diseases.


Assuntos
Longevidade , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Criação de Animais Domésticos , Animais , Restrição Calórica , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade da Espécie , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
17.
Am J Physiol Lung Cell Mol Physiol ; 306(4): L316-25, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24375794

RESUMO

Many signaling pathways are mediated by Shc adapter proteins that, in turn, are expressed as three isoforms with distinct functions. The p66(Shc) isoform antagonizes proliferation, regulates oxidative stress, and mediates apoptosis. It is highly expressed in the canalicular but not the later stages of mouse lung development, and its expression persists in bronchopulmonary dysplasia, a chronic disease associated with premature birth. These observations suggest that p66(Shc) has a developmental function. However, constitutive p66(Shc) deletion yields no morphological phenotype, and the structure of the Shc gene precludes its inducible deletion. To elucidate its function in lung development, we transfected p66(Shc) or nonsilencing small-interfering RNA (siRNA) into the epithelia of embryonic day 11 mouse lungs that were then cultured for 3 days and analyzed morphometrically. To assess cellular proliferation and epithelial differentiation, lung explants were immunostained and immunoblotted for p66(Shc), proliferating cell nuclear antigen (PCNA), the proximal airway differentiation antigens Clara cell 10-kDa protein (CC10) and thyroid transcription factor (TTF)-1, and the alveolar surfactant proteins (SP)-A, -B, and -C. Explants transfected with nonsilencing siRNA demonstrated specific epithelial uptake and normal morphological development relative to uninjected controls. In contrast, transfection with p66(Shc) siRNA significantly increased lumenal cross-sectional areas, decreased branching, and increased epithelial proliferation (P < 0.05 for all). Relative to controls, the expression of SP-B, SP-C, CC10, and TTF-1 was decreased by p66(Shc) knockdown. SP-A was not expressed in either control or treated lungs. These data suggest that p66(Shc) attenuates epithelial proliferation while promoting both distal and proximal epithelial maturation.


Assuntos
Células Epiteliais Alveolares/fisiologia , Pulmão/embriologia , Morfogênese , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Células 3T3 , Animais , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Pulmão/citologia , Pulmão/metabolismo , Camundongos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteína B Associada a Surfactante Pulmonar/metabolismo , RNA Interferente Pequeno/genética , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Técnicas de Cultura de Tecidos , Fatores de Transcrição , Uteroglobina/metabolismo
18.
Neuromolecular Med ; 16(1): 137-49, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24085465

RESUMO

We have previously shown the involvement of p66shc in mediating apoptosis. Here, we demonstrate the novel mechanism of ß-Amyloid-induced toxicity in the mammalian cells. ß-Amyloid leads to the phosphorylation of p66shc at the serine 36 residue and activates MKK6, by mediating the phosphorylation at serine 207 residue. Treatment of cells with antioxidants blocks ß-Amyloid-induced serine phosphorylation of MKK6, reactive oxygen species (ROS) generation, and hence protected cells against ß-Amyloid-induced cell death. Our results indicate that serine phosphorylation of p66shc is carried out by active MKK6. MKK6 knock-down resulted in decreased serine 36 phosphorylation of p66shc. Co-immunoprecipitation results demonstrate a direct physical association between p66shc and WT MKK6, but not with its mutants. Increase in ß-Amyloid-induced ROS production was observed in the presence of MKK6 and p66shc, when compared to triple mutant of MKK6 (inactive) and S36 mutant of p66shc. ROS scavengers and knock-down against p66shc, and MKK6 significantly decreased the endogenous level of active p66shc, ROS production, and cell death. Finally, we show that the MKK6-p66shc complex mediates ß-Amyloid-evoked apoptotic cell death.


Assuntos
Apoptose/fisiologia , MAP Quinase Quinase 6/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/efeitos dos fármacos , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Peptídeos beta-Amiloides/toxicidade , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Glioblastoma/patologia , Humanos , MAP Quinase Quinase 6/antagonistas & inibidores , MAP Quinase Quinase 6/genética , Sistema de Sinalização das MAP Quinases , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/genética , Neurônios/patologia , Estresse Oxidativo , Fragmentos de Peptídeos/toxicidade , Fosforilação , Fosfosserina/química , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Ratos , Espécies Reativas de Oxigênio , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
20.
Am J Nephrol ; 38(3): 226-32, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23988748

RESUMO

BACKGROUND/AIMS: Adult and childhood obesity is an independent risk factor in development of chronic kidney disease (CKD) and its progression to end-stage kidney disease. Pathologic consequences of obesity include non-esterified fatty acid-induced oxidative stress and consequent injury. Since the serine36-phosphorylated p66shc is a newly recognized mediator of oxidative stress and kidney injury, we studied its role in oleic acid (OA)-induced production of reactive oxygen species (ROS), mitochondrial depolarization and injury in cultured renal proximal tubule cells. METHODS: Renal proximal tubule cells were used and treated with OA: ROS production, mitochondrial depolarization as well as injury were determined. Transcriptional effects of OA on the p66shc gene were determined in a reporter luciferase assay. The role of p66shc in adverse effects of OA was determined using knockdown, p66shc serine36 phosphorylation and cytochrome c binding-deficient cells. RESULTS: We found that OA increased ROS production via the mitochondria - and to a less extent via the NADPH oxidase - resulting in ROS-dependent mitochondrial depolarization and consequent injury. Interestingly, OA also stimulated the promoter of p66shc. Hence, knockdown of p66shc, impairment its Ser36 phosphorylation (mutation of Ser36 residue to alanine) or cytochrome c binding (W134F mutation) significantly attenuated OA-dependent lipotoxicity. CONCLUSION: These results offer a novel mechanism by which obesity may lead to renal tubular injury and consequently development of CKD. Manipulation of this pathway may offer therapeutic means to ameliorate obesity-dependent renal lipotoxicity.


Assuntos
Rim/efeitos dos fármacos , Rim/metabolismo , Ácido Oleico/toxicidade , Proteínas Adaptadoras da Sinalização Shc/fisiologia , Animais , Linhagem Celular , Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Túbulos Renais/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Camundongos , Mitocôndrias/metabolismo , NADPH Oxidases/metabolismo , Obesidade/metabolismo , Fosforilação , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Espécies Reativas de Oxigênio , Fatores de Risco , Serina/química , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA