Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cell Signal ; 28(3): 177-189, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26739108

RESUMO

Extracellular signal-regulated kinases (ERKs) play important roles in proliferation, differentiation and gene expression. In our previous study, we demonstrated that both ERK5 and ERK1/2 were responsible for neurite outgrowth and tyrosine hydroxylase (TH) expression in rat pheochromocytoma cells (PC12) (J Biol Chem 284, 23,564-23,573, 2009). However, the functional differences between ERK5 and ERK1/2 signaling in neural differentiation remain unclear. In the present study, we show that ERK5, but not ERK1/2 regulates TH levels in rat sympathetic neurons. Furthermore, microarray analysis performed in PC12 cells using ERK5 and ERK1/2-specific inhibitors, identified ankyrin repeat domain 1 (ankrd1) as an ERK5-dependent and ERK1/2-independent gene. Here, we report a novel role of the ERK5/ankrd1 signaling in regulating TH levels and catecholamine biosynthesis. Ankrd1 mRNA was induced by nerve growth factor in time- and concentration-dependent manners. TH levels were reduced by ankrd1 knockdown with no changes in the mRNA levels, suggesting that ankrd1 was involved in stabilization of TH protein. Interestingly, ubiquitination of TH was enhanced and catecholamine biosynthesis was reduced by ankrd1 knockdown. Finally, we examined the relationship of ERK5 to TH levels in human adrenal pheochromocytomas. Whereas TH levels were correlated with ERK5 levels in normal adrenal medullas, ERK5 was down-regulated and TH was up-regulated in pheochromocytomas, indicating that TH levels are regulated by alternative mechanisms in tumors. Taken together, ERK5 signaling is required for catecholamine biosynthesis during neural differentiation, in part to induce ankrd1, and to maintain appropriate TH levels. This pathway is disrupted in pathological conditions.


Assuntos
Catecolaminas/biossíntese , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Proteínas Musculares/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Adolescente , Neoplasias das Glândulas Suprarrenais/metabolismo , Neoplasias das Glândulas Suprarrenais/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Catecolaminas/análise , Cromatografia Líquida de Alta Pressão , Regulação para Baixo/efeitos dos fármacos , Feminino , Humanos , Masculino , Microscopia de Fluorescência , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 7 Ativada por Mitógeno/genética , Proteínas Musculares/antagonistas & inibidores , Proteínas Musculares/genética , Fator de Crescimento Neural/farmacologia , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Células PC12 , Feocromocitoma/metabolismo , Feocromocitoma/patologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Proteínas Repressoras/antagonistas & inibidores , Proteínas Repressoras/genética , Transdução de Sinais/efeitos dos fármacos , Espectrometria de Massas em Tandem , Tirosina 3-Mono-Oxigenase/metabolismo , Regulação para Cima/efeitos dos fármacos
2.
Ann Clin Transl Neurol ; 1(11): 926-32, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25540807

RESUMO

A woman was isozygous for a novel mutation in the leukemia inhibitory factor receptor gene (LIFR) (c.2170C>G; p.Pro724Ala) which disrupts LIFR downstream signaling and results in Stüve-Wiedemann syndrome (STWS). She inherited two identical chromosomes 5 from her mother, heterozygous for the LIFR mutation. The presentation was typical for STWS, except there was no long bone dysplasia. Prominent cold-induced sweating and heat intolerance lead to an initial diagnosis of cold-induced sweating syndrome, excluded by exome sequencing. Skin biopsies provide the first human evidence of failed postnatal cholinergic differentiation of sympathetic neurons innervating sweat glands in cold-induced sweating, and of a neuropathy.

3.
Neurosci Lett ; 566: 1-5, 2014 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-24561183

RESUMO

The neurohormone leptin regulates energy homeostasis. Circulating levels of leptin secreted by adipose tissue act on hypothalamic neurons in the brain leading to decreased appetite and increased energy expenditure. Although leptin signaling in the central nervous system (CNS) is fundamental to its ability to regulate the body's metabolic balance, leptin also has a variety of effects in many peripheral tissues including the heart, the liver, and the sympathetic nervous system. Leptin stimulation of the hypothalamus can stimulate glucose uptake via the sympathetic nervous system in heart, muscle, and brown adipose tissue. Leptin receptors (Ob-Rb) are also expressed by peripheral sympathetic neurons, but their functional role is not clear. In this study, we found that leptin stimulates axonal growth of both adult and neonatal sympathetic neurons in vitro. Leptin stimulates acute activation of the transcription factor STAT3 via phosphorylation of tyrosine 705. STAT3 phosphorylation is required for leptin-stimulated sympathetic axon outgrowth. Thus, circulating levels of leptin may enhance sympathetic nerve innervation of peripheral tissues.


Assuntos
Axônios/fisiologia , Leptina/metabolismo , Gânglio Cervical Superior/fisiologia , Animais , Animais Recém-Nascidos , Leptina/farmacologia , Neurônios/fisiologia , Neurônios/ultraestrutura , Fosforilação , Ratos Sprague-Dawley , Fator de Transcrição STAT3/metabolismo , Gânglio Cervical Superior/crescimento & desenvolvimento , Gânglio Cervical Superior/ultraestrutura
4.
Exp Neurol ; 249: 111-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24013014

RESUMO

Development of cardiac sympathetic heterogeneity after myocardial infarction contributes to ventricular arrhythmias and sudden cardiac death. Regions of sympathetic hyperinnervation and denervation appear in the viable myocardium beyond the infarcted area. While elevated nerve growth factor (NGF) is implicated in sympathetic hyperinnervation, the mechanisms underlying denervation are unknown. Recent studies show that selective activation of the p75 neurotrophin receptor (p75(NTR)) in sympathetic neurons causes axon degeneration. We used mice that lack p75(NTR) to test the hypothesis that activation of p75(NTR) causes peri-infarct sympathetic denervation after cardiac ischemia-reperfusion. Wild type hearts exhibited sympathetic denervation adjacent to the infarct 24h and 3 days after ischemia-reperfusion, but no peri-infarct sympathetic denervation occurred in p75(NTR)-/- mice. Sympathetic hyperinnervation was found in the distal peri-infarct myocardium in both genotypes 3 days after MI, and hyperinnervation was increased in the p75(NTR)-/- mice. By 7 days after ischemia-reperfusion, cardiac sympathetic innervation density returned back to sham-operated levels in both genotypes, indicating that axonal pruning did not require p75(NTR). Prior studies revealed that proNGF is elevated in the damaged left ventricle after ischemia-reperfusion, as is mRNA encoding brain-derived neurotrophic factor (BDNF). ProNGF and BDNF preferentially bind p75(NTR) rather than TrkA on sympathetic neurons. Immunohistochemistry using Bdnf-HA mice confirmed the presence of BDNF or proBDNF in the infarct after ischemia-reperfusion. Thus, at least two p75(NTR) ligands are elevated in the left ventricle after ischemia-reperfusion where they may stimulate p75(NTR)-dependent denervation of peri-infarct myocardium. In contrast, NGF-induced sympathetic hyperinnervation in the distal peri-infarct ventricle is attenuated by p75(NTR).


Assuntos
Traumatismos Cardíacos/metabolismo , Ventrículos do Coração/inervação , Ventrículos do Coração/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Receptores de Fator de Crescimento Neural/deficiência , Simpatectomia/métodos , Animais , Feminino , Traumatismos Cardíacos/patologia , Ventrículos do Coração/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infarto do Miocárdio/patologia , Miocárdio/patologia
5.
Mol Cell Neurosci ; 56: 272-82, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23831387

RESUMO

The transcription factor STAT3 has been implicated in axon regeneration. Here we investigate a role for STAT3 in sympathetic nerve sprouting after myocardial infarction (MI) - a common injury in humans. We show that NGF stimulates serine phosphorylation (S727) of STAT3 in sympathetic neurons via ERK1/2, in contrast to cytokine phosphorylation of Y705. Maximal sympathetic axon regeneration in vitro requires phosphorylation of both S727 and Y705. Furthermore, cytokine signaling is necessary for NGF-induced sympathetic nerve sprouting in the heart after MI. Transfection studies in neurons lacking STAT3 suggest two independent pools of STAT3, phosphorylated on either S727 or Y705, that regulate sympathetic regeneration via both transcriptional and non-transcriptional means. Additional data identify STAT3-microtubule interactions that may complement the well-characterized role of STAT3 stimulating regeneration associated genes. These data show that STAT3 is critical for sympathetic axon regeneration in vitro and in vivo, and identify a novel non-transcriptional mode of action.


Assuntos
Fibras Adrenérgicas/fisiologia , Axônios/metabolismo , Citocinas/metabolismo , Fator de Crescimento Neural/farmacologia , Regeneração Nervosa , Fator de Transcrição STAT3/metabolismo , Fibras Adrenérgicas/efeitos dos fármacos , Fibras Adrenérgicas/metabolismo , Animais , Axônios/efeitos dos fármacos , Axônios/fisiologia , Células Cultivadas , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Microtúbulos/metabolismo , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Fator de Transcrição STAT3/genética , Transcrição Gênica
6.
Mol Cell Neurosci ; 46(3): 671-80, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21241805

RESUMO

Functional noradrenergic transmission requires the coordinate expression of enzymes involved in norepinephrine (NE) synthesis, as well as the norepinephrine transporter (NET) which removes NE from the synapse. Inflammatory cytokines acting through gp130 can suppress the noradrenergic phenotype in sympathetic neurons. This occurs in a subset of sympathetic neurons during development and also occurs in adult neurons after injury. For example, cytokines suppress noradrenergic function in sympathetic neurons after axotomy and during heart failure. The molecular basis for suppression of noradrenergic genes is not well understood, but previous studies implicated a reduction of Phox2a in cytokine suppression of dopamine beta hydroxylase. We used sympathetic neurons and neuroblastoma cells to investigate the role of Phox2a in cytokine suppression of NET transcription. Chromatin immunoprecipitation experiments revealed that Phox2a did not bind the NET promoter, and overexpression of Phox2a did not prevent cytokine suppression of NET transcription. Hand2 and Gata3 are transcription factors that induce noradrenergic genes during development and are present in mature sympathetic neurons. Both Hand2 and Gata3 were decreased by cytokines in sympathetic neurons and neuroblastoma cells. Overexpression of either Hand2 or Gata3 was sufficient to rescue NET transcription following suppression by cytokines. We examined expression of these genes following axotomy to determine if their expression was altered following nerve injury. NET and Hand2 mRNAs decreased significantly in sympathetic neurons 48 h after axotomy, but Gata3 mRNA was unchanged. These data suggest that cytokines can inhibit NET expression through downregulation of Hand2 or Gata3 in cultured sympathetic neurons, but axotomy in adult animals selectively suppresses Hand2 expression.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Citocinas/metabolismo , Neurônios/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Animais , Axotomia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Células Cultivadas , Fator Neurotrófico Ciliar/metabolismo , Fator de Transcrição GATA3/genética , Fator de Transcrição GATA3/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Neurônios/citologia , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/genética , Ratos , Ratos Sprague-Dawley
7.
J Neurochem ; 99(6): 1480-93, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17223854

RESUMO

The duration of intracellular signaling is thought to be a critical component in effecting specific biological responses. This paradigm is demonstrated by growth factor activation of the extracellular signal-regulated kinase (ERK) signaling cascade in the rat pheochromocytoma cell line (PC12 cells). In this model, sustained ERK activation induced by nerve growth factor (NGF) results in differentiation, whereas transient ERK activation induced by epidermal growth factor (EGF) results in proliferation in these cells. Recently, the immediate early gene product c-fos has been proposed to be a sensor for ERK signaling duration in fibroblasts. In this study, we ask whether this is true for NGF and EGF stimulation of PC12 cells. We show that NGF, but not EGF, can regulate both c-fos stability and activation in an ERK-dependent manner in PC12 cells. This is achieved through ERK-dependent phosphorylation of c-fos. Interestingly, distinct sites regulate enhanced stability and transactivation of c-fos. Phosphorylation of Thr325 and Thr331 are required for maximal NGF-dependent transactivation of c-fos. In addition, a consensus ERK binding site (DEF domain) is also required for c-fos transactivation. However, stability is controlled by ERK-dependent phosphorylation of Ser374, while phosphorylation of Ser362 can induce conformational changes in protein structure. We also provide evidence that sustained ERK activation is required for proper post-translational regulation of c-fos following NGF treatment of PC12 cells. Because these ERK-dependent phosphorylations are required for proper c-fos function, and occur sequentially, we propose that c-fos is a sensor for ERK signaling duration in the neuronal-like cell line PC12.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica/efeitos dos fármacos , Fator de Crescimento Neural/farmacologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ativação Transcricional/efeitos dos fármacos , Animais , Western Blotting/métodos , Butadienos/farmacologia , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Luciferases/metabolismo , Metaloproteinase 3 da Matriz/metabolismo , Mutagênese/fisiologia , Nitrilas/farmacologia , Células PC12 , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/genética , Ratos , Serina/metabolismo , Treonina/metabolismo , Fator de Transcrição AP-1/metabolismo , Transfecção/métodos
8.
J Neurophysiol ; 87(5): 2464-70, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11976383

RESUMO

Synaptic adaptations are thought to be an important component of the consequences of drug abuse. One such adaptation is an up-regulation of adenylyl cyclase that has been shown to increase transmitter release at several inhibitory synapses. In this study the effects of chronic morphine treatment were studied on mossy fiber synapses in the guinea pig hippocampus using extracellular field potential recordings. This opioid-sensitive synapse was chosen because of the known role of the adenylyl cyclase cascade in the regulation of glutamate release. Long-term potentiation (LTP) at the mossy fiber synapse was enhanced after chronic morphine treatment. In control animals, opioid antagonists increased LTP but had no effect in morphine-treated guinea pigs. In contrast, the long-lasting depression of transmission induced by a mGluR agonist and CA1 LTP were not altered. Chronic morphine treatment neither caused tolerance to mu- and kappa-receptor-mediated inhibition at the mossy fiber synapse nor modified total hippocampal dynorphin levels. The results suggest that the phasic inhibition of glutamate transmission mediated by endogenous opioids is reduced after chronic exposure to morphine.


Assuntos
Analgésicos Opioides/farmacologia , Morfina/farmacologia , Fibras Musgosas Hipocampais/efeitos dos fármacos , Peptídeos Opioides/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Animais , AMP Cíclico/metabolismo , Tolerância a Medicamentos/fisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Cobaias , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Fibras Musgosas Hipocampais/metabolismo , Síndrome de Abstinência a Substâncias/fisiopatologia , Transmissão Sináptica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA