Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Med Sci Monit ; 25: 1960-1969, 2019 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-30875363

RESUMO

BACKGROUND This study aimed to investigate the effects of abdominal aortic transplantation of bone marrow mesenchymal stem cells (BMMSCs) on the expression of inflammatory cytokines in a rat model of spinal cord ischemia-reperfusion injury. MATERIAL AND METHODS Adult female Sprague-Dawley rats (N=160) were divided into five groups: the sham operation group (N-32); the control group (N=32); the BMMSC transplanted group (N=32); the anti-ciliary neurotrophic factor (CNTF)-treated BMMSC transplanted group (N=32); and the CNTF small interfering RNA (siRNA)-treated BMMSC transplanted group (N=32). Motor behavior was assessed using the Basso, Beattie, and Bresnahan (BBB) locomotor scale. Motor evoked potentials (MEPs) and cortical somatosensory evoked potentials (CSEPs) were measured. Immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR), and Western blot analysis evaluated the expression of spinal inflammatory cytokines. RESULTS Following surgery, compared with the control group the findings in the BMMSC transplant groups included significantly increased BBB scores; the latency and the amplitude of MEP and CSEP were reduced and increased, respectively; spinal neuronal necrosis was reduced; the number of normal neurons increased; CNTF mRNA and protein expression levels increased; expression levels of interleukin-6 (IL-6) were reduced and IL-10 levels were significantly increased (P<0.05). The effects of abdominal aortic BMMSC transplantation were at least partially reversed by both anti-CNTF and CNTF siRNA treatment. CONCLUSIONS In a rat model of spinal cord ischemia-reperfusion injury, abdominal aortic transplantation of BMMSCs increased the expression of CNTF, which improved hindlimb locomotor recovery by regulating the expression of IL-6 and IL-10 to reduce inflammation of the spinal cord.


Assuntos
Fator Neurotrófico Ciliar/genética , Traumatismo por Reperfusão/fisiopatologia , Isquemia do Cordão Espinal/terapia , Animais , Aorta Abdominal/fisiologia , Células Cultivadas , Fator Neurotrófico Ciliar/fisiologia , Citocinas/genética , Modelos Animais de Doenças , Feminino , Inflamação , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Medula Espinal/metabolismo , Traumatismos da Medula Espinal/metabolismo , Isquemia do Cordão Espinal/genética , Isquemia do Cordão Espinal/metabolismo
2.
Brain Behav Immun ; 70: 325-334, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29548998

RESUMO

Ciliary neurotrophic factor (CNTF) potently decreases food intake and body weight in diet-induced obese mice by acting through neuronal circuits and pathways located in the arcuate nucleus (ARC) of the hypothalamus. CNTF also exerts pro-inflammatory actions within the brain. Here we tested whether CNTF modifies energy balance by inducing inflammatory responses in the ARC and whether these effects depend upon the mechanistic target of rapamycin complex 1 (mTORC1) pathway, which regulates both energy metabolism and inflammation. To this purpose, chow- and high fat diet (HFD)- fed mice lacking the S6 kinase 1 (S6K1-/-), a downstream target of mTORC1, and their wild-type (WT) littermates received 12 days continuous intracerebroventricular (icv) infusion of the CNTF analogue axokine (CNTFAx15). Behavioral, metabolic and molecular effects were evaluated. Central chronic administration of CNTFAx15 decreased body weight and feed efficiency in WT mice only, when fed HFD, but not chow. These metabolic effects correlated with increased number of iba-1 positive microglia specifically in the ARC and were accompanied by significant increases of IL-1ß and TNF-α mRNA expression in the hypothalamus. Hypothalamic iNOS and SOCS3 mRNA, molecular markers of pro-inflammatory response, were also increased by CNTFAx15. All these changes were absent in S6K1-/- mice. This study reveals that CNTFAx15 requires a functional S6K1 to modulate energy balance and hypothalamic inflammation in a diet-dependent fashion. Further investigations should determine whether S6K1 is a suitable target for the treatment of pathologies characterized by a high neuroinflammatory state.


Assuntos
Fator Neurotrófico Ciliar/metabolismo , Fator Neurotrófico Ciliar/fisiologia , Proteínas Quinases S6 Ribossômicas 70-kDa/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal , Dieta Hiperlipídica , Ingestão de Alimentos , Metabolismo Energético , Homeostase , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Leptina , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/fisiologia , Neuroglia/fisiologia , Neuroimunomodulação/fisiologia , Obesidade/fisiopatologia , Proteínas Quinases S6 Ribossômicas 70-kDa/genética
3.
J Neurosci ; 33(7): 3240-50, 2013 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-23407977

RESUMO

Ciliary neurotrophic factor (CNTF) has been shown to be expressed after brain lesions and in particular after demyelination. Here, we addressed the role of this cytokine in the regulation of neural progenitor migration in the adult rodent brain. Using an acute model of demyelination, we show that CNTF is strongly re-expressed after lesion and is involved in the postlesional mobilization of endogenous progenitors that participate in the myelin regenerative process. We show that CNTF controls the migration of subventricular zone (SVZ)-derived neural progenitors toward the demyelinated corpus callosum. Furthermore, an ectopic source of CNTF in adult healthy brains changes SVZ-derived neural progenitors' migratory behavior that migrate toward the source by activation of the Janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) pathway. Using various in vitro assays (Boyden chambers, explants, and video time-lapse imaging), we demonstrate that CNTF controls the directed migration of SVZ-derived progenitors and oligodendrocyte precursors. Altogether, these results demonstrate that in addition to its neuroprotective activity and its role in progenitor survival and maturation, CNTF acts as a chemoattractant and participates in the recruitment of endogenous progenitors during myelin repair.


Assuntos
Encéfalo/fisiologia , Movimento Celular/fisiologia , Fator Neurotrófico Ciliar/fisiologia , Bainha de Mielina/fisiologia , Células-Tronco Neurais/fisiologia , Animais , Antimetabólitos , Encéfalo/citologia , Bromodesoxiuridina , Proliferação de Células , Sobrevivência Celular/fisiologia , Células Cultivadas , Fatores Quimiotáticos/farmacologia , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/fisiologia , Transplante de Células-Tronco , Células-Tronco/fisiologia , Transfecção
4.
Front Neuroendocrinol ; 34(3): 167-78, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23684668

RESUMO

Adult-born new neurons are continuously added to the hippocampus and the olfactory bulb to serve aspects of learning and perceptual functions. Recent evidence establishes a third neurogenic niche in the ventral hypothalamic parenchyma surrounding the third ventricle that ensures the plasticity of specific brain circuits to stabilize physiological functions such as the energy-balance regulatory system. Hypothalamic lesion studies have demonstrated that regions associated with reproduction-related functions are also capable of recruiting newborn neurons to restore physiological functions and courtship behavior. Induced by lesion or other stimulation, elevated neurotrophic factors trigger neurogenic cascades that contribute to remodeling of certain neural circuits to meet specific transient functions. This insight raises the possibility that event-specific changes, such as increased GnRH, may be mediated by courtship-sensitive neurotrophic factors. We will discuss the potentially integral and ubiquitous roles of neurogenesis in physiological and biological phenomena, roles that await future experimental exploration.


Assuntos
Hipotálamo/fisiologia , Neurogênese , Animais , Aves , Diferenciação Celular , Movimento Celular , Proliferação de Células , Ventrículos Cerebrais/fisiologia , Fator Neurotrófico Ciliar/fisiologia , Corte , Hipocampo/fisiologia , Leptina/farmacologia , Masculino , Fatores de Crescimento Neural/fisiologia , Neurogênese/efeitos dos fármacos , Ratos , Comportamento Sexual Animal/efeitos dos fármacos
5.
J Anat ; 224(1): 3-14, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24007389

RESUMO

In motoneuron disease and other neurodegenerative disorders, the loss of synapses and axon branches occurs early but is compensated by sprouting of neighboring axon terminals. Defective local axonal signaling for maintenance and dynamics of the axonal microtubule and actin cytoskeleton plays a central role in this context. The molecular mechanisms that lead to defective cytoskeleton architecture in two mouse models of motoneuron disease are summarized and discussed in this manuscript. In the progressive motor neuropathy (pmn) mouse model of motoneuron disease that is caused by a mutation in the tubulin-specific chaperone E gene, death of motoneuron cell bodies appears as a consequence of axonal degeneration. Treatment with bcl-2 overexpression or with glial-derived neurotrophic factor prevents loss of motoneuron cell bodies but does not influence the course of disease. In contrast, treatment with ciliary neurotrophic factor (CNTF) significantly delays disease onset and prolongs survival of pmn mice. This difference is due to the activation of Stat-3 via the CNTF receptor complex in axons of pmn mutant motoneurons. Most of the activated Stat-3 protein is not transported to the nucleus to activate transcription, but interacts locally in axons with stathmin, a protein that destabilizes microtubules. This interaction plays a major role in CNTF signaling for microtubule dynamics in axons. In Smn-deficient mice, a model of spinal muscular atrophy, defects in axonal translocation of ß-actin mRNA and possibly other mRNA species have been observed. Moreover, the regulation of local protein synthesis in response to signals from neurotrophic factors and extracellular matrix proteins is altered in motoneurons from this model of motoneuron disease. These findings indicate that local signals are important for maintenance and plasticity of axonal branches and neuromuscular endplates, and that disturbances in these signaling mechanisms could contribute to the pathophysiology of motoneuron diseases.


Assuntos
Axônios/fisiologia , Doença dos Neurônios Motores/fisiopatologia , Neurônios Motores/fisiologia , Plasticidade Neuronal/fisiologia , Animais , Fator Neurotrófico Ciliar/fisiologia , Modelos Animais de Doenças , Camundongos , Placa Motora/fisiologia , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais/fisiologia
6.
Handb Exp Pharmacol ; 220: 283-307, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24668477

RESUMO

Complex interactions between the brain and peripheral tissues mediate the effective control of energy balance and body weight. Hypothalamic and hindbrain neural circuits integrate peripheral signals informing the nutritional status of the animal and in response regulate nutrient intake and energy utilization. Obesity and its many medical complications emerge from the dysregulation of energy homeostasis. Excessive weight gain might also arise from alterations in reward systems of the brain that drive consumption of calorie dense, palatable foods in the absence of an energy requirement. Several neurotrophins, most notably brain-derived neurotrophic factor, have been implicated in the molecular and cellular processes underlying body weight regulation. Here, we review investigations interrogating their roles in energy balance and reward centers of the brain impacting feeding behavior and energy expenditure.


Assuntos
Peso Corporal , Encéfalo/fisiologia , Metabolismo Energético , Fatores de Crescimento Neural/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/fisiologia , Fator Neurotrófico Ciliar/fisiologia , Comportamento Alimentar , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Humanos
7.
Handb Exp Pharmacol ; 220: 411-41, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24668481

RESUMO

Amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA) represent the two major forms of motoneuron disease. In both forms of disease, spinal and bulbar motoneurons become dysfunctional and degenerate. In ALS, cortical motoneurons are also affected, which contributes to the clinical phenotype. The gene defects for most familial forms of ALS and SMA have been discovered and they point to a broad spectrum of disease mechanisms, including defects in RNA processing, pathological protein aggregation, altered apoptotic signaling, and disturbed energy metabolism. Despite the fact that lack of neurotrophic factors or their corresponding receptors are not found as genetic cause of motoneuron disease, signaling pathways initiated by neurotrophic factors for motoneuron survival, axon growth, presynaptic development, and synaptic function are disturbed in ALS and SMA. Better understanding of how neurotrophic factors and downstream signaling pathways interfere with these disease mechanisms could help to develop new therapies for motoneuron disease and other neurodegenerative disorders.


Assuntos
Esclerose Lateral Amiotrófica/fisiopatologia , Atrofia Muscular Espinal/fisiopatologia , Esclerose Lateral Amiotrófica/genética , Animais , Fator Neurotrófico Ciliar/fisiologia , Proteínas de Ligação a DNA/fisiologia , Humanos , Neurônios Motores/fisiologia , Atrofia Muscular Espinal/genética , Fatores de Crescimento Neural/fisiologia , RNA/metabolismo , Transdução de Sinais/fisiologia , Superóxido Dismutase/genética , Superóxido Dismutase-1
8.
J Neurosci ; 31(37): 13028-38, 2011 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-21917786

RESUMO

Astrocytes undergo major phenotypic changes in response to injury and disease that directly influence repair in the CNS, but the mechanisms involved are poorly understood. Previously, we have shown that neurosphere-derived rat astrocytes plated on poly-L-lysine (PLL-astrocytes) support myelination in dissociated rat spinal cord cultures (myelinating cultures). It is hypothesized that astrocyte reactivity can affect myelination, so we have exploited this culture system to ascertain how two distinct astrocyte phenotypes influence myelination. Astrocytes plated on tenascin C (TnC-astrocytes), a method to induce quiescence, resulted in less myelinated fibers in the myelinating cultures when compared with PLL-astrocytes. In contrast, treatment of myelinating cultures plated on PLL-astrocytes with ciliary neurotrophic factor (CNTF), a cytokine known to induce an activated astrocyte phenotype, promoted myelination. CNTF could also reverse the effect of quiescent astrocytes on myelination. A combination of microarray gene expression analysis and quantitative real-time PCR identified CXCL10 as a potential candidate for the reduction in myelination in cultures on TnC-astrocytes. The effect of TnC-astrocytes on myelination was eliminated by neutralizing CXCL10 antibodies. Conversely, CXCL10 protein inhibited myelination on PLL-astrocytes. Furthermore, CXCL10 treatment of purified oligodendrocyte precursor cells did not affect proliferation, differentiation, or process extension compared with untreated controls, suggesting a role in glial/axonal ensheathment. These data demonstrate a direct correlation of astrocyte phenotypes with their ability to support myelination. This observation has important implications with respect to the development of therapeutic strategies to promote CNS remyelination in demyelinating diseases.


Assuntos
Astrócitos/metabolismo , Quimiocina CXCL10/fisiologia , Fibras Nervosas Mielinizadas/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/fisiologia , Células Cultivadas , Fator Neurotrófico Ciliar/fisiologia , Meios de Cultura , Feminino , Masculino , Fibras Nervosas Mielinizadas/fisiologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Fenótipo , Polilisina/fisiologia , Análise Serial de Proteínas/métodos , Ratos , Ratos Sprague-Dawley
9.
Nat Cell Biol ; 2(12): 906-14, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11146655

RESUMO

Cytokines that are related to ciliary neurotrophic factor (CNTF) are physiologically important survival factors for motoneurons, but the mechanisms by which they prevent neuronal cell death remain unknown. Reg-2/PAP I (pancreatitis-associated protein I), referred to here as Reg-2, is a secreted protein whose expression in motoneurons during development is dependent on cytokines. Here we show that CNTF-related cytokines induce Reg-2 expression in cultured motoneurons. Purified Reg-2 can itself act as an autocrine/paracrine neurotrophic factor for a subpopulation of motoneurons, by stimulating a survival pathway involving phosphatidylinositol-3-kinase, Akt kinase and NF-kappaB. Blocking Reg-2 expression in motoneurons using Reg-2 antisense adenovirus specifically abrogates the survival effect of CNTF on cultured motoneurons, indicating that Reg-2 expression is a necessary step in the CNTF survival pathway. Reg-2 shows a unique pattern of expression in late embryonic spinal cord: it is progressively upregulated in individual motoneurons on a cell-by-cell basis, indicating that only a fraction of motoneurons in a given motor pool may be exposed to cytokines. Thus, Reg-2 is a neurotrophic factor for motoneurons, and is itself an obligatory intermediate in the survival signalling pathway of CNTF-related cytokines.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Fator Neurotrófico Ciliar/fisiologia , Neurônios Motores/fisiologia , Fatores de Crescimento Neural/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Animais , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Células Cultivadas , Citocinas/fisiologia , Desenvolvimento Embrionário e Fetal/genética , Desenvolvimento Embrionário e Fetal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Litostatina , Neurônios Motores/citologia , Neurônios Motores/efeitos dos fármacos , Fatores de Crescimento Neural/antagonistas & inibidores , Fatores de Crescimento Neural/genética , Fatores de Crescimento Neural/farmacologia , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/farmacologia , Proteínas Associadas a Pancreatite , Ratos , Transdução de Sinais
10.
Nat Med ; 8(6): 620-4, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12042814

RESUMO

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS). So far, immunological mechanisms responsible for demyelination have been the focus of interest. However, mechanisms regulating axon maintenance as well as glial precursor-cell proliferation and oligodendrocyte survival might also influence disease outcome. The cytokine ciliary neurotrophic factor (CNTF), which was originally identified as a survival factor for isolated neurons, promotes differentiation, maturation and survival of oligodendrocytes. To investigate the role of endogenous CNTF in inflammatory demyelinating disease, we studied myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) in CNTF-deficient and wild-type C57BL/6 mice. Disease was more severe in CNTF-deficient mice and recovery was poor, with a 60% decrease in the number of proliferating oligodendrocyte precursor cells (OPCs) and a more than 50% increase in the rate of oligodendrocyte apoptosis. In addition, vacuolar dystrophy of myelin and axonal damage were more severe in CNTF-deficient mice. These specific pathological features could be prevented by treatment with an antiserum against tumor necrosis factor-alpha, suggesting that endogenous CNTF may counterbalance this effect of TNF-alpha (ref. 7). Here we identify a factor that modulates, in an inflammatory environment, glial cell survival and is an outcome determinant of EAE.


Assuntos
Fator Neurotrófico Ciliar/fisiologia , Doenças Desmielinizantes/prevenção & controle , Encefalomielite Autoimune Experimental/patologia , Esclerose Múltipla/prevenção & controle , Fatores de Crescimento Neural/fisiologia , Animais , Fator Neurotrófico Ciliar/deficiência , Fator Neurotrófico Ciliar/genética , Citocinas/fisiologia , Inflamação/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
Stem Cells ; 27(2): 431-41, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19023034

RESUMO

In the neurogenic areas of the adult rodent brain, neural stem cells (NSCs) proliferate and produce new neurons throughout the lifetime. This requires a permanent pool of NSCs, the size of which needs to be tightly controlled. The gp130-associated cytokines ciliary neurotrophic factor (CNTF) and leukemia inhibitory factor (LIF) have been implicated in regulating NSC self-renewal and differentiation during embryonic development and in the adult brain. To study the relevance of the two cytokines in vivo, we analyzed precursor cell proliferation and neurogenesis in the dentate gyrus of CNTF- and LIF-deficient mouse mutants. The number of radial glia-like NSCs, proliferative activity, and generation of new neurons were all reduced in CNTF(-/-) mutants but unaltered in LIF(-/-) animals. Conditional ablation of the signal transducer and activator of transcription 3 (STAT3) gene under the control of the human glial fibrillary acidic protein promoter resulted in a reduction of neurogenesis similar to that in CNTF(-/-) mice. The size of the granule cell layer was decreased in both mutants. Treatment of neurosphere cultures prepared from adult forebrain with CNTF inhibited overall proliferative activity but increased the number of NSCs as indicated by enhanced secondary neurosphere formation and upregulated expression of stem cell markers. Knockdown of STAT3 with short interfering RNA inhibited CNTF effects on neurospheres, and knockdown of suppressor of cytokine signaling 3 (SOCS3) enhanced them. Our results provide evidence that CNTF-induced STAT3 signaling is essential for the formation and/or maintenance of the neurogenic subgranular zone in the adult dentate gyrus and suggest that CNTF is required to keep the balance between NSC self-renewal and the generation of neuronal progenitors.


Assuntos
Fator Neurotrófico Ciliar/farmacologia , Fator Neurotrófico Ciliar/fisiologia , Giro Denteado/citologia , Giro Denteado/metabolismo , Neurogênese/fisiologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Fator Neurotrófico Ciliar/genética , Imuno-Histoquímica , Fator Inibidor de Leucemia/genética , Fator Inibidor de Leucemia/fisiologia , Camundongos , Camundongos Mutantes , Neurogênese/genética , Neurônios/citologia , Fator de Transcrição STAT3/genética , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
12.
J Neurosci ; 28(9): 2231-41, 2008 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-18305256

RESUMO

Neurogenesis continues in the adult forebrain subventricular zone (SVZ) and the dentate gyrus of the hippocampal formation. Degeneration of dopaminergic projections in Parkinson's disease and animals reduces, whereas ciliary neurotrophic factor (CNTF) promotes, neurogenesis. We tested whether the dopaminergic system promotes neurogenesis through CNTF. Astrocytes of the SVZ and dentate gyrus expressed CNTF and were close to dopaminergic terminals. Dopaminergic denervation in adult mice reduced CNTF mRNA by approximately 60%, whereas systemic treatment with the D2 agonist quinpirole increased CNTF mRNA in the SVZ and hippocampal formation, and in cultured astrocytes by 1.5-5 fold. The effect of quinpirole in vitro was blocked by the D2 antagonist eticlopride and did not cause astroglial proliferation or hypertrophy. Systemic quinpirole injections increased proliferation in wild-type mice by approximately 25-75% but not in CNTF-/- littermates or in the SVZ of mice infused with CNTF antibodies. Quinpirole increased the number of neuroblasts in wild-type but not in CNTF-/- littermates. Neurogenesis was reduced by approximately 20% in CNTF-/- mice, confirming the endogenous role of CNTF. Nigrostriatal denervation did not affect SVZ proliferation in CNTF-/- mice, suggesting that the dopaminergic innervation normally regulates neurogenesis through CNTF. Quinpirole acted on postsynaptic receptors as it reversed the reduced proliferation seen after dopaminergic denervation in wild-type mice. Thus, CNTF mediates dopaminergic innervation- and D2 receptor-induced neurogenesis in the adult forebrain. Because CNTF is predominantly expressed in the nervous system, this mechanism and the ability to pharmacologically modulate it have implications for Parkinson's disease and cell-replacement therapies for other disorders.


Assuntos
Proliferação de Células , Sistema Nervoso Central/citologia , Fator Neurotrófico Ciliar/fisiologia , Neurônios/fisiologia , Receptores de Dopamina D2/fisiologia , Animais , Anticorpos/farmacologia , Bromodesoxiuridina/metabolismo , Sistema Nervoso Central/efeitos dos fármacos , Fator Neurotrófico Ciliar/deficiência , Fator Neurotrófico Ciliar/imunologia , Técnicas de Cocultura/métodos , Agonistas de Dopamina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Ventrículos Laterais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuroglia/fisiologia , Oxidopamina/farmacologia , Quimpirol/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
13.
J Cell Biol ; 166(7): 963-8, 2004 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-15452140

RESUMO

Neural stem cell (NSC) differentiation is precisely controlled by a network of transcription factors, which themselves are regulated by extracellular signals (Bertrand, N., D.S. Castro, and F. Guillemot, 2002. Nat. Rev. Neurosci 3:517-530; Shirasaki, R. and S.L. Pfaff, 2002. Annu. Rev. Neurosci 25:251-281). One way that the activity of such transcription factors is controlled is by the regulation of their movement between the cytosol and nucleus (Vandromme, M., C. Gauthier-Rouviere, N. Lamb, and A. Fernandez, 1996. Trends Biochem.Sci. 21:59-64; Lei, E.P. and P.A. Silver, 2002. Dev. Cell 2:261-272). Here we show that the basic helix-loop-helix transcription factor OLIG2, which has been shown to be required for motor neuron and oligodendrocyte development, is found in the cytoplasm, but not the nucleus, of astrocytes in culture and of a subset of astrocytes in the subventricular zone. We demonstrate that the accumulation of OLIG2 in the nucleus of NSCs blocks the CNTF-induced astrocyte differentiation and that the translocation of OLIG2 to the cytoplasm is promoted by activated AKT. We propose that the AKT-stimulated export of OLIG2 from the nucleus of NSCs is essential for the astrocyte differentiation.


Assuntos
Astrócitos/metabolismo , Diferenciação Celular/genética , Sistema Nervoso Central/metabolismo , Fator Neurotrófico Ciliar/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/genética , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Diferenciação Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Sistema Nervoso Central/citologia , Sistema Nervoso Central/embriologia , Fator Neurotrófico Ciliar/metabolismo , Fator Neurotrófico Ciliar/farmacologia , Citoplasma/efeitos dos fármacos , Citoplasma/genética , Citoplasma/metabolismo , Carioferinas/genética , Carioferinas/metabolismo , Camundongos , Proteínas do Tecido Nervoso/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Neurônios/metabolismo , Fator de Transcrição 2 de Oligodendrócitos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Células-Tronco/citologia , Proteína Exportina 1
14.
Neurochem Res ; 34(1): 109-17, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18401707

RESUMO

Microglia, CNS-resident macrophages, serve as scavengers to remove cellular debris and facilitate tissue remodeling in the developing and injured CNS. Little is known as what and how microenvironmental factors mediate the phagocytotic ability of microglia. Our previous study has indicated that treatment with glial cell line-derived neurotrophic factor (GDNF) increased the phagocytotic activity of primary rat microglia possibly through the upregulation of alpha5 integrin. In the present study, ciliary neurotrophic factor (CNTF), which has been reported to be produced by glia, was shown to have stimulatory effect on the phagocytosis of primary rat microglia and mouse microglial cell line BV2. Ca2+ imaging analysis and the application of intracellular calcium chelator BAPTA-AM revealed that CNTF-induced increase in microglial phagocytosis was mediated by a calcium signaling pathway. Furthermore, treatment with CNTF led to an increase in the expression of alphav integrin, which has been reported to be involved in the phagocytosis of the apoptotic cells. In summary, we have provided evidence that CNTF can increase microglial phagocytosis through a calcium-mediated pathway. Our results also suggest that the upregulation of alphav integrin by CNTF could be involved in the increased phagocytotic activity of microglia.


Assuntos
Fator Neurotrófico Ciliar/fisiologia , Microglia/fisiologia , Fagocitose/fisiologia , Animais , Cálcio/metabolismo , Linhagem Celular , Células Cultivadas , Integrina alfaV/metabolismo , Interleucina-1beta/metabolismo , Camundongos , Fagocitose/efeitos dos fármacos , Ratos , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
15.
Psychoneuroendocrinology ; 100: 96-105, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30299260

RESUMO

Ciliary neurotrophic factor (CNTF) is produced by astrocytes and promotes neurogenesis and neuroprotection. Little is known about the role of CNTF in affective behavior. We investigated whether CNTF affects depressive- and anxiety-like behavior in adult mice as tested in the forced swim, sucrose preference and elevated-T maze tests. Female wild type CNTF+/+ mice more readily developed behavioral despair with increased immobility time and decreased latency to immobility in the forced swim test than male CNTF+/+ littermates. The lack of CNTF in CNTF-/- mice had an opposite effect on depressive-like behavior in female mice (reduced immobility time and increased sucrose preference) vs. male mice (increased immobility time). Female wildtype mice expressed more CNTF in the amygdala than male mice. Ovariectomy increased CNTF expression, as well as immobility time, which was significantly reduced in CNTF-/- mice, suggesting that CNTF mediates overiectomy-induced immobility time, possibly in the amygdala. Progesterone but not 17-ß estradiol inhibited CNTF expression in cultured C6 astroglioma cells. Progesterone treatment also reduced CNTF expression in the amygdala and decreased immobility time in female CNTF+/+ but not in CNTF-/- mice. Castration did not alter CNTF expression in males nor their behavior. Lastly, there were no effects of CNTF on the elevated T-maze, a behavioral test of anxiety, suggesting that a different mechanism may underlie anxiety-like behavior. This study reveals a novel CNTF-mediated mechanism in stress-induced depressive-like behavior and points to opportunities for sex-specific treatments for depression, e.g. progesterone in females and CNTF-stimulating drugs in males.


Assuntos
Fator Neurotrófico Ciliar/fisiologia , Depressão/genética , Animais , Astrócitos/metabolismo , Astrócitos/fisiologia , Comportamento Animal/fisiologia , Fator Neurotrófico Ciliar/genética , Depressão/patologia , Depressão/fisiopatologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurogênese/genética , Caracteres Sexuais , Células Tumorais Cultivadas
16.
J Neurosci ; 27(27): 7094-104, 2007 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-17611262

RESUMO

High energy demands of neurons make them vulnerable to adverse effects of energy impairment. Recently, astrocytes were shown to regulate the flux of energy substrates to neurons. In pathological situations, astrocytes are activated but the consequences on brain energy metabolism are still poorly characterized. We found that local lentiviral-mediated gene transfer of ciliary neurotrophic factor (CNTF), a cytokine known to activate astrocytes, induced a stable decrease in the glycolytic flux in the rat striatum in vivo as measured by 2-[18F]-2-deoxy-D-glucose autoradiography and micro-positron emission tomography imaging. The activity of the mitochondrial complex IV enzyme cytochrome oxidase was not modified, suggesting maintenance of downstream oxidative steps of energy production. CNTF significantly increased the phosphorylation level of the intracellular energy sensor AMP-activated protein kinase (AMPK), supporting a specific reorganization of brain energy pathways. Indeed, we found that different key enzymes/transporters of fatty acids beta-oxidation and ketolysis were overexpressed by CNTF-activated astrocytes within the striatum. In primary striatal neuron/astrocyte mixed cultures exposed to CNTF, the AMPK pathway was also activated, and the rate of oxidation of fatty acids and ketone bodies was significantly enhanced. This metabolic plasticity conferred partial glial and neuronal protection against prolonged palmitate exposure and glycolysis inhibition. We conclude that CNTF-activated astrocytes may have a strong protective potential to face severe metabolic insults.


Assuntos
Astrócitos/metabolismo , Fator Neurotrófico Ciliar/fisiologia , Metabolismo Energético/fisiologia , Plasticidade Neuronal/fisiologia , Animais , Astrócitos/citologia , Astrócitos/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Humanos , Masculino , Ratos , Ratos Endogâmicos Lew
17.
J Neurosci ; 27(36): 9664-9, 2007 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-17804627

RESUMO

The cytokine ciliary neurotrophic factor (CNTF) promotes the growth of neural processes from many kinds of neurons in the developing and regenerating adult nervous system, but the intracellular signaling mechanisms mediating this important function of CNTF are poorly understood. Here, we show that CNTF activates the nuclear factor-kappaB (NF-kappaB) transcriptional system in neonatal sensory neurons and that blocking NF-kappaB-dependent transcription inhibits CNTF-promoted neurite growth. Selectively blocking NF-kappaB activation by the noncanonical pathway that requires tyrosine phosphorylation of inhibitor kappaB-alpha (IkappaB-alpha), but not by the canonical pathway that requires serine phosphorylation of IkappaB-alpha, also effectively inhibits CNTF-promoted neurite growth. CNTF treatment activates spleen tyrosine kinase (SYK) whose substrates include IkappaB-alpha. CNTF-induced SYK phosphorylation is rapidly followed by increased tyrosine phosphorylation of IkappaB-alpha, and blocking SYK activation or tyrosine phosphorylation of IkappaB-alpha prevents CNTF-induced NF-kappaB activation and CNTF-promoted neurite growth. These findings demonstrate that NF-kappaB signaling by an unusual activation mechanism is essential for the ability of CNTF to promote the growth of neural processes in the developing nervous system.


Assuntos
Fator Neurotrófico Ciliar/fisiologia , Proteínas I-kappa B/metabolismo , NF-kappa B/metabolismo , Neuritos/fisiologia , Neurônios Aferentes/fisiologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Fator Neurotrófico Ciliar/farmacologia , Ativação Enzimática/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Inibidor de NF-kappaB alfa , Neuritos/efeitos dos fármacos , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/ultraestrutura , Gânglio Nodoso/citologia , Oligonucleotídeos/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Quinase Syk , Transcrição Gênica/efeitos dos fármacos , Tirosina/metabolismo
18.
FEBS Lett ; 582(27): 3832-8, 2008 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-18950628

RESUMO

Administration of CNTF durably reduces food intake and body weight in obese humans and rodent models. However, the involvement of endogenous CNTF in the central regulation of energy homeostasis needs to be elucidated. Here, we demonstrate that CNTF and its receptor are expressed in the arcuate nucleus, a key hypothalamic region controlling food intake, and that CNTF levels are inversely correlated to body weight in rats fed a high-sucrose diet. Thus endogenous CNTF may act, in some individuals, as a protective factor against weight gain during hypercaloric diet and could account for individual differences in the susceptibility to obesity.


Assuntos
Regulação do Apetite/genética , Núcleo Arqueado do Hipotálamo/metabolismo , Fator Neurotrófico Ciliar/fisiologia , Metabolismo Energético/genética , Aumento de Peso/genética , Animais , Regulação do Apetite/efeitos dos fármacos , Fator Neurotrófico Ciliar/administração & dosagem , Fator Neurotrófico Ciliar/genética , Subunidade alfa do Receptor do Fator Neutrófico Ciliar/genética , Subunidade alfa do Receptor do Fator Neutrófico Ciliar/fisiologia , Metabolismo Energético/efeitos dos fármacos , Feminino , Expressão Gênica , Predisposição Genética para Doença , Homeostase , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Obesidade/genética , Obesidade/prevenção & controle , Fosforilação , Ratos , Ratos Wistar , Proteínas Recombinantes/biossíntese , Aumento de Peso/efeitos dos fármacos
19.
J Neurosci Res ; 86(8): 1694-710, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18293415

RESUMO

To understand the characteristics of tsAM5D cells immortalized with the temperature-sensitive simian virus 40 large T-antigen, we first examined the responsiveness of the cells to ligands of the glial cell line-derived neurotrophic factor (GDNF) family. tsAM5D cells proliferated at the permissive temperature of 33 degrees C in response to either GDNF or neurturin, but not persephin or artemin. At the nonpermissive temperature of 39 degrees C, GDNF or neurturin caused tsAM5D cells to differentiate into neuron-like cells; however, the differentiated cells died in a time-dependent manner. Interestingly, ciliary neurotrophic factor (CNTF) did not affect the GDNF-mediated cell proliferation at 33 degrees C but promoted the survival and differentiation of GDNF-treated cells at 39 degrees C. In the presence of GDNF plus CNTF, the morphological change induced by the temperature shift was associated with up-regulated expression of various neuronal marker genes, indicating that the cells had undergone neuronal differentiation. In addition, tsAM5D cells caused to differentiate by GDNF plus CNTF at 39 degrees C became dependent solely on nerve growth factor (NGF) for their survival and neurite outgrowth. Moreover, upon treatment with GDNF plus CNTF, the dopaminergic phenotype was suppressed by the temperature shift. Thus, we demonstrated that tsAM5D cells had the capacity to differentiate terminally into neuron-like cells in response to GDNF plus CNTF when the oncogene was inactivated by the temperature shift. This cell line provides a useful model system for studying the role of a variety of signaling molecules for GDNF/CNTF-induced neuronal differentiation.


Assuntos
Antígenos Transformantes de Poliomavirus/fisiologia , Diferenciação Celular/fisiologia , Células Cromafins/citologia , Fator Neurotrófico Ciliar/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Neurônios/citologia , Glândulas Suprarrenais/citologia , Glândulas Suprarrenais/fisiologia , Animais , Morte Celular/fisiologia , Linhagem Celular Transformada , Células Cultivadas , Células Cromafins/fisiologia , Humanos , Fator de Crescimento Neural/fisiologia , Neurônios/fisiologia , Ratos , Temperatura , Fatores de Tempo
20.
J Neurosci Res ; 86(8): 1748-57, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18293418

RESUMO

Huntington's disease (HD) is a neurodegenerative disorder caused by an elongation of CAG repeats in the HD gene, which encodes a mutant copy of huntingtin with an expanded polyglutatmine repeat. Individuals who are affected by the disease suffer from motor, cognitive, and emotional impairments. Levels of certain striatal-enriched mRNAs decrease in both HD patients and transgenic HD mice prior to the development of motor symptoms and neuronal cell death. Ciliary neurotrophic factor (CNTF) has been shown to protect neurons against chemically induced toxic insults in vitro and in vivo. To test the hypothesis that CNTF might protect neurons from the negative effects of the mutant huntingtin protein in vivo, CNTF was continuously expressed following transduction of the striatum by recombinant adeno-associated viral vectors (rAAV2). Wild-type and R6/1 HD transgenic (R6/1) mice that received bilateral or unilateral intrastriatal injections of rAAV2-CNTF experienced weight loss. The CNTF-treated R6/1 HD transgenic mice experienced motor impairments at an earlier age than expected compared with age-matched control R6/1 HD transgenic animals. CNTF also caused abnormal behavior in WT mice. In addition to behavioral impairments, in situ hybridization showed that, in both WT and R6/1 mice, CNTF expression caused a significant decrease in the levels of striatal-enriched transcripts. Overall, continuous expression of striatal CNTF at the dose mediated by the expression cassette used in this study was detrimental to HD and wild-type mice.


Assuntos
Fator Neurotrófico Ciliar/biossíntese , Corpo Estriado/metabolismo , Regulação para Baixo/fisiologia , Transtornos das Habilidades Motoras/metabolismo , Animais , Comportamento Animal/fisiologia , Fator Neurotrófico Ciliar/genética , Fator Neurotrófico Ciliar/fisiologia , Regulação para Baixo/genética , Humanos , Doença de Huntington/complicações , Doença de Huntington/genética , Doença de Huntington/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Transtornos das Habilidades Motoras/etiologia , Transtornos das Habilidades Motoras/genética , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA