Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biol Reprod ; 103(6): 1300-1313, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32886743

RESUMO

Fibroblast growth factor 9 (FGF9) is an autocrine/paracrine growth factor that plays critical roles in embryonic and organ developments and is involved in diverse physiological events. Loss of function of FGF9 exhibits male-to-female sex reversal in the transgenic mouse model and gain of FGF9 copy number was found in human 46, XX sex reversal patient with disorders of sex development. These results suggested that FGF9 plays a vital role in male sex development. Nevertheless, how FGF9/Fgf9 expression is regulated during testis determination remains unclear. In this study, we demonstrated that human and mouse SRY bind to -833 to -821 of human FGF9 and -1010 to -998 of mouse Fgf9, respectively, and control FGF9/Fgf9 mRNA expression. Interestingly, we showed that mouse SRY cooperates with SF1 to regulate Fgf9 expression, whereas human SRY-mediated FGF9 expression is SF1 independent. Furthermore, using an ex vivo gonadal culture system, we showed that FGF9 expression is sufficient to switch cell fate from female to male sex development in 12-16 tail somite XX mouse gonads. Taken together, our findings provide evidence to support the SRY-dependent, fate-determining role of FGF9 in male sex development.


Assuntos
Transtornos do Desenvolvimento Sexual/genética , Fator 9 de Crescimento de Fibroblastos/metabolismo , Gônadas/fisiologia , Processos de Determinação Sexual/fisiologia , Proteína da Região Y Determinante do Sexo/metabolismo , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Feminino , Fator 9 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Masculino , Camundongos , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína da Região Y Determinante do Sexo/genética , Técnicas de Cultura de Tecidos , Regulação para Cima
2.
Mol Neurobiol ; 57(10): 4090-4105, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32666227

RESUMO

Mild hypothermia has promising effects in the treatment of acute brain insults and also affects cell cycle progression. Mitochondrial dynamics, fusion and fission, are changed along with the cell cycle and disrupted in neurodegenerative diseases, including Parkinson's disease (PD). However, the effects of hypothermia on aberrant mitochondrial dynamics in PD remain unknown. We hypothesized that mild hypothermia protects neurons by regulating cell cycle-dependent protein expression and mitochondrial dynamics in a 1-methyl-4-phenylpyridinium (MPP+)-induced cell model of PD. We found that the hypothermia treatment at 32 °C prevented MPP+-induced neuron death; however, 32 °C treatment itself also reduced cell viability. This reduction was associated with cell cycle arrest and downregulation of cyclin-dependent kinase 4 (CDK4) in proliferating human SK-N-SH neuroblastoma cells but upregulation in well-differentiated primary rat cortical neurons. In both types of neurons, hypothermia upregulated p27 (an endogenous inhibitor of CDKs) and p35 (CDK5 activator) protein expression. Treatment with hypothermia, or a selective CDK4 inhibitor, or roscovitine (CDK5 inhibitor) prevented MPP+-induced mitochondrial fission, upregulation of mitochondrial fission protein dynamin-related protein 1 (Drp1), and neuron death. In addition, overexpression of dominant negative mutant Drp1K38A improved MPP+-induced mitochondrial fission while overexpression of wild-type Drp1 blunted the prevention of mitochondrial fission by hypothermia as well as CDK4 inhibitor and roscovitine. These results elucidate that hypothermia may inhibit CDK4 and CDK5 activation by upregulating p27 and p35 expression to prevent Drp1-dependent mitochondrial fission and neuron loss after MPP+ treatment. CDK4 and CDK5 inhibition imitates the neuroprotective functions of hypothermia as a potential therapy for PD.


Assuntos
Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 5 Dependente de Ciclina/antagonistas & inibidores , Dinaminas/metabolismo , Hipotermia Induzida , Dinâmica Mitocondrial , Neurônios/patologia , 1-Metil-4-fenilpiridínio , Animais , Proteínas de Ciclo Celular/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Córtex Cerebral/patologia , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 5 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Dinâmica Mitocondrial/efeitos dos fármacos , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Ratos Sprague-Dawley , Roscovitina/farmacologia , Regulação para Cima/efeitos dos fármacos
3.
Theranostics ; 10(6): 2817-2831, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194837

RESUMO

Rationale: The formation of adipose-derived stem cells (ASCs) into spheres on a chitosan-coated microenvironment promoted ASCs differentiation into a mixed population of neural lineage-like cells (NLCs), but the underline mechanism is still unknown. Since the fibroblast growth factor 9 (FGF9) and fibroblast growth factor receptors (FGFRs) play as key regulators of neural cell fate during embryo development and stem cell differentiation, the current study aims to reveal the interplay of FGF9 and FGFRs for promoting peripheral nerve regeneration. Methods: Different concentration of FGF9 peptide (10, 25, 50, 100 ng/mL) were added during NLCs induction (FGF9-NLCs). The FGFR expressions and potential signaling were studied by gene and protein expressions as well as knocking down by specific FGFR siRNA or commercial inhibitors. FGF9-NLCs were fluorescent labeled and applied into a nerve conduit upon the injured sciatic nerves of experimental rats. Results: The FGFR2 and FGFR4 were significantly increased during NLCs induction. The FGF9 treated FGF9-NLCs spheres became smaller and changed into Schwann cells (SCs) which expressed S100ß and GFAP. The specific silencing of FGFR2 diminished FGF9-induced Akt phosphorylation and inhibited the differentiation of SCs. Transplanted FGF9-NLCs participated in myelin sheath formation, enhanced axonal regrowth and promoted innervated muscle regeneration. The knockdown of FGFR2 in FGF9-NLCs led to the abolishment of nerve regeneration. Conclusions: Our data therefore demonstrate the importance of FGF9 in the determination of SC fate via the FGF9-FGFR2-Akt pathway and reveal the therapeutic benefit of FGF9-NLCs.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Fator 9 de Crescimento de Fibroblastos/farmacologia , Células-Tronco Mesenquimais , Nervo Isquiático , Animais , Células Cultivadas , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Regeneração Nervosa/efeitos dos fármacos , Peptídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/lesões
4.
Free Radic Biol Med ; 130: 256-266, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30391672

RESUMO

Huntington's disease (HD) is a heritable neurodegenerative disorder, and has been characterized as an increase of oxidative stress in brain regions. In our previous results, we showed fibroblast growth factor 9 (FGF9) provides neuroprotective functions to suppress cell death in HD striatal cells dominantly through ERK signalling. However, whether the working mechanism of FGF9 is related to anti-oxidative stress in HD is still unknown. In this study, STHdhQ7/Q7 (Q7) and STHdhQ111/Q111 (Q111) striatal knock-in cell lines were used to examine the neuroprotective effects of FGF9 against oxidative stress in HD. Results show that FGF9 alleviates oxidative stress induced by starvation in Q7 and Q111 cells. The treatment of FGF9 not only induces upregulation and activation of nuclear factor erythroid 2-like 2 (Nrf2), a critical transcription factor for anti-oxidative stress, but also further upregulates its downstream targets, such as superoxide dismutase 2, gamma-glutamylcysteine synthetase and glutathione reductase. Furthermore, blockage of the Nrf2 pathway abolishes the anti-oxidative functions of FGF9, and inhibition of ERK signalling reduces the activation of the FGF9-Nrf2 pathway, resulting in higher level of oxidative stress in HD cells. These results support the neuroprotective effects of FGF9 against oxidative stress through the ERK-Nrf2 pathway, and imply one of potential strategies for therapy of HD.


Assuntos
Antioxidantes/farmacologia , Encéfalo/metabolismo , Fator 9 de Crescimento de Fibroblastos/genética , Doença de Huntington/tratamento farmacológico , Fator 2 Relacionado a NF-E2/genética , Animais , Encéfalo/patologia , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Glutamato-Cisteína Ligase/genética , Glutationa Redutase/genética , Humanos , Proteína Huntingtina/genética , Doença de Huntington/genética , Doença de Huntington/patologia , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Superóxido Dismutase/genética , Córtex Visual/metabolismo , Córtex Visual/patologia
5.
Cancer Sci ; 109(11): 3503-3518, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30191630

RESUMO

Fibroblast growth factor 9 (FGF9) promotes cancer progression; however, its role in cell proliferation related to tumorigenesis remains elusive. We investigated how FGF9 affected MA-10 mouse Leydig tumor cell proliferation and found that FGF9 significantly induced cell proliferation by activating ERK1/2 and retinoblastoma (Rb) phosphorylations within 15 minutes. Subsequently, the expressions of E2F1 and the cell cycle regulators: cyclin D1, cyclin E1 and cyclin-dependent kinase 4 (CDK4) in G1 phase and cyclin A1, CDK2 and CDK1 in S-G2 /M phases were increased at 12 hours after FGF9 treatment; and cyclin B1 in G2 /M phases were induced at 24 hours after FGF9 stimulation, whereas the phosphorylations of p53, p21 and p27 were not affected by FGF9. Moreover, FGF9-induced effects were inhibited by MEK inhibitor PD98059, indicating FGF9 activated the Rb/E2F pathway to accelerate MA-10 cell proliferation by activating ERK1/2. Immunoprecipitation assay and ChIP-quantitative PCR results showed that FGF9-induced Rb phosphorylation led to the dissociation of Rb-E2F1 complexes and thereby enhanced the transactivations of E2F1 target genes, Cyclin D1, Cyclin E1 and Cyclin A1. Silencing of FGF receptor 2 (FGFR2) using lentiviral shRNA inhibited FGF9-induced ERK1/2 phosphorylation and cell proliferation, indicating that FGFR2 is the obligate receptor for FGF9 to bind and activate the signaling pathway in MA-10 cells. Furthermore, in a severe combined immunodeficiency mouse xenograft model, FGF9 significantly promoted MA-10 tumor growth, a consequence of increased cell proliferation and decreased apoptosis. Conclusively, FGF9 interacts with FGFR2 to activate ERK1/2, Rb/E2F1 and cell cycle pathways to induce MA-10 cell proliferation in vitro and tumor growth in vivo.


Assuntos
Fator de Transcrição E2F1/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 9 de Crescimento de Fibroblastos/metabolismo , Tumor de Células de Leydig/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Proteína do Retinoblastoma/metabolismo , Neoplasias Testiculares/metabolismo , Animais , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Masculino , Camundongos , Fosforilação , Transdução de Sinais
6.
Cell Physiol Biochem ; 48(2): 605-617, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30021209

RESUMO

BACKGROUND/AIMS: Huntington's disease (HD) is a heritable neurodegenerative disorder, and there is no cure for HD to date. A type of fibroblast growth factor (FGF), FGF9, has been reported to play prosurvival roles in other neurodegenerative diseases, such as Parkinson's disease and Alzheimer's disease. However, the effects of FGF9 on HD is still unknown. With many similarities in the cellular and pathological mechanisms that eventually cause cell death in neurodegenerative diseases, we hypothesize that FGF9 might provide neuroprotective functions in HD. METHODS: In this study, STHdhQ7/Q7 (WT) and STHdhQ111/Q111 (HD) striatal knock-in cell lines were used to evaluate the neuroprotective effects of FGF9. Cell proliferation, cell death and neuroprotective markers were determined via the MTT assay, propidium iodide staining and Western blotting, respectively. The signaling pathways regulated by FGF9 were demonstrated using Western blotting. Additionally, HD transgenic mouse models were used to further confirm the neuroprotective effects of FGF9 via ELISA, Western blotting and immunostaining. RESULTS: Results show that FGF9 not only enhances cell proliferation, but also alleviates cell death as cells under starvation stress. In addition, FGF9 significantly upregulates glial cell line-derived neurotrophic factor (GDNF) and an anti-apoptotic marker, Bcl-xL, and decreases the expression level of an apoptotic marker, cleaved caspase 3. Furthermore, FGF9 functions through ERK, AKT and JNK pathways. Especially, ERK pathway plays a critical role to influence the effects of FGF9 toward cell survival and GDNF production. CONCLUSIONS: These results not only show the neuroprotective effects of FGF9, but also clarify the critical mechanisms in HD cells, further providing an insight for the therapeutic potential of FGF9 in HD.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 9 de Crescimento de Fibroblastos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Butadienos/farmacologia , Caspase 3/metabolismo , Linhagem Celular , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Doença de Huntington/metabolismo , Doença de Huntington/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Camundongos Transgênicos , Nitrilas/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Córtex Visual/citologia , Córtex Visual/efeitos dos fármacos , Córtex Visual/metabolismo , Proteína bcl-X/metabolismo
7.
Am J Transl Res ; 9(11): 4785-4806, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29218080

RESUMO

Glioblastoma multiforme (GBM) is one of the most malignant and aggressive brain tumors with great amount of hyaluronan (HA) secretion and CD44 overexpression (HA receptor). CD44 has been suggested as a cancer stem cells (CSCs) marker. However, several clinical studies have indicated that CD44low glioma cell exhibit CSCs traits. Additionally, our previous study indicated that more CD44 expression was associated with a better prognosis in GBM patients. To determine whether CD44 is an appropriate marker of glioma stem cells (GSCs), we manipulated CD44 expression using intrinsic (CD44 knockdown, CD44kd) and extrinsic (HA supplement, HA+) methods. Our results show that CD44kd suppressed cell proliferation by retarding cell cycle progression from G0/G1 to S phase. Furthermore, it caused GSCs traits, including lower expression of differentiation marker (glial fibrillary acidic protein, GFAP), a higher level of sphere formation and higher expression of stem cell markers (CD133, nestin and Oct4). The reduction of CD44 expression induced by HA+ was accompanied by an increase in GSCs properties. Interestingly, the presence of HA+ in glioma cells with GSC traits conversely facilitated differentiation. Our data indicated that the CD44 low-expressing cells exhibit more GSCs straits, suggesting that CD44 is not an appropriate marker for GSCs. Furthermore, the preferential expression of CD44 at the invasive rim in rat glioma specimen implies that CD44 may be more important for invasion and migration instead of GSCs marker in glioma.

8.
Free Radic Biol Med ; 89: 274-86, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26424114

RESUMO

Our previous studies demonstrated that fibroblast growth factor 9 (FGF9) protects cortical and dopaminergic neurons from 1-methyl-4-phenylpyridinium (MPP(+))-induced oxidative insult by upregulation of γ-glutamylcysteine synthetase (γ-GCS) and heme oxygenase-1 (HO-1). However, the mechanisms responsible for FGF9-induced γ-GCS and HO-1 upregulation remain uncharacterized. In the present study, we demonstrate the signaling pathways by which FGF9 upregulates HO-1 and γ-GCS expression. We found that FGF9-induced HO-1 and γ-GCS expression was prevented by PD173014, an inhibitor of the FGF receptor (FGFR). FGF9 treatment induced the phosphorylation of FGFR downstream signals of extracellular signal-regulated kinase 1/2 (ERK1/2) and AKT in a dose- and time-dependent manner. The inhibition of MEK/ERK1/2 or PI3K/AKT activity by U0126 or wortmannin, but not the inhibition of phospholipase Cγ by U73122, prevented FGF9-induced γ-GCS and HO-1 upregulation, changes in cellular redox status, and neuroprotection against MPP(+) toxicity in primary cortical and dopaminergic neurons. Furthermore, FGF9 treatment enhanced the promoter activity of the cAMP-response element binding protein (CREB) and nuclear factor erythroid-derived 2-like 2 (Nrf2), and this phenomenon was blocked by PD173014 or U0126 or wortmannin. Knockdown of CREB and Nrf2 by shRNA blocked FGF9-induced γ-GCS and HO-1 upregulation, but not ERK and AKT phosphorylation. An in vivo study consistently showed that FGF9 overexpression using a lentivirus delivery system induced ERK1/2 phosphorylation and HO-1 upregulation and protected dopaminergic neurons against MPP(+) toxicity in rat substantia nigra. These results indicate that FGF9-induced HO-1 and γ-GCS upregulation is mediated by binding to FGFR and activation of two parallel downstream signaling pathways, ERK and AKT, which reconverge to induce CREB and Nrf2 transcriptional activity.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 9 de Crescimento de Fibroblastos/metabolismo , Heme Oxigenase-1/metabolismo , Fator 2 Relacionado a NF-E2/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Western Blotting , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/genética , Fator 9 de Crescimento de Fibroblastos/genética , Regulação da Expressão Gênica , Heme Oxigenase-1/genética , Técnicas Imunoenzimáticas , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , RNA Mensageiro/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Fatores de Crescimento de Fibroblastos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Regulação para Cima
9.
Exp Neurol ; 263: 50-62, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25286336

RESUMO

Exercise induces oxidative stress, which may activate adaptive antioxidant responses. Nuclear factor erythroid 2-related factor 2 (Nrf2) plays an important role in the defense of oxidative stress by regulating the expression of antioxidant enzymes, gamma-glutamylcysteine ligase (γGCL) and heme oxygenase-1 (HO-1). We investigated whether treadmill exercise protects dopaminergic neurons by regulating the Nrf2 antioxidant system in a 1-methyl-4-phenylpyridine (MPP(+))-induced parkinsonian rat model. We found that MPP(+) induced early decreases in total glutathione level and Nrf2/γGCLC (catalytic subunit of γGCL) expression, but late upregulation of HO-1 expression in association with loss of nigral dopaminergic neurons and downregulation of tyrosine hydroxylase and dopamine transporter expression in the striatum. Treadmill exercise for 4weeks induced upregulation of Nrf2 and γGCLC expression, and also prevented the MPP(+)-induced downregulation of Nrf2/γGCLC/glutathione, HO-1 upregulation, and nigrostriatal dopaminergic neurodegeneration. Moreover, the protective effect of exercise was blocked by the knockdown of Nrf2 using a lentivirus-carried shNrf2 delivery system. These results demonstrate an essential role of Nrf2 in the exercise-mediated protective effect that exercise enhances the nigrostriatal Nrf2 antioxidant defense capacity to protect dopaminergic neurons against the MPP(+)-induced toxicity.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Intoxicação por MPTP/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Degeneração Neural/metabolismo , Condicionamento Físico Animal/fisiologia , Animais , Western Blotting , Corpo Estriado/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Glutamato-Cisteína Ligase/metabolismo , Heme Oxigenase-1/metabolismo , Imuno-Histoquímica , Masculino , Degeneração Neural/reabilitação , Estresse Oxidativo/fisiologia , Ratos , Ratos Wistar
10.
Respir Res ; 12: 147, 2011 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-22054060

RESUMO

BACKGROUND: Neonatal mice developed neurological disease and pulmonary dysfunction after an infection with a mouse-adapted human Enterovirus 71 (EV71) strain MP4. However, the hallmark of severe human EV71 infection, pulmonary edema (PE), was not evident. METHODS: To test whether EV71-induced PE required a proinflammatory cytokine response, exogenous pro-inflammatory cytokines were administered to EV71-infected mice during the late stage of infection. RESULTS: After intracranial infection of EV71/MP4, 7-day-old mice developed hind-limb paralysis, pulmonary dysfunction, and emphysema. A transient increase was observed in serum IL-6, IL-10, IL-13, and IFN-γ, but not noradrenaline. At day 3 post infection, treatment with IL-6, IL-13, and IFN-γ provoked mild PE and severe emphysema that were accompanied by pulmonary dysfunction in EV71-infected, but not herpes simplex virus-1 (HSV-1)-infected control mice. Adult mice did not develop PE after an intracerebral microinjection of EV71 into the nucleus tractus solitarii (NTS). While viral antigen accumulated in the ventral medulla and the NTS of intracerebrally injected mice, neuronal loss was observed in the ventral medulla only. CONCLUSIONS: Exogenous IL-6, IL-13, and IFN-γ treatment could induce mild PE and exacerbate pulmonary abnormality of EV71-infected mice. However, other factors such as over-activation of the sympathetic nervous system may also be required for the development of classic PE symptoms.


Assuntos
Enterovirus Humano A , Infecções por Enterovirus/patologia , Interferon gama/toxicidade , Interleucina-13/toxicidade , Interleucina-6/toxicidade , Pulmão/patologia , Edema Pulmonar/patologia , Enfisema Pulmonar/patologia , Animais , Animais Recém-Nascidos , Chlorocebus aethiops , Enterovirus Humano A/imunologia , Enterovirus Humano A/patogenicidade , Infecções por Enterovirus/sangue , Infecções por Enterovirus/imunologia , Humanos , Interferon gama/sangue , Interleucina-13/sangue , Interleucina-6/sangue , Pulmão/imunologia , Pulmão/virologia , Camundongos , Camundongos Endogâmicos ICR , Paralisia/imunologia , Paralisia/patologia , Paralisia/virologia , Edema Pulmonar/sangue , Edema Pulmonar/imunologia , Enfisema Pulmonar/imunologia , Enfisema Pulmonar/virologia , Células Vero
11.
J Pineal Res ; 51(2): 233-45, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21545521

RESUMO

The induction of oxidative stress and inflammation has been closely linked in traumatic brain injury (TBI). Transcriptional factors of signal transducers and activators of transcription (STAT) proteins are redox sensitive and participate in the regulation of cytokine signaling. Previous studies demonstrated that melatonin protects neurons through its antioxidative and anti-inflammatory effects in various neuropathological conditions. However, the effect of melatonin on STAT activity after TBI has not yet been explored. In this study, we used a controlled weight-drop TBI model and found that brain contusion induced oxidative stress (a decreased level of total glutathione and an increased ratio of oxidized glutathione to total glutathione), a reduction in STAT1 DNA-binding activity, and consequently neuronal loss in a contusion depth-dependent manner. A significant increased mRNA expression of suppressor of cytokine signaling (SOCS3), inducible nitric oxide synthetase (iNOS), and interleukine-6 (IL-6), but a decreased protein expression of protein inhibitor of activated STAT (PIAS1), was found 24 hr after brain contusion. SOCS3 and PIAS1 are endogenous negative regulators of STAT1. Moreover, the combination of intraperitoneal and local (presoaked in gelfoam and placed on the traumatic cortex) administration of melatonin had the most pronounced influence in inhibiting all effects except the PIAS1 downregulation induced by brain contusion. The results suggest that SOCS-3 upregulation and oxidative stress may contribute to the STAT1 inactivation after TBI. Melatonin protects neurons from TBI by reducing oxidative stress, STAT1 inactivation, and upregulation of SOCS-3 and pro-inflammatory cytokines.


Assuntos
Antioxidantes/farmacologia , Lesões Encefálicas/metabolismo , Melatonina/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fator de Transcrição STAT1/metabolismo , Proteínas Supressoras da Sinalização de Citocina/biossíntese , Ativação Transcricional/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Animais , Lesões Encefálicas/patologia , Interleucina-6/biossíntese , Masculino , Óxido Nítrico Sintase Tipo II/biossíntese , Proteínas Inibidoras de STAT Ativados/metabolismo , RNA Mensageiro/biossíntese , Ratos , Proteína 3 Supressora da Sinalização de Citocinas
12.
Neuro Oncol ; 12(10): 1050-60, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20663792

RESUMO

Malignant glioma is a common and severe primary brain tumor with a high recurrence rate and an extremely high mortality rate within 2 years of diagnosis, even when surgical, radiological, and chemotherapeutic interventions are applied. Intravenously administered drugs have limited use because of their adverse systemic effects and poor blood-brain barrier penetration. Here, we combine 2 methods to increase drug delivery to brain tumors. Focused ultrasound transiently permeabilizes the blood-brain barrier, increasing passive diffusion. Subsequent application of an external magnetic field then actively enhances localization of a chemotherapeutic agent immobilized on a novel magnetic nanoparticle. Combining these techniques significantly improved the delivery of 1,3-bis(2-chloroethyl)-1-nitrosourea to rodent gliomas. Furthermore, the physicochemical properties of the nanoparticles allowed their delivery to be monitored by magnetic resonance imaging (MRI). The resulting suppression of tumor progression without damaging the normal regions of the brain was verified by MRI and histological examination. This noninvasive, reversible technique promises to provide a more effective and tolerable means of tumor treatment, with lower therapeutic doses and concurrent clinical monitoring.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Glioma/tratamento farmacológico , Ondas de Choque de Alta Energia/uso terapêutico , Magnetismo , Nanopartículas/administração & dosagem , Animais , Barreira Hematoencefálica/fisiologia , Carmustina/administração & dosagem , Imageamento por Ressonância Magnética , Magnetismo/métodos , Masculino , Ratos , Ratos Sprague-Dawley
13.
Free Radic Biol Med ; 49(6): 1099-108, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20615462

RESUMO

Oxidative stress and lower levels of trophic factors involved in nigrostriatal dopaminergic neurodegeneration are a hallmark of Parkinson disease. Our previous studies found that fibroblast growth factor 9 (FGF9) prevented 1-methyl-4-phenylpyridinium (MPP(+))-induced nigral dopaminergic neuron death and was involved in the neuroprotection of the antioxidant melatonin. However, the protective mechanisms mediated by FGF9 remain unclear. Herein, we explored whether FGF9 regulated the cellular antioxidant defense protecting dopaminergic neurons against MPP(+) intoxication. We found that FGF9 treatment alone induced a decrease in hydrogen peroxide (H(2)O(2)) level, an increase in glutathione content, and an upregulation of gamma-glutamylcysteine synthetase (gamma-GCS) and heme oxygenase 1 (HO-1) expression in primary cortical neurons but not in astrocytes. Simultaneous treatment with FGF9 and MPP(+) prevented MPP(+)-induced neuron death and H(2)O(2) overproduction but did not affect the FGF9-increased gamma-GCS and HO-1 protein expression. Inhibition of gamma-GCS or HO-1 prevented the inhibitory effect of FGF9 on MPP(+)-induced H(2)O(2) production and death in mesencephalic dopaminergic and cortical neurons. However, in the absence of MPP(+), the FGF9-induced H(2)O(2) reduction was blocked by HO-1 inhibitors, but not by gamma-GCS inhibitors. These results indicate that FGF9 upregulates gamma-GCS and HO-1 expression to protect cortical and dopaminergic neurons from MPP(+)-induced oxidative insult.


Assuntos
Apoptose/efeitos dos fármacos , Fator 9 de Crescimento de Fibroblastos/farmacologia , Glutamato-Cisteína Ligase/metabolismo , Heme Oxigenase-1/metabolismo , Neurônios/efeitos dos fármacos , 1-Metil-4-fenilpiridínio/toxicidade , Animais , Técnicas de Cultura de Células , Células Cultivadas , Citoproteção/efeitos dos fármacos , Dopamina/metabolismo , Embrião de Mamíferos , Glutamato-Cisteína Ligase/genética , Glutationa/metabolismo , Heme Oxigenase-1/genética , Peróxido de Hidrogênio/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Regulação para Cima
14.
Eur J Neurosci ; 27(7): 1634-46, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18371080

RESUMO

WW domain-containing oxidoreductase (named WWOX, FOR or WOX1) is a pro-apoptotic protein and tumor suppressor. Animals treated with dopaminergic neurotoxin 1-methyl-4-phenyl-pyridinium (MPP+) develop Parkinson's disease (PD)-like symptoms. Here we investigated whether WOX1 is involved in MPP+-induced neurodegeneration. Upon insult with MPP+ in rat brains, WOX1 protein was upregulated and phosphorylated at Tyr33 (or activated) in the injured neurons in the striatum and cortex ipsilaterally to intoxication, as determined by immunohistochemistry and Western blotting. Also, WOX1 was present in the condensed nuclei and damaged mitochondria of degenerative neurons, as revealed by transmission immunoelectron microscopy. Time-lapse microscopy revealed that MPP+ induced membrane blebbing and shrinkage of neuroblastoma SK-N-SH cells. Dominant-negative WOX1, a potent inhibitor of Tyr33 phosphorylation, abolished this event, indicating a critical role of the phosphorylation in apoptosis. c-Jun N-terminal kinase (JNK1) is known to bind and counteract the apoptotic function of WOX1. Suppression of JNK1 function by a dominant-negative spontaneously induced WOX1 activation. WOX1 physically interacted with JNK1 in SK-N-SH cells and rat brain extracts. MPP+ rapidly increased the binding, followed by dissociation, which is probably needed for WOX1 to exert apoptosis. We synthesized a short Tyr33-phosphorylated WOX1 peptide (11 amino acid residues). Interestingly, this peptide blocked MPP+-induced neuronal death in the rat brains, whereas non-phospho-WOX1 peptide had no effect. Together, activated WOX1 plays an essential role in the MPP+-induced neuronal death. Our synthetic phospho-WOX1 peptide prevents neuronal death, suggestive of its therapeutic potential in mitigating the symptoms of PD.


Assuntos
1-Metil-4-fenilpiridínio/toxicidade , Apoptose/fisiologia , Neurônios/enzimologia , Oxirredutases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Tirosina/metabolismo , Animais , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular Tumoral , Humanos , Masculino , Neurônios/efeitos dos fármacos , Neurônios/patologia , Oxirredutases/genética , Doença de Parkinson/enzimologia , Doença de Parkinson/patologia , Doença de Parkinson/terapia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Proteínas Supressoras de Tumor/genética , Tirosina/genética , Oxidorredutase com Domínios WW
15.
Shock ; 27(5): 527-33, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17438458

RESUMO

Fluid resuscitation is vital for treating traumatic hemorrhagic shock (HS), but reperfusion is believed to have the adverse consequences of generating reactive oxygen species and inflammatory cytokines, both of which cause multiple organ dysfunctions. We investigated the effects of various resuscitation fluids on the changes of redox-sensitive molecules after HS and fluid resuscitation (HS/R). We induced HS by bleeding male Sprague-Dawley rats to a blood pressure of 30 to 40 mmHg for 60 minutes. Thirty minutes later, the rats were killed (HS group) or immediately resuscitated with shed blood (HS + BL group), L-isomer lactated Ringer's solution (HS + LR group), or hydroxyethyl starch (HS + HES group). After HS, we found a significant increase in nuclear factor kappaB DNA binding activity, which was effectively inhibited using HES solution or blood resuscitation. Moreover, resuscitation with blood or LR solution, but not HES solution, induced significant oxidative stress, manifested by a high ratio of oxidized glutathione to reduced glutathione in the lungs, liver, and spleen. HS alone, however, did not increase the ratio of the oxidized glutathione to reduced glutathione in all organs. Although the protein expression of anti-apoptotic Bcl-2 and pro-apoptotic Bax varied in different organs, we found that resuscitation using HES solution prevented the HS-induced reduction of the Bcl-2/Bax ratio in the heart. HES solution was an appropriate resuscitation fluid in reversing nuclear factor kappaB activation, maintaining the Bcl-2/Bax ratio, and preventing oxidative stress after acute HS.


Assuntos
Derivados de Hidroxietil Amido/uso terapêutico , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Ressuscitação/métodos , Choque Hemorrágico/terapia , Animais , Western Blotting , Ensaio de Desvio de Mobilidade Eletroforética , Hidratação/métodos , Glutationa/metabolismo , Dissulfeto de Glutationa/metabolismo , Derivados de Hidroxietil Amido/farmacologia , Soluções Isotônicas/farmacologia , Soluções Isotônicas/uso terapêutico , Masculino , Substitutos do Plasma/farmacologia , Substitutos do Plasma/uso terapêutico , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Lactato de Ringer , Choque Hemorrágico/metabolismo , Proteína X Associada a bcl-2/metabolismo , Proteína de Morte Celular Associada a bcl/metabolismo
16.
J Pineal Res ; 36(2): 117-25, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14962063

RESUMO

1-Methyl-4-phenylpyridinium (MPP(+)) ion, a toxic metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, is produced by monoamine oxidase B in astrocytes. MPP(+) causes a selective dopaminergic neurodegeneration, the pathophysiologic hallmark of Parkinson disease. However, the toxic effect of MPP(+) on astrocytes remains unclear. Here, we examined the effect of MPP(+) on human astrocytoma U373MG cells, with particular attention to the temporal interaction of glutathione (GSH) and reactive oxygen species (ROS) (H2O2 and O). MPP(+) induced astrocyte apoptosis in a dose-dependent manner 48 hr after treatment. Distinctive early (<6 hr) and late (24-48 hr) responses were observed. ROS production and the oxidized GSH (GSSG)/GSH ratio, indicators of oxidative stress, rose dramatically after 24 hr of MPP(+) exposure, whereas the H2O2 level transiently decreased at 6 hr. ROS overproduction and GSH dysfunction were concomitantly associated with caspase-3 activation and finally led to cell apoptosis. Moreover, GSH depletion by diethyl maleate, but not buthionine sulfoximine, caused cells to die quickly and potentiated the cytotoxicity of MPP(+). Co-treatment with melatonin, a known antioxidant secreted by the pineal gland, significantly prevented cell apoptosis by inhibiting oxidative stress and caspase-3 activation, but it did not affect that the early changes due to MPP(+) treatment. Our results demonstrate that in astrocytes, GSH is involved in the early decrease and late increase in ROS levels induced by MPP(+) treatment. Melatonin remedies the dysfunction of GSH system to block caspase-3 activation and cell apoptosis induced by oxidative stress during the long-term exposure of MPP(+).


Assuntos
1-Metil-4-fenilpiridínio/farmacologia , Astrócitos/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Glutationa/metabolismo , Melatonina/farmacologia , Espécies Reativas de Oxigênio/metabolismo , 1-Metil-4-fenilpiridínio/toxicidade , Relação Dose-Resposta a Droga , Humanos , Fatores de Tempo
17.
J Surg Res ; 106(1): 1-6, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12127800

RESUMO

BACKGROUND: Cyclosporine A (CsA) and morphine have neurotoxic and psychiatric side effects, respectively. Endogenous opiatelike peptides can elicit a number of behavioral responses that mimic the symptoms of psychiatric illness. The purpose of this study was to quantitiate the changes of Met-enkephalin (ME) and beta-endorphin (BE) after administration of CsA and morphine in surgery and to assess the antinociceptive effect. PATIENTS AND MATERIALS: Pain sensitivity, an antinociceptive indicator in rats, was determined with the hotplate test. Plasma ME and BE levels were measured with radioimmunoassays. RESULTS: In normal unoperated rats, CsA induced a profound analgesic effect concomitant with an increased plasma ME level on day 1. Morphine produced an analgesic effect on days 1 and 2, with decreased ME levels on days 2 and 3. Coadministration of CsA and morphine prolonged the analgesia from days 1 to 4 and increased the plasma ME level on day 1. No change in plasma BE level was found. In surgically operated rats, CsA induced an analgesic effect and higher ME levels than those in unoperated rats. Interestingly, the combined use of CsA and morphine prolonged the analgesia and increased plasma ME levels from days 1 to 4, with no significant change in plasma BE levels. CONCLUSIONS: Our results showed that CsA can induce antinociception and increase plasma ME levels. This induction can be potentiated by the addition of morphine. Acute neuropsychiatric manifestations in the early posttransplant period might, therefore, be due to induction of ME after coadministration of CsA and morphine.


Assuntos
Analgésicos Opioides/farmacologia , Ciclosporina/farmacologia , Encefalina Metionina/sangue , Imunossupressores/farmacologia , Morfina/farmacologia , Nociceptores/efeitos dos fármacos , Animais , Encefalina Metionina/metabolismo , Masculino , Transtornos Mentais/induzido quimicamente , Dor Pós-Operatória/tratamento farmacológico , Complicações Pós-Operatórias/psicologia , Ratos , Ratos Sprague-Dawley , Transplante/psicologia , beta-Endorfina/sangue , beta-Endorfina/metabolismo
18.
J Pineal Res ; 32(4): 262-9, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11982797

RESUMO

In this study we selected a rat model of Parkinson's disease (PD) by using intrastriatal infusion of the 1-methyl-4-phenyl-pyridinium ion (MPP+) to investigate the neuroprotective action of melatonin and its inhibitory activity on MPP+-impaired glutathione (GSH) system in the nigrostriatal system. Results show that MPP+ caused not only a severe neuronal injury in the striatum and in the ipsilateral substantia nigra (SN), but it also induced a significant decrease in GSH levels and an increase in the GSSG/GSH ratio 3 days after intrastriatal MPP+ infusion. Intraperitoneal co-administration of melatonin (10 mg/kg, five times) significantly attenuated MPP+-induced nigrostriatal neurotoxicity and GSH impairment. Depletion of cytosolic GSH by L-buthionine sulfoximine (BSO) did not cause neuronal damage by itself. It, however, when co-administrated with MPP+, potentiated the GSH reduction in the striatum, without aggravating nigrostriatal neurodegeneration induced by MPP+. Moreover, the MPP+-caused neuronal damage was positively correlated with a rising ratio of GSSG/GSH, but not with a drop of GSH. These results suggest that the MPP+-triggered oxidative stress may play a more important role than the loss of the antioxidant GSH in determining neuronal injury. Interestingly, the neuronal damage and oxidative stress elicited by co-treatment of BSO with MPP+ were effectively reduced by melatonin. Our results hence provide direct evidence showing that melatonin attenuates MPP+-induced nigrostriatal dopaminergic injury by its ability to impede the increase of GSSG/GSH ratio; therefore melatonin may have therapeutic implications in PD.


Assuntos
1-Metil-4-fenilpiridínio/farmacologia , Corpo Estriado/efeitos dos fármacos , Glutationa/fisiologia , Melatonina/farmacologia , Substância Negra/efeitos dos fármacos , Animais , Corpo Estriado/metabolismo , Dopamina/metabolismo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Ratos , Ratos Wistar , Substância Negra/enzimologia , Substância Negra/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo
19.
Neurochem Int ; 40(2): 169-79, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11738483

RESUMO

While there is growing evidence that Bcl-2 proto-oncogene and beta-amyloid precursor proteins (APP) are neuroprotective in function, our recent studies have demonstrated that Bcl-2 and APP may be co-expressed and co-regulated in retinal neurons or glia under normal or experimental conditions. Whether Bcl-2 and APP are functionally coupled in other neuronal systems is not clear. This issue was investigated further in the present experiments by examining the expression pattern of two molecules after unilateral intrastriatal injection of 1-methyl-4-phenyl-pyridinium (MPP(+)), a neurotoxic metabolite that selectively damages dopaminergic neurons. One hour to 2 months after MPP(+) injection into rat striatum, a significant increase in Bcl-2 expression was observed in distinct populations of neurons, astrocyte-like and OX-42-positive cells not only in traumatic regions but also in remote areas including the ipsilateral cortex and substantia nigra (SN). No detectable change was observed in the striatum, cortex or SN on the contralateral side of the brain. The immunoreactive pattern and time-dependent APP increase was similar to that of Bcl-2 in the severely injured striatum and cortex. However, an up-regulation of Bcl-2 expression, but not APP, appears in dopaminergic neurons in the ipsilateral SN pars compacta where there was retrograde degeneration. In contrast, APP immunoreactivity was decreased in the hippocampus following intrastriatal injury, whereas, no alteration in Bcl-2 expression was detected. The differential changes in Bcl-2 and APP expression in nigral neurons and some other brain tissues suggest that these proteins may not be co-regulated by a common mechanism, at least in certain neuronal pathways.


Assuntos
1-Metil-4-fenilpiridínio/farmacologia , Precursor de Proteína beta-Amiloide/genética , Córtex Cerebral/fisiologia , Corpo Estriado/fisiologia , Regulação da Expressão Gênica/fisiologia , Genes bcl-2/efeitos dos fármacos , Neurônios/fisiologia , Substância Negra/fisiologia , 1-Metil-4-fenilpiridínio/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Córtex Cerebral/efeitos dos fármacos , Corpo Estriado/efeitos dos fármacos , Lateralidade Funcional , Regulação da Expressão Gênica/efeitos dos fármacos , Injeções Intraperitoneais , Masculino , Microinjeções , Neurônios/classificação , Neurônios/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Substância Negra/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA