Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Neurobiol ; 58(5): 2396-2406, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33421017

RESUMEN

Proper development of neuronal cells is important for brain functions, and impairment of neuronal development may lead to neuronal disorders, implying that improvement in neuronal development may be a therapeutic direction for these diseases. Huntington's disease (HD) is a neurodegenerative disease characterized by impairment of neuronal structures, ultimately leading to neuronal death and dysfunctions of the central nervous system. Based on previous studies, fibroblast growth factor 9 (FGF9) may provide neuroprotective functions in HD, and FGFs may enhance neuronal development and neurite outgrowth. However, whether FGF9 can provide neuronal protective functions through improvement of neuronal morphology in HD is still unclear. Here, we study the effects of FGF9 on neuronal length in HD and attempt to understand the related working mechanisms. Taking advantage of striatal cell lines from HD knock-in mice, we found that FGF9 increases total neuronal length and upregulates several structural and synaptic proteins under HD conditions. In addition, activation of nuclear factor kappa B (NF-kB) signaling by FGF9 was observed to be significant in HD cells, and blockage of NF-kB leads to suppression of these structural and synaptic proteins induced by FGF9, suggesting the involvement of NF-kB signaling in these effects of FGF9. Taken these results together, FGF9 may enhance total neuronal length through upregulation of NF-kB signaling, and this mechanism could serve as an important mechanism for neuroprotective functions of FGF9 in HD.


Asunto(s)
Cuerpo Estriado/efectos de los fármacos , Factor 9 de Crecimiento de Fibroblastos/farmacología , Enfermedad de Huntington/metabolismo , FN-kappa B/metabolismo , Proyección Neuronal/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Línea Celular , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Ratones , Neuronas/metabolismo , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
2.
Life Sci ; 267: 118952, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33383048

RESUMEN

AIMS: Huntington's disease (HD) is a neurodegenerative disease that causes deficits in neurite outgrowth, which suggests that enhancement of neurite outgrowth is a potential direction by which to improve HD. Our previous publications showed that fibroblast growth factor 9 (FGF9) provides anti-apoptosis and anti-oxidative functions in striatal cell models of HD through the extracellular signal-regulated kinases (ERK) pathway, and FGF9 also stimulates cytoskeletons to enhance neurite outgrowth via nuclear factor kappa B (NF-kB) signaling. In this study, we further demonstrate the importance of the ERK pathway for the neurite outgrowth induced by FGF9 in HD striatal models. MATERIALS AND METHODS: FGF9 was treated with ERK (U0126) or NF-kB (BAY11-7082) inhibitors in STHdhQ7/Q7 and STHdhQ111/Q111 striatal knock-in cell lines to examine neurite outgrowth, cytoskeletal markers, and synaptic proteins via immunofluorescence staining and Western blotting. NF-kB activity was analyzed by NF-kB promoter reporter assay. KEY FINDINGS: Here, we show that suppression of ERK signaling significantly inhibits FGF9-induced neurite outgrowth, cytoskeletal markers, and synaptic proteins in HD striatal cells. In addition, we also show suppression of ERK signaling significantly decreases FGF9-induced NF-kB activation, whereas suppression of NF-kB does not decrease FGF9-induced ERK signaling. These results suggest that FGF9 activates ERK signaling first, stimulates NF-kB upregulation, and then enhances neurite outgrowth in HD striatal cells. SIGNIFICANCE: We elucidate the more detailed mechanisms of neurite outgrowth enhanced by FGF9 in these HD striatal cells. This study may provide insights into targeting neurite outgrowth for HD therapy.


Asunto(s)
Factor 9 de Crecimiento de Fibroblastos/metabolismo , Factor 9 de Crecimiento de Fibroblastos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neuritas/metabolismo , Animales , Butadienos/farmacología , Línea Celular , Células Cultivadas , Cuerpo Estriado/metabolismo , Inhibidores Enzimáticos/farmacología , Factor 9 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Ratones , Ratones Transgénicos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Neuritas/efectos de los fármacos , Proyección Neuronal/fisiología , Nitrilos/farmacología , Proteínas Nucleares/metabolismo , Estrés Oxidativo/efectos de los fármacos , Proteínas Recombinantes/farmacología , Transducción de Señal , Sulfonas/farmacología
3.
Front Neurosci ; 14: 573509, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33041765

RESUMEN

Moderate exercise and mild hypothermia have protective effects against brain injury and neurodegeneration. Running in a cold environment alters exercise-induced hyperthermia and outcomes; however, evaluations of post-exercise cold exposure related to exercise benefits for the brain are relatively rare. We investigated the effects of 4°C cold exposure after exercise on exercise-induced thermal responses and neuroprotection in an MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-induced Parkinsonian mouse model. Male C57BL/6J mice were pretreated with MPTP for five consecutive days and follow-up treadmill exercise for 4 weeks. After 1-h running at a 22°C temperature, the mice were exposed to a 4°C environment for 2 h. An MPTP injection induced a transient drop in body and brain temperatures, while mild brain hypothermia was found to last for 4 weeks after MPTP treatment. Preventing brain hypothermia by exercise or 4°C exposure was associated with an improvement in MPTP-induced striatal uncoupling protein 4 (UCP4) downregulation and nigrostriatal dopaminergic neurodegeneration. However, 4°C exposure after exercise abrogated the exercise-induced beneficial effects and thermal responses in MPTP-treated mice, including a low amplitude of exercise-induced brain hyperthermia and body temperature while at rest after exercise. Our findings elucidate that post-exercise thermoregulation and UCP4 expression are important in the neuroprotective effects of exercise against MPTP toxicity.

4.
Biol Reprod ; 103(6): 1300-1313, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32886743

RESUMEN

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.


Asunto(s)
Trastornos del Desarrollo Sexual/genética , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Gónadas/fisiología , Procesos de Determinación del Sexo/fisiología , Proteína de la Región Y Determinante del Sexo/metabolismo , Animales , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Femenino , Factor 9 de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica/fisiología , Humanos , Masculino , Ratones , Regiones Promotoras Genéticas , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína de la Región Y Determinante del Sexo/genética , Técnicas de Cultivo de Tejidos , Regulación hacia Arriba
5.
Mol Neurobiol ; 57(10): 4090-4105, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32666227

RESUMEN

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.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Dinaminas/metabolismo , Hipotermia Inducida , Dinámicas Mitocondriales , Neuronas/patología , 1-Metil-4-fenilpiridinio , Animales , Proteínas de Ciclo Celular/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Corteza Cerebral/patología , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 5 Dependiente de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Dinámicas Mitocondriales/efectos de los fármacos , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Ratas Sprague-Dawley , Roscovitina/farmacología , Regulación hacia Arriba/efectos de los fármacos
6.
Theranostics ; 10(6): 2817-2831, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32194837

RESUMEN

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.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Factor 9 de Crecimiento de Fibroblastos/farmacología , Células Madre Mesenquimatosas , Nervio Ciático , Animales , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Regeneración Nerviosa/efectos de los fármacos , Péptidos/farmacología , Ratas , Ratas Sprague-Dawley , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Nervio Ciático/efectos de los fármacos , Nervio Ciático/lesiones
7.
Free Radic Biol Med ; 130: 256-266, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30391672

RESUMEN

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.


Asunto(s)
Antioxidantes/farmacología , Encéfalo/metabolismo , Factor 9 de Crecimiento de Fibroblastos/genética , Enfermedad de Huntington/tratamiento farmacológico , Factor 2 Relacionado con NF-E2/genética , Animales , Encéfalo/patología , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Glutamato-Cisteína Ligasa/genética , Glutatión Reductasa/genética , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Superóxido Dismutasa/genética , Corteza Visual/metabolismo , Corteza Visual/patología
8.
Cancer Sci ; 109(11): 3503-3518, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30191630

RESUMEN

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.


Asunto(s)
Factor de Transcripción E2F1/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Tumor de Células de Leydig/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteína de Retinoblastoma/metabolismo , Neoplasias Testiculares/metabolismo , Animales , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Masculino , Ratones , Fosforilación , Transducción de Señal
9.
Cell Physiol Biochem ; 48(2): 605-617, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30021209

RESUMEN

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.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 9 de Crecimiento de Fibroblastos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Butadienos/farmacología , Caspasa 3/metabolismo , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Transgénicos , Nitrilos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Corteza Visual/citología , Corteza Visual/efectos de los fármacos , Corteza Visual/metabolismo , Proteína bcl-X/metabolismo
10.
Am J Transl Res ; 9(11): 4785-4806, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29218080

RESUMEN

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.

11.
J Pineal Res ; 61(2): 230-40, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27159033

RESUMEN

Mitochondrial dysfunction and oxidative stress are involved in the pathogenesis of Parkinson's disease (PD). Mitochondrial morphology is dynamic and precisely regulated by the mitochondrial fission and fusion machinery. Aberrant mitochondrial fragmentation controlled by the mitochondrial fission protein, dynamin-related protein 1 (Drp1), may result in cell death. Our previous results showed that melatonin protected neurons by inhibiting oxidative stress in a 1-methyl-4-phenylpyridinium (MPP(+) )-induced PD model. However, the effect of melatonin on mitochondrial dynamics remains uncharacterized. Herein, we investigated the effect of melatonin and the role of Drp1 on MPP(+) -induced mitochondrial fission in rat primary cortical neurons. We found that MPP(+) induced a rapid increase in the ratio of GSSG:total glutathione (a marker of oxidative stress) and mitochondrial fragmentation, Drp1 upregulation within 4 hours, and finally resulted in neuron loss 48 hours after the treatment. Neurons overexpressing wild-type Drp1 promoted mitochondrial and nuclear fragmentation; however, neurons overexpressing dominant-negative Drp1(K38A) or cotreated with melatonin exhibited significantly reduced MPP(+) -induced mitochondrial fragmentation and neuron death. Moreover, melatonin cotreatment prevented an MPP(+) -induced high ratio of GSSG and mitochondrial Drp1 upregulation. The prevention of mitochondrial fission by melatonin was not found in neurons transfected with wild-type Drp1. These results provide a new insight that the neuroprotective effect of melatonin against MPP(+) toxicity is mediated by inhibiting the oxidative stress and Drp1-mediated mitochondrial fragmentation.


Asunto(s)
Corteza Cerebral/metabolismo , Dinaminas/metabolismo , Intoxicación por MPTP/metabolismo , Melatonina/farmacología , Dinámicas Mitocondriales/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , 1-Metil-4-fenilpiridinio/toxicidad , Animales , Corteza Cerebral/patología , Dinaminas/genética , Intoxicación por MPTP/genética , Intoxicación por MPTP/patología , Dinámicas Mitocondriales/genética , Neuronas/patología , Estrés Oxidativo/genética , Ratas , Ratas Sprague-Dawley
12.
Organogenesis ; 12(2): 61-77, 2016 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-27078042

RESUMEN

An expressional lack of fibroblast growth factor 9 (FGF9) would cause male-to-female sex reversal in the mouse, implying the essential role of FGF9 in testicular organogenesis and maturation. However, the temporal expression of FGF9 and its receptors during testicular development remains elusive. In this study, immunohistochemistry was used to identify the localization of FGF9 and its receptors at different embryonic and postnatal stages in mice testes. Results showed that FGF9 continuously expressed in the testis during development. FGF9 had highest expression in the interstitial region at 17-18 d post coitum (dpc) and in the spermatocytes, spermatids and Leydig cell on postnatal days (pnd) 35-65. Regarding receptor expression, FGFR1 and FGFR4 were evenly expressed in the whole testis during the embryonic and postnatal stages. However, FGFR2 and FGFR3 were widely expressed during the embryonic testis development with higher FGFR2 expression in seminiferous tubules at 16-18 dpc and higher FGFR3 expression in interstitial region at 17-18 dpc. In postnatal stage, FGFR2 extensively expressed with higher expression at spermatids and Leydig cells on 35-65 pnd and FGFR3 widely expressed in the whole testis. Taken together, these results strongly suggest that FGF9 is correlated with the temporal expression profiles of FGFR2 and FGFR3 and possibly associated with testis development.


Asunto(s)
Factor 9 de Crecimiento de Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Embrión de Mamíferos/metabolismo , Factor 9 de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica , Masculino , Ratones Endogámicos C57BL , Proteínas Tirosina Quinasas Receptoras/genética , Testículo/citología , Testículo/crecimiento & desarrollo , Testículo/metabolismo
13.
Stress ; 19(1): 125-32, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26473638

RESUMEN

Psychological stress is an important global health problem. It is well documented that stress increases the incidences of various cardiovascular disorders. Regular exercise is known to reduce resting blood pressure (BP) and heart rate (HR). This study was designed to clarify the effects of long-term exercise on stress-evoked cardiovascular responses and to emphasize post-stress recovery effects. Male Wistar rats underwent 8 weeks of moderate treadmill training, with cardiovascular responses, autonomic nervous system activities and local Fos reactivity changes in the cardiovascular regulation center were monitored before, during and after immobilization stress. A spectral analysis of cardiovascular parameters was used to examine autonomic nervous activities. We found that long-term exercise (i) lowered resting BP, HR and sympathetic activity, but increased resting parasympathetic activity and baroreflex sensitivity (BRS); (ii) accelerated post-stress recovery of stress-evoked cardiovascular and sympathetic responses along with increased BRS and (iii) accelerated post-stress recovery of stress-evoked neuron activations in the paraventricular nucleus, but delayed it in the nucleus of the tractus solitarius. We conclude that, in rats, long-term exercise accelerated recovery of stress-evoked cardiovascular responses differentially altering hypothalamic and medullar neuron activities.


Asunto(s)
Sistema Nervioso Parasimpático/fisiopatología , Condicionamiento Físico Animal/fisiología , Estrés Fisiológico/fisiología , Estrés Psicológico/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Animales , Sistema Nervioso Autónomo/fisiopatología , Barorreflejo/fisiología , Presión Sanguínea/fisiología , Enfermedades Cardiovasculares , Sistema Cardiovascular , Prueba de Esfuerzo , Frecuencia Cardíaca/fisiología , Masculino , Núcleo Hipotalámico Paraventricular/fisiopatología , Ratas , Ratas Wistar , Descanso , Restricción Física , Núcleo Solitario/fisiopatología
14.
Free Radic Biol Med ; 89: 274-86, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26424114

RESUMEN

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.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Hemo-Oxigenasa 1/metabolismo , Factor 2 Relacionado con NF-E2/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Animales , Western Blotting , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Factor 9 de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/genética , Técnicas para Inmunoenzimas , Masculino , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/genética , Ratas , Ratas Wistar , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Factores de Crecimiento de Fibroblastos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Regulación hacia Arriba
15.
Exp Neurol ; 263: 50-62, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25286336

RESUMEN

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.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Intoxicación por MPTP/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Degeneración Nerviosa/metabolismo , Condicionamiento Físico Animal/fisiología , Animales , Western Blotting , Cuerpo Estriado/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Glutamato-Cisteína Ligasa/metabolismo , Hemo-Oxigenasa 1/metabolismo , Inmunohistoquímica , Masculino , Degeneración Nerviosa/rehabilitación , Estrés Oxidativo/fisiología , Ratas , Ratas Wistar
16.
J Appl Physiol (1985) ; 113(6): 889-95, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22837167

RESUMEN

Although exercise usually improves motor performance, the underlying cellular changes in the cerebellum remain to be elucidated. This study aimed to investigate whether and how chronic treadmill exercise in young rats induced Purkinje cell changes to improve motor performance and rendered the cerebellum less vulnerable to toxin insults. After 1-wk familiarization of treadmill running, 6-wk-old male Wistar rats were divided into exercise and sedentary groups. The exercise group was then subjected to 8 wk of exercise training at moderate intensity. The rotarod test was carried out to evaluate motor performance. Purkinje cells in cerebellar slices were visualized by lucifer yellow labeling in single neurons and by calbindin immunostaining in groups of neurons. Compared with sedentary control rats, exercised rats not only performed better in the rotarod task, but also showed finer Purkinje cell structure (higher dendritic volume and spine density with the same dendritic field). The exercise-improved cerebellar functions were further evaluated by monitoring the long-lasting effects of intraventricular application of OX7-saporin. In the sedentary group, OX7-saporin treatment retarded the rotarod performance and induced ∼60% Purkinje cell loss in 3 wk. As a comparison, the exercise group showed much milder injuries in the cerebellum by the same toxin treatment. In conclusion, exercise training in young rats increased the dendritic density of Purkinje cells, which might play an important role in improving the motor performance. Furthermore, as Purkinje cells in the exercise group were relatively toxin resistant, the exercised rats showed good motor performance, even under toxin-treated conditions.


Asunto(s)
Anticuerpos Monoclonales/toxicidad , Inmunoconjugados/toxicidad , Inmunotoxinas/toxicidad , Actividad Motora , Síndromes de Neurotoxicidad/prevención & control , Esfuerzo Físico , Células de Purkinje/efectos de los fármacos , Proteínas Inactivadoras de Ribosomas Tipo 1/toxicidad , Animales , Anticuerpos Monoclonales/administración & dosificación , Biomarcadores/metabolismo , Calbindinas , Forma de la Célula , Técnica del Anticuerpo Fluorescente , Inmunoconjugados/administración & dosificación , Inmunotoxinas/administración & dosificación , Inyecciones Intraventriculares , Masculino , Microscopía Fluorescente , Actividad Motora/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Síndromes de Neurotoxicidad/metabolismo , Síndromes de Neurotoxicidad/patología , Células de Purkinje/metabolismo , Células de Purkinje/patología , Ratas , Ratas Wistar , Proteínas Inactivadoras de Ribosomas Tipo 1/administración & dosificación , Carrera , Proteína G de Unión al Calcio S100/metabolismo , Saporinas , Conducta Sedentaria , Factores de Tiempo
17.
Neurobiol Learn Mem ; 97(1): 140-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22085720

RESUMEN

Different exercise paradigms show differential effects on various forms of memory. We hypothesize that the differential effects of exercises on memory performance are caused by different neuroplasticity changes in relevant brain regions in response to different exercise trainings. We examined the effects of treadmill running (TR) and wheel running (WR) on the Pavlovian fear conditioning task that assesses learning and memory performance associated with the amygdala (cued conditioning) and both the amygdala and hippocampus (contextual conditioning). The skeletal muscle citrate synthase activity, an indicator of aerobic capacity, was elevated in rats received 4 w of TR, but not WR. While both TR and WR elevated the contextual conditional response, only TR facilitated the cued conditional response. Using a single-neuron labeling technique, we found that while both TR and MR enlarged the dendritic field and increased the spine density in hippocampal CA3 neurons, only TR showed these effects in basolateral amygdalar neurons. Moreover, both types of exercise upregulated synaptic proteins (i.e., TrkB and SNAP-25) in the hippocampus; however only TR showed similar effects in the amygdala. Injection of K252a, a TrkB kinase inhibitor, in the dorsal hippocampus or basolateral amygdala abolished the exercise-facilitated contextual or cued fear learning and memory performance, respectively, regardless of the types of exercise. In summary, our results supported that different types of exercise affect the performance of learning and memory via BDNF-TrkB signaling and neuroplasticity in specific brain regions. The brain region-specific neuronal adaptations are possibly induced by various levels of intensity/stress elicited by different types of exercise.


Asunto(s)
Condicionamiento Clásico/fisiología , Espinas Dendríticas/fisiología , Memoria/fisiología , Neuronas/fisiología , Condicionamiento Físico Animal/métodos , Amígdala del Cerebelo/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Señales (Psicología) , Miedo/fisiología , Hipocampo/fisiología , Condicionamiento Físico Animal/fisiología , Ratas , Receptor trkB/metabolismo , Proteína 25 Asociada a Sinaptosomas/metabolismo , Sinaptotagmina I/metabolismo
18.
Respir Res ; 12: 147, 2011 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-22054060

RESUMEN

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.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus/patología , Interferón gamma/toxicidad , Interleucina-13/toxicidad , Interleucina-6/toxicidad , Pulmón/patología , Edema Pulmonar/patología , Enfisema Pulmonar/patología , Animales , Animales Recién Nacidos , Chlorocebus aethiops , Enterovirus Humano A/inmunología , Enterovirus Humano A/patogenicidad , Infecciones por Enterovirus/sangre , Infecciones por Enterovirus/inmunología , Humanos , Interferón gamma/sangre , Interleucina-13/sangre , Interleucina-6/sangre , Pulmón/inmunología , Pulmón/virología , Ratones , Ratones Endogámicos ICR , Parálisis/inmunología , Parálisis/patología , Parálisis/virología , Edema Pulmonar/sangre , Edema Pulmonar/inmunología , Enfisema Pulmonar/inmunología , Enfisema Pulmonar/virología , Células Vero
19.
J Hypertens ; 29(12): 2339-48, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22002337

RESUMEN

OBJECTIVE: The cardiovascular integration center not only sends out signals to offset the stimulus-induced responses but also resets the resting blood pressure. We hypothesize that GABAergic adaptations in the hypothalamus participate in the chronic exercise-induced cardiovascular resetting effects in conscious normotensive animals. METHODS: Male Wistar rats were subjected to chronic moderate exercise (CME, 8-week treadmill running at moderate intensity). A biotelemetry system was used to measure blood pressure, heart rate, autonomic nervous activities, baroreflex sensitivity and endogenous GABAergic activities in the paraventricular nucleus and the posterior hypothalamic area. Hypothalamic specimens were collected for quantifying GABA-related proteins and GABAergic neurons. RESULTS: CME reduced resting blood pressure, heart rate, sympathetic activity and enhanced parasympathetic activity and baroreflex sensitivity. Additionally, CME elevated the resting level of hypothalamic GABAergic activities, increased the percentage of GABAergic neurons in the hypothalamus and upregulated the hypothalamic protein levels of neuronal nitric oxide synthase, GAD67 and gephyrin, but not GABAA receptor. Moreover, a single bout of moderate exercise transiently elevated blood pressure and heart rate with prolonged high levels of neural controls (sympathetic activity, baroreflex sensitivity and hypothalamic GABAergic activities). CME accelerated the postexercise recovery in cardiovascular parameters and neural control alterations. CONCLUSION: Chronic treadmill running in normotensive rats augmented the GABAergic system in both paraventricular nucleus and posterior hypothalamic area, resulting in lower resting blood pressure, heart rate and sympathetic tone under conscious unrestraint conditions. This study provides insight into mechanisms important for explaining how chronic exercise resets the resting blood pressure.


Asunto(s)
Presión Sanguínea/fisiología , Prueba de Esfuerzo/veterinaria , Hipotálamo Posterior/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Condicionamiento Físico Animal/fisiología , Adaptación Fisiológica , Animales , Bicuculina/farmacología , Presión Sanguínea/efectos de los fármacos , Proteínas Portadoras/metabolismo , Corticosterona/sangre , Antagonistas del GABA/farmacología , Glutamato Descarboxilasa/metabolismo , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Hemodinámica , Hipotálamo Posterior/citología , Hipotálamo Posterior/efectos de los fármacos , Masculino , Proteínas de la Membrana/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Ratas , Ratas Wistar , Receptores de GABA/metabolismo , Receptores de GABA-A/metabolismo , Procesamiento de Señales Asistido por Computador , Telemetría
20.
J Pineal Res ; 51(2): 233-45, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21545521

RESUMEN

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.


Asunto(s)
Antioxidantes/farmacología , Lesiones Encefálicas/metabolismo , Melatonina/farmacología , Proteínas del Tejido Nervioso/metabolismo , Estrés Oxidativo/efectos de los fármacos , Factor de Transcripción STAT1/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/biosíntesis , Activación Transcripcional/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Animales , Lesiones Encefálicas/patología , Interleucina-6/biosíntesis , Masculino , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Proteínas Inhibidoras de STAT Activados/metabolismo , ARN Mensajero/biosíntesis , Ratas , Proteína 3 Supresora de la Señalización de Citocinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...