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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Brain Behav Immun ; 25(1): 135-46, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20851176

RESUMEN

Parkinson's disease (PD) is characterized by a progressive and selective loss of dopaminergic (DA) neurons in the substantia nigra (SN). Although the etiology of PD remains unclear, neuroinflammation has been implicated in the development of PD. Running exercise (Ex) promotes neuronal survival and facilitates the recovery of brain functions after injury. Therefore, we hypothesize that Ex protects the DA neurons against inflammation-induced injury in the SN. An intraperitoneal lipopolysaccharide (LPS, 1 mg/kg) injection induced microglia activation in the SN within hours, followed by a reduction in the number of DA neurons. LPS reduced the level of dopamine in the striatum and impaired the performance of motor coordination. Furthermore, the levels of the brain-derived neurotrophic factor (BDNF) were reduced in the SN by the LPS treatment. Four weeks of Ex before LPS treatment completely prevented the LPS-induced loss of DA neurons, reduction of dopamine levels and dysfunction of motor movement. Ex did not change the LPS-induced status of microglia activation or the levels of cytokines/chemokines, but restored the levels of LPS-reduced BDNF-TrkB signaling molecules. Blocking the action of BDNF, through its receptor TrkB antagonist, abolished the Ex-induced protection against LPS-induced DA neuron loss. Intrastriatal perfusion of BDNF alone was sufficient to counteract the LPS-induced DA neuron loss. Altogether, our results show that Ex protects DA neurons against inflammation-induced insults. The neuroprotective effects of Ex are not due to the modulation of inflammation status, but rather to the activation of the BDNF-TrkB signaling pathway.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/fisiología , Dopamina/fisiología , Inflamación/patología , Degeneración Nerviosa/patología , Neuronas/fisiología , Condicionamiento Físico Animal/fisiología , Carrera/fisiología , Sustancia Negra/patología , Animales , Carbazoles/farmacología , Recuento de Células , Supervivencia Celular/fisiología , Quimiocinas/biosíntesis , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Citocinas/biosíntesis , Inmunohistoquímica , Alcaloides Indólicos/farmacología , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/fisiología , Actividad Motora/fisiología , Desempeño Psicomotor/fisiología , Transducción de Señal/fisiología
9.
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
10.
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
11.
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
12.
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
13.
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.

14.
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
15.
J Physiol ; 587(Pt 13): 3221-31, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19451201

RESUMEN

Chronic exercise has been reported to improve cognitive function. However, whether and how different types of exercise affect various learning and memory tasks remain uncertain. To address this issue, male BALB/c mice were trained for 4 weeks under two different exercise protocols: moderate treadmill running or voluntary wheel running. After exercise training, their spatial memory and aversive memory were evaluated by a Morris water maze and by one-trial passive avoidance (PA), respectively. Levels of neural plasticity-related proteins, i.e. brain-derived neurotrophic factor (BDNF), tropomyosin-related kinase B (TrkB) and synaptotagmin I (Syt I), in hippocampus and amygdala were determined by ELISA or immunoblotting. Finally, the functional roles of these proteins in the basolateral amygdala were verified by locally blocking them with K252a (a TrkB kinase inhibitor), or lentivirus expressing Syt I shRNA. We found that (1) although both moderate treadmill running and wheel running improved the Morris water maze performance, only the former improved PA performance; (2) likewise, both exercise protocols upregulated the BDNF-TrkB pathway and Syt I in the hippocampus, whereas only treadmill exercise upregulated their expression levels in the amygdala; (3) local injection of K252a abolished the treadmill exercise-facilitated PA performance and upregulation of amygdalar TrkB and Syt I; and (4) local administration of Syt I shRNA abolished the treadmill exercise-facilitated PA performance and upregulation of amygdalar Syt I. Therefore, our results support the notion that different forms of exercise induce neuroplasticity changes in different brain regions, and thus exert diverse effects on various forms of learning and memory.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/fisiología , Aprendizaje/fisiología , Memoria/fisiología , Actividad Motora/fisiología , Sinaptotagmina I/fisiología , Amígdala del Cerebelo/fisiología , Animales , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Secuencia de Bases , Carbazoles/farmacología , Corticosterona/sangre , Hipocampo/fisiología , Alcaloides Indólicos/farmacología , Aprendizaje/efectos de los fármacos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Músculo Esquelético/fisiología , Plasticidad Neuronal/fisiología , ARN Interferente Pequeño/genética , Receptor trkB/antagonistas & inhibidores , Receptor trkB/fisiología , Carrera/fisiología , Sinaptotagmina I/antagonistas & inhibidores , Sinaptotagmina I/genética
16.
J Neurochem ; 109(5): 1400-12, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19476551

RESUMEN

Oxidative stress and down-regulated trophic factors are involved in the pathogenesis of nigrostriatal dopamine(DA)rgic neurodegeneration in Parkinson's disease. Fibroblast growth factor 9 (FGF9) is a survival factor for various cell types; however, the effect of FGF9 on DA neurons has not been studied. The antioxidant melatonin protects DA neurons against neurotoxicity. We used MPP(+) to induce neuron death in vivo and in vitro and investigated the involvement of FGF9 in MPP(+) intoxication and melatonin protection. We found that MPP(+) in a dose- and time-dependent manner inhibited FGF9 mRNA and protein expression, and caused death in primary cortical neurons. Treating neurons in the substantia nigra and mesencephalic cell cultures with FGF9 protein inhibited the MPP(+)-induced cell death of DA neurons. Melatonin co-treatment attenuated MPP(+)-induced FGF9 down-regulation and DA neuronal apoptosis in vivo and in vitro. Co-treating DA neurons with melatonin and FGF9-neutralizing antibody prevented the protective effect of melatonin. In the absence of MPP(+), the treatment of FGF9-neutralizing antibody-induced DA neuronal apoptosis whereas FGF9 protein reduced it indicating that endogenous FGF9 is a survival factor for DA neurons. We conclude that MPP(+) down-regulates FGF9 expression to cause DA neuron death and that the prevention of FGF9 down-regulation is involved in melatonin-provided neuroprotection.


Asunto(s)
1-Metil-4-fenilpiridinio/toxicidad , Dopamina/metabolismo , Factor 9 de Crecimiento de Fibroblastos/metabolismo , Herbicidas/toxicidad , Melatonina/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Animales Recién Nacidos , Anticuerpos/farmacología , Muerte Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática/métodos , Factor 9 de Crecimiento de Fibroblastos/genética , Factor 9 de Crecimiento de Fibroblastos/inmunología , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Mesencéfalo/citología , Proteínas Asociadas a Microtúbulos/metabolismo , Ratas , Ratas Wistar , Sales de Tetrazolio , Tiazoles , Factores de Tiempo , Tirosina 3-Monooxigenasa/metabolismo
17.
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
18.
Eur J Neurosci ; 27(7): 1634-46, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18371080

RESUMEN

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.


Asunto(s)
1-Metil-4-fenilpiridinio/toxicidad , Apoptosis/fisiología , Neuronas/enzimología , Oxidorreductasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Tirosina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular Tumoral , Humanos , Masculino , Neuronas/efectos de los fármacos , Neuronas/patología , Oxidorreductasas/genética , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/terapia , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteínas Supresoras de Tumor/genética , Tirosina/genética , Oxidorreductasa que Contiene Dominios WW
19.
Neurobiol Learn Mem ; 89(4): 489-96, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-17892954

RESUMEN

While serotonin (5-HT) may impair learning and memory, exercise has been reported to improve them. Whether chronic exercise can facilitate fear memory via regulating the serotonin system is unknown. We examined the effects of 4-week treadmill exercise training on levels of 5-HT and its metabolite 5-hydroxyindoleacetic acid (5-HIAA), the protein expression of its receptor 5-HT(1A) and transporter in the amygdala, hippocampus and prefrontal cortex of male Sprague-Dawley rats. Our results demonstrated that treadmill exercise (1) improved the passive avoidance learning performance; (2) decreased the 5-HT level in the hippocampus; (3) decreased the expression of 5-HT(1A) receptor in the amygdala without altering the transporter expression. Moreover, pretreatment with 0.1 mg/kg 8-hydroxy-di-n-propylamino tetralin, a selective 5-HT(1A) receptor agonist, impaired the passive avoidance performance and completely abolished the exercise-enhanced fear memory. Our results suggest that down-regulation of the 5-HT system in the limbic system, i.e., the reduction of the hippocampus 5-HT content and the amygdala 5-HT(1A) receptor expression, may be involved in the exercise-enhanced fear memory.


Asunto(s)
Reacción de Prevención/fisiología , Sistema Límbico/fisiología , Condicionamiento Físico Animal/fisiología , Serotonina/metabolismo , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Amígdala del Cerebelo/fisiología , Animales , Reacción de Prevención/efectos de los fármacos , Regulación hacia Abajo/fisiología , Prueba de Esfuerzo , Miedo/fisiología , Ácido Hidroxiindolacético/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Agonistas de Receptores de Serotonina/farmacología
20.
Neurobiol Learn Mem ; 90(1): 81-9, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18374609

RESUMEN

Cognitive functions usually involve various synaptic proteins and neurotrophic factors in the hippocampus. However, whether treadmill exercise can improve learning and memory by upregulating some of these molecules remain unraveled. To address this question, male BALB/c mice were divided into control and exercise groups, the latter group went through 4 weeks of treadmill exercise training. At the end of exercise training period, they were either tested for passive avoidance (PA) performance or sacrificed for quantifying the hippocampal levels of brain-derived neurotrophic factor (BDNF), tropomyosin-related kinase B (TrkB, the BDNF receptor), synaptotagmin (a Ca(2+)-dependent synaptic vesicle protein), and SNAP-25 (a presynaptic vesicular fusion protein). Our results showed that treadmill exercise training (1) increased the retention latency without affecting the fear acquisition in the PA test, (2) transiently increased the hippocampal BDNF level at 1, 2, and 4h after the completion of exercise training, and (3) persistently increased the hippocampal protein levels of full-length TrkB, phosphorylated TrkB and synaptotagmin, but not truncated TrkB or SNAP-25. Moreover, the protein expression level of full-length TrkB or synaptotagmin was positively correlated with PA performance in mice. Finally, inhibition of TrkB signaling by K252a abolished the exercise-facilitated PA performance and upregulation of TrkB and synaptotagmin. Taken together, these data suggest that the upregulation of TrkB and synaptotagmin in the hippocampus contributes to the exercise-facilitated aversive memory.


Asunto(s)
Reacción de Prevención/fisiología , Hipocampo/fisiología , Glicoproteínas de Membrana/metabolismo , Memoria/fisiología , Condicionamiento Físico Animal/fisiología , Proteínas Tirosina Quinasas/metabolismo , Sinaptotagmina I/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Carbazoles/farmacología , Corticosterona/sangre , Inhibidores Enzimáticos/farmacología , Alcaloides Indólicos/farmacología , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Ratones , Ratones Endogámicos BALB C , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Carrera/fisiología , Proteína 25 Asociada a Sinaptosomas/metabolismo , Regulación hacia Arriba/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA