Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Int J Mol Sci ; 25(12)2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38927995

RESUMEN

Neural precursor cells (NPCs) that persist in the postnatal/adult subventricular zone (SVZ) express connexins that form hemichannels and gap junctions. Gap junctional communication plays a role in NPC proliferation and differentiation during development, but its relevance on postnatal age remains to be elucidated. In this work we aimed to evaluate the effect of the blockade of gap junctional communication on proliferation and cell fate of NPCs obtained from the SVZ of postnatal rats. NPCs were isolated and expanded in culture as neurospheres. Electron microscopy revealed the existence of gap junctions among neurosphere cells. Treatment of cultures with octanol, a broad-spectrum gap junction blocker, or with Gap27, a specific blocker for gap junctions formed by connexin43, produced a significant decrease in bromodeoxyuridine incorporation. Octanol treatment also exerted a dose-dependent antiproliferative effect on glioblastoma cells. To analyze possible actions on NPC fate, cells were seeded in the absence of mitogens. Treatment with octanol led to an increase in the percentage of astrocytes and oligodendrocyte precursors, whereas the percentage of neurons remained unchanged. Gap27 treatment, in contrast, did not modify the differentiation pattern of SVZ NPCs. Our results indicate that general blockade of gap junctions with octanol induces significant effects on the behavior of postnatal SVZ NPCs, by reducing proliferation and promoting glial differentiation.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Uniones Comunicantes , Células-Madre Neurales , Neuroglía , Octanoles , Animales , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Proliferación Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Ratas , Octanoles/farmacología , Neuroglía/efectos de los fármacos , Neuroglía/metabolismo , Neuroglía/citología , Células Cultivadas , Ventrículos Laterales/citología , Ventrículos Laterales/metabolismo , Ventrículos Laterales/efectos de los fármacos , Conexina 43/metabolismo , Ratas Wistar , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/citología , Animales Recién Nacidos , Humanos
2.
Bull Exp Biol Med ; 171(3): 333-337, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34297290

RESUMEN

We studied the participation of JNK and p53 in the realization of the growth potential of different types of progenitors of the subventricular zone of mouse brain and secretion of neurotrophins by glial cells. The stimulating role of these signaling molecules in mitotic activity and specialization of multipotent neural stem cells was shown. It was found that JNK and p53 do not participate in the regulation of committed neuronal progenitor cells (clonogenic PSA-NCAM+ cells). A dependence of neurotrophic growth factors in individual populations of neuroglia on activity of these protein kinase and transcription factor was revealed. The role of JNK and p53 in astrocytes consists in stimulation of their secretion, and in microglial cells, on the contrary, in its inhibition. The secretory neurotrophic function of oligodendrogliocytes is not associated with JNK and p53 activity.


Asunto(s)
Astrocitos/metabolismo , MAP Quinasa Quinasa 4/genética , Células Madre Multipotentes/metabolismo , Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Proteína p53 Supresora de Tumor/genética , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Benzotiazoles/farmacología , Antígeno CD56/genética , Antígeno CD56/metabolismo , Medios de Cultivo Condicionados/farmacología , Regulación de la Expresión Génica , Ventrículos Laterales/citología , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Madre Multipotentes/citología , Células Madre Multipotentes/efectos de los fármacos , Factores de Crecimiento Nervioso/biosíntesis , Factores de Crecimiento Nervioso/genética , Moléculas de Adhesión de Célula Nerviosa/genética , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Neuroglía/citología , Neuroglía/efectos de los fármacos , Transducción de Señal , Tolueno/análogos & derivados , Tolueno/farmacología , Proteína p53 Supresora de Tumor/metabolismo
3.
Differentiation ; 119: 1-9, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33848959

RESUMEN

Taxol (paclitaxel), a chemotherapeutic agent for several cancers, can adversely affect the peripheral nervous system. Recently, its negative impact on cognitive function in cancer patients has become evident. In rodents, taxol impaired learning and memory, with other possible negative effects on the brain. In this study, we investigated the effects of taxol on cultured neural stem cells (NSCs) from the mouse neurogenic region, the subventricular zone (SVZ). Taxol significantly decreased both proliferation and neuronal differentiation of NSCs. Transient treatment with taxol for one day during a 4-day differentiation greatly decreased neurogenesis along with an abnormal cell cycle progression. Yet, taxol did not kill differentiated Tuj1+ neurons and those neurons had longer neurites than neurons under control conditions. For glial differentiation, taxol significantly reduced oligodendrogenesis as observed by immunostaining for Olig2 and O4. However, differentiation of astrocytes was not affected by taxol. In contrast, differentiated oligodendrocytes were extremely sensitive to taxol. Almost no Olig2-positive cells were observed after three days of treatment with taxol. Taxol has distinct effects on neurons and glial cells during their production through differentiation from NSCs as well as post-differentiation. Thus, we suggest that taxol might interfere with neurogenesis of NSCs possibly through a disturbance in the cell cycle and may eliminate differentiated oligodendrocytes.


Asunto(s)
Diferenciación Celular/genética , Células-Madre Neurales/citología , Neuronas/citología , Factor de Transcripción 2 de los Oligodendrocitos/genética , Tubulina (Proteína)/genética , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/crecimiento & desarrollo , Ratones , Proteínas del Tejido Nervioso/genética , Células-Madre Neurales/efectos de los fármacos , Neuritas/metabolismo , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Neuronas/metabolismo , Paclitaxel/farmacología
4.
Metab Brain Dis ; 36(5): 969-981, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33608831

RESUMEN

Intrauterine growth restriction (IUGR) affects brain neural stem cell (NSC) differentiation. In the present study, we investigated whether taurine supplementation may improve NSC differentiation in IUGR fetal rats via the protein kinase A-cyclic adenosine monophosphate (cAMP) response element protein-brain derived neurotrophic factor (PKA-CREB-BDNF) signaling pathway. The IUGR fetal rat model was established with a low-protein diet. Fresh subventricular zone (SVZ) tissue from the fetuses on the 14th day of pregnancy was microdissected and dissociated into single-cell suspensions, then was cultured to form neurospheres. The neurospheres were divided into the control group, the IUGR group, the IUGR+taurine (taurine) group, the IUGR+H89 (H89) group and the IUGR+taurine+H89 (taurine+H89) group. The mRNA and protein expression levels of PKA, CREB and BDNF were measured by reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting (WB). Tuj-1-positive neurons and GFAP-positive glial cells were detected by immunofluorescence. The total number of proliferating NSCs and the percentage of Tuj-1-positive neurons in the IUGR group were lower than those in the control group, but the percentage of GFAP-positive cells was higher in the IUGR group than in the control group. Taurine supplementation increased the total number of neural cells and the percentage of Tuj-1-positive neurons, and reduced the percentage of GFAP-positive cells among differentiated NSCs after IUGR. H89 reduced the total number and percentage of Tuj-1-positive neurons and increased the percentage of GFAP-positive cells. The mRNA and protein levels of PKA, CREB, and BDNF were lower in the IUGR group. The mRNA and protein expression levels of these factors were increased by taurine supplementation but reduced by the addition of H89. Taurine supplementation increased the ratio of neurons to glial cells and prevented gliosis in the differentiation of NSCs in IUGR fetal rats by activating the PKA-CREB-BDNF signaling pathway.


Asunto(s)
Retardo del Crecimiento Fetal/metabolismo , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Taurina/farmacología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
5.
Int J Mol Sci ; 22(2)2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33445804

RESUMEN

Neural progenitor cells (NPCs) are self-renewing and multipotent cells that persist in the postnatal and adult brain in the subventricular zone and the hippocampus. NPCs can be expanded in vitro to be used in cell therapy. However, expansion is limited, since the survival and proliferation of adult NPCs decrease with serial passages. Many signaling pathways control NPC survival and renewal. Among these, purinergic receptor activation exerts differential effects on the biology of adult NPCs depending on the cellular context. In this study, we sought to analyze the effect of a general blockade of purinergic receptors with suramin on the proliferation and survival of NPCs isolated from the subventricular zone of postnatal rats, which are cultured as neurospheres. Treatment of neurospheres with suramin induced a significant increase in neurosphere diameter and in NPC number attributed to a decrease in apoptosis. Proliferation and multipotency were not affected. Suramin also induced an increase in the gap junction protein connexin43 and in vascular endothelial growth factor, which might be involved in the anti-apoptotic effect. Our results offer a valuable tool for increasing NPC survival before implantation in the lesioned brain and open the possibility of using this drug as adjunctive therapy to NPC transplantation.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Antagonistas Purinérgicos/farmacología , Receptores Purinérgicos/metabolismo , Células Madre/efectos de los fármacos , Suramina/farmacología , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/metabolismo , Masculino , Células-Madre Neurales/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Células Madre/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
J Neurochem ; 156(4): 465-480, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32052426

RESUMEN

Neural stem/progenitor cells (NSPCs) persist in the mammalian subventricular zone throughout life, where they can be activated in response to physiological and pathophysiological stimuli. A recent study indicates metabotropic glutamate receptor 4 (mGluR4) is involved in regulating NSPCs behaviors. Therefore, defining mGluR4 function in NSPCs is necessary for determining novel strategies to enhance the intrinsic potential for brain regeneration after injuries. In this study, mGluR4 was functionally expressed in SVZ-derived NSPCs from male Sprague-Dawley rats, in which the cyclic adenosine monophosphate concentration was reduced after treatment with the mGluR4-specific agonist VU0155041. Additionally, lateral ventricle injection of VU0155041 significantly decreased 5-bromo-2'-deoxyuridine (BrdU)+ and Ki67+ cells, while increased Doublecortin (DCX)/BrdU double-positive cells in SVZ. In cultured NSPCs, mGluR4 activation decreased the ratio of BrdU+ cells, G2/M-phase cells, and inhibited Cyclin D1 expression, whereas it increased neuron-specific class III ß-tubulin (Tuj1) expression and the number of Tuj1, DCX, and PSA-NCAM-positive cells. However, pharmacological blocking mGluR4 with the antagonist MSOP or knockdown of mGluR4 abolished the effects of VU0155041 on NSPCs proliferation and neuronal differentiation. Further investigation demonstrated that VU0155041 treatment down-regulated AKT phosphorylation and up-regulated expression of the phosphatase and tensin homolog protein (PTEN) in NSPCs culture. Moreover VU0155041-induced proliferating inhibition and neuronal differentiating amplification in NSPCs were significantly hampered by VO-OHpic, a PTEN inhibitor. We conclude that activation of mGluR4 in SVZ-derived NSPCs suppresses proliferation and enhances their neuronal differentiation, and regulation of PTEN may be involved as a potential intracellular target of mGluR4 signal. Cover Image for this issue: https://doi.org/10.1111/jnc.15052.


Asunto(s)
Diferenciación Celular/fisiología , Ventrículos Laterales/metabolismo , Células-Madre Neurales/metabolismo , Fosfohidrolasa PTEN/biosíntesis , Receptores de Glutamato Metabotrópico/metabolismo , Anilidas/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Células Cultivadas , Ácidos Ciclohexanocarboxílicos/farmacología , Relación Dosis-Respuesta a Droga , Proteína Doblecortina , Expresión Génica , Ventrículos Laterales/citología , Ventrículos Laterales/efectos de los fármacos , Masculino , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Neurogénesis/fisiología , Fosfohidrolasa PTEN/genética , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/agonistas
7.
Neuro Oncol ; 22(8): 1150-1161, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32296841

RESUMEN

BACKGROUND: Imagining ways to prevent or treat glioblastoma (GBM) has been hindered by a lack of understanding of its pathogenesis. Although overexpression of platelet derived growth factor with two A-chains (PDGF-AA) may be an early event, critical details of the core biology of GBM are lacking. For example, existing PDGF-driven models replicate its microscopic appearance, but not its genomic architecture. Here we report a model that overcomes this barrier to authenticity. METHODS: Using a method developed to establish neural stem cell cultures, we investigated the effects of PDGF-AA on subventricular zone (SVZ) cells, one of the putative cells of origin of GBM. We microdissected SVZ tissue from p53-null and wild-type adult mice, cultured cells in media supplemented with PDGF-AA, and assessed cell viability, proliferation, genome stability, and tumorigenicity. RESULTS: Counterintuitive to its canonical role as a growth factor, we observed abrupt and massive cell death in PDGF-AA: wild-type cells did not survive, whereas a small fraction of null cells evaded apoptosis. Surviving null cells displayed attenuated proliferation accompanied by whole chromosome gains and losses. After approximately 100 days in PDGF-AA, cells suddenly proliferated rapidly, acquired growth factor independence, and became tumorigenic in immune-competent mice. Transformed cells had an oligodendrocyte precursor-like lineage marker profile, were resistant to platelet derived growth factor receptor alpha inhibition, and harbored highly abnormal karyotypes similar to human GBM. CONCLUSION: This model associates genome instability in neural progenitor cells with chronic exposure to PDGF-AA and is the first to approximate the genomic landscape of human GBM and the first in which the earliest phases of the disease can be studied directly.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Células-Madre Neurales , Factor de Crecimiento Derivado de Plaquetas , Proteína p53 Supresora de Tumor , Animales , Neoplasias Encefálicas/inducido químicamente , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Células Cultivadas , Glioblastoma/inducido químicamente , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/metabolismo , Ventrículos Laterales/patología , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/patología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/metabolismo
8.
Biosci Rep ; 40(1)2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31919522

RESUMEN

Adult neurogenesis, the production of newborn neurons from neural stem cells (NSCs) has been suggested to be decreased in patients with schizophrenia. A similar finding was observed in an animal model of schizophrenia, as indicated by decreased bromodeoxyuridine (BrdU) labelling cells in response to a non-competitive N-methyl-d-aspartate (NMDA) receptor antagonist. The antipsychotic drug clozapine was shown to counteract the observed decrease in BrdU-labelled cells in hippocampal dentate gyrus (DG). However, phenotypic determination by immunohistochemistry analysis could not reveal whether BrdU-positive cells were indeed NSCs. Using a previously established cell model for analysing NSC protection in vitro, we investigated a protective effect of clozapine on NSCs. Primary NSCs were isolated from the mouse subventricular zone (SVZ), we show that clozapine had a NSC protective activity alone, as evident by employing an ATP cell viability assay. In contrast, haloperidol did not show any NSC protective properties. Subsequently, cells were exposed to the non-competitive NMDA-receptor antagonist ketamine. Clozapine, but not haloperidol, had a NSC protective/anti-apoptotic activity against ketamine-induced cytotoxicity. The observed NSC protective activity of clozapine was associated with increased expression of the anti-apoptotic marker Bcl-2, decreased expression of the pro-apoptotic cleaved form of caspase-3 and associated with decreased expression of the autophagosome marker 1A/1B-light chain 3 (LC3-II). Collectively, our findings suggest that clozapine may have a protective/anti-apoptotic effect on NSCs, supporting previous in vivo observations, indicating a neurogenesis-promoting activity for clozapine. If the data are further confirmed in vivo, the results may encourage an expanded use of clozapine to restore impaired neurogenesis in schizophrenia.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Clozapina/farmacología , Antagonistas de Aminoácidos Excitadores/toxicidad , Ketamina/toxicidad , Ventrículos Laterales/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Células Madre Adultas/metabolismo , Células Madre Adultas/patología , Animales , Caspasa 3/metabolismo , Células Cultivadas , Haloperidol/farmacología , Ventrículos Laterales/metabolismo , Ventrículos Laterales/patología , Masculino , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Neurogénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal
9.
Neuropharmacology ; 162: 107813, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31628935

RESUMEN

The sister incretins glucagon-like peptide-1 (GLP-1) and glucagon dependent insulinotropic polypeptide (GIP) are growth factors responsible for re-sensitizing insulin signalling. Interestingly, their analogues, originally developed to treat type 2 diabetes (T2D), have demonstrated a range of neuroprotective and neurorestorative properties. Novel peptide GLP-1/GIP dual agonist (DA) shows good effects in diabetic patients, superior to the effects demonstrated by single GIP or GLP-1 mimetics. Furthermore, novel DAs have shown considerable neuroprotection in neurodegenerative models. Here, we investigated the neuroprotective and restorative involvement of the DA DA-JC1 and liraglutide (Lg), a single GLP-1 receptor analogue, in vitro using human neuroblastoma (SH-SY5Y) against oxidative stress induced by oxygen peroxide (H2O2), and in vivo, in a mouse model of Alzheimer's disease (AD), APP/PS1. First, we determined the ideal concentration of the peptides and demonstrated that DA-JC1 protects cells against oxidative stress more than Lg, improving cell viability, normalizing reactive oxygen species (ROS) and attenuating DNA damage generated by H2O2. Moreover, in 10-to-12-months-old APP/PS1 animals treated for 4 weeks, both Lg and DA-JC1 were very efficient in stimulating neurogenesis and reducing some important hallmarks of AD, but DA-JC1 was better than Lg in attenuating crucial neuroinflammatory markers, especially reactive astrocyte, in both wild-type (WT) and APP/PS1 hippocampal regions. Altogether, this study suggests an interactive role of GLP-1 and GIP receptors, enhancing the efficiency of single GLP-1 analogues, especially in attenuating oxidative stress and neuroinflammation. We confirm that combining GLP-1 and GIP results in a variety of beneficial effects, providing key evidences for the development of a promising therapeutic strategy for AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/agonistas , Incretinas/farmacología , Liraglutida/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Receptores de la Hormona Gastrointestinal/agonistas , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/efectos de los fármacos , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular Tumoral , Supervivencia Celular , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Peróxido de Hidrógeno , Técnicas In Vitro , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/metabolismo , Ventrículos Laterales/patología , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Neuronas/patología , Estrés Oxidativo/efectos de los fármacos , Péptidos/farmacología , Presenilina-1/genética
10.
J Appl Toxicol ; 39(11): 1557-1567, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31368586

RESUMEN

The developing brain is uniquely vulnerable to toxic chemical exposures. Studies indicate that neural stem cell (NSC) self-renewal is susceptible to oxidative stress caused by xenobiotics. However, the impact of antioxidants on NSC self-renewal and the potential mechanisms remain elusive. In this study, primary murine neural progenitor cells (mNPCs) from the subventricular zone were used as a research model. In addition, paraquat (PQ) was used to elicit oxidative stress and N-acetylcysteine (NAC) was used as a powerful antioxidant. mNPCs were treated with 80 µm PQ for 24 hours with or without 4 hours of NAC pretreatment. Our results showed that PQ treatment increased intracellular reactive oxygen species production, decreased cell viability and DNA synthesis, and promoted cell apoptosis. Meanwhile, pretreatment with NAC alleviated PQ-induced cytotoxicity in mNPCs. To elucidate the mechanisms further, we found that NAC pretreatment prevented PQ-induced reactive oxygen species production, mitochondrial fragmentation and autophagy in mNPCs. NAC-pretreated cells showed increased anti-apoptotic protein Bcl-2 and decreased pro-apoptotic protein Bax expression. Similarly, NAC pretreatment increased p-mTOR and decreased LC3B-II protein expression. Moreover, NAC decreased mitophagy related mRNA Pink1 and Parkin expression. Taken together, our results suggested that the antioxidant NAC treatment significantly attenuated PQ-induced mNPC self-renewal disruption through decreasing autophagy and salvaging mitochondrial morphology. These findings revealed a potential mechanism for neurological treatment relating to antioxidant and suggested potentially relevant implications for PQ-related neurodegenerative disorders. Thus, our study also provided insight into therapeutic strategies for the neurotoxic effects of oxidative stress-associated toxicants.


Asunto(s)
Acetilcisteína/farmacología , Antioxidantes/farmacología , Autofagia/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Paraquat/toxicidad , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/patología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/ultraestructura , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Estrés Oxidativo/efectos de los fármacos , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo
11.
J Med Virol ; 91(6): 1158-1167, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30701563

RESUMEN

BACKGROUND: Opioid-primed relapse is a global burden. Although current strategies have improved, optimal therapy is urgently needed. METHODS: A recombinant adenovirus (Ad-NEP) expressing ß-endorphin (ß-EP) was designed and injected intracerebroventricularly (icv) into the right lateral ventricle in rats. Spatial and temporal ß-EP expression in the lateral ventricle wall, subventricular zone and adjacent choroid plexus and the ß-EP concentration in the cerebrospinal fluid (CSF) were observed during a 21-day period. A morphine priming-induced conditioned place preference (CPP) rat model was established. The ß-EP-ir neuron counts, CSF ß-EP concentration, and CPP score, which were used to evaluate morphine-primed reinstatement following extinction, were recorded 7 days after the icv injection. Additionally, the rats were pretreated with the irreversible µ opioid receptor antagonist ß-funaltrexamine (ß-FNA) and the selective κ opioid receptor antagonist nor-binaltorphimine (nor-BNI) to identify the receptor-dependent mechanism. RESULTS: Both peak ß-EP expression in target neurons and the peak CSF ß-EP concentration occurred 7 to 8 days after Ad-NEP icv injection. The sustainable increase in the CSF ß-EP concentration was correlated with a decrease in the CPP score 7 days after the Ad-NEP icv injection. Furthermore, reinstatement was almost reversed by ß-FNA pretreatment 24 hours before the behavioral test, but nor-BNI had little effect. CONCLUSION: The increasing cerebrospinal fluid ß-endorphin concentrations showed that the therapeutic effect on opioid relapse occurred predominantly through a µ opioid receptor-dependent mechanism. The Ad-NEP adenovirus can be considered an alternative therapy for opioid relapse.


Asunto(s)
Conducta Animal/efectos de los fármacos , Morfina/administración & dosificación , Narcóticos/farmacología , Receptores Opioides mu/efectos de los fármacos , betaendorfina/líquido cefalorraquídeo , betaendorfina/genética , Adenoviridae/genética , Animales , Animales Modificados Genéticamente , Ventrículos Laterales/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley , Recurrencia , Prevención Secundaria
12.
Glia ; 66(11): 2456-2469, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30500112

RESUMEN

Astrocyte-derived ciliary neurotrophic factor (CNTF) promotes adult subventricular zone (SVZ) neurogenesis. We found that focal adhesion kinase (FAK) and JNK, but not ERK or P38, repress CNTF in vitro. Here, we defined the FAK-JNK pathway and its regulation of CNTF in mice, and the related leukemia inhibitory factor (LIF) and interleukin-6 (IL-6), which promote stem cell renewal at the expense of neurogenesis. Intrastriatal injection of FAK inhibitor, FAK14, in adult male C57BL/6 mice reduced pJNK and increased CNTF expression in the SVZ-containing periventricular region. Injection of a JNK inhibitor increased CNTF without affecting LIF and IL-6, and increased SVZ proliferation and neuroblast formation. The JNK inhibitor had no effect in CNTF-/- mice, suggesting that JNK inhibits SVZ neurogenesis by repressing CNTF. Inducible deletion of FAK in astrocytes increased SVZ CNTF and neurogenesis, but not LIF and IL-6. Intrastriatal injection of inhibitors suggested that P38 reduces LIF and IL-6 expression, whereas ERK induces CNTF and LIF. Intrastriatal FAK inhibition increased LIF, possibly through ERK, and IL-6 through another pathway that does not involve P38. Systemic injection of FAK14 also inhibited JNK while increasing CNTF, but did not affect P38 and ERK activation, or LIF and IL-6 expression. Importantly, systemic FAK14 increased SVZ neurogenesis in wild-type C57BL/6 and CNTF+/+ mice, but not in CNTF-/- littermates, indicating that it acts by upregulating CNTF. These data show a surprising differential regulation of related cytokines and identify the FAK-JNK-CNTF pathway as a specific target in astrocytes to promote neurogenesis and possibly neuroprotection in neurological disorders.


Asunto(s)
Astrocitos/metabolismo , Factor Neurotrófico Ciliar/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Ventrículos Laterales/citología , Sistema de Señalización de MAP Quinasas/fisiología , Neurogénesis/fisiología , Animales , Antracenos/farmacología , Astrocitos/efectos de los fármacos , Línea Celular Tumoral , Factor Neurotrófico Ciliar/genética , Citocinas/genética , Citocinas/metabolismo , Inhibidores Enzimáticos/farmacología , Quinasa 1 de Adhesión Focal/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Antígeno Ki-67/metabolismo , Ventrículos Laterales/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurogénesis/efectos de los fármacos , Fosforilación/efectos de los fármacos , Fosforilación/genética , Factores de Tiempo
13.
Sci Rep ; 8(1): 14947, 2018 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-30297722

RESUMEN

Chinese herbal medicines (CHMs) have been used to treat human diseases for thousands of years. Among them, Ginkgo biloba is reported to be beneficial to the nervous system and a potential treatment of neurological disorders. Since the presence of adult neural stem cells (NSCs) brings hope that the brain may heal itself, whether the effect of Ginkgo biloba is on NSCs remains elusive. In this study, we found that Ginkgo biloba extract (GBE) and one of its main ingredients, ginkgolide B (GB) promoted cell cycle exit and neuronal differentiation in NSCs derived from the postnatal subventricular zone (SVZ) of the mouse lateral ventricle. Furthermore, the administration of GB increased the nuclear level of ß-catenin and activated the canonical Wnt pathway. Knockdown of ß-catenin blocked the neurogenic effect of GB, suggesting that GB promotes neuronal differentiation through the Wnt/ß-catenin pathway. Thus, our data provide a potential mechanism underlying the therapeutic effect of GBE or GB on brain injuries and neurodegenerative disorders.


Asunto(s)
Ginkgólidos/farmacología , Lactonas/farmacología , Ventrículos Laterales/citología , Células-Madre Neurales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Animales , Línea Celular Tumoral , Células Cultivadas , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/metabolismo , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo
14.
Pharmazie ; 73(10): 585-588, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30223922

RESUMEN

Recent studies have shown that telmisartan (TMS) is effective for the protection against ischemia/brain damage in rat models. However, the specific underlying mechanism is poorly understood. In line with previous results, our data showed that TMS improves CBF and physiological variables, including pH, pCO2, pO2. Through CD31 immunofluorescence staining, reduction of blood vessel density was found in MCAO group, but TMS treatment could enhance the cerebral vascular density in the ischemic area. Meanwhile, TMS treatment could enhance the number of BrdU/lectin double-positive cells. Furthermore, the reduction of nestin-positive cells was identified in the brain of MCAO rats, while the number of nestin-positive cells was significantly increased after TMS administration. Furthermore, the expression of ERS-related proteins, including GRP78, CHOP/GADD153, Caspase12 was increased after MCAO, but was decreased after administration of TMS, thereby enhancing angiogenesis and neuron regeneration.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Telmisartán/farmacología , Animales , Apoptosis/efectos de los fármacos , Encéfalo/irrigación sanguínea , Encéfalo/metabolismo , Encéfalo/patología , Caspasa 12/metabolismo , Proteínas de Choque Térmico/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/metabolismo , Ventrículos Laterales/patología , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Factor de Transcripción CHOP/metabolismo
15.
World Neurosurg ; 115: e375-e385, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29678708

RESUMEN

OBJECTIVE: Several survival prediction models for patients with glioblastoma have been proposed, but none is widely used. This study aims to identify the predictors of overall survival (OS) and to conduct an independent comparative analysis of 5 prediction models. METHODS: Multi-institutional data from 159 patients with newly diagnosed glioblastoma who received adjuvant temozolomide concomitant chemoradiotherapy (CCRT) were collected. OS was assessed by Cox proportional hazards regression and adjusted for known prognostic factors. An independent CCRT patient cohort was used to externally validate the 1) RTOG (Radiation Therapy Oncology Group) recursive partitioning analysis (RPA) model, 2) Yang RPA model, and 3) Wee RPA model, Chaichana model, and the RTOG nomogram model. The predictive accuracy for each model at 12-month survival was determined by concordance indices. Calibration plots were performed to ascertain model prediction precision. RESULTS: The median OS for patients who received CCRT was 19.0 months compared with 12.7 months for those who did not (P < 0.001). Independent predictors were: 1) subventricular zone II tumors (hazard ratio [HR], 1.6; 95% confidence interval [CI], 1.0-2.5); 2) methylguanine methyltransferase promoter methylation (HR, 0.36; 95% CI, 0.2-0.6); and 3) extent of resection of >85% (HR, 0.59; 95% CI, 0.4-0.9). For 12-month OS prediction, the RTOG nomogram model was superior to the RPA models with a c-index of 0.70. Calibration plots for 12-month survival showed that none of the models was precise, but the RTOG nomogram performed relatively better. CONCLUSIONS: The RTOG nomogram best predicted 12-month OS. Methylguanine methyltransferase promoter methylation status, subventricular zone tumor location, and volumetric extent of resection should be considered when constructing prediction models.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Dacarbazina/análogos & derivados , Glioblastoma/mortalidad , Metiltransferasas/farmacología , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/terapia , Quimioradioterapia/métodos , Terapia Combinada/métodos , Metilación de ADN/efectos de los fármacos , Metilasas de Modificación del ADN/metabolismo , Dacarbazina/farmacología , Femenino , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/patología , Masculino , Persona de Mediana Edad , Temozolomida , Proteínas Supresoras de Tumor/metabolismo
16.
Arch Toxicol ; 92(1): 529-539, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28884398

RESUMEN

Polybrominated diphenyl ethers (PBDEs) are additive flame retardants widely used in various products (e.g., textiles, consumer electronics, and plastics). Strong evidence indicates that PBDEs are developmental neurotoxicants that can cause neurodevelopmental disabilities and cognitive defects. Currently, decabromodiphenyl ether (BDE 209) is the only PBDE permitted for production in most countries. This study investigated the impact of BDE 209 on postnatal neurogenesis in the subventricular zone (SVZ) of ICR mice. For this purpose, pregnant ICR mice were orally administrated a daily dose of 0, 20 or 100 mg/kg BDE 209 from gestation day 6 to postnatal day 16. Bromodeoxyuridine (BrdU) incorporation and in vivo postnatal electroporation were performed to label the newly generated cells in the SVZ. On PND 16, a reduction of type-B stem cells was found in the 100 mg/kg group. BDE 209 also decreased the number of newborn cells and Calretinin+ interneurons in granule cell layer at the dose of 100 mg/kg. In addition, we observed impaired neuronal migration and dendritic development of newborn olfactory granule cells in both 20 and 100 mg/kg groups. In conclusion, developmental exposure to BDE 209 produces adverse effects on SVZ neurogenesis and dendritic growth of mouse offspring. These findings suggest a potential risk of BDE 209 in human neurodevelopment.


Asunto(s)
Éteres Difenilos Halogenados/toxicidad , Ventrículos Laterales/efectos de los fármacos , Bulbo Olfatorio/efectos de los fármacos , Animales , Animales Recién Nacidos , Calreticulina/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dendritas/efectos de los fármacos , Dendritas/patología , Femenino , Retardadores de Llama/toxicidad , Ventrículos Laterales/patología , Masculino , Ratones Endogámicos ICR , Neurogénesis/efectos de los fármacos , Bulbo Olfatorio/patología , Embarazo , Efectos Tardíos de la Exposición Prenatal , Células Madre/efectos de los fármacos , Células Madre/patología
17.
Neuroscience ; 364: 202-211, 2017 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-28943249

RESUMEN

Neurogenesis in the subventricular zone (SVZ) plays a vital role in neurologic recovery after stroke. However, only a small fraction of newly generated neuroblasts from the SVZ will survive long-term. Successful migration and survival of neuroblasts requires angiogenesis, lesion-derived chemo-attractants, and appropriate local microenvironments, which are partly regulated by the platelet-derived growth factor receptor (PDGFR) signaling pathway. In this study, we investigated the effects of PDGFR inhibition in a mouse model of transient middle cerebral artery occlusion (MCAO). We blocked the pathway using a nonselective PDGFR inhibitor, crenolanib, during the acute post-MCAO phase (days 1-3) or during the sub-acute phase (days 7-9). Downregulating the PDGFR signaling pathway with crenolanib from day 1 to day 3 after MCAO significantly decreased the migration of neuroblasts from the SVZ to the peri-infarct region, decreased angiogenesis, and lowered expression of vascular endothelial growth factor, stromal cell-derived factor-1, and monocyte chemotactic protein-1. Downregulation of the PDGFR signaling pathway on days 7-9 with crenolanib significantly increased apoptosis of the neuroblasts that had migrated to the peri-infarct region, increased the number of activated microglia, and decreased the expression of brain-derived neurotrophic factor, neurotrophin-3, and interleukin-10. Crenolanib treatment increased the apoptosis of pericytes and decreased the pericyte/vascular coverage, but had no effects on apoptosis of astrocytes. We conclude that the PDGFR signaling pathway plays a vital role in the SVZ neurogenesis after stroke. It can also affect angiogenesis, lesion-derived chemo-attractants, and the local microenvironment, which are all important to stroke-induced neurogenesis.


Asunto(s)
Bencimidazoles/farmacología , Infarto de la Arteria Cerebral Media , Ventrículos Laterales/efectos de los fármacos , Neurogénesis/efectos de los fármacos , Piperidinas/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Bencimidazoles/administración & dosificación , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/inmunología , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Piperidinas/administración & dosificación
18.
J Mol Neurosci ; 63(2): 198-205, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28871488

RESUMEN

Neural stem cells are self-renewing, multipotent cells that can be found in subventricular (SVZ) and subgranular (SGZ) zones of the brain. These zones are susceptible to irradiation-induced apoptosis and oxidative stress. Melatonin (MLT) is a natural protector of neural cells against toxicity. The aim of this study was to evaluate the effects of MLT as a radio-protective material effective in reducing tissue lesions in the SVZ of the brain and changing local apoptotic potential in rats. Twenty-five Gray irradiation was applied on adult rat brain for this study. One hour before irradiation, 100 mg/kg/IP MLT was injected, and 6 h later, the animals were sacrificed. The antioxidant enzymes and MDA activity levels were measured post-sacrifice. Also, the expression level of Nestin and caspase 3 were studied by immunohistochemistry. Spectrophotometric analysis showed significant increases in the amount of malondialdehyde (MDA) levels in the irradiation-exposed (RAD) group compared to that of the control (Co) group (P < 0.05). Pre-treatment with MLT (100 mg/kg) ameliorates the harmful effects of the aforementioned 25 Gy irradiation by increasing antioxidant enzyme activity and decreasing MDA levels. A significant reduction in apoptotic cells was observed in rats treated with MLT 1 h before exposure (P < 0.001). Nestin-positive cells were also reduced in the RAD group (P < 0.001). Our results confirm the anti-apoptotic and antioxidant role of MLT. The MLT concentration used may serve as a threshold for significant protection against 25 Gy gamma irradiations on neural stem cells in SVZ.


Asunto(s)
Apoptosis , Ventrículos Laterales/efectos de los fármacos , Melatonina/farmacología , Nestina/metabolismo , Traumatismos por Radiación/prevención & control , Protectores contra Radiación/farmacología , Animales , Ventrículos Laterales/metabolismo , Ventrículos Laterales/efectos de la radiación , Masculino , Melatonina/administración & dosificación , Melatonina/uso terapéutico , Nestina/genética , Traumatismos por Radiación/tratamiento farmacológico , Radiación Ionizante , Protectores contra Radiación/administración & dosificación , Protectores contra Radiación/uso terapéutico , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
19.
Toxicology ; 380: 30-37, 2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28163110

RESUMEN

Cadmium (Cd) is a heavy metal with a long biological half-life in humans and is recognized as a toxic pollutant. Cd is also a potential neurotoxicant and its exposure is associated with olfactory impairment in humans. However, the molecular and cellular mechanisms of Cd neurotoxicity are not well defined. Adult neurogenesis is a process that generates functional neurons from adult neural stem/progenitor cells (aNPCs). It occurs in specific regions of the adult brain including the subventricular zone (SVZ) along the lateral ventricles in mammals, a process that is critical for olfaction. Various external stimuli can modulate adult neurogenesis and the effect of neurotoxicants on adult neurogenesis is just beginning to be elucidated. Since Cd exposure can impair olfaction in humans, the goal of this study is to investigate the effects of Cd on SVZ adult neurogenesis and underlying mechanisms using primary cultured SVZ-aNPCs. In this study, we report that low-level Cd exposure decreases cell number, induces apoptosis, and inhibits cell proliferation in SVZ-aNPCs. Furthermore, Cd exposure significantly increases phosphorylation of c-Jun NH2-terminal kinase (JNK), and p38 MAP kinase in these cells, indicative of JNK and p38 activation. Pharmacological inhibition of JNK or p38 MAPK kinases attenuated Cd-induced cell loss and apoptosis. Cd treatment did not cause cell loss or apoptosis in SVZ-aNPCs prepared from transgenic mice null for the neural-specific JNK3 isoform. These data suggest a critical role for p38 and JNK3 MAP kinases in Cd neurotoxicity. These results are, to our knowledge, the first demonstration that Cd impairs SVZ adult neurogenesis in vitro, which may contribute to its neurotoxicity in olfaction.


Asunto(s)
Cadmio/toxicidad , Proliferación Celular/efectos de los fármacos , Proteína Quinasa 10 Activada por Mitógenos/metabolismo , Células-Madre Neurales/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Ventrículos Laterales/citología , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Quinasa 10 Activada por Mitógenos/genética , Células-Madre Neurales/patología , Fosforilación , Proteínas Quinasas p38 Activadas por Mitógenos/genética
20.
Stem Cells ; 35(2): 458-472, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27538853

RESUMEN

The phosphodiesterase 7 (PDE7) enzyme is one of the enzymes responsible for controlling intracellular levels of cyclic adenosine 3',5'-monophosphate in the immune and central nervous system. We have previously shown that inhibitors of this enzyme are potent neuroprotective and anti-inflammatory agents. In addition, we also demonstrated that PDE7 inhibition induces endogenous neuroregenerative processes toward a dopaminergic phenotype. Here, we show that PDE7 inhibition controls stem cell expansion in the subgranular zone of the dentate gyrus of the hippocampus (SGZ) and the subventricular zone (SVZ) in the adult rat brain. Neurospheres cultures obtained from SGZ and SVZ of adult rats treated with PDE7 inhibitors presented an increased proliferation and neuronal differentiation compared to control cultures. PDE7 inhibitors treatment of neurospheres cultures also resulted in an increase of the levels of phosphorylated cAMP response element binding protein, suggesting that their effects were indeed mediated through the activation of the cAMP/PKA signaling pathway. In addition, adult rats orally treated with S14, a specific inhibitor of PDE7, presented elevated numbers of proliferating progenitor cells, and migrating precursors in the SGZ and the SVZ. Moreover, long-term treatment with this PDE7 inhibitor shows a significant increase in newly generated neurons in the olfactory bulb and the hippocampus. Also a better performance in memory tests was observed in S14 treated rats, suggesting a functional relevance for the S14-induced increase in SGZ neurogenesis. Taken together, our results indicate for the first time that inhibition of PDE7 directly regulates proliferation, migration and differentiation of neural stem cells, improving spatial learning and memory tasks. Stem Cells 2017;35:458-472.


Asunto(s)
Envejecimiento/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 7/antagonistas & inhibidores , Hipocampo/enzimología , Hipocampo/crecimiento & desarrollo , Ventrículos Laterales/enzimología , Ventrículos Laterales/crecimiento & desarrollo , Neurogénesis , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 7/metabolismo , Giro Dentado/citología , Hipocampo/efectos de los fármacos , Ventrículos Laterales/efectos de los fármacos , Masculino , Células-Madre Neurales/citología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neurogénesis/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fenotipo , Inhibidores de Fosfodiesterasa/farmacología , Ratas Wistar , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA