Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 674: 36-43, 2023 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-37393642

RESUMEN

Peripheral nerve injuries have common clinical problems that are often accompanied by sensory and motor dysfunction and failure of axonal regeneration. Although various therapeutic approaches have been attempted, full functional recovery and axonal regeneration are rarely achieved in patients. In this study, we investigated the effects of recombinant adeno-associated virus (AAV) of mesencephalic astrocyte-derived neurotrophic factor (AAV-MANF) or placental growth factor (AAV-PlGF) transduced into mesenchymal stem cells (hMSC-MANF and hMSC-PlGF), which were then transplanted using human decellularized nerves (HDN) into sciatic nerve injury model. Our results showed that both AAV-MANF and AAV-PlGF were expressed in MSCs transplanted into the injury site. Behavioral measurements performed 2, 4, 6, 8, and 12 weeks after injury indicated that MANF facilitated the rapid and improved recovery of sensory and motor functions than PlGF. In addition, immunohistochemical analysis was used to quantitatively analyze the myelination of neurofilaments, Schwann cells, and regrowth axons. Both hMSC-MANF and hMSC-PlGF increased axon numbers and immunoreactive areas of axons and Schwann cells compared with the hMSC-GFP group. However, hMSC-MANF significantly improved the thickness of axons and Schwann cells compared with hMSC-PlGF. G-ratio analysis also showed a marked increase in axon myelination in axons thicker than 2.0 µm treated with MANF than that treated with PlGF. Our study suggests that transplantation of hMSC transduced with AAV-MANF has a potential to provide a novel and efficient strategy for promoting functional recovery and axonal regeneration in peripheral nerve injury.


Asunto(s)
Traumatismos de los Nervios Periféricos , Neuropatía Ciática , Humanos , Femenino , Traumatismos de los Nervios Periféricos/metabolismo , Recuperación de la Función/fisiología , Astrocitos/metabolismo , Regeneración Nerviosa/fisiología , Factor de Crecimiento Placentario/metabolismo , Neuropatía Ciática/metabolismo , Axones/metabolismo , Factores de Crecimiento Nervioso/metabolismo , Células de Schwann/metabolismo , Nervio Ciático/metabolismo
2.
Mol Psychiatry ; 26(8): 3737-3750, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-32989244

RESUMEN

Guanine nucleotide binding protein (G protein) gamma 8 (Gng8) is a subunit of G proteins and expressed in the medial habenula (MHb) and interpeduncular nucleus (IPN). Recent studies have demonstrated that Gng8 is involved in brain development; however, the roles of Gng8 on cognitive function have not yet been addressed. In the present study, we investigated the expression of Gng8 in the brain and found that Gng8 was predominantly expressed in the MHb-IPN circuit of the mouse brain. We generated Gng8 knockout (KO) mice by CRISPR/Cas9 system in order to assess the role of Gng8 on cognitive function. Gng8 KO mice exhibited deficiency in learning and memory in passive avoidance and Morris water maze tests. In addition, Gng8 KO mice significantly reduced long-term potentiation (LTP) in the hippocampus compared to that of wild-type (WT) mice. Furthermore, we observed that levels of acetylcholine (ACh) and choline acetyltransferase (ChAT) in the MHb and IPN of Gng8 KO mice were significantly decreased, compared to WT mice. The administration of nAChR α4ß2 agonist A85380 rescued memory impairment in the Gng8 KO mice, suggesting that Gng8 regulates cognitive function via modulation of cholinergic activity. Taken together, Gng8 is a potential therapeutic target for memory-related diseases and/or neurodevelopmental diseases.


Asunto(s)
Habénula , Acetilcolina , Animales , Aprendizaje , Aprendizaje por Laberinto , Ratones , Ratones Noqueados , Agonistas Nicotínicos
3.
Mol Cell ; 51(3): 374-85, 2013 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-23871434

RESUMEN

WIP1 (wild-type p53-induced phosphatase 1) functions as a homeostatic regulator of the ataxia telangiectasia mutated (ATM)-mediated signaling pathway in response to ionizing radiation (IR). Here we identify homeodomain-interacting protein kinase 2 (HIPK2) as a protein kinase that targets WIP1 for phosphorylation and proteasomal degradation. In unstressed cells, WIP1 is constitutively phosphorylated by HIPK2 and maintained at a low level by proteasomal degradation. In response to IR, ATM-dependent AMPKα2-mediated HIPK2 phosphorylation promotes inhibition of WIP1 phosphorylation through dissociation of WIP1 from HIPK2, followed by stabilization of WIP1 for termination of the ATM-mediated double-strand break (DSB) signaling cascade. Notably, HIPK2 depletion impairs IR-induced γ-H2AX foci formation, cell-cycle checkpoint activation, and DNA repair signaling, and the survival rate of hipk2+/- mice upon γ-irradiation is markedly reduced compared to wild-type mice. Taken together, HIPK2 plays a critical role in the initiation of DSB repair signaling by controlling WIP1 levels in response to IR.


Asunto(s)
Proteínas Portadoras/metabolismo , Daño del ADN/efectos de la radiación , Reparación del ADN , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Daño del ADN/genética , Células HEK293 , Células HeLa , Histonas/metabolismo , Humanos , Ratones , Ratones Transgénicos , Fosforilación , Proteína Fosfatasa 2C , Radiación Ionizante , Transducción de Señal , Ubiquitinación
4.
Biochem Biophys Res Commun ; 431(4): 796-801, 2013 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-23313493

RESUMEN

Mesenchymal stem cells (MSCs) are effective vectors in delivering a gene of interest into degenerating brain. In ex vivo gene therapy, viability of transplanted MSCs is correlated with the extent of functional recovery. It has been reported that BDNF facilitates survival of MSCs but dividing MSCs do not express the BDNF receptor, TrkB. In this study, we found that the expression of TrkB is upregulated in human MSCs by the addition of forskolin (Fsk), an activator of adenylyl cyclase. To increase survival rate of MSCs and their secretion of tropic factors that enhance regeneration of endogenous cells, we pre-exposed hMSCs with Fsk and transduced with BDNF-adenovirus before transplantation into the brain of memory deficient rats, a degenerating brain disease model induced by ibotenic acid injection. Viability of MSCs and expression of a GABA synthesizing enzyme were increased. The pre-treatment improved learning and memory, as detected by the behavioral tests including Y-maze task and passive avoidance test. These results suggest that TrkB expression of hMSCs elevates the neuronal regeneration and efficiency of BDNF delivery for treating degenerative neurological diseases accompanying memory loss.


Asunto(s)
Colforsina/farmacología , Terapia Genética/métodos , Trastornos de la Memoria/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/efectos de los fármacos , Receptor trkB/biosíntesis , Regeneración/efectos de los fármacos , Adenilil Ciclasas/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Memoria/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Neuronas/fisiología , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Transducción Genética , Regulación hacia Arriba , Ácido gamma-Aminobutírico/metabolismo
5.
Exp Mol Med ; 54(11): 1955-1966, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36376495

RESUMEN

NANOG plays a key role in cellular plasticity and the acquisition of the stem cell state during reprogramming, but its role in the regenerative process remains unclear. Here, we show that the induction of NANOG in neuronal cells is necessary for the physiological initiation of neuronal regeneration in response to ischemic stress. Specifically, we found that NANOG was preferentially expressed in undifferentiated neuronal cells, and forced expression of Nanog in neural progenitor cells (NPCs) promoted their self-renewing expansion both in ex-vivo slice cultures and in vitro limiting dilution analysis. Notably, the upstream region of the Nanog gene contains sequence motifs for hypoxia-inducible factor-1 alpha (HIF-1α). Therefore, cerebral neurons exposed to hypoxia significantly upregulated NANOG expression selectively in primitive (CD133+) cells, but not in mature cells, leading to the expansion of NPCs. Notably, up to 80% of the neuronal expansion induced by hypoxia was attributed to NANOG-expressing neuronal cells, whereas knockdown during hypoxia abolished this expansion and was accompanied by the downregulation of other pluripotency-related genes. Moreover, the number of NANOG-expressing neuronal cells were transiently increased in response to ischemic insult, predominantly in the infarct area of brain regions undergoing neurogenesis, but not in non-neurogenic loci. Together, these findings reveal a functional effect of NANOG-induction for the initiation of adaptive neuronal regeneration among heterogeneous NPC subsets, pointing to cellular plasticity as a potential link between regeneration and reprogramming processes.


Asunto(s)
Proteína Homeótica Nanog , Células-Madre Neurales , Encéfalo/metabolismo , Hipoxia/metabolismo , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Neuronas/metabolismo , Animales
6.
Biochem Biophys Res Commun ; 412(2): 328-33, 2011 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21821000

RESUMEN

Deregulation of the expression of human papillomavirus (HPV) oncogenes E6 and E7 plays a pivotal role in cervical carcinogenesis because the E6 and E7 proteins neutralize p53 and Rb tumor suppressor pathways, respectively. In approximately 90% of all cervical carcinomas, HPVs are found to be integrated into the host genome. Following integration, the core-enhancer element and P105 promoter that control expression of E6 and E7 adopt a chromatin structure that is different from that of episomal HPV, and this has been proposed to contribute to activation of E6 and E7 expression. However, the molecular basis underlying this chromatin structural change remains unknown. Previously, BAF53 has been shown to be essential for the integrity of higher-order chromatin structure and interchromosomal interactions. Here, we examined whether BAF53 is required for activated expression of E6 and E7 genes. We found that BAF53 knockdown led to suppression of expression of E6 and E7 genes from HPV integrants in cervical carcinoma cell lines HeLa and SiHa. Conversely, expression of transiently transfected HPV18-LCR-Luciferase was not suppressed by BAF53 knockdown. The level of the active histone marks H3K9Ac and H4K12Ac on the P105 promoter of integrated HPV 18 was decreased in BAF53 knockdown cells. BAF53 knockdown restored the p53-dependent signaling pathway in HeLa and SiHa cells. These results suggest that activated expression of the E6 and E7 genes of integrated HPV is dependent on BAF53-dependent higher-order chromatin structure or nuclear motor activity.


Asunto(s)
Actinas/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Viral de la Expresión Génica , Papillomavirus Humano 18/genética , Proteínas Oncogénicas Virales/genética , Activación Transcripcional , Neoplasias del Cuello Uterino/virología , Actinas/genética , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/antagonistas & inhibidores , Femenino , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Proteínas Oncogénicas Virales/antagonistas & inhibidores , Regiones Promotoras Genéticas , Interferencia de ARN , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo
7.
Acta Neurochir (Wien) ; 153(10): 2021-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21656118

RESUMEN

BACKGROUND AND AIMS: Various techniques have been investigated to enhance peripheral nerve regeneration including the application of low-intensity electrical stimulation (ES) and the administration of growth factors, especially brain-derived neurotrophic factor (BDNF). The purpose of this study was to investigate the effects of combining short-term (ES) and recombinant adenoviral vector-mediated BDNF (BDNF-Ad) transfer, in comparison to each sole modality, on peripheral nerve regeneration in a rat model with crush-injured sciatic nerve. METHODS: Sixty male Sprague-Dawley rats (250-300 g) were equally distributed into four groups; the control group, the ES group, the BDNF-Ad group, and the combination group (n = 15 each). A standard crush injury was introduced to the sciatic nerve. The control group received no treatment after injury, the ES group received 30 minutes of low-intensity ES, the BDNF-Ad group received an injection of recombinant BDNF-Ad (concentration = 10(11) pfu/µl, 3 µl/rat) after injury, and the combination group received both ES and BDNF-Ad. The rats were followed-up for 3 weeks. RESULTS: At the end of the follow-up period, the sciatic function index (ES =-39, BDNF-Ad =-38) and number of the retrogradely labeled sensory neurons were significantly increased in the ES group and the BDNF-Ad group (ES = 326, BDNF-Ad = 264), but not in the combined treatment group, compared to the control group (SFI = -53, retrogradely labeled neurons = 229). Axonal counts were highest in the ES group (7,208 axons), axonal densities in the BDNF group (10,598 axons/mm(2)), and the myelin thickness was greater in both groups as compared to the control group. The combined treatment group showed no signs of superior recovery compared to the other groups. CONCLUSIONS: Both the ES and the BDNF-Ad treatments were effective techniques enhancing the sciatic nerve regeneration following a crush injury in rats. Nevertheless, the combined treatment with ES and BDNF-Ad produces neither a synergistic effect nor an improvement in this injury model.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Terapia por Estimulación Eléctrica/métodos , Técnicas de Transferencia de Gen , Regeneración Nerviosa/genética , Neuropatía Ciática/genética , Neuropatía Ciática/terapia , Adenoviridae/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/fisiología , Terapia Combinada/métodos , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen/tendencias , Vectores Genéticos/administración & dosificación , Vectores Genéticos/uso terapéutico , Masculino , Compresión Nerviosa/métodos , Regeneración Nerviosa/fisiología , Ratas , Ratas Sprague-Dawley , Neuropatía Ciática/fisiopatología
8.
J Clin Med ; 10(8)2021 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-33924095

RESUMEN

The sources of mesenchymal stromal cells (MSCs) for cell therapy trials are expanding, increasing the need for their characterization. Here, we characterized multi-donor, turbinate-derived MSCs (TB-MSCs) that develop from the neural crest, and compared them to bone marrow-derived MSCs (BM-MSCs). TB-MSCs had higher proliferation potential and higher self-renewal of colony forming cells, but lower potential for multi-lineage differentiation than BM-MSCs. TB-MSCs expressed higher levels of neural crest markers and lower levels of pericyte-specific markers. These neural crest-like properties of TB-MSCs were reflected by their propensity to differentiate into neuronal cells and proliferative response to nerve growth factors. Proteomics (LC-MS/MS) analysis revealed a distinct secretome profile of TB-MSCs compared to BM and adipose tissue-derived MSCs, exhibiting enrichments of factors for cell-extracellular matrix interaction and neurogenic signaling. However, TB-MSCs and BM-MSCs exhibited comparable suppressive effects on the allo-immune response and comparable stimulatory effects on hematopoietic stem cell self-renewal. In contrast, TB-MSCs stimulated growth and metastasis of breast cancer cells more than BM-MSCs. Altogether, our multi-donor characterization of TB-MSCs reveals distinct cell autonomous and paracrine properties, reflecting their unique developmental origin. These findings support using TB-MSCs as an alternative source of MSCs with distinct biological characteristics for optimal applications in cell therapy.

9.
Biochem Biophys Res Commun ; 402(1): 42-7, 2010 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-20888794

RESUMEN

Wogonin is a flavonoid isolated from Scutellaria baicalensis root, and has multiple pharmacological effects, including anti-inflammatory, anti-oxidant, and anti-cancer effects. It is also neuroprotective in the brain under many stress conditions, but wogonin does not elevate neuronal cell survival. Thus, the mechanisms controlling the neuroprotective effect of wogonin are not clear. Neural precursor cells (NPCs), present in the hippocampus and subventricular zone of adult brains, replace damaged cells. In this study we investigated the biological functions underlying the neuroprotective effect of wogonin on NPCs. We initially examined survival of NPCs but found it was slightly reduced at concentrations higher than 2µg/ml. When we explored differentiation of NPCs into neuronal cells, the number of differentiated cells expressing neurofilaments was increased remarkably (fourfold) in the hippocampal NPCs treated with wogonin. Wogonin maximally elevated the expressions of presynaptic protein, synapsin I and postsynaptic protein (PSD95) at a concentration of 0.7µg/ml. Differentiated cells containing longer neurites were significantly increased in cortical NPCs, primarily cultured from rat E14 embryonic brain. Wogonin also promoted differentiation of NPCs into mature neurons in vivo. When transplanted into the adult rat hippocampus, NPCs differentiated into cells expressing NeuN, the mature neuron marker, by 4weeks after transplantation. These data indicate that wogonin induces differentiation of NPCs both in culture and in vivo, and suggest that facilitation of NPC differentiation is a biological activity by which wogonin protects neurons in damaged brain.


Asunto(s)
Flavanonas/farmacología , Células-Madre Neurales/efectos de los fármacos , Neuritas/efectos de los fármacos , Neurogénesis , Animales , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Hipocampo/citología , Hipocampo/efectos de los fármacos , Masculino , Células-Madre Neurales/fisiología , Células-Madre Neurales/trasplante , Neuritas/fisiología , Ratas , Ratas Endogámicas , Trasplante de Células Madre
10.
Biol Pharm Bull ; 33(6): 1054-9, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20522977

RESUMEN

Although donepezil, a potent acetylcholinesterase (AChE) inhibitor, has been used to treat Alzheimer's disease (AD) due to its neuroprotective effects, its mode of action to inhibit the growth of cancer cells is poorly understood. In the present study, we investigated the pro-apoptotic activities of donepezil in HL-60 human promyelocytic leukemia cells and the underlying molecular mechanism involved. It was found that donepezil induced the apoptosis of HL-60 and U937 cells in a dose- and time-dependent manner, as evidenced by the formation of DNA fragmentation and the accumulation of positive cells for Annexin V. In addition, the activations of caspase-8, -9, and -3 were significantly increased 36 h after donepezil treatment. Furthermore, the broad caspase inhibitor (z-VAD-fmk) blocked donepezil-induced apoptosis. In addition, donepezil was found to cause the loss of mitochondrial membrane potential (DeltaPsi(m)), to increase the release of cytochrome c to the cytosol, and to alter the expressions of Bcl-2 family proteins. Taken together, these results demonstrate for the first time that donepezil displayed an induction of apoptosis in HL-60 cells via a mitochondria-mediated caspase-dependent pathway.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Inhibidores de la Colinesterasa/farmacología , Indanos/farmacología , Leucemia Promielocítica Aguda/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Piperidinas/farmacología , Anexina A5/metabolismo , Citocromos c/metabolismo , Citosol , Fragmentación del ADN , Donepezilo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Células HL-60 , Células HeLa , Humanos , Leucemia Promielocítica Aguda/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Células U937
11.
Mol Biol Cell ; 18(10): 4013-23, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17652455

RESUMEN

Chromosomes are compartmentalized into discrete chromosome territories during interphase in mammalian cells. A chromosome territory is generated by the tendency of chromatin to occupy the smallest shell volume, which is determined by the polymeric properties and interactions of the internal meshwork of the chromatin fiber. Here, we show that BAF53 knockdown by small interfering RNA interference led to the expansion of chromosome territories. This was accompanied by a reduction in chromatin compaction, an increase in the micrococcal nuclease sensitivity of the chromatin, and an alteration in H3-K9 and H3-K79 dimethylation. Interestingly, the BAF53 knockdown cells suffer a cell cycle defect. Despite the significant irregularity and decompaction of the polynucleosomes isolated from the BAF53 knockdown cells, the chromatin loading of H1 and core histones remained unaltered, as did the nucleosome spacing. The histone hyperacetylation and down-regulation of BRG-1, mBrm, and Tip49, the catalytic components of the SWI/SNF complex and the TIP60 complex, respectively, did not expand chromosome territories. These results indicate that BAF53 contributes to the polymeric properties and/or the internal meshwork interactions of the chromatin fiber probably via a novel mechanism.


Asunto(s)
Actinas/deficiencia , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/deficiencia , Cromosomas de los Mamíferos/metabolismo , Proteínas de Unión al ADN/deficiencia , Interfase , Acetilación , Animales , Proteínas de Ciclo Celular/genética , ADN Helicasas/genética , Regulación hacia Abajo/genética , Histonas/metabolismo , Lisina/metabolismo , Metilación , Ratones , Células 3T3 NIH , Proteínas Nucleares/genética , Nucleosomas/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos/genética , Factores de Transcripción/genética , Regulación hacia Arriba/genética
12.
Neurosci Lett ; 450(1): 45-50, 2009 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-19010391

RESUMEN

The p62 protein has been identified as a major component of the protein aggregations associated with neurodegenerative disease. Oxidative insult has also been identified as a principal cause of neurodegenerative disease. Thus, in the present study, we investigated the potential role of p62 in oxidative stress-induced cell death in SH-SY5Y human neuroblastoma cells. The results indicated that H(2)O(2) treatment induced p62 expression in SH-SY5Y cells. In addition, p62 showed neuroprotective effects against H(2)O(2)-induced cell death in differentiated SH-SY5Y cells. p62 expression prolonged Akt phosphorylation during the later stages of H(2)O(2)-induced cell death. Furthermore, coexpression of p62 and wild-type PDK1, the upstream kinase of Akt, further increased Akt phosphorylation and cell viability, whereas the expression of kinase-defective PDK1 reversed the cytoprotective effects of p62 under oxidative stress. Overexpression of p62 led to the dissociation of PDK1 from the 14-3-3theta protein, which is thought to be a negative regulator of PDK1 kinase activity. These findings suggest a mechanism that involves the p62-mediated modulation of the interaction between signaling molecules and results in cell survival.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Peróxido de Hidrógeno/toxicidad , Neuronas/fisiología , Estrés Oxidativo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/fisiología , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Expresión Génica/efectos de los fármacos , Humanos , Morfolinas/farmacología , Neuronas/efectos de los fármacos , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Proteína Quinasa C/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Proteína Sequestosoma-1 , Transducción de Señal , Transfección
13.
Schizophr Res ; 103(1-3): 201-8, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18541413

RESUMEN

PDGFRB is located on chromosome 5q31-q32, a chromosomal region identified by linkage analyses to contain a susceptibility gene for schizophrenia (SCZ). Recent research has focused on the role of the N-methyl-d-aspartate (NMDA) receptor in the pathogenesis of SCZ. D4 dopamine receptor-mediated transactivation of the gene encoding platelet-derived growth factor receptor beta (PDGFRB) has immediate effects on synaptic neurotransmission via calcium-dependent inactivation of NMDA receptors. In this study, we investigate the association between the PDGFRB gene and SCZ in a Korean population. We screened 6 single-nucleotide polymorphisms (SNPs) in the 5'-upstream region of PDGFRB and conducted a case-control study of 381 SCZ patients and 752 controls. The genotype and haplotype frequencies of 3 of the 6 SNPs [SNP1 (g.-1924T>C, rs3756314), SNP3 (g.-1772A>G, rs3756312) and SNP4 (rs3756311, g.-1658G>A)] were significantly associated with SCZ [SNP1, corrected p=0.012 (co-dominant model), 0.002 (Dominant model), and 0.506 (Recessive model); SNP3 and 4, corrected p=0.003, 0.009, and 0.049]. Haplotype analysis also revealed that ht1 (CGG) and ht2 (TAA) were significantly associated with SCZ (ht1, corrected p=0.018, 0.340, and 0.010; ht2, corrected p=0.002, 0.009, and 0.016). Transient transfection in neuronal cells revealed that ht1 had higher luciferase activity than the vector alone. Furthermore, Pdgfrb expression was increased in the frontal cortex and hippocampus in a mouse model of SCZ induced by MK801. We conclude that SNPs of the 5'-upstream region of PDGFRB are associated with SCZ in a Korean population. These are weak positives that require future studies to confirm these results.


Asunto(s)
Polimorfismo de Nucleótido Simple/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Esquizofrenia/genética , Adulto , Animales , Estudios de Casos y Controles , Cromosomas Humanos Par 5/genética , Modelos Animales de Enfermedad , Femenino , Lóbulo Frontal/patología , Frecuencia de los Genes/genética , Predisposición Genética a la Enfermedad/genética , Genotipo , Haplotipos , Hipocampo/patología , Humanos , Corea (Geográfico) , Masculino , Ratones , Ratones Endogámicos ICR , Persona de Mediana Edad
14.
Neurosci Lett ; 430(1): 60-3, 2008 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-17997036

RESUMEN

To further clarify schizophrenia (SCZ), disrupted in schizophrenia 1 (DISC1) is a promising candidate gene expressed predominantly within the hippocampus. Several lines of evidence suggest that DISC1 may be involved in susceptibility to SCZ. In this study, we investigated whether genetic polymorphisms in the coding region of DISC1 were associated with several SCZ clinical phenotypes in a Korean population. To examine any association between DISC1 and SCZ, we genotyped three clinical single nucleotide polymorphisms (SNPs) (rs3738401, R264Q; rs3738402, L465L; rs821616, S704C) in the coding region of the DISC1 gene using the Illumina Sentrix Array Matrix chip and direct sequencing in 303 patients with SCZ and 300 healthy controls. Our case-control analysis showed that none of these SNPs was associated with SCZ. In further endophenotype stratification, however, we found a significant association between rs821616 and the poor concentration subgroup of SCZ, determined using the Operational Criteria Checklist (codominant model, p=0.015). Our results suggest that DISC1 may be a susceptibility gene for poor concentration among Korean patients with SCZ.


Asunto(s)
Predisposición Genética a la Enfermedad , Proteínas del Tejido Nervioso/genética , Polimorfismo de Nucleótido Simple , Esquizofrenia/genética , Adulto , Pueblo Asiatico/genética , Estudios de Casos y Controles , Femenino , Humanos , Corea (Geográfico) , Masculino , Reacción en Cadena de la Polimerasa
15.
Phytother Res ; 22(10): 1324-9, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18693285

RESUMEN

Neurogenesis persists in the adult mammalian brain and can be a target for modulation for therapeutic purposes. This study investigated the effect of a Polygala tenuifolia root extract on the proliferation of a stem cell population in the rat hippocampus. The root extract of P. tenuifolia (2 mg/kg/day, 14 times intraperitoneal injections) increased the incorporation of bromodeoxyuridine (BrdU) into cells in the hippocampal CA1 region. This activity was enriched in the saponin-containing fraction. The majority of cells labelled with BrdU were immunoreactive to nestin or Tuj1 and the percentages of nestin/BrdU- and Tuj1/BrdU-double positive cells were increased by the P. tenuifolia root extract, suggesting that the P. tenuifolia root extract promotes the proliferation of neural stem cells. In addition, this extract promoted the neurite outgrowth of rat neuronal precursor cells, HiB5. These activities of P. tenuifolia root extract may contribute to the therapeutic benefits of herbal medicines containing P. tenuifolia root for the treatment of patients with insomnia, neurosis and dementia.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Hipocampo/efectos de los fármacos , Sistema Nervioso/efectos de los fármacos , Extractos Vegetales/farmacología , Raíces de Plantas/química , Polygala/química , Células Madre/efectos de los fármacos , Animales , Línea Celular , Hipocampo/citología , Inmunohistoquímica , Masculino , Sistema Nervioso/citología , Ratas , Ratas Sprague-Dawley
16.
Neuroreport ; 18(12): 1225-9, 2007 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-17632272

RESUMEN

Amyloid precursor protein generates the secreted amyloid precursor protein alpha, which protects hippocampal neurons from ischemic injury and facilitates neuronal survival and synaptogenesis in the developing nervous system. Here, we examined whether platelet-derived growth factor regulates the generation of secreted amyloid precursor protein alpha during the neuronal differentiation of hippocampal precursor cells, HiB5. We showed that platelet-derived growth factor promoted amyloid precursor protein production and secreted amyloid precursor protein alpha secretion. These effects of platelet-derived growth factor were diminished by the PI3K-specific inhibitor wortmannin and the protein kinase C-specific inhibitor GF109203X, suggesting the involvement of the PI3K and protein kinase C-signaling pathway. Furthermore, the conditioned media enriched with secreted amyloid precursor protein alpha promoted the survival of HiB5 cells during neuronal differentiation. These results suggest that the neurotrophic effect of platelet-derived growth factor is mediated in part via upregulation of the expression and release of secreted amyloid precursor protein alpha.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Diferenciación Celular/fisiología , Hipocampo/embriología , Neuronas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Madre/metabolismo , Precursor de Proteína beta-Amiloide/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Medios de Cultivo Condicionados/farmacología , Inhibidores Enzimáticos/farmacología , Hipocampo/citología , Hipocampo/efectos de los fármacos , Humanos , Neuronas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Madre/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
17.
Arch Pharm Res ; 30(6): 750-4, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17679554

RESUMEN

HiB5 is a multipotent hippocampal stem cell line whose differentiation into cells of a neuronal phenotype is promoted by neurotrophic factors such as PDGF and brain-derived neurotrophic factor (BDNF). We examined the potential role of Src homology 2 (SH2)-containing protein tyrosine phosphatase (Shp2) in this differentiation process. We found that Shp2 became tyrosine phosphorylated following PDGF treatment. Wild-type Shp2 enhanced the expression of neurofilament, synapsin I and PSD95 by PDGF and BDNF, whereas their expression was attenuated by the catalytically inactive mutants Shp2C/S and Shp2DeltaP. Formation of dendritic spine-like structures increased with wild-type Shp2, but diminished with Shp2C/S and Shp2DeltaP. PSD95, localized in the post-synaptic density region of dendritic spines, PDGFRbeta and TrkB were co-immunoprecipitated with Shp2 antibodies. These results suggest that Shp2 plays a positive role in mediating PDGF- and BDNF-activated signaling which promotes the formation of dendritic spines.


Asunto(s)
Diferenciación Celular/fisiología , Hipocampo/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Neuronas/fisiología , Proteínas Tirosina Fosfatasas/fisiología , Animales , Western Blotting , Factor Neurotrófico Derivado del Encéfalo/farmacología , Catálisis , Espinas Dendríticas/fisiología , Homólogo 4 de la Proteína Discs Large , Inmunohistoquímica , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/fisiología , Mutación/fisiología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Tirosina Fosfatasas/genética , Ratas , Ratas Sprague-Dawley , Células Madre/fisiología , Sinapsinas/fisiología , Transfección
18.
J Microbiol Biotechnol ; 17(12): 2033-41, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18167452

RESUMEN

For many years, it has been demonstrated that neurotrophins regulate the adult nervous system, implicating their potential as therapeutic agents for the treatment of neurodegenerative diseases. We generated adenoviral vectors encoding brain-derived neutotrophin factor (BDNF) and neurotrophin-3 (NT3) and tested either separately or together for the ability to induce differentiation of neuronal precursor cells with two different origins. Separate transduction of adenovirus delivering BDNF (BDNF-Ad) or NT3 (NT3-Ad) induced the neuronal differentiation in hippocampal and cortical precursor cells. NT3-Ad infected cells extended short neurites, whereas BDNFAd infected cells had longer neurites. In the early differentiation of hippocampal precursor cells, simultaneous infection of BDNFAd and NT3-Ad promoted further differentiation and neurite elongation compared with the separate infection of each virus. In contrast, simultaneous infection did not show the synergistic effect in the cortical precursor cells, suggesting that the neurotrophins play distinct roles in different regions of the brain. However, the numbers of neurites and spines per differentiated cells were markedly increased in cortical as well as hippocampal precursor cells, indicating the promotion of efficient neurite elongation and formation of dendritic spine, when BDNF-Ad and NT3-Ad were co-infected. These results suggest more studies in the effect of a combinatorial use of neurotrophins on different sites of brain need to be carried out to develop gene therapy protocols for neurodegenerative diseases.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/fisiología , Corteza Cerebral/fisiología , Hipocampo/fisiología , Neuronas/fisiología , Neurotrofina 3/fisiología , Adenoviridae/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/genética , Diferenciación Celular/fisiología , Línea Celular , Corteza Cerebral/citología , Vectores Genéticos/genética , Hipocampo/citología , Humanos , Immunoblotting , Inmunohistoquímica , Neuronas/citología , Neurotrofina 3/biosíntesis , Neurotrofina 3/genética , ARN/química , ARN/genética , Ratas , Ratas Sprague-Dawley , Receptor trkB/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología , Células Madre/fisiología
19.
FASEB J ; 16(10): 1310-2, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12154005

RESUMEN

Junctin is a 26-kDa integral membrane protein, colocalized with the ryanodine receptor (RyR) and calsequestrin at the junctional sarcoplasmic reticulum (SR) membrane in cardiac and skeletal muscles. To elucidate the functional role of junctin in heart, transgenic (TG) mice overexpressing canine junctin (24-29 folds) under the control of mouse a-myosin heavy chain promoter were generated. Overexpression of the junctin in mouse heart was associated with heart enlargements, bradycardia, atrial fibrillation, and increased fibrosis. Many ultrastructural alterations were observed in TG atria. The junctional SR cisternae facing transverse-tubules contained a dense matrix of calsequestrin in TG heart. According to echocardiography, TG mice showed enlarged left ventricles, dilated right atriums, and ventricles with paradoxical septal motion and impaired left ventricular systolic function. Overexpression of junctin led to down-regulation of triadin and RyR but to up-regulation of dihydropyridine receptor. The L-type Ca2+ current density and action potential durations increased, which could be the cause for the bradycardia in TG heart. This study provides an important example of pathogenesis leading to substantial cardiac remodeling and atrial fibrillation, which was caused by overexpression of junctin in heart.


Asunto(s)
Fibrilación Atrial/etiología , Fibrilación Atrial/patología , Proteínas de Unión al Calcio , Cardiomegalia/etiología , Proteínas Portadoras/genética , Proteínas de la Membrana , Oxigenasas de Función Mixta , Proteínas Musculares/genética , Potenciales de Acción , Animales , Fibrilación Atrial/fisiopatología , Bradicardia/etiología , Canales de Calcio Tipo L/fisiología , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Proteínas Portadoras/metabolismo , Células Cultivadas , Conductividad Eléctrica , Fibrosis , Corazón/fisiopatología , Cinética , Ratones , Ratones Transgénicos , Modelos Cardiovasculares , Contracción Muscular , Proteínas Musculares/metabolismo , Miocardio/patología , Miocardio/ultraestructura , Remodelación Ventricular
20.
Oncol Rep ; 14(6): 1475-9, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16273241

RESUMEN

To enhance the transduction efficiency (TE) of a recombinant adeno-associated virus 2 (rAAV2) in human cancer cells, we examined the combined effects of various chemicals known to influence the rAAV2 transduction process at distinct steps. Among the agents tested were trichostatin A, a histone deacetylase inhibitor, MG-132, a proteosome inhibitor, the genotoxic agents hydroxyurea, aphidicolin, etoposide and camptothecin, and tyrphostin-1, an epidermal growth factor receptor inhibitor. During or after chemical treatment, various human cancer cells were infected with rAAV2 expressing beta-galactosidase. Treatment with hydroxy-urea or etoposide plus tyrphostin-1 dramatically increased the TE in most cell lines. The combination of hydroxyurea plus tyrphostin-1 increased TE to 37.7+/-7.9%, 32.8+/-2.0% and 31.8+/-2.1% in SK-Hep1, HeLa, and HCT116 cells, respectively. In addition, following rAAV2 infection and treatment with hydroxyurea plus tyrphostin-1, long-term transgene expression was observed for up to 6 months, with no damage to the transduced cells. These results indicate that rAAV2 transgene expression can be significantly enhanced by a combination of chemical agents with distinct activity and prolonged gene expression can occur following rAAV2 gene transfer into human cancer cells.


Asunto(s)
Adenoviridae/genética , Expresión Génica/efectos de los fármacos , Hidroxiurea/farmacología , Tirfostinos/farmacología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , ADN Recombinante/genética , Citometría de Flujo , Células HeLa , Humanos , Immunoblotting , Operón Lac/genética , Neoplasias/genética , Neoplasias/patología , Factores de Tiempo , Transfección , beta-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA