Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Neurobiol ; 2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38676810

RESUMEN

It is established that neurogenesis of dentate gyrus is increased after ischemic insult, although the regulatory mechanisms have not yet been elucidated. In this study, we focused on Ezh2 which suppresses gene expression through catalyzing trimethylation of lysine 27 of histone 3. Male gerbils were injected with adeno-associated virus (AAV) carrying shRNA targeting to Ezh2 into right dentate gyrus 2 weeks prior to forebrain ischemia. One week after ischemia, animals were injected with thymidine analogue to label proliferating cells. Three weeks after ischemia, animals were killed for histological analysis. AAV-mediated knockdown of Ezh2 significantly decreased the ischemia-induced increment of proliferating cells, and the proliferated cells after ischemia showed significantly longer migration from subgranular zone (SGZ), compared to the control group. Furthermore, the number of neural stem cells in SGZ significantly decreased after ischemia with Ezh2 knockdown group. Of note, Ezh2 knockdown did not affect the number of proliferating cells or the migration from SGZ in the non-ischemic condition. Our data showed that, specifically after ischemia, Ezh2 knockdown shifted the balance between self-renewal and differentiation toward differentiation in adult dentate gyrus.

2.
J Gene Med ; 25(1): e3457, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36278965

RESUMEN

BACKGROUND: The delivery of adeno-associated virus (AAV) vectors via the cerebrospinal fluid (CSF) has emerged as a valuable method for widespread transduction in the central nervous system. Although infusion into the cerebral ventricles is a common protocol in preclinical studies of small animals, the cisterna magna has been recognized as an alternative target for clinical studies because it can be reached in a less invasive manner using an intrathecal catheter via the subarachnoid space from a lumbar puncture. METHODS: We evaluated the early distribution of fluorine-18-labeled AAV9 vectors infused into the lateral ventricle or cisterna magna of four non-human primates using positron emission tomography. The expression of the green fluorescent protein was immunohistochemically determined. RESULTS: In both approaches, the labeled vectors diffused into the broad arachnoid space around the brain stem and cervical spinal cord within 30 min. Both infusion routes efficiently transduced neurons in the cervical spinal cord. CONCLUSIONS: For gene therapy that primarily targets the cervical spinal cord and brainstem, such as amyotrophic lateral sclerosis, cisterna magna infusion would be a feasible and effective administration method.


Asunto(s)
Terapia Genética , Médula Espinal , Animales , Transducción Genética , Médula Espinal/metabolismo , Terapia Genética/métodos , Primates/genética , Vectores Genéticos/genética , Dependovirus/genética
3.
Hum Gene Ther ; 33(1-2): 76-85, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34348481

RESUMEN

The safety and high efficiency of adeno-associated virus (AAV) vectors has facilitated their wide-scale use to deliver therapeutic genes for experimental and clinical purposes in diseases affecting the central nervous system (CNS). AAV1, 2, 5, 8, 9, and rh10 are the most commonly used serotypes for CNS applications. Most AAVs are known to transduce genes predominantly into neurons. However, the precise tropism of AAVs in the dentate gyrus (DG), the region where persistent neurogenesis occurs in the adult brain, is not fully understood. We stereotaxically injected 1.5 × 1010 viral genomes of AAV2, 5, or rh10 carrying green fluorescent protein (GFP) into the right side of gerbil hippocampus, and performed immunofluorescent analysis using differentiation stage-specific markers 1 week after injection. We found that AAV5 showed a significantly larger number of double-positive cells for GFP and Sox2 in the DG, compared with the AAV2 and rh10 groups. On the contrary, AAVrh10 presented a substantially larger number of double-positive cells for GFP and NeuN in the DG, compared with AAV2 and AAV5. Our findings indicated that AAV5 showed high transduction efficiency to neural stem cells and precursor cells, whereas AAVrh10 showed much higher efficiency to mature neurons in the DG.


Asunto(s)
Dependovirus , Células-Madre Neurales , Animales , Giro Dentado , Dependovirus/genética , Vectores Genéticos/genética , Gerbillinae , Proteínas Fluorescentes Verdes/genética , Neuronas , Transducción Genética
4.
J Gene Med ; 24(3): e3402, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34897885

RESUMEN

BACKGROUND: Despite the increasing availability of effective drugs, around one-third of patients with epilepsy are still resistant to pharmacotherapy. Gene therapy has been suggested as a plausible approach to achieve seizure control, in particular for patients with focal epilepsy. Because seizures develop across wide spans of the brain in many forms of epilepsy, global delivery of the vectors is necessary to tackle such generalized seizures. Neuroligin 2 (NL2) is a postsynaptic cell adhesion molecule that induces or strengthens inhibitory synaptic function by specifically combining with neurexin 1. METHODS: In the present study, we applied an adeno-associated virus (AAV) type 9 vector expressing NL2 to modulate neuronal excitability in broad areas of the brain in epileptic (EL) mice, a model of polygene epilepsy. We administered the AAV vector expressing Flag-tagged NL2 under the synapsin I promoter (AAV-NL2) via cardiac injection 6 weeks after birth. RESULTS: Significant reductions in the duration, strength and frequency of seizure were observed during a 14-week observation period in NL2-treated EL mice compared to untreated or AAV-green fluorescent protein-treated EL mice. No behavioral abnormality was observed in NL2-treated EL mice in an open-field test. Immunohistochemical examination at 14 weeks after AAV-NL2 injection revealed the expression of exogenous NL2 in broad areas of the brain, including the hippocampus and, in these areas, NL2 co-localized with postsynaptic inhibitory molecule gephyrin. CONCLUSIONS: Global brain delivery of NL2 by systemic administration of AAV vector may provide a non-invasive therapeutic approach for generalized epilepsy.


Asunto(s)
Epilepsia , Sinapsis , Animales , Encéfalo , Moléculas de Adhesión Celular Neuronal , Epilepsia/genética , Epilepsia/metabolismo , Epilepsia/terapia , Humanos , Ratones , Proteínas del Tejido Nervioso , Convulsiones/genética , Convulsiones/metabolismo , Convulsiones/terapia , Sinapsis/metabolismo
5.
Mol Ther Methods Clin Dev ; 13: 180-186, 2019 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-30788386

RESUMEN

Gene therapy has been employed as a therapeutic approach for intractable focal epilepsies. Considering the potential of focal GABAergic neuromodulation in regulating epileptogenesis, the GABA-producing enzyme, γ-aminobutyric acid decarboxylase 67 (GAD67), is highly suitable for epilepsy therapy. The EL/Suz (EL) mouse is a model of multifactorial temporal lobe epilepsy. In the present study, we examined focal gene transduction in epileptic EL mice using recombinant adeno-associated virus serotype 8 (rAAV8) expressing human GAD67 to enhance GABA-mediated neural inhibition. Eight-week-old mice were bilaterally injected with rAAV8-GFP or rAAV8-GAD67 in the hippocampal CA3 region. After four weeks, the GAD67-transduced EL mice, but not the rAAV-GFP-treated EL mice, exhibited a significant reduction in seizure generation. The GAD67-mediated depression became stable after 14 weeks. The excitability of the CA3 region was markedly reduced in the GAD67-transduced EL mice, consistent with the results of the Ca2+ imaging using hippocampal slices. In addition, downregulation of c-Fos expression was observed in GAD67-transduced hippocampi. Our findings showed that rAAV8-GAD67 induced significant changes in the GABAergic system in the EL hippocampus. Thus, rAAV8-mediated GAD67 gene transfer is a promising therapeutic strategy for the treatment of epilepsies.

6.
Eur J Neurosci ; 48(12): 3466-3476, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30251448

RESUMEN

Survivin, a member of the inhibitors of apoptosis protein gene family, inhibits the activity of caspase, leading to a halt of the apoptotic process. Our study focused on the neuroprotective effect of survivin after transient middle cerebral artery occlusion (MCAO) with intraparenchymal administration of an adeno-associated virus (AAV) vector. His-tagged survivin was cloned and packaged into the AAV-rh10 vector. Four-week-old Sprague-Dawley rats were injected with 4 × 109  vg of AAV-GFP or AAV-His-survivin into the right striatum and were treated 3 weeks later with transient MCAO for 90 min. Twenty-four hours after MCAO, functional and histological analyses of the rats were performed. The result showed that rats that had been treated with AAV-green fluorescent protein (GFP) and those that had been treated with AAV-His-survivin did not show a significant difference in neurological scores 24 hr after MCAO, however, infarction volume was significantly reduced in the AAV-His-survivin group compared to that in the AAV-GFP group. Although the neutrophil marker myeloperoxidase did not show a significant difference in the ischemic boundary zone, cells positive for active caspase-3 and terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling were significantly decreased in the AAV-His-survivin group. In conclusion, survivin overexpression in the ischemic boundary zone induced by using an AAV vector has the potential for amelioration of ischemic damage via an antiapoptotic mechanism.


Asunto(s)
Isquemia Encefálica/virología , Infarto de la Arteria Cerebral Media/virología , Fármacos Neuroprotectores/farmacología , Survivin/metabolismo , Animales , Apoptosis/efectos de los fármacos , Isquemia Encefálica/metabolismo , Dependovirus/genética , Modelos Animales de Enfermedad , Etiquetado Corte-Fin in Situ/métodos , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Ratas Sprague-Dawley , Survivin/genética
7.
Neurosci Lett ; 682: 27-31, 2018 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-29885449

RESUMEN

Adeno-associated virus (AAV) is an ideal vector for gene transduction into the central nervous system because of its safety and efficiency. While it is currently widely used for clinical trials and is expected to become more widespread, the appropriate combination of viral serotypes and promoters have not been fully investigated. In this study, we compared the transduced gene expression of AAVrh10 to AAV5 in gerbil hippocampus using three different promoters, including cytomegalovirus (CMV), chicken ß-actin promoter with the CMV immediate-early enhancer (CAG), and the Synapsin 1 (Syn1) promoter. Four-week-old male gerbils underwent stereotaxic injection with 1 × 1010 viral genome of AAV carrying green fluorescent protein (GFP). Quantification of the GFP-positive areas 3 weeks after injection showed that AAVrh10-CMV and AAVrh10-CAG were the most efficient (p < 0.001, compared with the control) and AAVrh10-Syn1 and AAV5-CMV were the next most efficient (p < 0.05, compared with the control). On the other hand, AAV5-Syn1 showed little expression, which was only observed at the injected site. In conclusion, we should note that some combinations of viral capsids and promoters can result in failure of gene delivery, while most of them will work appropriately in the transgene expression in the brain.


Asunto(s)
Adenoviridae , Cápside , Vectores Genéticos/administración & dosificación , Hipocampo/química , Hipocampo/efectos de los fármacos , Regiones Promotoras Genéticas , Adenoviridae/genética , Animales , Pollos , Vectores Genéticos/genética , Gerbillinae , Masculino , Regiones Promotoras Genéticas/genética , Técnicas Estereotáxicas
8.
J Gene Med ; 20(4): e3013, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29624790

RESUMEN

BACKGROUND: We generated an adeno-associated virus (AAV) vector in which the human SLC2A1 gene, encoding glucose transporter type 1 (GLUT1), was expressed under the human endogenous GLUT1 promoter (AAV-GLUT1). We examined whether AAV-GLUT1 administration could lead to functional improvement in GLUT1-deficient mice. METHODS: We extrapolated human endogenous GLUT1 promoter sequences from rat minimal Glut1 promoter sequences. We generated a tyrosine-mutant AAV9/3 vector in which human SLC2A1-myc-DDK was expressed under the human GLUT1 promoter (AAV-GLUT1). AAV-GLUT1 was administered to GLUT1-deficient mice (GLUT1+/- mice) via intracerebroventricular injection (1.85 × 1010 vg/mouse or 6.5 × 1010 vg/mouse). We analyzed exogenous GLUT1 mRNA and protein expression in the brain and other major organs. We also examined improvements of cerebral microvasculature, motor function using rota-rod and footprint tests, as well as blood and cerebrospinal fluid (CSF) glucose levels. Additionally, we confirmed exogenous GLUT1 protein distribution in the brain and other organs after intracardiac injection (7.8 × 1011 vg/mouse). RESULTS: Exogenous GLUT1 protein was strongly expressed in the cerebral cortex, hippocampus and thalamus. It was mainly expressed in endothelial cells, and partially expressed in neural cells and oligodendrocytes. Motor function and CSF glucose levels were significantly improved following intracerebroventricular injection. Exogenous GLUT1 expression was not detected in other organs after intracerebroventricular injection of AAV-GLUT1, whereas it was detected in the liver and muscle tissue after intracardiac injection. CONCLUSIONS: Exogenous GLUT1 expression after AAV-GLUT1 injection approximated that of physiological human GLUT1 expression. Local central nervous system administration of AAV-GLUT1 improved CSF glucose levels and motor function of GLUT1-deficient mice and minimized off-target effects.


Asunto(s)
Dependovirus/genética , Terapia Genética , Transportador de Glucosa de Tipo 1/genética , Animales , Encéfalo/metabolismo , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Glucosa/líquido cefalorraquídeo , Transportador de Glucosa de Tipo 1/líquido cefalorraquídeo , Humanos , Hígado/metabolismo , Ratones , Regiones Promotoras Genéticas , Ratas , Transgenes
9.
Mol Genet Metab Rep ; 10: 67-74, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28119822

RESUMEN

OBJECTIVE: We generated an adeno-associated virus (AAV) vector in which the human SLC2A1 gene was expressed under the synapsin I promoter (AAV-hSLC2A1) and examined if AAV-hSLC2A1 administration can lead to functional improvement in GLUT1-deficient mice. METHODS: AAV-hSLC2A1 was injected into heterozygous knock-out murine Glut1 (GLUT1+/-) mice intraperitoneally (systemic; 1.85 × 1011 vg/mouse) or intra-cerebroventricularly (local; 1.85 × 1010 vg/mouse). We analyzed GLUT1 mRNA and protein expression, motor function using rota-rod and footprint tests, and blood and cerebrospinal fluid (CSF) glucose levels. RESULTS: Vector-derived RNA was detected in the cerebrum for both injection routes. In the intra-cerebroventricular injection group, exogenous GLUT1 protein was strongly expressed in the cerebral cortex and hippocampus near the injection site. In the intraperitoneal injection group, exogenous GLUT1 protein was mildly expressed in neural cells throughout the entire central nervous system. The motor function test and CSF/blood glucose ratio were significantly improved following intra-cerebroventricular injection. CONCLUSIONS: AAV-hSLC2A1 administration produced exogenous GLUT1 in neural cells and improved CSF glucose levels and motor function of heterozygous knock-out murine Glut1 mice.

10.
Brain Res ; 1592: 113-23, 2014 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-25451096

RESUMEN

The findings that antidepressive treatments increase hippocampal neurotrophins have led researchers to emphasize the importance of neurogenesis, formation of new dendrites, and survival of neurons in the brain. However, it is difficult to maintain neural plasticity just by enriching the environment to facilitate formation of new networks. Neural plasticity also requires a degradation process that clears off unnecessary and undesirable components. We have recently reported an increase in autophagy signaling (wherein the cell digests components of itself) that has the potential of enhancing neuronal and synaptic plasticity after multiple sessions of electroconvulsive seizure treatment. The present study revealed an increase in autophagy signaling in the rat hippocampus following 2 weeks of environmental enrichment (EE), a procedure known to elicit antidepressive and anxiolytic behavioral changes in various animal paradigms. Western blot analysis showed an increase in hippocampal expression of microtubule-associated protein light chain 3-II (LC3-II), which is lipidated from LC3-I, in rats in the EE group. The effectiveness of the 2-week EE housing condition was validated by anxiolytic effects observed in the elevated plus maze test, enhanced habituation in the open field test, and elevation of hippocampal brain-derived neurotrophic factor expression. In addition, we showed that the EE housing condition ameliorated numbing/avoidance behaviors, but not hypervigilant behaviors, in an animal model of post-traumatic stress disorder (PTSD). This is the first report to show that EE can increase autophagy signaling and improve numbing/avoidance behaviors in an animal model of PTSD.


Asunto(s)
Autofagia/fisiología , Ambiente , Hipocampo/fisiopatología , Vivienda para Animales , Trastornos por Estrés Postraumático/fisiopatología , Trastornos por Estrés Postraumático/terapia , Animales , Ansiedad/fisiopatología , Ansiedad/terapia , Western Blotting , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Electrochoque , Reacción de Fuga/fisiología , Conducta Exploratoria/fisiología , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Actividad Motora/fisiología , Ratas Wistar
11.
Artículo en Inglés | MEDLINE | ID: mdl-24316174

RESUMEN

The putative antidepressive mechanisms of a series of electroconvulsive seizures (ECS) are the following: 1) downregulation of monoaminergic receptor expression in several brain regions, 2) upregulation of the expression of brain-derived neurotrophic factor (BDNF), and 3) increased neurogenesis in the hippocampus. In this study, we used Western blot techniques to present another mechanism in which ECS enhances the autophagy signaling that is involved in the machinery related to synaptic and neural plasticity. Antibodies for conjugated Atg5-Atg12 (58kD) and cleaved light chain protein 3-II (LC3-II; 14 kD) were used to detect autophagy signals. An antibody for cleaved caspase-3 (17 kD) was used to detect alterations in apoptotic signals. Mature BDNF (14kD) expression in the hippocampus was evaluated in order to qualify the effectiveness of the ECS or stress-loading treatment. While significantly increased autophagy signals and no increases in apoptotic signals were detected in the ECS-treated rat hippocampus, the reverse (increased apoptotic signals and no altered autophagy signals) was observed in stressed rat hippocampus. No neuronal cell loss but new mossy fiber sprouting has been reported to accompany multiple ECS treatments, and recent studies have revealed that autophagy processes regulate the number of specific neurotransmitter receptors and the plasticity of synaptic components. The present study illustrated the neuroplastic and neurotrophic profiles of ECS and the neurotoxic impact of severe stress loading on hippocampal regions. This is the first report to demonstrate increased autophagy signals in ECS-treated rat hippocampus and no alterations in autophagy signals in stress-loaded rat hippocampus.


Asunto(s)
Autofagia/fisiología , Electrochoque , Hipocampo/metabolismo , Transducción de Señal/fisiología , Animales , Apoptosis/fisiología , Proteína 5 Relacionada con la Autofagia , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Caspasa 3/metabolismo , Hipocampo/fisiopatología , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Plasticidad Neuronal/fisiología , Proteínas/metabolismo , Ratas , Estrés Psicológico/metabolismo
12.
EMBO Mol Med ; 5(11): 1710-9, 2013 11.
Artículo en Inglés | MEDLINE | ID: mdl-24115583

RESUMEN

Amyotrophic lateral sclerosis (ALS) is the most common adult-onset motor neuron disease, and the lack of effective therapy results in inevitable death within a few years of onset. Failure of GluA2 RNA editing resulting from downregulation of the RNA-editing enzyme adenosine deaminase acting on RNA 2 (ADAR2) occurs in the majority of ALS cases and causes the death of motor neurons via a Ca(2+) -permeable AMPA receptor-mediated mechanism. Here, we explored the possibility of gene therapy for ALS by upregulating ADAR2 in mouse motor neurons using an adeno-associated virus serotype 9 (AAV9) vector that provides gene delivery to a wide array of central neurons after peripheral administration. A single intravenous injection of AAV9-ADAR2 in conditional ADAR2 knockout mice (AR2), which comprise a mechanistic mouse model of sporadic ALS, caused expression of exogenous ADAR2 in the central neurons and effectively prevented progressive motor dysfunction. Notably, AAV9-ADAR2 rescued the motor neurons of AR2 mice from death by normalizing TDP-43 expression. This AAV9-mediated ADAR2 gene delivery may therefore enable the development of a gene therapy for ALS.


Asunto(s)
Adenosina Desaminasa/genética , Esclerosis Amiotrófica Lateral/enzimología , Terapia Genética , Neuronas Motoras/enzimología , Proteínas de Unión al ARN/genética , Adenosina Desaminasa/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/terapia , Animales , Encéfalo/citología , Encéfalo/enzimología , Encéfalo/virología , Dependovirus/genética , Dependovirus/fisiología , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas de Unión al ARN/metabolismo
13.
Biomed Res Int ; 2013: 974819, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23762870

RESUMEN

Recombinant adeno-associated virus (AAV) vectors are powerful tools for both basic neuroscience experiments and clinical gene therapies for neurological diseases. Intravascularly administered self-complementary AAV9 vectors can cross the blood-brain barrier. However, AAV9 vectors are of limited usefulness because they mainly transduce astrocytes in adult animal brains and have restrictions on foreign DNA package sizes. In this study, we show that intracardiac injections of tyrosine-mutant pseudotype AAV9/3 vectors resulted in extensive and widespread transgene expression in the brains and spinal cords of adult mice. Furthermore, the usage of neuron-specific promoters achieved selective transduction of neurons. These results suggest that tyrosine-mutant AAV9/3 vectors may be effective vehicles for delivery of therapeutic genes, including miRNAs, into the brain and for treating diseases that affect broad areas of the central nervous system.


Asunto(s)
Envejecimiento/metabolismo , Dependovirus/genética , Vectores Genéticos/genética , Mutación/genética , Neuronas/metabolismo , Transducción Genética , Tirosina/genética , Animales , Encéfalo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Regiones Promotoras Genéticas/genética , Células de Purkinje/citología , Células de Purkinje/metabolismo , Médula Espinal/citología , Sustancia Negra/citología , Sustancia Negra/metabolismo
14.
Life Sci ; 89(1-2): 50-6, 2011 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-21620871

RESUMEN

AIMS: Transplantation of bone marrow stromal cells (MSCs) has been shown to ameliorate ischemic brain injury in animals. In the present study, we investigated whether the transplantation of MSCs combined with FK506, a clinically used immunosuppressant, enhanced neuroprotective effects in rat experimental stroke. MAIN METHODS: Male Sprague-Dawley rats underwent transient 90 min middle cerebral artery occlusion (MCAO). Two or 6h after ischemia onset, the rats were randomly assigned to receive intravenous administration of MSCs plus FK506, MSCs alone, FK506 alone, or vehicle. Infarct volume, and neurological and immunohistological assessments were performed to examine the effects of these therapies. KEY FINDINGS: In 2-hour post-ischemia treatment groups, significant improvement of infarct volume and neurological scores were observed 1 day after combination therapy compared with monotherapy, and this neuroprotection continued for 7 days. Combination therapy significantly reduced the number of TUNEL-positive apoptotic cells, increased Bcl-2 expression, decreased Bax expression, and suppressed neutrophil infiltration and microglia/macrophage activation compared to monotherapy. In 6-hour post-ischemia treatment groups, a significant reduction of infarct volume, edema index, and neurological score was observed only in the combination therapy group. Moreover, the number of engrafted MSCs on day 7 with combination therapy was significantly higher than with MSCs alone. SIGNIFICANCE: Combination therapy using FK506 enhanced the anti-apoptotic and anti-inflammatory effects of MSCs and increased the survival of transplanted cells, leading to expansion of the therapeutic time window for MSCs.


Asunto(s)
Trasplante de Médula Ósea/métodos , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/cirugía , Terapia Combinada/métodos , Tacrolimus/uso terapéutico , Análisis de Varianza , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Masculino , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Células del Estroma/trasplante , Tacrolimus/farmacología , Resultado del Tratamiento , Proteína X Asociada a bcl-2/metabolismo
15.
Behav Brain Res ; 221(1): 172-81, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21377499

RESUMEN

There has been a long-standing need to develop efficient and standardized behavioral test methods for evaluating higher-order brain functions in mice. Here, we developed and validated a behavioral flexibility test in mice using IntelliCage, a fully automated behavioral analysis system for mice in a group-housed environment. We first developed a "behavioral sequencing task" in the IntelliCage that enables us to assess the learning ability of place discrimination and behavioral sequence for reward acquisition. In the serial reversal learning using the task, the discriminated spatial patterns of the rewarded and never-rewarded places were serially reversed, and thus, mice were accordingly expected to realign the previously acquired behavioral sequence. In general, the tested mice showed rapid acquisition of the behavioral sequencing task and behavioral flexibility in the subsequent serial reversal stages both in intra- and inter-session analyses. It was found that essentially the same results were obtained among three different laboratories, which confirm the high stability of the present test protocol in different strains of mice (C57BL/6, DBA/2, and ICR). In particular, the most trained cohort of C57BL/6 mice achieved a markedly rapid adaptation to the reversal task in the final phase of the long-term serial reversal test, which possibly indicated that the mice adapted to the "reversal rule" itself. In conclusion, the newly developed behavioral test was shown to be a valid assay of behavioral flexibility in mice, and is expected to be utilized in tests of mouse models of cognitive deficits.


Asunto(s)
Automatización de Laboratorios/métodos , Modelos Animales , Aprendizaje Inverso , Aprendizaje Seriado , Animales , Aprendizaje Discriminativo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Endogámicos ICR , Recompensa , Especificidad de la Especie
16.
Dev Biol ; 336(2): 192-200, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19818338

RESUMEN

Neural recognition molecule NB-2/contactin 5 is expressed transiently during the first postnatal week in glutamatergic neurons of the central auditory system. Here, we investigated the effect of NB-2 deficiency on the auditory brainstem in mouse. While almost all principal neurons are wrapped with the calyces of Held in the medial nucleus of the trapezoid body (MNTB) in wild type, 8% of principal neurons in NB-2 knockout (KO) mice lack the calyces of Held at postnatal day (P) 6. At P10 and P15, apoptotic principal neurons were detected in NB-2 KO mice, but not in wild type. Apoptotic cells were also increased in the ventral cochlear nucleus (VCN) of NB-2 KO mice, which contains bushy neurons projecting to the MNTB and the lateral superior olive (LSO). At the age of 1 month, the number of principal neurons in the MNTB and of glutamatergic synapses in the LSO was reduced in NB-2 KO mice. Finally, interpeak latencies for auditory brainstem response waves II-III and III-IV were significantly increased in NB-2 KO mice. Together, these findings suggest that NB-2 deficiency causes a deficit in synapse formation and then induces apoptosis in MNTB and VCN neurons, affecting auditory brainstem function.


Asunto(s)
Vías Auditivas , Moléculas de Adhesión Celular Neuronal/fisiología , Núcleo Coclear/embriología , Glutamatos/metabolismo , Núcleo Olivar/embriología , Animales , Anticuerpos Monoclonales/inmunología , Apoptosis , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/inmunología , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología
17.
Cardiovasc Res ; 83(4): 682-7, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-19477969

RESUMEN

AIMS: Hcn4, which encodes the hyperpolarization-activated, cyclic nucleotide-sensitive channel (I(h)), is a well-established marker of the cardiac sino-atrial node. We aimed to identify cis-elements in the genomic locus of the Hcn4 gene that regulate the transcription of Hcn4. METHODS AND RESULTS: We screened evolutionarily conserved non-coding sequences (CNSs) that are often involved in the regulation of gene expression. The VISTA Enhancer Browser identified 16 regions, termed CNS 1-16, within the Hcn4 locus. Using the luciferase reporter assay in primary neonatal rat cardiomyocytes, we found that CNS13 conferred a prominent enhancer activity (more than 30-fold) on the Hcn4 promoter. Subsequent mutation analysis revealed that the Hcn4 enhancer function was dependent on myocyte enhancer factor-2 (MEF2) and activator protein-1 (AP1) binding sequences located in CNS13. Electrophoretic mobility shift assay and chromatin immunoprecipitation confirmed that MEF2 and AP1 proteins bound CNS13. Furthermore, overexpression of a dominant negative MEF2 mutant inhibited the enhancer activity of CNS13, decreased Hcn4 mRNA expression and also decreased the amplitude of I(h) current in myocytes isolated from the inflow tract of embryonic heart. CONCLUSION: These results suggest that the novel enhancer CNS13 and MEF2 may play a critical role in the transcription of Hcn4 in the heart.


Asunto(s)
Factores Reguladores Miogénicos/metabolismo , Canales de Potasio/genética , Nodo Sinoatrial/metabolismo , Animales , Secuencia de Bases , Sitios de Unión/genética , Células Cultivadas , ADN/genética , ADN/metabolismo , Cartilla de ADN/genética , Elementos de Facilitación Genéticos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Factores de Transcripción MEF2 , Mutación , Miocitos Cardíacos/metabolismo , Factores Reguladores Miogénicos/genética , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Factor de Transcripción AP-1/metabolismo , Transcripción Genética
18.
J Comp Neurol ; 513(4): 349-62, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19177518

RESUMEN

NB-2 is a neuronal cell recognition molecule that is preferentially expressed in auditory pathways. Mice deficient in the NB-2 gene exhibit aberrant responses to acoustic stimuli. Here we examined the expression and localization of NB-2 in the auditory brainstem during development in the rat. NB-2 was strongly expressed in the ventral cochlear nucleus (VCN), ventral acoustic stria, lateral and medial superior olivary complex (SOC), superior paraolivary nucleus, medial nucleus of the trapezoid body (MNTB), ventrolateral lemniscus, and central nucleus of the inferior colliculus (CIC). In the VCN and CIC, NB-2 was expressed in the regions that are known to respond to high frequencies. In situ hybridization combined with immunohistochemistry suggested that NB-2 is expressed only in neurons. NB-2 was colocalized with glutamatergic elements in the neuropil and the calyces of Held but not with glycinergic or GABAergic elements. NB-2 expression in the SOC was first detected at embryonic day (E)19, reached a maximum level at postnatal day (P)7, and declined thereafter. Immunolabeling with VGLUT1 and VGLUT2, markers for mature and premature glutamatergic synapses, respectively, in combination with NB-2 immunolabeling revealed that NB-2 is expressed at glutamatergic synapses. Collectively, our findings suggest that NB-2 plays a key role in maturation of glutamatergic synapses in the brainstem during the final stages of auditory development.


Asunto(s)
Vías Auditivas/metabolismo , Tronco Encefálico/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Ácido Glutámico/metabolismo , Neuronas/metabolismo , Animales , Vías Auditivas/embriología , Vías Auditivas/crecimiento & desarrollo , Western Blotting , Tronco Encefálico/embriología , Tronco Encefálico/crecimiento & desarrollo , Contactinas , Glutamato Descarboxilasa/metabolismo , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos C57BL , Neuronas/ultraestructura , Ratas , Ratas Wistar , Sinapsis/metabolismo , Proteína 1 de Transporte Vesicular de Glutamato/metabolismo , Proteína 2 de Transporte Vesicular de Glutamato/metabolismo
19.
Stroke ; 39(12): 3411-7, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18757288

RESUMEN

BACKGROUND AND PURPOSE: We have previously shown that the sphingosine 1-phosphate (S1P)/S1P receptor-1 (S1P(1)R) axis contributes to the migration of transplanted neural progenitor cells (NPCs) toward areas of spinal cord injury. In the current study, we examined a strategy to increase endogenous NPC migration toward the injured central nervous system to modify S1PR. METHODS: S1P concentration in the ischemic brain was measured in a mouse thrombosis model of the middle cerebral artery. NPC migration in vitro was assessed by a Boyden chamber assay. Endogenous NPC migration toward the insult was evaluated after ventricular administration of the S1P(2)R antagonist JTE-013. RESULTS: The concentration of S1P in the brain was increased after ischemia and was maximal 14 days after the insult. The increase in S1P in the infarcted brain was primarily caused by accumulation of microglia at the insult. Mouse NPCs mainly expressed S1P(1)R and S1P(2)R as S1PRs, and S1P significantly induced the migration of NPCs in vitro through activation of S1P(1)R. However, an S1P(1)R agonist failed to have any synergistic effect on S1P-mediated NPC migration, whereas pharmacologic or genetic inhibition of S1P(2)R by JTE-013 or short hairpin RNA expression enhanced S1P-mediated NPC migration but did not affect proliferation and differentiation. Interestingly, administration of JTE-013 into a brain ventricle significantly enhanced endogenous NPC migration toward the area of ischemia. CONCLUSIONS: Our findings suggest that S1P is a chemoattractant for NPCs released from an infarcted area and regulation of S1P(2)R function further enhances the migration of NPCs toward a brain infarction.


Asunto(s)
Encéfalo/citología , Infarto Cerebral/terapia , Quimiotaxis/efectos de los fármacos , Células Madre Embrionarias/trasplante , Lisofosfolípidos/fisiología , Pirazoles/uso terapéutico , Piridinas/uso terapéutico , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Esfingosina/análogos & derivados , Animales , Isquemia Encefálica/complicaciones , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Infarto Cerebral/tratamiento farmacológico , Infarto Cerebral/fisiopatología , Quimiotaxis/fisiología , Evaluación Preclínica de Medicamentos , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Femenino , Inyecciones Intraventriculares , Subgrupos Linfocitarios/efectos de los fármacos , Lisofosfolípidos/agonistas , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Pirazoles/farmacología , Piridinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Receptores de Lisoesfingolípidos/fisiología , Esfingosina/agonistas , Esfingosina/fisiología , Receptores de Esfingosina-1-Fosfato
20.
Mol Ther ; 16(3): 474-80, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18180779

RESUMEN

Since standard aminoglycoside treatment progressively causes hearing disturbance with hair cell degeneration, systemic use of the drugs is limited. Adeno-associated virus (AAV)-based vectors have been of great interest because they mediate stable transgene expression in a variety of postmitotic cells with minimal toxicity. In this study, we investigated the effects of regulated AAV1-mediated glial cell line-derived neurotrophic factor (GDNF) expression in the cochlea on aminoglycoside-induced damage. AAV1-based vectors encoding GDNF or vectors encoding GDNF with an rtTA2s-S2 Tet-on regulation system were directly microinjected into the rat cochleae through the round window at 5 x 10(10) genome copies/body. Seven days after the virus injection, a dose of 333 mg/kg of kanamycin was subcutaneously given twice daily for 12 consecutive days. GDNF expression in the cochlea was confirmed and successfully modulated by the Tet-on system. Monitoring of the auditory brain stem response revealed an improvement of cochlear function after GDNF transduction over the frequencies tested. Damaged spiral ganglion cells and hair cells were significantly reduced by GDNF expression. Our results suggest that AAV1-mediated expression of GDNF using a regulated expression system in the cochlea is a promising strategy to protect the cochlea from aminoglycoside-induced damage.


Asunto(s)
Aminoglicósidos/toxicidad , Cóclea/efectos de los fármacos , Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Animales , Cóclea/metabolismo , Cóclea/patología , Dependovirus/genética , Vectores Genéticos/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/genética , Pérdida Auditiva/terapia , Inmunohistoquímica , Masculino , Plásmidos/genética , Ratas , Ratas Sprague-Dawley , Transgenes/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...