Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 116(47): 23426-23436, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31685616

RESUMEN

As a central feature of neuroinflammation, microglial dysfunction has been increasingly considered a causative factor of neurodegeneration implicating an intertwined pathology with amyloidogenic proteins. Herein, we report the smallest synthetic molecule (N,N'-diacetyl-p-phenylenediamine [DAPPD]), simply composed of a benzene ring with 2 acetamide groups at the para position, known to date as a chemical reagent that is able to promote the phagocytic aptitude of microglia and subsequently ameliorate cognitive defects. Based on our mechanistic investigations in vitro and in vivo, 1) the capability of DAPPD to restore microglial phagocytosis is responsible for diminishing the accumulation of amyloid-ß (Aß) species and significantly improving cognitive function in the brains of 2 types of Alzheimer's disease (AD) transgenic mice, and 2) the rectification of microglial function by DAPPD is a result of its ability to suppress the expression of NLRP3 inflammasome-associated proteins through its impact on the NF-κB pathway. Overall, our in vitro and in vivo investigations on efficacies and molecular-level mechanisms demonstrate the ability of DAPPD to regulate microglial function, suppress neuroinflammation, foster cerebral Aß clearance, and attenuate cognitive deficits in AD transgenic mouse models. Discovery of such antineuroinflammatory compounds signifies the potential in discovering effective therapeutic molecules against AD-associated neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antiinflamatorios/farmacología , Cognición/efectos de los fármacos , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fagocitosis/efectos de los fármacos , Fenilendiaminas/farmacología , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Antiinflamatorios/uso terapéutico , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/genética , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Microglía/fisiología , Estructura Molecular , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Fármacos Neuroprotectores/uso terapéutico , Fragmentos de Péptidos/genética , Fenilendiaminas/química , Fenilendiaminas/uso terapéutico , Presenilina-1/genética , Memoria Espacial/efectos de los fármacos
2.
Mol Ther ; 27(8): 1507-1526, 2019 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-31138511

RESUMEN

Gliosis in Niemann-Pick type C (NP-C) disease is characterized by marked changes in microglia and astrocytes. However, the gliosis onset and progression in NP-C has not been systematically studied, nor has the mechanism underlying this finding. Here, we found early gliosis in the subventricular zone (SVZ) of NP-C mice. Neural progenitor damage by Npc1 mutation suppressed vascular endothelial growth factor (VEGF) expression and further induced microglia activation followed by astrogliosis. Interestingly, excessive astrogliosis in the SVZ induced neural progenitor retention and/or migration into thalamus via astrocyte-derived VEGF, resulting in acceleration of thalamic and cortical gliosis through thalamo-cortical pathways. Transplantation of VEGF-overexpressing neural stem cells into the SVZ improved whole-brain pathology of NP-C mice. Overall, our data provide a new pathological perspective on NP-C neural pathology, revealing abnormalities in the subventricular-thalamo-cortical circuit of NP-C mouse brain and highlighting the importance of the SVZ microenvironment as a therapeutic target for NP-C disease.


Asunto(s)
Corteza Cerebral/metabolismo , Ventrículos Laterales/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Transducción de Señal , Tálamo/metabolismo , Animales , Astrocitos/metabolismo , Biomarcadores , Movimiento Celular , Modelos Animales de Enfermedad , Gliosis/etiología , Gliosis/metabolismo , Gliosis/patología , Ratones , Microglía/metabolismo , Células-Madre Neurales/metabolismo , Enfermedad de Niemann-Pick Tipo C/etiología , Enfermedad de Niemann-Pick Tipo C/patología , Enfermedad de Niemann-Pick Tipo C/terapia , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Mol Cells ; 38(9): 806-13, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26282862

RESUMEN

Gaucher disease (GD) is an autosomal recessive lysosomal storage disorder caused by mutations in the glucocerebrosidase gene (GBA), which encodes the lysosomal enzyme glucosylceramidase (GCase). Deficiency in GCase leads to characteristic visceral pathology and lethal neurological manifestations in some patients. Investigations into neurogenesis have suggested that neurodegenerative disorders, such as GD, could be overcome or at least ameliorated by the generation of new neurons. Bone marrow-derived mesenchymal stem cells (BM-MSCs) are potential candidates for use in the treatment of neurodegenerative disorders because of their ability to promote neurogenesis. Our objective was to examine the mechanism of neurogenesis by BM-MSCs in GD. We found that neural stem cells (NSCs) derived from a neuronopathic GD model exhibited decreased ability for self-renewal and neuronal differentiation. Co-culture of GBA-deficient NSCs with BM-MSCs resulted in an enhanced capacity for self-renewal, and an increased ability for differentiation into neurons or oligodendrocytes. Enhanced proliferation and neuronal differentiation of GBA-deficient NSCs was associated with elevated release of macrophage colony-stimulating factor (M-CSF) from BM-MSCs. Our findings suggest that soluble M-CSF derived from BM-MSCs can modulate GBA-deficient NSCs, resulting in their improved proliferation and neuronal differentiation.


Asunto(s)
Diferenciación Celular , Autorrenovación de las Células , Factor Estimulante de Colonias de Macrófagos/farmacología , Células-Madre Neurales/fisiología , Animales , Células Cultivadas , Técnicas de Cocultivo , Evaluación Preclínica de Medicamentos , Enfermedad de Gaucher/tratamiento farmacológico , Glucosilceramidasa , Glucosilceramidas/metabolismo , Factor Estimulante de Colonias de Macrófagos/fisiología , Células Madre Mesenquimatosas/fisiología , Ratones Transgénicos , Factores de Crecimiento Nervioso/metabolismo , Comunicación Paracrina
4.
J Vet Med Sci ; 77(6): 693-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25715651

RESUMEN

Niemann-Pick type C disease (NP-C) is a rare and ultimately fatal lysosomal storage disorder with variable neurologic symptoms. Loss of neuronal function and neuronal cell death occur in the NP-C brain, similar to the findings for other neurodegenerative diseases. Targeting of neuronal cells in the brain therefore represents a potential clinical intervention strategy to reduce the rate of disease progression and improve the quality of life. We previously reported that bone marrow stem cells show a neurogenic effect through CCL2 (also known as monocyte chemoattractant protein-1, MCP-1) secretion in the brains of NP-C mice. However, the direct effect of CCL2 on neurogenesis has not been ascertained. Here, to define neurogenic effects of CCL2 in NP-C, we applied human recombinant CCL2 to neural stem cells (NSCs) derived from NP-C mice. CCL2-treated NSCs showed significantly increased capacity for self-renewal, proliferation and neuronal differentiation. Similar results were observed in the subventricular zone of NP-C mice after CCL2 treatment. Furthermore, infusion of CCL2 into the NP-C mouse brain resulted in reduction of neuroinflammation. Taken together, our results demonstrate that CCL2 is a potential new therapeutic agent for NP-C.


Asunto(s)
Quimiocina CCL2/farmacología , Evaluación Preclínica de Medicamentos/métodos , Células-Madre Neurales/fisiología , Neurogénesis/efectos de los fármacos , Enfermedad de Niemann-Pick Tipo C/tratamiento farmacológico , Enfermedad de Niemann-Pick Tipo C/metabolismo , Análisis de Varianza , Animales , Bromodesoxiuridina , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quimiocina CCL2/metabolismo , Cartilla de ADN/genética , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Células-Madre Neurales/efectos de los fármacos , Reacción en Cadena de la Polimerasa , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
Brain Res ; 1529: 113-24, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23831521

RESUMEN

Accumulation of amyloid-ß (Aß) is thought to be a central pathology in the brain of patients with Alzheimer's disease (AD). Neprilysin (NEP), a plasma membrane glycoprotein of the neutral zinc metalloendopeptidase family, is known as a major Aß-degrading enzyme in the brain. The level of NEP is reduced in the brains of patients with AD; therefore, NEP is under intense investigation as a potential therapeutic source for degradation of deposited Aß in AD. Previous studies have utilized viral vectors expressing NEP for reduction of Aß deposition in the brain. However, viral vectors have disadvantages regarding difficulty in control of insert size, expression desired (short- or long-term), and target cell type. Here, in order to overcome these disadvantages, we produced recombinant soluble NEP from insect cells using an NEP expression vector, which was administered by intracerebral injection into AD mice, resulting in significantly reduced accumulation of Aß. In addition, AD mice treated with NEP showed improved behavioral performance on the water maze test. These data support a role of recombinant soluble NEP in improving memory impairment by regulation of Aß deposition and suggest the possibility that approaches using protein therapy might have potential for development of alternative therapies for treatment of AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Antipsicóticos/uso terapéutico , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/terapia , Neprilisina/uso terapéutico , Fragmentos de Péptidos/metabolismo , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Cultivadas , Modelos Animales de Enfermedad , Embrión de Mamíferos , Hipocampo/citología , Humanos , Etiquetado Corte-Fin in Situ , Aprendizaje por Laberinto/efectos de los fármacos , Trastornos de la Memoria/etiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Presenilina-1/genética , Proteínas Recombinantes/uso terapéutico
6.
Eur Spine J ; 19(10): 1753-60, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20464431

RESUMEN

Bone marrow stem cells (BMSCs) can be obtained from the vertebral body (VB) and iliac crest (IC) for augmenting spinal arthrodesis. However, it is still not evaluated, which of the two sites would have a better BMSCs potential on Proliferation and osteoblastic differentiation is still not evaluated. Fourteen patients (10 men and 4 women) undergoing posterolateral lumbar arthrodesis and pedicle screw instrumentation were involved. The mean age was 54.7 years (range 31-75 years). Bone marrow aspirates were obtained from the vertebral body through the bilateral pedicle and were quantified relative to matched, bilateral aspirates from the iliac crest that were obtained from the same patient and at the same time. The mononuclear cell count and concentration of BMSCs were calculated and compared. Proliferation and osteoblastic differentiation of each of the BMSCs were characterized using biochemical and molecular biology techniques. Concentration (cells/mL) of BMSCs from VB and IC were 3.73 × 10(3) and 3.19 × 10(3), respectively (P > 0.05). VB and IC exhibited similar proliferation pattern at 3, 5 and 7 days, but BMSCs from the VB exhibited an increased mineralization staining with Alizarin Red S at 14 days. BMSCs from both anatomic sites expressed comparable levels of CD29, CD34, CD44, CD90 and CD105. VB and IC displayed similar levels of expression of ALP, type I collagen and osterix, but VB expressed higher level of osteocalcin and Runx-2, especially at 14 and 21 days. Our studies show that BMSCs from VB have osteogenic differentiation potential similar to IC. Based on these findings, we suggest that BMSCs from VB would be comparable candidates for osseous graft supplementation especially in spinal fusion procedures.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Médula Ósea/métodos , Diferenciación Celular/fisiología , Proliferación Celular , Ilion/citología , Osteoblastos/citología , Columna Vertebral/citología , Trasplante de Células Madre/métodos , Adulto , Anciano , Células de la Médula Ósea/fisiología , Células Cultivadas , Femenino , Humanos , Ilion/fisiología , Ilion/cirugía , Masculino , Persona de Mediana Edad , Osteoblastos/fisiología , Columna Vertebral/fisiología , Columna Vertebral/cirugía
7.
J Vet Sci ; 5(4): 295-302, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15613812

RESUMEN

This study was aimed to investigate the changes of orexin-A (OXA) and neuropeptide Y (NPY) expression in the hypothalamus of the fasted and high-fat diet fed rats. For the experiments, the male Sprague-Dawley (SD) rats were used as the model of high-fat diet-induced obesity. The mean loss of body weight (MLBW) did not show the linear pattern during the fasting; from 24 h to 84 h of fastings, the MLBW was not significantly changed. The numbers of OXA-immunoreactive (IR) neurons were decreased at 84 h of fasting compared with those in other five fasting subgroups. The NPY immunoreactivities in the arcuate nucleus (ARC) and the suprachiasmatic nucleus (SCN) observed at 84 h of fasting were higher than that observed at 24 h of fasting. The number of OXA-IR neurons of the LHA (lateral hypothalamic area) in the high-fat (HF) diet fed group was more increased than that of the same area in the normal-fat (NF) diet fed group. The NPY immunoreactivities of the ARC and the SCN were higher in HF group than those observed in the same areas of NF group. Based on these results, it is noteworthy that the decrease of the body weight during the fast was not proportionate to the time-course, implicating a possible adaptation of the body for survival against starvation. The HF diet might activate the OXA and the NPY in the LHA to enhance food intake.


Asunto(s)
Ayuno/fisiología , Hipotálamo/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neuropéptido Y/metabolismo , Neuropéptidos/metabolismo , Adaptación Fisiológica/fisiología , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Grasas de la Dieta , Ingestión de Alimentos , Área Hipotalámica Lateral/metabolismo , Inmunohistoquímica/veterinaria , Masculino , Obesidad , Orexinas , Ratas , Ratas Sprague-Dawley/fisiología , Núcleo Supraquiasmático/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA