Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cells Dev ; 32(13-14): 365-378, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37051687

RESUMEN

Articular cartilage injury is common in various conditions, including osteoarthritis, rheumatic diseases, and trauma. Current treatments for cartilage injury fail to completely regenerate the damaged cartilage. Mesenchymal stromal cells (MSCs) have emerged as potential candidates for cartilage regeneration. However, MSCs exhibit hypertrophic differentiation, and their chondrogenic ability is reduced in an inflammatory environment. In recent years, genetic modification has been proposed for optimizing MSC-based therapies, some of which are expected to enter clinical trials. This review summarizes recent research findings and developments in genetic engineering strategies to enhance stem cell-based therapy for cartilage regeneration. We also discuss the mechanisms of biofunctions of MSCs in cartilage regeneration and outline the efficacy and safety of the different genetic modification strategies, including viral and nonviral delivery transduction. Finally, we highlight the major challenges and prospects for clinical translation of genetically modified MSCs.


Asunto(s)
Cartílago Articular , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Osteoartritis , Humanos , Diferenciación Celular/genética , Condrogénesis/genética
2.
Dev Dyn ; 251(4): 714-728, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34622503

RESUMEN

BACKGROUND: Plasticity-related genes (Prgs/PRGs) or lipid phosphate phosphatase-related proteins (LPPRs) comprise five known members, which have been linked to neuronal differentiation processes, such as neurite outgrowth, axonal branching, or dendritic spine formation. PRGs are highly brain-specific and belong to the lipid phosphate phosphatases (LPPs) superfamily, which influence lipid metabolism by dephosphorylation of bioactive lipids. PRGs, however, do not possess enzymatic activity, but modify lipid metabolism in a way that is still under investigation. RESULTS: We analyzed mRNA expression levels of all Prgs during mouse brain development, in the hippocampus, neocortex, olfactory bulbs, and cerebellum. We found different spatio-temporal expression patterns for each of the Prgs, and identified a high expression of the uncharacterized Prg4 throughout brain development. Unlike its close family members PRG3 and PRG5, PRG4 did not induce filopodial outgrowth in non-neuronal cell lines, and does not localize to the plasma membrane of filopodia. CONCLUSION: We showed PRG4 to be highly expressed in the developing and the adult brain, suggesting that it is of vital importance for normal brain function. Despite its similarities to other family members, it seems not to be involved in changes of cell morphology; instead, it is more likely to be associated with intracellular signaling.


Asunto(s)
Encéfalo , Monoéster Fosfórico Hidrolasas , Animales , Encéfalo/metabolismo , Membrana Celular/metabolismo , Hipocampo/metabolismo , Ratones , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Proteoglicanos/metabolismo , Seudópodos
3.
Int J Mol Sci ; 21(12)2020 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-32599915

RESUMEN

Niemann-Pick type C1 (NPC1) is a lysosomal storage disorder, inherited as an autosomal-recessive trait. Mutations in the Npc1 gene result in malfunction of the NPC1 protein, leading to an accumulation of unesterified cholesterol and glycosphingolipids. Beside visceral symptoms like hepatosplenomegaly, severe neurological symptoms such as ataxia occur. Here, we analyzed the sphingosine-1-phosphate (S1P)/S1P receptor (S1PR) axis in different brain regions of Npc1-/- mice and evaluated specific effects of treatment with 2-hydroxypropyl-ß-cyclodextrin (HPßCD) together with the iminosugar miglustat. Using high-performance thin-layer chromatography (HPTLC), mass spectrometry, quantitative real-time PCR (qRT-PCR) and western blot analyses, we studied lipid metabolism in an NPC1 mouse model and human skin fibroblasts. Lipid analyses showed disrupted S1P metabolism in Npc1-/- mice in all brain regions, together with distinct changes in S1pr3/S1PR3 and S1pr5/S1PR5 expression. Brains of Npc1-/- mice showed only weak treatment effects. However, side effects of the treatment were observed in Npc1+/+ mice. The S1P/S1PR axis seems to be involved in NPC1 pathology, showing only weak treatment effects in mouse brain. S1pr expression appears to be affected in human fibroblasts, induced pluripotent stem cells (iPSCs)-derived neural progenitor and neuronal differentiated cells. Nevertheless, treatment-induced side effects make examination of further treatment strategies indispensable.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Péptidos y Proteínas de Señalización Intracelular/fisiología , Lisofosfolípidos/metabolismo , Mutación , Enfermedad de Niemann-Pick Tipo C/tratamiento farmacológico , Esfingosina/análogos & derivados , 1-Desoxinojirimicina/farmacología , Adulto , Animales , Encéfalo/metabolismo , Encéfalo/patología , Inhibidores de Glicósido Hidrolasas/farmacología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Ratones , Ratones Noqueados , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/metabolismo , Enfermedad de Niemann-Pick Tipo C/patología , Esfingosina/metabolismo , Adulto Joven
4.
Cereb Cortex ; 30(3): 1465-1480, 2020 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-31599924

RESUMEN

Niemann-Pick type C1 (NPC1) disease is characterized by neurodegeneration caused by cholesterol accumulation in the late endosome/lysosome. In this study, a defective basal and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-stimulated internalization of GluR2-containing AMPA receptors in NPC1-/- cortical neurons was detected. Our results show that the amount of cholesterol and group I metabotropic glutamate receptors (mGluR1/5) in lipid rafts of NPC1-/- cortical tissue and neurons are decreased and their downstream signals of p-ERK are defective, which are restored by a rebalance of cholesterol homeostasis through ß-cyclodextrin (ß-CD) treatment. Application of 3,5-dihydroxyphenylglycine (DHPG)-a mGluR1/5 agonist-and ß-CD markedly increases the internalization of AMPA receptors and decreases over-influx of calcium in NPC1-/- neurons, respectively. Furthermore, the defective phosphorylated GluR2 and protein kinase C signals are ameliorated by the treatment with DHPG and ß-CD, respectively, suggesting an involvement of them in internalization dysfunction. Taken together, our data imply that abnormal internalization of AMPA receptors is a critical mechanism for neuronal dysfunction and the correction of dysfunctional mGluR1/5 is a potential therapeutic strategy for NPC1 disease.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Receptores AMPA/metabolismo , Receptores de Ácido Kaínico/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones Transgénicos , Neuronas/fisiología , Proteína Niemann-Pick C1
5.
Int J Mol Sci ; 20(20)2019 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-31635081

RESUMEN

Niemann-Pick Type C (NP-C) is a rare disorder of lipid metabolism caused by mutations within the NPC1 and NPC2 genes. NP-C is a neurovisceral disease leading to a heterogeneous, multisystemic spectrum of symptoms in those affected. Until now, there is no investigative tool to demonstrate the significance of single variants within the NPC genes. Hence, the aim of the study was to establish a test that allows for an objective assessment of the pathological potential of NPC1 gene variants. Chinese hamster ovary cells defective in the NPC1 gene accumulate cholesterol in lysosomal storage organelles. The cells were transfected with NPC1-GFP plasmid vectors carrying distinct sequence variants. Filipin staining was used to test for complementation of the phenotype. The known variant p.Ile1061Thr showed a significantly impaired cholesterol clearance after 12 and 24 h compared to the wild type. Among the investigated variants, p.Ser954Leu and p.Glu1273Lys showed decelerated cholesterol clearance as well. The remaining variants p.Gln60His, p.Val494Met, and p.Ile787Val showed a cholesterol clearance indistinguishable from wild type. Further, p.Ile1061Thr acquired an enhanced clearance ability upon 25-hydroxycholesterol treatment. We conclude that the variants that caused an abnormal clearance phenotype are highly likely to be of clinical relevance. Moreover, we present a system that can be utilized to screen for new drugs.


Asunto(s)
Prueba de Complementación Genética , Variación Genética , Péptidos y Proteínas de Señalización Intracelular/genética , Alelos , Secuencia de Aminoácidos , Animales , Células CHO , Células Cultivadas , Colesterol/metabolismo , Mapeo Cromosómico , Cricetulus , Frecuencia de los Genes , Estudios de Asociación Genética , Genotipo , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Moleculares , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/diagnóstico , Enfermedad de Niemann-Pick Tipo C/genética , Enfermedad de Niemann-Pick Tipo C/metabolismo , Fenotipo , Conformación Proteica , Relación Estructura-Actividad
6.
Mol Brain ; 12(1): 17, 2019 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-30866987

RESUMEN

Hypomyelination in the central nerves system (CNS) is one of the most obviously pathological features in Niemann-Pick Type C disease (NPC), which is a rare neurodegenerative disorder caused by mutations in the NPC intracellular cholesterol transporter 1 or 2 (Npc1 or Npc2). Npc1 plays key roles in both neurons and oligodendrocytes during myelination, however, the linkage between the disturbed cholesterol transport and inhibited myelination is unrevealed. In this study, mass spectrometry (MS)-based differential quantitative proteomics was applied to compare protein composition in the corpus callosum between wild type (WT) and NPC mice. In total, 3009 proteins from both samples were identified, including myelin structural proteins, neuronal proteins, and astrocyte-specific proteins. In line to hypomyelination, our data revealed downregulation of myelin structural and indispensable proteins in Npc1 mutant mice. Notably, the reduced ceramide synthase 2 (Cers2), UDP glycosyltransferase 8 (Ugt8), and glycolipid transfer protein (Gltp) indicate the altered sphingolipid metabolism in the disease and the involvement of Gltp in myelination. The identification of most reported myelin structural proteins and proteins from other cell types advocates the use of the corpus callosum to investigate proteins in different cell types that regulate myelination.


Asunto(s)
Cuerpo Calloso/metabolismo , Vaina de Mielina/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Proteómica , Animales , Ontología de Genes , Ratones Endogámicos BALB C , Ratones Mutantes , Proteínas del Tejido Nervioso/metabolismo
7.
Dev Dyn ; 248(5): 375-395, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30847983

RESUMEN

BACKGROUND: LPA is a small bioactive phospholipid that acts as an extracellular signaling molecule and is involved in cellular processes, including cell proliferation, migration, and differentiation. LPA acts by binding and activating at least six known G protein-coupled receptors: LPA1-6 . In recent years, LPA has been suggested to play an important role both in normal neuronal development and under pathological conditions in the nervous system. RESULTS: We show the expression pattern of LPA receptors during mouse brain development by using qRT-PCR, in situ hybridization, and immunocytochemistry. Only LPA 1 , LPA 2, LPA 4, and LPA 6 mRNA transcripts were detected throughout development stages from embryonic day 16 until postnatal day 30 of hippocampus, neocortex, cerebellum, and bulbus olfactorius in our experiments, while expression of LPA 3 and LPA 5 genes was below detection level. In addition to our qRT-PCR results, we also analyzed the cellular protein expression of endogenous LPA receptors, with focus on LPA1 and LPA2 within postnatal brain slices and primary neuron differentiation with and without cytoskeleton stabilization and destabilization. CONCLUSIONS: The expression of LPA receptors changes depends on the developmental stage in mouse brain and in cultured hippocampal primary neurons. Interestingly, we found that commercially available antibodies for LPA receptors are largely unspecific.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Receptores del Ácido Lisofosfatídico/metabolismo , Animales , Encéfalo/metabolismo , Células Cultivadas , Hipocampo/citología , Ratones , Neuronas/citología , ARN Mensajero/análisis , Receptores del Ácido Lisofosfatídico/genética
8.
Mol Metab ; 18: 15-24, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30348590

RESUMEN

OBJECTIVE: The liver performs a central role in regulating energy homeostasis by increasing glucose output during fasting. Recent studies on Argonaute2 (Ago2), a key RNA-binding protein mediating the microRNA pathway, have illustrated its role in adaptive mechanisms according to changes in metabolic demand. Here we sought to characterize the functional role of Ago2 in the liver in the maintenance of systemic glucose homeostasis. METHODS: We first analyzed Ago2 expression in mouse primary hepatocyte cultures after modulating extracellular glucose concentrations and in the presence of activators or inhibitors of glucokinase activity. We then characterized a conditional loss-of-function mouse model of Ago2 in liver for alterations in systemic energy metabolism. RESULTS: Here we show that Ago2 expression in liver is directly correlated to extracellular glucose concentrations and that modulating glucokinase activity is adequate to affect hepatic Ago2 levels. Conditional deletion of Ago2 in liver resulted in decreased fasting glucose levels in addition to reducing hepatic glucose production. Moreover, loss of Ago2 promoted hepatic expression of AMP-activated protein kinase α1 (AMPKα1) by de-repressing its targeting by miR-148a, an abundant microRNA in the liver. Deletion of Ago2 from hyperglycemic, obese, and insulin-resistant Lepob/ob mice reduced both random and fasted blood glucose levels and body weight and improved insulin sensitivity. CONCLUSIONS: These data illustrate a central role for Ago2 in the adaptive response of the liver to fasting. Ago2 mediates the suppression of AMPKα1 by miR-148a, thereby identifying a regulatory link between non-coding RNAs and a key stress regulator in the hepatocyte.


Asunto(s)
Proteínas Argonautas/metabolismo , Gluconeogénesis , Hígado/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Proteínas Argonautas/genética , Glucemia/metabolismo , Células Cultivadas , Ayuno/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Quinasas/metabolismo
9.
Histol Histopathol ; 33(12): 1311-1321, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29956298

RESUMEN

Niemann-Pick type C1 (NPC1) disease is a lysosomal storage disorder caused by mutation of Npc1 or Npc2 gene, resulting in various progressive pathological features. Myelin defection is a major pathological problem in Npc1 mutant mice; however, impairment of myelin proteins in the developing brain is still incompletely understood. In this study, we showed that the expression of myelin genes and proteins is strongly inhibited from postnatal day 35 onwards including reduced myelin basic protein (MBP) expression in the brain. Furthermore, myelination characterized by MBP immunohistochemistry was strongly perturbed in the forebrain, moderately in the midbrain and cerebellum, and slightly in the hindbrain. Our results demonstrate that mutation of the Npc1 gene is sufficient to cause severe and progressive defects in myelination in the mouse brain.


Asunto(s)
Encéfalo/patología , Vaina de Mielina/patología , Enfermedad de Niemann-Pick Tipo C/patología , Animales , Modelos Animales de Enfermedad , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Mutantes , Vaina de Mielina/genética , Proteína Niemann-Pick C1 , Proteínas/genética
10.
Brain Res ; 1694: 87-93, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29753706

RESUMEN

Niemann-Pick Type C1 (NPC1) disease is an autosomal recessive neurodegenerative disease characterized by an excessive accumulation of unesterified cholesterol in late endosomes/lysosomes. Patients with NPC1 disease show a series of symptoms in neuropathology, including a gradually increased loss of motor control and seizures. However, mechanism of the neurological manifestations in NPC1 disease is not fully understood yet. In this study, we utilized the micro-electrode array (MEA) to analyze the spontaneous extracellular electrical activity in cultivated cortical neurons of the NPC1 mutant (NPC1-/-) mouse. Our results show a decrease of the spontaneous electrical activity in NPC1-/- neuronal network when compared to wild type neurons, as indicated by the decreased spike rate, burst rate, event rate, and the increased burst period and event period. Application of 3,5-dihydroxyphenylglycine (DHPG), a specific agonist of group I metabotropic glutamate receptors, improved the electrical activity of the NPC1-/- neuronal network, suggesting that DHPG can be used as a potential therapeutic strategy for recovery of the electrical activity in NPC1 disease.


Asunto(s)
Metoxihidroxifenilglicol/análogos & derivados , Neuronas/efectos de los fármacos , Proteínas/efectos de los fármacos , Proteínas/genética , Animales , Proteínas Portadoras/efectos de los fármacos , Proteínas Portadoras/genética , Endosomas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Glicoproteínas de Membrana/metabolismo , Metoxihidroxifenilglicol/farmacología , Ratones Transgénicos , Neuronas/fisiología , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/tratamiento farmacológico , Proteínas/metabolismo
11.
J Neurol Sci ; 386: 56-63, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29406968

RESUMEN

Niemann-Pick Type C (NPC) disease is a rare neurovisceral disorder caused by mutations of either NPC1 or NPC2 gene and characterized by defective intracellular transport of cholesterol and glycosphingolipids, leading to neuron loss and myelin aberration in the central nervous system. In this study, by comparing protein expression in the cortical white matter tracts from mice at different postnatal days, we identified that in the NPC1 mutant (NPC1-/-) mice, the onset of myelination is delayed and the amount of the major myelin protein MBP and PLP, and oligodendrocyte regulatory factor Olig1 and Olig2, but not NG2 and Sox10, decreased significantly, suggesting a disruption of oligodendrocyte differentiation. Furthermore, in in vitro oligodendrocyte cultivation, NPC1-/- oligodendrocytes showed less response to the stimulation of neuron-conditioned medium (CdM), indicating a defect of oligodendrocyte per se. Interestingly, lovastatin restores the number of mature myelin-forming oligodendrocytes by increasing Olig1 and Olig2 expressions. Our data suggest a potential strategy for improving myelination using lovastatin in NPC disease.


Asunto(s)
Anticolesterolemiantes/uso terapéutico , Lovastatina/uso terapéutico , Mutación/genética , Vaina de Mielina/fisiología , Proteína Niemann-Pick C1/genética , Enfermedad de Niemann-Pick Tipo C , Oligodendroglía/patología , Animales , Animales Recién Nacidos , Antivirales/uso terapéutico , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Corteza Cerebral/citología , Medios de Cultivo Condicionados/farmacología , Modelos Animales de Enfermedad , Filipina/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Proteínas de la Mielina/genética , Proteínas de la Mielina/metabolismo , Proteína Niemann-Pick C1/metabolismo , Enfermedad de Niemann-Pick Tipo C/tratamiento farmacológico , Enfermedad de Niemann-Pick Tipo C/genética , Enfermedad de Niemann-Pick Tipo C/patología , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo
12.
Cell Mol Biol Lett ; 22: 16, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28828010

RESUMEN

BACKGROUND: ADAM23 is widely expressed in the embryonic central nervous system and plays an important role in tissue formation. RESULTS: In this study, we showed that ADAM23 contributes to cell survival and is involved in neuronal differentiation during the differentiation of human neural progenitor cells (hNPCs). Upregulation of ADAM23 in hNPCs was found to increase the number of neurons and the length of neurite, while its downregulation decreases them and triggers cell apoptosis. RNA microarray analysis revealed mechanistic insights into genes and pathways that may become involved in multiple cellular processes upon up- or downregulation of ADAM23. CONCLUSIONS: Our results suggest that ADAM23 regulates neuronal differentiation by triggering specific signaling pathways during hNPC differentiation.


Asunto(s)
Proteínas ADAM/fisiología , Células-Madre Neurales/metabolismo , Neurogénesis , Transducción de Señal , Proteínas ADAM/metabolismo , Humanos , Células-Madre Neurales/fisiología
13.
Biochem Biophys Res Commun ; 491(1): 40-46, 2017 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-28694190

RESUMEN

Wnt/ß-catenin and Wnt/Ca2+ pathways are involved in cellular processes during embryonic development and the interaction between them in the same cell decides the outcome of cellular functions. In this study, we showed that Wnt3a triggers the Wnt/Ca2+ signaling pathway, indicated by an increase of cytosolic free calcium ([Ca2+]i) and activation of calmodulin dependent kinase II (CaMKII) during the differentiation of human neuronal progenitor cells (hNPCs). Wnt3a via the increase of [Ca2+]i activates proline-rich tyrosine kinase 2 (Pyk2), which subsequently regulates phosphorylation of glycogen synthase kinase 3ß (GSK3ß) and ß-catenin stabilization. Our findings suggest that Pyk2 plays an important role in the coordination of stabilization of ß-catenin in the crosstalk between Wnt/ß-catenin and Wnt/Ca2+ signaling pathways upon Wnt3a stimulation in differentiating hNPCs.


Asunto(s)
Quinasa 2 de Adhesión Focal/metabolismo , Células-Madre Neurales/fisiología , Neuronas/fisiología , Vía de Señalización Wnt/fisiología , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica/fisiología , Humanos , Células-Madre Neurales/citología , Neuronas/citología , Receptor Cross-Talk/fisiología
14.
BMC Neurosci ; 15: 126, 2014 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-25472750

RESUMEN

BACKGROUND: Niemann-Pick type C1 (NPC1) disease is an inherited lysosomal storage disease caused by mutation of the Npc1 gene, resulting in a progressive accumulation of unesterified cholesterol and glycolipids in lysosomes of multiple tissues and leading to neurodegeneration and other disease. In Npc1 mutant mice, retinal degeneration including impaired visual function, lipofuscin accumulation in the pigment epithelium and ganglion cells as well as photoreceptor defects has been found. However, the pathologies of other individual cell types of the retina in Npc1 mutant mice are still not fully clear. We hypothesized that horizontal cells, amacrine cells, bipolar cells and glial cells are also affected in the retina of Npc1 mutant mice. RESULTS: Immunohistochemistry and electron microscopy were used to investigate pathologies of ganglion cells, horizontal cells, amacrine cells, bipolar cells, and optic nerves as well as altered activity of glial cells in Npc1 mutant mice. Electron microscopy reveals that electron-dense inclusions are generally accumulated in ganglion cells, bipolar cells, Müller cells, and in the optic nerve. Furthermore, abnormal arborisation and ectopic processes of horizontal and amacrine cells as well as defective bipolar cells are observed by immunohistochemistry for specific cellular markers. Furthermore, hyperactivity of glial cells, including astrocytes, microglial cells, and Müller cells, is also revealed. CONCLUSIONS: Our data extend previous findings to show multiple defects in the retina of Npc1 mutant mice, suggesting an important role of Npc1 protein in the normal function of the retina.


Asunto(s)
Neuroglía/patología , Neuronas/patología , Enfermedad de Niemann-Pick Tipo C/patología , Retina/patología , Animales , Apoptosis , Modelos Animales de Enfermedad , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular , Ratones Noqueados , Microscopía Electrónica , Neuroglía/metabolismo , Neuronas/metabolismo , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/metabolismo , Nervio Óptico/metabolismo , Nervio Óptico/patología , Proteínas/genética , Proteínas/metabolismo , Retina/metabolismo
15.
Glia ; 62(7): 1024-40, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24644136

RESUMEN

Niemann-Pick disease type C1 (NPC1) is a neurodegenerative disease with various progressive pathological features, for example, neuronal loss, dysmyelination, abnormal axon swelling, and gliosis, in the brain. Pathological activation of p38-mitogen-activated protein kinase (MAPK) results in hyperphosphorylation of tau protein, which contributes to the development of neurodegenerative diseases. In this study, axonal varicosities or spheroids and presynaptic aggregates in the spinal cord of the Npc1 mutant mice were found from postnatal day (P) 35 onwards, as indicated by the increased hyperphosphorylated neurofilament and synaptophysin immunoreactivity as well as the findings from electron microscopy. However, activities of astrocytes and microglia in the Npc1 mutant spinal cord were progressively increased earlier from P10 onwards, accompanied by increased expression of interleukin-1ß and apolipoprotein E, as well as up-regulated p38-MAPK activity and enhanced phosphorylated tau protein, but not cyclin-dependent kinase 5/p35 complex and glycogen synthase kinase-3ß. Taken together, our data suggest that the axonal pathologies in the Npc1 mutant spinal cord are strongly correlated with the increase of activated glial cells, which produce IL-1ß and ApoE, resulting in the activation of p38-MAPK signaling pathway and enhanced phosphorylated tau protein.


Asunto(s)
Axones/fisiología , Enfermedades Neurodegenerativas/fisiopatología , Neuroglía/fisiología , Proteínas/metabolismo , Médula Espinal/fisiopatología , Factores de Edad , Animales , Apolipoproteínas E/metabolismo , Astrocitos/patología , Astrocitos/fisiología , Axones/patología , Quinasa 5 Dependiente de la Ciclina/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Interleucina-1beta/metabolismo , Filamentos Intermedios/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones Endogámicos BALB C , Ratones Noqueados , Microglía/patología , Microglía/fisiología , Enfermedades Neurodegenerativas/patología , Neuroglía/patología , Proteína Niemann-Pick C1 , Fosforilación , Fosfotransferasas/metabolismo , Proteínas/genética , Médula Espinal/patología , Sinaptofisina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas tau/metabolismo
16.
Histol Histopathol ; 29(9): 1185-99, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24671883

RESUMEN

Niemann-Pick type C1 (NPC1) disease is an autosomal recessive disorder characterized by dysmyelination and neurodegeneration, which can result in the death of patients in early childhood in some cases. Members of the delta-protocadherins (Pcdhs) play important roles in neurogenesis and brain development. In this study, we compared expression profiles of Pcdhs in the brain of both wild-type and Npc1 mutant mice from postnatal day (P) 9 onwards by in situ hybridization. Our data show that laminar distribution of some Pcdhs in the cerebral cortex of Npc1 mutated mice is different from that of wild-type mice. Furthermore, expressions of Pcdhs by oligodendrocytes in the corpus callosum and by Purkinje cells and granular cells in the cerebellum are strongly decreased in Npc1 mutated mice at later stages. Taken together, our data suggest that aberrant expression of Pcdhs is a pathological process accompanied by neurodegeneration in Npc1 mutant mice.


Asunto(s)
Encéfalo/metabolismo , Encéfalo/patología , Cadherinas/biosíntesis , Enfermedad de Niemann-Pick Tipo C/metabolismo , Proteínas/genética , Animales , Western Blotting , Cadherinas/análisis , Modelos Animales de Enfermedad , Inmunohistoquímica , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Mutantes , Mutación , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/patología , Análisis de Secuencia por Matrices de Oligonucleótidos
17.
Histol Histopathol ; 29(10): 1287-93, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24515304

RESUMEN

Nanog is a potential stem cell marker and is considered a regeneration factor during tissue repair. In the present study, we investigated expression patterns of nanog in the rat heart after acute myocardial infarction by semi-quantitative RT-PCR, immunohistochemistry and Western blot analyses. Our results show that nanog at both mRNA and protein levels is positively expressed in myocardial cells, fibroblasts and small round cells in different myocardial zones at different stages after myocardial infarction, showing a spatio-temporal and dynamic change. After myocardial infarction, the nanog expression in fibroblasts and small round cells in the infarcted zone (IZ) is much stronger than that in the margin zone (MZ) and remote infarcted zone (RIZ). From day 7 after myocardial infarction, the fibroblasts and small cells strongly expressed nanog protein in the IZ, and a few myocardial cells in the MZ and the RIZ and the numbers of nanog-positive fibroblasts and small cells reached the highest peak at 21 days after myocardial infarction, but in this period the number of nanog-positive myocardial cells decreased gradually. At 28 days after myocardial infarction, the numbers of all nanog-positive cells decreased into a low level. Therefore, our data suggest that all myocardial cells, fibroblasts and small round cells are involved in myocardial reconstruction after cardiac infarction. The nanog-positive myocardial cells may respond to early myocardial repair, and the nanog-positive fibroblasts and small round cells are the main source for myocardial reconstruction after cardiac infarction.


Asunto(s)
Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Factores de Transcripción/biosíntesis , Animales , Western Blotting , Diferenciación Celular , Colorantes , Inmunohistoquímica , Proteína Homeótica Nanog , Reacción en Cadena de la Polimerasa , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Células Madre
18.
PLoS One ; 9(1): e84617, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24404179

RESUMEN

Members of the ADAM (a disintegrin and metalloprotease) family are involved in embryogenesis and tissue formation via their proteolytic function, cell-cell and cell-matrix interactions. ADAM10 is expressed temporally and spatially in the developing chicken spinal cord, but its function remains elusive. In the present study, we address this question by electroporating ADAM10 specific morpholino antisense oligonucleotides (ADAM10-mo) or dominant-negative ADAM10 (dn-ADAM10) plasmid into the developing chicken spinal cord as well as by in vitro cell culture investigation. Our results show that downregulation of ADAM10 drives precocious differentiation of neural progenitor cells and radial glial cells, resulting in an increase of neurons in the developing spinal cord, even in the prospective ventricular zone. Remarkably, overexpression of the dn-ADAM10 plasmid mutated in the metalloprotease domain (dn-ADAM10-me) mimics the phenotype as found by the ADAM10-mo transfection. Furthermore, in vitro experiments on cultured cells demonstrate that downregulation of ADAM10 decreases the amount of the cleaved intracellular part of Notch1 receptor and its target, and increases the number of ßIII-tubulin-positive cells during neural progenitor cell differentiation. Taken together, our data suggest that ADAM10 negatively regulates neuronal differentiation, possibly via its proteolytic effect on the Notch signaling during development of the spinal cord.


Asunto(s)
Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Diferenciación Celular , Proteínas de la Membrana/metabolismo , Neuronas/citología , Neuronas/metabolismo , Médula Espinal/embriología , Médula Espinal/metabolismo , Proteínas ADAM/química , Proteínas ADAM/genética , Proteína ADAM10 , Secretasas de la Proteína Precursora del Amiloide/química , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Diferenciación Celular/genética , Proliferación Celular , Embrión de Pollo , Regulación de la Expresión Génica , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Neuroglía/citología , Neuroglía/metabolismo , Dominios y Motivos de Interacción de Proteínas , Receptores Notch/metabolismo , Transducción de Señal
19.
Dev Growth Differ ; 55(3): 368-76, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23496030

RESUMEN

Members of the ADAM (a disintegrin and metalloprotease) family are type I transmembrane proteins involved in biological processes of proteolysis, cell adhesion, cell-matrix interaction, as well as in the intracellular signaling transduction. In the present study, expression patterns of seven members of the ADAM family were investigated at the early stages of the developing cochlea by in situ hybridization. The results show that each individual ADAM is expressed and regulated in the early developing cochlea. ADAM9, ADAM10, ADAM17, and ADAM23 are initially and widely expressed in the otic vesicle at embryonic day 2.5 (E2.5) and in the differential elements of the cochlear duct at E9, while ADAM12 is expressed in acoustic ganglion cells at E7. ADAM22 is detectable in cochlear ganglion cells as early as from E4 and in the basilar papilla from E7. Therefore, the present study extends our previous results and suggests that ADAMs also play a role in the early cochlear development.


Asunto(s)
Proteínas ADAM/metabolismo , Cóclea/embriología , Cóclea/metabolismo , Animales , Embrión de Pollo , Pollos , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ
20.
PLoS One ; 8(12): e82216, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24391715

RESUMEN

BACKGROUND: Niemann-Pick type C disease (NPC) is a rare autosomal recessive lipid storage disease characterized by progressive neurodegeneration. As only a few studies have been conducted on the impact of NPC on sensory systems, we used a mutant mouse model (NPC1(-/-)) to examine the effects of this disorder to morphologically distinct regions of the olfactory system, namely the olfactory epithelium (OE) and olfactory bulb (OB). METHODOLOGY/PRINCIPAL FINDINGS: For structural and functional analysis immunohistochemistry, electron microscopy, western blotting, and electrophysiology have been applied. For histochemistry and western blotting, we used antibodies against a series of neuronal and glia marker proteins, as well as macrophage markers. NPC1(-/-) animals present myelin-like lysosomal deposits in virtually all types of cells of the peripheral and central olfactory system. Especially supporting cells of the OE and central glia cells are affected, resulting in pronounced astrocytosis and microgliosis in the OB and other olfactory cortices. Up-regulation of Galectin-3, Cathepsin D and GFAP in the cortical layers of the OB underlines the critical role and location of the OB as a possible entrance gate for noxious substances. Unmyelinated olfactory afferents of the lamina propria seem less affected than ensheathing cells. Supporting the structural findings, electro-olfactometry of the olfactory mucosa suggests that NPC1(-/-) animals exhibit olfactory and trigeminal deficits. CONCLUSIONS/SIGNIFICANCE: Our data demonstrate a pronounced neurodegeneration and glia activation in the olfactory system of NPC1(-/-), which is accompanied by sensory deficits.


Asunto(s)
Enfermedad de Niemann-Pick Tipo C/genética , Enfermedad de Niemann-Pick Tipo C/fisiopatología , Proteínas/genética , Proteínas/fisiología , Olfato/genética , Olfato/fisiología , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Biomarcadores/metabolismo , Catepsina D/metabolismo , Modelos Animales de Enfermedad , Femenino , Galectina 3/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lisosomas/metabolismo , Lisosomas/patología , Activación de Macrófagos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Microscopía Electrónica de Transmisión , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Degeneración Nerviosa/fisiopatología , Neuroglía/patología , Neuroglía/fisiología , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/patología , Bulbo Olfatorio/patología , Bulbo Olfatorio/fisiopatología , Mucosa Olfatoria/patología , Mucosa Olfatoria/fisiopatología , Vías Olfatorias/patología , Vías Olfatorias/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...