Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Neuropathol Appl Neurobiol ; 47(5): 625-639, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33345400

RESUMEN

AIMS: Amyloid-ß (Aß) oligomers trigger synaptic degeneration that precedes plaque and tangle pathology. However, the signalling molecules that link Aß oligomers to synaptic pathology remain unclear. Here, we addressed the potential role of RAPGEF2 as a novel signalling molecule in Aß oligomer-induced synaptic and cognitive impairments in human-mutant amyloid precursor protein (APP) mouse models of Alzheimer's disease (AD). METHODS: To investigate the role of RAPGEF2 in Aß oligomer-induced synaptic and cognitive impairments, we utilised a combination of approaches including biochemistry, molecular cell biology, light and electron microscopy, behavioural tests with primary neuron cultures, multiple AD mouse models and post-mortem human AD brain tissue. RESULTS: We found significantly elevated RAPGEF2 levels in the post-mortem human AD hippocampus. RAPGEF2 levels also increased in the transgenic AD mouse models, generating high levels of Aß oligomers before exhibiting synaptic and cognitive impairment. RAPGEF2 upregulation activated the downstream effectors Rap2 and JNK. In cultured hippocampal neurons, oligomeric Aß treatment increased the fluorescence intensity of RAPGEF2 and reduced the number of dendritic spines and the intensities of synaptic marker proteins, while silencing RAPGEF2 expression blocked Aß oligomer-induced synapse loss. Additionally, the in vivo knockdown of RAPGEF2 expression in the AD hippocampus prevented cognitive deficits and the loss of excitatory synapses. CONCLUSIONS: These findings demonstrate that the upregulation of RAPGEF2 levels mediates Aß oligomer-induced synaptic and cognitive disturbances in the AD hippocampus. We propose that an early intervention regarding RAPGEF2 expression may have beneficial effects on early synaptic pathology and memory loss in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/patología , Disfunción Cognitiva/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Disfunción Cognitiva/genética , Disfunción Cognitiva/patología , Modelos Animales de Enfermedad , Femenino , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Masculino , Ratones Transgénicos , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Sinapsis/metabolismo , Sinapsis/patología
2.
FASEB J ; 34(5): 6965-6983, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32237183

RESUMEN

Microtubule-associated protein (MAP) 2 has been perceived as a static cytoskeletal protein enriched in neuronal dendritic shafts. Emerging evidence indicates dynamic functions for various MAPs in activity-dependent synaptic plasticity. However, it is unclear how MAP2 is associated with synaptic plasticity mechanisms. Here, we demonstrate that specific silencing of high-molecular-weight MAP2 in vivo abolished induction of long-term potentiation (LTP) in the Schaffer collateral pathway of CA1 pyramidal neurons and in vitro blocked LTP-induced surface delivery of AMPA receptors and spine enlargement. In mature hippocampal neurons, we observed rapid translocation of a subpopulation of MAP2, present in dendritic shafts, to spines following LTP stimulation. Time-lapse confocal imaging showed that spine translocation of MAP2 was coupled with LTP-induced spine enlargement. Consistently, immunogold electron microscopy revealed that LTP stimulation of the Schaffer collateral pathway promoted MAP2 labeling in spine heads of CA1 neurons. This translocation depended on NMDA receptor activation and Ras-MAPK signaling. Furthermore, LTP stimulation led to an increase in surface-expressed AMPA receptors specifically in the neurons with MAP2 spine translocation. Altogether, this study indicates a novel role for MAP2 in LTP mechanisms and suggests that MAP2 participates in activity-dependent synaptic plasticity in mature hippocampal networks.


Asunto(s)
Región CA1 Hipocampal/citología , Región CA1 Hipocampal/metabolismo , Potenciación a Largo Plazo/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Células Piramidales/metabolismo , Animales , Células Cultivadas , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Técnicas In Vitro , Sistema de Señalización de MAP Quinasas , Masculino , Ratones Endogámicos C57BL , Microscopía Inmunoelectrónica , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/genética , Plasticidad Neuronal/fisiología , Transporte de Proteínas , Células Piramidales/ultraestructura , Interferencia de ARN , ARN Interferente Pequeño/genética , Ratas , Receptores AMPA/metabolismo
4.
Int J Mol Med ; 52(6)2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37888610

RESUMEN

Exosomes are used as innovative treatment options for repairing skin defects, such as aging, atopic dermatitis and wounds. However, the effects of exosomes obtained from human foreskin fibroblasts BJ­5ta (BJ­5ta Exo) on ultraviolet B (UVB)­mediated photoaging have not been previously reported, at least to the best of our knowledge. Therefore, the present study aimed to investigate the anti­photoaging effects of BJ­5ta Exo on UVB radiation in human skin fibroblasts and SKH­1 hairless mice. The results revealed that BJ­5ta Exo decreased the production of reactive oxygen species and inhibited the decrease in the expression levels of superoxide dismutase 1 and 2, glutathione peroxidase and catalase following UVB exposure. In addition, BJ­5ta Exo attenuated the decrease in nuclear factor erythroid 2­related factor 2 levels induced by UVB rays, indicating its scavenging activity against oxidative stress. Moreover, BJ­5ta Exo inhibited the UVB­induced increase in the levels of γH2AX, p53/21 and cleaved PARP, whereas it promoted DNA double­strand break repair through radiation sensitive 52 and effectively activated the TGF­ß1/Smad pathway. BJ­5ta Exo also protected against UVB­induced senescence, as indicated by the downregulation in the levels of senescence­associated ß­galactosidase and p16. In a mouse model of photoaging, BJ­5ta Exo prevented the UVB­induced increase in transepidermal water loss, wrinkle formation and MMP­1 expression, while also suppressing the UVB­mediated decrease in collagen type I and elastin levels in the dorsal skin. Overall, the findings of the present study suggest that BJ­5ta Exo represent an effective anti­photoaging agent, which can be used as a component in cosmetic products.


Asunto(s)
Exosomas , Envejecimiento de la Piel , Animales , Ratones , Humanos , Exosomas/metabolismo , Piel/metabolismo , Colágeno Tipo I/metabolismo , Fibroblastos/metabolismo , Rayos Ultravioleta/efectos adversos , Especies Reactivas de Oxígeno/metabolismo
5.
J Neurosci ; 29(18): 5974-84, 2009 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-19420263

RESUMEN

Neural-cadherin (N-cadherin), a member of the classical cadherin family of transmembrane glycoproteins, mediates cellular recognition and cell-cell adhesion through calcium-dependent homophilic interactions and plays important roles in the development and maintenance of the nervous system. Metalloproteinase is known to cleave N-cadherin, which is further cleaved by gamma-secretase. The intracellular domain of N-cadherin interacts with beta-catenin, and beta-catenin stability is critical for cell-cell adhesion and cell survival. In the present study, we showed that N-cadherin is cleaved specifically by calpain, resulting in the generation of a novel 110 kDa fragment. The cleavage occurred in ischemic brain lesions and in vitro neural cells in the presence of NMDA and ionomycin, and was restored by calpain inhibitors but not matrix metalloproteinase or gamma-secretase inhibitors. Calpain directly cleaved N-cadherin in in vitro calpain assays, and calpain inhibitors prevented its cleavage in a dose-dependent manner. Using N-cadherin deletion mutants, we found that calpain cleavage sites exist in at least four regions of the cytoplasmic domain. Treatment with NMDA induced neuronal death, and it suppressed the expression of surface N-cadherin and the N-cadherin/beta-catenin interaction, effects that were prevented by calpain inhibitor. Furthermore, calpain-mediated N-cadherin cleavage significantly affected cell-cell adhesion, AKT signaling, the N-cadherin/beta-catenin interaction and the Wnt target gene expressions through the accumulation of nuclear beta-catenin.


Asunto(s)
Lesiones Encefálicas/metabolismo , Cadherinas/metabolismo , Calpaína/metabolismo , Animales , Animales Recién Nacidos , Biotinilación/métodos , Lesiones Encefálicas/patología , Cadherinas/genética , Calcio/metabolismo , Calpaína/farmacología , Células Cultivadas , Modelos Animales de Enfermedad , Maleato de Dizocilpina/farmacología , Embrión de Mamíferos , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Inhibidores Enzimáticos/farmacología , Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/genética , Técnicas In Vitro , Ionomicina/farmacología , Ionóforos/farmacología , Ratones , Modelos Biológicos , Mutación/genética , N-Metilaspartato/farmacología , Neuronas/efectos de los fármacos , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Fracciones Subcelulares/efectos de los fármacos , Transfección/métodos , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
6.
PPAR Res ; 2020: 3785137, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32099540

RESUMEN

Cells can shift their metabolism between glycolysis and oxidative phosphorylation to enact their cell fate program in response to external signals. Widely distributed α 1-adrenergic receptors (ARs) are physiologically stimulated during exercise, were reported to associate with the activating energetic AMPK pathway, and are expected to have biological effects beyond their hemodynamic effects. To investigate the effects and mechanism of AR stimulation on the physiology of the whole body, various in vitro and in vivo experiments were conducted using the AR agonist midodrine, 2-amino-N-[2-(2,5-dimethoxyphenyl)-2-hydroxy-ethyl]-acetamide. The expression of various biomarkers involved in ATP production was estimated through Western blotting, reverse transcription polymerase chain reaction, oxygen consumption rate, enzyme-linked immunosorbent assay (ELISA), fluorescence staining, and Oil red O staining in several cell lines (skeletal muscle, cardiac muscle, liver, macrophage, vascular endothelial, and adipose cells). In spontaneously hypertensive rats, blood pressure, blood analysis, organ-specific biomarkers, and general biomolecules related to ATP production were measured with Western blot analysis, immunohistochemistry, ELISA, and echocardiography. Pharmacological activation of α 1-adrenergic receptors in C2C12 skeletal muscle cells promoted mitochondrial oxidative phosphorylation and ATP production by increasing the expression of catabolic molecules, including PPARδ, AMPK, and PGC-1α, through cytosolic calcium signaling and increased GLUT4 expression, as seen in exercise. It also activated those energetic molecules and mitochondrial oxidative phosphorylation with cardiomyocytes, endothelial cells, adipocytes, macrophages, and hepatic cells and affected their relevant cell-specific biological functions. All of those effects occurred around 3 h (and peaked 6 h) after midodrine treatment. In spontaneously hypertensive rats, α 1-adrenergic receptor stimulation affected mitochondrial oxidative phosphorylation and ATP production by activating PPARδ, AMPK, and PGC-1α and the relevant biologic functions of multiple organs, suggesting organ crosstalk. The treatment lowered blood pressure, fat and body weight, cholesterol levels, and inflammatory activity; increased ATP content and insulin sensitivity in skeletal muscles; and increased cardiac contractile function without exercise training. These results suggest that the activation of α 1-adrenergic receptor stimulates energetic reprogramming via PPARδ that increases mitochondrial oxidative phosphorylation and has healthy and organ-specific biological effects in multiple organs, including skeletal muscle, beyond its vasomotion effect. In addition, the action mechanism of α 1-adrenergic receptor may be mainly exerted via PPARδ.

7.
Autophagy ; 16(8): 1396-1412, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-31690171

RESUMEN

TARDBP/TDP-43 (TAR DNA binding protein) proteinopathies are a common feature in a variety of neurodegenerative disorders, including amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration (FTLD), and Alzheimer disease (AD). However, the molecular mechanisms underlying TARDBP-induced neurotoxicity are largely unknown. In this study, we demonstrated that TARDBP proteinopathies induce impairment in the ubiquitin proteasome system (UPS), as evidenced by an accumulation of ubiquitinated proteins and a reduction in proteasome activity in neuronal cells. Through kinase inhibitor screening, we identified PTK2/FAK (PTK2 protein tyrosine kinase 2) as a suppressor of neurotoxicity induced by UPS impairment. Importantly, PTK2 inhibition significantly reduced ubiquitin aggregates and attenuated TARDBP-induced cytotoxicity in a Drosophila model of TARDBP proteinopathies. We further identified that phosphorylation of SQSTM1/p62 (sequestosome 1) at S403 (p-SQSTM1 [S403]), a key component in the autophagic degradation of poly-ubiquitinated proteins, is increased upon TARDBP overexpression and is dependent on the activation of PTK2 in neuronal cells. Moreover, expressing a non-phosphorylated form of SQSTM1 (SQSTM1S403A) significantly repressed the accumulation of insoluble poly-ubiquitinated proteins and neurotoxicity induced by TARDBP overexpression in neuronal cells. In addition, TBK1 (TANK binding kinase 1), a kinase that phosphorylates S403 of SQSTM1, was found to be involved in the PTK2-mediated phosphorylation of SQSTM1. Taken together, our data suggest that the PTK2-TBK1-SQSTM1 axis plays a critical role in the pathogenesis of TARDBP by regulating neurotoxicity induced by UPS impairment. Therefore, targeting the PTK2-TBK1-SQSTM1 axis may represent a novel therapeutic intervention for neurodegenerative diseases with TARDBP proteinopathies.Abbreviations: ALP: macroautophagy/autophagy lysosomal pathway; ALS: amyotrophic lateral sclerosis; ATXN2: ataxin 2; BafA1: bafilomycin A1; cCASP3: cleaved caspase 3; CSNK2: casein kinase 2; FTLD: frontotemporal lobar degeneration; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; OPTN: optineurin; PTK2/FAK: PTK2 protein tyrosine kinase 2; SQSTM1/p62: sequestosome 1; TARDBP/TDP-43: TAR DNA binding protein; TBK1: TANK binding kinase 1; ULK1: unc-51 like autophagy activating kinase 1; UPS: ubiquitin-proteasome system.


Asunto(s)
Quinasa 1 de Adhesión Focal/metabolismo , Proteína Sequestosoma-1/metabolismo , Proteinopatías TDP-43/metabolismo , Respuesta de Proteína Desplegada , Animales , Autofagia/efectos de los fármacos , Conducta Animal/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Drosophila melanogaster/metabolismo , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/genética , Ratones , Modelos Biológicos , Mutación/genética , Neurotoxinas/toxicidad , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Solubilidad , Proteínas Ubiquitinadas/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos
8.
PLoS One ; 13(4): e0195339, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29608598

RESUMEN

α-Synuclein (α-syn) is a major component of Lewy bodies found in synucleinopathies including Parkinson's disease (PD) and Dementia with Lewy Bodies (DLB). Under the pathological conditions, α-syn tends to generate a diverse form of aggregates showing toxicity to neuronal cells and able to transmit across cells. However, mechanisms by which α-syn aggregates affect cytotoxicity in neurons have not been fully elucidated. Here we report that α-syn aggregates preferentially sequester specific synaptic proteins such as vesicle-associated membrane protein 2 (VAMP2) and synaptosomal-associated protein 25 (SNAP25) through direct binding which is resistant to SDS. The sequestration effect of α-syn aggregates was shown in a cell-free system, cultured primary neurons, and PD mouse model. Furthermore, we identified a specific blocking peptide derived from VAMP2 which partially inhibited the sequestration by α-syn aggregates and contributed to reduced neurotoxicity. These results provide a mechanism of neurotoxicity mediated by α-syn aggregates and suggest that the blocking peptide interfering with the pathological role of α-syn aggregates could be useful for designing a potential therapeutic drug for the treatment of PD.


Asunto(s)
Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Péptidos/farmacología , Agregación Patológica de Proteínas/tratamiento farmacológico , Proteína 2 de Membrana Asociada a Vesículas/metabolismo , alfa-Sinucleína/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Escherichia coli , Humanos , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Neuronas/metabolismo , Neuronas/patología , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/patología , Agregación Patológica de Proteínas/metabolismo , Unión Proteica/efectos de los fármacos , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo
9.
JAKSTAT ; 2(2): e23282, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24058805

RESUMEN

Myogenic differentiation plays an important role in muscle regeneration and is regulated by two transcription factor families, MRFs and MEF2, which induce differentiation of myoblasts through expression of the muscle-specific gene, myogenin. In addition, many intracellular signaling pathways are also involved in myogenic differentiation, including p38 MAPK, ERK/MAPK and PI3K/AKT. The JAK-STAT pathway is activated by various cytokines and positively or negatively regulates the differentiation of myoblasts. JAK1 plays a notable role in proliferation; whereas, JAK2 and JAK3 function mainly in differentiation. The STATs, molecules downstream of JAK, regulate myogenesis. With JAK1, STAT1 promotes proliferation, while STAT3 has a dual effect on proliferation and differentiation. The JAK-STAT negative regulator, SOCS, is also associated with myogenesis; although, its role is controversial. In this review, we will discuss the role of the JAK-STAT pathway on myogenic differentiation.

10.
Cell Signal ; 24(3): 742-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22120524

RESUMEN

Skeletal muscle differentiation is regulated by transcription factors, including members of the myogenic regulatory factor (MRF) family and many signaling pathways. The JAK1 and JAK2 pathways are known to each have different effects on myoblast proliferation and differentiation; however, the role of JAK3 in myoblast differentiation remains unclear. In this study, we investigated the effect of JAK3 inhibition on myogenic differentiation in the C2C12 mouse myoblast cell line. During myogenic differentiation, treatment with the JAK3 inhibitor WHIp154 significantly increased the number of MHC-positive multinucleated myotubes and the expressions of myosin heavy chain (MHC), myogenin (MGN), MyoD, and myogenic enhancer factor 2 (MEF2). Knockdown of the JAK3 gene using siJAK3 also significantly increased MHC, MGN and MyoD mRNA expressions as well as insulin-like growth factor-II (IGF-II) gene expression. During differentiation, JAK3 was initially activated and later decreased. Differentiation decreased STAT1, which was further decreased by WHIp154. In contrast, STAT3 gradually was elevated during differentiation, and was increased by JAK3 inhibition. Moreover, we found that up-regulation of AKT activity and down-regulation of ERK activity cooperated to accelerate myogenic differentiation. Taken together, these data indicate that JAK3 inhibition potently facilitates myoblast differentiation through antagonistic STAT1/STAT3 activities. Additionally, JAK3 inhibition induced precocious differentiation and played important roles for terminal differentiation, including fusion, which is involved with regulation of AKT and ERK pathways.


Asunto(s)
Diferenciación Celular , Janus Quinasa 3/metabolismo , Mioblastos/citología , Mioblastos/enzimología , Animales , Línea Celular , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Janus Quinasa 3/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas , Ratones , Proteína MioD/genética , Proteína MioD/metabolismo , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Miogenina/genética , Miogenina/metabolismo , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Quinazolinas/química , Quinazolinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA