Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Gene Ther ; 30(1-2): 75-87, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-35132206

RESUMO

Traumatic brain injury (TBI) survivors suffer from long-term disability and neuropsychiatric sequelae due to irreparable brain tissue destruction. However, there are still few efficient therapies to promote neurorestoration in damaged brain tissue. This study aimed to investigate whether the pro-oncogenic gene ski can promote neurorestoration after TBI. We established a ski-overexpressing experimental TBI mouse model using adenovirus-mediated overexpression through immediate injection after injury. Hematoxylin-eosin staining, MRI-based 3D lesion volume reconstruction, neurobehavioral tests, and analyses of neuronal regeneration and astrogliosis were used to assess neurorestorative efficiency. The effects of ski overexpression on the proliferation of cultured immature neurons and astrocytes were evaluated using imaging flow cytometry. The Ski protein level increased in the perilesional region at 3 days post injury. ski overexpression further elevated Ski protein levels up to 14 days post injury. Lesion volume was attenuated by approximately 36-55% after ski overexpression, with better neurobehavioral recovery, more newborn immature and mature neurons, and less astrogliosis in the perilesional region. Imaging flow cytometry results showed that ski overexpression elevated the proliferation rate of immature neurons and reduced the proliferation rate of astrocytes. These results show that ski can be considered a novel neurorestoration-related gene that effectively promotes neurorestoration, facilitates neuronal regeneration, and reduces astrogliosis after TBI.


Assuntos
Lesões Encefálicas Traumáticas , Gliose , Camundongos , Animais , Gliose/genética , Gliose/metabolismo , Gliose/patologia , Neurônios/metabolismo , Lesões Encefálicas Traumáticas/terapia , Encéfalo/metabolismo , Regeneração
2.
Chin J Traumatol ; 2023 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-37679245

RESUMO

Adenosine exerts its dual functions of homeostasis and neuromodulation in the brain by acting at mainly the two G-protein coupled receptors, called A1 and A2A receptors. The adenosine A2A receptor (A2AR) antagonists have clinically pursued for the last two decades, leading to final approval of the istradefylline, an A2AR antagonist, for the treatment of OFF-Parkinson's disease (PD) patients. The approval paves the way to develop novel therapeutic methods for A2AR antagonists to address two major unmet medical needs in PD and traumatic brain injury (TBI), namely neuroprotection or improving cognition. In this review, we first consider the evidence for aberrantly increased adenosine signaling in PD and TBI and the sufficiency of the increased A2AR signaling to trigger neurotoxicity and cognitive impairment. We further discuss the increasing preclinical data on the reversal of cognitive deficits in PD and TBI by A2AR antagonists through control of degenerative proteins and synaptotoxicity, and on protection against TBI and PD pathologies by A2AR antagonists through control of neuroinflammation. Moreover, we provide the supporting evidence from multiple human prospective epidemiological studies which revealed an inverse relation between the consumption of caffeine and the risk of developing PD and cognitive decline in aging population and Alzheimer's disease patients. Collectively, the convergence of clinical, epidemiological and experimental evidence supports the validity of A2AR as a new therapeutic target and facilitates the design of A2AR antagonists in clinical trial for disease-modifying and cognitive benefit in PD and TBI patients.

3.
Sheng Li Xue Bao ; 74(4): 505-512, 2022 Aug 25.
Artigo em Zh | MEDLINE | ID: mdl-35993201

RESUMO

The purpose of the present study was to investigate the effect of glutamate scavenger oxaloacetate (OA) combined with CGS21680, an adenosine A2A receptor (A2AR) agonist, on acute traumatic brain injury (TBI), and to elucidate the underlying mechanisms. C57BL/6J mice were subjected to moderate-level TBI by controlled cortical impact, and then were treated with OA, CGS21680, or OA combined with CGS21680 at acute stage of TBI. At 24 h post TBI, neurological severity score, brain water content, glutamate concentration in cerebrospinal fluid (CSF), mRNA and protein levels of IL-1ß and TNF-α, mRNA level and activity of glutamate oxaloacetate aminotransferase (GOT), and ATP level of brain tissue were detected. The results showed that neurological deficit, brain water content, glutamate concentration in CSF, and the inflammatory cytokine IL-1ß and TNF-α production were exacerbated in CGS21680 treated mice. Administrating OA suppressed the rise of both glutamate concentration in CSF and brain water content, and elevated the ATP level of cerebral tissue. More interestingly, neurological deficit, brain edema, glutamate concentration, IL-1ß and TNF-α levels were ameliorated significantly in mice treated with OA combined with CGS21680. The combined treatment exhibited better therapeutic effects than single OA treatment. We also observed that GOT activity was enhanced in single CGS21680 treatment group, and both the GOT mRNA level and GOT activity were up-regulated in early-stage combined treatment group. These results suggest that A2AR can improve the efficiency of GOT and potentiate the ability of OA to metabolize glutamate. This may be the mechanism that A2AR activation in combination group augmented the neuroprotective effect of OA rather than aggravated the brain damages. Taken together, the present study provides a new insight for the clinical treatment of TBI with A2AR agonists and OA.


Assuntos
Agonistas do Receptor A2 de Adenosina , Lesões Encefálicas Traumáticas , Fármacos Neuroprotetores , Ácido Oxaloacético , Receptor A2A de Adenosina , Agonistas do Receptor A2 de Adenosina/farmacologia , Agonistas do Receptor A2 de Adenosina/uso terapêutico , Trifosfato de Adenosina , Animais , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/genética , Lesões Encefálicas/metabolismo , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/genética , Lesões Encefálicas Traumáticas/metabolismo , Ácido Glutâmico , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ácido Oxaloacético/farmacologia , Ácido Oxaloacético/uso terapêutico , RNA Mensageiro , Receptor A2A de Adenosina/genética , Receptor A2A de Adenosina/metabolismo , Fator de Necrose Tumoral alfa/genética , Água
4.
J Neuroinflammation ; 18(1): 241, 2021 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-34666797

RESUMO

BACKGROUND: Cognitive impairment in the late stage of traumatic brain injury (TBI) is associated with the NOD-, LRR and pyrin domain-containing protein 3 (NLRP3) inflammasome, which plays an important role in neuroinflammation. Although classical inflammatory pathways have been well-documented in the late stage of TBI (4-8 weeks post-injury), the mechanism by which the NLRP3 inflammasome impairs cognition is still unclear. METHODS: Mice lacking the gene encoding for NLRP3 (NLRP3-knockout mice) and their wild-type littermates were used in a controlled cortical impact model of TBI. Levels of NLRP3 inflammasome and inflammatory factors such as IL-1ß and HMGB1 were detected in post-injury hippocampal tissue, as well as long-term potentiation. Behaviors were assessed by T-maze test, novel object recognition, and nesting tests. Glycyrrhizin was used to antagonize HMGB1. Calcium imaging were performed on primary neuronal cultures. RESULTS: By using the NLRP3-knockout TBI model, we found that the continuous activation of the NLRP3 inflammasome and high mobility group box 1 (HMGB1) release were closely related to cognitive impairment. We also found that inhibition of HMGB1 improved LTP reduction and cognitive function by increasing the phosphorylation level of the NMDAR1 subunit at serine 896 while reducing NLRP3 inflammasome activation. CONCLUSION: NLRP3 inflammasome damages memory in the late stage of TBI primarily through HMGB1 upregulation and provides an explanation for the long-term progression of cognitive dysfunction.


Assuntos
Lesões Encefálicas Traumáticas/metabolismo , Disfunção Cognitiva/metabolismo , Proteína HMGB1/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Lesões Encefálicas Traumáticas/patologia , Células Cultivadas , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Técnicas de Cocultura , Disfunção Cognitiva/patologia , Potenciais Pós-Sinápticos Excitadores/fisiologia , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Cultura de Órgãos
5.
J Cell Mol Med ; 24(12): 7000-7014, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32394486

RESUMO

Spatial recognition memory impairment is an important complication after traumatic brain injury (TBI). We previously found that spatial recognition memory impairment can be alleviated in adenosine A2A receptor knockout (A2A R KO) mice after TBI, but the mechanism remains unclear. In the current study, we used manganese-enhanced magnetic resonance imaging and the Y-maze test to determine whether the electrical activity of neurons in the retrosplenial cortex (RSC) was reduced and spatial recognition memory was impaired in wild-type (WT) mice after moderate TBI. Furthermore, spatial recognition memory was damaged by optogenetically inhibiting the electrical activity of RSC neurons in WT mice. Additionally, the electrical activity of RSC neurons was significantly increased and spatial recognition memory impairment was reduced in A2A R KO mice after moderate TBI. Specific inhibition of A2A R in the ipsilateral RSC alleviated the impairment in spatial recognition memory in WT mice. In addition, A2A R KO improved autophagic flux in the ipsilateral RSC after injury. In primary cultured neurons, activation of A2A R reduced lysosomal-associated membrane protein 1 and cathepsin D (CTSD) levels, increased phosphorylated protein kinase A and phosphorylated extracellular signal-regulated kinase 2 levels, reduced transcription factor EB (TFEB) nuclear localization and impaired autophagic flux. These results suggest that the impairment of spatial recognition memory after TBI may be associated with impaired autophagic flux in the RSC and that A2A R activation may reduce lysosomal biogenesis through the PKA/ERK2/TFEB pathway to impair autophagic flux.


Assuntos
Antagonistas do Receptor A2 de Adenosina/uso terapêutico , Autofagia , Lesões Encefálicas Traumáticas/complicações , Giro do Cíngulo/patologia , Giro do Cíngulo/fisiopatologia , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/etiologia , Memória Espacial/efeitos dos fármacos , Antagonistas do Receptor A2 de Adenosina/farmacologia , Animais , Autofagia/efeitos dos fármacos , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Giro do Cíngulo/efeitos dos fármacos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Transtornos da Memória/fisiopatologia , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/patologia , Biogênese de Organelas
6.
J Neuroinflammation ; 16(1): 235, 2019 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-31771656

RESUMO

BACKGROUND: Vacuolar sorting protein 35 (VPS35), a critical component of retromer, is essential for selective endosome-to-Golgi retrieval of membrane proteins. It is highly expressed in microglial cells, in addition to neurons. We have previously demonstrated microglial VPS35's functions in preventing hippocampal, but not cortical, microglial activation, and in promoting adult hippocampal neurogenesis. However, microglial VPS35's role in the cortex in response to ischemic stroke remains largely unclear. METHODS: We used mice with VPS35 cKO (conditional knockout) in microglial cells and examined and compared their responses to ischemic stroke with control mice. The brain damage, cell death, changes in glial cells and gene expression, and sensorimotor deficits were assessed by a combination of immunohistochemical and immunofluorescence staining, RT-PCR, Western blot, and neurological functional behavior tests. RESULTS: We found that microglial VPS35 loss results in an increase of anti-inflammatory microglia in mouse cortex after ischemic stroke. The ischemic stroke-induced brain injury phenotypes, including brain damage, neuronal death, and sensorimotor deficits, were all attenuated by microglial VPS35-deficiency. Further analysis of protein expression changes revealed a reduction in CX3CR1 (CX3C chemokine receptor 1) in microglial VPS35-deficient cortex after ischemic stroke, implicating CX3CR1 as a potential cargo of VPS35 in this event. CONCLUSION: Together, these results reveal an unrecognized function of microglial VPS35 in enhancing ischemic brain injury-induced inflammatory microglia, but suppressing the injury-induced anti-inflammatory microglia. Consequently, microglial VPS35 cKO mice exhibit attenuation of ischemic brain injury response.


Assuntos
Isquemia Encefálica/metabolismo , Polaridade Celular/fisiologia , Microglia/metabolismo , Córtex Sensório-Motor/metabolismo , Acidente Vascular Cerebral/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Receptor 1 de Quimiocina CX3C/genética , Receptor 1 de Quimiocina CX3C/metabolismo , Morte Celular/fisiologia , Modelos Animais de Doenças , Gliose/genética , Gliose/metabolismo , Gliose/patologia , Camundongos , Camundongos Knockout , Destreza Motora/fisiologia , Córtex Sensório-Motor/patologia , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/patologia , Proteínas de Transporte Vesicular/genética
7.
Neurochem Res ; 44(12): 2755-2764, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31650360

RESUMO

Pathogens such as bacterial lipopolysaccharide (LPS) play an important role in promoting the production of the inflammatory cytokines interleukin-1 beta (IL-1ß) and tumour necrosis factor-α (TNF-α) in response to infection or damage in microglia. However, whether different signalling pathways regulate these two inflammatory factors remains unclear. The protein kinase C (PKC) family is involved in the regulation of inflammation, and our previous research showed that the activation of the PKC pathway played a key role in the LPS-induced transformation of the adenosine A2A receptor (A2AR) from anti-inflammatory activity to pro-inflammatory activity under high glutamate concentrations. Therefore, in the current study, we investigated the role of PKC in the LPS-induced production of these inflammatory cytokines in mouse primary microglia. GF109203X, a specific PKC inhibitor, inhibited the LPS-induced expression of IL-1ß messenger ribonucleic acid and intracellular protein in a dose-dependent manner. Moreover, 5 µM GF109203X prevented LPS-induced IL-1ß expression but did not significantly affect LPS-induced TNF-α expression. PKC promoted IL-1ß expression by regulating the activity of NF-κB but did not significantly impact the activity of ERK1/2. A2AR activation by CGS21680, an A2AR agonist, facilitated LPS-induced IL-1ß expression through the PKC pathway at high glutamate concentrations but did not significantly affect LPS-induced TNF-α expression. Taken together, these results suggest a new direction for specific intervention with LPS-induced inflammatory factors in response to specific signalling pathways and provide a mechanism for A2AR targeting, especially after brain injury, to influence inflammation by interfering with A2AR.


Assuntos
Ácido Glutâmico/metabolismo , Interleucina-1beta/metabolismo , Microglia/metabolismo , Proteína Quinase C/metabolismo , Receptor A2A de Adenosina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Adenosina/análogos & derivados , Adenosina/farmacologia , Agonistas do Receptor A2 de Adenosina/farmacologia , Animais , Indóis/farmacologia , Inflamação/induzido quimicamente , Lipopolissacarídeos , Maleimidas/farmacologia , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fenetilaminas/farmacologia , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição RelA/metabolismo
8.
Biochem Biophys Res Commun ; 498(4): 795-802, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29526759

RESUMO

Excitatory amino acid transporters (EAATs) on cerebral vascular endothelial cells play an important role in maintaining glutamate homeostasis in the brain. The dysfunction of endothelial EAATs is an important reason for the dramatically elevated brain glutamate levels after brain injury, such as traumatic brain injury (TBI). The adenosine A2A receptor (A2AR) plays an important role in regulating the brain glutamate level after brain injury; however, researchers have not clearly determined whether this role was related to its ability to regulate endothelial EAATs. Activation of A2AR in vitro not only decreased the PKA- and glutamate level-dependent strengthening of the interaction between NKA-α1 and the FXYD1 subunit and the subsequent decrease in the activity of Na+/K+-ATPases (NKAs) but also enhanced its interaction with EAATs and ultimately aggravated the reverse transport function of endothelial EAATs under oxygen-glucose deprivation (OGD) conditions. Conversely, inhibition of A2AR restored the normal transport of EAAT. Moreover, A2AR inhibition increased NKA activity and decreased its interaction with EAATs in isolated brain capillaries after TBI, further confirming its role in endothelial EAATs in vivo. Based on our results, A2AR played an important role in regulating endothelial EAAT function, and strategies that restore the normal transport of endothelial EAATs through the inhibition of A2AR might serve as an effective treatment for brain injury.


Assuntos
Antagonistas do Receptor A2 de Adenosina/farmacologia , Lesões Encefálicas Traumáticas/metabolismo , Encéfalo/metabolismo , Proteínas de Transporte de Glutamato da Membrana Plasmática/metabolismo , Ácido Glutâmico/metabolismo , Receptor A2A de Adenosina/metabolismo , Agonistas do Receptor A2 de Adenosina/farmacologia , Animais , Encéfalo/efeitos dos fármacos , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/genética , Endotélio/efeitos dos fármacos , Endotélio/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor A2A de Adenosina/genética , ATPase Trocadora de Sódio-Potássio/metabolismo
9.
Mol Cell Biochem ; 440(1-2): 189-197, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28828564

RESUMO

Glucocorticoids are commonly used for the treatment of pancreatitis and complicated acute lung injury and help to reduce the mortality rates of both. The effect of gene variants in heat shock protein 90 (Hsp90), a key chaperone molecule of the glucocorticoid receptor (GR), on the therapeutic effect of glucocorticoids is unclear. Our study aims to investigate the different susceptibility to glucocorticoid treatment in BALB/c and C57BL/6 mice carrying different Hsp90 genotypes in an animal model of pancreatitis-induced lung injury. Compared with BALB/c mice, C57BL/6 mice have lower mortality rates, decreased water content in their lungs, and a lower level of IL-1 beta in an animal model of acute pancreatitis. C57BL/6 mice show a greater therapeutic effect and increased GR binding activities with glucocorticoid responsive element compared to BALB/c mice after a 0.4 mg/kg dexamethasone (DEX) treatment. Treatment with a higher dose of DEX (4 mg/kg) significantly reduced mortality rates and increased GR-GRE binding activity in both strains of mice, and there was no significant difference between the two strains. DEX did not exert a protective role after geldanamycin, a specific inhibitor of Hsp90, was administered in both strains of mice. Our study revealed that Hsp90 gene variants are responsible for the greater therapeutic effect of DEX in C57BL/6 mice compared to BALB/c mice, which implies that combining DEX treatment with Hsp90 regulation would promote the efficiency of DEX and would be an effective way to alleviate the side effects of hormone therapy.


Assuntos
Dexametasona/farmacologia , Proteínas de Choque Térmico HSP90/metabolismo , Lesão Pulmonar , Pulmão/metabolismo , Pancreatite , Receptores de Glucocorticoides/metabolismo , Animais , Interleucina-1beta/metabolismo , Pulmão/patologia , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/etiologia , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pancreatite/complicações , Pancreatite/tratamento farmacológico , Pancreatite/metabolismo , Pancreatite/patologia
10.
Growth Factors ; 34(3-4): 119-27, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27418111

RESUMO

We previously demonstrated that cellular Sloan-Kettering Institute (c-Ski) played a dual role, both promoting wound healing and alleviating scar formation. However, its mechanism and therapeutic effects are not clear, especially compared with widely used treatments, such as basic fibroblast growth factor (bFGF) administration. However, Ski treatment led to an even shorter healing time and a more significant reduction in scar area than bFGF treatment. The mechanism underlying this difference was related to a reduced inflammatory response, more rapid re-epithelialization, less collagen after healing and a greater reduction in the proportion of alpha-smooth muscle actin and SMemb-positive cells after Ski treatment. These results not only confirm that Ski plays a dual role in promoting healing and reducing scarring but also suggest that Ski yields better treatment effects than bFGF, indicating better potential therapeutic effects in wound repair.


Assuntos
Fator 2 de Crescimento de Fibroblastos/farmacologia , Terapia Genética/métodos , Proteínas Proto-Oncogênicas/genética , Cicatrização/genética , Actinas/genética , Actinas/metabolismo , Animais , Colágeno/genética , Colágeno/metabolismo , Feminino , Terapia Genética/efeitos adversos , Masculino , Proteínas Proto-Oncogênicas/metabolismo , Ratos , Ratos Wistar , Proteínas Recombinantes , Cicatrização/efeitos dos fármacos
11.
Cell Physiol Biochem ; 38(4): 1354-64, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27007335

RESUMO

BACKGROUND/AIMS: Although it has been reported that somatostatin (SOM) upregulated the level of 90-kD heat shock protein (Hsp90), which participates in the inflammatory regulation by its client proteins, such as glucocorticoid receptor (GR), it remains unclear if it has a protective role against acute lung injury (ALI). METHODS: ALI model was established by the injection of oleic acid (OA) into the tail vein of mice. Lung injury was assessed by histological analysis, lung water content and arterial blood gases. The levels of Hsp90 and GR, the binding capacity and the affinity of GR were examined. RESULTS: It was showed that pretreatment with SOM significantly increased Hsp90 levels and alleviated lung injuries in OA-injected mice. Furthermore, SOM increased the GR expression and improved the affinity of the GR in animals with lung injury. However, little alteration was found in the maximum binding capacity of the GR in mice with or without SOM. CONCLUSION: The data indicate SOM exerts a protective effect by increasing Hsp90 abundant and further enhancing the affinity of the GR. The beneficial effects of SOM treatment provide a new strategy for modulation of GR efficiency and alleviation of acute lung injury.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Receptores de Glucocorticoides/metabolismo , Somatostatina/uso terapêutico , Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Gasometria , Western Blotting , Modelos Animais de Doenças , Proteínas de Choque Térmico HSP90/química , Proteínas de Choque Térmico HSP90/metabolismo , Hormônios/farmacologia , Hormônios/uso terapêutico , Ligantes , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Ligação Proteica , RNA Mensageiro/metabolismo , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/genética , Somatostatina/farmacologia , Regulação para Cima/efeitos dos fármacos
12.
Cell Tissue Res ; 366(2): 411-425, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27324125

RESUMO

Increased circulating syncytiotrophoblast microparticles (STBMs) are often associated with preeclampsia (PE) but the molecular mechanisms regulating STBM shedding remain elusive. Experimental evidence has shown that actin plays a key role in STBM shedding and that Rho/ROCK is important in regulating actin rearrangement. To investigate the role of RhoB/ROCK-regulated actin arrangement in STBM shedding in PE, chorionic villous explants were prepared from placenta of patients with normotensive or PE pregnancies and BeWo cells were fused to imitate syncytiotrophoblasts. The oxygen-glucose deprivation (OGD) conditions were applied to imitate the pathophysiology of PE in vitro. The results showed that RhoB and ROCK were activated in the preeclamptic placenta, accompanied by increased actin polymerization and decreased outgrowing microvilli. In villous tissue cultures or BeWo cells, OGD activated RhoB, ROCK1 and ROCK2 and promoted STBM shedding and actin stress fibers formation. In BeWo cells, RhoB overexpression activated ROCK1 and ROCK2, leading to F-actin redistribution and STBM shedding and the OGD-induced actin polymerization and STBM shedding could be reversed by RhoB or ROCK knockdown. These results reveal that RhoB and ROCK play a key role in PE by targeting STBM shedding through actin rearrangement and that RhoB/ROCK intervention may be a potential therapeutic strategy for PE.


Assuntos
Micropartículas Derivadas de Células/metabolismo , Glucose/deficiência , Oxigênio/farmacologia , Pré-Eclâmpsia/metabolismo , Pré-Eclâmpsia/patologia , Trofoblastos/metabolismo , Quinases Associadas a rho/metabolismo , Proteína rhoB de Ligação ao GTP/metabolismo , Actinas/metabolismo , Apoptose , Linhagem Celular Tumoral , Ativação Enzimática , Feminino , Humanos , Microvilosidades/metabolismo , Polimerização , Gravidez
13.
Cell Physiol Biochem ; 35(2): 616-26, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25612921

RESUMO

BACKGROUND: Nogo-A, a major myelin-associated inhibitor, can inhibit injured optic nerve regeneration. However, whether Amino-Nogo is the most important functional domain of Nogo-A remains unknown. This study aimed to identify the role of Amino-Nogo following optic nerve injury, and the mechanism of the Amino-Nogo-integrin αv signaling pathway in vivo. METHODS: Sprague-Dawley rats with optic nerve crush injury were injected with Nogo-A siRNA (Nogo-A-siRNA), the Nogo-66 functional domain antagonist peptide of Nogo-A (Nep1-40) or a recombinant rat Amino-Nogo-A protein (∆20) into the vitreous cavity to knock down Nogo-A, inhibit Nogo-66 or activate the Amino-Nogo, resparately. Retinal ganglion cell (RGC) density, axon regeneration and the pattern of NPN of visual electrophysiology (flash visual evoked potentials [F-VEP]) at different times post-injury were investigated. RESULTS: Our study revealed a lower RGC survival rate; shorter axonal outgrowth; longer N1, P1 and N2 waves latencies; and lower N1-P1 and P1-N2 amplitudes in the Δ20 group, and Δ20 treatment significantly attenuated integrin αv expression and phosphorylated focal adhesion kinase (p-FAK) levels. In the Nep1-40 and Nogo-A siRNA groups, there were higher RGC survival rates, longer axonal outgrowth, shorter N1 and P1 wave latencies, and higher N1-P1 and P1-N2amplitudes. Nogo-A siRNA treatment significantly increased integrin αv expression and p-FAK levels. Nepl-40 treatment did not alter integrin αv expression. In addition, there was no significant change in integrin α5 in any group. CONCLUSION: These results suggest that the integrin signaling pathway is regulated by Amino-Nogo, which inhibits optic nerve regeneration and functional recovery, and that the integrin subunit involved might be integrin αv but not integrin α5.


Assuntos
Integrina alfaV/metabolismo , Proteínas da Mielina/antagonistas & inibidores , Proteínas da Mielina/química , Regeneração Nervosa , Nervo Óptico/fisiopatologia , Transdução de Sinais , Animais , Potenciais Evocados Visuais , Técnicas de Silenciamento de Genes , Proteínas da Mielina/metabolismo , Proteínas Nogo , Nervo Óptico/citologia , Traumatismos do Nervo Óptico/metabolismo , Traumatismos do Nervo Óptico/fisiopatologia , Fragmentos de Peptídeos/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Células Ganglionares da Retina/metabolismo
14.
Chin J Traumatol ; 18(4): 187-93, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26764538

RESUMO

Explosive blast injury has become the most prevalent injury in recent military conflicts and terrorist attacks. The magnitude of this kind of polytrauma is complex due to the basic physics of blast and the surrounding environments. Therefore, development of stable, reproducible and controllable animal model using an ideal blast simulation device is the key of blast injury research. The present review addresses the modeling of blast injury and applications of shock tubes.


Assuntos
Traumatismos por Explosões/etiologia , Modelos Animais de Doenças , Animais , Ondas de Choque de Alta Energia , Pesquisa
15.
Chin J Traumatol ; 18(4): 194-200, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26764539

RESUMO

With the increasing incidence of blast injury, the research on its mechanisms and protective measures draws more and more attention. Blast injury has many characteristics different from general war injuries or trauma. For example, soldiers often have various degrees of visceral injury without significant surface damage, combined injuries and arterial air embolism. Researchers in China began to investigate blast injury later than the United States and Sweden, but the development is so fast that lots of achievements have been gained, including the development of biological shock tube, the mechanisms and characteristics of blast injury in various organs, as well as protective measures under special environments. This article reviews the past and current situation of blast injury research in China.


Assuntos
Traumatismos por Explosões/etiologia , Animais , Traumatismos por Explosões/diagnóstico , Traumatismos por Explosões/prevenção & controle , Traumatismos por Explosões/terapia , Lesões Encefálicas Traumáticas/etiologia , China , Modelos Animais de Doenças , Orelha/lesões , Traumatismos Oculares/etiologia , Ondas de Choque de Alta Energia , Humanos , Lesão Pulmonar/etiologia , Pesquisa
16.
Chin J Traumatol ; 18(4): 204-11, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26764541

RESUMO

OBJECTIVE: To investigate the effects of three different ways of chronic caffeine administration on blast- induced memory dysfunction and to explore the underlying mechanisms. METHODS: Adult male C57BL/6 mice were used and randomly divided into five groups: control: without blast exposure, con-water: administrated with water continuously before and after blast-induced traumatic brain injury (bTBI), con-caffeine: administrated with caffeine continuously for 1 month before and after bTBI, pre-caffeine: chronically administrated with caffeine for 1 month before bTBI and withdrawal after bTBI, post-caffeine: chronically administrated with caffeine after bTBI. After being subjected to moderate intensity of blast injury, mice were recorded for learning and memory performance using Morris water maze (MWM) paradigms at 1, 4, and 8 weeks post-blast injury. Neurological deficit scoring, glutamate concentration, proinflammatory cytokines production, and neuropathological changes at 24 h, 1, 4, and 8 weeks post-bTBI were examined to evaluate the brain injury in early and prolonged stages. Adenosine A1 receptor expression was detected using qPCR. RESULTS: All of the three ways of chronic caffeine exposure ameliorated blast-induced memory deficit, which is correlated with the neuroprotective effects against excitotoxicity, inflammation, astrogliosis and neuronal loss at different stages of injury. Continuous caffeine treatment played positive roles in both early and prolonged stages of bTBI; pre-bTBI and post-bTBI treatment of caffeine tended to exert neuroprotective effects at early and prolonged stages of bTBI respectively. Up-regulation of adenosine A1 receptor expression might contribute to the favorable effects of chronic caffeine consumption. CONCLUSION: Since caffeinated beverages are widely consumed in both civilian and military personnel and are convenient to get, the results may provide a promising prophylactic strategy for blast-induced neurotrauma and the consequent cognitive impairment.


Assuntos
Traumatismos por Explosões/complicações , Lesões Encefálicas Traumáticas/complicações , Cafeína/farmacologia , Transtornos da Memória/prevenção & controle , Animais , Córtex Cerebral/patologia , Hipocampo/patologia , Masculino , Transtornos da Memória/etiologia , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/análise , Receptor A1 de Adenosina/genética
17.
CNS Neurosci Ther ; 30(1): e14408, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37564004

RESUMO

AIMS: Adenosine 2A receptor (A2A R) is widely expressed in the brain and plays important roles in neuroinflammation, and the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome is a crucial component of the innate immune system while the regulation of A2A R on it in the central nervous system (CNS) has not been clarified. METHODS: The effects of microglial A2A R on NLRP3 inflammasome assembly and activation were investigated in wild-type, A2A R- or NLRP3-knockout primary microglia with pharmacological treatment. Microglial A2A R or NLRP3 conditional knockout mice were used to interrogate the effects of this regulation on neuroinflammation posttraumatic brain injury (TBI). RESULTS: We found that A2A R directly interacted with NLRP3 and facilitated NLRP3 inflammasome assembly and activation in primary microglia while having no effects on mRNA levels of inflammasome components. Inhibition of the interaction via A2A R agonist or knockout attenuated inflammasome assembly and activation in vitro. In the TBI model, microglial A2A R and NLRP3 were co-expressed at high levels in microglia next to the peri-injured cortex, and abrogating of this interaction by microglial NLRP3 or A2A R conditional knockout attenuated the neurological deficits and neuropathology post-TBI via reducing the NLRP3 inflammasome activation. CONCLUSION: Our results demonstrated that inhibition of the interaction between A2A R and NLRP3 in microglia could mitigate the NLRP3 inflammasome assembly and activation and ameliorate the neuroinflammation post-TBI. It provides new insights into the effects of A2A R on neuroinflammation regulation post-TBI and offers a potential target for the treatment of NLRP3 inflammasome-related CNS diseases.


Assuntos
Lesões Encefálicas Traumáticas , Inflamassomos , Animais , Camundongos , Adenosina/metabolismo , Lesões Encefálicas Traumáticas/metabolismo , Inflamassomos/metabolismo , Camundongos Knockout , Microglia , Doenças Neuroinflamatórias , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
18.
BMC Cell Biol ; 14: 26, 2013 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-23711233

RESUMO

BACKGROUND: H19 is a paternally imprinted gene that has been shown to be highly expressed in the trophoblast tissue. Results from previous studies have initiated a debate as to whether noncoding RNA H19 acts as a tumor suppressor or as a tumor promotor in trophoblast tissue. In the present study, we developed lentiviral vectors expressing H19-specific small interfering RNA (siRNA) to specifically block the expression of H19 in the human choriocarcinoma cell line JAR. Using this approach, we investigated the impact of the H19 gene on the proliferation, invasion and apoptosis of JAR cells. Moreover, we examined the effect of H19 knockdown on the expression of insulin-like growth factor 2 (IGF2), hairy and enhancer of split homologue-1 (HES-1) and dual-specific phosphatase 5 (DUSP5) genes. RESULTS: H19 knockdown inhibited apoptosis and proliferation of JAR cells, but had no significant impact on cell invasion. In addition, H19 knockdown resulted in significant upregulation of HES-1 and DUSP5 expression, but not IGF2 expression in JAR cells. CONCLUSIONS: The finding that H19 downregulation could simultaneously inhibit proliferation and apoptosis of JAR cells highlights a putative dual function for H19 in choriocarcinoma and may explain the debate on whether H19 acts as a tumor suppressor or a tumor promotor in trophoblast tissue. Furthermore, upregulation of HES-1 and DUSP5 may mediate H19 downregulation-induced suppression of proliferation and apoptosis of JAR cells.


Assuntos
Apoptose/fisiologia , Proliferação de Células , Coriocarcinoma/patologia , Lentivirus/genética , Proteínas Nucleares/genética , Interferência de RNA/fisiologia , Proteínas Supressoras de Tumor/genética , Neoplasias Uterinas/patologia , Apoptose/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem Celular Tumoral , Coriocarcinoma/metabolismo , Fosfatases de Especificidade Dupla/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Vetores Genéticos/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Fator de Crescimento Insulin-Like II/metabolismo , Proteínas Nucleares/fisiologia , Gravidez , Fatores de Transcrição HES-1 , Proteínas Supressoras de Tumor/fisiologia , Neoplasias Uterinas/metabolismo
19.
Neurochem Res ; 38(7): 1365-74, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23579387

RESUMO

Nogo-A is a myelin-derived inhibitor playing a pivotal role in the prevention of axonal regeneration. A functional domain of Nogo-A, Amino-Nogo, exerts an inhibitory effect on axonal regeneration, although the mechanism is unclear. The present study investigated the role of the Amino-Nogo-integrin signaling pathway in primary retinal ganglion cells (RGCs) with respect to axonal outgrowth, which is required for axonal regeneration. Immunohistochemistry showed that integrin αv, integrin α5 and FAK were widely expressed in the visual system. Thy-1 and GAP-43 immunofluorescence showed that axonal outgrowth of RGCs was promoted by Nogo-A siRNA and a peptide antagonist of the Nogo-66 functional domain of Nogo-A (Nep1-40), and inhibited by a recombinant rat Nogo-A-Fc chimeric protein (Δ20). Western blotting revealed increased integrin αv and p-FAK expression in Nogo-A siRNA group, decreased integrin αv expression in Δ20 group and decreased p-FAK expression in Nep1-40 group. Integrin α5 expression was not changed in any group. RhoA G-LISA showed that RhoA activation was inhibited by Nogo-A siRNA and Δ20, but increased by Nep1-40 treatment. These results suggest that Amino-Nogo inhibits RGC axonal outgrowth primarily through the integrin αv signaling pathway.


Assuntos
Axônios , Proteínas da Mielina/metabolismo , Células Ganglionares da Retina/citologia , Transdução de Sinais , Animais , Sequência de Bases , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Técnicas de Silenciamento de Genes , Integrina alfa5/metabolismo , Proteínas da Mielina/genética , Proteínas Nogo , Nervo Óptico/enzimologia , Fosforilação , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Células Ganglionares da Retina/metabolismo , Córtex Visual/enzimologia , Proteína rhoA de Ligação ao GTP/metabolismo
20.
Int Rev Neurobiol ; 170: 225-265, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37741693

RESUMO

Accumulating evidence has revealed the adenosine 2A receptor is a key tuner for neuropathological and neurobehavioral changes following traumatic brain injury by experimental animal models and a few clinical trials. Here, we highlight recent data involving acute/sub-acute and chronic alterations of adenosine and adenosine 2A receptor-associated signaling in pathological conditions after trauma, with an emphasis of traumatic brain injury, including neuroinflammation, cognitive and psychiatric disorders, and other severe consequences. We expect this would lead to the development of therapeutic strategies for trauma-related disorders with novel mechanisms of action.


Assuntos
Lesões Encefálicas Traumáticas , Transtornos Mentais , Animais , Humanos , Adenosina , Modelos Animais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA