Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Adv Res ; 51: 73-90, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-36402285

RESUMO

INTRODUCTION: Spontaneous recovery after CNS injury is often very limited and incomplete, leaving most stroke patients with permanent disability. Maf1 is known as a key growth suppressor in proliferating cells. However, its role in neuronal cells after stroke remains unclear. OBJECTIVE: We aimed to investigate the mechanistic role of Maf1 in spontaneous neural repair and evaluated the therapeutic effect of targeting Maf1 on stroke recovery. METHODS: We used mouse primary neurons to determine the signaling mechanism of Maf1, and the cleavage-under-targets-and-tagmentation-sequencing to map the whole-genome promoter binding sites of Maf1 in isolated mature cortical neurons. Photothrombotic stroke model was used to determine the therapeutic effect on neural repair and functional recovery by AAV-mediated Maf1 knockdown. RESULTS: We found that Maf1 mediates mTOR signaling to regulate RNA polymerase III (Pol III)-dependent rRNA and tRNA transcription in mouse cortical neurons. mTOR regulates neuronal Maf1 phosphorylation and subcellular localization. Maf1 knockdown significantly increases Pol III transcription, neurite outgrowth and dendritic spine formation in neurons. Conversely, Maf1 overexpression suppresses such activities. In response to photothrombotic stroke in mice, Maf1 expression is increased and accumulates in the nucleus of neurons in the peripheral region of infarcted cortex, which is the key region for neural remodeling and repair during spontaneous recovery. Intriguingly, Maf1 knockdown in the peri-infarct cortex significantly enhances neural plasticity and functional recovery. Mechanistically, Maf1 not only interacts with the promoters and represses Pol III-transcribed genes, but also those of CREB-associated genes that are critical for promoting plasticity during neurodevelopment and neural repair. CONCLUSION: These findings indicate Maf1 as an intrinsic neural repair suppressor against regenerative capability of mature CNS neurons, and suggest that Maf1 is a potential therapeutic target for enhancing functional recovery after ischemic stroke and other CNS injuries.


Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Animais , Camundongos , Transcrição Gênica , Serina-Treonina Quinases TOR/genética , Fosforilação , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo
2.
Neuroimmunomodulation ; 29(4): 282-295, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35580556

RESUMO

Seizures are a very common manifestation of autoimmune encephalitis (AE), ranging from 33% to 100% depending on the antigen, most often accompanied by other clinical features such as behavioral changes, movement disorders, memory deficits, autoimmune disturbances, and altered levels of consciousness. Unusual seizure frequency, resistance to antiepileptic treatment, and often, definitive response to immunotherapy emphasize the importance for neurologists to consider the probable etiology of immune disorders. Studies on pathogenic mechanisms of autoantibodies have improved the understanding of different pathophysiologies and clinical characteristics of different AE groups. In encephalitis with antibodies to neuronal extracellular antigens, autoantibodies play a direct role in disease pathogenesis. They have access to target antigens and can potentially alter the structure and function of antigens but induce relatively little neuronal death. Prompt immunotherapy is usually very effective, and long-term antiepileptic treatment may not be needed. In contrast, in encephalitis with antibodies against intracellular antigens, autoantibodies may not be directly pathogenic but serve as tumor markers. These autoantibodies cannot reach intracellular target antigens and are considered to result from a T-cell-mediated immune response against antigens released by apoptotic tumor cells, which contain nerve tissue or express neuronal proteins. Neuronal loss is frequently described and predominantly induced through cytotoxic T-cell mechanisms. They often exhibit an inadequate response to immunotherapy and require early tumor treatment. Long-term antiepileptic treatment is usually needed. In conclusion, each neural autoantibody can specifically precipitate seizures. Early proper management of these cases may help prevent neurological deterioration and manage the occurrence of seizures. Consequently, confirmation of the presence of neuronal autoantibodies is strongly recommended even in patients with confirmed AE, as they are not only essential in achieving a good outcome but also may provide evidence for underlying neoplasia.


Assuntos
Doenças Autoimunes do Sistema Nervoso , Encefalite , Humanos , Anticonvulsivantes , Convulsões/etiologia , Convulsões/terapia , Autoanticorpos , Doenças Autoimunes do Sistema Nervoso/complicações , Doenças Autoimunes do Sistema Nervoso/terapia
3.
Front Pharmacol ; 13: 790136, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35418858

RESUMO

Epilepsy is a chronic neurological disorder. Current pharmacological therapies for epilepsy have limited efficacy that result in refractory epilepsy (RE). Owing to the limitations of conventional therapies, it is needed to develop new anti-epileptic drugs. The beneficial effects of polysaccharides from Chinese medicines, such as Lycium barbarum polysaccharides (COP) and Ganoderma lucidum polysaccharides (GLP), for treatment of epilepsy include regulation of inflammatory factors, neurotransmitters, ion channels, and antioxidant reactions. Especially, polysaccharides could be digested by intestinal microbial flora, referred as "intestinal brain organ" or "adult's second brain", may be the target for treatment of epilepsy. Actually, polysaccharides can effectively improve the type and quantity of intestinal flora such as bifidobacteria and lactic acid bacteria and achieve the purpose of treating epilepsy. Therefore, polysaccharides are hypothesized and discussed as potential agent for treatment of epilepsy.

4.
J Neurochem ; 159(4): 690-709, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34532857

RESUMO

After a sublethal ischemic preconditioning (IPC) stimulus, the brain has a remarkable capability of acquiring tolerance to subsequent ischemic insult by establishing precautionary self-protective mechanism. Understanding this endogenous mechanism would reveal novel and effective neuroprotective targets for ischemic brain injury. Our previous study has implied that telomerase reverse transcriptase (TERT) is associated with IPC-induced tolerance. Here, we investigated the mechanism of TERT-mediated ischemic tolerance. Preconditioning was modeled by oxygen-glucose deprivation (OGD) and by TERT inhibitor BIBR1532 in primary neurons. We found that ischemic tolerance was conferred by BIBR1532 preconditioning. We used the Cleavage-Under-Targets-And-Tagmentation approach, a recently developed method with superior signal-to-noise ratio, to comprehensively map the genomic binding sites of TERT in primary neurons, and showed that more than 50% of TERT-binding sites were located at the promoter regions. Mechanistically, we demonstrated that under normal conditions TERT physically bound to many previously unknown genomic loci in neurons, whereas BIBR1532 preconditioning significantly altered TERT-chromatin-binding profile. Intriguingly, we found that BIBR1532-preconditioned neurons showed significant up-regulation of promoter binding of TERT to the mitochondrial anti-oxidant genes, which were correlated with their elevated expression. Functional analysis further indicated that BIBR1532-preconditioning significantly reduced ROS levels and enhanced tolerance to severe ischemia-induced mitochondrial oxidative stress in neurons in a TERT-dependent manner. Together, these results demonstrate that BIBR1532 confers neuronal ischemic tolerance through TERT-mediated transcriptional reprogramming for up-regulation of mitochondrial anti-oxidation gene expression, suggesting the translational potential of BIBR1532 as a therapeutic agent for the treatment of cerebral ischemic injury and oxidative stress-induced neurological disorders.


Assuntos
Aminobenzoatos/uso terapêutico , Isquemia Encefálica/tratamento farmacológico , Naftalenos/uso terapêutico , Neurônios , Inibidores da Transcriptase Reversa/farmacologia , Telomerase/metabolismo , Animais , Antioxidantes/metabolismo , Sítios de Ligação/genética , Isquemia Encefálica/patologia , Cromatina/metabolismo , Mapeamento Cromossômico , Feminino , Técnicas de Silenciamento de Genes , Glucose/deficiência , Hipóxia , Precondicionamento Isquêmico , Camundongos , Camundongos Endogâmicos C57BL , Neuroproteção , Gravidez , Cultura Primária de Células , Espécies Reativas de Oxigênio , Razão Sinal-Ruído , Telomerase/antagonistas & inibidores , Telomerase/genética , Ativação Transcricional
5.
Front Neurosci ; 15: 655412, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33867929

RESUMO

Objective: Deep brain stimulation (DBS) has shown promising outcomes as new therapeutic opportunities for patients with treatment-resistant depression (TRD) who do not respond adequately to several consecutive treatments. This study aims to systematically review and conduct a meta-analysis on the efficacy and safety of DBS for TRD. Method: The literature was comprehensively reviewed using Medline, Google scholar, Cochrane library, Embase, and World Health Organization International Clinical Trials Registry Platform until January 2019. The studied outcomes included response, remission, recurrence, and adverse events (AEs) rates, and were reported as the rate ratio (RR) or pooled estimate with a 95% confidence interval (95% CI). Heterogeneity was measured by an I-square test and a sensitive analysis. Results: A total of 17 studies involving 7 DBS targets were included. For efficacy, DBS treatment was statistically beneficial for TRD, and the response, remission, and recurrence rates were 56% (ranging from 43 to 69%), 35% (ranging from 27 to 44%), and 14% (ranging from 4 to 25%), respectively. However, only two randomized-controlled trials (RCTs) considered the invalidity of DBS (RR = 1.45, 95% CI = 0.50-4.21). For safety, the AEs rate was 67% (ranging from 54 to 80%). The AEs were common and moderate, but the problems related to suicide and suicidal ideation should not be underestimated. Conclusion: These findings suggest that DBS for TRD is considered promising, which should be confirmed by well-designed and large sample studies. Future basic research and comprehensive clinical trials are needed to reach better understanding on the mechanisms of action and optimal targeted structure.

6.
Medicine (Baltimore) ; 100(15): e25314, 2021 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-33847630

RESUMO

BACKGROUND: Prolonged hospitalization and immobility of critical care patients elevate the risk of long-term physical and cognitive impairments. However, the therapeutic effects of early mobilization have been difficult to interpret due to variations in study populations, interventions, and outcome measures. We conducted a meta-analysis to assess the effects of early mobilization therapy on cardiac surgery patients in the intensive care unit (ICU). METHODS: PubMed, Excerpta Medica database (EMBASE), Cumulative Index of Nursing and Allied Health Literature (CINAHL), Physiotherapy Evidence Database (PEDro), and the Cochrane Library were comprehensively searched from their inception to September 2018. Randomized controlled trials were included if patients were adults (≥18 years) admitted to any ICU for cardiac surgery due to cardiovascular disease and who were treated with experimental physiotherapy initiated in the ICU (pre, post, or peri-operative). Data were extracted by 2 reviewers independently using a pre-constructed data extraction form. Length of ICU and hospital stay was evaluated as the primary outcomes. Physical function and adverse events were assessed as the secondary outcomes. Review Manager 5.3 (RevMan 5.3) was used for statistical analysis. For all dichotomous variables, relative risks or odds ratios with 95% confidence intervals (CI) were presented. For all continuous variables, mean differences (MDs) or standard MDs with 95% CIs were calculated. RESULTS: The 5 studies with a total of 652 patients were included in the data synthesis final meta-analysis. While a slight favorable effect was detected in 3 out of the 5 studies, the overall effects were not significant, even after adjusting for heterogeneity. CONCLUSIONS: This population-specific evaluation of the efficacy of early mobilization to reduce hospitalization duration suggests that intervention may not universally justify the labor barriers and resource costs in patients undergoing non-emergency cardiac surgery. PROSPERO RESEARCH REGISTRATION IDENTIFYING NUMBER: CRD42019135338.


Assuntos
Procedimentos Cirúrgicos Cardíacos/reabilitação , Deambulação Precoce/métodos , Deambulação Precoce/estatística & dados numéricos , Unidades de Terapia Intensiva/estatística & dados numéricos , Modalidades de Fisioterapia/estatística & dados numéricos , Humanos , Tempo de Internação/estatística & dados numéricos , Desempenho Físico Funcional , Ensaios Clínicos Controlados Aleatórios como Assunto
7.
Medicine (Baltimore) ; 99(4): e18843, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31977881

RESUMO

BACKGROUND: Prolonged hospitalization and immobility of critical care patients elevates the risk of long-term physical and cognitive impairments. However, the therapeutic effects of early mobilization have been difficult to interpret due to variations in study populations, interventions, and outcome measures. This systematic review and meta-analysis aims to assess the effects of early mobilization therapy for non-emergency cardiac surgery patients in the intensive care unit (ICU). METHODS: The following databases will be used to search for relevant keywords: PubMed, Embase, CINAHL, PEDro, and the Cochrane Library from inception to September 2018 by 2 researchers independently. Randomized controlled trials (RCTs), will be included if patients are adults (≥18 years) admitted to any ICU for cardiac surgery due to cardiovascular disease and who are treated with experimental physiotherapy initiated in the ICU (pre, post, or perioperative). The Review Manager 5.3 will be used for meta-analysis and the evidence level will be assessed by using the method for Grading of Recommendations Assessment, Development, and Evaluation (GRADE). Continuous outcomes will be presented as the weighted mean difference (WMD) or standardized mean difference (SMD) with 95% confidence interval (CI), while dichotomous data will be expressed as relative risk (RR) with 95% CI. If the included studies have existing heterogeneity (P < 0.1), a random-effects model will be used. Otherwise, we will calculate using a fixed effects model. RESULTS: This review will evaluate the effects of early mobilization on length of ICU and hospital stay, physical function and adverse events in patients with cardiac surgery patients in the ICU. CONCLUSION: This systematic review will comprehensively provide conclusive evidence of the therapeutic effect of early mobilization on cardiac surgery patients in the ICU.PROSPERO Research registration identifying number: CRD42019135338.


Assuntos
Procedimentos Cirúrgicos Cardíacos/reabilitação , Deambulação Precoce/métodos , Humanos , Unidades de Terapia Intensiva/organização & administração , Tempo de Internação , Metanálise em Rede , Revisões Sistemáticas como Assunto , Resultado do Tratamento
8.
Front Cell Neurosci ; 14: 540669, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33584203

RESUMO

Ischemic cerebral infarction represents a significant cause of disability and death worldwide. Caspase-1 is activated by the NLRP3/ASC pathway and inflammasomes, thus triggering pyroptosis, a programmed cell death. In particular, this death is mediated by gasdermin D (GSDMD), which induces secretion of interleukin (IL)-1ß and IL-18. Accordingly, inhibition of caspase-1 prevents the development and worsening of multiple neurodegenerative diseases. However, it is not clear whether inhibition of caspase-1 can preserve blood-brain barrier (BBB) integrity following cerebral infarction. This study therefore aimed at understanding the effect of caspase-1 on BBB dysfunction and its underlying mechanisms in permanent middle cerebral artery occlusion (MCAO). Our findings in rat models revealed that expression of caspase-1 was upregulated following MCAO-induced injury in rats. Consequently, pharmacologic inhibition of caspase-1 using vx-765 ameliorated ischemia-induced infarction, neurological deficits, and neuronal injury. Furthermore, inhibition of caspase-1 enhanced the encapsulation rate of pericytes at the ischemic edge, decreased leakage of both Evans Blue (EB) and matrix metalloproteinase (MMP) proteins, and upregulated the levels of tight junctions (TJs) and tissue inhibitors of metalloproteinases (TIMPs) in MCAO-injured rats. This in turn improved the permeability of the BBB. Meanwhile, vx-765 blocked the activation of ischemia-induced pyroptosis and reduced the expression level of inflammatory factors such as caspase-1, NLRP3, ASC, GSDMD, IL-1ß, and IL-18. Similarly, vx-765 treatment significantly reduced the expression levels of inflammation-related receptor for advanced glycation end products (RAGE), high-mobility family box 1 (HMGB1), mitogen-activated protein kinase (MAPK), and nuclear factor-κB (NF-κB). Evidently, inhibition of caspase-1 significantly improves ischemia-associated BBB permeability and integrity by suppressing pyroptosis activation and the RAGE/MAPK pathway.

9.
Neurosci Lett ; 690: 61-68, 2019 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-30300683

RESUMO

Neural stem cells (NSCs) as sources of new neurons in brain injuries or diseases are required to not only elicit neurons for neuronal repair, but also to enhance neurite outgrowth for neuronal network reestablishment. Various trophic or chemotropic factors have been shown to cooperatively improve NSC neurogenesis. However, effects of combined treatment of all-trans-retinoic acid (RA) with GF (Basic fibroblast growth factor and epidermal growth factor, bFGF/EGF) on neurogenesis of NSCs are poorly understood. To address this question, NSCs were isolated from the forebrains of embryonic mice, and treated with GF and RA either alone or in combination for differentiation in vitro. Neurons and astrocytes differentiated from NSCs were stained for MAP2 and GFAP separately by immunofluorescence. The results indicated that GF displayed superior efficacy in promoting neuronal differentiation, and RA showed better efficacy in advancing neurite outgrowth by increasing both neurite length and number. In addition, higher differentiation efficiency of neurons to astrocytes in RA or GF, or both acted at the early stage. However, more importantly, compared with RA alone, GF and RA in combination enhanced neuronal differentiation. Moreover, the combined use of GF and RA increased the length and number of neurites compared with GF, as well as the relative expression level of Smurf1. In addition, astrocytes induced by GF, RA, or both exhibited a radial glia-like morphology with long processes differing from serum effects, which might in part attribute to the total numbers of neurons. These findings for the first time unveil the roles of combined use of GF and RA on the neurogenesis of NSCs, suggesting that the use of this combination could be a comprehensive strategy for the functional repair of the nervous system through promoting neuronal differentiation, and advancing neurite outgrowth.


Assuntos
Fator de Crescimento Epidérmico/farmacologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Neuritos/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Tretinoína/farmacologia , Animais , Astrócitos/metabolismo , Células Cultivadas , Sinergismo Farmacológico , Fator 2 de Crescimento de Fibroblastos/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/metabolismo , Prosencéfalo , Ubiquitina-Proteína Ligases/metabolismo
10.
Oncotarget ; 8(55): 93516-93529, 2017 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-29212169

RESUMO

CCCTC-binding factor (CTCF) is an important epigenetic regulator implicated in multiple cellular processes, including growth, proliferation, differentiation, and apoptosis. Although CTCF deletion or mutation has been associated with human breast cancer, the role of CTCF in breast cancer is questionable. We investigated the biological functions of CTCF in breast cancer and the underlying mechanism. The results showed that CTCF expression in human breast cancer cells and tissues was significantly lower than that in normal breast cells and tissues. In addition, CTCF expression correlated significantly with cancer stage (P = 0.043) and pathological differentiation (P = 0.029). Furthermore, CTCF overexpression resulted in the inhibition of proliferation, migration, and invasion, while CTCF knockdown induced these processes in breast cancer cells. Transcriptome analysis and further experimental confirmation in MDA-MD-231 cells revealed that forced overexpression of CTCF might attenuate the DNA-binding ability of nuclear factor-kappaB (NF-κB) p65 subunit and inhibit activation of NF-κB and its target pro-oncogenes (tumor necrosis factor alpha-induced protein 3 [TNFAIP3]) and genes for growth-related proteins (early growth response protein 1 [EGR1] and growth arrest and DNA-damage-inducible alpha [GADD45a]). The present study provides a new insight into the tumor suppressor roles of CTCF in breast cancer development and suggests that the CTCF/NF-κB pathway is a potential target for breast cancer therapy.

11.
Sci Rep ; 7: 46204, 2017 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-28393844

RESUMO

Accumulating data from genome-wide association studies (GWAS) have provided a collection of novel candidate genes associated with complex diseases, such as atherosclerosis. We identified an atherosclerosis-associated single-nucleotide polymorphism (SNP) located in the intron of the long noncoding RNA (lncRNA) LINC00305 by searching the GWAS database. Although the function of LINC00305 is unknown, we found that LINC00305 expression is enriched in atherosclerotic plaques and monocytes. Overexpression of LINC00305 promoted the expression of inflammation-associated genes in THP-1 cells and reduced the expression of contractile markers in co-cultured human aortic smooth muscle cells (HASMCs). We showed that overexpression of LINC00305 activated nuclear factor-kappa beta (NF-κB) and that inhibition of NF-κB abolished LINC00305-mediated activation of cytokine expression. Mechanistically, LINC00305 interacted with lipocalin-1 interacting membrane receptor (LIMR), enhanced the interaction of LIMR and aryl-hydrocarbon receptor repressor (AHRR), and promoted protein expression as well as nuclear localization of AHRR. Moreover, LINC00305 activated NF-κB exclusively in the presence of LIMR and AHRR. In light of these findings, we propose that LINC00305 promotes monocyte inflammation by facilitating LIMR and AHRR cooperation and the AHRR activation, which eventually activates NF-κB, thereby inducing HASMC phenotype switching.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Inflamação/genética , Inflamação/patologia , Monócitos/metabolismo , Monócitos/patologia , NF-kappa B/metabolismo , RNA Longo não Codificante/metabolismo , Proteínas Repressoras/metabolismo , Aorta/patologia , Aterosclerose/genética , Aterosclerose/patologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem Celular , Núcleo Celular/metabolismo , Estudo de Associação Genômica Ampla , Humanos , Miócitos de Músculo Liso/metabolismo , Fenótipo , Transporte Proteico , RNA Longo não Codificante/genética , Receptores de Superfície Celular/metabolismo , Proteínas Repressoras/genética , Transdução de Sinais/genética , Regulação para Cima
12.
Eur Heart J ; 38(18): 1389-1398, 2017 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-27099261

RESUMO

AIMS: Oxidative stress contributes to the development of cardiac hypertrophy and heart failure. One of the mitochondrial sirtuins, Sirt4, is highly expressed in the heart, but its function remains unknown. The aim of the present study was to investigate the role of Sirt4 in the pathogenesis of pathological cardiac hypertrophy and the molecular mechanism by which Sirt4 regulates mitochondrial oxidative stress. METHODS AND RESULTS: Male C57BL/6 Sirt4 knockout mice, transgenic (Tg) mice exhibiting cardiac-specific overexpression of Sirt4 (Sirt4-Tg) and their respective controls were treated with angiotensin II (Ang II, 1.1 mg/kg/day). At 4 weeks, hypertrophic growth of cardiomyocytes, fibrosis and cardiac function were analysed. Sirt4 deficiency conferred resistance to Ang II infusion by significantly suppressing hypertrophic growth, and the deposition of fibrosis. In Sirt4-Tg mice, aggravated hypertrophy and reduced cardiac function were observed compared with non-Tg mice following Ang II treatment. Mechanistically, Sirt4 inhibited the binding of manganese superoxide dismutase (MnSOD) to Sirt3, another member of the mitochondrial sirtuins, and increased MnSOD acetylation levels to reduce its activity, resulting in elevated reactive oxygen species (ROS) accumulation upon Ang II stimulation. Furthermore, inhibition of ROS with manganese 5, 10, 15, 20-tetrakis-(4-benzoic acid) porphyrin, a mimetic of SOD, blocked the Sirt4-mediated aggravation of the hypertrophic response in Ang II-treated Sirt4-Tg mice. CONCLUSIONS: Sirt4 promotes hypertrophic growth, the generation of fibrosis and cardiac dysfunction by increasing ROS levels upon pathological stimulation. These findings reveal a role of Sirt4 in pathological cardiac hypertrophy, providing a new potential therapeutic strategy for this disease.


Assuntos
Cardiomegalia/enzimologia , Proteínas Mitocondriais/fisiologia , Sirtuínas/fisiologia , Superóxido Dismutase/antagonistas & inibidores , Angiotensina II/farmacologia , Animais , Técnicas de Silenciamento de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias Cardíacas/enzimologia , Miócitos Cardíacos/enzimologia , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/antagonistas & inibidores , Remodelação Vascular/fisiologia , Vasoconstritores/farmacologia
13.
Sci China Life Sci ; 59(10): 1024-1033, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27614752

RESUMO

Safe and efficient gene transfer systems are the basis of gene therapy applications. Non-integrating lentiviral (NIL) vectors are among the most promising candidates for gene transfer tools, because they exhibit high transfer efficiency in both dividing and non-dividing cells and do not present a risk of insertional mutagenesis. However, non-integrating lentiviral vectors cannot introduce stable exogenous gene expression to dividing cells, thereby limiting their application. Here, we report the design of a non-integrating lentiviral vector that contains the minimal scaffold/matrix attachment region (S/MAR) sequence (SNIL), and this SNIL vector is able to retain episomal transgene expression in dividing cells. Using SNIL vectors, we detected the expression of the eGFP gene for 61 days in SNIL-transduced stable CHO cells, either with selection or not. In the NIL group without the S/MAR sequence, however, the transduced cells died under selection for the transient expression of NIL vectors. Furthermore, Southern blot assays demonstrated that the SNIL vectors were retained extrachromosomally in the CHO cells. In conclusion, the minimal S/MAR sequence retained the non-integrating lentiviral vectors in dividing cells, which indicates that SNIL vectors have the potential for use as a gene transfer tool.


Assuntos
Expressão Gênica , Lentivirus/genética , Regiões de Interação com a Matriz/genética , Transgenes/genética , Animais , Células CHO , Divisão Celular/genética , Cricetinae , Cricetulus , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Plasmídeos/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Reprodutibilidade dos Testes , Transfecção/métodos
14.
Exp Ther Med ; 9(5): 1962-1966, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-26136923

RESUMO

The aim of this study was to investigate the method and significance of the application of direct electrical stimulation (DES) to the brain mapping of language functions during glioma surgery. A retrospective analysis of clinical data was performed for 91 cases of brain functional area glioma surgery under DES from January 2003 until January 2012. Following cortical electrical stimulation, 88 patients exhibited seizures involving facial or hand movements and 91 cases experienced language disorders such as counting interruption, naming errors or anomia. The most commonly observed areas of counting interruption were distributed on the posterior part of the left anterior central gyrus (47.7%), the operculum of the left inferior frontal gyrus (24.4%) and the triangular part of the left inferior frontal gyrus (12.8%). Postoperative magnetic resonance imaging demonstrated that overall excision was achieved in 53 cases and sub-overall excision was performed in 31 cases. A total of 42 cases (46.2%) exhibited no postoperative neurological dysfunction, 39 cases (42.9%) exhibited brief language dysfunction, 27 cases (29.7%) experienced brief limb movement disorder, and one case appeared to have permanent neurological dysfunction. DES was indicated to be a reliable and noninvasive method for the intraoperative positioning of language areas, and was able to resect gliomas in the language area with maximal safety.

15.
Sci China Life Sci ; 57(5): 488-94, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24740453

RESUMO

Studies on the chaperone protein α-hemoglobin stabilizing protein (AHSP) reveal that abundant AHSP in erythroid cells enhance the cells' tolerance to oxidative stress imposed by excess α-hemoglobin in pathological conditions. However, the potential intracellular modulation of AHSP expression itself in response to oxidative stress is still unknown. The present study examined the effect and molecular mechanism of STAT3, an oxidative regulator, on the expression of AHSP. AHSP expression increased in K562 cells upon cytokine IL-6-induced STAT3 activation and decreased in STAT3 knock-down K562 cells. Regulation of AHSP in oxidative circumstance was then examined in α-globin-overloaded K562 cells, and real-time PCR showed strengthened expression of both AHSP and STAT3. ChIP analysis showed binding of STAT3 to AHSP promoter and binding was significantly augmented with IL6 stimulation and upon α-globin overexpression. Dual luciferase reporter assays of the wildtype and mutated SB3 element, an IL-6RE site, in the AHSP promoter in K562 cells highlighted the direct regulatory effect of STAT3 on AHSP gene. Finally, direct binding of STAT3 to SB3 site of AHSP promoter was confirmed with EMSA assays. Our work reveals an adaptive AHSP regulation mediated by the redox-sensitive STAT3 signaling pathway, and provides clues to the therapeutic strategy for AHSP enhancement.


Assuntos
Proteínas Sanguíneas/genética , Chaperonas Moleculares/genética , Fator de Transcrição STAT3/metabolismo , Sítios de Ligação/genética , Humanos , Interleucina-6/metabolismo , Células K562 , Regiões Promotoras Genéticas , Ligação Proteica , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Regulação para Cima , alfa-Globinas/genética
16.
Blood ; 123(2): 261-70, 2014 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-24255919

RESUMO

Promyelocytic leukemia protein (PML) has been implicated as a participant in multiple cellular processes including senescence, apoptosis, proliferation, and differentiation. Studies of PML function in hematopoietic differentiation previously focused principally on its myeloid activities and also indicated that PML is involved in erythroid colony formation. However, the exact role that PML plays in erythropoiesis is essentially unknown. In this report, we found that PML4, a specific PML isoform expressed in erythroid cells, promotes endogenous erythroid genes expression in K562 and primary human erythroid cells. We show that the PML4 effect is GATA binding protein 1 (GATA-1) dependent using GATA-1 knockout/rescued G1E/G1E-ER4 cells. PML4, but not other detected PML isoforms, directly interacts with GATA-1 and can recruit it into PML nuclear bodies. Furthermore, PML4 facilitates GATA-1 trans-activation activity in an interaction-dependent manner. Finally, we present evidence that PML4 enhances GATA-1 occupancy within the globin gene cluster and stimulates cooperation between GATA-1 and its coactivator p300. These results demonstrate that PML4 is an important regulator of GATA-1 and participates in erythroid differention by enhancing GATA-1 trans-activation activity.


Assuntos
Diferenciação Celular/fisiologia , Células Eritroides/citologia , Células Eritroides/metabolismo , Fator de Transcrição GATA1/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Proteínas Supressoras de Tumor/metabolismo , Acetilação , Proteína p300 Associada a E1A/metabolismo , Fator de Transcrição GATA1/química , Fator de Transcrição GATA1/metabolismo , Expressão Gênica , Humanos , Células K562 , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteína da Leucemia Promielocítica , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/genética , Transcrição Gênica , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/genética , Dedos de Zinco
17.
J Biol Chem ; 287(36): 30641-52, 2012 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-22825848

RESUMO

Matrix attachment region (MAR)-binding protein (MARBP) has profound influence on gene transcriptional control by tethering genes to the nuclear scaffold. MARBP SATB2 is recently known as a versatile regulator functioning in the differentiation of multiple cell types including embryonic stem cells, osteoblasts and immunocytes. Roles of SATB2 in erythroid cells and its working mechanism in orchestrating target gene expression are largely unexplored. We show here that SATB2 is expressed in erythroid cells and activates γ-globin genes by binding to MARs in their promoters and recruiting histone acetylase PCAF. Further analysis in higher-order chromatin structure shows that SATB2 affects physical proximity of human (G)γ- and (A)γ-globin promoters via self-association. We also found that SATB2 interacts with SATB1, which specifically activates ε-globin gene expression. Our results establish SATB2 as a novel γ-globin gene regulator and provide a glimpse of the differential and cooperative roles of SATB family proteins in modulating clustered genes transcription and mediating higher-order chromatin structures.


Assuntos
Células Eritroides/metabolismo , Regulação da Expressão Gênica/fisiologia , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Regiões de Interação com a Matriz/fisiologia , Família Multigênica/fisiologia , Fatores de Transcrição/metabolismo , gama-Globinas/biossíntese , Animais , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células Eritroides/citologia , Humanos , Células K562 , Proteínas de Ligação à Região de Interação com a Matriz/genética , Camundongos , Fatores de Transcrição/genética , Transcrição Gênica/fisiologia , gama-Globinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...