Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Int J Mol Sci ; 24(13)2023 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-37445774

RESUMO

Repetitive low-level blast (rLLB) exposure is a potential risk factor for the health of soldiers or workers who are exposed to it as an occupational characteristic. Alveolar macrophages (AMs) are susceptible to external blast waves and produce pro-inflammatory or anti-inflammatory effects. However, the effect of rLLB exposure on AMs is still unclear. Here, we generated rLLB waves through a miniature manual Reddy-tube and explored their effects on MH-S cell morphology, phenotype transformation, oxidative stress status, and apoptosis by immunofluorescence, real-time quantitative PCR (qPCR), western blotting (WB) and flow cytometry. Ipatasertib (GDC-0068) or PDTC was used to verify the role of the Akt/NF-κB signaling pathway in these processes. Results showed that rLLB treatment could cause morphological irregularities and cytoskeletal disorders in MH-S cells and promote their polarization to the M1 phenotype by increasing iNOS, CD86 and IL-6 expression. The molecular mechanism is through the Akt/NF-κB signaling pathway. Moreover, we found reactive oxygen species (ROS) burst, Ca2+ accumulation, mitochondrial membrane potential reduction, and early apoptosis of MH-S cells. Taken together, our findings suggest rLLB exposure may cause M1 polarization and early apoptosis of AMs. Fortunately, it is blocked by specific inhibitors GDC-0068 or PDTC. This study provides a new treatment strategy for preventing and alleviating health damage in the occupational population caused by rLLB exposure.


Assuntos
Macrófagos Alveolares , NF-kappa B , Camundongos , Animais , NF-kappa B/metabolismo , Macrófagos Alveolares/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
2.
Eur J Trauma Emerg Surg ; 48(1): 455-470, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32488449

RESUMO

BACKGROUND: Crush injury/crush syndrome (CI/CS) is the second most common cause of death during earthquakes. Most studies of CI/CS have mainly focused on kidney injury after decompression. Few studies have focused on myocardial injury caused by crush injury and its potential mechanisms. METHODS: We first verified cardiomyocyte injury during compression in rats with a crush injury. The survival rate, electrocardiographic results, histological results, catecholamine changes and cardiac ß1-AR expression were evaluated. Next, we explored the effects of pretreatment with a selective ß1-blocker (bisoprolol) with or without fluid resuscitation on rats with a crush injury. In addition to evaluating the survival rates, biochemical and histological analyses and echocardiographic measurements were also performed. RESULTS: Reduced heart rates, elevated ST segments, and tall-peaked T waves were observed in the rats with a crush injury. The changes in the myocardial enzymes and pathological results demonstrated that myocardial damage occurred during compression in rats with a crush injury. The levels of the catecholamine norepinephrine in both the serum and myocardial tissue were elevated during compression. Pretreatment with a selective ß1-blocker combined with fluid resuscitation significantly improved the survival rates of the rats with lethal crush injury. The myocardial enzymes and pathological results showed that the combined therapy decreased myocardial damage. The echocardiography measurements showed that the rats that received the combined therapy exhibited decreased left ventricular mass (LVM), left ventricular volume at end-systole (LVVs) and left ventricular internal diameter (LVID) compared with the rats with a crush injury. CONCLUSIONS: Our findings demonstrated the presence of myocardial injury in the early stage of compression in rats with a crush injury. Pretreatment with a ß1-blocker (bisoprolol) with fluid resuscitation significantly reduced mortality, decreased myocardial tissue damage, and improved ventricular remodelling in rats with a lethal crush injury.


Assuntos
Síndrome de Esmagamento , Remodelação Ventricular , Animais , Eletrocardiografia , Ventrículos do Coração , Miocárdio , Ratos
3.
Mil Med Res ; 8(1): 37, 2021 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-34148549

RESUMO

BACKGROUND: Acute kidney injury (AKI) is the main life-threatening complication of crush syndrome (CS), and myoglobin is accepted as the main pathogenic factor. The pattern recognition receptor retinoicacid-inducible gene I (RIG-I) has been reported to exert anti-viral effects function in the innate immune response. However, it is not clear whether RIG-I plays a role in CS-AKI. The present research was carried out to explore the role of RIG-I in CS-AKI. METHODS: Sprague-Dawley rats were randomly divided into two groups: the sham and CS groups (n = 12). After administration of anesthesia, the double hind limbs of rats in the CS group were put under a pressure of 3 kg for 16 h to mimic crush conditions. The rats in both groups were denied access to food and water. Rats were sacrificed at 12 h or 36 h after pressure was relieved. The successful establishment of the CS-AKI model was confirmed by serum biochemical analysis and renal histological examination. In addition, RNA sequencing was performed on rat kidney tissue to identify molecular pathways involved in CS-AKI. Furthermore, NRK-52E cells were treated with 200 µmol/L ferrous myoglobin to mimic CS-AKI at the cellular level. The cells and cell supernatant samples were collected at 6 h or 24 h. Small interfering RNAs (siRNA) was used to knock down RIG-I expression. The relative expression levels of molecules involved in the RIG-I pathway in rat kidney or cells samples were measured by quantitative Real-time PCR (qPCR), Western blotting analysis, and immunohistochemistry (IHC) staining. Tumor necrosis factor-α (TNF-α) was detected by ELISA. Co-Immunoprecipitation (Co-IP) assays were used to detect the interaction between RIG-I and myoglobin. RESULTS: RNA sequencing of CS-AKI rat kidney tissue revealed that the different expression of RIG-I signaling pathway. qPCR, Western blotting, and IHC assays showed that RIG-I, nuclear factor kappa-B (NF-κB) P65, p-P65, and the apoptotic marker caspase-3 and cleaved caspase-3 were up-regulated in the CS group (P < 0.05). However, the levels of interferon regulatory factor 3 (IRF3), p-IRF3 and the antiviral factor interferon-beta (IFN-ß) showed no significant changes between the sham and CS groups. Co-IP assays showed the interaction between RIG-I and myoglobin in the kidneys of the CS group. Depletion of RIG-I could alleviate the myoglobin induced expression of apoptosis-associated molecules via the NF-κB/caspase-3 axis. CONCLUSION: RIG-I is a novel damage-associated molecular patterns (DAMPs) sensor for myoglobin and participates in the NF-κB/caspase-3 signaling pathway in CS-AKI. In the development of CS-AKI, specific intervention in the RIG-I pathway might be a potential therapeutic strategy for CS-AKI.


Assuntos
Caspase 3/efeitos dos fármacos , NF-kappa B/efeitos dos fármacos , RNA Helicases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/fisiopatologia , Alarminas , Animais , China , Síndrome de Esmagamento/sangue , Síndrome de Esmagamento/complicações , Modelos Animais de Doenças , Masculino , Mioglobina/farmacologia , Mioglobina/uso terapêutico , RNA Helicases/uso terapêutico , Ratos , Ratos Sprague-Dawley
4.
Mol Med Rep ; 23(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33880578

RESUMO

Acute kidney injury (AKI) has become a global public health problem with high morbidity and mortality rates, as well as high healthcare costs. Immune cells, particularly macrophages, which regulate tissue development, destroy pathogens, control homeostasis and repair wounds, play crucial and complex roles in AKI. In various types of AKI, numerous rapidly recruited monocytes and tissue­resident macrophages act in a coordinated manner. Thus, elucidating the phenotypic and functional characteristics of macrophages in AKI is essential for identifying potential therapeutic targets. Macrophage­sensing mediators and macrophage­derived mediators participate in the major macrophage­related signaling pathways in AKI, which regulate macrophage polarization and determine disease progression. In conclusion, macrophages change their roles and regulatory mechanisms during the occurrence and development of AKI. The aim of the present review was to contribute to an improved understanding of AKI and to the identification of novel therapeutic targets for this condition.


Assuntos
Injúria Renal Aguda/metabolismo , Macrófagos/metabolismo , Transdução de Sinais , Animais , Citocinas , Homeostase , Humanos , Rim/metabolismo , Ativação de Macrófagos , Monócitos/metabolismo
5.
Int J Mol Sci ; 21(17)2020 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-32878118

RESUMO

Primary blast lung injury (PBLI) is a common cause of casualties in wars, terrorist attacks, and explosions. It can exist in the absence of any other outward signs of trauma, and further develop into acute lung injury (ALI) or a more severe acute respiratory distress syndrome (ARDS). The pathogenesis of PBLI at the cellular and molecular level has not been clear. Damage-associated molecular pattern (DAMP) is a general term for endogenous danger signals released by the body after injury, including intracellular protein molecules (HMGB1, histones, s100s, heat shock proteins, eCIRP, etc.), secretory protein factors (IL-1ß, IL-6, IL-10, TNF-α, VEGF, complements, etc.), purines and pyrimidines and their derived degradation products (nucleic acids, ATP, ADP, UDPG, uric acid, etc.), and extracellular matrix components (hyaluronic acid, fibronectin, heparin sulfate, biglycan, etc.). DAMPs can be detected by multiple receptors including pattern recognition receptors (PRRs). The study of DAMPs and their related signaling pathways, such as the mtDNA-triggered cGAS-YAP pathway, contributes to revealing the molecular mechanism of PBLI, and provides new therapeutic targets for controlling inflammatory diseases and alleviating their symptoms. In this review, we focus on the recent progress of research on DAMPs and their signaling pathways, as well as the potential therapeutic targets and future research directions in PBLI.


Assuntos
Alarminas/metabolismo , Traumatismos por Explosões/patologia , Lesão Pulmonar/patologia , Animais , Traumatismos por Explosões/metabolismo , Humanos , Lesão Pulmonar/metabolismo , Transdução de Sinais
6.
Neurosci Lett ; 714: 134617, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31705924

RESUMO

Magnetic resonance molecular imaging, as a safe imaging technology, provides a new idea for the early qualitative and hierarchical diagnosis of gliomas. The purpose of this study was to design and evaluate the value of neuropilin-1 (NRP-1) targeting molecular probes in the hierarchical diagnosis of gliomas. First, we created an NRP-1 targeted magnetic resonance molecular probe (USPIO-PEG-tLyP-1) by combining the polypeptide tLyP-1 with ultra-small superparamagnetic iron oxide nanoparticles (USPIONs), detecting the physical properties by transmission electron microscopy (TEM) and dynamic light scattering (DLS). Second, in vivo experiments, we established two different degrees of malignant gliomas in-situ in nude mice by injecting U87 and CHG-5 cells. Then, to detect the binding ability of the probe with different grades of tumour tissues, we injected the probe into the tumour-bearing mice through the tail vein. Next, MRI was performed before injection, and 6 h, 12 h, 24 h after injection, and we found significantly more iron particles in the tumour tissues of U87 tumour-bearing mice than in tumour tissues of CHG-5 tumour-bearing mice. The signal intensities of the T2-weighted images of the tumour tissues of each group as well as microscopic observations by Prussian blue staining indicated that the binding ability of this molecular probe to U87 glioma (HGG) with high NRP-1 expression was significantly greater than that of CHG-5 glioma (LGG) with low NRP-1 expression (P < 0.01). Therefore, this study confirms that the novel molecular probe USPIO-PEG-tLyP-1 can be used for the grading diagnosis by MRI for gliomas of high and low grade with different NRP-1 expression levels.


Assuntos
Meios de Contraste , Dextranos , Glioma/diagnóstico por imagem , Nanopartículas de Magnetita , Neuropilina-1/metabolismo , Peptídeos Cíclicos , Polietilenoglicóis , Animais , Linhagem Celular Tumoral , Peptídeos Penetradores de Células , Dextranos/ultraestrutura , Difusão Dinâmica da Luz , Glioma/metabolismo , Glioma/patologia , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita/ultraestrutura , Camundongos , Camundongos Nus , Microscopia Eletrônica de Transmissão , Sondas Moleculares/ultraestrutura , Gradação de Tumores , Interferência de RNA , Transfecção
7.
Oncol Lett ; 17(6): 5821-5829, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31186810

RESUMO

Nervous system polycomb 1 (NSPc1) is a member of the polycomb group (PcG) family of proteins and has been demonstrated to maintain the differentiation and pluripotency of stem cells. Long non-coding RNAs (lncRNAs) have been demonstrated to be involved in the control of pluripotency and differentiation in embryonic and pluripotent cells. In the present study, the expression levels of NSPc1 were associated with the malignant potential of various glioma cell lines. Additionally, lncRNAs were differentially expressed in glioblastoma cell lines. Following induced differentiation of U87 glioblastoma cells with all-trans retinoic acid, the expression levels of NSPc1 decreased initially, reaching its lowest point on day 6, but then subsequently increased until day 10. The expression of lncRNA candidates decreased in the cell differentiation stage. Additionally, the expression of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), sex-determining region of the Y chromosome-box 2 overlapping transcript (SOX2OT) and antisense non-coding RNA in the INK4 locus (ANRIL) was significantly altered relative to the expression levels of NSPc1. RNA immunoprecipitation (RIP) assays demonstrated that MALAT1, SOX2OT and ANRIL bind to NSPc1 in U87 glioblastoma cells and the enrichment of ANRIL in anti-NSPc1 antibody group was associated with the expression levels of NSPc1 during U87 cell differentiation. Small interfering RNA mediated downregulation of NSPc1 expression with MALAT1, SOX2OT and ANRIL, inhibited the proliferation, and promoted apoptosis in U87 cells. The results of the present study demonstrate that MALAT1, SOX2OT and ANRIL combine and crosstalk with NSPc1 in U87 cells to affect proliferation and apoptosis.

8.
Oncol Rep ; 41(4): 2575-2584, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30720120

RESUMO

Recently, emerging evidence shows that a number of long non­coding RNAs (lncRNAs) recruit polycomb group (PcG) proteins to specific chromatin loci to silence relevant gene expression. In the present study, we provided evidence that lncRNA candidates, selected by bioinformatic analysis and nervous system polycomb 1 (NSPc1), a key polycomb repressive complex 1 (PRC1) member, were highly expressed in glioma H4 cells in contrast to that noted in non­cancerous cells. RNA binding protein immunoprecipitation (RIP) assays demonstrated that metastasis associated lung adenocarcinoma transcript 1 (MALAT1), SOX2 overlapping transcript (SOX2OT) and maternally expressed 3 (MEG3) among the 8 candidates bound to the NSPc1 protein complex in glioma H4 cells. Furthermore, overexpression of NSPc1 caused a decrease in the expression of MALAT1 and MEG3 and increased expression of SOX2OT, while NSPc1 downregulation caused the levels of all three genes to increase. Meanwhile, suppression of the expression of MALAT1 increased the expression levels of mRNA and protein of NSPc1, whereas downregulation of the expression of SOX2OT decreased NSPc1 expression. Moreover, a significant decrease in cell growth and increased cell apoptosis were observed in the transfected H4 cells by MTT assay and flow cytometric analysis. The results showed that the reduced co­expression between NSPc1 and MALAT1/SOX2OT decreased the proliferation and promoted the death of H4 cells more obviously than the respectively decrease in expression of NSPc1, MALAT1 and SOX2OT. Remarkably, the influence of a simultaneously decreased expression of NSPc1 and SOX2OT on promoting cell apoptosis was more obvious than the total effect of the separate downregulation of NSPc1 and SOX2OT on accelerating cell death. However, that impact was partially counteracted in the silencing of the co­expression of MALAT1 and NSPc1. Furthermore, they cooperated to affect transcription of p21 and OCT4.Briefly, these data suggest NSPc1 polycomb protein complex binding and cross­talk to lncRNAs in glioma H4 cells, offering new insight into the important function of polycomb protein complex and lncRNA interactions in glioma cells and provide a novel view of potential biomarkers and targets for the diagnosis and therapy of glioma.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glioma/genética , Complexo Repressor Polycomb 1/metabolismo , RNA Longo não Codificante/metabolismo , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação para Baixo , Glioma/patologia , Humanos , Complexo Repressor Polycomb 1/genética , RNA Longo não Codificante/genética , RNA Interferente Pequeno/metabolismo
9.
Neurosci Lett ; 656: 1-8, 2017 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-28694091

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal disease that selectively involves motor neurons. Neurotrophic factor supplementation and neural stem cell (NSC) alternative therapy have been used to treat ALS. The two approaches can affect each other in their pathways of action, and there is a possibility for synergism. However, to date, there have been no studies demonstrating the effects of combined therapy in the treatment of ALS. In this study, for the first time, we adopted a method involving the intranasal administration of nerve growth factor combined with lateral ventricle NSC transplantation using G93A-SOD1 transgenic mice as experimental subjects to explore the treatment effect of this combined therapy in ALS. We discover that the combined therapy increase the quantity of TrkA receptors, broaden the migration of exogenous NSCs, further promote active proliferation in neurogenic regions of the brain and enhance the preservation of motor neurons in the spinal cord. Regarding physical activity, the combined therapy improved motor functions, further postponed ALS onset and extended the survival time of the mice.


Assuntos
Esclerose Lateral Amiotrófica/terapia , Fator de Crescimento Neural/uso terapêutico , Células-Tronco Neurais/transplante , Administração Intranasal , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Proliferação de Células , Terapia Combinada , Ventrículos Laterais/citologia , Camundongos Transgênicos , Neurônios Motores/patologia , Fator de Crescimento Neural/metabolismo , Receptor trkA/metabolismo , Medula Espinal/patologia , Superóxido Dismutase/genética
10.
Biochem Biophys Res Commun ; 440(4): 527-32, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24113379

RESUMO

Nspc1 is an identified transcription repressor. However, transiently up-regulated or down-regulated Nspc1 in P19 embryonal carcinoma cells affects expression levels of Oct4, Sox2 and Nanog in a positive correlation. Luciferase activity assays verified that Nspc1 regulates the Oct4 promoter in a dose dependent manner. ChIP assay shows that Nspc1 activates Oct4 by directly binding to the (-1021 to -784) region of Oct4 promoter. Dominant negative analysis indicated the activation is dependent on the retinoid acid response element (RARE). We demonstrated Nspc1 has a positive role in maintaining the pluripotency of P19 cells by directly regulating Oct4.


Assuntos
Proteínas de Homeodomínio/genética , Neurogênese/genética , Fator 3 de Transcrição de Octâmero/genética , Células-Tronco Pluripotentes/citologia , Complexo Repressor Polycomb 1/metabolismo , Fatores de Transcrição SOXB1/genética , Ativação Transcricional , Animais , Linhagem Celular Tumoral , Camundongos , Proteína Homeobox Nanog , Células-Tronco Pluripotentes/metabolismo , Complexo Repressor Polycomb 1/genética , Regiões Promotoras Genéticas
11.
J Thromb Thrombolysis ; 36(3): 317-23, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23271305

RESUMO

Polymorphisms of the upstream stimulatory factor 1 (USF1) have been associated with carotid artery intima-media thickness and coronary atherosclerotic lesions. Unstable carotid plaque is an atherosclerotic change of vascular morphology that has been correlated with cerebrovascular ischemic symptoms. Associations of three single nucleotide polymorphisms of the USF1 gene with total unstable carotid plaque area (CPA) were investigated in Chinese atherosclerotic stroke patients. We recruited 668 atherosclerotic stroke patients and 602 controls. Total unstable CPA values were measured by ultrasound. Genotypes were analyzed using polymerase chain reaction-based restriction fragment length polymorphism (PCR-RFLP) or mismatched PCR-RFLP. A significant difference in total unstable CPA was found for rs2516838 and rs2516839 genotypes (P = 0.039 and 0.046, respectively) in atherosclerotic stroke patients with unstable carotid plaque. Furthermore, in multiple logistic regression analysis adjusted by age, sex, BMI, hypertension, smoking status, glucose, total cholesterol, triglycerides, high-density lipoprotein-cholesterols, low-density lipoprotein-cholesterols and high-sensitivity C-reactive protein, significant associations were seen between the total unstable CPA values and genotypes of the rs2516838 or the rs2516839 in these patients. The rare allele C of rs2516838 or rare allele A of rs2516839 could predict relative low total unstable CPA values. The rs2516838 and rs2516839 polymorphisms of USF1 influence total unstable CPA in atherosclerotic stroke patients, which might be new markers to predict the risk of recurrence for this disease.


Assuntos
Doenças das Artérias Carótidas/genética , Arteriosclerose Intracraniana/genética , Placa Aterosclerótica/genética , Polimorfismo de Fragmento de Restrição , Polimorfismo de Nucleotídeo Único , Acidente Vascular Cerebral/genética , Fatores Estimuladores Upstream/genética , Idoso , Doenças das Artérias Carótidas/metabolismo , Feminino , Humanos , Arteriosclerose Intracraniana/metabolismo , Masculino , Pessoa de Meia-Idade , Placa Aterosclerótica/metabolismo , Acidente Vascular Cerebral/metabolismo , Fatores Estimuladores Upstream/metabolismo
12.
Neurobiol Aging ; 33(3): 522-34, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20619502

RESUMO

Alzheimer's disease (AD) is a progressive neurodegenerative disorder mainly characterized by amyloid-beta (Aß) deposition and neurofibrillary tangles (NFTs). The abnormal enrichment of amyloid protein precursor (APP) leads to a high risk of AD. One of the plausible age-associated AD animal models, senescence-accelerated mouse prone 8 (SAMP8), have age-related learning and memory deficits. We found APP protein significantly increased in the hippocampus of aged SAMP8 mice. The 20 to 25 nucleotide (nt) tiny regulators, known as micro ribonucleic acids (miRNAs), have been found to play crucial roles in neurodegenerative diseases. Here, we examined the post-transcriptional regulation mechanism of APP mediated by micro ribonucleic acids and found that miR-16 was one of the post-transcriptional regulators of APP in SAMP8 mice. Overexpression of miR-16, both in vitro and in vivo, led to reduced APP protein expression. Furthermore, miR-16 and APP displayed complementary expression patterns in SAMP8 mice and BALb/c mice embryos. Taken together, these findings demonstrate that APP is a target of miR-16 and the abnormally low expression of miR-16 could potentially lead to APP protein accumulation in AD mice.


Assuntos
Senilidade Prematura/genética , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/metabolismo , MicroRNAs/genética , Senilidade Prematura/metabolismo , Doença de Alzheimer/etiologia , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/fisiologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes Neurológicos , Células NIH 3T3 , Neuroblastoma/genética
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 27(1): 71-3, 2011 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-21208569

RESUMO

AIM: To express the human recombinant PCGF1 protein and prepare monoclonal antibody (mAb) against it. METHODS: The recombinant expression plasmid pET32a-His-PCGF1-128/189 was made and transformed into E.coli (BL21), and then the recombinant fusion protein His-PCGF1-128/189 was expressed and purified. The BALB/c mice were immuned with purified protein His-PCGF1-128/189 as antigen. The mAbs against PCGF1 were prepared by using standard hybridoma technique. The hybridoma cell lines were obtained by ELISA and Western blot screening procedure, the isotype of the mAbs were further identified by immune-double diffusion. Ascites were prepared from one propagated hybridoma cell line and mAbs were purified by using the Kit from Millipore. The valence of mAb was detected by Western blot. RESULTS: The recombinant protein His-PCGF1-128/189 was expressed and purified. Two hybfidmas producing antibodies against PCGF1 were obtained, the isotypes of two mAbs were IgG1, Western blot showed that the antibodies were high sensitive(1:6 000) and high specific for PCGF1. CONCLUSION: The anti-PCGF1 mAb prepared by using recombinant His-PCGF1-128/189 protein as antigen can be used for detecting PCGF1 proteins which are either endogenous or exogenous.


Assuntos
Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/imunologia , Proteínas Repressoras/biossíntese , Proteínas Repressoras/imunologia , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/isolamento & purificação , Western Blotting/métodos , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Vetores Genéticos/genética , Células HeLa , Humanos , Hibridomas/imunologia , Imunoglobulina G/imunologia , Imunoglobulina G/isolamento & purificação , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos/genética , Proteínas do Grupo Polycomb , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/isolamento & purificação , Proteínas Repressoras/genética , Proteínas Repressoras/isolamento & purificação
14.
J Mol Cell Cardiol ; 47(6): 761-9, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19800350

RESUMO

The insulin receptor substrate (IRS) family plays important roles in cellular growth, signaling, and survival in the brain. We identified IRS6/Dok-5, a member of the IRS family, also expressed in heart. Dok-5 expression level significantly increased during cardiomyocyte differentiation of P19CL6 cells. To understand the mechanism of Dok-5 gene expression and regulation during cardiomyocyte differentiation, we first mapped the transcription start site of the mouse Dok-5 gene and characterized its promoter regions. Truncation and mutation analysis of the Dok-5 promoter identified the forkhead binding element responsible for the repression of Dok-5 promoter activation. The co-localization of FOXO3a and Dok-5 in the mouse heart allows FOXO3a to be a transcriptional regulator of Dok-5. Electrophoretic mobility shift assay and chromatin immunoprecipitation assay confirmed that FOXO3a could bind to the Dok-5 promoter, accompanied by FOXO3a translocation from the nucleus to cytoplasm. FOXO3a overexpression could inhibit Dok-5 promoter activity. Silencing FOXO3a expression by siRNA upregulated the expression of Dok-5 and enhanced cardiomyocyte differentiation. Moreover, Dok-5 siRNA attenuated cardiomyocyte differentiation. Our results provide the first evidence that FOXO3a, the PI3K/PKB downstream substrate, acts as a transcriptional repressor to inhibit the expression of Dok-5. Dok-5 is involved in cardiomyocyte differentiation by a PI3K/PKB/FOXO3a signaling pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Diferenciação Celular , Fatores de Transcrição Forkhead/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/enzimologia , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular Tumoral , Ensaio de Desvio de Mobilidade Eletroforética , Proteína Forkhead Box O3 , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Miocárdio/metabolismo , Técnicas de Amplificação de Ácido Nucleico , Fosfoproteínas/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , RNA Interferente Pequeno/metabolismo , Sítio de Iniciação de Transcrição , Transcrição Gênica
15.
J Mol Biol ; 390(3): 530-7, 2009 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-19481096

RESUMO

The conserved DPY-30 is an essential component of the dosage compensation complex that balances the X-linked gene expression by regulation of the complex formation in Caenorhabditis elegans. The human DPY-30-like protein (DPY-30L) homolog is a conserved member of certain histone methyltransferase (HMT) complexes. In the human MLL1 (mixed-lineage leukemia-1) HMT complex, DPY-30L binds to the BRE2 homolog ASH2L in order to regulate histone 3-lysine 4 trimethylation. We have determined the 1.2-A crystal structure of the human DPY-30L C-terminal domain (DPY-30L(C)). The DPY-30L(C) structure, harboring the conserved DPY-30 motif, is composed of two alpha-helices linked by a sharp loop and forms a typical X-type four-helix bundle required for dimer formation. DPY-30L(C) dimer formation is largely mediated by an extensive hydrophobic interface with some additional polar interactions. The oligomerization of DPY-30L(C) in solution, together with its reported binding to ASH2L, leads us to propose that the hydrophobic surface of the dimer may provide a platform for interaction with ASH2L in the MLL1 HMT complex.


Assuntos
Proteínas Nucleares/química , Estrutura Quaternária de Proteína , Sequência de Aminoácidos , Cristalografia por Raios X , Dimerização , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Alinhamento de Sequência , Fatores de Transcrição
16.
Chin Med J (Engl) ; 122(20): 2405-11, 2009 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-20079150

RESUMO

BACKGROUND: MicroRNAs (miRNAs) are small noncoding regulatory RNAs whose aberrant expression may be observed in many malignancies. However, few data are yet available on human primary medulloblastomas. This work aimed to identify that whether miRNAs would be aberrantly expressed in tumor tissues compared with non-tumorous cerebellum tissues from same patients, and to explore a possible role during carcinogenesis. METHODS: A high throughput microRNA microarray was performed in human primary medulloblastoma specimens to investigate differentially expressed miRNAs, and some miRNAs were validated using real-time quantitative RT-PCR method. In addition, the predicted target genes for the most significantly down- or up-regulated miRNAs were analyzed by using a newly modified ensemble algorithm. RESULTS: Nine miRNA species were differentially expressed in medulloblastoma specimens versus normal non-tumorous cerebellum tissues. Of these, 4 were over expressed and 5 were under expressed. The changes ranged from 0.02-fold to 6.61-fold. These findings were confirmed using real-time quantitative RT-PCR for most significant deregulated miRNAs (miR-17, miR-100, miR-106b, and miR-218) which are novel and have not been previously published. Interestingly, most of the predicted target genes for these miRNAs were involved in medulloblastoma carcinogenesis. CONCLUSIONS: MiRNAs are differentially expressed between human medulloblastoma and non-tumorous cerebellum tissue. MiRNAs may play a role in the tumorigenesis of medulloblastoma and maybe serve as potential targets for novel therapeutic strategies in future.


Assuntos
Meduloblastoma/genética , MicroRNAs/metabolismo , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Masculino , MicroRNAs/genética , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
J Mol Med (Berl) ; 87(1): 43-51, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18810376

RESUMO

MicroRNAs are approximately 21nt single-stranded RNAs and function as regulators of gene expression. Previous studies have shown that microRNAs play crucial roles in tumorigenesis by targeting the mRNAs of oncogenes or tumor suppressors. Here we show that brain-enriched miR-128 is down-regulated in glioma tissues and cell lines when compared to normal brain tissues. Overexpression of miR-128 in glioma cells inhibited cell proliferation. A bioinformatics search revealed a conserved target site within the 3'untranslated region (UTR) of E2F3a, a transcription factor that regulates cell cycle progression. The protein levels of E2F3a in gliomas and normal brain tissues were negatively correlated to the expression levels of miR-128 in these tissues. Overexpression of miR-128 suppressed a luciferase-reporter containing the E2F3a-3'UTR and reduced the level of E2F3a protein in T98G cells. Moreover, knocking down of E2F3a had similar effect as overexpression of miR-128, and overexpression of E2F3a can partly rescue the proliferation inhibition caused by miR-128. Taken together, our study demonstrates that miR-128 can inhibit proliferation of glioma cells through one of its targets, E2F3a.


Assuntos
Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , MicroRNAs/biossíntese , Proteínas de Neoplasias/biossíntese , Linhagem Celular , Fator de Transcrição E2F3 , Glioma/genética , Humanos , MicroRNAs/genética , Proteínas de Neoplasias/genética
18.
Glia ; 57(9): 989-99, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19062177

RESUMO

Nectin-like molecule 1 (NECL1)/CADM3/IGSF4B/TSLL1/SynCAM3 is a neural tissue-specific immunoglobulin-like cell-cell adhesion molecule downregulated at the mRNA level in 12 human glioma cell lines. Here we found that the expression of NECL1 was lost in six glioma cell lines and 15 primary glioma tissues at both RNA and protein levels. Re-expression of NECL1 into glioma cell line U251 would repress cell proliferation in vitro by inducing cell cycle arrest. And also NECL1 could decrease the growth rate of tumors in nude mice in vivo. To further investigate the mechanism why NECL1 was silenced in glioma, the basic promoter region located at -271 to +81 in NECL1 genomic sequence was determined. DNA bisulfite sequencing was performed to study the methylation status of CpG islands in NECL1 promoter; however, no hypermethylated CpG site was found. Additionally, the activity of histone deacetylase (HDACs) in glioma was higher than that in normal brain tissues, and the expression of NECL1 in glioma cell lines could be reactivated by HDACs inhibitor-Trichostatin A (TSA). So the loss of NECL1 in glioma was at least partly caused by histone deacetylation. Luciferase reporter assays, chromatin immunoprecipitation and co-immunoprecipitation (co-IP) assays indicated that Sp1 played an important role in this process by binding to either HDAC1 in untreated glioma cells or p300/CBP in TSA treated cells. Our finding suggests that NECL1 may act as a tumor suppressor in glioma and loss of it in glioma may be caused by histone deacetylation.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Ácidos Hidroxâmicos/farmacologia , Imunoglobulinas/metabolismo , Proteínas de Membrana/metabolismo , Acetilação , Animais , Sítios de Ligação , Encéfalo/metabolismo , Moléculas de Adesão Celular , Linhagem Celular Tumoral , Proliferação de Células , Ilhas de CpG , Metilação de DNA , Feminino , Inibidores de Histona Desacetilases , Histona Desacetilases/metabolismo , Humanos , Imunoglobulinas/genética , Proteínas de Membrana/genética , Metilação , Camundongos , Camundongos Nus , Transplante de Neoplasias , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Análise de Sequência de DNA , Fator de Transcrição Sp1/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo
19.
Cancer Lett ; 272(2): 197-205, 2008 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-19013014

RESUMO

MicroRNAs have been linked to different cancer-related processes. The microRNA miR-21 appears to function as an anti-apoptosis factor in glioblastomas. However, the functional target genes of miR-21 are largely unknown in glioblastomas. In this study, bioinformatics analysis was used to identify miR-21 target sites in various genes. Luciferase activity assay showed that a number of genes involved in apoptosis, PDCD4, MTAP, and SOX5, carry putative miR-21 binding sites. Expression of PDCD4 protein correlates inversely with expression of miR-21 in a number of human glioblastoma cell lines such as T98G, A172, U87, and U251. Inhibition of miR-21 increases endogenous levels of PDCD4 in cell line T98G and over-expression miR-21 inhibits PDCD4-dependent apoptosis. Together, these results indicate that miR-21 expression plays a key role in regulating cellular processes in glioblastomas and may serve as a target for effective therapies.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Neoplasias Encefálicas/genética , Regulação para Baixo/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Glioblastoma/genética , MicroRNAs/fisiologia , Proteínas de Ligação a RNA/genética , Sequência de Bases , Northern Blotting , Western Blotting , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Primers do DNA , Glioblastoma/patologia , Humanos , Marcação In Situ das Extremidades Cortadas , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 30(3): 268-74, 2008 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-18686603

RESUMO

OBJECTIVE: To detect the expression of CBX7 in human glioma and investigate the potential regulatory effect of abnormally expressed microRNAs on CBX7 expression. METHODS: Semi-quantitative reverse transcription polymerase chain reaction (RT-PCR) and Western blot were applied to detect the expression pattern of CBX7 in 2 human normal brain tissues, 9 glioma tissues, and 3 glioma cell lines. Miranda algorithm and Ensemble Machine Learning algorithm were combined to predict miRNAs that target human CBX7. The expression of miR-9 in those tissues and cell lines were detected by real-time PCR. After miR-9 overexpression in 293ET and miR-9 knock-down in T98G, luciferase assay and Western blot were used to confirm the effect of miR-9 on CBX7 expression. MTT assay and flow cytometry were applied to detect the effect of miR-9 knock-down on T98G cells. RESULTS: No obvious difference in the CBX7 mRNA level between normal and tumor tissues was observed, while the protein level of CBX7 was abrogated or markedly reduced in glioma tissues and cell lines. Several miRNAs including miR-9 may target CBX7 by bioinformatics prediction. MiR-9 was up-regulated in glioma tissues and cell lines. In 293ET cell, luciferase activity of CBX7-3'UTR reporter was decreased to 24% after miR-9 overexpression. After miR-9 knock-down in T98G cell, the luciferase activity was increased by 1.8 fold and there was no change of CBX7 mRNA, while the protein level of endogenous CBX7 was significantly increased. The number of survival T98G cells increased and cells in G1 phase decreased after miR-9 knock-down. CONCLUSION: In human glioma, CBX7 is down-regulated by the inhibition of miR-9 at posttranscriptional level.


Assuntos
Glioma/metabolismo , MicroRNAs/fisiologia , Proteínas Repressoras/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Algoritmos , Western Blotting , Encéfalo/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células , Criança , Feminino , Citometria de Fluxo , Glioma/genética , Humanos , Técnicas In Vitro , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Complexo Repressor Polycomb 1 , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA