Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Zhonghua Yi Xue Za Zhi ; 104(10): 758-765, 2024 Mar 12.
Artigo em Chinês | MEDLINE | ID: mdl-38462356

RESUMO

Objective: To investigate the effects of lncRNA SNHG11 on proliferation, migration, invasion and apoptosis of colorectal cancer cancer cells and possible mechanisms. Methods: qRT-PCR was performed to detect the expression level of lncRNA SNHG11 in colorectal cancer tissues and its related cell lines. The correlation between SNHG11 expression and clinical prognosis of patients was assessed by bioinformatics techniques. Cultured CRC cell lines were transfected with shCtrl (shCtrl group), shSNHG11#1 (shSNHG11#1 group), shSNHG11#2 (shSNHG11#2 group), Control cDNA (Control cDNA group), and SNHG11 cDNA (SNHG11 cDNA), respectively. Thiazolyl blue (MTT), clone formation assay, Transwell assay, cell scratch assay, and flow cytometry were used to detect the proliferation, migration, invasion, and apoptosis of CRC cells in each group. Western protein blotting was used to detect the expression of relevant proteins in each group, and the effect of lncRNA SNHG11 knockdown on the growth of tumour cells in vivo was analysed by nude mice tumouring assay. Phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) signalling pathway inhibitor LY294002 was used for rescue experiments. Results: The expression of lncRNA SNHG11 was significantly higher in colorectal cancer cells and tissues than in normal tissues (P<0.05). Survival analysis showed that the expression level of SNHG11 was not statistically associated with CRC survival (P>0.05). shSNHG11#2 group compared with shCtrl group. MTT OD490/570 values decreased, the number of CRC cell clones decreased, the number of Transwell cells decreased, the area of cell scratch decreased, and the apoptosis rate increased (P<0.05). The mesenchymal markers matrix metalloproteinase (MMP9), N-cadherin and vimentin were significantly reduced, and the expression of the epithelial marker E-cadherin was upregulated. The expression of anti-apoptotic proteins Bcl-2 and Bcl-xl was decreased, and the expression of pro-apoptotic protein Bax was increased (P<0.05).In vivo experiments showed that lncRNA SNHG11 knockdown inhibited the growth of colorectal cancer cells, and the expression of Ki67 was reduced in tumours (P<0.05). LncRNA SNHG11 knockdown inhibited the expression of p-PI3K, p-Akt and p-mTOR.The PI3K/Akt/mTOR signaling pathway inhibitor LY294002 was able to restore the malignant cytological progression of colorectal cancer cells induced by the overexpression of lncRNA SNHG11. Conclusions: LncRNA SNHG11 is highly expressed in colorectal cancer. lncRNA SNHG11 can promote the malignant progression of colorectal cancer cells by regulating the PI3K/Akt/mTOR signaling pathway, and this finding provides a new theoretical basis for targeted therapy of colorectal cancer.


Assuntos
Neoplasias Colorretais , RNA Longo não Codificante , Animais , Camundongos , Humanos , Fosfatidilinositol 3-Quinase/metabolismo , Fosfatidilinositol 3-Quinase/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/farmacologia , RNA Longo não Codificante/genética , Camundongos Nus , DNA Complementar/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Serina-Treonina Quinases TOR/farmacologia , Neoplasias Colorretais/genética , Mamíferos/genética , Mamíferos/metabolismo
2.
Biomed Pharmacother ; 168: 115792, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37924789

RESUMO

Ulipristal acetate (UPA) is a selective progesterone receptor modulator and is used for the treatment of uterine leiomyoma (a benign tumor). Uterine sarcoma which is highly malignant cancer with a poor prognosis is clinically resembled with uterine leiomyoma. There has been no experimental research on the effect of UPA on uterine sarcoma. In this study, we examined the efficacy of UPA in uterine sarcoma with in vitro and in vivo animal models. Cytotoxicity of UPA was determined in uterine sarcoma cell lines (MES-SA, SK-UT-1, and SK-LMS-1). Apoptotic genes and signaling pathways affected by UPA were analyzed by complementary DNA (cDNA) microarray of uterine sarcoma cell lines and western blot, respectively. An in vivo efficacy of UPA was examined with uterine sarcoma cell line- and patient-derived xenograft (PDX) mice models. UPA inhibited cell growth in uterine sarcoma cell lines and primary culture cells from a PDX mouse (PDX-C). cDNA microarray analysis revealed that CCL2 was highly down-regulated by UPA. Phosphorylation and the total expression of STAT3 were inhibited by UPA. UPA also inhibited CCL2 and STAT3 in PDX-C. The inhibitory effect of UPA had not changed in the overexpression of PR and treatment of progesterone. In vivo efficacy studies with cell line-derived xenografts and a PDX model with leiomyosarcoma, a typical uterine sarcoma, demonstrated that UPA significantly decreased tumor growth. UPA had significant anti-tumor effects in uterine sarcoma through the inhibition of STAT3/CCL2 signaling pathway and might be a potential therapeutic agent to treat this disease.


Assuntos
Leiomioma , Sarcoma , Neoplasias Uterinas , Feminino , Humanos , Animais , Camundongos , Receptores de Progesterona/metabolismo , DNA Complementar/farmacologia , DNA Complementar/uso terapêutico , Neoplasias Uterinas/patologia , Leiomioma/patologia , Transdução de Sinais , Morte Celular , Sarcoma/tratamento farmacológico , Quimiocina CCL2/metabolismo , Fator de Transcrição STAT3/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 42(11): 1378-1397, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36172865

RESUMO

BACKGROUND: Migration of human aortic smooth muscle cells (HASMCs) contributes to the pathogenesis of atherosclerosis. This study aims to functionally characterize long noncoding RNA TPRG1-AS1 (tumor protein p63 regulated 1, antisense 1) in HASMCs and reveal the underlying mechanism of TPRG1-AS1 in HASMCs migration, neointima formation, and subsequent atherosclerosis. METHODS: The expression of TPRG1-AS1 in atherosclerotic plaques was verified a series of in silico analysis and quantitative real-time polymerase chain reaction analysis. Northern blot, rapid amplification of cDNA ends and Sanger sequencing were used to determine its full length. In vitro transcription-translation assay was used to investigate the protein-coding capacity of TPRG1-AS1. RNA fluorescent in situ hybridization was used to confirm its subcellular localization. Loss- and gain-of-function studies were used to investigate the function of TPRG1-AS1. Furthermore, the effect of TPRG1-AS1 on the pathological response was evaluated in carotid balloon injury model, wire injury model, and atherosclerosis model, respectively. RESULTS: TPRG1-AS1 was significantly increased in atherosclerotic plaques. TPRG1-AS1 did not encode any proteins and its full length was 1279nt, which was bona fide a long noncoding RNA. TPRG1-AS1 was mainly localized in cytoplasmic and perinuclear regions in HASMCs. TPRG1-AS1 directly interacted with MYH9 (myosin heavy chain 9) protein in HASMCs, promoted MYH9 protein degradation through the proteasome pathway, hindered F-actin stress fiber formation, and finally inhibited HASMCs migration. Vascular smooth muscle cell-specific transgenic overexpression of TPRG1-AS1 significantly reduced neointima formation, and attenuated atherosclerosis in apolipoprotein E knockout (Apoe-/-) mice. CONCLUSIONS: This study demonstrated that TPRG1-AS1 inhibited HASMCs migration through interacting with MYH9 protein and consequently suppressed neointima formation and atherosclerosis.


Assuntos
Aterosclerose , MicroRNAs , Placa Aterosclerótica , RNA Longo não Codificante , Humanos , Camundongos , Animais , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Músculo Liso Vascular/metabolismo , Neointima/metabolismo , Placa Aterosclerótica/metabolismo , Actinas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , DNA Complementar/metabolismo , DNA Complementar/farmacologia , Hibridização in Situ Fluorescente , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Proliferação de Células , Miócitos de Músculo Liso/metabolismo , Movimento Celular , Aterosclerose/genética , Aterosclerose/prevenção & controle , Aterosclerose/metabolismo , MicroRNAs/genética , Proteínas do Citoesqueleto/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas/metabolismo
4.
J Microencapsul ; 33(4): 391-9, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27530524

RESUMO

Drug delivery to corneal epithelial cells is challenging due to the intrinsic mechanisms that protect the eye. Here, we report a novel liposomal formulation to encapsulate and deliver a short sequence peptide into human corneal epithelial cells (hTCEpi). Using a mixture of Phosphatidylcholine/Caproylamine/Dioleoylphosphatidylethanolamine (PC/CAP/DOPE), we encapsulated a fluorescent peptide, resulting in anionic liposomes with an average size of 138.8 ± 34 nm and a charge of -18.2 ± 1.3 mV. After 2 h incubation with the peptide-encapsulated liposomes, 66% of corneal epithelial (hTCEpi) cells internalised the FITC-labelled peptide, demonstrating the ability of this formulation to effectively deliver peptide to hTCEpi cells. Additionally, lipoplexes (liposomes complexed with plasmid DNA) were also able to transfect hTCEpi cells, albeit at a modest level (8% of the cells). Here, we describe this novel anionic liposomal formulation intended to enhance the delivery of small cargo molecules in situ.


Assuntos
DNA Complementar , Epitélio Corneano/metabolismo , Peptídeos , Transfecção/métodos , Células Cultivadas , DNA Complementar/química , DNA Complementar/farmacologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Epitélio Corneano/citologia , Humanos , Lipossomos , Peptídeos/química , Peptídeos/farmacologia , Plasmídeos/química , Plasmídeos/farmacologia
5.
J Control Release ; 196: 184-96, 2014 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-25312541

RESUMO

A genetically engineered apoferritin variant consisting of 24 heavy-chain subunits (HFn) was produced to achieve a cumulative delivery of an antitumor drug, which exerts its cytotoxic action by targeting the DNA at the nucleus of human cancer cells with subcellular precision. The rationale of our approach is based on exploiting the natural arsenal of defense of cancer cells to stimulate them to recruit large amounts of HFn nanoparticles loaded with doxorubicin inside their nucleus in response to a DNA damage, which leads to a programmed cell death. After demonstrating the selectivity of HFn for representative cancer cells compared to healthy fibroblasts, doxorubicin-loaded HFn was used to treat the cancer cells. The results from confocal microscopy and DNA damage assays proved that loading of doxorubicin in HFn nanoparticles increased the nuclear delivery of the drug, thus enhancing doxorubicin efficacy. Doxorubicin-loaded HFn acts as a "Trojan Horse": HFn was internalized in cancer cells faster and more efficiently compared to free doxorubicin, then promptly translocated into the nucleus following the DNA damage caused by the partial release in the cytoplasm of encapsulated doxorubicin. This self-triggered translocation mechanism allowed the drug to be directly released in the nuclear compartment, where it exerted its toxic action. This approach was reliable and straightforward providing an antiproliferative effect with high reproducibility. The particular self-assembling nature of HFn nanocage makes it a versatile and tunable nanovector for a broad range of molecules suitable both for detection and treatment of cancer cells.


Assuntos
Antineoplásicos/administração & dosagem , Apoferritinas/administração & dosagem , Núcleo Celular/efeitos dos fármacos , DNA de Neoplasias/efeitos dos fármacos , Terapia de Alvo Molecular/métodos , Nanoestruturas/química , Antineoplásicos/química , Apoferritinas/química , Linhagem Celular Tumoral , Dano ao DNA , DNA Complementar/administração & dosagem , DNA Complementar/farmacologia , Escherichia coli/metabolismo , Células HeLa , Humanos , Translocação Genética
6.
J Tissue Eng Regen Med ; 8(10): 763-70, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22888035

RESUMO

In the field of bone regeneration, BMP-2 is considered one of the most important growth factors because of its strong osteogenic activity, and is therefore extensively used in clinical practice. However, the short half-life of BMP-2 protein necessitates the use of supraphysiological doses, leading to severe side-effects. This study investigated the efficiency of bone formation at ectopic and orthotopic sites as a result of a low-cost, prolonged presence of BMP-2 in a large animal model. Constructs consisting of alginate hydrogel and BMP-2 cDNA, together acting as a non-viral gene-activated matrix, were combined with goat multipotent stromal cells (gMSCs) and implanted in spinal cassettes or, together with ceramic granules, intramuscularly in goats, both for 16 weeks. Bone formation occurred in all cell-seeded ectopic constructs, but the constructs containing both gMSCs and BMP-2 plasmid DNA showed higher collagen I and bone levels, indicating an osteogenic effect of the BMP-2 plasmid DNA. This was not seen in unseeded constructs, even though transfected, BMP-2-producing cells were detected in all constructs containing plasmid DNA. Orthotopic constructs showed mainly bone formation in the unseeded groups. Besides bone, calcified alginate was present in these groups, acting as a surface for new bone formation. In conclusion, transfection of seeded or resident cells from this DNA delivery system led to stable expression of BMP-2 during 16 weeks, and promoted osteogenic differentiation and subsequent bone formation in cell-seeded constructs at an ectopic location and in cell-free constructs at an orthotopic location in a large animal model.


Assuntos
Proteína Morfogenética Óssea 2 , DNA Complementar , Técnicas de Transferência de Genes , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Plasmídeos , Alginatos/farmacologia , Animais , Autoenxertos , Proteína Morfogenética Óssea 2/biossíntese , Proteína Morfogenética Óssea 2/genética , Células Cultivadas , Células Imobilizadas/metabolismo , Células Imobilizadas/transplante , DNA Complementar/genética , DNA Complementar/farmacologia , Cabras , Hidrogéis/farmacologia , Plasmídeos/genética , Plasmídeos/farmacologia
7.
Int J Mol Sci ; 13(2): 2133-2147, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22408443

RESUMO

RPL23A gene encodes a ribosomal protein that is a component of the 60S subunit. The protein belongs to the L23P family of ribosomal proteins, which is located in the cytoplasm. The purpose of this paper was to explore the structure and anti-cancer function of ribosomal protein L23A (RPL23A) gene of the Giant Panda (Ailuropoda melanoleuca). The cDNA of RPL23A was cloned successfully from the Giant Panda using RT-PCR technology. We constructed a recombinant expression vector containing RPL23A cDNA and over-expressed it in Escherichia coli using pET28a plasmids. The expression product obtained was purified by using Ni chelating affinity chromatography. Recombinant protein of RPL23A obtained from the experiment acted on Hep-2 cells and human HepG-2 cells, then the growth inhibitory effect of these cells was observed by MTT (3-[4,5-dimethyl-2-thiazolyl]-2,5-diphenyl-2H-tetrazolium bromide) assay. The result indicated that the length of the fragment cloned is 506 bp, and it contains an open-reading frame (ORF) of 471 bp encoding 156 amino acids. Primary structure analysis revealed that the molecular weight of the putative RPL23A protein is 17.719 kDa with a theoretical pI 11.16. The molecular weight of the recombinant protein RPL23A is 21.265 kDa with a theoretical pI 10.57. The RPL23A gene can be really expressed in E. coli and the RPL23A protein, fusioned with the N-terminally His-tagged protein, gave rise to the accumulation of an expected 22 KDa polypeptide. The data showed that the recombinant protein RPL23A had a time- and dose-dependency on the cell growth inhibition rate. The data also indicated that the effect at low concentrations was better than at high concentrations on Hep-2 cells, and that the concentration of 0.185 µg/mL had the best rate of growth inhibition of 36.31%. All results of the experiment revealed that the recombinant protein RPL23A exhibited anti-cancer function on the Hep-2 cells. The study provides a scientific basis and aids orientation for the research and development of cancer protein drugs as well as possible anti-cancer mechanisms. Further research is on going to determine the bioactive principle(s) of recombinant protein RPL23A responsible for its anticancer activity.


Assuntos
Antineoplásicos , DNA Complementar , Proteínas Ribossômicas , Ursidae/genética , Sequência de Aminoácidos , Animais , Antineoplásicos/isolamento & purificação , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Sequência de Bases , Clonagem Molecular , DNA Complementar/genética , DNA Complementar/isolamento & purificação , DNA Complementar/farmacologia , Avaliação Pré-Clínica de Medicamentos , Células Hep G2 , Humanos , Dados de Sequência Molecular , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/isolamento & purificação , Proteínas Ribossômicas/farmacologia , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Transfecção , Células Tumorais Cultivadas
9.
Biomed Pharmacother ; 63(3): 241-8, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18617357

RESUMO

Overexpression of cyclooxygenase (COX)-2 is associated with the progression of various malignancies, but the contribution of COX-2 expression, bioactivity or their cooperation to bladder cancer growth calls for further clarification. In this study, we investigated the inhibitory effect of COX-2 inhibitors, antisense COX-2 nucleotide, and their combination on the growth of bladder cancer cells (5637, 5637-P and 5637-AS). Suppression of either COX-2 expression or activity caused reduced cell proliferation, enhanced cell numbers in G(1) phase, and increased apoptosis; the joint suppression of COX-2 expression and bioactivity enhanced the degree of cell growth inhibition. COX-2 antisense-expressing 5637-AS tumors showed a 41.42+/-3.08% growth inhibition as compared with 5637 controls. Oral administration of indomethacin (3mg/kg) or celecoxib (15 mg/kg) caused tumor growth inhibition by 31.5+/-14.87% or 83.17+/-1.17%, respectively. When COX-2 antisense cDNA and COX-2 inhibitor celecoxib were combined, the tumor growth inhibition rate was further increased up to 88.78+/-3.10%. These results provide evidence that celecoxib has potential therapeutic effect on bladder cancer, and the joint use of COX-2 antisense cDNA with celecoxib may improve their individual therapeutic effect, especially significantly increase the growth inhibitory effect of COX-2 antisense cDNA.


Assuntos
Inibidores de Ciclo-Oxigenase 2/farmacologia , DNA Antissenso/farmacologia , DNA Complementar/farmacologia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Celecoxib , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclo-Oxigenase 2/efeitos dos fármacos , Ciclo-Oxigenase 2/genética , Inibidores de Ciclo-Oxigenase 2/administração & dosagem , Inibidores de Ciclo-Oxigenase/administração & dosagem , Inibidores de Ciclo-Oxigenase/farmacologia , DNA Antissenso/administração & dosagem , DNA Complementar/administração & dosagem , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Indometacina/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pirazóis/administração & dosagem , Sulfonamidas/administração & dosagem , Neoplasias da Bexiga Urinária/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Chin Med J (Engl) ; 121(15): 1433-8, 2008 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-18959122

RESUMO

BACKGROUND: Cyclin B1 (CLB1) is necessary for mitotic initiation in mammalian cells and plays important roles in cancer development. Therefore, a potential strategy in cancer therapy is to suppress the activity of CLB1 by delivering antisense constructs of CLB1 into tumor cells. In previous CLB1 studies, antisense constructs with a short half life were often used and these constructs might not persistently inhibit CLB1. METHODS: We successfully created a recombinant plasmid encoding the full-length antisense cDNA of mouse cyclin B1 (AS-mCLB1) and transfected this construct to the murine Lewis lung carcinoma (LL/2) and CT-26 colon carcinoma (CT-26) cells. We isolated clones of LL/2 and CT-26 transfectants with stable expression of AS-mCLB1. Reverse transcriptional polymerase chain reaction (RT-PCR) and Western blot were applied to detect the expression of the mRNA and protein levels of CLB1. To further test the efficacy of this strategy in vivo, AS-mCLB1-expressing LL/2 and CT-26 transfectants were implanted into mice. RESULTS: We found the expression of the mRNA and protein levels of CLB1 decrease in these transfectants. The inhibition of CLB1 caused prominent G1 arrest, abnormal morphology, retarded cell growth and an increase in apoptosis. In AS-mCLB1-expressing LL/2 and CT-26 transfectants implanted mice, tumorigenicity was effectively suppressed compared with the controls. In addition, the expression of AS-mCLB1 also significantly increases the survival duration of implanted animals. CONCLUSION: AS-mCLB1 is likely to be useful in future cancer therapy, which may be associated with its ability to down-regulate the expression of CLB1 and then induce G1arrest and apoptosis in tumor cells.


Assuntos
Ciclina B/antagonistas & inibidores , DNA Antissenso/farmacologia , DNA Complementar/farmacologia , Neoplasias Experimentais/terapia , Animais , Apoptose , Proliferação de Células , Sobrevivência Celular , Ciclina B/genética , Ciclina B1 , Fase G1 , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neoplasias Experimentais/patologia
11.
Neuromuscul Disord ; 18(3): 248-58, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18343113

RESUMO

At present there is no satisfactory treatment for McArdle's disease, deficiency of myophosphorylase. Injection of modified adenovirus 5 (AdV5) and adeno-associated virus 2 (AAV2) vectors containing myophosphorylase expression cassettes, into semitendinosus muscle of sheep with McArdle's disease, produced expression of functional myophosphorylase and some re-expression of the non-muscle glycogen phosphorylase isoforms (both liver and brain) in regenerating fibres. Expression of both non-muscle isoforms was also seen after control injections of AdV5LacZ vectors. There was up to an order of magnitude greater expression of phosphorylase after myophosphorylase vector injection than after LacZ controls (62% of sections with over 1000 positive muscle fibres, versus 7%). The results presented here suggest that the use of viral vector-mediated phosphorylase gene transfer may be applicable to the treatment of McArdle's disease and that sustained re-expression of the brain and liver isoforms should also be investigated as a possible treatment.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética/métodos , Glicogênio Fosforilase Muscular/genética , Doença de Depósito de Glicogênio Tipo V/terapia , Músculo Esquelético/fisiologia , Adenoviridae/genética , Animais , Biópsia , DNA Complementar/genética , DNA Complementar/farmacologia , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica , Glicogênio Fosforilase Muscular/metabolismo , Doença de Depósito de Glicogênio Tipo V/genética , Doença de Depósito de Glicogênio Tipo V/patologia , Humanos , Óperon Lac , Fibras Musculares Esqueléticas/enzimologia , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/patologia , Reação do Ácido Periódico de Schiff , Ovinos , beta-Galactosidase/genética
12.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 39(6): 940-3, 2008 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-19253831

RESUMO

OBJECTIVE: To investigate the effect of antisense MMP-2 by transfecting the Ad-aMMP-2 on the vascular endothelial cells (VEC) derived from human proferating hemangiomas in vitro. METHODS: Three groups. Group M199, Group Ad-GFP and Group Ad-aMMP-2 were tested in this study with the 100 multiplicity of infection (MOI). Reverse transcription PCR (RT-PCR), Western Blotting and Gelatin Zymography analyses were applied to evaluate the level of endogenous MMP-2 expression. RESULTS: The decreased expression level of endogenous MMP-2 mRNA and protein excretion of MMP-2 and Gelatin Zymography analyses of liveness of Group Ad-aMMP-2 were observed by comparison with Group M199 and Group Ad-GFP (P<0.05). CONCLUSION: Ad-aMMP-2 could inhibit the secretion of MMP-2 from VEC in vitro.


Assuntos
Células Endoteliais/metabolismo , Hemangioma Cavernoso/patologia , Metaloproteinase 2 da Matriz/genética , Oligonucleotídeos Antissenso/farmacologia , Transfecção , Células Cultivadas , DNA Complementar/genética , DNA Complementar/farmacologia , Células Endoteliais/patologia , Feminino , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Pessoa de Meia-Idade , Oligonucleotídeos Antissenso/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Neoplasias Cutâneas/patologia
13.
Mol Cell Neurosci ; 36(3): 392-407, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17888676

RESUMO

A cDNA encoding a novel protein was cloned from ischemic rat brain and found to be homologous to testis Mea-2 Golgi-associated protein (Golga3). The sequence predicted a 165-kDa protein, and in vitro translated protein exhibited a molecular mass of 165-170 kDa. Because brain ischemia induced the mRNA, and the protein localized to the Golgi apparatus, this protein was designated Ischemia-Inducible Golgin Protein 165 (IIGP165). In HeLa cells, serum and glucose deprivation-induced caspase-dependent cleavage of the IIGP165 protein, after which the IIGP165 fragments translocated to the nucleus. The C-terminus of IIGP165, which contains a LXXLL motif, appears to function as a transcriptional co-regulator. Akt co-localizes with IIGP165 protein in the Golgi in vivo, and phosphorylates IIGP165 on serine residues 345 and 134. Though transfection of IIGP165 cDNA alone does not protect HeLa cells from serum deprivation or Brefeldin-A-triggered cell death, co-transfection of both Akt and IIGP165 cDNA or combined IIGP165-transfection with PDGF treatment significantly protects HeLa cells better than either treatment alone. These data show that Akt phosphorylation of IIGP165 protects against apoptotic cell death, and add to evidence that the Golgi apparatus also plays a role in regulating apoptosis.


Assuntos
Apoptose/fisiologia , Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Citoproteção/genética , Proteínas de Membrana/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Motivos de Aminoácidos/fisiologia , Sequência de Aminoácidos/fisiologia , Animais , Autoantígenos/química , Autoantígenos/genética , Autoantígenos/metabolismo , Sequência de Bases , Encéfalo/fisiopatologia , Isquemia Encefálica/genética , Isquemia Encefálica/fisiopatologia , Células COS , Chlorocebus aethiops , Citoproteção/efeitos dos fármacos , DNA Complementar/farmacologia , Proteínas da Matriz do Complexo de Golgi , Células HeLa , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Dados de Sequência Molecular , Células PC12 , Fosforilação , Proteínas Proto-Oncogênicas c-akt/genética , Ratos , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/isolamento & purificação , Transfecção/métodos
14.
J Gen Physiol ; 129(4): 267-83, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17389247

RESUMO

Elucidation of the role of PtdIns(4,5)P(2) in epithelial function has been hampered by the inability to selectively manipulate the cellular content of this phosphoinositide. Here we report that SigD, a phosphatase derived from Salmonella, can effectively hydrolyze PtdIns(4,5)P(2), generating PtdIns(5)P. When expressed by microinjecting cDNA into epithelial cells forming confluent monolayers, wild-type SigD induced striking morphological and functional changes that were not mimicked by a phosphatase-deficient SigD mutant (C462S). Depletion of PtdIns(4,5)P(2) in intact SigD-injected cells was verified by detachment from the membrane of the pleckstrin homology domain of phospholipase Cdelta, used as a probe for the phosphoinositide by conjugation to green fluorescent protein. Single-cell measurements of cytosolic pH indicated that the Na(+)/H(+) exchange activity of epithelia was markedly inhibited by depletion of PtdIns(4,5)P(2). Similarly, anion permeability, measured using two different halide-sensitive probes, was depressed in cells expressing SigD. Depletion of PtdIns(4,5)P(2) was associated with marked alterations in the actin cytoskeleton and its association with the plasma membrane. The junctional complexes surrounding the injected cells gradually opened and the PtdIns(4,5)P(2)-depleted cells eventually detached from the monolayer, which underwent rapid restitution. Similar observations were made in intestinal and renal epithelial cultures. In addition to its effects on phosphoinositides, SigD has been shown to convert inositol 1,3,4,5,6-pentakisphosphate (IP(5)) into inositol 1,4,5,6-tetrakisphosphate (IP(4)), and the latter has been postulated to mediate the diarrhea caused by Salmonella. However, the effects of SigD on epithelial cells were not mimicked by microinjection of IP(4). In contrast, the cytoskeletal and ion transport effects were replicated by hydrolyzing PtdIns(4,5)P(2) with a membrane-targeted 5-phosphatase or by occluding the inositide using high-avidity tandem PH domain constructs. We therefore suggest that opening of the tight junctions and inhibition of Na(+)/H(+) exchange caused by PtdIns(4,5)P(2) hydrolysis combine to account, at least in part, for the fluid loss observed during Salmonella-induced diarrhea.


Assuntos
Proteínas de Bactérias/metabolismo , Células Epiteliais/patologia , Fosfatidilinositol 4,5-Difosfato/metabolismo , Infecções por Salmonella/metabolismo , Salmonella typhimurium/enzimologia , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/patologia , Animais , Ânions/metabolismo , Apoptose/fisiologia , Proteínas de Bactérias/genética , DNA Complementar/farmacologia , Diarreia/metabolismo , Diarreia/microbiologia , Diarreia/patologia , Células Epiteliais/enzimologia , Células Epiteliais/microbiologia , Células HeLa , Humanos , Hidrólise , Intestino Delgado/citologia , Mutagênese , Fosfatos de Fosfatidilinositol/biossíntese , Ratos , Infecções por Salmonella/microbiologia , Infecções por Salmonella/patologia , Trocadores de Sódio-Hidrogênio/metabolismo , Junções Íntimas/metabolismo , Junções Íntimas/patologia , Vacúolos/metabolismo , Vacúolos/patologia
15.
Methods Mol Biol ; 403: 87-109, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18827989

RESUMO

Automated electrophysiological assays are of great importance for modern drug discovery, and various approaches have been developed into practical devices. Here, we describe the automation of two-electrode voltage-clamp (TEVC) recording from Xenopus oocytes using the Roboocyte automated workstation, jointly developed by Multi Channel Systems and Bayer Technology Services. We briefly discuss the technology, including its advantages and limitations relative to patch clamp and other TEVC systems. We provide a step-by-step description of typical operating procedures and show that the Roboocyte represents a practical and highly effective way to perform automated electrophysiology in an industrial setting.


Assuntos
Automação/métodos , Eletrofisiologia/métodos , Oócitos/fisiologia , Robótica/métodos , Xenopus laevis , Animais , DNA Complementar/administração & dosagem , DNA Complementar/farmacologia , Relação Dose-Resposta a Droga , Eletrodos , Injeções , Ativação do Canal Iônico/efeitos dos fármacos , Ligantes , Oócitos/efeitos dos fármacos , Oócitos/enzimologia , Técnicas de Patch-Clamp , Linguagens de Programação , RNA Mensageiro/administração & dosagem , RNA Mensageiro/farmacologia , ATPase Trocadora de Sódio-Potássio/metabolismo
16.
Oral Oncol ; 42(9): 907-13, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16730219

RESUMO

The newly identified MMP-28 has been shown to be expressed in several types of carcinomas, however, its functional role in transformation events is unknown. This study was to assess whether this proteinase plays a role in oral tumor malignancy. By using RT-PCR, we found that expression of MMP-28 was significantly higher in 92 oral squamous cell carcinomas (OSCCs) (52/92, 56.5%) than in seven oral premalignant lesions (OPMLs) (0/7, 0%) (P=0.004). No statistically significant correlation was found between MMP-28 expression and tumor stage, thickness, size, and metastasis. Both mRNA and protein of MMP-28 were preferentially concentrated in OSCC specimens than in neighboring tissues as analyzed by semi-quantitative RT-PCR (P=0.015) and immunohistochemistry, respectively. Transfection of OSCC and esophageal carcinoma cell lines with MMP-28 antisense oligodeoxynucleotide (AODN) resulted in the reduced secretion of MMP-28 protein and the ability of colony formation in soft agar without affecting cell growth. Our findings show the close correlation between MMP-28 and OSCC, and support a role for MMP-28 in the anchorage-independent growth of both OSCC and esophageal carcinomas.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Regulação Neoplásica da Expressão Gênica , Metaloproteinases da Matriz Secretadas/metabolismo , Neoplasias Bucais/enzimologia , Proteínas de Neoplasias/metabolismo , Lesões Pré-Cancerosas/enzimologia , Western Blotting/métodos , Carcinoma de Células Escamosas/patologia , Proliferação de Células , DNA Complementar/farmacologia , Neoplasias Esofágicas/enzimologia , Humanos , Imuno-Histoquímica , Hibridização In Situ , Neoplasias Bucais/patologia , Oligonucleotídeos Antissenso/farmacologia , Lesões Pré-Cancerosas/patologia , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Inibidor Tecidual de Metaloproteinase-2/análise , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Transfecção/métodos , Células Tumorais Cultivadas
17.
Endocrine ; 30(3): 313-23, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17526944

RESUMO

In this study, we investigated the effect of dexamethasone on the synthesis of steroidogenic acute regulatory protein (StAR) and the expression of DAX-1 (dosage sensitive sex reversal adrenal hypoplasia congenita critical region on the X chromosome, gene 1) and SF-1 (steroidogenic factor-1) in vivo. Male rats were treated with dexamethasone (0.4 and 4 mg/kg body wt per day) by intraperitoneal injections using phosphate-buffered saline as the vehicle for 7 d. At the end of 7 d, serum testosterone levels were decreased. Response to luteinizing hormone (LH) and 8-bromo-cyclic-AMP (8-Br-cAMP) in vitro was reduced in testicular cells isolated from dexamethasone-treated rat testes. Dexamethasone decreased LH-stimulated cAMP production. The conversion of 22(R)-hydroxycholesterol, pregnenolone, 17-hydroxypregnenolone, dehydroepiandrosterone, and androstenedione to testosterone was not affected by dexamethasone. Dexamethasone increased DAX-1 expression and concordantly decreased StAR protein and mRNA in testicular cells. The increase in DAX-1 protein corresponded to a 57% reduction in StAR mRNA levels concomitant with a 79% reduction in serum testosterone levels. Dexamethasone had no effect on the level of SF-1, but increased the amount of complexed DAX-1-SF-1. Dexamethasone in vitro suppressed StAR promoter activity when an increasing amount of DAX-1 cDNA was transfected. These results demonstrate that dexamethasone increases expression of DAX-1, which results in increased amounts of complexed DAX-1-SF-1, in the absence of any change in the expression of SF-1. These observations strongly support the concept that dexamethasone suppresses rat testicular testosterone production, at least in part, by increasing the amount of complexed DAX-1-SF-1 in these cells, which leads directly to decreased availability of free SF-1 and, therefore, decreased activation of transcription of the rat StAR gene.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Dexametasona/farmacologia , Glucocorticoides/metabolismo , Proteínas de Homeodomínio/metabolismo , Fosfoproteínas/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores do Ácido Retinoico/metabolismo , Proteínas Repressoras/metabolismo , Testosterona/biossíntese , Fatores de Transcrição/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Linhagem Celular Tumoral , Receptor Nuclear Órfão DAX-1 , DNA Complementar/farmacologia , Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Masculino , Regiões Promotoras Genéticas/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Fator Esteroidogênico 1 , Testículo/efeitos dos fármacos , Testículo/metabolismo , Testosterona/sangue , Testosterona/metabolismo
18.
J Endovasc Ther ; 12(4): 469-78, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16048379

RESUMO

PURPOSE: To evaluate the efficiency of recombinant adeno-associated virus (rAAV)-mediated CD151 gene delivery in promoting neovascularization and improving blood perfusion in the skeletal muscle of the rat hind-limb ischemia model. METHODS: CD151 was constructed into the rAAV vector. Twelve Wistar rats were randomly divided into 2 groups of 6 rats each and then intramuscularly injected with rAAV-CD151 or rAAV-GFP, respectively, in one hind limb. Two weeks after gene delivery, the femoral arteries in the treated limbs were excised to establish the model of hind-limb ischemia. Expression of the transgene product CD151 was confirmed by Western blot and the reverse transcription polymerase chain reaction. The skin temperature, angiographic score, and capillary density of the hind limb were measured to assess blood perfusion and neovascularization 6 weeks after transfection. RESULTS: Compared to the group transfected with GFP, the CD151 group showed a 63% higher angiographic score (p<0.05) and an 18% increase in capillary density (p<0.05). In addition, the mean skin temperature of the AAV-CD151-transfected hind limbs was equivalent to the level of the contralateral nonischemic limb, whereas the limb temperature in the GFP-transfected rats was significantly lower than the nonischemic control. The expression of CD151 in the ischemic hind limb injected with rAAV-CD151 was significantly higher than limbs injected with rAAV-GFP. The CD151 mRNA and protein expression was persistently observed in the injected muscle for at least 6 weeks after injection, while no human CD151 mRNA could be detected in remote organs or rAAV-GFP-injected muscles. CONCLUSIONS: This study demonstrates that the rAAV-mediated CD151 gene transfer into rat skeletal muscles is efficient, stable, and has no ectopic expression. Moreover, rAAV-mediated CD151 gene transfer stimulates neovascularization, especially arteriogenesis, and thereby improves blood perfusion in a rat hind-limb ischemia model. These findings suggest that CD151 could be a new target for neovascularization therapy in ischemic disease, and rAAV-mediated CD151 gene transfer may be useful for treatment of ischemic disease.


Assuntos
Antígenos CD/farmacologia , Terapia Genética/métodos , Vetores Genéticos/farmacologia , Membro Posterior/irrigação sanguínea , Isquemia/terapia , Neovascularização Fisiológica/efeitos dos fármacos , Angiografia , Animais , Antígenos CD/genética , Biópsia por Agulha , Citomegalovirus , DNA Complementar/farmacologia , Modelos Animais de Doenças , Endotélio Vascular/patologia , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde , Humanos , Imuno-Histoquímica , Isquemia/diagnóstico por imagem , Isquemia/fisiopatologia , Masculino , Neovascularização Fisiológica/fisiologia , Probabilidade , Distribuição Aleatória , Ratos , Ratos Wistar , Valores de Referência , Fluxo Sanguíneo Regional/fisiologia , Tetraspanina 24 , Resultado do Tratamento
19.
J Cell Physiol ; 204(2): 437-44, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15672448

RESUMO

Extracellular signal-regulated kinase (ERK) plays a central role in regulating cell growth, differentiation, and apoptosis. We previously found that 2-(2-mercaptoethanol)-3-methyl-1,4-napthoquinone or Compound 5 (Cpd 5), is a Cdc25A protein phosphatase inhibitor and causes prolonged, strong ERK phosphorylation which is triggered by epidermal growth factor receptor (EGFR) activation. We now report that Cpd 5 can directly cause ERK phosphorylation by inhibiting Cdc25A activity independently of the EGFR pathway. We found that Cdc25A physically interacted with and de-phosphorylated phospho-ERK both in vitro and in cell culture. Inhibition of Cdc25A activity by Cpd 5 resulted in ERK hyper-phosphorylation. Transfection of Hep3B human hepatoma cells with inactive Cdc25A mutant enhanced Cpd 5 action on ERK phosphorylation, whereas over-expression of Cdc25A attenuated this Cpd 5 action. Furthermore, endogenous Cdc25A knock-down by Cdc25A siRNA resulted in a constitutive-like ERK phosphorylation and Cpd 5 treatment further enhanced it. In EGFR-devoid NR6 fibroblasts and MEK (ERK kinase) mutated MCF7 cells, Cpd 5 treatment also resulted in ERK phosphorylation, providing support for the idea that Cpd 5 can directly act on ERK phosphorylation by inhibiting Cdc25A activity. These data suggest that phospho-ERK is likely another Cdc25A substrate, and Cpd 5-caused ERK phosphorylation is probably regulated by both EGFR-dependent and EGFR-independent pathways.


Assuntos
Inibidores Enzimáticos/farmacologia , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Mercaptoetanol/análogos & derivados , Naftoquinonas/farmacologia , Fosfatases cdc25/metabolismo , Linhagem Celular Tumoral , Sistema Livre de Células , DNA Complementar/farmacologia , Interações Medicamentosas , Ativação Enzimática/fisiologia , Humanos , Mercaptoetanol/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-raf/metabolismo , RNA Interferente Pequeno/farmacologia , Transfecção , Fosfatases cdc25/genética
20.
J Cell Physiol ; 203(3): 510-9, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15534860

RESUMO

We have previously shown that Compound 5 (Cpd 5), an inhibitor of protein phosphatase Cdc25A, inhibits Hep3B human hepatoma cell growth. We now show that hepatocyte growth factor (HGF), a hepatocyte growth stimulant, can strongly enhance Cpd 5-induced growth inhibition in Hep3B cells, and this enhancement in cell growth inhibition is correlated with a much stronger ERK phosphorylation when compared to cells treated with Cpd 5 or HGF separately. We found that HGF/Cpd 5-induced ERK phosphorylation and cell growth inhibition were mediated by Akt (protein kinase B) pathway, since combination HGF/Cpd 5 treatment of Hep3B cells inhibited Akt phosphorylation at Ser-473 and its kinase activity, which led to the suppression of Raf-1 phosphorylation at Ser-259. The suppression of Raf-1 Ser-259 phosphorylation caused the induction of Raf-1 kinase activity, as well as hyper-ERK phosphorylation. Transient transfection of Hep3B cells with dominant negative Akt c-DNA further enhanced both Cpd 5- and HGF/Cpd 5-induced ERK phosphorylation, while over-expression of wild-type Akt c-DNA diminished their effects. In contrast, HGF antagonized the growth inhibitory actions of Cpd 5 on normal rat hepatocytes, thus showing a selective effect on tumor cells compared to normal cells. Our data suggest that Akt kinase negatively regulates MAPK activity at the Akt-Raf level. Suppression of Akt activity by either combination HGF/Cpd 5 treatment or by dominant negative Akt c-DNA transfection antagonizes the Akt inhibitory effect on Raf-1, resulting in an enhancement of Cpd 5-induced MAPK activation and cell growth inhibition.


Assuntos
Inibidores do Crescimento/farmacologia , Fator de Crescimento de Hepatócito/farmacologia , Hepatócitos/efeitos dos fármacos , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Vitamina K/análogos & derivados , Vitamina K/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , DNA Complementar/genética , DNA Complementar/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator de Crescimento de Hepatócito/uso terapêutico , Hepatócitos/enzimologia , Humanos , Neoplasias Hepáticas Experimentais/enzimologia , Sistema de Sinalização das MAP Quinases/fisiologia , Mutação/genética , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-akt , Proteínas Proto-Oncogênicas c-raf/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-raf/metabolismo , Ratos , Ratos Endogâmicos F344 , Células Tumorais Cultivadas , Fosfatases cdc25/antagonistas & inibidores , Fosfatases cdc25/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA