Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 9(1): 17449, 2019 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-31767948

RESUMO

MicroRNAs (miRNAs) are a class of small non-coding RNAs that regulate gene or protein expression by targeting mRNAs and triggering either translational repression or mRNA degradation. Distinct expression levels of miRNAs, including miR-29b, have been detected in various biological fluids and tissues from a large variety of disease models. However, how miRNAs "react" and function in different cellular environments is still largely unknown. In this study, the regulation patterns of miR-29b between human and mouse cell lines were compared for the first time. CRISPR/Cas9 gene editing was used to stably knockdown miR-29b in human cancer HeLa cells and mouse fibroblast NIH/3T3 cells with minimum off-targets. Genome editing revealed mir-29b-1, other than mir-29b-2, to be the main source of generating mature miR-29b. The editing of miR-29b decreased expression levels of its family members miR-29a/c via changing the tertiary structures of surrounding nucleotides. Comparing transcriptome profiles of human and mouse cell lines, miR-29b displayed common regulation pathways involving distinct downstream targets in macromolecular complex assembly, cell cycle regulation, and Wnt and PI3K-Akt signalling pathways; miR-29b also demonstrated specific functions reflecting cell characteristics, including fibrosis and neuronal regulations in NIH/3T3 cells and tumorigenesis and cellular senescence in HeLa cells.


Assuntos
Regulação da Expressão Gênica , Células HeLa/metabolismo , MicroRNAs/genética , Células NIH 3T3/metabolismo , Animais , Sequência de Bases , Sistemas CRISPR-Cas , Transformação Celular Neoplásica , Senescência Celular , Células Clonais , Edição de Genes , Técnicas de Silenciamento de Genes , Ontologia Genética , Humanos , Camundongos , MicroRNAs/biossíntese , Modelos Moleculares , Conformação de Ácido Nucleico , RNA Guia de Cinetoplastídeos/genética , Alinhamento de Sequência , Transdução de Sinais , Transcriptoma
2.
J Cell Biochem ; 119(2): 1501-1510, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28777484

RESUMO

DNA methylation plays a crucial role in lots of biological processes and cancer. 5-azacytidine (5-AC), a DNA methylation inhibitor, has been used as a potential chemotherapeutic agent for cancer. In this study, we used 5-AC treatment to investigate whether DNA methylation was involved in regulation of programmed cell death (PCD) in mouse embryo fibroblast NIH-3T3 cells which could undergo PCD after treatment with TNF-α and cycloheximide (CHX). The results showed that the genomic DNA of NIH-3T3 cells was hypermethylated during PCD induced by TNF-α and CHX, and 5-AC might prevent this PCD process. However, treatment with the other three DNA methylation inhibitors, 5-aza-deoxycytidine, 6-thioguanine and RG108, did not interfere with the NIH-3T3 cell PCD process. Additionally, knockdown of DNMT1 did not affect the apoptosis process. The present results and observations indicated that 5-AC specifically inhibited the NIH-3T3 apoptosis process via a genomic DNA methylation-independent pathway. During the TNF-α and CHX-inducing apoptosis process, the PCD related BCL-2 family proteins were significantly down-regulated. Furthermore, after the small interference RNA-mediated knockdown of BCL-XL, one of the BCL-2 family proteins, 5-AC did not inhibit the apoptosis process, suggesting that 5-AC inhibited the PCD process induced by TNF-α and CHX by affecting the anti-apoptotic protein BCL-XL.


Assuntos
Azacitidina/farmacologia , Cicloeximida/farmacologia , Células NIH 3T3/citologia , Fator de Necrose Tumoral alfa/farmacologia , Proteína bcl-X/metabolismo , Células A549 , Animais , Apoptose/efeitos dos fármacos , Sobrevivência Celular , Metilação de DNA/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células HeLa , Humanos , Camundongos , Células NIH 3T3/efeitos dos fármacos , Células NIH 3T3/metabolismo , Células RAW 264.7 , Proteína bcl-X/genética
3.
PLoS One ; 12(5): e0178182, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28542481

RESUMO

Hyperbaric oxygen therapy (HBOT) is a noninvasive widely applied treatment that increases the oxygen pressure in tissues. In cochlear implant (CI) research, intracochlear application of neurotrophic factors (NTFs) is able to improve survival of spiral ganglion neurons (SGN) after deafness. Cell-based delivery of NTFs such as brain-derived neurotrophic factor (BDNF) may be realized by cell-coating of the surface of the CI electrode. Human mesenchymal stem cells (MSC) secrete a variety of different neurotrophic factors and may be used for the development of a biohybrid electrode in order to release endogenously-derived neuroprotective factors for the protection of residual SGN and for a guided outgrowth of dendrites in the direction of the CI electrode. HBOT could be used to influence cell behaviour after transplantation to the inner ear. The aim of this study was to investigate the effect of HBOT on the proliferation, BDNF-release and secretion of neuroprotective factors. Thus, model cells (an immortalized fibroblast cell line (NIH3T3)-native and genetically modified) and MSCs were repeatedly (3 x - 10 x) exposed to 100% oxygen at different pressures. The effects of HBO on cell proliferation were investigated in relation to normoxic and normobaric conditions (NOR). Moreover, the neuroprotective and neuroregenerative effects of HBO-treated cells were analysed by cultivation of SGN in conditioned medium. Both, the genetically modified NIH3T3/BDNF and native NIH3T3 fibroblasts, showed a highly significant increased proliferation after five days of HBOT in comparison to normoxic controls. By contrast, the number of MSCs was decreased in MSCs treated with 2.0 bar of HBO. Treating SGN cultures with supernatants of fibroblasts and MSCs significantly increased the survival rate of SGN. HBO treatment did not influence (increase / reduce) this effect. Secretome analysis showed that HBO treatment altered the protein expression pattern in MSCs.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Oxigenoterapia Hiperbárica , Neuroproteção/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Terapia Baseada em Transplante de Células e Tecidos , Meios de Cultivo Condicionados , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , Células NIH 3T3/metabolismo , Células NIH 3T3/transplante , Regeneração Nervosa/fisiologia , Crescimento Neuronal/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Gânglio Espiral da Cóclea/citologia , Gânglio Espiral da Cóclea/fisiologia , Adulto Jovem
4.
Proc Natl Acad Sci U S A ; 114(20): E3882-E3891, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28461498

RESUMO

Cells in physiology integrate local soluble and mechanical signals to regulate genomic programs. Whereas the individual roles of these signals are well studied, the cellular responses to the combined chemical and physical signals are less explored. Here, we investigated the cross-talk between cellular geometry and TNFα signaling. We stabilized NIH 3T3 fibroblasts into rectangular anisotropic or circular isotropic geometries and stimulated them with TNFα and analyzed nuclear translocation of transcription regulators -NFκB (p65) and MKL and downstream gene-expression patterns. We found that TNFα induces geometry-dependent actin depolymerization, which enhances IκB degradation, p65 nuclear translocation, nuclear exit of MKL, and sequestration of p65 at the RNA-polymerase-II foci. Further, global transcription profile of cells under matrix-TNFα interplay reveals a geometry-dependent gene-expression pattern. At a functional level, we find cell geometry affects TNFα-induced cell proliferation. Our results provide compelling evidence that fibroblasts, depending on their geometries, elicit distinct cellular responses for the same cytokine.


Assuntos
Expressão Gênica/fisiologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Animais , Núcleo Celular/metabolismo , Forma Celular/genética , Tamanho Celular , Fibroblastos/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas I-kappa B/metabolismo , Camundongos , Células NIH 3T3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transativadores/metabolismo , Fator de Transcrição RelA/metabolismo
5.
Chembiochem ; 16(3): 472-6, 2015 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-25586136

RESUMO

Phospholipids with a choline head group are abundant components of all biological membranes, performing critical functions in cellular structure, metabolism, and signaling. In spite of their importance, our ability to visualize choline phospholipids in vivo remains very limited. We present a simple and robust chemical strategy to image choline phospholipids, based on the metabolic incorporation of azidocholine analogues, that accurately reflects the normal biosynthetic incorporation of choline into cellular phospholipids. Azidocholine-labeled phospholipids can be imaged in cells with high sensitivity and resolution, following derivatization with fluorophores, by bio-orthogonal chemical reactions compatible with live-cell imaging. We used this method to visualize the subcellular localization of choline phospholipids. We also demonstrate that double metabolic labeling with azidocholine and propargylcholine allows sensitive two-color imaging of choline phospholipids. Our method represents a powerful approach to directly image phospholipids, and to study their dynamics in cells and tissues.


Assuntos
Química Click , Imagem Molecular/métodos , Fosfolipídeos/análise , Fosfolipídeos/química , Animais , Azidas/química , Colina/análogos & derivados , Colina/química , Corantes Fluorescentes/química , Camundongos , Estrutura Molecular , Células NIH 3T3/metabolismo , Fosfatidilcolinas/análise , Fosfatidilcolinas/química , Fosfolipídeos/metabolismo , Sensibilidade e Especificidade
6.
Wound Repair Regen ; 23(1): 14-21, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25571764

RESUMO

Diabetic patients exhibit dysfunction of the normal wound healing process, leading to local ischemia by vascular occlusive disease as well as sustained increases in the proinflammatory cytokines and overproduction of reactive oxygen species (ROS). Of the many sources of ROS, the enzyme xanthine oxidase (XO) has been linked to overproduction of ROS in diabetic environment, and studies have shown that treatment with XO inhibitors decreases XO overactivity and XO-generated ROS. This study evaluates the role of XO in the diabetic wound and the impact of specifically inhibiting its activity on wound healing. Treatment of diabetic wounds with siXDH (xanthine dehydrogenase siRNA) decreased XDH mRNA expression by 51.6%, XO activity by 35.9%, ROS levels by 78.1%, pathologic wound burden by 31.5%, and accelerated wound healing by 7 days (23.3%). Polymerase chain reaction analysis showed that increased XO activity in wild-type wound may be due to XDH to XO conversion and/or XO phosphorylation, but not to gene transcription, whereas increased XO activity in diabetic wounds may also be from gene transcription. These results suggest that XO may be responsible for large proportion of elevated oxidative stress in the diabetic wound environment and that normalizing the metabolic activity of XO using targeted delivery of siXDH may decrease overproduction of ROS and accelerate wound healing in diabetic patients.


Assuntos
Inibidores Enzimáticos/farmacologia , Células NIH 3T3/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Purinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Cicatrização/efeitos dos fármacos , Xantina Oxidase/antagonistas & inibidores , Animais , Linhagem Celular , Células Cultivadas , Expressão Gênica , Homeostase , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , RNA Mensageiro , Reação em Cadeia da Polimerase em Tempo Real
7.
Biomolecules ; 4(4): 1070-92, 2014 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-25513747

RESUMO

A sensitive, versatile and economical method to extract and quantify cyclic nucleotide monophosphates (cNMPs) using LC-MS/MS, including both 3',5'-cNMPs and 2',3'-cNMPs, in mammalian tissues and cellular systems has been developed. Problems, such as matrix effects from complex biological samples, are addressed and have been optimized. This protocol allows for comparison of multiple cNMPs in the same system and was used to examine the relationship between tissue levels of cNMPs in a panel of rat organs. In addition, the study reports the first identification and quantification of 2',3'-cIMP. The developed method will allow for quantification of cNMPs levels in cells and tissues with varying disease states, which will provide insight into the role(s) and interplay of cNMP signalling pathways.


Assuntos
Nucleotídeos Cíclicos/análise , Espectrometria de Massas em Tandem/métodos , Animais , Calibragem , Cromatografia Líquida/métodos , IMP Cíclico/análise , Limite de Detecção , Mamíferos/metabolismo , Camundongos , Células NIH 3T3/metabolismo , Nucleotídeos Cíclicos/metabolismo , Ratos Sprague-Dawley , Reprodutibilidade dos Testes
8.
Exp Eye Res ; 112: 79-92, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23623979

RESUMO

Genetic predisposition and senescence of retinal pigment epithelium induced by oxidative stress are major contributors to age-related macular degeneration (AMD). Single-nucleotide polymorphisms in HTRA1 are strongly linked to the onset of AMD. In this study, we examine the role of HtrA1 in premature senescence and cell death induced by oxidative stress. HtrA1 mRNA and protein were up-regulated during premature senescence induced by H2O2 in both mouse embryonic fibroblasts (MEFs) and ARPE-19 cells. Expression of the senescence markers p21(CIP1/WAF1) and p16(INK4a), and SA-ß-galactosidase activity, were higher in HtrA1+/- MEFs than in HtrA1-/- MEFs. HtrA1+/+ and HtrA1+/- MEFs were more resistant than HtrA1-/- MEFs to H2O2-induced cell death. Activation of p38 MAPK by oxidative stress was quicker in HtrA1+/- MEFs than in HtrA1-/- MEFs. The effects of excess HtrA1 were examined by transient transfection of cells with HtrA1 expression vectors or by addition of recombinant proteins. Excess wild type HtrA1 accelerated premature senescence of MEFs and ARPE-19 cells, while the protease-inactive HtrA1 S328A did not. HtrA1-induced senescence was abrogated by inhibition of p38 MAPK. We conclude that HtrA1 is induced by oxidative stress and promotes premature cell senescence through p38 MAPK in a protease activity-dependent manner.


Assuntos
Apoptose/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Peróxido de Hidrogênio/toxicidade , Estresse Oxidativo , Serina Endopeptidases/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Western Blotting , Feminino , Serina Peptidase 1 de Requerimento de Alta Temperatura A , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células NIH 3T3/efeitos dos fármacos , Células NIH 3T3/metabolismo , Plasmídeos , RNA Mensageiro/metabolismo , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina Endopeptidases/metabolismo , Transfecção , Regulação para Cima
9.
PLoS One ; 8(3): e60528, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555988

RESUMO

Podosomes are cellular "feet," characterized by F-actin-rich membrane protrusions, which drive cell migration and invasion into the extracellular matrix. Small GTPases that regulate the actin cytoskeleton, such as Cdc42 and Rac are central regulators of podosome formation. The adaptor protein IRSp53 contains an I-BAR domain that deforms membranes into protrusions and binds to Rac, a CRIB motif that interacts with Cdc42, an SH3 domain that binds to many actin cytoskeletal regulators with proline-rich peptides including VASP, and the C-terminal variable region by splicing. However, the role of IRSp53 and VASP in podosome formation had been unclear. Here we found that the knockdown of IRSp53 by RNAi attenuates podosome formation and migration in Src-transformed NIH3T3 (NIH-Src) cells. Importantly, the differences in the IRSp53 C-terminal splicing isoforms did not affect podosome formation. Overexpression of IRSp53 deletion mutants suggested the importance of linking small GTPases to SH3 binding partners. Interestingly, VASP physically interacted with IRSp53 in NIH-Src cells and was essential for podosome formation. These data highlight the role of IRSp53 as a linker of small GTPases to VASP for podosome formation.


Assuntos
Moléculas de Adesão Celular/metabolismo , Movimento Celular , Proteínas dos Microfilamentos/metabolismo , Células NIH 3T3/citologia , Proteínas do Tecido Nervoso/metabolismo , Fosfoproteínas/metabolismo , Pseudópodes/metabolismo , Actinas/metabolismo , Animais , Camundongos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Células NIH 3T3/metabolismo , Proteínas do Tecido Nervoso/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Regulação para Cima , Domínios de Homologia de src , Quinases da Família src/metabolismo
10.
Cancer Sci ; 103(8): 1460-6, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22497681

RESUMO

CD98 is a heterodimeric glycoprotein of 125-kDa, which consists of a 90-kDa heavy chain (hc) subunit and 35-kDa to 55-kDa light chain (lc) subunits. It is strongly expressed on the surface of proliferating normal cells and almost all tumor cells. To investigate the participation of CD98 in cellular proliferation and malignant transformation, we analyzed cell-cycle progression of NIH3T3 clones transfected with cDNA of human CD98hc. Although NIH3T3 and control transfectant cells grown to the subconfluent state were arrested in the G0/G1 phase by serum starvation, considerable portions of CD98hc-transfected cells resided at S and G2/M phases. Under serum-starved and confluent conditions, significant fractions (20-25%) of NIH3T3 and control transfectant cells contained less than 2n content DNA, indicating occurrence of apoptosis, whereas no apoptotic cells were detected in CD98hc-transfectant cells. Under serum-starved conditions, a marked increase in the levels of cyclin D1 and cyclin E and a decrease in p16 were observed in CD98hc-transfectant cells. The reverse was true for NIH3T3 and control transfectant cells. Our results suggest that resistance to G1 arrest and apoptosis by CD98 overexpression are associated with high G1-cyclins and low p16 levels.


Assuntos
Apoptose/genética , Proteínas de Ciclo Celular/genética , Ciclo Celular/genética , Cadeia Pesada da Proteína-1 Reguladora de Fusão/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular/fisiologia , Células NIH 3T3/metabolismo , Animais , Técnicas de Cultura de Células , Proteínas de Ciclo Celular/metabolismo , Transformação Celular Neoplásica , Pontos de Checagem da Fase G1 do Ciclo Celular/genética , Humanos , Técnicas Imunológicas , Camundongos , Soro
11.
Toxicol In Vitro ; 25(3): 623-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21195159

RESUMO

Previous studies have shown that activities of tyrosine kinases and secretion of the active form of matrix metalloproteinase-2 (MMP-2) are correlated with promotion of tumor growth, while apoptotic cell death in cancer cells is correlated with anti-cancer effects. Although arsenic has been reported to have both cancer-promoting and anti-cancer effects, the mechanisms of the arsenic-mediated bidirectional effects remain unknown. We examined the effects of arsenic on both proto-oncogene c-RET-transfected NIH3T3 cells with benign characters and oncogenic RET-MEN2A-transfected NIH3T3 cells with malignant characters. Arsenic promoted not only c-RET tyrosine kinase activity but also genetically activated RET-MEN2A kinase activity with promotion of dimer formation of RET proteins. Arsenic also increased secretion of the active form of MMP-2 in both RET-MEN2A-transfectants and c-RET-transfectants. On the other hand, arsenic promoted poly-(ADP-ribose) polymerase (PARP) degradation and cell death in both malignant and non-malignant cells. Interestingly, l-cysteine inhibited the arsenic-mediated tumor-promoting effects (activation of kinases and MMP-2 secretion) but not arsenic-mediated anti-cancer effects (PARP degradation and cell death). Our results suggest redox-linked regulation of arsenic-mediated activities of kinases and MMP-2 secretion but not arsenic-mediated cell death. Our results also suggest that l-cysteine is an ideal supplement that inhibits arsenic-mediated tumor-promoting effects without affecting arsenic-mediated anti-cancer effects.


Assuntos
Anticarcinógenos/toxicidade , Arsenitos/toxicidade , Carcinógenos Ambientais/toxicidade , Cisteína/farmacologia , Inibidores Enzimáticos/toxicidade , Células NIH 3T3/efeitos dos fármacos , Compostos de Sódio/toxicidade , Animais , Sobrevivência Celular/efeitos dos fármacos , Interações Medicamentosas , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Neoplasia Endócrina Múltipla Tipo 2a/genética , Células NIH 3T3/metabolismo , Células NIH 3T3/patologia , Poli(ADP-Ribose) Polimerases/metabolismo , Transfecção
12.
Hybridoma (Larchmt) ; 29(1): 31-6, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20199149

RESUMO

Recombinant human myxovirus resistance protein A (MxA) was successfully expressed by an Escherichia coli expression system. After immunization and cell fusion, a mouse hybridoma (3C2) producing MAbs to MxA was established. Hybridoma 3C2 was further characterized using indirect ELISA, Western blot analysis, immunofluorescent staining, and immunoprecipitation. The ELISA results showed that the titer of 3C2 was between 1:6400 and 1:12800 in ascitic fluids. The isotype of the monoclonal antibody was tested to be IgG1kappa. 3C2 can also specifically recognize human MxA protein in various formats by Western blot analysis, immunofluorescent staining, and immunoprecipitation assay. We further demonstrated that 3C2 could be used to detected MxA expression induce by type I interferon in A549 cell line and human peripheral blood mononuclear cells by Western blot in a dose-dependent manner.


Assuntos
Anticorpos Monoclonais/imunologia , Proteínas de Ligação ao GTP/imunologia , Proteínas Recombinantes/imunologia , Animais , Anticorpos Monoclonais/isolamento & purificação , Antivirais/farmacologia , Líquido Ascítico , Western Blotting , Cães , Ensaio de Imunoadsorção Enzimática , Feminino , Imunofluorescência , Proteínas de Ligação ao GTP/genética , Humanos , Hibridomas/metabolismo , Imunização , Imunoprecipitação , Interferon-alfa/farmacologia , Interferon gama/farmacologia , Rim/citologia , Rim/efeitos dos fármacos , Rim/metabolismo , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Proteínas de Resistência a Myxovirus , Células NIH 3T3/efeitos dos fármacos , Células NIH 3T3/imunologia , Células NIH 3T3/metabolismo , Plasmídeos , Proteínas Recombinantes/genética
13.
Cancer Biol Ther ; 9(2): 122-33, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19923925

RESUMO

A common metabolic change in cancer is the acquisition of glycolytic phenotypes. Increased expression of glycolytic enzymes is considered as one contributing factor. The role of mitochondrial defects in acquisition of glycolytic phenotypes has been postulated but remains controversial. Here we show that functional defects in mitochondrial respiration could be induced by oncogenic H-Ras(Q61L) transformation, even though the mitochondrial contents or mass was not reduced in the transformed cells. First, mitochondrial respiration, as measured by mitochondrial oxygen consumption, was suppressed in NIH-3T3 cells transformed with H-Ras(Q61L). Second, oligomycin or rotenone did not reduce the cellular ATP levels in the H-Ras(Q61L) transformed cells, suggesting a diminished role of mitochondrial respiration in the cellular energy metabolism. Third, inhibition of glycolysis with iodoacetic acid reduced ATP levels at a much faster rate in H-Ras(Q61L) transformed cells than in the vector control cells. The reduction of cellular ATP levels was reversed by exogenously added pyruvate in the vector control cells but not in H-Ras(Q61L) transformed cells. Finally when compared to the HRas(Q61L) transformed cells, the vector control cells had increased resistance toward glucose deprivation. The increased resistance was dependent on mitochondrial oxidative phosphorylation since rotenone or oligomycin abolished the increased survival of the vector control cells under glucose deprivation. The results also suggest an inability of the H-Ras(Q61L) transformed cells to reactivate mitochondrial respiration under glucose deprivation. Taken together, the data suggest that mitochondrial respiration can be impaired during transformation of NIH-3T3 cells by oncogeneic H-Ras(Q61L).


Assuntos
Transformação Celular Neoplásica , Fibroblastos/metabolismo , Genes ras , Mitocôndrias/metabolismo , Proteína Oncogênica p21(ras)/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Antimicina A/farmacologia , Transporte de Elétrons/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Glucose/metabolismo , Glicólise/efeitos dos fármacos , Ácido Iodoacético/farmacologia , Camundongos , Mitocôndrias/efeitos dos fármacos , Mutação de Sentido Incorreto , Células NIH 3T3/metabolismo , Oligomicinas/farmacologia , Proteína Oncogênica p21(ras)/genética , Fosforilação Oxidativa/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Mutação Puntual , Ácido Pirúvico/farmacologia , Rotenona/farmacologia
14.
Int J Cancer ; 126(3): 640-50, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19662655

RESUMO

Proteoglycans play a key role in cancer development and progression by participating in the constitution of a specific fertile tumor microenvironment. As they are largely overexpressed in the malignant stroma, proteoglycans provide a reservoir of potential new targets for anticancer therapies, because they can serve to convey toxic payloads in the close proximity of cancer cells and subsequently destroy them. In this context, versican, a proteoglycan largely overexpressed in several solid cancers, bears the potential to be such an ideal target. As 4 main versican isoforms have been characterized, we sought to determine which isoform could represent the best target in human breast cancer. We used a series of 10 primary breast cancer lesions that were characterized as overexpressing the versican protein, when compared with matched normal breast tissues, using shotgun mass spectrometry and immunohistochemistry experiments. Quantitative polymerase chain reaction and western-blotting experiments were used to evaluate versican isoform expression in breast cancer/normal tissue pairs for which ARN quality was excellent. All known isoforms were significantly overexpressed in the malignant lesions, both at the mRNA and at the protein levels. In the course of this study, we also identified and cloned a new alternatively spliced versican isoform, referred to as V4, which was also found to be upregulated in human breast cancer. This study provides for the first time a comprehensive mRNA and protein analysis of versican isoforms expression in human breast tissues, and offers insights into which therapeutic strategy would be best suited to target versican in human breast cancer lesions.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Neoplasias/biossíntese , Versicanas/biossíntese , Animais , Western Blotting , Linhagem Celular Tumoral/metabolismo , Clonagem Molecular , Sistemas de Liberação de Medicamentos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Células NIH 3T3/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Espectrometria de Massas em Tandem/métodos , Fator de Crescimento Transformador beta1/farmacologia , Regulação para Cima , Versicanas/química , Versicanas/genética , Versicanas/metabolismo
15.
Viral Immunol ; 22(6): 353-69, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19951173

RESUMO

Vesicular stomatitis virus (VSV) replication is highly sensitive to interferon (IFN)-induced antiviral responses. VSV infection of well-known cell lines pretreated with IFN-beta results in a 10(4)-fold reduction in the release of infectious particles, with a concomitant abrogation in viral transcript and/or protein levels. However, in cell lines of neuronal lineage only a threefold reduction in viral transcript and protein levels was observed, despite the same 10(4)-fold reduction in released infectious virions, suggesting an assembly defect. Examination of VSV matrix (M) protein ubiquitination yielded no differences between mock- and IFN-beta-treated neuronal cells. Further analysis of potential post-translational modification events, by scintillation and two-dimensional electrophoretic methods, revealed IFN-beta-induced alterations in M protein and phosphoprotein (P) phosphorylation. Hypophosphorylated P protein was demonstrated by reduced (32)P counts, normalized by (35)S-cysteine/methionine incorporation, and by a shift in isoelectric focusing. Hypophosphorylation of VSV P protein was found to occur in neuronal cell lysates, but not within budded virions from the same IFN-beta-treated cells. In contrast, hyperphosphorylation of VSV M protein was observed in both cell lysates and viral particles from IFN-beta-treated neuronal cells. Hyperphosphorylated M protein was demonstrated by increased (32)P counts relative to (35)S-cysteine/methionine normalization, and by altered isoelectric focusing in protein populations from cell and viral lysates. Hyperphosphorylated VSV M protein was found to inhibit its association with VSV nucleocapsid, suggesting a possible mechanism for type I IFN-mediated misassembly through disruption of the interactions between ribonucleoprotein cores, and hyperphosphorylated M protein bound to the plasma membrane inner leaflet.


Assuntos
Interferon beta/farmacologia , Neurônios/efeitos dos fármacos , Fosfoproteínas/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Vírus da Estomatite Vesicular Indiana/efeitos dos fármacos , Proteínas da Matriz Viral/metabolismo , Proteínas Estruturais Virais/metabolismo , Montagem de Vírus/efeitos dos fármacos , Animais , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Linhagem Celular Tumoral/virologia , Humanos , Células L/efeitos dos fármacos , Células L/metabolismo , Células L/virologia , Camundongos , Células NIH 3T3/efeitos dos fármacos , Células NIH 3T3/metabolismo , Células NIH 3T3/virologia , Neuroblastoma/patologia , Neurônios/metabolismo , Neurônios/virologia , Especificidade de Órgãos , Fosforilação/efeitos dos fármacos , Ubiquitinação/efeitos dos fármacos , Vírus da Estomatite Vesicular Indiana/fisiologia , Montagem de Vírus/fisiologia
16.
BMC Biol ; 7: 81, 2009 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-19939239

RESUMO

BACKGROUND: Bacteria-triggered signaling events in infected host cells are key elements in shaping the host response to pathogens. Within the eukaryotic cell, signaling complexes are spatially organized. However, the investigation of protein-protein interactions triggered by bacterial infection in the cellular context is technically challenging. Here, we provide a methodological approach to exploit fluorescence resonance energy transfer (FRET) to visualize pathogen-initiated signaling events in human cells. RESULTS: Live-cell microscopy revealed the transient recruitment of the Src family tyrosine kinase Hck upon bacterial engagement of the receptor carcinoembryonic antigen-related cell adhesion molecule 3 (CEACAM3). In cells expressing a CEACAM3 variant lacking the cytoplasmic domain, the Src homology 2 (SH2) domain of Hck (Hck-SH2) was not recruited, even though bacteria still bound to the receptor. FRET measurements on the basis of whole cell lysates revealed intimate binding between Hck-SH2 (using enhanced yellow fluorescent protein (YPet)-Hck-SH2) and the tyrosine-phosphorylated enhanced cyan fluorescent protein-labeled cytoplasmic domain of wild-type CEACAM3 (CEACAM3 WT-CyPet) and a flow cytometry-based FRET approach verified this association in intact cells. Using confocal microscopy and acceptor photobleaching, FRET between Hck-SH2 and CEACAM3 was localized to the sites of bacteria-host cell contact. CONCLUSION: These data demonstrate not only the intimate binding of the SH2 domain of Hck to the tyrosine-phosphorylated cytoplasmic domain of CEACAM3 in intact cells, but furthermore, FRET measurements allow the subcellular localization of this process during bacterial infection. FRET-based assays are valuable tools to resolve bacteria-induced protein-protein interactions in the context of the intact host cell.


Assuntos
Transferência Ressonante de Energia de Fluorescência/métodos , Rim/metabolismo , Células NIH 3T3/metabolismo , Transdução de Sinais , Animais , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/metabolismo , Citoplasma/metabolismo , Interações Hospedeiro-Patógeno , Rim/citologia , Rim/microbiologia , Camundongos , Microscopia Confocal , Células NIH 3T3/citologia , Células NIH 3T3/microbiologia , Neisseria gonorrhoeae/fisiologia , Domínios e Motivos de Interação entre Proteínas , Proteínas Proto-Oncogênicas c-hck/metabolismo , Transfecção , Domínios de Homologia de src/fisiologia
17.
J Neurosci Res ; 87(13): 3024-32, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19405101

RESUMO

Acetylcholine (ACh) is known to be a key neurotransmitter in the central and peripheral nervous systems, but it is also produced in a variety of non-neuronal tissues and cells, including lymphocytes, placenta, amniotic membrane, vascular endothelial cells, keratinocytes, and epithelial cells in the digestive and respiratory tracts. To investigate contribution made by the high-affinity choline transporter (CHT1) to ACh synthesis in both cholinergic neurons and nonneuronal cells, we transfected rat CHT1 cDNA into NIH3T3ChAT cells, a mouse fibroblast line expressing mouse choline acetyltransferase (ChAT), to establish the NIH3T3ChAT 112-1 cell line, which stably expresses both CHT1 and ChAT. NIH3T3ChAT 112-1 cells showed increased binding of the CHT1 inhibitor [(3)H]hemicholinium-3 (HC-3) and greater [(3)H]choline uptake and ACh synthesis than NIH3T3ChAT 103-1 cells, a CHT1-negative control cell line. HC-3 significantly inhibited ACh synthesis in NIH3T3ChAT 112-1 cells but did not affect synthesis in NIH3T3ChAT 103-1 cells. ACh synthesis in NIH3T3ChAT 112-1 cells was also reduced by amiloride, an inhibitor of organic cation transporters (OCTs) involved in low-affinity choline uptake, and by procaine and lidocaine, two local anesthetics that inhibit plasma membrane phospholipid metabolism. These results suggest that CHT1 plays a key role in ACh synthesis in NIH3T3ChAT 112-1 cells and that choline taken up by OCTs or derived from the plasma membrane is also utilized for ACh synthesis in both cholinergic neurons and nonneuronal cholinergic cells, such as lymphocytes.


Assuntos
Acetilcolina/biossíntese , Proteínas de Transporte de Cátions/fisiologia , Colina/metabolismo , Acetilcolina/metabolismo , Amilorida/farmacologia , Animais , Proteínas de Transporte de Cátions/genética , Colina O-Acetiltransferase/metabolismo , Hemicolínio 3/farmacologia , Lidocaína/farmacologia , Lipídeos de Membrana/metabolismo , Camundongos , Células NIH 3T3/enzimologia , Células NIH 3T3/metabolismo , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Fosfolipídeos/metabolismo , Procaína/farmacologia , Ratos , Proteínas Recombinantes de Fusão/fisiologia , Sódio/metabolismo
18.
J Biomed Mater Res B Appl Biomater ; 91(1): 109-21, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19360887

RESUMO

In this study, the polyester urethane Degrapol (DP) was explored for medical applications. Electrospun DP-fiber fleeces were characterized with regard to fiber morphology, swelling, and interconnectivity of interfiber spaces. Moreover, DP was assayed for cell proliferation and hemocompatibility being a prerequisite to any further in vivo application. It was shown that DP-fiber fleeces produced at different humidity while spinning affects interconnectivity of interfiber spaces, such that the higher the humidity the looser the resulting fiber fleeces. When the spinning target was cooled with dry ice, the resulting DP-fibers remained less fused to each other. However, permeability for fluorescent beads was not significantly increased. Fibroblast adhesion and proliferation occurred in a comparable manner on native as well as on fibronectin or collagen I adsorbed DP-fiber fleeces. On DP-surfaces fibroblasts proliferated equally well as compared with glass or PLGA surfaces or DP-surfaces adsorbed with fibronectin or collagen I. In contrast, human umbilical vein endothelial cells proliferated only after adsorption of DP-surfaces with fibronectin or collagen I, indicating that different cell types respond differently to DP-surfaces. Furthermore, hemocompatibility of DP-surfaces was found to be similar or better to PLGA or stainless steel, both medically used materials. These experiments indicate that DP-fiber fleeces or surfaces might be useful for tissue engineering.


Assuntos
Poliésteres/química , Poliuretanos/química , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/metabolismo , Técnicas de Cultura de Células/métodos , Proliferação de Células , Ativação do Complemento , Humanos , Teste de Materiais , Camundongos , Células NIH 3T3/citologia , Células NIH 3T3/metabolismo , Poliésteres/metabolismo , Poliuretanos/metabolismo , Propriedades de Superfície , Engenharia Tecidual/instrumentação
19.
Cell Signal ; 21(4): 502-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19146951

RESUMO

Protein kinase C delta (PKCdelta) modulates cell survival and apoptosis in diverse cellular systems. We recently reported that PKCdelta functions as a critical anti-apoptotic signal transducer in cells containing activated p21(Ras) and results in the activation of AKT, thereby promoting cell survival. How PKCdelta is regulated by p21(Ras), however, remains incompletely understood. In this study, we show that PKCdelta, as a transducer of anti-apoptotic signals, is activated by phosphotidylinositol 3' kinase/phosphoinositide-dependent kinase 1 (PI(3)K-PDK1) to deliver the survival signal to Akt in the environment of activated p21(Ras). PDK1 is upregulated in cells containing an activated p21Ras. Knock-down of PDK1, PKCdelta, or AKT forces cells containing activated p21(Ras) to undergo apoptosis. PDK1 regulates PKCdelta activity, and constitutive expression of PDK1 increases PKCdelta activity in different cell types. Conversely, expression of a kinase-dead (dominant-negative) PDK1 significantly suppresses PKCdelta activity. p21(Ras)-mediated survival signaling is therefore regulated by via a PI(3)K-AKT pathway, which is dependent upon both PDK1 and PKCdelta, and PDK1 activates and regulates PKCdelta to determine the fate of cells containing a mutated, activated p21(Ras).


Assuntos
Proteína Oncogênica p21(ras)/fisiologia , Proteína Quinase C-delta/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Apoptose , Células 3T3 BALB/metabolismo , Divisão Celular , Linhagem Celular Tumoral/metabolismo , Sobrevivência Celular , Ativação Enzimática , Técnicas de Silenciamento de Genes , Genes ras , Humanos , Camundongos , Células NIH 3T3/metabolismo , Proteína Oncogênica p21(ras)/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteína Quinase C-delta/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/fisiologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , RNA Interferente Pequeno/farmacologia , Proteínas Recombinantes de Fusão/fisiologia
20.
Leukemia ; 23(3): 501-9, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19005479

RESUMO

In acute myeloid leukaemia (AML), nucleophosmin-1 (NPM1) mutations create a nuclear export signal (NES) motif and disrupt tryptophans at NPM1 C-terminus, leading to nucleophosmin accumulation in leukaemic cell cytoplasm. We investigated how nucleophosmin NES motifs (two physiological and one created by the mutation) regulate traffic and interaction of mutated NPM1, NPM1wt and p14(ARF). Nucleophosmin export into cytoplasm was maximum when the protein contained all three NES motifs, as naturally occurs in NPM1-mutated AML. The two physiological NES motifs mediated NPM1 homo/heterodimerization, influencing subcellular distribution of NPM1wt, mutated NPM1 and p14(ARF) in a 'dose-dependent tug of war' fashion. In transfected cells, excess doses of mutant NPM1 relocated completely NPM1wt (and p14(ARF)) from the nucleoli to the cytoplasm. This distribution pattern was also observed in a proportion of NPM1-mutated AML patients. In transfected cells, excess of NPM1wt (and p14(ARF)) relocated NPM1 mutant from the cytoplasm to the nucleoli. Notably, this distribution pattern was not observed in AML patients where the mutant was consistently cytoplasmic restricted. These findings reinforce the concept that NPM1 mutants are naturally selected for most efficient cytoplasmic export, pointing to this event as critical for leukaemogenesis. Moreover, they provide a rationale basis for designing small molecules acting at the interface between mutated NPM1 and other interacting proteins.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Leucemia Mieloide/genética , Proteínas de Neoplasias/genética , Sinais de Exportação Nuclear/genética , Proteínas Nucleares/genética , Mapeamento de Interação de Proteínas , Proteína Supressora de Tumor p14ARF/química , Transporte Ativo do Núcleo Celular/genética , Doença Aguda , Animais , Nucléolo Celular/metabolismo , Transformação Celular Neoplásica/genética , Citoplasma/metabolismo , Dimerização , Sistemas de Liberação de Medicamentos , Humanos , Leucemia Mieloide/metabolismo , Camundongos , Células NIH 3T3/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Sinais de Exportação Nuclear/fisiologia , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Nucleofosmina , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Relação Estrutura-Atividade , Transfecção , Proteína Supressora de Tumor p14ARF/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...