Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34810252

RESUMO

Vascular endothelial cells are exposed to shear stresses with disturbed vs. laminar flow patterns, which lead to proinflammatory vs. antiinflammatory phenotypes, respectively. Effective treatment against endothelial inflammation and the consequent atherogenesis requires the identification of new therapeutic molecules and the development of drugs targeting these molecules. Using Connectivity Map, we have identified vitexin, a natural flavonoid, as a compound that evokes the gene-expression changes caused by pulsatile shear, which mimics laminar flow with a clear direction, vs. oscillatory shear (OS), which mimics disturbed flow without a clear direction. Treatment with vitexin suppressed the endothelial inflammation induced by OS or tumor necrosis factor-α. Administration of vitexin to mice subjected to carotid partial ligation blocked the disturbed flow-induced endothelial inflammation and neointimal formation. In hyperlipidemic mice, treatment with vitexin ameliorated atherosclerosis. Using SuperPred, we predicted that apurinic/apyrimidinic endonuclease1 (APEX1) may directly interact with vitexin, and we experimentally verified their physical interactions. OS induced APEX1 nuclear translocation, which was inhibited by vitexin. OS promoted the binding of acetyltransferase p300 to APEX1, leading to its acetylation and nuclear translocation. Functionally, knocking down APEX1 with siRNA reversed the OS-induced proinflammatory phenotype, suggesting that APEX1 promotes inflammation by orchestrating the NF-κB pathway. Animal experiments with the partial ligation model indicated that overexpression of APEX1 negated the action of vitexin against endothelial inflammation, and that endothelial-specific deletion of APEX1 ameliorated atherogenesis. We thus propose targeting APEX1 with vitexin as a potential therapeutic strategy to alleviate atherosclerosis.


Assuntos
Apigenina/genética , Apigenina/fisiologia , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Células Endoteliais/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Aterosclerose , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação , Camundongos , Fenótipo , Fosforilação , Ligação Proteica , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo
2.
Front Cell Dev Biol ; 9: 642150, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33898431

RESUMO

The transition of flow microenvironments from veins to arteries in vein graft surgery induces "peel-off" of venous endothelial cells (vECs) and results in restenosis. Recently, arterial laminar shear stress (ALS) and oscillatory shear stress (OS) have been shown to affect the cell cycle and inflammation through epigenetic controls such as histone deacetylation by histone deacetylases (HDACs) and trimethylation on lysine 9 of histone 3 (H3K9me3) in arterial ECs. However, the roles of H3K9me3 and HDAC in vEC damage under ALS are not known. We hypothesized that the different responses of HDACs and H3K9me3 might cause vEC damage under the transition of venous flow to arterial flow. We found that arterial ECs showed high expression of H3K9me3 protein and were retained in the G0 phase of the cell cycle after being subjected to ALS. vECs became round under ALS with a decrease in the expression of H3K9me3, HDAC3, and HDAC5, and an increase in the expression of vascular cell adhesion molecule 1 (VCAM-1). Inhibition of HDACs activity by a specific inhibitor, phenylbutyrate, in arterial ECs caused similar ALS-induced inflammation and cell loss as observed in vECs. Activation of HDACs and H3K9me3 by ITSA-1, an HDAC activator, could prevent ALS-induced peel-off and reduced VCAM-1 expression in vECs. Moreover, shear stress modulates EC morphology by the regulation of focal adhesion kinase (FAK) expression. ITSA-1 or EGF could increase phosphorylated (p)-FAK expression in vECs under ALS. We found that perturbation of the activity of p-FAK and increase in p-FAK expression restored ALS-induced H3K9me3 expression in vECs. Hence, the abnormal mechanoresponses of H3K9me3 and HDAC in vECs after being subjected to ALS could be reversed by ITSA-1 or EGF treatment: this offers a strategy to prevent vein graft failure.

3.
Arterioscler Thromb Vasc Biol ; 39(12): 2492-2504, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31597449

RESUMO

OBJECTIVE: Understanding message delivery among vascular cells is essential for deciphering the intercellular communications in cardiovascular diseases. MicroRNA (miR)-92a is enriched in endothelial cells (ECs) and circulation under atheroprone conditions. Macrophages are the primary immune cells in atherosclerotic lesions that modulate lesion development. Therefore, we hypothesize that, in response to atheroprone stimuli, ECs export miR-92a to macrophages to regulate their functions and enhance atherosclerotic progression. Approach and Results: We investigated the macrophage functions that are regulated by EC miR-92a under atheroprone microenvironments. We first determined the distributions of functional extracellular miR-92a by fractionating the intravesicular and extravesicular compartments from endothelial conditioned media and mice serum. The results indicate that extracellular vesicles are the primary vehicles for EC miR-92a transportation. Overexpression of miR-92a in ECs enhanced the proinflammatory responses and low-density lipoprotein uptake, while impaired the migration, of cocultured macrophage. Opposite effects were found in macrophages cocultured with ECs with miR-92a knockdown. Further analyses demonstrated that intravesicular miR-92a suppressed the expression of target gene KLF4 (Krüppel-like factor 4) in macrophages, suggesting a mechanism by which intravesicular miR-92a regulates recipient cell functions. Indeed, the overexpression of KLF4 rescued the EC miR-92a-induced macrophage atheroprone phenotypes. Furthermore, an inverse correlation of intravesicular miR-92a in blood serum and KLF4 expression in lesions was observed in atherosclerotic animals, indicating the potential function of extracellular miR-92a in regulating vascular diseases. CONCLUSIONS: EC miR-92a can be transported to macrophages through extracellular vesicles to regulate KLF4 levels, thus leading to the atheroprone phenotypes of macrophage and, hence, atherosclerotic lesion formation.


Assuntos
Aterosclerose/genética , Células Endoteliais da Veia Umbilical Humana/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Macrófagos/metabolismo , MicroRNAs/genética , Animais , Aterosclerose/metabolismo , Aterosclerose/patologia , Comunicação Celular , Células Cultivadas , Líquido Extracelular/metabolismo , Células Endoteliais da Veia Umbilical Humana/ultraestrutura , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/biossíntese , Macrófagos/ultraestrutura , Camundongos , MicroRNAs/biossíntese , Microscopia Eletrônica de Transmissão
4.
Biomaterials ; 204: 59-69, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30884320

RESUMO

The use of biochemical signaling to derive smooth muscle cells (SMCs) from mesenchymal stem cells (MSCs) has been explored, but the induction of a fully functional SMC phenotype remains to be a major challenge. Cell morphology has been shown to regulate MSC differentiation into various lineages, including SMCs. We engineered substrates with microgrooves to induce cell elongation to study the mechanism underlying the MSC shape modulation in SMC differentiation. In comparison to those on flat substrates, MSCs cultured on engineered substrates were elongated with increased aspect ratios for both cell body and nucleus, as well as augmented cytoskeletal tensions. Biochemical studies indicated that the microgroove-elongated cells expressed significantly higher levels of SMC markers. MicroRNA analyses showed that up-regulation of miR-145 and the consequent repression of KLF4 in these elongated cells promoted MSC-to-SMC differentiation. Rho/ROCK inhibitions, which impair cytoskeletal tension, attenuated cell and nuclear elongations and disrupted the miR-145/KLF4 regulation for SMC differentiation. Furthermore, cell traction force measurements showed that miR-145 is essential for the functional contractility in the microgroove-induced SMC differentiation. Collectively, our findings demonstrate that, through a Rho-ROCK/miR-145/KLF4 pathway, the elongated cell shape serves as a decisive geometric cue to direct MSC differentiation into functional SMCs.


Assuntos
Diferenciação Celular , Forma Celular , Células-Tronco Mesenquimais/citologia , MicroRNAs/metabolismo , Miócitos de Músculo Liso/citologia , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Forma Celular/efeitos dos fármacos , Forma Celular/genética , Dimetilpolisiloxanos/farmacologia , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , Modelos Biológicos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
5.
Sci Rep ; 7(1): 14996, 2017 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-29118325

RESUMO

The earliest atherosclerotic lesions preferentially develop in arterial regions experienced disturbed blood flow, which induces endothelial expression of pro-atherogenic genes and the subsequent endothelial dysfunction. Our previous study has demonstrated an up-regulation of DNA methyltransferase 1 (DNMT1) and a global hypermethylation in vascular endothelium subjected to disturbed flow. Here, we determined that DNMT1-specific inhibition in arterial wall ameliorates the disturbed flow-induced atherosclerosis through, at least in part, targeting cell cycle regulator cyclin A and connective tissue growth factor (CTGF). We identified the signaling pathways mediating the flow-induction of DNMT1. Inhibition of the mammalian target of rapamycin (mTOR) suppressed the DNMT1 up-regulation both in vitro and in vivo. Together, our results demonstrate that disturbed flow influences endothelial function and induces atherosclerosis in an mTOR/DNMT1-dependent manner. The conclusions obtained from this study might facilitate further evaluation of the epigenetic regulation of endothelial function during the pathological development of atherosclerosis and offer novel prevention and therapeutic targets of this disease.


Assuntos
Aterosclerose/patologia , Endotélio Vascular/patologia , Epigênese Genética/fisiologia , Hemorreologia/fisiologia , Animais , Artérias/patologia , Artérias/fisiopatologia , Aterosclerose/genética , Aterosclerose/fisiopatologia , Bovinos , Fator de Crescimento do Tecido Conjuntivo/genética , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Ciclina A/genética , Ciclina A/metabolismo , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Metilação de DNA/fisiologia , Modelos Animais de Doenças , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , Regiões Promotoras Genéticas/genética , Serina-Treonina Quinases TOR/metabolismo
6.
Physiol Genomics ; 49(7): 339-345, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28500253

RESUMO

The long noncoding RNAs (lncRNAs), which constitute a large portion of the transcriptome, have gained intense research interest because of their roles in regulating physiological and pathophysiological functions in the cell. We identified from RNA-Seq profiling a set of lncRNAs in cultured human umbilical vein endothelial cells (HUVECs) that are differentially regulated by atheroprotective vs. atheroprone shear flows. Among the comprehensively annotated lncRNAs, including both known and novel transcripts, LINC00341 is one of the most abundant lncRNAs in endothelial cells. Moreover, its expression level is enhanced by atheroprotective pulsatile shear flow and atorvastatin. Overexpression of LINC00341 suppresses the expression of vascular cell adhesion molecule 1 (VCAM1) and the adhesion of monocytes induced by atheroprone flow and tumor necrosis factor-alpha. Underlying this anti-inflammatory role, LINC00341 guides enhancer of zest homolog 2, a core histone methyltransferase of polycomb repressive complex 2, to the promoter region of the VCAM1 gene to suppress VCAM1. Network analysis reveals that the key signaling pathways (e.g., Rho and PI3K/AKT) are co-regulated with LINC00341 in endothelial cells in response to pulsatile shear. Together, these findings suggest that LINC00341, as an example of lncRNAs, plays important roles in modulating endothelial function in health and disease.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Inflamação/genética , RNA Longo não Codificante/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Atorvastatina/farmacologia , Adesão Celular , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Inflamação/patologia , Monócitos/patologia , Complexo Repressor Polycomb 2/metabolismo , RNA Longo não Codificante/genética , Fator de Necrose Tumoral alfa/metabolismo
7.
Proc Natl Acad Sci U S A ; 113(41): 11525-11530, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27671657

RESUMO

The focal nature of atherosclerotic lesions suggests an important role of local hemodynamic environment. Recent studies have demonstrated significant roles of Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) in mediating mechanotransduction and vascular homeostasis. The objective of this study is to investigate the functional role of YAP/TAZ in the flow regulation of atheroprone endothelial phenotypes and the consequential development of atherosclerotic lesions. We found that exposure of cultured endothelial cells (ECs) to the atheroprone disturbed flow resulted in YAP/TAZ activation and translocation into EC nucleus to up-regulate the target genes, including cysteine-rich angiogenic inducer 61 (CYR61), connective tissue growth factor (CTGF), and ankyrin repeat domain 1 (ANKRD1). In contrast, the athero-protective laminar flow suppressed YAP/TAZ activities. En face analysis of mouse arteries demonstrated an increased nuclear localization of YAP/TAZ and elevated levels of the target genes in the endothelium in atheroprone areas compared with athero-protective areas. YAP/TAZ knockdown significantly attenuated the disturbed flow induction of EC proliferative and proinflammatory phenotypes, whereas overexpression of constitutively active YAP was sufficient to promote EC proliferation and inflammation. In addition, treatment with statin, an antiatherosclerotic drug, inhibited YAP/TAZ activities to diminish the disturbed flow-induced proliferation and inflammation. In vivo blockade of YAP/TAZ translation by morpholino oligos significantly reduced endothelial inflammation and the size of atherosclerotic lesions. Our results demonstrate a critical role of the activation of YAP/TAZ by disturbed flow in promoting atheroprone phenotypes and atherosclerotic lesion development. Therefore, inhibition of YAP/TAZ activation is a promising athero-protective therapeutic strategy.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Aterosclerose/metabolismo , Aterosclerose/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfoproteínas/metabolismo , Reologia , Animais , Apolipoproteínas E/deficiência , Aterosclerose/tratamento farmacológico , Artéria Carótida Primitiva/patologia , Ciclo Celular , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Proteína Rica em Cisteína 61/metabolismo , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Inflamação/patologia , Camundongos , Proteínas Musculares/metabolismo , Proteínas Nucleares/metabolismo , Fenótipo , Proteínas Repressoras/metabolismo , Transativadores , Fatores de Transcrição , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas de Sinalização YAP
8.
J Virol Methods ; 212: 8-11, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25446515

RESUMO

Human papillomavirus (HPV) types 16 and 18 are known to be high-risk viruses that cause cervical cancer. An HPV rapid testing kit that could help physicians to make early and more informed decisions regarding patient care is needed urgently but not yet available. This study aimed to develop a multiplex nested polymerase chain reaction-immunochromatographic test (PCR-ICT) for the rapid identification of HPV 16 and 18. A multiplex nested PCR was constructed to amplify the HPV 16 and 18 genotype-specific L1 gene fragments and followed by ICT which coated with antibodies to identify rapidly the different PCR products. The type-specific gene regions of high-risk HPV 16 and 18 could be amplified successfully by multiplex nested PCR at molecular sizes of approximately 99 and 101bp, respectively. The capture antibodies raised specifically against the moleculars labeled on the PCR products could be detected simultaneously both HPV 16 and 18 in one strip. Under optimal conditions, this PCR-ICT assay had the capability to detect HPV in a sample with as low as 100 copies of HPV viral DNA. The PCR-ICT system has the advantage of direct and simultaneous detection of two high-risk HPV 16 and 18 DNA targets in one sample, which suggested a significant potential of this assay for clinical application.


Assuntos
Cromatografia de Afinidade/métodos , Papillomavirus Humano 16/isolamento & purificação , Papillomavirus Humano 18/isolamento & purificação , Reação em Cadeia da Polimerase Multiplex/métodos , Infecções por Papillomavirus/diagnóstico , Infecções por Papillomavirus/virologia , Reação em Cadeia da Polimerase/métodos , Humanos , Sensibilidade e Especificidade
9.
Clin Chim Acta ; 433: 284-9, 2014 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-24685572

RESUMO

Colorectal cancer (CRC) patients with KRAS mutations do not benefit from epidermal growth factor receptor (EGFR) targeted therapy. In clinical practice, identifying patients with KRAS mutations is critical prior to EGFR targeting therapy, and gene testing is generally performed using the DNA extracted from tumor tissue. The aim of this study was to compare the presence of KRAS mutations in circulating cell-free DNA (cfDNA) and primary tumor tissue using a peptide nucleic acid mediated polymerase chain reaction. We extracted and analyzed the DNA from plasmas and corresponding primary tumor samples from 52 patients with CRC. The results demonstrated that the detection rate of KRAS sequence variations was 50% (26 of 52) in plasma samples and 28.8% (15 of 52) in resected primary tumor tissue samples. The majority of KRAS mutations detected in tumors were also found in matched plasma specimens with an agreement rate of 78.8%. Eleven plasma cfDNA were found positive for KRAS mutation but not in their corresponding tissue. In conclusion, our results suggest that circulating cfDNA provides a better representation of the malignant disease as a whole and could be a reliable source of diagnostic DNA to replace the tumor tissue in a diagnostic setting.


Assuntos
Neoplasias Colorretais/sangue , Neoplasias Colorretais/genética , Análise Mutacional de DNA , DNA/sangue , DNA/genética , Proteínas Proto-Oncogênicas/genética , Proteínas ras/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas p21(ras)
10.
PLoS One ; 8(8): e71342, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23967196

RESUMO

Current research in pulmonary pathology has focused on inflammatory reactions initiated by immunological responses to allergens and irritants. In addition to these biochemical stimuli, physical forces also play an important role in regulating the structure, function, and metabolism of the lung. Hyperstretch of lung tissues can contribute to the inflammatory responses in asthma, but the mechanisms of mechanically induced inflammation in the lung remain unclear. Our results demonstrate that excessive stretch increased the secretion of inflammatory cytokines by human bronchial epithelial cells (hBECs), including IL-8. This increase of IL-8 secretion was due to an elevated microRNA-155 (miR-155) expression, which caused the suppression of Src homology 2 domain-containing inositol 5-phosphatase 1 (SHIP1) production and the subsequent activation of JNK signaling. In vivo studies in our asthmatic mouse model also showed such changes in miR-155, IL-8, and SHIP1 expressions that reflect inflammatory responses. Co-culture with human mesenchymal stem cells (hMSCs) reversed the stretch-induced hBEC inflammatory responses as a result of IL-10 secretion by hMSCs to down-regulate miR-155 expression in hBECs. In summary, we have demonstrated that mechanical stretch modulates the homeostasis of the hBEC secretome involving miR-155 and that hMSCs can be used as a potential therapeutic approach to reverse bronchial epithelial inflammation in asthma.


Assuntos
Brônquios/metabolismo , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , Mucosa Respiratória/metabolismo , Animais , Asma/induzido quimicamente , Asma/metabolismo , Linhagem Celular , Técnicas de Cocultura , Modelos Animais de Doenças , Expressão Gênica , Humanos , Inflamação/metabolismo , Inositol Polifosfato 5-Fosfatases , Pulmão/metabolismo , Pulmão/patologia , Fenômenos Mecânicos , Camundongos , MicroRNAs/metabolismo , Ovalbumina/efeitos adversos , Comunicação Parácrina , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatases , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo
11.
Proc Natl Acad Sci U S A ; 110(32): 13174-9, 2013 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-23878263

RESUMO

Endothelial cells (ECs) are constantly exposed to xenobiotics and endobiotics or their metabolites, which perturb EC function, as well as to shear stress, which plays a crucial role in vascular homeostasis. Pregnane X receptor (PXR) is a nuclear receptor and a key regulator of the detoxification of xeno- and endobiotics. Here we show that laminar shear stress (LSS), the atheroprotective flow, activates PXR in ECs, whereas oscillatory shear stress, the atheroprone flow, suppresses PXR. LSS activation of PXR in cultured ECs led to the increased expression of a PXR target gene, multidrug resistance 1 (MDR1). An in vivo study using rats showed that the expression of MDR1 was significantly higher in the endothelium from the descending thoracic aorta, where flow is mostly laminar, than from the inner curvature of aortic arch, where flow is disturbed. Functionally, LSS-activated PXR protects ECs from apoptosis triggered by doxorubicin via the induction of MDR1 and other detoxification genes. PXR also suppressed the expression of proinflammatory adhesion molecules and monocyte adhesion in response to TNF-α and lipopolysaccharide. Overexpression of a constitutively active PXR in rat carotid arteries potently attenuated proinflammatory responses. In addition, cDNA microarray revealed a large number of the PXR-activated endothelial genes whose products are responsible for major steps of detoxification, including phase I and II metabolizing enzymes and transporters. These detoxification genes in ECs are induced by LSS in ECs in a PXR-dependent manner. In conclusion, our results indicate that PXR represents a flow-activated detoxification system to protect ECs against damage by xeno- and endobiotics.


Assuntos
Perfilação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Inativação Metabólica/genética , Receptores de Esteroides/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Apoptose/efeitos dos fármacos , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Western Blotting , Artérias Carótidas/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Citocromo P-450 CYP1B1 , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Receptor de Pregnano X , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Receptores de Esteroides/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estresse Mecânico , Xenobióticos/metabolismo , Xenobióticos/farmacologia
12.
Circ Res ; 113(1): 40-51, 2013 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-23603512

RESUMO

RATIONALE: Endothelial microRNA-126 (miR-126) modulates vascular development and angiogenesis. However, its role in the regulation of smooth muscle cell (SMC) function is unknown. OBJECTIVE: To elucidate the role of miR-126 secreted by endothelial cells (ECs) in regulating SMC turnover in vitro and in vivo, as well as the effects of shear stress on the regulation. METHODS AND RESULTS: Coculture of SMCs with ECs or treatment of SMCs with conditioned media from static EC monoculture (EC-CM) increased SMC miR-126 level and SMC turnover; these effects were abolished by inhibition of endothelial miR-126 and by the application of laminar shear stress to ECs. SMC miR-126 did not increase when treated with EC-CM from ECs subjected to inhibition of miR biogenesis, or with CM from sheared ECs. Depletion of extracellular/secreted vesicles in EC-CM did not affect the increase of SMC miR-126 by EC-CM. Biotinylated miR-126 or FLAG (DYKDDDDK epitope)-tagged Argonaute2 transfected into ECs was detected in the cocultured or EC-CM-treated SMCs, indicating a direct EC-to-SMC transmission of miR-126 and Argonaute2. Endothelial miR-126 represses forkhead box O3, B-cell lymphoma 2, and insulin receptor substrate 1 mRNAs in the cocultured SMCs, suggesting the functional roles of the transmitted miR-126. Systemic depletion of miR-126 in mice inhibited neointimal lesion formation of carotid arteries induced by cessation of blood flow. Administration of EC-CM or miR-126 mitigated the inhibitory effect. CONCLUSIONS: Endothelial miR-126 acts as a key intercellular mediator to increase SMC turnover, and its release is reduced by atheroprotective laminar shear stress.


Assuntos
Células Endoteliais/metabolismo , Regulação da Expressão Gênica , MicroRNAs/fisiologia , Miócitos de Músculo Liso/citologia , Animais , Proteínas Argonautas/genética , Proteínas Argonautas/fisiologia , Artéria Carótida Primitiva/patologia , Técnicas de Cultura de Células/instrumentação , Linhagem Celular , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/biossíntese , Fatores de Transcrição Forkhead/genética , Inativação Gênica , Genes bcl-2 , Hemorreologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Proteínas Substratos do Receptor de Insulina/biossíntese , Proteínas Substratos do Receptor de Insulina/genética , Ligadura , Camundongos , MicroRNAs/genética , MicroRNAs/uso terapêutico , Músculo Liso Vascular/citologia , Neointima , Comunicação Parácrina , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Recombinantes de Fusão/fisiologia , Artérias Umbilicais/citologia
13.
PLoS One ; 7(10): e46889, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23118862

RESUMO

Endothelial proliferation, which is an important process in vascular homeostasis, can be regulated by the extracellular microenvironment. In this study we demonstrated that proliferation of endothelial cells (ECs) was enhanced on hydrogels with high stiffness (HSG, 21.5 kPa) in comparison to those with low stiffness (LSG, 1.72 kPa). ECs on HSG showed markedly prominent stress fibers and a higher RhoA activity than ECs on LSG. Blockade of RhoA attenuated stress fiber formation and proliferation of ECs on HSG, but had little effect on ECs on LSG; enhancement of RhoA had opposite effects. The phosphorylations of Src and Vav2, which are positive RhoA upstream effectors, were higher in ECs on HSG. The inhibition of Src/Vav2 attenuated the HSG-mediated RhoA activation and EC proliferation but exhibited nominal effects on ECs on LSG. Septin 9 (SEPT9), the negative upstream effector for RhoA, was significantly higher in ECs on LSG. The inhibition of SEPT9 increased RhoA activation, Src/Vav2 phosphorylations, and EC proliferation on LSG, but showed minor effects on ECs on HSG. We further demonstrated that the inactivation of integrin α(v)ß(3) caused an increase of SEPT9 expression in ECs on HSG to attenuate Src/Vav2 phosphorylations and inhibit RhoA-dependent EC proliferation. These results demonstrate that the SEPT9/Src/Vav2/RhoA pathway constitutes an important molecular mechanism for the mechanical regulation of EC proliferation.


Assuntos
Proliferação de Células , Células Endoteliais , Septinas , Pontos de Checagem do Ciclo Celular , Citoesqueleto/metabolismo , Células Endoteliais/citologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Integrina alfaVbeta3/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Proto-Oncogênicas c-vav/metabolismo , RNA Interferente Pequeno , Septinas/genética , Septinas/metabolismo , Transdução de Sinais , Especificidade por Substrato , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
14.
Annu Rev Biomed Eng ; 12: 1-27, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20415587

RESUMO

microRNAs (miRNAs) are small RNAs 18 to 24 nucleotides in length that serve the pivotal function of regulating gene expression. Instead of being translated into proteins, the mature single-stranded miRNA binds to messenger RNAs (mRNAs) to interfere with the translational process. It is estimated that whereas only 1% of the genomic transcripts in mammalian cells encode miRNA, nearly one-third of the encoded genes are regulated by miRNA. Various bioinformatics databases, tools, and algorithms have been developed to predict the sequences of miRNAs and their target genes. In combination with the in silico approaches in systems biology, experimental studies on miRNA provide a new bioengineering approach for understanding the mechanism of fine-tuning gene regulation. This review aims to provide state-of-the-art information on this important mechanism of gene regulation for researchers working in biomedical engineering and related fields. Particular emphases are placed on summarizing the current tools and strategies for miRNA study from a bioengineering perspective and the possible applications of miRNAs (such as antagomirs and miRNA sponges) in biomedical engineering research.


Assuntos
Engenharia Biomédica , Fenômenos Fisiológicos Celulares/genética , Biologia Computacional , Regulação da Expressão Gênica , MicroRNAs/genética , Animais , Sistema Cardiovascular/metabolismo , Bases de Dados Genéticas , Humanos , Masculino , Camundongos , MicroRNAs/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
15.
Proc Natl Acad Sci U S A ; 107(7): 3240-4, 2010 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-20133739

RESUMO

Endothelial cells (ECs) respond to changes in mechanical forces, leading to the modulation of signaling networks and cell function; an example is the inhibition of EC proliferation by steady laminar flow. MicroRNAs (miRs) are short noncoding 20-22 nucleotide RNAs that negatively regulate the expression of target genes at the posttranscriptional level. This study demonstrates that miRs are involved in the flow regulation of gene expression in ECs. With the use of microRNA chip array, we found that laminar shear stress (12 dyn/cm(2), 12 h) regulated the EC expression of many miRs, including miR-19a. We further showed that stable transfection of miR-19a significantly decreased the expression of a reporter gene controlled by a conserved 3'-untranslated region of the cyclinD1 gene and also the protein level of cyclin D1, leading to an arrest of cell cycle at G1/S transition. Laminar flow suppressed cyclin D1 protein level, and this suppressive effect was diminished when the endogenous miR-19a was inhibited. In conclusion, we demonstrated that miR-19a plays an important role in the flow regulation of cyclin D1 expression. These results revealed a mechanism by which mechanical forces modulate endothelial gene expression.


Assuntos
Ciclina D1/metabolismo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica/fisiologia , MicroRNAs/metabolismo , Fluxo Sanguíneo Regional/fisiologia , Veias Umbilicais/citologia , Pareamento de Bases , Sequência de Bases , Fenômenos Biomecânicos , Western Blotting , Primers do DNA/genética , Citometria de Fluxo , Humanos , MicroRNAs/genética , Análise em Microsséries , Dados de Sequência Molecular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Veias Umbilicais/fisiologia
16.
Proc Natl Acad Sci U S A ; 107(7): 3234-9, 2010 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-20133741

RESUMO

MicroRNAs (miRs) can regulate many cellular functions, but their roles in regulating responses of vascular endothelial cells (ECs) to mechanical stimuli remain unexplored. We hypothesize that the physiological responses of ECs are regulated by not only mRNA and protein signaling networks, but also expression of the corresponding miRs. EC growth arrest induced by pulsatile shear (PS) flow is an important feature for flow regulation of ECs. miR profiling showed that 21 miRs are differentially expressed (8 up- and 13 downregulated) in response to 24-h PS as compared to static condition (ST). The mRNA expression profile indicates EC growth arrest under 24-h PS. Analysis of differentially expressed miRs yielded 68 predicted mRNA targets that overlapped with results of microarray mRNA profiling. Functional analysis of miR profile indicates that the cell cycle network is highly regulated. The upregulation of miR-23b and miR-27b was found to correlate with the PS-induced EC growth arrest. Inhibition of miR-23b using antagomir-23b oligonucleotide (AM23b) reversed the PS-induced E2F1 reduction and retinoblastoma (Rb) hypophosphorylation and attenuated the PS-induced G1/G0 arrest. Antagomir AM27b regulated E2F1 expression, but did not affect Rb and growth arrest. Our findings indicate that PS suppresses EC proliferation through the regulation of miR-23b and provide insights into the role of miRs in mechanotransduction.


Assuntos
Células Endoteliais/fisiologia , Regulação da Expressão Gênica/fisiologia , MicroRNAs/fisiologia , Fluxo Pulsátil/fisiologia , Proteína do Retinoblastoma/metabolismo , Análise de Variância , Bromodesoxiuridina , Proliferação de Células , Citometria de Fluxo , Humanos , MicroRNAs/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Oligonucleotídeos/genética , Fosforilação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/fisiologia
17.
J Biol Chem ; 285(1): 30-42, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19889638

RESUMO

Interstitial flow in and around bone tissue is oscillatory in nature and affects the mechanical microenvironment for bone cell growth and formation. We investigated the role of oscillatory shear stress (OSS) in modulating the proliferation of human osteoblast-like MG63 cells and its underlying mechanisms. Application of OSS (0.5 +/- 4 dynes/cm(2)) to MG63 cells induced sustained activation of phosphatidylinositol 3-kinase (PI3K)/Akt/mTOR/p70S6K (p70S6 kinase) signaling cascades and hence cell proliferation, which was accompanied by increased expression of cyclins A and D1, cyclin-dependent protein kinases-2, -4, and -6, and bone formation-related genes (c-fos, Egr-1, and Cox-2) and decreased expression of p21(CIP1) and p27(KIP1). OSS-induced activation of PI3K/Akt/mTOR/p70S6K and cell proliferation were inhibited by specific antibodies or small interference RNAs of alpha(v)beta(3) and beta(1) integrins and by dominant-negative mutants of Shc (Shc-SH2) and focal adhesion kinase (FAK) (FAK(F397Y)). Co-immunoprecipitation assay showed that OSS induces sustained increases in association of Shc and FAK with alpha(v)beta(3) and beta(1) integrins and PI3K subunit p85, which were abolished by transfecting the cells with FAK(F397Y) or Shc-SH2. OSS also induced sustained activation of ERK, which was inhibited by the specific PI3K inhibitor LY294002 and was required for OSS-induced activation of mTOR/p70S6K and proliferation in MG63 cells. Our findings provide insights into the mechanisms by which OSS induces osteoblast-like cell proliferation through activation of alpha(v)beta(3) and beta(1) integrins and synergistic interactions of FAK and Shc with PI3K, leading to the modulation of downstream ERK and Akt/mTOR/p70S6K pathways.


Assuntos
Integrina alfaVbeta3/metabolismo , Integrina beta1/metabolismo , Osteoblastos/citologia , Osteoblastos/enzimologia , Reologia , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Transdução de Sinais , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Proliferação de Células , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Proteína 1 de Resposta de Crescimento Precoce/genética , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Regulação da Expressão Gênica , Humanos , Complexos Multiproteicos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Estresse Mecânico , Serina-Treonina Quinases TOR
18.
Cell Mol Bioeng ; 2(3): 341-350, 2009 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20011623

RESUMO

Vascular endothelial cells (ECs) are continuously exposed to mechanical stimuli (e.g., shear stress). Our previous study has shown that the shear-induced nuclear factor-κB (NF-κB) activation is mediated by integrins [Bhullar, I. S., Y. S. Li, H. Miao, E. Zandi, M. Kim, et al. J. Biol. Chem. 273:30544-30549, 1998]. The shear-activated integrins can also transactivate Flk-1 (a receptor for vascular endothelial growth factor (VEGF)) [Wang, Y., H. Miao, S. Li, K. D. Chen, Y. S. Li, et al. Am. J. Physiol. Cell Physiol. 283:C1540-C1547, 2002], which subsequently recruits Casitas B-lineage lymphoma (Cbl) to regulate inhibitor of κB protein kinase (IKK) [Wang, Y., J. Chang, Y. C. Li, Y. S. Li, J. Y. Shyy, and S. Chien. Am. J. Physiol. Heart Circ. Physiol. 286:H685-H692, 2004], an upstream molecule of NF-κB. Therefore, shear stress may likely utilize the Flk-1/Cbl pathway in regulating NF-κB. In this paper, we confirmed that the inhibition of Flk-1 by its specific inhibitor SU1498 blocked the shear-induced NF-κB translocation. The inhibition of Cbl (an adaptor protein which binds to Flk-1 upon shear) by using a negative mutant (Cbl(nm)) also blocked the promoter activity of NF-κB, and the inhibition of the Cbl-downstream molecule phosphatidylinositol-3-kinase (PI3K) abolished the NF-κB translocation. Further experiments revealed that the disruption of actin cytoskeleton inhibited the Flk-1 and Cbl interaction and NF-κB translocation. The inhibition of focal adhesion kinase (FAK) and Src family kinases, which are involved in the integrin-mediated focal adhesion complex, also blocked the shear-induced NF-κB translocation. Together with our previous findings that integrins mediate the shear-induced activation of Flk-1 and NF-κB [Bhullar, I. S., Y. S. Li, H. Miao, E. Zandi, M. Kim, et al. J. Biol. Chem. 273:30544-30549, 1998; Wang, Y., H. Miao, S. Li, K. D. Chen, Y. S. Li, et al. Am. J. Physiol. Cell Physiol. 283:C1540-C1547, 2002], the present results suggest that Flk-1, Cbl, and PI3K act upstream to NF-κB in response to shear stress. This Flk-1/Cbl/PI3K/NF-κB signaling pathway may be originated from integrins and transmitted by key tyrosine kinases and actin cytoskeleton. These results shed new lights on the molecular mechanism by which mechanical shear stress activates the NF-κB signaling pathway, which is critical for vascular inflammatory responses and atherosclerosis.

19.
J Biomed Sci ; 16: 91, 2009 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-19799797

RESUMO

Bacterial adherence to epithelial cells is a key virulence trait of pathogenic bacteria. The type 1 fimbriae and the P-fimbriae of uropathogenic Escherichia coli (UPEC) have both been described to be important for the establishment of urinary tract infections (UTI). To explore the interactions between the host and bacterium responsible for the different environments of UPEC invasion, we examined the effect of pH and osmolarity on UPEC strain J96 fimbrial expression, and subsequent J96-induced interleukin-8 (IL-8) expression in different uroepithelial cells. The J96 strain grown in high pH with low osmolarity condition was favorable for the expression of type 1 fimbriae; whereas J96 grown in low pH with high osmolarity condition was beneficial for P fimbriae expression. Type 1 fimbriated J96 specifically invaded bladder 5637 epithelial cells and induced IL-8 expression. On the contrary, P fimbriated J96 invaded renal 786-O epithelial cells and induced IL-8 expression effectively. Type 1 fimbriated J96-induced IL-8 induction involved the p38, as well as ERK, JNK pathways, which leads to AP-1-mediated gene expression. P fimbriated J96-induced augmentation of IL-8 expression mainly involved p38-mediated AP-1 and NF-kappaB transcriptional activation. These results indicate that different expression of fimbriae in J96 trigger differential IL-8 gene regulation pathways in different uroepithelial cells.


Assuntos
Células Epiteliais/metabolismo , Células Epiteliais/microbiologia , Regulação Bacteriana da Expressão Gênica , Interleucina-8/biossíntese , Sistema Urinário/metabolismo , Sistema Urinário/microbiologia , Escherichia coli Uropatogênica/metabolismo , Adesinas Bacterianas/química , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Concentração de Íons de Hidrogênio , NF-kappa B/metabolismo , Invasividade Neoplásica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição AP-1/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA