Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Circ Res ; 129(12): 1158-1174, 2021 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-34747636

RESUMO

RATIONALE: Disturbed flow occurring in arterial branches and curvatures induces vascular endothelial cell (EC) dysfunction and atherosclerosis. We postulated that disturbed flow plays important role in modulating phosphoprotein expression profiles to regulate endothelial functions and atherogenesis. OBJECTIVE: The goal of this study is to discover novel site-specific phosphorylation alterations induced by disturbed flow in ECs to contribute to atherosclerosis. METHODS AND RESULTS: Quantitative phosphoproteomics analysis of ECs exposed to disturbed flow with low and oscillatory shear stress (0.5±4 dynes/cm2) versus pulsatile shear stress (12±4 dynes/cm2) revealed that oscillatory shear stress induces phospho-YY1S118 (serine [S]118 phosphorylation of Yin Yang 1) in ECs. Elevated phospho-YY1S118 level in ECs was further confirmed to be present in the disturbed flow regions in experimental animals and human atherosclerotic arteries. This disturbed flow-induced EC phospho-YY1S118 is mediated by CK2α (casein kinase 2α) through its direct interaction with YY1. Yeast 2-hybrid library screening and in situ proximity ligation assays demonstrate that phospho-YY1S118 directly binds ZKSCAN4 (zinc finger with KRAB [krüppel-associated box] and SCAN [SRE-ZBP, CTfin51, AW-1 and Number 18 cDNA] domains 4) to induce promoter activity and gene expression of HDM2 (human double minute 2), which consequently induces EC proliferation through downregulation of p53 and p21CIP1. Administration of apoE-deficient (ApoE-/-) mice with CK2-specific inhibitor tetrabromocinnamic acid or atorvastatin inhibits atherosclerosis formation through downregulations of EC phospho-YY1S118 and HDM2. Generation of novel transgenic mice bearing EC-specific overexpression of S118-nonphosphorylatable mutant of YY1 in ApoE-/- mice confirms the critical role of phospho-YY1S118 in promoting atherosclerosis through EC HDM2. CONCLUSIONS: Our findings provide new insights into the mechanisms by which disturbed flow induces endothelial phospho-YY1S118 to promote atherosclerosis, thus indicating phospho-YY1S118 as a potential molecular target for atherosclerosis treatment.


Assuntos
Aterosclerose/metabolismo , Células Endoteliais/metabolismo , Fator de Transcrição YY1/metabolismo , Animais , Aterosclerose/fisiopatologia , Sítios de Ligação , Circulação Sanguínea , Caseína Quinase II/metabolismo , Linhagem Celular , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Ligação Proteica , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Transcrição YY1/química , Fator de Transcrição YY1/genética , Dedos de Zinco
2.
Front Cell Dev Biol ; 9: 697539, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34262908

RESUMO

BACKGROUND: Pathophysiological vascular remodeling in response to disturbed flow with low and oscillatory shear stress (OSS) plays important roles in atherosclerosis progression. Pomegranate extraction (PE) was reported having anti-atherogenic effects. However, whether it can exert a beneficial effect against disturbed flow-induced pathophysiological vascular remodeling to inhibit atherosclerosis remains unclear. The present study aims at investigating the anti-atherogenic effects of pomegranate peel polyphenols (PPP) extraction and its purified compound punicalagin (PU), as well as their protective effects on disturbed flow-induced vascular dysfunction and their underlying molecular mechanisms. METHODS: The anti-atherogenic effects of PPP/PU were examined on low-density lipoprotein receptor knockout mice fed with a high fat diet. The vaso-protective effects of PPP/PU were examined in rat aortas using myograph assay. A combination of in vivo experiments on rats and in vitro flow system with human endothelial cells (ECs) was used to investigate the pharmacological actions of PPP/PU on EC dysfunction induced by disturbed flow. In addition, the effects of PPP/PU on vascular smooth muscle cell (VSMC) dysfunction were also examined. RESULTS: PU is the effective component in PPP against atherosclerosis. PPP/PU evoked endothelium-dependent relaxation in rat aortas. PPP/PU inhibited the activation of Smad1/5 in the EC layers at post-stenotic regions of rat aortas exposed to disturbed flow with OSS. PPP/PU suppressed OSS-induced expression of cell cycle regulatory and pro-inflammatory genes in ECs. Moreover, PPP/PU inhibited inflammation-induced VSMC dysfunction. CONCLUSION: PPP/PU protect against OSS-induced vascular remodeling through inhibiting force-specific activation of Smad1/5 in ECs and this mechanism contributes to their anti-atherogenic effects.

3.
J Mol Cell Biol ; 6(6): 458-72, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25548371

RESUMO

T-lymphocyte migration under flow is critical for immune responses, but the mechanisms by which flow modulates the migratory behaviors of T-lymphocytes remain unclear. Human peripheral blood T-lymphocytes (PBTLs), when stimulated with phorbol 12-myristate 13-acetate (PMA), stretched their cell bodies dramatically and moved along the flow direction. In contrast, stromal cell-derived factor-1α-stimulated PBTLs deformed and migrated in a random manner. Here we elucidated the molecular mechanisms underlying flow-induced directionality and deformation of PMA-stimulated PBTLs. PMA primed PBTLs for polarization under flow, with protein kinase C (PKC)-δ enriched in the leading edge, PKC-ßI in the microtubule organizing center, and PKC-ßII in the uropod and peripheral region. PKC-δ regulated cell protrusions in the leading edge through Tiam1/Rac1/calmodulin, whereas PKC-ß regulated RhoA/Rho-associated kinase activity and microtubule stability to modulate uropod contractility and detachment. Our findings indicate that PKC-δ and -ß coordinate in the cell leading edge and uropod, respectively, to modulate the directionality and deformability of migratory T-lymphocytes under flow.


Assuntos
Movimento Celular/fisiologia , Proteína Quinase C beta/metabolismo , Proteína Quinase C-delta/metabolismo , Linfócitos T/enzimologia , Carcinógenos/farmacologia , Movimento Celular/efeitos dos fármacos , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Linfócitos T/citologia , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T , Acetato de Tetradecanoilforbol/farmacologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
4.
Biochim Biophys Acta ; 1833(12): 3124-3133, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24021264

RESUMO

Mechanical forces induced by interstitial fluid flow in and surrounding tissues and by blood/lymphatic flow in vessels may modulate cancer cell invasion and metastasis and anticancer drug delivery. Our previous study demonstrated that laminar flow-induced shear stress induces G2/M arrest in tumor cells. However, whether shear stress modulates final cell fate remains unclear. In this study, we investigated the role of flow-induced shear stress in modulating the survival of four human tumor cell lines, i.e., Hep3B hepatocarcinoma cells, MG63 osteosarcoma cells, SCC25 oral squamous carcinoma cells, and A549 carcinomic alveolar basal epithelial cells. Laminar shear stress (LSS) ranging from 0.5 to 12dyn/cm(2) induced death of these four tumor cell lines. In contrast to LSS at 0.5dyn/cm(2), oscillatory shear stress (OSS) at 0.5±4dyn/cm(2) cannot induce cancer cell death. Both LSS and OSS had no effect on human normal hepatocyte, lung epithelial, and endothelial cells. Application of LSS to these four cell lines increased the percentage of cells stained positively for annexin V-FITC, with up-regulations of cleaved caspase-8, -9, and -3, and PARP. In addition, LSS also induced Hep3B cell autophagy, as detected by acidic vesicular organelle formation, LC3B transformation, and p62/SQSTM1 degradation. By transfecting with small interfering RNA, we found that the shear-induced apoptosis and autophagy are mediated by bone morphogenetic protein receptor type (BMPR)-IB, BMPR-specific Smad1 and Smad5, and p38 mitogen-activated protein kinase in Hep3B cells. Our findings provide insights into the molecular mechanisms by which shear stress induces apoptosis and autophagy in tumor cells.


Assuntos
Apoptose , Autofagia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Neoplasias/patologia , Proteínas Smad/metabolismo , Estresse Mecânico , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Linhagem Celular Tumoral , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Neoplasias/enzimologia , Transdução de Sinais , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo
5.
Cardiovasc Res ; 96(2): 296-307, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22865639

RESUMO

AIMS: The implication of circulating haematopoietic CD34(+) progenitors in the vasculature is unclear due to the lack of understanding of their characteristics and plasticity mediated by their cellular microenvironment. We investigated how vascular smooth muscle cells (SMCs) and their interactions with endothelial cells (ECs) affect the behaviour and plasticity of CD34(+)CD31(+) progenitors and the underlying mechanisms. METHODS AND RESULTS: Human peripheral blood-derived CD34(+)CD31(+) cells were directly transplanted into injured arteries in vivo and co-cultured with ECs and SMCs in vitro. CD34(+)CD31(+) progenitors injected into wire-injured mouse arteries differentiate into ECs and macrophages in the neoendothelial layer and neointima, respectively. SMC-co-culture increases CD34(+)CD31(+) cell mobility and adhesion to and transmigration across ECs. Sorted CD34(+)CD31(+) progenitors that adhered to ECs co-cultured with SMCs have the capacity to form capillary-like structures in Matrigel and chimeric blood vessels in vivo. Sorted transmigrated progenitors give rise to macrophages with increased pro-angiogenic activity. These differentiations of CD34(+)CD31(+) progenitors into ECs and macrophages are mediated by ß(2)-integrin and Notch-1, respectively. ß(2)-Integrin and Notch-1 are activated by their counterligands, intercellular adhesion molecule-1 (ICAM-1) and jagged-1, which are highly expressed in the neoendothelium and neointima in injured arteries. Intra-arterial injection of ß(2)-integrin-activated CD34(+)CD31(+) progenitors into wire-injured mouse arteries inhibits neointima formation. CONCLUSION: Our findings indicate that the peripheral vascular niches composed of ECs and SMCs may predispose haematopoietic CD34(+)CD31(+) progenitors to differentiate into ECs and macrophages through the activations of the ICAM-1/ß(2)-integrin and jagged-1/Notch-1 cascades, respectively.


Assuntos
Antígenos CD18/metabolismo , Células Endoteliais/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Miócitos de Músculo Liso/fisiologia , Receptor Notch1/metabolismo , Animais , Apolipoproteínas E/genética , Proteínas de Ligação ao Cálcio/metabolismo , Adesão Celular , Movimento Celular , Células Endoteliais/citologia , Endotélio Vascular/fisiologia , Artéria Femoral/lesões , Células Endoteliais da Veia Umbilical Humana , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteína Jagged-1 , Macrófagos/citologia , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos SCID , Músculo Liso Vascular/fisiologia , Neointima/prevenção & controle , Neovascularização Fisiológica , Proteínas Serrate-Jagged , Migração Transendotelial e Transepitelial
6.
Proc Natl Acad Sci U S A ; 109(20): 7770-5, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22550179

RESUMO

Vascular endothelial cells (ECs) are constantly exposed to blood flow-induced shear stress, but the mechanism of force-specific activation of their signaling to modulate cellular function remains unclear. We have demonstrated that bone morphogenetic protein receptor (BMPR)-specific Smad1/5 can be force-specifically activated by oscillatory shear stress (OSS) in ECs to cause cell cycle progression. Smad1/5 is highly activated in ECs of atherosclerotic lesions in diseased human coronary arteries from patients with end-stage heart failure undergoing heart transplantation and from apolipoprotein E-deficient mice. Application of OSS (0.5 ± 4 dyn/cm(2)) causes the sustained activation of Smad1/5 in ECs through activations of mammalian target of rapamycin and p70S6 kinase, leading to up-regulation of cyclin A and down-regulations of p21(CIP1) and p27(KIP1) and, hence, EC cycle progression. En face examination of rat aortas reveals high levels of phospho-Smad1/5 in ECs of the inner, but not the outer, curvature of aortic arch, nor the straight segment of thoracic aorta [corrected]. Immunohistochemical and en face examinations of the experimentally stenosed abdominal aorta in rats show high levels of phospho-Smad1/5 in ECs at poststenotic sites, where OSS occurs. These OSS activations of EC Smad1/5 in vitro and in vivo are not inhibited by the BMP-specific antagonist Noggin and, hence, are independent of BMP ligand. Transfecting ECs with Smad1/5-specific small interfering RNAs inhibits the OSS-induced EC cycle progression. Our findings demonstrate the force-specificity of the activation of Smad1/5 and its contribution to cell cycle progression in ECs induced by disturbed flow.


Assuntos
Aterosclerose/fisiopatologia , Ciclo Celular/fisiologia , Células Endoteliais/fisiologia , Regulação da Expressão Gênica/fisiologia , Fluxo Sanguíneo Regional/fisiologia , Proteína Smad1/metabolismo , Estresse Mecânico , Animais , Aorta Abdominal/citologia , Aorta Abdominal/patologia , Apolipoproteínas E/genética , Fenômenos Biomecânicos , Vasos Coronários/citologia , Vasos Coronários/patologia , Ciclina A/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Ratos , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Taiwan
7.
Biomaterials ; 32(26): 6164-73, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21641031

RESUMO

In order to increase the absorption of hydrophilic macromolecules in the small intestine, permeation enhancers such as chitosan (CS) and its derivatives have been evaluated. The aim of the current work was to investigate, on molecular levels, the effect of CS on tight junction (TJ) integrity in Caco-2 cells. The observed changes in transepithelial-electrical-resistance measurements and the staining patterns of the monolayer Caco-2 cells demonstrate that CS can transiently and reversibly open the TJs between cells, thus enhancing the paracellular permeability. TJ ultra-structures examined by transmission electron microscopy support the concept that CS did induce transient opening of TJs. We then assessed TJ disruption at the gene and protein expression levels. Our data indicate that exposure to CS followed by recovery resulted in a significant increase in claudin-4 (Cldn4) gene transcription. Additionally, CS treatment induced redistribution of the TJ protein CLDN4 intracellularly following by its degradation in lysosomes, which represented an important contributing factor in TJ weakening, leading to the opening of TJs. The recovery of TJ after CS disruption required CLDN4 protein synthesis. These results suggest that CS regulates TJs by inducing changes in transmembrane CLDN4 protein. Understanding the mechanism of interaction between CS and epithelial cells is of paramount importance and needs to be established to aid further development in the use of CS to mediate the trans-epithelial drug delivery.


Assuntos
Junções Íntimas/metabolismo , Western Blotting , Células CACO-2 , Quitosana/farmacologia , Claudina-4 , Claudinas/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Imuno-Histoquímica , Microscopia Eletrônica de Transmissão , Reação em Cadeia da Polimerase , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/ultraestrutura
8.
J Bone Miner Res ; 25(3): 627-39, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19821775

RESUMO

Estrogen and mechanical forces are positive regulators for osteoblast proliferation and bone formation. We investigated the synergistic effect of estrogen and flow-induced shear stress on signal transduction and gene expression in human osetoblast-like MG63 cells and primary osteoblasts (HOBs) using activations of extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK) and expressions of c-fos and cyclooxygenase-2 (I) as readouts. Estrogen (17beta-estradiol, 10 nM) and shear stress (12 dyn/cm(2)) alone induced transient phosphorylations of ERK and p38 MAPK in MG63 cells. Pretreating MG63 cells with 17beta-estradiol for 6 hours before shearing augmented these shear-induced MAPK phosphorylations. Western blot and flow cytometric analyses showed that treating MG63 cells with 17beta-estradiol for 6 hrs induced their beta(1)-integrin expression. This estrogen-induction of beta(1)-integrin was inhibited by pretreating the cells with a specific antagonist of estrogen receptor ICI 182,780. Both 17beta-estradiol and shear stress alone induced c-fos and Cox-2 gene expressions in MG63 cells. Pretreating MG63 cells with 17beta-estradiol for 6 hrs augmented the shear-induced c-fos and Cox-2 expressions. The augmented effects of 17beta-estradiol on shear-induced MAPK phosphorylations and c-fos and Cox-2 expressions were inhibited by pretreating the cells with ICI 182,780 or transfecting the cells with beta(1)-specific small interfering RNA. Similar results on the augmented effect of estrogen on shear-induced signaling and gene expression were obtained with HOBs. Our findings provide insights into the mechanism by which estrogen augments shear stress responsiveness of signal transduction and gene expression in bone cells via estrogen receptor-mediated increases in beta(1)-integrin expression.


Assuntos
Estrogênios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Integrina beta1/metabolismo , Osteoblastos/efeitos dos fármacos , Receptores de Estrogênio/efeitos dos fármacos , Western Blotting , Linhagem Celular , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Citometria de Fluxo , Humanos , Integrina beta1/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Resistência ao Cisalhamento , Transdução de Sinais/efeitos dos fármacos
9.
Proc Natl Acad Sci U S A ; 105(10): 3927-32, 2008 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-18310319

RESUMO

Interstitial flow in and around tumor tissue affects the mechanical microenvironment to modulate tumor cell growth and metastasis. We investigated the roles of flow-induced shear stress in modulating cell cycle distribution in four tumor cell lines and the underlying mechanisms. In all four cell lines, incubation under static conditions for 24 or 48 h led to G(0)/G(1) arrest; in contrast, shear stress (12 dynes/cm(2)) induced G(2)/M arrest. The molecular basis of the shear effect was analyzed, and the presentation on molecular mechanism is focused on human MG63 osteosarcoma cells. Shear stress induced increased expressions of cyclin B1 and p21(CIP1) and decreased expressions of cyclins A, D1, and E, cyclin-dependent protein kinases (Cdk)-1, -2, -4, and -6, and p27(KIP1) as well as a decrease in Cdk1 activity. Using specific antibodies and small interfering RNA, we found that the shear-induced G(2)/M arrest and corresponding changes in G(2)/M regulatory protein expression and activity were mediated by alpha(v)beta(3) and beta(1) integrins through bone morphogenetic protein receptor type IA-specific Smad1 and Smad5. Shear stress also down-regulated runt-related transcription factor 2 (Runx2) binding activity and osteocalcin and alkaline phosphatase expressions in MG63 cells; these responses were mediated by alpha(v)beta(3) and beta(1) integrins through Smad5. Our findings provide insights into the mechanism by which shear stress induces G(2)/M arrest in tumor cells and inhibits cell differentiation and demonstrate the importance of mechanical microenvironment in modulating molecular signaling, gene expression, cell cycle, and functions in tumor cells.


Assuntos
Ciclo Celular , Integrinas/metabolismo , Neoplasias/patologia , Proteínas Smad/metabolismo , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/metabolismo , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Fase G2 , Humanos , Integrina alfaVbeta3/metabolismo , Integrina beta1/metabolismo , Mitose , Modelos Biológicos , Fosforilação , Ligação Proteica , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Estresse Mecânico
10.
J Bone Miner Res ; 23(7): 1140-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18333755

RESUMO

Integrins play significant roles in mechanical responses of cells on extracellular matrix (ECM). We studied the roles of integrins and ECM proteins (fibronectin [FN], type I collagen [COL1], and laminin [LM]) in shear-mediated signaling and the expression of bone formation-related genes (early growth response-1 [Egr-1], c-fos, cyclooxygenase-2 [Cox-2], and osteopontin [OPN]) in human osteosarcoma MG63 cells. MG63 cells on FN, COL1, and LM were kept as controls or subjected to shear stress (12 dynes/cm(2)), and the association of alpha(v)beta(3) and beta(1) integrins with Shc, phosphorylation of mitogen-activated protein kinases (MAPKs, i.e., extracellular signal-regulated kinase [ERK], c-jun-NH(2)-terminal kinase [JNK], and p38), and expressions of Egr-1, c-fos, Cox-2, and OPN were determined. In MG63 cells, shear stress induces sustained associations of alpha(v)beta(3) and beta(1) with Shc when seeded on FN, but sustained associations of only beta(1) with Shc when seeded on COL1/LM. Shear inductions of MAPKs and bone formation-related genes were sustained (24 h) in cells on FN, but some of these responses were transient in cells on COL1/LM. The shear activations of ERK, JNK, and p38 were mediated by integrins and Shc, and these pathways differentially modulated the downstream bone formation-related gene expression. Our findings showed that beta(1) integrin plays predominant roles for shear-induced signaling and gene expression in osteoblast-like MG63 cells on FN, COL1, and LM and that alpha(v)beta(3) also plays significant roles for such responses in cells on FN. The beta(1)/Shc association leads to the activation of ERK, which is critical for shear induction of bone formation-related genes in osteoblast-like cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Desenvolvimento Ósseo/genética , Matriz Extracelular/metabolismo , Integrinas/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Osteoblastos/metabolismo , Sequência de Bases , Western Blotting , Linhagem Celular , Primers do DNA , Humanos , Imunoprecipitação , Osteoblastos/citologia , Osteoblastos/enzimologia , Fosforilação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Adaptadoras da Sinalização Shc , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src
11.
Blood ; 110(2): 519-28, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17371946

RESUMO

E-selectin is a major adhesion molecule expressed by endothelial cells (ECs), which are exposed to shear stress and neighboring smooth muscle cells (SMCs). We investigated the mechanisms underlying the modulation of EC E-selectin expression by SMCs and shear stress. SMC coculture induced rapid and sustained increases in expression of E-selectin and phosphorylation of interleukin-1 (IL-1) receptor-associated kinase glycoprotein-130, as well as the downstream mitogen-activated protein kinases (MAPKs) and Akt. By using specific inhibitors, dominant-negative mutants, and small interfering RNA, we demonstrated that activations of c-Jun-NH(2)-terminal kinase (JNK) and p38 of the MAPK pathways are critical for the coculture-induced E-selectin expression. Gel shifting and chromatin immunoprecipitation assays showed that SMC coculture increased the nuclear factor-kappaB (NF-kappaB)-promoter binding activity in ECs; inhibition of NF-kappaB activation by p65-antisense, lactacystin, and N-acetyl-cysteine blocked the coculture-induced E-selectin promoter activity. Protein arrays and blocking assays using neutralizing antibodies demonstrated that IL-1beta and IL-6 produced by EC/SMC cocultures are major contributors to the coculture induction of EC signaling and E-selectin expression. Preshearing of ECs at 12 dynes/cm(2) inhibited the coculture-induced EC signaling and E-selectin expression. Our findings have elucidated the molecular mechanisms underlying the SMC induction of EC E-selectin expression and the shear stress protection against this SMC induction.


Assuntos
Selectina E/genética , Endotélio Vascular/fisiologia , Músculo Liso Vascular/fisiologia , Animais , Técnicas de Cocultura , Endotélio Vascular/citologia , Regulação da Expressão Gênica , Humanos , Interleucina-1/genética , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/citologia , Transdução de Sinais , Estresse Mecânico , Veias Umbilicais
12.
Biomaterials ; 28(7): 1355-66, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17126899

RESUMO

Chitooligosaccharides (COS) have been shown to regulate various cellular and biological functions. However, the effect of COS on inflammatory responses of the cells remains unclear. We investigated the regulatory effect of highly N-acetylated COS (NACOS) on tumor necrosis factor-alpha (TNF-alpha)-induced endothelial cell (EC) E-selectin expression, which is crucial for leukocyte recruitment. ECs were kept as controls or pre-treated with NACOS for different times, and then stimulated with TNF-alpha for 4h. The results show that pre-treating ECs with NACOS inhibited the TNF-alpha-induced E-selectin expression in a dose- and time-dependent manner. This NACOS-mediated inhibition in E-selectin expression was regulated at the transcriptional level, but not due to changes in mRNA stability. Stimulation of ECs with TNF-alpha-induced rapid increases in the phosphorylation of their mitogen-activated protein kinases (MAPKs) [extracellular signal-regulated kinase (ERK), c-Jun-NH2-terminal kinase (JNK), and p38 MAPK]; the inhibitor for JNK (i.e., SP600125), but not those for ERK (i.e., PD98059) and p38 MAPK (i.e., SB203580), attenuated this TNF-alpha-induced E-selectin expression. Pre-treating ECs with NACOS inhibited the TNF-alpha-induced JNK activation, suggesting that JNK was involved in the inhibitory effect of NACOS on TNF-alpha-induced E-selectin expression. Pre-treating ECs with NACOS inhibited the TNF-alpha-induced p65 and p50 mRNA expressions. Gel shifting and chromatin immunoprecipitation assays showed that NACOS blocked the TNF-alpha-induced increases in the binding activity and in vivo promoter binding of nuclear factor-kappaB (NF-kappaB) in ECs. Our findings provide a molecular mechanism by which NACOS inhibit TNF-alpha-induced E-selectin expression in ECs, and a basis for using NACOS in pharmaceutical therapy against inflammation.


Assuntos
Selectina E/genética , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Oligossacarídeos/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Sequência de Bases , Materiais Biocompatíveis , Adesão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , DNA/genética , Células Endoteliais/citologia , Expressão Gênica/efeitos dos fármacos , Humanos , Teste de Materiais , Oligossacarídeos/química , Oligossacarídeos/toxicidade , Fosforilação , Estabilidade de RNA/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células U937
13.
Proc Natl Acad Sci U S A ; 103(8): 2665-70, 2006 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-16477012

RESUMO

The phenotype of smooth muscle cells (SMCs) plays an important role in vascular function in health and disease. We investigated the mechanism of modulation of SMC phenotype (from contractile to synthetic) induced by the synergistic action of a growth factor (platelet-derived growth factor, PDGF-BB) and a cytokine (interleukin, IL-1beta). Human aortic SMCs grown on polymerized collagen showed high expression levels of contractile markers (smooth muscle alpha-actin, myosin heavy chain, and calponin). These levels were not significantly affected by PDGF-BB and IL-1beta individually, but decreased markedly after the combined usage of PDGF-BB and IL-1beta. PDGF/IL-1beta costimulation also induced a sustained phosphorylation of Akt and p70 ribosomal S6 kinase (p70S6K). The effects of PDGF/IL-1beta costimulation on contractile marker expression and Akt and p70S6K phosphorylation were blocked by the phosphatidylinositol 3-kinase inhibitors wortmannin and LY294002 and by adenovirus expressing a dominant-negative Akt, and they were mimicked by constitutively active Akt. PDGF-BB/IL-1beta induced a sustained phosphorylation of PDGF receptor (PDGFR)-beta and its association with IL-1 receptor (IL-1R1). Such activation and association of receptors were blocked by a PDGFR-beta neutralizing antibody (AF385), an IL-1R1 antagonist (IL-1ra), as well as a specific inhibitor of PDGFR-beta phosphorylation (AG1295); these agents also eliminated the PDGF-BB/IL-1beta-induced signaling and phenotypic modulation. PDGF-BB/IL-1beta inhibited the polymerized collagen-induced serum response factor DNA binding activity in the nucleus, and this effect was mediated by the PDGFR-beta/IL-1R1 association and phosphatidylinositol 3-kinase/Akt/p70S6K pathway. Our findings provide insights into the mechanism of SMC phenotypic modulation from contractile to synthetic, e.g., in atherosclerosis.


Assuntos
Aorta/enzimologia , Interleucina-1/farmacologia , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Aorta/citologia , Aorta/efeitos dos fármacos , Doenças da Aorta/enzimologia , Aterosclerose/enzimologia , Becaplermina , Células Cultivadas , Colágeno/metabolismo , Sinergismo Farmacológico , Humanos , Contração Muscular , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Fenótipo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-sis , Proteínas Quinases S6 Ribossômicas 70-kDa/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Fator de Resposta Sérica/metabolismo
14.
J Biomed Sci ; 12(3): 481-502, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15971008

RESUMO

We investigate the role of shear stress in regulating the gene expression in endothelial cells (ECs) in response to tumor necrosis factor-alpha (TNF-alpha). ECs were kept in static condition or pre-exposed to a high level (HSS, 20 dynes/cm2) or a low level of shear stress (LSS, 0.5 dynes/cm2) for 24 h, and TNF-alpha was added under static condition for 4 h. In static ECs, DNA microarray showed that TNF-alpha caused a significant increase in expression of 102 genes and a significant decrease in expression of 12 genes. Pre-shearing of ECs decreased the TNF-alpha-responsiveness of many pro-inflammatory, pro-coagulant, proliferative, and pro-apoptotic genes, whereas it increased the responsiveness of some antioxidant, anti-coagulant, and anti-apoptotic genes. LSS showed less regulatory effects than HSS on EC gene expression in response to TNF-alpha. The microarray data were confirmed by reverse-transcription polymerase chain reaction for 64 selected genes. Pre-shearing of ECs at HSS significantly inhibited the TNF-alpha-induced p65 and p50 mRNA expressions and nuclear factor-kappaB (NF-kappaB)-DNA binding activity. Inhibition of NF-kappaB activity with the p65-antisense or lactacystin under static condition blocked the expression of most of the genes that are TNF-alpha-inducible and shear stress-down-regulated. Our findings suggest that laminar shear stress serves protective functions against atherogenesis.


Assuntos
Células Endoteliais/metabolismo , Estresse Mecânico , Transcrição Gênica , Fator de Necrose Tumoral alfa/farmacologia , Adesão Celular , Células Cultivadas , Ensaio de Desvio de Mobilidade Eletroforética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Monócitos/fisiologia , NF-kappa B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Oligonucleotídeos Antissenso/metabolismo , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Arterioscler Thromb Vasc Biol ; 25(5): 963-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15718492

RESUMO

OBJECTIVES: Vascular endothelial cells (ECs) are influenced by shear stress and neighboring smooth muscle cells (SMCs). We investigated the inflammation-relevant gene expression in EC/SMC cocultures under static condition and in response to shear stress. MATERIALS AND METHODS: Under static condition, DNA microarrays and reverse-transcription polymerase chain reaction identified 23 inflammation-relevant genes in ECs whose expression was significantly affected by coculture with SMCs, with 18 upregulated and 5 downregulated. Application of shear stress (12 dynes/cm2) to the EC side of the coculture for 6 hours inhibited most of the proinflammatory gene expressions in ECs induced by coculture with SMCs. Inhibition of nuclear factor-kappaB (NF-kappaB) activation by the p65-antisense, lactacystin, and N-acetyl-cysteine blocked the coculture-induced EC expression of proinflammatory genes, indicating that the NF-kappaB binding sites in the promoters of these genes play a significant role in their expression as a result of coculture with SMCs. Chromatin immunoprecipitation assays demonstrated the in vivo regulation of NF-kappaB recruitment to selected target promoters. Shear stress inhibited the SMC coculture-induced NF-kappaB activation in ECs and monocytic THP-1 cell adhesion to ECs. CONCLUSIONS: Our findings suggest that shear stress plays an inhibitory role in the proinflammatory gene expression in ECs located in close proximity to SMCs.


Assuntos
Endotélio Vascular/fisiologia , Músculo Liso Vascular/fisiologia , Subunidade p50 de NF-kappa B/metabolismo , Fator de Transcrição RelA/metabolismo , Vasculite/fisiopatologia , Adesão Celular/imunologia , Comunicação Celular/fisiologia , Células Cultivadas , Quimiocina CCL2/genética , Cromatina/fisiologia , Técnicas de Cocultura , Endotélio Vascular/citologia , Regulação da Expressão Gênica/imunologia , Humanos , Imunoprecipitação , Molécula 1 de Adesão Intercelular/genética , Monócitos/citologia , Músculo Liso Vascular/citologia , Subunidade p50 de NF-kappa B/genética , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas/fisiologia , RNA Mensageiro/análise , Estresse Mecânico , Fator de Transcrição RelA/genética , Vasculite/genética , Vasculite/imunologia
16.
J Biomech ; 37(4): 531-9, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14996565

RESUMO

Vascular endothelial cells (ECs) are constantly subjected to blood flow-induced shear stress and the influences of neighboring smooth muscle cells (SMCs). In the present study, a coculture flow system was developed to study the effect of shear stress on EC-SMC interactions. ECs and SMCs were separated by a porous membrane with only the EC side subjected to the flow condition. When ECs were exposed to a shear stress of 12 dynes/cm2 for 24 h, the cocultured SMCs tended to orient perpendicularly to the flow direction. This perpendicular orientation of the cocultured SMCs to flow direction was not observed when ECs were exposed to a shear stress of 2 dynes/cm2. Under the static condition, long and parallel actin bundles were observed in the central regions of the cocultured SMCs, whereas the actin filaments localized mainly at the periphery of the cocultured ECs. After 24 h of flow application, the cocultured ECs displayed very long, well-organized, parallel actin stress fibers aligned with the flow direction in the central regions of the cells. Immunostaining of platelet endothelial cell adhesion molecule-1 confirmed the elongation and alignment of the cocultured ECs with the flow direction. Coculture with SMCs under static condition induced EC gene expressions of growth-related oncogene-alpha and monocyte chemotactic protein-1, and shear stress was found to abolish these SMC-induced gene expressions. Our results suggest that shear stress may serve as a down-regulator for the pathophysiologically relevant gene expression in ECs cocultured with SMCs.


Assuntos
Células Endoteliais/fisiologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , Citoesqueleto de Actina/fisiologia , Quimiocina CCL2/antagonistas & inibidores , Quimiocina CCL2/genética , Técnicas de Cocultura , Células Endoteliais/metabolismo , Humanos , Técnicas Imunológicas , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Oncogenes/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Coloração e Rotulagem , Estresse Mecânico , Fatores de Tempo
17.
Arterioscler Thromb Vasc Biol ; 24(1): 73-9, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14615388

RESUMO

OBJECTIVE: Vascular endothelial cells (ECs) are subjected to shear stress and cytokine stimulation. We studied the interplay between shear stress and cytokine in modulating the expression of adhesion molecule genes in ECs. METHODS AND RESULTS: Shear stress (20 dynes/cm2) was applied to ECs prior to and/or following the addition of tumor necrosis factor (TNF)-alpha. Shear stress increased the TNF-alpha-induced expression of intercellular adhesion molecule-1 (ICAM-1) at both mRNA and surface protein levels, but decreased the TNF-alpha-induced expression of vascular adhesion molecule-1 (VCAM-1) and E-selectin. Transfection studies using promoter reporter gene constructs of ICAM-1, VCAM-1, and E-selectin demonstrated that these shear stress modulations of gene expression occur at the transcriptional levels. After 24-hour preshearing followed by 1 hour of static incubation, the effect of preshearing on TNF-alpha-induced ICAM-1 mRNA expression vanished. The recovery of the TNF-alpha-induced VCAM-1 and E-selectin mRNA expressions following preshearing, however, required a static incubation time of >6 hours (complete recovery at 24 hours). Pre- and postshearing caused a reduction in the nuclear factor-kappaB-DNA binding activity induced by TNF-alpha in the EC nucleus. CONCLUSIONS: Our findings suggest that shear stress plays differential roles in modulating the TNF-alpha-induced expressions of ICAM-1 versus VCAM-1 and E-selectin genes in ECs.


Assuntos
Selectina E/biossíntese , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Regulação da Expressão Gênica , Molécula 1 de Adesão Intercelular/biossíntese , Estresse Mecânico , Fator de Necrose Tumoral alfa/farmacologia , Molécula 1 de Adesão de Célula Vascular/biossíntese , Núcleo Celular/metabolismo , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , DNA/genética , DNA/metabolismo , Selectina E/genética , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Molécula 1 de Adesão Intercelular/genética , NF-kappa B/metabolismo , Molécula 1 de Adesão de Célula Vascular/genética
18.
J Biomech ; 36(12): 1883-95, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14614942

RESUMO

The preferential adhesion of monocytes to vascular endothelial cells (ECs) at regions near branches and curvatures of the arterial tree, where flow is disturbed, suggests that hemodynamic conditions play significant roles in monocyte adhesion. The present study aims to elucidate the effects of disturbed flow on monocyte adhesion to ECs and the adhesive properties of ECs. We applied, for the first time, the micron-resolution particle image velocimetry (microPIV) technique to analyze the characteristics of the disturbed flow produced in our vertical-step flow (VSF) chamber. The results demonstrated the existence of a higher near-wall concentration and a longer residence time of the monocytic analog THP-1 cells near the step and the reattachment point. THP-1 cells showed prominent adhesion to ECs pretreated with TNFalpha in the regions near the step and the reattachment point, but they showed virtually no adhesion to un-stimulated ECs. Pre-incubation of the TNFalpha-treated ECs with antibodies against intercellular adhesion molecule-1 (ICAM-1), vascular adhesion molecule-1 (VCAM-1), and E-selectin inhibited the THP-1 adhesion; the maximal inhibition was observed with a combination of these antibodies. Pre-exposure of ECs to disturbed flow in VSF for 24 h led to significant increases in their surface expressions of ICAM-1 and E-selectin, but not VCAM-1, and in the adhesion of THP-1 cells. Our findings demonstrate the importance of complex flow environment in modulating the adhesive properties of vascular endothelium and consequently monocyte adhesion in regions of prevalence of atherosclerotic lesions.


Assuntos
Movimento Celular/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/fisiologia , Citometria de Fluxo/métodos , Microfluídica/métodos , Monócitos/citologia , Monócitos/fisiologia , Reologia/métodos , Velocidade do Fluxo Sanguíneo , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Técnicas de Cultura de Células/métodos , Movimento Celular/efeitos dos fármacos , Selectina E , Endotélio Vascular/efeitos dos fármacos , Citometria de Fluxo/instrumentação , Hemorreologia/instrumentação , Hemorreologia/métodos , Humanos , Molécula 1 de Adesão Intercelular/metabolismo , Microfluídica/instrumentação , Reologia/instrumentação , Fator de Necrose Tumoral alfa/farmacologia , Molécula 1 de Adesão de Célula Vascular/metabolismo
19.
Chin J Physiol ; 45(4): 169-76, 2002 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-12817708

RESUMO

The interplay between shear stress and cytokines in regulating vascular endothelial function remains largely unexplored. In the present study, the potential role of shear stress in regulating tumor necrosis factor-alpha (TNF-alpha)-induced gene expression in endothelial cells (ECs) was investigated. The TNF-alpha-induced monocyte chemotactic protein-1 (MCP-1) mRNA expressions were significantly attenuated in ECs subjected to a high level of shear stress (20 dynes/cm2) for 4 or 24 h prior to the addition of TNF-alpha in the presence of flow. Less inhibition of TNF-alpha-induced MCP-1 mRNA expression was found in ECs pre-exposed to a low level of shear stress (1.2 dynes/cm2) for 24 h as compared with the cells presheared (pre-exposed to shear stress) for 4 h. Simultaneous exposure of ECs to TNF-alpha and a high or low level of shear stress down-regulated TNF-alpha-induced MCP-1 gene expressions, suggesting that the post-flow condition modulates endothelial responses to cytokine stimulation. Individually or combined, an endothelial nitric oxide synthase (eNOS) inhibitor and a glutathione (GSH) biosynthesis inhibitor had no effect on this shear stress-mediated inhibition. Moreover, in ECs either presheared or remained in a static condition prior to stimulation by TNF-alpha while under shear flow, the ability of TNF-alpha to induce AP-1-DNA binding activity in the nucleus was reduced. Our findings suggest that shear stress plays a protective role in vascular homeostasis by inhibiting endothelial responses to cytokine stimulation.


Assuntos
Antineoplásicos/farmacologia , Quimiocina CCL2/genética , Endotélio Vascular/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Núcleo Celular/metabolismo , Endotélio Vascular/citologia , Inibidores Enzimáticos/farmacologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Glutationa/metabolismo , Humanos , Monócitos/fisiologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo III , Oxirredução , Estresse Mecânico , Fator de Transcrição AP-1/metabolismo , Cordão Umbilical/citologia , ômega-N-Metilarginina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA