Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 139
Filtrar
1.
Aging (Albany NY) ; 13(23): 25241-25255, 2021 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-34887361

RESUMO

The abnormal proliferation of vascular smooth muscle cells (VSMCs) plays an important role in the development and progression of diabetic vascular complications. In high-glucose (HG) conditions, endothelial cells (ECs) act as the first barrier to damaging stimuli and trigger a multi-response, including EC and VSMC crosstalk. However, the crosstalk pathways between ECs and VSMCs under HG conditions remain unclear. This study aimed to explore the roles and underlying mechanism of exosomes derived from ECs in the crosstalk between ECs and VSMCs. Our results showed that mouse aortic endothelial cell (MAEC)-secreted exosomes could promote the proliferation and inhibit the apoptosis of VSMCs induced by HG. Furthermore, we isolated the exosomes secreted by MAECs and found that exosomes derived from MAECs that were exposed to HG could transfer circHIPK3, which is enriched in MAEC-derived exosomes, to VSMCs. Exosomal circHIPK3 promoted the proliferation and inhibited the apoptosis of VSMCs. circHIPK3 sponged miR-106a-5p to relieve its repression of forkhead box O1 (Foxo1) expression. The increased expression of Foxo1 acted as a transcription factor to promote Vcam1 expression, thus facilitating the uptake of MAEC-derived exosomes by VSMCs. The results of this study suggested that exosomal circHIPK3 derived from MAECs promotes the proliferation of VSMCs induced by HG via the miR-106a-5p/Foxo1/Vcam1 pathway.


Assuntos
Proliferação de Células , Proteína Forkhead Box O1/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , MicroRNAs/metabolismo , Músculo Liso Vascular/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Molécula 1 de Adesão de Célula Vascular/metabolismo , Animais , Apoptose , Proliferação de Células/efeitos dos fármacos , Exossomos/metabolismo , Glucose/farmacologia , Células HEK293 , Humanos , Camundongos , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/crescimento & desenvolvimento , Transdução de Sinais/efeitos dos fármacos
2.
J Biomed Mater Res A ; 109(12): 2493-2505, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34096176

RESUMO

Long-term in vivo observation in large animal model is critical for evaluating the potential of small diameter tissue engineering vascular graft (SDTEVG) in clinical application, but is rarely reported. In this study, a SDTEVG is fabricated by the electrospinning of poly(ε-caprolactone) and subsequent heparin modification. SDTEVG is implanted into canine's abdominal aorta for 511 days in order to investigate its clinical feasibility. An active and robust remodeling process was characterized by a confluent endothelium, macrophage infiltrate, extracellular matrix deposition and remodeling on the explanted graft. The immunohistochemical and immunofluorescence analysis further exhibit the regeneration of endothelium and smooth muscle layer on tunica intima and tunica media, respectively. Thus, long-term follow-up reveals viable neovessel formation beyond graft degradation. Furthermore, the von Kossa staining exhibits no occurrence of calcification. However, although no TEVG failure or rupture happens during the follow-up, the aneurysm is found by both Doppler ultrasonic and gross observation. Consequently, as-prepared TEVG shows promising potential in vascular tissue engineering if it can be appropriately strengthened to prevent the occurrence of aneurysm.


Assuntos
Prótese Vascular , Vasos Sanguíneos/transplante , Heparina/química , Poliésteres/química , Aneurisma/prevenção & controle , Animais , Aorta Abdominal/transplante , Cães , Endotélio Vascular/crescimento & desenvolvimento , Matriz Extracelular/ultraestrutura , Humanos , Macrófagos , Músculo Liso Vascular/crescimento & desenvolvimento , Projetos Piloto , Alicerces Teciduais , Túnica Íntima , Túnica Média
3.
Mol Med Rep ; 24(1)2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33982767

RESUMO

Accumulating evidence suggests that pulmonary expression of a disintegrin and metalloproteinase­33 (ADAM33) serves a key role in the pathogenesis of airway remodeling­related diseases, including asthma. Airway vascular proliferation has been recognized as a key feature of airway remodeling. Our previous study showed that ADAM33 is constitutively expressed in airway vascular smooth muscle cells in patients with asthma, suggesting a potential role of ADAM33 in regulating airway vascular remodeling. Using in vitro human aortic smooth muscle cells (HASMCs) and lentiviral vector carrying short hairpin RNA for ADAM33, the present study aimed to evaluate the influence of ADAM33 silencing on the proliferation and apoptosis of HASMCs and the underlying molecular pathways. Cellular proliferation was observed using the Cell Counting Kit­8 method. Cellular apoptosis was evaluated with Annexin V­PE/7­AAD staining and flow cytometry. Reverse transcription­quantitative PCR and western blotting were used to evaluate the changes in mRNA and protein levels of involved signaling molecules. It was found that silencing of ADAM33 expression in HASMCs significantly inhibited proliferation, but induced the apoptosis of HASMCs. These changes were accompanied by inhibition of the PI3K/AKT/ERK pathway and Bcl­2, but an increase in Bax expression. These results suggested that constitutive expression of ADAM33 may be important to maintain a proliferative phenotype in HASMCs. The influences of ADAM33 on proliferation and apoptosis of HASMCs may involve regulation of PI3K/AKT/ERK and Bax/Bcl­2 pathways. These findings suggested an important role of ADAM33 in airway vascular remodeling and potential therapeutic significance of ADAM33 inhibition in airway remodeling­related diseases.


Assuntos
Proteínas ADAM/genética , Músculo Liso Vascular/enzimologia , Músculo Liso Vascular/crescimento & desenvolvimento , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Remodelação Vascular/genética , Proteínas ADAM/biossíntese , Idoso , Apoptose/genética , Ciclo Celular/genética , Linhagem Celular , Proliferação de Células/genética , Feminino , Inativação Gênica , Humanos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/genética , Proteína X Associada a bcl-2/metabolismo
4.
Int J Mol Sci ; 21(24)2020 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-33322781

RESUMO

Amine-coated biodegradable materials based on synthetic polymers have a great potential for tissue remodeling and regeneration because of their excellent processability and bioactivity. In the present study, we have investigated the influence of various chemical compositions of amine plasma polymer (PP) coatings and the influence of the substrate morphology, represented by polystyrene culture dishes and polycaprolactone nanofibers (PCL NFs), on the behavior of vascular smooth muscle cells (VSMCs). Although all amine-PP coatings improved the initial adhesion of VSMCs, 7-day long cultivation revealed a clear preference for the coating containing about 15 at.% of nitrogen (CPA-33). The CPA-33 coating demonstrated the ideal combination of good water stability, a sufficient amine group content, and favorable surface wettability and morphology. The nanostructured morphology of amine-PP-coated PCL NFs successfully slowed the proliferation rate of VSMCs, which is essential in preventing restenosis of vascular replacements in vivo. At the same time, CPA-33-coated PCL NFs supported the continuous proliferation of VSMCs during 7-day long cultivation, with no significant increase in cytokine secretion by RAW 264.7 macrophages. The CPA-33 coating deposited on biodegradable PCL NFs therefore seems to be a promising material for manufacturing small-diameter vascular grafts, which are still lacking on the current market.


Assuntos
Aminas/química , Materiais Revestidos Biocompatíveis/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Nanofibras/química , Plasma/química , Polímeros/química , Aminas/efeitos adversos , Aminas/imunologia , Aminas/farmacologia , Animais , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Materiais Revestidos Biocompatíveis/efeitos adversos , Materiais Revestidos Biocompatíveis/química , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/crescimento & desenvolvimento , Miócitos de Músculo Liso/metabolismo , Nanofibras/efeitos adversos , Espectroscopia Fotoeletrônica , Plasma/imunologia , Poliésteres/química , Polímeros/efeitos adversos , Polímeros/farmacologia , Células RAW 264.7 , Ratos , Propriedades de Superfície/efeitos dos fármacos , Alicerces Teciduais/efeitos adversos , Alicerces Teciduais/química
5.
Sci Rep ; 10(1): 13858, 2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32807822

RESUMO

The aim of this study is to investigate the therapeutic role of Tanshinone II A, a key integrant from salvia miltiorrhiza, against pathological vascular remodeling. Completed ligation of mouse left common carotid arteries animal model and rat smooth muscle cells used to investigate the role of Tanshinone II A in regulating pathological vascular remodeling through hematoxylin and eosin staining, immunohistochemistry staining, immunofluorescence staining, adenovirus infection, real time PCR and western blotting. Our data demonstrated that Tanshinone II A treatment suppresses vascular injury-induced neointima formation. In vitro studies on rat smooth muscle cell indicated that Tanshinone II A treatment attenuates PDGF-BB induced cell growth, and promotes smooth muscle cell differentiated marker genes expression that induced by rapamycin treatment. Tanshinone II A treatment significant inhibits rat smooth muscle cell proliferation and migration. Tanshinone II A promotes KLF4 expression during smooth muscle phenotypic switching. Overexpression of KLF4 exacerbates Tanshinone II A mediated smooth muscle cell growth inhibition. Tanshinone II A plays a pivotal role in regulating pathological vascular remodeling through KLF4 mediated smooth muscle cell phenotypic switching. This study demonstrated that Tanshinone II A is a potential therapeutic agent for vascular diseases.


Assuntos
Abietanos/farmacologia , Diferenciação Celular/genética , Expressão Gênica/efeitos dos fármacos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Músculo Liso Vascular/crescimento & desenvolvimento , Remodelação Vascular/efeitos dos fármacos , Abietanos/uso terapêutico , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Fator 4 Semelhante a Kruppel , Camundongos , Fenótipo , Fitoterapia , Ratos Sprague-Dawley , Salvia miltiorrhiza , Doenças Vasculares/tratamento farmacológico
6.
Mol Med Rep ; 22(2): 886-894, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32467985

RESUMO

Increasing evidence suggests that T­cell immunoglobulin and mucin domain 3 (TIM­3) displays anti­atherosclerotic effects, but its role in vascular smooth muscle cells (VSMCs) has not been reported. The present study aimed to investigate the function of TIM­3 and its roles in human artery VSMCs (HASMCs). A protein array was used to investigate the TIM­3 protein expression profile, which indicated that TIM­3 expression was increased in the serum of patients with lower extremity arteriosclerosis obliterans disease (LEAOD) compared with healthy individuals. Immunohistochemistry and western blotting of arterial tissue further revealed that TIM­3 expression was increased in LEAOD artery tissue compared with normal artery tissue. Additionally, platelet­derived growth factor­BB (PDGF­BB) displayed a positive correlation with TIM­3 expression in HASMCs. TIM­3 decreased the migration and proliferation of PDGF­BB­induced HASMCs, and anti­TIM­3 blocked the effects of TIM­3. The effect of TIM­3 on the proliferation and migration of HASMCs was further investigated using LV­TIM­3­transduced cells. The results revealed that TIM­3 also inhibited PDGF­BB­induced expression of the inflammatory factors interleukin­6 and tumor necrosis factor­α by suppressing NF­κB activation. In summary, the present study revealed that TIM­3 displayed a regulatory role during the PDGF­BB­induced inflammatory reaction in HASMCs, which indicated that TIM­3 may display anti­atherosclerotic effects.


Assuntos
Artérias/metabolismo , Aterosclerose/metabolismo , Becaplermina/antagonistas & inibidores , Receptor Celular 2 do Vírus da Hepatite A/biossíntese , Receptor Celular 2 do Vírus da Hepatite A/sangue , Músculo Liso Vascular/metabolismo , Idoso , Artérias/citologia , Artérias/crescimento & desenvolvimento , Arteriosclerose Obliterante/sangue , Aterosclerose/induzido quimicamente , Becaplermina/efeitos adversos , Linhagem Celular , Movimento Celular , Proliferação de Células , Feminino , Humanos , Interleucina-6/metabolismo , Extremidade Inferior/irrigação sanguínea , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Músculo Liso Vascular/crescimento & desenvolvimento , NF-kappa B/metabolismo , Análise Serial de Proteínas , Transcriptoma , Fator de Necrose Tumoral alfa/metabolismo
7.
Cell Death Differ ; 26(12): 2790-2806, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31024075

RESUMO

TEAD1 (TEA domain transcription factor 1), a transcription factor known for the functional output of Hippo signaling, is important for tumorigenesis. However, the role of TEAD1 in the development of vascular smooth muscle cell (VSMC) is unknown. To investigate cell-specific role of Tead1, we generated cardiomyocyte (CMC) and VSMC-specific Tead1 knockout mice. We found CMC/VSMC-specific deletion of Tead1 led to embryonic lethality by E14.5 in mice due to hypoplastic cardiac and vascular walls, as a result of impaired CMC and VSMC proliferation. Whole transcriptome analysis revealed that deletion of Tead1 in CMCs/VSMCs downregulated expression of muscle contractile genes and key transcription factors including Pitx2c and myocardin. In vitro studies demonstrated that PITX2c and myocardin rescued TEAD1-dependent defects in VSMC differentiation. We further identified Pitx2c as a novel transcriptional target of TEAD1, and PITX2c exhibited functional synergy with myocardin by directly interacting with myocardin, leading to augment the differentiation of VSMC. In summary, our study reveals a critical role of Tead1 in cardiovascular development in mice, but also identifies a novel regulatory mechanism, whereby Tead1 functions upstream of the genetic regulatory hierarchy for establishing smooth muscle contractile phenotype.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Músculo Liso Vascular/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/genética , Feminino , Deleção de Genes , Masculino , Camundongos , Camundongos Knockout , Músculo Liso Vascular/citologia , Músculo Liso Vascular/crescimento & desenvolvimento , Fatores de Transcrição de Domínio TEA , Fatores de Transcrição/genética
8.
Am J Physiol Lung Cell Mol Physiol ; 316(6): L999-L1012, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30908936

RESUMO

Lung diseases with hypoxia are complicated by pulmonary hypertension, leading to heart failure and death. No pharmacological treatment exists. Increased proinflammatory cytokines are found in hypoxic patients, suggesting an inflammatory pathogenesis. Caspase-1, the effector of the inflammasome, mediates inflammation through activation of the proinflammatory cytokines interleukin (IL)-18 and IL-1ß. Here, we investigate inflammasome-related mechanisms that can trigger hypoxia-induced pulmonary hypertension. Our aim was to examine whether caspase-1 induces development of hypoxia-related pulmonary hypertension and is a suitable target for therapy. Wild-type (WT) and caspase-1-/- mice were exposed to 10% oxygen for 14 days. Hypoxic caspase-1-/- mice showed lower pressure and reduced muscularization in pulmonary arteries, as well as reduced right ventricular remodeling compared with WT. Smooth muscle cell (SMC) proliferation was reduced in caspase-1-deficient pulmonary arteries and in WT arteries treated with a caspase-1 inhibitor. Impaired inflammation was shown in hypoxic caspase-1-/- mice by abolished pulmonary influx of immune cells and lower levels of IL-18, IL-1ß, and IL-6, which were also reduced in the medium surrounding caspase-1 abrogated pulmonary arteries. By adding IL-18 or IL-1ß to caspase-1-deficient pulmonary arteries, SMC proliferation was retained. Furthermore, inhibition of both IL-6 and phosphorylated STAT3 reduced proliferation of SMC in vitro, indicating IL-18, IL-6, and STAT3 as downstream mediators of caspase-1-induced SMC proliferation in pulmonary arteries. Caspase-1 induces SMC proliferation in pulmonary arteries through the caspase-1/IL-18/IL-6/STAT3 pathway, leading to pulmonary hypertension in mice exposed to hypoxia. We propose that caspase-1 inhibition is a potential target for treatment of pulmonary hypertension.


Assuntos
Caspase 1/genética , Hipóxia Celular/fisiologia , Hipertensão Pulmonar/patologia , Miócitos de Músculo Liso/fisiologia , Função Ventricular Direita/fisiologia , Animais , Linhagem Celular , Proliferação de Células/genética , Humanos , Inflamassomos/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/citologia , Músculo Liso Vascular/crescimento & desenvolvimento , Artéria Pulmonar/citologia , Artéria Pulmonar/patologia , Fator de Transcrição STAT3/metabolismo
9.
J Anat ; 234(4): 465-479, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30793310

RESUMO

Teeth are richly supported by blood vessels and peripheral nerves. The aim of this study was to describe in detail the developmental time-course and localization of blood vessels during early tooth formation and to compare that to innervation, as well as to address the putative role of vascular endothelial growth factor (VEGF), which is an essential regulator of vasculature development, in this process. The localization of blood vessels and neurites was compared using double immunofluorescence staining on sections at consecutive stages of the embryonic (E) and postnatal (PN) mandibular first molar tooth germ (E11-PN7). Cellular mRNA expression domains of VEGF and its signaling receptor VEGFR2 were studied using sectional radioactive in situ hybridization. Expression of VEGF mRNA and the encoded protein were studied by RT-PCR and western blot analysis, respectively, in the cap and early bell stage tooth germs, respectively. VEGFR2 was immunolocalized on tooth tissue sections. Smooth muscle cells were investigated by anti-alpha smooth muscle actin (αSMA) antibodies. VEGF showed developmentally regulated epithelial and mesenchymal mRNA expression domains including the enamel knot signaling centers that correlated with the growth and navigation of the blood vessels expressing Vegfr2 and VEGFR2 to the dental papilla and enamel organ. Developing blood vessels were present in the jaw mesenchyme including the presumptive dental mesenchyme before the appearance of the epithelial dental placode and dental neurites. Similarly, formation of a blood vessel plexus around the bud stage tooth germ and ingrowth of vessels into dental papilla at E14 preceded ingrowth of neurites. Subsequently, pioneer blood vessels in the dental papilla started to receive smooth muscle coverage at the early embryonic bell stage. Establishment and patterning of the blood vessels and nerves during tooth formation are developmentally regulated, stepwise processes that likely involve differential patterning mechanisms. Development of tooth vascular supply is proposed to be regulated by local, tooth-specific regulation by epithelial-mesenchymal tissue interactions and involving tooth target expressed VEGF signaling. Further investigations on tooth vascular development by local VEGF signaling, as well as how tooth innervation and development of blood vessels are integrated with advancing tooth organ formation by local signaling mechanisms, are warranted.


Assuntos
Vasos Sanguíneos/crescimento & desenvolvimento , Dente , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Animais , Padronização Corporal , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Músculo Liso Vascular/crescimento & desenvolvimento , Odontogênese , Transdução de Sinais , Dente/embriologia , Dente/inervação , Dente/metabolismo , Germe de Dente/embriologia , Germe de Dente/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
10.
IUBMB Life ; 71(1): 125-133, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30291803

RESUMO

The long-term failure of vein grafts due to neointimal hyperplasia remains a difficult problem in cardiovascular surgery. Exploring novel approaches to prevent neointimal hyperplasia is important. MicroRNA-146a (miR-146a) plays an essential role in promoting vascular smooth muscle cell (VSMC) proliferation. Thus, the aim of the present study is to investigate whether adenovirus-mediated miR-146a sponge (Ad-miR-146a-SP) gene therapy could attenuate neointimal formation in rat vein grafts. (Ad-miR-146a-SP) was constructed to transfect cultured VSMCs and grafted veins. To improve the efficiency of transferring the miR-146a sponge gene into the grafted veins, 20% poloxamer F-127 gel incorporated with 0.25% trypsin was used to increase adenovirus contact time and penetration. miR-146a-SP transduction significantly reduced the expression of miR-146a both in cultured VSMCs and vein grafts. miR-146a sponge markedly attenuated VSMC proliferation and migration. Consistent with this, miR-146a sponge gene therapy significantly attenuated neointimal formation and also improved blood flow in the vein grafts. Mechanistically, we identified the Krüppel-like factor 4(KLF4) as a potential downstream target gene of miR-146a in vein grafts. Our data show that miR-146a sponge gene therapy could effectively reduce miR-146a activity and attenuate neointimal formation in vein grafts, suggesting its potential therapeutic application for prevention of vein graft failure. © 2018 IUBMB Life, 71(1):125-133, 2019.


Assuntos
Terapia Genética , MicroRNAs/genética , Neointima/terapia , Veias/crescimento & desenvolvimento , Adenoviridae/genética , Animais , Prótese Vascular , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Masculino , MicroRNAs/farmacologia , Músculo Liso Vascular/crescimento & desenvolvimento , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Neointima/genética , Ratos , Veias/fisiopatologia
11.
Am J Physiol Cell Physiol ; 315(6): C850-C862, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30257103

RESUMO

Vascular smooth muscle cell (VSMC) migration contributes to neointimal formation after vascular injury. We previously demonstrated that copper (Cu) transporter ATP7A is involved in platelet-derived growth factor (PDGF)-induced VSMC migration in a Cu- and Rac1-dependent manner. The underlying mechanism is still unknown. Here we show that ATP7A interacts with IQGAP1, a Rac1 and receptor tyrosine kinase binding scaffolding proteins, which mediates PDGF-induced VSMC migration and vascular remodeling. In cultured rat aortic SMCs, PDGF stimulation rapidly promoted ATP7A association with IQGAP1 and Rac1 and their translocation to the lipid rafts and leading edge. Cotransfection assay revealed that ATP7A directly bound to NH2-terminal domain of IQGAP1. Functionally, either ATP7A or IQGAP1 depletion using siRNA significantly inhibited PDGF-induced VSMC migration without additive effects, suggesting that IQGAP1 and ATP7A are in the same axis to promote migration. Furthermore, IQGAP1 siRNA blocked PDGF-induced ATP7A association with Rac1 as well as its translocation to leading edge, while PDGF-induced IQGAP1 translocation was not affected by ATP7A siRNA or Cu chelator. Overexpression of mutant IQGAP1 lacking a Rac1 binding site prevented PDGF-induced translocation of Rac1, but not ATP7A, to the leading edge, thereby inhibiting lamellipodia formation and VSMC migration. In vivo, ATP7A colocalized with IQGAP1 at neointimal VSMCs in a mice wire injury model, while neointimal formation and extracellular matrix deposition induced by vascular injury were inhibited in ATP7A mutant mice with reduced Cu transporter function. In summary, IQGAP1 functions as ATP7A and Rac1 binding scaffolding protein to organize PDGF-dependent ATP7A translocation to the lamellipodial leading edge, thereby promoting VSMC migration and vascular remodeling.


Assuntos
ATPases Transportadoras de Cobre/genética , Fator de Crescimento Derivado de Plaquetas/genética , Remodelação Vascular/genética , Proteínas rac1 de Ligação ao GTP/genética , Proteínas Ativadoras de ras GTPase/genética , Animais , Aorta/citologia , Aorta/metabolismo , Movimento Celular/genética , Cobre/química , Cobre/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Microdomínios da Membrana/genética , Microdomínios da Membrana/metabolismo , Camundongos , Músculo Liso Vascular/crescimento & desenvolvimento , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Neointima/genética , Fosforilação , Ligação Proteica , Ratos
12.
Exp Biol Med (Maywood) ; 242(5): 487-496, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28056547

RESUMO

Vascular remodeling, a pathogenic hallmark in pulmonary hypertension, is mainly driven by a dysbalance between proliferation and apoptosis of human pulmonary artery smooth muscle cells. It has previously been shown that microRNAs are involved in the pathogenesis of pulmonary hypertension. However, the role of long noncoding RNAs has not been evaluated. long noncoding RNA expression was quantified in human pulmonary artery smooth muscle cells using PCR arrays and quantitative PCR. Knockdown of genes was performed by transfection of siRNA or GapmeR. Proliferation and migration were measured using BrdU incorporation and wound healing assays. The mouse model of hypoxia-induced PH was used to determine the physiological meaning of identified long noncoding RNAs. The expression of 84 selected long noncoding RNAs was assessed in hypoxic human pulmonary artery smooth muscle cells and the levels of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) were significantly increased. Depletion of hypoxia-inducible factor 1α abolished the hypoxia-induced upregulation of metastasis-associated lung adenocarcinoma transcript 1 expression. Silencing of MALAT1 significantly decreased proliferation and migration of human pulmonary artery smooth muscle cells. In vivo, MALAT1 expression was significantly increased in lungs of hypoxic mice. Of note, targeting of MALAT1 by GapmeR ameliorated heart hypertrophy in mice with pulmonary hypertension. This is the first report on functional characterization of MALAT1 in the pulmonary vasculature. Our data provide evidence that MALAT1 expression is significantly increased by hypoxia, probably by hypoxia-inducible factor 1α. Intervention experiments confirmed that MALAT1 regulates the proliferative phenotype of smooth muscle cells and silencing of MALAT1 reduced heart hypertrophy in mice with pulmonary hypertension. These data indicate a potential role of MALAT1 in the pathogenesis of pulmonary hypertension. Impact statement Metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is a long noncoding RNA that mediates several biological processes. In the context of vascular biology, MALAT1 has been shown to be inducible by hypoxia and to control cell proliferation. These processes are of major importance for the pathophysiology of hypoxia-induced pulmonary hypertension (PH). Until now, the physiological role of MALAT1 in PH remains unclear. By using smooth muscle cells and by employing an established PH mouse model, we provide evidence that hypoxia induces MALAT1 expression. Moreover, depletion of MALAT1 inhibited migration and proliferation of smooth muscle cells, probably by the induction of cyclin-dependent kinase inhibitors. Of note, MALAT1 was significantly increased in mice exposed to hypoxia and silencing of MALAT1 ameliorated heart hypertrophy in mice with hypoxia-induced PH. Since vascular remodeling and right heart failure as a consequence of pulmonary pressure overload is a major problem in PH, these data have implications for our pathogenetic understanding.


Assuntos
Proliferação de Células/fisiologia , Músculo Liso Vascular/crescimento & desenvolvimento , RNA Longo não Codificante/fisiologia , RNA não Traduzido/fisiologia , Animais , Western Blotting , Células Cultivadas , Humanos , Hipóxia/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/fisiologia , Artéria Pulmonar/citologia , Reação em Cadeia da Polimerase em Tempo Real
13.
Mol Med Rep ; 14(5): 4551-4558, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27748810

RESUMO

Vascular smooth muscle cell (VSMC) proliferation is a key event in the development of in­stent restenosis. Evodiamine is an indole alkaloid extracted from the Chinese medicine, evodia, and has been shown to inhibit tumor cell proliferation and protect the cardiovascular system. However, whether evodiamine affects VSMC proliferation remains to be elucidated. Therefore, the present study examined the effects and the mechanisms of action of evodiamine on the proliferation of rat VSMCs. The cells were treated with evodiamine alone or in combination with platelet­derived growth factor­BB (PDGF­BB) stimulation. It was found that evodiamine inhibited PDGF­BB­induced VSMC proliferation in a dose­dependent manner, without inducing cell death. Evodiamine also retarded cell cycle progression, evidenced by the suppression of the expression of cell cycle­promoting cyclin proteins and cyclin­dependent kinases. In addition, evodiamine attenuated the PDGF­BB­induced phosphorylation of mitogen­activated protein kinases p38 and extracellular signal­regulated kinases 1/2, however, it had no effect on the phosphorylation of Akt. Evodiamine also inhibited the increase of reactive oxygen species generation and upregulated the mRNA expression levels of genes encoding antioxidant enzymes. These findings provide important insights into the mechanisms underlying the vasoprotective actions of evodiamine and suggest that it may be a useful therapeutic agent for the treatment of vascular occlusive disease.


Assuntos
Proliferação de Células/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Quinazolinas/administração & dosagem , Animais , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Evodia/química , Regulação da Expressão Gênica/efeitos dos fármacos , Medicina Tradicional Chinesa , Músculo Liso Vascular/crescimento & desenvolvimento , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-sis/administração & dosagem , Proteínas Proto-Oncogênicas c-sis/metabolismo , Ratos , Espécies Reativas de Oxigênio/metabolismo , Doenças Vasculares/tratamento farmacológico , Doenças Vasculares/genética , Doenças Vasculares/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/genética
14.
Hypertension ; 68(5): 1225-1235, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27698063

RESUMO

Estrogens are important regulators of cardiovascular function. Some of estrogen's cardiovascular effects are mediated by a G-protein-coupled receptor mechanism, namely, G-protein-coupled estrogen receptor (GPER). Estradiol-mediated regulation of vascular cell programmed cell death reflects the balance of the opposing actions of GPER versus estrogen receptor α (ERα). However, the significance of these opposing actions on the regulation of vascular smooth muscle cell proliferation or migration in vitro is unclear, and the significance in vivo is unknown. To determine the effects of GPER activation in vitro, we studied rat aortic vascular smooth muscle cells maintained in primary culture. GPER was reintroduced using adenoviral gene transfer. Both estradiol and G1, a GPER agonist, inhibited both proliferation and cell migration effects that were blocked by the GPER antagonist, G15. To determine the importance of the GPER-ERα balance in regulating vascular remodeling in a rat model of carotid ligation, we studied the effects of upregulation of GPER expression versus downregulation of ERα. Reintroduction of GPER significantly attenuated the extent of medial hypertrophy and attenuated the extent of CD45 labeling. Downregulation of ERα expression comparably attenuated the extent of medial hypertrophy and inflammation after carotid ligation. These studies demonstrate that the balance between GPER and ERα regulates vascular remodeling. Receptor-specific modulation of estrogen's effects may be an important new approach in modifying vascular remodeling in both acute settings like vascular injury and perhaps in longer term regulation like in hypertension.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Regulação da Expressão Gênica , Músculo Liso Vascular/crescimento & desenvolvimento , Receptores Acoplados a Proteínas G/genética , Remodelação Vascular/genética , Análise de Variância , Animais , Artérias Carótidas/cirurgia , Movimento Celular/genética , Proliferação de Células/genética , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Ligadura/métodos , Masculino , Músculo Liso Vascular/citologia , Valor Preditivo dos Testes , RNA Mensageiro/genética , Distribuição Aleatória , Ratos , Ratos Wistar , Regulação para Cima
15.
Usp Fiziol Nauk ; 47(2): 45-61, 2016.
Artigo em Russo | MEDLINE | ID: mdl-27530043

RESUMO

Literature and our data suggest the regulatory action of a number of biologically active substances (catecholamines, cardiac glycosides, ß-blockers, angiotensin-converting-enzyme inhibitor) on the growth and proliferation of heart cells. By using of organotypic tissue culture has proved that the basis of this regulation is the ability of test substances, receptor- or transducer-mediated signaling to modulate the function of Na⁺, K⁺-ATPase. There is a delay in the development of vascular smooth muscle in the late postnatal period in rats with the blockade of the sympathetic nervous system in the prenatal period. The relationship between vascular remodeling and contractile activity is described. It seems that one of the causes of high blood pressure is a remodeling of the cardiovascular system, which precedes the development of hypertension.


Assuntos
Sistema Cardiovascular/fisiopatologia , Hipertensão/fisiopatologia , Miócitos Cardíacos/metabolismo , Sistema Nervoso Simpático/fisiopatologia , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Animais , Sistema Cardiovascular/crescimento & desenvolvimento , Coração/fisiopatologia , Humanos , Hipertensão/tratamento farmacológico , Hipertensão/metabolismo , Músculo Liso Vascular/crescimento & desenvolvimento , Músculo Liso Vascular/fisiopatologia , Miócitos Cardíacos/patologia , Ratos , ATPase Trocadora de Sódio-Potássio/metabolismo
16.
Ann Anat ; 201: 79-90, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26232584

RESUMO

The porcine aorta is often used in studies on morphology, pathology, transplantation surgery, vascular and endovascular surgery, and biomechanics of the large arteries. Using quantitative histology and stereology, we estimated the area fraction of elastin, collagen, alpha-smooth muscle actin, vimentin, and desmin within the tunica media in 123 tissue samples collected from five segments (thoracic ascending aorta; aortic arch; thoracic descending aorta; suprarenal abdominal aorta; and infrarenal abdominal aorta) of porcine aortae from growing domestic pigs (n=25), ranging in age from 0 to 230 days. The descending thoracic aorta had the greatest elastin fraction, which decreased proximally toward the aortic arch as well as distally toward the abdominal aorta. Abdominal aortic segments had the highest fraction of actin, desmin, and vimentin positivity and all of these vascular smooth muscle markers were lower in the thoracic aortic segments. No quantitative differences were found when comparing the suprarenal abdominal segments with the infrarenal abdominal segments. The area fraction of actin within the media was comparable in all age groups and it was proportional to the postnatal growth. Thicker aortic segments had more elastin and collagen with fewer contractile cells. The collagen fraction decreased from ascending aorta and aortic arch toward the descending aorta. By revealing the variability of the quantitative composition of the porcine aorta, the results are suitable for planning experiments with the porcine aorta as a model, i.e. power test analyses and estimating the number of samples necessary to achieving a desirable level of precision. The complete primary morphometric data, in the form of continuous variables, are made publicly available for biomechanical modeling of site-dependent distensibility and compliance of the porcine aorta.


Assuntos
Envelhecimento/fisiologia , Aorta/crescimento & desenvolvimento , Aorta/ultraestrutura , Colágeno/metabolismo , Elastina/metabolismo , Músculo Liso Vascular/crescimento & desenvolvimento , Músculo Liso Vascular/ultraestrutura , Túnica Média/crescimento & desenvolvimento , Túnica Média/ultraestrutura , Actinas/metabolismo , Animais , Animais Recém-Nascidos , Aorta Abdominal/crescimento & desenvolvimento , Aorta Abdominal/ultraestrutura , Aorta Torácica/crescimento & desenvolvimento , Aorta Torácica/ultraestrutura , Desmina/metabolismo , Imuno-Histoquímica , Contração Muscular/fisiologia , Sus scrofa , Suínos , Vimentina/metabolismo
17.
Biochem Soc Trans ; 43(3): 495-501, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26009197

RESUMO

Pannexin channels are newly discovered ATP release channels expressed throughout the body. Pannexin 1 (Panx1) channels have become of great interest as they appear to participate in a multitude of signalling cascades, including regulation of vascular function. Although numerous Panx1 pharmacological inhibitors have been discovered, these inhibitors are not specific for Panx1 and have additional effects on other proteins. Therefore, molecular tools, such as RNA interference and knockout animals, are needed to demonstrate the role of pannexins in various cellular functions. This review focuses on the known roles of Panx1 related to purinergic signalling in the vasculature focusing on post-translational modifications and channel gating mechanisms that may participate in the regulated release of ATP.


Assuntos
Conexinas/genética , Músculo Liso Vascular/metabolismo , Proteínas do Tecido Nervoso/genética , Processamento de Proteína Pós-Traducional/genética , Receptores Purinérgicos/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Conexinas/metabolismo , Humanos , Músculo Liso Vascular/crescimento & desenvolvimento , Proteínas do Tecido Nervoso/metabolismo , Interferência de RNA , Receptores Purinérgicos/genética , Transdução de Sinais
18.
Int J Med Sci ; 11(8): 765-70, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24936138

RESUMO

MicroRNA-365 (miR-365) plays crucial roles in regulating cell proliferation, apoptosis and differentiation in various cell types. However, its function in vascular smooth muscle cells (VSMCs) is largely unknown. In our study, we found miR-365 was highly expressed in adult rat carotid arteries, but was significantly decreased in rat carotid arteries after balloon injury, a process involving neointima formation and VSMC proliferation. In vitro, the miR-365 significantly inhibited cell proliferation of isolated primary rat aortic VSMCs. Furthermore, we identified that cyclin D1 was a direct target of miR-365 in VSMCs. The miR-365 suppressed cyclin D1 expression on both mRNA and protein level. Luciferase reporter assay demonstrated that miR-365 inhibited cyclin D1 through targeting its 3'UTR. Importantly, cyclin D1 overexpression rescued the inhibitory effect of miR-365 on VSMCs proliferation. Taken together, by our studies, we identified a new MicroRNA, miR-365, involving in the pathological process of vascular injury, which inhibits VSMC proliferation through targeting cyclinD1.


Assuntos
Diferenciação Celular/genética , Proliferação de Células/genética , Ciclina D1/genética , MicroRNAs/genética , Animais , Apoptose/genética , Artérias Carótidas/citologia , Artérias Carótidas/metabolismo , Ciclina D1/antagonistas & inibidores , Músculo Liso Vascular/crescimento & desenvolvimento , Músculo Liso Vascular/metabolismo , Neointima/genética , Ratos , Transdução de Sinais
19.
J Cell Biochem ; 115(10): 1752-61, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24819721

RESUMO

Abnormal proliferation of vascular smooth muscle cells (VSMCs) is a common feature of disease progression in atherosclerosis. Cell proliferation is regulated by cell cycle regulatory proteins. MicroRNAs (miR) have been reported to act as important gene regulators and play essential roles in the proliferation and migration of VSMCs in a cardiovascular disease. However, the roles and mechanisms of miRs in VSMCs and neointimal formation are far from being fully understood. In this study, cell cycle-specific cyclin D1 was found to be a potential target of miR-365 by direct binding. Through an in vitro experiment, we showed that exogenous miR-365 overexpression reduced VSMC proliferation and proliferating cell nuclear antigen (PCNA) expression, while miR-365 was observed to block G1/S transition in platelet-derived growth factor-bb (PDGF-bb)-induced VSMCs. In addition, the proliferation of VSMCs by various stimuli, including PDGF-bb, angiotensin II (Ang II), and serum, led to the downregulation of miR-365 expression levels. The expression of miR-365 was confirmed in balloon-injured carotid arteries. Taken together, our results suggest an anti-proliferative role for miR-365 in VSMC proliferation, at least partly via modulating the expression of cyclin D1. Therefore, miR-365 may influence neointimal formation in atherosclerosis patients.


Assuntos
Aterosclerose/patologia , Ciclina D1/biossíntese , MicroRNAs/genética , Músculo Liso Vascular/crescimento & desenvolvimento , Neointima/genética , Angiotensina II/farmacologia , Animais , Becaplermina , Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/metabolismo , Divisão Celular/genética , Movimento Celular/genética , Proliferação de Células/genética , Células Cultivadas , Regulação para Baixo , MicroRNAs/biossíntese , Músculo Liso Vascular/citologia , Antígeno Nuclear de Célula em Proliferação/biossíntese , Ligação Proteica , Proteínas Proto-Oncogênicas c-sis/farmacologia , Proteínas de Ligação a RNA , Ratos , Pontos de Checagem da Fase S do Ciclo Celular/genética
20.
Cell Physiol Biochem ; 34(6): 2049-60, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25562153

RESUMO

BACKGROUND: Vascular calcification (VC), in which vascular smooth muscle cells (VSMCs) undergo a phenotypic transformation into osteoblast-like cells, is one of the emergent risk factors for the accelerated atherosclerosis process characteristic of chronic kidney disease (CKD). Phosphate is an important regulator of VC. METHODS: The expression of different smooth muscle cell or osteogenesis markers in response to high concentrations of phosphate or exogenous bone morphogenetic protein 2 (BMP-2) was examined by qRT-PCR and western blotting in rat VSMCs. Osteocalcin secretion was measured by radioimmunoassay. Differentiation and calcification of VSMCs were examined by alkaline phosphatase (ALP) activity assay and Alizarin staining. Short hairpin RNA-mediated silencing of ß-catenin was performed to examine the involvement of Wnt/ß-catenin signaling in VSMC calcification and osteoblastic differentiation induced by high phosphate or BMP-2. Apoptosis was determined by TUNEL assay and immunofluorescence imaging. RESULTS: BMP-2 serum levels were significantly higher in CKD patients than in controls. High phosphate concentrations and BMP-2 induced VSMC apoptosis and upregulated the expression of ß-catenin, Msx2, Runx2 and the phosphate cotransporter Pit1, whereas a BMP-2 neutralization antibody reversed these effects. Knockdown of ß-catenin abolished the effect of high phosphate and BMP-2 on VSMC apoptosis and calcification. CONCLUSIONS: BMP-2 plays a crucial role in calcium deposition in VSMCs and VC in CKD patients via a mechanism involving the Wnt/ß-catenin pathway.


Assuntos
Proteína Morfogenética Óssea 2/biossíntese , Calcinose/genética , Insuficiência Renal Crônica/genética , beta Catenina/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Anticorpos Neutralizantes/administração & dosagem , Apoptose/genética , Proteína Morfogenética Óssea 2/antagonistas & inibidores , Diferenciação Celular/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/crescimento & desenvolvimento , Músculo Liso Vascular/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patologia , Ratos , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética , beta Catenina/antagonistas & inibidores , beta Catenina/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA