Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Lipid Res ; 65(2): 100499, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38218337

RESUMO

Ferroptosis is a novel cell death mechanism that is mediated by iron-dependent lipid peroxidation. It may be involved in atherosclerosis development. Products of phospholipid oxidation play a key role in atherosclerosis. 1-palmitoyl-2-glutaroyl-sn-glycero-3-phosphocholine (PGPC) is a phospholipid oxidation product present in atherosclerotic lesions. It remains unclear whether PGPC causes atherosclerosis by inducing endothelial cell ferroptosis. In this study, human umbilical vein endothelial cells (HUVECs) were treated with PGPC. Intracellular levels of ferrous iron, lipid peroxidation, superoxide anions (O2•-), and glutathione were detected, and expression of fatty acid binding protein-3 (FABP3), glutathione peroxidase 4 (GPX4), and CD36 were measured. Additionally, the mitochondrial membrane potential (MMP) was determined. Aortas from C57BL6 mice were isolated for vasodilation testing. Results showed that PGPC increased ferrous iron levels, the production of lipid peroxidation and O2•-, and FABP3 expression. However, PGPC inhibited the expression of GPX4 and glutathione production and destroyed normal MMP. These effects were also blocked by ferrostatin-1, an inhibitor of ferroptosis. FABP3 silencing significantly reversed the effect of PGPC. Furthermore, PGPC stimulated CD36 expression. Conversely, CD36 silencing reversed the effects of PGPC, including PGPC-induced FABP3 expression. Importantly, E06, a direct inhibitor of the oxidized 1-palmitoyl-2-arachidonoyl-phosphatidylcholine IgM natural antibody, inhibited the effects of PGPC. Finally, PGPC impaired endothelium-dependent vasodilation, ferrostatin-1 or FABP3 inhibitors inhibited this impairment. Our data demonstrate that PGPC impairs endothelial function by inducing endothelial cell ferroptosis through the CD36 receptor to increase FABP3 expression. Our findings provide new insights into the mechanisms of atherosclerosis and a therapeutic target for atherosclerosis.


Assuntos
Aterosclerose , Cicloexilaminas , Ferroptose , Fenilenodiaminas , Animais , Camundongos , Humanos , Fosfolipídeos , Fosforilcolina , Éteres Fosfolipídicos/metabolismo , Éteres Fosfolipídicos/farmacologia , Camundongos Endogâmicos C57BL , Células Endoteliais da Veia Umbilical Humana/metabolismo , Endotélio/metabolismo , Glutationa/metabolismo , Ferro/metabolismo , Proteína 3 Ligante de Ácido Graxo
2.
J Mol Cell Cardiol ; 173: 154-168, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36367517

RESUMO

Vascular calcification is an important risk factor for cardiovascular events, accompanied by DNA damage during the process. The sirtuin 6 (SIRT6) has been reported to alleviate atherosclerosis, which is related to the reduction of DNA damage. However, whether smooth muscle cell SIRT6 mediates vascular calcification involving DNA damage remains unclear. Western blot and immunofluorescence revealed that SIRT6 expression was decreased in human vascular smooth muscle cells (HVSMCs), human and mouse arteries during vascular calcification. Alizarin red staining and calcium content assay showed that knockdown or deletion of SIRT6 significantly promoted HVSMC calcification induced by high phosphorus and calcium, accompanied by upregulation of osteogenic differentiation markers including Runx2 and BMP2. By contrast, adenovirus-mediated SIRT6 overexpression attenuated osteogenic differentiation and calcification of HVSMCs. Moreover, ex vivo study revealed that SIRT6 overexpression inhibited calcification of mouse and human arterial rings. Of note, smooth muscle cell-specific knockout of SIRT6 markedly aggravated Vitamin D3-induced aortic calcification in mice. Mechanistically, overexpression of SIRT6 reduced DNA damage and upregulated p-ATM during HVSMCs calcification, whereas knockdown of SIRT6 showed the opposite effects. Knockdown of ATM in HVSMCs abrogated the inhibitory effect of SIRT6 overexpression on calcification and DNA damage. This study for the first time demonstrates that vascular smooth muscle cell-specific deletion of SIRT6 facilitates vascular calcification via suppression of DNA damage repair. Therefore, modulation of SIRT6 and DNA damage repair may represent a therapeutic strategy for vascular calcification.


Assuntos
Sirtuínas , Calcificação Vascular , Humanos , Cálcio/metabolismo , Dano ao DNA , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Osteogênese/genética , Sirtuínas/genética , Sirtuínas/metabolismo , Calcificação Vascular/genética , Reparo do DNA
3.
Kidney Int ; 102(6): 1259-1275, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36063875

RESUMO

Vascular calcification is a common pathologic condition in patients with chronic kidney disease (CKD). Cell death such as apoptosis plays a critical role in vascular calcification. Ferroptosis is a type of iron-catalyzed and regulated cell death resulting from excessive iron-dependent reactive oxygen species and lipid peroxidation. However, it is unclear whether ferroptosis of vascular smooth muscle cells (VSMCs) regulates vascular calcification in CKD. Our results showed that high calcium and phosphate concentrations induced ferroptosis in rat VSMCs in vitro. Inhibition of ferroptosis by ferrostatin-1 dose-dependently reduced mineral deposition in rat VSMCs under pro-osteogenic conditions, as indicated by alizarin red staining and quantification of calcium content. In addition, gene expression analysis revealed that ferrostatin-1 inhibited osteogenic differentiation of rat VSMCs. Similarly, ferrostatin-1 remarkably attenuated calcification of rat and human arterial rings ex vivo and aortic calcification in vitamin D3-overloaded mice in vivo. Moreover, inhibition of ferroptosis by either ferrostatin-1 or deferoxamine attenuated aortic calcification in rats with CKD. Mechanistically, high calcium and phosphate downregulated expression of SLC7A11 (a cystine-glutamate antiporter), and reduced glutathione (GSH) content in VSMCs. Additionally, GSH depletion induced by erastin (a small molecule initiating ferroptotic cell death) significantly promoted calcification of VSMCs under pro-osteogenic conditions, whereas GSH supplement by N-acetylcysteine reduced calcification of VSMCs. Consistently, knockdown of SLC7A11 by siRNA markedly promoted VSMC calcification. Furthermore, high calcium and phosphate downregulated glutathione peroxidase 4 (GPX4) expression, and reduced glutathione peroxidase activity. Inhibition of GPX4 by RSL3 promoted VSMC calcification. Thus, repression of the SLC7A11/GSH/GPX4 axis triggers ferroptosis of VSMCs to promote vascular calcification under CKD conditions, providing a novel targeting strategy for vascular calcification.


Assuntos
Ferroptose , Insuficiência Renal Crônica , Calcificação Vascular , Humanos , Ratos , Camundongos , Animais , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Músculo Liso Vascular , Osteogênese , Cálcio/metabolismo , Antiporters/metabolismo , Miócitos de Músculo Liso/metabolismo , Calcificação Vascular/genética , Calcificação Vascular/prevenção & controle , Ferro/metabolismo , Glutationa/metabolismo , Insuficiência Renal Crônica/patologia , Fosfatos/metabolismo , Sistema y+ de Transporte de Aminoácidos/genética , Sistema y+ de Transporte de Aminoácidos/metabolismo
4.
J Pathol ; 249(4): 461-471, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31397884

RESUMO

Vascular calcification is prevalent in patients with chronic kidney disease (CKD) and a major risk factor of cardiovascular disease. Vascular calcification is now recognised as a biological process similar to bone formation involving osteogenic differentiation of vascular smooth muscle cells (VSMCs). Cell division cycle 42 (CDC42), a Rac1 family member GTPase, is essential for cartilage development during endochondral bone formation. However, whether CDC42 affects osteogenic differentiation of VSMCs and vascular calcification remains unknown. In the present study, we observed a significant increase in the expression of CDC42 both in rat VSMCs and in calcified arteries during vascular calcification. Alizarin red staining and calcium content assay revealed that adenovirus-mediated CDC42 overexpression led to an apparent VSMC calcification in the presence of calcifying medium, accompanied with up-regulation of bone-related molecules including RUNX2 and BMP2. By contrast, inhibition of CDC42 by ML141 significantly blocked calcification of VSMCs in vitro and aortic rings ex vivo. Moreover, ML141 markedly attenuated vascular calcification in rats with CKD. Furthermore, pharmacological inhibition of AKT signal was shown to block CDC42-induced VSMC calcification. These findings demonstrate for the first time that CDC42 contributes to vascular calcification through a mechanism involving AKT signalling; this uncovered a new function of CDC42 in regulating vascular calcification. This may provide a potential therapeutic target for the treatment of vascular calcification in the context of CKD. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Osteogênese , Insuficiência Renal Crônica/complicações , Calcificação Vascular/etiologia , Proteína cdc42 de Ligação ao GTP/metabolismo , Idoso , Animais , Estudos de Casos e Controles , Células Cultivadas , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Osteogênese/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirazóis/farmacologia , Ratos Sprague-Dawley , Insuficiência Renal Crônica/sangue , Insuficiência Renal Crônica/enzimologia , Insuficiência Renal Crônica/patologia , Transdução de Sinais , Sulfonamidas/farmacologia , Técnicas de Cultura de Tecidos , Calcificação Vascular/sangue , Calcificação Vascular/enzimologia , Calcificação Vascular/patologia
5.
Cell Rep ; 28(1): 190-201.e3, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31269439

RESUMO

The regeneration capacity of neonatal mouse heart is controversial. In addition, whether epicardial cells provide a progenitor pool for de novo heart regeneration is incompletely defined. Following apical resection of the neonatal mouse heart, we observed limited regeneration potential. Fate-mapping of Tbx18MerCreMer mice revealed that newly formed coronary vessels and a limited number of cardiomyocytes were derived from the T-box transcription factor 18 (Tbx18) lineage. However, further lineage tracing with SM-MHCCreERT2 and Nfactc1Cre mice revealed that the new smooth muscle and endothelial cells are in fact derivatives of pre-existing coronary vessels. Our data show that neonatal mouse heart can regenerate but that its potential is limited. Moreover, although epicardial cells are multipotent during embryogenesis, their contribution to heart repair through "stem" or "progenitor" cell conversion is minimal after birth. These observations suggest that early embryonic heart development and postnatal heart regeneration are distinct biological processes. Multipotency of epicardial cells is significantly decreased after birth.


Assuntos
Vasos Coronários/citologia , Traumatismos Cardíacos/metabolismo , Coração/fisiologia , Miócitos Cardíacos/citologia , Pericárdio/citologia , Regeneração/fisiologia , Células-Tronco/citologia , Proteínas com Domínio T/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem da Célula , Vasos Coronários/metabolismo , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Técnicas de Introdução de Genes , Traumatismos Cardíacos/genética , Camundongos , Miócitos Cardíacos/metabolismo , Neovascularização Fisiológica , Pericárdio/metabolismo , Regeneração/genética , Células-Tronco/metabolismo , Proteínas com Domínio T/genética
6.
Circulation ; 138(25): 2919-2930, 2018 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-30566018

RESUMO

BACKGROUND: For more than a decade, Sca-1+ cells within the mouse heart have been widely recognized as a stem cell population with multipotency that can give rise to cardiomyocytes, endothelial cells, and smooth muscle cells in vitro and after cardiac grafting. However, the developmental origin and authentic nature of these cells remain elusive. METHODS: Here, we used a series of high-fidelity genetic mouse models to characterize the identity and regenerative potential of cardiac resident Sca-1+ cells. RESULTS: With these novel genetic tools, we found that Sca-1 does not label cardiac precursor cells during early embryonic heart formation. Postnatal cardiac resident Sca-1+ cells are in fact a pure endothelial cell population. They retain endothelial properties and exhibit minimal cardiomyogenic potential during development, normal aging and upon ischemic injury. CONCLUSIONS: Our study provides definitive insights into the nature of cardiac resident Sca-1+ cells. The observations challenge the current dogma that cardiac resident Sca-1+ cells are intrinsic stem cells for myocardial development, renewal, and repair, and suggest that the mechanisms of transplanted Sca-1+ cells in heart repair need to be reassessed.


Assuntos
Células-Tronco Adultas/fisiologia , Antígenos Ly/metabolismo , Células Endoteliais/fisiologia , Coração/embriologia , Proteínas de Membrana/metabolismo , Miócitos Cardíacos/fisiologia , Animais , Antígenos Ly/genética , Diferenciação Celular , Linhagem da Célula , Autorrenovação Celular , Células Cultivadas , Desenvolvimento Embrionário , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Modelos Animais , Regeneração , Transplante de Células-Tronco , Cicatrização
7.
J Biol Chem ; 293(24): 9162-9175, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29735531

RESUMO

Chondrocyte hypertrophy is the terminal step in chondrocyte differentiation and is crucial for endochondral bone formation. How signaling pathways regulate chondrocyte hypertrophic differentiation remains incompletely understood. In this study, using a Tbx18:Cre (Tbx18Cre/+) gene-deletion approach, we selectively deleted the gene for the signaling protein SMAD family member 4 (Smad4f/f ) in the limbs of mice. We found that the Smad4-deficient mice develop a prominent shortened limb, with decreased expression of chondrocyte differentiation markers, including Col2a1 and Acan, in the humerus at mid-to-late gestation. The most striking defects in these mice were the absence of stylopod elements and failure of chondrocyte hypertrophy in the humerus. Moreover, expression levels of the chondrocyte hypertrophy-related markers Col10a1 and Panx3 were significantly decreased. Of note, we also observed that the expression of runt-related transcription factor 2 (Runx2), a critical mediator of chondrocyte hypertrophy, was also down-regulated in Smad4-deficient limbs. To determine how the skeletal defects arose in the mouse mutants, we performed RNA-Seq with ChIP-Seq analyses and found that Smad4 directly binds to regulatory elements in the Runx2 promoter. Our results suggest a new mechanism whereby Smad4 controls chondrocyte hypertrophy by up-regulating Runx2 expression during skeletal development. The regulatory mechanism involving Smad4-mediated Runx2 activation uncovered here provides critical insights into bone development and pathogenesis of chondrodysplasia.


Assuntos
Desenvolvimento Ósseo , Condrócitos/patologia , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Proteína Smad4/genética , Animais , Diferenciação Celular , Proliferação de Células , Condrócitos/citologia , Condrócitos/metabolismo , Condrogênese , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Hipertrofia/genética , Hipertrofia/metabolismo , Hipertrofia/patologia , Camundongos , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patologia , Proteína Smad4/metabolismo
8.
Lab Invest ; 98(10): 1320-1332, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29785051

RESUMO

Vascular calcification is a highly regulated biological process similar to bone formation involving osteogenic differentiation of vascular smooth muscle cells (VSMCs). Hyaluronan (HA), a major structural component of the extracellular matrix in cartilage, has been shown to inhibit osteoblast differentiation. However, whether HA affects osteogenic differentiation and calcification of VSMCs remains unclear. In the present study, we used in vitro and ex vivo models of vascular calcification to investigate the role of HA in vascular calcification. Both high and low molecular weight HA treatment significantly reduced calcification of rat VSMCs in a dose-dependent manner, as detected by alizarin red staining and calcium content assay. Ex vivo study further confirmed the inhibitory effect of HA on vascular calcification. Similarly, HA treatment decreased ALP activity and expression of bone-related molecules including Runx2, BMP2 and Msx2. By contrast, inhibition of HA synthesis by 4-methylumbelliferone (4MU) promoted calcification of rat VSMCs. In addition, adenovirus-mediated overexpression of HA synthase 2 (HAS2), a major HA synthase in VSMCs, also inhibited calcification of VSMCs, whereas CRISPR/Cas9-mediated HAS2 knockout promoted calcification of rat A10 cells. Furthermore, we found that BMP2 signaling was inhibited in VSMCs after HA treatment. Recombinant BMP2 enhanced high calcium and phosphate-induced VSMC calcification, which can be blocked by HA treatment. Taken together, these findings suggest that HA inhibits vascular calcification involving BMP2 signaling.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Ácido Hialurônico/metabolismo , Calcificação Vascular/etiologia , Animais , Linhagem Celular , Técnicas de Inativação de Genes , Hialuronan Sintases/genética , Hialuronan Sintases/metabolismo , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Cultura Primária de Células , Ratos Sprague-Dawley , Calcificação Vascular/metabolismo
9.
Cell Physiol Biochem ; 45(1): 88-107, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29310113

RESUMO

Human neuropeptide Y (hNPY) is one of the most widely expressed neurotransmitters in the human central and peripheral nervous systems. It consists of 36 highly conserved amino acid residues, and was first isolated from the porcine hypothalamus in 1982. While it is the most recently discovered member of the pancreatic polypeptide family (which includes neuropeptide Y, gut-derived hormone peptide YY, and pancreatic polypeptide), NPY is the most abundant peptide found in the mammalian brain. In order to exert particular functions, NPY needs to bind to the NPY receptor to activate specific signaling pathways. NPY receptors belong to the class A or rhodopsin-like G-protein coupled receptor (GPCR) family and signal via cell-surface receptors. By binding to GPCRs, NPY plays a crucial role in various biological processes, including cortical excitability, stress response, food intake, circadian rhythms, and cardiovascular function. Abnormal regulation of NPY is involved in the development of a wide range of diseases, including obesity, hypertension, atherosclerosis, epilepsy, metabolic disorders, and many cancers. Thus far, five receptors have been cloned from mammals (Y1, Y2, Y4, Y5, and y6), but only four of these (hY1, hY2, hY4, and hY5) are functional in humans. In this review, we summarize the structural characteristics of human NPY receptors and their role in metabolic diseases.


Assuntos
Doenças Metabólicas/etiologia , Receptores de Neuropeptídeo Y/metabolismo , Diabetes Mellitus/etiologia , Diabetes Mellitus/metabolismo , Diabetes Mellitus/terapia , Dislipidemias/etiologia , Dislipidemias/metabolismo , Dislipidemias/terapia , Humanos , Hipertensão/etiologia , Hipertensão/metabolismo , Hipertensão/terapia , Doenças Metabólicas/metabolismo , Doenças Metabólicas/terapia , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/terapia , Receptores de Neuropeptídeo Y/química , Receptores de Neuropeptídeo Y/genética , Transdução de Sinais
10.
J Cardiovasc Pharmacol ; 70(3): 184-193, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28678055

RESUMO

The aim of this study was to explore the effect and mechanism of action of resveratrol (RSV) on cardiac function in diabetic cardiomyopathy (DCM). Hyperglycemia-induced apoptosis contributes to the pathogenic changes in DCM. RSV treatment inhibited high glucose-induced apoptosis of neonatal rat ventricular myocytes. Additionally, high glucose decreased cell viability, prevented serine-threonine kinase (Akt) and FoxO3a phosphorylation, and suppressed cytoplasmic translocation of FoxO3a. However, these effects of apoptosis were reversed by 10 µM of RSV. The PI3K inhibitor LY294002 abolished the RSV protective effect in vitro. RSV (5 or 50 mg·kg·d orally for 8 weeks) prevented the deterioration of cardiac function and structural cardiomyopathy in a streptozotocin-induced rat model of diabetes and reduced apoptosis in diabetic myocardium. Furthermore, it restored streptozotocin-impaired phosphorylation of Akt and FoxO3a (p-Akt and p-FoxO3a) and suppressed nuclear translocation of FoxO3a in vivo. Together, these data indicate that RSV has therapeutic potential against DCM by inhibiting apoptosis via the PI3K/Akt/FoxO3a pathway.


Assuntos
Apoptose/efeitos dos fármacos , Cardiomiopatias Diabéticas/tratamento farmacológico , Proteína Forkhead Box O3/antagonistas & inibidores , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Estilbenos/uso terapêutico , Animais , Apoptose/fisiologia , Cardiomiopatias Diabéticas/metabolismo , Modelos Animais de Doenças , Proteína Forkhead Box O3/metabolismo , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Resveratrol , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Estilbenos/farmacologia
11.
Eur J Pharmacol ; 794: 45-51, 2017 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-27876618

RESUMO

Vascular calcification is a major feature of advanced atherosclerosis and highly associated with cardiovascular diseases. Oxidized low density lipoprotein (Ox-LDL) has been recognized as a critical risk factor for atherosclerosis and osteogenic differentiation of vascular smooth muscle cells (VSMCs). Previous studies have demonstrated that toll like receptor 4 (TLR4) is highly expressed in atherosclerotic lesions and participates in the progression of atherosclerosis. However, the role of TLR4 in vascular calcification remains unknown. In this study, we investigated whether TLR4 modulates vascular calcification induced by Ox-LDL. TLR4 expression was up-regulated in cultured human VSMCs treated with Ox-LDL. Knockdown of TLR4 by small interfering RNA (siRNA) significantly reduced Ox-LDL-induced calcification, detected by alizarin red staining and calcium content assay. TLR4 siRNA also decreased the mRNA expression of bone-related proteins including Msx2, osterix, BMP2 and KLF4, but increased the expression of VSMC contractile proteins including SMA and SM22α in VSMCs. In addition, Ox-LDL stimulated nuclear translocation of nuclear factor kappa B (NK-κB) p65. These effects of Ox-LDL on VSMCs were reversed by TLR4 siRNA. Furthermore, NK-κB inhibitor, pyrrolidine dithiocarbamate (PDTC), attenuated Ox-LDL-induced VSMC calcification, which was rescued by C2-ceramide treatment. In conclusion, these findings suggest that TLR4 regulates VSMC calcification induced by Ox-LDL through activation of NK-κB, highlighting the critical role of TLR4/NK-κB signaling in vascular calcification.


Assuntos
Calcinose/induzido quimicamente , Calcinose/patologia , Ceramidas/metabolismo , Lipoproteínas LDL/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , NF-kappa B/metabolismo , Receptor 4 Toll-Like/metabolismo , Calcinose/metabolismo , Diferenciação Celular/efeitos dos fármacos , Humanos , Fator 4 Semelhante a Kruppel , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Osteogênese/efeitos dos fármacos
12.
Eur J Pharmacol ; 788: 90-97, 2016 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-27321870

RESUMO

Proliferation and migration of vascular smooth muscle cells (VSMCs) play crucial roles in the development of vascular restenosis. Our previous study showed that CCN4, namely Wnt1 inducible signaling pathway protein 1 (WISP1), significantly promotes proliferation and migration of rat VSMCs, but its mechanism remains unclear. This study aims to investigate whether and how WISP1 stimulates proliferation and migration of human VSMCs. Western blot analysis showed that FBS treatment increased WISP1 protein levels in human VSMCs in a dose-dependent manner. Overexpression of WISP1 using adenovirus encoding WISP1 (AD-WISP1) significantly increased proliferation rate of human VSMCs by 2.98-fold compared with empty virus (EV)-transfected cells, shown by EdU incorporation assay. Additionally, Scratch-induced wound healing assay revealed that adenovirus-mediated overexpression of WISP1 significantly increased cell migration compared with EV-transfected cells from 6h (4.56±1.14% vs. 11.23±2.25%, P<0.05) to 48h (25.25±5.51% vs. 97.54±13.12%, P<0.01) after injury. Transwell Migration Assay confirmed that WISP1 overexpression significantly promoted human VSMC migration by 2.25-fold compared with EV. Furthermore, WISP1 overexpression stimulated Akt signaling activation in human VSMCs. Blockage of Akt signaling by Akt inhibitor AZD5363 or PI3K inhibitor LY294002, led to an inhibitory effect of WISP1-induced proliferation and migration in human VSMCs. Moreover, we found that WISP1 overexpression stimulated GSK3α/ß phosphorylation, and increased expression of cyclin D1 and MMP9 in human VSMCs, and this effect was abolished by AZD5363. Collectively, we demonstrated that Akt signaling pathway mediates WISP1-induced migration and proliferation of human VSMCs, suggesting that WISP1 may act as a novel potential therapeutic target for vascular restenosis.


Assuntos
Proteínas de Sinalização Intercelular CCN/genética , Movimento Celular , Músculo Liso Vascular/citologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas/genética , Transdução de Sinais , Movimento Celular/genética , Proliferação de Células/genética , Ciclina D1/metabolismo , Expressão Gênica , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Metaloproteinase 9 da Matriz/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/genética , Transdução de Sinais/genética , Regulação para Cima
13.
Nat Commun ; 6: 8701, 2015 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-26515110

RESUMO

Identifying a bona fide population of cardiac stem cells (CSCs) is a critical step for developing cell-based therapies for heart failure patients. Previously, cardiac c-kit(+) cells were reported to be CSCs with a potential to become myocardial, endothelial and smooth muscle cells in vitro and after cardiac injury. Here we provide further insights into the nature of cardiac c-kit(+) cells. By targeting the c-kit locus with multiple reporter genes in mice, we find that c-kit expression rarely co-localizes with the expression of the cardiac progenitor and myogenic marker Nkx2.5, or that of the myocardial marker, cardiac troponin T (cTnT). Instead, c-kit predominantly labels a cardiac endothelial cell population in developing and adult hearts. After acute cardiac injury, c-kit(+) cells retain their endothelial identity and do not become myogenic progenitors or cardiomyocytes. Thus, our work strongly suggests that c-kit(+) cells in the murine heart are endothelial cells and not CSCs.


Assuntos
Infarto do Miocárdio/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Células-Tronco/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Camundongos , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Células-Tronco/citologia , Fatores de Transcrição/metabolismo
14.
Genesis ; 53(6): 377-86, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26010701

RESUMO

Tnnt2, encoding thin-filament sarcomeric protein cardiac troponin T, plays critical roles in heart development and function in mammals. To develop an inducible genetic deletion strategy in myocardial cells, we generated a new Tnnt2:MerCreMer (Tnnt2(MerCreMer/+)) knock-in mouse. Rosa26 reporter lines were used to examine the specificity and efficiency of the inducible Cre recombinase. We found that Cre was specifically and robustly expressed in the cardiomyocytes at embryonic and adult stages following tamoxifen induction. The knock-in allele on Tnnt2 locus does not impact cardiac function. These results suggest that this new Tnnt2(MerCreMer/+) mouse could be applied towards the temporal genetic deletion of genes of interests in cardiomyocytes with Cre-LoxP technology. The Tnnt2(MerCreMer/+) mouse model also provides a useful tool to trace myocardial lineage during development and repair after cardiac injury.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Miocárdio/metabolismo , Tamoxifeno/farmacologia , Troponina T/genética , Actinas/metabolismo , Animais , Antineoplásicos Hormonais/farmacologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Feminino , Coração/embriologia , Coração/crescimento & desenvolvimento , Coração/fisiologia , Imuno-Histoquímica , Integrases/genética , Integrases/metabolismo , Masculino , Camundongos Transgênicos , Modelos Animais , Músculo Liso/química , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , RNA não Traduzido/genética , Fatores de Tempo , Troponina T/metabolismo , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
15.
Atherosclerosis ; 219(2): 440-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21920521

RESUMO

OBJECTIVES: Vascular calcification is a major clinical problem and elucidating the underlying mechanism is important to improve the prognosis of patients with cardiovascular disease. We aimed to elucidate the role and mechanism of action of Hepatocyte Growth Factor (HGF)/c-Met signalling in vascular calcification and establish whether blocking this pathway could prevent mineralisation of vascular smooth muscle cells (VSMCs) in vitro. METHODS AND RESULTS: We demonstrate increased HGF secretion and c-Met up-regulation and phosphorylation during VSMC osteogenic differentiation. Adenoviral-mediated over-expression of HGF (AdHGF) in VSMCs accelerated mineralisation, shown by alizarin red staining, and significantly increased (45)Calcium incorporation (1.96 ± 0.54-fold [P < 0.05]) and alkaline phosphatase (ALP) activity (3.01 ± 0.8-fold [P < 0.05]) compared to controls. AdHGF also significantly elevated mRNA expression of bone-related proteins, Runx2, osteocalcin, BMP2 and osterix in VSMCs. AdHGF-accelerated mineralisation correlated with increased Akt phosphorylation, nuclear translocation of Notch3 intracellular domain (N3IC) and up-regulation of the Notch3 target protein, HES1. In contrast, adenoviral-mediated over-expression of the HGF antagonist, NK4, markedly attenuated VSMC mineralisation, and reduced c-Met phosphorylation, Akt activation and HES1 protein expression compared to AdHGF-treated cells. Furthermore, the Notch inhibitor, DAPT, attenuated N3IC nuclear translocation and AdHGF-induced mineralisation. CONCLUSION: We demonstrate HGF induces VSMC osteogenic differentiation via c-Met/Akt/Notch3 signalling, highlighting these pathways as potential targets for intervention of vascular calcification.


Assuntos
Diferenciação Celular , Fator de Crescimento de Hepatócito/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Osteogênese , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Calcificação Vascular/metabolismo , Adenoviridae/genética , Fosfatase Alcalina/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteína Morfogenética Óssea 2/genética , Cálcio/metabolismo , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Vetores Genéticos , Fator de Crescimento de Hepatócito/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Receptor Notch3 , Fator de Transcrição Sp7 , Fatores de Tempo , Fatores de Transcrição HES-1 , Fatores de Transcrição/genética , Transfecção , Regulação para Cima , Calcificação Vascular/genética , Calcificação Vascular/patologia
16.
Arterioscler Thromb Vasc Biol ; 31(3): 608-15, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21205989

RESUMO

OBJECTIVE: Decorin and oxidized low-density lipoprotein (Ox-LDL) independently induce osteogenic differentiation of vascular smooth muscle cells (VSMCs). We aimed to determine whether decorin glycosaminoglycan (GAG) chain synthesis contributes to Ox-LDL-induced differentiation and calcification of human VSMCs in vitro. METHODS AND RESULTS: Human VSMCs treated with Ox-LDL to induce oxidative stress showed increased alkaline phosphatase (ALP) activity, accelerated mineralization, and a difference in both decorin GAG chain biosynthesis and CS/DS structure compared with untreated controls. Ox-LDL increased mRNA abundance of both xylosyltransferase (XT)-I, the key enzyme responsible for GAG chain biosynthesis and Msx2, a marker of osteogenic differentiation. Furthermore, downregulation of XT-I expression using small interfering RNA blocked Ox-LDL-induced VSMC mineralization. Adenoviral-mediated overexpression of decorin, but not a mutated unglycanated form, accelerated mineralization of VSMCs, suggesting GAG chain addition on decorin is crucial for the process of differentiation. The decorin-induced VSMC osteogenic differentiation involved activation of the transforming growth factor (TGF)-ß pathway, because it was attenuated by blocking of TGF-ß receptor signaling and because decorin overexpression potentiated phosphorylation of the downstream signaling molecule smad2. CONCLUSIONS: These studies provide direct evidence that oxidative stress-mediated decorin GAG chain synthesis triggers TGF-ß signaling and mineralization of VSMCs in vitro.


Assuntos
Calcinose/metabolismo , Decorina/biossíntese , Lipoproteínas LDL/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Osteogênese , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Fosfatase Alcalina/metabolismo , Células Cultivadas , Decorina/genética , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Estresse Oxidativo , Pentosiltransferases/genética , Pentosiltransferases/metabolismo , Fosforilação , Interferência de RNA , Proteína Smad2/metabolismo , Fatores de Tempo , UDP Xilose-Proteína Xilosiltransferase
17.
Int J Cardiol ; 134(1): 82-90, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18619688

RESUMO

BACKGROUND: The mechanisms underlying heart failure induced by adriamycin are very complicated and still unclear. The aim of this study was to investigate whether autophagy was involved in the progression of heart failure induced by adriamycin, so that we can develop a novel treatment strategy for heart failure. METHODS: 3-methyladenine (3MA), a specific inhibitor on autophagy was used in a heart failure model of rats induced by adriamycin. Neonatal cardiomyocytes were isolated from Sprague-Dawley rat hearts and randomly divided into controls, an adriamycin-treated group, and a 3MA plus adriamycin-treated group. We then examined the morphology, expression of beclin 1 gene, mitochondrial permeability transition (MPT), and Na+-K+ ATPase activity in vivo. We also assessed cell viability, mitochondrial membrane potential changes and counted autophagic vacuoles in cultured cardiomyocytes. In addition, we analyzed the expression of autophagy associated gene, beclin 1 using RT-PCR and Western blotting in an animal model. RESULTS: 3MA significantly improved cardiac function and reduced mitochondrial injury. Furthermore, adriamycin induced the formation of autophagic vacuoles, and 3MA strongly downregulated the expression of beclin 1 in adriamycin-induced failing heart and inhibited the formation of autophagic vacuoles. CONCLUSION: Autophagic cardiomyocyte death plays an important role in the pathogenesis of heart failure in rats induced by adriamycin. Mitochondrial injury may be involved in the progression of heart failure caused by adriamycin via the autophagy pathway.


Assuntos
Autofagia/fisiologia , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/patologia , Miócitos Cardíacos/patologia , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Antibióticos Antineoplásicos/toxicidade , Proteínas Reguladoras de Apoptose/genética , Autofagia/efeitos dos fármacos , Proteína Beclina-1 , Pressão Sanguínea , Modelos Animais de Doenças , Doxorrubicina/toxicidade , Expressão Gênica/efeitos dos fármacos , Frequência Cardíaca , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , ATPase Trocadora de Sódio-Potássio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA