Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
RSC Adv ; 14(30): 21241-21249, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38974227

RESUMO

Cardiovascular diseases caused by atherosclerosis (AS) seriously damage human health. Nano-photothermal technology has been proven to inhibit the development of vascular inflammation by inhibiting the proliferation of inflammatory macrophages. However, photothermal therapy can inhibit the enrichment of AS macrophages in the early stage, but the inhibitory effect is insufficient in the later stage. Herein, we designed and prepared CoS1.097 nanocrystals by a simple hydrothermal method as new nanoplatforms for efficient photothermal therapy of arterial inflammation. CoS1.097 nanocrystals exhibited the degradability to release the cobalt ions, and can inhibit the proliferation of macrophages both in vitro and in vivo resulting from the slowly released cobalt ions. Moreover, CoS1.097 nanocrystals showed intense absorption in the NIR region, thus showing excellent photothermal performance. When irradiated by an 808 nm laser, the photothermal effect of CoS1.097 nanocrystals can more efficiently kill the macrophages which play an important role in the development of atherosclerosis. As far as we know, this is the first work on CoS1.097 nanocrystals for photothermal therapy of arterial inflammation.

2.
Biosci Biotechnol Biochem ; 88(7): 776-783, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38714325

RESUMO

Atherosclerosis (AS) is the major cause of multiple cardiovascular diseases. In addition, the lipid accumulation of human vascular smooth muscle cells (HVSMCs) can cause the occurrence of AS. Secreted frizzled-related protein 5 (Sfrp5) was known to be downregulated in AS; however, the detailed function of Sfrp5 in HVSMCs remains unclear. Specifically, we found that Sfrp5 expression in oxLDL-treated HVSMCs was downregulated. Sfrp5 overexpression inhibited the viability of HVSMCs induced by oxLDL. In addition, oxLDL-induced proliferation and migration in HVSMCs were abolished by Sfrp5 overexpression. Sfrp5 overexpression reduced oxLDL-caused oxidative stress, lipid accumulation, and inflammation in HVSMCs. Meanwhile, oxLDL treatment increased the expressions of Wnt5a, c-Myc, and ß-catenin in HVSMCs, while this phenomenon was rescued by Sfrp5 overexpression. Furthermore, the inhibitory effect of Sfrp5 upregulation on the viability and migration of HVSMCs was reversed by R-spondin 1. These results indicate that Sfrp5 overexpression could reverse oxLDL-induced lipid accumulation in HVSMCs through inactivating Wnt5a/ß-catenin signaling pathway.


Assuntos
Movimento Celular , Metabolismo dos Lipídeos , Lipoproteínas LDL , Músculo Liso Vascular , Miócitos de Músculo Liso , Proteína Wnt-5a , Humanos , Lipoproteínas LDL/metabolismo , Lipoproteínas LDL/farmacologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/citologia , Proteína Wnt-5a/metabolismo , Proteína Wnt-5a/genética , Movimento Celular/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Estresse Oxidativo , beta Catenina/metabolismo , beta Catenina/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Aterosclerose/metabolismo , Aterosclerose/genética , Aterosclerose/patologia , Transdução de Sinais
3.
J Cell Mol Med ; 27(22): 3431-3442, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37596794

RESUMO

The high rates of misdiagnosis and untreated mortality with regard to Budd-Chiari syndrome (BCS) indicated the need to screen effective biomarkers. The aim of this study was to explore the function of extracellular vesicles (EVs) in patients with BCS as well as associated mechanisms. First, differentially expressed long non-coding RNAs (lncRNAs) from EVs separated from serum between BCS and healthy controls were screened using microarray analysis. Second, the proliferation, migration and tube formation of human vascular endothelial cells (HUVECs) were detected after EVs treatment, along with vascular endothelial growth factor (VEGF) levels and inflammatory factors from the cell supernatant. Last, the overexpressed lncRNA was transfected into the cells to further explore the mechanisms involved. Extracellular vesicles of BCS patients have significantly higher levels of lncRNA MTUS2-5 than healthy controls. Apparently, treatment with EVs from BCS or the ones transfected with plasmids that overexpress lncRNA MTUS2-5 enhances proliferation, migration and angiogenesis capacity. The results were considerably better than those obtained from treatment with EVs from healthy controls or transfection with the normal control plasmid, which also elevated the level of VEGF and inflammatory factors. Furthermore, FOS and PTGS2 were potentially regulated by the lncRNA MTUS2-5 transmitted by EVs. The lncRNA MTUS2-5 in EVs plays an important role in angiogenesis in the Budd-Chiari syndrome.


Assuntos
Síndrome de Budd-Chiari , Vesículas Extracelulares , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Células Endoteliais/metabolismo , Síndrome de Budd-Chiari/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Vesículas Extracelulares/genética , Vesículas Extracelulares/metabolismo , Neovascularização Patológica/metabolismo , Proliferação de Células/genética
4.
Vasc Med ; 28(1): 6-17, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36759934

RESUMO

BACKGROUND: Circular RNAs (circRNAs) have been implicated in the pathogenesis of atherosclerosis (AS) and the migration and proliferation of vascular smooth muscle cells (VSMCs) under oxidized low-density lipoprotein (ox-LDL). Here, we defined the exact action of human circ_0007478 in VSMC migration and proliferation induced by ox-LDL. METHODS: Human VSMCs (HVSMCs) were exposed to ox-LDL. Circ_0007478, microRNA (miR)-638, and rho-associated protein kinase 2 (ROCK2) levels were gauged by quantitative real-time PCR (qRT-PCR) and western blot. Cell viability and proliferation were assessed by MTT and EdU assays, respectively. Transwell assays were used to detect cell migration and invasion. Dual-luciferase reporter and RNA immunoprecipitation (RIP) assays were used to evaluate the direct relationship between miR-638 and circ_0007478 or ROCK2. RESULTS: Our data indicated that circ_0007478 expression was augmented in AS serum samples and ox-LDL-treated HVSMCs. Depletion of circ_0007478 attenuated HVSMC proliferation, migration, and invasion induced by ox-LDL. Mechanistically, circ_0007478 targeted miR-638 by directly pairing to miR-638. Reduction of miR-638 reversed the effects of circ_0007478 depletion on ox-LDL-evoked proliferation, migration, and invasion in HVSMCs. ROCK2 was a direct miR-638 target and miR-638-mediated inhibition of ROCK2 relieved ox-LDL-evoked HVSMC proliferation, migration, and invasion. Furthermore, circ_0007478 was identified as a competing endogenous RNA (ceRNA) for miR-638 to modulate ROCK2 expression. CONCLUSION: Our present study establishes an undescribed ceRNA regulatory network, in which circ_0007478 targets miR-638 to upregulate ROCK2, thereby contributing to ox-LDL-induced proliferation and migration in HVSMCs.


Assuntos
Aterosclerose , MicroRNAs , Humanos , Músculo Liso Vascular , Aterosclerose/genética , Movimento Celular , Lipoproteínas LDL/farmacologia , Proliferação de Células , MicroRNAs/genética , Apoptose , Células Cultivadas , Miócitos de Músculo Liso , Quinases Associadas a rho/genética
5.
DNA Cell Biol ; 42(2): 97-104, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36730754

RESUMO

To investigate the effect of paired box protein 5 (PAX5)/integrin subunit alpha X (ITGAX) in atherosclerosis (AS). AS model was established using ApoE-/- mice (C57BL/6). Human vascular smooth muscle cells (HVSMCs) were stimulated with ox-LDL. Quantitative reverse transcription polymerase chain reaction and Western blotting were used to detect the expression levels of genes and proteins. Reporter constructs and luciferase assays were used to investigate the role of ITGAX and PAX5. Cells proliferation and inflammation factors were detected. The results presented that aortic plaque area, lipid content, serum triglyceride, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol levels were significantly increased in the high-fat diet group (p < 0.05). ITGAX was upregulated in atherosclerotic tissues. In addition, ox-LDL treatment induced HVSMCs proliferation, migration, and invasion. Reporter constructs and luciferase assays indicated ITGAX interaction with PAX5. Furthermore, siITGAX and siPAX5 cotransfection restored the rate of HVSMCs in G1 and S and G2/M phases, decreased the content of tumor necrosis factor-alpha (TNF-ɑ), interleukin (IL)-6, and IL-8 (p < 0.05). Interestingly, siITGAX and siPAX5 cotransfection also decreased the expression levels of TNF-α, TNF-R1, TNF-R2, CD19, and CD86 (p < 0.05). Our results suggest that ITGAX may be a potential therapeutic target for AS.


Assuntos
Aterosclerose , Fator de Necrose Tumoral alfa , Animais , Humanos , Camundongos , Aterosclerose/metabolismo , Diferenciação Celular , Colesterol/metabolismo , Interleucina-6 , Lipoproteínas LDL/farmacologia , Lipoproteínas LDL/metabolismo , Camundongos Endogâmicos C57BL , Fator de Transcrição PAX5/metabolismo , Transdução de Sinais
6.
Eur J Histochem ; 66(3)2022 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-35855629

RESUMO

Thrombomodulin (TM) is involved in the pathological process of atherosclerosis; however, the underlying mechanism remains unclear. Oxidised low-density lipoprotein (Ox-LDL; 100 µg/mL) was used to induce human vascular smooth muscle cells (HVSMCs) into a stable atherosclerotic cell model. The expression levels of miR-550a-3p and TM were detected by real-time reverse transcription-polymerase chain reaction. Cell proliferation was estimated using CCK8 and EDU assays. Wound scratch and transwell assays were used to measure the ability of cells to invade and migrate. Propidium iodide fluorescence-activated cell sorting was used to detect apoptosis and cell cycle changes. A dual-luciferase reporter assay was performed to determine the binding of miR-550a-3p to TM. Our results suggested the successful development of a cellular atherosclerosis model. Our data revealed that TM overexpression significantly promoted the proliferation, invasion, migration, and apoptosis of HVSMCs as well as cell cycle changes. Upregulation of miR-550a-3p inhibited the growth and metastasis of HVSMCs. Furthermore, miR-550a-3p was confirmed to be a direct target of TM. Restoration of miR-550a-3p expression rescued the effects of TM overexpression. Thus, miR-550a-3p might play a role in atherosclerosis and, for the first time, normalised the function of injured vascular endothelial cells by simultaneous transfection of TM and miR-550a-3p. These results suggest that the miR-550a-3p/TM axis is a potential therapeutic target for atherosclerosis.


Assuntos
Aterosclerose , MicroRNAs , Apoptose/fisiologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Proliferação de Células/fisiologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Trombomodulina/genética , Trombomodulina/metabolismo
7.
Exp Ther Med ; 22(2): 791, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34093747

RESUMO

Excessive proliferation and migration of vascular smooth muscle cells (VSMCs) is the main cause of arteriosclerosis obliterans (ASO). The present study aimed to investigate the role of microRNA (miR)-125b on the proliferation and migration of VSMCs. Platelet-derived growth factor-BB (PDGF-BB; 20 ng/ml) was used to treat VSMCs to establish an in vitro model of ASO. VSMCs were transfected with miR-125b mimic to overexpress miR-125. Cell Counting kit-8 (CCK-8) and BrdU assays were performed to assess the proliferative ability of VSMCs, while Transwell and wound healing assays were performed to assess the migratory ability of VSMCs. Western blot and immunofluorescence analyses were performed to detect the expression levels of angio-associated migratory cell protein (AAMP) and serum response factor (SRF) in VSMCs following transfection with miR-125b mimic or inhibitor. The results demonstrated that miR-125b expression decreased following treatment with PDGF-BB, the effects of which were reversed following transfection with miR-125b mimic. According to the CCK-8 assay, the cell proliferative ability decreased by ~50% compared with the negative control (NC) group, and ~40% at day 4 based on the BrdU assay. The results of the Transwell and wound healing assays indicated that the migratory ability of VSMCs significantly decreased in the miR-125b mimic group compared with the NC group. Furthermore, western blot and immunofluorescence analyses demonstrated that AAMP and SRF expression levels decreased following transfection with miR-125b mimic compared with the NC group, the effects of which were reversed following transfection with miR-125 inhibitor. Taken together, the results of the present study suggested that miR-125b inhibits the proliferative and migratory abilities of VSMCs by regulating the expression levels of AAMP and SRF.

8.
Exp Ther Med ; 18(1): 77-84, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31258640

RESUMO

As an atherosclerotic disease, the process of peripheral arterial disease (PAD) is complicated and includes the abnormal proliferation of vascular smooth muscle. The current study aimed to determine the role of microRNA-15b (miR-15b) in the development of PAD and its associated mechanisms. Human vascular smooth muscle cells (hVSMCs) were used in the current study. To assess the effects of miR-15b on hVSMCs, miR-15b was up- or downregulated in hVSMCs using miR-15b mimics or miR-15b inhibitors respectively. Cell viability, migration and apoptosis were then determined via MTT, transwell and flow cytometry assays, respectively. TargetScan bioinformatics software was utilized to predict the targets of miR-15b, and the binding sites between insulin growth factor 1 receptor (IGF1R) and miR-15b were confirmed by dual-luciferase reporter assay. The results reveled that the miR-15b mimic significantly reduced hVSMC cell viability and migration, and promoted cell apoptosis. However, the opposite effect was observed following miR-15b inhibitor transfection. It was also determined that miR-15b directly targeted IGF1R and negatively regulated its expression in hVSMCs. Additionally, the results demonstrated that the miR-15b mimic inhibited the PI3K/AKT signaling pathway in hVSMCs, whereas the miR-15b inhibitor promoted it. Furthermore, the results indicated that the effect of the miR-15b mimic on hVSMCs was reversed by IGF1R overexpression. In conclusion, the data indicated that miR-15b participated in the occurrence and development of PAD by modulating hVSMC proliferation, apoptosis and migration via the regulation of IGF1R expression.

9.
J Cell Biochem ; 120(9): 14405-14413, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30989723

RESUMO

The main aim of our study was to investigate the roles and molecular basis of long noncoding RNA myocardial infarction associated transcript (MIAT) in the development of thoracic aortic aneurysm. RT-qPCR assay was performed to measure the expressions of MIAT, microRNA-145 (miR-145), along with Bcl-2 and Bcl-xl messenger RNAs. Western blot assay was conducted to determine protein levels of Bcl-2, Bcl-xl, phosphorylated-Akt (p-Akt), and total Akt (t-Akt). Cell viability was detected by the (3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay. The relationship of MIAT and miR-145 was examined by bioinformatics analysis and luciferase reporter assay. MIAT expression was significantly increased, and miR-145 expression was markedly reduced in thoracic aortic aneurysms compared with normal thoracic aortic tissues. MIAT overexpression or miR-145 depletion improved cell viability and inhibited cell apoptosis in human aortic vascular smooth muscle cells (h-VSMCs). Further exploration revealed that MIAT could inhibit miR-145 expression by direct interaction. And miR-145 upregulation abrogated MIAT-induced viability increase and apoptosis inhibition in h-VSMCs. Moreover, MIAT inhibited the activation of Akt signaling, while this effect was abated by miR-145 overexpression in h-VSMCs. The inhibition of the Akt pathway by MK-22062HCl resulted in the reduction of cell viability and the increase of cell apoptotic activity in h-VSMCs. Akt activation by HY-18749 improved cell viability and suppressed cell apoptosis in h-VSMCs. And the introduction of HY-18749 raised cell viability and curbed cell apoptosis in h-VSMCs cotransfected with MIAT overexpression plasmid and miR-145 mimic. lncRNA-MIAT could target miR-145 to affect the viability and apoptosis of h-VSMCs, which was implicated in the regulation of the PI3K/Akt signaling pathway.


Assuntos
Aorta Torácica/metabolismo , Aneurisma Aórtico/genética , MicroRNAs/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/genética , Aorta Torácica/patologia , Aneurisma Aórtico/metabolismo , Apoptose/genética , Sobrevivência Celular/genética , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Transdução de Sinais/genética
10.
Med Sci Monit ; 25: 128-134, 2019 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-30610777

RESUMO

BACKGROUND Inferior vena cava (IVC) filters have proven valuable for the prevention of pulmonary embolism. However, retrieval of IVC filters can be challenging and complicated techniques are needed. The aim of this study was to retrospectively review the outcomes after retrieval of retrievable IVC filters which required complicated retrieval techniques. MATERIAL AND METHODS This study was a single-center retrospective observational study. All patients who underwent complicated IVC filter retrieval from September 2012 to May 2016 were included. Patient demographics and filter retrieval procedure were documented. Clinical outcomes and procedure-related complications were evaluated. Villalta score and VEINES-QOL/Sym were recorded to assess post-thrombotic syndrome. RESULTS A total of 79 consecutive patients, 37 males and 42 were female, with a mean age of 46.5 years (age range: 22-65 years) were included in this study. IVC filters, with mean indwell time of 108 days (range: 74-157 days), were refractory to standard treatment and underwent complicated IVC filter retrieval. There were 6 serious procedure-related complications: 4 popliteal puncture area hematoma complications and 2 hematuria complications. With a mean follow­up of 20.5 months (range: 18-24 months), no pulmonary embolisms occurred, and 2 patients experienced recurrent deep venous thrombosis. Twenty-seven patients developed post-thrombotic syndrome within the first 2 years after IVC retrieval. CONCLUSIONS Complicated methods can be used to safely remove IVC filters, alleviate filter-related morbidity, and reduce risk for post-thrombotic syndrome. The application of these techniques was safe and effective for patients with refractory IVC filters.


Assuntos
Remoção de Dispositivo/métodos , Filtros de Veia Cava/efeitos adversos , Trombose Venosa/complicações , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Embolia Pulmonar/prevenção & controle , Estudos Retrospectivos , Fatores de Risco , Resultado do Tratamento , Trombose Venosa/etiologia , Trombose Venosa/cirurgia
11.
Int J Clin Exp Pathol ; 12(5): 1599-1608, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31933977

RESUMO

BACKGROUND: Abnormal proliferation of PASMCs is the main phenotype of pulmonary arterial hypertension (PAH). MicroRNAs (miRNAs) were reported to participate in regulating the progression of PAH. Here, we aimed to investigate the impact of miR-107 on proliferation and migration of PASMCs and potential mechanism. METHODS: MTT assay was carried out to examine the cell viability of PASMCs. PASMC migration ability was verified through Transwell assay. RT-qPCR was performed to detect the expression of miR-107 and NOR1. Western blot was conducted to detect the expression of cell proliferation markers Ki-67, p27 and Cyclin D1, as well as NOR1. Bioinformatics analysis was conducted to verify whether the 3'-untranslated region (3'-UTR) of NOR1 contains a binding site for miR-107, and luciferase reporter assay and RNA immunoprecipitation (RIP) were employed to confirm the relationship between miR-107 and NOR1. RESULTS: Platelet-derived growth factor (PDGF)-BB promoted the cell viability and migration of PASMCs, and suppressed miR-107 expression in a time-dependent and concentration-dependent manner. Introduction of miR-107 inhibited the promotion of proliferation and migration of PASMCs stimulated by PDGF-BB, while loss of miR-107 facilitated PDGF-BB-induced promoted effects. NOR1 was identified as a downstream gene of miR-107 and down-regulated by miR-107. Knockout of NOR1 also repressed the promotion of proliferation and migration of PASMCs stimulated by PDGF-BB. Additionally, restoration of NOR1 attenuated the inhibition of miR-107 on the cell viability and migration ability of PASMCs. CONCLUSION: miR-107 inhibits PDGF-BB-induced PASMCs proliferation and migration through targeting NOR1.

12.
Open Med (Wars) ; 14: 663-672, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31989041

RESUMO

BACKGROUND: Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) accelerated vascular diseases progression, like atherosclerosis and restenosis. MicroRNAs were reported to participate in modulating diverse cellular processes. Here, we focused on exploring the role of miR-638 in VSMCs glycolysis and underlying mechanism. METHODS: Cell Counting Kit-8 (CCK-8) assay was used to measure cell viability. Western blot assay was conducted to determine the expression of cell proliferation markers proliferating cell nuclear antigen (PCNA) and Ki-67, as well as Lactate dehydrogenase A (LDHA). VSMCs migration and invasion were evaluated by Transwell assay. Luciferase reporter gene assay and RNA immunoprecipitation were performed to validate the target relationship between miR-638 and LDHA. LDHA and miR-638 expression were also determined. Glycolysis of VSMCs was tested by corresponding Kits. RESULTS: Platelet-derived growth factor-bb (PDGF-bb) promoted the VSMCs viability and down-regulated miR-638. Overexpression of miR-638 inhibited cell proliferation, migration and invasion of VSMCs. LDHA was identified as a target of miR-638, and counter-regulated by miR-638. Loss of miR-638 attenuated the suppressor effects on the proliferation, migration and invasion of VSMCs induced by LDHA down-regulation. MiR-638 inhibited the glycolysis of VSMCs by targeting LDHA. CONCLUSION: MiR-638 is down-regulated by PDGF-bb treatment and suppressed the glycolysis of VSMCs via targeting LDHA.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...