Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Int J Cardiol ; 366: 11-18, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-35716948

RESUMO

BACKGROUND: Vascular calcification (VC), as a prevalent feature of atherosclerosis (AS), is a life-threatening pathological change. Mitofusin 2 (MFN2) has been reported to be down-regulated and participate in the pathogenesis of AS. Here, we explored the feasible impacts of MFN2 on VC in AS. METHODS: Atherosclerotic lesion was evaluated by Oil Red O staining. The VC was detected by Alizarin Red S staining, ALP staining, and calcium content in vascular smooth muscle cells (VSMCs) or atherosclerotic mice. The chondrocyte differentiation of VSMCs was measured by Alcian blue staining. Western blotting and qRT-PCR were used to determine the protein and mRNA expression of associated molecules. Intermolecular interaction was measured by ChIP and dual luciferase assays. RESULTS: The expression of MFN2 and E2F1 was reduced in the aorta tissues of AS patients and mice. Silencing of MFN2 drove calcification in VSMCs and aortas of atherosclerotic mice as confirmed by up-regulating RUNX2, OPG levels, and down-regulating SM22α, α-SMA levels. The chondrocyte differentiation of VSMCs was accelerated by MFN2 knockdown through inducing the expression of Aggrecan, Collagen II, and SOX9. In addition, E2F1 promoted the transcription and expression of MFN2 in VSMCs. Overexpression of MFN2 or E2F1 suppressed ox-LDL-induced VSMC calcification. Finally, MFN2 depletion enhanced VSMC calcification via activating RAS-RAF-ERK1/2 pathway. CONCLUSION: Our results suggest that silencing of MFN2 drives VC via activating RAS-RAF-ERK1/2 pathway in the progression of AS, thus MFN2 may be a therapeutic target for AS.


Assuntos
Aterosclerose , Calcificação Vascular , Animais , Aterosclerose/metabolismo , Diferenciação Celular , Células Cultivadas , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Calcificação Vascular/metabolismo
2.
J Cardiovasc Transl Res ; 15(6): 1315-1326, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35467292

RESUMO

Atherosclerosis is the most common arterial disease and is closely related to vascular calcification. CircHIPK3 has been implicated in atherosclerosis development, but the possible downstream regulatory mechanisms remain unclear. The levels of circHIPK3, miR-106a and MFN2 in tissues and blood samples of patients with atherosclerosis were detected by RT-qPCR. The levels of circHIPK3, miR-106a and MFN2 were detected by RT-qPCR and the expression levels of MFN2, osteogenic and cartilage differentiation marker proteins were detected by western blot in vitro. ALP staining, Alizarin Red staining, and calcium content detection evaluated the degree of osteogenic differentiation of cells. Alcian blue staining detected the level of cell cartilage differentiation. Luciferase detected the targeting relationship between circHIPK3 and miR-106a-5p, as well as miR-106a-5p and MFN2. CircHIPK3 and MFN2 were low expressed and miR-106a-5p was highly expressed in tissues and blood samples of patients with atherosclerosis, as well as vascular smooth muscle cell (VSMC) with osteogenic and cartilage differentiation. Overexpression of circHIPK3 reduced the cell mineralization and calcium content. Overexpression of circHIPK3 inhibited osteogenic differentiation by decreasing ALP activity, RUNX2, and OPG expression, and increasing SM22α and SMA level. What's more, overexpression of circHIPK3 decreased the chondrogenic differentiation by inhibiting the protein level of SOX9, aggrecan, and collagen II. CircHIPK3 targeted miR-106a-5p and miR-106a-5p targeted MFN2. MiR-106a-5p overexpression or MFN2 depletion repressed the effect of circHIPK3 overexpression on VSMC calcification. CircHIPK3 regulated osteogenic and cartilage differentiation of VSMC via miR-106a-5p/MFN2 axis, indicating a target for treating vascular calcification.


Assuntos
Aterosclerose , GTP Fosfo-Hidrolases , MicroRNAs , RNA Circular , Calcificação Vascular , Humanos , Aterosclerose/genética , Cálcio , Diferenciação Celular , GTP Fosfo-Hidrolases/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Mitocondriais/genética , Músculo Liso Vascular/metabolismo , Osteogênese/genética , Calcificação Vascular/genética , RNA Circular/genética
3.
Ann Vasc Surg ; 45: 231-238, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28687504

RESUMO

BACKGROUND: Infected abdominal aortic aneurysms (iAAAs) are rare but life-threatening diseases. The purpose of the present study was to report our experience of extra-anatomic prosthesis bypass in the retroperitoneum as a treatment for iAAAs. METHODS: Data of 8 consecutive patients diagnosed with iAAAs and treated by an extra-anatomic prosthesis bypass in the retroperitoneum were retrospectively collected. Operative details were as follows: one side of the retroperitoneal space was selected to build a track, and a bifurcated expanded polytetrafluoroethylene prosthesis was placed through the track. The proximal end of the prosthesis was sutured with the normal segment of abdominal aorta proximal to the infected aneurysm by end-to-end anostomosis. The 2 distal ends of the prosthesis were, respectively, sutured with the external iliac artery distal to the aneurysm. The anastomoses were then consolidated with the nearby connective tissue. After the closure of the retroperitoneum, the infected aneurysm was incised, and the infected tissue was debrided. Drainage tubes were placed in the aneurysm sac, which was packed with an omentum flap. All patients received perioperative antibiotic therapy for a period of time. All 8 patients were regularly followed up by outpatient observation. RESULTS: Eight patients with iAAAs underwent an extra-anatomic prosthesis bypass in the retroperitoneum and debridement of the infected aneurysm. An emergency operation was performed for 1 patient who underwent concomitant gastrointestinal procedures for aortoduodenal fistula. All 8 patients were definitively diagnosed by one or more sequential computed tomography scans combined with other methods. The blood or tissue cultures of all cases were positive in the perioperative period, with Salmonella (5 cases) being the most common pathogens. Other pathogens included Burkholderia pseudomallei (2 cases) and Escherichia coli (1 case). All patients survived and were discharged in 4-5 weeks after their operations. All patients were free from graft infection during the follow-up period. CONCLUSIONS: The extra-anatomic prosthesis bypass in the retroperitoneum for treating iAAAs was safe and effective. Our experience with the procedure may provide a new approach for the treatment of this disease.


Assuntos
Aneurisma Infectado/cirurgia , Aneurisma da Aorta Abdominal/cirurgia , Implante de Prótese Vascular/métodos , Idoso , Anastomose Cirúrgica , Aneurisma Infectado/diagnóstico por imagem , Aneurisma Infectado/microbiologia , Antibacterianos/administração & dosagem , Aneurisma da Aorta Abdominal/diagnóstico por imagem , Aneurisma da Aorta Abdominal/microbiologia , Aortografia/métodos , Prótese Vascular , Implante de Prótese Vascular/efeitos adversos , Implante de Prótese Vascular/instrumentação , China , Angiografia por Tomografia Computadorizada , Drenagem , Feminino , Hospitais Gerais , Humanos , Masculino , Pessoa de Meia-Idade , Omento/cirurgia , Politetrafluoretileno , Desenho de Prótese , Espaço Retroperitoneal/cirurgia , Estudos Retrospectivos , Retalhos Cirúrgicos , Fatores de Tempo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...