Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.387
Filtrar
1.
J Cell Mol Med ; 28(9): e18351, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38693854

RESUMO

Coronary artery bypass grafting (CABG) is an effective treatment for coronary heart disease, with vascular transplantation as the key procedure. Intimal hyperplasia (IH) gradually leads to vascular stenosis, seriously affecting the curative effect of CABG. Mesenchymal stem cells (MSCs) were used to alleviate IH, but the effect was not satisfactory. This work aimed to investigate whether lncRNA MIR155HG could improve the efficacy of MSCs in the treatment of IH and to elucidate the role of the competing endogenous RNA (ceRNA). The effect of MIR155HG on MSCs function was investigated, while the proteins involved were assessed. IH was detected by HE and Van Gieson staining. miRNAs as the target of lncRNA were selected by bioinformatics analysis. qRT-PCR and dual-luciferase reporter assay were performed to verify the binding sites of lncRNA-miRNA. The apoptosis, Elisa and tube formation assay revealed the effect of ceRNA on the endothelial protection of MIR155HG-MSCs. We observed that MIR155HG improved the effect of MSCs on IH by promoting viability and migration. MIR155HG worked as a sponge for miR-205. MIR155HG/miR-205 significantly improved the function of MSCs, avoiding apoptosis and inducing angiogenesis. The improved therapeutic effects of MSCs on IH might be due to the ceRNA role of MIR155HG/miR-205.


Assuntos
Apoptose , Hiperplasia , Células-Tronco Mesenquimais , MicroRNAs , RNA Longo não Codificante , MicroRNAs/genética , MicroRNAs/metabolismo , Células-Tronco Mesenquimais/metabolismo , Humanos , RNA Longo não Codificante/genética , Apoptose/genética , Movimento Celular/genética , Animais , Transplante de Células-Tronco Mesenquimais/métodos , Túnica Íntima/patologia , Túnica Íntima/metabolismo , Regulação da Expressão Gênica , Proliferação de Células/genética , Masculino , Sobrevivência Celular/genética , RNA Endógeno Competitivo
2.
Int J Mol Sci ; 25(9)2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38732029

RESUMO

Neointimal hyperplasia is the main cause of vascular graft failure in the medium term. Vitamin D receptor activation modulates the biology of vascular smooth muscle cells and has been reported to protect from neointimal hyperplasia following endothelial injury. However, the molecular mechanisms are poorly understood. We have now explored the impact of the selective vitamin D receptor activator, paricalcitol, on neointimal hyperplasia, following guidewire-induced endothelial cell injury in rats, and we have assessed the impact of paricalcitol or vehicle on the expression of key cell stress factors. Guidewire-induced endothelial cell injury caused neointimal hyperplasia and luminal stenosis and upregulated the expression of the growth factor growth/differentiation factor-15 (GDF-15), the cytokine receptor CD74, NFκB-inducing kinase (NIK, an upstream regulator of the proinflammatory transcription factor NFκB) and the chemokine monocyte chemoattractant protein-1 (MCP-1/CCL2). Immunohistochemistry confirmed the increased expression of the cellular proteins CD74 and NIK. Paricalcitol (administered in doses of 750 ng/kg of body weight, every other day) had a non-significant impact on neointimal hyperplasia and luminal stenosis. However, it significantly decreased GDF-15, CD74, NIK and MCP-1/CCL2 mRNA expression, which in paricalcitol-injured arteries remained within the levels found in control vehicle sham arteries. In conclusion, paricalcitol had a dramatic effect, suppressing the stress response to guidewire-induced endothelial cell injury, despite a limited impact on neointimal hyperplasia and luminal stenosis. This observation identifies novel molecular targets of paricalcitol in the vascular system, whose differential expression cannot be justified as a consequence of improved tissue injury.


Assuntos
Anti-Inflamatórios , Quimiocina CCL2 , Ergocalciferóis , Hiperplasia , Animais , Ratos , Ergocalciferóis/farmacologia , Masculino , Quimiocina CCL2/metabolismo , Quimiocina CCL2/genética , Anti-Inflamatórios/farmacologia , Neointima/metabolismo , Neointima/patologia , Neointima/tratamento farmacológico , Fator 15 de Diferenciação de Crescimento/metabolismo , Fator 15 de Diferenciação de Crescimento/genética , Túnica Íntima/patologia , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/metabolismo , Antígenos de Diferenciação de Linfócitos B/metabolismo , Antígenos de Diferenciação de Linfócitos B/genética , Células Endoteliais/metabolismo , Células Endoteliais/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe II
3.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167170, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38631407

RESUMO

Intimal hyperplasia (IH) is a common pathological feature of vascular proliferative diseases, such as atherosclerosis and restenosis after angioplasty. Urotensin II (UII) and its receptor (UTR) are widely expressed in cardiovascular tissues. However, it remains unclear whether the UII/UTR system is involved in IH. Right unilateral common carotid artery ligation was performed and maintained for 21 days to induce IH in UTR knockout (UTR-/-) and wild-type (WT) mice. Histological analysis revealed that compared with WT mice, UTR-deficient mice exhibited a decreased neointimal area, angiostenosis and intima-media ratio. Immunostaining revealed fewer smooth muscle cells (SMCs), endothelial cells and macrophages in the lesions of UTR-/- mice than in those of WT mice. Protein interaction analysis suggested that the UTR may affect cell proliferation by regulating YAP and its downstream target genes. In vitro experiments revealed that UII can promote the proliferation and migration of SMCs, and western blotting also revealed that UII increased the protein expression of RhoA, CTGF, Cyclin D1 and PCNA and downregulated p-YAP protein expression, while these effects could be partly reversed by urantide. To evaluate the translational value of UTRs in IH management, WT mice were also treated with two doses of urantide, a UTR antagonist, to confirm the benefit of UTR blockade in IH progression. A high dose of urantide (600 µg/kg/day), rather than a low dose (60 µg/kg/day), successfully improved ligation-induced IH compared with that in mice receiving vehicle. The results of the present study suggested that the UII/UTR system may regulate IH partly through the RhoA-YAP signaling pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proliferação de Células , Hiperplasia , Camundongos Knockout , Receptores Acoplados a Proteínas G , Transdução de Sinais , Proteínas de Sinalização YAP , Proteína rhoA de Ligação ao GTP , Animais , Proteínas de Sinalização YAP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Camundongos , Hiperplasia/metabolismo , Hiperplasia/patologia , Ligadura , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Masculino , Túnica Íntima/patologia , Túnica Íntima/metabolismo , Urotensinas/metabolismo , Urotensinas/genética , Urotensinas/farmacologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Camundongos Endogâmicos C57BL , Movimento Celular , Neointima/metabolismo , Neointima/patologia , Neointima/genética
4.
J Mol Cell Cardiol ; 190: 62-75, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38583797

RESUMO

Intimal hyperplasia is a complicated pathophysiological phenomenon attributable to in-stent restenosis, and the underlying mechanism remains unclear. Interleukin enhancer-binding factor 3 (ILF3), a double-stranded RNA-binding protein involved in regulating mRNA stability, has been recently demonstrated to assume a crucial role in cardiovascular disease; nevertheless, its impact on intimal hyperplasia remains unknown. In current study, we used samples of human restenotic arteries and rodent models of intimal hyperplasia, we found that vascular smooth muscle cell (VSMC) ILF3 expression was markedly elevated in human restenotic arteries and murine ligated carotid arteries. SMC-specific ILF3 knockout mice significantly suppressed injury induced neointimal formation. In vitro, platelet-derived growth factor type BB (PDGF-BB) treatment elevated the level of VSMC ILF3 in a dose- and time-dependent manner. ILF3 silencing markedly inhibited PDGF-BB-induced phenotype switching, proliferation, and migration in VSMCs. Transcriptome sequencing and RNA immunoprecipitation sequencing depicted that ILF3 maintained its stability upon binding to the mRNA of the high-mobility group box 1 protein (HMGB1), thereby exerting an inhibitory effect on the transcription of dual specificity phosphatase 16 (DUSP16) through enhanced phosphorylation of signal transducer and activator of transcription 3 (STAT3). Therefore, the results both in vitro and in vivo indicated that the loss of ILF3 in VSMC ameliorated neointimal hyperplasia by regulating the STAT3/DUSP16 axis through the degradation of HMGB1 mRNA. Our findings revealed that vascular injury activates VSMC ILF3, which in turn promotes intima formation. Consequently, targeting specific VSMC ILF3 may present a potential therapeutic strategy for ameliorating cardiovascular restenosis.


Assuntos
Proteína HMGB1 , Hiperplasia , Camundongos Knockout , Músculo Liso Vascular , Miócitos de Músculo Liso , Proteínas do Fator Nuclear 90 , Estabilidade de RNA , Fator de Transcrição STAT3 , Túnica Íntima , Animais , Humanos , Masculino , Camundongos , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Regulação da Expressão Gênica , Proteína HMGB1/metabolismo , Proteína HMGB1/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Neointima/metabolismo , Neointima/patologia , Proteínas do Fator Nuclear 90/metabolismo , Proteínas do Fator Nuclear 90/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Fator de Transcrição STAT3/metabolismo , Túnica Íntima/metabolismo , Túnica Íntima/patologia
5.
Arterioscler Thromb Vasc Biol ; 44(4): 883-897, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38328936

RESUMO

BACKGROUND: Myeloid cells (MCs) reside in the aortic intima at regions predisposed to atherosclerosis. Systemic inflammation triggers reverse transendothelial migration (RTM) of intimal MCs into the arterial blood, which orchestrates a protective immune response that clears intracellular pathogens from the arterial intima. Molecular pathways that regulate RTM remain poorly understood. S1P (sphingosine-1-phosphate) is a lipid mediator that regulates immune cell trafficking by signaling via 5 G-protein-coupled receptors (S1PRs [S1P receptors]). We investigated the role of S1P in the RTM of aortic intimal MCs. METHODS: Intravenous injection of lipopolysaccharide was used to model a systemic inflammatory stimulus that triggers RTM. CD11c+ intimal MCs in the lesser curvature of the ascending aortic arch were enumerated by en face confocal microscopy. Local gene expression was evaluated by transcriptomic analysis of microdissected intimal cells. RESULTS: In wild-type C57BL/6 mice, lipopolysaccharide induced intimal cell expression of S1pr1, S1pr3, and Sphk1 (a kinase responsible for S1P production). Pharmacological modulation of multiple S1PRs blocked lipopolysaccharide-induced RTM and modulation of S1PR1 and S1PR3 reduced RTM in an additive manner. Cre-mediated deletion of S1pr1 in MCs blocked lipopolysaccharide-induced RTM, confirming a role for myeloid-specific S1PR1 signaling. Global or hematopoietic deficiency of Sphk1 reduced plasma S1P levels, the abundance of CD11c+ MCs in the aortic intima, and blunted lipopolysaccharide-induced RTM. In contrast, plasma S1P levels, the abundance of intimal MCs, and lipopolysaccharide-induced RTM were rescued in Sphk1-/- mice transplanted with Sphk1+/+ or mixed Sphk1+/+ and Sphk1-/- bone marrow. Stimulation with lipopolysaccharide increased endothelial permeability and intimal MC exposure to circulating factors such as S1P. CONCLUSIONS: Functional and expression studies support a novel role for S1P signaling in the regulation of lipopolysaccharide-induced RTM and the homeostatic maintenance of aortic intimal MCs. Our data provide insight into how circulating plasma mediators help orchestrate intimal MC dynamics.


Assuntos
Receptores de Lisoesfingolipídeo , Migração Transendotelial e Transepitelial , Camundongos , Animais , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/metabolismo , Lipopolissacarídeos/toxicidade , Camundongos Endogâmicos C57BL , Esfingosina/metabolismo , Células Mieloides/metabolismo , Lisofosfolipídeos/metabolismo , Túnica Íntima/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo
6.
Int J Mol Sci ; 24(12)2023 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-37373529

RESUMO

Saphenous vein bypass grafting is an effective technique used to treat peripheral arterial disease (PAD). However, restenosis is the major clinical challenge for the graft vessel among people with PAD postoperation. We hypothesize that there is a common culprit behind arterial occlusion and graft restenosis. To investigate this hypothesis, we found TGF-ß, a gene specifically upregulated in PAD arteries, by bioinformatics analysis. TGF-ß has a wide range of biological activities and plays an important role in vascular remodeling. We discuss the molecular pathway of TGF-ß and elucidate its mechanism in vascular remodeling and intimal hyperplasia, including EMT, extracellular matrix deposition, and fibrosis, which are the important pathways contributing to stenosis. Additionally, we present a case report of a patient with graft restenosis linked to the TGF-ß pathway. Finally, we discuss the potential applications of targeting the TGF-ß pathway in the clinic to improve the long-term patency of vein grafts.


Assuntos
Doença Arterial Periférica , Túnica Íntima , Humanos , Túnica Íntima/metabolismo , Veia Safena/metabolismo , Remodelação Vascular , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Doença Arterial Periférica/cirurgia , Doença Arterial Periférica/metabolismo
7.
FASEB J ; 37(1): e22701, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36520031

RESUMO

Calcification of the medial layer, inducing arterial stiffness, contributes significantly to cardiovascular mortality in patients with chronic kidney disease (CKD). Extracellular nucleotides block the mineralization of arteries by binding to purinergic receptors including the P2Y2 receptor. This study investigates whether deletion of the P2Y2 receptor influences the development of arterial media calcification in CKD mice. Animals were divided into: (i) wild type mice with normal renal function (control diet) (n = 8), (ii) P2Y2 R-/- mice with normal renal function (n = 8), (iii) wild type mice with CKD (n = 27), and (iv) P2Y2 R-/- mice with CKD (n = 22). To induce CKD, animals received an alternating (0.2-0.3%) adenine diet for 7 weeks. All CKD groups developed a similar degree of chronic renal failure as reflected by high serum creatinine and phosphorus levels. Also, the presence of CKD induced calcification in the heart and medial layer of the aortic wall. However, deletion of the P2Y2 receptor makes CKD mice more susceptible to the development of calcification in the heart and aorta (aortic calcium scores (median ± IQR), CKD-wild type: 0.34 ± 4.3 mg calcium/g wet tissue and CKD-P2Y2 R-/- : 4.0 ± 13.2 mg calcium/g wet tissue). As indicated by serum and aortic mRNA markers, this P2Y2 R-/- mediated increase in CKD-related arterial media calcification was associated with an elevation of calcification stimulators, including alkaline phosphatase and inflammatory molecules interleukin-6 and lipocalin 2. The P2Y2 receptor should be considered as an interesting therapeutic target for tackling CKD-related arterial media calcification.


Assuntos
Fosfatase Alcalina , Lipocalina-2 , Insuficiência Renal Crônica , Túnica Íntima , Calcificação Vascular , Animais , Camundongos , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Cálcio/metabolismo , Lipocalina-2/genética , Lipocalina-2/metabolismo , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/genética , Insuficiência Renal Crônica/metabolismo , Túnica Íntima/metabolismo , Túnica Íntima/patologia , Regulação para Cima , Calcificação Vascular/etiologia , Calcificação Vascular/genética , Calcificação Vascular/metabolismo
8.
Cell Transplant ; 31: 9636897221122999, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36066039

RESUMO

Coronary artery bypass grafting remains the gold standard in the therapy of advanced-stage patients. But the vein grafts are prone to restenosis or failure. Pentoxifylline (PTX) is a methylxanthine derivative with a function of inhibiting cell proliferation. We thus applied PTX locally to the vein grafts to study its effect on the inhibition of graft restenosis using a rat vein graft model. Morphometric results showed a significant decrease in the thickness of vein grafts intimal and medial at day 28 after the bypass operation. Results from Western blot and immunohistochemistry showed that PTX also significantly reduced the proliferating cell nuclear antigen (PCNA), alpha-smooth muscle actin (α-SMA) expression, and phosphorylation of p38 in vein grafts. These results firstly discovered the positive role of PTX in preventing the vein grafts restenosis and the mechanism may be inhibition of vascular smooth muscle cells (VSMCs) proliferation via the p38MAPK pathway.


Assuntos
Pentoxifilina , Proteínas Quinases p38 Ativadas por Mitógeno , Animais , Proliferação de Células , Ponte de Artéria Coronária , Hiperplasia/metabolismo , Hiperplasia/patologia , Pentoxifilina/metabolismo , Pentoxifilina/farmacologia , Pentoxifilina/uso terapêutico , Ratos , Túnica Íntima/metabolismo , Túnica Íntima/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Cardiovasc Diabetol ; 21(1): 72, 2022 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-35549955

RESUMO

BACKGROUND: Individuals with type 2 diabetes mellitus (T2DM) have an increased risk for developing macrovascular disease (MVD) manifested by atherosclerosis. Phenotypically and functionally different monocyte subsets (classical; CD14++CD16-, non-classical; CD14+CD16++, and intermediate; CD14++CD16+) including pro-angiogenic monocytes expressing Tie2 (TEMs) can be identified. Here we investigated monocyte heterogeneity and its association with T2DM and MVD. METHODS: Individuals with (N = 51) and without (N = 56) T2DM were recruited and allocated to "non-MVD" or "with MVD" (i.e., peripheral or coronary artery disease) subgroups. Blood monocyte subsets were quantified based on CD14, CD16 and Tie2 expression levels. Plasma levels of Tie2-ligands angiopoietin-1 and angiopoietin-2 were determined using ELISA. Carotid endarterectomy samples from individuals with (N = 24) and without (N = 22) T2DM were stained for intraplaque CD68+ macrophages (inflammation) and CD34+ (angiogenesis), as plaque vulnerability markers. RESULTS: Monocyte counts were similar between individuals with T2DM and healthy controls (non-diabetic, non-MVD). Non-classical monocytes were reduced (p < 0.05) in T2DM, whereas the percentage of TEMs within the intermediate subset was increased (p < 0.05). T2DM was associated with increased angiopoietin-1 (p < 0.05) and angiopoietin-2 (p = 0.0001) levels. Angiopoietin-2 levels were higher in T2DM individuals with MVD compared with non-MVD (p < 0.01). Endarterectomized plaques showed no differences in macrophage influx and microvessel number between individuals with and without T2DM. CONCLUSIONS: Monocyte subset distribution is altered in T2DM with reduced non-classical monocytes and increased TEM percentage in the intermediate monocyte subset. Increased angiopoietin-2 levels together with increased frequency of TEMs might promote plaque vulnerability in T2DM which could however not be confirmed at tissue level in advanced atherosclerotic lesions.


Assuntos
Aterosclerose , Diabetes Mellitus Tipo 2 , Placa Aterosclerótica , Angiopoietina-1/metabolismo , Angiopoietina-2/metabolismo , Aterosclerose/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Monócitos/metabolismo , Placa Aterosclerótica/patologia , Receptor TIE-2 , Túnica Íntima/química , Túnica Íntima/metabolismo , Túnica Íntima/patologia
10.
Pharmacol Ther ; 235: 108157, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35183591

RESUMO

Intimal hyperplasia (IH) occurs in a considerable number of cases of blood vessel reconstruction by stenting or balloon angioplasty, venous bypass grafting, and arteriovenous dialysis accesses. It is triggered by endothelial injury during the vascular intervention and leads to vessel restenosis with life-threatening consequences for patients. To date, the drugs used for IH prevention in clinics-paclitaxel and rapalog drugs-have been focusing primarily on the vascular smooth muscle cell (VSMC) proliferation pathway of IH development. Limitations, such as endothelial toxicity and inappropriate drug administration timing, have spurred the search for new and efficient pharmacological approaches to control IH. In this state-of-the-art review, we present the pathways of IH development, focusing on the key events and actors involved in IH. Subsequently, we discuss different drugs and drug combinations interfering with these pathways based on their effect on peripheral circulation IH models in animal studies, or on clinical reports. The reports were obtained through an extensive search of peer-reviewed publications in Pubmed, Embase, and Google Scholar, with search equations composed based on five concepts around IH and their various combinations. To improve vascular intervention outcomes, rethinking of conventional therapeutic approaches to IH prevention is needed. Exploring local application of drugs and drug combinations acting on different pathophysiological pathways of IH development has the potential to provide effective and safe restenosis prevention.


Assuntos
Túnica Íntima , Animais , Humanos , Hiperplasia/metabolismo , Hiperplasia/patologia , Hiperplasia/prevenção & controle , Túnica Íntima/metabolismo , Túnica Íntima/patologia
11.
Gene ; 819: 146233, 2022 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-35121027

RESUMO

BACKGROUND: Due to permanent aortic dilation, thoracic aortic aneurysm (TAA) is a life-threatening disease. Once ruptured, TAA has a high lethality and disability rate. Although studies have focused on transcriptomic alterations in TAA, more detailed analysis is still lacking, especially the different aortic intima-media and adventitia roles. This study aimed to identify the different co-expression patterns between the aortic intima-media and the adventitia underlying the aortic dilation. METHODS: We analyzed the gene expression profiles obtained from Gene Expression Omnibus (GEO, GSE26155) database. With a false discovery rate (FDR) < 0.05 and |log2FC| ≥ 1, 56 and 33 differential genes in the intima-media and adventitia, respectively, between the non-dilated and dilated status. Gene ontology (GO) and gene set enrichment analysis revealed that degranulation and activation of neutrophils play an essential role in the intima-media of dilated aortas. Through weighted gene co-expression network analysis (WGCNA), we identified essential co-expressed modules and hub genes to explore the biological functions of the dysregulated genes. RESULTS: Functional pathway analysis suggested that lipid metabolism, C-C motif chemokine pathways were significantly enriched in the adventitia, whereas ribosome proteins and related mRNA translation pathways were closely related to intima and media. Furthermore, the ssGSEA analysis indicated that macrophages, helper T cells, and neutrophils were higher in the intima-media of the dilated thoracic aorta. Finally, we validated the critical findings of the study with the murine model of TAA. CONCLUSION: This study identified and verified hub genes and pathways in aortic intima-media and adventitia prominently associated with aortic dilation, providing practical understanding in the perspective of searching for new molecular targets.


Assuntos
Túnica Adventícia/metabolismo , Aneurisma da Aorta Torácica/genética , Aneurisma da Aorta Torácica/metabolismo , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Transcriptoma , Túnica Íntima/metabolismo , Animais , Quimiocinas/metabolismo , Dilatação Patológica/genética , Dilatação Patológica/metabolismo , Perfilação da Expressão Gênica/métodos , Humanos , Inflamação , Metabolismo dos Lipídeos , RNA Mensageiro/metabolismo , Proteínas Ribossômicas/metabolismo
12.
Int J Mol Sci ; 23(3)2022 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-35163232

RESUMO

Despite all the advances of modern medicine, atherosclerosis continues to be one of the most important medical and social problems. Atherosclerosis is the cause of several cardiovascular diseases, which are associated with high rates of disability and mortality. The development of atherosclerosis is associated with the accumulation of lipids in the arterial intima and the disruption of mechanisms that maintain the balance between the development and resolution of inflammation. Fatty acids are involved in many mechanisms of inflammation development and maintenance. Endothelial cells demonstrate multiple cross-linkages between lipid metabolism and innate immunity. In addition, these processes are linked to hemodynamics and the function of other cells in the vascular wall, highlighting the central role of the endothelium in vascular biology.


Assuntos
Aterosclerose/imunologia , Aterosclerose/metabolismo , Ácidos Graxos/metabolismo , Animais , Doenças Cardiovasculares/metabolismo , Eicosanoides/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Ácidos Graxos/imunologia , Hemodinâmica , Humanos , Imunidade Inata , Inflamação/imunologia , Inflamação/metabolismo , Metabolismo dos Lipídeos/imunologia , Metabolismo dos Lipídeos/fisiologia , Lipídeos/fisiologia , Túnica Íntima/metabolismo
13.
Oxid Med Cell Longev ; 2021: 3010577, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34853629

RESUMO

Mediator complex subunit 1 (MED1) is a component of the mediator complex and functions as a coactivator involved in the regulated transcription of nearly all RNA polymerase II-dependent genes. Previously, we showed that MED1 in macrophages has a protective effect on atherosclerosis; however, the effect of MED1 on intimal hyperplasia and mechanisms regulating proinflammatory cytokine production after macrophage MED1 deletion are still unknown. In this study, we report that MED1 macrophage-specific knockout (MED1 ΔMac) mice showed aggravated neointimal hyperplasia, vascular smooth muscle cells (VSMCs), and macrophage accumulation in injured arteries. Moreover, MED1 ΔMac mice showed increased proinflammatory cytokine production after an injury to the artery. After lipopolysaccharide (LPS) treatment, MED1 ΔMac macrophages showed increased generation of reactive oxygen species (ROS) and reduced expression of peroxisome proliferative activated receptor gamma coactivator-1α (PGC1α) and antioxidant enzymes, including catalase and glutathione reductase. The overexpression of PGC1α attenuated the effects of MED1 deficiency in macrophages. In vitro, conditioned media from MED1 ΔMac macrophages induced more proliferation and migration of VSMCs. To explore the potential mechanisms by which MED1 affects inflammation, macrophages were treated with BAY11-7082 before LPS treatment, and the results showed that MED1 ΔMac macrophages exhibited increased expression of phosphorylated-p65 and phosphorylated signal transducer and activator of transcription 1 (p-STAT1) compared with the control macrophages, suggesting the enhanced activation of NF-κB and STAT1. In summary, these data showed that MED1 deficiency enhanced inflammation and the proliferation and migration of VSMCs in injured vascular tissue, which may result from the activation of NF-κB and STAT1 due to the accumulation of ROS.


Assuntos
Inflamação/metabolismo , Macrófagos/metabolismo , Subunidade 1 do Complexo Mediador/deficiência , Espécies Reativas de Oxigênio/metabolismo , Túnica Íntima/metabolismo , Animais , Hiperplasia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos , Túnica Íntima/patologia
14.
Int J Mol Sci ; 22(22)2021 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-34830334

RESUMO

An association between high serum calcium/phosphate and cardiovascular events or death is well-established. However, a mechanistic explanation of this correlation is lacking. Here, we examined the role of calciprotein particles (CPPs), nanoscale bodies forming in the human blood upon its supersaturation with calcium and phosphate, in cardiovascular disease. The serum of patients with coronary artery disease or cerebrovascular disease displayed an increased propensity to form CPPs in combination with elevated ionised calcium as well as reduced albumin levels, altogether indicative of reduced Ca2+-binding capacity. Intravenous administration of CPPs to normolipidemic and normotensive Wistar rats provoked intimal hyperplasia and adventitial/perivascular inflammation in both balloon-injured and intact aortas in the absence of other cardiovascular risk factors. Upon the addition to primary human arterial endothelial cells, CPPs induced lysosome-dependent cell death, promoted the release of pro-inflammatory cytokines, stimulated leukocyte adhesion, and triggered endothelial-to-mesenchymal transition. We concluded that CPPs, which are formed in the blood as a result of altered mineral homeostasis, cause endothelial dysfunction and vascular inflammation, thereby contributing to the development of cardiovascular disease.


Assuntos
Angina Pectoris/fisiopatologia , Isquemia Encefálica/fisiopatologia , Cloreto de Cálcio/sangue , Doença da Artéria Coronariana/fisiopatologia , Células Endoteliais/patologia , Infarto do Miocárdio/fisiopatologia , Fosfatos/sangue , Angina Pectoris/sangue , Angina Pectoris/genética , Animais , Aorta/metabolismo , Aorta/patologia , Isquemia Encefálica/sangue , Isquemia Encefálica/genética , Cloreto de Cálcio/química , Estudos de Casos e Controles , Morte Celular , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/genética , Células Endoteliais/metabolismo , Transição Epitelial-Mesenquimal , Floculação , Regulação da Expressão Gênica , Humanos , Inflamação , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Leucócitos/metabolismo , Leucócitos/patologia , Lisossomos/metabolismo , Lisossomos/patologia , Masculino , Infarto do Miocárdio/sangue , Infarto do Miocárdio/genética , Fosfatos/química , Cultura Primária de Células , Ratos , Ratos Wistar , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Túnica Íntima/metabolismo , Túnica Íntima/patologia , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
15.
J Nanobiotechnology ; 19(1): 374, 2021 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-34789284

RESUMO

BACKGROUND: Intimal hyperplasia caused by vascular injury is an important pathological process of many vascular diseases, especially occlusive vascular disease. In recent years, Nano-drug delivery system has attracted a wide attention as a novel treatment strategy, but there are still some challenges such as high clearance rate and insufficient targeting. RESULTS: In this study, we report a biomimetic ROS-responsive MM@PCM/RAP nanoparticle coated with macrophage membrane. The macrophage membrane with the innate "homing" capacity can superiorly regulate the recruitment of MM@PCM/RAP to inflammatory lesion to enhance target efficacy, and can also disguise MM@PCM/RAP nanoparticle as the autologous cell to avoid clearance by the immune system. In addition, MM@PCM/RAP can effectively improve the solubility of rapamycin and respond to the high concentration level of ROS accumulated in pathological lesion for controlling local cargo release, thereby increasing drug availability and reducing toxic side effects. CONCLUSIONS: Our findings validate that the rational design, biomimetic nanoparticles MM@PCM/RAP, can effectively inhibit the pathological process of intimal injury with excellent biocompatibility.


Assuntos
Hiperplasia/metabolismo , Macrófagos/citologia , Sistemas de Liberação de Fármacos por Nanopartículas , Espécies Reativas de Oxigênio/metabolismo , Túnica Íntima , Animais , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Materiais Biomiméticos/farmacologia , Membrana Celular/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistemas de Liberação de Fármacos por Nanopartículas/química , Sistemas de Liberação de Fármacos por Nanopartículas/metabolismo , Sirolimo/química , Sirolimo/farmacocinética , Sirolimo/farmacologia , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/metabolismo , Peixe-Zebra
16.
Ann Biomed Eng ; 49(12): 3425-3437, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34734362

RESUMO

Macrophage to foam cell transition and their accumulation in the arterial intima are the key events that trigger atherosclerosis, a multifactorial inflammatory disease. Previous studies have linked arterial stiffness and cardiovascular disease and have highlighted the use of arterial stiffness as a potential early-stage marker. Yet the relationship between arterial stiffness and atherosclerosis in terms of macrophage function is poorly understood. Thus, it is pertinent to understand the mechanobiology of macrophages to clarify their role in plaque advancement. We explore how substrate stiffness affects proliferation of macrophages and foam cells, traction forces exerted by macrophages and uptake of native and oxidized low-density lipoproteins. We demonstrate that stiffness influences foam cell proliferation under both naïve and inflammatory conditions. Naïve foam cells proliferated faster on the 4 kPa polyacrylamide gel and glass whereas under inflammatory conditions, maximum proliferation was recorded on glass. Macrophage and foam cell traction forces were positively correlated to the substrate stiffness. Furthermore, the influence of stiffness was demonstrated on the uptake of lipoproteins on macrophages treated with lipopolysaccharide + interferon gamma. Cells on softer 1 kPa substrates had a significantly higher uptake of low-density lipoproteins and oxidized low-density lipoproteins compared to stiffer substrates. The results herein indicate that macrophage function is modulated by stiffness and help better understand ways in which macrophages and foam cells could contribute to the development and progression of atherosclerotic plaque.


Assuntos
Proliferação de Células , Células Espumosas/metabolismo , Inflamação/fisiopatologia , Lipoproteínas/metabolismo , Macrófagos/metabolismo , Placa Aterosclerótica/fisiopatologia , Rigidez Vascular/fisiologia , Células Cultivadas , Humanos , Inflamação/metabolismo , Placa Aterosclerótica/metabolismo , Túnica Íntima/metabolismo
17.
J Cell Mol Med ; 25(19): 9427-9433, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34459107

RESUMO

No study has examined the differential value of arterial intima thickness in the subtypes of acute ischaemic stroke. This study aimed to assess whether intima thickness of carotid artery (CIT), radial artery (RIT) and dorsalis pedis artery (PIT) have an independent and additive value in differentiating ischaemic stroke subtypes due to large-artery atherosclerosis (LAA) or small-vessel occlusion (SVO). One hundred and sixty-one patients with LAA and 79 patients with SVO were recruited. CIT, RIT and PIT were measured with a 24-MHz ultrasound transducer. Binary logistic regression analysis was used to evaluate the differential values of the different parameters in the two subtypes. ROC curve analyses were plotted to compare the differential performance of different parameters and the combination model. Both RIT and PIT were substantially thicker in LAA than in SVO stroke patients. RIT and carotid intima-media thickness had similar performances in differentiating stroke subtypes. Introduction of RIT to traditional atherosclerotic associated risk factors had a marginal satisfactory differential performance for LAA and SVO stroke patients (AUC 0.775). RIT is a promising parameter for LAA and SVO subgroup classification. The combination of RIT and traditional risk factors might be a promising tool for differentiating ischaemic stroke subgroups.


Assuntos
Aterosclerose/complicações , Aterosclerose/patologia , AVC Isquêmico/etiologia , AVC Isquêmico/patologia , Túnica Íntima/metabolismo , Idoso , Aterosclerose/diagnóstico por imagem , Aterosclerose/metabolismo , Biomarcadores , Espessura Intima-Media Carotídea , Suscetibilidade a Doenças , Feminino , Humanos , AVC Isquêmico/diagnóstico por imagem , AVC Isquêmico/metabolismo , Metabolismo dos Lipídeos , Lipídeos/sangue , Masculino , Pessoa de Meia-Idade , Razão de Chances , Curva ROC , Fatores de Risco , Ultrassonografia
18.
FASEB J ; 35(9): e21831, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34383982

RESUMO

The nuclear factor of activated T-cells 5 (NFAT5) is a transcriptional regulator of macrophage activation and T-cell development, which controls stabilizing responses of cells to hypertonic and biomechanical stress. In this study, we detected NFAT5 in the media layer of arteries adjacent to human arteriosclerotic plaques and analyzed its role in vascular smooth muscle cells (VSMCs) known to contribute to arteriosclerosis through the uptake of lipids and transformation into foam cells. Exposure of both human and mouse VSMCs to cholesterol stimulated the nuclear translocation of NFAT5 and increased the expression of the ATP-binding cassette transporter Abca1, required to regulate cholesterol efflux from cells. Loss of Nfat5 promoted cholesterol accumulation in these cells and inhibited the expression of genes involved in the management of oxidative stress or lipid handling, such as Sod1, Plin2, Fabp3, and Ppard. The functional relevance of these observations was subsequently investigated in mice fed a high-fat diet upon induction of a smooth muscle cell-specific genetic ablation of Nfat5 (Nfat5(SMC)-/- ). Under these conditions, Nfat5(SMC)-/- but not Nfat5fl/fl mice developed small, focal lipid-rich lesions in the aorta after 14 and 25 weeks, which were formed by intracellular lipid droplets deposited in the sub-intimal VSMCs layer. While known for being activated by external stimuli, NFAT5 was found to mediate the expression of VSMC genes associated with the handling of lipids in response to a cholesterol-rich environment. Failure of this protective function may promote the formation of lipid-laden arterial VSMCs and pro-atherogenic vascular responses.


Assuntos
Aorta/metabolismo , Metabolismo dos Lipídeos/fisiologia , Lipídeos/fisiologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Fatores de Transcrição/metabolismo , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Idoso , Animais , Aterosclerose/metabolismo , Células Cultivadas , Colesterol/metabolismo , Feminino , Células Espumosas/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Hipercolesterolemia/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Estresse Oxidativo/fisiologia , Túnica Íntima/metabolismo
19.
Circulation ; 144(6): 455-470, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34111946

RESUMO

BACKGROUND: Coronary allograft vasculopathy (CAV) is a devastating sequela of heart transplant in which arterial intimal thickening limits coronary blood flow. There are currently no targeted therapies to prevent or reduce this pathology that leads to transplant failure. Vascular smooth muscle cell (VSMC) phenotypic plasticity is critical in CAV neointima formation. TET2 (TET methylcytosine dioxygenase 2) is an important epigenetic regulator of VSMC phenotype, but the role of TET2 in the progression of CAV is unknown. METHODS: We assessed TET2 expression and activity in human CAV and renal transplant samples. We also used the sex-mismatched murine aortic graft model of graft arteriopathy (GA) in wild-type and inducible smooth muscle-specific Tet2 knockout mice; and in vitro studies in murine and human VSMCs using knockdown, overexpression, and transcriptomic approaches to assess the role of TET2 in VSMC responses to IFNγ (interferon γ), a cytokine elaborated by T cells that drives CAV progression. RESULTS: In the present study, we found that TET2 expression and activity are negatively regulated in human CAV and renal transplant samples and in the murine aortic graft model of GA. IFNγ was sufficient to repress TET2 and induce an activated VSMC phenotype in vitro. TET2 depletion mimicked the effects of IFNγ, and TET2 overexpression rescued IFNγ-induced dedifferentiation. VSMC-specific TET2 depletion in aortic grafts, and in the femoral wire restenosis model, resulted in increased VSMC apoptosis and medial thinning. In GA, this apoptosis was tightly correlated with proliferation. In vitro, TET2-deficient VSMCs undergo apoptosis more readily in response to IFNγ and expressed a signature of increased susceptibility to extrinsic apoptotic signaling. Enhancing TET2 enzymatic activity with high-dose ascorbic acid rescued the effect of GA-induced VSMC apoptosis and intimal thickening in a TET2-dependent manner. CONCLUSIONS: TET2 is repressed in CAV and GA, likely mediated by IFNγ. TET2 serves to protect VSMCs from apoptosis in the context of transplant vasculopathy or IFNγ stimulation. Promoting TET2 activity in vivo with systemic ascorbic acid reduces VSMC apoptosis and intimal thickening. These data suggest that promoting TET2 activity in CAV may be an effective strategy for limiting CAV progression.


Assuntos
Apoptose/genética , Proteínas de Ligação a DNA/genética , Dioxigenases/genética , Miócitos de Músculo Liso/metabolismo , Túnica Íntima/metabolismo , Túnica Íntima/patologia , Doenças Vasculares/etiologia , Doenças Vasculares/metabolismo , Aloenxertos , Animais , Biomarcadores , Proteínas de Ligação a DNA/metabolismo , Dioxigenases/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Transplante de Coração/efeitos adversos , Humanos , Imuno-Histoquímica , Interferon gama/metabolismo , Camundongos , Camundongos Knockout , Fator de Transcrição STAT1 , Transdução de Sinais , Doenças Vasculares/patologia
20.
Am J Physiol Heart Circ Physiol ; 320(6): H2438-H2447, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33961504

RESUMO

Adventitial abnormalities including enhanced vasa vasorum malformation are associated with development and vulnerability of atherosclerotic plaque. However, the mechanisms of vasa vasorum malformation and its role in vascular remodeling have not been fully clarified. We recently reported that ninjurin-1 (Ninj1) is a crucial adhesion molecule for pericytes to form matured neovessels. The purpose is to examine if Ninj1 regulates adventitial angiogenesis and affects the vascular remodeling of injured vessels using pericyte-specific Ninj1 deletion mouse model. Mouse femoral arteries were injured by insertion of coiled wire. Four weeks after vascular injury, fixed arteries were decolorized. Vascular remodeling, including intimal hyperplasia and adventitial microvessel formation were estimated in a three-dimensional view. Vascular fragility, including blood leakiness was estimated by extravasation of fluorescein isothiocyanate (FITC)-lectin or FITC-dextran from microvessels. Ninj1 expression was increased in pericytes in response to vascular injury. NG2-CreER/Ninj1loxp mice were treated with tamoxifen (Tam) to induce deletion of Ninj1 in pericyte (Ninj1 KO). Tam-treated NG2-CreER or Tam-nontreated NG2-CreER/Ninj1loxp mice were used as controls. Intimal hyperplasia was significantly enhanced in Ninj1 KO compared with controls. Vascular leakiness was significantly enhanced in Ninj1 KO. In Ninj1 KO, the number of infiltrated macrophages in adventitia was increased, along with the expression of inflammatory cytokines. In conclusion, deletion of Ninj1 in pericytes induces the immature vasa vasorum formation of injured vasculature and exacerbates adventitial inflammation and intimal hyperplasia. Thus, Ninj1 contributes to the vasa vasorum maturation in response to vascular injury and to reduction of vascular remodeling.NEW & NOTEWORTHY Although abnormalities of adventitial vasa vasorum are associated with vascular remodeling such as atherosclerosis, the mechanisms of vasa vasorum malformation and its role in vascular remodeling have not been fully clarified. The present study provides a line of novel evidence that ninjurin-1 contributes to adventitial microvascular maturation during vascular injury and regulates vascular remodeling.


Assuntos
Moléculas de Adesão Celular Neuronais/genética , Artéria Femoral/metabolismo , Neointima/genética , Fatores de Crescimento Neural/genética , Pericitos/metabolismo , Vasa Vasorum/metabolismo , Remodelação Vascular/genética , Túnica Adventícia/metabolismo , Túnica Adventícia/patologia , Animais , Artéria Femoral/lesões , Artéria Femoral/patologia , Técnicas de Inativação de Genes , Hiperplasia/genética , Inflamação/genética , Inflamação/metabolismo , Macrófagos/patologia , Camundongos , Neointima/patologia , Neovascularização Fisiológica/genética , Transcriptoma , Túnica Íntima/metabolismo , Túnica Íntima/patologia , Vasa Vasorum/patologia , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/metabolismo , Lesões do Sistema Vascular/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA