Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.112
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Nat Immunol ; 21(10): 1194-1204, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32895539

RESUMO

Early atherosclerosis depends upon responses by immune cells resident in the intimal aortic wall. Specifically, the healthy intima is thought to be populated by vascular dendritic cells (DCs) that, during hypercholesterolemia, initiate atherosclerosis by being the first to accumulate cholesterol. Whether these cells remain key players in later stages of disease is unknown. Using murine lineage-tracing models and gene expression profiling, we reveal that myeloid cells present in the intima of the aortic arch are not DCs but instead specialized aortic intima resident macrophages (MacAIR) that depend upon colony-stimulating factor 1 and are sustained by local proliferation. Although MacAIR comprise the earliest foam cells in plaques, their proliferation during plaque progression is limited. After months of hypercholesterolemia, their presence in plaques is overtaken by recruited monocytes, which induce MacAIR-defining genes. These data redefine the lineage of intimal phagocytes and suggest that proliferation is insufficient to sustain generations of macrophages during plaque progression.


Assuntos
Aorta/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Placa Aterosclerótica/imunologia , Túnica Íntima/imunologia , Animais , Diferenciação Celular , Linhagem da Célula , Movimento Celular , Proliferação de Células , Células Cultivadas , Colesterol/metabolismo , Progressão da Doença , Humanos , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Parabiose , Fagocitose
2.
Nat Immunol ; 17(11): 1263-1272, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27668800

RESUMO

Regions of the normal arterial intima predisposed to atherosclerosis are sites of ongoing monocyte trafficking and also contain resident myeloid cells with features of dendritic cells. However, the pathophysiological roles of these cells are poorly understood. Here we found that intimal myeloid cells underwent reverse transendothelial migration (RTM) into the arterial circulation after systemic stimulation of pattern-recognition receptors (PRRs). This process was dependent on expression of the chemokine receptor CCR7 and its ligand CCL19 by intimal myeloid cells. In mice infected with the intracellular pathogen Chlamydia muridarum, blood monocytes disseminated infection to the intima. Subsequent CCL19-CCR7-dependent RTM was critical for the clearance of intimal C. muridarum. This process was inhibited by hypercholesterolemia. Thus, RTM protects the normal arterial intima, and compromised RTM during atherogenesis might contribute to the intracellular retention of pathogens in atherosclerotic lesions.


Assuntos
Quimiocina CCL19/metabolismo , Chlamydia muridarum/imunologia , Células Mieloides/imunologia , Células Mieloides/metabolismo , Receptores CCR7/metabolismo , Migração Transendotelial e Transepitelial , Túnica Íntima/imunologia , Túnica Íntima/metabolismo , Animais , Antígeno CD11c/metabolismo , Infecções por Chlamydia/imunologia , Infecções por Chlamydia/metabolismo , Infecções por Chlamydia/virologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Lipopolissacarídeos/imunologia , Masculino , Camundongos , Camundongos Knockout , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/microbiologia , RNA Mensageiro/genética , Transdução de Sinais , Receptores Toll-Like/metabolismo , Túnica Íntima/microbiologia
3.
Arterioscler Thromb Vasc Biol ; 44(4): 883-897, 2024 04.
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
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.
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 , Animais , Humanos , Apoptose/genética , Movimento Celular/genética , Proliferação de Células/genética , Sobrevivência Celular/genética , Regulação da Expressão Gênica , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Endógeno Competitivo , RNA Longo não Codificante/genética , Túnica Íntima/patologia , Túnica Íntima/metabolismo , Ratos
6.
J Vasc Res ; 61(5): 244-251, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39033742

RESUMO

INTRODUCTION: This study investigated the combination of venous stasis and inflammation in varicose vein development. METHODS: The study included patients with primary varicose veins operated using high ligation and stripping of greater saphenous vein. All of them showed reflux at sapheno-femoral junction on preoperative Doppler ultrasound. Mesenteric veins from early or advanced gastric cancer specimens were used as control group. Inflammatory mediators expressed in the venous wall were measured via immunohistochemistry and compared between the two groups. RESULTS: Thirty-five (59.3%) men and 24 women with a mean age of 52.8 years (range, 23-77 years) were included and 29 (49.2%) patients had edema or skin changes according to Clinical-Etiology-Anatomy-Pathophysiology (CEAP) classification and reporting standards for chronic venous disorders. The expression of interleukin 6 (IL-6) and transforming growth factor ß1 (TGF-ß1) in intima and those of IL-6 in media of greater saphenous veins increased, with statistically significant differences between the two groups (p < 0.001). IL-6 in media and TGF-ß1 levels in intima were independent predictors of varicose veins (adjusted odds ratios 74.62 and 66.69, respectively). CONCLUSION: Elevated venous pressure represented by reflux on Doppler ultrasound and increased expression of inflammatory cytokines including IL-6 in media and TGF-ß1 in intima are associated with the development of varicose veins.


Assuntos
Mediadores da Inflamação , Interleucina-6 , Veia Safena , Fator de Crescimento Transformador beta1 , Túnica Média , Varizes , Humanos , Varizes/diagnóstico por imagem , Varizes/cirurgia , Feminino , Pessoa de Meia-Idade , Masculino , Adulto , Veia Safena/diagnóstico por imagem , Veia Safena/metabolismo , Idoso , Interleucina-6/metabolismo , Adulto Jovem , Mediadores da Inflamação/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Túnica Média/diagnóstico por imagem , Túnica Média/patologia , Túnica Íntima/diagnóstico por imagem , Túnica Íntima/patologia , Inflamação
7.
Strahlenther Onkol ; 200(8): 725-729, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38866999

RESUMO

Pulmonary artery intimal sarcoma (PAIS) is a rare and aggressive malignancy originating from the intimal layer of the pulmonary artery with poor prognosis due to its aggressive nature. The management of PAIS poses both diagnostic and therapeutic challenges. It presents with nonspecific symptoms and is often misdiagnosed as pulmonary embolism. While surgical resection is the primary treatment modality, the role of adjuvant chemotherapy and radiotherapy remains uncertain. However, given the high recurrence rate, adjuvant chemotherapy and/or radiotherapy have been utilized in a limited number of case reports. We present the case of a 46-year-old woman who was diagnosed with PAIS and underwent surgical resection followed by adjuvant chemotherapy (ChT) and radiotherapy (RT), demonstrating good tolerance to this multimodal treatment approach.


Assuntos
Artéria Pulmonar , Sarcoma , Túnica Íntima , Neoplasias Vasculares , Humanos , Feminino , Pessoa de Meia-Idade , Artéria Pulmonar/patologia , Sarcoma/terapia , Sarcoma/patologia , Neoplasias Vasculares/terapia , Neoplasias Vasculares/patologia , Neoplasias Vasculares/diagnóstico por imagem , Terapia Combinada , Túnica Íntima/patologia , Quimioterapia Adjuvante , Radioterapia Adjuvante , Diagnóstico Diferencial
8.
Curr Atheroscler Rep ; 26(8): 353-366, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38822987

RESUMO

PURPOSE OF REVIEW: This review investigates the relationship between myocardial bridges (MBs), intimal thickening in coronary arteries, and Atherosclerotic cardiovascular disease. It focuses on the role of mechanical forces, such as circumferential strain, in arterial wall remodeling and aims to clarify how MBs affect coronary artery pathology. REVIEW FINDINGS: MBs have been identified as influential in modulating coronary artery intimal thickness, demonstrating a protective effect against thickening within the MB segment and an increase in thickness proximal to the MB. This is attributed to changes in mechanical stress and hemodynamics. Research involving arterial hypertension models and vein graft disease has underscored the importance of circumferential strain in vascular remodeling and intimal hyperplasia. Understanding the complex dynamics between MBs, mechanical strain, and vascular remodeling is crucial for advancing our knowledge of coronary artery disease mechanisms. This could lead to improved management strategies for cardiovascular diseases, highlighting the need for further research into MB-related vascular changes.


Assuntos
Ponte Miocárdica , Humanos , Ponte Miocárdica/fisiopatologia , Ponte Miocárdica/complicações , Vasos Coronários/fisiopatologia , Vasos Coronários/patologia , Doença da Artéria Coronariana/fisiopatologia , Túnica Íntima/patologia , Animais , Aterosclerose/fisiopatologia , Remodelação Vascular/fisiologia , Estresse Mecânico
9.
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
10.
Inflamm Res ; 73(6): 929-943, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38642079

RESUMO

OBJECTIVES: Intimal hyperplasia is a serious clinical problem associated with the failure of therapeutic methods in multiple atherosclerosis-related coronary heart diseases, which are initiated and aggravated by the polarization of infiltrating macrophages. The present study aimed to determine the effect and underlying mechanism by which tumor necrosis factor receptor-associated factor 5 (TRAF5) regulates macrophage polarization during intimal hyperplasia. METHODS: TRAF5 expression was detected in mouse carotid arteries subjected to wire injury. Bone marrow-derived macrophages, mouse peritoneal macrophages and human myeloid leukemia mononuclear cells were also used to test the expression of TRAF5 in vitro. Bone marrow-derived macrophages upon to LPS or IL-4 stimulation were performed to examine the effect of TRAF5 on macrophage polarization. TRAF5-knockout mice were used to evaluate the effect of TRAF5 on intimal hyperplasia. RESULTS: TRAF5 expression gradually decreased during neointima formation in carotid arteries in a time-dependent manner. In addition, the results showed that TRAF5 expression was reduced in classically polarized macrophages (M1) subjected to LPS stimulation but was increased in alternatively polarized macrophages (M2) in response to IL-4 administration, and these changes were demonstrated in three different types of macrophages. An in vitro loss-of-function study with TRAF5 knockdown plasmids or TRAF5-knockout mice revealed high expression of markers associated with M1 macrophages and reduced expression of genes related to M2 macrophages. Subsequently, we incubated vascular smooth muscle cells with conditioned medium of polarized macrophages in which TRAF5 expression had been downregulated or ablated, which promoted the proliferation, migration and dedifferentiation of VSMCs. Mechanistically, TRAF5 knockdown inhibited the activation of anti-inflammatory M2 macrophages by directly inhibiting PPARγ expression. More importantly, TRAF5-deficient mice showed significantly aggressive intimal hyperplasia. CONCLUSIONS: Collectively, this evidence reveals an important role of TRAF5 in the development of intimal hyperplasia through the regulation of macrophage polarization, which provides a promising target for arterial restenosis-related disease management.


Assuntos
Hiperplasia , Macrófagos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PPAR gama , Fator 5 Associado a Receptor de TNF , Animais , Macrófagos/metabolismo , Fator 5 Associado a Receptor de TNF/genética , Fator 5 Associado a Receptor de TNF/metabolismo , PPAR gama/metabolismo , PPAR gama/genética , Masculino , Camundongos , Humanos , Artérias Carótidas/patologia , Neointima/patologia , Neointima/metabolismo , Interleucina-4/genética , Células Cultivadas , Túnica Íntima/patologia , Lipopolissacarídeos/farmacologia
11.
Cerebrovasc Dis ; 53(2): 224-232, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39250893

RESUMO

Research on the pathophysiological mechanism of carotid artery dissection and its clinical translation is limited due to the lack of effective animal models to simulate the occurrence of this condition. Assuming that intimal injury is an important factor in the formation of carotid dissection, we established a novel method for inducing carotid dissection models by scraping the carotid intima using a fine needle. Scraping the carotid intima with fine needles can induce the rapid formation of carotid dissection. Magnetic resonance imaging and hematoxylin-eosin staining suggest the presence of false lumens and mural hematomas in the vessels. Our model-induction technique, inspired by iatrogenic catheter-induced artery dissections (carotid, coronary, aortic), significantly mimics the pathological process of clinical carotid dissection. The results suggest that mechanical injury may be a significant cause of carotid dissection and that intimal injury is a major factor in the formation of arterial dissections. This approach will provide assistance in the understanding of medically induced arterial dissection.


Assuntos
Modelos Animais de Doenças , Túnica Íntima , Animais , Túnica Íntima/patologia , Túnica Íntima/lesões , Masculino , Artérias Carótidas/patologia , Artérias Carótidas/diagnóstico por imagem , Dissecação da Artéria Carótida Interna/diagnóstico por imagem , Dissecação da Artéria Carótida Interna/etiologia , Dissecação da Artéria Carótida Interna/patologia , Lesões das Artérias Carótidas/patologia , Lesões das Artérias Carótidas/diagnóstico por imagem , Lesões das Artérias Carótidas/etiologia , Dissecção Aórtica/diagnóstico por imagem , Dissecção Aórtica/patologia , Dissecção Aórtica/etiologia
12.
J Vasc Interv Radiol ; 35(2): 285-292, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37871832

RESUMO

PURPOSE: To determine whether inhibition of the F11 receptor/JAM-A (F11R) using F11R-specific antagonist peptide 4D results in inhibition of smooth muscle cell (SMC) proliferation and migration in vivo, known as neointimal hyperplasia (NIH), using a mouse focal carotid artery stenosis model (FCASM). MATERIALS AND METHODS: The mouse FCASM was chosen to test the hypothesis because the dominant cell type at the site of stenosis is SMC, similar to that in vascular access stenosis. Fourteen C57BL/6 mice underwent left carotid artery (LCA) partial ligation to induce stenosis, followed by daily injection of peptide 4D in 7 mice and saline in the remaining 7 mice, and these mice were observed for 21 days and then euthanized. Bilateral carotid arteries were excised for histologic analysis of the intima and media areas. RESULTS: The mean intimal area was significantly larger in control mice compared with peptide 4D-treated mice (0.031 mm2 [SD ± 0.024] vs 0.0082 mm2 [SD ± 0.0103]; P = .011). The mean intima-to-intima + media area ratio was significantly larger in control mice compared with peptide 4D-treated mice (0.27 [SD ± 0.13] vs 0.089 [SD ± 0.081]; P = .0079). NIH was not observed in the right carotid arteries in both groups. CONCLUSIONS: Peptide 4D, an F11R antagonist, significantly inhibited NIH in C57BL/6 mice in a FCASM.


Assuntos
Estenose das Carótidas , Molécula A de Adesão Juncional , Animais , Camundongos , Hiperplasia/metabolismo , Hiperplasia/patologia , Molécula A de Adesão Juncional/metabolismo , Túnica Íntima/patologia , Modelos Animais de Doenças , Constrição Patológica/patologia , Camundongos Endogâmicos C57BL , Neointima/metabolismo , Neointima/patologia , Artérias Carótidas , Peptídeos/farmacologia , Peptídeos/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 43(3): 456-473, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36700427

RESUMO

BACKGROUND: Late vein graft failure is caused by intimal thickening resulting from endothelial cell (EC) damage and inflammation which promotes vascular smooth muscle cell (VSMC) dedifferentiation, migration, and proliferation. Nonphosphorylatable PRH (proline-rich homeodomain) S163C:S177C offers enhanced stability and sustained antimitotic effect. Therefore, we investigated whether adenovirus-delivered PRH S163C:S177C protein attenuates intimal thickening via VSMC phenotype modification without detrimental effects on ECs. METHODS: PRH S163C:S177C was expressed in vitro (human saphenous vein-VSMCs and human saphenous vein-ECs) and in vivo (ligated mouse carotid arteries) by adenoviruses. Proliferation, migration, and apoptosis were quantified and phenotype was assessed using Western blotting for contractile filament proteins and collagen gel contraction. EC inflammation was quantified using VCAM (vascular cell adhesion protein)-1, ICAM (intercellular adhesion molecule)-1, interleukin-6, and monocyte chemotactic factor-1 measurement and monocyte adhesion. Next Generation Sequencing was utilized to identify novel downstream mediators of PRH action and these and intimal thickening were investigated in vivo. RESULTS: PRH S163C:S177C inhibited proliferation, migration, and apoptosis and promoted contractile phenotype (enhanced contractile filament proteins and collagen gel contraction) compared with virus control in human saphenous vein-VSMCs. PRH S163C:S177C expression in human saphenous vein-ECs significantly reduced apoptosis, without affecting cell proliferation and migration, while reducing TNF (tumor necrosis factor)-α-induced VCAM-1 and ICAM-1 and monocyte adhesion and suppressing interleukin-6 and monocyte chemotactic factor-1 protein levels. PRH S163C:S177C expression in ligated murine carotid arteries significantly impaired carotid artery ligation-induced neointimal proliferation and thickening without reducing endothelial coverage. Next Generation Sequencing revealed STAT-1 (signal transducer and activator of transcription 1) and HDAC-9 (histone deacetylase 9) as mediators of PRH action and was supported by in vitro and in vivo analyses. CONCLUSIONS: We observed PRH S163C:S177C attenuated VSMC proliferation, and migration and enhanced VSMC differentiation at least in part via STAT-1 and HDAC-9 signaling while promoting endothelial repair and anti-inflammatory properties. These findings highlight the potential for PRH S163C:S177C to preserve endothelial function whilst suppressing intimal thickening, and reducing late vein graft failure.


Assuntos
Interleucina-6 , Túnica Íntima , Camundongos , Animais , Humanos , Interleucina-6/metabolismo , Túnica Íntima/patologia , Proliferação de Células , Neointima/patologia , Fatores Quimiotáticos/metabolismo , Fatores Quimiotáticos/farmacologia , Miócitos de Músculo Liso/metabolismo , Movimento Celular
14.
Mol Biol Rep ; 51(1): 568, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38656400

RESUMO

BACKGROUND: Tumor embolism is a very rare primary manifestation of cancers and the diagnosis is challenging, especially if located in the pulmonary arteries, where it can mimic nonmalignant pulmonary embolism. Intimal sarcoma is one of the least commonly reported primary tumors of vessels with only a few cases reported worldwide. A typical location of this malignancy is the pulmonary artery. Herein, we present a case report of an intimal sarcoma with primary manifestation in the pulmonary arteries. A 53-year-old male initially presented with dyspnea. On imaging, a pulmonary artery embolism was detected and was followed by thrombectomy of the right ventricular outflow tract, main pulmonary artery trunk, and right pulmonary artery after ineffective lysis therapy. Complementary imaging of the chest and abdomen including a PET-CT scan demonstrated no evidence of a primary tumor. Subsequent pathology assessment suggested an intimal sarcoma further confirmed by DNA methylation based molecular analysis. We initiated adjuvant chemotherapy with doxorubicin. Four months after the completion of adjuvant therapy a follow-up scan revealed a local recurrence without distant metastases. DISCUSSION: Primary pulmonary artery intimal sarcoma (PAS) is an exceedingly rare entity and pathological diagnosis remains challenging. Therefore, the detection of entity-specific molecular alterations is a supporting argument in the diagnostic spectrum. Complete surgical resection is the prognostically most important treatment for intimal cardiac sarcomas. Despite adjuvant chemotherapy, the prognosis of cardiac sarcomas remains very poor. This case of a PAS highlights the difficulty in establishing a diagnosis and the aggressive natural course of the disease. CONCLUSION: In case of atypical presentation of a pulmonary embolism, a tumor originating from the great vessels should be considered. Molecular pathology techniques support in establishing a reliable diagnosis.


Assuntos
Artéria Pulmonar , Sarcoma , Trombose , Humanos , Masculino , Pessoa de Meia-Idade , Artéria Pulmonar/patologia , Sarcoma/diagnóstico , Sarcoma/patologia , Túnica Íntima/patologia , Neoplasias Vasculares/diagnóstico , Neoplasias Vasculares/patologia , Embolia Pulmonar/diagnóstico , Diagnóstico Diferencial
15.
BMC Cardiovasc Disord ; 24(1): 34, 2024 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-38184530

RESUMO

Coronary heavy calcification (HC) poses a sturdy challenge to percutaneous coronary intervention (PCI). Scores considering calcification length, thickness, or circumferential extent, are widely accepted to dictate upfront calcium modification to improve PCI outcomes. Although often marginalized, calcification shape (morphology) may require consideration during procedure planning in selected cases. This case demonstrates how a focal but spur-shaped calcification led to a massive proximal left anterior descending (LAD) dissecting intramural hematoma.


Assuntos
Calcinose , Intervenção Coronária Percutânea , Humanos , Cálcio , Intervenção Coronária Percutânea/efeitos adversos , Túnica Íntima , Vasos Coronários/diagnóstico por imagem , Vasos Coronários/cirurgia , Calcinose/diagnóstico por imagem , Calcinose/etiologia , Calcinose/cirurgia , Hematoma/diagnóstico por imagem , Hematoma/etiologia , Hematoma/terapia
16.
Ann Vasc Surg ; 101: 29-40, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38128692

RESUMO

BACKGROUND: To compare the 30-day and long-term outcomes between patients with concomitant type B intramural hematoma and intimal disruption upon admission who underwent endovascular repair in the acute or subacute phases. METHODS: Data were extracted from January 1, 2010, to December 31, 2019. Logistic regression and Cox regression were performed to evaluate the impact of timing of intervention on 30-day and long-term outcomes, respectively. RESULTS: The study included 241 patients, among which 159 were in the acute group. No significant difference was observed in 30-day mortality (0.6% vs. 0%, P = 1), 30-day complication rate (2.5% vs. 1.2%, P = 0.664), long-term all-cause mortality (10.7% vs. 7.3%, P = 0.540), and aortic reintervention rate (2.5% vs. 2.4%, P = 1) between the acute and subacute group. In multivariable analysis, the timing of intervention was not associated with 30-day mortality (odds ratio (OR) = 0, 95% confidence interval CI: 0-Inf, P = 0.999), 30-day complication (OR = 0.30, 95% CI: 0.02-3.77, P = 0.348), long-term mortality (hazard ratio = 0.56, 95% CI: 0.20-1.61, P = 0.283), and aortic reintervention (OR = 0.97, 95% CI: 0.15-6.08, P = 0.970). CONCLUSIONS: For patients with concomitant type B intramural hematoma and intimal disruption upon admission, there is no significant difference in 30-day and long-term outcomes between those who undergo endovascular treatment in the acute or subacute phase.


Assuntos
Aneurisma da Aorta Torácica , Doenças da Aorta , Implante de Prótese Vascular , Procedimentos Endovasculares , Humanos , Resultado do Tratamento , Hematoma Intramural Aórtico , Doenças da Aorta/complicações , Doenças da Aorta/diagnóstico por imagem , Doenças da Aorta/cirurgia , Túnica Íntima , Estudos Retrospectivos , Procedimentos Endovasculares/efeitos adversos , Hematoma/diagnóstico por imagem , Hematoma/etiologia , Hematoma/cirurgia , Fatores de Risco , Implante de Prótese Vascular/efeitos adversos , Aneurisma da Aorta Torácica/cirurgia
17.
Nanomedicine ; 61: 102763, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38897395

RESUMO

The primary challenge in percutaneous coronary interventions for vascular restenosis is the occurrence of restenosis, which is defined by the excessive proliferation of neointimal tissue. Herein, our research team suggests that exosomes obtained from PSC, when paired with quercetin (Q@PSC-E), successfully reduce neointimal hyperplasia in a Sprague-Dawley rat model. Furthermore, the physical properties of the synthesized Q@PSC-E were examined using UV-vis, DLS, and FT-IR characterization techniques. The rats were subjected to balloon injury (BI) utilizing a 2-Fr Fogarty arterial embolectomy balloon catheter. Intimal hyperplasia and the degree of VSMC proliferation were evaluated using histological analysis in the rat groups that received a dosage of Q@PSC-E at 30 mg/kg/d. Significantly, Q@PSC-E inhibited cell proliferation through a pathway that does not include lipoxygenase, as demonstrated by [3H] thymidine incorporation, MTT, and flow cytometry studies. Additionally, the data indicate that Q@PSC-E hinders cell proliferation by targeting particular events that promote cell growth, including the activation of Akt and NF-κB, disruption of cell-cycle progression and also obstructs the ERK signaling pathway.


Assuntos
Proliferação de Células , Exossomos , Hiperplasia , Proteínas Proto-Oncogênicas c-akt , Quercetina , Transdução de Sinais , Animais , Masculino , Ratos , Lesões das Artérias Carótidas/patologia , Lesões das Artérias Carótidas/tratamento farmacológico , Lesões das Artérias Carótidas/metabolismo , Proliferação de Células/efeitos dos fármacos , Exossomos/metabolismo , Exossomos/efeitos dos fármacos , Hiperplasia/patologia , Hiperplasia/tratamento farmacológico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quercetina/farmacologia , Quercetina/química , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Túnica Íntima/patologia , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/metabolismo
18.
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
19.
Morphologie ; 108(362): 100779, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38608628

RESUMO

BACKGROUND: One of the most recent hormones to be identified and isolated is irisin, extracted from mouse skeletal muscle in 2012. Irisin has been proven to alter blood pressure, which has an impact on blood vessels, enhance endothelial functions, and prevent injury to endothelial cells. The current study aimed to study the effect of irisin on the ultrastructure of the rat thoracic aorta using the transmission electron microscope (TEM). MATERIALS AND METHODS: Twenty female rats were recruited for this study and divided into a control group (non-injected), and four experimental groups (injected groups) each consisting of 4 rats. The experimental groups were injected intraperitoneally with different doses of irisin (250ng/mL, 500ng/mL, 1000ng/mL, and 2000ng/mL) twice a week for 4weeks. Then, the descending thoracic aorta of all experimental rats were resected and proceeded with imaging. RESULTS: The results of this study showed a change in the thickness of the tunica intima, internal elastic lamina, elastic lamellae, and external elastic lamina concerning increasing injected irisin concentration. While there was a significant increase in the thickness of tunica media (P<0.0001) and smooth muscle cells (P<0.05). Also, the results showed a significant increase in the number of elastic lamellae in the tunica media (P<0.0001). CONCLUSION: Irisin had a major impact on the elasticity of the rat thoracic aorta wall, suggesting that it influences the growth factors of the wall and activates smooth muscle cells in addition to endothelial cells.


Assuntos
Aorta Torácica , Fibronectinas , Microscopia Eletrônica de Transmissão , Animais , Fibronectinas/farmacologia , Aorta Torácica/efeitos dos fármacos , Aorta Torácica/ultraestrutura , Ratos , Feminino , Túnica Íntima/ultraestrutura , Túnica Íntima/efeitos dos fármacos , Ratos Sprague-Dawley , Túnica Média/efeitos dos fármacos , Túnica Média/ultraestrutura
20.
Circ Res ; 128(4): 530-543, 2021 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-33397122

RESUMO

RATIONALE: Bone marrow transplantation (BMT) is used frequently to study the role of hematopoietic cells in atherosclerosis, but aortic arch lesions are smaller in mice after BMT. OBJECTIVE: To identify the earliest stage of atherosclerosis inhibited by BMT and elucidate potential mechanisms. METHODS AND RESULTS: Ldlr-/- mice underwent total body γ-irradiation, bone marrow reconstitution, and 6-week recovery. Atherosclerosis was studied in the ascending aortic arch and compared with mice without BMT. In BMT mice, neutral lipid and myeloid cell topography were lower in lesions after feeding a cholesterol-rich diet for 3, 6, and 12 weeks. Lesion coalescence and height were suppressed dramatically in mice post-BMT, whereas lateral growth was inhibited minimally. Targeted radiation to the upper thorax alone reproduced the BMT phenotype. Classical monocyte recruitment, intimal myeloid cell proliferation, and apoptosis did not account for the post-BMT phenotype. Neutral lipid accumulation was reduced in 5-day lesions, thus we developed quantitative assays for LDL (low-density lipoprotein) accumulation and paracellular leakage using DiI-labeled human LDL and rhodamine B-labeled 70 kD dextran. LDL accumulation was dramatically higher in the intima of Ldlr-/- relative to Ldlr+/+ mice, and was inhibited by injection of HDL mimics, suggesting a regulated process. LDL, but not dextran, accumulation was lower in mice post-BMT both at baseline and in 5-day lesions. Since the transcript abundance of molecules implicated in LDL transcytosis was not significantly different in the post-BMT intima, transcriptomics from whole aortic arch intima, and at single-cell resolution, was performed to give insights into pathways modulated by BMT. CONCLUSIONS: Radiation exposure inhibits LDL entry into the aortic intima at baseline and the earliest stages of atherosclerosis. Single-cell transcriptomic analysis suggests that LDL uptake by endothelial cells is diverted to lysosomal degradation and reverse cholesterol transport pathways. This reduces intimal accumulation of lipid and impacts lesion initiation and growth.


Assuntos
Aterosclerose/metabolismo , Raios gama , Lipoproteínas LDL/metabolismo , Túnica Íntima/efeitos da radiação , Animais , Aorta/metabolismo , Aorta/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Receptores de LDL/deficiência , Receptores de LDL/genética , Transcriptoma , Túnica Íntima/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA