Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 43(9): 1639-1652, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37409527

RESUMO

BACKGROUND: Treatment of occluded vessels can involve angioplasty, stenting, and bypass grafting, which can be limited by restenosis and thrombosis. Drug-eluting stents attenuate restenosis, but the current drugs used are cytotoxic, causing smooth muscle cell (SMC) and endothelial cell (EC) death that may lead to late thrombosis. N-cadherin is a junctional protein expressed by SMCs, which promotes directional SMC migration contributing to restenosis. We propose that engaging N-cadherin with mimetic peptides can act as a cell type-selective therapeutic strategy to inhibit polarization and directional migration of SMCs without negatively impacting ECs. METHODS: We designed a novel N-cadherin-targeting chimeric peptide with a histidine-alanine-valine cadherin-binding motif, combined with a fibronectin-binding motif from Staphylococcus aureus. This peptide was tested in SMC and EC culture assays of migration, viability, and apoptosis. Rat carotid arteries were balloon injured and treated with the N-cadherin peptide. RESULTS: Treating scratch-wounded SMCs with the N-cadherin-targeting peptide inhibited migration and reduced polarization of wound-edge cells. The peptide colocalized with fibronectin. Importantly, EC junction, permeability, or migration was not impacted by peptide treatment in vitro. We also demonstrated that the chimeric peptide persisted for 24 hours after transient delivery in the balloon-injured rat carotid artery. Treatment with the N-cadherin-targeting chimeric peptide reduced intimal thickening in balloon-injured rat carotid arteries at 1 and 2 weeks after injury. Reendothelialization of injured vessels after 2 weeks was unimpaired by peptide treatment. CONCLUSIONS: These studies show that an N-cadherin-binding and fibronectin-binding chimeric peptide is effective in inhibiting SMC migration in vitro and in vivo and limiting neointimal hyperplasia after balloon angioplasty without affecting EC repair. These results establish the potential of an advantageous SMC-selective strategy for antirestenosis therapy.


Assuntos
Lesões das Artérias Carótidas , Trombose , Ratos , Animais , Fibronectinas/farmacologia , Lesões das Artérias Carótidas/patologia , Caderinas , Artérias Carótidas/patologia , Hiperplasia/patologia , Peptídeos/farmacologia , Trombose/patologia
2.
Arterioscler Thromb Vasc Biol ; 43(7): 1096-1110, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37226729

RESUMO

Despite recent advancements in vascular disease treatments, thrombosis and poor long-term vessel patency remain significant barriers to effective endovascular intervention. Current balloon angioplasty and stenting techniques effectively restore acute blood flow in occluded vessels but have persistent limitations. Damage to the arterial endothelium caused by injury during catheter tracking triggers neointimal hyperplasia and the release of proinflammatory factors leading to increased risk of thrombosis and restenosis. Antirestenotic agents commonly delivered on angioplasty balloons and stents have lowered arterial restenosis rates, but the absence of cell type selectivity significantly delays critical endothelium repair. Targeted delivery of biomolecular therapeutics, coupled with engineered nanoscale excipients, has the potential to redefine cardiovascular interventions by improving long-term efficacy, limiting off-target effects, and reducing costs compared with conventional clinical standards of care. This review analyzes current forms of localized vascular drug delivery, emerging nanoscale therapeutic and excipient strategies, and provides recommendations for future areas of study to advance the treatment of vascular disease through innovations in nanotechnology.


Assuntos
Angioplastia com Balão , Trombose , Doenças Vasculares , Humanos , Angioplastia com Balão/efeitos adversos , Angioplastia com Balão/métodos , Stents , Constrição Patológica/etiologia , Doenças Vasculares/etiologia , Trombose/etiologia , Nanotecnologia , Resultado do Tratamento
3.
Am J Physiol Heart Circ Physiol ; 324(4): H391-H410, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36607797

RESUMO

This study reports a new methodology for right heart imaging by ultrasound in mice under right ventricular (RV) pressure overload. Pulmonary artery constriction (PAC) or sham surgeries were performed on C57BL/6 male mice at 8 wk of age. Ultrasound imaging was conducted at 2, 4, and 8 wk postsurgery using both classical and advanced ultrasound imaging modalities including electrocardiogram (ECG)-based kilohertz visualization, anatomical M-mode, and strain imaging. Based on pulsed Doppler, the PAC group demonstrated dramatically enhanced pressure gradient in the main pulmonary artery (MPA) as compared with the sham group. By the application of advanced imaging modalities in novel short-axis views of the ventricles, the PAC group demonstrated increased thickness of RV free wall, enlarged RV chamber, and reduced RV fractional shortening compared with the sham group. The PAC group also showed prolonged RV contraction, asynchronous interplay between RV and left ventricle (LV), and passive leftward motion of the interventricular septum (IVS) at early diastole. Consequently, the PAC group exhibited prolongation of LV isovolumic relaxation time, without change in LV wall thickness or systolic function. Significant correlations were found between the maximal pressure gradient in MPA measured by Doppler and the RV systolic pressure by catheterization, as well as the morphological and functional parameters of RV by ultrasound.NEW & NOTEWORTHY The established protocol overcomes the challenges in right heart imaging in mice, thoroughly elucidating the changes of RV, the dynamics of IVS, and the impact on LV and provides new insights into the pathophysiological mechanism of RV remodeling.


Assuntos
Disfunção Ventricular Direita , Remodelação Ventricular , Masculino , Animais , Camundongos , Camundongos Endogâmicos C57BL , Coração , Ventrículos do Coração/diagnóstico por imagem , Ultrassonografia , Disfunção Ventricular Direita/diagnóstico por imagem , Disfunção Ventricular Direita/etiologia , Pressão Ventricular/fisiologia , Função Ventricular Direita
4.
Am J Respir Cell Mol Biol ; 67(5): 562-573, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35926106

RESUMO

Pulmonary hypertension (PH) is a multifaceted condition characterized by elevated pulmonary arterial pressure, which can result in right ventricular dysfunction and failure. Disorders of lung development can present with secondary PH, which is a leading cause of mortality in infants with bronchopulmonary dysplasia (BPD). DDR1 (discoidin domain receptor 1) is a collagen-binding receptor that regulates tissue fibrosis and inflammation and controls cellular growth and migration. However, the roles of DDR1 in lung development or the pathogenesis of PH are unknown. Studying mice with a DDR1 deletion (Ddr1-/-), we have noted 35% mortality between 1 and 4 months of age, and we demonstrate that DDR1 deficiency results in reduced right ventricular contractility and muscularization of distal pulmonary arteries, consistent with PH. Pathology analysis revealed enlarged alveolar spaces in Ddr1-/- mice by Postnatal Day 7, consistent with impaired alveolar development. Gene expression analysis showed that Ddr1-/- mice have reduced concentrations of alveologenesis factors and epithelial-to-mesenchymal transition markers. Mechanistic studies in vitro confirmed that DDR1 mediated epithelial-to-mesenchymal transition, migration, and growth of alveolar epithelial cells. Taken together, these data suggest that DDR1 plays important roles mediating alveolarization during lung development. Our studies also describe a new model of spontaneous PH and bronchopulmonary dysplasia in mice.


Assuntos
Displasia Broncopulmonar , Receptor com Domínio Discoidina 1 , Hipertensão Pulmonar , Animais , Humanos , Recém-Nascido , Camundongos , Receptor com Domínio Discoidina 1/genética , Receptor com Domínio Discoidina 1/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Fibrose
5.
Arterioscler Thromb Vasc Biol ; 40(7): 1763-1776, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32493168

RESUMO

OBJECTIVE: Vascular calcification is a pathology characterized by arterial mineralization, which is a common late-term complication of atherosclerosis that independently increases the risk of adverse cardiovascular events by fourfold. A major source of calcifying cells is transdifferentiating vascular smooth muscle cells (VSMCs). Previous studies showed that deletion of the collagen-binding receptor, DDR1 (discoidin domain receptor-1), attenuated VSMC calcification. Increased matrix stiffness drives osteogenesis, and DDR1 has been implicated in stiffness sensing in other cell types; however, the role of DDR1 as a mechanosensor in VSMCs has not been investigated. Here, we test the hypothesis that DDR1 senses increased matrix stiffness and promotes VSMC transdifferentiation and calcification. Approach and Results: Primary VSMCs isolated from Ddr1+/+ (wild-type) and Ddr1-/- (knockout) mice were studied on collagen-I-coated silicon substrates of varying stiffness, culturing in normal or calcifying medium. DDR1 expression and phosphorylation increased with increasing stiffness, as did in vitro calcification, nuclear localization of Runx2 (Runt-related transcription factor 2), and expression of other osteochondrocytic markers. By contrast, DDR1 deficient VSMCs were not responsive to stiffness and did not undergo transdifferentiation. DDR1 regulated stress fiber formation and RhoA (ras homolog family member A) activation through the RhoGEF (rho guanine nucleotide exchange factor), Vav2. Inhibition of actomyosin contractility reduced Runx2 activation and attenuated in vitro calcification in wild-type VSMCs. Finally, a novel positive feedforward loop was uncovered between DDR1 and actomyosin contractility, important in regulating DDR1 expression, clustering, and activation. CONCLUSIONS: This study provides mechanistic insights into DDR1 mechanosignaling and shows that DDR1 activity and actomyosin contractility are interdependent in mediating stiffness-dependent increases in VSMC calcification.


Assuntos
Aterosclerose/enzimologia , Transdiferenciação Celular , Receptor com Domínio Discoidina 1/metabolismo , Matriz Extracelular/enzimologia , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Osteogênese , Calcificação Vascular/enzimologia , Proteína rhoA de Ligação ao GTP/metabolismo , Actomiosina/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/patologia , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Receptor com Domínio Discoidina 1/deficiência , Receptor com Domínio Discoidina 1/genética , Modelos Animais de Doenças , Matriz Extracelular/patologia , Mecanotransdução Celular , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Proto-Oncogênicas c-vav/metabolismo , Calcificação Vascular/genética , Calcificação Vascular/patologia
8.
J Biol Chem ; 293(8): 2841-2849, 2018 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-29298894

RESUMO

Discoidin domain receptor 1 (DDR1) is a collagen receptor that mediates cell communication with the extracellular matrix (ECM). Aberrant expression and activity of DDR1 in tumor cells are known to promote tumor growth. Although elevated DDR1 levels in the stroma of breast tumors are associated with poor patient outcome, a causal role for tumor-extrinsic DDR1 in cancer promotion remains unclear. Here we report that murine mammary tumor cells transplanted to syngeneic recipient mice in which Ddr1 has been knocked out (KO) grow less robustly than in WT mice. We also found that the tumor-associated stroma in Ddr1-KO mice exhibits reduced collagen deposition compared with the WT controls, supporting a role for stromal DDR1 in ECM remodeling of the tumor microenvironment. Furthermore, the stromal-vascular fraction (SVF) of Ddr1 knockout adipose tissue, which contains committed adipose stem/progenitor cells and preadipocytes, was impaired in its ability to stimulate tumor cell migration and invasion. Cytokine array-based screening identified interleukin 6 (IL-6) as a cytokine secreted by the SVF in a DDR1-dependent manner. SVF-produced IL-6 is important for SVF-stimulated tumor cell invasion in vitro, and, using antibody-based neutralization, we show that tumor promotion by IL-6 in vivo requires DDR1. In conclusion, our work demonstrates a previously unrecognized function of DDR1 in promoting tumor growth.


Assuntos
Tecido Adiposo/metabolismo , Neoplasias da Mama/metabolismo , Receptor com Domínio Discoidina 1/metabolismo , Interleucina-6/metabolismo , Células Estromais/metabolismo , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/imunologia , Tecido Adiposo/patologia , Animais , Anticorpos Neutralizantes/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colágeno/metabolismo , Receptor com Domínio Discoidina 1/genética , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Invasividade Neoplásica/imunologia , Invasividade Neoplásica/patologia , Invasividade Neoplásica/prevenção & controle , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias , Células Estromais/efeitos dos fármacos , Células Estromais/imunologia , Células Estromais/patologia , Transplante Isogênico , Carga Tumoral/efeitos dos fármacos , Células Tumorais Cultivadas , Microambiente Tumoral/efeitos dos fármacos
9.
Arterioscler Thromb Vasc Biol ; 38(8): 1878-1889, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29930002

RESUMO

Objective- Vascular calcification is a common and severe complication in patients with atherosclerosis which is exacerbated by type 2 diabetes mellitus. Our laboratory recently reported that the collagen receptor discoidin domain receptor 1 (DDR1) mediates vascular calcification in atherosclerosis; however, the underlying mechanisms are unknown. During calcification, vascular smooth muscle cells transdifferentiate into osteoblast-like cells, in a process driven by the transcription factor RUNX2 (runt-related transcription factor 2). DDR1 signals via the phosphoinositide 3-kinase/Akt pathway, which is also central to insulin signaling, and upstream of RUNX2, and this led us to investigate whether DDR1 promotes vascular calcification in diabetes mellitus via this pathway. Approach and Results- Ddr1+/+ ; Ldlr-/- (single knock-out) and Ddr1-/- ; Ldlr-/- (double knock-out) mice were placed on high-fat diet for 12 weeks to induce atherosclerosis and type 2 diabetes mellitus. Von Kossa staining revealed reduced vascular calcification in the aortic arch of double knock-out compared with single knock-out mice. Immunofluorescent staining for RUNX2 was present in calcified plaques of single knock-out but not double knock-out mice. Primary vascular smooth muscle cells obtained from Ddr1+/+ and Ddr1-/- mice were cultured in calcifying media. DDR1 deletion resulted in reduced calcification, a 74% reduction in p-Akt levels, and an 88% reduction in RUNX2 activity. Subcellular fractionation revealed a 77% reduction in nuclear RUNX2 levels in Ddr1-/- vascular smooth muscle cells. DDR1 associated with phosphoinositide 3-kinase, and treatment with the inhibitor wortmannin attenuated calcification. Finally, we show that DDR1 is important to maintain the microtubule cytoskeleton which is required for the nuclear localization of RUNX2. Conclusions- These novel findings demonstrate that DDR1 promotes RUNX2 activity and atherosclerotic vascular calcification in diabetes mellitus via phosphoinositide 3-kinase/Akt signaling.


Assuntos
Aterosclerose/enzimologia , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Diabetes Mellitus Tipo 2/enzimologia , Angiopatias Diabéticas/enzimologia , Receptor com Domínio Discoidina 1/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Calcificação Vascular/enzimologia , Transporte Ativo do Núcleo Celular , Animais , Aterosclerose/genética , Aterosclerose/patologia , Células Cultivadas , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Angiopatias Diabéticas/genética , Angiopatias Diabéticas/patologia , Dieta Hiperlipídica , Receptor com Domínio Discoidina 1/deficiência , Receptor com Domínio Discoidina 1/genética , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Fosforilação , Receptores de LDL/deficiência , Receptores de LDL/genética , Transdução de Sinais , Calcificação Vascular/genética , Calcificação Vascular/patologia
10.
Am J Physiol Heart Circ Physiol ; 312(5): H943-H958, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28283548

RESUMO

Atherosclerotic plaque rupture with subsequent embolic events is a major cause of sudden death from myocardial infarction or stroke. Although smooth muscle cells (SMCs) produce and respond to collagens in vitro, there is no direct evidence in vivo that SMCs are a crucial source of collagens and that this impacts lesion development or fibrous cap formation. We sought to determine how conditional SMC-specific knockout of collagen type XV (COL15A1) in SMC lineage tracing mice affects advanced lesion formation given that 1) we have previously identified a Col15a1 sequence variant associated with age-related atherosclerosis, 2) COL15A1 is a matrix organizer enhancing tissue structural integrity, and 3) small interfering RNA-mediated Col15a1 knockdown increased migration and decreased proliferation of cultured human SMCs. We hypothesized that SMC-derived COL15A1 is critical in advanced lesions, specifically in fibrous cap formation. Surprisingly, we demonstrated that SMC-specific Col15a1 knockout mice fed a Western diet for 18 wk failed to form advanced lesions. SMC-specific Col15a1 knockout resulted in lesions reduced in size by 78%, with marked reductions in numbers and proliferating SMCs, and lacked a SMC and extracellular matrix-rich lesion or fibrous cap. In vivo RNA-seq analyses on SMC Col15a1 knockout and wild-type lesions suggested that a mechanism for these effects is through global repression of multiple proatherogenic inflammatory pathways involved in lesion development. These results provide the first direct evidence that a SMC-derived collagen, COL15A1, is critical during lesion pathogenesis, but, contrary to expectations, its loss resulted in marked attenuation rather than exacerbation of lesion pathogenesis.NEW & NOTEWORTHY We report the first direct in vivo evidence that a smooth muscle cell (SMC)-produced collagen, collagen type XV (COL15A1), is critical for atherosclerotic lesion development. SMC Col15a1 knockout markedly attenuated advanced lesion formation, likely through reducing SMC proliferation and impairing multiple proatherogenic inflammatory processes.


Assuntos
Aterosclerose/genética , Aterosclerose/patologia , Colágeno/genética , Miócitos de Músculo Liso/patologia , Envelhecimento/patologia , Animais , Aorta/citologia , Linhagem da Célula , Dieta Aterogênica , Feminino , Técnicas de Silenciamento de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miografia , Rigidez Vascular
11.
Arterioscler Thromb Vasc Biol ; 36(3): 525-33, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26800565

RESUMO

OBJECTIVE: Collagen accumulation and calcification are major determinants of atherosclerotic plaque stability. Extracellular vesicle (EV)-derived microcalcifications in the collagen-poor fibrous cap may promote plaque rupture. In this study, we hypothesize that the collagen receptor discoidin domain receptor-1 (DDR-1) regulates collagen deposition and release of calcifying EVs by vascular smooth muscle cells (SMCs) through the transforming growth factor-ß (TGF-ß) pathway. APPROACH AND RESULTS: SMCs from the carotid arteries of DDR-1(-/-) mice and wild-type littermates (n=5-10 per group) were cultured in normal or calcifying media. At days 14 and 21, SMCs were harvested and EVs isolated for analysis. Compared with wild-type, DDR-1(-/-) SMCs exhibited a 4-fold increase in EV release (P<0.001) with concomitantly elevated alkaline phosphatase activity (P<0.0001) as a hallmark of EV calcifying potential. The DDR-1(-/-) phenotype was characterized by increased mineralization (Alizarin Red S and Osteosense, P<0.001 and P=0.002, respectively) and amorphous collagen deposition (P<0.001). We further identified a novel link between DDR-1 and the TGF-ß pathway previously implicated in both fibrotic and calcific responses. An increase in TGF-ß1 release by DDR-1(-/-) SMCs in calcifying media (P<0.001) stimulated p38 phosphorylation (P=0.02) and suppressed activation of Smad3. Inhibition of either TGF-ß receptor-I or phospho-p38 reversed the fibrocalcific DDR-1(-/-) phenotype, corroborating a causal relationship between DDR-1 and TGF-ß in EV-mediated vascular calcification. CONCLUSIONS: DDR-1 interacts with the TGF-ß pathway to restrict calcifying EV-mediated mineralization and fibrosis by SMCs. We therefore establish a novel mechanism of cell-matrix homeostasis in atherosclerotic plaque formation.


Assuntos
Aterosclerose/metabolismo , Colágeno/metabolismo , Vesículas Extracelulares/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Calcificação Vascular/metabolismo , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Células Cultivadas , Receptor com Domínio Discoidina 1 , Modelos Animais de Doenças , Feminino , Fibrose , Predisposição Genética para Doença , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Osteogênese , Fenótipo , Fosforilação , Placa Aterosclerótica , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Proteína Tirosina Quinases/deficiência , Receptores Proteína Tirosina Quinases/genética , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteína Smad3/metabolismo , Fatores de Tempo , Calcificação Vascular/genética , Calcificação Vascular/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Am J Physiol Endocrinol Metab ; 311(2): E335-45, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27221119

RESUMO

It has been argued whether insulin accelerates or prevents atherosclerosis. Although results from in vitro studies have been conflicting, recent in vivo mice studies demonstrated antiatherogenic effects of insulin. Insulin is a known activator of endothelial nitric oxide synthase (NOS), leading to increased production of NO, which has potent antiatherogenic effects. We aimed to examine the role of NOS in the protective effects of insulin against atherosclerosis. Male apolipoprotein E-null mice (8 wk old) fed a high-cholesterol diet (1.25% cholesterol) were assigned to the following 12-wk treatments: control, insulin (0.05 U/day via subcutaneous pellet), N(ω)-nitro-l-arginine methyl ester hydrochloride (l-NAME, via drinking water at 100 mg/l), and insulin plus l-NAME. Insulin reduced atherosclerotic plaque burden in the descending aorta by 42% compared with control (plaque area/aorta lumen area: control, 16.5 ± 1.9%; insulin, 9.6 ± 1.3%, P < 0.05). Although insulin did not decrease plaque burden in the aortic sinus, macrophage accumulation in the plaque was decreased by insulin. Furthermore, insulin increased smooth muscle actin and collagen content and decreased plaque necrosis, consistent with increased plaque stability. In addition, insulin treatment increased plasma NO levels, decreased inducible NOS staining, and tended to increase phosphorylated vasodilator-stimulated phosphoprotein staining in the plaques of the aortic sinus. All these effects of insulin were abolished by coadministration of l-NAME, whereas l-NAME alone showed no effect. Insulin also tended to increase phosphorylated endothelial NOS and total neuronal NOS staining, effects not modified by l-NAME. In conclusion, we demonstrate that insulin treatment decreases atherosclerotic plaque burden and increases plaque stability through NOS-dependent mechanisms.


Assuntos
Aorta/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Hipoglicemiantes/farmacologia , Insulina/farmacologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/efeitos dos fármacos , Placa Aterosclerótica/metabolismo , Actinas/efeitos dos fármacos , Actinas/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , Apolipoproteínas E/genética , Colágeno/efeitos dos fármacos , Colágeno/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Necrose , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo I/efeitos dos fármacos , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico Sintase Tipo III/efeitos dos fármacos , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfoproteínas/efeitos dos fármacos , Fosfoproteínas/metabolismo , Placa Aterosclerótica/patologia , Seio Aórtico/efeitos dos fármacos , Seio Aórtico/metabolismo , Seio Aórtico/patologia
13.
Am J Pathol ; 182(3): 628-39, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23260773

RESUMO

The rapid proliferation of smooth muscle cells (SMCs) contributes to atherosclerotic plaque formation and neointimal thickening in other occlusive vascular diseases. In cancer cells, rapid cell proliferation is often accompanied by DNA damage, division aberrations, elevated cell apoptosis, or accumulation of abnormal cells. However, little is known about division fidelity in vascular disorders. We have analyzed the cell division fidelity during the rapid SMC proliferation that occurs after balloon injury of the rat carotid artery using en face confocal microscopy of the full thickness of the vessel wall. SMCs newly migrated to the neointima had increased division defects and increased apoptosis compared with SMCs in the subjacent media, despite comparable mitosis rates. Protein kinase Cα and the receptor for hyaluronic acid-mediated motility (RHAMM) regulate division fidelity in cultured neointimal SMCs. The centrosomal targeting sequence of RHAMM was required for localization to the mitotic spindle and spindle organization. Dynein and RHAMM colocalized in the spindle area and were part of a complex. Dynein inhibition caused spindle defects similar to RHAMM or protein kinase C inhibition. Our study uncovered abnormalities in rapidly proliferating SMCs after arterial injury that could contribute to the growth of atherosclerotic plaques and reduce plaque stability by triggering apoptosis, and it described a mechanism by which RHAMM and dynein coordinate division fidelity in neointimal SMCs.


Assuntos
Aterosclerose/patologia , Artérias Carótidas/patologia , Divisão Celular , Progressão da Doença , Animais , Apoptose , Artérias Carótidas/metabolismo , Divisão do Núcleo Celular , Proliferação de Células , Centrossomo/metabolismo , Segregação de Cromossomos , Citocinese , Dineínas/metabolismo , Proteínas da Matriz Extracelular/química , Proteínas da Matriz Extracelular/metabolismo , Receptores de Hialuronatos/química , Receptores de Hialuronatos/metabolismo , Masculino , Índice Mitótico , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/patologia , Neointima/metabolismo , Neointima/patologia , Ligação Proteica , Proteína Quinase C-alfa/antagonistas & inibidores , Proteína Quinase C-alfa/metabolismo , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Fuso Acromático/metabolismo , Túnica Média/metabolismo , Túnica Média/patologia
14.
Am J Pathol ; 182(6): 2241-53, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23567639

RESUMO

Collagens in the atherosclerotic plaque signal regulation of cell behavior and provide tensile strength to the fibrous cap. Type VIII collagen, a short-chain collagen, is up-regulated in atherosclerosis; however, little is known about its functions in vivo. We studied the response to arterial injury and the development of atherosclerosis in type VIII collagen knockout mice (Col8(-/-) mice). After wire injury of the femoral artery, Col8(-/-) mice had decreased vessel wall thickening and outward remodeling when compared with Col8(+/+) mice. We discovered that apolipoprotein E (ApoE) is an endogenous repressor of the Col8a1 chain, and, therefore, in ApoE knockout mice, type VIII collagen was up-regulated. Deficiency of type VIII collagen in ApoE(-/-) mice (Col8(-/-);ApoE(-/-)) resulted in development of plaques with thin fibrous caps because of decreased smooth muscle cell migration and proliferation and reduced accumulation of fibrillar type I collagen. In contrast, macrophage accumulation was not affected, and the plaques had large lipid-rich necrotic cores. We conclude that in atherosclerosis, type VIII collagen is up-regulated in the absence of ApoE and functions to increase smooth muscle cell proliferation and migration. This is an important mechanism for formation of a thick fibrous cap to protect the atherosclerotic plaque from rupture.


Assuntos
Aterosclerose/patologia , Colágeno Tipo VIII/fisiologia , Placa Aterosclerótica/patologia , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/fisiologia , Aterosclerose/metabolismo , Movimento Celular/fisiologia , Proliferação de Células , Células Cultivadas , Colágeno/metabolismo , Colágeno Tipo VIII/deficiência , Colágeno Tipo VIII/genética , Elastina/metabolismo , Feminino , Artéria Femoral/lesões , Gelatinases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Necrose , Placa Aterosclerótica/metabolismo , RNA Mensageiro/genética , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia
15.
J Cell Sci ; 124(Pt 12): 2013-20, 2011 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-21610088

RESUMO

Cadherins aggregate and stabilize cell-cell junctions through interactions with adjacent cells. In addition, N-cadherin and E-cadherin concentrate at free edges or at the lamellipodia of migrating cells and are found within large vesicles called macropinosomes, which develop from membrane ruffles. The binding properties of cadherins have not previously been associated with the localization of cadherins at membrane ruffles; however, we report that the dorsal, ventral and lateral membrane contacts that occur as a result of the overlap of membrane ruffles aggregate N-cadherin, and that both N-cadherin and E-cadherin promote macropinosome closure and fluid-phase uptake in macropinosomes. These data reveal a previously unsuspected function for cadherin-mediated cell-cell adhesion molecules in the closure of cell-autonomous membrane contacts at membrane ruffles, resulting in macropinocytosis.


Assuntos
Caderinas/fisiologia , Comunicação Celular/fisiologia , Pinocitose/fisiologia , Animais , Becaplermina , Caderinas/metabolismo , Adesão Celular/fisiologia , Comunicação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Dextranos/metabolismo , Humanos , Camundongos , Músculo Liso/citologia , Músculo Liso/metabolismo , Pinocitose/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Proto-Oncogênicas c-sis , Pseudópodes/efeitos dos fármacos , Pseudópodes/metabolismo
16.
J Vasc Res ; 50(4): 279-88, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23988659

RESUMO

UNLABELLED: In vitro, insulin has both growth-promoting and vasculoprotective effects. In vivo, the effect of insulin is mainly protective. Insulin treatment (3 U/day) decreases smooth muscle cell (SMC) migration and neointimal growth after carotid angioplasty in normal rats maintained at normoglycemia by oral glucose. SMC migration requires limited proteolysis of the extracellular matrix, which is mediated by matrix metalloproteinases (MMPs). In this study, we investigated the effects of normoglycemic hyperinsulinemia on MMP activity after balloon angioplasty. Rats were divided into three groups: (1) control implants and tap water; (2) control implants and oral glucose, and (3) insulin implants (3 U/day) and oral glucose. RESULTS: Gelatin zymography revealed that insulin reduced the gelatinolytic activity of pro-MMP-2 by 46% (p < 0.05), MMP-2 by 44% (p < 0.05) and MMP-9 by 51% (p < 0.05) compared to controls after arterial injury. Insulin also reduced mRNA levels of MMP-2 (p < 0.05) and MMP-9 (p < 0.05) and protein levels of MMP-2 (p < 0.05). In contrast, there were no significant changes in membrane-type 1 MMP protein and tissue inhibitors of MMP activity after insulin treatment. Thus, these results suggest a mechanism by which insulin inhibits SMC migration and supports a vasculoprotective role for insulin in vivo.


Assuntos
Lesões das Artérias Carótidas/tratamento farmacológico , Artéria Carótida Primitiva/efeitos dos fármacos , Insulina/farmacologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Lesões do Sistema Vascular/tratamento farmacológico , Administração Oral , Angioplastia com Balão , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Lesões das Artérias Carótidas/enzimologia , Lesões das Artérias Carótidas/etiologia , Artéria Carótida Primitiva/enzimologia , Modelos Animais de Doenças , Regulação para Baixo , Implantes de Medicamento , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glucose/administração & dosagem , Insulina/administração & dosagem , Insulina/sangue , Masculino , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Inibidores Teciduais de Metaloproteinases/metabolismo , Lesões do Sistema Vascular/enzimologia , Lesões do Sistema Vascular/etiologia
18.
Am J Pathol ; 178(2): 895-910, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21281821

RESUMO

Directed migration of smooth muscle cells (SMCs) from the media to the intima in arteries occurs during atherosclerotic plaque formation and during restenosis after angioplasty or stent application. The polarized orientation of the microtubule-organizing center (MTOC) is a key determinant of this process, and we therefore investigated factors that regulate MTOC polarity in vascular SMCs. SMCs migrating in vivo from the medial to the intimal layer of the rat carotid artery following balloon catheter injury were rear polarized, with the MTOC located posterior of the nucleus. In tissue culture, migrating neointimal cells maintained rear polarization, whereas medial cells were front polarized. Using phosphoproteomic screening and mass spectrometry, we identified ARPC5 and RHAMM as protein kinase C (PKC)-phosphorylated proteins associated with rear polarization of the MTOC in neointimal SMCs. RNA silencing of ARPC5 and RHAMM, PKC inhibition, and transfection with a mutated nonphosphorylatable ARPC5 showed that these proteins regulate rear polarization by organizing the actin and microtubule cytoskeletons in neointimal SMCs. Both ARPC5 and RHAMM, in addition to PKC, were required for migration of neointimal SMCs.


Assuntos
Complexo 2-3 de Proteínas Relacionadas à Actina/metabolismo , Polaridade Celular , Proteínas da Matriz Extracelular/metabolismo , Receptores de Hialuronatos/metabolismo , Centro Organizador dos Microtúbulos/metabolismo , Miócitos de Músculo Liso/patologia , Proteína Quinase C-alfa/metabolismo , Túnica Íntima/patologia , Complexo 2-3 de Proteínas Relacionadas à Actina/antagonistas & inibidores , Animais , Artérias/metabolismo , Artérias/patologia , Contagem de Células , Movimento Celular , Núcleo Celular/metabolismo , Proteínas da Matriz Extracelular/antagonistas & inibidores , Inativação Gênica , Masculino , Modelos Biológicos , Mutação/genética , Fosforilação , Proteína Quinase C-alfa/antagonistas & inibidores , Pseudópodes/metabolismo , Ratos , Ratos Sprague-Dawley , Túnica Íntima/metabolismo , Túnica Média/metabolismo , Túnica Média/patologia
19.
Circ Res ; 106(11): 1775-83, 2010 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-20448217

RESUMO

RATIONALE: Discoidin domain receptor (DDR)1 is a collagen receptor expressed on both smooth muscle cells (SMCs) and macrophages, where it plays important roles regulating cell and matrix accumulation during atherogenesis. Systemic deletion of DDR1 resulted in attenuated plaque growth but accelerated matrix accumulation in LDLR-deficient mice. Deletion of DDR1 solely on bone marrow-derived cells resulted in decreased macrophage accumulation and plaque growth but no change in matrix accumulation. OBJECTIVE: These findings led us to hypothesize that accelerated matrix accumulation was attributable to the increased synthetic ability of Ddr1(-/-) resident vascular wall SMCs. METHODS AND RESULTS: We used bone marrow transplantation to generate chimeric mice and investigate the role of SMC DDR1 during atherogenesis. Mice with deficiency of DDR1 in vessel wall-derived cells (Ddr1(+/+-->-/-)) or control mice (Ddr1(+/+-->+/+)) were fed an atherogenic diet for 12 weeks. We observed a 3.8-fold increase in the size of aortic sinus plaques in Ddr1(+/+-->-/-) compared to Ddr1(+/+-->+/+) mice. This was attributed to pronounced accumulation of collagen, elastin, proteoglycans, and fibronectin and resulted in a thickened fibrous cap. The enhanced matrix accumulation decreased the proportion of plaque area occupied by cells but was associated with a shift in the cellular composition of the lesions toward increased numbers of vessel wall-derived SMCs compared to bone marrow-derived macrophages. In vitro studies confirmed that Ddr1(-/-) SMCs expressed more matrix, proliferated more, and migrated farther than Ddr1(+/+) SMCs. CONCLUSIONS: DDR1 expression on resident vessel wall SMCs limits proliferation, migration and matrix accumulation during atherogenesis.


Assuntos
Doenças da Aorta/enzimologia , Aterosclerose/enzimologia , Proteínas da Matriz Extracelular/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Receptores Proteína Tirosina Quinases/deficiência , Animais , Doenças da Aorta/genética , Doenças da Aorta/patologia , Aterosclerose/genética , Aterosclerose/patologia , Transplante de Medula Óssea , Movimento Celular , Proliferação de Células , Colágeno/metabolismo , Receptor com Domínio Discoidina 1 , Modelos Animais de Doenças , Elastina/metabolismo , Proteínas da Matriz Extracelular/genética , Feminino , Fibronectinas/metabolismo , Fibrose , Macrófagos/metabolismo , Masculino , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Proteoglicanas/metabolismo , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/genética , Fatores de Tempo , Quimeras de Transplante , Regulação para Cima
20.
Matrix Biol ; 110: 129-140, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35562016

RESUMO

OBJECTIVE: Increased matrix stiffness is sensed by the collagen-binding receptor tyrosine kinase discoidin domain receptor 1 (DDR1). We have previously shown that DDR1 stimulates a positive feedback loop to increase its own expression in vascular smooth muscle cells (VSMCs). The transcriptional co-factors YAP/TAZ are stiffness sensing molecules that have not previously been investigated in DDR1 signaling. Here, we test the hypothesis that DDR1 signals through YAP/TAZ to auto-regulate its own expression. APPROACH AND RESULTS: We used vascular smooth muscle cells (VSMCs) from wild-type and DDR1 knockout mice stimulated with collagen and/or substrates of different stiffness. We show that DDR1 controls YAP/TAZ nuclear localization and activity, whereas knockdown of YAP/TAZ attenuates DDR1 expression. In response to increased substrate stiffness, collagen stimulation, or RhoA activation, YAP/TAZ translocate to the nucleus and bind to chromatin. Finally, collagen stimulation promotes increased YAP/TAZ association with the Ddr1 promoter. CONCLUSIONS: These findings reveal the mechanism by which DDR1 regulates YAP/TAZ activity which can then mediate positive feedback regulation of DDR1 expression by promoting transcription of the DDR1 gene.


Assuntos
Receptor com Domínio Discoidina 1/metabolismo , Miócitos de Músculo Liso , Aciltransferases/metabolismo , Animais , Receptor com Domínio Discoidina 1/genética , Retroalimentação , Homeostase , Camundongos , Miócitos de Músculo Liso/metabolismo , Fatores de Transcrição/genética , Proteínas de Sinalização YAP/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa