Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Vasc Res ; 55(4): 210-223, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30071538

RESUMO

Age-associated structural and functional remodeling of the arterial wall produces a productive environment for the initiation and progression of hypertension and atherosclerosis. Chronic aging stress induces low-grade proinflammatory signaling and causes cellular proinflammation in arterial walls, which triggers the structural phenotypic shifts characterized by endothelial dysfunction, diffuse intimal-medial thickening, and arterial stiffening. Microscopically, aged arteries exhibit an increase in arterial cell senescence, proliferation, invasion, matrix deposition, elastin fragmentation, calcification, and amyloidosis. These characteristic cellular and matrix alterations not only develop with aging but can also be induced in young animals under experimental proinflammatory stimulation. Interestingly, these changes can also be attenuated in old animals by reducing low-grade inflammatory signaling. Thus, mitigating age-associated proinflammation and arterial phenotype shifts is a potential approach to retard arterial aging and prevent the epidemic of hypertension and atherosclerosis in the elderly.


Assuntos
Envelhecimento/fisiologia , Artérias/fisiopatologia , Inflamação/fisiopatologia , Rigidez Vascular/fisiologia , Animais , Artérias/patologia , Aterosclerose/fisiopatologia , Aterosclerose/prevenção & controle , Células Endoteliais/fisiologia , Endotelina-1/fisiologia , Endotélio Vascular/fisiopatologia , Humanos , Hipertensão/fisiopatologia , Hipertensão/prevenção & controle , Inflamação/prevenção & controle , Fenótipo , Sistema Renina-Angiotensina/fisiologia , Sistema Nervoso Simpático/fisiopatologia , Síndrome
2.
Gerontology ; 60(6): 519-29, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25171100

RESUMO

Arterial aging is a cornerstone of organismal aging. The central arterial wall structurally and functionally remodels under chronic proinflammatory stress over a lifetime. The low-grade proinflammation that accompanies advancing age causes arterial wall thickening and stiffening. These structural and functional alterations are consequences of adverse molecular and cellular events, e.g. an increase in local angiotensin II signaling that induces an inflammatory phenotypic shift of endothelial and smooth muscle cells. Thus, interventions to restrict proinflammatory signaling are a rational approach to delay or prevent age-associated adverse arterial remodeling.


Assuntos
Envelhecimento/patologia , Artérias/patologia , Túnica Íntima/patologia , Túnica Média/patologia , Angiotensina II/fisiologia , Animais , Artérias/fisiopatologia , Aterosclerose/etiologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Haplorrinos , Humanos , Hipertensão/etiologia , Hipertensão/metabolismo , Hipertensão/patologia , Miócitos de Músculo Liso/fisiologia , Coelhos , Ratos , Transdução de Sinais/fisiologia , Túnica Íntima/fisiopatologia , Túnica Média/fisiopatologia , Remodelação Vascular/fisiologia
3.
Aging Med (Milton) ; 7(3): 414-423, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38975316

RESUMO

Arterial stiffening is a critical risk factor contributing to the exponential rise in age-associated cardiovascular disease incidence. This process involves age-induced arterial proinflammation, collagen deposition, and calcification, which collectively contribute to arterial stiffening. The primary driver of proinflammatory processes leading to collagen deposition in the arterial wall is the transforming growth factor-beta1 (TGF-ß1) signaling. Activation of this signaling is pivotal in driving vascular extracellular remodeling, eventually leading to arterial fibrosis and calcification. Interestingly, the glycosylated protein vasorin (VASN) physically interacts with TGF-ß1, and functionally restraining its proinflammatory fibrotic signaling in arterial walls and vascular smooth muscle cells (VSMCs). Notably, as age advances, matrix metalloproteinase type II (MMP-2) is activated, which effectively cleaves VASN protein in both arterial walls and VSMCs. This age-associated/MMP-2-mediated decrease in VASN levels exacerbates TGF-ß1 activation, amplifying arterial fibrosis and calcification in the arterial wall. Importantly, TGF-ß1 is a downstream molecule of the angiotensin II (Ang II) signaling pathway in the arterial wall and VSMCs, which is modulated by VASN. Indeed, chronic administration of Ang II to young rats significantly activates MMP-2 and diminishes the VASN expression to levels comparable to untreated older control rats. This review highlights and discusses the role played by VASN in mitigating fibrosis and calcification by alleviating TGF-ß1 activation and signaling in arterial walls and VSMCs. Understanding these molecular physical and functional interactions may pave the way for establishing VASN-based therapeutic strategies to counteract adverse age-associated cardiovascular remodeling, eventually reducing the risk of cardiovascular diseases.

4.
Cells ; 12(2)2023 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-36672188

RESUMO

Medin, a small 50-amino acid peptide, is an internal cleaved product from the second discoidin domain of milk fat globule epidermal growth factor VIII (MFG-E8) protein. Medin has been reported as the most common amylogenic protein in the upper part of the arterial system, including aortic, temporal, and cerebral arterial walls in the elderly. Medin has a high affinity to elastic fibers and is closely associated with arterial degenerative inflammation, elastic fiber fragmentation, calcification, and amyloidosis. In vitro, treating with the medin peptide promotes the inflammatory phenotypic shift of both endothelial cells and vascular smooth muscle cells. In vitro, ex vivo, and in vivo studies demonstrate that medin enhances the abundance of reactive oxygen species and reactive nitrogen species produced by both endothelial cells and vascular smooth muscle cells and promotes vascular endothelial dysfunction and arterial stiffening. Immunostaining and immunoblotting analyses of human samples indicate that the levels of medin are increased in the pathogenesis of aortic aneurysm/dissection, temporal arteritis, and cerebrovascular dementia. Thus, medin peptide could be targeted as a biomarker diagnostic tool or as a potential molecular approach to curbing the arterial degenerative inflammatory remodeling that accompanies aging and disease.


Assuntos
Fator de Crescimento Epidérmico , Doenças Vasculares , Humanos , Idoso , Fator de Crescimento Epidérmico/metabolismo , Células Endoteliais/metabolismo , Artérias/metabolismo , Glicoproteínas/metabolismo , Doenças Vasculares/metabolismo
5.
J Am Heart Assoc ; 11(17): e022574, 2022 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-36000422

RESUMO

Background Age-associated aortic remodeling includes a marked increase in intimal medial thickness (IMT), associated with signs of inflammation. Although aortic wall milk fat globule-epidermal growth factor VIII (MFG-E8) increases with age, and is associated with aortic inflammation, it is not known whether MFG-E8 is required for the age-associated increase in aortic IMT. Here, we tested whether MFG-E8 is required for the age-associated increase in aortic IMT. Methods and Results To determine the role of MFG-E8 in the age-associated increase of IMT, we compared aortic remodeling in adult (20-week) and aged (96-week) MFG-E8 (-/-) knockout and age matched wild-type (WT) littermate mice. The average aortic IMT increased with age in the WT from 50±10 to 70±20 µm (P<0.0001) but did not significantly increase with age in MFG-E8 knockout mice. Because angiotensin II signaling is implicated as a driver of age-associated increase in IMT, we infused 30-week-old MFG-E8 knockout and age-matched littermate WT mice with angiotensin II or saline via osmotic mini-pumps to determine whether MFG-E8 is required for angiotensin II-induced aortic remodeling. (1) In WT mice, angiotensin II infusion substantially increased IMT, elastic lamina degradation, collagen deposition, and the proliferation of vascular smooth muscle cells; in contrast, these effects were significantly reduced in MFG-E8 KO mice; (2) On a molecular level, angiotensin II treatment significantly increased the activation and expression of matrix metalloproteinase type 2, transforming growth factor beta 1, and its downstream signaling molecule phosphorylated mother against decapentaplegic homolog 2, and collagen type I production in WT mice; however, in the MFG-E8 knockout mice, these molecular effects were significantly reduced; and (3) in WT mice, angiotensin II increased levels of aortic inflammatory markers phosphorylated nuclear factor-kappa beta p65, monocyte chemoattractant protein 1, tumor necrosis factor alpha, intercellular adhesion molecule 1, and vascular cell adhesion molecule 1 molecular expression, while in contrast, these inflammatory markers did not change in knockout mice. Conclusions Thus, MFG-E8 is required for both age-associated proinflammatory aortic remodeling and also for the angiotensin II-dependent induction in younger mice of an aortic inflammatory phenotype observed in advanced age. Targeting MFG-E8 would be a novel molecular approach to curb adverse arterial remodeling.


Assuntos
Angiotensina II , Fator de Crescimento Epidérmico , Angiotensina II/farmacologia , Animais , Glicolipídeos , Glicoproteínas , Inflamação/metabolismo , Gotículas Lipídicas , Camundongos , Camundongos Knockout , Proteínas do Leite/genética , Proteínas do Leite/metabolismo
6.
Circ Res ; 104(12): 1337-46, 2009 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-19443842

RESUMO

Advancing age induces aortic wall thickening that results from the concerted effects of numerous signaling proteins, many of which have yet to be identified. To search for novel proteins associated with aortic wall thickening, we have performed a comprehensive quantitative proteomic study to analyze aortic proteins from young (8 months) and old (30 months) rats and identified 50 proteins that significantly change in abundance with aging. One novel protein, the milk fat globule protein epidermal growth factor 8 (MFG-E8), increases 2.3-fold in abundance in old aorta. Transcription and translation analysis demonstrated that aortic MFG-E8 mRNA and protein levels increase with aging in several mammalian species including humans. Dual immunolabeling shows that MFG-E8 colocalizes with both angiotensin II and monocyte chemoattractant protein (MCP)-1 within vascular smooth muscle cells (VSMCs) of the thickened aged aortic wall. Exposure of early passage VSMCs from young aorta to angiotensin II markedly increases MFG-E8 and enhances invasive capacity to levels observed in VSMCs from old rats. Treatment of VSMCs with MFG-E8 increases MCP-1 expression and VSMCs invasion that are inhibited by the MCP-1 receptor blocker vCCI. Silencing MFG-E8 RNA substantially reduces MFG-E8 expression and VSMCs invasion capacity. The data indicate that arterial MFG-E8 significantly increases with aging and is a pivotal relay element within the angiotensin II/MCP-1/VSMC invasion signaling cascade. Thus, targeting of MFG-E8 within this signaling axis pathway is a potential novel therapy for the prevention and treatment of the age-associated vascular diseases such as atherosclerosis.


Assuntos
Angiotensina II/farmacologia , Antígenos de Superfície/biossíntese , Movimento Celular/efeitos dos fármacos , Quimiocina CCL2/biossíntese , Proteínas do Leite/biossíntese , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Transdução de Sinais/efeitos dos fármacos , Vasoconstritores/farmacologia , Adolescente , Adulto , Idoso , Envelhecimento/efeitos dos fármacos , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Antígenos de Superfície/genética , Aorta/metabolismo , Aorta/patologia , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Movimento Celular/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Inativação Gênica , Humanos , Macaca mulatta , Masculino , Pessoa de Meia-Idade , Proteínas do Leite/genética , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Ratos , Ratos Endogâmicos F344 , Receptores CCR2/genética , Receptores CCR2/metabolismo , Proteínas Virais/farmacologia , Fatores de Virulência/farmacologia
7.
Mech Ageing Dev ; 196: 111490, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33839189

RESUMO

Elastic fibers are the main components of the extracellular matrix of the large arterial wall. Elastic fiber remodeling is an intricate process of synthesis and degradation of the core elastin protein and microfibrils accompanied by the assembly and disassembly of accessory proteins. Age-related morphological, structural, and functional proinflammatory remodeling within the elastic fiber has a profound effect upon the integrity, elasticity, calcification, amyloidosis, and stiffness of the large arterial wall. An age-associated increase in arterial stiffness is a major risk factor for the pathogenesis of diseases of the large arteries such as hypertensive and atherosclerotic vasculopathy. This mini review is an update on the key molecular, cellular, functional, and structural mechanisms of elastic fiber proinflammatory remodeling in large arteries with aging. Targeting structural and functional integrity of the elastic fiber may be an effective approach to impede proinflammatory arterial remodeling with advancing age.


Assuntos
Envelhecimento/fisiologia , Artérias , Tecido Elástico , Remodelação Vascular/imunologia , Artérias/patologia , Artérias/fisiopatologia , Tecido Elástico/imunologia , Tecido Elástico/patologia , Tecido Elástico/fisiopatologia , Fatores de Risco de Doenças Cardíacas , Humanos , Inflamação/patologia , Inflamação/fisiopatologia
8.
Curr Opin Nephrol Hypertens ; 19(2): 201-7, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20040868

RESUMO

PURPOSE OF REVIEW: Age-associated arterial alterations in cells, matrix, and biomolecules are the foundation for the initiation and progression of cardiovascular diseases in older persons. This review focuses on the latest advances on the intertwining of aging and disease within the arterial wall at the cell and molecular levels. RECENT FINDINGS: Endothelial dysfunction, vascular smooth muscle cell (VSMC) proliferation/invasion/secretion, matrix fragmentation, collagenization and glycation are characteristics of an age-associated arterial phenotype that creates a microenvironment enriched with reactive oxygen species (ROS) for the pathogenesis of arterial disease. This niche creates an age-associated arterial secretory phenotype (AAASP), which is orchestrated by the concerted effects of numerous age-modified angiotensin II signaling molecules. Most of these biomolecular, cell, and matrix modifications that constitute the AAASP can be elicited by experimental hypertension or atherosclerosis at a younger age. The arterial AAASP also shares features of a senescence-associated secretory phenotype (SASP) identified in other mesenchymocytes, that is, fibroblasts. SUMMARY: A subclinical AAASP evolves during aging. Targeting this subclinical AAASP may reduce the incidence and progression of the quintessential age-associated arterial diseases, that is, hypertension and atherosclerosis.


Assuntos
Envelhecimento/fisiologia , Artérias/crescimento & desenvolvimento , Artérias/fisiologia , Doenças Vasculares/patologia , Angiotensina II/fisiologia , Animais , Aterosclerose/patologia , Humanos , Transdução de Sinais/fisiologia
9.
Aging Med (Milton) ; 3(3): 159-168, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33103036

RESUMO

Aging is a major risk factor for quintessential cardiovascular diseases, which are closely related to arterial proinflammation. The age-related alterations of the amount, distribution, and properties of the collagen fibers, such as cross-links and degradation in the arterial wall, are the major sequelae of proinflammation. In the aging arterial wall, collagen types I, II, and III are predominant,  and are mainly produced by stiffened vascular smooth muscle cells (VSMCs) governed by proinflammatory signaling, leading to profibrosis. Profibrosis is regulated by an increase in the proinflammatory molecules angiotensin II, milk fat globule-EGF-VIII, and transforming growth factor-beta 1 (TGF-ß1) signaling and a decrease in the vasorin signaling cascade. The release of these proinflammatory factors triggers the activation of matrix metalloproteinase type II (MMP-2) and activates profibrogenic TGF-ß1 signaling, contributing to profibrosis. The age-associated increase in activated MMP-2 cleaves latent TGF-ß and subsequently increases TGF-ß1 activity leading to collagen deposition in the arterial wall. Furthermore, a blockade of the proinflammatory signaling pathway alleviates the fibrogenic signaling, reduces profibrosis, and prevents arterial stiffening with aging. Thus, age-associated proinflammatory-profibrosis coupling is the underlying molecular mechanism of arterial stiffening with advancing age.

10.
PLoS One ; 14(12): e0225911, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31805124

RESUMO

Collagen accumulation and remodeling in the vascular wall is a cardinal feature of vascular fibrosis that exacerbates the complications of hypertension, aging, diabetes and atherosclerosis. With no specific therapy available to date, identification of mechanisms underlying vascular fibrogenesis is an important clinical goal. Here, we tested the hypothesis that Discoidin Domain Receptor 2 (DDR2), a collagen-specific receptor tyrosine kinase, is a determinant of arterial fibrosis. We report a significant increase in collagen type 1 levels along with collagen and ECM remodeling, degradation of elastic laminae, enhanced fat deposition and calcification in the abdominal aorta in a non-human primate model of high-fat, high-sucrose diet (HFS)-induced metabolic syndrome. These changes were associated with a marked increase in DDR2. Resveratrol attenuated collagen type I deposition and remodeling induced by the HFS diet, with a concomintant reduction in DDR2. Further, in isolated rat vascular adventitial fibroblasts and VSMCs, hyperglycemia increased DDR2 and collagen type I expression via TGF-ß1/SMAD2/3, which was attenuated by resveratrol. Notably, gene knockdown and overexpression approaches demonstrated an obligate role for DDR2 in hyperglycemia-induced increase in collagen type I expression in these cells. Together, our observations point to DDR2 as a hitherto unrecognized molecular link between metabolic syndrome and arterial fibrosis, and hence a therapeutic target.


Assuntos
Artérias/metabolismo , Artérias/patologia , Receptor com Domínio Discoidina 2/genética , Síndrome Metabólica/complicações , Síndrome Metabólica/genética , Doenças Vasculares/etiologia , Doenças Vasculares/patologia , Animais , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Receptor com Domínio Discoidina 2/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Fibroblastos/metabolismo , Fibrose , Masculino , Síndrome Metabólica/metabolismo , Primatas
11.
J Am Heart Assoc ; 7(18): e009112, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30371211

RESUMO

Background Aging exponentially increases the incidence of morbidity and mortality of quintessential cardiovascular disease mainly due to arterial proinflammatory shifts at the molecular, cellular, and tissue levels within the arterial wall. Calorie restriction ( CR ) in rats improves arterial function and extends both health span and life span. How CR affects the proinflammatory landscape of molecular, cellular, and tissue phenotypic shifts within the arterial wall in rats, however, remains to be elucidated. Methods and Results Aortae were harvested from young (6-month-old) and old (24-month-old) Fischer 344 rats, fed ad libitum and a second group maintained on a 40% CR beginning at 1 month of age. Histopathologic and morphometric analysis of the arterial wall demonstrated that CR markedly reduced age-associated intimal medial thickening, collagen deposition, and elastin fractionation/degradation within the arterial walls. Immunostaining/blotting showed that CR effectively prevented an age-associated increase in the density of platelet-derived growth factor, matrix metalloproteinase type II activity, and transforming growth factor beta 1 and its downstream signaling molecules, phospho-mothers against decapentaplegic homolog-2/3 (p- SMAD -2/3) in the arterial wall. In early passage cultured vascular smooth muscle cells isolated from AL and CR rat aortae, CR alleviated the age-associated vascular smooth muscle cell phenotypic shifts, profibrogenic signaling, and migration/proliferation in response to platelet-derived growth factor. Conclusions CR reduces matrix and cellular proinflammation associated with aging that occurs within the aortic wall and that are attributable to platelet-derived growth factor signaling. Thus, CR reduces the platelet-derived growth factor-associated signaling cascade, contributing to the postponement of biological aging and preservation of a more youthful aortic wall phenotype.


Assuntos
Envelhecimento/fisiologia , Aorta Torácica/metabolismo , Restrição Calórica , Inflamação/metabolismo , Músculo Liso Vascular/metabolismo , Doenças Vasculares/prevenção & controle , Animais , Aorta Torácica/patologia , Células Cultivadas , Modelos Animais de Doenças , Inflamação/patologia , Masculino , Músculo Liso Vascular/patologia , Fenótipo , Ratos , Ratos Endogâmicos F344 , Doenças Vasculares/metabolismo , Doenças Vasculares/patologia
12.
Sci Rep ; 8(1): 2668, 2018 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-29422510

RESUMO

Here we report exquisitely distinct material properties of primary vascular smooth muscle (VSM) cells isolated from the thoracic aorta of adult (8 months) vs. aged (30 months) F344XBN rats. Individual VSM cells derived from the aged animals showed a tense internal network of the actin cytoskeleton (CSK), exhibiting increased stiffness (elastic) and frictional (loss) moduli than those derived from the adult animals over a wide frequency range of the imposed oscillatory deformation. This discrete mechanical response was long-lived in culture and persistent across a physiological range of matrix rigidity. Strikingly, the pro-fibrotic transforming growth factor ß1 (TGFß1) emerged as a specific modifier of age-associated VSM stiffening in vitro. TGFß1 reinforced the mechanical phenotype of arterial aging in VSM cells on multiple time and length scales through clustering of mechanosensitive α5ß1 and αvß3 integrins. Taken together, these studies identify a novel nodal point for the long-range regulation of VSM stiffness and serve as a proof-of-concept that the broad-based inhibition of TGFß1 expression, or TGFß1 signal transduction in VSM, may be a useful therapeutic approach to mitigate the pathologic progression of central arterial wall stiffening associated with aging.


Assuntos
Músculo Liso Vascular/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Rigidez Vascular/fisiologia , Actinas/metabolismo , Fatores Etários , Animais , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Artérias/metabolismo , Citoesqueleto/metabolismo , Elasticidade/fisiologia , Hipertensão/metabolismo , Masculino , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Ratos , Ratos Endogâmicos BN , Transdução de Sinais
13.
Oncotarget ; 9(43): 27117-27132, 2018 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-29930755

RESUMO

The glycosylated protein vasorin physically interacts with the transforming growth factor-beta1 (TGF-ß1) and functionally attenuates its fibrogenic signaling in the vascular smooth muscle cells (VSMCs) of the arterial wall. Angiotensin II (Ang II) amplifies TGF-ß1 activation in the VSMCs of the arterial wall with aging. In this study, we hypothesized that a reduced expression of the protein vasorin plays a contributory role in magnifying Ang II-associated fibrogenic signaling in the VSMCs of the arterial wall with aging. The current study shows that vasorin mRNA and protein expression were significantly decreased both in aortic wall and VSMCs from old (30 mo) vs. young (8 mo) FXBN rats. Exposing young VSMCs to Ang II reduced vasorin protein expression to the levels of old untreated cells while treating old VSMCs with the Ang II type AT1 receptor antagonist Losartan upregulated vasorin protein expression up to the levels of young. The physical interaction between vasorin and TGF-ß1 was significantly decreased in old vs. young VSMCs. Further, exposing young VSMCs to Ang II increased the levels of matrix metalloproteinase type II (MMP-2) activation and TGF-ß1 downstream molecules p-SMAD-2/3 and collagen type I production up to the levels of old untreated VSMCs, and these effects were substantially inhibited by overexpressing vasorin. Administration of Ang II to young rats (8 mo) for 28 days via an osmotic minipump markedly reduced the expression of vasorin. Importantly, vasorin protein was effectively cleaved by activated MMP-2 both in vitro and in vivo. Administration of the MMP inhibitor, PD 166793, for 6 mo to young adult (18 mo) via a daily gavage markedly increased levels of vasorin in the aortic wall. Thus, reduced vasorin amplifies Ang II profibrotic signaling via an activation of MMP-2 in VSMCs within the aging arterial wall.

14.
Arterioscler Thromb Vasc Biol ; 26(7): 1503-9, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16690877

RESUMO

OBJECTIVE: To study matrix metalloproteinase 2 (MMP-2) effects on transforming growth factor-beta1 (TGF-beta1) activation status and downstream signaling during arterial aging. METHODS AND RESULTS: Western blotting and immunostaining showed that latent and activated TGF-beta1 are markedly increased within the aorta of aged Fisher 344 cross-bred Brown Norway (30 months of age) rats compared with adult (8 months of age) rats. Aortic TGF-beta1-type II receptor (TbetaRII), its downstream molecules p-similar to mad-mother against decapentaplegic (SMAD)2/3 and SMAD4, fibronectin, and collagen also increased with age. Moreover, TGF-beta1 staining is colocalized with that of activated MMP-2 within the aged arterial wall and vascular smooth muscle cell (VSMC) in vitro, and this physical association was confirmed by coimmunoprecipitation. Incubation of young aortic rings ex vivo or VSMCs in vitro with activated MMP-2 enhanced active TGF-beta1, collagen, and fibronectin expression to the level of untreated old counterparts, and this effect was abolished via inhibitors of MMP-2. Interestingly, in old untreated rings or VSMCs, the increased TGF-beta1, fibronectin, and collagen were also substantially reduced by inhibition of MMP-2. CONCLUSIONS: Active TGF-beta1, its receptor, and receptor-mediated signaling increase within the aortic wall with aging. TGF-beta1 activation is dependent, in part at least, by a concomitant age-associated increase in MMP-2 activity. Thus, MMP-2-activated TGF-beta1, and subsequently TbetaRII signaling, is a novel molecular mechanism for arterial aging.


Assuntos
Envelhecimento/fisiologia , Aorta/fisiologia , Metaloproteinase 2 da Matriz/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta/metabolismo , Envelhecimento/metabolismo , Animais , Aorta/metabolismo , Biomarcadores/metabolismo , Colágeno/biossíntese , Endotélio Vascular/metabolismo , Fibronectinas/biossíntese , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Serina-Treonina Quinases , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos F344 , Receptor do Fator de Crescimento Transformador beta Tipo II , Proteínas Smad/metabolismo , Distribuição Tecidual , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta1
15.
Arterioscler Thromb Vasc Biol ; 24(8): 1397-402, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15178559

RESUMO

OBJECTIVE: With age, rat arterial walls thicken and vascular smooth muscle cells (VSMCs) exhibit enhanced migration and proliferation. Monocyte chemotactic protein-1 (MCP-1) affects these VSMC properties in vitro. Because arterial angiotensin II, which induces MCP-1 expression, increases with age, we hypothesized that aortic MCP-1 and its receptor CCR2 are also upregulated and affect VSMC properties. METHODS AND RESULTS: Both MCP-1 and CCR2 mRNAs and proteins increased in old (30-month) versus young (8-month) F344xBN rat aortas in vivo. Cellular MCP-1 and CCR2 staining colocalized with that of alpha-smooth muscle actin in the thickened aortas of old rats and were expressed by early-passage VSMCs isolated from old aortas, which, relative to young VSMCs, exhibited increased invasion, and the age difference was abolished by vCCI, an inhibitor of CCR2 signaling. MCP-1 treatment of young VSMCs induced migration and increased their ability to invade a synthetic basement membrane. The MCP-1-dependent VSMC invasiveness was blocked by vCCI. After MCP-1 treatment, migration and invasion capacities of VSMCs from young aortas no longer differed from those of VSMCs isolated from older rats. CONCLUSIONS: Arterial wall and VSMC MCP-1/CCR2 increase with aging. MCP-1 enhances VSMC migration and invasion, and thus, MCP-1/CCR2 signaling may play a role in age-associated arterial remodeling.


Assuntos
Envelhecimento/metabolismo , Aorta/crescimento & desenvolvimento , Quimiocina CCL2/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Músculo Liso Vascular/crescimento & desenvolvimento , Receptores de Quimiocinas/biossíntese , Envelhecimento/patologia , Angiotensina II , Animais , Aorta/metabolismo , Aorta/patologia , Divisão Celular , Movimento Celular , Células Cultivadas/citologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Quimiocina CCL2/genética , Quimiocina CCL2/farmacologia , Quimiocina CCL2/fisiologia , Quimiotaxia/efeitos dos fármacos , Cruzamentos Genéticos , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Masculino , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , RNA Mensageiro/biossíntese , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos F344 , Receptores CCR2 , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/fisiologia , Proteínas Virais/farmacologia , Fatores de Virulência
16.
J Gerontol A Biol Sci Med Sci ; 57(6): B239-46, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12023260

RESUMO

We examined the in vitro proliferative potential of 669 cell cultures established from skin biopsies of members of the Baltimore Longitudinal Study of Aging. The colony size distribution was used to estimate the proliferative life span of the cultures. A significant decline in proliferative potential with donor age was observed for female but not male donors. For both male and female donors, the proliferative potential was significantly greater for donors under the age of 30 years compared with all donors over the age of 30 years. In an attempt to reduce genetic heterogeneity, we examined the proliferative potential of cultures derived at different ages from the same donor. These studies revealed a trend (approaching statistical significance) toward low proliferative potential as donors aged. Interestingly, samples obtained from donors who had a history of skin cancer at the time of biopsy had a significantly lower doubling potential than those from donors who did not. The implications of these results for the use of cells derived from donors of different ages for aging research are discussed.


Assuntos
Envelhecimento/patologia , Envelhecimento/fisiologia , Fibroblastos/patologia , Fibroblastos/fisiologia , Adulto , Fatores Etários , Idoso , Análise de Variância , Biópsia por Agulha , Divisão Celular/fisiologia , Sobrevivência Celular , Células Cultivadas , Feminino , Humanos , Técnicas In Vitro , Modelos Lineares , Estudos Longitudinais , Masculino , Pessoa de Meia-Idade , Sensibilidade e Especificidade , Fatores Sexuais , Pele/citologia
17.
Trends Endocrinol Metab ; 25(2): 72-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24365513

RESUMO

Arterial aging is the major contributing factor to increases in the incidence and prevalence of cardiovascular disease, due mainly to the presence of chronic, low-grade, 'sterile' arterial inflammation. Inflammatory signaling driven by the angiotensin II cascade perpetrates adverse age-associated arterial structural and functional remodeling. The aged artery is characterized by endothelial disruption, enhanced vascular smooth muscle cell (VMSC) migration and proliferation, extracellular matrix (ECM) deposition, elastin fracture, and matrix calcification/amyloidosis/glycation. Importantly, the molecular mechanisms of arterial aging are also relevant to the pathogenesis of hypertension and atherosclerosis. Age-associated arterial proinflammation is to some extent mutable, and interventions to suppress or delay it may have the potential to ameliorate or retard age-associated arterial diseases.


Assuntos
Envelhecimento/fisiologia , Aterosclerose/etiologia , Hipertensão/fisiopatologia , Inflamação/fisiopatologia , Idoso , Angiotensina II/fisiologia , Animais , Antígenos de Superfície/fisiologia , Artérias , Arterite/fisiopatologia , Calpaína/fisiologia , Quimiocina CCL2/fisiologia , Endotelina-1/fisiologia , Humanos , Mediadores da Inflamação/fisiologia , Metaloproteinase 2 da Matriz/fisiologia , Pessoa de Meia-Idade , Proteínas do Leite , Músculo Liso Vascular/patologia , Óxido Nítrico/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Receptores CCR2/fisiologia , Fator de Crescimento Transformador beta1/fisiologia
18.
Hypertension ; 60(2): 459-66, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22689745

RESUMO

Age-associated arterial remodeling involves arterial wall collagen deposition and elastin fragmentation, as well as an increase in arterial pressure. This arterial remodeling is linked to proinflammatory signaling, including transforming growth factor-ß1, monocyte chemoattractant protein 1, and proendothelin 1, activated by extracellular matrix metalloproteinases (MMPs) and orchestrated, in part, by the transcriptional factor ets-1. We tested the hypothesis that inhibition of MMP activation can decelerate the age-associated arterial proinflammation and its attendant increase in arterial pressure. Indeed, chronic administration of a broad-spectrum MMP inhibitor, PD166739, via a daily gavage, to 16-month-old rats for 8 months markedly blunted the expected age-associated increases in arterial pressure. This was accompanied by the following: (1) inhibition of the age-associated increases in aortic gelatinase and interstitial collagenase activity in situ; (2) preservation of the elastic fiber network integrity; (3) a reduction of collagen deposition; (4) a reduction of monocyte chemoattractant protein 1 and transforming growth factor-ß1 activation; (5) a diminution in the activity of the profibrogenic signaling molecule SMAD-2/3 phosphorylation; (6) inhibition of proendothelin 1 activation; and (7) downregulation of expression of ets-1. Acute exposure of cultured vascular smooth muscle cells in vitro to proendothelin 1 increased both the transcription and translation of ets-1, and these effects were markedly reduced by MMP inhibition. Furthermore, infection of vascular smooth muscle cells with an adenovirus harboring a full-length ets-1 cDNA increased activities of both transforming growth factor-ß1 and monocyte chemoattractant protein 1. Collectively, our results indicate that MMP inhibition retards age-associated arterial proinflammatory signaling, and this is accompanied by preservation of intact elastin fibers, a reduction in collagen, and blunting of an age-associated increase in blood pressure.


Assuntos
Envelhecimento/metabolismo , Arterite/prevenção & controle , Inibidores Enzimáticos/farmacologia , Ácidos Hidroxâmicos/farmacologia , Hipertensão/prevenção & controle , Inibidores de Metaloproteinases de Matriz , Metaloproteinases da Matriz/efeitos dos fármacos , Oligopeptídeos/farmacologia , Animais , Arterite/metabolismo , Arterite/fisiopatologia , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Quimiocina CCL2/metabolismo , Colágeno/metabolismo , Modelos Animais de Doenças , Elastina/metabolismo , Endotelina-1/metabolismo , Gelatinases/metabolismo , Hipertensão/fisiopatologia , Masculino , Precursores de Proteínas/metabolismo , Proteína Proto-Oncogênica c-ets-1/metabolismo , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos F344 , Fator de Crescimento Transformador beta1/metabolismo
19.
Hypertension ; 60(5): 1192-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23006733

RESUMO

Age-associated central arterial wall stiffness is linked to extracellular matrix remodeling, including fibrosis and vascular calcification. Angiotensin II induces both matrix metalloproteinase 2 (MMP2) and calpain-1 expression and activity in the arterial wall. However, the role of calpain-1 in MMP2 activation and extracellular matrix remodeling remains unknown. Dual histo-immunolabeling demonstrates colocalization of calpain-1 and MMP2 within old rat vascular smooth muscle cells. Overexpression of calpain-1 induces MMP2 transcripts, protein levels, and activity, in part, by increasing the ratio of membrane type 1 MMPs to tissue inhibitor of metalloproteinases 2. These effects of calpain-1 overexpression-induced MMP2 activation are linked to increased collagen I and III production and vascular calcification. In addition, overexpression of calpain-1 also induces transforming growth factor-ß1/Smad signaling, elastin degradation, alkaline phosphatase activation, and total calcium content but reduces the expression of calcification inhibitors, osteopontin, and osteonectin, in cultured vascular smooth muscle cells in vitro and in carotid artery rings ex vivo. Furthermore, both calpain-1 and collagen II increase with aging within human aortic intima. Interestingly, in aged human aortic wall, both calpain-1 and collagen II are highly expressed in artherosclerotic plaque areas compared with grossly normal areas. Cross-talk of 2 proteases, calpain-1 and MMP2, leads to secretion of active MMP2, which modulates extracellular matrix remodeling via enhancing collagen production and facilitating vascular calcification. These results establish calpain-1 as a novel molecular candidate to retard age-associated extracellular matrix remodeling and its attendant risk for hypertension and atherosclerosis.


Assuntos
Envelhecimento , Aorta/metabolismo , Calpaína/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Miócitos de Músculo Liso/metabolismo , Adolescente , Idoso , Animais , Aorta/patologia , Western Blotting , Calcinose/genética , Calcinose/metabolismo , Calpaína/genética , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Elastina/genética , Elastina/metabolismo , Ativação Enzimática , Fibrose , Humanos , Masculino , Metaloproteinase 2 da Matriz/genética , Pessoa de Meia-Idade , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Inibidor Tecidual de Metaloproteinase-2/genética , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Adulto Jovem
20.
Aging Cell ; 11(3): 500-8, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22385834

RESUMO

An accumulation of milk fat globule EGF-8 protein (MFG-E8) occurs within the context of arterial wall inflammatory remodeling during aging, hypertension, diabetes mellitus, or atherosclerosis. MFG-E8 induces VSMC invasion, but whether it affects VSMC proliferation, a salient feature of arterial inflammation, is unknown. Here, we show that in the rat arterial wall in vivo, PCNA and Ki67, markers of cell cycle activation, increase with age between 8 and 30 months. In fresh and early passage VSMC isolated from old aortae, an increase in CDK4 and PCNA, an increase in the acceleration of cell cycle S and G2 phases, decrease in the G1/G0 phase, and an increase in PDGF and its receptors confer elevated proliferative capacity, compared to young VSMC. Increased coexpression and physical interaction of MFG-E8 and integrin αvß5 occur with aging in both the rat aortic wall in vivo and in VSMC in vitro. In young VSMC in vitro, MFG-E8 added exogenously, or overexpressed endogenously, triggers phosphorylation of ERK1/2, augmented levels of PCNA and CDK4, increased BrdU incorporation, and promotes proliferation, via αvß5 integrins. MFG-E8 silencing, or its receptor inhibition, or the blockade of ERK1/2 phosphorylation in these cells reduces PCNA and CDK4 levels and decelerates the cell cycle S phase, conferring a reduction in proliferative capacity. Collectively, these results indicate that MFG-E8 in a dose-dependent manner coordinates the expression of cell cycle molecules and facilitates VSMC proliferation via integrin/ERK1/2 signaling. Thus, an increase in MFG-E8 signaling is a mechanism of the age-associated increase in aortic VSMC proliferation.


Assuntos
Antígenos de Superfície/metabolismo , Integrinas/metabolismo , Proteínas do Leite/metabolismo , Músculo Liso Vascular/citologia , Fatores Etários , Animais , Antígenos de Superfície/biossíntese , Antígenos de Superfície/genética , Processos de Crescimento Celular/fisiologia , Imuno-Histoquímica , Sistema de Sinalização das MAP Quinases , Masculino , Proteínas do Leite/biossíntese , Proteínas do Leite/genética , Músculo Liso Vascular/metabolismo , Fosforilação , Fator de Crescimento Derivado de Plaquetas/metabolismo , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos F344 , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA