Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Sci ; 136(19)2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37737020

RESUMO

The Rho family GTPases Rac and Rho play critical roles in transmitting mechanical information contained within the extracellular matrix (ECM) to the cell. Rac and Rho have well-described roles in regulating stiffness-dependent actin remodeling, proliferation and motility. However, much less is known about the relative roles of these GTPases in stiffness-dependent transcription, particularly at the genome-wide level. Here, we selectively inhibited Rac and Rho in mouse embryonic fibroblasts cultured on deformable substrata and used RNA sequencing to elucidate and compare the contribution of these GTPases to the early transcriptional response to ECM stiffness. Surprisingly, we found that the stiffness-dependent activation of Rac was dominant over Rho in the initial transcriptional response to ECM stiffness. We also identified activating transcription factor 3 (ATF3) as a major target of stiffness- and Rac-mediated signaling and show that ATF3 repression by ECM stiffness helps to explain how the stiffness-dependent activation of Rac results in the induction of cyclin D1.


Assuntos
Fator 3 Ativador da Transcrição , Fibroblastos , Animais , Camundongos , Fator 3 Ativador da Transcrição/genética , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Proteínas rac de Ligação ao GTP/genética , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Transdução de Sinais
2.
Am J Pathol ; 190(5): 1118-1136, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32084369

RESUMO

Fibroblast activation protein (FAP) has been established as an inducible and mesenchymal cell-specific mediator of disease progression in cancer and fibrosis. Atherosclerosis is a fibroinflammatory disease, and FAP was previously reported to be up-regulated in human atherosclerotic plaques compared with normal vessel. We investigated the spatial and temporal distribution of Fap-expressing cells in a murine model of atherosclerosis and used a genetic approach to determine if and how Fap affected disease progression. Fap was found to be expressed predominantly on vascular smooth muscle cells in lesions of athero-prone Apoe-/- mice. Global deletion of Fap (Fap-/-) in Apoe-/- mice accelerated atherosclerotic disease progression in both males and females, with the effect observed earlier in males. Sex-specific effects on lesion morphology were observed. Relative levels of extracellular matrix, fibrotic, and inflammatory cell content were comparable in lesions in male mice regardless of Fap status. In contrast, lesions in Fap-/- female mice were characterized by a more fibrotic composition due to a reduction in inflammation, specifically a reduction in Mox macrophages. Combined, these data suggest that Fap restrains the progression of atherosclerosis and may contribute to the sexually dimorphic susceptibility to atherosclerosis by regulating the balance between inflammation (an indicator of vulnerability to plaque rupture) and fibrosis (an indicator of plaque stability).


Assuntos
Aterosclerose/metabolismo , Fibrose/metabolismo , Gelatinases/metabolismo , Inflamação/metabolismo , Proteínas de Membrana/metabolismo , Serina Endopeptidases/metabolismo , Caracteres Sexuais , Animais , Apolipoproteínas E/deficiência , Endopeptidases , Feminino , Masculino , Camundongos , Camundongos Knockout para ApoE
3.
Proc Natl Acad Sci U S A ; 114(33): 8758-8763, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28765370

RESUMO

The integrity of the endothelial barrier between circulating blood and tissue is important for blood vessel function and, ultimately, for organ homeostasis. Here, we developed a vessel-on-a-chip with perfused endothelialized channels lined with human bone marrow stromal cells, which adopt a mural cell-like phenotype that recapitulates barrier function of the vasculature. In this model, barrier function is compromised upon exposure to inflammatory factors such as LPS, thrombin, and TNFα, as has been observed in vivo. Interestingly, we observed a rapid physical withdrawal of mural cells from the endothelium that was accompanied by an inhibition of endogenous Rac1 activity and increase in RhoA activity in the mural cells themselves upon inflammation. Using a system to chemically induce activity in exogenously expressed Rac1 or RhoA within minutes of stimulation, we demonstrated RhoA activation induced loss of mural cell coverage on the endothelium and reduced endothelial barrier function, and this effect was abrogated when Rac1 was simultaneously activated. We further showed that N-cadherin expression in mural cells plays a key role in barrier function, as CRISPR-mediated knockout of N-cadherin in the mural cells led to loss of barrier function, and overexpression of N-cadherin in CHO cells promoted barrier function. In summary, this bicellular model demonstrates the continuous and rapid modulation of adhesive interactions between endothelial and mural cells and its impact on vascular barrier function and highlights an in vitro platform to study the biology of perivascular-endothelial interactions.


Assuntos
Caderinas/metabolismo , Endotélio Vascular/metabolismo , Endotélio/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Biomimética/métodos , Células CHO , Cricetulus , Humanos , Inflamação/metabolismo , Trombina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 38(1): 164-173, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29051139

RESUMO

OBJECTIVE: Vascular extracellular matrix stiffening is a risk factor for aortic and coronary artery disease. How matrix stiffening regulates the transcriptome profile of human aortic and coronary vascular smooth muscle cells (VSMCs) is not well understood. Furthermore, the role of long noncoding RNAs (lncRNAs) in the cellular response to stiffening has never been explored. This study characterizes the stiffness-sensitive (SS) transcriptome of human aortic and coronary VSMCs and identifies potential key lncRNA regulators of stiffness-dependent VSMC functions. APPROACH AND RESULTS: Aortic and coronary VSMCs were cultured on hydrogel substrates mimicking physiological and pathological extracellular matrix stiffness. Total RNAseq was performed to compare the SS transcriptome profiles of aortic and coronary VSMCs. We identified 3098 genes (2842 protein coding and 157 lncRNA) that were stiffness sensitive in both aortic and coronary VSMCs (false discovery rate <1%). Hierarchical clustering revealed that aortic and coronary VSMCs grouped by stiffness rather than cell origin. Conservation analyses also revealed that SS genes were more conserved than stiffness-insensitive genes. These VSMC SS genes were less tissue-type specific and expressed in more tissues than stiffness-insensitive genes. Using unbiased systems analyses, we identified MALAT1 as an SS lncRNA that regulates stiffness-dependent VSMC proliferation and migration in vitro and in vivo. CONCLUSIONS: This study provides the transcriptomic landscape of human aortic and coronary VSMCs in response to extracellular matrix stiffness and identifies novel SS human lncRNAs. Our data suggest that the SS transcriptome is evolutionarily important to VSMCs function and that SS lncRNAs can act as regulators of stiffness-dependent phenotypes.


Assuntos
Biologia Computacional/métodos , Mineração de Dados/métodos , Matriz Extracelular/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , RNA Longo não Codificante/metabolismo , Transcriptoma , Rigidez Vascular , Aorta/metabolismo , Aorta/patologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Análise por Conglomerados , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Matriz Extracelular/genética , Matriz Extracelular/patologia , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Humanos , Hidrogéis , Mecanotransdução Celular , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , RNA Longo não Codificante/genética
5.
J Biomech Eng ; 141(8)2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-30917195

RESUMO

Arterial stiffening is a hallmark of aging, but how aging affects the arterial response to pressure is still not completely understood, especially with regard to specific matrix metalloproteinases (MMPs). Here, we performed biaxial inflation-extension tests on C57BL/6 mice to study the effects of age and MMP12, a major arterial elastase, on arterial biomechanics. Aging from 2 to 24 months leads to both circumferential and axial stiffening with stretch, and these changes are associated with an increased wall thickness, a decreased inner radius-wall thickness ratio, and a decreased in vivo axial stretch. Analysis of in vivo stretch and stress-stretch curves with arteries from age- and sex-matched wild-type (WT) and MMP12-null arteries demonstrates that MMP12 deletion attenuates age-dependent arterial stiffening, mostly in the axial direction. MMP12 deletion also prevents the aging-associated decrease in the in vivo stretch and, in general, leads to an axial mechanics phenotype characteristic of much younger mice. Circumferential arterial mechanics were much less affected by deletion of MMP12. We conclude that the induction of MMP12 during aging preferentially promotes axial arterial stiffening.

6.
Biophys J ; 114(6): 1467-1476, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29590603

RESUMO

We show that substrates with nonzero Gaussian curvature influence the organization of stress fibers and direct the migration of cells. To study the role of Gaussian curvature, we developed a sphere-with-skirt surface in which a positive Gaussian curvature spherical cap is seamlessly surrounded by a negative Gaussian curvature draping skirt, both with principal radii similar to cell-length scales. We find significant reconfiguration of two subpopulations of stress fibers when fibroblasts are exposed to these curvatures. Apical stress fibers in cells on skirts align in the radial direction and avoid bending by forming chords across the concave gap, whereas basal stress fibers bend along the convex direction. Cell migration is also strongly influenced by the Gaussian curvature. Real-time imaging shows that cells migrating on skirts repolarize to establish a leading edge in the azimuthal direction. Thereafter, they migrate in that direction. This behavior is notably different from migration on planar surfaces, in which cells typically migrate in the same direction as the apical stress fiber orientation. Thus, this platform reveals that nonzero Gaussian curvature not only affects the positioning of cells and alignment of stress fiber subpopulations but also directs migration in a manner fundamentally distinct from that of migration on planar surfaces.


Assuntos
Movimento Celular , Fibras de Estresse/metabolismo , Animais , Linhagem Celular , Polaridade Celular , Camundongos , Distribuição Normal
7.
J Cell Sci ; 129(6): 1093-100, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26919980

RESUMO

Cadherins and integrins are intrinsically linked through the actin cytoskeleton and share common signaling molecules. Although mechanosensing by the integrin-actin axis has long been appreciated, a growing body of literature now demonstrates that cadherins also transduce and respond to mechanical forces. Mounting evidence shows that mechanically driven crosstalk between integrins and cadherins regulates the spatial distribution of these receptors, their signaling intermediates, the actin cytoskeleton and intracellular forces. This interplay between integrins and cadherins can control fibronectin matrix assembly and signaling, and a fine balance between traction forces at focal adhesions and intercellular tension at adherens junctions is crucial for directional collective cell migration. In this Commentary, we discuss two central ideas: (1) how the dynamic interplay between integrins and cadherins regulates the spatial organization of intracellular signals and the extracellular matrix, and (2) the emerging consensus that intracellular force is a central mechanism that dictates cell behavior, guides tissue development and ultimately drives physiology.


Assuntos
Caderinas/metabolismo , Células/metabolismo , Integrinas/metabolismo , Transdução de Sinais , Animais , Caderinas/genética , Movimento Celular , Células/química , Células/citologia , Humanos , Integrinas/genética
8.
Soft Matter ; 14(33): 6867-6874, 2018 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-30079410

RESUMO

Boundaries play an important role in the emergence of nematic order in classical liquid crystal systems; we explore their importance in adhesive cells that form active nematics. In particular, we study how cells are affected by an edge, which in our experiments is a boundary between adhesive and non-adhesive domains on a planar surface. We find that such edges induce elongation and direct the migration of isolated fibroblasts. In confluent monolayers, these elongated cells co-align and migrate to form an active, two-dimensional nematic structure in which edges enforce planar alignment and provide local slip to streams of cells that move along them. On an adhesive square island of dimensions 1 mm × 1 mm, cells near the edges in confluent nematic monolayers have enhanced alignment and velocity. The corners of the adhesive island seed defects with signs that depend on the direction of the motion of the streams of cells that meet there. Distortions emerge with rotations of -π/2 to form a -1/4 defect for streams that move clockwise or counterclockwise, and +π/2 to form a +1/4 defect for converging streams. We explore how cells transmit alignment information to each other in the absence of an edge by studying cell pairs and find that while such pairs do co-align, this alignment is only transient and short lived. These results shed light on the importance of edges in imposing nematic order in confluent monolayers and how edges can be used as tools to pattern the long-range organization of cells for tissue engineering applications.

9.
Nat Mater ; 15(12): 1297-1306, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27525568

RESUMO

During mesenchymal development, the microenvironment gradually transitions from one that is rich in cell-cell interactions to one that is dominated by cell-ECM (extracellular matrix) interactions. Because these cues cannot readily be decoupled in vitro or in vivo, how they converge to regulate mesenchymal stem cell (MSC) mechanosensing is not fully understood. Here, we show that a hyaluronic acid hydrogel system enables, across a physiological range of ECM stiffness, the independent co-presentation of the HAVDI adhesive motif from the EC1 domain of N-cadherin and the RGD adhesive motif from fibronectin. Decoupled presentation of these cues revealed that HAVDI ligation (at constant RGD ligation) reduced the contractile state and thereby nuclear YAP/TAZ localization in MSCs, resulting in altered interpretation of ECM stiffness and subsequent changes in downstream cell proliferation and differentiation. Our findings reveal that, in an evolving developmental context, HAVDI/N-cadherin interactions can alter stem cell perception of the stiffening extracellular microenvironment.


Assuntos
Caderinas/metabolismo , Adesão Celular , Fenômenos Mecânicos , Células-Tronco Mesenquimais/citologia , Animais , Fenômenos Biomecânicos , Bovinos , Adesão Celular/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Metilação
10.
Circ Res ; 112(3): 432-40, 2013 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-23250985

RESUMO

RATIONALE: Human genetics have implicated the 5-lipoxygenase enzyme in the pathogenesis of cardiovascular disease, and an inhibitor of the 5-lipoxygenase activating protein (FLAP) is in clinical development for asthma. OBJECTIVE: Here we determined whether FLAP deletion modifies the response to vascular injury. METHODS AND RESULTS: Vascular remodeling was characterized 4 weeks after femoral arterial injury in FLAP knockout mice and wild-type controls. Both neointimal hyperplasia and the intima/media ratio of the injured artery were significantly reduced in the FLAP knockouts, whereas endothelial integrity was preserved. Lesional myeloid cells were depleted and vascular smooth muscle cell (VSMC) proliferation, as reflected by bromodeoxyuridine incorporation, was markedly attenuated by FLAP deletion. Inflammatory cytokine release from FLAP knockout macrophages was depressed, and their restricted ability to induce VSMC migration ex vivo was rescued with leukotriene B(4). FLAP deletion restrained injury and attenuated upregulation of the extracellular matrix protein, tenascin C, which affords a scaffold for VSMC migration. Correspondingly, the phenotypic modulation of VSMC to a more synthetic phenotype, reflected by morphological change, loss of α-smooth muscle cell actin, and upregulation of vascular cell adhesion molecule-1 was also suppressed in FLAP knockout mice. Transplantation of FLAP-replete myeloid cells rescued the proliferative response to vascular injury. CONCLUSIONS: Expression of lesional FLAP in myeloid cells promotes leukotriene B(4)-dependent VSMC phenotypic modulation, intimal migration, and proliferation.


Assuntos
Proteínas Ativadoras de 5-Lipoxigenase/metabolismo , Movimento Celular , Proliferação de Células , Músculo Liso Vascular/enzimologia , Células Mieloides/enzimologia , Miócitos de Músculo Liso/enzimologia , Lesões do Sistema Vascular/prevenção & controle , Proteínas Ativadoras de 5-Lipoxigenase/deficiência , Proteínas Ativadoras de 5-Lipoxigenase/genética , Animais , Transplante de Medula Óssea , Células Cultivadas , Cisteína/metabolismo , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Artéria Femoral/enzimologia , Artéria Femoral/lesões , Artéria Femoral/patologia , Genótipo , Hiperplasia , Mediadores da Inflamação/metabolismo , Leucotrieno B4/metabolismo , Leucotrienos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/imunologia , Músculo Liso Vascular/lesões , Músculo Liso Vascular/patologia , Células Mieloides/imunologia , Células Mieloides/transplante , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/patologia , Neointima , Fenótipo , Tenascina/metabolismo , Fatores de Tempo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Lesões do Sistema Vascular/enzimologia , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/imunologia , Lesões do Sistema Vascular/patologia
11.
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
12.
Differentiation ; 86(3): 112-20, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23969122

RESUMO

Changes in tissue stiffness are frequently associated with diseases such as cancer, fibrosis, and atherosclerosis. Several recent studies suggest that, in addition to resulting from pathology, mechanical changes may play a role akin to soluble factors in causing the progression of disease, and similar mechanical control might be essential for normal tissue development and homeostasis. Many cell types alter their structure and function in response to exogenous forces or as a function of the mechanical properties of the materials to which they adhere. This review summarizes recent progress in identifying intracellular signaling pathways, and especially transcriptional programs, that are differentially activated when cells adhere to materials with different mechanical properties or when they are subject to tension arising from external forces. Several cytoplasmic or cytoskeletal signaling pathways involving small GTPases, focal adhesion kinase and transforming growth factor beta as well as the transcriptional regulators MRTF-A, NFκB, and Yap/Taz have emerged as important mediators of mechanical signaling.


Assuntos
Fibroblastos/metabolismo , Mecanotransdução Celular , Estresse Mecânico , Fatores de Transcrição/metabolismo , Transcrição Gênica , Animais , Citoesqueleto/metabolismo , Adesões Focais/metabolismo , Humanos , Fatores de Transcrição/genética
13.
Nat Rev Cancer ; 4(12): 948-55, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15573116

RESUMO

Cyclin-dependent kinase inhibitors (CKIs) are well known inhibitors of cell proliferation. Their activity is disrupted in many tumour types. Recent studies show that some of these proteins have interesting alternative functions, acting in the cytoplasm to regulate Rho signalling and thereby controlling cytoskeletal organization and cell migration. The upregulation of CKIs in the cytoplasm of many cancer cells indicates that although loss of nuclear CKIs is important for cancer cell proliferation, gain of cytoplasmic CKI function might be involved in tumour invasion and metastasis.


Assuntos
Quinases Ciclina-Dependentes/antagonistas & inibidores , Citoesqueleto/patologia , Inibidores Enzimáticos/farmacologia , Neoplasias/patologia , Divisão Celular , Citoesqueleto/efeitos dos fármacos , Humanos , Neoplasias/enzimologia , Transdução de Sinais , Proteínas rho de Ligação ao GTP/metabolismo
14.
bioRxiv ; 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-38014108

RESUMO

Stiffening of the extracellular matrix (ECM) occurs after vascular injury and contributes to the injury-associated proliferation of vascular smooth muscle cells (SMCs). ECM stiffness also activates Rac-GTP, and SMC Rac1 deletion strongly reduces the proliferative response to injury in vivo . However, ECM stiffening and Rac can affect SMC differentiation, which, in itself, can influence ECM stiffness and proliferation. Here, we used pressure myography and immunofluorescence analysis of mouse carotid arteries to ask if the reported effect of Rac1 deletion on in vivo SMC proliferation might be secondary to a Rac effect on basal arterial stiffness or SMC differentiation. The results show that Rac1 deletion does not affect the abundance of arterial collagen-I, -III, or -V, the integrity of arterial elastin, or the arterial responses to pressure, including the axial and circumferential stretch-strain relationships that are assessments of arterial stiffness. Medial abundance of alpha-smooth muscle actin and smooth muscle-myosin heavy chain, markers of the SMC differentiated phenotype, were not statistically different in carotid arteries containing or deficient in Rac1. Nor did Rac1 deficiency have a statistically significant effect on carotid artery contraction to KCl. Overall, these data argue that the inhibitory effect of Rac1 deletion on in vivo SMC proliferation reflects a primary effect of Rac1 signaling to the cell cycle rather than a secondary effect associated with altered SMC differentiation or arterial stiffness.

15.
J Clin Invest ; 133(24)2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-37824206

RESUMO

Metabolic syndrome, today affecting more than 20% of the US population, is a group of 5 conditions that often coexist and that strongly predispose to cardiovascular disease. How these conditions are linked mechanistically remains unclear, especially two of these: obesity and elevated blood pressure. Here, we show that high fat consumption in mice leads to the accumulation of lipid droplets in endothelial cells throughout the organism and that lipid droplet accumulation in endothelium suppresses endothelial nitric oxide synthase (eNOS), reduces NO production, elevates blood pressure, and accelerates atherosclerosis. Mechanistically, the accumulation of lipid droplets destabilizes eNOS mRNA and activates an endothelial inflammatory signaling cascade that suppresses eNOS and NO production. Pharmacological prevention of lipid droplet formation reverses the suppression of NO production in cell culture and in vivo and blunts blood pressure elevation in response to a high-fat diet. These results highlight lipid droplets as a critical and unappreciated component of endothelial cell biology, explain how lipids increase blood pressure acutely, and provide a mechanistic account for the epidemiological link between obesity and elevated blood pressure.


Assuntos
Hipertensão , Gotículas Lipídicas , Óxido Nítrico Sintase Tipo III , Animais , Camundongos , Pressão Sanguínea , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Hipertensão/metabolismo , Gotículas Lipídicas/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Obesidade/metabolismo , Dieta Hiperlipídica/efeitos adversos
16.
Circulation ; 123(6): 631-9, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21282500

RESUMO

BACKGROUND: Microsomal (m) prostaglandin (PG) E2 synthase (S)-1 catalyzes the formation of PGE2 from PGH2, a cyclooxygenase product that is derived from arachidonic acid. Previous studies in mice suggest that targeting mPGES-1 may be less likely to cause hypertension or thrombosis than cyclooxygenase-2-selective inhibition or deletion in vivo. Indeed, deletion of mPGES-1 retards atherogenesis and angiotensin II-induced aortic aneurysm formation. The role of mPGES-1 in the response to vascular injury is unknown. METHODS AND RESULTS: Mice were subjected to wire injury of the femoral artery. Both neointimal area and vascular stenosis were significantly reduced 4 weeks after injury in mPGES-1 knockout mice compared with wild-type controls (65.6 ± 5.7 versus 37.7 ± 5.1 × 10³ pixel area and 70.5 ± 13.4% versus 47.7 ± 17.4%, respectively; P < 0.01). Induction of tenascin-C, a proproliferative and promigratory extracellular matrix protein, after injury was attenuated in the knockouts. Consistent with in vivo rediversion of PG biosynthesis, mPGES-1-deleted vascular smooth muscle cells generated less PGE2 but more PGI2 and expressed reduced tenascin-C compared with wild-type cells. Both suppression of PGE2 and augmentation of PGI2 attenuate tenascin-C expression and vascular smooth muscle cell proliferation and migration in vitro. CONCLUSIONS: Deletion of mPGES-1 in mice attenuates neointimal hyperplasia after vascular injury, in part by regulating tenascin-C expression. This raises for consideration the therapeutic potential of mPGES-1 inhibitors as adjuvant therapy for percutaneous coronary intervention.


Assuntos
Artéria Femoral/enzimologia , Artéria Femoral/lesões , Oxirredutases Intramoleculares/metabolismo , Microssomos/enzimologia , Animais , Movimento Celular , Proliferação de Células , Constrição Patológica/enzimologia , Constrição Patológica/patologia , Dinoprostona/biossíntese , Epoprostenol/biossíntese , Oxirredutases Intramoleculares/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/enzimologia , Prostaglandina H2/metabolismo , Prostaglandina-E Sintases , Tenascina/metabolismo , Túnica Íntima/enzimologia , Túnica Íntima/lesões , Túnica Íntima/patologia
17.
J Cell Biol ; 178(5): 741-7, 2007 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-17724117

RESUMO

We describe a self-amplifying feedback loop that autoinduces Skp2 during G1 phase progression. This loop, which contains Skp2 itself, p27(kip1) (p27), cyclin E-cyclin dependent kinase 2, and the retinoblastoma protein, is closed through a newly identified, conserved E2F site in the Skp2 promoter. Interference with the loop, by knockin of a Skp2-resistant p27 mutant (p27(T187A)), delays passage through the restriction point but does not interfere with S phase entry under continuous serum stimulation. Skp2 knock down inhibits S phase entry in nontransformed mouse embryonic fibroblasts but not in human papilloma virus-E7 expressing fibroblasts. We propose that the essential role for Skp2-dependent degradation of p27 is in the formation of an autoinduction loop that selectively controls the transition to mitogen-independence, and that Skp2-dependent proteolysis may be dispensable when pocket proteins are constitutively inactivated.


Assuntos
Retroalimentação Fisiológica , Fase G1/fisiologia , Regulação da Expressão Gênica , Proteínas Quinases Associadas a Fase S/metabolismo , Animais , Células Cultivadas , Ciclina E/genética , Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina/genética , Quinase 2 Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Humanos , Camundongos , Camundongos Knockout , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/metabolismo , Interferência de RNA , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Fase S/fisiologia , Proteínas Quinases Associadas a Fase S/genética
18.
J Cell Biol ; 176(4): 535-44, 2007 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-17296798

RESUMO

High molecular weight (HMW) hyaluronan (HA) is widely distributed in the extracellular matrix, but its biological activities remain incompletely understood. We previously reported that HMW-HA binding to CD44 antagonizes mitogen-induced S-phase entry in vascular smooth muscle cells (SMCs; Cuff, C.A., D. Kothapalli, I. Azonobi, S. Chun, Y. Zhang, R. Belkin, C. Yeh, A. Secreto, R.K. Assoian, D.J. Rader, and E. Puré. 2001. J. Clin. Invest. 108:1031-1040); we now characterize the underlying molecular mechanism and document its relevance in vivo. HMW-HA inhibits the mitogen-dependent induction of cyclin D1 and down-regulation of p27(kip1) in vascular SMCs. p27(kip1) messenger RNA levels were unaffected by HMW-HA, but the expression of Skp2, the rate-limiting component of the SCF complex that degrades p27(kip1), was reduced. Rescue experiments identified cyclin D1 as the primary target of HMW-HA. Similar results were observed in fibroblasts, and these antimitogenic effects were not detected in CD44-null cells. Analysis of arteries from wild-type and CD44-null mice showed that the effects of HMW-HA/CD44 on cyclin D1 and Skp2 gene expression are detected in vivo and are associated with altered SMC proliferation after vascular injury.


Assuntos
Artérias/metabolismo , Ciclinas/metabolismo , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/metabolismo , Mesoderma/metabolismo , Músculo Liso Vascular/metabolismo , Animais , Artérias/citologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ciclina D , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Ciclinas/efeitos dos fármacos , Ciclinas/genética , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Fibroblastos/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , Humanos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/farmacologia , Ácido Hialurônico/farmacologia , Mesoderma/citologia , Mesoderma/efeitos dos fármacos , Camundongos , Camundongos Knockout , Mitógenos/metabolismo , Mitógenos/farmacologia , Músculo Liso Vascular/citologia , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Proteínas Quinases Associadas a Fase S/genética , Proteínas Quinases Associadas a Fase S/metabolismo
19.
Vasc Biol ; 4(1): 1-10, 2022 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-35441125

RESUMO

Hutchinson-Guilford Progeria syndrome (HGPS) is a rare genetic disease of premature aging and early death due to cardiovascular disease. The arteries of HGPS children and mice are pathologically stiff, and HGPS mice also display reduced arterial contractility. We recently showed that reduced contractility is an early event in HGPS and linked to an aberrantly low expression of smooth muscle myosin heavy chain (smMHC). Here, we have explored the basis for reduced smMHC abundance and asked whether it is a direct effect of progerin expression or a longer-term adaptive response. Myh11, the gene encoding for smMHC, is regulated by myocardin-related transcription factors (MRTFs), and we show that HGPS aortas have a reduced MRTF signature. Additionally, smooth muscle cells (SMCs) isolated from HGPS mice display reduced MRTF nuclear localization. Acute progerin expression in WT SMCs phenocopied both the decrease in MRTF nuclear localization and expression of Myh11 seen in HGPS. Interestingly, RNA-mediated depletion of MRTF-A in WT SMCs reproduced the preferential inhibitory effect of progerin on Myh11 mRNA relative to Acta2 mRNA. Our results show that progerin expression acutely disrupts MRTF localization to the nucleus and suggest that the consequent decrease in nuclear coactivator activity can help to explain the reduction in smMHC abundance and SMC contractility seen in HGPS.

20.
Vasc Biol ; 4(1): 28-39, 2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-36222505

RESUMO

Arterial stiffening is a hallmark of aging and cardiovascular disease. While it is well established that vascular smooth muscle cells (SMCs) contribute to arterial stiffness by synthesizing and remodeling the arterial extracellular matrix, the direct contributions of SMC contractility and mechanosensors to arterial stiffness, and particularly the arterial response to pressure, remain less well understood despite being a long-standing question of biomedical importance. Here, we have examined this issue by combining the use of pressure myography of intact carotid arteries, pharmacologic inhibition of contractility, and genetic deletion of SMC focal adhesion kinase (FAK). Biaxial inflation-extension tests performed at physiological pressures showed that acute inhibition of cell contractility with blebbistatin or EGTA altered vessel geometry and preferentially reduced circumferential, as opposed to axial, arterial stiffness in wild-type mice. Similarly, genetic deletion of SMC FAK, which attenuated arterial contraction to KCl, reduced vessel wall thickness and circumferential arterial stiffness in response to pressure while having minimal effect on axial mechanics. Moreover, these effects of FAK deletion were lost by treating arteries with blebbistatin or by inhibiting myosin light-chain kinase. The expression of arterial fibrillar collagens, the integrity of arterial elastin, or markers of SMC differentiation were not affected by the deletion of SMC FAK. Our results connect cell contractility and SMC FAK to the regulation of arterial wall thickness and directionally specific arterial stiffening.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA