Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(23)2023 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-38069178

RESUMO

We have previously shown that an excess of deoxycorticosterone acetate and high sodium chloride intake (DOCA/salt) in one-renin gene mice induces a high urinary Na/K ratio, hypokalemia, and cardiac and renal hypertrophy in the absence of hypertension. Dietary potassium supplementation prevents DOCA/salt-induced pathological processes. In the present study, we further study whether DOCA/salt-treated mice progressively develop chronic inflammation and fibrosis in the kidney and whether dietary potassium supplementation can reduce the DOCA/salt-induced renal pathological process. Results showed that (1) long-term DOCA/salt-treated one-renin gene mice developed severe kidney injuries including tubular/vascular hypertrophy, mesangial/interstitial/perivascular fibrosis, inflammation (lymphocyte's immigration), proteinuria, and high serum creatinine in the absence of hypertension; (2) there were over-expressed mRNAs of plasminogen activator inhibitor-1 (PAI-1), fibronectin, collagen type I and III, interferon-inducible protein-10 (IP-10), monocyte chemotactic protein-1 (MCP1), transforming growth factor-ß (TGF-ß), tumor necrosis factor-alpha (TNF-α), osteopontin, Nuclear factor kappa B (NF-κB)/P65, and intercellular adhesion molecule (ICAM)-1; and (3) dietary potassium supplementation normalized urinary Na/K ratio, hypokalemia, proteinuria, and serum creatinine, reduced renal hypertrophy, inflammations, and fibrosis, and down-regulated mRNA expression of fibronectin, Col-I and III, TGF-ß, TNF-α, osteopontin, and ICAM without changes in the blood pressure. The results provide new evidence that potassium and sodium may modulate proinflammatory and fibrotic genes, leading to chronic renal lesions independent of blood pressure.


Assuntos
Acetato de Desoxicorticosterona , Glomerulonefrite , Hipertensão , Hipopotassemia , Camundongos , Animais , Pressão Sanguínea , Cloreto de Sódio/metabolismo , Fibronectinas/metabolismo , Osteopontina/metabolismo , Potássio na Dieta/metabolismo , Acetato de Desoxicorticosterona/efeitos adversos , Cloretos/metabolismo , Renina/metabolismo , Hipopotassemia/patologia , Fator de Necrose Tumoral alfa/metabolismo , Creatinina/metabolismo , Hipertensão/metabolismo , Rim/metabolismo , Cloreto de Sódio na Dieta/metabolismo , Glomerulonefrite/patologia , Inflamação/metabolismo , Suplementos Nutricionais , Fator de Crescimento Transformador beta/metabolismo , Proteinúria/metabolismo , Hipertrofia/metabolismo , Fibrose , Acetatos/metabolismo
2.
Proc Natl Acad Sci U S A ; 120(23): e2221742120, 2023 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-37252964

RESUMO

Fibrillin-1 is an extracellular matrix protein that assembles into microfibrils which provide critical functions in large blood vessels and other tissues. Mutations in the fibrillin-1 gene are associated with cardiovascular, ocular, and skeletal abnormalities in Marfan syndrome. Here, we reveal that fibrillin-1 is critical for angiogenesis which is compromised by a typical Marfan mutation. In the mouse retina vascularization model, fibrillin-1 is present in the extracellular matrix at the angiogenic front where it colocalizes with microfibril-associated glycoprotein-1, MAGP1. In Fbn1C1041G/+ mice, a model of Marfan syndrome, MAGP1 deposition is reduced, endothelial sprouting is decreased, and tip cell identity is impaired. Cell culture experiments confirmed that fibrillin-1 deficiency alters vascular endothelial growth factor-A/Notch and Smad signaling which regulate the acquisition of endothelial tip cell/stalk cell phenotypes, and we showed that modulation of MAGP1 expression impacts these pathways. Supplying the growing vasculature of Fbn1C1041G/+ mice with a recombinant C-terminal fragment of fibrillin-1 corrects all defects. Mass spectrometry analyses showed that the fibrillin-1 fragment alters the expression of various proteins including ADAMTS1, a tip cell metalloprotease and matrix-modifying enzyme. Our data establish that fibrillin-1 is a dynamic signaling platform in the regulation of cell specification and matrix remodeling at the angiogenic front and that mutant fibrillin-1-induced defects can be rescued pharmacologically using a C-terminal fragment of the protein. These findings, identify fibrillin-1, MAGP1, and ADAMTS1 in the regulation of endothelial sprouting, and contribute to our understanding of how angiogenesis is regulated. This knowledge may have critical implications for people with Marfan syndrome.


Assuntos
Fibrilina-1 , Síndrome de Marfan , Animais , Camundongos , Matriz Extracelular/metabolismo , Fibrilina-1/genética , Fibrilina-1/metabolismo , Síndrome de Marfan/genética , Síndrome de Marfan/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Methods Mol Biol ; 2608: 345-364, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36653717

RESUMO

Angiogenesis is the formation of new blood vessels from the existing vasculature. It is a fundamental process in developmental biology but also a pathological event that initiates or aggravates many diseases. In this complex multistep process, endothelial cells are activated by angiogenic stimuli; undergo specialization in response to VEGF/Notch signaling; degrade the basement membrane of the parent vessel; sprout, migrate, and proliferate to form capillary tubes that branch; and ultimately anastomose with adjacent vessels. Here we describe an assay that mimics the invasion step in vitro. Human microvascular endothelial cells are confronted by a VEGF-enriched basement membrane material in a three-dimensional environment that promotes endothelial cell sprouting, tube formation, and anastomosis. After a few hours, endothelial cells have become tip cells, and vascular sprouts can be observed by phase contrast, fluorescence, or time-lapse microscopy. Sprouting endothelial cells express tip cell markers, display podosomes and filopodia, and exhibit cell dynamics similar to those of angiogenic endothelial cells in vivo. This model provides a system that can be manipulated genetically to study physiological or pathological angiogenesis and that can be used to screen compounds for pro-/anti-angiogenic properties. In this chapter, we describe the key steps in setting up this assay.


Assuntos
Células Endoteliais , Podossomos , Humanos , Células Endoteliais/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Neovascularização Fisiológica/fisiologia , Podossomos/metabolismo , Neovascularização Patológica/metabolismo
4.
Cells ; 11(24)2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36552862

RESUMO

Tissue engineering strategies aim at characterizing and at optimizing the cellular component that is combined with biomaterials, for improved tissue regeneration. Here, we present the immunoMap of apical papilla, the native tissue from which SCAPs are derived. We characterized stem cell niches that correspond to a minority population of cells expressing Mesenchymal stromal/Stem Cell (CD90, CD105, CD146) and stemness (SSEA4 and CD49f) markers as well as endothelial cell markers (VWF, CD31). Based on the colocalization of TKS5 and cortactin markers, we detected migration-associated organelles, podosomes-like structures, in specific regions and, for the first time, in association with stem cell niches in normal tissue. From six healthy teenager volunteers, each with two teeth, we derived twelve cell banks, isolated and amplified under 21 or 3% O2. We confirmed a proliferative advantage of all banks when cultured under 3% versus 21% O2. Interestingly, telomerase activity was similar to that of the highly proliferative hiPSC cell line, but unrelated to O2 concentration. Finally, SCAPs embedded in a thixotropic hydrogel and implanted subcutaneously in immunodeficient mice were protected from cell death with a slightly greater advantage for cells preconditioned at 3% O2.


Assuntos
Células-Tronco Mesenquimais , Células-Tronco , Animais , Camundongos , Células Cultivadas , Diferenciação Celular , Oxigênio/metabolismo
5.
Int J Mol Sci ; 23(6)2022 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-35328350

RESUMO

Connexin37 (Cx37) and Cx40 form intercellular channels between endothelial cells (EC), which contribute to the regulation of the functions of vessels. We previously documented the participation of both Cx in developmental angiogenesis and have further shown that loss of Cx40 decreases the growth of different tumors. Here, we report that loss of Cx37 reduces (1) the in vitro proliferation of primary human EC; (2) the vascularization of subcutaneously implanted matrigel plugs in Cx37-/- mice or in WT using matrigel plugs supplemented with a peptide targeting Cx37 channels; (3) tumor angiogenesis; and (4) the growth of TC-1 and B16 tumors, resulting in a longer mice survival. We further document that Cx37 and Cx40 function in a collaborative manner to promote tumor growth, inasmuch as the injection of a peptide targeting Cx40 into Cx37-/- mice decreased the growth of TC-1 tumors to a larger extent than after loss of Cx37. This loss did not alter vessel perfusion, mural cells coverage and tumor hypoxia compared to tumors grown in WT mice. The data show that Cx37 is relevant for the control of EC proliferation and growth in different tumor models, suggesting that it may be a target, alone or in combination with Cx40, in the development of anti-tumoral treatments.


Assuntos
Células Endoteliais , Neoplasias , Animais , Proliferação de Células , Conexinas/genética , Células Endoteliais/fisiologia , Endotélio Vascular/patologia , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia
6.
Cell Mol Life Sci ; 79(4): 208, 2022 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-35347477

RESUMO

Angiogenesis involves cell specification orchestrated by regulatory interactions between the vascular endothelial growth factor and Notch signaling pathways. However, the role of microRNAs in these regulations remains poorly explored. Here we show that a controlled level of miR-155 is essential for proper angiogenesis. In the mouse retina angiogenesis model, antimiR-155 altered neovascularization. In vitro assays established that endogenous miR-155 is involved in podosome formation, activation of the proteolytic machinery and cell migration but not in morphogenesis. The role of miR-155 was explored using miR-155 mimics. In vivo, exposing the developing vasculature to miR-155 promoted hypersprouting, thus phenocopying defects associated with Notch deficiency. Mechanistically, miR-155 overexpression weakened Notch signaling by reducing Smad1/5 expression, leading to the formation of tip cell-like cells which did not reach full invasive capacity and became unable to undergo morphogenesis. These results identify miR-155 as a novel regulator of physiological angiogenesis and as a novel actor of pathological angiogenesis.


Assuntos
MicroRNAs , Neovascularização Fisiológica , Animais , Camundongos , MicroRNAs/metabolismo , Neovascularização Patológica/genética , Neovascularização Fisiológica/genética , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/genética
8.
Eur J Cell Biol ; 99(4): 151084, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32439218

RESUMO

Invadosomes are specialised actin-based dynamic microdomains of the plasma membrane. Their occurrence has been associated with cell adhesion, matrix degrading and mechanosensory functions that make them crucial regulators of cell migration and invasion. Monocytic, cancer cell and Src-transformed cell invadosomes have been extensively described. Less well defined are the structures which form in other cell types, i.e., non-haematopoietic and non-transformed cells, exposed to specific stimuli. We herein describe the specificities of podosomes induced in aortic endothelial cells stimulated with TGFß in vitro and in conditions that more closely resemble the in vivo situation. These podosomes display the typical architecture of monocytic podosomes. They organise into large rosette-shape superstructures where they exhibit collective dynamic behavior consisting in cycles of formation and regression. At the ultrastructural level, microfilament arrangements in individual podosomes were revealed. Oxygen levels and hemodynamic forces, which are key players in endothelial cell biology, both influence the process. In 3D environment, podosomes appear as globular structures along cellular extensions. A better characterization of endothelial podosomes has far-reaching implications in the understanding and, possibly, in the treatment of some vascular diseases.


Assuntos
Aorta/anatomia & histologia , Células Endoteliais/metabolismo , Podossomos/metabolismo , Humanos
9.
Biochim Biophys Acta Mol Cell Res ; 1866(4): 545-553, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30594495

RESUMO

Extensive in vitro studies have described podosomes as actin-based structures at the plasma membrane, connecting the cell with its extracellular matrix and endowed with multiple capabilities. Contractile actin-myosin cables assemble them into a network that constitutes a multifaceted cellular superstructure taking different forms - with common characteristics - but manifesting different properties depending on the context of study. Their morphology and their role in cell functioning and behavior are therefore now apprehended in in vivo or in vitro situations relevant to physiological processes. We focus here on three of them, namely: macrophage migration, antigen presentation by dendritic cells and endothelial cell sprouting during angiogenesis to highlight the characteristics of podosomes and their functioning shaped by the microenvironment.


Assuntos
Podossomos/fisiologia , Apresentação de Antígeno , Membrana Celular/metabolismo , Movimento Celular , Células Dendríticas/imunologia , Endotélio Vascular/fisiologia , Expressão Gênica , Macrófagos/fisiologia , Neovascularização Fisiológica , Transdução de Sinais
10.
Arterioscler Thromb Vasc Biol ; 37(11): 2136-2146, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28982669

RESUMO

OBJECTIVE: Cx40 (Connexin40) forms intercellular channels that coordinate the electric conduction in the heart and the vasomotor tone in large vessels. The protein was shown to regulate tumoral angiogenesis; however, whether Cx40 also contributes to physiological angiogenesis is still unknown. APPROACH AND RESULTS: Here, we show that Cx40 contributes to physiological angiogenesis. Genetic deletion of Cx40 leads to a reduction in vascular growth and capillary density in the neovascularization model of the mouse neonatal retina. At the angiogenic front, vessel sprouting is reduced, and the mural cells recruited along the sprouts display an altered phenotype. These alterations can be attributed to disturbed endothelial cell functions as selective reexpression of Cx40 in these cells restores normal angiogenesis. In vitro, targeting Cx40 in microvascular endothelial cells, by silencing its expression or by blocking gap junction channels, decreases their proliferation. Moreover, loss of Cx40 in these cells also increases their release of PDGF (platelet-derived growth factor) and promotes the chemoattraction of mural cells. In vivo, an intravitreal injection of a Cx40 inhibitory peptide, phenocopies the loss of Cx40 in the retinal vasculature of wild-type mice. CONCLUSIONS: Collectively, our data show that endothelial Cx40 contributes to the early stages of physiological angiogenesis in the developing retina, by regulating vessel growth and maturation. Cx40 thus represents a novel therapeutic target for treating pathological ocular angiogenesis.


Assuntos
Capilares/metabolismo , Conexinas/metabolismo , Células Endoteliais/metabolismo , Neovascularização Fisiológica , Vasos Retinianos/metabolismo , Animais , Animais Recém-Nascidos , Capilares/crescimento & desenvolvimento , Linhagem Celular , Proliferação de Células , Quimiotaxia , Conexinas/deficiência , Conexinas/genética , Regulação para Baixo , Junções Comunicantes/metabolismo , Genótipo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Interferência de RNA , Vasos Retinianos/crescimento & desenvolvimento , Transdução de Sinais , Transfecção , Proteína alfa-5 de Junções Comunicantes
11.
Cardiovasc Res ; 113(7): 805-816, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28449099

RESUMO

AIMS: Intimal hyperplasia (IH) is an abnormal response to vessel injury characterized by the dedifferentiation, migration, and proliferation of quiescent vascular smooth muscle cells (VSMC) to form a neointima layer. Vascular connexins (Cx) are involved in the pathophysiology of various vascular diseases, and Cx43, the main Cx expressed in VSMC, has been shown to promote VSMC proliferation and IH. The aim of this study was to investigate the participation of another Cx, namely Cx37, in the formation of the neointima layer. METHODS AND RESULTS: Wild-type (WT) and Cx37-deficient (Cx37-/-) C57BL/6J mice were subjected to carotid artery ligation (CAL), a model of vessel injury and IH. The neointima developed linearly in WT until 28 days post surgery. In contrast, the neointima layer was almost absent 14 days after surgery in Cx37-/- mice, and twice as more developed after 28 days compared to WT mice. This large neointima formation correlated with a two-fold increase in cell proliferation in the media and neointima regions between 14 and 28 days in Cx37-/- mice compared to WT mice. The CAL triggered Cx43 overexpression in the media and neointima layers of ligated carotids in WT mice, and selectively up-regulated Cx37 expression in the media layer, but not in the neointima layer. The de novo expression of Cx37 in human primary VSMC reduced cell proliferation and P-Akt levels, in association with lower Cx43 levels, whereas Cx43 overexpression increased P-Akt levels. CONCLUSION: The presence of Cx37 in the media layer of injured arteries restrains VSMC proliferation and limits the development of IH, presumably by interfering with the pro-proliferative effect of Cx43 and the Akt pathway.


Assuntos
Lesões das Artérias Carótidas/metabolismo , Estenose das Carótidas/metabolismo , Proliferação de Células , Conexinas/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Neointima , Idoso , Animais , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Artérias Carótidas/cirurgia , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Estenose das Carótidas/genética , Estenose das Carótidas/patologia , Células Cultivadas , Conexina 43/metabolismo , Conexinas/deficiência , Conexinas/genética , Modelos Animais de Doenças , Feminino , Humanos , Hiperplasia , Ligadura , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fatores de Tempo , Proteína alfa-4 de Junções Comunicantes
12.
Cell Rep ; 17(2): 484-500, 2016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27705796

RESUMO

During angiogenic sprouting, endothelial tip cells emerge from existing vessels in a process that requires vascular basement membrane degradation. Here, we show that F-actin/cortactin/P-Src-based matrix-degrading microdomains called podosomes contribute to this step. In vitro, VEGF-A/Notch signaling regulates the formation of functional podosomes in endothelial cells. Using a retinal neovascularization model, we demonstrate that tip cells assemble podosomes during physiological angiogenesis in vivo. In the retina, podosomes are also part of an interconnected network that surrounds large microvessels and impinges on the underlying basement membrane. Consistently, collagen-IV is scarce in podosome areas. Moreover, Notch inhibition exacerbates podosome formation and collagen-IV loss. We propose that the localized proteolytic action of podosomes on basement membrane collagen-IV facilitates endothelial cell sprouting and anastomosis within the developing vasculature. The identification of podosomes as key components of the sprouting machinery provides another opportunity to target angiogenesis therapeutically.


Assuntos
Colágeno Tipo IV/genética , Microvasos/metabolismo , Neovascularização Fisiológica/genética , Podossomos/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Actinas/genética , Animais , Membrana Basal/metabolismo , Colágeno Tipo IV/metabolismo , Cortactina/genética , Células Endoteliais/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Microvasos/crescimento & desenvolvimento , Morfogênese/genética , Neovascularização Patológica/metabolismo , Proteólise , Receptores Notch/metabolismo , Retina/crescimento & desenvolvimento , Retina/metabolismo , Transdução de Sinais/genética , Quinases da Família src/genética
13.
J Cell Sci ; 129(13): 2586-98, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27231093

RESUMO

Podosomes are dynamic cell-matrix contact structures that combine several key abilities, including adhesion, matrix degradation and mechanosensing. These actin-based cytoskeletal structures have been mostly studied in monocytic cells, but much less is known about those formed in other lineages. In this study, we characterise podosomes in capillary-derived microvascular endothelial cells. We identify two types of podosomes: constitutive podosomes that form in the absence of specific stimulation and induced podosomes that arise in response to the angiogenic factor VEGF-A. Constitutive and VEGF-A-induced podosomes share similar components but exhibit marked differences in terms of gelatinolytic activity. We also show that the extracellular matrix proteins laminin and collagen-IV are key determinants of the VEGF-A response, but neither collagen-I nor fibronectin are conducive for podosome induction. Moreover, only collagen-IV elicits the formation of proteolytically active podosomes through a mechanism involving increased Src phosphorylation, p190RhoGAP-B (also known as ARHGAP5) relocalisation and MT1-MMP (also known as MMP14) cell surface exposure at podosome sites. We hypothesise that by promoting podosome formation, VEGF-A enables endothelial cells to overcome the basement membrane barrier to allow sprouting outwards from the existing vasculature.


Assuntos
Colágeno Tipo IV/genética , Proteínas Ativadoras de GTPase/genética , Metaloproteinase 14 da Matriz/genética , Podossomos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Actinas/genética , Colágeno Tipo IV/biossíntese , Citoesqueleto/genética , Citoesqueleto/metabolismo , Células Endoteliais/metabolismo , Proteínas Ativadoras de GTPase/biossíntese , Regulação da Expressão Gênica , Humanos , Metaloproteinase 14 da Matriz/biossíntese , Fosforilação , Podossomos/genética , Proteólise , Fator A de Crescimento do Endotélio Vascular/administração & dosagem
14.
Oncotarget ; 7(12): 14015-28, 2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-26883111

RESUMO

Endothelial connexin40 (Cx40) contributes to regulate the structure and function of vessels. We have examined whether the protein also modulates the altered growth of vessels in tumor models established in control mice (WT), mice lacking Cx40 (Cx40-/-), and mice expressing the protein solely in endothelial cells (Tie2-Cx40). Tumoral angiogenesis and growth were reduced, whereas vessel perfusion, smooth muscle cell (SMC) coverage and animal survival were increased in Cx40-/- but not Tie2-Cx40 mice, revealing a critical involvement of endothelial Cx40 in transformed tissues independently of the hypertensive status of Cx40-/- mice. As a result, Cx40-/- mice bearing tumors survived significantly longer than corresponding controls, including after a cytotoxic administration. Comparable observations were made in WT mice injected with a peptide targeting Cx40, supporting the Cx40 involvement. This involvement was further confirmed in the absence of Cx40 or by peptide-inhibition of this connexin in aorta-sprouting, matrigel plug and SMC migration assays, and associated with a decreased expression of the phosphorylated form of endothelial nitric oxide synthase. The data identify Cx40 as a potential novel target in cancer treatment.


Assuntos
Vasos Sanguíneos/fisiologia , Conexinas/antagonistas & inibidores , Endotélio Vascular/metabolismo , Neoplasias Pulmonares/prevenção & controle , Melanoma Experimental/prevenção & controle , Neovascularização Patológica/prevenção & controle , Neoplasias da Bexiga Urinária/prevenção & controle , Animais , Aorta/patologia , Apoptose , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Conexinas/metabolismo , Endotélio Vascular/patologia , Feminino , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/patologia , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Invasividade Neoplásica , Perfusão , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/irrigação sanguínea , Neoplasias da Bexiga Urinária/patologia , Proteína alfa-5 de Junções Comunicantes
15.
PLoS One ; 10(9): e0138847, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26398895

RESUMO

Venous bypass grafts often fail following arterial implantation due to excessive smooth muscle cells (VSMC) proliferation and consequent intimal hyperplasia (IH). Intercellular communication mediated by Connexins (Cx) regulates differentiation, growth and proliferation in various cell types. Microarray analysis of vein grafts in a model of bilateral rabbit jugular vein graft revealed Cx43 as an early upregulated gene. Additional experiments conducted using an ex-vivo human saphenous veins perfusion system (EVPS) confirmed that Cx43 was rapidly increased in human veins subjected ex-vivo to arterial hemodynamics. Cx43 knock-down by RNA interference, or adenoviral-mediated overexpression, respectively inhibited or stimulated the proliferation of primary human VSMC in vitro. Furthermore, Cx blockade with carbenoxolone or the specific Cx43 inhibitory peptide 43gap26 prevented the burst in myointimal proliferation and IH formation in human saphenous veins. Our data demonstrated that Cx43 controls proliferation and the formation of IH after arterial engraftment.


Assuntos
Conexina 43/antagonistas & inibidores , Hiperplasia/prevenção & controle , Túnica Íntima/patologia , Enxerto Vascular , Veias/transplante , Animais , Proliferação de Células , Conexina 43/fisiologia , Humanos , Masculino , Músculo Liso Vascular/patologia , Coelhos
16.
Biomaterials ; 35(9): 2588-99, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24429385

RESUMO

The saphenous vein is the conduit of choice in bypass graft procedures. Haemodynamic factors play a major role in the development of intimal hyperplasia (IH), and subsequent bypass failure. To evaluate the potential protective effect of external reinforcement on such a failure, we developed an ex vivo model for the perfusion of segments of human saphenous veins under arterial shear stress. In veins submitted to pulsatile high pressure (mean pressure at 100 mmHg) for 3 or 7 days, the use of an external macroporous polyester mesh 1) prevented the dilatation of the vessel, 2) decreased the development of IH, 3) reduced the apoptosis of smooth muscle cells, and the subsequent fibrosis of the media layer, 4) prevented the remodelling of extracellular matrix through the up-regulation of matrix metalloproteinases (MMP-2, MMP-9) and plasminogen activator type I. The data show that, in an experimental ex vivo setting, an external scaffold decreases IH and maintains the integrity of veins exposed to arterial pressure, via increase in shear stress and decrease wall tension, that likely contribute to trigger selective molecular and cellular changes.


Assuntos
Veia Safena/patologia , Alicerces Teciduais/química , Túnica Íntima/patologia , Idoso , Idoso de 80 Anos ou mais , Caspase 3/metabolismo , Regulação para Baixo , Efrina-B2/genética , Efrina-B2/metabolismo , Feminino , Heme Oxigenase (Desciclizante)/metabolismo , Humanos , Hiperplasia , Técnicas In Vitro , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Pessoa de Meia-Idade , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Óxido Nítrico Sintase Tipo III/metabolismo , Perfusão , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Pressão , Receptor EphB4/genética , Receptor EphB4/metabolismo , Veia Safena/enzimologia , Estresse Mecânico , Fator de Crescimento Transformador beta/metabolismo
17.
Cell Physiol Biochem ; 31(1): 166-78, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23407022

RESUMO

BACKGROUND/AIMS: Smooth muscle tone is controlled by Ca(2+) signaling in the endothelial layer. Mouse endothelial cells are interconnected by gap junctions made of Connexin40 (Cx40) and Cx37, which allow the exchange of signaling molecules to coordinate their activity. Here, we investigated the role of Cx40 in the endothelial Ca(2+) signaling of the mouse aorta. METHODS: Ca(2+) imaging was performed on intact aortic endothelium from both wild type (Cx40+/+) and Connexin40-deficient (Cx40 -/-) mice. RESULTS: Acetylcholine (ACh) induced early fast and high amplitude Ca(2+) transients in a fraction of endothelial cells expressing the M3 muscarinic receptors. Inhibition of intercellular communication using carbenoxolone or octanol fully blocked the propagation of ACh-induced Ca(2+) transients toward adjacent cells in WT and Cx40-/- mice. As compared to WT, Cx40-/- mice displayed a reduced propagation of ACh-induced Ca(2+) waves, indicating that Cx40 contributes to the spreading of Ca(2+) signals. The propagation of those Ca(2+) responses was not blocked by suramin, a blocker of purinergic ATP receptors, indicating that there is no paracrine effect of ATP release on the Ca(2+) waves. CONCLUSIONS: Altogether our data show that Cx40 and Cx37 contribute to the propagation and amplification of the Ca(2+) signaling triggered by ACh in endothelial cells expressing the M3 muscarinic receptors.


Assuntos
Cálcio/metabolismo , Conexinas/metabolismo , Células Endoteliais/metabolismo , Receptor Muscarínico M3/metabolismo , Acetilcolina/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Antiulcerosos/farmacologia , Aorta/citologia , Sinalização do Cálcio/efeitos dos fármacos , Carbenoxolona/farmacologia , Comunicação Celular/efeitos dos fármacos , Células Cultivadas , Conexinas/genética , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Junções Comunicantes/metabolismo , Camundongos , Camundongos Knockout , Octanóis/farmacologia , Proteína alfa-5 de Junções Comunicantes , Proteína alfa-4 de Junções Comunicantes
18.
J Vasc Surg ; 57(5): 1371-82, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23351647

RESUMO

BACKGROUND: Human saphenous vein grafts are one of the salvage bypass conduits when endovascular procedures are not feasible or fail. Understanding the remodeling process that venous grafts undergo during exposure to arterial conditions is crucial to improve their patency, which is often compromised by intimal hyperplasia. The precise role of hemodynamic forces such as shear stress and arterial pressure in this remodeling is not fully characterized. The aim of this study was to determine the involvement of arterial shear stress and pressure on vein wall remodeling and to unravel the underlying molecular mechanisms. METHODS: An ex vivo vein support system was modified for chronic (up to 1 week), pulsatile perfusion of human saphenous veins under controlled conditions that permitted the separate control of arterial shear stress and different arterial pressure (7 mm Hg or 70 mm Hg). RESULTS: Veins perfused for 7 days under high pressure (70 mm Hg) underwent significant development of a neointima compared with veins exposed to low pressure (7 mm Hg). These structural changes were associated with altered expression of several molecular markers. Exposure to an arterial shear stress under low pressure increased the expression of matrix metalloproteinase (MMP)-2 and MMP-9 and tissue inhibitor of metalloproteinase (TIMP)-1 at the transcript, protein, and activity levels. This increase was enhanced by high pressure, which also increased TIMP-2 protein expression despite decreased levels of the cognate transcript. In contrast, the expression of plasminogen activator inhibitor-1 increased with shear stress but was not modified by pressure. Levels of the venous marker Eph-B4 were decreased under arterial shear stress, and levels of the arterial marker Ephrin-B2 were downregulated under high-pressure conditions. CONCLUSIONS: This model is a valuable tool to identify the role of hemodynamic forces and to decipher the molecular mechanisms leading to failure of human saphenous vein grafts. Under ex vivo conditions, arterial perfusion is sufficient to activate the remodeling of human veins, a change that is associated with the loss of specific vein markers. Elevation of pressure generates intimal hyperplasia, even though veins do not acquire arterial markers. CLINICAL RELEVANCE: The pathological remodeling of the venous wall, which leads to stenosis and ultimately graft failure, is the main limiting factor of human saphenous vein graft bypass. This remodeling is due to the hemodynamic adaptation of the vein to the arterial environment and cannot be prevented by conventional therapy. To develop a more targeted therapy, a better understanding of the molecular mechanisms involved in intimal hyperplasia is essential, which requires the development of ex vivo models of chronic perfusion of human veins.


Assuntos
Hemodinâmica , Veia Safena/patologia , Veia Safena/fisiopatologia , Idoso , Idoso de 80 Anos ou mais , Pressão Arterial , Fenômenos Biomecânicos , Proliferação de Células , Efrina-B2/genética , Efrina-B2/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Hiperplasia , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Pessoa de Meia-Idade , Neointima , Perfusão , Inibidor 1 de Ativador de Plasminogênio/genética , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Fluxo Pulsátil , RNA Mensageiro/metabolismo , Receptor EphB4/genética , Receptor EphB4/metabolismo , Veia Safena/metabolismo , Estresse Mecânico , Fatores de Tempo , Técnicas de Cultura de Tecidos , Inibidor Tecidual de Metaloproteinase-1/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Inibidor Tecidual de Metaloproteinase-2/genética , Inibidor Tecidual de Metaloproteinase-2/metabolismo , Grau de Desobstrução Vascular
19.
Endocr Relat Cancer ; 20(1): 137-49, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23207293

RESUMO

The acquisition of neuroendocrine (NE) characteristics by prostate cancer (PCa) cells is closely related to tumour progression and hormone resistance. The mechanisms by which NE cells influence PCa growth and progression are not fully understood. Macrophage migration inhibitory factor (MIF) is a pro-inflammatory cytokine involved in oncogenic processes, and MIF serum levels correlate with aggressiveness of PCa. Here, we investigated the regulation and the functional consequences of MIF expression during NE transdifferentiation of PCa cells. NE differentiation (NED) of LNCaP cells, initiated either by increasing intracellular levels of cAMP or by culturing cells in an androgen-depleted medium, was associated with markedly increased MIF release. Yet, intracellular MIF protein and mRNA levels and MIF gene promoter activity decreased during NED of LNCaP cells, suggesting that NED favours MIF release despite decreasing MIF synthesis. Adenoviral-mediated forced MIF expression in NE-differentiated LNCaP cells increased cell proliferation without affecting the expression of NE markers. Addition of exogenous recombinant MIF to LNCaP and PC-3 cells stimulated the AKT and ERK1/2 signalling pathways, the expression of genes involved in PCa, as well as proliferation and resistance to paclitaxel and thapsigargin-induced apoptosis. Altogether, these data provide evidence that increased MIF release during NED in PCa may facilitate cancer progression or recurrence, especially following androgen deprivation. Thus, MIF could represent an attractive target for PCa therapy.


Assuntos
Apoptose , Diferenciação Celular , Proliferação de Células , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Células Neuroendócrinas/patologia , Neoplasias da Próstata/patologia , Northern Blotting , Western Blotting , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Oxirredutases Intramoleculares/genética , Fatores Inibidores da Migração de Macrófagos/genética , Masculino , Células Neuroendócrinas/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
20.
Cell Commun Adhes ; 19(2): 25-37, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22642233

RESUMO

Intercellular Ca(2+) wave propagation between vascular smooth muscle cells (SMCs) is associated with the propagation of contraction along the vessel. Here, we characterize the involvement of gap junctions (GJs) in Ca(2+) wave propagation between SMCs at the cellular level. Gap junctional communication was assessed by the propagation of intercellular Ca(2+) waves and the transfer of Lucifer Yellow in A7r5 cells, primary rat mesenteric SMCs (pSMCs), and 6B5N cells, a clone of A7r5 cells expressing higher connexin43 (Cx43) to Cx40 ratio. Mechanical stimulation induced an intracellular Ca(2+) wave in pSMC and 6B5N cells that propagated to neighboring cells, whereas Ca(2+) waves in A7r5 cells failed to progress to neighboring cells. We demonstrate that Cx43 forms the functional GJs that are involved in mediating intercellular Ca(2+) waves and that co-expression of Cx40 with Cx43, depending on their expression ratio, may interfere with Cx43 GJ formation, thus altering junctional communication.


Assuntos
Sinalização do Cálcio , Conexina 43/metabolismo , Artérias Mesentéricas/citologia , Miócitos de Músculo Liso/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Carbenoxolona/farmacologia , Comunicação Celular , Células Cultivadas , Conexina 43/genética , Conexinas/genética , Conexinas/metabolismo , Corantes Fluorescentes/metabolismo , Junções Comunicantes/efeitos dos fármacos , Junções Comunicantes/metabolismo , Expressão Gênica , Isoquinolinas/metabolismo , Masculino , Octanóis/farmacologia , Peptídeos/farmacologia , Cultura Primária de Células , Ligação Proteica , Transporte Proteico , Ratos , Ratos Wistar , Análise de Célula Única , Proteína alfa-5 de Junções Comunicantes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA