Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Biomed Eng ; 5(12): 1437-1456, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34031559

RESUMO

The fibrotic encapsulation of implants involves the mechanical activation of myofibroblasts and of pro-fibrotic transforming growth factor beta 1 (TGF-ß1). Here, we show that both softening of the implant surfaces and inhibition of the activation of TGF-ß1 reduce the fibrotic encapsulation of subcutaneous silicone implants in mice. Conventionally stiff silicones (elastic modulus, ~2 MPa) coated with a soft silicone layer (elastic modulus, ~2 kPa) reduced collagen deposition as well as myofibroblast activation without affecting the numbers of macrophages and their polarization states. Instead, fibroblasts around stiff implants exhibited enhanced intracellular stress, increased the recruitment of αv and ß1 integrins, and activated TGF-ß1 signalling. In vitro, the recruitment of αv integrin to focal adhesions and the activation of ß1 integrin and of TGF-ß were higher in myofibroblasts grown on latency-associated peptide (LAP)-coated stiff silicones than on soft silicones. Antagonizing αv integrin binding to LAP through the small-molecule inhibitor CWHM-12 suppressed active TGF-ß signalling, myofibroblast activation and the fibrotic encapsulation of stiff subcutaneous implants in mice.


Assuntos
Próteses e Implantes , Silicones , Fator de Crescimento Transformador beta , Animais , Fibroblastos , Fibrose , Reação a Corpo Estranho , Camundongos , Miofibroblastos/patologia
2.
Cells ; 9(12)2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33348602

RESUMO

We identify the focal adhesion protein kindlin-2 as player in a novel mechanotransduction pathway that controls profibrotic cardiac fibroblast to myofibroblast activation. Kindlin-2 is co-upregulated with the myofibroblast marker α-smooth muscle actin (α-SMA) in fibrotic rat hearts and in human cardiac fibroblasts exposed to fibrosis-stiff culture substrates and pro-fibrotic TGF-ß1. Stressing fibroblasts using ferromagnetic microbeads, stretchable silicone membranes, and cell contraction agonists all result in kindlin-2 translocation to the nucleus. Overexpression of full-length kindlin-2 but not of kindlin-2 missing a putative nuclear localization sequence (∆NLS kindlin-2) results in increased α-SMA promoter activity. Downregulating kindlin-2 with siRNA leads to decreased myofibroblast contraction and reduced α-SMA expression, which is dependent on CC(A/T)-rich GG(CArG) box elements in the α-SMA promoter. Lost myofibroblast features under kindlin-2 knockdown are rescued with wild-type but not ∆NLS kindlin-2, indicating that myofibroblast control by kindlin-2 requires its nuclear translocation. Because kindlin-2 can act as a mechanotransducer regulating the transcription of α-SMA, it is a potential target to interfere with myofibroblast activation in tissue fibrosis.


Assuntos
Mecanotransdução Celular , Proteínas de Membrana/metabolismo , Miofibroblastos/metabolismo , Proteínas de Neoplasias/metabolismo , Actinas/genética , Actinas/metabolismo , Adulto , Animais , Núcleo Celular/metabolismo , Células Cultivadas , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Masculino , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Microscopia de Fluorescência , Miofibroblastos/citologia , Miofibroblastos/efeitos dos fármacos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Fator de Crescimento Transformador beta1/farmacologia
3.
Sci Signal ; 12(564)2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30647145

RESUMO

Macrophages contribute to the activation of fibroblastic cells into myofibroblasts, which secrete collagen and contract the collagen matrix to acutely repair injured tissue. Persistent myofibroblast activation leads to the accumulation of fibrotic scar tissue that impairs organ function. We investigated the key processes that turn acute beneficial repair into destructive progressive fibrosis. We showed that homotypic cadherin-11 interactions promoted the specific binding of macrophages to and persistent activation of profibrotic myofibroblasts. Cadherin-11 was highly abundant at contacts between macrophages and myofibroblasts in mouse and human fibrotic lung tissues. In attachment assays, cadherin-11 junctions mediated specific recognition and strong adhesion between macrophages and myofibroblasts. One functional outcome of cadherin-11-mediated adhesion was locally restricted activation of latent transforming growth factor-ß (TGF-ß) between macrophage-myofibroblast pairs that was not observed in cocultures of macrophages and myofibroblasts that were not in contact with one another. Our data suggest that cadherin-11 junctions maintain latent TGF-ß-producing macrophages and TGF-ß-activating myofibroblasts in close proximity to one another. Inhibition of homotypic cadherin-11 interactions could be used to cause macrophage-myofibroblast separation, thereby destabilizing the profibrotic niche.


Assuntos
Caderinas/metabolismo , Macrófagos/metabolismo , Miofibroblastos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Caderinas/genética , Adesão Celular , Células Cultivadas , Técnicas de Cocultura , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibrose , Humanos , Macrófagos/citologia , Macrófagos/ultraestrutura , Masculino , Camundongos Endogâmicos C57BL , Microscopia Eletrônica/métodos , Miofibroblastos/citologia , Ligação Proteica , Interferência de RNA , Transdução de Sinais
4.
J Am Heart Assoc ; 7(7)2018 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-29599211

RESUMO

BACKGROUND: Development of right ventricular (RV) hypertension eventually contributes to RV and left ventricular (LV) myocardial fibrosis and dysfunction. The molecular mechanisms are not fully elucidated. METHODS AND RESULTS: Pulmonary artery banding was used to induce RV hypertension in rats in vivo. Then, we evaluated cardiac function and regional remodeling 6 weeks after pulmonary artery banding. To further elucidate mechanisms responsible for regional cardiac remodeling, we also mimicked RV hypertensive stress by cyclic mechanical stretching applied to confluent cultures of cardiac fibroblasts, isolated from the RV free wall, septal hinge points, and LV free wall. Echocardiography and catheter evaluation demonstrated that rats in the pulmonary artery banding group developed RV hypertension with leftward septal displacement, LV compression, and increased LV end-diastolic pressures. Picrosirius red staining indicated that pulmonary artery banding induced marked RV fibrosis and dysfunction, with prominent fibrosis and elastin deposition at the septal hinge points but less LV fibrosis. These changes were associated with proportionally increased expressions of integrin-ß1 and profibrotic signaling proteins, including phosphorylated Smad2/3 and transforming growth factor-ß1. Moreover, mechanically stretched fibroblasts also expressed significantly increased levels of α-smooth muscle actin, integrin-ß1, transforming growth factor-ß1, collagen I deposition, and wrinkle formation on gel assays, consistent with myofibroblast transformation. These changes were not observed in parallel cultures of mechanically stretched fibroblasts, preincubated with the integrin inhibitor (BTT-3033). CONCLUSIONS: Experimentally induced RV hypertension triggers regional RV, hinge-point, and LV integrin ß1-dependent mechanotransduction signaling pathways that eventually trigger myocardial fibrosis via transforming growth factor-ß1 signaling. Reduced LV fibrosis and preserved global function, despite geometrical and pressure aberrations, suggest a possible elastin-mediated protective mechanism at the septal hinge points.


Assuntos
Pressão Arterial , Ventrículos do Coração/metabolismo , Hipertensão Pulmonar/complicações , Hipertrofia Ventricular Esquerda/etiologia , Hipertrofia Ventricular Direita/etiologia , Integrina beta1/metabolismo , Artéria Pulmonar/fisiopatologia , Função Ventricular Esquerda , Função Ventricular Direita , Remodelação Ventricular , Animais , Células Cultivadas , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Elastina/metabolismo , Fibrose , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/patologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Hipertrofia Ventricular Direita/metabolismo , Hipertrofia Ventricular Direita/patologia , Hipertrofia Ventricular Direita/fisiopatologia , Masculino , Mecanotransdução Celular , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta1/metabolismo
5.
J Cell Sci ; 131(5)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29361522

RESUMO

Dysregulated secretion and extracellular activation of TGF-ß1 stimulates myofibroblasts to accumulate disordered and stiff extracellular matrix (ECM) leading to fibrosis. Fibronectin immobilizes latent TGF-ß-binding protein-1 (LTBP-1) and thus stores TGF-ß1 in the ECM. Because the ED-A fibronectin splice variant is prominently expressed during fibrosis and supports myofibroblast activation, we investigated whether ED-A promotes LTBP-1-fibronectin interactions. Using stiffness-tuneable substrates for human dermal fibroblast cultures, we showed that high ECM stiffness promotes expression and colocalization of LTBP-1 and ED-A-containing fibronectin. When rescuing fibronectin-depleted fibroblasts with specific fibronectin splice variants, LTBP-1 bound more efficiently to ED-A-containing fibronectin than to ED-B-containing fibronectin and fibronectin lacking splice domains. Function blocking of the ED-A domain using antibodies and competitive peptides resulted in reduced LTBP-1 binding to ED-A-containing fibronectin, reduced LTBP-1 incorporation into the fibroblast ECM and reduced TGF-ß1 activation. Similar results were obtained by blocking the heparin-binding stretch FNIII12-13-14 (HepII), adjacent to the ED-A domain in fibronectin. Collectively, our results suggest that the ED-A domain enhances association of the latent TGF-ß1 by promoting weak direct binding to LTBP-1 and by enhancing heparin-mediated protein interactions through HepII in fibronectin.


Assuntos
Fibronectinas/genética , Fibrose/genética , Proteínas de Ligação a TGF-beta Latente/genética , Fator de Crescimento Transformador beta1/genética , Animais , Proteínas de Transporte , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fibronectinas/química , Fibrose/patologia , Células HEK293 , Humanos , Proteínas de Ligação a TGF-beta Latente/química , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Ligação Proteica/genética , Domínios Proteicos/genética , Isoformas de Proteínas/genética , Ratos
6.
Am J Transl Res ; 9(6): 2723-2735, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28670364

RESUMO

Lipocalin-2 (also known as NGAL) levels are elevated in obesity and diabetes yet relatively little is known regarding effects on the heart. We induced pressure overload (PO) in mice and found that lipocalin-2 knockout (LKO) mice exhibited less PO-induced autophagy and NLRP3 inflammasome activation than Wt. PO-induced mitochondrial damage was reduced and autophagic flux greater in LKO mice, which correlated with less cardiac dysfunction. All of these observations were negated upon adenoviral-mediated restoration of normal lipocalin-2 levels in LKO. Studies in primary cardiac fibroblasts indicated that lipocalin-2 enhanced priming and activation of NLRP3-inflammasome, detected by increased IL-1ß, IL-18 and Caspase-1 activation. This was attenuated in cells isolated from NLRP3-deficient mice or upon pharmacological inhibition of NLRP3. Furthermore, lipocalin-2 induced release of HMGB1 from cells and NLRP3-inflammasome activation was attenuated by TLR4 inhibition. We also found evidence of increased inflammasome activation and reduced autophagy in cardiac biopsy samples from heart failure patients. Overall, this study provides new mechanistic insight on the detrimental role of lipocalin-2 in the development of cardiac dysfunction.

7.
Cell Rep ; 18(7): 1774-1790, 2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28199848

RESUMO

Discoidin domain receptor 1 (DDR1) is a tyrosine kinase collagen adhesion receptor that mediates cell migration through association with non-muscle myosin IIA (NMIIA). Because DDR1 is implicated in cancer fibrosis, we hypothesized that DDR1 interacts with NMIIA to enable collagen compaction by traction forces. Mechanical splinting of rat dermal wounds increased DDR1 expression and collagen alignment. In periodontal ligament of DDR1 knockout mice, collagen mechanical reorganization was reduced >30%. Similarly, cultured cells with DDR1 knockdown or expressing kinase-deficient DDR1d showed 50% reduction of aligned collagen. Tractional remodeling of collagen was dependent on DDR1 clustering, activation, and interaction of the DDR1 C-terminal kinase domain with NMIIA filaments. Collagen remodeling by traction forces, DDR1 tyrosine phosphorylation, and myosin light chain phosphorylation were increased on stiff versus soft substrates. Thus, DDR1 clustering, activation, and interaction with NMIIA filaments enhance the collagen tractional remodeling that is important for collagen compaction in fibrosis.


Assuntos
Colágeno/metabolismo , Receptor com Domínio Discoidina 1/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Animais , Adesão Celular/fisiologia , Movimento Celular/fisiologia , Feminino , Fibrose/metabolismo , Camundongos , Camundongos Knockout , Cadeias Leves de Miosina/metabolismo , Fosforilação/fisiologia , Ratos , Ratos Wistar , Receptores Proteína Tirosina Quinases/metabolismo
8.
Nat Mater ; 16(3): 379-389, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27798620

RESUMO

Expansion on stiff culture substrates activates pro-fibrotic cell programs that are retained by mechanical memory. Here, we show that priming on physiologically soft silicone substrates suppresses fibrogenesis and desensitizes mesenchymal stem cells (MSCs) against subsequent mechanical activation in vitro and in vivo, and identify the microRNA miR-21 as a long-term memory keeper of the fibrogenic program in MSCs. During stiff priming, miR-21 levels were gradually increased by continued regulation through the acutely mechanosensitive myocardin-related transcription factor-A (MRTF-A/MLK-1) and remained high over 2 weeks after removal of the mechanical stimulus. Knocking down miR-21 once by the end of the stiff-priming period was sufficient to erase the mechanical memory and sensitize MSCs to subsequent exposure to soft substrates. Soft priming and erasing mechanical memory following cell culture expansion protects MSCs from fibrogenesis in the host wound environment and increases the chances for success of MSC therapy in tissue-repair applications.


Assuntos
Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Estresse Mecânico , Fatores de Transcrição/metabolismo , Animais , Fibrose , Células-Tronco Mesenquimais/patologia , MicroRNAs/genética , Ratos , Ratos Wistar , Fatores de Transcrição/genética
9.
Int J Cardiol ; 216: 32-42, 2016 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-27140334

RESUMO

BACKGROUND: The cardiac remodelling process in advanced heart failure due to pressure overload has not been clearly defined but likely involves mechanisms of cardiac fibrosis and cardiomyocyte hypertrophy. The aim of this study was to examine pressure overload (PO)-induced cardiac remodelling processes and their reversibility after unloading in both humans with heart failure and a mouse model of PO induced by aortic constriction. METHODS & RESULTS: Speckle tracking echocardiography showed PO-induced cardiac dysfunction in mice was reversible after removal of aortic constriction to unload. Masson's Trichrome staining suggested that PO-induced myocardial fibrosis was reversible, however detailed analysis of 3-dimensional collagen architecture by scanning electron microscopy demonstrated that matrix remodelling was not completely normalised as a disorganised network of thin collagen fibres was evident. Analysis of human left ventricular biopsy samples from HF patients revealed increased presence of large collagen fibres which were greatly reduced in paired samples from the same individuals after unloading by left ventricular assist device implantation. Again, an extensive network of small collagen fibres was still clearly seen to closely surround cardiomyocytes after unloading. Other features of PO-induced remodelling including increased myofibroblast content, cardiomyocyte disarray and hypertrophy were largely reversed upon unloading in both humans and mouse model. Previous work in humans demonstrated that receptors for adiponectin, an important mediator of cardiac fibrosis and hypertrophy, decreased in heart failure patients and returned to normal after unloading. Here we provide novel data showing a similar trend for adiponectin receptor adaptor protein APPL1, but not APPL2 isoform. CONCLUSIONS: LV unloading diminishes PO-induced cardiac remodelling and improves function. These findings add new insights into the cardiac remodelling process, and provide novel targets for future pharmacologic therapies.


Assuntos
Estenose da Valva Aórtica/complicações , Cardiomiopatia Hipertrófica/diagnóstico por imagem , Colágeno/ultraestrutura , Insuficiência Cardíaca/patologia , Ventrículos do Coração/diagnóstico por imagem , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Cardiomiopatia Hipertrófica/etiologia , Cardiomiopatia Hipertrófica/metabolismo , Modelos Animais de Doenças , Ecocardiografia , Ventrículos do Coração/metabolismo , Humanos , Masculino , Camundongos , Microscopia Eletrônica de Varredura , Remodelação Ventricular
10.
J Am Soc Nephrol ; 27(10): 3117-3128, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26961347

RESUMO

Like many organs, the kidney stiffens after injury, a process that is increasingly recognized as an important driver of fibrogenesis. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are related mechanosensory proteins that bind to Smad transcription factors, the canonical mediators of profibrotic TGF-ß responses. Here, we investigated the role of YAP/TAZ in the matrix stiffness dependence of fibroblast responses to TGF-ß In contrast to growth on a stiff surface, fibroblast growth on a soft matrix led to YAP/TAZ sequestration in the cytosol and impaired TGF-ß-induced Smad2/3 nuclear accumulation and transcriptional activity. YAP knockdown or treatment with verteporfin, a drug that was recently identified as a potent YAP inhibitor, elicited similar changes. Furthermore, verteporfin reduced YAP/TAZ levels and decreased the total cellular levels of Smad2/3 after TGF-ß stimulation. Verteporfin treatment of mice subjected to unilateral ureteral obstruction similarly reduced YAP/TAZ levels and nuclear Smad accumulation in the kidney, and attenuated renal fibrosis. Our data suggest that organ stiffening cooperates with TGF-ß to induce fibrosis in a YAP/TAZ- and Smad2/3-dependent manner. Interference with this YAP/TAZ and TGF-ß/Smad crosstalk likely underlies the antifibrotic activity of verteporfin. Finally, through repurposing of a clinically used drug, we illustrate the therapeutic potential of a novel mechanointerference strategy that blocks TGF-ß signaling and renal fibrogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Rim/patologia , Fosfoproteínas/fisiologia , Proteína Smad2/fisiologia , Proteína Smad3/fisiologia , Fatores de Transcrição/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Aciltransferases , Animais , Proteínas de Ciclo Celular , Fibrose/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Proteínas de Sinalização YAP
11.
Am J Pathol ; 185(12): 3326-37, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26458763

RESUMO

Dupuytren disease is a fibrotic disorder characterized by contraction of myofibroblast-rich cords and nodules in the hands. The Hippo member Yes-associated protein 1 (YAP1) is activated by tissue stiffness and the profibrotic transforming growth factor-ß1, but its role in cell fibrogenesis is yet unclear. We hypothesized that YAP1 regulates the differentiation of dermal fibroblasts into highly contractile myofibroblasts and that YAP1 governs the maintenance of a myofibroblast phenotype in primary Dupuytren cells. Knockdown of YAP1 in transforming growth factor-ß1-stimulated dermal fibroblasts decreased the formation of contractile smooth muscle α-actin stress fibers and the deposition of collagen type I, which are hallmark features of myofibroblasts. Translating our findings to a clinically relevant model, we found that YAP1 deficiency in Dupuytren disease myofibroblasts resulted in decreased expression of ACTA2, COL1A1, and CCN2 mRNA, but this did not result in decreased protein levels. YAP1-deficient Dupuytren myofibroblasts showed decreased contraction of a collagen hydrogel. Finally, we showed that YAP1 levels and nuclear localization were elevated in affected Dupuytren disease tissue compared with matched control tissue and partly co-localized with smooth muscle α-actin-positive cells. In conclusion, our data show that YAP1 is a regulator of myofibroblast differentiation and contributes to the maintenance of a synthetic and contractile phenotype, in both transforming growth factor-ß1-induced myofibroblast differentiation and primary Dupuytren myofibroblasts.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Contratura de Dupuytren/patologia , Miofibroblastos/patologia , Fosfoproteínas/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Colágeno/metabolismo , Contratura de Dupuytren/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Miofibroblastos/efeitos dos fármacos , Fosfoproteínas/genética , Fatores de Transcrição , Fator de Crescimento Transformador beta1/farmacologia , Proteínas de Sinalização YAP
12.
PLoS One ; 10(4): e0121049, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25910275

RESUMO

Adiponectin, circulating levels of which are reduced in obesity and diabetes, mediates cardiac extracellular matrix (ECM) remodeling in response to pressure overload (PO). Here, we performed a detailed temporal analysis of progressive cardiac ECM remodelling in adiponectin knockout (AdKO) and wild-type (WT) mice at 3 days and 1, 2, 3 and 4 weeks following the induction of mild PO via minimally invasive transverse aortic banding. We first observed that myocardial adiponectin gene expression was reduced after 4 weeks of PO, whereas increased adiponectin levels were detected in cardiac homogenates at this time despite decreased circulating levels of adiponectin. Scanning electron microscopy and Masson's trichrome staining showed collagen accumulation increased in response to 2 and 4 weeks of PO in WT mice, while fibrosis in AdKO mice was notably absent after 2 weeks but highly apparent after 4 weeks of PO. Time and intensity of fibroblast appearance after PO was not significantly different between AdKO and WT animals. Gene array analysis indicated that MMP2, TIMP2, collagen 1α1 and collagen 1α3 were induced after 2 weeks of PO in WT but not AdKO mice. After 4 weeks MMP8 was induced in both genotypes, MMP9 only in WT mice and MMP1α only in AdKO mice. Direct stimulation of primary cardiac fibroblasts with adiponectin induced a transient increase in total collagen detected by picrosirius red staining and collagen III levels synthesis, as well as enhanced MMP2 activity detected via gelatin zymography. Adiponectin also enhanced fibroblast migration and attenuated angiotensin-II induced differentiation to a myofibroblast phenotype. In conclusion, these data indicate that increased myocardial bioavailability of adiponectin mediates ECM remodeling following PO and that adiponectin deficiency delays these effects.


Assuntos
Adiponectina/genética , Matriz Extracelular/metabolismo , Miocárdio/metabolismo , Remodelação Ventricular/genética , Adiponectina/deficiência , Adiponectina/metabolismo , Animais , Colágeno/genética , Colágeno/metabolismo , Modelos Animais de Doenças , Matriz Extracelular/genética , Fibrose , Expressão Gênica , Masculino , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Knockout , Miocárdio/patologia , Miocárdio/ultraestrutura , Ratos , Inibidores Teciduais de Metaloproteinases/genética , Inibidores Teciduais de Metaloproteinases/metabolismo
13.
Biomaterials ; 54: 136-47, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25907047

RESUMO

Over the past decade, various implantable devices have been developed to treat diseases that were previously difficult to manage such diabetes, chronic pain, and neurodegenerative disorders. However, translation of these novel technologies into clinical practice is often difficult because fibrotic encapsulation and/or rejection impairs device function after body implantation. Ideally, cells of the host tissue should perceive the surface of the implant being similar to the normal extracellular matrix. Here, we developed an innovative approach to provide implant surfaces with adhesive protein micropatterns. The patterns were designed to promote adhesion of fibroblasts and macrophages by simultaneously suppressing fibrogenic activation of both cell types. In a rat model, subcutaneously implanted silicone pads provided with the novel micropatterns caused 6-fold lower formation of inflammatory giant cells compared with clinical grade, uncoated, or collagen-coated silicone implants. We further show that micropatterning of implants resulted in 2-3-fold reduced numbers of pro-fibrotic myofibroblast by inhibiting their mechanical activation. Our novel approach allows controlled cell attachment to implant surfaces, representing a critical advance for enhanced biointegration of implantable medical devices.


Assuntos
Fibrose/etiologia , Fibrose/prevenção & controle , Próteses e Implantes/efeitos adversos , Silício/efeitos adversos , Silício/química , Animais , Análise de Falha de Equipamento , Fibrose/patologia , Masculino , Desenho de Prótese , Ratos , Ratos Wistar , Propriedades de Superfície
14.
J Invest Dermatol ; 135(3): 885-894, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25268583

RESUMO

Cutaneous responses to injury, infection, and tumor formation involve the activation of resident dermal fibroblasts and subsequent transition to myofibroblasts. The key for induction of myofibroblast differentiation is the activation of transforming growth factor-ß (TGF-ß) receptors and stimulation of integrins and their associated proteins, including integrin-linked kinase (ILK). Cross-talk processes between TGF-ß and ILK are crucial for myofibroblast formation, as ILK-deficient dermal fibroblasts exhibit impaired responses to TGF-ß receptor stimulation. We now show that ILK associates with type II TGF-ß receptors (TßRII) in ligand- and receptor kinase activity-independent manners. In cells with targeted Ilk gene inactivation, cellular levels of TßRII are decreased, through mechanisms that involve enhanced ubiquitination and proteasomal degradation. Partitioning of TGF-ß receptors into membrane has been linked to proteasome-dependent receptor degradation. We found that interfering with membrane raft formation in ILK-deficient cells restored TßRII levels and signaling. These observations support a model whereby ILK functions in fibroblasts to direct TßRII away from degradative pathways during their differentiation into myofibroblasts.


Assuntos
Fibroblastos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Pele/metabolismo , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Fibroblastos/patologia , Camundongos , Camundongos Mutantes , Modelos Animais , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Transdução de Sinais/fisiologia , Pele/patologia
15.
J Cell Biol ; 207(2): 283-97, 2014 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-25332161

RESUMO

Integrin-mediated force application induces a conformational change in latent TGF-ß1 that leads to the release of the active form of the growth factor from the extracellular matrix (ECM). Mechanical activation of TGF-ß1 is currently understood as an acute process that depends on the contractile force of cells. However, we show that ECM remodeling, preceding the activation step, mechanically primes latent TGF-ß1 akin to loading a mechanical spring. Cell-based assays and unique strain devices were used to produce a cell-derived ECM of controlled organization and prestrain. Mechanically conditioned ECM served as a substrate to measure the efficacy of TGF-ß1 activation after cell contraction or direct force application using magnetic microbeads. The release of active TGF-ß1 was always higher from prestrained ECM as compared with unorganized and/or relaxed ECM. The finding that ECM prestrain regulates the bioavailability of TGF-ß1 is important to understand the context of diseases that involve excessive ECM remodeling, such as fibrosis or cancer.


Assuntos
Matriz Extracelular/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Diferenciação Celular , Células Cultivadas , Células HEK293 , Humanos , Integrinas/metabolismo , Integrinas/fisiologia , Mecanotransdução Celular , Miofibroblastos/citologia , Miofibroblastos/metabolismo , Ratos Wistar
16.
Adv Wound Care (New Rochelle) ; 2(5): 238-246, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24527345

RESUMO

SIGNIFICANCE: Abnormal wound repair results from disorders in granulation tissue remodeling, and can lead to hypertrophic scarring and fibrosis. Excessive scarring can compromise tissue function and decrease tissue resistance to additional injuries. The development of potential therapies to minimize scarring is, thus, necessary to address an important clinical problem. RECENT ADVANCES: It has been clearly established that multiple cytokines and growth factors participate in the regulation of cutaneous wound healing. More recently, it has become apparent that these factors do not necessarily activate isolated signaling pathways. Rather, in some cases, there is cross-modulation of several cellular pathways involved in this process. Two of the key pathways that modulate each other during wound healing are activated by transforming growth factor-ß and by extracellular matrix proteins acting through integrins. CRITICAL ISSUES: The pathogenesis of excessive scarring upon wound healing is not fully understood, as a result of the complexity of this process. However, the fact that many pathways combine to produce fibrosis provides multiple potential therapeutic targets. Some of them have been identified, such as focal adhesion kinase and integrin-linked kinase. Currently, a major challenge is to develop pharmacological inhibitors of these proteins with therapeutic value to promote efficient wound repair. FUTURE DIRECTIONS: The ability to better understand how different pathways crosstalk during wound repair and to identify and pharmacologically modulate key factors that contribute to the regulation of multiple wound-healing pathways could potentially provide effective therapeutic targets to decrease or prevent excessive scar formation and/or development of fibrosis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...