Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Methods Mol Biol ; 2299: 1-5, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34028731

RESUMO

Myofibroblasts are key cells in mediating normal wound contraction and promoting connective tissue deformations characteristic of fibrosis and scarring. Five decades ago, myofibroblasts were discovered in electron micrographs of wound granulation tissue as fibroblastic cells containing microfilaments that are organized in bundles like those present in smooth muscle. The contractile function of myofibroblasts was demonstrated by measuring the contraction of strips of granulation tissue in response to smooth muscle agonists and in cell culture. Although formation of contractile bundles already defines the myofibroblast, neo-expression of α-smooth muscle actin (α-SMA) in fibroblastic cells has become the most widely used myofibroblast marker. Because α-SMA incorporation into stress fibers mediates enhanced fibroblast contraction, it has been proposed and successfully tested as a drug target in therapeutic approaches to reduce tissue contractures. Other anti-fibrosis strategies target growth factor-, extracellular matrix-, and mechanical stress-induced pathways of myofibroblast activation from various precursors or aim to induce myofibroblast apoptosis. To understand the involved mechanisms of myofibroblast formation and function, critical experimental tools and animal models have been developed, which are made available in this collection of protocols by experts in the field.


Assuntos
Actinas/metabolismo , Miofibroblastos/fisiologia , Animais , Fibrose , Humanos , Contração Muscular , Cicatrização
3.
Front Physiol ; 10: 336, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31001134

RESUMO

Fascial tissues form a ubiquitous network throughout the whole body, which is usually regarded as a passive contributor to biomechanical behavior. We aimed to answer the question, whether fascia may possess the capacity for cellular contraction which, in turn, could play an active role in musculoskeletal mechanics. Human and rat fascial specimens from different body sites were investigated for the presence of myofibroblasts using immunohistochemical staining for α-smooth muscle actin (n = 31 donors, n = 20 animals). In addition, mechanographic force registrations were performed on isolated rat fascial tissues (n = 8 to n = 18), which had been exposed to pharmacological stimulants. The density of myofibroblasts was increased in the human lumbar fascia in comparison to fasciae from the two other regions examined in this study: fascia lata and plantar fascia [H(2) = 14.0, p < 0.01]. Mechanographic force measurements revealed contractions in response to stimulation by fetal bovine serum, the thromboxane A2 analog U46619, TGF-ß1, and mepyramine, while challenge by botulinum toxin type C3-used as a Rho kinase inhibitor- provoked relaxation (p < 0.05). In contrast, fascial tissues were insensitive to angiotensin II and caffeine (p < 0.05). A positive correlation between myofibroblast density and contractile response was found (r s = 0.83, p < 0.001). The hypothetical application of the registered forces to human lumbar tissues predicts a potential impact below the threshold for mechanical spinal stability but strong enough to possibly alter motoneuronal coordination in the lumbar region. It is concluded that tension of myofascial tissue is actively regulated by myofibroblasts with the potential to impact active musculoskeletal dynamics.

4.
Int J Cardiol ; 264: 1-6, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29776555

RESUMO

BACKGROUND: Culprit coronary atherosclerotic plaques (APs) from young sudden cardiac death (SCD) victims are mostly non-atheromatous, i.e., consisting of proliferative smooth muscle cells (SMCs). Coronary vasospasm has been advocated to explain plaque instability in the absence of thrombosis. Our aim was to characterize the SMC phenotype in the intima and media of coronary arteries from young SCD victims. METHODS AND RESULTS: A total of 38 coronary artery segments were studied: (a) 18 APs from young (≤40 years old) SCD patients, (b) 9 APs from old (>40 years old) SCD patients, (c) 11 non-atherosclerotic coronary arteries from young patients (≤40 years old). Markers of differentiated SMCs such as α-smooth muscle actin (α-SMA), smooth muscle myosin heavy chains (SMMHCs), and heavy-caldesmon (h-CaD), were assessed in intima and media by immunohistochemistry and quantified morphometrically. In the intima, their expression was higher in non-atherosclerotic arteries (44.37 ±â€¯3.03% for α-SMA, 14.21 ±â€¯2.01% for SMMHCs, 8.90 ±â€¯1.33% for h-CaD) and APs from young SCD victims (38.95 ±â€¯2.29% for α-SMA, 11.92 ±â€¯1.92% for SMMHCs, 8.93 ±â€¯1.12% for h-CaD) compared with old patients (22.01 ±â€¯3.56% for α-SMA, 6.39 ±â€¯0.7% for SMMHCs, 3.00 ±â€¯0.57% for h-CaD; all P statistically significant). The media of non-atherosclerotic arteries and APs from young SCD victims exhibited strong positivity for the differentiation markers unlike that of old patients. CONCLUSIONS: SMCs of coronary APs as well as from the underlying media from young SCD victims exhibit strong contractile phenotype. In the setting of critical stenosis, both intima and media SMC contractility might contribute to transient coronary spasm leading to myocardial ischemia and SCD.


Assuntos
Actinas/metabolismo , Proteínas de Ligação a Calmodulina/metabolismo , Vasos Coronários , Morte Súbita Cardíaca , Cadeias Pesadas de Miosina/metabolismo , Placa Aterosclerótica , Adulto , Fatores Etários , Biomarcadores/metabolismo , Vasoespasmo Coronário/metabolismo , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Vasos Coronários/fisiopatologia , Morte Súbita Cardíaca/etiologia , Morte Súbita Cardíaca/patologia , Feminino , Humanos , Imuno-Histoquímica , Itália , Masculino , Pessoa de Meia-Idade , Contração Muscular/fisiologia , Miócitos de Músculo Liso/metabolismo , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Placa Aterosclerótica/fisiopatologia , Túnica Íntima/metabolismo , Túnica Íntima/patologia
5.
F1000Res ; 5: 416, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27335638

RESUMO

Higher vertebrates express six different highly conserved actin isoforms that can be classified in three subgroups: 1) sarcomeric actins, α-skeletal (α-SKA) and α-cardiac (α-CAA), 2) smooth muscle actins (SMAs), α-SMA and γ-SMA, and 3) cytoplasmic actins (CYAs), ß-CYA and γ-CYA. The variations among isoactins, in each subgroup, are due to 3-4 amino acid differences located in their acetylated N-decapeptide sequence. The first monoclonal antibody (mAb) against an actin isoform (α-SMA) was produced and characterized in our laboratory in 1986 (Skalli  et al., 1986). We have further obtained mAbs against the 5 other isoforms. In this report, we focus on the mAb anti-α-SKA and anti-α-CAA obtained after immunization of mice with the respective acetylated N-terminal decapeptides using the Repetitive Immunizations at Multiple Sites Strategy (RIMMS). In addition to the identification of their epitope by immunoblotting, we describe the expression of the 2 sarcomeric actins in mature skeletal muscle and during muscle repair after micro-lesions. In particular, we analyze the expression of α-CAA, α-SKA and α-SMA by co-immunostaining in a time course frame during the muscle repair process. Our results indicate that a restricted myocyte population expresses α-CAA and suggest a high capacity of self-renewal in muscle cells. These antibodies may represent a helpful tool for the follow-up of muscle regeneration and pathological changes.

6.
F1000Res ; 52016.
Artigo em Inglês | MEDLINE | ID: mdl-27158462

RESUMO

The discovery of the myofibroblast has allowed definition of the cell responsible for wound contraction and for the development of fibrotic changes. This review summarizes the main features of the myofibroblast and the mechanisms of myofibroblast generation. Myofibroblasts originate from a variety of cells according to the organ and the type of lesion. The mechanisms of myofibroblast contraction, which appear clearly different to those of smooth muscle cell contraction, are described. Finally, we summarize the possible strategies in order to reduce myofibroblast activities and thus influence several pathologies, such as hypertrophic scars and organ fibrosis.

7.
Cytoskeleton (Hoboken) ; 72(6): 257-67, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26147585

RESUMO

α-Smooth Muscle Actin (α-SMA), a widely characterized cytoskeletal protein, represents the hallmark of myofibroblast differentiation. Transforming growth factorß1 (TGFß1) stimulates α-SMA expression and incorporation into stress fibers, thus providing an increased myofibroblast contractile force that participates in tissue remodeling. We have addressed the molecular mechanism by which α-SMA is stably incorporated into stress fibers in human myofibroblasts following exposure to TGFß1. The unique N-terminal sequence AcEEED, which is critical for α-SMA incorporation into stress fibers, was used to screen for AcEEED binding proteins. Tropomyosins were identified as candidate binding proteins. We find that after TGFß1 treatment elevated levels of the Tpm1.6/7 isoforms, and to a lesser extent Tpm2.1, precede the increase in α-SMA. RNA interference experiments demonstrate that α-SMA fails to stably incorporate into stress fibers of TGFß1 treated fibroblasts depleted of Tpm1.6/7, but not other tropomyosins. This does not appear to be due to exclusive interactions between α-SMA and just the Tpm1.6/7 isoforms. We propose that an additional AcEEED binding factor may be required to generate α-SMA filaments containing just Tpm1.6/7 which result in stable incorporation of the resulting filaments into stress fibers.


Assuntos
Fibroblastos/metabolismo , Músculo Liso/metabolismo , Miofibroblastos/metabolismo , Isoformas de Proteínas/metabolismo , Tropomiosina/metabolismo , Humanos , Proteômica , Fibras de Estresse
8.
Cytoskeleton (Hoboken) ; 71(8): 464-71, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24864014

RESUMO

Here we discuss how the concept and the name of cytoskeleton were generated and started to evolve over the last two centuries into what is presently a basic topic of modern biology. We also attempt to describe some facets of the emergence of cytoskeleton component characterization in which our laboratory was in part involved.


Assuntos
Citoesqueleto/metabolismo , Actinas/metabolismo , Animais , Biologia/história , História do Século XX , História do Século XXI , Humanos , Miofibroblastos/metabolismo , Cicatrização/fisiologia
9.
Ideggyogy Sz ; 67(3-4): 124-5, 2014 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-26118255

RESUMO

The present writing is a recollection of Hans Selye, as an educator of graduate and post-graduate students. His main aim in teaching his students was to encourage originality and significance of all scientific research.


Assuntos
Criatividade , Docentes de Medicina/história , Relações Interprofissionais , Mentores , Observação , Pesquisadores/história , Pesquisa/história , Academias e Institutos/história , Canadá , Tecido de Granulação/patologia , História do Século XX , Humanos , Hungria , Microscopia Eletrônica , Miofibroblastos/patologia , Observação/métodos , Pesquisa/educação , Pesquisa/normas , Projetos de Pesquisa/normas , Pesquisadores/educação , Pesquisadores/normas , Faculdades de Medicina/história
10.
Hum Mol Genet ; 22(15): 3123-37, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23591991

RESUMO

Mutations in ACTA2, encoding the smooth muscle cell (SMC)-specific isoform of α-actin (α-SMA), cause thoracic aortic aneurysms and dissections and occlusive vascular diseases, including early onset coronary artery disease and stroke. We have shown that occlusive arterial lesions in patients with heterozygous ACTA2 missense mutations show increased numbers of medial or neointimal SMCs. The contribution of SMC hyperplasia to these vascular diseases and the pathways responsible for linking disruption of α-SMA filaments to hyperplasia are unknown. Here, we show that the loss of Acta2 in mice recapitulates the SMC hyperplasia observed in ACTA2 mutant SMCs and determine the cellular pathways responsible for SMC hyperplasia. Acta2(-/-) mice showed increased neointimal formation following vascular injury in vivo, and SMCs explanted from these mice demonstrated increased proliferation and migration. Loss of α-SMA induced hyperplasia through focal adhesion (FA) rearrangement, FA kinase activation, re-localization of p53 from the nucleus to the cytoplasm and increased expression and ligand-independent activation of platelet-derived growth factor receptor beta (Pdgfr-ß). Disruption of α-SMA in wild-type SMCs also induced similar cellular changes. Imatinib mesylate inhibited Pdgfr-ß activation and Acta2(-/-) SMC proliferation in vitro and neointimal formation with vascular injury in vivo. Loss of α-SMA leads to SMC hyperplasia in vivo and in vitro through a mechanism involving FAK, p53 and Pdgfr-ß, supporting the hypothesis that SMC hyperplasia contributes to occlusive lesions in patients with ACTA2 missense mutations.


Assuntos
Actinas/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Actinas/genética , Animais , Movimento Celular/genética , Núcleo Celular/metabolismo , Proliferação de Células , Ativação Enzimática , Hiperplasia , Camundongos , Camundongos Knockout , Modelos Biológicos , Fenótipo , Transporte Proteico , Espécies Reativas de Oxigênio/metabolismo
11.
Cardiovasc Pathol ; 22(5): 339-44, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23466011

RESUMO

OBJECTIVES: Characterize the phenotypic features of smooth muscle cells (SMCs) in the wall of human saccular intracranial aneurysms (sIAs). METHODS AND RESULTS: We investigated by means of immunohistochemistry the expression of the cytoskeletal differentiation markers α-smooth muscle actin (α-SMA), smooth muscle myosin heavy chains (SMMHCs), and smoothelin in 26 sIAs and 15 nonaneurysmal cerebral arteries. In addition, S100A4, a recently identified marker of dedifferentiated SMCs in atherosclerotic plaques, was also investigated. Six sIAs and 5 nonaneurysmal arteries were used for morphometric analysis. sIAs displayed a significant medial atrophy compared with nonaneurysmal cerebral arteries; moreover, sIA SMCs showed marked decrease of α-SMA and SMMHCs expression and disappearance of smoothelin. Unexpectedly, S100A4 was strongly up-regulated in media SMCs of sIAs. CONCLUSIONS: In sIAs, media SMCs acquire a dedifferentiated phenotype and show de novo expression of S100A4, characteristic features of atherosclerotic plaque SMCs.


Assuntos
Aneurisma Intracraniano/metabolismo , Aneurisma Intracraniano/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Actinas/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Estudos de Casos e Controles , Desdiferenciação Celular , Diferenciação Celular , Artérias Cerebrais/metabolismo , Artérias Cerebrais/patologia , Proteínas do Citoesqueleto/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Proteínas Musculares/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Fenótipo , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/metabolismo
12.
Cardiovasc Pathol ; 22(1): 33-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-22770861

RESUMO

INTRODUCTION: Venous abnormalities have been associated with different neurological conditions, and the presence of a vascular involvement in multiple sclerosis (MS) has long been anticipated. In view of the recent debate regarding the existence of cerebral venous outflow impairment in MS due to abnormalities of the azygos or internal jugular veins (IJVs), we have studied the morphological and biological features of IJVs in MS patients. METHODS: We examined (a) IJVs specimens from MS patients who underwent surgical reconstruction of the IJV and specimens of the great saphenous vein used for surgical reconstruction, (b) different vein specimens from an MS patient dead of an unrelated cause, and (c) autoptical and surgical IJV specimens from patients without MS. Collagen deposition was assessed by means of Sirius red staining followed by polarized light examination. The expression of collagen type I and III, cytoskeletal proteins (α-smooth muscle actin and smooth muscle myosin heavy chains), and inflammatory markers (CD3 and CD68) was investigated. RESULTS: The extracranial veins of MS patients showed focal thickenings of the wall characterized by a prevailing yellow-green birefringence (corresponding to thin, loosely packed collagen fibers) correlated to a higher expression of type III collagen. No differences in cytoskeletal protein and inflammatory marker expression were observed. DISCUSSION: The IJVs of MS patients presenting a focal thickening of the vein wall are characterized by the prevalence of loosely packed type III collagen fibers in the adventitia. Further studies are required to determine whether the observed venous alterations play a role in MS pathogenesis.


Assuntos
Colágeno Tipo III/análise , Colágeno Tipo I/análise , Veias Jugulares/química , Esclerose Múltipla Crônica Progressiva/metabolismo , Esclerose Múltipla Recidivante-Remitente/metabolismo , Actinas/análise , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos CD/análise , Antígenos de Diferenciação Mielomonocítica/análise , Biomarcadores/análise , Complexo CD3/análise , Estudos de Casos e Controles , Feminino , Humanos , Mediadores da Inflamação/análise , Veias Jugulares/patologia , Masculino , Microscopia de Polarização , Pessoa de Meia-Idade , Esclerose Múltipla Crônica Progressiva/patologia , Esclerose Múltipla Recidivante-Remitente/patologia , Cadeias Pesadas de Miosina/análise , Coloração e Rotulagem/métodos
14.
PLoS One ; 7(6): e39836, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22768138

RESUMO

Emerging evidence suggests that both adult cardiac cell and the cardiac stem/progenitor cell (CSPC) compartments are involved in the patho-physiology of diabetic cardiomyopathy (DCM). We evaluated whether early administration of Resveratrol, a natural antioxidant polyphenolic compound, in addition to improving cardiomyocyte function, exerts a protective role on (i) the progenitor cell pool, and (ii) the myocardial environment and its impact on CSPCs, positively interfering with the onset of DCM phenotype. Adult Wistar rats (n = 128) with streptozotocin-induced type-1 diabetes were either untreated (D group; n = 54) or subjected to administration of trans-Resveratrol (i.p. injection: 2.5 mg/Kg/day; DR group; n = 64). Twenty-five rats constituted the control group (C). After 1, 3 or 8 weeks of hyperglycemia, we evaluated cardiac hemodynamic performance, and cardiomyocyte contractile properties and intracellular calcium dynamics. Myocardial remodeling and tissue inflammation were also assessed by morphometry, immunohistochemistry and immunoblotting. Eventually, the impact of the diabetic "milieu" on CSPC turnover was analyzed in co-cultures of healthy CSPCs and cardiomyocytes isolated from D and DR diabetic hearts. In untreated animals, cardiac function was maintained during the first 3 weeks of hyperglycemia, although a definite ventricular remodeling was already present, mainly characterized by a marked loss of CSPCs and adult cardiac cells. Relevant signs of ventricular dysfunction appeared after 8 weeks of diabetes, and included: 1) a significant reduction in ±dP/dt in comparison with C group, 2) a prolongation of isovolumic contraction/relaxation times, 3) an impaired contraction of isolated cardiomyocytes associated with altered intracellular calcium dynamics. Resveratrol administration reduced atrial CSPC loss, succeeded in preserving the functional abilities of CSPCs and mature cardiac cells, improved cardiac environment by reducing inflammatory state and decreased unfavorable ventricular remodeling of the diabetic heart, leading to a marked recovery of ventricular function. These findings indicate that RSV can constitute an adjuvant therapeutic option in DCM prevention.


Assuntos
Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/fisiopatologia , Miocárdio/patologia , Estilbenos/farmacologia , Estilbenos/uso terapêutico , Remodelação Ventricular/efeitos dos fármacos , Actinas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Contagem de Células , Técnicas de Cocultura , Diabetes Mellitus Tipo 1/sangue , Diabetes Mellitus Tipo 1/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Proteína HMGB1/metabolismo , Hemodinâmica/efeitos dos fármacos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Masculino , Miocárdio/enzimologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Ratos , Ratos Wistar , Resveratrol , Células-Tronco/efeitos dos fármacos , Células-Tronco/patologia
15.
Cell Adh Migr ; 6(3): 203-19, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22568985

RESUMO

Since its first description in wound granulation tissue, the myofibroblast has been recognized to be a key actor in the epithelial-mesenchymal cross-talk that plays a crucial role in many physiological and pathological situations, such as regulation of prostate development, ventilation-perfusion in lung alveoli or organ fibrosis. The presence of myofibroblasts in the stroma reaction to epithelial tumors is well established and many data are accumulating which suggest that the stroma compartment is an active participant in tumor onset and/or evolution. In this review we summarize the evidence in favor of this concept, the main mechanisms that regulate myofibroblast differentiation and function, as well as the biophysical and biochemical factors possibly involved in epithelial-stroma interactions, using liver carcinoma as main model, in view of achieving a better understanding of tumor progression mechanisms and of tools directed toward stroma as eventual therapeutic target.


Assuntos
Matriz Extracelular/metabolismo , Miofibroblastos/fisiologia , Neoplasias/patologia , Microambiente Tumoral , Animais , Fenômenos Biomecânicos , Comunicação Celular , Matriz Extracelular/patologia , Matriz Extracelular/fisiologia , Humanos , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Neoplasias/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/fisiologia
16.
J Pathol ; 228(2): 131-47, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22570261

RESUMO

The incidence of chronic kidney diseases (CKD) is constantly rising, reaching epidemic proportions in the western world and leading to an enormous threat, even to modern health-care systems, in industrialized countries. Therapies of CKD have greatly improved following the introduction of drugs targeting the renin-angiotensin system (RAAS) but even this refined pharmacological approach has failed to stop progression to end-stage renal disease (ESRD) in many individuals. In vitro historical data and recent new findings have suggested that progression of renal fibrosis might occur as a result of an altered tubulo-interstitial microenvironment and, more specifically, as a result of an altered epithelial-mesenchymal crosstalk. Here we the review biological findings that support the hypothesis of an altered cellular crosstalk in an injured local tubulo-interstitial microenvironment leading to renal disease progression. Copyright © 2012 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Comunicação Celular/fisiologia , Transição Epitelial-Mesenquimal/fisiologia , Rim/patologia , Microambiente Celular , Progressão da Doença , Células Epiteliais/patologia , Retroalimentação Fisiológica/fisiologia , Fibrose , Humanos , Falência Renal Crônica/diagnóstico , Falência Renal Crônica/fisiopatologia , Túbulos Renais/patologia , Mesoderma/patologia , Sistema Renina-Angiotensina/fisiologia
17.
Pathol Int ; 62(4): 246-53, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22449228

RESUMO

Retinoic acid (RA) is a vitamin A derivative that exerts pleiotropic biological effects. Intracellular transport and metabolism of RA are regulated by cellular retinol-binding proteins (CRBP). CRBP-1 is transiently expressed in granulation tissue fibroblasts during wound healing; however, its role in cardiac remodeling remains unknown. A rat myocardial infarction (MI) model was established by ligation of the left coronary artery, and hearts were obtained at 3, 6, 15, 30 and 45 days after operation. Heart sections were examined immunohistochemically using anti-vimentin, anti-α-smooth muscle actin (α-SMA), anti-matrix metalloproteinase (MMP)-2, anti-MMP-9 and anti-CRBP-1 antibodies. Infarction involved 48.8 ± 3.6% of the left ventricle and was followed by an important cardiac remodeling. Vimentin-positive fibroblastic cells including α-SMA-positive myofibroblasts expressed CRBP-1 at 3-, 6-, and 15-days after MI. Expression of CRBP-1 reached a maximum at 6-days after infarction. Thereafter, CRBP-1 expression was dramatically decreased, showing a similar tendency to MMP expression. Human heart specimens of individuals with a recent myocardial infarction demonstrated presence of CRBP-1-positive fibroblasts by immunohistochemistry. We have demonstrated that CRBP-1 is transiently expressed by fibroblasts during cardiac remodeling. Our results suggest that CRBP-1 plays a role in ventricular remodeling after MI allegedly through its RA binding activity.


Assuntos
Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Proteínas Celulares de Ligação ao Retinol/metabolismo , Remodelação Ventricular/fisiologia , Actinas/metabolismo , Idoso , Animais , Biomarcadores/metabolismo , Modelos Animais de Doenças , Fibroblastos/metabolismo , Fibroblastos/patologia , Ventrículos do Coração/metabolismo , Humanos , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Ratos , Ratos Wistar , Vimentina/metabolismo
18.
Int Orthop ; 36(8): 1733-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22460820

RESUMO

PURPOSE: The myofibroblast, a contractile fibroblastic cell expressing α-smooth muscle actin (α-SMA), has been reported to play a role in ligament healing. The aim of this study was to evaluate the feasibility of transplanting culture-derived myofibroblasts in injured rabbit medial collateral ligaments (MCL) and in intact anterior cruciate ligaments (ACL). METHODS: Fibroblasts isolated from the iliotibial band were cultured in the presence of transforming growth factor beta-1 (TGF-ß1) for five days and analysed for α-SMA expression. In a concentration of TGF-ß1 ≥ 10 ng/ml, the differentiation rate into myofibroblast was 90%. After labelling with PKH26, α-SMA -positive cells were transplanted in intact ACL and in injured MCL of ten rabbits. RESULTS: Survival of PKH-26+ cells was seen in all intact and damaged ligaments one day after injection. The density of PKH-26+ cells had decreased at seven days postinjection in both ligaments. Double-positive PKH-26+/α-SMA+ cells were only observed in injured MCL at seven days postinjection. Moreover, we found that genetically modified fibroblasts differentiate into myofibroblasts and can be transplanted into ligaments. CONCLUSIONS: Our data demonstrate that culture-born myofibroblasts survive and maintain α-SMA expression up to one week after transplantation. This study provides the first insight into the feasibility of transplanted mechanically active cells for ligament reconstruction.


Assuntos
Ligamento Cruzado Anterior/cirurgia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Ligamento Colateral Médio do Joelho/lesões , Ligamento Colateral Médio do Joelho/cirurgia , Miofibroblastos/transplante , Actinas/metabolismo , Animais , Sobrevivência Celular , Células Cultivadas , Estudos de Viabilidade , Feminino , Modelos Animais , Miofibroblastos/citologia , Miofibroblastos/metabolismo , Coelhos , Procedimentos de Cirurgia Plástica , Transplante Autólogo , Resultado do Tratamento
19.
Am J Pathol ; 180(4): 1340-55, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22387320

RESUMO

The discovery of the myofibroblast has opened new perspectives for the comprehension of the biological mechanisms involved in wound healing and fibrotic diseases. In recent years, many advances have been made in understanding important aspects of myofibroblast basic biological characteristics. This review summarizes such advances in several fields, such as the following: i) force production by the myofibroblast and mechanisms of connective tissue remodeling; ii) factors controlling the expression of α-smooth muscle actin, the most used marker of myofibroblastic phenotype and, more important, involved in force generation by the myofibroblast; and iii) factors affecting genesis of the myofibroblast and its differentiation from precursor cells, in particular epigenetic factors, such as DNA methylation, microRNAs, and histone modification. We also review the origin and the specific features of the myofibroblast in diverse fibrotic lesions, such as systemic sclerosis; kidney, liver, and lung fibrosis; and the stromal reaction to certain epithelial tumors. Finally, we summarize the emerging strategies for influencing myofibroblast behavior in vitro and in vivo, with the ultimate goal of an effective therapeutic approach for myofibroblast-dependent diseases.


Assuntos
Tecido Conjuntivo/fisiologia , Miofibroblastos/fisiologia , Actinas/genética , Actinas/metabolismo , Animais , Diferenciação Celular/fisiologia , Epigênese Genética/fisiologia , Fibrose , Regulação da Expressão Gênica/fisiologia , Humanos , Miofibroblastos/citologia , Miofibroblastos/metabolismo , Microambiente Tumoral/fisiologia , Cicatrização/fisiologia
20.
Arterioscler Thromb Vasc Biol ; 31(11): 2391-6, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21868702

RESUMO

The arterial adventitia has been long considered an essentially supportive tissue; however, more and more data suggest that it plays a major role in the modulation of the vascular tone by complex interactions with structures located within intima and media. The purpose of this review is to summarize these data and to describe the mechanisms involved in adventitia/media and adventitia/intima cross-talk. In response to a plethora of stimuli, the adventitia undergoes remodeling processes, resulting in positive (adaptive) remodeling, negative (constrictive) remodeling, or both. The differentiation of the adventitial fibroblast into myofibroblast (MF), a key player of wound healing and fibrosis development, is a hallmark of negative remodeling; this can lead to vessel stenosis and thus contribute to major cardiovascular diseases. The mechanisms of fibroblast-to-MF differentiation and the role of the MF in adventitial remodeling are highlighted herein.


Assuntos
Tecido Conjuntivo/patologia , Miofibroblastos/patologia , Túnica Íntima/patologia , Túnica Média/patologia , Animais , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Comunicação Celular/fisiologia , Diferenciação Celular/fisiologia , Tecido Conjuntivo/fisiopatologia , Fibroblastos/patologia , Fibroblastos/fisiologia , Humanos , Miofibroblastos/fisiologia , Túnica Íntima/fisiopatologia , Túnica Média/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA