Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 295(16): 5278-5291, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32144206

RESUMO

Inter-α-inhibitor is a proteoglycan essential for mammalian reproduction and also plays a less well-characterized role in inflammation. It comprises two homologous "heavy chains" (HC1 and HC2) covalently attached to chondroitin sulfate on the bikunin core protein. Before ovulation, HCs are transferred onto the polysaccharide hyaluronan (HA) to form covalent HC·HA complexes, thereby stabilizing an extracellular matrix around the oocyte required for fertilization. Additionally, such complexes form during inflammatory processes and mediate leukocyte adhesion in the synovial fluids of arthritis patients and protect against sepsis. Here using X-ray crystallography, we show that human HC1 has a structure similar to integrin ß-chains, with a von Willebrand factor A domain containing a functional metal ion-dependent adhesion site (MIDAS) and an associated hybrid domain. A comparison of the WT protein and a variant with an impaired MIDAS (but otherwise structurally identical) by small-angle X-ray scattering and analytical ultracentrifugation revealed that HC1 self-associates in a cation-dependent manner, providing a mechanism for HC·HA cross-linking and matrix stabilization. Surprisingly, unlike integrins, HC1 interacted with RGD-containing ligands, such as fibronectin, vitronectin, and the latency-associated peptides of transforming growth factor ß, in a MIDAS/cation-independent manner. However, HC1 utilizes its MIDAS motif to bind to and inhibit the cleavage of complement C3, and small-angle X-ray scattering-based modeling indicates that this occurs through the inhibition of the alternative pathway C3 convertase. These findings provide detailed structural and functional insights into HC1 as a regulator of innate immunity and further elucidate the role of HC·HA complexes in inflammation and ovulation.


Assuntos
alfa-Globulinas/química , Matriz Extracelular/metabolismo , Imunidade Inata , Simulação de Dinâmica Molecular , Ovulação , Humanos , Cadeias beta de Integrinas/química , Domínios Proteicos , Fator de von Willebrand/química
2.
Hum Mol Genet ; 27(21): 3675-3687, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30060141

RESUMO

Fibrillin microfibrils are extracellular matrix assemblies that form the template for elastic fibres, endow blood vessels, skin and other elastic tissues with extensible properties. They also regulate the bioavailability of potent growth factors of the TGF-ß superfamily. A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)10 is an essential factor in fibrillin microfibril function. Mutations in fibrillin-1 or ADAMTS10 cause Weill-Marchesani syndrome (WMS) characterized by short stature, eye defects, hypermuscularity and thickened skin. Despite its importance, there is poor understanding of the role of ADAMTS10 and its function in fibrillin microfibril assembly. We have generated an ADAMTS10 WMS mouse model using Clustered Regularly Spaced Interspaced Short Palindromic Repeats and CRISPR associated protein 9 (CRISPR-Cas9) to introduce a truncation mutation seen in WMS patients. Homozygous WMS mice are smaller and have shorter long bones with perturbation to the zones of the developing growth plate and changes in cell proliferation. Furthermore, there are abnormalities in the ciliary apparatus of the eye with decreased ciliary processes and abundant fibrillin-2 microfibrils suggesting perturbation of a developmental expression switch. WMS mice have increased skeletal muscle mass and more myofibres, which is likely a consequence of an altered skeletal myogenesis. These results correlated with expression data showing down regulation of Growth differentiation factor (GDF8) and Bone Morphogenetic Protein (BMP) growth factor genes. In addition, the mitochondria in skeletal muscle are larger with irregular shape coupled with increased phospho-p38 mitogen-activated protein kinase (MAPK) suggesting muscle remodelling. Our data indicate that decreased SMAD1/5/8 and increased p38/MAPK signalling are associated with ADAMTS10-induced WMS. This model will allow further studies of the disease mechanism to facilitate the development of therapeutic interventions.


Assuntos
Proteínas ADAMTS/genética , Modelos Animais de Doenças , Microfibrilas/metabolismo , Mutação , Transdução de Sinais , Síndrome de Weill-Marchesani/metabolismo , Proteínas ADAMTS/metabolismo , Animais , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Transgênicos , Proteínas Smad Reguladas por Receptor/metabolismo , Síndrome de Weill-Marchesani/genética
3.
Int J Exp Pathol ; 98(4): 172-190, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28905442

RESUMO

Fibrillin microfibrils are indispensable structural elements of connective tissues in multicellular organisms from early metazoans to humans. They have an extensible periodic beaded organization, and support dynamic tissues such as ciliary zonules that suspend the lens. In tissues that express elastin, including blood vessels, skin and lungs, microfibrils support elastin deposition and shape the functional architecture of elastic fibres. The vital contribution of microfibrils to tissue form and function is underscored by the heritable fibrillinopathies, especially Marfan syndrome with severe elastic, ocular and skeletal tissue defects. Research since the early 1990s has advanced our knowledge of biology of microfibrils, yet understanding of their mechanical and homeostatic contributions to tissues remains far from complete. This review is a personal reflection on key insights, and puts forward the conceptual hypothesis that microfibrils are structural 'tensometers' that direct cells to monitor and respond to altered tissue mechanics.


Assuntos
Tecido Elástico/patologia , Matriz Extracelular/patologia , Fibrilinas/metabolismo , Síndrome de Marfan/patologia , Microfibrilas/patologia , Proteínas dos Microfilamentos/metabolismo , Animais , Tecido Elástico/metabolismo , Matriz Extracelular/metabolismo , Humanos , Síndrome de Marfan/metabolismo , Microfibrilas/metabolismo
4.
Methods ; 96: 85-96, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26608109

RESUMO

Induced pluripotent stem cells (iPSCs) provide invaluable opportunities for future cell therapies as well as for studying human development, modelling diseases and discovering therapeutics. In order to realise the potential of iPSCs, it is crucial to comprehensively characterise cells generated from large cohorts of healthy and diseased individuals. The human iPSC initiative (HipSci) is assessing a large panel of cell lines to define cell phenotypes, dissect inter- and intra-line and donor variability and identify its key determinant components. Here we report the establishment of a high-content platform for phenotypic analysis of human iPSC lines. In the described assay, cells are dissociated and seeded as single cells onto 96-well plates coated with fibronectin at three different concentrations. This method allows assessment of cell number, proliferation, morphology and intercellular adhesion. Altogether, our strategy delivers robust quantification of phenotypic diversity within complex cell populations facilitating future identification of the genetic, biological and technical determinants of variance. Approaches such as the one described can be used to benchmark iPSCs from multiple donors and create novel platforms that can readily be tailored for disease modelling and drug discovery.


Assuntos
Fibronectinas/química , Ensaios de Triagem em Larga Escala , Células-Tronco Pluripotentes Induzidas/ultraestrutura , Imagem Molecular/métodos , Fenótipo , Sequência de Aminoácidos , Adesão Celular , Diferenciação Celular , Linhagem Celular , Células Alimentadoras/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Dados de Sequência Molecular , Peptídeos/química
5.
J Cell Sci ; 127(Pt 1): 158-71, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24190885

RESUMO

Here, we show that epithelial-mesenchymal status influences how cells deposit extracellular matrix. Retinal pigmented epithelial (RPE) cells that expressed high levels of E-cadherin and had cell-cell junctions rich in zona occludens (ZO)-1, ß-catenin and heparan sulfate, required syndecan-4 but not fibronectin or protein kinase C α (PKCα) to assemble extracellular matrix (fibrillin microfibrils and perlecan). In contrast, RPE cells that strongly expressed mesenchymal smooth muscle α-actin but little ZO-1 or E-cadherin, required fibronectin (like fibroblasts) and PKCα, but not syndecan-4. Integrins α5ß1 and/or α8ß1 and actomyosin tension were common requirements for microfibril deposition, as was heparan sulfate biosynthesis. TGFß, which stimulates epithelial-mesenchymal transition, altered gene expression and overcame the dependency on syndecan-4 for microfibril deposition in epithelial RPE cells, whereas blocking cadherin interactions disrupted microfibril deposition. Renal podocytes had a transitional phenotype with pericellular ß-catenin but little ZO-1; they required syndecan-4 and fibronectin for efficient microfibril deposition. Thus, epithelial-mesenchymal status modulates microfibril deposition.


Assuntos
Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/genética , Microfibrilas/metabolismo , Proteínas dos Microfilamentos/genética , Actinas/genética , Actinas/metabolismo , Actomiosina/genética , Actomiosina/metabolismo , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular , Células Epiteliais/ultraestrutura , Feminino , Fibrilina-1 , Fibrilinas , Fibroblastos/metabolismo , Fibroblastos/ultraestrutura , Fibronectinas/genética , Fibronectinas/metabolismo , Regulação da Expressão Gênica , Heparitina Sulfato/metabolismo , Humanos , Integrinas/genética , Integrinas/metabolismo , Junções Intercelulares/metabolismo , Junções Intercelulares/ultraestrutura , Glândulas Mamárias Humanas/metabolismo , Glândulas Mamárias Humanas/ultraestrutura , Microfibrilas/ultraestrutura , Proteínas dos Microfilamentos/metabolismo , Especificidade de Órgãos , Podócitos/metabolismo , Podócitos/ultraestrutura , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/ultraestrutura , Sindecana-4/genética , Sindecana-4/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
6.
Ann Rheum Dis ; 74(6): 1249-56, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24442880

RESUMO

OBJECTIVES: Leri's pleonosteosis (LP) is an autosomal dominant rheumatic condition characterised by flexion contractures of the interphalangeal joints, limited motion of multiple joints, and short broad metacarpals, metatarsals and phalanges. Scleroderma-like skin thickening can be seen in some individuals with LP. We undertook a study to characterise the phenotype of LP and identify its genetic basis. METHODS AND RESULTS: Whole-genome single-nucleotide polymorphism genotyping in two families with LP defined microduplications of chromosome 8q22.1 as the cause of this condition. Expression analysis of dermal fibroblasts from affected individuals showed overexpression of two genes, GDF6 and SDC2, within the duplicated region, leading to dysregulation of genes that encode proteins of the extracellular matrix and downstream players in the transforming growth factor (TGF)-ß pathway. Western blot analysis revealed markedly decreased inhibitory SMAD6 levels in patients with LP. Furthermore, in a cohort of 330 systemic sclerosis cases, we show that the minor allele of a missense SDC2 variant, p.Ser71Thr, could confer protection against disease (p<1×10(-5)). CONCLUSIONS: Our work identifies the genetic cause of LP in these two families, demonstrates the phenotypic range of the condition, implicates dysregulation of extracellular matrix homoeostasis genes in its pathogenesis, and highlights the link between TGF-ß/SMAD signalling, growth/differentiation factor 6 and syndecan-2. We propose that LP is an additional member of the growing 'TGF-ß-pathies' group of musculoskeletal disorders, which includes Myhre syndrome, acromicric dysplasia, geleophysic dysplasias, Weill-Marchesani syndromes and stiff skin syndrome. Identification of a systemic sclerosis-protective SDC2 variant lays the foundation for exploration of the role of syndecan-2 in systemic sclerosis in the future.


Assuntos
Cromossomos Humanos Par 8/genética , Duplicação Gênica , Fator 6 de Diferenciação de Crescimento/genética , Deformidades Congênitas da Mão/genética , Artropatias/congênito , Ossificação Heterotópica/genética , Escleroderma Sistêmico/genética , Sindecana-2/genética , Adulto , Idoso , Pré-Escolar , Matriz Extracelular/metabolismo , Fácies , Feminino , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Fator 6 de Diferenciação de Crescimento/metabolismo , Deformidades Congênitas da Mão/metabolismo , Deformidades Congênitas da Mão/fisiopatologia , Humanos , Lactente , Artropatias/genética , Artropatias/metabolismo , Artropatias/fisiopatologia , Masculino , Pessoa de Meia-Idade , Ossificação Heterotópica/metabolismo , Ossificação Heterotópica/fisiopatologia , Fenótipo , Transdução de Sinais , Sindecana-2/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Adulto Jovem
7.
J Cell Sci ; 124(Pt 8): 1288-300, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21429937

RESUMO

Cell migration during vascular remodelling is regulated by crosstalk between growth factor receptors and integrin receptors, which together coordinate cytoskeletal and motogenic changes. Here, we report extracellular matrix (ECM)-directed crosstalk between platelet-derived growth factor receptor (PDGFR)-ß and α5ß1-integrin, which controls the migration of mesenchymal stem (stromal) cells (MSCs). Cell adhesion to fibronectin induced α5ß1-integrin-dependent phosphorylation of PDGFR-ß in the absence of growth factor stimulation. Phosphorylated PDGFR-ß co-immunoprecipitated with α5-integrin and colocalised with α5ß1-integrin in the transient tidemarks of focal adhesions. Adhesion to fibronectin also strongly potentiated PDGF-BB-induced PDGFR-ß phosphorylation and focal adhesion kinase (FAK) activity, in an α5ß1-integrin-dependent manner. PDGFR-ß-induced phosphoinositide 3-kinase (PI3K) and Akt activity, actin reorganisation and cell migration were all regulated by fibronectin and α5ß1-integrin. This synergistic relationship between α5ß1-integrin and PDGFR-ß is a fundamental determinant of cell migration. Thus, fibronectin-rich matrices can prime PDGFR-ß to recruit mesenchymal cells at sites of vascular remodelling.


Assuntos
Movimento Celular , Fibronectinas/metabolismo , Integrina alfa5beta1/metabolismo , Células-Tronco Mesenquimais/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Adulto , Becaplermina , Adesão Celular , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Humanos , Integrina alfa5beta1/genética , Masculino , Células-Tronco Mesenquimais/citologia , Proteínas Proto-Oncogênicas c-sis , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Adulto Jovem
8.
Expert Rev Mol Med ; 15: e8, 2013 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-23962539

RESUMO

Elastic fibres are insoluble components of the extracellular matrix of dynamic connective tissues such as skin, arteries, lungs and ligaments. They are laid down during development, and comprise a cross-linked elastin core within a template of fibrillin-based microfibrils. Their function is to endow tissues with the property of elastic recoil, and they also regulate the bioavailability of transforming growth factor ß. Severe heritable elastic fibre diseases are caused by mutations in elastic fibre components; for example, mutations in elastin cause supravalvular aortic stenosis and autosomal dominant cutis laxa, mutations in fibrillin-1 cause Marfan syndrome and Weill-Marchesani syndrome, and mutations in fibulins-4 and -5 cause autosomal recessive cutis laxa. Acquired elastic fibre defects include dermal elastosis, whereas inflammatory damage to fibres contributes to pathologies such as pulmonary emphysema and vascular disease. This review outlines the latest understanding of the composition and assembly of elastic fibres, and describes elastic fibre diseases and current therapeutic approaches.


Assuntos
Doença , Tecido Elástico , Saúde , Animais , Tecido Elástico/química , Tecido Elástico/metabolismo , Humanos
9.
Stem Cells ; 30(3): 548-60, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22213560

RESUMO

Defining the signaling mechanisms that regulate the fate of adult stem cells is an essential step toward their use in regenerative medicine. Platelet-derived growth factor receptor (PDGFR) signaling plays a crucial role in specifying mesenchymal stem cell (MSC) commitment to mesenchymal lineages. Based on the hypothesis that selective inhibition of signaling pathways involved in differentiation may increase stem cell potency, we examined the role of PDGFR signaling in controlling the fate of human MSCs. Using a small molecular PDGFR inhibitor that induced MSCs toward a more rounded shape, expression of Oct4 and Nanog were markedly upregulated. In these PDGFR inhibitor-treated MSCs, Oct4 and Nanog expression and cell shape were regulated by janus kinase (JAK), MAPK kinase (MEK), and epidermal growth factor receptor (EGFR) signaling. Under defined differentiation conditions, these PDGFR-inhibited MSCs expressed definitive endodermal, ectodermal, and mesodermal markers. We also confirmed that depletion of individual PDGF receptors upregulated expression of Oct4A and Nanog. This study identifies PDGFR signaling as a key regulator of Oct4 and Nanog expression and of MSC potency. Thus, inhibiting these specific receptor tyrosine kinases, which play essential roles in tissue formation, offers a novel approach to unlock the therapeutic capacity of MSCs.


Assuntos
Forma Celular/efeitos dos fármacos , Proteínas de Homeodomínio/metabolismo , Indanos/farmacologia , Células-Tronco Mesenquimais/fisiologia , Fator 3 de Transcrição de Octâmero/metabolismo , Pirazóis/farmacologia , Quinolinas/farmacologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Actomiosina/metabolismo , Adulto , Diferenciação Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Tamanho do Núcleo Celular/efeitos dos fármacos , Células Cultivadas , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Proteínas de Homeodomínio/genética , Humanos , Janus Quinases/antagonistas & inibidores , Janus Quinases/metabolismo , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-abl/genética , Proteínas Proto-Oncogênicas c-abl/metabolismo , Interferência de RNA , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Adulto Jovem
10.
J Biol Chem ; 286(46): 40266-75, 2011 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-21908605

RESUMO

Collagen VI is a ubiquitous extracellular matrix protein that assembles into beaded microfibrils that form networks linking cells to the matrix. Collagen VI microfibrils are typically formed from a heterotrimer of the α1, α2, and α3 chains. The α3 chain is distinct as it contains an extended N terminus with up to 10 consecutive von Willebrand factor type A-domains (VWA). Here, we use solution small angle x-ray scattering (SAXS) and single particle analysis EM to determine the nanostructure of nine of these contiguous A-domains. Both techniques reveal a tight C-shape conformation for the A-domains. Furthermore, using biophysical approaches, we demonstrate that the N-terminal region undergoes a conformational change and a proportion forms dimers in the presence of Zn(2+). This is the first indication that divalent cations interact with collagen VI A-domains. A three-dimensional reconstruction of tissue-purified collagen VI microfibrils was generated using EM and single particle image analysis. The reconstruction showed the intricate architecture of the collagen VI globular regions, in particular the highly structurally conserved C-terminal region and variations in the appearance of the N-terminal region. The N-terminal domains project out from the globular beaded region like angled radial spokes. These could potentially provide interactive surfaces for other cell matrix molecules.


Assuntos
Colágeno Tipo VI/química , Microfibrilas/química , Multimerização Proteica/fisiologia , Colágeno Tipo VI/genética , Colágeno Tipo VI/metabolismo , Células HEK293 , Humanos , Microfibrilas/genética , Microfibrilas/metabolismo , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína
11.
J Cell Sci ; 123(Pt 17): 3006-18, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20699357

RESUMO

Control of the bioavailability of the growth factor TGFbeta is essential for tissue formation and homeostasis, yet precisely how latent TGFbeta is incorporated into the extracellular matrix is unknown. Here, we show that deposition of a large latent TGFbeta complex (LLC), which contains latent TGFbeta-binding protein 1 (LTBP-1), is directly dependent on the pericellular assembly of fibrillin microfibrils, which interact with fibronectin during higher-order fibrillogenesis. LTBP-1 formed pericellular arrays that colocalized with microfibrils, whereas fibrillin knockdown inhibited fibrillar LTBP-1 and/or LLC deposition. Blocking alpha5beta1 integrin or supplementing cultures with heparin, which both inhibited microfibril assembly, disrupted LTBP-1 deposition and enhanced Smad2 phosphorylation. Full-length LTBP-1 bound only weakly to N-terminal pro-fibrillin-1, but this association was strongly enhanced by heparin. The microfibril-associated glycoprotein MAGP-1 (MFAP-2) inhibited LTBP-1 binding to fibrillin-1 and stimulated Smad2 phosphorylation. By contrast, fibulin-4, which interacted strongly with full-length LTBP-1, did not induce Smad2 phosphorylation. Thus, LTBP-1 and/or LLC deposition is dependent on pericellular microfibril assembly and is governed by complex interactions between LTBP-1, heparan sulfate, fibrillin-1 and microfibril-associated molecules. In this way, microfibrils control TGFbeta bioavailability.


Assuntos
Proteínas de Ligação a TGF-beta Latente/metabolismo , Microfibrilas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Proteínas Contráteis/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fibrilina-1 , Fibrilinas , Fibroblastos/metabolismo , Heparina/farmacologia , Heparitina Sulfato/metabolismo , Humanos , Microscopia de Fluorescência , Modelos Moleculares , Dados de Sequência Molecular , Fatores de Processamento de RNA
12.
J Cell Biol ; 177(3): 489-500, 2007 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-17470632

RESUMO

Vascular endothelial growth factor (VEGF-A) is a crucial stimulator of vascular cell migration and proliferation. Using bone marrow-derived human adult mesenchymal stem cells (MSCs) that did not express VEGF receptors, we provide evidence that VEGF-A can stimulate platelet-derived growth factor receptors (PDGFRs), thereby regulating MSC migration and proliferation. VEGF-A binds to both PDGFRalpha and PDGFRbeta and induces tyrosine phosphorylation that, when inhibited, results in attenuation of VEGF-A-induced MSC migration and proliferation. This mechanism was also shown to mediate human dermal fibroblast (HDF) migration. VEGF-A/PDGFR signaling has the potential to regulate vascular cell recruitment and proliferation during tissue regeneration and disease.


Assuntos
Células da Medula Óssea/metabolismo , Células-Tronco Mesenquimais/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adolescente , Adulto , Células da Medula Óssea/citologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Células Cultivadas , Derme/citologia , Derme/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Recém-Nascido , Masculino , Células-Tronco Mesenquimais/citologia , Ligação Proteica/fisiologia , Regeneração/efeitos dos fármacos , Regeneração/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/farmacologia
13.
J Cell Biol ; 176(3): 355-67, 2007 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-17242066

RESUMO

We have discovered that fibrillin-1, which forms extracellular microfibrils, can regulate the bioavailability of transforming growth factor (TGF) beta1, a powerful cytokine that modulates cell survival and phenotype. Altered TGFbeta signaling is a major contributor to the pathology of Marfan syndrome (MFS) and related diseases. In the presence of cell layer extracellular matrix, a fibrillin-1 sequence encoded by exons 44-49 releases endogenous TGFbeta1, thereby stimulating TGFbeta receptor-mediated Smad2 signaling. This altered TGFbeta1 bioavailability does not require intact cells, proteolysis, or the altered expression of TGFbeta1 or its receptors. Mass spectrometry revealed that a fibrillin-1 fragment containing the TGFbeta1-releasing sequence specifically associates with full-length fibrillin-1 in cell layers. Solid-phase and BIAcore binding studies showed that this fragment interacts strongly and specifically with N-terminal fibrillin-1, thereby inhibiting the association of C-terminal latent TGFbeta-binding protein 1 (a component of the large latent complex [LLC]) with N-terminal fibrillin-1. By releasing LLC from microfibrils, the fibrillin-1 sequence encoded by exons 44-49 can contribute to MFS and related diseases.


Assuntos
Microfibrilas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Transdução de Sinais/fisiologia , Fator de Crescimento Transformador beta1/metabolismo , Receptores de Ativinas Tipo I/metabolismo , Linhagem Celular , Fibrilina-1 , Fibrilinas , Expressão Gênica/fisiologia , Humanos , Síndrome de Marfan/metabolismo , Espectrometria de Massas , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/genética , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Proteínas Serina-Treonina Quinases , Estrutura Terciária de Proteína , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína Smad2/metabolismo
14.
Matrix Biol ; 107: 24-39, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35122964

RESUMO

TGFß superfamily members are potent growth factors in the extracellular matrix with essential roles in all aspects of cellular behaviour. Latent TGFß binding proteins (LTBPs) are co-expressed with TGFß, essential for correct folding and secretion of the growth factor, to form large latent complexes. These large latent complexes bind extracellular proteins such as fibrillin for sequestration of TGFß in the matrix, essential for normal tissue function, and dysregulated TGFß signalling is a hallmark of many fibrillinopathies. Transglutaminase-2 (TG2) cross-linking of LTBPs is known to play a role in TGFß activation but the underlying molecular mechanisms are not resolved. Here we show that fibrillin is a matrix substrate for TG2 and that TG2 cross-linked complexes can be formed between fibrillin and LTBP-1 and -3, and their latent TGFß complexes. The structure of the fibrillin-LTBP1 complex shows that the two elongated proteins interact in a perpendicular arrangement which would allow them to form distal interactions between the matrix and the cell surface. Formation of the cross-link with fibrillin does not change the interaction between latent TGFß and integrin αVß6 but does increase TGFß activation in cell-based assays. The activating effect may be due to direction of the latent complexes to the cell surface by fibrillin, as competition with heparan sulphate can ameliorate the activating effect. Together, these data support that TGFß activation can be enhanced by covalent tethering of LTBPs to the matrix via fibrillin.


Assuntos
Proteínas dos Microfilamentos , Transglutaminases , Matriz Extracelular/metabolismo , Fibrilina-1/genética , Fibrilina-1/metabolismo , Fibrilina-2/metabolismo , Fibrilinas/metabolismo , Proteínas de Ligação a TGF-beta Latente/genética , Proteínas de Ligação a TGF-beta Latente/metabolismo , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Transglutaminases/genética , Transglutaminases/metabolismo
15.
Mol Cell Proteomics ; 8(12): 2715-32, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19755719

RESUMO

Deciphering interacting networks of the extracellular matrix is a major challenge. We describe an affinity purification and mass spectrometry strategy that has provided new insights into the molecular interactions of elastic fibers, essential extracellular assemblies that provide elastic recoil in dynamic tissues. Using cell culture models, we defined primary and secondary elastic fiber interaction networks by identifying molecular interactions with the elastic fiber molecules fibrillin-1, MAGP-1, fibulin-5, and lysyl oxidase. The sensitivity and validity of our method was confirmed by identification of known interactions with the bait proteins. Our study revealed novel extracellular protein interactions with elastic fiber molecules and delineated secondary interacting networks with fibronectin and heparan sulfate-associated molecules. This strategy is a novel approach to define the macromolecular interactions that sustain complex extracellular matrix assemblies and to gain insights into how they are integrated into their surrounding matrix.


Assuntos
Cromatografia de Afinidade/métodos , Tecido Elástico/metabolismo , Espectrometria de Massas/métodos , Proteínas de Ligação ao Cálcio/metabolismo , Fibrilina-1 , Fibrilinas , Heparina/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/metabolismo , Peptídeos/metabolismo , Progranulinas , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Reprodutibilidade dos Testes , Software
16.
Biochem J ; 427(1): 29-40, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20102335

RESUMO

Using human MSCs (mesenchymal stem cells) lacking VEGF (vascular endothelial growth factor) receptors, we show that the pro-angiogenic receptor neuropilin-1 associates with phosphorylated PDGFRs [PDGF (platelet-derived growth factor) receptors], thereby regulating cell signalling, migration, proliferation and network assembly. Neuropilin-1 co-immunoprecipitated and co-localized with phosphorylated PDGFRs in the presence of growth factors. Neuropilin-1 knockdown blocked PDGF-AA-induced PDGFRalpha phosphorylation and migration, reduced PDGF-BB-induced PDGFRbeta activation and migration, blocked VEGF-A activation of both PDGFRs, and attenuated proliferation. Neuropilin-1 prominently co-localized with both PDGFRs within MSC networks assembled in Matrigel and in the chorioallantoic membrane vasculature microenvironment, and its knockdown grossly disrupted network assembly and decreased PDGFR signalling. Thus neuropilin-1 regulates MSCs by forming ligand-specific receptor complexes that direct PDGFR signalling, especially the PDGFRalpha homodimer. This receptor cross-talk may control the mobilization of MSCs in neovascularization and tissue remodelling.


Assuntos
Movimento Celular , Células-Tronco Mesenquimais/metabolismo , Neuropilina-1/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Becaplermina , Western Blotting , Proliferação de Células , Galinhas , Membrana Corioalantoide/metabolismo , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Neuropilina-1/antagonistas & inibidores , Neuropilina-1/genética , Fosforilação , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-sis , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
J Biol Chem ; 284(38): 25938-43, 2009 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-19617354

RESUMO

Fibulin 5 is a 52-kDa calcium-binding epidermal growth factor (cbEGF)-rich extracellular matrix protein that is essential for the formation of elastic tissues. Missense mutations in fibulin 5 cause the elastin disorder cutis laxa and have been associated with age-related macular degeneration, a leading cause of blindness. We investigated the structure, hydrodynamics, and oligomerization of fibulin 5 using small angle x-ray scattering, EM, light scattering, circular dichroism, and sedimentation. Compact structures for the monomer were determined by small angle x-ray scattering and EM, and are supported by close agreement between the theoretical sedimentation of the structures and the experimental sedimentation of the monomer in solution. EM showed that monomers associate around a central cavity to form a dimer. Light scattering and equilibrium sedimentation demonstrated that the equilibrium between the monomer and the dimer is dependent upon NaCl and Ca2+ concentrations and that the dimer is dominant under physiological conditions. The dimerization of fragments containing just the cbEGF domains suggests that intermolecular interactions between cbEGFs cause dimerization of fibulin 5. It is possible that fibulin 5 functions as a dimer during elastinogenesis or that dimerization may provide a method for limiting interactions with binding partners such as tropoelastin.


Assuntos
Proteínas da Matriz Extracelular/química , Multimerização Proteica/fisiologia , Cloreto de Sódio/química , Cálcio/química , Cálcio/metabolismo , Dicroísmo Circular , Cútis Laxa/genética , Cútis Laxa/metabolismo , Tecido Elástico/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Humanos , Degeneração Macular/genética , Degeneração Macular/metabolismo , Mutação de Sentido Incorreto , Ligação Proteica/fisiologia , Estrutura Quaternária de Proteína/fisiologia , Espalhamento de Radiação , Cloreto de Sódio/metabolismo , Tropoelastina/química , Tropoelastina/genética , Tropoelastina/metabolismo , Raios X
18.
Methods Mol Biol ; 522: 123-41, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19247600

RESUMO

The atomic force microscope (AFM) may be used to collect quantitative height data from extracellular matrix molecules and macro-molecular assemblies adsorbed to a wide range of solid substrates. The advantages of atomic force microscopy include rapid specimen preparation, which does not rely on chemical fixation, dehydration or heavy-metal staining, and sub-nanometre resolution imaging with a high signal-noise ratio. In combination with complimentary techniques such as molecular combing and by exploiting the ability to act as a force spectrometer, the AFM can provide valuable information on the nano-mechanical properties of extracellular matrix components.


Assuntos
Matriz Extracelular/química , Microscopia de Força Atômica/métodos , Nanotecnologia
19.
Methods Mol Biol ; 522: 151-61, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19247598

RESUMO

In the scanning transmission electron microscope, the degree of electron scattering induced by biological specimens, such as ECM macromolecules, is dependent on the molecular mass. By calibrating the ratio of scattered to non-scattered electrons against a known mass standard, such as tobacco mosaic virus, it is possible to quantify absolute changes in both mass and mass distribution. These mass mapping approaches can provide important information on ECM assembly, organisation, and interactions which is not obtainable by other means.


Assuntos
Proteínas da Matriz Extracelular/química , Microscopia Eletrônica de Transmissão e Varredura/métodos , Peso Molecular
20.
Methods Mol Biol ; 522: 175-81, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19247619

RESUMO

Conventional preparation techniques for electron microscopy employ contrast enhancing heavy metal stains in solution to visualize isolated macromolecules. In rotary shadowing electron microscopy, the heavy metal is evaporated onto surface adsorbed molecules and macromolecular assemblies. High resolution shadowing remains a valuable method for the visualization and characterization of extracellular matrix macromolecules including fibrillar collagens, microfibrillar elements, and glycoproteins.


Assuntos
Proteínas da Matriz Extracelular/ultraestrutura , Microscopia Eletrônica de Transmissão/métodos , Colágeno/química , Fibrilinas , Proteínas dos Microfilamentos/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA