Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
Matrix Biol ; 107: 24-39, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35122964

RESUMEN

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.


Asunto(s)
Proteínas de Microfilamentos , Transglutaminasas , Matriz Extracelular/metabolismo , Fibrilina-1/genética , Fibrilina-1/metabolismo , Fibrilina-2/metabolismo , Fibrilinas/metabolismo , Proteínas de Unión a TGF-beta Latente/genética , Proteínas de Unión a TGF-beta Latente/metabolismo , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Transglutaminasas/genética , Transglutaminasas/metabolismo
2.
J Biol Chem ; 295(16): 5278-5291, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32144206

RESUMEN

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.


Asunto(s)
alfa-Globulinas/química , Matriz Extracelular/metabolismo , Inmunidad Innata , Simulación de Dinámica Molecular , Ovulación , Humanos , Cadenas beta de Integrinas/química , Dominios Proteicos , Factor de von Willebrand/química
3.
Hum Mol Genet ; 27(21): 3675-3687, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30060141

RESUMEN

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.


Asunto(s)
Proteínas ADAMTS/genética , Modelos Animales de Enfermedad , Microfibrillas/metabolismo , Mutación , Transducción de Señal , Síndrome de Weill-Marchesani/metabolismo , Proteínas ADAMTS/metabolismo , Animales , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Transgénicos , Proteínas Smad Reguladas por Receptores/metabolismo , Síndrome de Weill-Marchesani/genética
4.
Methods Mol Biol ; 1722: 249-260, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29264810

RESUMEN

We present a method to capture mesenchymal stromal cells (MSCs) by adhesion to extracellular matrix (ECM) molecules under flow conditions. The technique simulates a physiological system and exploits the natural biological interactions of cells, through integrin receptors, with their ECM. The system offers an insight into how MSCs could be targeted/localized to the site of interest (graft) following intravenous injection.


Asunto(s)
Matriz Extracelular/fisiología , Citometría de Flujo/métodos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Adhesión Celular/fisiología , Diferenciación Celular/fisiología , Células Inmovilizadas/metabolismo , Fibronectinas/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Procesamiento de Imagen Asistido por Computador , Integrinas/metabolismo , Laminina/metabolismo , Células Madre Mesenquimatosas/clasificación , Microscopía por Video , ARN , Análisis de la Célula Individual
5.
Tissue Eng Part A ; 24(11-12): 968-978, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29279011

RESUMEN

We previously developed a 14-day culture protocol under potentially GMP, chemically defined conditions, to generate chondroprogenitors from human embryonic stem cells (hESCs). In vivo work has confirmed the cartilage repair capacity of these cells in a nude rat osteochondral defect model. Aiming to enhance hESC-chondrogenesis, we screened a range of extracellular matrix (ECM) molecules for their ability to support differentiation of hESCs toward chondrocytes. We identified two novel ECM protein fragments that supported hESC-chondrogenesis: Fibronectin III (fibronectin 7-14 protein fragments, including the RGD domain, syndecan-binding domain, and heparin-binding domain) and fibrillin-1 (FBN1) fragment PF8 (encoded by exons 30-38, residues 1238-1605, which contains the RGD motif but not heparin-binding site). These two protein fragments support hESC-chondrogenesis compared with the substrates routinely used previously (a mixture of fibronectin and gelatin) in our directed chondrogenic protocol. We have identified recombinant fibronectin fragment (FN III) and FBNI fragment (PF8) as alternative coating substrates to promote expression of genes known to regulate chondrocytes and code for chondrocyte ECM components. These recombinant protein fragments are likely to have better batch to batch stability than full-length molecules, especially where extracted from tissue/serum.


Asunto(s)
Condrogénesis/efectos de los fármacos , Proteínas de la Matriz Extracelular/química , Cartílago/efectos de los fármacos , Cartílago/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Fibronectinas/metabolismo , Humanos
6.
Int J Exp Pathol ; 98(4): 172-190, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28905442

RESUMEN

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.


Asunto(s)
Tejido Elástico/patología , Matriz Extracelular/patología , Fibrilinas/metabolismo , Síndrome de Marfan/patología , Microfibrillas/patología , Proteínas de Microfilamentos/metabolismo , Animales , Tejido Elástico/metabolismo , Matriz Extracelular/metabolismo , Humanos , Síndrome de Marfan/metabolismo , Microfibrillas/metabolismo
7.
Stem Cell Res Ther ; 8(1): 79, 2017 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-28412968

RESUMEN

BACKGROUND: Inflammation is a key contributor to central nervous system (CNS) injury such as stroke, and is a major target for therapeutic intervention. Effective treatments for CNS injuries are limited and applicable to only a minority of patients. Stem cell-based therapies are increasingly considered for the treatment of CNS disease, because they can be used as in-situ regulators of inflammation, and improve tissue repair and recovery. One promising option is the use of bone marrow-derived mesenchymal stem cells (MSCs), which can secrete anti-inflammatory and trophic factors, can migrate towards inflamed and injured sites or can be implanted locally. Here we tested the hypothesis that pre-treatment with inflammatory cytokines can prime MSCs towards an anti-inflammatory and pro-trophic phenotype in vitro. METHODS: Human MSCs from three different donors were cultured in vitro and treated with inflammatory mediators as follows: interleukin (IL)-1α, IL-1ß, tumour necrosis factor alpha (TNF-α) or interferon-γ. After 24 h of treatment, cell supernatants were analysed by ELISA for expression of granulocyte-colony stimulating factor (G-CSF), IL-10, brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), IL-1 receptor antagonist (IL-1Ra) and vascular endothelial growth factor (VEGF). To confirm the anti-inflammatory potential of MSCs, immortalised mouse microglial BV2 cells were treated with bacterial lipopolysaccharide (LPS) and exposed to conditioned media (CM) of naïve or IL-1-primed MSCs, and levels of secreted microglial-derived inflammatory mediators including TNF-α, IL-10, G-CSF and IL-6 were measured by ELISA. RESULTS: Unstimulated MSCs constitutively expressed anti-inflammatory cytokines and trophic factors (IL-10, VEGF, BDNF, G-CSF, NGF and IL-1Ra). MSCs primed with IL-1α or IL-1ß showed increased secretion of G-CSF, which was blocked by IL-1Ra. Furthermore, LPS-treated BV2 cells secreted less inflammatory and apoptotic markers, and showed increased secretion of the anti-inflammatory IL-10 in response to treatment with CM of IL-1-primed MSCs compared with CM of unprimed MSCs. CONCLUSIONS: Our results demonstrate that priming MSCs with IL-1 increases expression of trophic factor G-CSF through an IL-1 receptor type 1 (IL-1R1) mechanism, and induces a reduction in the secretion of inflammatory mediators in LPS-activated microglial cells. The results therefore support the potential use of preconditioning treatments of stem cells in future therapies.


Asunto(s)
Antiinflamatorios/farmacología , Interleucina-1alfa/farmacología , Interleucina-1beta/farmacología , Células Madre Mesenquimatosas/citología , Adulto , Animales , Biomarcadores/metabolismo , Medios de Cultivo Condicionados/farmacología , Femenino , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Microglía/citología , Microglía/efectos de los fármacos , Factores de Crecimiento Nervioso/farmacología , Fenotipo , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
8.
Sci Rep ; 6: 35956, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27779234

RESUMEN

ADAMTS10 and ADAMTS6 are homologous metalloproteinases with ill-defined roles. ADAMTS10 mutations cause Weill-Marchesani syndrome (WMS), implicating it in fibrillin microfibril biology since some fibrillin-1 mutations also cause WMS. However little is known about ADAMTS6 function. ADAMTS10 is resistant to furin cleavage, however we show that ADAMTS6 is effectively processed and active. Using siRNA, over-expression and mutagenesis, it was found ADAMTS6 inhibits and ADAMTS10 is required for focal adhesions, epithelial cell-cell junction formation, and microfibril deposition. Either knockdown of ADAMTS6, or disruption of its furin processing or catalytic sites restores focal adhesions, implicating its enzyme activity acts on targets in the focal adhesion complex. In ADAMTS10-depleted cultures, expression of syndecan-4 rescues focal adhesions and cell-cell junctions. Recombinant C-termini of ADAMTS10 and ADAMTS6, both of which induce focal adhesions, bind heparin and syndecan-4. However, cells overexpressing full-length ADAMTS6 lack heparan sulphate and focal adhesions, whilst depletion of ADAMTS6 induces a prominent glycocalyx. Thus ADAMTS10 and ADAMTS6 oppositely affect heparan sulphate-rich interfaces including focal adhesions. We previously showed that microfibril deposition requires fibronectin-induced focal adhesions, and cell-cell junctions in epithelial cultures. Here we reveal that ADAMTS6 causes a reduction in heparan sulphate-rich interfaces, and its expression is regulated by ADAMTS10.


Asunto(s)
Proteínas ADAMTS/metabolismo , Células Epiteliales/fisiología , Adhesiones Focales , Uniones Intercelulares , Proteínas ADAMTS/genética , Análisis Mutacional de ADN , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos
9.
Sci Rep ; 6: 34347, 2016 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-27677855

RESUMEN

TGFß plays key roles in fibrosis and cancer progression, and latency is conferred by covalent linkage to latent TGFß binding proteins (LTBPs). LTBP1 is essential for TGFß folding, secretion, matrix localization and activation but little is known about its structure due to its inherent size and flexibility. Here we show that LTBP1 adopts an extended conformation with stable matrix-binding N-terminus, extended central array of 11 calcium-binding EGF domains and flexible TGFß-binding C-terminus. Moreover we demonstrate that LTBP1 forms short filament-like structures independent of other matrix components. The termini bind to each other to facilitate linear extension of the filament, while the N-terminal region can serve as a branch-point. Multimerization is enhanced in the presence of heparin and stabilized by the matrix cross-linking enzyme transglutaminase-2. These assemblies will extend the span of LTBP1 to potentially allow simultaneous N-terminal matrix and C-terminal fibrillin interactions providing tethering for TGFß activation by mechanical force.

10.
Methods ; 96: 85-96, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26608109

RESUMEN

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.


Asunto(s)
Fibronectinas/química , Ensayos Analíticos de Alto Rendimiento , Células Madre Pluripotentes Inducidas/ultraestructura , Imagen Molecular/métodos , Fenotipo , Secuencia de Aminoácidos , Adhesión Celular , Diferenciación Celular , Línea Celular , Células Nutrientes/citología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Datos de Secuencia Molecular , Péptidos/química
11.
Stem Cell Reports ; 4(3): 473-88, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25684225

RESUMEN

Mesenchymal progenitor cells have great therapeutic potential, yet incomplete characterization of their cell-surface interface limits their clinical exploitation. We have employed subcellular fractionation with quantitative discovery proteomics to define the cell-surface interface proteome of human bone marrow mesenchymal stromal/stem cells (MSCs) and human umbilical cord perivascular cells (HUCPVCs). We compared cell-surface-enriched fractions from MSCs and HUCPVCs (three donors each) with adult mesenchymal fibroblasts using eight-channel isobaric-tagging mass spectrometry, yielding relative quantification on >6,000 proteins with high confidence. This approach identified 186 upregulated mesenchymal progenitor biomarkers. Validation of 10 of these markers, including ROR2, EPHA2, and PLXNA2, confirmed upregulated expression in mesenchymal progenitor populations and distinct roles in progenitor cell proliferation, migration, and differentiation. Our approach has delivered a cell-surface proteome repository that now enables improved selection and characterization of human mesenchymal progenitor populations.


Asunto(s)
Antígenos de Superficie/metabolismo , Células Madre Mesenquimatosas/metabolismo , Células Madre Multipotentes/metabolismo , Proteoma , Proteómica , Adulto , Biomarcadores , Linaje de la Célula/genética , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Multipotentes/citología , Fenotipo , Proteómica/métodos , Interferencia de ARN , ARN Interferente Pequeño/genética , Reproducibilidad de los Resultados , Nicho de Células Madre , Adulto Joven
12.
Ann Rheum Dis ; 74(6): 1249-56, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24442880

RESUMEN

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.


Asunto(s)
Cromosomas Humanos Par 8/genética , Duplicación de Gen , Factor 6 de Diferenciación de Crecimiento/genética , Deformidades Congénitas de la Mano/genética , Artropatías/congénito , Osificación Heterotópica/genética , Esclerodermia Sistémica/genética , Sindecano-2/genética , Adulto , Anciano , Preescolar , Matriz Extracelular/metabolismo , Facies , Femenino , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Factor 6 de Diferenciación de Crecimiento/metabolismo , Deformidades Congénitas de la Mano/metabolismo , Deformidades Congénitas de la Mano/fisiopatología , Humanos , Lactante , Artropatías/genética , Artropatías/metabolismo , Artropatías/fisiopatología , Masculino , Persona de Mediana Edad , Osificación Heterotópica/metabolismo , Osificación Heterotópica/fisiopatología , Fenotipo , Transducción de Señal , Sindecano-2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Adulto Joven
13.
Colloids Surf B Biointerfaces ; 116: 544-52, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24572497

RESUMEN

Current vascular biomaterials exhibit poor biocompatibility characterised by failure to promote endothelialisation, predisposition to neoinitmal hyperplasia and excessive thrombogenicity. Fibrillin-1, a major constituent of microfibrils is associated with elastic fibres in the arterial wall. Fibrillin-1 binds to endothelial cells through an RGD cell adhesion motif in the fourth TB module. The RGD motif is present in PF8, a recombinant fibrillin-1 fragment. We investigated the potential of PF8 to improve the biocompatibility of PTFE. PF8 enhanced endothelial cell attachment and cell proliferation to a greater extent than fibronectin (p<0.01). PF8 immobilised on PTFE using plasma immersion ion implantation (PIII), retained these favourable cell interactive properties, again promoting endothelial cell attachment and proliferation. The thrombogenicity of covalently bound PF8 on PTFE was assessed in both static and dynamic conditions. In static conditions, uncoated PIII treated PTFE was more thrombogenic than untreated PTFE, while PF8 coating reduced thrombogenicity. Under flow, there was no difference in the thrombogenicity of PF8 coated PTFE and untreated PTFE. Immobilised PF8 shows a striking ability to promote attachment and growth of endothelial cells on PTFE, while providing a non-thrombogenic surface. These features make PF8 a promising candidate to improve the biocompatibility of current synthetic vascular grafts.


Asunto(s)
Materiales Biocompatibles/química , Células Endoteliales/química , Proteínas de Microfilamentos/química , Politetrafluoroetileno/química , Adhesión Celular , Proliferación Celular , Fibrilina-1 , Fibrilinas , Células HEK293 , Humanos
14.
J Cell Sci ; 127(Pt 1): 158-71, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24190885

RESUMEN

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.


Asunto(s)
Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/genética , Microfibrillas/metabolismo , Proteínas de Microfilamentos/genética , Actinas/genética , Actinas/metabolismo , Actomiosina/genética , Actomiosina/metabolismo , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular , Células Epiteliales/ultraestructura , Femenino , Fibrilina-1 , Fibrilinas , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Fibronectinas/genética , Fibronectinas/metabolismo , Regulación de la Expresión Génica , Heparitina Sulfato/metabolismo , Humanos , Integrinas/genética , Integrinas/metabolismo , Uniones Intercelulares/metabolismo , Uniones Intercelulares/ultraestructura , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/ultraestructura , Microfibrillas/ultraestructura , Proteínas de Microfilamentos/metabolismo , Especificidad de Órganos , Podocitos/metabolismo , Podocitos/ultraestructura , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/ultraestructura , Sindecano-4/genética , Sindecano-4/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Proteína de la Zonula Occludens-1/genética , Proteína de la Zonula Occludens-1/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
15.
Expert Rev Mol Med ; 15: e8, 2013 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-23962539

RESUMEN

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.


Asunto(s)
Enfermedad , Tejido Elástico , Salud , Animales , Tejido Elástico/química , Tejido Elástico/metabolismo , Humanos
16.
PLoS One ; 7(11): e48634, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23133647

RESUMEN

The extracellular glycoprotein fibrillin-1 forms microfibrils that act as the template for elastic fibers. Most mutations in fibrillin-1 cause Marfan syndrome with severe cardiovascular and ocular symptoms, and tall stature. This is in contrast to mutations within a heparin-binding TB domain (TB5), which is downstream of the arg-gly-asp cell adhesion domain, which can cause Weill-Marchesani syndrome (WMS) or Acromicric (AD) and Geleophysic Dysplasias (GD). WMS is characterized by short limbs, joint stiffness and ocular defects, whilst fibrillin-1 AD and GD have severe short stature, joint defects and thickened skin. We previously showed that TB5 binds heparin. Here, we show that the corresponding region of fibrillin-2 binds heparin very poorly, highlighting a novel functional difference between the two isoforms. This finding enabled us to map heparin/heparan sulfate binding to two sites on fibrillin-1 TB5 using a mutagenesis approach. Once these sites were mapped, we were able to investigate whether disease-causing mutations in this domain disrupt binding to HS. We show that a WMS deletion mutant, and five AD and GD point mutants all have disrupted heparin binding to TB5. These data provide insights into the biology of fibrillins and the pathologies of WMS, AD and GD.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Heparitina Sulfato/metabolismo , Deformidades Congénitas de las Extremidades/genética , Proteínas de Microfilamentos/genética , Mutación , Síndrome de Weill-Marchesani/genética , Sitios de Unión , Línea Celular , Fibrilina-1 , Fibrilina-2 , Fibrilinas , Fibroblastos/citología , Eliminación de Gen , Células HEK293 , Humanos , Mutagénesis , Oligosacáridos/química , Isoformas de Proteínas , Estructura Terciaria de Proteína , Proteínas Recombinantes/química
17.
Stem Cells ; 30(3): 548-60, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22213560

RESUMEN

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.


Asunto(s)
Forma de la Célula/efectos de los fármacos , Proteínas de Homeodominio/metabolismo , Indanos/farmacología , Células Madre Mesenquimatosas/fisiología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Pirazoles/farmacología , Quinolinas/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Actomiosina/metabolismo , Adulto , Diferenciación Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Tamaño del Núcleo Celular/efectos de los fármacos , Células Cultivadas , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Humanos , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/metabolismo , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-abl/genética , Proteínas Proto-Oncogénicas c-abl/metabolismo , Interferencia de ARN , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Adulto Joven
18.
J Biol Chem ; 286(46): 40266-75, 2011 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-21908605

RESUMEN

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.


Asunto(s)
Colágeno Tipo VI/química , Microfibrillas/química , Multimerización de Proteína/fisiología , Colágeno Tipo VI/genética , Colágeno Tipo VI/metabolismo , Células HEK293 , Humanos , Microfibrillas/genética , Microfibrillas/metabolismo , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína
19.
J Cell Sci ; 124(Pt 8): 1288-300, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21429937

RESUMEN

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.


Asunto(s)
Movimiento Celular , Fibronectinas/metabolismo , Integrina alfa5beta1/metabolismo , Células Madre Mesenquimatosas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal , Adulto , Becaplermina , Adhesión Celular , Células Cultivadas , Femenino , Regulación de la Expresión Génica , Humanos , Integrina alfa5beta1/genética , Masculino , Células Madre Mesenquimatosas/citología , Proteínas Proto-Oncogénicas c-sis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Adulto Joven
20.
Stem Cell Res ; 6(3): 238-50, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21420373

RESUMEN

Mechanisms underlying the vascular differentiation of human bone marrow stromal cells (HBMSCs) and their contribution to neovascularisation are poorly understood. We report the essential role of cell density-induced signals in directing HBMSCs along endothelial or smooth muscle lineages. Plating HBMSCs at high density rapidly induced Notch signaling, which initiated HBMSC commitment to a vascular progenitor cell population expressing markers for both vascular lineages. Notch also induced VEGF-A, which inhibited vascular smooth muscle commitment while consolidating differentiation to endothelial cells with cobblestone morphology and characteristic endothelial markers and functions. These mechanisms can be exploited therapeutically to regulate HBMSCs during neovascularisation.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular , Linaje de la Célula , Músculo Liso Vascular/citología , Células del Estroma/citología , Células de la Médula Ósea/metabolismo , Recuento de Células , Células Cultivadas , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Humanos , Músculo Liso Vascular/metabolismo , Células del Estroma/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...