Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Eur J Hum Genet ; 31(5): 596-601, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36599937

RESUMEN

Pathogenic variants in TGFBR1 are a common cause of Loeys-Dietz syndrome (LDS) characterized by life-threatening aortic and arterial disease. Generally, these are missense changes in highly conserved amino acids in the serine-threonine kinase domain. Conversely, nonsense, frameshift, or specific missense changes in the ligand-binding extracellular domain cause multiple self-healing squamous epithelioma (MSSE) lacking the cardiovascular phenotype. Here, we report on two novel variants in the penultimate exon 8 of TGFBR1 were identified in 3 patients from two unrelated LDS families: both were predicted to cause frameshift and premature stop codons (Gln448Profs*15 and Cys446Asnfs*4) resulting in truncated TGFBR1 proteins lacking the last 43 and 56 amino acid residues, respectively. These were classified as variants of uncertain significance based on current criteria. Transcript expression analyses revealed both mutant alleles escaped nonsense-mediated mRNA decay. Functional characterization in patient's dermal fibroblasts showed paradoxically enhanced TGFß signaling, as observed for pathogenic missense TGFBR1 changes causative of LDS. In summary, we expanded the allelic repertoire of LDS-associated TGFBR1 variants to include truncating variants escaping nonsense-mediated mRNA decay. Our data highlight the importance of functional studies in variants interpretation for correct clinical diagnosis.


Asunto(s)
Síndrome de Loeys-Dietz , Humanos , Exones , Síndrome de Loeys-Dietz/genética , Síndrome de Loeys-Dietz/patología , Degradación de ARNm Mediada por Codón sin Sentido , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo
2.
Cells ; 11(24)2022 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-36552803

RESUMEN

Hypermobile Ehlers-Danlos syndrome (hEDS) and hypermobility spectrum disorders (HSD) are clinically overlapping connective tissue disorders of unknown etiology and without any validated diagnostic biomarker and specific therapies. Herein, we in-depth characterized the cellular phenotype and gene expression profile of hEDS and HSD dermal fibroblasts by immunofluorescence, amplicon-based RNA-seq, and qPCR. We demonstrated that both cell types show a common cellular trait, i.e., generalized extracellular matrix (ECM) disarray, myofibroblast differentiation, and dysregulated gene expression. Functional enrichment and pathway analyses clustered gene expression changes in different biological networks that are likely relevant for the disease pathophysiology. Specifically, the complex gene expression dysregulation (mainly involving growth factors, structural ECM components, ECM-modifying enzymes, cytoskeletal proteins, and different signal transducers), is expected to perturb many ECM-related processes including cell adhesion, migration, proliferation, and differentiation. Based on these findings, we propose a disease model in which an unbalanced ECM remodeling triggers a vicious cycle with a synergistic contribution of ECM degradation products and proinflammatory mediators leading to a functional impairment of different connective tissues reflecting the multisystemic presentation of hEDS/HSD patients. Our results offer many promising clues for translational research aimed to define molecular bases, diagnostic biomarkers, and specific therapies for these challenging connective tissue disorders.


Asunto(s)
Síndrome de Ehlers-Danlos , Inestabilidad de la Articulación , Humanos , RNA-Seq , Inestabilidad de la Articulación/diagnóstico , Inestabilidad de la Articulación/genética , Inestabilidad de la Articulación/metabolismo , Síndrome de Ehlers-Danlos/genética , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo
3.
Cells ; 10(11)2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34831458

RESUMEN

Hypermobile Ehlers-Danlos syndrome (hEDS) is the most frequent type of EDS and is characterized by generalized joint hypermobility and musculoskeletal manifestations which are associated with chronic pain, and mild skin involvement along with the presence of more than a few comorbid conditions. Despite numerous research efforts, no causative gene(s) or validated biomarkers have been identified and insights into the disease-causing mechanisms remain scarce. Variability in the spectrum and severity of symptoms and progression of hEDS patients' phenotype likely depend on a combination of age, gender, lifestyle, and the probable multitude of genes involved in hEDS. However, considering the clinical overlap with other EDS forms, which lead to abnormalities in extracellular matrix (ECM), it is plausible that the mechanisms underlying hEDS pathogenesis also affect the ECM to a certain extent. Herein, we performed a series of in vitro studies on the secretome of hEDS dermal fibroblasts that revealed a matrix metalloproteinases (MMPs) dysfunction as one of the major disease drivers by causing a detrimental feedback loop of excessive ECM degradation coupled with myofibroblast differentiation. We demonstrated that doxycycline-mediated inhibition of MMPs rescues in hEDS cells a control-like ECM organization and induces a partial reversal of their myofibroblast-like features, thus offering encouraging clues for translational studies confirming MMPs as a potential therapeutic target in hEDS with the expectation to improve patients' quality of life and alleviate their disabilities.


Asunto(s)
Diferenciación Celular , Dermis/patología , Doxiciclina/farmacología , Síndrome de Ehlers-Danlos/patología , Matriz Extracelular/metabolismo , Fibroblastos/patología , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Miofibroblastos/patología , Diferenciación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Matriz Extracelular/efectos de los fármacos , Ontología de Genes , Humanos , Terapia Molecular Dirigida , Miofibroblastos/efectos de los fármacos , Fenotipo , Mapas de Interacción de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proteómica , Secretoma
4.
Biochim Biophys Acta Mol Basis Dis ; 1867(4): 166051, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33383104

RESUMEN

Hypermobile Ehlers-Danlos syndrome (hEDS), mainly characterized by generalized joint hypermobility and its complications, minor skin changes, and apparently segregating with an autosomal dominant pattern, is still without a known molecular basis. Hence, its diagnosis is only clinical based on a strict set of criteria defined in the revised EDS nosology. Moreover, the hEDS phenotypic spectrum is wide-ranging and comprises multiple associated signs and symptoms shared with other heritable or acquired connective tissue disorders and chronic inflammatory diseases. In this complex scenario, we previously demonstrated that hEDS patients' skin fibroblasts show phenotypic features of myofibroblasts, widespread extracellular matrix (ECM) disarray, perturbation of ECM-cell contacts, and dysregulated expression of genes involved in connective tissue architecture and related to inflammatory and pain responses. Herein, the cellular proteome of 6 hEDS dermal myofibroblasts was compared to that of 12 control fibroblasts to deepen the knowledge on mechanisms involved in the disease pathogenesis. Qualitative and quantitative differences were assessed based on top-down and bottom-up approaches and some differentially expressed proteins were proofed by biochemical analyses. Proteomics disclosed the differential expression of proteins principally implicated in cytoskeleton organization, energy metabolism and redox balance, proteostasis, and intracellular trafficking. Our findings offer a comprehensive view of dysregulated protein networks and related pathways likely associated with the hEDS pathophysiology. The present results can be regarded as a starting point for future in-depth investigations aimed to decipher the functional impact of potential bioactive molecules for the development of targeted management and therapies.


Asunto(s)
Síndrome de Ehlers-Danlos/patología , Fibroblastos/patología , Miofibroblastos/patología , Proteoma/análisis , Adulto , Células Cultivadas , Citoesqueleto/metabolismo , Citoesqueleto/patología , Síndrome de Ehlers-Danlos/metabolismo , Metabolismo Energético , Femenino , Fibroblastos/metabolismo , Humanos , Persona de Mediana Edad , Miofibroblastos/metabolismo , Proteoma/metabolismo , Proteostasis , Transducción de Señal , Piel/metabolismo , Piel/patología
5.
Biochim Biophys Acta Mol Basis Dis ; 1866(6): 165742, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32105826

RESUMEN

Transforming growth factor beta-activated kinase 1 (TAK1) is a highly conserved kinase protein encoded by MAP3K7, and activated by multiple extracellular stimuli, growth factors and cytokines. Heterozygous variants in MAP3K7 cause the cardiospondylocarpofacial syndrome (CSCFS) which is characterized by short stature, dysmorphic facial features, cardiac septal defects with valve dysplasia, and skeletal anomalies. CSCFS has been described in seven patients to date and its molecular pathogenesis is only partially understood. Here, the functional effects of the MAP3K7 c.737-7A > G variant, previously identified in a girl with CSCFS and additional soft connective tissue features, were explored. This splice variant generates an in-frame insertion of 2 amino acid residues in the kinase domain of TAK1. Computational analysis revealed that this in-frame insertion alters protein dynamics in the kinase activation loop responsible for TAK1 autophosphorylation after binding with its interactor TAB1. Co-immunoprecipitation studies demonstrate that the ectopic expression of TAK1-mutated protein impairs its ability to physically bind TAB1. In patient's fibroblasts, MAP3K7 c.737-7A > G variant results in reduced TAK1 autophosphorylation and dysregulation of the downstream TAK1-dependent signaling pathway. TAK1 loss-of-function is associated with an impaired TGFß-mediated α-SMA cytoskeleton assembly and cell migration, and defective autophagy process. These findings contribute to our understanding of the molecular pathogenesis of CSCFS and might offer the rationale for the design of novel therapeutic targets.


Asunto(s)
Anomalías Múltiples/genética , Actinas/genética , Autofagia/genética , Pérdida Auditiva Bilateral/genética , Quinasas Quinasa Quinasa PAM/genética , Insuficiencia de la Válvula Mitral/genética , Osteosclerosis/genética , Anomalías Múltiples/fisiopatología , Proteínas Adaptadoras Transductoras de Señales/genética , Niño , Citoesqueleto/genética , Femenino , Fibroblastos/metabolismo , Pérdida Auditiva Bilateral/fisiopatología , Humanos , Mutación con Pérdida de Función/genética , Insuficiencia de la Válvula Mitral/fisiopatología , Mutación/genética , Osteosclerosis/fisiopatología , Fosforilación/genética , Polimorfismo de Nucleótido Simple/genética , Unión Proteica/genética , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/genética
6.
Clin Genet ; 97(3): 396-406, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31794058

RESUMEN

The 2017 classification of Ehlers-Danlos syndromes (EDS) identifies three types associated with causative variants in COL1A1/COL1A2 and distinct from osteogenesis imperfecta (OI). Previously, patients have been described with variable features of both disorders, and causative variants in COL1A1/COL1A2; but this phenotype has not been included in the current classification. Here, we expand and re-define this OI/EDS overlap as a missing EDS type. Twenty-one individuals from 13 families were reported, in whom COL1A1/COL1A2 variants were found after a suspicion of EDS. None of them could be classified as affected by OI or by any of the three recognized EDS variants associated with COL1A1/COL1A2. This phenotype is dominated by EDS-related features. OI-related features were limited to mildly reduced bone mass, occasional fractures and short stature. Eight COL1A1/COL1A2 variants were novel and five recurrent with a predominance of glycine substitutions affecting residues within the procollagen N-proteinase cleavage site of α1(I) and α2(I) procollagens. Selected variants were investigated by biochemical, ultrastructural and immunofluorescence studies. The pattern of observed changes in the dermis and in vitro for selected variants was more typical of EDS rather than OI. Our findings indicate the existence of a wider recognizable spectrum associated with COL1A1/COL1A2.


Asunto(s)
Colágeno Tipo I/genética , Enfermedades del Tejido Conjuntivo/clasificación , Síndrome de Ehlers-Danlos/clasificación , Variación Genética , Osteogénesis Imperfecta/clasificación , Adolescente , Adulto , Niño , Preescolar , Colágeno Tipo I/ultraestructura , Cadena alfa 1 del Colágeno Tipo I , Tejido Conectivo/ultraestructura , Enfermedades del Tejido Conjuntivo/genética , Demografía , Síndrome de Ehlers-Danlos/genética , Femenino , Heterocigoto , Humanos , Lactante , Masculino , Persona de Mediana Edad , Osteogénesis Imperfecta/genética , Fenotipo , Adulto Joven
7.
Sci Immunol ; 4(41)2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31784499

RESUMEN

Genetic etiologies of chronic mucocutaneous candidiasis (CMC) disrupt human IL-17A/F-dependent immunity at mucosal surfaces, whereas those of connective tissue disorders (CTDs) often impair the TGF-ß-dependent homeostasis of connective tissues. The signaling pathways involved are incompletely understood. We report a three-generation family with an autosomal dominant (AD) combination of CMC and a previously undescribed form of CTD that clinically overlaps with Ehlers-Danlos syndrome (EDS). The patients are heterozygous for a private splice-site variant of MAPK8, the gene encoding c-Jun N-terminal kinase 1 (JNK1), a component of the MAPK signaling pathway. This variant is loss-of-expression and loss-of-function in the patients' fibroblasts, which display AD JNK1 deficiency by haploinsufficiency. These cells have impaired, but not abolished, responses to IL-17A and IL-17F. Moreover, the development of the patients' TH17 cells was impaired ex vivo and in vitro, probably due to the involvement of JNK1 in the TGF-ß-responsive pathway and further accounting for the patients' CMC. Consistently, the patients' fibroblasts displayed impaired JNK1- and c-Jun/ATF-2-dependent induction of key extracellular matrix (ECM) components and regulators, but not of EDS-causing gene products, in response to TGF-ß. Furthermore, they displayed a transcriptional pattern in response to TGF-ß different from that of fibroblasts from patients with Loeys-Dietz syndrome caused by mutations of TGFBR2 or SMAD3, further accounting for the patients' complex and unusual CTD phenotype. This experiment of nature indicates that the integrity of the human JNK1-dependent MAPK signaling pathway is essential for IL-17A- and IL-17F-dependent mucocutaneous immunity to Candida and for the TGF-ß-dependent homeostasis of connective tissues.


Asunto(s)
Candidiasis Mucocutánea Crónica/inmunología , Enfermedades del Tejido Conjuntivo/inmunología , Interleucina-17/inmunología , Proteína Quinasa 8 Activada por Mitógenos/inmunología , Factor de Crecimiento Transformador beta/inmunología , Alelos , Células Cultivadas , Femenino , Humanos , Masculino , Proteína Quinasa 8 Activada por Mitógenos/genética , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Mutación
8.
Genes (Basel) ; 10(11)2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31731524

RESUMEN

The Ehlers-Danlos syndromes (EDS) constitute a clinically and genetically heterogeneous group of connective tissue disorders. Tenascin X (TNX) deficiency is a rare type of EDS, defined as classical-like EDS (clEDS), since it phenotypically resembles the classical form of EDS, though lacking atrophic scarring. Although most patients display a well-defined phenotype, the diagnosis of TNX-deficiency is often delayed or overlooked. Here, we described an additional patient with clEDS due to a homozygous null-mutation in the TNXB gene. A review of the literature was performed, summarizing the most important and distinctive clinical signs of this disorder. Characterization of the cellular phenotype demonstrated a distinct organization of the extracellular matrix (ECM), whereby clEDS distinguishes itself from most other EDS subtypes by normal deposition of fibronectin in the ECM and a normal organization of the α5ß1 integrin.


Asunto(s)
Síndrome de Ehlers-Danlos/genética , Mutación con Pérdida de Función , Tenascina/genética , Adulto , Células Cultivadas , Síndrome de Ehlers-Danlos/patología , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/metabolismo , Humanos , Tenascina/metabolismo
9.
Genes (Basel) ; 10(8)2019 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-31409039

RESUMEN

The Ehlers‒Danlos syndromes (EDS) constitute a heterogenous group of connective tissue disorders characterized by joint hypermobility, skin abnormalities, and vascular fragility. The latest nosology recognizes 13 types caused by pathogenic variants in genes encoding collagens and other molecules involved in collagen processing and extracellular matrix (ECM) biology. Classical (cEDS), vascular (vEDS), and hypermobile (hEDS) EDS are the most frequent types. cEDS and vEDS are caused respectively by defects in collagen V and collagen III, whereas the molecular basis of hEDS is unknown. For these disorders, the molecular pathology remains poorly studied. Herein, we review, expand, and compare our previous transcriptome and protein studies on dermal fibroblasts from cEDS, vEDS, and hEDS patients, offering insights and perspectives in their molecular mechanisms. These cells, though sharing a pathological ECM remodeling, show differences in the underlying pathomechanisms. In cEDS and vEDS fibroblasts, key processes such as collagen biosynthesis/processing, protein folding quality control, endoplasmic reticulum homeostasis, autophagy, and wound healing are perturbed. In hEDS cells, gene expression changes related to cell-matrix interactions, inflammatory/pain responses, and acquisition of an in vitro pro-inflammatory myofibroblast-like phenotype may contribute to the complex pathogenesis of the disorder. Finally, emerging findings from miRNA profiling of hEDS fibroblasts are discussed to add some novel biological aspects about hEDS etiopathogenesis.


Asunto(s)
Síndrome de Ehlers-Danlos/etiología , Animales , Autofagia , Síndrome de Ehlers-Danlos/patología , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Transcriptoma , Cicatrización de Heridas
10.
Hum Mutat ; 40(10): 1886-1898, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31250519

RESUMEN

Transforming growth factor ß-activated kinase 1 (TAK1) mediates multiple biological processes through the nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) and the mitogen-activated protein kinase (MAPK) signaling pathways. TAK1 activation is tightly regulated by its binding partners (TABs). In particular, binding with TAB2 is crucial for cardiovascular development and extracellular matrix (ECM) homeostasis. In our previous work, we reported a novel multisystem disorder associated with the heterozygous TAB2 c.1398dup variant. Here, we dissect the functional effects of this variant in order to understand its molecular pathogenesis. We demonstrate that TAB2 c.1398dup considerably undergoes to nonsense-mediated messenger RNA decay and encodes a truncated protein that loses its ability to bind TAK1. We also show an alteration of the TAK1 autophosphorylation status and of selected downstream signaling pathways in patients' fibroblasts. Immunofluorescence analyses and ECM-related polymerase chain reaction-array panels highlight that patient fibroblasts display ECM disorganization and altered expression of selected ECM components and collagen-related pathways. In conclusion, we deeply dissect the molecular pathogenesis of the TAB2 c.1398dup variant and show that the resulting phenotype is well explained by TAB2 loss-of-function. Our data also offer initial insights on the ECM homeostasis impairment as a molecular mechanism probably underlying a multisystem disorder linked to TAB2.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Matriz Extracelular/metabolismo , Variación Genética , Haploinsuficiencia , Homeostasis , Proteínas Adaptadoras Transductoras de Señales/química , Secuencia de Aminoácidos , Línea Celular , Proliferación Celular , Análisis Mutacional de ADN , Fibroblastos/metabolismo , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , Mutación , Degradación de ARNm Mediada por Codón sin Sentido , Fosforilación , Unión Proteica , Transducción de Señal
11.
PLoS One ; 14(2): e0211647, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30716086

RESUMEN

Classical Ehlers-Danlos syndrome (cEDS) is a dominant inherited connective tissue disorder mainly caused by mutations in the COL5A1 and COL5A2 genes encoding type V collagen (COLLV), which is a fibrillar COLL widely distributed in a variety of connective tissues. cEDS patients suffer from skin hyperextensibility, abnormal wound healing/atrophic scars, and joint hypermobility. Most of the causative variants result in a non-functional COL5A1 allele and COLLV haploinsufficiency, whilst COL5A2 mutations affect its structural integrity. To shed light into disease mechanisms involved in cEDS, we performed gene expression profiling in skin fibroblasts from four patients harboring haploinsufficient and structural mutations in both disease genes. Transcriptome profiling revealed significant changes in the expression levels of different extracellular matrix (ECM)-related genes, such as SPP1, POSTN, EDIL3, IGFBP2, and C3, which encode both matricellular and soluble proteins that are mainly involved in cell proliferation and migration, and cutaneous wound healing. These gene expression changes are consistent with our previous protein findings on in vitro fibroblasts from other cEDS patients, which exhibited reduced migration and poor wound repair owing to COLLV disorganization, altered deposition of fibronectin into ECM, and an abnormal integrin pattern. Microarray analysis also indicated the decreased expression of DNAJB7, VIPAS39, CCPG1, ATG10, SVIP, which encode molecular chaperones facilitating protein folding, enzymes regulating post-Golgi COLLs processing, and proteins acting as cargo receptors required for endoplasmic reticulum (ER) proteostasis and implicated in the autophagy process. Patients' cells also showed altered mRNA levels of many cell cycle regulating genes including CCNE2, KIF4A, MKI67, DTL, and DDIAS. Protein studies showed that aberrant COLLV expression causes the disassembly of itself and many structural ECM constituents including COLLI, COLLIII, fibronectin, and fibrillins. Our findings provide the first molecular evidence of significant gene expression changes in cEDS skin fibroblasts highlighting that defective ECM remodeling, ER homeostasis and autophagy might play a role in the pathogenesis of this connective tissue disorder.


Asunto(s)
Síndrome de Ehlers-Danlos/genética , Síndrome de Ehlers-Danlos/patología , Fibroblastos/fisiología , Anomalías Cutáneas/genética , Anomalías Cutáneas/patología , Piel/patología , Transcriptoma/genética , Adulto , Alelos , Estudios de Casos y Controles , Ciclo Celular/genética , Células Cultivadas , Colágeno/genética , Tejido Conectivo/fisiología , Retículo Endoplásmico/genética , Matriz Extracelular/genética , Femenino , Fibronectinas/genética , Perfilación de la Expresión Génica/métodos , Haploinsuficiencia/genética , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , ARN Mensajero/genética
12.
Int J Mol Sci ; 19(4)2018 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-29587413

RESUMEN

The αvß3 integrin, an endothelial cells' receptor-binding fibronectin (FN) in the extracellular matrix (ECM) of blood vessels, regulates ECM remodeling during migration, invasion, angiogenesis, wound healing and inflammation, and is also involved in the epithelial mesenchymal transition. In vitro-grown human control fibroblasts organize a fibrillar network of FN, which is preferentially bound on the entire cell surface to its canonical α5ß1 integrin receptor, whereas the αvß3 integrin is present only in rare patches in focal contacts. We report on the preferential recruitment of the αvß3 integrin, due to the lack of FN-ECM and its canonical integrin receptor, in dermal fibroblasts from Ehlers-Danlos syndromes (EDS) and arterial tortuosity syndrome (ATS), which are rare multisystem connective tissue disorders. We review our previous findings that unraveled different biological mechanisms elicited by the αvß3 integrin in fibroblasts derived from patients affected with classical (cEDS), vascular (vEDS), hypermobile EDS (hEDS), hypermobility spectrum disorders (HSD), and ATS. In cEDS and vEDS, respectively, due to defective type V and type III collagens, αvß3 rescues patients' fibroblasts from anoikis through a paxillin-p60Src-mediated cross-talk with the EGF receptor. In hEDS and HSD, without a defined molecular basis, the αvß3 integrin transduces to the ILK-Snail1-axis inducing a fibroblast-to-myofibroblast-transition. In ATS cells, the deficiency of the dehydroascorbic acid transporter GLUT10 leads to redox imbalance, ECM disarray together with the activation of a non-canonical αvß3 integrin-TGFBRII signaling, involving p125FAK/p60Src/p38MAPK. The characterization of these different biological functions triggered by αvß3 provides insights into the multifaced nature of this integrin, at least in cultured dermal fibroblasts, offering future perspectives for research in this field.


Asunto(s)
Arterias/anomalías , Síndrome de Ehlers-Danlos/metabolismo , Integrina alfaVbeta3/metabolismo , Inestabilidad de la Articulación/metabolismo , Enfermedades Cutáneas Genéticas/metabolismo , Malformaciones Vasculares/metabolismo , Arterias/metabolismo , Colágeno Tipo III/genética , Colágeno Tipo V/genética , Síndrome de Ehlers-Danlos/genética , Humanos , Inestabilidad de la Articulación/genética , Transducción de Señal , Enfermedades Cutáneas Genéticas/genética , Malformaciones Vasculares/genética
13.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt A): 1010-1023, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29309923

RESUMEN

Hypermobile Ehlers-Danlos syndrome (hEDS) is a heritable connective tissue disorder with unknown molecular basis mainly characterized by generalized joint hypermobility, joint instability complications, and minor skin changes. The phenotypic spectrum is broad and includes multiple associated symptoms shared with chronic inflammatory systemic diseases. The stricter criteria defined in the 2017 EDS nosology leave without an identity many individuals with symptomatic joint hypermobility and/or features of hEDS; for these patients, the term Hypermobility Spectrum Disorders (HSD) was introduced. We previously reported that in vitro cultured hEDS and HSD patients' skin fibroblasts show a disarray of several extracellular matrix (ECM) components and dysregulated expression of genes involved in connective tissue homeostasis and inflammatory/pain/immune responses. Herein, we report that hEDS and HSD skin fibroblasts exhibit in vitro a similar myofibroblast-like phenotype characterized by the organization of α-smooth muscle actin cytoskeleton, expression of OB-cadherin/cadherin-11, enhanced migratory capability associated with augmented levels of the ECM-degrading metalloproteinase-9, and altered expression of the inflammation mediators CCN1/CYR61 and CCN2/CTGF. We demonstrate that in hEDS and HSD cells this fibroblast-to-myofibroblast transition is triggered by a signal transduction pathway that involves αvß3 integrin-ILK complexes, organized in focal adhesions, and the Snail1/Slug transcription factor, thus providing insights into the molecular mechanisms related to the pathophysiology of these protean disorders. The indistinguishable phenotype identified in hEDS and HSD cells resembles an inflammatory-like condition, which correlates well with the systemic phenotype of patients, and suggests that these multisystemic disorders might be part of a phenotypic continuum rather than representing distinct clinical entities.


Asunto(s)
Dermis/metabolismo , Síndrome de Ehlers-Danlos/metabolismo , Integrina alfaVbeta3/metabolismo , Inestabilidad de la Articulación/metabolismo , Miofibroblastos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Factores de Transcripción de la Familia Snail/metabolismo , Dermis/patología , Síndrome de Ehlers-Danlos/genética , Síndrome de Ehlers-Danlos/patología , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Humanos , Integrina alfaVbeta3/genética , Inestabilidad de la Articulación/genética , Inestabilidad de la Articulación/patología , Masculino , Miofibroblastos/patología , Proteínas Serina-Treonina Quinasas/genética , Factores de Transcripción de la Familia Snail/genética
14.
PLoS One ; 13(1): e0191220, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29346445

RESUMEN

Vascular Ehlers-Danlos syndrome (vEDS) is a dominantly inherited connective tissue disorder caused by mutations in the COL3A1 gene that encodes type III collagen (COLLIII), which is the major expressed collagen in blood vessels and hollow organs. The majority of disease-causing variants in COL3A1 are glycine substitutions and in-frame splice mutations in the triple helix domain that through a dominant negative effect are associated with the severe clinical spectrum potentially lethal of vEDS, characterized by fragility of soft connective tissues with arterial and organ ruptures. To shed lights into molecular mechanisms underlying vEDS, we performed gene expression profiling in cultured skin fibroblasts from three patients with different structural COL3A1 mutations. Transcriptome analysis revealed significant changes in the expression levels of several genes involved in maintenance of cell redox and endoplasmic reticulum (ER) homeostasis, COLLs folding and extracellular matrix (ECM) organization, formation of the proteasome complex, and cell cycle regulation. Protein analyses showed that aberrant COLLIII expression is associated with the disassembly of many structural ECM constituents, such as fibrillins, EMILINs, and elastin, as well as with the reduction of the proteoglycans perlecan, decorin, and versican, all playing an important role in the vascular system. Furthermore, the altered distribution of the ER marker protein disulfide isomerase PDI and the strong reduction of the COLLs-modifying enzyme FKBP22 are consistent with the disturbance of ER-related homeostasis and COLLs biosynthesis and post-translational modifications, indicated by microarray analysis. Our findings add new insights into the pathophysiology of this severe vascular disorder, since they provide a picture of the gene expression changes in vEDS skin fibroblasts and highlight that dominant negative mutations in COL3A1 also affect post-translational modifications and deposition into the ECM of several structural proteins crucial to the integrity of soft connective tissues.


Asunto(s)
Colágeno Tipo III/genética , Síndrome de Ehlers-Danlos/genética , Mutación , Sustitución de Aminoácidos , Ciclo Celular/genética , Células Cultivadas , Colágeno Tipo III/química , Colágeno Tipo III/metabolismo , Síndrome de Ehlers-Danlos/etiología , Síndrome de Ehlers-Danlos/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Humanos , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación Missense , Pliegue de Proteína , Procesamiento Proteico-Postraduccional , Piel/metabolismo
15.
Int J Mol Sci ; 18(8)2017 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-28829359

RESUMEN

GLUT10 belongs to a family of transporters that catalyze the uptake of sugars/polyols by facilitated diffusion. Loss-of-function mutations in the SLC2A10 gene encoding GLUT10 are responsible for arterial tortuosity syndrome (ATS). Since subcellular distribution of the transporter is dubious, we aimed to clarify the localization of GLUT10. In silico GLUT10 localization prediction suggested its presence in the endoplasmic reticulum (ER). Immunoblotting showed the presence of GLUT10 protein in the microsomal, but not in mitochondrial fractions of human fibroblasts and liver tissue. An even cytosolic distribution with an intense perinuclear decoration of GLUT10 was demonstrated by immunofluorescence in human fibroblasts, whilst mitochondrial markers revealed a fully different decoration pattern. GLUT10 decoration was fully absent in fibroblasts from three ATS patients. Expression of exogenous, tagged GLUT10 in fibroblasts from an ATS patient revealed a strict co-localization with the ER marker protein disulfide isomerase (PDI). The results demonstrate that GLUT10 is present in the ER.


Asunto(s)
Arterias/anomalías , Retículo Endoplásmico/metabolismo , Fibroblastos/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Inestabilidad de la Articulación/metabolismo , Enfermedades Cutáneas Genéticas/metabolismo , Malformaciones Vasculares/metabolismo , Arterias/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Espacio Intracelular/metabolismo , Inestabilidad de la Articulación/genética , Microsomas/metabolismo , Unión Proteica , Transporte de Proteínas , Enfermedades Cutáneas Genéticas/genética , Malformaciones Vasculares/genética
16.
Brain ; 140(3): 555-567, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28073787

RESUMEN

Itch is thought to represent the peculiar response to stimuli conveyed by somatosensory pathways shared with pain through the activation of specific neurons and receptors. It can occur in association with dermatological, systemic and neurological diseases, or be the side effect of certain drugs. However, some patients suffer from chronic idiopathic itch that is frequently ascribed to psychological distress and for which no biomarker is available to date. We investigated three multigenerational families, one of which diagnosed with joint hypermobility syndrome/Ehlers-Danlos syndrome hypermobility type (JHS/EDS-HT), characterized by idiopathic chronic itch with predominantly proximal distribution. Skin biopsy was performed in all eight affected members and revealed in six of them reduced intraepidermal nerve fibre density consistent with small fibre neuropathy. Whole exome sequencing identified two COL6A5 rare variants co-segregating with chronic itch in eight affected members and absent in non-affected members, and in one unrelated sporadic patient with type 1 painless diabetic neuropathy and chronic itch. Two families and the diabetic patient carried the nonsense c.6814G>T (p.Glu2272*) variant and another family carried the missense c.6486G>C (p.Arg2162Ser) variant. Both variants were predicted as likely pathogenic by in silico analyses. The two variants were rare (minor allele frequency < 0.1%) in 6271 healthy controls and absent in 77 small fibre neuropathy and 167 JHS/EDS-HT patients without itch. Null-allele test on cDNA from patients' fibroblasts of both families carrying the nonsense variant demonstrated functional haploinsufficiency due to activation of nonsense mediated RNA decay. Immunofluorescence microscopy and western blotting revealed marked disorganization and reduced COL6A5 synthesis, respectively. Indirect immunofluorescence showed reduced COL6A5 expression in the skin of patients carrying the nonsense variant. Treatment with gabapentinoids provided satisfactory itch relief in the patients carrying the mutations. Our findings first revealed an association between COL6A5 gene and familiar chronic itch, suggesting a new contributor to the pathogenesis of neuropathic itch and identifying a new candidate therapeutic target.


Asunto(s)
Colágeno Tipo VI/genética , Salud de la Familia , Variación Genética/genética , Enfermedades del Sistema Nervioso Periférico/genética , Prurito/genética , Adulto , Análisis Mutacional de ADN , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedades del Sistema Nervioso Periférico/complicaciones , Prurito/complicaciones , Prurito/patología , Piel/inervación , Piel/metabolismo , Piel/patología
17.
PLoS One ; 11(8): e0161347, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27518164

RESUMEN

Joint hypermobility syndrome/Ehlers-Danlos syndrome hypermobility type (JHS/EDS-HT), is likely the most common systemic heritable connective tissue disorder, and is mostly recognized by generalized joint hypermobility, joint instability complications, minor skin changes and a wide range of satellite features. JHS/EDS-HT is considered an autosomal dominant trait but is still without a defined molecular basis. The absence of (a) causative gene(s) for JHS/EDS-HT is likely attributable to marked genetic heterogeneity and/or interaction of multiple loci. In order to help in deciphering such a complex molecular background, we carried out a comprehensive immunofluorescence analysis and gene expression profiling in cultured skin fibroblasts from five women affected with JHS/EDS-HT. Protein study revealed disarray of several matrix structural components such as fibrillins, tenascins, elastin, collagens, fibronectin, and their integrin receptors. Transcriptome analysis indicated perturbation of different signaling cascades that are required for homeostatic regulation either during development or in adult tissues as well as altered expression of several genes involved in maintenance of extracellular matrix architecture and homeostasis (e.g., SPON2, TGM2, MMP16, GPC4, SULF1), cell-cell adhesion (e.g., CDH2, CHD10, PCDH9, CLDN11, FLG, DSP), immune/inflammatory/pain responses (e.g., CFD, AQP9, COLEC12, KCNQ5, PRLR), and essential for redox balance (e.g., ADH1C, AKR1C2, AKR1C3, MAOB, GSTM5). Our findings provide a picture of the gene expression profile and dysregulated pathways in JHS/EDS-HT skin fibroblasts that correlate well with the systemic phenotype of the patients.


Asunto(s)
Biomarcadores/metabolismo , Síndrome de Ehlers-Danlos/genética , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Inestabilidad de la Articulación/genética , Anomalías Cutáneas/genética , Adulto , Estudios de Casos y Controles , Células Cultivadas , Síndrome de Ehlers-Danlos/patología , Femenino , Fibroblastos/patología , Proteínas Filagrina , Humanos , Inestabilidad de la Articulación/patología , Persona de Mediana Edad , Fenotipo , Anomalías Cutáneas/patología , Síndrome
18.
FEBS Lett ; 590(11): 1630-40, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27153185

RESUMEN

Loss-of-function mutations in the gene encoding GLUT10 are responsible for arterial tortuosity syndrome (ATS), a rare connective tissue disorder. In this study GLUT10-mediated dehydroascorbic acid (DAA) transport was investigated, supposing its involvement in the pathomechanism. GLUT10 protein produced by in vitro translation and incorporated into liposomes efficiently transported DAA. Silencing of GLUT10 decreased DAA transport in immortalized human fibroblasts whose plasma membrane was selectively permeabilized. Similarly, the transport of DAA through endomembranes was markedly reduced in fibroblasts from ATS patients. Re-expression of GLUT10 in patients' fibroblasts restored DAA transport activity. The present results demonstrate that GLUT10 is a DAA transporter and DAA transport is diminished in the endomembranes of fibroblasts from ATS patients.


Asunto(s)
Arterias/anomalías , Ácido Deshidroascórbico/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Inestabilidad de la Articulación/genética , Enfermedades Cutáneas Genéticas/genética , Malformaciones Vasculares/genética , Ácido Ascórbico/metabolismo , Transporte Biológico/efectos de los fármacos , Transporte Biológico/genética , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Facilitadoras del Transporte de la Glucosa/antagonistas & inhibidores , Humanos , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/farmacología
19.
Am J Med Genet A ; 170A(1): 103-15, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26373698

RESUMEN

The musculocontractural type of Ehlers-Danlos syndrome (MC-EDS) has been recently recognized as a clinical entity. MC-EDS represents a differential diagnosis within the congenital neuromuscular and connective tissue disorders spectrum. Thirty-one and three patients have been reported with MC-EDS so far with bi-allelic mutations identified in CHST14 and DSE, respectively, encoding two enzymes necessary for dermatan sulfate (DS) biosynthesis. We report seven additional patients with MC-EDS from four unrelated families, including the follow-up of a sib-pair originally reported with the kyphoscoliotic type of EDS in 1975. Brachycephaly, a characteristic facial appearance, an asthenic build, hyperextensible and bruisable skin, tapering fingers, instability of large joints, and recurrent formation of large subcutaneous hematomas are always present. Three of seven patients had mildly elevated serum creatine kinase. The oldest patient was blind due to retinal detachment at 45 years and died at 59 years from intracranial bleeding; her affected brother died at 28 years from fulminant endocarditis. All patients in this series harbored homozygous, predicted loss-of-function CHST14 mutations. Indeed, DS was not detectable in fibroblasts from two unrelated patients with homozygous mutations. Patient fibroblasts produced higher amounts of chondroitin sulfate, showed intracellular retention of collagen types I and III, and lacked decorin and thrombospondin fibrils compared with control. A great proportion of collagen fibrils were not integrated into fibers, and fiber bundles were dispersed into the ground substance in one patient, all of which is likely to contribute to the clinical phenotype. This report should increase awareness for MC-EDS.


Asunto(s)
Enfermedades del Tejido Conjuntivo/patología , Dermis/patología , Síndrome de Ehlers-Danlos/patología , Fibroblastos/patología , Mutación/genética , Sulfotransferasas/genética , Adolescente , Adulto , Niño , Preescolar , Enfermedades del Tejido Conjuntivo/genética , Dermis/metabolismo , Síndrome de Ehlers-Danlos/genética , Femenino , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Lactante , Masculino , Persona de Mediana Edad , Adulto Joven
20.
Orphanet J Rare Dis ; 10: 145, 2015 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-26560304

RESUMEN

BACKGROUND: Brittle cornea syndrome (BCS) is a rare, generalized connective tissue disorder associated with extreme corneal thinning and a high risk of corneal rupture. Recessive mutations in transcription factors ZNF469 and PRDM5 cause BCS. Both transcription factors are suggested to act on a common pathway regulating extracellular matrix genes, particularly fibrillar collagens. We identified bilateral myopic choroidal neovascularization as the presenting feature of BCS in a 26-year-old-woman carrying a novel PRDM5 mutation (p.Glu134*). We performed immunohistochemistry of anterior and posterior segment ocular tissues, as expression of PRDM5 in the eye has not been described, or the effects of PRDM5-associated disease on the retina, particularly the extracellular matrix composition of Bruch's membrane. METHODS: Immunohistochemistry using antibodies against PRDM5, collagens type I, III, and IV was performed on the eyes of two unaffected controls and two patients (both with Δ9-14 PRDM5). Expression of collagens, integrins, tenascin and fibronectin in skin fibroblasts of a BCS patient with a novel p.Glu134* PRDM5 mutation was assessed using immunofluorescence. RESULTS: PRDM5 is expressed in the corneal epithelium and retina. We observe reduced expression of major components of Bruch's membrane in the eyes of two BCS patients with a PRDM5 Δ9-14 mutation. Immunofluorescence performed on skin fibroblasts from a patient with p.Glu134* confirms the generalized nature of extracellular matrix abnormalities in BCS. CONCLUSIONS: PDRM5-related disease is known to affect the cornea, skin and joints. Here we demonstrate, to the best of our knowledge for the first time, that PRDM5 localizes not only in the human cornea, but is also widely expressed in the retina. Our findings suggest that ECM abnormalities in PRDM5-associated disease are more widespread than previously reported.


Asunto(s)
Lámina Basal de la Coroides/metabolismo , Lámina Basal de la Coroides/patología , Proteínas de Unión al ADN/biosíntesis , Anomalías del Ojo/diagnóstico , Anomalías del Ojo/metabolismo , Inestabilidad de la Articulación/congénito , Anomalías Cutáneas/diagnóstico , Anomalías Cutáneas/metabolismo , Factores de Transcripción/biosíntesis , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Células Cultivadas , Niño , Proteínas de Unión al ADN/genética , Anomalías del Ojo/genética , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Inestabilidad de la Articulación/diagnóstico , Inestabilidad de la Articulación/genética , Inestabilidad de la Articulación/metabolismo , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Anomalías Cutáneas/genética , Factores de Transcripción/genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...