Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Clin Genet ; 97(3): 396-406, 2020 03.
Article in English | MEDLINE | ID: mdl-31794058

ABSTRACT

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.


Subject(s)
Collagen Type I/genetics , Connective Tissue Diseases/classification , Ehlers-Danlos Syndrome/classification , Genetic Variation , Osteogenesis Imperfecta/classification , Adolescent , Adult , Child , Child, Preschool , Collagen Type I/ultrastructure , Collagen Type I, alpha 1 Chain , Connective Tissue/ultrastructure , Connective Tissue Diseases/genetics , Demography , Ehlers-Danlos Syndrome/genetics , Female , Heterozygote , Humans , Infant , Male , Middle Aged , Osteogenesis Imperfecta/genetics , Phenotype , Young Adult
2.
Hum Mutat ; 40(10): 1886-1898, 2019 10.
Article in English | MEDLINE | ID: mdl-31250519

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Extracellular Matrix/metabolism , Genetic Variation , Haploinsufficiency , Homeostasis , Adaptor Proteins, Signal Transducing/chemistry , Amino Acid Sequence , Cell Line , Cell Proliferation , DNA Mutational Analysis , Fibroblasts/metabolism , Humans , MAP Kinase Kinase Kinases/metabolism , Mutation , Nonsense Mediated mRNA Decay , Phosphorylation , Protein Binding , Signal Transduction
3.
Biochim Biophys Acta Mol Basis Dis ; 1864(4 Pt A): 1010-1023, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29309923

ABSTRACT

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.


Subject(s)
Dermis/metabolism , Ehlers-Danlos Syndrome/metabolism , Integrin alphaVbeta3/metabolism , Joint Instability/metabolism , Myofibroblasts/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Snail Family Transcription Factors/metabolism , Dermis/pathology , Ehlers-Danlos Syndrome/genetics , Ehlers-Danlos Syndrome/pathology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Female , Humans , Integrin alphaVbeta3/genetics , Joint Instability/genetics , Joint Instability/pathology , Male , Myofibroblasts/pathology , Protein Serine-Threonine Kinases/genetics , Snail Family Transcription Factors/genetics
4.
Brain ; 140(3): 555-567, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28073787

ABSTRACT

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.


Subject(s)
Collagen Type VI/genetics , Family Health , Genetic Variation/genetics , Peripheral Nervous System Diseases/genetics , Pruritus/genetics , Adult , DNA Mutational Analysis , Female , Humans , Male , Middle Aged , Peripheral Nervous System Diseases/complications , Pruritus/complications , Pruritus/pathology , Skin/innervation , Skin/metabolism , Skin/pathology
5.
Int J Mol Sci ; 19(4)2018 Mar 26.
Article in English | MEDLINE | ID: mdl-29587413

ABSTRACT

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.


Subject(s)
Arteries/abnormalities , Ehlers-Danlos Syndrome/metabolism , Integrin alphaVbeta3/metabolism , Joint Instability/metabolism , Skin Diseases, Genetic/metabolism , Vascular Malformations/metabolism , Arteries/metabolism , Collagen Type III/genetics , Collagen Type V/genetics , Ehlers-Danlos Syndrome/genetics , Humans , Joint Instability/genetics , Signal Transduction , Skin Diseases, Genetic/genetics , Vascular Malformations/genetics
6.
Hum Mol Genet ; 24(23): 6769-87, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26376865

ABSTRACT

Arterial tortuosity syndrome (ATS) is an autosomal recessive connective tissue disorder caused by loss-of-function mutations in SLC2A10, which encodes facilitative glucose transporter 10 (GLUT10). The role of GLUT10 in ATS pathogenesis remains an enigma, and the transported metabolite(s), i.e. glucose and/or dehydroascorbic acid, have not been clearly elucidated. To discern the molecular mechanisms underlying the ATS aetiology, we performed gene expression profiling and biochemical studies on skin fibroblasts. Transcriptome analyses revealed the dysregulation of several genes involved in TGFß signalling and extracellular matrix (ECM) homeostasis as well as the perturbation of specific pathways that control both the cell energy balance and the oxidative stress response. Biochemical and functional studies showed a marked increase in ROS-induced lipid peroxidation sustained by altered PPARγ function, which contributes to the redox imbalance and the compensatory antioxidant activity of ALDH1A1. ATS fibroblasts also showed activation of a non-canonical TGFß signalling due to TGFBRI disorganization, the upregulation of TGFBRII and connective tissue growth factor, and the activation of the αvß3 integrin transduction pathway, which involves p125FAK, p60Src and p38 MAPK. Stable GLUT10 expression in patients' fibroblasts normalized redox homeostasis and PPARγ activity, rescued canonical TGFß signalling and induced partial ECM re-organization. These data add new insights into the ATS dysregulated biological pathways and definition of the pathomechanisms involved in this disorder.


Subject(s)
Arteries/abnormalities , Fibroblasts/metabolism , Glucose Transport Proteins, Facilitative/deficiency , Integrin alphaVbeta3/metabolism , Joint Instability/metabolism , Oxidative Stress , Signal Transduction , Skin Diseases, Genetic/metabolism , Transforming Growth Factor beta/physiology , Vascular Malformations/metabolism , Arteries/metabolism , Arteries/physiopathology , Extracellular Matrix/physiology , Fibroblasts/physiology , Gene Expression Profiling , Glucose Transport Proteins, Facilitative/genetics , Homeostasis , Humans , Joint Instability/physiopathology , Mutation , Skin/metabolism , Skin/physiopathology , Skin Diseases, Genetic/physiopathology , Vascular Malformations/physiopathology
7.
Int J Mol Sci ; 18(8)2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28829359

ABSTRACT

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.


Subject(s)
Arteries/abnormalities , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Joint Instability/metabolism , Skin Diseases, Genetic/metabolism , Vascular Malformations/metabolism , Arteries/metabolism , Fluorescent Antibody Technique , Humans , Intracellular Space/metabolism , Joint Instability/genetics , Microsomes/metabolism , Protein Binding , Protein Transport , Skin Diseases, Genetic/genetics , Vascular Malformations/genetics
8.
Am J Med Genet A ; 170A(1): 103-15, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26373698

ABSTRACT

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.


Subject(s)
Connective Tissue Diseases/pathology , Dermis/pathology , Ehlers-Danlos Syndrome/pathology , Fibroblasts/pathology , Mutation/genetics , Sulfotransferases/genetics , Adolescent , Adult , Child , Child, Preschool , Connective Tissue Diseases/genetics , Dermis/metabolism , Ehlers-Danlos Syndrome/genetics , Female , Fibroblasts/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Infant , Male , Middle Aged , Young Adult
9.
Am J Hum Genet ; 90(2): 201-16, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22265013

ABSTRACT

We report on an autosomal-recessive variant of Ehlers-Danlos syndrome (EDS) characterized by severe muscle hypotonia at birth, progressive scoliosis, joint hypermobility, hyperelastic skin, myopathy, sensorineural hearing impairment, and normal pyridinoline excretion in urine. Clinically, the disorder shares many features with the kyphoscoliotic type of EDS (EDS VIA) and Ullrich congenital muscular dystrophy. Linkage analysis in a large Tyrolean kindred identified a homozygous frameshift mutation in FKBP14 in two affected individuals. Based on the cardinal clinical characteristics of the disorder, four additional individuals originating from different European countries were identified who carried either homozygous or compound heterozygous mutations in FKBP14. FKBP14 belongs to the family of FK506-binding peptidyl-prolyl cis-trans isomerases (PPIases). ER-resident FKBPs have been suggested to act as folding catalysts by accelerating cis-trans isomerization of peptidyl-prolyl bonds and to act occasionally also as chaperones. We demonstrate that FKBP14 is localized in the endoplasmic reticulum (ER) and that deficiency of FKBP14 leads to enlarged ER cisterns in dermal fibroblasts in vivo. Furthermore, indirect immunofluorescence of FKBP14-deficient fibroblasts indicated an altered assembly of the extracellular matrix in vitro. These findings suggest that a disturbance of protein folding in the ER affecting one or more components of the extracellular matrix might cause the generalized connective tissue involvement in this disorder. FKBP14 mutation analysis should be considered in all individuals with apparent kyphoscoliotic type of EDS and normal urinary pyridinoline excretion, in particular in conjunction with sensorineural hearing impairment.


Subject(s)
Abnormalities, Multiple/genetics , Ehlers-Danlos Syndrome/genetics , Frameshift Mutation , Hearing Loss/genetics , Peptidylprolyl Isomerase/genetics , Adolescent , Amino Acids/urine , Child , Child, Preschool , Ehlers-Danlos Syndrome/urine , Endoplasmic Reticulum/genetics , Extracellular Matrix/genetics , Female , Fibroblasts/metabolism , Genetic Variation , Hearing Loss/urine , Heterozygote , Homozygote , Humans , Male , Middle Aged , Phenotype , Protein Folding , cis-trans-Isomerases/genetics
10.
Nat Genet ; 38(4): 452-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16550171

ABSTRACT

Arterial tortuosity syndrome (ATS) is an autosomal recessive disorder characterized by tortuosity, elongation, stenosis and aneurysm formation in the major arteries owing to disruption of elastic fibers in the medial layer of the arterial wall. Previously, we used homozygosity mapping to map a candidate locus in a 4.1-Mb region on chromosome 20q13.1 (ref. 2). Here, we narrowed the candidate region to 1.2 Mb containing seven genes. Mutations in one of these genes, SLC2A10, encoding the facilitative glucose transporter GLUT10, were identified in six ATS families. GLUT10 deficiency is associated with upregulation of the TGFbeta pathway in the arterial wall, a finding also observed in Loeys-Dietz syndrome, in which aortic aneurysms associate with arterial tortuosity. The identification of a glucose transporter gene responsible for altered arterial morphogenesis is notable in light of the previously suggested link between GLUT10 and type 2 diabetes. Our data could provide new insight on the mechanisms causing microangiopathic changes associated with diabetes and suggest that therapeutic compounds intervening with TGFbeta signaling represent a new treatment strategy.


Subject(s)
Arteries/pathology , Glucose Transport Proteins, Facilitative/physiology , Mutation , Neovascularization, Pathologic/genetics , Vascular Diseases/genetics , Amino Acid Sequence , Chromosome Mapping , Chromosomes, Human, Pair 20 , Female , Glucose Transport Proteins, Facilitative/chemistry , Glucose Transport Proteins, Facilitative/genetics , Humans , Male , Molecular Sequence Data , Pedigree , Sequence Homology, Amino Acid
11.
Am J Hum Genet ; 88(6): 767-777, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21664999

ABSTRACT

Extreme corneal fragility and thinning, which have a high risk of catastrophic spontaneous rupture, are the cardinal features of brittle cornea syndrome (BCS), an autosomal-recessive generalized connective tissue disorder. Enucleation is frequently the only management option for this condition, resulting in blindness and psychosocial distress. Even when the cornea remains grossly intact, visual function could also be impaired by a high degree of myopia and keratoconus. Deafness is another common feature and results in combined sensory deprivation. Using autozygosity mapping, we identified mutations in PRDM5 in families with BCS. We demonstrate that regulation of expression of extracellular matrix components, particularly fibrillar collagens, by PRDM5 is a key molecular mechanism that underlies corneal fragility in BCS and controls normal corneal development and maintenance. ZNF469, encoding a zinc finger protein of hitherto undefined function, has been identified as a quantitative trait locus for central corneal thickness, and mutations in this gene have been demonstrated in Tunisian Jewish and Palestinian kindreds with BCS. We show that ZNF469 and PRDM5, two genes that when mutated cause BCS, participate in the same regulatory pathway.


Subject(s)
DNA-Binding Proteins/genetics , Extracellular Matrix/genetics , Transcription Factors/genetics , Child , DNA Mutational Analysis , Ehlers-Danlos Syndrome/genetics , Ehlers-Danlos Syndrome/pathology , Extracellular Matrix/physiology , Eye Abnormalities , Female , Humans , Joint Instability/congenital , Male , Mutation , Pedigree , Skin Abnormalities
12.
Biochim Biophys Acta ; 1820(10): 1576-87, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22705941

ABSTRACT

BACKGROUND: Alternative splicing of EDA fibronectin (FN) region is a cell type- and development-regulated mechanism controlled by pathological processes, growth factors and extracellular matrix (ECM). Classic and vascular Ehlers-Danlos syndrome (cEDS and vEDS) are connective tissue disorders caused by COL5A1/COL5A2 and COL3A1 gene mutations, leading to an in vivo abnormal collagen fibrillogenesis and to an in vitro defective organisation in the ECM of type V (COLLV) and type III collagen (COLLIII). These defects induce the FN-ECM disarray and the decrease of COLLs and FN receptors, the α2ß1 and α5ß1 integrins. Purified COLLV and COLLIII restore the COLL-FN-ECMs in both EDS cell strains. METHODS: Real-time PCR, immunofluorescence microscopy, and Western blotting were used to investigate the effects of COLLs on FN1 gene expression, EDA region alternative splicing, EDA(+)-FN-ECM assembly, α5ß1 integrin and EDA(+)-FN-specific α9 integrin subunit organisation, α5ß1 integrin and FAK co-regulation in EDS fibroblasts. RESULTS: COLLV-treated cEDS and COLLIII-treated vEDS fibroblasts up-regulate the FN1 gene expression, modulate the EDA(+) mRNA maturation and increase the EDA(+)-FN levels, thus restoring a control-like FN-ECM, which elicits the EDA(+)-FN-specific α9ß1 integrin organisation, recruits the α5ß1 integrin and switches on the FAK binding and phosphorylation. CONCLUSION: COLLs regulate the EDA(+)-FN-ECM organisation at transcriptional and post-transcriptional level and activate the α5ß1-FAK complexes. COLLs also recruit the α9ß1 integrin involved in the assembly of the EDA(+)-FN-ECM in EDS cells. GENERAL SIGNIFICANCE: The knowledge of the COLLs-ECM role in FN isotype expression and in EDA(+)-FN-ECM-mediated signal transduction adds insights in the ECM remodelling mechanisms in EDS cells.


Subject(s)
Collagen Type III/pharmacology , Collagen Type V/pharmacology , Ehlers-Danlos Syndrome/pathology , Extracellular Matrix/drug effects , Fibroblasts/drug effects , Fibronectins/genetics , Fibronectins/metabolism , Case-Control Studies , Cells, Cultured , Ehlers-Danlos Syndrome/metabolism , Extracellular Matrix/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/physiology , Gene Expression Regulation/drug effects , Humans , Integrin alpha5beta1/metabolism , Integrins/metabolism , Male , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization/drug effects
13.
Eur J Hum Genet ; 31(5): 596-601, 2023 05.
Article in English | MEDLINE | ID: mdl-36599937

ABSTRACT

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.


Subject(s)
Loeys-Dietz Syndrome , Humans , Exons , Loeys-Dietz Syndrome/genetics , Loeys-Dietz Syndrome/pathology , Nonsense Mediated mRNA Decay , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I/genetics , Receptor, Transforming Growth Factor-beta Type I/metabolism
14.
Am J Med Genet A ; 158A(5): 1164-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22488877

ABSTRACT

Arterial tortuosity syndrome (ATS) is an autosomal recessive connective tissue disorder, mainly characterized by tortuosity and elongation of the large- and medium-sized arteries with predisposition to stenoses and aneurysms. ATS is caused by mutations in the SLC2A10 gene, encoding for the facilitative glucose transporter 10 (GLUT10) and is described typically in pediatric patients. We report on a 51-year-old woman, originally ascertained because of unexplained widespread chronic pain and positive family history of aortic malformation. The main findings included aged appearance, congenital joint hypermobility, joint instability complications, chronic fatigue syndrome, progressive painful joint stiffness, abdominal hernias, pelvic prolapses, multiple cardiac valve prolapses, varicose veins, easy bruising, and gingival recession. Vascular imaging revealed kinking and anomalous origin of the aortic arch branches, marked tortuosity of the aorta, pulmonary and most middle arteries, and a small aneurysm of the splenic artery. SLC2A10 analysis disclosed homozygosity for the novel c.1411+1G>A splice mutation, leading to a 41 amino acids GLUT10 internal deletion. Expression study by immunofluorescence using healthy control cells showed lack of membrane internalization of GLUT10 in patient's skin fibroblasts. This report describes the first splice-site SLC2A10 mutation and increases to 19 the repertoire of known mutations in this gene. Comparison with the few previously published adult patients with ATS contributes to the natural history of this condition, which is probably under diagnosed within the expanding family of inherited connective tissue disorders.


Subject(s)
Glucose Transport Proteins, Facilitative/genetics , Point Mutation , Arteries/abnormalities , Cells, Cultured , Connective Tissue Diseases/genetics , Female , Fibroblasts/pathology , Homozygote , Humans , Middle Aged , Skin/pathology
15.
Cells ; 11(24)2022 12 14.
Article in English | MEDLINE | ID: mdl-36552803

ABSTRACT

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.


Subject(s)
Ehlers-Danlos Syndrome , Joint Instability , Humans , RNA-Seq , Joint Instability/diagnosis , Joint Instability/genetics , Joint Instability/metabolism , Ehlers-Danlos Syndrome/genetics , Extracellular Matrix/metabolism , Fibroblasts/metabolism
16.
Cells ; 10(11)2021 11 19.
Article in English | MEDLINE | ID: mdl-34831458

ABSTRACT

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.


Subject(s)
Cell Differentiation , Dermis/pathology , Doxycycline/pharmacology , Ehlers-Danlos Syndrome/pathology , Extracellular Matrix/metabolism , Fibroblasts/pathology , Matrix Metalloproteinase Inhibitors/pharmacology , Myofibroblasts/pathology , Cell Differentiation/drug effects , Culture Media, Conditioned/pharmacology , Extracellular Matrix/drug effects , Gene Ontology , Humans , Molecular Targeted Therapy , Myofibroblasts/drug effects , Phenotype , Protein Interaction Maps/drug effects , Proteolysis/drug effects , Proteomics , Secretome
17.
Biochim Biophys Acta Mol Basis Dis ; 1867(4): 166051, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33383104

ABSTRACT

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.


Subject(s)
Ehlers-Danlos Syndrome/pathology , Fibroblasts/pathology , Myofibroblasts/pathology , Proteome/analysis , Adult , Cells, Cultured , Cytoskeleton/metabolism , Cytoskeleton/pathology , Ehlers-Danlos Syndrome/metabolism , Energy Metabolism , Female , Fibroblasts/metabolism , Humans , Middle Aged , Myofibroblasts/metabolism , Proteome/metabolism , Proteostasis , Signal Transduction , Skin/metabolism , Skin/pathology
18.
Biochim Biophys Acta Mol Basis Dis ; 1866(6): 165742, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32105826

ABSTRACT

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.


Subject(s)
Abnormalities, Multiple/genetics , Actins/genetics , Autophagy/genetics , Hearing Loss, Bilateral/genetics , MAP Kinase Kinase Kinases/genetics , Mitral Valve Insufficiency/genetics , Osteosclerosis/genetics , Abnormalities, Multiple/physiopathology , Adaptor Proteins, Signal Transducing/genetics , Child , Cytoskeleton/genetics , Female , Fibroblasts/metabolism , Hearing Loss, Bilateral/physiopathology , Humans , Loss of Function Mutation/genetics , Mitral Valve Insufficiency/physiopathology , Mutation/genetics , Osteosclerosis/physiopathology , Phosphorylation/genetics , Polymorphism, Single Nucleotide/genetics , Protein Binding/genetics , Signal Transduction/genetics , Transforming Growth Factor beta/genetics
19.
Biochim Biophys Acta ; 1783(6): 1177-88, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18405669

ABSTRACT

Extracellular matrix (ECM) binding to integrin receptors regulates cell cycle progression and survival. In adherent cells, ECM disassembly induces anoikis, the apoptotic pathway switched on by loss of adhesion. ECM-deficient Ehlers-Danlos syndrome (EDS) fibroblasts, to adhere to rare fibronectin (FN) fibrils, and to proliferate, only organize, as FN receptor, the alphavbeta3 integrin. We report that in EDS cells the alphavbeta3 integrin is bound to talin and vinculin, but not to tensin, and that actin cytoskeleton is disorganized. Furthermore, in EDS cells Bcl-2 is down-regulated and caspases are active. We provide evidence that the antibody-mediated alphavbeta3 integrin or the FN inhibition induces anoikis in EDS cells. The alphavbeta3 integrin transduces survival signals to pp60src-mediated tyrosine phosphorylated paxillin, instead than to FAK, and interacts with EGF receptor (EGFR). This complex, when activated by EGF and FN, signals for the rescue of EDS cells from anoikis. Therefore, EDS cells, through the alphavbeta3 integrin-EGFR complexes, engage a paxillin- but not FAK-mediated pathway of cell survival.


Subject(s)
Anoikis/physiology , Ehlers-Danlos Syndrome/pathology , ErbB Receptors/metabolism , Fibroblasts/metabolism , Fibronectins/metabolism , Focal Adhesion Kinase 1/metabolism , Integrin alphaVbeta3/metabolism , Actins/metabolism , Caspases/metabolism , Cell Adhesion/physiology , Cell Membrane/metabolism , Cells, Cultured , Cytoskeleton/metabolism , Ehlers-Danlos Syndrome/metabolism , Extracellular Matrix/metabolism , Fibroblasts/cytology , Humans , Integrin alpha5beta1/metabolism , Paxillin/metabolism , Phosphorylation , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , Skin/cytology , Skin/metabolism , Talin/metabolism , Tyrosine/metabolism , Vinculin/metabolism
20.
Genes (Basel) ; 10(8)2019 08 12.
Article in English | MEDLINE | ID: mdl-31409039

ABSTRACT

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.


Subject(s)
Ehlers-Danlos Syndrome/etiology , Animals , Autophagy , Ehlers-Danlos Syndrome/pathology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Transcriptome , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL