Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
2.
J Invest Dermatol ; 144(2): 284-295.e16, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37716648

ABSTRACT

Desmosomes are dynamic complex protein structures involved in cellular adhesion. Disruption of these structures by loss-of-function variants in desmosomal genes leads to a variety of skin- and heart-related phenotypes. In this study, we report TUFT1 as a desmosome-associated protein, implicated in epidermal integrity. In two siblings with mild skin fragility, woolly hair, and mild palmoplantar keratoderma but without a cardiac phenotype, we identified a homozygous splice-site variant in the TUFT1 gene, leading to aberrant mRNA splicing and loss of TUFT1 protein. Patients' skin and keratinocytes showed acantholysis, perinuclear retraction of intermediate filaments, and reduced mechanical stress resistance. Immunolabeling and transfection studies showed that TUFT1 is positioned within the desmosome and that its location is dependent on the presence of the desmoplakin carboxy-terminal tail. A Tuft1-knockout mouse model mimicked the patients' phenotypes. Altogether, this study reveals TUFT1 as a desmosome-associated protein, whose absence causes skin fragility, woolly hair, and palmoplantar keratoderma.


Subject(s)
Hair Diseases , Keratoderma, Palmoplantar , Skin Abnormalities , Animals , Humans , Mice , Desmoplakins/genetics , Desmoplakins/metabolism , Desmosomes/metabolism , Hair/metabolism , Hair Diseases/genetics , Hair Diseases/metabolism , Keratoderma, Palmoplantar/genetics , Keratoderma, Palmoplantar/metabolism , Skin/metabolism , Skin Abnormalities/metabolism
3.
Front Immunol ; 12: 773720, 2021.
Article in English | MEDLINE | ID: mdl-34899726

ABSTRACT

Anti-laminin 332 mucous membrane pemphigoid (MMP) is an autoimmune blistering disease characterized by predominant mucosal lesions and autoantibodies against laminin 332. The exact diagnosis of anti-laminin 332 MMP is important since nearly 30% of patients develop solid cancers. This study compared two independently developed diagnostic indirect immunofluorescence (IF) tests based on recombinant laminin 332 expressed in HEK239 cells (biochip mosaic assay) and the migration trails of cultured keratinocytes rich in laminin 332 (footprint assay). The sera of 54 anti-laminin 332 MMP, 35 non-anti-laminin 332 MMP, and 30 pemphigus vulgaris patients as well as 20 healthy blood donors were analyzed blindly and independently. Fifty-two of 54 and 54/54 anti-laminin 332 MMP sera were positive in the biochip mosaic and the footprint assay, respectively. In the 35 non-anti-laminin 332 MMP sera, 3 were positive in both tests and 4 others showed weak reactivity in the footprint assay. In conclusion, both assays are easy to perform, highly sensitive, and specific, which will further facilitate the diagnosis of anti-laminin 332 MMP.


Subject(s)
Autoantibodies/blood , Autoantigens/immunology , Biological Assay , Cell Adhesion Molecules/immunology , Pemphigoid, Benign Mucous Membrane/diagnosis , Adult , Aged , Aged, 80 and over , Autoantigens/genetics , Autoantigens/metabolism , Biomarkers/blood , Case-Control Studies , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Female , Fluorescent Antibody Technique, Indirect , Germany , HEK293 Cells , Humans , Japan , Keratinocytes/immunology , Keratinocytes/metabolism , Male , Middle Aged , Netherlands , Pemphigoid, Benign Mucous Membrane/blood , Pemphigoid, Benign Mucous Membrane/immunology , Predictive Value of Tests , Reproducibility of Results , Kalinin
4.
Methods Mol Biol ; 1559: 107-118, 2017.
Article in English | MEDLINE | ID: mdl-28063041

ABSTRACT

Particle bombardment of gold microparticles coated with plasmids, which are accelerated to high velocity, is used for transfection of cells within tissue. Using this method, cDNA encoding proteins of interest introduced into ex vivo living human skin enables studying of proteins of interest in real time. Here, technical aspects of particle bombardment of ex vivo skin are described using green fluorescent protein (GFP) as readout for efficiency. This method can be applied on numerous tissues, including in living model animals.


Subject(s)
Biolistics/methods , Green Fluorescent Proteins/genetics , Plasmids/metabolism , Skin/metabolism , Tissue Culture Techniques/methods , Animals , Biolistics/instrumentation , DNA, Complementary/genetics , DNA, Complementary/metabolism , Gene Expression , Genes, Reporter , Gold/chemistry , Green Fluorescent Proteins/metabolism , Helium , Humans , Microspheres , Particle Size , Plasmids/chemistry , Spectrometry, Fluorescence
6.
Hum Mol Genet ; 24(11): 3155-62, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25712130

ABSTRACT

PLEC, the gene encoding the cytolinker protein plectin, has eight tissue-specific isoforms in humans, arising by alternate splicing of the first exon. To date, all PLEC mutations that cause epidermolysis bullosa simplex (EBS) were found in exons common to all isoforms. Due to the ubiquitous presence of plectin in mammalian tissues, EBS from recessive plectin mutations is always associated with extracutaneous involvement including muscular dystrophy, pyloric atresia and cardiomyopathy. We studied a consanguineous family with sisters having isolated blistering suggesting EBS. Skin disease started with foot blisters at walking age and became generalized at puberty while sparing mucous membranes. DNA sequencing revealed a homozygous nonsense mutation (c.46C>T; p.Arg16X) in the first exon of the plectin variant encoding plectin isoform 1a (P1a). Immunofluorescence antigen mapping, transmission electron microscopy, western blot analysis and qRT-PCR were performed on patient skin and cultured keratinocytes, control myocardium and striated muscle samples. We found hypoplastic hemidesmosomes and intra-epidermal 'pseudo-junctional' cleavage fitting EBS. Screening for cardiomyopathy and muscle dystrophy showed no abnormalities. We report the first cases of autosomal-recessive EBS from P1a deficiency affecting skin, while mucous membranes, heart and muscle are spared. The dominant expression of the P1a isoform in epidermal basal cell layer and cultured keratinocytes suggests that mutations in the first exon of isoform 1a cause skin-only EBS without extracutaneous involvement. Our study characterizes yet another of the eight isoforms of plectin and adds a tissue-specific phenotype to the spectrum of 'plectinopathies' produced by mutations of unique first exons of this gene.


Subject(s)
Epidermolysis Bullosa Simplex/genetics , Plectin/genetics , Adult , Amino Acid Sequence , Base Sequence , Cells, Cultured , Consanguinity , DNA Mutational Analysis , Epidermolysis Bullosa Simplex/metabolism , Exons , Female , Genetic Association Studies , Humans , Molecular Sequence Data , Pedigree , Plectin/metabolism
8.
J Invest Dermatol ; 132(5): 1374-83, 2012 May.
Article in English | MEDLINE | ID: mdl-22318390

ABSTRACT

Mutations in the type XVII collagen gene (COL17A1) result in the blistering disorder non-Herlitz junctional epidermolysis bullosa (JEB-nH). The incidence of revertant mosaicism, also called "natural gene therapy", was identified in a cohort of 14 patients with JEB-nH caused by COL17A1 mutations in the Netherlands. Five different in vivo reversions, all correcting the germ-line COL17A1 mutation c.2237delG in exon 30, were found in four mosaic JEB-nH patients. The correcting DNA changes involved a wide variety of somatic mutations, from which an indel mutation (c.2228-101_2263+70delins15) and a large deletion of 2,165 base pairs (c.2227+153_2336-318del) have not been previously observed in patients with revertant mosaicism. Our results show that there is no preference for a repair mechanism. Moreover, revertant mosaicism was confirmed on a DNA level in 6 out of 10 generalized JEB-nH patients. Further, photo-material and clinical history of the other four generalized JEB-nH patients demonstrated that each patient has revertant skin patches. In contrast, revertant mosaicism was not detected in the four localized JEB-nH patients. The fact that so many, if not all, generalized JEB-nH COL17A1 patients have revertant patches offers opportunities for cell therapies in which the patient's own naturally corrected cells are used as a source.


Subject(s)
Autoantigens/genetics , DNA Repair , Epidermolysis Bullosa/genetics , Epidermolysis Bullosa/pathology , Non-Fibrillar Collagens/genetics , Adult , Aged , Autoantigens/metabolism , Child , Epidermolysis Bullosa/metabolism , Female , Genetic Therapy , Germ-Line Mutation , Humans , INDEL Mutation , Male , Middle Aged , Mosaicism , Non-Fibrillar Collagens/metabolism , Remission, Spontaneous , Sequence Analysis, DNA , Collagen Type XVII
9.
Arch Dermatol ; 148(2): 213-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22004882

ABSTRACT

BACKGROUND: Dystrophic epidermolysis bullosa is a genetic blistering disorder caused by mutations in the type VII collagen gene, COL7A1. In revertant mosaicism, germline mutations are corrected by somatic events resulting in a mosaic disease distribution. This "natural gene therapy" phenomenon long has been recognized in other forms of epidermolysis bullosa but only recently in dystrophic epidermolysis bullosa. OBSERVATIONS: We describe a 21-year-old man with recessive dystrophic epidermolysis bullosa carrying the homozygous c.6508C>T (p.Gln2170X) nonsense mutation who reported an unaffected skin patch on his neck where blisters never had occurred. Immunofluorescent type VII collagen staining was normal in 80% of the unaffected skin biopsy; however, it was strongly reduced in the affected skin. In the unaffected skin, the somatic nucleotide substitution c.6510G>T reverted the germline nonsense codon to tyrosine (p.Gln2170Tyr), thereby restoring functional protein production. CONCLUSIONS: Revertant mosaicism is considered rare in recessive dystrophic epidermolysis bullosa. However, it might be more common than previously anticipated because our patient is the third in whom revertant mosaicism was identified in a short period of time. The correction mechanism is different than that previously reported. Systematic examination of patients with recessive dystrophic epidermolysis bullosa, therefore, will likely reveal more patients with revertant patches. This is important because the natural gene therapy phenomenon may provide opportunities for revertant cell therapy.


Subject(s)
Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/genetics , Mosaicism , Adult , Codon, Nonsense , Collagen Type VII/analysis , DNA Mutational Analysis , Epidermolysis Bullosa Dystrophica/metabolism , Homozygote , Humans , Male , Young Adult
10.
J Dermatol Sci ; 56(1): 9-18, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19665875

ABSTRACT

BACKGROUND: The current classification of recessive dystrophic epidermolysis bullosa (RDEB) comprises two major subtypes: 'severe generalized RDEB' (RDEB-sev gen) with early-onset, extensive, generalized blistering and scarring, complete absence of type VII collagen, and bi-allelic COL7A1 null mutations; milder 'generalized other RDEB' (RDEB-O) with reduced-to-normal type VII collagen expression, and non-null genotypes. OBJECTIVE: To search for previously unrecognized phenotype-genotype correlations in 33 Dutch RDEB families. METHODS: We analyzed extensive clinical follow-up data, available for all patients up to 19 years, detailed type VII collagen immunostaining and genotypes, and correlated clinical phenotype to molecular phenotype and genotype. RESULTS: We identified 20 novel COL7A1 mutations. In 14 of 15 RDEB-sev gen patients type VII collagen was completely absent, one had strongly reduced type VII collagen, and all carried bi-allelic null mutations. Five of 11 RDEB-O patients developed pseudosyndactyly of the fingers preceded by skin atrophy and flexion contractures later in childhood and adolescence. All five had esophageal involvement and growth retardation. Type VII collagen immunostaining ranged from strongly reduced to slightly reduced in RDEB-O patients with pseudosyndactyly, whereas RDEB-O patients without pseudosyndactyly had slightly reduced to normal type VII collagen staining. There was no difference in genotypes between both groups, although we unexpectedly found bi-allelic null mutations in two of five RDEB-O patients with pseudosyndactyly. CONCLUSION: Pseudosyndactyly occurs in approximately half of RDEB-O patients when type VII collagen is strongly reduced. The prognosis in RDEB cannot always be simply predicted from the COL7A1 genotype.


Subject(s)
Collagen Type VII/genetics , Epidermolysis Bullosa Dystrophica/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Collagen Type VII/metabolism , Epidermolysis Bullosa Dystrophica/metabolism , Epidermolysis Bullosa Dystrophica/pathology , Exons/genetics , Female , Follow-Up Studies , Genes, Recessive , Genetic Predisposition to Disease , Genotype , Humans , Infant , Male , Microscopy, Electron, Transmission , Middle Aged , Mutation , Netherlands , Pedigree , Phenotype , Skin/pathology , Skin/ultrastructure , Young Adult
11.
J Dermatol Sci ; 43(1): 11-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16580182

ABSTRACT

BACKGROUND: Type XVII collagen is an important structural component of keratinocyte hemidesmosomes and its functional loss in genetic or autoimmune disease results in blistering of the skin. In neoplastic tissue aberrant expression is seen dependent on the stage of the tumor. While the sequence of the type XVII collagen encoding gene -COL17A1 - is now completely elucidated, the sequence of the 5'-untranslated region (UTR) of the mRNA is still unknown. Since UTRs can modulate translation efficiency, the determination of the UTR sequence is indispensable for understanding the regulation of translation of type XVII collagen mRNA. OBJECTIVE: To resolve the sequence of the 5'UTR of type XVII collagen mRNA and to analyse the promoter region for transcription motifs. METHODS: 5' Rapid amplification of cDNA ends (RACE) followed by sequence analysis and ribonuclease protection assays (RPA) were performed. RESULTS: RACE and sequence analysis revealed the presence of six different 5'UTRs for the type XVII collagen mRNA. The start points of these six transcripts differ but no alternative exons are used. The longest 5'UTR starts 220 nucleotides before the open reading frame, whereas the shortest UTR is only 89 nucleotides in length. RPA confirmed the RACE results and furthermore demonstrated that the 5'UTRs with lengths of 102 and 220 nucleotides are the two major transcripts. Transcription motif analysis of the 5' region of the COL17A gene demonstrated several binding sites for transcription factors including the Sp1 and activating protein-1 (AP-1) families. CONCLUSION: Type XVII collagen mRNA is alternatively transcribed, which may result in complex regulation of type XVII collagen.


Subject(s)
5' Untranslated Regions , Autoantigens/genetics , Non-Fibrillar Collagens/genetics , RNA, Messenger/genetics , 3' Untranslated Regions , Alternative Splicing , Base Sequence , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cells, Cultured , DNA Primers/genetics , Humans , Keratinocytes/metabolism , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Neoplasm/genetics , Collagen Type XVII
12.
Am J Hum Genet ; 77(5): 727-40, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16252234

ABSTRACT

Revertant mosaicism by somatic reversion of inherited mutations has been described for a number of genetic diseases. Several mechanisms can underlie this reversion process, such as gene conversion, crossing-over, true back mutation, and second-site mutation. Here, we report the occurrence of multiple corrections in two unrelated probands with revertant mosaicism of non-Herlitz junctional epidermolysis bullosa, an autosomal recessive genodermatosis due to mutations in the COL17A1 gene. Immunofluorescence microscopy and laser dissection microscopy, followed by DNA and RNA analysis, were performed on skin biopsy specimens. In patient 1, a true back mutation, 3781T-->C, was identified in the specimen from the arm, and a second-site mutation, 4463-1G-->A, which compensated for the frameshift caused by the inherited 4424-5insC mutation, was identified in the 3' splice site of exon 55 in a specimen from the middle finger. Patient 2 showed--besides two distinct gene conversion events in specimens from the arm and hand sites, both of which corrected the 1706delA mutation--a second-site mutation (3782G-->C) in an ankle specimen, which prevented the premature ending of the protein by the 3781C-->T nonsense mutation (R1226X). Thus, both inherited mutations, paternal as well as maternal, reverted at least once by different reversion events in distinct cell clusters in the described patients. The occurrence of multiple correcting mutations within the same patient indicates that in vivo reversion is less unusual than was generally thought. Furthermore, in the male patient, mosaic patterns of type XVII collagen-positive keratinocytes were present in clinically unaffected and affected skin. This latter observation makes it likely that reversion may be overlooked and may happen more often than expected.


Subject(s)
Autoantigens/genetics , Epidermolysis Bullosa/genetics , Gene Conversion , Non-Fibrillar Collagens/genetics , Aged , Autoantigens/physiology , Epidermolysis Bullosa/pathology , Female , Humans , Male , Microscopy, Confocal , Middle Aged , Mutation , Non-Fibrillar Collagens/physiology , Sequence Analysis, DNA , Skin/pathology , Collagen Type XVII
13.
J Invest Dermatol ; 119(6): 1275-81, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12485428

ABSTRACT

Integrin alpha6beta4 is a hemidesmosomal transmembrane molecule involved in maintaining basal cell-matrix adhesion through interaction of the large intracytoplasmic tail of the beta4 subunit with the keratin intermediate filament network, at least in part through its binding with plectin and BP180/type XVII collagen. Here we report a patient with predominant features of epidermolysis bullosa simplex due to a mutation in the integrin beta4 gene. The patient, a 49-y-old female, had mild blistering of hands and feet from birth on, dystrophy of the nails with onychogryposis, and enamel hypoplasia. She had no alopecia and no history of pyloric atresia. Electron microscopy and antigen mapping of a skin blister revealed that the level of separation was intraepidermal, low in the basal keratinocytes through the attachment plaque of the hemidesmosome. Immuno-fluorescence microscopy revealed absent binding of monoclonal antibody 450-11 A against the third fibronectin III repeat on the intracellular domain of integrin beta4, whereas binding was reduced with monoclonal antibodies recognizing epitopes on amino-terminal and carboxy-terminal ends of the polypeptide. At the molecular level the phenotype was caused by a novel 2 bp deletion 4733delCT in ITGB4, resulting in in-frame skipping of exon 36 and a deduced 50 amino acid deletion (1450-1499) within the third fibronectin type III repeat in the cytoplasmic domain of the integrin beta4 polypeptide. Immunoblot analysis demonstrated a 5 kDa shorter beta4 polypeptide. The 4733delCT mutation was heterozygously present in the DNA. The patient is also expected to be heterozygous for a null allele, as no full-size protein was detected in vitro and the epitope 450-11 A was absent in vivo. These data show that deletion of the third fibronectin type III repeat in the cytoplasmic domain of integrin beta4, which is thought to interact with BP180/type XVII collagen, is clinically pathogenic and results in a mild phenotype with predominant features of epidermolysis bullosa simplex.


Subject(s)
Epidermolysis Bullosa Simplex/genetics , Gene Deletion , Integrin beta4/chemistry , Integrin beta4/genetics , Base Sequence , Blister/genetics , Blister/pathology , Cytoplasm/chemistry , Cytoplasm/ultrastructure , DNA Mutational Analysis , Epidermolysis Bullosa Simplex/pathology , Exons , Female , Fluorescent Antibody Technique , Hemidesmosomes/chemistry , Hemidesmosomes/pathology , Heterozygote , Humans , Integrin alpha6beta4/analysis , Microscopy, Electron , Middle Aged , Molecular Sequence Data , Phenotype , Protein Structure, Tertiary , Skin/pathology
14.
J Invest Dermatol ; 118(4): 626-30, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11918708

ABSTRACT

A patient with recessive epidermolysis bullosa simplex due to a previously described homozygous KRT14 1842-2A-->C splice-site mutation was re-examined, because we unexpectedly found signs of revertant mosaicism. The germline mutation resulted in different aberrant transcripts containing premature termination codons, all leading to truncated keratin 14 proteins. Basal keratinocytes in skin and in culture completely lacked keratin 14 and intermediate filaments. From this keratin 14-/- patient we started cultures from a new skin biopsy and here, we serendipitously found keratinocytes that spontaneously expressed keratin 14. This biopsy had been taken from an area of skin that was clinically affected, because blisters could simply be evoked by gentle rubbing. Immunofluorescence and electron microscopy of additional biopsies from this skin area revealed a mosaic expression of keratin 14 and reappearance of intermediate filaments in basal keratinocytes. Immunoblotting showed a revertant keratin 14 polypeptide with seemingly normal molecular weight. DNA analysis of exon 2 and its flanking intron borders showed no additional mutations in the genomic KRT14 sequence. Analysis of mRNA isolated from mosaic skin keratinocytes revealed an additional in-frame transcript (1844T-->G, 1845Delta6) that codes for an abnormal keratin 14 polypeptide with a two residue deletion and one amino acid change. The re-expression of a revertant, albeit abnormal, keratin 14 polypeptide, so-called partial revertant mosaicism, accounts for the antibody staining pattern and for the reappearance of intermediate filaments, which however, are semifunctional and not able to revert the clinical phenotype. The combination of a keratin 14-positive and a keratin 14-negative cell population in epidermis as well as in cultured keratinocytes suggests that the cellular reversion might be caused by an endogenous factor. We hypothesize that a second somatic modulating factor in the genome that affects the processing of the mutant KRT14 pre-mRNA may underlie this phenomenon.


Subject(s)
Epidermolysis Bullosa Simplex/genetics , Epidermolysis Bullosa Simplex/pathology , Keratins/genetics , Mosaicism/pathology , Aged , Amino Acid Sequence , Cytoskeleton/genetics , Female , Genes, Recessive , Haplotypes , Humans , Keratin-14 , Keratinocytes/pathology , Molecular Sequence Data , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...