Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Invest Dermatol ; 144(6): 1334-1343.e14, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38157931

ABSTRACT

Junctional epidermolysis bullosa (JEB) is a rare autosomal recessive genodermatosis with a broad spectrum of phenotypes. Current genotype-phenotype paradigms are insufficient to accurately predict JEB subtype and characteristics from genotype, particularly for splice site variants, which account for over a fifth of disease-causing variants in JEB. This study evaluated the genetic and clinical findings from a JEB cohort, investigating genotype-phenotype correlations through bioinformatic analyses and comparison with previously reported variants. Eighteen unique variants in LAMB3, LAMA3, LAMC2, or COL17A1 were identified from 17 individuals. Seven had severe JEB, 9 had intermediate JEB, and 1 had laryngo-onycho-cutaneous syndrome. Seven variants were previously unreported. Deep phenotyping was completed for all intermediate JEB cases and demonstrated substantial variation between individuals. Splice site variants underwent analysis with SpliceAI, a state-of-the-art artificial intelligence tool, to predict resultant transcripts. Predicted functional effects included exon skipping and cryptic splice site activation, which provided potential explanations for disease severity and in most cases correlated with laminin-332 immunofluorescence. RT-PCR was performed for 1 case to investigate resultant transcripts produced from the splice site variant. This study expands the JEB genomic and phenotypic landscape. Artificial intelligence tools show potential for predicting the functional effects of splice site variants and may identify candidates for confirmatory laboratory investigation. Investigation of RNA transcripts will help to further elucidate genotype-phenotype correlations for novel variants.


Subject(s)
Collagen Type XVII , Epidermolysis Bullosa, Junctional , Genetic Association Studies , Kalinin , Laminin , Non-Fibrillar Collagens , Severity of Illness Index , Humans , Epidermolysis Bullosa, Junctional/genetics , Epidermolysis Bullosa, Junctional/pathology , Laminin/genetics , Male , Female , Non-Fibrillar Collagens/genetics , Child , Phenotype , Cell Adhesion Molecules/genetics , Child, Preschool , Autoantigens/genetics , RNA Splice Sites/genetics , Infant , Adolescent , Adult , Mutation , Young Adult , Genotype
3.
Pediatr Dermatol ; 38(5): 1094-1101, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34514630

ABSTRACT

BACKGROUND/OBJECTIVES: Laryngo-onycho-cutaneous syndrome (LOC) is a rare subtype of junctional epidermolysis bullosa (JEB), featuring aberrant granulation tissue formation in the skin, larynx, and eyes. So far, three mutations including the specific (founder) mutation in exon 39 of LAMA3 (c.151dup) have been identified, but sparse data exists regarding the natural history, the genotype-phenotype correlation, and its differentiation from other JEB types. METHODS: We reviewed our pediatric EB database to identify English children with clinical and genetically diagnosed LOC within the last 15 years. Their demographic, clinical, and laboratory data were examined. We searched three databases for case reports of LOC between January 1986 and November 2020 and extracted clinical and molecular details. RESULTS: We identified 6 LOC patients, all female (mean age 5.4 years). Periungual hypergranulation and skin fragility were the earliest presenting signs (0-3 months), followed by laryngeal stenosis, symblepharon (mean onset 10.7 and 11.8 months, respectively), and dental abnormalities. Five children developed anemia at an average of 19.2 months. We identified 22 published studies in English with 31 cases. CONCLUSIONS: This study delineates the disease course of LOC and highlights the overlap with some forms of JEB. Classical signs/symptoms including anemia appear early in life. Genetic analysis revealed three new LOC-associated variants and underscores the finding that interpretation of skin immunolabeling and molecular diagnostics can be challenging. We provide recommendations on management of this complex syndrome.


Subject(s)
Conjunctival Diseases , Epidermolysis Bullosa, Junctional , Laryngeal Diseases , Skin Abnormalities , Child , Child, Preschool , Female , Humans , Skin
4.
J Am Acad Dermatol ; 83(2): 447-454, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31786163

ABSTRACT

BACKGROUND: Recessive dystrophic epidermolysis bullosa (RDEB) is a hereditary blistering disorder due to a lack of type VII collagen. At present, treatment is mainly supportive. OBJECTIVES: To determine whether intravenous allogeneic bone marrow-derived mesenchymal stromal/stem cells (BM-MSCs) are safe in RDEB adults and if the cells improve wound healing and quality of life. METHODS: We conducted a prospective, phase I/II, open-label study recruiting 10 RDEB adults to receive 2 intravenous infusions of BM-MSCs (on day 0 and day 14; each dose 2-4 × 106 cells/kg). RESULTS: BM-MSCs were well tolerated with no serious adverse events to 12 months. Regarding efficacy, there was a transient reduction in disease activity scores (8/10 subjects) and a significant reduction in itch. One individual showed a transient increase in type VII collagen. LIMITATIONS: Open-label trial with no placebo. CONCLUSIONS: MSC infusion is safe in RDEB adults and can have clinical benefits for at least 2 months.


Subject(s)
Epidermolysis Bullosa Dystrophica/therapy , Mesenchymal Stem Cell Transplantation/methods , Pruritus/therapy , Adolescent , Adult , Aged , Epidermolysis Bullosa Dystrophica/complications , Epidermolysis Bullosa Dystrophica/diagnosis , Female , Humans , Infusions, Intravenous , Male , Middle Aged , Prospective Studies , Pruritus/diagnosis , Pruritus/etiology , Quality of Life , Severity of Illness Index , Transplantation, Homologous/methods , Treatment Outcome , Wound Healing , Young Adult
5.
JCI Insight ; 4(11)2019 06 06.
Article in English | MEDLINE | ID: mdl-31167965

ABSTRACT

BACKGROUNDRecessive dystrophic epidermolysis bullosa (RDEB) is a severe form of skin fragility disorder due to mutations in COL7A1 encoding basement membrane type VII collagen (C7), the main constituent of anchoring fibrils (AFs) in skin. We developed a self-inactivating lentiviral platform encoding a codon-optimized COL7A1 cDNA under the control of a human phosphoglycerate kinase promoter for phase I evaluation.METHODSIn this single-center, open-label phase I trial, 4 adults with RDEB each received 3 intradermal injections (~1 × 106 cells/cm2 of intact skin) of COL7A1-modified autologous fibroblasts and were followed up for 12 months. The primary outcome was safety, including autoimmune reactions against recombinant C7. Secondary outcomes included C7 expression, AF morphology, and presence of transgene in the injected skin.RESULTSGene-modified fibroblasts were well tolerated, without serious adverse reactions or autoimmune reactions against recombinant C7. Regarding efficacy, there was a significant (P < 0.05) 1.26-fold to 26.10-fold increase in C7 mean fluorescence intensity in the injected skin compared with noninjected skin in 3 of 4 subjects, with a sustained increase up to 12 months in 2 of 4 subjects. The presence of transgene (codon-optimized COL7A1 cDNA) was demonstrated in the injected skin at month 12 in 1 subject, but no new mature AFs were detected.CONCLUSIONTo our knowledge, this is the first human study demonstrating safety and potential efficacy of lentiviral fibroblast gene therapy with the presence of COL7A1 transgene and subsequent C7 restoration in vivo in treated skin at 1 year after gene therapy. These data provide a rationale for phase II studies for further clinical evaluation.TRIAL REGISTRATIONClincalTrials.gov NCT02493816.FUNDINGCure EB, Dystrophic Epidermolysis Bullosa Research Association (UK), UK NIHR Biomedical Research Centre at Guy's and St Thomas' NHS Foundation Trust and King's College London, and Fondation René Touraine Short-Exchange Award.


Subject(s)
Epidermolysis Bullosa Dystrophica/therapy , Fibroblasts , Genetic Therapy , Lentivirus/genetics , Adult , Collagen Type VII/genetics , Female , Fibroblasts/metabolism , Fibroblasts/transplantation , Genetic Therapy/adverse effects , Genetic Therapy/methods , Humans , Male , Middle Aged , Treatment Outcome
6.
Int J Dermatol ; 57(9): 1058-1067, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30011071

ABSTRACT

BACKGROUND: Epidermolysis bullosa (EB) is a clinically and genetically heterogeneous blistering skin disease, but in countries such as Kuwait, there are very limited data on the clinical and molecular pathology of EB. To improve understanding of EB in Kuwait, we report the experience of a local tertiary referral center over a 17.5 year period (January 2000-June 2017) in establishing clinical and molecular diagnoses. METHODS: Review of hospital records and diagnostic reports. Individual cases were diagnosed by combinations of clinical assessment, skin biopsy (immunohistochemistry and transmission electron microscopy), Sanger sequencing of EB genes, and whole exome sequencing. RESULTS: Fifty-four families with EB were registered with the clinic over this period, 41 of whom (84 patients) participated in diagnostic studies. Thirty-seven of these 41 families had consanguineous marriages; 34 had recessive forms of EB, while only seven had dominant subtypes. Recurrent mutations were observed in epidermal dystonin, transglutaminase 5, and type VII collagen. CONCLUSIONS: The prevalence of EB in Kuwait is approximately three times that of internationally cited rates with an over-representation of autosomal recessive variants. Establishing the molecular basis of EB in Kuwait with accurate diagnostic subtyping provides a basis for determining healthcare requirements and improving patient management of EB.


Subject(s)
Epidermolysis Bullosa/diagnosis , Epidermolysis Bullosa/genetics , Skin/pathology , Biopsy , Cell Adhesion Molecules/genetics , Collagen Type VII/genetics , Consanguinity , Desmoplakins/genetics , Dystonin/genetics , Epidermolysis Bullosa/pathology , Exome , Female , Genes, Dominant , Genes, Recessive , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Integrin beta4/genetics , Keratin-14/genetics , Keratin-5/genetics , Kuwait , Male , Transglutaminases/genetics , gamma Catenin/genetics , Kalinin
8.
BMC Dermatol ; 18(1): 1, 2018 01 20.
Article in English | MEDLINE | ID: mdl-29352809

ABSTRACT

BACKGROUND: Epidermolysis bullosa simplex with muscular dystrophy (EBS-MD; OMIM #226670) is an autosomal recessive disease, characterized mainly by skin blistering at birth or shortly thereafter, progressive muscle weakness, and rarely by alopecia. EBS-MD is caused by mutations in the PLEC gene (OMIM *601282), which encodes plectin, a structural protein expressed in several tissues, including epithelia and muscle. We describe a patient affected with EBS-MD and diffuse alopecia in which we identified a novel pathogenic mutation by PCR amplification of all coding exons and exon-intron boundaries of PLEC gene, followed by bidirectional Sanger sequencing. CASE PRESENTATION: The patient, a 28-year-old female and only child of consanguineous healthy parents, was born after uneventful pregnancy. At 2 days of age, she developed skin and oral mucosal blistering, accompanied by voice hoarseness. On physical examination as an adult, we observed diffuse non-scarring alopecia on the scalp, onychodystrophy (pachyonychia) in all 20 nails, dental decay, mild dysphonia, and severe muscle atrophy mainly affecting the extremities. Neurological examination showed profoundly diminished reflexes. Mutation analysis revealed the patient to be homozygous for the novel PLEC nonsense mutation - c.7159G > T (p.Glu2387*) - located in exon 31. Thismutation predicts the lack of expression of the full-length plectin isoform. CONCLUSION: The present case appears to be the second association of EBS-MD with diffuse alopecia, both cases having different mutations involving PLEC exon 31. It remains to be elucidated whether diffuse alopecia results from PLEC mutations and/or from environmental factors.


Subject(s)
Alopecia Areata/genetics , Epidermolysis Bullosa Simplex/genetics , Muscular Dystrophies, Limb-Girdle/genetics , Plectin/genetics , Adult , Child , Codon, Nonsense , Female , Homozygote , Humans , Male
11.
Am J Hum Genet ; 95(3): 308-14, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25152456

ABSTRACT

Grainyhead-like 2, encoded by GRHL2, is a member of a highly conserved family of transcription factors that play essential roles during epithelial development. Haploinsufficiency for GRHL2 has been implicated in autosomal-dominant deafness, but mutations have not yet been associated with any skin pathology. We investigated two unrelated Kuwaiti families in which a total of six individuals have had lifelong ectodermal defects. The clinical features comprised nail dystrophy or nail loss, marginal palmoplantar keratoderma, hypodontia, enamel hypoplasia, oral hyperpigmentation, and dysphagia. In addition, three individuals had sensorineural deafness, and three had bronchial asthma. Taken together, the features were consistent with an unusual autosomal-recessive ectodermal dysplasia syndrome. Because of consanguinity in both families, we used whole-exome sequencing to search for novel homozygous DNA variants and found GRHL2 mutations common to both families: affected subjects in one family were homozygous for c.1192T>C (p.Tyr398His) in exon 9, and subjects in the other family were homozygous for c.1445T>A (p.Ile482Lys) in exon 11. Immortalized keratinocytes (p.Ile482Lys) showed altered cell morphology, impaired tight junctions, adhesion defects, and cytoplasmic translocation of GRHL2. Whole-skin transcriptomic analysis (p.Ile482Lys) disclosed changes in genes implicated in networks of cell-cell and cell-matrix adhesion. Our clinical findings of an autosomal-recessive ectodermal dysplasia syndrome provide insight into the role of GRHL2 in skin development, homeostasis, and human disease.


Subject(s)
Cleft Palate/genetics , DNA-Binding Proteins/genetics , Ectodermal Dysplasia/genetics , Genes, Recessive/genetics , Intellectual Disability/genetics , Mutation/genetics , Skin/pathology , Syndactyly/genetics , Transcription Factors/genetics , Blotting, Western , Child , DNA-Binding Proteins/metabolism , Exons/genetics , Female , Humans , Male , Pedigree , Phenotype , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Skin/metabolism , Syndrome , Transcription Factors/metabolism
12.
Exp Dermatol ; 22(12): 825-31, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24279917

ABSTRACT

Individuals with inherited skin diseases often pose one of the most difficult diagnostic challenges in dermatology. The hunt for the underlying molecular pathology may involve candidate gene screening or linkage analysis, which is usually determined by the initial history, the physical findings and laboratory tests. Recent technical advances in DNA sequencing, however, are shifting the diagnostic paradigm. Notably, next-generation sequencing allows a more comprehensive approach to diagnosing inherited diseases, with potential savings of both time and money. In the setting of a paediatric dermatology genetics clinic in Kuwait, we therefore performed whole-exome sequencing on seven individuals without a priori detailed knowledge of the patients' disorders: from these sequencing data, we diagnosed X-linked hypohidrotic ectodermal dysplasia (two cases), acrodermatitis enteropathica, recessive erythropoietic protoporphyria (two siblings) and localized recessive dystrophic epidermolysis bullosa (two siblings). All these groups of disorders are clinically and genetically heterogeneous, but the sequencing data proved inherently useful in improving patient care and avoiding unnecessary investigations. Our observations highlight the value of whole-exome sequencing, in combination with robust bioinformatics analysis, in determining the precise molecular pathology and clinical diagnosis in patients with genetic skin disorders, notably at an early stage in the clinical evaluation of these often complex disorders and thereby support a new paradigm for future diagnostics.


Subject(s)
Dermatology/methods , Exome , High-Throughput Nucleotide Sequencing , Skin Diseases/genetics , Adolescent , Base Sequence , Child, Preschool , Computational Biology , Female , Genetic Predisposition to Disease , Genome, Human , Humans , Male , Molecular Diagnostic Techniques , Sequence Analysis, DNA
14.
J Invest Dermatol ; 130(6): 1551-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20164846

ABSTRACT

Epidermolysis bullosa (EB) is a group of autosomal dominant and recessive blistering skin diseases in which pathogenic mutations have been reported in 13 different genes encoding structural proteins involved in keratinocyte integrity, as well as cell-matrix or cell-cell adhesion. We now report an inherited skin fragility disorder with a homozygous nonsense mutation in the dystonin gene (DST) that encodes the coiled-coil domain of the epithelial isoform of bullous pemphigoid antigen 1, BPAG1-e (also known as BP230). The mutation, p.Gln1124X, leads to the loss of hemidesmosomal inner plaques and a complete absence of skin immunostaining for BPAG1-e, as well as reduced labeling for plectin, the beta4 integrin subunit, and for type XVII collagen. The 38-year-old affected individual has lifelong generalized trauma-induced spontaneous blisters and erosions, particularly around the ankles. In addition, he experiences episodic numbness in his limbs, which started at the age of 37 years. These neurological symptoms may also be due to DST gene mutation, although he has a concomitant diagnosis of CADASIL (cerebral arteriopathy, autosomal dominant, with subcortical infarcts and leukoencephalopathy), a cerebral small-vessel arteriopathy, which thus complicates the genotype-phenotype interpretation. With regard to skin blistering, the clinicopathological findings expand the molecular basis of EB by identifying BPAG1-e pathology in a new form of autosomal recessive EB simplex.


Subject(s)
Carrier Proteins/genetics , Codon, Nonsense/genetics , Cytoskeletal Proteins/genetics , Epidermolysis Bullosa Simplex/diagnosis , Epidermolysis Bullosa Simplex/genetics , Homozygote , Nerve Tissue Proteins/genetics , Adult , Blister/metabolism , Carrier Proteins/metabolism , Collagen Type VII/metabolism , Cytoskeletal Proteins/metabolism , Dystonin , Epidermolysis Bullosa Simplex/metabolism , Hemidesmosomes/metabolism , Humans , Integrin beta4/metabolism , Male , Nerve Tissue Proteins/metabolism , Plectin/metabolism , Protein Isoforms , Skin/metabolism
15.
J Pediatr Gastroenterol Nutr ; 47(5): 585-91, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18955862

ABSTRACT

BACKGROUND: Mutations in alpha6 or beta4 integrins (ITGA6, ITGB4) are known to cause junctional epidermolysis bullosa with pyloric atresia (JEB-PA), often lethal in infancy through skin desquamation. There is 1 report of pyloric atresia associated with a desquamatory enteropathy but without skin disease, of unknown molecular basis. PATIENTS AND METHODS: We report 2 Kuwaiti siblings with pyloric atresia and life-threatening intestinal desquamation without significant skin abnormality. The older sibling died of intractable diarrhoea, and the younger sibling suffered episodes of massive protein-losing enteropathy, triggered by viral infections, in addition to obstructive uropathy. Mutation analysis was performed for ITGA6 and ITGB4 and expression of ITGA6 and ITGB4 protein was examined in skin and intestinal biopsies. Her serum also was incubated with normal intestine. RESULTS: We identified a novel mutation in ITGB4, with homozygous deletion of a single residue (isoleucine 1314) within the intracellular plectin-binding domain. Expression of ITGA6 and ITGB4 within skin, duodenal, and colonic epithelium was normal or minimally reduced, in contrast to previous reports. Biopsies taken during relapse showed accumulation of immunoglobulin G and C1q within intestinal basement membrane, whereas immunoglobulin G from her serum bound to basement membrane of normal small intestine. Immunomodulatory therapy induced significant improvement following relapses. CONCLUSIONS: ITGB4 mutation may induce a desquamative enteropathy in infancy without significant skin disease. A history of pyloric atresia is important in infants with severe chronic diarrhoeal disease and should prompt investigation for JEB-PA associated mutations. Acquired immune responses may exacerbate primary genetic disorders of epithelial adhesion and immunomodulatory therapy may be beneficial.


Subject(s)
Digestive System Abnormalities/genetics , Enteritis/genetics , Epidermolysis Bullosa, Junctional/genetics , Integrin beta4/genetics , Mutation , Pylorus/pathology , Diarrhea/genetics , Diarrhea/pathology , Diarrhea/therapy , Digestive System Abnormalities/pathology , Digestive System Abnormalities/therapy , Enteritis/pathology , Enteritis/therapy , Epidermolysis Bullosa, Junctional/pathology , Epidermolysis Bullosa, Junctional/therapy , Female , Humans , Infant , Intestinal Mucosa/pathology , Intestines/pathology , Intestines/physiopathology , Parenteral Nutrition , Pylorus/abnormalities , Pylorus/physiopathology , Skin/pathology , Skin/physiopathology
17.
J Invest Dermatol ; 126(9): 2039-43, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16710310

ABSTRACT

The mechanobullous disease Hallopeau-Siemens recessive dystrophic epidermolysis bullosa (HS-RDEB) results from mutations in the type VII collagen gene (COL7A1) on chromosome 3p21.31. Typically, there are frameshift, splice site, or nonsense mutations on both alleles. In this report, we describe a patient with HS-RDEB, who was homozygous for a new frameshift mutation, 345insG, in exon 3 of COL7A1. However, sequencing of parental DNA showed that although the patient's mother was a heterozygous carrier of this mutation, the father's DNA contained only wild-type sequence. Microsatellite marker analysis confirmed paternity and genotyping of 28 microsatellites spanning chromosome 3 revealed that the affected child was homozygous for every marker tested with all alleles originating from a single maternal chromosome 3. Thus, the HS-RDEB phenotype in this patient is due to complete maternal isodisomy of chromosome 3 and reduction to homozygosity of the mutant COL7A1 gene locus. To our knowledge, there are no published reports of uniparental disomy (UPD) in HS-RDEB; moreover, this case represents only the third example of UPD of chromosome 3 to be reported. The severity of the HS-RDEB in this case was similar to other affected individuals and no additional phenotypic abnormalities were observed, suggesting an absence of maternally imprinted genes on chromosome 3.


Subject(s)
Chromosomes, Human, Pair 3 , Epidermolysis Bullosa Dystrophica/genetics , Epidermolysis Bullosa Dystrophica/pathology , Uniparental Disomy , Adult , Base Sequence , Collagen Type VII/genetics , Exons/genetics , Female , Frameshift Mutation , Genes, Recessive , Homozygote , Humans , Infant, Newborn , Male , Microsatellite Repeats , Molecular Sequence Data , Phenotype
18.
Am J Med Genet A ; 140(8): 887-91, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16532463

ABSTRACT

We report on the clinical and molecular abnormalities in a 7-month-old girl and her mother with an ectodermal dysplasia disorder that most closely resembles Rapp-Hodgkin syndrome (RHS). At birth, the child had bilateral cleft palate, a narrow pinched nose, small chin, and hypoplastic nipples, and suffered from respiratory distress, feeding difficulties, and poor weight gain, although developmental progress was normal. Her mother had a cleft palate, sparse hair, high forehead, dental anomalies, a narrow nose, dysplastic nails, and reduced sweating. Sequencing of the p63 gene in genomic DNA from both individuals revealed a heterozygous frameshift mutation, 1721delC, in exon 14. This mutation has not been described previously and is the seventh report of a pathogenic p63 gene mutation in RHS. The frameshift results in changes to the tail of p63 with the addition of 90 missense amino acids downstream and a delayed termination codon that extends the protein by 21 amino acids. This mutation is predicted to disrupt the normal repressive function of the transactivation inhibitory domain leading to gain-of-function for at least two isoforms of the p63 transcription factor. The expanding p63 mutation database demonstrates that there is considerable overlap between the molecular pathology of RHS and Hay-Wells syndrome, with identical mutations in some cases, and that these two disorders may in fact be synonymous.


Subject(s)
Ectodermal Dysplasia/genetics , Adult , Biomarkers , Chin/abnormalities , Cleft Palate/genetics , Female , Frameshift Mutation , Humans , Infant , Membrane Proteins/genetics , Nails, Malformed/genetics , Nose/abnormalities , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...