Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters

Publication year range
1.
J Pak Med Assoc ; 74(9): 1687-1689, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39279078

ABSTRACT

Erythrokeratodermia variabilis (EKV) is a rare inherited genodermatosis characterised by migratory and erythematous patches changing over the course of hours to days and fixed keratotic plaques. The disease begins mostly at birth or within the first year of life; it very rarely starts after childhood. We present here a sporadic case with adult onset EKV that was aggravated by pregnancy with spontaneous resolution, and later on, after crash diet more persistent patches and plaques appeared with no spontaneous resolution. The patient showed excellent response to systemic retinoids.


Subject(s)
Erythrokeratodermia Variabilis , Humans , Female , Pregnancy , Adult , Erythrokeratodermia Variabilis/genetics , Pregnancy Complications
2.
Int J Mol Sci ; 24(18)2023 Sep 11.
Article in English | MEDLINE | ID: mdl-37762265

ABSTRACT

Ichthyoses are genetically determined cornification disorders of the epidermis characterized by the presence of different degrees of scaling, hyperkeratosis, and erythroderma often associated with palmoplantar keratoderma. Different classifications of these diseases have been proposed, often based upon the involved genes and/or the clinical presentation. The clinical features of these diseases present some overlap of phenotypes among distinct genetic entities, depending mainly on the penetrance of mutations. In this study, using a clinical, genetic, and molecular approach, we analyzed a family with two affected members who had clinical and histological features resembling erythrokeratodermia variabilis (EKV) or a type of erythrodermic hyperkeratosis with palmoplantar keratoderma. Despite of the clinical presentation, we demonstrated that the affected patients were genetically double heterozygous for two different mutations in the ABCA12 gene, known to be responsible for harlequin ichthyosis. To explain the mild phenotype of our patients, we performed a molecular characterization of the skin. In the upper layers of the epidermis, the results showed a patchy presence of the glucosyl-ceramides (GlcCer), which is the lipid transported by ABCA12, fundamental in contributing to skin impermeability. Indeed, the two mutations detected do not completely abolish ABCA12 activity, indicating that the mild phenotype is due to a partial loss of function of the enzyme, thus giving rise to an intermediate phenotype resembling EKVP, due to a partial depletion of GlcCer deposition.


Subject(s)
Erythrokeratodermia Variabilis , Ichthyosis, Lamellar , Ichthyosis , Keratoderma, Palmoplantar , Humans , Erythrokeratodermia Variabilis/genetics , Ichthyosis, Lamellar/genetics , Ichthyosis/genetics , Mutation , Glucosylceramides , ATP-Binding Cassette Transporters/genetics
3.
Exp Dermatol ; 31(4): 594-599, 2022 04.
Article in English | MEDLINE | ID: mdl-34717022

ABSTRACT

Erythrokeratodermia variabilis et progressiva (EKVP) is a rare genodermatosis of clinical and genetic heterogeneity, characterized by the manifestations of localized or disseminated persistent hyperkeratotic plagues and stationary to migratory transient erythematous patches. The majority of EKVP cases display an autosomal dominant mode of inheritance with incomplete penetrance, although recessive transmission has also been described. Mutations associated with EKVP have been primarily detected in connexin (Cx) genes. We herein reported a Chinese sporadic case of late-onset EKVP with a novel heterozygous missense mutation c.109G>A (p.V37M) in GJB4 (Cx30.3) gene, which resulted in a significant reduction of GJB4 expression in the epidermis of the patient. In accordance, while wild-type GJB4 localized at the cell membrane of HeLa cells forming intercellular junctions and intracellular puncta, V37M mutant variant was diffusely expressed within HeLa cells at a considerably lower level. Our findings reveal an essential role of GJB4 in the pathogenesis of EKVP and provides insights into the therapeutic potential of the disease.


Subject(s)
Connexins , Erythrokeratodermia Variabilis , Connexins/genetics , Erythrokeratodermia Variabilis/genetics , Erythrokeratodermia Variabilis/pathology , HeLa Cells , Heterozygote , Humans , Mutation, Missense
4.
Int J Mol Sci ; 23(1)2022 Jan 01.
Article in English | MEDLINE | ID: mdl-35008913

ABSTRACT

Although inherited GJA1 (encoding Cx43) gene mutations most often lead to oculodentodigital dysplasia and related disorders, four variants have been linked to erythrokeratodermia variabilis et progressiva (EKVP), a skin disorder characterized by erythematous and hyperkeratotic lesions. While two autosomal-dominant EKVP-linked GJA1 mutations have been shown to lead to augmented hemichannels, the consequence(s) of keratinocytes harboring a de novo P283L variant alone or in combination with a de novo T290N variant remain unknown. Interestingly, these variants reside within or adjacent to a carboxy terminus polypeptide motif that has been shown to be important in regulating the internalization and degradation of Cx43. Cx43-rich rat epidermal keratinocytes (REKs) or Cx43-ablated REKs engineered to express fluorescent protein-tagged P283L and/or T290N variants formed prototypical gap junctions at cell-cell interfaces similar to wildtype Cx43. Dye coupling and dye uptake studies further revealed that each variant or a combination of both variants formed functional gap junction channels, with no evidence of augmented hemichannel function or induction of cell death. Tracking the fate of EKVP-associated variants in the presence of the protein secretion blocker brefeldin A, or an inhibitor of protein synthesis cycloheximide, revealed that P283L or the combination of P283L and T290N variants either significantly extended Cx43 residency on the cell surface of keratinocytes or delayed its degradation. However, caution is needed in concluding that this modest change in the Cx43 life cycle is sufficient to cause EKVP, or whether an additional underlying mechanism or another unidentified gene mutation is contributing to the pathogenesis found in patients. This question will be resolved if further patients are identified where whole exome sequencing reveals a Cx43 P283L variant alone or, in combination with a T290N variant, co-segregates with EKVP across several family generations.


Subject(s)
Connexin 43/chemistry , Connexin 43/genetics , Erythrokeratodermia Variabilis/genetics , Mutation/genetics , Animals , Coloring Agents , Endoplasmic Reticulum/metabolism , Gap Junctions/metabolism , HeLa Cells , Humans , Mutant Proteins/metabolism , Proteolysis , Rats
5.
Int J Mol Sci ; 22(5)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33807656

ABSTRACT

Glycine is an amino acid with unique properties because its side chain is composed of a single hydrogen atom. It confers conformational flexibility to proteins and conserved glycines are often indicative of protein domains involving tight turns or bends. All six beta-type connexins expressed in human epidermis (Cx26, Cx30, Cx30.3, Cx31, Cx31.1 and Cx32) contain a glycine at position 12 (G12). G12 is located about halfway through the cytoplasmic amino terminus and substitutions alter connexin function in a variety of ways, in some cases altering protein interactions and leading to cell death. There is also evidence that alteration of G12 changes the structure of the amino terminus in connexin- and amino acid- specific ways. This review integrates structural, functional and physiological information about the role of G12 in connexins, focusing on beta-connexins expressed in human epidermis. The importance of G12 substitutions in these beta-connexins is revealed in two hereditary skin disorders, keratitis ichthyosis and erythrokeratodermia variabilis, both of which result from missense mutations affecting G12.


Subject(s)
Connexins/metabolism , Epidermis/metabolism , Erythrokeratodermia Variabilis/metabolism , Gap Junctions/metabolism , Ichthyosis/metabolism , Mutation, Missense , Amino Acid Substitution , Connexins/genetics , Epidermis/pathology , Erythrokeratodermia Variabilis/genetics , Erythrokeratodermia Variabilis/pathology , Gap Junctions/genetics , Gap Junctions/pathology , Glycine/genetics , Glycine/metabolism , Humans , Ichthyosis/genetics , Ichthyosis/pathology
6.
Zhonghua Yi Xue Za Zhi ; 101(16): 1128-1131, 2021 Apr 27.
Article in Zh | MEDLINE | ID: mdl-33902244

ABSTRACT

Progressive symmetric erythrokeratodermia (PSEK) comprises a group of clinically and genetically heterogeneous diseases. Previous research have identified GJB3 and GJB4 as the leading genetic causes of this disorder. With the rapid development of genetics, GJA1, KDSR, KRT83 and TRPM4 have been identified as the new causative genes for PSEK, leading to a further understanding of its clinical features and genetic mechanisms. It's worth noting that Nagashima-type palmoplantar keratosis was often misdiagnosed as PSEK by our domestic dermatologists. Due to the identification of SERPINB7 as the causative gene of Nagashima-type palmoplantar keratosis recently, differentiation between the two disorders could be easily distinguished.


Subject(s)
Erythrokeratodermia Variabilis , Keratoderma, Palmoplantar , Erythrokeratodermia Variabilis/diagnosis , Erythrokeratodermia Variabilis/genetics , Humans , Keratoderma, Palmoplantar/diagnosis , Keratoderma, Palmoplantar/genetics
7.
J Hum Genet ; 65(4): 397-410, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31911611

ABSTRACT

Molecular diagnosis of rare inherited palmoplantar keratoderma (PPK) is still challenging. We investigated at the clinical and genetic level a consanguineous Tunisian family presenting an autosomal dominant atypical form of transgrediens and progrediens PPK to better characterize this ultrarare disease and to identify its molecular etiology. Whole-exome sequencing (WES), filtering strategies, and bioinformatics analysis have been achieved. Clinical investigation and follow up over 13 years of this Tunisian family with three siblings formerly diagnosed as an autosomal recessive form of Mal de Melela-like conducted us to reconsider its initial phenotype. Indeed, the three patients presented clinical features that overlap both Mal de Meleda and progressive symmetric erythrokeratoderma (PSEK). The mode of inheritance was also reconsidered, since the mother, initially classified as unaffected, exhibited a similar expression of the disease. WES analysis showed the absence of potentially functional rare variants in known PPKs or PSEK-related genes. Results revealed a novel heterozygous nonsynonymous variant in cadherin-12 gene (CDH12, NM_004061, c.1655C > A, p.Thr552Asn) in all affected family members. This variant is absent in dbSNP and in 50 in-house control exomes. In addition, in silico analysis of the mutated 3D domain structure predicted that this variant would result in cadherin-12 protein destabilization and thermal instability. Functional annotation and biological network construction data provide further supporting evidence for the potential role of CDH12 in the maintenance of skin integrity. Taken together, these results suggest that CDH12 gene is a potential candidate gene for an atypical presentation of an autosomal dominant form of transgrediens and progrediens PPK.


Subject(s)
Cadherins , Chromosome Disorders , Erythrokeratodermia Variabilis , Genes, Dominant , Mutation, Missense , Adult , Aged , Cadherin Related Proteins , Cadherins/chemistry , Cadherins/genetics , Chromosome Disorders/genetics , Chromosome Disorders/pathology , Computer Simulation , Erythrokeratodermia Variabilis/genetics , Erythrokeratodermia Variabilis/pathology , Female , Humans , Male , Protein Domains , Skin/pathology , Exome Sequencing
8.
Am J Med Genet A ; 182(8): 1933-1938, 2020 08.
Article in English | MEDLINE | ID: mdl-32449269

ABSTRACT

Pathogenic germline variants in the gap junction protein alpha 1 (GJA1) gene have been identified in several congenital disorders affecting cutaneous, skeletal, and cardiac tissues. Here, we describe a 12-year-old patient with a GJA1 c.113G>A, p.(Gly38Glu) variant, who presented with fulminant myocarditis following recurrent generalized erythrokeratoderma. His mother and younger sister had the same clinical manifestations with the same GJA1 variant, but did not have cardiac dysfunction. GJA1 variants have been reported in patients with congenital cardiac malformations, while acute myocarditis in GJA1-related disorders has not been reported so far.


Subject(s)
Connexin 43/genetics , Erythrokeratodermia Variabilis/genetics , Heart Defects, Congenital/genetics , Myocarditis/genetics , Adult , Child , Erythrokeratodermia Variabilis/complications , Erythrokeratodermia Variabilis/pathology , Female , Germ-Line Mutation/genetics , Heart Defects, Congenital/complications , Heart Defects, Congenital/pathology , Heterozygote , Humans , Male , Myocarditis/complications , Myocarditis/pathology , Siblings
9.
J Hepatol ; 71(2): 344-356, 2019 08.
Article in English | MEDLINE | ID: mdl-30965071

ABSTRACT

BACKGROUND & AIMS: Hepatocyte polarity is essential for the development of bile canaliculi and for safely transporting bile and waste products from the liver. Functional studies of autologous mutated proteins in the context of the polarized hepatocyte have been challenging because of the lack of appropriate cell models. The aims of this study were to obtain a patient-specific hepatocyte model that recapitulated hepatocyte polarity and to employ this model to study endogenous mutant proteins in liver diseases that involve hepatocyte polarity. METHODS: Urine cell-derived pluripotent stem cells, taken from a patient with a homozygous mutation in ATP7B and a patient with a heterozygous mutation, were differentiated towards hepatocyte-like cells (hiHeps). HiHeps were also derived from a patient with MEDNIK syndrome. RESULTS: Polarized hiHeps that formed in vivo-like bile canaliculi could be generated from embryonic and patient urine cell-derived pluripotent stem cells. HiHeps recapitulated polarized protein trafficking processes, exemplified by the Cu2+-induced redistribution of the copper transporter protein ATP7B to the bile canalicular domain. We demonstrated that, in contrast to the current dogma, the most frequent yet enigmatic Wilson disease-causing ATP7B-H1069Q mutation per se did not preclude trafficking of ATP7B to the trans-Golgi Network. Instead, it prevented its Cu2+-induced polarized redistribution to the bile canalicular domain, which could not be reversed by pharmacological folding chaperones. Finally, we demonstrate that hiHeps from a patient with MEDNIK syndrome, suffering from liver copper overload of unclear etiology, showed no defect in the Cu2+-induced redistribution of ATP7B to the bile canaliculi. CONCLUSIONS: Functional cell polarity can be achieved in patient pluripotent stem cell-derived hiHeps, enabling, for the first time, the study of the endogenous mutant proteins, patient-specific pathogenesis and drug responses for diseases where hepatocyte polarity is a key factor. LAY SUMMARY: This study demonstrates that cells that are isolated from urine can be reprogrammed in a dish towards hepatocytes that display architectural characteristics similar to those seen in the intact liver. The application of this methodology to cells from patients diagnosed with inherited copper metabolism-related liver diseases (that is, Wilson disease and MEDNIK syndrome) revealed unexpected and novel insights into patient mutation-specific disease mechanisms and drug responses.


Subject(s)
Bile Canaliculi/pathology , Cell Polarity/genetics , Erythrokeratodermia Variabilis/genetics , Hepatocytes/metabolism , Hepatolenticular Degeneration/genetics , Pluripotent Stem Cells/metabolism , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex sigma Subunits/genetics , Bile Canaliculi/metabolism , Cells, Cultured , Copper/metabolism , Copper-Transporting ATPases/genetics , Erythrokeratodermia Variabilis/pathology , Hepatolenticular Degeneration/pathology , Humans , Mutant Proteins/metabolism , Mutation , Protein Transport
10.
Exp Dermatol ; 28(10): 1106-1113, 2019 10.
Article in English | MEDLINE | ID: mdl-29570224

ABSTRACT

Erythrokeratodermia variabilis et progressiva (EKV-P) is caused by mutations in either the GJB3 (Cx31) or GJB4 genes (Cx30.3). We identified a rare GJB3 missense mutation, c.134G>A (p.G45E), in two unrelated patients and investigated its cellular characteristics. Expression of Cx31G45E-GFP caused previously undescribed changes within HeLa cells and HaCaT cells, a model human keratinocyte cell line. Cx31WT-GFP localised to the plasma membrane, but expression of Cx31G45E-GFP caused vacuolar expansion of the endoplasmic reticulum (ER), the mutant protein accumulated within the ER membrane and disassembly of the microtubular network occurred. No ER stress responses were evoked. Cx31WT-myc-myc-6xHis and Cx31G45E-GFP co-immunoprecipitated, indicative of heteromeric interaction, but co-expression with Cx31WT-mCherry, Cx26 or Cx30.3 did not mitigate the phenotype. Cx31 and Cx31G45E both co-immunoprecipitated with Cx43, indicating the ability to form heteromeric connexons. WT-Cx31 and Cx43 assembled into large gap junction plaques at points of cell-to-cell contact; Cx31G45E restricted the ability of Cx43 to reach the plasma membrane in both HaCaT cells and HeLa cells stably expressing Cx43 where the proteins strongly co-localised with the vacolourised ER. Cell viability assays identified an increase in cell death in cells expressing Cx31G45E-GFP, which FACS analysis determined was necrotic. Blocking connexin channel function with 18α-glycyrrhetinic acid did not completely rescue necrosis or prevent propidium iodide uptake, suggesting that expression of Cx31G45E-GFP damages the cellular membrane independent of its channel function. Our data suggest that entrapment of Cx43 and necrotic cell death in the epidermis could underlie the EKV skin phenotype.


Subject(s)
Connexins/genetics , Erythrokeratodermia Variabilis/genetics , Mutation, Missense , Cell Death , Cell Membrane/drug effects , Cells, Cultured , Connexin 43/biosynthesis , Connexin 43/genetics , Endoplasmic Reticulum/ultrastructure , Epidermis/pathology , Erythrokeratodermia Variabilis/pathology , Genes, Dominant , Genetic Association Studies , Glycyrrhetinic Acid/analogs & derivatives , Glycyrrhetinic Acid/pharmacology , HeLa Cells , Humans , Keratinocytes , Necrosis , Protein Transport
12.
J Med Genet ; 54(3): 186-189, 2017 03.
Article in English | MEDLINE | ID: mdl-27965375

ABSTRACT

BACKGROUND: Progressive symmetric erythrokeratoderma (PSEK) is a rare skin disorder characterised by symmetrically distributed demarcated hyperkeratotic plaques, often with associated palmoplantar hyperkeratosis, with new plaques appearing over time. Most cases are inherited in an autosomal dominant manner, although a few cases exhibit apparent autosomal recessive inheritance. OBJECTIVE: To identify the gene underlying autosomal recessive PSEK in a large Pakistani kindred. METHODS: We first carried out autozygosity mapping using microsatellite markers in candidate regions of the genome. We then carried out exome sequencing of five family members, autozygosity mapping and mutation analysis using the exome data and verification by Sanger sequencing. RESULTS: Autozygosity mapping and exome sequencing identified a homozygous frameshift deletion (c.811delA; p.Ser271fs) in KRT83, which co-segregated with the PSEK phenotype in the family and which is expected to abolish keratin 83, a type II keratin of hair and skin. CONCLUSIONS: At least some cases of PSEK result from loss-of-function mutations in KRT83. Heterozygous missense substitutions in KRT83 have been implicated in autosomal dominant monilethrix, a rare hair disorder. Our findings indicate that at least some cases of autosomal recessive PSEK and autosomal dominant monilethrix are allelic, respectively resulting from loss-of-function and missense mutations in the KRT83 gene. Together, these findings indicate that different types of mutations in KRT83 can result in quite different skin and hair phenotypes.


Subject(s)
Erythrokeratodermia Variabilis/genetics , Keratins, Hair-Specific/genetics , Keratins, Type II/genetics , Monilethrix/genetics , Alleles , Erythrokeratodermia Variabilis/pathology , Exome/genetics , Female , Hair/metabolism , Hair/pathology , Heterozygote , Homozygote , Humans , Male , Monilethrix/pathology , Mutation, Missense , Pakistan , Pedigree , Phenotype , Sequence Deletion , Skin/metabolism , Skin/pathology
13.
Hum Mol Genet ; 24(21): 6054-65, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26251042

ABSTRACT

Mutations in connexin-31 (Cx31) are associated with multiple human diseases, including familial erythrokeratodermia variabilis (EKV). The pathogenic mechanism of EKV-associated Cx31 mutants remains largely elusive. Here, we show that EKV-pathogenic Cx31 mutants are un/misfolded and temperature sensitive. In Drosophila, expression of pathogenic Cx31, but not wild-type Cx31, causes depigmentation and degeneration of ommatidia that are rescued by expression of either dBip or dHsp70. Ectopic expression of Cx31 in mouse skin results in skin abnormalities resembling human EKV. The affected tissues show remarkable disrupted gap junction formation and significant upregulation of chaperones Bip and Hsp70 as well as AP-1 proteins c-Fos and JunB, in addition to molecular signatures of skin diseases. Consistently, c-Fos, JunB, Bip and Hsp70 are strikingly higher in keratinocytes of EKV patients than their matched control individuals. Furthermore, a druggable AP-1 inhibitory small molecule suppresses skin phenotype and pathological abnormalities of transgenic Cx31 mice. The study suggests that Cx31 mutant proteins are un/misfolded to cause EKV likely via an AP-1-mediated mechanism and identifies a small molecule with therapeutic potential of the disease.


Subject(s)
Connexins/metabolism , Erythrokeratodermia Variabilis/metabolism , Protein Folding , Animals , Animals, Genetically Modified , Benzophenones/pharmacology , Compound Eye, Arthropod/pathology , Connexins/antagonists & inhibitors , Connexins/genetics , Drosophila , Drosophila Proteins/genetics , Erythrokeratodermia Variabilis/drug therapy , Erythrokeratodermia Variabilis/genetics , Erythrokeratodermia Variabilis/pathology , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Isoxazoles/pharmacology , Mice , Mutation , Pigmentation/genetics , Protein Unfolding , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Recombinant Fusion Proteins , Skin/pathology , Stress, Physiological , Temperature , Transcription Factor TFIID/genetics , Transcription Factors/metabolism , Up-Regulation
16.
Genes (Basel) ; 15(3)2024 02 24.
Article in English | MEDLINE | ID: mdl-38540347

ABSTRACT

Erythrokeratodermia variabilis (EKV) is a rare genodermatosis characterized by well-demarcated erythematous patches and hyperkeratotic plaques. EKV is most often transmitted in an autosomal dominant manner. Until recently, only mutations in connexins such as GJB3 (connexin 31), GJB4 (connexin 30.3), and occasionally GJA1 (connexin 43) were known to cause EKV. In recent years, mutations in other genes have been described as rare causes of EKV, including the genes KDSR, KRT83, and TRPM4. Features of the EKV phenotype can also appear with other genodermatoses: for example, in Netherton syndrome, which hampers correct diagnosis. However, in autosomal recessive congenital ichthyosis (ARCI), an EKV phenotype has rarely been described. Here, we report on seven patients who clinically show a clear EKV phenotype, but in whom molecular genetic analysis revealed biallelic mutations in ABCA12, which is why the patients are classified in the ARCI group. Our study indicates that ARCI should be considered as a differential diagnosis in EKV.


Subject(s)
Erythrokeratodermia Variabilis , Humans , Erythrokeratodermia Variabilis/genetics , Mutation , Phenotype , Pedigree , ATP-Binding Cassette Transporters/genetics
17.
Biochim Biophys Acta ; 1818(8): 1952-61, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21933662

ABSTRACT

The expression of multiple connexin (Cx) types in the epidermis, their differential expression during wound closure and the association of skin pathology with specific Cx gene mutations, are indicative of important functions for Cxs in the skin. In this review, we focus on the role of Cx proteins in the epidermis and during wound healing and discuss mutations in Cx genes which cause skin disease. This article is part of a Special Issue entitled: The Communicating junctions, composition, structure and characteristics.


Subject(s)
Connexins/metabolism , Epidermis/metabolism , Skin Diseases/metabolism , Animals , Connexin 26 , Connexin 30 , Connexin 43/metabolism , Craniofacial Abnormalities/genetics , Ectodermal Dysplasia/genetics , Erythrokeratodermia Variabilis/genetics , Eye Abnormalities/genetics , Foot Deformities, Congenital/genetics , Homeostasis , Humans , Keratinocytes/cytology , Models, Biological , Mutation , Syndactyly/genetics , Tooth Abnormalities/genetics , Wound Healing , Xenopus
19.
Eur J Dermatol ; 22(2): 182-6, 2012.
Article in English | MEDLINE | ID: mdl-22266302

ABSTRACT

Erythrokeratodermia variabilis (EKV) is characterized by migrating red patches resembling a geographical map, and by localized or generalized hyperkeratosis with scaling of the skin. The onset is usually at birth or during infancy, and the disease persists throughout life. EKV is mainly inherited as an autosomal dominant disease, although recessive transmission has occasionally been reported. Mutations associated with EKV have been identified in the connexin (Cx) genes GJB3 (Cx31) and GJB4 (Cx30.3), however, several cases of EKV have been tested negative for mutations in these two Cx genes. Here, we report our findings of the clinical, histological, and molecular examinations performed in two unrelated sporadic cases of EKV. The molecular screening involved bidirectional sequencing of the coding regions of the GJB3 and GJB4 genes and revealed the existence of a novel c.295G>A missense variant in the GJB4 gene found in homozygosity in one case. The substitution was found to result in a p.E99K change of the Cx30.3 protein, an alteration predicted to have a benign rather than a damaging effect on the protein function.


Subject(s)
Connexins/genetics , Erythrokeratodermia Variabilis/genetics , Erythrokeratodermia Variabilis/pathology , Adult , Child, Preschool , Female , Humans , Mutation, Missense
SELECTION OF CITATIONS
SEARCH DETAIL