Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 578
Filter
1.
Annu Rev Genet ; 51: 123-141, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29178821

ABSTRACT

Genetic mosaicism arises when a zygote harbors two or more distinct genotypes, typically due to de novo, somatic mutation during embryogenesis. The clinical manifestations largely depend on the differentiation status of the mutated cell; earlier mutations target pluripotent cells and generate more widespread disease affecting multiple organ systems. If gonadal tissue is spared-as in somatic genomic mosaicism-the mutation and its effects are limited to the proband, whereas mosaicism also affecting the gametes, such as germline or gonosomal mosaicism, is transmissible. Mosaicism is easily appreciated in cutaneous disorders, as phenotypically distinct mutant cells often give rise to lesions in patterns determined by the affected cell type. Genetic investigation of cutaneous mosaic disorders has identified pathways central to disease pathogenesis, revealing novel therapeutic targets. In this review, we discuss examples of cutaneous mosaicism, approaches to gene discovery in these disorders, and insights into molecular pathobiology that have potential for clinical translation.


Subject(s)
Gene Expression Regulation, Developmental , Mosaicism , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Skin Diseases, Genetic/genetics , Ectoderm/metabolism , Ectoderm/pathology , Embryo, Mammalian , Endoderm/metabolism , Endoderm/pathology , Humans , Keratin-1/genetics , Keratin-1/metabolism , Keratin-10/genetics , Keratin-10/metabolism , Laser Capture Microdissection , Mesoderm/metabolism , Mesoderm/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Skin Diseases, Genetic/metabolism , Skin Diseases, Genetic/pathology , Time Factors , Exome Sequencing
2.
Br J Dermatol ; 190(5): 617-627, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38149939

ABSTRACT

Patients living with inherited skin diseases have benefited from recent advances in DNA sequencing technologies that provide new or improved diagnostics. However, developing and delivering new treatments for the 'genodermatoses' remains challenging. The goal of creating topical preparations that can recover the inherent gene pathology remains largely aspirational. However, recent progress in two fields - the chemistry of topical delivery formulations (lipid nanoparticles) and the molecular biology of gene repair (CRISPR-Cas9, base and prime editing) - presents new opportunities to address this unmet need. In this review, we discuss how lipid nanoparticle delivery vehicles could be used to deliver gene-editing tools to formulate topical 'gene creams' suitable for the treatment of genodermatoses. We summarize the historical landscape of topical therapeutics and advances in gene editing that may herald an era of new therapies for patients with inherited skin disorders.


Subject(s)
Liposomes , Nanoparticles , Skin Diseases, Genetic , Humans , Gene Editing , CRISPR-Cas Systems , Gene Transfer Techniques , Skin Diseases, Genetic/genetics
3.
Am J Dermatopathol ; 46(10): 637-647, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39293663

ABSTRACT

ABSTRACT: We present the histopathology of 12 skin biopsies from 6 patients with vacuoles, enzyme E1, X-linked, autoinflammatory, somatic syndrome and review the literature. The age of these 6 men ranges from 62 to 83 years (median of 70 years). UBA1 mutation was documented in all 6 patients. Multiple organ systems were involved with constitutional symptoms noted in 4 of 6 patients (67%), cutaneous involvement in 6 of 6 patients (100%), hematologic abnormalities in 6 of 6 patients (100%), pulmonary involvement in 4 of 6 patients (67%), musculoskeletal abnormalities in 3 of 6 patients (50%), vascular thrombosis in 2 of 6 patients (33%), ocular involvement in 2 of 6 patients (33%), and gastrointestinal involvement in 5 of 6 patients (83%). Of the 6 presented patients, neutrophilic dermatosis was seen in 3 biopsies, histiocytoid neutrophilic dermatosis in 1 biopsy, neutrophilic dermatosis with vasculitis in 1 biopsy, neutrophilic and granulomatous dermatitis in 2 biopsies, septal panniculitis consistent with erythema nodosum in 2 biopsies, and nonspecific patterns in 3 biopsies. In summary, neutrophilic dermatosis, small-vessel vasculitis, and panniculitis are frequent histopathologic patterns noted in decreasing frequency in skin biopsies of the patients with vacuoles, enzyme E1, X-linked, autoinflammatory, somatic syndrome. However, the histopathologic findings can be diverse, nonspecific in some instances, and varied among different biopsies obtained from the same patient.


Subject(s)
Myelodysplastic Syndromes , Skin Diseases, Genetic , Skin , Aged , Aged, 80 and over , Humans , Male , Middle Aged , Biopsy , Mutation , Skin/pathology , Syndrome , Ubiquitin-Activating Enzymes/genetics , Skin Diseases, Genetic/diagnosis , Skin Diseases, Genetic/genetics , Skin Diseases, Genetic/pathology , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology
4.
Int J Mol Sci ; 25(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38892036

ABSTRACT

The extracellular matrix is a complex network of proteins and other molecules that are essential for the support, integrity, and structure of cells and tissues within the human body. The genes ZNF469 and PRDM5 each produce extracellular-matrix-related proteins that, when mutated, have been shown to result in the development of brittle cornea syndrome. This dysfunction results from aberrant protein function resulting in extracellular matrix disruption. Our group recently identified and published the first known associations between variants in these genes and aortic/arterial aneurysms and dissection diseases. This paper delineates the proposed effects of mutated ZNF469 and PRDM5 on various essential extracellular matrix components, including various collagens, TGF-B, clusterin, thrombospondin, and HAPLN-1, and reviews our recent reports associating single-nucleotide variants to these genes' development of aneurysmal and dissection diseases.


Subject(s)
Extracellular Matrix , Transcription Factors , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/genetics , Joint Instability/genetics , Joint Instability/congenital , Histone-Lysine N-Methyltransferase/genetics , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/pathology , Ehlers-Danlos Syndrome/genetics , Ehlers-Danlos Syndrome/pathology , Aortic Aneurysm/genetics , Mutation , DNA-Binding Proteins/genetics , Skin Diseases, Genetic/genetics , Skin Diseases, Genetic/pathology , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Eye Abnormalities , Skin Abnormalities
5.
Exp Dermatol ; 32(9): 1334-1343, 2023 09.
Article in English | MEDLINE | ID: mdl-37353900

ABSTRACT

Dyschromatosis universalis hereditaria (DUH) is characterized by diffuse symmetrically distributed hypopigmented macules mixed with hyperpigmentation. DUH is divided into three types by Online Mendelian inheritance in man (OMIM) that is, DUH1 (OMIM 127500), DUH2 (OMIM 612715) and DUH3 (OMIM 615402) according to the different linkage regions. Although each condition possesses corresponding phenotypic characteristics and the prognosis for each is somewhat different, these disorders are highly overlapped and difficult to differentiate in the clinical setting. Our latest study reveals a novel DUH subtype that presents a mild phenotype of pigmentation anomalies and is named PER3rs772027021 SNP related DUH or DUH4 by us, which make the DUH subtype can be further retyped. Heterozygous distribution or mosaic-like distribution of melanin is a newly discovered pathological features that is uniquely demonstrated in the affected layers of DUH1 and DUH4 patients. In this review, DUH is further divided into four subtypes according the causative genes and their mutational sites, and the mutation regions described in the previous reports. To make an accurate diagnosis, we suggest that Sanger sequencing or the target region sequencing (TRS) to the candidate causative genes related melanogenesis may be the most effective and convenient method of clinical diagnosis or/and prenatal diagnosis for DUH and DUH-like patients. More importantly, heterozygous distribution or mosaic-like distribution of melanin can be utilized for differential diagnosis of DUH. We also investigate the underlying molecular mechanism to form mosaic-like melanin in the affected layers of hyper- and/or hypo-pigmented macules from DUH1 and DUH4 patients. This review provides a molecular and pathological delineation of four types of DUH and aims to establish a concise diagnostic strategy to allow clinical dermatologists to make an accurate diagnosis.


Subject(s)
Hyperpigmentation , Skin Diseases, Genetic , Humans , Pathology, Molecular , Melanins/genetics , Skin Diseases, Genetic/diagnosis , Skin Diseases, Genetic/genetics , Skin Diseases, Genetic/pathology , Hyperpigmentation/diagnosis , Hyperpigmentation/genetics , Pedigree
6.
Am J Med Genet A ; 191(3): 899-901, 2023 03.
Article in English | MEDLINE | ID: mdl-36448706

ABSTRACT

Sarcoidosis is a chronic granulomatous disorder affecting the lungs, skin, and many other organs. Twin studies suggest that genetic factors account, to a large degree, for the etiology of the disorder. Hence, theoretically, we could postulate that the phenomenon of superimposed mosaicism in the form of a pronounced segmental involvement, overlaying the disseminated non-segmental lesions, should also occur in sarcoidosis. Indeed, one case suggesting superimposed mosaicism in cutaneous sarcoidosis was found in the literature and is reassessed here.


Subject(s)
Sarcoidosis , Skin Diseases, Genetic , Skin Diseases , Humans , Mosaicism , Skin Diseases, Genetic/diagnosis , Skin Diseases, Genetic/genetics , Skin Diseases, Genetic/pathology , Skin/pathology , Sarcoidosis/diagnosis , Sarcoidosis/genetics , Skin Diseases/diagnosis , Skin Diseases/genetics , Skin Diseases/pathology
7.
J Eur Acad Dermatol Venereol ; 37(3): 488-500, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36502512

ABSTRACT

Genodermatoses are a complex and heterogeneous group of genetic skin disorders characterized by variable expression and clinical and genetic heterogeneity, rendering their diagnosis challenging. DNA-based techniques, like whole-exome sequencing, can establish a diagnosis in 50% of cases. RNA-sequencing is emerging as an attractive tool that can obtain information regarding gene expression while integrating functional genomic data with regard to the interpretation of variants. This increases the diagnostic rate by an additional 10-15%. In the present review, we detail the clinical steps involved in the diagnosis of genodermatoses, as well as the current DNA-based technologies available to clinicians. Herein, the intention is to facilitate a better understanding of the possibilities and limitations of these diagnostic technologies. In addition, this review could guide dermatologists through new emerging techniques, such as RNA-sequencing and its applications to familiarizing them with future techniques. Currently, this multi-omics approach is likely the best strategy designed to promote the diagnosis of patients with genodermatoses and discover new skin disease genes that could result in novel targeted therapies.


Subject(s)
Skin Diseases, Genetic , Humans , Skin Diseases, Genetic/diagnosis , Skin Diseases, Genetic/genetics , Skin Diseases, Genetic/therapy , Exome Sequencing , RNA , DNA
8.
J Allergy Clin Immunol ; 149(1): 432-439.e4, 2022 01.
Article in English | MEDLINE | ID: mdl-34048852

ABSTRACT

BACKGROUND: A novel autoinflammatory syndrome was recently described in male patients who harbored somatic mutations in the X-chromosomal UBA1 gene. These patients were characterized by adult-onset, treatment-refractory inflammation with fever, cytopenia, dysplastic bone marrow, vacuoles in myeloid and erythroid progenitor cells, cutaneous and pulmonary inflammation, chondritis, and vasculitis, which is abbreviated as VEXAS. OBJECTIVE: This study aimed to (retrospectively) diagnose VEXAS in patients who had previously been registered as having unclassified autoinflammation. We furthermore aimed to describe clinical experiences with this multifaceted, complex disease. METHODS: A systematic reanalysis of whole-exome sequencing data from a cohort of undiagnosed patients with autoinflammation from academic hospitals in The Netherlands was performed. When no sequencing data were available, targeted Sanger sequencing was applied in cases with high clinical suspicion of VEXAS. RESULTS: A total of 12 male patients who carried mutations in UBA1 were identified. These patients presented with adult-onset (mean age 67 years, range 47-79 years) autoinflammation with systemic symptoms, elevated inflammatory parameters, and multiorgan involvement, most typically involving the skin and bone marrow. Novel features of VEXAS included interstitial nephritis, cardiac involvement, stroke, and intestinal perforation related to treatment with tocilizumab. Although many types of treatment were initiated, most patients became treatment-refractory, with a high mortality rate of 50%. CONCLUSION: VEXAS should be considered in the differential diagnosis of males with adult-onset autoinflammation characterized by systemic symptoms and multiorgan involvement. Early diagnosis can prevent unnecessary diagnostic procedures and provide better prognostic information and more suitable treatment options, including stem cell transplantation.


Subject(s)
Hereditary Autoinflammatory Diseases/genetics , Myelodysplastic Syndromes/genetics , Skin Diseases, Genetic/genetics , Ubiquitin-Activating Enzymes/genetics , Adult , Age of Onset , Aged , Hereditary Autoinflammatory Diseases/diagnosis , Humans , Male , Middle Aged , Mutation , Myelodysplastic Syndromes/diagnosis , Netherlands , Retrospective Studies , Skin Diseases, Genetic/diagnosis
9.
JAMA ; 329(4): 318-324, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36692560

ABSTRACT

Importance: VEXAS (vacuoles, E1-ubiquitin-activating enzyme, X-linked, autoinflammatory, somatic) syndrome is a disease with rheumatologic and hematologic features caused by somatic variants in UBA1. Pathogenic variants are associated with a broad spectrum of clinical manifestations. Knowledge of prevalence, penetrance, and clinical characteristics of this disease have been limited by ascertainment biases based on known phenotypes. Objective: To determine the prevalence of pathogenic variants in UBA1 and associated clinical manifestations in an unselected population using a genomic ascertainment approach. Design, Setting, and Participants: This retrospective observational study evaluated UBA1 variants in exome data from 163 096 participants within the Geisinger MyCode Community Health Initiative. Clinical phenotypes were determined from Geisinger electronic health record data from January 1, 1996, to January 1, 2022. Exposures: Exome sequencing was performed. Main Outcomes and Measures: Outcome measures included prevalence of somatic UBA1 variation; presence of rheumatologic, hematologic, pulmonary, dermatologic, and other findings in individuals with somatic UBA1 variation on review of the electronic health record; review of laboratory data; bone marrow biopsy pathology analysis; and in vitro enzymatic assays. Results: In 163 096 participants (mean age, 52.8 years; 94% White; 61% women), 11 individuals harbored likely somatic variants at known pathogenic UBA1 positions, with 11 of 11 (100%) having clinical manifestations consistent with VEXAS syndrome (9 male, 2 female). A total of 5 of 11 individuals (45%) did not meet criteria for rheumatologic and/or hematologic diagnoses previously associated with VEXAS syndrome; however, all individuals had anemia (hemoglobin: mean, 7.8 g/dL; median, 7.5 g/dL), which was mostly macrocytic (10/11 [91%]) with concomitant thrombocytopenia (10/11 [91%]). Among the 11 patients identified, there was a pathogenic variant in 1 male participant prior to onset of VEXAS-related signs or symptoms and 2 female participants had disease with heterozygous variants. A previously unreported UBA1 variant (c.1861A>T; p.Ser621Cys) was found in a symptomatic patient, with in vitro data supporting a catalytic defect and pathogenicity. Together, disease-causing UBA1 variants were found in 1 in 13 591 unrelated individuals (95% CI, 1:7775-1:23 758), 1 in 4269 men older than 50 years (95% CI, 1:2319-1:7859), and 1 in 26 238 women older than 50 years (95% CI, 1:7196-1:147 669). Conclusions and Relevance: This study provides an estimate of the prevalence and a description of the clinical manifestations of UBA1 variants associated with VEXAS syndrome within a single regional health system in the US. Additional studies are needed in unselected and genetically diverse populations to better define general population prevalence and phenotypic spectrum.


Subject(s)
Myelodysplastic Syndromes , Skin Diseases, Genetic , Ubiquitin-Activating Enzymes , Female , Humans , Male , Biopsy , Electronic Health Records , Prevalence , Myelodysplastic Syndromes/complications , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/epidemiology , Myelodysplastic Syndromes/genetics , Ubiquitin-Activating Enzymes/genetics , Mutation , Retrospective Studies , Exome , Middle Aged , Skin Diseases, Genetic/complications , Skin Diseases, Genetic/diagnosis , Skin Diseases, Genetic/epidemiology , Skin Diseases, Genetic/genetics , United States/epidemiology
10.
Hum Mol Genet ; 29(9): 1476-1488, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32307537

ABSTRACT

Arterial tortuosity syndrome (ATS) is a recessively inherited connective tissue disorder, mainly characterized by tortuosity and aneurysm formation of the major arteries. ATS is caused by loss-of-function mutations in SLC2A10, encoding the facilitative glucose transporter GLUT10. Former studies implicated GLUT10 in the transport of dehydroascorbic acid, the oxidized form of ascorbic acid (AA). Mouse models carrying homozygous Slc2a10 missense mutations did not recapitulate the human phenotype. Since mice, in contrast to humans, are able to intracellularly synthesize AA, we generated a novel ATS mouse model, deficient for Slc2a10 as well as Gulo, which encodes for L-gulonolactone oxidase, an enzyme catalyzing the final step in AA biosynthesis in mouse. Gulo;Slc2a10 double knock-out mice showed mild phenotypic anomalies, which were absent in single knock-out controls. While Gulo;Slc2a10 double knock-out mice did not fully phenocopy human ATS, histological and immunocytochemical analysis revealed compromised extracellular matrix formation. Transforming growth factor beta signaling remained unaltered, while mitochondrial function was compromised in smooth muscle cells derived from Gulo;Slc2a10 double knock-out mice. Altogether, our data add evidence that ATS is an ascorbate compartmentalization disorder, but additional factors underlying the observed phenotype in humans remain to be determined.


Subject(s)
Arteries/abnormalities , Ascorbic Acid Deficiency/genetics , Glucose Transport Proteins, Facilitative/genetics , Joint Instability/genetics , L-Gulonolactone Oxidase/genetics , Skin Diseases, Genetic/genetics , Vascular Malformations/genetics , Animals , Arteries/metabolism , Arteries/pathology , Ascorbic Acid/biosynthesis , Ascorbic Acid/genetics , Ascorbic Acid Deficiency/metabolism , Ascorbic Acid Deficiency/pathology , Disease Models, Animal , Homozygote , Humans , Joint Instability/metabolism , Joint Instability/pathology , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Respiration/genetics , Signal Transduction/genetics , Skin Diseases, Genetic/metabolism , Skin Diseases, Genetic/pathology , Vascular Malformations/metabolism , Vascular Malformations/pathology
11.
BMC Endocr Disord ; 22(1): 70, 2022 Mar 16.
Article in English | MEDLINE | ID: mdl-35296306

ABSTRACT

BACKGROUND: The GNAS gene on chromosome 20q13.3, encodes the alpha-subunit of the stimulatory G protein, which is expressed in most tissues and regulated through reciprocal genomic imprinting. Disorders of GNAS inactivation produce several different clinical phenotypes including pseudohypoparathyroidism (PHP), pseudopseudohypoparathyroidism (PPHP), progressive osseous heteroplasia (POH), and osteoma cutis (OC). The clinical and biochemical characteristics overlap of PHP subtypes and other related disorders presents challenges for differential diagnosis. METHODS: We enrolled a total of 11 Chinese children with PHP in our study and analyzed their clinical characteristics, laboratory results, and genetic mutations. RESULTS: Among these 11 patients, nine of them (9/11) presented with resistance to parathyroid hormone (PTH); and nine (9/11) presented with an Albright's hereditary osteodystrophy (AHO) phenotype. GNAS abnormalities were detected in all 11 patients, including nine cases with GNAS gene variations and two cases with GNAS methylation defects. These GNAS variations included an intronic mutation (c.212 + 3_212 + 6delAAGT), three missense mutations (c.314C > T, c.308 T > C, c.1123G > T), two deletion mutations (c.565_568delGACT*2, c.74delA), and two splicing mutations (c.721 + 1G > A, c.432 + 1G > A). Three of these mutations, namely, c.314C > T, c.1123G > T, and c.721 + 1G > A, were found to be novel. This data was then used to assign a GNAS subtype to each of these patients with six cases diagnosed as PHP1a, two cases as PHP1b, one as PPHP, and two as POH. CONCLUSIONS: Evaluating patients with PTH resistance and AHO phenotype improved the genetic diagnosis of GNAS mutations significantly. In addition, our results suggest that when GNAS gene sequencing is negative, GNAS methylation study should be performed. Early genetic detection is required for the differential diagnosis of GNAS disorders and is critical to the clinician's ability to distinguish between heterotopic ossification in the POH and AHO phenotype.


Subject(s)
Bone Diseases, Metabolic , Chromogranins/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Ossification, Heterotopic , Pseudohypoparathyroidism , Skin Diseases, Genetic , Adolescent , Bone Diseases, Metabolic/diagnosis , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/pathology , Child , Child, Preschool , China , Female , Humans , Infant , Male , Ossification, Heterotopic/diagnosis , Ossification, Heterotopic/genetics , Ossification, Heterotopic/pathology , Pseudohypoparathyroidism/diagnosis , Pseudohypoparathyroidism/genetics , Pseudohypoparathyroidism/pathology , Pseudopseudohypoparathyroidism/diagnosis , Pseudopseudohypoparathyroidism/genetics , Pseudopseudohypoparathyroidism/pathology , Skin Diseases, Genetic/diagnosis , Skin Diseases, Genetic/genetics , Skin Diseases, Genetic/pathology
12.
Pediatr Dermatol ; 39(2): 268-272, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35178752

ABSTRACT

Generalized inflammatory peeling skin syndrome (PSS) is a rare autosomal recessive genodermatosis caused by loss-of-function disease-causing variants of the corneodesmosin gene (CDSN), resulting in excessive shedding of the superficial layers of the epidermis. We describe a case of generalized inflammatory PSS in an infant, presenting at day two of life with ichthyosiform erythroderma and superficial peeling of the skin. Hair microscopy showed trichorrhexis invaginata. Normal amounts of skin LEKT1, a product of SPINK5 on immunohistochemical staining excluded a diagnosis of Netherton syndrome. Genetic analysis revealed a homozygous novel complete CDSN deletion, estimated 4.6 kb in size, supporting the diagnosis of generalized inflammatory PSS.


Subject(s)
Dermatitis, Exfoliative , Eosinophilia , Hair Diseases , Netherton Syndrome , Skin Diseases, Genetic , Dermatitis, Exfoliative/diagnosis , Dermatitis, Exfoliative/genetics , Dermatitis, Exfoliative/pathology , Humans , Infant , Intercellular Signaling Peptides and Proteins , Netherton Syndrome/diagnosis , Netherton Syndrome/genetics , Skin Diseases, Genetic/diagnosis , Skin Diseases, Genetic/genetics
13.
Rev Med Suisse ; 18(776): 652-659, 2022 Apr 06.
Article in French | MEDLINE | ID: mdl-35385616

ABSTRACT

VEXAS syndrome was recently discovered in patients who developed late in adulthood an inflammatory syndrome with fever, cytopenias, dysplastic bone marrow, cutaneous and pulmonary neutrophilic inflammation, arthritis, chondritis, or vasculitis. It is the result of an inactivating somatic mutation affecting methionine codon 41 of the UBA1 gene which encodes an ubiquitin activating enzyme (E1). Systemic corticosteroids generally reduce symptoms, while other immunosuppressive drugs only have limited long-term effects. Azacitidine is a promising treatment, but further studies are warranted. Here, we describe 2 new cases including one associated with pyoderma gangrenosum and cryoglobulinemia.


Le syndrome VEXAS (Vacuoles, E1 Enzyme, X-Linked, Auto- Inflammatory, Somatic Syndrome) a été récemment découvert chez des patients développant tardivement à l'âge adulte un syndrome inflammatoire associé à de la fièvre, des cytopénies, une moelle osseuse dysplasique, une inflammation neutrophilique cutanée et pulmonaire, des arthrites, des chondrites ou des vasculites. Il est le résultat d'une mutation somatique inactivatrice affectant le codon méthionine 41 du gène UBA1 qui encode une enzyme E1 activant l'ubiquitine. Les corticostéroïdes systémiques permettent généralement de diminuer les symptômes alors que les autres immunosuppresseurs ont un effet limité à long terme. L'azacitidine est l'un des traitements ayant démontré une efficacité, cependant de nouvelles études sont souhaitables. Nous décrivons ici 2 cas dont l'un est associé à un pyoderma gangrenosum et une cryoglobulinémie.


Subject(s)
Myelodysplastic Syndromes , Skin Diseases, Genetic , Vasculitis , Adult , Humans , Inflammation , Mutation , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Pyoderma Gangrenosum , Skin Diseases, Genetic/diagnosis , Skin Diseases, Genetic/drug therapy , Skin Diseases, Genetic/genetics , Ubiquitin-Activating Enzymes/genetics
14.
Am J Hum Genet ; 102(2): 219-232, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29336782

ABSTRACT

Amyloidosis cutis dyschromica (ACD) is a distinct form of primary cutaneous amyloidosis characterized by generalized hyperpigmentation mottled with small hypopigmented macules on the trunks and limbs. Affected families and sporadic case subjects have been reported predominantly in East and Southeast Asian ethnicities; however, the genetic cause has not been elucidated. We report here that the compound heterozygosity or homozygosity of GPNMB truncating alleles is the cause of autosomal-recessive ACD. Six nonsense or frameshift mutations were identified in nine individuals diagnosed with ACD. Immunofluorescence analysis of skin biopsies showed that GPNMB is expressed in all epidermal cells, with the highest staining observed in melanocytes. GPNMB staining is significantly reduced in the lesional skin of affected individuals. Hyperpigmented lesions exhibited significantly increased amounts of DNA/keratin-positive amyloid deposits in the papillary dermis and infiltrating macrophages compared with hypo- or depigmented macules. Depigmentation of the lesions was attributable to loss of melanocytes. Intracytoplasmic fibrillary aggregates were observed in keratinocytes scattered in the lesional epidermis. Thus, our analysis indicates that loss of GPNMB, which has been implicated in melanosome formation, autophagy, phagocytosis, tissue repair, and negative regulation of inflammation, underlies autosomal-recessive ACD and provides insights into the etiology of amyloidosis and pigment dyschromia.


Subject(s)
Amyloidosis, Familial/genetics , Genes, Recessive , Genetic Predisposition to Disease , Membrane Glycoproteins/genetics , Skin Diseases, Genetic/genetics , Adolescent , Adult , Amino Acid Sequence , Base Sequence , Cell Count , Child , Child, Preschool , Dermis/pathology , Dermis/ultrastructure , Epidermis/metabolism , Epidermis/pathology , Female , HeLa Cells , Humans , Hyperpigmentation/genetics , Keratinocytes/pathology , Keratinocytes/ultrastructure , Macrophages/metabolism , Male , Melanocytes/metabolism , Membrane Glycoproteins/chemistry , Mutation/genetics , Pedigree
15.
Clin Genet ; 99(4): 572-576, 2021 04.
Article in English | MEDLINE | ID: mdl-33410500

ABSTRACT

We describe an 11-year-old girl with PLACK Syndrome (peeling skin, leukonychia, acral punctate keratosis, cheilitis, and knuckle pads), who was found to have a novel homozygous variant in CAST, the pathogenicity of which was confirmed using blood-derived RNA. There is no established treatment for PLACK syndrome. However, we demonstrate for the first time that this condition is associated with low levels of vitamin A and essential fatty acids, which prompted us to consider a potential treatment strategy. Indeed, we initiated this patient on intravenous lipid infusion (Vitalipid®; an emulsion of fat-soluble vitamins and lipofundin-MCT/LCT 20%) and the response was dramatic. Following the fourth monthly course of treatment, pruritis disappeared and the skin lesions showed remarkable objective improvement. PLACK syndrome is a very rare genodermatosis and only six families have been described to date with pathogenic CAST variants. This is the first report of an objective response to a therapeutic agent, which suggests that PLACK is a potentially treatable condition. The remarkable response we report and the relative safety of the intervention should prompt healthcare providers who care for PLACK syndrome patients to explore this as a potential treatment strategy in future studies.


Subject(s)
Dermatitis, Exfoliative/drug therapy , Hypopigmentation/drug therapy , Nail Diseases/congenital , Phospholipids/therapeutic use , Skin Diseases, Genetic/drug therapy , Soybean Oil/therapeutic use , Blister/etiology , Calcium-Binding Proteins/genetics , Cheilitis/drug therapy , Cheilitis/genetics , Child , Consanguinity , Dermatitis, Exfoliative/genetics , Emulsions/administration & dosage , Emulsions/therapeutic use , Female , Humans , Hypopigmentation/genetics , Infusions, Intravenous , Keratosis/drug therapy , Keratosis/genetics , Nail Diseases/drug therapy , Nail Diseases/genetics , Pedigree , Phospholipids/administration & dosage , Pruritus/drug therapy , Pruritus/genetics , Remission Induction , Skin Diseases, Genetic/genetics , Soybean Oil/administration & dosage , Syndrome , Treatment Outcome
16.
Exp Eye Res ; 209: 108671, 2021 08.
Article in English | MEDLINE | ID: mdl-34133966

ABSTRACT

Hereditary connective tissue diseases form a heterogeneous group of disorders that affect collagen and extracellular matrix components. The cornea and the skin are among the major forms of connective tissues, and syndromes affecting both organs are often due to mutations in single genes. Brittle cornea syndrome is one of the pathologies that illustrates this association well. Furthermore, sex hormones are known to play a role in the maintenance of the structure and the integrity of the connective tissue including the skin and cornea, and may be involved in pathogenesis of oculocutaneous diseases. Herein, a double consanguineous family of Moroccan origin with two affected siblings, with suspected brittle cornea syndrome, was recruited. Ophthalmic examinations and genetic testing were performed in all the nuclear family individuals. Clinical examinations showed that the two affected boys presented with thinning of the cornea, blue sclera, keratoconus, hyperelasticity of the skin, joint hypermobility, muscle weakness, hearing loss and dental abnormalities that are compatible with the diagnosis of BCS disease. They showed however additional clinical signs including micropenis, hypospadias and cryptorchidism, suggesting abnormalities in endocrine pathways. Using a duo exome sequencing analysis performed in the mother and the propositus, we identified the novel homozygous missense mutation c.461G > A (p.Arg154Gln) in the short-chain dehydrogenase/reductase family 42E member 1 (SDR42E1) gene. This novel mutation, which co-segregated with the disease in the family, was predicted to be pathogenic by bioinformatics tools. SDR42E1 stability analysis using DynaMut web-server showed that the p.Arg154Gln mutations has a destabilizing effect with a ΔΔG value of -1.039 kcal/mol. As this novel gene belongs to the large family of short-chain dehydrogenases/reductases (SDR) thought to be involved in steroid biosynthesis, endocrinological investigations subsequently revealed that the two patients also had low levels of cholesterol. Karyotyping revealed a normal 46,XY karyotype for the two boys, excluding other causes of disorders of sex development due to chromosomal rearrangements. In conclusion, our study reveals that mutation in the novel SDR42E1 gene alters the steroid hormone synthesis and associated with a new syndrome we named oculocutaneous genital syndrome. In addition, this study highlights the role of SDR42E1 in the regulation of cholesterol metabolism in the maintenance of connective tissue and sexual maturation in humans.


Subject(s)
Abnormalities, Multiple , Eye Abnormalities/genetics , Eye Diseases, Hereditary/genetics , Joint Instability/congenital , Mutation , Short Chain Dehydrogenase-Reductases/genetics , Skin Abnormalities/genetics , Skin Diseases, Genetic/genetics , Steroids/biosynthesis , Child , Child, Preschool , DNA/genetics , DNA Mutational Analysis , Eye Abnormalities/metabolism , Eye Diseases, Hereditary/metabolism , Humans , Joint Instability/genetics , Joint Instability/metabolism , Male , Pedigree , Short Chain Dehydrogenase-Reductases/metabolism , Skin Abnormalities/metabolism , Skin Diseases, Genetic/metabolism
17.
Hum Genomics ; 14(1): 45, 2020 12 07.
Article in English | MEDLINE | ID: mdl-33287903

ABSTRACT

BACKGROUND: Germline variants of ten keratin genes (K1, K2, K5, K6A, K6B, K9, K10, K14, K16, and K17) have been reported for causing different types of genodermatoses with an autosomal dominant mode of inheritance. Among all the variants of these ten keratin genes, most of them are missense variants. Unlike pathogenic and likely pathogenic variants, understanding the clinical importance of novel missense variants or variants of uncertain significance (VUS) is the biggest challenge for clinicians or medical geneticists. Functional characterization is the only way to understand the clinical association of novel missense variants or VUS but it is time consuming, costly, and depends on the availability of patient's samples. Existing databases report the pathogenic variants of the keratin genes, but never emphasize the systematic effects of these variants on keratin protein structure and genotype-phenotype correlation. RESULTS: To address this need, we developed a comprehensive database KVarPredDB, which contains information of all ten keratin genes associated with genodermatoses. We integrated and curated 400 reported pathogenic missense variants as well as 4629 missense VUS. KVarPredDB predicts the pathogenicity of novel missense variants as well as to understand the severity of disease phenotype, based on four criteria; firstly, the difference in physico-chemical properties between the wild type and substituted amino acids; secondly, the loss of inter/intra-chain interactions; thirdly, evolutionary conservation of the wild type amino acids and lastly, the effect of the substituted amino acids in the heptad repeat. Molecular docking simulations based on resolved crystal structures were adopted to predict stability changes and get the binding energy to compare the wild type protein with the mutated one. We use this basic information to determine the structural and functional impact of novel missense variants on the keratin coiled-coil heterodimer. KVarPredDB was built under the integrative web application development framework SSM (SpringBoot, Spring MVC, MyBatis) and implemented in Java, Bootstrap, React-mutation-mapper, MySQL, Tomcat. The website can be accessed through http://bioinfo.zju.edu.cn/KVarPredDB . The genomic variants and analysis results are freely available under the Creative Commons license. CONCLUSIONS: KVarPredDB provides an intuitive and user-friendly interface with computational analytical investigation for each missense variant of the keratin genes associated with genodermatoses.


Subject(s)
Computational Biology/methods , Databases, Genetic , Genetic Predisposition to Disease/genetics , Keratins/genetics , Mutation, Missense , Skin Diseases, Genetic/genetics , Humans , Phenotype , Polymorphism, Single Nucleotide , Protein Isoforms/genetics , Reproducibility of Results
18.
Br J Dermatol ; 184(6): 1123-1131, 2021 06.
Article in English | MEDLINE | ID: mdl-32926582

ABSTRACT

BACKGROUND: Peeling skin syndrome type 1 (PSS1) is a rare and severe autosomal recessive form of congenital ichthyosis. Patients are affected by pronounced erythroderma accompanied by pruritus and superficial generalized peeling of the skin. The disease is caused by nonsense mutations or complete deletion of the CDSN gene encoding for corneodesmosin (CDSN). PSS1 severely impairs quality of life and therapeutic approaches are totally unsatisfactory. OBJECTIVES: The objective of this study was to develop the first steps towards a specific protein replacement therapy for CDSN deficiency. Using this approach, we aimed to restore the lack of CDSN and improve cell-cell cohesion in the transition area of the stratum granulosum (SG) to the stratum corneum. METHODS: Human CDSN was recombinantly expressed in Escherichia coli. A liposome-based carrier system, prepared with a cationic lipopeptide to mediate the transport to the outer membrane of keratinocytes, was developed. This formulation was chosen for CDSN delivery into the skin. The liposomal carrier system was characterized with respect to size, stability and toxicity. Furthermore, the interaction with primary keratinocytes and human epidermal equivalents was investigated. RESULTS: The liposomes showed an accumulation at the membranes of keratinocytes. CDSN-deficient epidermal equivalents that were treated with liposomal encapsulated CDSN demonstrated presence of CDSN in the SG. Finally, the penetration assay and histological examinations revealed an improved epidermal integrity for CDSN-deficient epidermal equivalents, if they were treated with liposomal encapsulated CDSN. CONCLUSIONS: This study presents the first preclinical in vitro experiments for a future specific protein replacement therapy for patients affected by PSS1.


Subject(s)
Dermatitis, Exfoliative , Skin Diseases, Genetic , Glycoproteins , Humans , Intercellular Signaling Peptides and Proteins , Quality of Life , Skin Diseases, Genetic/drug therapy , Skin Diseases, Genetic/genetics
19.
J Cutan Pathol ; 48(1): 77-80, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32519343

ABSTRACT

Buschke-Ollendorff syndrome (BOS; OMIM 166700) is a rare autosomal dominant disorder characterized by the existence of connective tissue nevus and/or osteopoikilosis. The skin lesions usually present as firm, yellow, or flesh-colored papules and nodules, which may coalesce into plaques and increase in size and number over time. We present a case of a 26-year-old male with multiple subcutaneous nodules on the waist and thigh for more than 20 years. Being deeply seated, his skin lesions were not visible and could only be appreciated by palpation. Accordingly, pathology showed an increase in thick, crossed, or paralleled, elastic fibers arranged between the collagen bundles in the lower part of the reticular dermis and the subcutaneous fat with mucin deposition. Heterozygous point mutation in exon 8 of the LEMD3 gene was detected, which confirmed the diagnosis of BOS. The deeply situated nature of skin lesions noted in our case has not been reported in the literature of BOS. Our case thus expands the clinical and pathological features of the disease.


Subject(s)
DNA-Binding Proteins/genetics , Membrane Proteins/genetics , Osteopoikilosis/genetics , Osteopoikilosis/pathology , Skin Diseases, Genetic/genetics , Skin Diseases, Genetic/pathology , Subcutaneous Tissue/pathology , Adult , Germ-Line Mutation , Humans , Male , Mucins
20.
J Pediatr Hematol Oncol ; 43(4): e558-e560, 2021 05 01.
Article in English | MEDLINE | ID: mdl-32941296

ABSTRACT

Type 1 plasminogen deficiency is a rare genetic disorder. Type 1 plasminogen deficiency is characterized by fibrin-rich pseudomembrane formation on mucosal surfaces, particularly the conjunctiva. Tracheobronchial tree involvement is a less common reported manifestation of type 1 plasminogen deficiency. Pseudomembranes in the tracheobronchial tree may result in respiratory compromise and ultimately fail if not recognized and treated. Currently, there is no specific replacement therapy approved for the treatment of congenital plasminogen deficiency. In the present paper, we report that type 1 plasminogen deficiency with novel frameshift mutation and pulmonary involvement was treated initially with systemic fresh frozen plasma followed by pulmonary lavage with fresh frozen plasma and tissue plasminogen activator.


Subject(s)
Conjunctivitis/genetics , Frameshift Mutation , Plasminogen/deficiency , Plasminogen/genetics , Skin Diseases, Genetic/genetics , Blood Component Transfusion , Conjunctivitis/pathology , Conjunctivitis/therapy , Humans , Infant , Lung/pathology , Male , Skin Diseases, Genetic/pathology , Skin Diseases, Genetic/therapy , Tissue Plasminogen Activator/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL