Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters

Publication year range
1.
J Clin Immunol ; 43(6): 1040-1051, 2023 08.
Article in English | MEDLINE | ID: mdl-37204644

ABSTRACT

Hidradenitis suppurativa (HS), also known as Verneuil's disease and acne inversa, is a prevalent, debilitating, and understudied inflammatory skin disease. It is marked by repeated bouts of pathological inflammation causing pain, hyperplasia, aberrant healing, and fibrosis. HS is difficult to manage and has many unmet medical needs. There is clinical and pharmacological evidence for extensive etiological heterogeneity with HS, suggesting that this clinical diagnosis is capturing a spectrum of disease entities. Human genetic studies provide robust insight into disease pathogenesis. They also can be used to resolve etiological heterogeneity and to identify drug targets. However, HS has not been extensively investigated with well-powered genetic studies. Here, we review what is known about its genetic architecture. We identify overlap in molecular, cellular, and clinical features between HS and inborn errors of immunity (IEI). This evidence indicates that HS may be an underrecognized component of IEI and suggests that undiagnosed IEI are present in HS cohorts. Inborn errors of immunity represent a salient opportunity for rapidly resolving the immunological landscape of HS pathogenesis, for prioritizing drug repurposing studies, and for improving the clinical management of HS.


Subject(s)
Dermatitis , Hidradenitis Suppurativa , Humans , Hidradenitis Suppurativa/etiology , Hidradenitis Suppurativa/genetics , Dermatitis/complications , Cost of Illness
2.
Bioinformatics ; 2021 Jan 30.
Article in English | MEDLINE | ID: mdl-33515242

ABSTRACT

MOTIVATION: Predicting regulatory effects of genetic variants is a challenging but important problem in functional genomics. Given the relatively low sensitivity of functional assays, and the pervasiveness of class imbalance in functional genomic data, popular statistical prediction models can sharply underestimate the probability of a regulatory effect. We describe here the presence-only model (PO-EN), a type of semi-supervised model, to predict regulatory effects of genetic variants at sequence-level resolution in a context of interest by integrating a large number of epigenetic features and massively parallel reporter assays (MPRAs). RESULTS: Using experimental data from a variety of MPRAs we show that the presence-only model produces better calibrated predicted probabilities and has increased accuracy relative to state-of-the-art prediction models. Furthermore, we show that the predictions based on pre-trained PO-EN models are useful for prioritizing functional variants among candidate eQTLs and significant SNPs at GWAS loci. In particular, for the costimulatory locus, associated with multiple autoimmune diseases, we show evidence of a regulatory variant residing in an enhancer 24.4 kb downstream of CTLA4, with evidence from capture Hi-C of interaction with CTLA4. Furthermore, the risk allele of the regulatory variant is on the same risk increasing haplotype as a functional coding variant in exon 1 of CTLA4, suggesting that the regulatory variant acts jointly with the coding variant leading to increased risk to disease. AVAILABILITY: The presence-only model is implemented in the R package 'PO.EN', freely available on CRAN. A vignette describing a detailed demonstration of using the proposed PO-EN model can be found on github at https://github.com/Iuliana-Ionita-Laza/PO.EN/. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.

4.
J Am Soc Nephrol ; 32(8): 2031-2047, 2021 08.
Article in English | MEDLINE | ID: mdl-33941608

ABSTRACT

BACKGROUND: Genetic variants in complement genes have been associated with a wide range of human disease states, but well-powered genetic association studies of complement activation have not been performed in large multiethnic cohorts. METHODS: We performed medical records-based genome-wide and phenome-wide association studies for plasma C3 and C4 levels among participants of the Electronic Medical Records and Genomics (eMERGE) network. RESULTS: In a GWAS for C3 levels in 3949 individuals, we detected two genome-wide significant loci: chr.1q31.3 (CFH locus; rs3753396-A; ß=0.20; 95% CI, 0.14 to 0.25; P=1.52x10-11) and chr.19p13.3 (C3 locus; rs11569470-G; ß=0.19; 95% CI, 0.13 to 0.24; P=1.29x10-8). These two loci explained approximately 2% of variance in C3 levels. GWAS for C4 levels involved 3998 individuals and revealed a genome-wide significant locus at chr.6p21.32 (C4 locus; rs3135353-C; ß=0.40; 95% CI, 0.34 to 0.45; P=4.58x10-35). This locus explained approximately 13% of variance in C4 levels. The multiallelic copy number variant analysis defined two structural genomic C4 variants with large effect on blood C4 levels: C4-BS (ß=-0.36; 95% CI, -0.42 to -0.30; P=2.98x10-22) and C4-AL-BS (ß=0.25; 95% CI, 0.21 to 0.29; P=8.11x10-23). Overall, C4 levels were strongly correlated with copy numbers of C4A and C4B genes. In comprehensive phenome-wide association studies involving 102,138 eMERGE participants, we cataloged a full spectrum of autoimmune, cardiometabolic, and kidney diseases genetically related to systemic complement activation. CONCLUSIONS: We discovered genetic determinants of plasma C3 and C4 levels using eMERGE genomic data linked to electronic medical records. Genetic variants regulating C3 and C4 levels have large effects and multiple clinical correlations across the spectrum of complement-related diseases in humans.


Subject(s)
Complement C3/genetics , Complement C3/metabolism , Complement C4/genetics , Complement C4/metabolism , Genetic Variation , Medical Records , Adult , Aged , Alleles , Complement Activation/genetics , Databases, Genetic , Epidemiologic Studies , Female , Gene Dosage , Genetic Loci , Genome-Wide Association Study , Humans , Male , Medical Record Linkage , Middle Aged , Young Adult
5.
Exp Dermatol ; 29(3): 243-253, 2020 03.
Article in English | MEDLINE | ID: mdl-31169925

ABSTRACT

Alopecia areata (AA) is a highly prevalent autoimmune disease that attacks the hair follicle and leads to hair loss that can range from small patches to complete loss of scalp and body hair. Our previous linkage and genome-wide association studies (GWAS) generated strong evidence for aetiological contributions from inherited genetic variants at different population frequencies, including both rare mutations and common polymorphisms. Additionally, we conducted gene expression (GE) studies on scalp biopsies of 96 patients and controls to establish signatures of active disease. In this study, we performed an integrative analysis on these two datasets to test the hypothesis that rare CNVs in patients with AA could be leveraged to identify drivers of disease in our AA GE signatures. We analysed copy number variants (CNVs) in a case-control cohort of 673 patients with AA and 16 311 controls independent of the case-control cohort of 96 research participants used in our GE study. Using an integrative computational analysis, we identified 14 genes whose expression levels were altered by CNVs in a consistent direction of effect, corresponding to gene expression changes in lesional skin of patients. Four of these genes were affected by CNVs in three or more unrelated patients with AA, including ATG4B and SMARCA2, which are involved in autophagy and chromatin remodelling, respectively. Our findings identified new classes of genes with potential contributions to AA pathogenesis.


Subject(s)
Alopecia Areata/genetics , Alopecia Areata/immunology , Autophagy , DNA Copy Number Variations , Gene Dosage , Autophagy-Related Proteins/genetics , Cysteine Endopeptidases/genetics , Gene Expression Profiling , Genome-Wide Association Study , Genotype , Hair/pathology , Hair Follicle/physiology , Humans , Mutation , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Scalp/pathology , Transcription Factors/genetics
6.
J Investig Dermatol Symp Proc ; 20(1): S22-S27, 2020 11.
Article in English | MEDLINE | ID: mdl-33099379

ABSTRACT

Human genetic studies of diseases that are multifactorial and prevalent have generated a wealth of knowledge about the genetic architecture of chronic diseases. Generalizable attributes are shaping the development of models to explain how the human genome influences our health and can be leveraged to improve it. Importantly, both rare and common genetic variants contribute to disease risk and provide complementary information. Although initial genetic studies of alopecia areata have yielded insight with high clinical impact, there remains a number of important unanswered questions pertaining to disease biology and patient care that could be addressed by further genetic investigations.


Subject(s)
Alopecia Areata/genetics , Chronic Disease , Mutation , Genome-Wide Association Study , Humans , Models, Genetic , Polymorphism, Single Nucleotide , Translational Research, Biomedical
7.
World J Surg ; 44(1): 84-94, 2020 01.
Article in English | MEDLINE | ID: mdl-31605180

ABSTRACT

BACKGROUND: The extent to which obesity and genetics determine postoperative complications is incompletely understood. METHODS: We performed a retrospective study using two population cohorts with electronic health record (EHR) data. The first included 736,726 adults with body mass index (BMI) recorded between 1990 and 2017 at Vanderbilt University Medical Center. The second cohort consisted of 65,174 individuals from 12 institutions contributing EHR and genome-wide genotyping data to the Electronic Medical Records and Genomics (eMERGE) Network. Pairwise logistic regression analyses were used to measure the association of BMI categories with postoperative complications derived from International Classification of Disease-9 codes, including postoperative infection, incisional hernia, and intestinal obstruction. A genetic risk score was constructed from 97 obesity-risk single-nucleotide polymorphisms for a Mendelian randomization study to determine the association of genetic risk of obesity on postoperative complications. Logistic regression analyses were adjusted for sex, age, site, and race/principal components. RESULTS: Individuals with overweight or obese BMI (≥25 kg/m2) had increased risk of incisional hernia (odds ratio [OR] 1.7-5.5, p < 3.1 × 10-20), and people with obesity (BMI ≥ 30 kg/m2) had increased risk of postoperative infection (OR 1.2-2.3, p < 2.5 × 10-5). In the eMERGE cohort, genetically predicted BMI was associated with incisional hernia (OR 2.1 [95% CI 1.8-2.5], p = 1.4 × 10-6) and postoperative infection (OR 1.6 [95% CI 1.4-1.9], p = 3.1 × 10-6). Association findings were similar after limitation of the cohorts to those who underwent abdominal procedures. CONCLUSIONS: Clinical and Mendelian randomization studies suggest that obesity, as measured by BMI, is associated with the development of postoperative incisional hernia and infection.


Subject(s)
Mendelian Randomization Analysis/methods , Obesity/complications , Postoperative Complications/genetics , Adult , Body Mass Index , Female , Humans , Logistic Models , Male , Middle Aged , Polymorphism, Single Nucleotide , Postoperative Complications/etiology , Retrospective Studies , Risk Factors
8.
Am J Hum Genet ; 98(5): 934-955, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27153397

ABSTRACT

Haplotype-dependent allele-specific methylation (hap-ASM) can impact disease susceptibility, but maps of this phenomenon using stringent criteria in disease-relevant tissues remain sparse. Here we apply array-based and Methyl-Seq approaches to multiple human tissues and cell types, including brain, purified neurons and glia, T lymphocytes, and placenta, and identify 795 hap-ASM differentially methylated regions (DMRs) and 3,082 strong methylation quantitative trait loci (mQTLs), most not previously reported. More than half of these DMRs have cell type-restricted ASM, and among them are 188 hap-ASM DMRs and 933 mQTLs located near GWAS signals for immune and neurological disorders. Targeted bis-seq confirmed hap-ASM in 12/13 loci tested, including CCDC155, CD69, FRMD1, IRF1, KBTBD11, and S100A(∗)-ILF2, associated with immune phenotypes, MYT1L, PTPRN2, CMTM8 and CELF2, associated with neurological disorders, NGFR and HLA-DRB6, associated with both immunological and brain disorders, and ZFP57, a trans-acting regulator of genomic imprinting. Polymorphic CTCF and transcription factor (TF) binding sites were over-represented among hap-ASM DMRs and mQTLs, and analysis of the human data, supplemented by cross-species comparisons to macaques, indicated that CTCF and TF binding likelihood predicts the strength and direction of the allelic methylation asymmetry. These results show that hap-ASM is highly tissue specific; an important trans-acting regulator of genomic imprinting is regulated by this phenomenon; and variation in CTCF and TF binding sites is an underlying mechanism, and maps of hap-ASM and mQTLs reveal regulatory sequences underlying supra- and sub-threshold GWAS peaks in immunological and neurological disorders.


Subject(s)
DNA Methylation , Genomic Imprinting , Haplotypes/genetics , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci , Trans-Activators/genetics , Alleles , Animals , Brain/metabolism , Brain/pathology , Female , Genome-Wide Association Study , Humans , Immune System Diseases/genetics , Macaca mulatta , Macaca radiata , Nervous System Diseases/genetics , Placenta/metabolism , Placenta/pathology , Pregnancy , Species Specificity , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
11.
J Investig Dermatol Symp Proc ; 19(1): S57-S61, 2018 01.
Article in English | MEDLINE | ID: mdl-29273109

ABSTRACT

Autoimmune diseases create a substantial burden of disease, and alopecia areata is among the more prevalent forms. Comorbidities are medical conditions that tend to occur together and may provide etiologic insights, suggest novel therapeutic strategies, and help patients and family members understand the risk of other health conditions. It is well established that having one autoimmune disease increases risk for others because of an underlying shared biology. Precision medicine initiatives are creating vast amounts of data that allow us to efficiently identify comorbidities. A survey across various datasets suggests that patients with autoimmune disease, and patients with alopecia areata in particular, may have comorbid neuropsychiatric and metabolic conditions.


Subject(s)
Alopecia Areata/epidemiology , Animals , Autoimmune Diseases/epidemiology , Big Data , Comorbidity , Female , Humans , Male , Mental Disorders/epidemiology , Metabolic Diseases/epidemiology , Mice , Risk Factors
12.
J Immunol ; 197(4): 1089-99, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27412416

ABSTRACT

Alopecia areata (AA) is an autoimmune disease of the hair follicle that results in hair loss of varying severity. Recently, we showed that IFN-γ-producing NKG2D(+)CD8(+) T cells actively infiltrate the hair follicle and are responsible for its destruction in C3H/HeJ AA mice. Our transcriptional profiling of human and mouse alopecic skin showed that the IFN pathway is the dominant signaling pathway involved in AA. We showed that IFN-inducible chemokines (CXCL9/10/11) are markedly upregulated in the skin of AA lesions, and further, that the IFN-inducible chemokine receptor, CXCR3, is upregulated on alopecic effector T cells. To demonstrate whether CXCL9/10/11 chemokines were required for development of AA, we treated mice with blocking Abs to CXCR3, which prevented the development of AA in the graft model, inhibiting the accumulation of NKG2D(+)CD8(+) T cells in the skin and cutaneous lymph nodes. These data demonstrate proof of concept that interfering with the Tc1 response in AA via blockade of IFN-inducible chemokines can prevent the onset of AA. CXCR3 blockade could be approached clinically in human AA with either biologic or small-molecule inhibition, the latter being particularly intriguing as a topical therapeutic.


Subject(s)
Alopecia Areata/immunology , Receptors, CXCR3/antagonists & inhibitors , T-Lymphocytes/immunology , Animals , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Mice , Mice, Inbred C3H , Polymerase Chain Reaction , Receptors, CXCR3/biosynthesis , Skin/immunology
13.
Exp Dermatol ; 26(6): 536-541, 2017 06.
Article in English | MEDLINE | ID: mdl-27306922

ABSTRACT

Alopecia areata (AA) is a common hair loss disorder of autoimmune aetiology, which often results in pronounced psychological distress. Understanding of the pathophysiology of AA is increasing, due in part to recent genetic findings implicating common variants at several genetic loci. To date, no study has investigated the contribution of copy number variants (CNVs) to AA, a prominent class of genomic variants involved in other autoimmune disorders. Here, we report a genomewide- and a candidate gene-focused CNV analysis performed in a cohort of 585 patients with AA and 1340 controls of Central European origin. A nominally significant association with AA was found for CNVs in the following five chromosomal regions: 4q35.2, 6q16.3, 9p23, 16p12.1 and 20p12.1. The most promising finding was a 342.5-kb associated region in 6q16.3 (duplications in 4/585 patients; 0/1340 controls). The duplications spanned the genes MCHR2 and MCHR2-AS1, implicated in melanin-concentrating hormone (MCH) signalling. These genes have not been implicated in previous studies of AA pathogenesis. However, previous research has shown that MCHR2 affects the scale colour of barfin flounder fish via the induction of melanin aggregation. AA preferentially affects pigmented hairs, and the hair of patients with AA frequently shows a change in colour when it regrows following an acute episode of AA. This might indicate a relationship between AA, pigmentation and MCH signalling. In conclusion, the present results provide suggestive evidence for the involvement of duplications in MCHR2 in AA pathogenesis.


Subject(s)
Alopecia Areata/genetics , DNA Copy Number Variations , Genome-Wide Association Study , Receptors, G-Protein-Coupled/genetics , Receptors, Pituitary Hormone/genetics , Adult , Belgium , Chromosome Mapping , Cohort Studies , Europe , Female , Genotype , Germany , Humans , Hypothalamic Hormones/metabolism , Male , Melanins/metabolism , Netherlands , Pigmentation , Pituitary Hormones/metabolism , Polymorphism, Single Nucleotide , Signal Transduction
15.
Pediatr Dermatol ; 34(5): e271-e272, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28884897

ABSTRACT

Alopecia areata (AA) is a common autoimmune disease and it is challenging to predict which patients will have severe disease. The purpose of this retrospective study was to identify comorbidities in children enrolled in the National Alopecia Areata Registry. Atopic dermatitis was more common in patients with severe AA than in those with mild disease. The most common autoimmune comorbidities were vitiligo, psoriasis, thyroid disease, and juvenile idiopathic arthritis.


Subject(s)
Alopecia Areata/epidemiology , Autoimmune Diseases/epidemiology , Child , Child, Preschool , Comorbidity , Female , Humans , Male , Registries , Retrospective Studies
16.
Proc Natl Acad Sci U S A ; 111(29): 10648-53, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-24989505

ABSTRACT

Mechanisms that regulate the growth of eyelashes have remained obscure. We ascertained two families from Pakistan who presented with familial trichomegaly, or extreme eyelash growth. Using a combination of whole exome sequencing and homozygosity mapping, we identified distinct pathogenic mutations within fibroblast growth factor 5 (FGF5) that underlie the disorder. Subsequent sequencing of this gene in several additional trichomegaly families identified an additional mutation in FGF5. We further demonstrated that hair fibers from forearms of these patients were significantly longer than hairs from control individuals, with an increased proportion in the growth phase, anagen. Using hair follicle organ cultures, we show that FGF5 induces regression of the human hair follicle. We have identified FGF5 as a crucial regulator of hair growth in humans for the first time, to our knowledge, and uncovered a therapeutic target to selectively regulate eyelash growth.


Subject(s)
Fibroblast Growth Factor 5/metabolism , Hair/anatomy & histology , Amino Acid Sequence , Exons/genetics , Female , Fibroblast Growth Factor 5/chemistry , Fibroblast Growth Factor 5/genetics , Hair/growth & development , Hair Follicle/metabolism , Homozygote , Humans , Male , Molecular Sequence Data , Mutation, Missense/genetics , Pedigree , Phenotype , Protein Transport , Sequence Analysis, DNA
17.
Nature ; 464(7291): 1043-7, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20393562

ABSTRACT

Hereditary hypotrichosis simplex is a rare autosomal dominant form of hair loss characterized by hair follicle miniaturization. Using genetic linkage analysis, we mapped a new locus for the disease to chromosome 18p11.22, and identified a mutation (Leu9Arg) in the adenomatosis polyposis down-regulated 1 (APCDD1) gene in three families. We show that APCDD1 is a membrane-bound glycoprotein that is abundantly expressed in human hair follicles, and can interact in vitro with WNT3A and LRP5-two essential components of Wnt signalling. Functional studies show that APCDD1 inhibits Wnt signalling in a cell-autonomous manner and functions upstream of beta-catenin. Moreover, APCDD1 represses activation of Wnt reporters and target genes, and inhibits the biological effects of Wnt signalling during both the generation of neurons from progenitors in the developing chick nervous system, and axis specification in Xenopus laevis embryos. The mutation Leu9Arg is located in the signal peptide of APCDD1, and perturbs its translational processing from the endoplasmic reticulum to the plasma membrane. APCDD1(L9R) probably functions in a dominant-negative manner to inhibit the stability and membrane localization of the wild-type protein. These findings describe a novel inhibitor of the Wnt signalling pathway with an essential role in human hair growth. As APCDD1 is expressed in a broad repertoire of cell types, our findings indicate that APCDD1 may regulate a diversity of biological processes controlled by Wnt signalling.


Subject(s)
Hypotrichosis/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Point Mutation/genetics , Wnt Proteins/antagonists & inhibitors , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Chick Embryo , Chromosome Mapping , Chromosomes, Human, Pair 18/genetics , Genes, Dominant/genetics , Genes, Reporter/genetics , Hair/growth & development , Hair/metabolism , Hair Follicle/growth & development , Hair Follicle/metabolism , Hair Follicle/pathology , Humans , Hypotrichosis/metabolism , Hypotrichosis/pathology , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/deficiency , Membrane Proteins , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Scalp , Signal Transduction , Skin , Spinal Cord/cytology , Stem Cells/cytology , Stem Cells/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , Xenopus Proteins/deficiency , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/genetics , Xenopus laevis/metabolism , beta Catenin/metabolism
18.
Nature ; 466(7302): 113-7, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20596022
19.
Proc Natl Acad Sci U S A ; 110(19): 7790-5, 2013 May 07.
Article in English | MEDLINE | ID: mdl-23603273

ABSTRACT

X-linked congenital generalized hypertrichosis (Online Mendelian Inheritance in Man 307150) is an extremely rare condition of hair overgrowth on different body sites. We previously reported linkage in a large Mexican family with X-linked congenital generalized hypertrichosis cosegregating with deafness and with dental and palate anomalies to Xq24-27. Using SNP oligonucleotide microarray analysis and whole-genome sequencing, we identified a 389-kb interchromosomal insertion at an extragenic palindrome site at Xq27.1 that completely cosegregates with the disease. Among the genes surrounding the insertion, we found that Fibroblast Growth Factor 13 (FGF13) mRNA levels were significantly reduced in affected individuals, and immunofluorescence staining revealed a striking decrease in FGF13 localization throughout the outer root sheath of affected hair follicles. Taken together, our findings suggest a role for FGF13 in hair follicle growth and in the hair cycle.


Subject(s)
Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/physiology , Gene Expression Regulation , Hypertrichosis/congenital , Alternative Splicing , Animals , Chromosome Mapping , Female , Genetic Linkage , Genome, Human , Hair Follicle/growth & development , Hair Follicle/physiology , Heterozygote , Humans , Hypertrichosis/genetics , Keratinocytes/metabolism , Male , Mice , Mutagenesis, Insertional , Pedigree , Polymorphism, Single Nucleotide , RNA, Messenger/metabolism , Sequence Analysis, DNA
20.
Am J Hum Genet ; 88(6): 839-844, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21665001

ABSTRACT

Hereditary leukonychia (porcelain nails or white nails) is a rare nail disorder with an unknown genetic basis. To identify variants in a gene underlying this phenotype, we identified four families of Pakistani origin showing features of hereditary leukonychia. All 20 nails of each affected individual were chalky and white in appearance, consistent with total leukonychia, with no other cutaneous, appendageal, or systemic findings. By using Affymetrix 10K chip, we established linkage to chromosome 3p21.3-p22 with a LOD score (Z) of 5.1. We identified pathogenic mutations in PLCD1 in all four families, which encodes phosphoinositide-specific phospholipase C delta 1 subunit, a key enzyme in phosphoinositide metabolism. We then identified localization of PLCD1 in the nail matrix. It was recently shown that PLCD1 is a component of the human nail plate by proteomic analysis and is localized in the matrix of human nails. Furthermore, mutations detected in PLCD1 resulted in reduced enzymatic activity in vitro. Our data show that mutations in PLCD1 underlie hereditary leukonychia, revealing a gene involved in molecular control of nail growth.


Subject(s)
Chromosomes, Human, Pair 3/genetics , Nails/pathology , Phospholipase C delta/genetics , Humans , Hypopigmentation/enzymology , Hypopigmentation/genetics , Hypopigmentation/pathology , Mutation , Nail Diseases/congenital , Nail Diseases/enzymology , Nail Diseases/genetics , Nail Diseases/pathology , Nails/embryology , Nails/enzymology , Pedigree , Phospholipase C delta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL