Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
2.
JAMA Dermatol ; 158(1): 16-25, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34851365

ABSTRACT

IMPORTANCE: Ichthyoses are clinically and genetically heterogeneous disorders characterized by scaly skin. Despite decades of investigation identifying pathogenic variants in more than 50 genes, clear genotype-phenotype associations have been difficult to establish. OBJECTIVE: To expand the genotypic and phenotypic spectra of ichthyosis and delineate genotype-phenotype associations. DESIGN, SETTING, AND PARTICIPANTS: This cohort study recruited an international group of individuals with ichthyosis and describes characteristic and distinguishing features of common genotypes, including genotype-phenotype associations, during a 10-year period from June 2011 to July 2021. Participants of all ages, races, and ethnicities were included and were enrolled worldwide from referral centers and patient advocacy groups. A questionnaire to assess clinical manifestations was completed by those with a genetic diagnosis. MAIN OUTCOMES AND MEASURES: Genetic analysis of saliva or blood DNA, a phenotyping questionnaire, and standardized clinical photographs. Descriptive statistics, such as frequency counts, were used to describe the cases in the cohort. Fisher exact tests identified significant genotype-phenotype associations. RESULTS: Results were reported for 1000 unrelated individuals enrolled from around the world (mean [SD] age, 50.0 [34.0] years; 524 [52.4%] were female, 427 [42.7%] were male, and 49 [4.9%] were not classified); 75% were from the US, 12% from Latin America, 4% from Canada, 3% from Europe, 3% from Asia, 2% from Africa, 1% from the Middle East, and 1% from Australia and New Zealand. A total of 266 novel disease-associated variants in 32 genes were identified among 869 kindreds. Of these, 241 (91%) pathogenic variants were found through multiplex amplicon sequencing and 25 (9%) through exome sequencing. Among the 869 participants with a genetic diagnosis, 304 participants (35%) completed the phenotyping questionnaire. Analysis of clinical manifestations in these 304 individuals revealed that pruritus, hypohydrosis, skin pain, eye problems, skin odor, and skin infections were the most prevalent self-reported features. Genotype-phenotype association analysis revealed that the presence of a collodion membrane at birth (odds ratio [OR], 6.7; 95% CI, 3.0-16.7; P < .001), skin odor (OR, 2.8; 95% CI, 1.1-6.8; P = .02), hearing problems (OR, 2.9; 95% CI, 1.6-5.5; P < .001), eye problems (OR, 3.0; 95% CI, 1.5-6.0; P < .001), and alopecia (OR, 4.6; 95% CI, 2.4-9.0; P < .001) were significantly associated with TGM1 variants compared with other ichthyosis genotypes studied. Skin pain (OR, 6.8; 95% CI, 1.6-61.2; P = .002), odor (OR, 5.7; 95% CI, 2.0-19.7; P < .001), and infections (OR, 3.1; 95% CI, 1.4-7.7; P = .03) were significantly associated with KRT10 pathogenic variants compared with disease-associated variants in other genes that cause ichthyosis. Pathogenic variants were identified in 869 (86.9%) participants. Most of the remaining individuals had unique phenotypes, enabling further genetic discovery. CONCLUSIONS AND RELEVANCE: This cohort study expands the genotypic and phenotypic spectrum of ichthyosis, establishing associations between clinical manifestations and genotypes. Collectively, the findings may help improve clinical assessment, assist with developing customized management plans, and improve clinical course prognostication.


Subject(s)
Ichthyosis, Lamellar , Ichthyosis , Cohort Studies , Female , Genomics , Humans , Ichthyosis/pathology , Ichthyosis, Lamellar/genetics , Male , Phenotype
3.
Matrix Biol ; 99: 43-57, 2021 05.
Article in English | MEDLINE | ID: mdl-34004352

ABSTRACT

Epidermolysis bullosa (EB) is a genotypically heterogeneous group of disorders characterized by cutaneous blistering and erosions with a tremendous spectrum of severity. One of the distinct forms of EB, Kindler EB (KEB), manifests with blistering and poikiloderma; this subtype of EB is caused by mutations in the FERMT1 gene encoding kindlin-1. In this study, we investigated a patient clinically diagnosed as KEB with reduced FERMT1 gene expression and intensity of immunostaining for kindlin-1. Transmission electron microscopy showed lamina densa reduplication, frequently observed in KEB. However, no mutations were identified in FERMT1 in this patient with consanguineous parents, and this gene resided outside of genomic regions of homozygosity (ROH). Instead, whole-exome sequencing and homozygosity mapping identified a homozygous sequence variant at the +4 position of intron 2 in the USB1 gene, encoding an exoribonuclease required for processing of U6 snRNA, a critical component of spliceosomes. Examination of the patient's RNA by RNA-Seq confirmed the pathogenicity of this variant, causing aberrant splicing predicted to result in loss of function of USB1. Mutations in this gene have been reported in patients with poikiloderma and neutropenia, with a few reported cases in association with skin fragility, a condition distinct from the KEB phenotype. Transcriptome analysis revealed that several genes, expressed in the cutaneous basement membrane zone and previously associated with different subtypes of EB, were differentially downregulated at the mRNA level. EB-associated mRNA downregulation was confirmed at protein levels by skin immunofluorescence. These observations provide a novel mechanism for blistering and erosions in the skin as a result reduced presence of adhesion complexes critical for stable association of epidermis and dermis at the level of cutaneous basement membrane zone.


Subject(s)
Epidermolysis Bullosa , Neutropenia , Basement Membrane , Epidermolysis Bullosa/genetics , Gene Expression , Humans , Membrane Proteins , Mutation , Neoplasm Proteins/genetics , Neutropenia/genetics , Phenotype , Phosphoric Diester Hydrolases , Skin Abnormalities
4.
Am J Hum Genet ; 107(1): 158-163, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32516568

ABSTRACT

The discovery of genetic causes of inherited skin disorders has been pivotal to the understanding of epidermal differentiation, function, and renewal. Here we show via exome sequencing that mutations in ASPRV1 (aspartic peptidase retroviral-like 1) cause a dominant Mendelian disorder featuring palmoplantar keratoderma and lamellar ichthyosis, a phenotype that has otherwise been exclusively recessive. ASPRV1 encodes a mammalian-specific and stratified epithelia-specific protease important in processing of filaggrin, a critical component of the uppermost epidermal layer. Three different heterozygous ASPRV1 missense mutations in four unrelated ichthyosis kindreds segregate with disease and disrupt protein residues within close proximity to each other and autocatalytic cleavage sites. Expression of mutant ASPRV1 proteins demonstrates that all three mutations alter ASPRV1 auto-cleavage and filaggrin processing, a function vital to epidermal barrier integrity.


Subject(s)
Aspartic Acid Endopeptidases/genetics , Heredity/genetics , Ichthyosis, Lamellar/genetics , Mutation, Missense/genetics , Skin Diseases/genetics , Filaggrin Proteins , Heterozygote , Humans , Intermediate Filament Proteins/genetics , Phenotype , Exome Sequencing/methods
5.
Am J Hum Genet ; 105(5): 1023-1029, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31630788

ABSTRACT

We describe unrelated individuals with ichthyosis, failure to thrive, thrombocytopenia, photophobia, and progressive hearing loss. Each have bi-allelic mutations in AP1B1, the gene encoding the ß subunit of heterotetrameric adaptor protein 1 (AP-1) complexes, which mediate endomembrane polarization, sorting, and transport. In affected keratinocytes the AP-1 ß subunit is lost, and the γ subunit is greatly reduced, demonstrating destabilization of the AP-1 complex. Affected cells and tissue contain an abundance of abnormal vesicles and show hyperproliferation, abnormal epidermal differentiation, and derangement of intercellular junction proteins. Transduction of affected cells with wild-type AP1B1 rescues the vesicular phenotype, conclusively establishing that loss of AP1B1 function causes this disorder.


Subject(s)
Adaptor Protein Complex 1/genetics , Adaptor Protein Complex beta Subunits/genetics , Deafness/genetics , Genes, Recessive/genetics , Ichthyosis/genetics , Mutation/genetics , Photophobia/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Female , Hearing Loss/genetics , Humans , Male , Phenotype , Protein Subunits/genetics , Protein Transport/genetics , Thrombocytopenia/genetics
6.
J Invest Dermatol ; 139(2): 380-390, 2019 02.
Article in English | MEDLINE | ID: mdl-30321533

ABSTRACT

Investigation of genetic determinants of Mendelian skin disorders has substantially advanced understanding of epidermal biology. Here we show that mutations in PERP, encoding a crucial component of desmosomes, cause both dominant and recessive human keratoderma. Heterozygosity for a C-terminal truncation, which produces a protein that appears to be unstably incorporated into desmosomes, causes Olmsted syndrome with severe periorificial and palmoplantar keratoderma in multiple unrelated kindreds. Homozygosity for an N-terminal truncation ablates expression and causes widespread erythrokeratoderma, with expansion of epidermal differentiation markers. Both exhibit epidermal hyperproliferation, immature desmosomes lacking a dense midline observed via electron microscopy, and impaired intercellular adhesion upon mechanical stress. Localization of other desmosomal components appears normal, which is in contrast to other conditions caused by mutations in genes encoding desmosomal proteins. These discoveries highlight the essential role of PERP in human desmosomes and epidermal homeostasis and further expand the heterogeneous spectrum of inherited keratinization disorders.


Subject(s)
Desmosomes/pathology , Epidermis/pathology , Keratoderma, Palmoplantar/genetics , Membrane Proteins/genetics , Adult , Cell Adhesion/genetics , Child , Child, Preschool , Codon, Nonsense , DNA Mutational Analysis , Desmosomes/ultrastructure , Epidermis/ultrastructure , Exons/genetics , Female , Frameshift Mutation , Genes, Tumor Suppressor , Heterozygote , Homozygote , Humans , Keratoderma, Palmoplantar/pathology , Male , Membrane Proteins/metabolism , Microscopy, Electron , Young Adult
7.
J Am Acad Dermatol ; 79(3): 487-494, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29477734

ABSTRACT

BACKGROUND: Heterozygous mutations in caspase recruitment domain family member 14 gene (CARD14) have been shown to be associated with psoriasis and familial pityriasis rubra pilaris (PRP). Many subjects with CARD14 mutations display features of both disorders, which can result in diagnostic uncertainty. In addition, these eruptions are often recalcitrant to conventional psoriasis therapies such as methotrexate, oral retinoids, and tumor necrosis factor-α inhibitors. OBJECTIVE: We sought to describe the clinical characteristics, family history, and response to therapy in subjects with papulosquamous eruptions due to mutations in CARD14. METHODS: Subjects were referred for genetic testing as part of a registry of subjects with inherited disorders of keratinization. DNA was isolated from blood or saliva, and multiplex targeted sequencing or whole exome sequencing was performed. Clinical histories of subjects with CARD14 mutations were reviewed. RESULTS: We identified 15 kindreds with CARD14-associated papulosquamous eruption (CAPE). Characteristic features of CAPE include early age of onset; prominent involvement of the cheeks, chin, and ears; family history of psoriasis or PRP; minimal response to conventional topical and systemic psoriasis therapies; and improvement with ustekinumab. LIMITATIONS: Relatively small sample size. CONCLUSIONS: Many subjects with CARD14 mutations display characteristics of both psoriasis and PRP. We propose the term CARD14-associated papulosquamous eruption to describe this spectrum of disease. Subjects with clinical features suggestive of CAPE should undergo CARD14 sequencing and may benefit from treatment with ustekinumab.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , Dermatologic Agents/therapeutic use , Facial Dermatoses/genetics , Guanylate Cyclase/genetics , Membrane Proteins/genetics , Skin Diseases, Papulosquamous/drug therapy , Skin Diseases, Papulosquamous/genetics , Ustekinumab/therapeutic use , Age of Onset , Child , Child, Preschool , Genetic Testing , Humans , Infant , Infant, Newborn , Phenotype , Pityriasis Rubra Pilaris/genetics , Psoriasis/genetics , Psoriasis/therapy , Retreatment
8.
Am J Hum Genet ; 100(6): 978-984, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28575652

ABSTRACT

The discovery of new genetic determinants of inherited skin disorders has been instrumental to the understanding of epidermal function, differentiation, and renewal. Here, we show that mutations in KDSR (3-ketodihydrosphingosine reductase), encoding an enzyme in the ceramide synthesis pathway, lead to a previously undescribed recessive Mendelian disorder in the progressive symmetric erythrokeratoderma spectrum. This disorder is characterized by severe lesions of thick scaly skin on the face and genitals and thickened, red, and scaly skin on the hands and feet. Although exome sequencing revealed several of the KDSR mutations, we employed genome sequencing to discover a pathogenic 346 kb inversion in multiple probands, and cDNA sequencing and a splicing assay established that two mutations, including a recurrent silent third base change, cause exon skipping. Immunohistochemistry and yeast complementation studies demonstrated that the mutations cause defects in KDSR function. Systemic isotretinoin therapy has achieved nearly complete resolution in the two probands in whom it has been applied, consistent with the effects of retinoic acid on alternative pathways for ceramide generation.


Subject(s)
Alcohol Oxidoreductases/genetics , Genes, Recessive , Genetic Predisposition to Disease , Keratosis/enzymology , Keratosis/genetics , Mutation/genetics , Ceramides/biosynthesis , Filaggrin Proteins , Genetic Complementation Test , Heterozygote , Humans , Intermediate Filament Proteins/metabolism , Polymorphism, Single Nucleotide/genetics , RNA Splicing/genetics , Saccharomyces cerevisiae/metabolism
9.
JAMA Dermatol ; 153(6): 537-543, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28403434

ABSTRACT

Importance: Bathing suit ichthyosis (BSI) is a rare congenital disorder of keratinization characterized by restriction of scale to sites of relatively higher temperature such as the trunk, with cooler areas remaining unaffected. Fewer than 40 cases have been reported in the literature. Bathing suit ichthyosis is caused by recessive, temperature-sensitive mutations in the transglutaminase-1 gene (TGM1). Clear genotype-phenotype correlations have been difficult to establish because several of the same TGM1 mutations have been reported in BSI and other forms of congenital ichthyosis. We identify novel and recurrent mutations in 16 participants with BSI. Objective: To expand the genotypic spectrum of BSI, identifying novel TGM1 mutations in patients with BSI, and to use BSI genotypes to draw inferences about the temperature sensitivity of TGM1 mutations. Design, Setting, and Participants: A total of 16 participants with BSI from 13 kindreds were identified from 6 academic medical centers. A detailed clinical history was obtained from each participant, including phenotypic presentation at birth and disease course. Each participant underwent targeted sequencing of TGM1. Main Outcomes and Measures: Phenotypic and genotypic characteristics in these patients from birth onward. Results: Of the 16 participants, 7 were male, and 9 were female (mean age, 12.6 years; range, 1-39 years). We found 1 novel TGM1 indel mutation (Ile469_Cys471delinsMetLeu) and 8 TGM1 missense mutations that to our knowledge have not been previously reported in BSI: 5 have been previously described in non-temperature-sensitive forms of congenital ichthyosis (Arg143Cys, Gly218Ser, Gly278Arg, Arg286Gln, and Ser358Arg), and 3 (Tyr374Cys, Phe495Leu, and Ser772Arg) are novel mutations. Three probands were homozygous for Arg264Trp, Arg286Gln, or Arg315Leu, indicating that these mutations are temperature sensitive. Seven of 10 probands with a compound heterozygous TGM1 genotype had a mutation at either arginine 307 or 315, providing evidence that mutations at these sites are temperature sensitive and highlighting the importance of these residues in the pathogenesis of BSI. Conclusions and Relevance: Our findings expand the genotypic spectrum of BSI and the understanding of temperature sensitivity of TGM1 mutations. Increased awareness of temperature-sensitive TGM1 genotypes should aid in genetic counseling and provide insights into the pathophysiology of TGM1 ichthyoses, transglutaminase-1 enzymatic activity, and potential therapeutic approaches.


Subject(s)
Body Temperature/genetics , Ichthyosis, Lamellar/genetics , Transglutaminases/genetics , Academic Medical Centers , Adolescent , Adult , Child , Child, Preschool , Female , Genotype , Humans , INDEL Mutation , Ichthyosis, Lamellar/physiopathology , Infant , Male , Mutation, Missense , Phenotype , Young Adult
11.
Hum Mol Genet ; 25(2): 348-57, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26604139

ABSTRACT

Disorders of keratinization (DOK) show marked genotypic and phenotypic heterogeneity. In most cases, disease is primarily cutaneous, and further clinical evaluation is therefore rarely pursued. We have identified subjects with a novel DOK featuring erythrokeratodermia and initially-asymptomatic, progressive, potentially fatal cardiomyopathy, a finding not previously associated with erythrokeratodermia. We show that de novo missense mutations clustered tightly within a single spectrin repeat of DSP cause this novel cardio-cutaneous disorder, which we term erythrokeratodermia-cardiomyopathy (EKC) syndrome. We demonstrate that DSP mutations in our EKC syndrome subjects affect localization of desmosomal proteins and connexin 43 in the skin, and result in desmosome aggregation, widening of intercellular spaces, and lipid secretory defects. DSP encodes desmoplakin, a primary component of desmosomes, intercellular adhesion junctions most abundant in the epidermis and heart. Though mutations in DSP are known to cause other disorders, our cohort features the unique clinical finding of severe whole-body erythrokeratodermia, with distinct effects on localization of desmosomal proteins and connexin 43. These findings add a severe, previously undescribed syndrome featuring erythrokeratodermia and cardiomyopathy to the spectrum of disease caused by mutation in DSP, and identify a specific region of the protein critical to the pathobiology of EKC syndrome and to DSP function in the heart and skin.


Subject(s)
Cardiomyopathies/genetics , Desmoplakins/genetics , Desmosomes/metabolism , Mutation, Missense , Skin Diseases, Genetic/genetics , Amino Acid Sequence , Cardiomyopathies/metabolism , Child , Child, Preschool , Connexin 43/metabolism , Desmoplakins/metabolism , Female , Humans , Infant , Male , Molecular Sequence Data , Myocardium/metabolism , Protein Transport , Sequence Alignment , Skin/metabolism , Skin Diseases, Genetic/metabolism , Syndrome
12.
J Invest Dermatol ; 135(6): 1540-1547, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25398053

ABSTRACT

Genetic investigation of inherited skin disorders has informed the understanding of skin self-renewal, differentiation, and barrier function. Erythrokeratodermia variabilis et progressiva (EKVP) is a rare, inherited skin disease that is characterized by transient figurate patches of erythema, localized or generalized scaling, and frequent palmoplantar keratoderma. By using exome sequencing, we show that de novo missense mutations in GJA1 (gap junction protein alpha 1) cause EKVP. The severe, progressive skin disease in EKVP subjects with GJA1 mutations is distinct from limited cutaneous findings rarely found in the systemic disorder oculodentodigital dysplasia, also caused by dominant GJA1 mutations. GJA1 encodes connexin 43 (Cx43), the most widely expressed gap junction protein. We show that the GJA1 mutations in EKVP subjects lead to disruption of Cx43 membrane localization and aggregation within the Golgi. These findings reveal a critical role for Cx43 in epidermal homeostasis, and they provide evidence of organ-specific pathobiology resulting from different mutations within GJA1.


Subject(s)
Connexin 43/genetics , Craniofacial Abnormalities/genetics , Erythrokeratodermia Variabilis/genetics , Eye Abnormalities/genetics , Foot Deformities, Congenital/genetics , Mutation , Skin Diseases/genetics , Syndactyly/genetics , Tooth Abnormalities/genetics , Amino Acid Sequence , Cell Membrane/metabolism , Child , Child, Preschool , Connexins/genetics , Disease Progression , Exome , Female , Golgi Apparatus/metabolism , HeLa Cells , Humans , Immunohistochemistry , Male , Molecular Sequence Data , Mutagenesis, Site-Directed , Phenotype , Sequence Analysis, DNA , Sequence Homology, Amino Acid
13.
Pediatr Dermatol ; 30(4): 409-15, 2013.
Article in English | MEDLINE | ID: mdl-23662773

ABSTRACT

Capillary malformation-arteriovenous malformation syndrome is an autosomal dominant disorder caused by mutations in the RASA1 gene and characterized by multiple small, round to oval capillary malformations with or without arteriovenous malformations. Ateriovenous malformations occur in up to one-third of patients and may involve the brain and spine. Although making the diagnosis is straightforward in some patients, there are other patients for whom diagnostic criteria may be helpful in their evaluation. Here we review the literature regarding capillary malformation-arteriovenous malformation syndrome, propose diagnostic criteria, and discuss the care of patients with this condition.


Subject(s)
Arteriovenous Malformations/diagnosis , Arteriovenous Malformations/therapy , Capillaries/abnormalities , Port-Wine Stain/diagnosis , Port-Wine Stain/therapy , Skin Diseases/diagnosis , Skin Diseases/therapy , Vascular Malformations/diagnosis , Vascular Malformations/therapy , Child , Dermatology , Humans , Pediatrics
16.
Nature ; 482(7383): 98-102, 2012 Jan 22.
Article in English | MEDLINE | ID: mdl-22266938

ABSTRACT

Hypertension affects one billion people and is a principal reversible risk factor for cardiovascular disease. Pseudohypoaldosteronism type II (PHAII), a rare Mendelian syndrome featuring hypertension, hyperkalaemia and metabolic acidosis, has revealed previously unrecognized physiology orchestrating the balance between renal salt reabsorption and K(+) and H(+) excretion. Here we used exome sequencing to identify mutations in kelch-like 3 (KLHL3) or cullin 3 (CUL3) in PHAII patients from 41 unrelated families. KLHL3 mutations are either recessive or dominant, whereas CUL3 mutations are dominant and predominantly de novo. CUL3 and BTB-domain-containing kelch proteins such as KLHL3 are components of cullin-RING E3 ligase complexes that ubiquitinate substrates bound to kelch propeller domains. Dominant KLHL3 mutations are clustered in short segments within the kelch propeller and BTB domains implicated in substrate and cullin binding, respectively. Diverse CUL3 mutations all result in skipping of exon 9, producing an in-frame deletion. Because dominant KLHL3 and CUL3 mutations both phenocopy recessive loss-of-function KLHL3 mutations, they may abrogate ubiquitination of KLHL3 substrates. Disease features are reversed by thiazide diuretics, which inhibit the Na-Cl cotransporter in the distal nephron of the kidney; KLHL3 and CUL3 are expressed in this location, suggesting a mechanistic link between KLHL3 and CUL3 mutations, increased Na-Cl reabsorption, and disease pathogenesis. These findings demonstrate the utility of exome sequencing in disease gene identification despite the combined complexities of locus heterogeneity, mixed models of transmission and frequent de novo mutation, and establish a fundamental role for KLHL3 and CUL3 in blood pressure, K(+) and pH homeostasis.


Subject(s)
Carrier Proteins/genetics , Cullin Proteins/genetics , Hypertension/genetics , Mutation/genetics , Pseudohypoaldosteronism/genetics , Water-Electrolyte Imbalance/genetics , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Base Sequence , Blood Pressure/genetics , Carrier Proteins/chemistry , Cohort Studies , Cullin Proteins/chemistry , Electrolytes , Exons/genetics , Female , Gene Expression Profiling , Genes, Dominant/genetics , Genes, Recessive/genetics , Genotype , Homeostasis/genetics , Humans , Hydrogen-Ion Concentration , Hypertension/complications , Hypertension/physiopathology , Male , Mice , Microfilament Proteins , Models, Molecular , Molecular Sequence Data , Phenotype , Potassium/metabolism , Pseudohypoaldosteronism/complications , Pseudohypoaldosteronism/physiopathology , Sodium Chloride/metabolism , Water-Electrolyte Imbalance/complications , Water-Electrolyte Imbalance/physiopathology
17.
Proc Natl Acad Sci U S A ; 108(8): 3330-5, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21300860

ABSTRACT

αß T-cell repertoire selection is mediated by peptide-MHC complexes presented by thymic epithelial or myeloid cells, and by lipid-CD1 complexes expressed by thymocytes. γδ T-cell repertoire selection, by contrast, is largely unresolved. Mice mutant for Skint-1, a unique Ig superfamily gene, do not develop canonical Vγ5Vδ1(+) dendritic epidermal T cells. This study shows that transgenic Skint-1, across a broad range of expression levels, precisely and selectively determines the Vγ5Vδ1(+) dendritic epidermal T-cell compartment. Skint-1 is expressed by medullary thymic epithelial cells, and unlike lipid-CD1 complexes, must be expressed by stromal cells to function efficiently. Its unusual transmembrane-cytoplasmic regions severely limit cell surface expression, yet increasing this or, conversely, retaining Skint1 intracellularly markedly compromises function. Each Skint1 domain appears nonredundant, including a unique decamer specifying IgV-domain processing. This investigation of Skint-1 biology points to complex events underpinning the positive selection of an intraepithelial γδ repertoire.


Subject(s)
Epidermis/immunology , Immunoglobulins/immunology , T-Lymphocytes/immunology , Animals , Dendritic Cells , Epidermal Cells , Epithelial Cells , Gene Expression/immunology , Immunoglobulins/genetics , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocyte Subsets
18.
Nat Genet ; 40(5): 656-62, 2008 May.
Article in English | MEDLINE | ID: mdl-18408721

ABSTRACT

B cells, alphabeta T cells and gammadelta T cells are conserved lymphocyte subtypes encoding their antigen receptors from somatically rearranged genes. alphabeta T cells undergo positive selection in the thymus by engagement of their T cell receptors (TCRs) with self-peptides presented by major histocompatibility complex molecules. The molecules that select gammadelta T cells are unknown. Vgamma5+Vdelta1+ cells comprise 90% of mouse epidermal gammadelta T cells. By mapping and genetic complementation using a strain showing loss of Vgamma5+Vdelta1+ cells due to a failure of thymic selection, we show that this defect is caused by mutation in Skint1, a newly identified gene expressed in thymus and skin that encodes a protein with immunoglobulin-like and transmembrane domains. Skint1 is the prototypic member of a rapidly evolving family of at least 11 genes in mouse, with greatest similarity to the butyrophilin genes. These findings define a new family of proteins mediating key epithelial-immune interactions.


Subject(s)
Epidermis/immunology , Immunoglobulins/genetics , Immunoglobulins/immunology , Multigene Family , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Codon, Nonsense , Genetic Linkage , Haplotypes , Immunoglobulins/chemistry , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Protein Conformation , Receptors, Antigen, T-Cell, gamma-delta/analysis
19.
N Engl J Med ; 346(20): 1513-21, 2002 May 16.
Article in English | MEDLINE | ID: mdl-12015390

ABSTRACT

BACKGROUND: Osteoporosis is a major public health problem of largely unknown cause. Loss-of-function mutations in the gene for low-density lipoprotein receptor-related protein 5 (LRP5), which acts in the Wnt signaling pathway, have been shown to cause osteoporosis-pseudoglioma. METHODS: We performed genetic and biochemical analyses of a kindred with an autosomal dominant syndrome characterized by high bone density, a wide and deep mandible, and torus palatinus. RESULTS: Genetic analysis revealed linkage of the syndrome to chromosome 11q12-13 (odds of linkage, >1 million to 1), an interval that contains LRP5. Affected members of the kindred had a mutation in this gene, with valine substituted for glycine at codon 171 (LRP5V171). This mutation segregated with the trait in the family and was absent in control subjects. The normal glycine lies in a so-called propeller motif that is highly conserved from fruit flies to humans. Markers of bone resorption were normal in the affected subjects, whereas markers of bone formation such as osteocalcin were markedly elevated. Levels of fibronectin, a known target of signaling by Wnt, a developmental protein, were also elevated. In vitro studies showed that the normal inhibition of Wnt signaling by another protein, Dickkopf-1 (Dkk-1), was defective in the presence of LRP5V171 and that this resulted in increased signaling due to unopposed Wnt activity. CONCLUSIONS: The LRP5V171 mutation causes high bone density, with a thickened mandible and torus palatinus, by impairing the action of a normal antagonist of the Wnt pathway and thus increasing Wnt signaling. These findings demonstrate the role of altered LRP5 function in high bone mass and point to Dkk as a potential target for the prevention or treatment of osteoporosis.


Subject(s)
Bone Density/genetics , Point Mutation , Proto-Oncogene Proteins/metabolism , Receptors, LDL/genetics , Zebrafish Proteins , Biomarkers/blood , Case-Control Studies , Chromosomes, Human, Pair 11 , Female , Genes, Dominant , Genotype , Humans , Intercellular Signaling Peptides and Proteins , LDL-Receptor Related Proteins , Low Density Lipoprotein Receptor-Related Protein-5 , Male , Mandible/diagnostic imaging , Mandible/pathology , Mutation, Missense , Osteogenesis/physiology , Palate/pathology , Pedigree , Proteins/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Radiography , Receptors, LDL/metabolism , Signal Transduction/physiology , Syndrome , Wnt Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...