Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cancer Lett ; 577: 216427, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37838280

ABSTRACT

Tumor cell migration, specifically epithelial-mesenchymal transition (EMT), serves as a key contributor to treatment failure in colon cancer patients. However, the limited comprehension of its genetic and biological aspects presents challenges for its investigation. EDAR-associated death domain (EDARADD), an important TNFR superfamily member, is elevated in colon cancer. However, it remains unclear about the exact role of EDARADD in the progression of colon cancer metastasis. In this study, we initially demonstrated that both protein and mRNA levels of EDDARADD are elevated in colon cancer tissues and cells, associated with reduced overall survival. Furthermore, functional experiments demonstrated that EDARADD promotes colon cancer cell proliferation and participates in EMT both in vitro and vivo. Mechanistically, Co-IP verified EDARADD could stabilize Snail1 by interacting with E3 ubiquitin ligase Trim21 to inhibit ubiquitination of Snail1. Interestingly, RNA-seq and ubiquitination assay revealed EDARADD's dual downregulation of Trim21 expression at the translational level via Cul1-mediated ubiquitin degradation, and at the transcriptional level through PPARa regulation. Moreover, EDARADD activates NF-κB signaling and experiences feedback transcriptional regulation by p65. In conclusion, this study highlights the signal pathway of EDARADD-PPARa-Trim21-Snail1-EMT and a feedback regulation of NF-κB signaling on EDARADD, which indicated EDARADD as an emerging therapeutic target for colon cancer.


Subject(s)
Colonic Neoplasms , Ubiquitin-Protein Ligases , Humans , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Cell Line, Tumor , Ubiquitination , Colonic Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Edar-Associated Death Domain Protein/genetics , Edar-Associated Death Domain Protein/metabolism
2.
Urol Oncol ; 40(8): 382.e15-382.e24, 2022 08.
Article in English | MEDLINE | ID: mdl-35637063

ABSTRACT

PURPOSE: Bladder cancer is a kind of common malignant cancer in the urinary system. The expression of EDARADD (ectodysplasin-A receptor-associated death domain) in bladder cancer is higher than the normal samples. However, its role in bladder cancer remains unknown. In the present study, we analyzed the expression of EDARADD in 81 bladder cancer samples by immunohistochemistry as well as its correlation with clinical characteristics. In addition, the role of EDARADD was also explored through loss of function. MATERIALS AND METHODS: Cell proliferation assay and MTT assay were conducted to assess the proliferation of bladder cancer cells and transwell assay and wound healing assay were conducted to assess the migration of bladder cancer cells. On the other hand, the levels of epithelial-mesenchymal transition (EMT) associated proteins and the key molecules in the MAPK signaling pathway were detected by western blot. In vivo experiments were also conducted to determine the effect of EDARADD silencing on the metastasis of bladder cancer cells and the MAPK signaling pathway. RESULTS: EDARADD was highly expressed in bladder cancer samples, especially in high-grade bladder cancer samples. The high EDARADD level indicated a poor survival. Interestingly, EDARADD silencing suppressed the proliferation, migration and EMT of bladder cancer cells. Furthermore, the MAPK signaling pathway was repressed by EDARADD silencing. Additionally, silencing EDARADD also inhibited the metastasis of bladder cancer and the MAPK signaling pathway in vivo. It is indicated that silencing EDARADD may suppress the proliferation and metastasis of bladder cancer cells through the MAPK signaling pathway. CONCLUSION: These results indicate that EDARADD may become a probable target for the treatment of bladder cancer.


Subject(s)
Urinary Bladder Neoplasms , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Edar-Associated Death Domain Protein/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness/genetics , Urinary Bladder Neoplasms/pathology
3.
J Forensic Sci ; 66(4): 1524-1532, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33942892

ABSTRACT

It has already been proposed that a combined use of different molecular and morphological markers of aging in multivariate models may result in a greater accuracy of age estimation. However, such an approach can be complex and expensive, and not every combination may be useful. The significance and usefulness of combined analyses of D-aspartic acid in dentine, pentosidine in dentine, DNA methylation in buccal swabs at five genomic regions (PDE4C, RPA2, ELOVL2, DDO, and EDARADD), and third molar mineralization were tested by investigating a sample of 90 oral surgery patients. Machine learning models for age estimation were trained and evaluated, and the contribution of each parameter to multivariate models was tested by assessment of the predictor importance. For models based on D-aspartic acid, pentosidine, and the combination of both, mean absolute errors (MAEs) of 2.93, 3.41, and 2.68 years were calculated, respectively. The additional inclusion of the five DNAm markers did not improve the results. The sole DNAm-based model revealed a MAE of 4.14 years. In individuals under 28 years of age, the combination of the DNAm markers with the third molar mineralization stages reduced the MAE from 3.85 to 2.81 years. Our findings confirm that the combination of parameters in multivariate models may be very useful for age estimation. However, the inclusion of many parameters does not necessarily lead to better results. It is a task for future research to identify the best selection of parameters for the different requirements in forensic practice.


Subject(s)
Age Determination by Teeth/methods , Adolescent , Adult , Aged , Arginine/analogs & derivatives , Arginine/metabolism , Biomarkers/metabolism , Child , CpG Islands/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , D-Aspartate Oxidase/metabolism , D-Aspartic Acid/metabolism , DNA Methylation , Dentin/metabolism , Edar-Associated Death Domain Protein/metabolism , Fatty Acid Elongases/metabolism , Humans , Lysine/analogs & derivatives , Lysine/metabolism , Machine Learning , Middle Aged , Molar, Third/growth & development , Multivariate Analysis , Replication Protein A/metabolism , Tooth Calcification , Young Adult
4.
J Dermatol ; 46(8): 710-715, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31245878

ABSTRACT

Hypohidrotic ectodermal dysplasia (HED) is a rare genetic disorder characterized by hypotrichosis, hypohidrosis and hypodontia. The disease shows X-linked recessive, autosomal dominant or autosomal recessive inheritance traits. The X-linked form of HED is caused by mutations in the EDA gene, while autosomal forms result from mutations in either EDAR or EDARADD genes. Regarding recessive mutations in the EDAR gene, the pathomechanisms have been well characterized. However, it has remained largely unknown how dominant mutations in the EDAR cause HED. In this study, we performed in vitro analyses for a dominant EDAR gene mutation, p.F398*, as a representative. We showed that the p.F398* mutant EDAR completely lost its affinity to EDARADD, and suppressed the downstream nuclear factor-κB activation induced by wild-type EDAR in a dominant-negative manner. Furthermore, we demonstrated that the mutant EDAR was capable of binding with the wild-type EDAR, which led to reduced interaction between the wild-type EDAR and EDARADD. Our findings not only underscore an essential role of the interaction between EDAR and EDARADD in ectodermal development, but also disclose, in part, the molecular basis of autosomal dominant HED.


Subject(s)
Ectodermal Dysplasia 1, Anhidrotic/genetics , Edar Receptor/genetics , Edar-Associated Death Domain Protein/metabolism , Genes, Dominant/genetics , Ectodermal Dysplasia 1, Anhidrotic/pathology , Edar Receptor/metabolism , HEK293 Cells , Humans , Mutation
5.
EMBO J ; 36(1): 61-78, 2017 01 04.
Article in English | MEDLINE | ID: mdl-27908934

ABSTRACT

Cell proliferation is essential to rapid tissue growth and repair, but can result in replication-associated genome damage. Here, we implicate the transcription factor Gata6 in adult mouse hair follicle regeneration where it controls the renewal of rapidly proliferating epithelial (matrix) progenitors and hence the extent of production of terminally differentiated lineages. We find that Gata6 protects against DNA damage associated with proliferation, thus preventing cell cycle arrest and apoptosis. Furthermore, we show that in vivo Gata6 stimulates EDA-receptor signaling adaptor Edaradd level and NF-κB pathway activation, known to be important for DNA damage repair and stress response in general and for hair follicle growth in particular. In cultured keratinocytes, Edaradd rescues DNA damage, cell survival, and proliferation of Gata6 knockout cells and restores MCM10 expression. Our data add to recent evidence in embryonic stem and neural progenitor cells, suggesting a model whereby developmentally regulated transcription factors protect from DNA damage associated with proliferation at key stages of rapid tissue growth. Our data may add to understanding why Gata6 is a frequent target of amplification in cancers.


Subject(s)
Cell Proliferation , GATA6 Transcription Factor/metabolism , Hair Follicle/cytology , Stem Cells/physiology , Animals , Cell Survival , DNA Repair , Edar-Associated Death Domain Protein/metabolism , Mice , Minichromosome Maintenance Proteins/metabolism , NF-kappa B/metabolism
6.
Am J Med Genet A ; 170A(1): 249-53, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26440664

ABSTRACT

Hypohidrotic ectodermal dysplasia (HED) is a rare disorder characterized by deficient development of structures derived from the ectoderm including hair, nails, eccrine glands, and teeth. HED forms that are caused by mutations in the genes EDA, EDAR, or EDARADD may show almost identical phenotypes, explained by a common signaling pathway. Proper interaction of the proteins encoded by these three genes is important for the activation of the NF-κB signaling pathway and subsequent transcription of the target genes. Mutations in the gene EDARADD are most rarely implicated in HED. Here we describe a novel missense mutation, c.367G>A (p.Asp123Asn), in this gene which did not appear to influence the interaction between EDAR and EDARADD proteins, but led to an impaired ability to activate NF-κB signaling. Female members of the affected family showed either unilateral or bilateral amazia. In addition, an affected girl developed bilateral ovarian teratomas, possibly associated with her genetic condition.


Subject(s)
Ectodermal Dysplasia 1, Anhidrotic/genetics , Edar Receptor/genetics , Edar-Associated Death Domain Protein/genetics , Mutation, Missense/genetics , Ovarian Neoplasms/genetics , Teratoma/genetics , Adolescent , Breast Diseases/genetics , Edar Receptor/metabolism , Edar-Associated Death Domain Protein/metabolism , Female , Hair/growth & development , Humans , Male , NF-kappa B/metabolism , Ovarian Neoplasms/pathology , Pedigree , Signal Transduction/genetics , Teratoma/pathology
7.
BMC Evol Biol ; 15: 129, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26134525

ABSTRACT

BACKGROUND: Only a handful of signaling pathways are major actors of development and responsible for both the conservation and the diversification of animal morphologies. To explain this twofold nature, gene duplication and enhancer evolution were predominantly put forth as tinkering mechanisms whereas the evolution of alternative isoforms has been, so far, overlooked. We investigate here the role of gain and loss of isoforms using Edaradd, a gene of the Ecodysplasin pathway, implicated in morphological evolution. A previous study had suggested a scenario of isoform gain and loss with an alternative isoform (A) newly gained in mammals but secondarily lost in mouse lineage. RESULTS: For a comprehensive view of A and B Edaradd isoforms history during mammal evolution, we obtained sequences for both isoforms in representative mammals and performed in vitro translations to support functional predictions. We showed that the ancestral B isoform is well conserved, whereas the mammal-specific A isoform was lost at least 7 times independently in terminal lineages throughout mammal phylogeny. Then, to gain insights into the functional relevance of this evolutionary pattern, we compared the biological function of these isoforms: i) In cellulo promoter assays showed that they are transcribed from two alternative promoters, only B exhibiting feedback regulation. ii) RT-PCR in various tissues and ENCODE data suggested that B isoform is systematically expressed whereas A isoform showed a more tissue-specific expression. iii) Both isoforms activated the NF-κB pathway in an in cellulo reporter assay, albeit at different levels and with different dynamics since A isoform exhibited feedback regulation at the protein level. Finally, only B isoform could rescue a zebrafish edaradd knockdown. CONCLUSIONS: These results suggest that the newly evolved A isoform enables modulating EDA signaling in specific conditions and with different dynamics. We speculate that during mammal diversification, A isoform regulation may have evolved rapidly, accompanying and possibly supporting the diversity of ectodermal appendages, while B isoform may have ensured essential roles. This study makes the case to pay greater attention to mosaic loss of evolutionarily speaking "young" isoforms as an important mechanism underlying phenotypic diversity and not simply as a manifestation of neutral evolution.


Subject(s)
Edar-Associated Death Domain Protein/genetics , Evolution, Molecular , Mammals/genetics , Protein Isoforms/genetics , Signal Transduction , Animals , Edar-Associated Death Domain Protein/metabolism , Gene Duplication , Mammals/classification , Mice , Phylogeny , Promoter Regions, Genetic , Rats , Zebrafish/genetics , Zebrafish/metabolism
9.
Trends Genet ; 30(1): 24-31, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24070496

ABSTRACT

The ectodysplasin (EDA) pathway, which is active during the development of ectodermal organs, including teeth, hairs, feathers, and mammary glands, and which is crucial for fine-tuning the developmental network controlling the number, size, and density of these structures, was discovered by studying human patients affected by anhidrotic/hypohidrotic ectodermal dysplasia. It comprises three main gene products: EDA, a ligand that belongs to the tumor necrosis factor (TNF)-α family, EDAR, a receptor related to the TNFα receptors, and EDARADD, a specific adaptor. This core pathway relies on downstream NF-κB pathway activation to regulate target genes. The pathway has recently been found to be associated with specific adaptations in natural populations: the magnitude of armor plates in sticklebacks and the hair structure in Asian human populations. Thus, despite its role in human disease, the EDA pathway is a 'hopeful pathway' that could allow adaptive changes in ectodermal appendages which, as specialized interfaces with the environment, are considered hot-spots of morphological evolution.


Subject(s)
Adaptation, Physiological/genetics , Ectodermal Dysplasia 1, Anhidrotic/genetics , Ectodysplasins/genetics , Animals , Ectodysplasins/metabolism , Edar Receptor/genetics , Edar Receptor/metabolism , Edar-Associated Death Domain Protein/genetics , Edar-Associated Death Domain Protein/metabolism , Gene Expression Regulation , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Vertebrates/genetics
10.
Stem Cell Reports ; 1(1): 19-27, 2013.
Article in English | MEDLINE | ID: mdl-24052938

ABSTRACT

Current models of how mouse tail interfollicular epidermis (ife) is maintained overlook the coexistence of two distinct terminal differentiation programs: parakeratotic (scale) and orthokeratotic (interscale). lineage tracing and clonal analysis revealed that scale and interscale are maintained by unipotent cells in the underlying basal layer, with scale progenitors dividing more rapidly than interscale progenitors. Although scales are pigmented and precisely aligned with hair follicles, melanocytes and follicles were not necessary for scale differentiation. Epidermal Wnt signaling was required for scale enlargement during development and for postnatal maintenance of scale-interscale boundaries. Loss of Edaradd inhibited ventral scale formation, whereas loss of Lrig1 led to scale enlargement and fusion. In wild-type skin, Lrig1 was not expressed in IFE but was selectively upregulated in dermal fibroblasts underlying the interscale. We conclude that the different IFE differentiation compartments are maintained by distinct stem cell populations and are regulated by epidermal and dermal signals.


Subject(s)
Cell Lineage , Edar-Associated Death Domain Protein/metabolism , Epidermis/metabolism , Hair Follicle/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Wnt Proteins/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Cell Cycle , Cell Differentiation , Edar-Associated Death Domain Protein/genetics , Epidermal Cells , Fibroblasts/cytology , Fibroblasts/metabolism , Hair Follicle/cytology , Melanocytes/cytology , Melanocytes/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Tail , Wnt Proteins/genetics , Wnt Signaling Pathway
11.
Br J Dermatol ; 168(3): 629-33, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22924441

ABSTRACT

BACKGROUND: Hypohidrotic ectodermal dysplasia (HED) is a rare condition characterized by hypotrichosis, hypohidrosis and hypodontia. A de novo heterozygous mutation in the tumour necrosis factor receptor-associated factor 6 gene (TRAF6) was recently identified in a patient with HED, while functional consequences resulting from the mutation remained unknown. OBJECTIVES: To determine the mechanism by which the TRAF6 mutation results in HED. METHODS: We performed coimmunoprecipitation (co-IP) studies to determine whether the mutation would affect the interaction of TRAF6 with transforming growth factor ß-activated kinase 1 (TAK1), TAK1-binding protein 2 (TAB 2) and ectodysplasin-A receptor-associated death domain protein (EDARADD). We then performed co-IP and glutathione S-transferase-pulldown assays to determine the TRAF6 binding sequences in EDARADD. In addition, we analysed the effect of the mutant TRAF6 protein on the affinity between wild-type TRAF6 and EDARADD, as well as on EDARADD-mediated nuclear factor (NF)-κB activation. RESULTS: The mutant TRAF6 protein was capable of forming a complex with TAK1 and TAB 2 in a similar way to wild-type TRAF6. However, the mutant TRAF6 protein completely lost the affinity to EDARADD, while the wild-type TRAF6 bound to the N-terminal domain of EDARADD. Furthermore, the mutant TRAF6 inhibited the interaction between the wild-type TRAF6 and EDARADD, and also potentially reduced the EDARADD-mediated NF-κB activity. CONCLUSIONS: We conclude that the mutant TRAF6 protein shows a dominant negative effect against the wild-type TRAF6 protein, which is predicted to affect the EDARADD-mediated activation of NF-κB during the development of ectoderm-derived organs, and to lead to the HED phenotype.


Subject(s)
Ectodermal Dysplasia, Hypohidrotic, Autosomal Recessive/genetics , Mutation/genetics , TNF Receptor-Associated Factor 6/genetics , Adaptor Proteins, Signal Transducing/metabolism , Drug Interactions , Edar Receptor/genetics , Edar Receptor/metabolism , Edar-Associated Death Domain Protein/genetics , Edar-Associated Death Domain Protein/metabolism , Humans , Immunoprecipitation , NF-kappa B/metabolism , TNF Receptor-Associated Factor 6/metabolism
12.
BMC Genet ; 12: 91, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-22013926

ABSTRACT

BACKGROUND: Hypohidrotic ectodermal dysplasia (HED) is a congenital disorder characterized by sparse hair, oligodontia, and inability to sweat. It is caused by mutations in any of three Eda pathway genes: ectodysplasin (Eda), Eda receptor (Edar), and Edar-associated death domain (Edaradd), which encode ligand, receptor, and intracellular adaptor molecule, respectively. The Eda signaling pathway activates NF-κB, which is central to ectodermal differentiation. Although the causative genes and the molecular pathway affecting HED have been identified, no curative treatment for HED has been established. Previously, we found a rat spontaneous mutation that caused defects in hair follicles and named it sparse-and-wavy (swh). Here, we have established the swh rat as the first rat model of HED and successfully identified the swh mutation. RESULTS: The swh/swh rat showed sparse hair, abnormal morphology of teeth, and absence of sweat glands. The ectoderm-derived glands, meibomian, preputial, and tongue glands, were absent. We mapped the swh mutation to the most telomeric part of rat Chr 7 and found a Pro153Ser missense mutation in the Edaradd gene. This mutation was located in the death domain of EDARADD, which is crucial for signal transduction and resulted in failure to activate NF-κB. CONCLUSIONS: These findings suggest that swh is a loss-of-function mutation in the rat Edaradd and indicate that the swh/swh rat would be an excellent animal model of HED that could be used to investigate the pathological basis of the disease and the development of new therapies.


Subject(s)
Ectodermal Dysplasia, Hypohidrotic, Autosomal Recessive/genetics , Edar-Associated Death Domain Protein/genetics , Edar-Associated Death Domain Protein/metabolism , NF-kappa B/metabolism , Animals , Chromosome Mapping , Disease Models, Animal , Ectodermal Dysplasia, Hypohidrotic, Autosomal Recessive/pathology , HEK293 Cells , Humans , Hypohidrosis/genetics , Mice , Mutation, Missense/genetics , NF-kappa B/genetics , Rats , Sweat Glands/pathology
13.
Dermatology ; 223(1): 74-9, 2011.
Article in English | MEDLINE | ID: mdl-21876339

ABSTRACT

BACKGROUND: Hypohidrotic ectodermal dysplasia (HED) is a rare condition characterized by hypotrichosis, hypohidrosis and hypodontia. The disease shows X-linked recessive, autosomal-dominant or autosomal-recessive inheritance trait. X-linked form of HED is caused by mutations in the EDA gene, while autosomal forms are caused by mutations in either EDAR or EDARADD genes. METHODS: We analyzed the DNA from a Japanese patient with HED through direct sequencing, and also performed functional studies for the mutation. RESULTS: We identified a homozygous missense mutation c.1073G>A (p.R358Q) in the EDAR gene of the patient, which was a nonconservative amino acid substitution within the death domain of EDAR protein. We demonstrated that the p.R358Q mutant EDAR protein lost its affinity to EDARADD, leading to reduced activation of the downstream NF-κB. CONCLUSION: Our data further suggest the crucial role of the EDAR signaling in development of hair, teeth, and sweat gland in humans.


Subject(s)
Ectodermal Dysplasia 1, Anhidrotic/genetics , Edar Receptor/genetics , Edar-Associated Death Domain Protein/metabolism , Mutation, Missense , Ectodermal Dysplasia 1, Anhidrotic/metabolism , Female , Humans , Infant , NF-kappa B/metabolism , Sequence Analysis, DNA , Signal Transduction
14.
Dev Dyn ; 239(10): 2674-84, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20803597

ABSTRACT

Hypohidrotic ectodermal dysplasia (HED) is characterized by defective ectodermal organ development. This includes the salivary glands (SGs), which have an important role in lubricating the oral cavity. In humans and mice, HED is caused by mutations in Ectodysplasin A (Eda) pathway genes. Various phenotypes of the mutant mouse Eda(Ta/Ta), which lacks the ligand Eda, can be rescued by maternal injection or in vitro culture supplementation with recombinant EDA. However, the response of the SGs to this treatment has not been investigated. Here, we show that the submandibular glands (SMGs) of Eda(Ta/Ta) mice exhibit impaired branching morphogenesis, and that supplementation of Eda(Ta/Ta) SMG explants with recombinant EDA rescues the defect. Supplementation of Edar(dlJ/dlJ) SMGs with recombinant Sonic hedgehog (Shh) also rescues the defect, whereas treatment with recombinant Fgf8 does not. This work is the first to test the ability of putative Eda target molecules to rescue Eda pathway mutant SMGs.


Subject(s)
Ectodysplasins/metabolism , Hedgehog Proteins/metabolism , Salivary Glands/metabolism , Animals , Ectodysplasins/genetics , Edar Receptor/genetics , Edar Receptor/metabolism , Edar-Associated Death Domain Protein/genetics , Edar-Associated Death Domain Protein/metabolism , Genotype , Hedgehog Proteins/genetics , In Situ Hybridization , Mice , Mice, Mutant Strains , Morphogenesis/genetics , Morphogenesis/physiology , Organ Culture Techniques , Salivary Glands/embryology , Signal Transduction/genetics , Signal Transduction/physiology
15.
Arch Dermatol Res ; 302(4): 307-10, 2010 May.
Article in English | MEDLINE | ID: mdl-20033817

ABSTRACT

Hypohidrotic ectodermal dysplasia (HED) is a genetic disorder characterised by sparse hair, lack of sweat glands and malformation of teeth. The X-linked form of the disease, caused by mutations in the EDA gene, represents the majority of HED cases. Autosomal forms result from mutations in either the EDAR or the EDARADD gene. The X-linked and autosomal forms are phenotypically indistinguishable. For the purpose of genetic counselling, it is, therefore, important to know which gene is involved. In this study, we ascertained a Spanish family demonstrating the autosomal recessive form of HED. Affected individuals in the family showed the characteristic features of HED, including fine and sparse scalp hair, sparse eyebrows and eyelashes, periorbital hyperpigmentation, prominent lips, hypodontia and conical teeth, reduced sweating, and dry and thin skin. Sequence analysis of the EDAR gene revealed a novel compound heterozygous mutation [c.52-2A>G; c.212G>A (p.Cys71Tyr)]. Our finding extends the body of evidence that supports the significance of the EDAR signalling pathway in the ectodermal morphogenesis.


Subject(s)
DNA Mutational Analysis , Ectodermal Dysplasia, Hypohidrotic, Autosomal Recessive/genetics , Edar Receptor/genetics , Adult , Anodontia , Ectodermal Dysplasia, Hypohidrotic, Autosomal Recessive/diagnosis , Ectodermal Dysplasia, Hypohidrotic, Autosomal Recessive/physiopathology , Ectodysplasins/genetics , Ectodysplasins/metabolism , Edar Receptor/metabolism , Edar-Associated Death Domain Protein/genetics , Edar-Associated Death Domain Protein/metabolism , Family , Female , Heterozygote , Humans , Hyperpigmentation , Male , Mutation/genetics , Pedigree , Spain
16.
Am J Med Genet A ; 149A(9): 2031-6, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19681132

ABSTRACT

Hypohidrotic (anhidrotic) ectodermal dysplasia (HED) is a congenital syndrome characterized by sparse hair, oligodontia, and reduced sweating. It is caused by mutations in any of the three Eda pathway genes: ectodysplasin (Eda), Edar, and Edaradd which encode a ligand, a receptor, and an intracellular signal mediator of a single linear pathway, respectively. In rare cases, HED is associated with immune deficiency caused by mutations in further downstream components of the Eda pathway that are necessary for the activation of the transcription factor NF-kappaB. Here I present a brief research update on the molecular aspects of this evolutionarily conserved pathway. The developmental role of Eda will be discussed in light of loss- and gain-of-function mouse models with emphasis on the past few years.


Subject(s)
Ectodermal Dysplasia , Ectodysplasins/genetics , Edar Receptor/genetics , Edar-Associated Death Domain Protein/genetics , Gene Expression Regulation , Hypohidrosis , NF-kappa B/genetics , Animals , Disease Models, Animal , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/pathology , Ectodermal Dysplasia, Hypohidrotic, Autosomal Recessive , Ectodysplasins/metabolism , Edar Receptor/metabolism , Edar-Associated Death Domain Protein/metabolism , Humans , Hypohidrosis/genetics , Hypohidrosis/pathology , Mice , Mutation , NF-kappa B/metabolism
17.
J Invest Dermatol ; 129(10): 2358-64, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19387480

ABSTRACT

The hair follicles (HFs) and the interfollicular epidermis (IFE) of intact mature skin are maintained by distinct stem cell populations. Upon wounding, however, emigration of HF keratinocytes to the IFE plays a role in acute stages of healing. In addition to this repair function, rapidly cycling cells of the upper HF have been observed transiting to the IFE in neonatal skin. Here we report that an absence of HF development leads to shortening and kinking of the mouse tail. These skeletal defects are reduced by stimulating keratinocyte proliferation, suggesting that they arise from impaired epidermal expansion. We confirm that rapidly cycling cells of the HF emigrate to the IFE of the neonatal tail. These results suggest that an absence of HFs results in impaired skin growth that is unable to keep pace with the rapidly elongating axial skeleton of the tail. Thus, in addition to their role in wound repair, HFs can make a significant contribution to lateral expansion of the IFE in the absence of trauma.


Subject(s)
Cell Movement/physiology , Cell Proliferation , Hair Follicle/physiology , Keratinocytes/cytology , Skin/cytology , Skin/growth & development , Animals , Animals, Newborn , Bone and Bones/pathology , Edar-Associated Death Domain Protein/genetics , Edar-Associated Death Domain Protein/metabolism , Keratinocytes/metabolism , Mice , Mice, Mutant Strains , Mutation/genetics , Skin/embryology , Tail/pathology , Wound Healing/physiology
18.
Dev Biol ; 305(1): 232-45, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17362907

ABSTRACT

A subgroup of the TNF receptor family, composed of Edar, Troy and Xedar, are implicated in the development of ectodermal appendages, such as hair follicles, teeth and sweat glands. We have isolated chicken orthologues of these three receptors and analysed their roles in early feather development. Conservation of protein sequences between mammalian and avian proteins is variable, with avian Edar showing the greatest degree of sequence identity. cXedar differs from its mammalian orthologue in that it contains an intracellular death domain. All three receptors are expressed during early feather morphogenesis and dominant negative forms of each receptor impair the epithelial contribution to feather bud morphogenesis, while the dermal contribution appears unaffected. Hyperactivation of each receptor leads to more widespread assumption of placode fate, though in different regions of the skin. Receptor signaling converges on NF-kappaB, and inhibiting this transcription factor alters feather bud number and size in a stage-specific manner. Our findings illustrate the roles of these three receptors during avian skin morphogenesis and also suggest that activators of feather placode fate undergo mutual regulation to reach a decision on skin appendage location and size.


Subject(s)
Chickens , Edar-Associated Death Domain Protein/genetics , Edar-Associated Death Domain Protein/metabolism , Feathers/growth & development , Morphogenesis/genetics , Signal Transduction/physiology , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Conserved Sequence/genetics , In Situ Hybridization , Molecular Sequence Data , NF-kappa B/metabolism , Oligonucleotides/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Signal Transduction/genetics , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...