Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Trends Cell Biol ; 34(5): 360-362, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38461099

RESUMO

Mutations and polymorphisms in A20/TNFAIP3 have been linked to various inflammatory disorders. However, in addition to its well-known role in inflammation, A20 also controls EDAR- and receptor activator of NF-κB (RANK)-induced NF-κB signaling, regulating the development of epidermal skin appendages and bone, respectively. Furthermore, A20 regulates synapse remodeling through a mechanism dependent on NF-κB.


Assuntos
NF-kappa B , Transdução de Sinais , Proteína 3 Induzida por Fator de Necrose Tumoral alfa , Humanos , Animais , Proteína 3 Induzida por Fator de Necrose Tumoral alfa/metabolismo , NF-kappa B/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Receptor Edar/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo
2.
J Dermatol ; 50(10): 1357-1362, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37269152

RESUMO

Pathogenic variants in the EDARADD gene result in autosomal recessive and autosomal dominant ectodermal dysplasia. This article reports on the fourth family in the world with ectodermal dysplasia 11A (ECTD11A) cause from a novel splicing variant in the EDARADD gene, identified by whole exome sequencing and confirmed by Sanger sequencing. The proband and his mother were heterozygous for the detected variant (NM_145861.4:c.161-2A>T). The proband manifests unusual symptoms including hyperkeratotic plaques, slow-growing hair, recurrent infection, and pectus excavatum. His mother presents hypohidrosis, extensive tooth decay, fragile nails, and sparse hair. Further studies on ECTD11A patients could be useful to characterizing the phenotype features more precisely.


Assuntos
Displasia Ectodérmica , Receptor Edar , Feminino , Humanos , Receptor Edar/genética , Receptor Edar/metabolismo , Linhagem , Displasia Ectodérmica/diagnóstico , Displasia Ectodérmica/genética , Fenótipo , Mães , Proteína de Domínio de Morte Associada a Edar/genética
3.
J Dermatol ; 50(3): 349-356, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36258277

RESUMO

Hypohidrotic ectodermal dysplasia is a rare condition characterized by hypohidrosis, hypodontia, and hypotrichosis. The disease can show X-linked recessive, autosomal dominant or autosomal recessive inheritance trait. Of these, the autosomal forms are caused by mutations in either EDAR or EDARADD. To date, the underlying pathomechanisms or genotype-phenotype correlations for autosomal forms have not completely been disclosed. In this study, we performed a series of in vitro studies for four missense mutations in the death domain of EDAR protein: p.R358Q, p.G382S, p.I388T, and p.T403M. The results revealed that p.R358Q- and p.T403M-mutant EDAR showed different expression patterns from wild-type EDAR in both western blots and immunostainings. NF-κB reporter assays demonstrated that all the mutant EDAR showed reduced activation of NF-κB, but the reduction by p.G382S- and p.I388T-mutant EDAR was moderate. Co-immunoprecipitation assays showed that p.R358Q- and p.T403M-mutant EDAR did not bind with EDARADD at all, whereas p.G382S- and p.I388T-mutant EDAR maintained the affinity to some extent. Furthermore, we demonstrated that all the mutant EDAR proteins analyzed aberrantly bound with TRAF6. Sum of the data suggest that the degree of loss-of-function is different among the mutant EDAR proteins, which may be associated with the severity of the disease.


Assuntos
Displasia Ectodérmica Anidrótica Tipo 1 , Displasia Ectodérmica , Humanos , Mutação de Sentido Incorreto , Displasia Ectodérmica Anidrótica Tipo 1/diagnóstico , Displasia Ectodérmica Anidrótica Tipo 1/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Receptor Edar/genética , Receptor Edar/metabolismo , Displasia Ectodérmica/genética , Mutação
4.
Mol Genet Genomic Med ; 9(1): e1555, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33205897

RESUMO

BACKGROUND: Hypohidrotic ectodermal dysplasia (HED) is the most common form of ectodermal dysplasia and is mainly associated with mutations in the EDA, EDAR, and EDARADD responsible for the development of ectodermal-derived structures. HED displays different modes of inheritance according to the gene that is involved, with X-linked EDA-related HED being the most frequent form of the disease. METHODS: Two families with tooth agenesis and manifestations of HED underwent clinical examination and EDA, EDAR, and EDARADD genetic analysis. The impact of the novel variant on the protein was evaluated through bioinformatics tools, whereas molecular modeling was used to predict the effect on the protein structure. RESULTS: A novel missense variant was identified in the EDAR (c.287T>C, p.Phe96Ser) of a female child proband and her mother, accounting for autosomal dominant HED. The genetic variant c.866G>A (p.Arg289His) in EDA, which has been previously described, was observed in the male proband of another family confirming its role in X-linked HED. The inheritance model of the missense mutation showed a different relationship with X-linked HED and non-syndromic tooth agenesis. CONCLUSION: Our findings provide evidence of variable expression of HED in heterozygous females, which should be considered for genetic counseling, and different modes of inheritance related to tooth development.


Assuntos
Anodontia/genética , Displasia Ectodérmica/genética , Ectodisplasinas/genética , Receptor Edar/genética , Adulto , Anodontia/patologia , Pré-Escolar , Displasia Ectodérmica/patologia , Ectodisplasinas/química , Ectodisplasinas/metabolismo , Receptor Edar/química , Receptor Edar/metabolismo , Feminino , Genes Dominantes , Humanos , Masculino , Mutação de Sentido Incorreto , Linhagem , Ligação Proteica , Domínios Proteicos , Estabilidade Proteica , Síndrome
5.
Methods Mol Biol ; 2248: 167-183, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33185875

RESUMO

Genetic deficiency of ectodysplasin A (EDA) causes X-linked hypohidrotic ectodermal dysplasia, a congenital condition characterized by the absence or abnormal formation of sweat glands, teeth, and several skin appendages. Stimulation of the EDA receptor (EDAR) with agonists in the form of recombinant EDA or anti-EDAR antibodies can compensate for the absence of Eda in a mouse model of Eda deficiency, provided that agonists are administered in a timely manner during fetal development. Here we provide detailed protocols for the administration of EDAR agonists or antagonists, or other proteins, by the intravenous, intraperitoneal, and intra-amniotic routes as well as protocols to collect blood, to visualize sweat gland function, and to prepare skulls in mice.


Assuntos
Receptor Edar/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Vias de Administração de Medicamentos , Displasia Ectodérmica/tratamento farmacológico , Displasia Ectodérmica/genética , Displasia Ectodérmica/metabolismo , Receptor Edar/genética , Camundongos , Fenótipo , Proteínas Recombinantes/administração & dosagem , Resultado do Tratamento
6.
Gene Expr Patterns ; 38: 119131, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32755633

RESUMO

Development of the scleral ossicles, a ring of bony elements within the sclera, is directed by a series of papillae that arise from placodes in the conjunctival epithelium over a 1.5-day induction period in the chicken embryo. The regular spacing of the papillae around the corneal-scleral limbus suggests that their induction may be regulated by a reaction-diffusion mechanism, similar to other epithelial appendages. Some key placode signalling molecules, including ß-catenin, are known to be expressed throughout the induction period. However, others have been studied only at certain stages or have not been successfully detected. Here we use qPCR to study the gene expression patterns of the wingless integration (WNT)/ß-catenin, bone morphogenetic protein (BMP), ectodysplasin (EDA), fibroblast growth factor (FGF) and hedgehog (HH) signalling families in discrete regions of the eye throughout the complete conjunctival placode and papillae induction period. This comprehensive analysis revealed a variable level of gene expression within specific eye regions, with some genes exhibiting high, moderate or low changes. Most genes exhibited an initial increase in gene expression, followed by a decrease or plateau as development proceeded, suggesting that some genes are important for a brief initial period whilst the sustained elevated expression level of other genes is needed for developmental progression. The timing or magnitude of these changes, and/or the overall gene expression trend differed in the temporal, nasal and/or dorsal eye regions for some, but not all genes, demonstrating that gene expression may vary across different eye regions. Temporal and nasal EDA receptor (EDAR) had the greatest number of strong correlations (r > 0.700) with other genes and ß-catenin had the greatest number of moderate correlations (r = 0.400-0.700), while EDA had the greatest range in correlation strengths. Among the strongly correlated genes, two distinct signalling modules were identified, connected by some intermediate genes. The dynamic gene expression patterns of the five signalling pathways studied here from conjunctival placode formation through to papillae development is consistent with other epithelial appendages and confirms the presence of a conserved induction and patterning signalling network. Two unique gene expression patterns and corresponding gene interaction modules suggest functionally distinct roles throughout placode development. Furthermore, spatial differences in gene expression patterns among the temporal, nasal and dorsal regions of the eye may indicate that the expression of certain genes is influenced by mechanical forces exerted throughout development. Therefore, this study identifies key placode signalling factors and their interactions, as well as some potential region-specific features of gene expression in the scleral ossicle system and provides a basis for further exploration of the spatial expression of these genes and the patterning mechanism(s) active throughout conjunctival placode and papillae formation.


Assuntos
Proteínas Aviárias/genética , Regulação da Expressão Gênica no Desenvolvimento , Esclera/metabolismo , Via de Sinalização Wnt , Animais , Proteínas Aviárias/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Embrião de Galinha , Túnica Conjuntiva/embriologia , Túnica Conjuntiva/metabolismo , Córnea/embriologia , Córnea/metabolismo , Receptor Edar/genética , Receptor Edar/metabolismo , Esclera/embriologia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
7.
Eur J Hum Genet ; 28(12): 1694-1702, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32499598

RESUMO

Ectodysplasin A1 receptor (EDAR) is a TNF receptor family member with roles in the development and growth of hair, teeth and glands. A derived allele of EDAR, single-nucleotide variant rs3827760, encodes EDAR:p.(Val370Ala), a receptor with more potent signalling effects than the ancestral EDAR370Val. This allele of rs3827760 is at very high frequency in modern East Asian and Native American populations as a result of ancient positive selection and has been associated with straighter, thicker hair fibres, alteration of tooth and ear shape, reduced chin protrusion and increased fingertip sweat gland density. Here we report the characterisation of another SNV in EDAR, rs146567337, encoding EDAR:p.(Ser380Arg). The derived allele of this SNV is at its highest global frequency, of up to 5%, in populations of southern China, Vietnam, the Philippines, Malaysia and Indonesia. Using haplotype analyses, we find that the rs3827760 and rs146567337 SNVs arose on distinct haplotypes and that rs146567337 does not show the same signs of positive selection as rs3827760. From functional studies in cultured cells, we find that EDAR:p.(Ser380Arg) displays increased EDAR signalling output, at a similar level to that of EDAR:p.(Val370Ala). The existence of a second SNV with partly overlapping geographic distribution, the same in vitro functional effect and similar evolutionary age as the derived allele of rs3827760, but of independent origin and not exhibiting the same signs of strong selection, suggests a northern focus of positive selection on EDAR function in East Asia.


Assuntos
Receptor Edar/genética , Mutação com Ganho de Função , Frequência do Gene , Sudeste Asiático , Receptor Edar/química , Receptor Edar/metabolismo , Evolução Molecular , Células HEK293 , Células HaCaT , Haplótipos , Humanos , Simulação de Dinâmica Molecular , Polimorfismo de Nucleotídeo Único , Seleção Genética
8.
Int J Mol Sci ; 20(21)2019 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-31652981

RESUMO

The dental abnormalities are the typical features of many ectodermal dysplasias along with congenital malformations of nails, skin, hair, and sweat glands. However, several reports of non-syndromic/isolated tooth agenesis have also been found in the literature. The characteristic features of hypohidrotic ectodermal dysplasia (HED) comprise of hypodontia/oligodontia, along with hypohidrosis/anhidrosis, and hypotrichosis. Pathogenic variants in EDA, EDAR, EDARADD, and TRAF6, cause the phenotypic expression of HED. Genetic alterations in EDA and WNT10A cause particularly non-syndromic/isolated oligodontia. In the current project, we recruited 57 patients of 17 genetic pedigrees (A-Q) from different geographic regions of the world, including Pakistan, Egypt, Saudi Arabia, and Syria. The molecular investigation of different syndromic and non-syndromic dental conditions, including hypodontia, oligodontia, generalized odontodysplasia, and dental crowding was carried out by using exome and Sanger sequencing. We have identified a novel missense variant (c.311G>A; p.Arg104His) in WNT10A in three oligodontia patients of family A, two novel sequence variants (c.207delinsTT, p.Gly70Trpfs*25 and c.1300T>G; p.Try434Gly) in EDAR in three patients of family B and four patients of family C, respectively. To better understand the structural and functional consequences of missense variants in WNT10A and EDAR on the stability of the proteins, we have performed extensive molecular dynamic (MD) simulations. We have also identified three previously reported pathogenic variants (c.1076T>C; p.Met359Thr), (c.1133C>T; p.Thr378Met) and (c.594_595insC; Gly201Argfs*39) in EDA in family D (four patients), E (two patients) and F (one patient), correspondingly. Presently, our data explain the genetic cause of 18 syndromic and non-syndromic tooth agenesis patients in six autosomal recessive and X-linked pedigrees (A-F), which expand the mutational spectrum of these unique clinical manifestations.


Assuntos
Displasia Ectodérmica Anidrótica Tipo 1/patologia , Ectodisplasinas/genética , Receptor Edar/genética , Simulação de Dinâmica Molecular , Proteínas Wnt/genética , Displasia Ectodérmica Anidrótica Tipo 1/genética , Ectodisplasinas/química , Ectodisplasinas/metabolismo , Receptor Edar/química , Receptor Edar/metabolismo , Humanos , Mutação de Sentido Incorreto , Linhagem , Fenótipo , Estabilidade Proteica , Estrutura Terciária de Proteína , Sequenciamento do Exoma , Proteínas Wnt/química , Proteínas Wnt/metabolismo
9.
J Dermatol ; 46(8): 710-715, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31245878

RESUMO

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.


Assuntos
Displasia Ectodérmica Anidrótica Tipo 1/genética , Receptor Edar/genética , Proteína de Domínio de Morte Associada a Edar/metabolismo , Genes Dominantes/genética , Displasia Ectodérmica Anidrótica Tipo 1/patologia , Receptor Edar/metabolismo , Células HEK293 , Humanos , Mutação
10.
PLoS Biol ; 17(2): e3000132, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30789897

RESUMO

Feathers are arranged in a precise pattern in avian skin. They first arise during development in a row along the dorsal midline, with rows of new feather buds added sequentially in a spreading wave. We show that the patterning of feathers relies on coupled fibroblast growth factor (FGF) and bone morphogenetic protein (BMP) signalling together with mesenchymal cell movement, acting in a coordinated reaction-diffusion-taxis system. This periodic patterning system is partly mechanochemical, with mechanical-chemical integration occurring through a positive feedback loop centred on FGF20, which induces cell aggregation, mechanically compressing the epidermis to rapidly intensify FGF20 expression. The travelling wave of feather formation is imposed by expanding expression of Ectodysplasin A (EDA), which initiates the expression of FGF20. The EDA wave spreads across a mesenchymal cell density gradient, triggering pattern formation by lowering the threshold of mesenchymal cells required to begin to form a feather bud. These waves, and the precise arrangement of feather primordia, are lost in the flightless emu and ostrich, though via different developmental routes. The ostrich retains the tract arrangement characteristic of birds in general but lays down feather primordia without a wave, akin to the process of hair follicle formation in mammalian embryos. The embryonic emu skin lacks sufficient cells to enact feather formation, causing failure of tract formation, and instead the entire skin gains feather primordia through a later process. This work shows that a reaction-diffusion-taxis system, integrated with mechanical processes, generates the feather array. In flighted birds, the key role of the EDA/Ectodysplasin A receptor (EDAR) pathway in vertebrate skin patterning has been recast to activate this process in a quasi-1-dimensional manner, imposing highly ordered pattern formation.


Assuntos
Padronização Corporal , Plumas/citologia , Plumas/embriologia , Transdução de Sinais , Animais , Fenômenos Biomecânicos , Aves/embriologia , Agregação Celular , Contagem de Células , Movimento Celular , Forma Celular , Ectodisplasinas/metabolismo , Receptor Edar/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Voo Animal/fisiologia , Mesoderma/citologia , Mesoderma/embriologia , Pele/citologia , Pele/embriologia , beta Catenina/metabolismo
11.
PLoS Biol ; 17(2): e3000064, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30730874

RESUMO

When patterns are set during embryogenesis, it is expected that they are straightly established rather than subsequently modified. The patterning of the three mouse molars is, however, far from straight, likely as a result of mouse evolutionary history. The first-formed tooth signaling centers, called MS and R2, disappear before driving tooth formation and are thought to be vestiges of the premolars found in mouse ancestors. Moreover, the mature signaling center of the first molar (M1) is formed from the fusion of two signaling centers (R2 and early M1). Here, we report that broad activation of Edar expression precedes its spatial restriction to tooth signaling centers. This reveals a hidden two-step patterning process for tooth signaling centers, which was modeled with a single activator-inhibitor pair subject to reaction-diffusion (RD). The study of Edar expression also unveiled successive phases of signaling center formation, erasing, recovering, and fusion. Our model, in which R2 signaling center is not intrinsically defective but erased by the broad activation preceding M1 signaling center formation, predicted the surprising rescue of R2 in Edar mutant mice, where activation is reduced. The importance of this R2-M1 interaction was confirmed by ex vivo cultures showing that R2 is capable of forming a tooth. Finally, by introducing chemotaxis as a secondary process to RD, we recapitulated in silico different conditions in which R2 and M1 centers fuse or not. In conclusion, pattern formation in the mouse molar field relies on basic mechanisms whose dynamics produce embryonic patterns that are plastic objects rather than fixed end points.


Assuntos
Padronização Corporal , Receptor Edar/metabolismo , Modelos Biológicos , Transdução de Sinais , Dente/embriologia , Dente/metabolismo , Animais , Quimiotaxia , Receptor Edar/genética , Epitélio/embriologia , Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Cabelo/embriologia , Camundongos , Camundongos Mutantes , Germe de Dente/embriologia , Germe de Dente/metabolismo
12.
Cell Death Differ ; 26(3): 443-454, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-29855541

RESUMO

Ectodysplasin receptor EDAR is seen as a typical Tumor Necrosis Factor receptor (TNFR) family member known to interact with its ligand Eda-A1, and signaling mainly through the nuclear factor-kappaB (NF-κB) and c-jun N-terminal kinases pathways. Mutations in genes that encode proteins involved in EDAR transduction cascade cause anhidrotic ectodermal dysplasia. Here, we report an unexpected pro-apoptotic activity of EDAR when unbound to its ligand Eda-A1, which is independent of NF-κB pathway. Contrarily to other death receptors, EDAR does recruit caspase-8 to trigger apoptosis but solely upon ligand withdrawal, thereby behaving as the so-called dependence receptors. We propose that pro-apoptotic activity of unbound EDAR confers it a tumor suppressive activity. Along this line, we identified loss-of-pro-apoptotic function mutations in EDAR gene in human melanoma. Moreover, we show that the invalidation of EDAR in mice promotes melanoma progression in a B-Raf mutant background. Together, these data support the view that EDAR constrains melanoma progression by acting as a dependence receptor.


Assuntos
Receptor Edar/genética , Melanoma/genética , Animais , Morte Celular/genética , Linhagem Celular Tumoral , Ectodisplasinas/metabolismo , Receptor Edar/metabolismo , Feminino , Células HEK293 , Humanos , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Mutação
13.
Yi Chuan ; 40(11): 1024-1032, 2018 Nov 20.
Artigo em Chinês | MEDLINE | ID: mdl-30465535

RESUMO

The ectodysplasinA receptor gene (EDAR) plays an important role in the development of ectoderm. The derived G allele of its key missense variant EDARV370A is prevalent in East Asians and Americans, but rare in Africans and Europeans. This leads to distinct ectodermal-derived phenotypes between different continental groups, such as the straighter and thicker hair, more eccrine sweat glands, feminine smaller breasts, shovel incisors characteristic of East Asians. At present, we know little about the association between EDARV370A and facial and ear morphology characteristics. To better understand the effect of EDARV370A on craniofacial phenotypes, we systematically examined the association between EDARV370A and 136 facial quantitative phenotypes, one chin ordinal phenotype and six ear ordinal phenotypes in 715 Uyghurs. The quantitative phenotypes were derived by applying our automated landmark annotation method to facial 3D photos and the ordinal phenotypes were manually graded from facial 2D photos. The analysis identified significant association (P<0.05 after multiple testing correction) between EDARV370A and eight facial phenotypes, one chin phenotype and three ear morphology phenotypes. Our study thus elucidated the pleotropic effect of EDARV370A on craniofacial phenotypes in a European-Asian admixed Uyghur population.


Assuntos
Povo Asiático/genética , Orelha/anatomia & histologia , Receptor Edar/genética , Face/anatomia & histologia , Mutação de Sentido Incorreto , Adolescente , Adulto , Alelos , Povo Asiático/etnologia , China/etnologia , Orelha/crescimento & desenvolvimento , Receptor Edar/metabolismo , Feminino , Humanos , Masculino , Desenvolvimento Maxilofacial , Pessoa de Meia-Idade , Fenótipo , Adulto Jovem
14.
Proc Natl Acad Sci U S A ; 115(32): 8173-8178, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30037996

RESUMO

Ectodysplasin A (Eda) signaling activates NF-κB during skin appendage formation, but how Eda controls specific gene transcription remains unclear. Here, we find that Eda triggers the formation of an NF-κB-associated SWI/SNF (BAF) complex in which p50/RelB recruits a linker protein, Tfg, that interacts with BAF45d in the BAF complex. We further reveal that Tfg is initially induced by Eda-mediated RelB activation and then bridges RelB and BAF for subsequent gene regulation. The BAF component BAF250a is particularly up-regulated in skin appendages, and epidermal knockout of BAF250a impairs skin appendage development, resulting in phenotypes similar to those of Eda-deficient mouse models. Transcription profiling identifies several target genes regulated by Eda, RelB, and BAF. Notably, RelB and the BAF complex are indispensable for transcription of Eda target genes, and both BAF complex and Eda signaling are required to open chromatin of Eda targets. Our studies thus suggest that Eda initiates a signaling cascade and recruits a BAF complex to specific gene loci to facilitate transcription during organogenesis.


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Ectodisplasinas/metabolismo , Organogênese/genética , Pele/embriologia , Fator de Transcrição RelB/genética , Fatores de Transcrição/fisiologia , Transcrição Gênica/fisiologia , Animais , Cromatina/metabolismo , Ectodisplasinas/genética , Receptor Edar/genética , Receptor Edar/metabolismo , Feminino , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Proteínas/genética , Proteínas/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/fisiologia , Fator de Transcrição RelB/metabolismo , Ativação Transcricional/fisiologia , Regulação para Cima
15.
Proc Natl Acad Sci U S A ; 115(19): E4426-E4432, 2018 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-29686092

RESUMO

Because of the ubiquitous adaptability of our material culture, some human populations have occupied extreme environments that intensified selection on existing genomic variation. By 32,000 years ago, people were living in Arctic Beringia, and during the Last Glacial Maximum (LGM; 28,000-18,000 y ago), they likely persisted in the Beringian refugium. Such high latitudes provide only very low levels of UV radiation, and can thereby lead to dangerously low levels of biosynthesized vitamin D. The physiological effects of vitamin D deficiency range from reduced dietary absorption of calcium to a compromised immune system and modified adipose tissue function. The ectodysplasin A receptor (EDAR) gene has a range of pleiotropic effects, including sweat gland density, incisor shoveling, and mammary gland ductal branching. The frequency of the human-specific EDAR V370A allele appears to be uniquely elevated in North and East Asian and New World populations due to a bout of positive selection likely to have occurred circa 20,000 y ago. The dental pleiotropic effects of this allele suggest an even higher occurrence among indigenous people in the Western Hemisphere before European colonization. We hypothesize that selection on EDAR V370A occurred in the Beringian refugium because it increases mammary ductal branching, and thereby may amplify the transfer of critical nutrients in vitamin D-deficient conditions to infants via mothers' milk. This hypothesized selective context for EDAR V370A was likely intertwined with selection on the fatty acid desaturase (FADS) gene cluster because it is known to modulate lipid profiles transmitted to milk from a vitamin D-rich diet high in omega-3 fatty acids.


Assuntos
Clima Frio , Receptor Edar , Ácidos Graxos/metabolismo , Troca Materno-Fetal/fisiologia , Leite Humano/metabolismo , Seleção Genética/fisiologia , Vitamina D/metabolismo , Alelos , Receptor Edar/genética , Receptor Edar/metabolismo , Feminino , Humanos , Masculino , Glândulas Mamárias Humanas/anatomia & histologia , Glândulas Mamárias Humanas/metabolismo , Gravidez
16.
J Cell Biol ; 214(6): 753-67, 2016 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-27621364

RESUMO

During organogenesis, cell fate specification and patterning are regulated by signaling centers, specialized clusters of morphogen-expressing cells. In many organs, initiation of development is marked by bud formation, but the cellular mechanisms involved are ill defined. Here, we use the mouse incisor tooth as a model to study budding morphogenesis. We show that a group of nonproliferative epithelial cells emerges in the early tooth primordium and identify these cells as a signaling center. Confocal live imaging of tissue explants revealed that although these cells reorganize dynamically, they do not reenter the cell cycle or contribute to the growing tooth bud. Instead, budding is driven by proliferation of the neighboring cells. We demonstrate that the activity of the ectodysplasin/Edar/nuclear factor κB pathway is restricted to the signaling center, and its inactivation leads to fewer quiescent cells and a smaller bud. These data functionally link the signaling center size to organ size and imply that the early signaling center is a prerequisite for budding morphogenesis.


Assuntos
Movimento Celular , Proliferação de Células , Células Epiteliais/fisiologia , Incisivo/embriologia , Animais , Ectodisplasinas/genética , Ectodisplasinas/metabolismo , Receptor Edar/genética , Receptor Edar/metabolismo , Células Epiteliais/metabolismo , Fase G1 , Regulação da Expressão Gênica no Desenvolvimento , Genótipo , Idade Gestacional , Incisivo/metabolismo , Camundongos Transgênicos , Microscopia Confocal , Morfogênese , NF-kappa B/genética , NF-kappa B/metabolismo , Tamanho do Órgão , Fenótipo , Transdução de Sinais
18.
Clin Exp Immunol ; 184(2): 183-96, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26724675

RESUMO

Sjögren's syndrome (SS) is an autoimmune disease and the second most common chronic systemic rheumatic disorder. Prevalence of primary SS in the general population has been estimated to be approximately 1-3%, whereas secondary SS has been observed in 10-20% of patients with rheumatoid arthritis, systemic lupus erythematosus (SLE) and scleroderma. Despite this, its exact aetiology and pathogenesis are largely unexplored. Nuclear factor-kappa B (NF-κB) signalling mechanisms provide central controls in SS, but how these pathways intersect the pathological features of this disease is unclear. The ubiquitin-editing enzyme A20 (tumour necrosis factor-α-induced protein 3, TNFAIP3) serves as a critical inhibitor on NF-κB signalling. In humans, polymorphisms in the A20 gene or a deregulated expression of A20 are often associated with several inflammatory disorders, including SS. Because A20 controls the ectodysplasin-A1 (EDA-A1)/ectodysplasin receptor (EDAR) signalling negatively, and the deletion of A20 results in excessive EDA1-induced NF-κB signalling, this work investigates the expression levels of EDA-A1 and EDAR in SS human salivary glands epithelial cells (SGEC) and evaluates the hypothesis that SS SGEC-specific deregulation of A20 results in excessive EDA1-induced NF-κB signalling in SS. Our approach, which combines the use of siRNA-mediated gene silencing and quantitative pathway analysis, was used to elucidate the role of the A20 target gene in intracellular EDA-A1/EDAR/NF-κB pathway in SS SGEC, holding significant promise for compound selection in drug discovery.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Ectodisplasinas/metabolismo , Receptor Edar/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , NF-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Síndrome de Sjogren/patologia , Proteínas de Ligação a DNA/genética , Células Epiteliais/metabolismo , Humanos , Proteínas I-kappa B/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Inibidor de NF-kappaB alfa , Proteínas Nucleares/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Glândulas Salivares/citologia , Glândulas Salivares/metabolismo , Transdução de Sinais , Proteína 3 Induzida por Fator de Necrose Tumoral alfa
19.
Am J Med Genet A ; 170A(1): 249-53, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26440664

RESUMO

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.


Assuntos
Displasia Ectodérmica Anidrótica Tipo 1/genética , Receptor Edar/genética , Proteína de Domínio de Morte Associada a Edar/genética , Mutação de Sentido Incorreto/genética , Neoplasias Ovarianas/genética , Teratoma/genética , Adolescente , Doenças Mamárias/genética , Receptor Edar/metabolismo , Proteína de Domínio de Morte Associada a Edar/metabolismo , Feminino , Cabelo/crescimento & desenvolvimento , Humanos , Masculino , NF-kappa B/metabolismo , Neoplasias Ovarianas/patologia , Linhagem , Transdução de Sinais/genética , Teratoma/patologia
20.
Histochem Cell Biol ; 143(5): 481-96, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25366125

RESUMO

Mice with skin and hair follicle (HF) defects are common models of human skin disorders. A mutant strain with the we/we wal/wal genotype develops alopecia. We found the hair shaft structure in the pelage of mutant mice to have significant defects. Although these mice lose their hair at 21 days, a label-retaining cell population persists in HFs until at least day 54. Depilation-induced anagen was accomplished in we/we wal/wal mutants but the resulting hair shafts were short and extremely deformed. Serious abnormalities in epidermis stratification and HF morphogenesis exist in we/we wal/wal homozygous E18.5 embryos. There were significantly fewer HF primordia in this mutant compared with wild type. We discovered specific structures, identified as invalid placodes, positive for ectodysplasin A1 receptor, nuclear ß-catenin, and LEF1, which failed to invaginate, produced a double basal-like layer of epidermal cells, and lacked cylindrical keratinocytes. Specification of dermal papillae (DP) was impaired, and the papillary dermis expressed alkaline phosphatase and LEF1. We also detected DP-like groups of intensively stained cells in the absence of visible signs of folliculogenesis in the epidermis. We showed differentiation disturbances in the mutant embryonic E18.5 epidermis and HFs: The cornified layer was absent, the width of the spinous layer was reduced, and HFs lacked LEF1-positive precortex cells. In this study, we used a very interesting and useful mouse model of alopecia. The presence of symptoms of skin disorders in we/we wal/wal murine embryos correlates with the postnatal skin phenotype. This correlation may help to evaluate reasons of alopecia.


Assuntos
Alopecia/patologia , Epiderme/anormalidades , Folículo Piloso/anormalidades , Fatores Etários , Alopecia/embriologia , Alopecia/genética , Alopecia/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Receptor Edar/metabolismo , Epiderme/metabolismo , Genótipo , Idade Gestacional , Folículo Piloso/metabolismo , Remoção de Cabelo , Queratinócitos/metabolismo , Queratinócitos/patologia , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Morfogênese , Fenótipo , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...