Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Lett ; 577: 216427, 2023 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-37838280

RESUMEN

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.


Asunto(s)
Neoplasias del Colon , Ubiquitina-Proteína Ligasas , Humanos , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Línea Celular Tumoral , Ubiquitinación , Neoplasias del Colon/genética , Transición Epitelial-Mesenquimal/genética , Proteína de Dominio de Muerte Asociada a Edar/genética , Proteína de Dominio de Muerte Asociada a Edar/metabolismo
2.
J Dermatol ; 50(10): 1357-1362, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37269152

RESUMEN

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.


Asunto(s)
Displasia Ectodérmica , Receptor Edar , Femenino , Humanos , Receptor Edar/genética , Receptor Edar/metabolismo , Linaje , Displasia Ectodérmica/diagnóstico , Displasia Ectodérmica/genética , Fenotipo , Madres , Proteína de Dominio de Muerte Asociada a Edar/genética
3.
Genes (Basel) ; 12(9)2021 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-34573371

RESUMEN

Ectodermal dysplasia (ED) is a diverse group of genetic disorders caused by congenital defects of two or more ectodermal-derived body structures, namely, hair, teeth, nails, and some glands, e.g., sweat glands. Molecular pathogenesis of ED involves mutations of genes encoding key proteins of major developmental pathways, including ectodysplasin (EDA) and wingless-type (WNT) pathways. The most common ED phenotype is hypohidrotic/anhidrotic ectodermal dysplasia (HED) featuring hypotrichosis, hypohidrosis/anhidrosis, and hypodontia. Molecular diagnosis is fundamental for disease management and emerging treatments. We used targeted next generation sequencing to study EDA, EDAR, EDARADD, and WNT10A genes in 45 Egyptian ED patients with or without hypohidrosis. We present genotype and phenotype data of 28 molecularly-characterized patients demonstrating genetic heterogeneity, variable expressivity, and intrafamilial phenotypic variability. Thirteen mutations were reported, including four novel EDA mutations, two novel EDARADD, and one novel EDAR mutations. Identified mutations congregated in exons encoding key functional domains. EDA is the most common gene contributing to 85% of the identified Egyptian ED genetic spectrum, followed by EDARADD (10%) and EDAR (5%). Our cohort represents the first and largest cohort from North Africa where more than 60% of ED patients were identified emphasizing the need for exome sequencing to explore unidentified cases.


Asunto(s)
Displasia Ectodérmica/genética , Ectodisplasinas/genética , Receptor Edar/genética , Proteína de Dominio de Muerte Asociada a Edar/genética , Mutación , Adulto , Niño , Preescolar , Displasia Ectodérmica/etiología , Egipto , Femenino , Heterocigoto , Humanos , Lactante , Masculino , Persona de Mediana Edad , Proteínas Wnt/genética
4.
J Dermatol ; 48(10): 1533-1541, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34219261

RESUMEN

Hypohidrotic ectodermal dysplasia (HED) is a genetic disorder characterized by hypohidrosis, hypodontia, and hypotrichosis. Autosomal forms of the disease are caused by mutations in either EDAR or EDARADD. To date, the underlying pathomechanisms for HED resulting from EDARADD mutations have not fully been disclosed. In this study, we performed detailed in vitro analyses in order to characterize three dominantly inherited missense mutations, p.D120Y, p.L122R, and p.D123N, and one recessively inherited missense mutation, p.E152K, in the EDARADD gene. Nuclear factor (NF)-κB reporter assays demonstrated that all the mutant EDARADD showed reduction in activation of NF-κB. Importantly, p.D120Y-, p.L122R-, and p.D123N-mutant EDARADD slightly reduced the NF-κB activity induced by wild-type EDARADD in a dominant negative manner. Co-immunoprecipitation assays showed that all of the mutant EDARADD were capable of binding to EDAR and wild-type EDARADD. Additional co-immunoprecipitation assays revealed that p.D120Y-, p.L122R-, and p.D123N-mutant EDARADD markedly prevented the interaction between EDAR and wild-type EDARADD, which further indicated a dominant negative effect by these mutations. Finally, we found that p.D120Y-, p.L122R-, and p.D123N-mutant EDARADD completely lost the ability to bind with TRAF6, while p.E152K-mutant EDARADD showed a mild reduction in the affinity. Our findings will provide crucial information toward unraveling the molecular mechanisms how EDARADD gene mutations cause the disease.


Asunto(s)
Anodoncia , Displasia Ectodermal Anhidrótica Tipo 1 , Proteína de Dominio de Muerte Asociada a Edar , Hipohidrosis , Deformidades Congénitas de las Extremidades , Ectodisplasinas , Proteína de Dominio de Muerte Asociada a Edar/genética , Humanos , Mutación
5.
Aging (Albany NY) ; 13(5): 6459-6484, 2021 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-33707346

RESUMEN

DNA methylation is known as a biomarker for age with applications in forensics. Here we describe the VISAGE (VISible Attributes through GEnomics) Consortium's enhanced tool for epigenetic age estimation in somatic tissues. The tool is based on eight DNA methylation markers (44 CpGs), bisulfite multiplex PCR followed by sequencing on the MiSeq FGx platform, and three statistical prediction models for blood, buccal cells and bones. The model for blood is based on six CpGs from ELOVL2, MIR29B2CHG, KLF14, FHL2, TRIM59 and PDE4C, and predicts age with a mean absolute error (MAE) of 3.2 years, while the model for buccal cells includes five CpGs from PDE4C, MIR29B2CHG, ELOVL2, KLF14 and EDARADD and predicts age with MAE of 3.7 years, and the model for bones has six CpGs from ELOVL2, KLF14, PDE4C and ASPA and predicts age with MAE of 3.4 years. The VISAGE enhanced tool for age estimation in somatic tissues enables reliable collection of DNA methylation data from small amounts of DNA using a sensitive multiplex MPS assay that provides accurate estimation of age in blood, buccal swabs, and bones using the statistical model tailored to each tissue.


Asunto(s)
Envejecimiento/genética , Islas de CpG , Modelos Estadísticos , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Amidohidrolasas/genética , Análisis Químico de la Sangre , Huesos/química , Niño , Preescolar , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Metilación de ADN , Proteína de Dominio de Muerte Asociada a Edar/genética , Epigénesis Genética , Elongasas de Ácidos Grasos/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Persona de Mediana Edad , Mucosa Bucal/química , Reacción en Cadena de la Polimerasa Multiplex , Análisis de Secuencia de ADN , Adulto Joven
6.
Eur J Dermatol ; 30(4): 408-416, 2020 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32819890

RESUMEN

BACKGROUND: Hypohidrotic ectodermal dysplasia (HED) is a congenital anomaly characterized by hypohydrosis, hypotrichosis and hypodontia. Mutations in at least four genes (EDAR, EDARADD, WNT10A, TRAF6) have been reported to cause both autosomal recessive and autosomal dominant forms of HED. Mutations in two other genes (EDA and IKBKG) have been reported to cause X-linked HED. OBJECTIVES: To clinically characterize three consanguineous families (A-C) segregating with autosomal recessive HED and identify possible disease-causing variants of EDAR and EDARADD genes. MATERIALS AND METHODS: The genes, EDAR and EDARADD, were sequenced in Family A and C, and exome sequencing was performed in Family B. Additionally, in Family A and C, the effect of the identified variants was examined by analysis of EDAR mRNA, extracted from hair follicles from both affected and unaffected members. RESULTS: Sequence analysis revealed three possible disease-causing EDAR variants including a novel splice acceptor site variant (IVS3-1G > A) in Family A and two previously reported mutations (p.[Ala26Val], p.[Arg25*]) in the two other families. Previously, the nonsense variant p.(Arg25*) was reported only in the heterozygous state. Analysis of the RNA, extracted from hair follicles, revealed skipping of a downstream exon in EDAR and complete degradation of EDAR mRNA in affected members in family A and C, respectively. Computational modelling validated the pathogenic effect of the two variants identified in Family B and C. CONCLUSION: The three variants reported here expand the spectrum of EDAR mutations associated with HED which may further facilitate genetic counselling of families segregating with similar disorders in the Pakistani population.


Asunto(s)
Consanguinidad , Displasia Ectodérmica/genética , Receptor Edar/genética , Proteína de Dominio de Muerte Asociada a Edar/genética , Adolescente , Niño , Codón sin Sentido , Displasia Ectodérmica/patología , Femenino , Genes Recesivos , Homocigoto , Humanos , Masculino , Mutación Missense , Pakistán , Linaje , Mutación Puntual , Sitios de Empalme de ARN/genética , Análisis de Secuencia de ARN , Adulto Joven
7.
Clin Epigenetics ; 12(1): 48, 2020 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-32188493

RESUMEN

BACKGROUND: Prostate cancer changes the phenotype of cells within the stromal microenvironment, including fibroblasts, which in turn promote tumour progression. Functional changes in prostate cancer-associated fibroblasts (CAFs) coincide with alterations in DNA methylation levels at loci-specific regulatory regions. Yet, it is not clear how these methylation changes compare across CAFs from different patients. Therefore, we examined the consistency and prognostic significance of genome-wide DNA methylation profiles between CAFs from patients with different grades of primary prostate cancer. RESULTS: We used Infinium MethylationEPIC BeadChips to evaluate genome-wide DNA methylation profiles from 18 matched CAFs and non-malignant prostate tissue fibroblasts (NPFs) from men with moderate to high grade prostate cancer, as well as five unmatched benign prostate tissue fibroblasts (BPFs) from men with benign prostatic hyperplasia. We identified two sets of differentially methylated regions (DMRs) in patient CAFs. One set of DMRs reproducibly differed between CAFs and fibroblasts from non-malignant tissue (NPFs and BPFs). Indeed, more than 1200 DMRs consistently changed in CAFs from every patient, regardless of tumour grade. The second set of DMRs varied between CAFs according to the severity of the tumour. Notably, hypomethylation of the EDARADD promoter occurred specifically in CAFs from high-grade tumours and correlated with increased transcript abundance and increased EDARADD staining in patient tissue. Across multiple cohorts, tumours with low EDARADD DNA methylation and high EDARADD mRNA expression were consistently associated with adverse clinical features and shorter recurrence free survival. CONCLUSIONS: We identified a large set of DMRs that are commonly shared across CAFs regardless of tumour grade and outcome, demonstrating highly consistent epigenome changes in the prostate tumour microenvironment. Additionally, we found that CAFs from aggressive prostate cancers have discrete methylation differences compared to CAFs from moderate risk prostate cancer. Together, our data demonstrates that the methylome of the tumour microenvironment reflects both the presence and the severity of the prostate cancer and, therefore, may provide diagnostic and prognostic potential.


Asunto(s)
Fibroblastos Asociados al Cáncer/patología , Metilación de ADN , Proteína de Dominio de Muerte Asociada a Edar/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Hiperplasia Prostática/patología , Neoplasias de la Próstata/patología , Anciano , Fibroblastos Asociados al Cáncer/química , Estudios de Casos y Controles , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Pronóstico , Regiones Promotoras Genéticas , Hiperplasia Prostática/genética , Neoplasias de la Próstata/genética , Análisis de Supervivencia , Células Tumorales Cultivadas , Microambiente Tumoral , Regulación hacia Arriba
8.
J Forensic Sci ; 65(2): 465-470, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31490551

RESUMEN

Age estimation using DNA methylation levels has been widely investigated in recent years because of its potential application in forensic genetics. The main aim of this study was to develop an age predictor model (APM) for blood samples of deceased individuals based in five age-correlated genes. Fifty-one samples were analyzed through the bisulfite polymerase chain reaction (PCR) sequencing method for DNA methylation evaluation in genes ELOVL2, FHL2, EDARADD, PDE4C, and C1orf132. Linear regression was used to analyze relationships between methylation levels and age. The model using the highest age-correlated CpG from each locus revealed a correlation coefficient of 0.888, explaining 76.3% of age variation, with a mean absolute deviation from the chronological age (MAD) of 6.08 years. The model was validated in an independent test set of 19 samples producing a MAD of 8.84 years. The developed APM seems to be informative and could have potential application in forensic analysis.


Asunto(s)
Envejecimiento/genética , Metilación de ADN , Análisis de Secuencia de ADN/métodos , Adulto , Anciano , Anciano de 80 o más Años , Islas de CpG/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Proteína de Dominio de Muerte Asociada a Edar/genética , Elongasas de Ácidos Grasos/genética , Femenino , Genética Forense/métodos , Marcadores Genéticos , Humanos , Proteínas con Homeodominio LIM/genética , Modelos Lineales , Masculino , Persona de Mediana Edad , Proteínas Musculares/genética , Reacción en Cadena de la Polimerasa/métodos , Sulfitos , Factores de Transcripción/genética , Adulto Joven
9.
Orphanet J Rare Dis ; 14(1): 281, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31796081

RESUMEN

BACKGROUND: Ectodermal dysplasias (ED) are a group of genetic conditions affecting the development and/or homeostasis of two or more ectodermal derivatives. An attenuated phenotype is considered a non-syndromic trait when the patient is affected by only one impaired ectodermal structure, such as in non-syndromic tooth agenesis (NSTA) disorder. Hypohidrotic ectodermal dysplasia (HED) is the most highly represented ED. X-linked hypohidrotic ectodermal dysplasia (XLHED) is the most common subtype, with an incidence of 1/50,000-100,000 males, and is associated with the EDA gene (Xq12-q13.1); the dominant and recessive subtypes involve the EDAR (2q13) and EDARADD (1q42.3) genes, respectively. The WNT10A gene (2q35) is associated more frequently with NSTA. Our goal was to determine the mutational spectrum in a cohort of 72 Spanish patients affected by one or more ectodermal derivative impairments referred to as HED (63/72) or NSTA (9 /72) to establish the prevalence of the allelic variants of the four most frequently associated genes. Sanger sequencing of the EDA, EDAR, EDARADD and WNT10A genes and multiplex ligation-dependent probe amplification (MLPA) were performed. RESULTS: A total of 61 children and 11 adults, comprising 50 males and 22 females, were included. The average ages were 5.4 and 40.2 years for children and adults, respectively. A molecular basis was identified in 51/72 patients, including 47/63 HED patients, for whom EDA was the most frequently involved gene, and 4/9 NSTA patients, most of whom had variants of WNT10A. Among all the patients, 37/51 had variants of EDA, 8/51 had variants of the WNT10A gene, 4/51 had variants of EDAR and 5/51 had variants of EDARADD. In 42/51 of cases, the variants were inherited according to an X-linked pattern (27/42), with the remaining showing an autosomal dominant (10/42) or autosomal recessive (5/42) pattern. Among the NSTA patients, 3/9 carried pathogenic variants of WNT10A and 1/9 carried EDA variants. A total of 60 variants were detected in 51 patients, 46 of which were different, and out of these 46 variants, 12 were novel. CONCLUSIONS: This is the only molecular study conducted to date in the Spanish population affected by ED. The EDA, EDAR, EDARADD and WNT10A genes constitute the molecular basis in 70.8% of patients with a 74.6% yield in HED and 44.4% in NSTA. Twelve novel variants were identified. The WNT10A gene has been confirmed as the second molecular candidate that has been identified and accounts for one-half of non-EDA patients and one-third of NSTA patients. Further studies using next generation sequencing (NGS) will help to identify other contributory genes in the remaining uncharacterized Spanish patients.


Asunto(s)
Displasia Ectodermal Anhidrótica Tipo 1/genética , Displasia Ectodérmica/genética , Receptor Edar/genética , Proteína de Dominio de Muerte Asociada a Edar/genética , Proteínas Wnt/genética , Adolescente , Adulto , Anodoncia/genética , Niño , Preescolar , Variaciones en el Número de Copia de ADN/genética , Exones/genética , Femenino , Humanos , Lactante , Recién Nacido , Intrones/genética , Masculino , Persona de Mediana Edad , España , Adulto Joven
10.
Cytogenet Genome Res ; 157(4): 189-196, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30974434

RESUMEN

Hypohidrotic or anhidrotic ectodermal dysplasia (HED/EDA) is characterized by impaired development of the hair, teeth, or sweat glands. HED/EDA is inherited in an X-linked, autosomal dominant, or autosomal recessive pattern and caused by the pathogenic variants in 4 genes: EDA, EDAR, EDARADD, and WNT10A. The aim of the present study was to perform molecular screening of these 4 genes in a cohort of Turkish individuals diagnosed with HED/EDA. We screened for pathogenic variants of WNT10A, EDA, EDAR, and EDARADD through Sanger sequencing. We further assessed the clinical profiles of the affected individuals in order to establish phenotype-genotype correlation. In 17 (63%) out of 27 families, 17 pathogenic variants, 8 being novel, were detected in the 4 well-known ectodermal dysplasia genes. EDAR and EDA variants were identified in 6 families each, WNT10A variants in 4, and an EDARADD variant in 1, accounting for 35.3, 35.3, 23.5, and 5.9% of mutation-positive families, respectively. The low mutation detection rate of the cohort and the number of the EDAR pathogenic variants being as high as the EDA ones were the most noteworthy findings which could be attributed to the high consanguinity rate.


Asunto(s)
Displasia Ectodérmica/genética , Ectodisplasinas/genética , Receptor Edar/genética , Proteína de Dominio de Muerte Asociada a Edar/genética , Mutación , Análisis de Secuencia de ADN/métodos , Proteínas Wnt/genética , Consanguinidad , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Linaje , Fenotipo , Turquía
11.
Oral Dis ; 25(3): 646-651, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29969831

RESUMEN

Tooth agenesis (TA) is one of the most common developmental anomalies that affects the number of teeth. An extensive analysis of publicly accessible databases revealed 15 causative genes responsible for nonsyndromic TA, along with their signaling pathways in Wnt/ß-catenin, TGF-ß/BMP, and Eda/Edar/NF-κB. However, genotype-phenotype correlation analysis showed that most of the causal genes are also responsible for syndromic TA or other conditions. In a total of 198 different mutations of the 15 genes responsible for nonsyndromic TA, 182 mutations (91.9%) are derived from seven genes (AXIN2, EDA, LRP6, MSX1, PAX9, WNT10A, and WNT10B) compared with the remaining 16 mutations (8.1%) identified in the remaining eight genes (BMP4, DKK1, EDAR, EDARADD, GREM2, KREMEN1, LTBP3, and SMOC2). Furthermore, specificity analysis in terms of the ratio of nonsyndromic TA mutations versus syndromic mutations in each of the aforementioned seven genes showed a 98.2% specificity rate in PAX9, 58.9% in WNT10A, 56.6% in MSX1, 41.2% in WNT10B, 31.4% in LRP6, 23.8% in AXIN2%, and 8.4% in EDA. These findings underscore an important role of the Wnt and Wnt-associated pathways in the genetic etiology of this heterozygous disease and shed new lights on the discovery of novel molecular mechanisms associated with tooth agenesis.


Asunto(s)
Anodoncia/genética , Proteína Morfogenética Ósea 4/genética , Ectodisplasinas/genética , Receptor Edar/genética , Factor de Crecimiento Transformador beta/genética , Vía de Señalización Wnt/genética , Animales , Proteína Axina/genética , Proteínas de Unión al Calcio/genética , Citocinas , Proteína de Dominio de Muerte Asociada a Edar/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de Unión a TGF-beta Latente/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Factor de Transcripción MSX1/genética , Proteínas de la Membrana/genética , Mutación , FN-kappa B/genética , Factor de Transcripción PAX9/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Wnt/genética
13.
Arch Oral Biol ; 97: 131-136, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30384154

RESUMEN

OBJECTIVE: Tooth agenesis (TA) is featured by congenital loss of teeth, and can be divided into two subtypes, non-syndromic TA (NSTA) and syndromic TA (STA). Although 12 candidate genes of NSTA have been revealed, the genetic basis of NSTA needs to be further studied. We noticed an overlap of candidate genes between NSTA and STA, and hypothesized that some candidate genes of STA may be new candidate genes of NSTA. METHODS: Sanger sequencing, whole exome sequencing, bioinformatics analyses and immunohistochemical staining were performed to reveal the genetic basis of the patients in a family with NSTA. RESULTS: No pathogenic mutation was found in the 12 candidate genes of NSTA. We screened the variants of 76 STA candidate genes and identified a novel pathogenic mutation c.G908C (p.R303 P) in Keratinocyte Differentiation Factor 1 (KDF1). This mutation was cosegregated with the disease in the family. Bioinformatics analyses predicted the mutation to be pathogenic. Immunohistochemical staining of kdf1 in developing tooth germs indicated that kdf1 expression is important for the development of teeth. CONCLUSIONS: This study identified KDF1 as a novel candidate gene for NSTA. STA candidate genes may be a promising source of new NSTA genes.


Asunto(s)
Anodoncia/genética , Proteínas/genética , Anodoncia/diagnóstico por imagen , Niño , Biología Computacional , Ectodisplasinas/genética , Receptor Edar/genética , Proteína de Dominio de Muerte Asociada a Edar/genética , Femenino , Humanos , Inmunohistoquímica , Masculino , Mutación , Linaje , Reacción en Cadena de la Polimerasa , Radiografía Panorámica , Proteínas Wnt/genética
14.
Int J Dermatol ; 57(8): 965-972, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29855039

RESUMEN

Hypohidrotic Ectodermal Dysplasia (HED) is a genetic human disorder which affects structures of ectodermal origin. Although there are autosomal recessive and dominant forms, X-linked (XL) is the most frequent form of the disease. This XL-HED phenotype is associated with mutations in the gene encoding the transmembrane protein ectodysplasin-1 (EDA1), a member of the TNFα-related signaling pathway. The proteins from this pathway are involved in signal transduction from ectoderm to mesenchyme leading to the development of ectoderm-derived structures in the fetus such as hair, teeth, skin, nails, and eccrine sweat glands. The aim of this review was to update the main clinical characteristics of HED regarding to recent molecular advances in the comprehension of all the possible genes involved in this group of disorders since it is known that Eda-A1-Edar signaling has multiple roles in ectodermal organ development, regulating their initiation, morphogenesis, and differentiation steps. The knowledge of the biological mechanisms that generate HED is needed for both a better detection of possible cases and for the design of efficient prevention and treatment approaches.


Asunto(s)
Displasia Ectodermal Anhidrótica Tipo 1/genética , Ectodisplasinas/genética , Receptor Edar/genética , Proteína de Dominio de Muerte Asociada a Edar/genética , Quinasa I-kappa B/genética , Anodoncia/etiología , Displasia Ectodermal Anhidrótica Tipo 1/complicaciones , Displasia Ectodermal Anhidrótica Tipo 1/diagnóstico , Displasia Ectodermal Anhidrótica Tipo 1/patología , Humanos , Hipohidrosis/etiología , Hipotricosis/etiología , Mutación , Transducción de Señal
15.
Forensic Sci Int Genet ; 36: 50-59, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29933125

RESUMEN

DNA methylation is the most extensively studied epigenetic signature, with a large number of studies reporting age-correlated CpG sites in overlapping genes. However, most of these studies lack sample coverage of individuals under 18 years old and therefore little is known about the progression of DNA methylation patterns in children and adolescents. In the present study we aimed to select candidate age-correlated DNA methylation markers based on public datasets from Illumina BeadChip arrays and previous publications, then to explore the resulting markers in 209 blood samples from donors aged between 2 to 18 years old using the EpiTYPER® DNA methylation analysis system. Results from our analyses identified six genes highly correlated with age in the young, in particular the gene KCNAB3, which indicates its potential as a highly informative and specific age biomarker for childhood and adolescence. We outline a preliminary age prediction model based on quantile regression that uses data from the six CpG sites most strongly correlated with age ranges extended to include children and adolescents.


Asunto(s)
Envejecimiento/genética , Metilación de ADN , Genética Forense/métodos , Marcadores Genéticos , Acetiltransferasas/genética , Adolescente , Amidohidrolasas/genética , Niño , Preescolar , Islas de CpG/genética , Proteína Quinasa Dependiente de GMP Cíclico Tipo II/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Proteína de Dominio de Muerte Asociada a Edar/genética , Elongasas de Ácidos Grasos , Humanos , Proteínas con Homeodominio LIM/genética , Proteínas Musculares/genética , Proteínas del Tejido Nervioso/genética , Reacción en Cadena de la Polimerasa , Canales de Potasio de la Superfamilia Shaker , Canales de Potasio Shaw/genética , Programas Informáticos , Factores de Transcripción/genética
16.
Arch Oral Biol ; 91: 96-102, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29705498

RESUMEN

OBJECTIVE: The aim of this study was the clinical and molecular characterization of a family segregating a trait consisting of a phenotype specifically involving the maxillary canines, including agenesis, impaction and ectopic eruption, characterized by incomplete penetrance and variable expressivity. DESIGN: Clinical standardized assessment of 14 family members and a whole-exome sequencing (WES) of three affected subjects were performed. WES data analyses (sequence alignment, variant calling, annotation and prioritization) were carried out using an in-house implemented pipeline. Variant filtering retained coding and splice-site high quality private and rare variants. Variant prioritization was performed taking into account both the disruptive impact and the biological relevance of individual variants and genes. Sanger sequencing was performed to validate the variants of interest and to carry out segregation analysis. RESULTS: Prioritization of variants "by function" allowed the identification of multiple variants contributing to the trait, including two concomitant heterozygous variants in EDARADD (c.308C>T, p.Ser103Phe) and COL5A1 (c.1588G>A, p.Gly530Ser), specifically associated with a more severe phenotype (i.e. canine agenesis). Differently, heterozygous variants in genes encoding proteins with a role in the WNT pathway were shared by subjects showing a phenotype of impacted/ectopic erupted canines. CONCLUSIONS: This study characterized the genetic contribution underlying a complex trait consisting of isolated canine anomalies in a medium-sized family, highlighting the role of WNT and EDA cell signaling pathways in tooth development.


Asunto(s)
Anodoncia/genética , Diente Canino/anomalías , Secuenciación del Exoma/métodos , Erupción Dental , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Proteínas de Unión al Calcio , Colágeno Tipo V/genética , Diente Canino/diagnóstico por imagen , Análisis Mutacional de ADN , Ectodisplasinas/metabolismo , Proteína de Dominio de Muerte Asociada a Edar/genética , Femenino , Proteínas Fetales/genética , Frecuencia de los Genes , Heterocigoto , Humanos , Italia , Masculino , Maxilar , Persona de Mediana Edad , Mutación , Proteínas del Tejido Nervioso/genética , Linaje , Fenotipo , Radiografía Panorámica , Alineación de Secuencia , Proteínas de Dominio T Box/genética , Trombospondinas/genética , Vía de Señalización Wnt , Adulto Joven
17.
Forensic Sci Rev ; 29(2): 121-144, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28691915

RESUMEN

Individual age estimation is a key factor in forensic science analysis that can provide very useful information applicable to criminal, legal, and anthropological investigations. Forensic age inference was initially based on morphological inspection or radiography and only later began to adopt molecular approaches. However, a lack of accuracy or technical problems hampered the introduction of these DNA-based methodologies in casework analysis. A turning point occurred when the epigenetic signature of DNA methylation was observed to gradually change during an individual´s lifespan. In the last four years, the number of publications reporting DNA methylation age-correlated changes has gradually risen and the forensic community now has a range of age methylation tests applicable to forensic casework. Most forensic age predictor models have been developed based on blood DNA samples, but additional tissues are now also being explored. This review assesses the most widely adopted genes harboring methylation sites, detection technologies, statistical age-predictive analyses, and potential causes of variation in age estimates. Despite the need for further work to improve predictive accuracy and establishing a broader range of tissues for which tests can analyze the most appropriate methylation sites, several forensic age predictors have now been reported that provide consistency in their prediction accuracies (predictive error of ±4 years); this makes them compelling tools with the potential to contribute key information to help guide criminal investigations.


Asunto(s)
Envejecimiento/genética , Metilación de ADN , Genética Forense/métodos , Acetiltransferasas/genética , Amidohidrolasas/genética , Islas de CpG/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/genética , Proteína de Dominio de Muerte Asociada a Edar/genética , Epigenómica , Elongasas de Ácidos Grasos , Humanos , Integrina alfa2/genética , Proteínas con Homeodominio LIM/genética , Proteínas Musculares/genética , Análisis de Secuencia de ADN , Sulfitos/química , Factores de Transcripción/genética
18.
Int J Mol Med ; 38(5): 1338-1348, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27665865

RESUMEN

Dental agenesis is one of the most common congenital craniofacial abnormalities. Dental agenesis can be classified, relative to the number of missing teeth (excluding third molars), as hypodontia (1 to 5 missing teeth), oligodontia (6 or more missing teeth), or anodontia (lack of all teeth). Tooth agenesis may occur either in association with genetic syndromes, based on the presence of other inherited abnormalities, or as a non-syndromic trait, with both familiar and sporadic cases reported. In this study, we enrolled 16 individuals affected by tooth agenesis, prevalently hypodontia, and we carried out direct Sanger sequencing of paired box 9 (PAX9) and Msh homeobox 1 (MSX1) genes in 9 subjects. Since no mutations were identified, we performed whole exome sequencing (WES) in the members of 5 families to identify causative gene mutations either novel or previously described. Three individuals carried a known homozygous disease mutation in the Wnt family member 10A (WNT10A) gene (rs121908120). Interestingly, two of these individuals were siblings and also carried a heterozygous functional variant in EDAR-associated death domain (EDARADD) (rs114632254), another disease causing gene, generating a combination of genetic variants never described until now. The analysis of exome sequencing data in the members of other 3 families highlighted new candidate genes potentially involved in tooth agenesis and considered suitable for future studies. Overall, our study confirmed the major role played by WNT10A in tooth agenesis and the genetic heterogeneity of this disease. Moreover, as more genes are shown to be involved in tooth agenesis, WES analysis may be an effective approach to search for genetic variants in familiar or sporadic tooth agenesis, at least in more severe clinical manifestations.


Asunto(s)
Anodoncia/genética , Análisis Mutacional de ADN/métodos , Exoma/genética , Mutación , Adolescente , Adulto , Secuencia de Bases , Niño , Proteína de Dominio de Muerte Asociada a Edar/genética , Salud de la Familia , Femenino , Humanos , Factor de Transcripción MSX1/genética , Masculino , Persona de Mediana Edad , Factor de Transcripción PAX9/genética , Polimorfismo de Nucleótido Simple , Proteínas Wnt/genética , Adulto Joven
19.
Hum Mol Genet ; 25(16): 3564-3577, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27378689

RESUMEN

Hypohidrotic ectodermal dysplasia (HED) results from mutation of the EDA, EDAR or EDARADD genes and is characterized by reduced or absent eccrine sweat glands, hair follicles and teeth, and defective formation of salivary, mammary and craniofacial glands. Mouse models with HED also carry Eda, Edar or Edaradd mutations and have defects that map to the same structures. Patients with HED have ear, nose and throat disease, but this has not been investigated in mice bearing comparable genetic mutations. We report that otitis media, rhinitis and nasopharyngitis occur at high frequency in Eda and Edar mutant mice and explore the pathogenic mechanisms related to glandular function, microbial and immune parameters in these lines. Nasopharynx auditory tube glands fail to develop in HED mutant mice and the functional implications include loss of lysozyme secretion, reduced mucociliary clearance and overgrowth of nasal commensal bacteria accompanied by neutrophil exudation. Heavy nasopharynx foreign body load and loss of gland protection alters the auditory tube gating function and the auditory tubes can become pathologically dilated. Accumulation of large foreign body particles in the bulla stimulates granuloma formation. Analysis of immune cell populations and myeloid cell function shows no evidence of overt immune deficiency in HED mutant mice. Our findings using HED mutant mice as a model for the human condition support the idea that ear and nose pathology in HED patients arises as a result of nasal and nasopharyngeal gland deficits, reduced mucociliary clearance and impaired auditory tube gating function underlies the pathological sequelae in the bulla.


Asunto(s)
Displasia Ectodermal Anhidrótica Tipo 1/genética , Ectodisplasinas/genética , Receptor Edar/genética , Proteína de Dominio de Muerte Asociada a Edar/genética , Animales , Modelos Animales de Enfermedad , Oído Medio/patología , Displasia Ectodermal Anhidrótica Tipo 1/fisiopatología , Predisposición Genética a la Enfermedad , Humanos , Ratones , Muramidasa/genética , Muramidasa/metabolismo , Mutación , FN-kappa B/genética , Nariz/patología , Fenotipo
20.
Am J Med Genet A ; 170A(1): 249-53, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26440664

RESUMEN

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.


Asunto(s)
Displasia Ectodermal Anhidrótica Tipo 1/genética , Receptor Edar/genética , Proteína de Dominio de Muerte Asociada a Edar/genética , Mutación Missense/genética , Neoplasias Ováricas/genética , Teratoma/genética , Adolescente , Enfermedades de la Mama/genética , Receptor Edar/metabolismo , Proteína de Dominio de Muerte Asociada a Edar/metabolismo , Femenino , Cabello/crecimiento & desarrollo , Humanos , Masculino , FN-kappa B/metabolismo , Neoplasias Ováricas/patología , Linaje , Transducción de Señal/genética , Teratoma/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...