Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 25(3)2024 Jan 27.
Article in English | MEDLINE | ID: mdl-38338866

ABSTRACT

Papillary thyroid carcinoma (PTC) is the most common histological category of thyroid cancer. In recent years, there has been an increasing number of studies on lncRNAs in PTC. Long intergenic non-protein coding RNA 887 (LINC00887) is a critical oncogene in developing other cancers. LINC00887 is upregulated in PTC samples but its role in PTC is currently unclear. This study aimed to investigate the impact the disruption of LINC00887 expression has on PTC progression. We performed a CRISPR/Cas9 strategy for the truncation of LINC00887 in BCPAP and TPC1 cell lines. Functional assays showed that LINC00887 knockdown in both TPC1 and BCPAP cells reduced cell proliferation, colony formation and migration, delayed the cell cycle, and increased apoptosis. These results strengthened the role of LINC00887 in cancer and showed for the first time that this lncRNA could be a potential oncogene in PTC, acting as a tumor promoter. Modulation of the immune system may be one of the etiopathogenic mechanisms of LINC00887 in PTC, as shown by the observed influence of this lncRNA on PD-L1 expression. In addition, the biological pathways of LINC00887 identified to date, such as EMT, the Wnt/ß-catenin signaling pathway or the FRMD6-Hippo signaling pathway may also be relevant regulatory mechanisms operating in PTC.


Subject(s)
Carcinoma, Papillary , RNA, Long Noncoding , Thyroid Cancer, Papillary , Thyroid Neoplasms , Humans , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , RNA, Long Noncoding/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Up-Regulation/genetics
2.
Nucleic Acids Res ; 49(D1): D1130-D1137, 2021 01 08.
Article in English | MEDLINE | ID: mdl-32990755

ABSTRACT

The knowledge of the genetic variability of the local population is of utmost importance in personalized medicine and has been revealed as a critical factor for the discovery of new disease variants. Here, we present the Collaborative Spanish Variability Server (CSVS), which currently contains more than 2000 genomes and exomes of unrelated Spanish individuals. This database has been generated in a collaborative crowdsourcing effort collecting sequencing data produced by local genomic projects and for other purposes. Sequences have been grouped by ICD10 upper categories. A web interface allows querying the database removing one or more ICD10 categories. In this way, aggregated counts of allele frequencies of the pseudo-control Spanish population can be obtained for diseases belonging to the category removed. Interestingly, in addition to pseudo-control studies, some population studies can be made, as, for example, prevalence of pharmacogenomic variants, etc. In addition, this genomic data has been used to define the first Spanish Genome Reference Panel (SGRP1.0) for imputation. This is the first local repository of variability entirely produced by a crowdsourcing effort and constitutes an example for future initiatives to characterize local variability worldwide. CSVS is also part of the GA4GH Beacon network. CSVS can be accessed at: http://csvs.babelomics.org/.


Subject(s)
Crowdsourcing , Databases, Genetic , Genetics, Population/methods , Genome, Human , Software , Alleles , Chromosome Mapping , Exome , Gene Frequency , Genetic Variation , Genomics , Humans , Internet , Precision Medicine/methods , Spain
3.
PLoS Genet ; 16(11): e1009106, 2020 11.
Article in English | MEDLINE | ID: mdl-33151932

ABSTRACT

Hirschsprung disease (HSCR, OMIM 142623) involves congenital intestinal obstruction caused by dysfunction of neural crest cells and their progeny during enteric nervous system (ENS) development. HSCR is a multifactorial disorder; pathogenetic variants accounting for disease phenotype are identified only in a minority of cases, and the identification of novel disease-relevant genes remains challenging. In order to identify and to validate a potential disease-causing relevance of novel HSCR candidate genes, we established a complementary study approach, combining whole exome sequencing (WES) with transcriptome analysis of murine embryonic ENS-related tissues, literature and database searches, in silico network analyses, and functional readouts using candidate gene-specific genome-edited cell clones. WES datasets of two patients with HSCR and their non-affected parents were analysed, and four novel HSCR candidate genes could be identified: ATP7A, SREBF1, ABCD1 and PIAS2. Further rare variants in these genes were identified in additional HSCR patients, suggesting disease relevance. Transcriptomics revealed that these genes are expressed in embryonic and fetal gastrointestinal tissues. Knockout of these genes in neuronal cells demonstrated impaired cell differentiation, proliferation and/or survival. Our approach identified and validated candidate HSCR genes and provided further insight into the underlying pathomechanisms of HSCR.


Subject(s)
Hirschsprung Disease/genetics , ATP Binding Cassette Transporter, Subfamily D, Member 1/genetics , Animals , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Cell Survival/genetics , Computer Simulation , Copper-Transporting ATPases/genetics , Disease Models, Animal , Gene Expression Profiling , Gene Knockout Techniques , Humans , Infant , Male , Mice , Protein Inhibitors of Activated STAT/genetics , Sterol Regulatory Element Binding Protein 1/genetics , Exome Sequencing
4.
Int J Mol Sci ; 24(9)2023 Apr 25.
Article in English | MEDLINE | ID: mdl-37175550

ABSTRACT

Thyroid carcinoma (TC) can be classified as medullary (MTC) and non-medullary (NMTC). While most TCs are sporadic, familial forms of MTC and NMTC also exist (less than 1% and 3-9% of all TC cases, respectively). Germline mutations in RET are found in more than 95% of familial MTC, whereas familial NMTC shows a high degree of genetic heterogeneity. Herein, we aimed to identify susceptibility genes for familial NMTC and non-RET MTC by whole exome sequencing in 58 individuals belonging to 18 Spanish families with these carcinomas. After data analysis, 53 rare candidate segregating variants were identified in 12 of the families, 7 of them located in previously TC-associated genes. Although no common mutated genes were detected, biological processes regulating functions such as cell proliferation, differentiation, survival and adhesion were enriched. The reported functions of the identified genes together with pathogenicity and structural predictions, reinforced the candidacy of 36 of them, suggesting new loci related to TC and novel genotype-phenotype correlations. Therefore, our strategy provides clues to possible molecular mechanisms underlying familial forms of MTC and NMTC. These new molecular findings and clinical data of patients may be helpful for the early detection, development of tailored therapies and optimizing patient management.


Subject(s)
Carcinoma , Thyroid Neoplasms , Humans , Exome Sequencing , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Germ-Line Mutation
5.
J Transl Med ; 18(1): 73, 2020 02 12.
Article in English | MEDLINE | ID: mdl-32050993

ABSTRACT

BACKGROUND: Retinitis Pigmentosa (RP) is a clinically and genetically heterogeneous disorder that results in inherited blindness. Despite the large number of genes identified, only ~ 60% of cases receive a genetic diagnosis using targeted-sequencing. The aim of this study was to design a whole genome sequencing (WGS) based approach to increase the diagnostic yield of complex Retinitis Pigmentosa cases. METHODS: WGS was conducted in three family members, belonging to one large apparent autosomal dominant RP family that remained unsolved by previous studies, using Illumina TruSeq library preparation kit and Illumina HiSeq X platform. Variant annotation, filtering and prioritization were performed using a number of open-access tools and public databases. Sanger sequencing of candidate variants was conducted in the extended family members. RESULTS: We have developed and optimized an algorithm, based on the combination of different open-access tools, for variant prioritization of WGS data which allowed us to reduce significantly the number of likely causative variants pending to be manually assessed and segregated. Following this algorithm, four heterozygous variants in one autosomal recessive gene (USH2A) were identified, segregating in pairs in the affected members. Additionally, two pathogenic alleles in ADGRV1 and PDZD7 could be contributing to the phenotype in one patient. CONCLUSIONS: The optimization of a diagnostic algorithm for WGS data analysis, accompanied by a hypothesis-free approach, have allowed us to unmask the genetic cause of the disease in one large RP family, as well as to reassign its inheritance pattern which implies differences in the clinical management of these cases. These results contribute to increasing the number of cases with apparently dominant inheritance that carry causal mutations in recessive genes, as well as the possible involvement of various genes in the pathogenesis of RP in one patient. Moreover, our WGS-analysis approach, based on open-access tools, can easily be implemented by other researchers and clinicians to improve the diagnostic yield of additional patients with inherited retinal dystrophies.


Subject(s)
Retinitis Pigmentosa , Algorithms , DNA Mutational Analysis , Humans , Mutation/genetics , Pedigree , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/genetics , Whole Genome Sequencing
6.
Clin Genet ; 97(1): 114-124, 2020 01.
Article in English | MEDLINE | ID: mdl-31355911

ABSTRACT

Hirschsprung disease (HSCR) is a rare congenital disorder caused by an incorrect enteric nervous system development due to a failure in migration, proliferation, differentiation and/or survival of enteric neural crest cells. HSCR is a complex genetic disease, where alterations at different molecular levels are required for the manifestation of the disease. In addition, a wide spectrum of mutations affecting many different genes cause HSCR, although the occurrence and severity of HSCR from many cases still remain unexplained. This review summarizes the current knowledge about molecular genetic basis of HSCR.


Subject(s)
Hirschsprung Disease/genetics , Nervous System Diseases/genetics , Nervous System/pathology , Proto-Oncogene Proteins c-ret/genetics , Hirschsprung Disease/pathology , Humans , Mutation/genetics , Nervous System/metabolism , Nervous System Diseases/pathology , Neural Crest/growth & development , Neural Crest/pathology , Signal Transduction/genetics
7.
Int J Mol Sci ; 21(15)2020 Aug 02.
Article in English | MEDLINE | ID: mdl-32748823

ABSTRACT

Hirschsprung disease (HSCR) is a neurocristopathy defined by intestinal aganglionosis due to alterations during the development of the Enteric Nervous System (ENS). A wide spectrum of molecules involved in different signaling pathways and mechanisms have been described in HSCR onset. Among them, epigenetic mechanisms are gaining increasing relevance. In an effort to better understand the epigenetic basis of HSCR, we have performed an analysis for the identification of long non-coding RNAs (lncRNAs) by qRT-PCR in enteric precursor cells (EPCs) from controls and HSCR patients. We aimed to test the presence of a set lncRNAs among 84 lncRNAs in human EPCs, which were previously related with crucial cellular processes for ENS development, as well as to identify the possible differences between HSCR patients and controls. As a result, we have determined a set of lncRNAs with positive expression in human EPCs that were screened for mutations using the exome data from our cohort of HSCR patients to identify possible variants related to this pathology. Interestingly, we identified three lncRNAs with different levels of their transcripts (SOCS2-AS, MEG3 and NEAT1) between HSCR patients and controls. We propose such lncRNAs as possible regulatory elements implicated in the onset of HSCR as well as potential biomarkers of this pathology.


Subject(s)
Biomarkers/metabolism , Enteric Nervous System/metabolism , Gene Expression Regulation , Genetic Predisposition to Disease/genetics , Hirschsprung Disease/genetics , RNA, Long Noncoding/genetics , Cells, Cultured , Enteric Nervous System/cytology , Female , Genetic Variation , Hirschsprung Disease/diagnosis , Humans , Male , Reverse Transcriptase Polymerase Chain Reaction
8.
Int J Mol Sci ; 21(23)2020 Nov 28.
Article in English | MEDLINE | ID: mdl-33260622

ABSTRACT

Hirschsprung disease (HSCR) is a neurocristopathy characterized by intestinal aganglionosis which is attributed to a failure in neural crest cell (NCC) development during the embryonic stage. The colonization of the intestine by NCCs is a process finely controlled by a wide and complex gene regulatory system. Several genes have been associated with HSCR, but many aspects still remain poorly understood. The present study is focused on deciphering the PAX6 interaction network during enteric nervous system (ENS) formation. A combined experimental and computational approach was performed to identify PAX6 direct targets, as well as gene networks shared among such targets as potential susceptibility factors for HSCR. As a result, genes related to PAX6 either directly (RABGGTB and BRD3) or indirectly (TGFB1, HRAS, and GRB2) were identified as putative genes associated with HSCR. Interestingly, GRB2 is involved in the RET/GDNF/GFRA1 signaling pathway, one of the main pathways implicated in the disease. Our findings represent a new contribution to advance in the knowledge of the genetic basis of HSCR. The investigation of the role of these genes could help to elucidate their implication in HSCR onset.


Subject(s)
Chromatin Immunoprecipitation Sequencing , Enteric Nervous System/embryology , Hirschsprung Disease/embryology , Hirschsprung Disease/genetics , Animals , Base Sequence , Gene Expression Regulation , Genetic Predisposition to Disease , Genome , Mice , Nucleotide Motifs/genetics , PAX6 Transcription Factor/metabolism , Spheroids, Cellular/pathology
9.
Int J Mol Sci ; 21(24)2020 Dec 08.
Article in English | MEDLINE | ID: mdl-33302505

ABSTRACT

The management of unsolved inherited retinal dystrophies (IRD) cases is challenging since no standard pipelines have been established. This study aimed to define a diagnostic algorithm useful for the diagnostic routine and to address unsolved cases. Here, we applied a Next-Generation Sequencing-based workflow, including a first step of panel sequencing (PS) followed by clinical-exome sequencing (CES) and whole-exome sequencing (WES), in 46 IRD patients belonging to 42 families. Twenty-six likely causal variants in retinal genes were found by PS and CES. CES and WES allowed proposing two novel candidate loci (WDFY3 and a X-linked region including CITED1), both abundantly expressed in human retina according to RT-PCR and immunohistochemistry. After comparison studies, PS showed the best quality and cost values, CES and WES involved similar analytical efforts and WES presented the highest diagnostic yield. These results reinforce the relevance of panels as a first step in the diagnostic routine and suggest WES as the next strategy for unsolved cases, reserving CES for the simultaneous study of multiple conditions. Standardizing this algorithm would enhance the efficiency and equity of clinical genetics practice. Furthermore, the identified candidate genes could contribute to increase the diagnostic yield and expand the mutational spectrum in these disorders.


Subject(s)
Exome Sequencing/methods , Genetic Testing/methods , Retinal Dystrophies/genetics , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Autophagy-Related Proteins/genetics , Genetic Testing/standards , Humans , Mutation , Retinal Dystrophies/diagnosis , Trans-Activators/genetics , Exome Sequencing/standards , Workflow
10.
Int J Mol Sci ; 20(13)2019 Jun 26.
Article in English | MEDLINE | ID: mdl-31247956

ABSTRACT

Hirschsprung disease (HSCR, OMIM 142623) is due to a failure of enteric precursor cells derived from neural crest (EPCs) to proliferate, migrate, survive or differentiate during Enteric Nervous System (ENS) formation. This is a complex process which requires a strict regulation that results in an ENS specific gene expression pattern. Alterations at this level lead to the onset of neurocristopathies such as HSCR. Gene expression is regulated by different mechanisms, such as DNA modifications (at the epigenetic level), transcriptional mechanisms (transcription factors, silencers, enhancers and repressors), postranscriptional mechanisms (3'UTR and ncRNA) and regulation of translation. All these mechanisms are finally implicated in cell signaling to determine the migration, proliferation, differentiation and survival processes for correct ENS development. In this review, we have performed an overview on the role of epigenetic mechanisms at transcriptional and posttranscriptional levels on these cellular events in neural crest cells (NCCs), ENS development, as well as in HSCR.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , Genetic Association Studies , Genetic Predisposition to Disease , Hirschsprung Disease/genetics , Adenosine Triphosphate/metabolism , Animals , Biomarkers , Chromatin Assembly and Disassembly/genetics , DNA Methylation , Genetic Association Studies/methods , Hirschsprung Disease/diagnosis , Hirschsprung Disease/metabolism , Hirschsprung Disease/therapy , Histones/metabolism , Humans , Polycomb-Group Proteins/metabolism , RNA, Untranslated/genetics
11.
Hum Mol Genet ; 25(23): 5265-5275, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27702942

ABSTRACT

Hirschsprung disease (HSCR) is the most common cause of neonatal intestinal obstruction. It is characterized by the absence of ganglia in the nerve plexuses of the lower gastrointestinal tract. So far, three common disease-susceptibility variants at the RET, SEMA3 and NRG1 loci have been detected through genome-wide association studies (GWAS) in Europeans and Asians to understand its genetic etiologies. Here we present a trans-ethnic meta-analysis of 507 HSCR cases and 1191 controls, combining all published GWAS results on HSCR to fine-map these loci and narrow down the putatively causal variants to 99% credible sets. We also demonstrate that the effects of RET and NRG1 are universal across European and Asian ancestries. In contrast, we detected a European-specific association of a low-frequency variant, rs80227144, in SEMA3 [odds ratio (OR) = 5.2, P = 4.7 × 10-10]. Conditional analyses on the lead SNPs revealed a secondary association signal, corresponding to an Asian-specific, low-frequency missense variant encoding RET p.Asp489Asn (rs9282834, conditional OR = 20.3, conditional P = 4.1 × 10-14). When in trans with the RET intron 1 enhancer risk allele, rs9282834 increases the risk of HSCR from 1.1 to 26.7. Overall, our study provides further insights into the genetic architecture of HSCR and has profound implications for future study designs.


Subject(s)
Genetic Predisposition to Disease , Hirschsprung Disease/genetics , Neuregulin-1/genetics , Proto-Oncogene Proteins c-ret/genetics , Semaphorin-3A/genetics , Alleles , Asian People/genetics , Ethnicity/genetics , Female , Genome-Wide Association Study , Genotype , Hirschsprung Disease/pathology , Humans , Introns/genetics , Male , Polymorphism, Single Nucleotide , White People/genetics
12.
Am J Hum Genet ; 96(4): 581-96, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25839327

ABSTRACT

Innervation of the gut is segmentally lost in Hirschsprung disease (HSCR), a consequence of cell-autonomous and non-autonomous defects in enteric neuronal cell differentiation, proliferation, migration, or survival. Rare, high-penetrance coding variants and common, low-penetrance non-coding variants in 13 genes are known to underlie HSCR risk, with the most frequent variants in the ret proto-oncogene (RET). We used a genome-wide association (220 trios) and replication (429 trios) study to reveal a second non-coding variant distal to RET and a non-coding allele on chromosome 7 within the class 3 Semaphorin gene cluster. Analysis in Ret wild-type and Ret-null mice demonstrates specific expression of Sema3a, Sema3c, and Sema3d in the enteric nervous system (ENS). In zebrafish embryos, sema3 knockdowns show reduction of migratory ENS precursors with complete ablation under conjoint ret loss of function. Seven candidate receptors of Sema3 proteins are also expressed within the mouse ENS and their expression is also lost in the ENS of Ret-null embryos. Sequencing of SEMA3A, SEMA3C, and SEMA3D in 254 HSCR-affected subjects followed by in silico protein structure modeling and functional analyses identified five disease-associated alleles with loss-of-function defects in semaphorin dimerization and binding to their cognate neuropilin and plexin receptors. Thus, semaphorin 3C/3D signaling is an evolutionarily conserved regulator of ENS development whose dys-regulation is a cause of enteric aganglionosis.


Subject(s)
Epistasis, Genetic/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation , Hirschsprung Disease/genetics , Proto-Oncogene Proteins c-ret/genetics , Semaphorins/genetics , Animals , Base Sequence , Genome-Wide Association Study , Mice , Molecular Sequence Data , Semaphorins/deficiency , Semaphorins/metabolism , Sequence Analysis, DNA
13.
Dev Biol ; 417(2): 198-208, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27426273

ABSTRACT

Abnormal development or disturbed functioning of the enteric nervous system (ENS), the intrinsic innervation of the gastrointestinal tract, is associated with the development of neuropathic gastrointestinal motility disorders. Here, we review the underlying molecular basis of these disorders and hypothesize that many of them have a common defective biological mechanism. Genetic burden and environmental components affecting this common mechanism are ultimately responsible for disease severity and symptom heterogeneity. We believe that they act together as the fulcrum in a seesaw balanced with harmful and protective factors, and are responsible for a continuum of symptoms ranging from neuronal hyperplasia to absence of neurons.


Subject(s)
Enteric Nervous System/pathology , Gastrointestinal Motility/physiology , Gastrointestinal Tract/innervation , Gastrointestinal Tract/pathology , Gene-Environment Interaction , Enteric Nervous System/growth & development , Gastrointestinal Motility/genetics , Hirschsprung Disease/genetics , Humans , Myocytes, Smooth Muscle/physiology
14.
Mol Biol Evol ; 33(5): 1205-18, 2016 05.
Article in English | MEDLINE | ID: mdl-26764160

ABSTRACT

Recent results from large-scale genomic projects suggest that allele frequencies, which are highly relevant for medical purposes, differ considerably across different populations. The need for a detailed catalog of local variability motivated the whole-exome sequencing of 267 unrelated individuals, representative of the healthy Spanish population. Like in other studies, a considerable number of rare variants were found (almost one-third of the described variants). There were also relevant differences in allelic frequencies in polymorphic variants, including ∼10,000 polymorphisms private to the Spanish population. The allelic frequencies of variants conferring susceptibility to complex diseases (including cancer, schizophrenia, Alzheimer disease, type 2 diabetes, and other pathologies) were overall similar to those of other populations. However, the trend is the opposite for variants linked to Mendelian and rare diseases (including several retinal degenerative dystrophies and cardiomyopathies) that show marked frequency differences between populations. Interestingly, a correspondence between differences in allelic frequencies and disease prevalence was found, highlighting the relevance of frequency differences in disease risk. These differences are also observed in variants that disrupt known drug binding sites, suggesting an important role for local variability in population-specific drug resistances or adverse effects. We have made the Spanish population variant server web page that contains population frequency information for the complete list of 170,888 variant positions we found publicly available (http://spv.babelomics.org/), We show that it if fundamental to determine population-specific variant frequencies to distinguish real disease associations from population-specific polymorphisms.


Subject(s)
Disease/genetics , Exome , Databases, Nucleic Acid , Drug Resistance/genetics , Gene Frequency , Genetic Predisposition to Disease , Genetic Variation , Genetics, Population/methods , Humans , Internet , Pharmacogenomic Testing , Polymorphism, Genetic , Spain/epidemiology
15.
Am J Med Genet A ; 170A(5): 1268-73, 2016 May.
Article in English | MEDLINE | ID: mdl-26762557

ABSTRACT

Monosomy 10p is a rare chromosomal disorder with a prevalence <1/1,000,000, in which a terminal or interstitial distal region of chromosome 10 is deleted resulting in a variable phenotype depending on the size of the deletion. Two main phenotypes have been defined depending on the location of the deletion: HDR syndrome (Hypoparathyroidism, sensorineural Deafness, and Renal disease), and DGS2 (DiGeorge syndrome type 2). The vast majority of cases reported so far have resulted from de novo events. Here, we present the first familial presentation of this contiguous gene deletion syndrome, affecting two family members in different generations: a child and his maternal uncle. In both cases, the deletion was due to a malsegregation of a maternal balanced rearrangement, ins(16;10)(q22;p13p15.2). The identification and characterization of this rearrangement was possible using a combination of different genetic analyses such as karyotype, MLPA, FISH, and array CGH. We underline the importance of the present results in terms of genetic and reproductive counseling for the carriers of the balanced rearrangement within the family, and demonstrate again the utility of expanding the genetic studies to the relatives of the affected patients.


Subject(s)
Chromosome Disorders/genetics , Chromosomes, Human, Pair 10/genetics , DiGeorge Syndrome/genetics , Hearing Loss, Sensorineural/genetics , Hypoparathyroidism/genetics , Nephrosis/genetics , Adult , Child , Chromosome Deletion , Chromosome Disorders/physiopathology , Comparative Genomic Hybridization , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , DiGeorge Syndrome/physiopathology , Gene Deletion , Hearing Loss, Sensorineural/physiopathology , Humans , Hypoparathyroidism/physiopathology , In Situ Hybridization, Fluorescence , Karyotyping , Male , Nephrosis/physiopathology
16.
BMC Med Genet ; 16: 89, 2015 Oct 05.
Article in English | MEDLINE | ID: mdl-26437850

ABSTRACT

BACKGROUND: The development of next-generation sequencing (NGS) technologies has a great impact in the human variation detection given their high-throughput. These techniques are particularly helpful for the evaluation of the genetic background in disorders of complex genetic etiology such as Hirschsprung disease (HSCR). The purpose of this study was the design of a panel of HSCR associated genes as a rapid and efficient tool to perform genetic screening in a series of patients. METHODS: We have performed NGS-based targeted sequencing (454-GS Junior) using a panel containing 26 associated or candidate genes for HSCR in a group of 11 selected HSCR patients. RESULTS: The average percentage of covered bases was of 97%, the 91.4% of the targeted bases were covered with depth above 20X and the mean coverage was 422X. In addition, we have found a total of 13 new coding variants and 11 new variants within regulatory regions among our patients. These outcomes allowed us to re-evaluate the genetic component associated to HSCR in these patients. CONCLUSIONS: Our validated NGS panel constitutes an optimum method for the identification of new variants in our patients. This approach could be used for a fast, reliable and more thorough genetic screening in future series of patients.


Subject(s)
Genetic Testing/methods , High-Throughput Nucleotide Sequencing/methods , Hirschsprung Disease/genetics , Phenotype , Base Sequence , Computational Biology , Female , Gene Library , Hirschsprung Disease/pathology , Humans , Male , Molecular Sequence Data , Sensitivity and Specificity , Spain
17.
Am J Med Genet A ; 167(7): 1597-600, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25823529

ABSTRACT

Mutations in USH2A are a common cause of Retinitis Pigmentosa (RP). Among the most frequently reported USH2A variants, c.2276G>T (p.C759F) has been found in both affected and healthy individuals. The pathogenicity of this variant remains controversial since it was detected in homozygosity in two healthy siblings of a Spanish family (S23), eleven years ago. The fact that these individuals remain asymptomatic today, prompted us to study the presence of other pathogenic variants in this family using targeted resequencing of 26 retinal genes in one of the affected individuals. This approach allowed us to identify one novel pathogenic homozygous mutation in exon 13 of PDE6B (c.1678C>T; p.R560C). This variant cosegregated with the disease and was absent in 200 control individuals. Remarkably, the identified variant in PDE6B corresponds to the mutation responsible of the retinal degeneration in the naturally occurring rd10 mutant mice. To our knowledge, this is the first report of the identification of the rd10 mice mutation in a RP family. These findings, together with a review of the literature, support the hypothesis that homozygous p.C759F mutations are not pathogenic and led us to exclude the implication of p.C759F in the RP of family S23. Our results indicate the need of re-evaluating all families genetically diagnosed with this mutation.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Extracellular Matrix Proteins/genetics , Retinitis Pigmentosa/genetics , Base Sequence , Extracellular Matrix Proteins/adverse effects , Gene Library , Humans , Molecular Sequence Data , Mutation, Missense/genetics , Pedigree , Retinitis Pigmentosa/pathology , Sequence Analysis, DNA , Spain
18.
Hum Mutat ; 35(3): 303-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24357527

ABSTRACT

A deletion encompassing several SOX10 enhancers was recently identified in a patient presenting with Waardenburg syndrome type 4 (WS4), which is defined as a combination of Hirschsprung disease (HSCR, intestinal aganglionosis) and WS (deafness and pigmentation defects). The expression patterns of some of the known SOX10 enhancers in animal models led to the speculation that endophenotypes of WS4 may be linked to mutations within some of these sequences. The present study investigated deletions and point mutations within four SOX10 enhancers in 144 unexplained isolated HSCR cases. One deletion and two point mutations affecting binding sites for known neural crest transcription factors were identified. In vitro functional analysis revealed that the first point mutation disrupts autoregulation by SOX10, whereas the second affects AP2a and SOX10 synergistic activity. The present findings suggest that the mutations within SOX10 enhancers contribute to isolated HSCR.


Subject(s)
Regulatory Sequences, Nucleic Acid , SOXE Transcription Factors/genetics , Waardenburg Syndrome/genetics , Base Sequence , Female , Hirschsprung Disease , Humans , Infant , Male , Molecular Sequence Data , Point Mutation , Sequence Deletion , Transcription Factors/genetics
19.
Dev Biol ; 382(1): 320-9, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23707863

ABSTRACT

Finding genes for complex diseases has been the goal of many genetic studies. Most of these studies have been successful by searching for genes and mutations in rare familial cases, by screening candidate genes and by performing genome wide association studies. However, only a small fraction of the total genetic risk for these complex genetic diseases can be explained by the identified mutations and associated genetic loci. In this review we focus on Hirschsprung disease (HSCR) as an example of a complex genetic disorder. We describe the genes identified in this congenital malformation and postulate that both common 'low penetrant' variants in combination with rare or private 'high penetrant' variants determine the risk on HSCR, and likely, on other complex diseases. We also discuss how new technological advances can be used to gain further insights in the genetic background of complex diseases. Finally, we outline a few steps to develop functional assays in order to determine the involvement of these variants in disease development.


Subject(s)
Genetic Variation , Hirschsprung Disease/genetics , Models, Biological , Animals , Genetic Association Studies , Genetic Predisposition to Disease , Hirschsprung Disease/pathology , Humans
20.
Genet Med ; 16(9): 703-10, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24577265

ABSTRACT

PURPOSE: Hirschsprung disease (OMIM 142623) is a neurocristopathy attributed to a failure of cell proliferation or migration and/or failure of the enteric precursors along the gut to differentiate during embryonic development. Although some genes involved in this pathology are well characterized, many aspects remain poorly understood. In this study, we aimed to identify novel genes implicated in the pathogenesis of Hirschsprung disease. METHODS: We compared the expression patterns of genes involved in human stem cell pluripotency between enteric precursors from controls and Hirschsprung disease patients. We further evaluated the role of DNMT3B in the context of Hirschsprung disease by inmunocytochemistry, global DNA methylation assays, and mutational screening. RESULTS: Seven differentially expressed genes were identified. We focused on DNMT3B, which encodes a DNA methyltransferase that performs de novo DNA methylation during embryonic development. DNMT3B mutational analysis in our Hirschsprung disease series revealed the presence of potentially pathogenic mutations (p.Gly25Arg, p.Arg190Cys, and p.Gly198Trp). CONCLUSION: DNMT3B may be regulating enteric nervous system development through DNA methylation in the neural crest cells, suggesting that aberrant methylation patterns could have a relevant role in Hirschsprung disease. Moreover, the synergistic effect of mutations in both DNMT3B and other Hirschsprung disease-related genes may be contributing to a more severe phenotype in our Hirschsprung disease patients.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Enteric Nervous System , Hirschsprung Disease/genetics , Neurogenesis/genetics , Biomarkers , Case-Control Studies , Child, Preschool , Cluster Analysis , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Female , Gene Expression Profiling , Gene Expression Regulation , Gene Order , Genetic Loci , Hirschsprung Disease/metabolism , Humans , Infant , Male , Mutation , Neural Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , DNA Methyltransferase 3B
SELECTION OF CITATIONS
SEARCH DETAIL