Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Hum Mol Genet ; 26(15): 2838-2849, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28449119

RESUMEN

CblX (MIM309541) is an X-linked recessive disorder characterized by defects in cobalamin (vitamin B12) metabolism and other developmental defects. Mutations in HCFC1, a transcriptional co-regulator which interacts with multiple transcription factors, have been associated with cblX. HCFC1 regulates cobalamin metabolism via the regulation of MMACHC expression through its interaction with THAP11, a THAP domain-containing transcription factor. The HCFC1/THAP11 complex potentially regulates genes involved in diverse cellular functions including cell cycle, proliferation, and transcription. Thus, it is likely that mutation of THAP11 also results in biochemical and other phenotypes similar to those observed in patients with cblX. We report a patient who presented with clinical and biochemical phenotypic features that overlap cblX, but who does not have any mutations in either MMACHC or HCFC1. We sequenced THAP11 by Sanger sequencing and discovered a potentially pathogenic, homozygous variant, c.240C > G (p.Phe80Leu). Functional analysis in the developing zebrafish embryo demonstrated that both THAP11 and HCFC1 regulate the proliferation and differentiation of neural precursors, suggesting important roles in normal brain development. The loss of THAP11 in zebrafish embryos results in craniofacial abnormalities including the complete loss of Meckel's cartilage, the ceratohyal, and all of the ceratobranchial cartilages. These data are consistent with our previous work that demonstrated a role for HCFC1 in vertebrate craniofacial development. High throughput RNA-sequencing analysis reveals several overlapping gene targets of HCFC1 and THAP11. Thus, both HCFC1 and THAP11 play important roles in the regulation of cobalamin metabolism as well as other pathways involved in early vertebrate development.


Asunto(s)
Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Vitamina B 12/metabolismo , Animales , Secuencia de Bases , Región Branquial/metabolismo , Diferenciación Celular , Niño , Anomalías Craneofaciales/genética , Fibroblastos , Regulación de la Expresión Génica/genética , Factor C1 de la Célula Huésped/química , Factor C1 de la Célula Huésped/genética , Factor C1 de la Célula Huésped/metabolismo , Humanos , Mutación , Cultivo Primario de Células , Transcripción Genética , Vitamina B 12/genética , Pez Cebra/genética
2.
Hum Mutat ; 38(5): 511-516, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28185376

RESUMEN

Deleterious variants in the same gene present in two or more families with overlapping clinical features provide convincing evidence of a disease-gene association; this can be a challenge in the study of ultrarare diseases. To facilitate the identification of additional families, several groups have created "matching" platforms. We describe four individuals from three unrelated families "matched" by GeneMatcher and MatchMakerExchange. Individuals had microcephaly, developmental delay, epilepsy, and recessive mutations in TRIT1. A single homozygous mutation in TRIT1 associated with similar features had previously been reported in one family. The identification of these individuals provides additional evidence to support TRIT1 as the disease-causing gene and interprets the variants as "pathogenic." TRIT1 functions to modify mitochondrial tRNAs and is necessary for protein translation. We show that dysfunctional TRIT1 results in decreased levels of select mitochondrial proteins. Our findings confirm the TRIT1 disease association and advance the phenotypic and molecular understanding of this disorder.


Asunto(s)
Transferasas Alquil y Aril/genética , Alelos , Genes Recesivos , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/genética , Mutación , Adolescente , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Niño , Preescolar , Facies , Femenino , Pruebas Genéticas , Homocigoto , Humanos , Imagen por Resonancia Magnética , Masculino , Fenotipo
3.
J Med Genet ; 53(10): 705-9, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27330106

RESUMEN

BACKGROUND: Osteonecrosis of the femoral head is a debilitating disease that involves impaired blood supply to the femoral head and leads to femoral head collapse. METHODS: We use whole-exome sequencing and Sanger sequencing to analyse a family with inherited osteonecrosis of the femoral head and fluorescent Ca(2+) imaging to functionally characterise the variant protein. RESULTS: We report a family with four siblings affected with inherited osteonecrosis of the femoral head and the identification of a c.2480_2483delCCCG frameshift deletion followed by a c.2486T>A substitution in one allele of the transient receptor potential vanilloid 4 (TRPV4) gene. TRPV4 encodes a Ca(2+)-permeable cation channel known to play a role in vasoregulation and osteoclast differentiation. While pathogenic TRPV4 mutations affect the skeletal or nervous systems, association with osteonecrosis of the femoral head is novel. Functional measurements of Ca(2+) influx through mutant TRPV4 channels in HEK293 cells and patient-derived dermal fibroblasts identified a TRPV4 gain of function. Analysis of channel open times, determined indirectly from measurement of TRPV4 activity within a cluster of TRPV4 channels, revealed that the TRPV4 gain of function was caused by longer channel openings. CONCLUSIONS: These findings identify a novel TRPV4 mutation implicating TRPV4 and altered calcium homeostasis in the pathogenesis of osteonecrosis while reinforcing the importance of TRPV4 in bone diseases and vascular endothelium.


Asunto(s)
Necrosis de la Cabeza Femoral/metabolismo , Mutación , Canales Catiónicos TRPV/genética , Secuencia de Aminoácidos , Calcio/metabolismo , Femenino , Necrosis de la Cabeza Femoral/genética , Humanos , Masculino , Linaje , Alineación de Secuencia , Canales Catiónicos TRPV/química , Canales Catiónicos TRPV/metabolismo , Adulto Joven
4.
Hum Mutat ; 37(9): 976-82, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27349184

RESUMEN

Vitamin B12 (cobalamin, Cbl) cofactors adenosylcobalamin (AdoCbl) and methylcobalamin (MeCbl) are required for the activity of the enzymes methylmalonyl-CoA mutase (MCM) and methionine synthase (MS). Inborn errors of Cbl metabolism are rare Mendelian disorders associated with hematological and neurological manifestations, and elevations of methylmalonic acid and/or homocysteine in the blood and urine. We describe a patient whose fibroblasts had decreased functional activity of MCM and MS and decreased synthesis of AdoCbl and MeCbl (3.4% and 1.0% of cellular Cbl, respectively). The defect in cultured patient fibroblasts complemented those from all known complementation groups. Patient cells accumulated transcobalamin-bound-Cbl, a complex which usually dissociates in the lysosome to release free Cbl. Whole-exome sequencing identified putative disease-causing variants c.851T>G (p.L284*) and c.1019C>T (p.T340I) in transcription factor ZNF143. Proximity biotinylation analysis confirmed the interaction between ZNF143 and HCFC1, a protein that regulates expression of the Cbl trafficking enzyme MMACHC. qRT-PCR analysis revealed low MMACHC expression levels both in patient fibroblasts, and in control fibroblasts incubated with ZNF143 siRNA.


Asunto(s)
Citoplasma/metabolismo , Errores Innatos del Metabolismo/genética , Transactivadores/genética , Transcobalaminas/metabolismo , Vitamina B 12/metabolismo , Proteínas Portadoras/metabolismo , Células Cultivadas , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Lactante , Masculino , Errores Innatos del Metabolismo/metabolismo , Errores Innatos del Metabolismo/patología , Mutación , Oxidorreductasas , Linaje
5.
Mol Genet Metab ; 117(3): 363-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26827111

RESUMEN

Next generation sequencing (NGS) based gene panel testing is increasingly available as a molecular diagnostic approach for inborn errors of metabolism. Over the past 40 years patients have been referred to the Vitamin B12 Clinical Research Laboratory at McGill University for diagnosis of inborn errors of cobalamin metabolism by functional studies in cultured fibroblasts. DNA samples from patients in which no diagnosis was made by these studies were tested by a NGS gene panel to determine whether any molecular diagnoses could be made. 131 DNA samples from patients with elevated methylmalonic acid and no diagnosis following functional studies of cobalamin metabolism were analyzed using the 24 gene extended cobalamin metabolism NGS based panel developed by Baylor Miraca Genetics Laboratories. Gene panel testing identified two or more variants in a single gene in 16/131 patients. Eight patients had pathogenic findings, one had a finding of uncertain significance, and seven had benign findings. Of the patients with pathogenic findings, five had mutations in ACSF3, two in SUCLG1 and one in TCN2. Thus, the NGS gene panel allowed for the presumptive diagnosis of 8 additional patients for which a diagnosis was not made by the functional assays.


Asunto(s)
Errores Innatos del Metabolismo/diagnóstico , Errores Innatos del Metabolismo/genética , Ácido Metilmalónico/metabolismo , Deficiencia de Vitamina B 12/diagnóstico , Deficiencia de Vitamina B 12/genética , Vitamina B 12/metabolismo , Complejo Vitamínico B/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/genética , Preescolar , Coenzima A Ligasas/genética , Femenino , Pruebas Genéticas/métodos , Variación Genética , Genotipo , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Lactante , Recién Nacido , Masculino , Técnicas de Diagnóstico Molecular , Mutación , Fenotipo , Succinato-CoA Ligasas/genética
6.
Mol Genet Metab ; 118(4): 264-71, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27233228

RESUMEN

Mutations in the MUT gene, which encodes the mitochondrial enzyme methylmalonyl-CoA mutase, are responsible for the mut form of methylmalonic aciduria (MMA). In this study, a next generation sequencing (NGS) based gene panel was used to analyze 53 patients that had been diagnosed with mut MMA by somatic cell complementation analysis. A total of 54 different mutations in MUT were identified in 48 patients; 16 novel mutations were identified, including 1 initiation site mutation (c.2T>C [p.M1?]), 1 missense mutation (c.566A>T [p.N189I]), 2 nonsense mutations (c.129G>A [p.W43*] and c.1975C>T [p.Q659*]), 2 mutations affecting splice sites (c.753+3A>G and c.754-2A>G), 8 small insertions, deletions, and duplications (c.29dupT [p.L10Ffs*39], c.55dupG [p.V19Gfs*30], c.631_633delGAG [p.E211del], c.795_796insT [p.M266Yfs*7], c.1061delCinsGGA [p.S354Wfs*20], c.1065_1068dupATGG [p.S357Mfs*5], c.1181dupT [p.L394Ffs*30], c.1240delG [p.E414Kfs*17]), a large insertion (c.146_147ins279), and a large deletion involving exon 13. Phenotypic rescue and cDNA analysis were used to confirm that the c.146_147ins279 and c.631_633delGAG mutations were associated with the decreased methylmalonyl-CoA mutase function observed in the patient fibroblasts. In five patients, the NGS panel did not confirm the diagnosis made by complementation analysis. One of these patients was found to carry 2 novel mutations (c.433G > A [p.E145K] and c.511A>C [p.N171H]) in the SUCLG1 gene.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Metilmalonil-CoA Mutasa/genética , Adolescente , Adulto , Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Células Cultivadas , Niño , Preescolar , Codón sin Sentido/genética , Exones/genética , Femenino , Fibroblastos/metabolismo , Humanos , Mutación INDEL/genética , Lactante , Masculino , Ácido Metilmalónico/metabolismo , Mitocondrias/enzimología , Mutación Missense/genética , Fenotipo , Eliminación de Secuencia/genética , Succinato-CoA Ligasas/genética , Adulto Joven
7.
Hum Mol Genet ; 22(22): 4591-601, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23825108

RESUMEN

The cblG and cblC disorders of cobalamin (Cbl) metabolism are two inherited causes of megaloblastic anaemia. In cblG, mutations in methionine synthase (MTR) decrease conversion of hydroxocobalamin  (HOCbl) to methylcobalamin, while in cblC, mutations in MMACHC disrupt formation of cob(II)alamin (detected as HOCbl). Cases with undetectable methionine synthase (MS) activity are extremely rare and classified as 'cblG-variant'. In four 'cblG-variant' cases, we observed a decreased conversion of cyanocobalamin to HOCbl that is also seen in cblC cases. To explore this observation, we studied the gene defects, splicing products and expression of MS, as well as MS/MMACHC protein interactions in cblG-variant, cblG, cblC and control fibroblasts. We observed a full-size MS encoded by MTR-001 and a 124 kDa truncated MS encoded by MTR-201 in cblG, cblC, control fibroblasts and HEK cells, but only the MTR-201 transcript and inactive truncated MS in cblG-variant cells. Co-immunoprecipitation and proximity ligation assay showed interaction between truncated MS and MMACHC in cblG-variant cells. This interaction decreased 2.2, 1.5 and 5.0-fold in the proximity ligation assay of cblC cells with p.R161Q and p.R206W mutations, and HEK cells with knock down expression of MS by siRNA, respectively, when compared with control cells. In 3D modelling and docking analysis, both truncated and full-size MS provide a loop anchored to MMACHC, which makes contacts with R-161 and R-206 residues. Our data suggest that the interaction of MS with MMACHC may play a role in the regulation of the cellular processing of Cbls that is required for Cbl cofactor synthesis.


Asunto(s)
5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/metabolismo , Anemia Megaloblástica/genética , Proteínas Portadoras/metabolismo , Isoformas de Proteínas/metabolismo , Deficiencia de Vitamina B 12/metabolismo , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/química , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética , Sitios de Unión/genética , Proteínas Portadoras/química , Proteínas Portadoras/genética , Células Cultivadas , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Hidroxocobalamina/metabolismo , Modelos Moleculares , Simulación del Acoplamiento Molecular , Oxidorreductasas , Unión Proteica/genética , Isoformas de Proteínas/genética , Estructura Secundaria de Proteína , Vitamina B 12/análogos & derivados , Vitamina B 12/metabolismo , Deficiencia de Vitamina B 12/genética
8.
Mol Genet Metab ; 112(3): 198-204, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24889031

RESUMEN

Patients with mutations in MMACHC have the autosomal recessive disease of cobalamin metabolism known as cblC. These patients are unable to convert cobalamin into the two active forms, methylcobalamin and adenosylcobalamin and consequently have elevated homocysteine and methylmalonic acid in blood and urine. In addition, some cblC patients have structural abnormalities, including congenital heart defects. MMACHC is conserved in the mouse and shows tissue and stage-specific expression pattern in midgestation stage embryos. To create a mouse model of cblC we generated a line of mice with a gene-trap insertion in intron 1 of the Mmachc gene, (Mmachc(Gt(AZ0348)Wtsi)). Heterozygous mice show a 50% reduction of MMACHC protein, and have significantly higher levels of homocysteine and methylmalonic acid in their blood. The Mmachc(Gt) allele was inherited with a transmission ratio distortion in matings with heterozygous animals. Furthermore, homozygous Mmachc(Gt) embryos were not found after embryonic day 3.5 and these embryos were unable to generate giant cells in outgrowth assays. Our findings confirm that cblC is modeled in mice with reduced levels of Mmachc and suggest an early requirement for Mmachc in mouse development.


Asunto(s)
Proteínas Portadoras/genética , Desarrollo Embrionario/genética , Alelos , Errores Innatos del Metabolismo de los Aminoácidos/genética , Animales , Femenino , Orden Génico , Marcación de Gen , Vectores Genéticos/genética , Genotipo , Hiperhomocisteinemia/genética , Masculino , Ratones , Oxidorreductasas , Fenotipo
10.
Clin Epigenetics ; 14(1): 52, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35440018

RESUMEN

BACKGROUND: epi-cblC is a recently discovered inherited disorder of intracellular vitamin B12 metabolism associating hematological, neurological, and cardiometabolic outcomes. It is produced by an epimutation at the promoter common to CCDC163P and MMACHC, which results from an aberrant antisense transcription due to splicing mutations in the antisense PRDX1 gene neighboring MMACHC. We studied whether the aberrant transcription produced a second epimutation by encompassing the CpG island of the TESK2 gene neighboring CCDC163P. METHODS: We unraveled the methylome architecture of the CCDC163P-MMACHC CpG island (CpG:33) and the TESK2 CpG island (CpG:51) of 17 epi-cblC cases. We performed an integrative analysis of the DNA methylome profiling, transcriptome reconstruction of RNA-sequencing (RNA-seq), chromatin immunoprecipitation sequencing (ChIP-Seq) of histone H3, and transcription expression of MMACHC and TESK2. RESULTS: The PRDX1 splice mutations and activation of numerous cryptic splice sites produced antisense readthrough transcripts encompassing the bidirectional MMACHC/CCDC163P promoter and the TESK2 promoter, resulting in the silencing of both the MMACHC and TESK2 genes through the deposition of SETD2-dependent H3K36me3 marks and the generation of epimutations in the CpG islands of the two promoters. CONCLUSIONS: The antisense readthrough transcription of the mutated PRDX1 produces an epigenetic silencing of MMACHC and TESK2. We propose using the term 'epi-digenism' to define this epigenetic disorder that affects two genes. Epi-cblC is an entity that differs from cblC. Indeed, the PRDX1 and TESK2 altered expressions are observed in epi-cblC but not in cblC, suggesting further evaluating the potential consequences on cancer risk and spermatogenesis.


Asunto(s)
Homocistinuria , Vitamina B 12 , Metilación de ADN , Homocistinuria/genética , Homocistinuria/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Mutación , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Proteínas Serina-Treonina Quinasas , Vitaminas
11.
Mol Genet Metab ; 103(4): 401-5, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21536470

RESUMEN

To examine whether Mmachc and Mmadhc, two genes involved in vitamin B(12) (cobalamin) metabolism, show tissue-specific expression during mouse embryogenesis, we determined their sites of expression at 11.5days post conception by in situ hybridization. There was ubiquitous expression of Mmadhc, but tissue and cell type-specific expression of Mmachc in the developing lung, heart, cardiovascular and nervous system. This suggests that during organogenesis Mmachc and Mmadhc may interact in only a subset of cells.


Asunto(s)
Proteínas Portadoras/genética , Animales , Proteínas Portadoras/metabolismo , Embrión de Mamíferos/metabolismo , Hibridación in Situ , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos , Especificidad de Órganos , Organogénesis/genética , Oxidorreductasas , Vitamina B 12/metabolismo
12.
Nat Commun ; 9(1): 67, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29302025

RESUMEN

To date, epimutations reported in man have been somatic and erased in germlines. Here, we identify a cause of the autosomal recessive cblC class of inborn errors of vitamin B12 metabolism that we name "epi-cblC". The subjects are compound heterozygotes for a genetic mutation and for a promoter epimutation, detected in blood, fibroblasts, and sperm, at the MMACHC locus; 5-azacytidine restores the expression of MMACHC in fibroblasts. MMACHC is flanked by CCDC163P and PRDX1, which are in the opposite orientation. The epimutation is present in three generations and results from PRDX1 mutations that force antisense transcription of MMACHC thereby possibly generating a H3K36me3 mark. The silencing of PRDX1 transcription leads to partial hypomethylation of the epiallele and restores the expression of MMACHC. This example of epi-cblC demonstrates the need to search for compound epigenetic-genetic heterozygosity in patients with typical disease manifestation and genetic heterozygosity in disease-causing genes located in other gene trios.


Asunto(s)
Proteínas Portadoras/genética , Epistasis Genética , Errores Innatos del Metabolismo/genética , Mutación , Peroxirredoxinas/genética , Vitamina B 12/metabolismo , Alelos , Azacitidina/farmacología , Secuencia de Bases , Inhibidores Enzimáticos/farmacología , Salud de la Familia , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Heterocigoto , Humanos , Masculino , Errores Innatos del Metabolismo/metabolismo , Oxidorreductasas , Linaje , Secuenciación Completa del Genoma
13.
Nat Commun ; 9(1): 554, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29396438

RESUMEN

The original version of this Article contained an error in the title, which was incorrectly given as 'APRDX1 mutant allele causes a MMACHC secondary epimutation in cblC patients'. This has now been corrected in both the PDF and HTML versions of the Article to read 'A PRDX1 mutant allele causes a MMACHC secondary epimutation in cblC patients'.

14.
J Taibah Univ Med Sci ; 12(3): 194-198, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31435239

RESUMEN

In the past several years, rare disease consortia have embarked on the discovery of disease-causing genes for Mendelian diseases using next generation sequencing approaches. Despite the success of these large-scale initiatives, many diseases still have no identified genetic cause. The Rare Disease Collaboration for Autosomal Loci (RaDiCAL) studies the rarest diseases, where occasionally only a single proband is available to identify putative disease-causing genes. This article reviews how "RaDiCAL" addressed some of the challenges in generating informed consent documents for international participants and considers the emerging topic of the "right not to know" in study design.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA