Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
1.
Hum Mol Genet ; 33(12): 1074-1089, 2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38520741

RESUMEN

We have generated using CRISPR/Cas9 technology a partially humanized mouse model of the neurometabolic disease phenylketonuria (PKU), carrying the highly prevalent PAH variant c.1066-11G>A. This variant creates an alternative 3' splice site, leading to the inclusion of 9 nucleotides coding for 3 extra amino acids between Q355 and Y356 of the protein. Homozygous Pah c.1066-11A mice, with a partially humanized intron 10 sequence with the variant, accurately recapitulate the splicing defect and present almost undetectable hepatic PAH activity. They exhibit fur hypopigmentation, lower brain and body weight and reduced survival. Blood and brain phenylalanine levels are elevated, along with decreased tyrosine, tryptophan and monoamine neurotransmitter levels. They present behavioral deficits, mainly hypoactivity and diminished social interaction, locomotor deficiencies and an abnormal hind-limb clasping reflex. Changes in the morphology of glial cells, increased GFAP and Iba1 staining signals and decreased myelinization are observed. Hepatic tissue exhibits nearly absent PAH protein, reduced levels of chaperones DNAJC12 and HSP70 and increased autophagy markers LAMP1 and LC3BII, suggesting possible coaggregation of mutant PAH with chaperones and subsequent autophagy processing. This PKU mouse model with a prevalent human variant represents a useful tool for pathophysiology research and for novel therapies development.


Asunto(s)
Modelos Animales de Enfermedad , Fenilalanina Hidroxilasa , Fenilcetonurias , Animales , Ratones , Fenilcetonurias/genética , Fenilcetonurias/patología , Fenilcetonurias/metabolismo , Humanos , Fenilalanina Hidroxilasa/genética , Fenilalanina Hidroxilasa/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Sistemas CRISPR-Cas , Autofagia/genética , Mutación , Hígado/metabolismo , Hígado/patología
2.
Mol Genet Metab ; 142(3): 108514, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38905920

RESUMEN

Phenylketonuria (PKU) is a genetic disorder caused by variations in the phenylalanine hydroxylase (PAH) gene. Among the 3369 reported PAH variants, 33.7% are missense alterations. Unfortunately, 30% of these missense variants are classified as variants of unknown significance (VUS), posing challenges for genetic risk assessment. In our study, we focused on analyzing 836 missense PAH variants following the American College of Medical Genetics and Genomics/Association for Molecular Pathology (ACMG/AMP) guidelines specified by ClinGen PAH Variant Curation Expert Panel (VCEP) criteria. We utilized and compared variant annotator tools like Franklin and Varsome, conducted 3D structural analysis of PAH, and examined active and regulatory site hotspots. In addition, we assessed potential splicing effect of apparent missense variants. By evaluating phenotype data from 22962 PKU patients, our aim was to reassess the pathogenicity of missense variants. Our comprehensive approach successfully reclassified 309 VUSs out of 836 missense variants as likely pathogenic or pathogenic (37%), upgraded 370 likely pathogenic variants to pathogenic, and reclassified one previously considered likely benign variant as likely pathogenic. Phenotypic information was available for 636 missense variants, with 441 undergoing 3D structural analysis and active site hotspot identification for 180 variants. After our analysis, only 6% of missense variants were classified as VUSs, and three of them (c.23A>C/p.Asn8Thr, c.59_60delinsCC/p.Gln20Pro, and c.278A >T/p.Asn93Ile) may be influenced by abnormal splicing. Moreover, a pathogenic variant (c.168G>T/p.Glu56Asp) was identified to have a risk exceeding 98% for modifications of the consensus splice site, with high scores indicating a donor loss of 0.94. The integration of ACMG/AMP guidelines with in silico structural analysis and phenotypic data significantly reduced the number of missense VUSs, providing a strong basis for genetic counseling and emphasizing the importance of metabolic phenotype information in variant curation. This study also sheds light on the current landscape of PAH variants.


Asunto(s)
Mutación Missense , Fenotipo , Fenilalanina Hidroxilasa , Fenilcetonurias , Humanos , Fenilalanina Hidroxilasa/genética , Fenilalanina Hidroxilasa/química , Fenilcetonurias/genética , Fenilcetonurias/patología , Simulación por Computador
3.
Am J Hum Genet ; 107(2): 234-250, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32668217

RESUMEN

Phenylketonuria (PKU), caused by variants in the phenylalanine hydroxylase (PAH) gene, is the most common autosomal-recessive Mendelian phenotype of amino acid metabolism. We estimated that globally 0.45 million individuals have PKU, with global prevalence 1:23,930 live births (range 1:4,500 [Italy]-1:125,000 [Japan]). Comparing genotypes and metabolic phenotypes from 16,092 affected subjects revealed differences in disease severity in 51 countries from 17 world regions, with the global phenotype distribution of 62% classic PKU, 22% mild PKU, and 16% mild hyperphenylalaninemia. A gradient in genotype and phenotype distribution exists across Europe, from classic PKU in the east to mild PKU in the southwest and mild hyperphenylalaninemia in the south. The c.1241A>G (p.Tyr414Cys)-associated genotype can be traced from Northern to Western Europe, from Sweden via Norway, to Denmark, to the Netherlands. The frequency of classic PKU increases from Europe (56%) via Middle East (71%) to Australia (80%). Of 758 PAH variants, c.1222C>T (p.Arg408Trp) (22.2%), c.1066-11G>A (IVS10-11G>A) (6.4%), and c.782G>A (p.Arg261Gln) (5.5%) were most common and responsible for two prevalent genotypes: p.[Arg408Trp];[Arg408Trp] (11.4%) and c.[1066-11G>A];[1066-11G>A] (2.6%). Most genotypes (73%) were compound heterozygous, 27% were homozygous, and 55% of 3,659 different genotypes occurred in only a single individual. PAH variants were scored using an allelic phenotype value and correlated with pre-treatment blood phenylalanine concentrations (n = 6,115) and tetrahydrobiopterin loading test results (n = 4,381), enabling prediction of both a genotype-based phenotype (88%) and tetrahydrobiopterin responsiveness (83%). This study shows that large genotype databases enable accurate phenotype prediction, allowing appropriate targeting of therapies to optimize clinical outcome.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Fenilcetonurias/epidemiología , Fenilcetonurias/genética , Alelos , Biopterinas/análogos & derivados , Biopterinas/genética , Europa (Continente) , Frecuencia de los Genes/genética , Estudios de Asociación Genética/métodos , Genotipo , Homocigoto , Humanos , Mutación/genética , Fenotipo , Fenilalanina/sangre , Fenilalanina Hidroxilasa/genética , Fenilcetonurias/sangre
4.
Mol Genet Metab ; 136(4): 289-295, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35849888

RESUMEN

RNA-based therapies are a new, rapidly growing class of drugs that until a few years ago were being used mainly in research in rare diseases. However, the clinical efficacy of recently approved oligonucleotide drugs and the massive success of COVID-19 RNA vaccines has boosted the interest in this type of molecules of both scientists and industry, as wells as of the lay public. RNA drugs are easy to design and cost effective, with greatly improved pharmacokinetic properties thanks to progress in oligonucleotide chemistry over the years. Depending on the type of strategy employed, RNA therapies offer the versatility to replace, supplement, correct, suppress, or eliminate the expression of a targeted gene. Currently, there are more than a dozen RNA-based drugs approved for clinical use, including some for specific inborn errors of metabolism (IEM), and many other in different stages of development. New initiatives in n-of-1 RNA drug development offer new hope for patients with rare diseases and/or ultra-rare mutations. RNA-based therapeutics include antisense oligonucleotides, aptamers, small interfering RNAs, small activating RNAs, microRNAs, lncRNAs and messenger RNAs. Further research and collaborations in the fields of chemistry, biology and medicine will help to overcome major challenges in their delivery to target tissues. Herein, we review the mechanism of action of the different therapeutic approaches using RNA drugs, focusing on those approved or in clinical trials to treat IEM.


Asunto(s)
COVID-19 , Errores Innatos del Metabolismo , Humanos , Errores Innatos del Metabolismo/tratamiento farmacológico , Errores Innatos del Metabolismo/terapia , Oligonucleótidos/uso terapéutico , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/uso terapéutico , Enfermedades Raras/tratamiento farmacológico , Enfermedades Raras/genética
5.
PLoS Genet ; 14(4): e1007360, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29684050

RESUMEN

Phenylketonuria (PKU), one of the most common inherited diseases of amino acid metabolism, is caused by mutations in the phenylalanine hydroxylase (PAH) gene. Recently, PAH exon 11 was identified as a vulnerable exon due to a weak 3' splice site, with different exonic mutations affecting exon 11 splicing through disruption of exonic splicing regulatory elements. In this study, we report a novel intron 11 regulatory element, which is involved in exon 11 splicing, as revealed by the investigated pathogenic effect of variants c.1199+17G>A and c.1199+20G>C, identified in PKU patients. Both mutations cause exon 11 skipping in a minigene system. RNA binding assays indicate that binding of U1snRNP70 to this intronic region is disrupted, concomitant with a slightly increased binding of inhibitors hnRNPA1/2. We have investigated the effect of deletions and point mutations, as well as overexpression of adapted U1snRNA to show that this splicing regulatory motif is important for regulation of correct splicing at the natural 5' splice site. The results indicate that U1snRNP binding downstream of the natural 5' splice site determines efficient exon 11 splicing, thus providing a basis for development of therapeutic strategies to correct PAH exon 11 splicing mutations. In this work, we expand the functional effects of non-canonical intronic U1 snRNP binding by showing that it may enhance exon definition and that, consequently, intronic mutations may cause exon skipping by a novel mechanism, where they disrupt stimulatory U1 snRNP binding close to the 5' splice site. Notably, our results provide further understanding of the reported therapeutic effect of exon specific U1 snRNA for splicing mutations in disease.


Asunto(s)
Mutación , Fenilalanina Hidroxilasa/genética , Sitios de Empalme de ARN/genética , Empalme del ARN/genética , Ribonucleoproteína Nuclear Pequeña U1/genética , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Secuencia de Bases , Simulación por Computador , Exones , Células Hep G2 , Humanos , Intrones , Fenilcetonurias/genética , Fenilcetonurias/metabolismo , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo
6.
Hum Mutat ; 41(7): 1329-1338, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32333439

RESUMEN

Biallelic variants of the gene DNAJC12, which encodes a cochaperone, were recently described in patients with hyperphenylalaninemia (HPA). This paper reports the retrospective genetic analysis of a cohort of unsolved cases of HPA. Biallelic variants of DNAJC12 were identified in 20 patients (generally neurologically asymptomatic) previously diagnosed with phenylalanine hydroxylase (PAH) deficiency (phenylketonuria [PKU]). Further, mutations of DNAJC12 were identified in four carriers of a pathogenic variant of PAH. The genetic spectrum of DNAJC12 in the present patients included four new variants, two intronic changes c.298-2A>C and c.502+1G>C, presumably affecting the splicing process, and two exonic changes c.309G>T (p.Trp103Cys) and c.524G>A (p.Trp175Ter), classified as variants of unknown clinical significance (VUS). The variant p.Trp175Ter was detected in 83% of the mutant alleles, with 14 cases homozygous, and was present in 0.3% of a Spanish control population. Functional analysis indicated a significant reduction in PAH and its activity, reduced tyrosine hydroxylase stability, but no effect on tryptophan hydroxylase 2 stability, classifying the two VUS as pathogenic variants. Additionally, the effect of the overexpression of DNAJC12 on some destabilizing PAH mutations was examined and a mutation-specific effect on stabilization was detected suggesting that the proteostasis network could be a genetic modifier of PAH deficiency and a potential target for developing mutation-specific treatments for PKU.


Asunto(s)
Fenilcetonurias/genética , Proteínas Represoras/genética , Adolescente , Adulto , Alelos , Niño , Preescolar , Análisis Mutacional de ADN , Exones , Humanos , Lactante , Recién Nacido , Intrones , Empalme del ARN , Estudios Retrospectivos , España
7.
Mol Genet Metab ; 125(3): 266-275, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30274917

RESUMEN

Propionic acidemia (PA) is caused by mutations in the PCCA and PCCB genes, encoding α and ß subunits, respectively, of the mitochondrial enzyme propionyl-CoA carboxylase (PCC). Up to date, >200 pathogenic mutations have been identified, mostly missense defects. Genetic analysis in PA patients referred to the laboratory for the past 15 years identified 20 novel variants in the PCCA gene and 14 in the PCCB gene. 21 missense variants were predicted as probably disease-causing by different bioinformatics algorithms. Structural analysis in the available 3D model of the PCC enzyme indicated potential instability for most of them. Functional analysis in a eukaryotic system confirmed the pathogenic effect for the missense variants and for one amino acid deletion, as they all exhibited reduced or null PCC activity and protein levels compared to wild-type constructs. PCCB variants p.E168del, p.Q58P and p.I460T resulted in medium-high protein levels and no activity. Variants p.R230C and p.C712S in PCCA, and p.G188A, p.R272W and p.H534R in PCCB retained both partial PCC activity and medium-high protein levels. Available patients-derived fibroblasts carriers of some of these mutations were grown at 28 °C or 37 °C and a slight increase in PCC activity or protein could be detected in some cases at the folding-permissive conditions. Examination of available clinical data showed correlation of the results of the functional analysis with disease severity for most mutations, with some notable exceptions, confirming the notion that the final phenotypic outcome in PA is not easily predicted.


Asunto(s)
Predisposición Genética a la Enfermedad , Metilmalonil-CoA Descarboxilasa/genética , Acidemia Propiónica/genética , Relación Estructura-Actividad , Adolescente , Adulto , Niño , Preescolar , Femenino , Estudios de Asociación Genética , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Metilmalonil-CoA Descarboxilasa/química , Mitocondrias/enzimología , Mitocondrias/genética , Mutación Missense/genética , Tamizaje Neonatal , Acidemia Propiónica/patología , Conformación Proteica , Pliegue de Proteína , Adulto Joven
8.
Biochem Genet ; 56(5): 533-541, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29654578

RESUMEN

Treatment with tetrahydrobiopterin (BH4) is the latest therapeutic option approved for patients with phenylketonuria (PKU)-one of the most frequent inborn metabolic diseases. PKU or phenylalanine hydroxylase (PAH) deficiency is caused by mutations in the PAH gene. Given that some PAH mutations are responsive to BH4 treatment while others are non-responsive, for every novel mutation that is discovered it is essential to confirm its pathogenic effect and to assess its responsiveness to a BH4 treatment in vitro, before the drug is administered to patients. We found a c.676C>A (p.Gln226Lys) mutation in the PAH gene in two unrelated patients with PKU. The corresponding aberrant protein has never been functionally characterized in vitro and its response to BH4 treatment is unknown. Computational analyses proposed that glutamine at position 226 is an important, evolutionary conserved amino acid while the substitution with lysine probably disturbs tertiary protein structure and impacts posttranslational PAH modifications. Using hepatoma cellular model, we demonstrated that the amount of mutant p.Gln226Lys PAH detected by Western blot was only 1.2% in comparison to wild-type PAH. The addition of sepiapterin, intracellular precursor of BH4, did not increase PAH protein yield thus marking p.Gln226Lys as BH4-non-responsive mutation. Therefore, computational, experimental, and clinical data were all in accordance showing that p.Gln226Lys is a severe pathogenic PAH mutation. Its non-responsiveness to BH4 treatment in hepatoma cellular model should be considered when deciding treatment options for PKU patients carrying this mutation. Consequently, our study will facilitate clinical genetic practice, particularly genotype-based stratification of PKU treatment.


Asunto(s)
Biopterinas/análogos & derivados , Fenilalanina Hidroxilasa/genética , Fenilalanina Hidroxilasa/metabolismo , Fenilcetonurias/genética , Biopterinas/farmacología , Línea Celular Tumoral , Glutamina/genética , Humanos , Lisina/genética , Modelos Biológicos , Modelos Moleculares , Fenilalanina Hidroxilasa/química , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/metabolismo , Mutación Puntual , Estructura Terciaria de Proteína , Análisis de Secuencia de ADN
9.
Hum Mutat ; 38(2): 160-168, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27774737

RESUMEN

The congenital disorder of glycosylation (CDG) due to phosphomannomutase 2 deficiency (PMM2-CDG), the most common N-glycosylation disorder, is a multisystem disease for which no effective treatment is available. The recent functional characterization of disease-causing mutations described in patients with PMM2-CDG led to the idea of a therapeutic strategy involving pharmacological chaperones (PC) to rescue PMM2 loss-of-function mutations. The present work describes the high-throughput screening, by differential scanning fluorimetry, of 10,000 low-molecular-weight compounds from a commercial library, to search for possible PCs for the enzyme PMM2. This exercise identified eight compounds that increased the thermal stability of PMM2. Of these, four compounds functioned as potential PCs that significantly increased the stability of several destabilizing and oligomerization mutants and also increased PMM activity in a disease model of cells overexpressing PMM2 mutations. Structural analysis revealed one of these compounds to provide an excellent starting point for chemical optimization since it passed tests based on a number of pharmacochemical quality filters. The present results provide the first proof-of-concept of a possible treatment for PMM2-CDG and describe a promising chemical structure as a starting point for the development of new therapeutic agents for this severe orphan disease.


Asunto(s)
Trastornos Congénitos de Glicosilación/genética , Fosfotransferasas (Fosfomutasas)/genética , Alelos , Trastornos Congénitos de Glicosilación/tratamiento farmacológico , Descubrimiento de Drogas , Activación Enzimática , Fibroblastos/metabolismo , Genotipo , Ensayos Analíticos de Alto Rendimiento , Humanos , Mutación con Pérdida de Función , Terapia Molecular Dirigida , Mutación , Fosfotransferasas (Fosfomutasas)/química , Fosfotransferasas (Fosfomutasas)/aislamiento & purificación , Estabilidad Proteica , Proteolisis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad
10.
Mol Genet Metab ; 122(1-2): 43-50, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28774709

RESUMEN

Oxidative stress contributes to the pathogenesis of propionic acidemia (PA), a life threatening disease caused by the deficiency of propionyl CoA-carboxylase, in the catabolic pathway of branched-chain amino acids, odd-number chain fatty acids and cholesterol. Patients develop multisystemic complications including seizures, extrapyramidal symptoms, basal ganglia deterioration, pancreatitis and cardiomyopathy. The accumulation of toxic metabolites results in mitochondrial dysfunction, increased reactive oxygen species and oxidative damage, all of which have been documented in patients' samples and in a hypomorphic mouse model. Here we set out to investigate whether treatment with a mitochondria-targeted antioxidant, MitoQ, or with the natural polyphenol resveratrol, which is reported to have antioxidant and mitochondrial activation properties, could ameliorate the altered redox status and its functional consequences in the PA mouse model. The results show that oral treatment with MitoQ or resveratrol decreases lipid peroxidation and the expression levels of DNA repair enzyme OGG1 in PA mouse liver, as well as inducing tissue-specific changes in the expression of antioxidant enzymes. Notably, treatment decreased the cardiac hypertrophy marker BNP that is found upregulated in the PA mouse heart. Overall, the results provide in vivo evidence to justify more in depth investigations of antioxidants as adjuvant therapy in PA.


Asunto(s)
Antioxidantes/uso terapéutico , Compuestos Organofosforados/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Acidemia Propiónica/tratamiento farmacológico , Estilbenos/uso terapéutico , Ubiquinona/análogos & derivados , Administración Oral , Aminoácidos de Cadena Ramificada , Animales , Antioxidantes/administración & dosificación , Modelos Animales de Enfermedad , Corazón/efectos de los fármacos , Humanos , Peroxidación de Lípido/efectos de los fármacos , Ratones , Compuestos Organofosforados/administración & dosificación , Acidemia Propiónica/fisiopatología , Resveratrol , Estilbenos/administración & dosificación , Ubiquinona/administración & dosificación , Ubiquinona/uso terapéutico
11.
J Inherit Metab Dis ; 40(4): 471-480, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28229250

RESUMEN

MicroRNAs (miRNAs) are short, noncoding RNAs that regulate gene expression posttranscriptionally by base pairing with target messenger RNAs (mRNAs). They are estimated to target ∼60% of all human protein-coding genes and are involved in regulating key physiological processes and intracellular signaling pathways. They also exhibit tissue specificity, and their dysregulation is linked to the progression of pathology. Identifying disease associated miRNAs and their respective targets provides novel molecular insight into disease, enabling the design of new therapeutic strategies. Notably, miRNAs are present in stable form in biological fluids, making them amenable to routine clinical processing and analysis, which has paved the way for their use as novel biomarkers of disease and response to therapy. One of the most relevant findings in miRNA research concerns the therapeutic modulation of specific miRNA levels in vitro and in vivo, which has led to miRNA-based drugs entering clinical trials. Most studies relative to miRNA profiling, association with pathology, and therapeutical modulation have been conducted for cancer, cardiovascular and neurodegenerative diseases. However, for different monogenic diseases, including inborn errors of metabolism (IEM), research contributing to alterations to physiopathology caused by miRNAs is steadily increasing. Herein, we review the biogenesis pathway and mode of miRNA action, their known roles in disease states, and use of circulating miRNAs as biomarkers, describing the available research tools for basic and clinical studies. In addition, we summarize recent literature on miRNA studies in inherited metabolic diseases.


Asunto(s)
Biomarcadores de Tumor/genética , Enfermedades Metabólicas/genética , Errores Innatos del Metabolismo/genética , MicroARNs/genética , Neoplasias/genética , Animales , Ensayos Clínicos como Asunto , Progresión de la Enfermedad , Humanos , MicroARNs/sangre , Enfermedades Neurodegenerativas , Resultado del Tratamiento
12.
Biochim Biophys Acta ; 1852(12): 2712-21, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26407519

RESUMEN

Mucopolysaccharidosis II is a lysosomal storage disorder caused by mutations in the IDS gene, including exonic alterations associated with aberrant splicing. In the present work, cell-based splicing assays were performed to study the effects of two splicing mutations in exon 3 of IDS, i.e., c.241C>T and c.257C>T, whose presence activates a cryptic splice site in exon 3 and one in exon 8, i.e., c.1122C>T that despite being a synonymous mutation is responsible for the creation of a new splice site in exon 8 leading to a transcript shorter than usual. Mutant minigene analysis and overexpression assays revealed that SRSF2 and hnRNP E1 might be involved in the use and repression of the constitutive 3' splice site of exon 3 respectively. For the c.1122C>T the use of antisense therapy to correct the splicing defect was explored, but transfection of patient fibroblasts with antisense morpholino oligonucleotides (n=3) and a locked nucleic acid failed to abolish the abnormal transcript; indeed, it resulted in the appearance of yet another aberrant splicing product. Interestingly, the oligonucleotides transfection in control fibroblasts led to the appearance of the aberrant transcript observed in patients' cells after treatment, which shows that the oligonucleotides are masking an important cis-acting element for 5' splice site regulation of exon 8. These results highlight the importance of functional studies for understanding the pathogenic consequences of mis-splicing and highlight the difficulty in developing antisense therapies involving gene regions under complex splicing regulation.

13.
Genet Med ; 18(10): 1037-43, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26913919

RESUMEN

PURPOSE: Glycogen storage disease (GSD) is an umbrella term for a group of genetic disorders that involve the abnormal metabolism of glycogen; to date, 23 types of GSD have been identified. The nonspecific clinical presentation of GSD and the lack of specific biomarkers mean that Sanger sequencing is now widely relied on for making a diagnosis. However, this gene-by-gene sequencing technique is both laborious and costly, which is a consequence of the number of genes to be sequenced and the large size of some genes. METHODS: This work reports the use of massive parallel sequencing to diagnose patients at our laboratory in Spain using either a customized gene panel (targeted exome sequencing) or the Illumina Clinical-Exome TruSight One Gene Panel (clinical exome sequencing (CES)). Sequence variants were matched against biochemical and clinical hallmarks. RESULTS: Pathogenic mutations were detected in 23 patients. Twenty-two mutations were recognized (mostly loss-of-function mutations), including 11 that were novel in GSD-associated genes. In addition, CES detected five patients with mutations in ALDOB, LIPA, NKX2-5, CPT2, or ANO5. Although these genes are not involved in GSD, they are associated with overlapping phenotypic characteristics such as hepatic, muscular, and cardiac dysfunction. CONCLUSIONS: These results show that next-generation sequencing, in combination with the detection of biochemical and clinical hallmarks, provides an accurate, high-throughput means of making genetic diagnoses of GSD and related diseases.Genet Med 18 10, 1037-1043.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno/diagnóstico , Enfermedad del Almacenamiento de Glucógeno/genética , Glucógeno/genética , Patología Molecular , Adolescente , Adulto , Anoctaminas , Niño , Preescolar , Canales de Cloruro/genética , Exoma/genética , Femenino , Fructosa-Bifosfato Aldolasa/genética , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno/fisiopatología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Masculino , Mutación , Proteínas Nucleares/genética , Esterol Esterasa/genética , Factor Nuclear Tiroideo 1 , Factores de Transcripción/genética , Adulto Joven
14.
J Hum Genet ; 61(8): 731-44, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27121329

RESUMEN

Phenylketonuria (PKU), the most common inborn error of amino acid metabolism, is caused by mutations in the phenylalanine-4-hydroxylase (PAH) gene. This study aimed to assess the genotype-phenotype correlation in the PKU Spanish population and the usefulness in establishing genotype-based predictions of BH4 responsiveness in our population. It involved the molecular characterization of 411 Spanish PKU patients: mild hyperphenylalaninemia non-treated (mild HPA-NT) (34%), mild HPA (8.8%), mild-moderate (20.7%) and classic (36.5%) PKU. BH4 responsiveness was evaluated using a 6R-BH4 loading test. We assessed genotype-phenotype associations and genotype-BH4 responsiveness in our population according to literature and classification of the mutations. The mutational spectrum analysis showed 116 distinct mutations, most missense (70.7%) and located in the catalytic domain (62.9%). The most prevalent mutations were c.1066-11G>A (9.7%), p.Val388Met (6.6%) and p.Arg261Gln (6.3%). Three novel mutations (c.61-13del9, p.Ile283Val and p.Gly148Val) were reported. Although good genotype-phenotype correlation was observed, there was no exact correlation for some genotypes. Among the patients monitored for the 6R-BH4 loading test: 102 were responders (87, carried either one or two BH4-responsive alleles) and 194 non-responders (50, had two non-responsive mutations). More discrepancies were observed in non-responders. Our data reveal a great genetic heterogeneity in our population. Genotype is quite a good predictor of phenotype and BH4 responsiveness, which is relevant for patient management, treatment and follow-up.


Asunto(s)
Estudios de Asociación Genética , Genotipo , Mutación , Fenotipo , Fenilalanina Hidroxilasa/genética , Fenilcetonurias/epidemiología , Fenilcetonurias/genética , Alelos , Terapia de Reemplazo Enzimático , Frecuencia de los Genes , Heterogeneidad Genética , Humanos , Epidemiología Molecular , Fenilalanina Hidroxilasa/metabolismo , Fenilcetonurias/diagnóstico , Fenilcetonurias/terapia , España/epidemiología
15.
Hum Mutat ; 36(9): 851-60, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26014514

RESUMEN

Congenital disorder of glycosylation type Ia (PMM2-CDG), the most common form of CDG, is caused by mutations in the PMM2 gene that reduce phosphomannomutase 2 (PMM2) activity. No curative treatment is available. The present work describes the functional analysis of nine human PMM2 mutant proteins frequently found in PMM2-CDG patients and also two murine Pmm2 mutations carried by the unique PMM2-CDG mouse model described to overcome embryonic lethality. The effects of the mutations on PMM2/Pmm2 stability, oligomerization, and enzyme activity were explored in an optimized bacterial system. The mutant proteins were associated with an enzymatic activity of up to 47.3% as compared with wild type (WT). Stability analysis performed using differential scanning fluorimetry and a bacterial transcription-translation-coupled system allowed the identification of several destabilizing mutations (p.V44A, p.D65Y, p.R123Q, p.R141H, p.R162W, p.F207S, p.T237M, p.C241S). Exclusion chromatography identified one mutation, p.P113L, that affected dimer interaction. Expression analysis of the p.V44A, p.D65Y, p.R162W, and p.T237M mutations in a eukaryotic expression system under permissive folding conditions showed the possibility of recovering their associated PMM2 activity. Together, the results suggest that some loss-of-function mutations detected in PMM2-CDG patients could be destabilizing, and therefore PMM2 activity could be, in certain cases, rescuable via the use of synergetic proteostasis modulators and/or chaperones.


Asunto(s)
Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/metabolismo , Mutación , Fosfotransferasas (Fosfomutasas)/genética , Fosfotransferasas (Fosfomutasas)/metabolismo , Pliegue de Proteína , Animales , Activación Enzimática/genética , Estabilidad de Enzimas/genética , Fibroblastos , Expresión Génica , Humanos , Ratones , Fosfotransferasas (Fosfomutasas)/química , Multimerización de Proteína , Proteolisis , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
16.
Hum Mol Genet ; 22(18): 3680-9, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23674520

RESUMEN

Methylmalonic aciduria (MMA) cblB type is caused by mutations in the MMAB gene. This encodes the enzyme ATP:cob(I)alamin adenosyltransferase (ATR), which converts reduced cob(I)alamin to an active adenosylcobalamin cofactor. We recently reported the presence of destabilizing pathogenic mutations that retain some residual ATR activity. The aim of the present study was to seek pharmacological chaperones as a tailored therapy for stabilizing the ATR protein. High-throughput ligand screening of over 2000 compounds was performed; six were found to enhance the thermal stability of purified recombinant ATR. Further studies using a well-established bacterial system in which the recombinant ATR protein was expressed in the presence of these six compounds, showed them all to increase the stability of the wild-type ATR and the p.Ile96Thr mutant proteins. Compound V (N-{[(4-chlorophenyl)carbamothioyl]amino}-2-phenylacetamide) significantly increased this stability and did not act as an inhibitor of the purified protein. Importantly, compound V increased the activity of ATR in patient-derived fibroblasts harboring the destabilizing p.Ile96Thr mutation in a hemizygous state to within control range. When cobalamin was coadministrated with compound V, mutant ATR activity further improved. Oral administration of low doses of compound V to C57BL/6J mice for 12 days, led to increase in steady-state levels of ATR protein in liver and brain (disease-relevant organs). These results hold promise for the clinical use of pharmacological chaperones in MMA cblB type patients harboring chaperone-responsive mutations.


Asunto(s)
Transferasas Alquil y Aril/genética , Errores Innatos del Metabolismo de los Aminoácidos/tratamiento farmacológico , Bencenoacetamidas/química , Bencenoacetamidas/farmacología , Tiourea/análogos & derivados , Transferasas Alquil y Aril/química , Transferasas Alquil y Aril/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Animales , Bencenoacetamidas/administración & dosificación , Sitios de Unión , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Estabilidad de Enzimas , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Hígado/efectos de los fármacos , Hígado/enzimología , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Tiourea/administración & dosificación , Tiourea/química , Tiourea/farmacología , Vitamina B 12/administración & dosificación , Vitamina B 12/farmacología
17.
Biochem Biophys Res Commun ; 452(3): 457-61, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25159844

RESUMEN

Propionic acidemia (PA), caused by a deficiency of the mitochondrial biotin dependent enzyme propionyl-CoA carboxylase (PCC) is one of the most frequent organic acidurias in humans. Most PA patients present in the neonatal period with metabolic acidosis and hyperammonemia, developing different neurological symptoms, movement disorders and cardiac complications. There is strong evidence indicating that oxidative damage could be a pathogenic factor in neurodegenerative, mitochondrial and metabolic diseases. Recently, we identified an increase in ROS levels in PA patients-derived fibroblasts. Here, we analyze the capability of seven antioxidants to scavenge ROS production in PA patients' cells. Tiron, trolox, resveratrol and MitoQ significantly reduced ROS content in patients and controls' fibroblasts. In addition, changes in the expression of two antioxidant enzymes, superoxide dismutase and glutathione peroxidase, were observed in PA patients-derived fibroblasts after tiron and resveratrol treatment. Our results in PA cellular models establish the proof of concept of the potential of antioxidants as an adjuvant therapy for PA and pave the way for future assessment of antioxidant strategies in the murine model of PA.


Asunto(s)
Antioxidantes/farmacología , Fibroblastos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Sal Disódica del Ácido 1,2-Dihidroxibenceno-3,5-Disulfónico/farmacología , Cromanos/farmacología , Fibroblastos/metabolismo , Fibroblastos/patología , Expresión Génica/efectos de los fármacos , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Humanos , Metilmalonil-CoA Descarboxilasa/genética , Mitocondrias/metabolismo , Mutación , Compuestos Organofosforados/farmacología , Cultivo Primario de Células , Acidemia Propiónica/genética , Acidemia Propiónica/metabolismo , Acidemia Propiónica/patología , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Resveratrol , Estilbenos/farmacología , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/farmacología , Glutatión Peroxidasa GPX1
18.
Gene ; 893: 147902, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-37839763

RESUMEN

Next-generation sequencing has improved the diagnosis of inborn errors of metabolism, allowing rapid confirmation of cases detected by clinical/biochemical studies or newborn screening. The challenge, however, remains for establishing the pathogenicity of the identified variants, especially for novel missense changes or small in-frame deletions. In this work we report a propionic acidemia patient exhibiting a severe neonatal form with coma and hyperammonaemia. Genetic analysis identified the previously described pathogenic PCCB variant p.R512C in the maternal allele and two novel PCCB variants in cis in the paternal allele, p.G246del and p.S322F. Expression analysis in a eukaryotic system confirmed the deleterious effect of the novel missense variant and of the one amino acid deletion, as they both exhibited reduced protein levels and reduced or null PCC activity compared to the wild-type construct. Accordingly, the double mutant resulted in no residual activity. This study increases the knowledge of the genotype-phenotype correlations in the rare disease propionic acidemia and highlights the necessity of functional analysis of novel variants to understand their contribution to disease severity and to accurately classify their pathogenic status. In conclusion, two novel PCCB pathogenic variants have been identified, expanding the current mutational spectrum of propionic acidemia.


Asunto(s)
Liasas de Carbono-Carbono , Acidemia Propiónica , Humanos , Recién Nacido , Liasas de Carbono-Carbono/genética , Mutación Missense , Acidemia Propiónica/genética , Eliminación de Secuencia
19.
Nucleic Acid Ther ; 34(3): 134-142, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38591802

RESUMEN

The PAH gene encodes the hepatic enzyme phenylalanine hydroxylase (PAH), and its deficiency, known as phenylketonuria (PKU), leads to neurotoxic high levels of phenylalanine. PAH exon 11 is weakly defined, and several missense and intronic variants identified in patients affect the splicing process. Recently, we identified a novel intron 11 splicing regulatory element where U1snRNP binds, participating in exon 11 definition. In this work, we describe the implementation of an antisense strategy targeting intron 11 sequences to correct the effect of PAH mis-splicing variants. We used an in vitro assay with minigenes and identified splice-switching antisense oligonucleotides (SSOs) that correct the exon skipping defect of PAH variants c.1199+17G>A, c.1199+20G>C, c.1144T>C, and c.1066-3C>T. To examine the functional rescue induced by the SSOs, we generated a hepatoma cell model with variant c.1199+17G>A using CRISPR/Cas9. The edited cell line reproduces the exon 11 skipping pattern observed from minigenes, leading to reduced PAH protein levels and activity. SSO transfection results in an increase in exon 11 inclusion and corrects PAH deficiency. Our results provide proof of concept of the potential therapeutic use of a single SSO for different exonic and intronic splicing variants causing PAH exon 11 skipping in PKU.


Asunto(s)
Exones , Intrones , Oligonucleótidos Antisentido , Fenilalanina Hidroxilasa , Fenilcetonurias , Empalme del ARN , Humanos , Fenilcetonurias/genética , Fenilcetonurias/terapia , Fenilcetonurias/patología , Fenilalanina Hidroxilasa/genética , Fenilalanina Hidroxilasa/metabolismo , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/uso terapéutico , Oligonucleótidos Antisentido/farmacología , Exones/genética , Empalme del ARN/genética , Intrones/genética , Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Empalme Alternativo/genética
20.
Hum Mutat ; 34(2): 355-62, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23086801

RESUMEN

This article describes a hitherto unreported involvement of the phosphatase PP2Cm, a recently described member of the branched-chain α-keto acid dehydrogenase (BCKDH) complex, in maple syrup urine disease (MSUD). The disease-causing mutation was identified in a patient with a mild variant phenotype, involving a gene not previously associated with MSUD. SNP array-based genotyping showed a copy-neutral homozygous pattern for chromosome 4 compatible with uniparental isodisomy. Mutation analysis of the candidate gene, PPM1K, revealed a homozygous c.417_418delTA change predicted to result in a truncated, unstable protein. No PP2Cm mutant protein was detected in immunocytochemical or Western blot expression analyses. The transient expression of wild-type PPM1K in PP2Cm-deficient fibroblasts recovered 35% of normal BCKDH activity. As PP2Cm has been described essential for cell survival, apoptosis and metabolism, the impact of its deficiency on specific metabolic stress variables was evaluated in PP2Cm-deficient fibroblasts. Increases were seen in ROS levels along with the activation of specific stress-signaling MAP kinases. Similar to that described for the pyruvate dehydrogenase complex, a defect in the regulation of BCKDH caused the aberrant metabolism of its substrate, contributing to the patient's MSUD phenotype--and perhaps others.


Asunto(s)
3-Metil-2-Oxobutanoato Deshidrogenasa (Lipoamida)/genética , Enfermedad de la Orina de Jarabe de Arce/genética , Fosfoproteínas Fosfatasas/genética , Apoptosis , Western Blotting , Supervivencia Celular , Análisis Mutacional de ADN , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Frecuencia de los Genes , Genotipo , Humanos , Lactante , Isoleucina/sangre , Leucina/sangre , Enfermedad de la Orina de Jarabe de Arce/diagnóstico , Microscopía Fluorescente , Mutación , Fenotipo , Proteína Fosfatasa 2C , Complejo Piruvato Deshidrogenasa/genética , Especies Reactivas de Oxígeno , Análisis de Secuencia de ADN , Piel/citología , Piel/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda