Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Int J Mol Sci ; 23(3)2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35163085

RESUMEN

The human albumin gene, the most abundant serum protein, is located in the long arm of chromosome 4, near the centromere, position 4q11-3. It is divided by 14 intervening introns into 15 exons, the last of which is untranslated. To date, 74 nucleotide substitutions (mainly missense) have been reported, determining the circulating variants of albumin or pre-albumin. In a heterozygous state, this condition is known as alloalbuminaemia or bisalbuminaemia (OMIM # 103600). The genetic variants are not associated with disease, neither in the heterozygous nor in the homozygous form. Only the variants resulting in familial dysalbuminaemic hyperthyroxinaemia and hypertriiodothyroninaemia are of clinical relevance because affected individuals are at risk of inappropriate treatment or may have adverse drug effects. In 28 other cases, the pathogenic variants (mainly affecting splicing, nonsense, and deletions), mostly in the homozygous form, cause a premature stop in the synthesis of the protein and lead to the condition known as congenital analbuminaemia. In this review, we will summarize the current knowledge of genetic and molecular aspects, functional consequences and potential therapeutic uses of the variants. We will also discuss the molecular defects resulting in congenital analbuminaemia, as well as the biochemical and clinical features of this rare condition.


Asunto(s)
Homocigoto , Hipoalbuminemia/genética , Hipoalbuminemia/patología , Intrones , Mutación , Albúmina Sérica Humana/genética , Exones , Humanos , Hipoalbuminemia/metabolismo , Albúmina Sérica Humana/metabolismo
2.
Clin Immunol ; 229: 108776, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34118401

RESUMEN

Early-onset ataxia with ocular motor apraxia and hypoalbuminemia (EAOH) is a neurodegenerative disorder caused by mutation in the aprataxin (APTX)-coding gene APTX, which is involved in DNA single-strand break repair (SSBR). The neurological abnormalities associated with EAOH are similar to those observed in patients with ataxia-telangiectasia. However, the immunological abnormalities in patients with EAOH have not been described. In this study, we report that EAOH patients have immunological abnormalities, including lymphopenia; decreased levels of CD4+ T-cells, CD8+ T-cells, and B-cells; hypogammaglobulinemia; low T-cell recombination excision circles and kappa-deleting element recombination circles; and oligoclonality of T-cell receptor ß-chain variable repertoire. These immunological abnormalities vary among the EAOH patients. Additionally, mild radiosensitivity in the lymphocytes obtained from the patients with EAOH was demonstrated. These findings suggested that the immunological abnormalities and mild radiosensitivity evident in patients with EAOH could be probably caused by the DNA repair defects.


Asunto(s)
Apraxias/inmunología , Ataxia Cerebelosa/congénito , Hipoalbuminemia/inmunología , Adolescente , Adulto , Apraxias/genética , Apraxias/metabolismo , Estudios de Casos y Controles , Ataxia Cerebelosa/genética , Ataxia Cerebelosa/inmunología , Ataxia Cerebelosa/metabolismo , Niño , Roturas del ADN de Cadena Simple , Reparación del ADN/genética , Reparación del ADN/efectos de la radiación , Proteínas de Unión al ADN/genética , Femenino , Genes Codificadores de los Receptores de Linfocitos T , Variación Genética , Humanos , Hipoalbuminemia/genética , Hipoalbuminemia/metabolismo , Masculino , Persona de Mediana Edad , Mutación , Proteínas Nucleares/genética , Tolerancia a Radiación/genética , Tolerancia a Radiación/inmunología , Linfocitos T/inmunología , Adulto Joven
3.
BMC Med Genet ; 21(1): 239, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33261563

RESUMEN

BACKGROUND: Early-onset chronic diarrhoea often indicates a congenital disorder. Mutation in diacylglycerol o-acyltransferase 1 (DGAT1) has recently been linked to early-onset chronic diarrhoea. To date, only a few cases of DGAT1 deficiency have been reported. Diarrhoea in those cases was severe and developed in the neonatal period or within 2 months after birth. CASE PRESENTATION: Here, we report a female patient with DGAT1 mutations with delayed-onset chronic diarrhoea. The patient had vomiting, hypoalbuminemia, hypertriglyceridemia, and failure to thrive at early infancy. Her intractable chronic diarrhoea occurred until she was 8 months of age. A compound heterozygous DGAT1 mutation was found in the patient, which was first found in the Chinese population. Her symptoms and nutrition status improved after nutritional therapy, including a fat restriction diet. CONCLUSIONS: This case expanded our knowledge of the clinical features of patients with DGAT1 mutations. Intractable diarrhoea with delayed onset could also be a congenital disorder.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/genética , Diarrea/genética , Insuficiencia de Crecimiento/genética , Hipertrigliceridemia/genética , Hipoalbuminemia/genética , Mutación , Vómitos/genética , Edad de Inicio , Secuencia de Bases , Diacilglicerol O-Acetiltransferasa/deficiencia , Diarrea/dietoterapia , Diarrea/metabolismo , Diarrea/fisiopatología , Dieta con Restricción de Grasas , Insuficiencia de Crecimiento/dietoterapia , Insuficiencia de Crecimiento/metabolismo , Insuficiencia de Crecimiento/fisiopatología , Femenino , Expresión Génica , Heterocigoto , Humanos , Hipertrigliceridemia/dietoterapia , Hipertrigliceridemia/metabolismo , Hipertrigliceridemia/fisiopatología , Hipoalbuminemia/dietoterapia , Hipoalbuminemia/metabolismo , Hipoalbuminemia/fisiopatología , Lactante , Índice de Severidad de la Enfermedad , Vómitos/dietoterapia , Vómitos/metabolismo , Vómitos/fisiopatología
4.
Nature ; 506(7486): 111-5, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24362567

RESUMEN

Faithful maintenance and propagation of eukaryotic genomes is ensured by three-step DNA ligation reactions used by ATP-dependent DNA ligases. Paradoxically, when DNA ligases encounter nicked DNA structures with abnormal DNA termini, DNA ligase catalytic activity can generate and/or exacerbate DNA damage through abortive ligation that produces chemically adducted, toxic 5'-adenylated (5'-AMP) DNA lesions. Aprataxin (APTX) reverses DNA adenylation but the context for deadenylation repair is unclear. Here we examine the importance of APTX to RNase-H2-dependent excision repair (RER) of a lesion that is very frequently introduced into DNA, a ribonucleotide. We show that ligases generate adenylated 5' ends containing a ribose characteristic of RNase H2 incision. APTX efficiently repairs adenylated RNA-DNA, and acting in an RNA-DNA damage response (RDDR), promotes cellular survival and prevents S-phase checkpoint activation in budding yeast undergoing RER. Structure-function studies of human APTX-RNA-DNA-AMP-Zn complexes define a mechanism for detecting and reversing adenylation at RNA-DNA junctions. This involves A-form RNA binding, proper protein folding and conformational changes, all of which are affected by heritable APTX mutations in ataxia with oculomotor apraxia 1. Together, these results indicate that accumulation of adenylated RNA-DNA may contribute to neurological disease.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Genoma Humano/genética , Proteínas Nucleares/metabolismo , ARN/metabolismo , Adenosina Monofosfato/metabolismo , Apraxias/genética , Ataxia Telangiectasia/genética , Supervivencia Celular , Ataxia Cerebelosa/congénito , ADN/química , Reparación del ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Exodesoxirribonucleasas/química , Exodesoxirribonucleasas/metabolismo , Humanos , Hipoalbuminemia/genética , Modelos Moleculares , Mutación/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Conformación de Ácido Nucleico , Conformación Proteica , Pliegue de Proteína , ARN/química , Ribonucleasa H/metabolismo , Puntos de Control de la Fase S del Ciclo Celular , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/metabolismo , Relación Estructura-Actividad , Zinc/metabolismo
5.
Gastroenterology ; 155(1): 130-143.e15, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29604290

RESUMEN

BACKGROUND & AIMS: Congenital diarrheal disorders are rare inherited intestinal disorders characterized by intractable, sometimes life-threatening, diarrhea and nutrient malabsorption; some have been associated with mutations in diacylglycerol-acyltransferase 1 (DGAT1), which catalyzes formation of triacylglycerol from diacylglycerol and acyl-CoA. We investigated the mechanisms by which DGAT1 deficiency contributes to intestinal failure using patient-derived organoids. METHODS: We collected blood samples from 10 patients, from 6 unrelated pedigrees, who presented with early-onset severe diarrhea and/or vomiting, hypoalbuminemia, and/or (fatal) protein-losing enteropathy with intestinal failure; we performed next-generation sequencing analysis of DNA from 8 patients. Organoids were generated from duodenal biopsies from 3 patients and 3 healthy individuals (controls). Caco-2 cells and patient-derived dermal fibroblasts were transfected or transduced with vectors that express full-length or mutant forms of DGAT1 or full-length DGAT2. We performed CRISPR/Cas9-guided disruption of DGAT1 in control intestinal organoids. Cells and organoids were analyzed by immunoblot, immunofluorescence, flow cytometry, chromatography, quantitative real-time polymerase chain reaction, and for the activity of caspases 3 and 7. RESULTS: In the 10 patients, we identified 5 bi-allelic loss-of-function mutations in DGAT1. In patient-derived fibroblasts and organoids, the mutations reduced expression of DGAT1 protein and altered triacylglycerol metabolism, resulting in decreased lipid droplet formation after oleic acid addition. Expression of full-length DGAT2 in patient-derived fibroblasts restored formation of lipid droplets. Organoids derived from patients with DGAT1 mutations were more susceptible to lipid-induced cell death than control organoids. CONCLUSIONS: We identified a large cohort of patients with congenital diarrheal disorders with mutations in DGAT1 that reduced expression of its product; dermal fibroblasts and intestinal organoids derived from these patients had altered lipid metabolism and were susceptible to lipid-induced cell death. Expression of full-length wildtype DGAT1 or DGAT2 restored normal lipid metabolism in these cells. These findings indicate the importance of DGAT1 in fat metabolism and lipotoxicity in the intestinal epithelium. A fat-free diet might serve as the first line of therapy for patients with reduced DGAT1 expression. It is important to identify genetic variants associated with congenital diarrheal disorders for proper diagnosis and selection of treatment strategies.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/genética , Duodeno/metabolismo , Fibroblastos/metabolismo , Hipoalbuminemia/genética , Trastornos del Metabolismo de los Lípidos/genética , Organoides/metabolismo , Enteropatías Perdedoras de Proteínas/genética , Células CACO-2 , Estudios de Casos y Controles , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Niño , Preescolar , Consanguinidad , Dermis/citología , Diacilglicerol O-Acetiltransferasa/deficiencia , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Países Bajos , Forboles , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN , Turquía
6.
BMC Med Genet ; 16: 28, 2015 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-25925991

RESUMEN

BACKGROUND: Mutations in CCBE1 have been found to be responsible for a subset of families with autosomal recessive Hennekam syndrome. Hennekam syndrome is defined as the combination of generalized lymphatic dysplasia (ie. lymphedema and lymphangiectasia), variable intellectual disability and characteristic dysmorphic features. The patient we describe here has a lymphatic dysplasia without intellectual disability or dysmorphism caused by mutation in CCBE1, highlighting the phenotypic variability that can be seen with abnormalities in this gene. CASE PRESENTATION: Our patient is a 5 week old child of Pakistani descent who presented to our center with generalized edema, ascites, and hypoalbuminemia. She was diagnosed with a protein losing enteropathy secondary to segmental primary intestinal lymphangiectasia. As the generalized edema resolved, it became clear that she had mild persistent lymphedema in her hands and feet. No other abnormalities were noted on examination and development was unremarkable at 27 months of age. Given the suspected genetic etiology and the consanguinity in the family, we used a combination of SNP genotyping and exome sequencing to identify the underlying cause of her disease. We identified several large stretches of homozygosity in the patient that allowed us to sort the variants found in the patient's exome to identify p.C98W in CCBE1 as the likely pathogenic variant. CONCLUSIONS: CCBE1 mutation analysis should be considered in all patients with unexplained lymphatic dysplasia even without the other features of classic Hennekam syndrome.


Asunto(s)
Proteínas de Unión al Calcio/genética , Anomalías Craneofaciales/genética , Enfermedades de los Genitales Masculinos/genética , Linfangiectasia Intestinal/genética , Sistema Linfático/embriología , Linfedema/genética , Proteínas Supresoras de Tumor/genética , Consanguinidad , Análisis Mutacional de ADN , Femenino , Genotipo , Humanos , Hipoalbuminemia/genética , Lactante , Pakistán , Polidactilia/genética , Polimorfismo de Nucleótido Simple , Enteropatías Perdedoras de Proteínas/genética
7.
J Chemother ; 36(3): 179-189, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-37599449

RESUMEN

Voriconazole (VRC) displays highly variable pharmacokinetics impacting treatment efficacy and safety. To provide evidence for optimizing VRC therapy regimens, the authors set out to determine the factors impacting VRC steady-state trough concentration (Cmin) in patients with various albumin (Alb) level. A total of 275 blood samples of 120 patients and their clinical characteristics and genotypes of CYP2C19, CYP3A4, CYP3A5, CYP2C9, FMO3, ABCB1, POR, NR1I2 and NR1I3 were included in this study. Results of multivariate linear regression analysis demonstrated that C-reactive protein (CRP) and total bilirubin (T-Bil) were predictors of the VRC Cmin adjusted for dose in patients with hypoalbuminemia (Alb < 35 g/L) (R2 = 0.16, P < 0.001). Additionally, in patients with normal albumin level (Alb ≥ 35 g/L), it resulted in a significant model containing factors of the poor metabolizer (PM) CYP2C19 genotype and CRP level (R2 = 0.26, P < 0.001). Therefore, CRP and T-Bil levels ought to receive greater consideration than genetic factors in patients with hypoalbuminemia.


Asunto(s)
Antifúngicos , Hipoalbuminemia , Humanos , Voriconazol/efectos adversos , Antifúngicos/efectos adversos , Citocromo P-450 CYP2C19/genética , Variantes Farmacogenómicas , Hipoalbuminemia/genética , Hipoalbuminemia/inducido químicamente , Hipoalbuminemia/tratamiento farmacológico , Proteína C-Reactiva , Genotipo , China
8.
Ann Biol Clin (Paris) ; 81(2): 204-209, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37184254

RESUMEN

Congenital analbuminemia (CAA) is a very rare genetic disorder characterized by a significant reduced or even complete absence of human serum albumin. Our data describe the clinical features and laboratory results of a case confirmed by mutation analysis of the albumin gene in a 35-year-old man presenting recurrent acute coronary syndrome. To the best of our knowledge, only two cases of coronary artery disease have been reported worldwide without recurrence. Our findings contribute to shed light on the clinical characteristics and biochemical parameters of this disease and confirm that cardiovascular complications must be taken seriously in this pathology. Mutational screening disclosed two novel compound heterozygous nucleotide variations located in intron 12 and in 3'UTR. The prediction of the functional and structural impact of the reported variations using different bioinformatics tools demonstrates that these genetics variations affect RNA transcription and mRNA folding.


Asunto(s)
Trombosis Coronaria , Hipoalbuminemia , Masculino , Humanos , Adulto Joven , Adulto , Albúmina Sérica , Nucleótidos , Trombosis Coronaria/complicaciones , Albúmina Sérica Humana/genética , Hipoalbuminemia/complicaciones , Hipoalbuminemia/diagnóstico , Hipoalbuminemia/genética , Mutación
9.
Biomolecules ; 13(3)2023 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-36979342

RESUMEN

Albumin is a highly abundant plasma protein with multiple functions, including the balance of fluid between body compartments and fatty acid trafficking. Humans with congenital analbuminemia (CAA) do not express albumin due to homozygosity for albumin gene mutation. Lessons about physiological control could be learned from CAA. Remarkably, these patients exhibit an apparently normal lifespan, without substantial impairments in physical functionality. There was speculation that tolerance to albumin deficiency would be characterized by significant upregulation of other plasma proteins to compensate for analbuminemia. It is unknown but possible that changes in plasma protein expression observed in CAA are required for the well-documented survival and general wellness. A systematic review of published case reports was performed to assess plasma protein pattern remodeling in CAA patients who were free of other illnesses that would confound interpretation. From a literature search in Pubmed, Scopus, and Purdue Libraries (updated October 2022), concentration of individual plasma proteins and protein classes were assessed. Total plasma protein concentration was below the reference range in the vast majority of CAA patients in the analysis, as upregulation of other proteins was not sufficient to prevent the decline of total plasma protein when albumin was absent. Nonetheless, an impressive level of evidence in the literature indicated upregulated plasma levels of multiple globulin classes and various specific proteins which may have metabolic functions in common with albumin. The potential role of this altered plasma protein expression pattern in CAA is discussed, and the findings may have implications for other populations with hypoalbuminemia.


Asunto(s)
Hipoalbuminemia , Humanos , Hipoalbuminemia/genética , Hipoalbuminemia/congénito , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Albúminas , Mutación , Plasma/metabolismo
10.
Hum Mutat ; 33(2): 351-4, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22065524

RESUMEN

Autosomal recessive ataxias are heterogeneous group of disorders characterized by cerebellar atrophy and peripheral sensorimotor neuropathy. Molecular characterization of this group of disorders identified a number of genes contributing to these overlapping phenotypes. Ataxia with oculomotor apraxia type 2 (AOA2) is an autosomal recessive form of ataxia caused by mutations in the SETX gene. We report on a consanguineous family with autosomal recessive inheritance and clinical characteristics of AOA2, and no mutations in the SETX gene. We mapped the AOA locus in this family to chromosome 17p12-p13. Sequencing of all genes in the refined region identified a homozygous missense mutation in PIK3R5 that was absent in 477 normal controls. Our characterization of the PIK3R5 protein and findings suggest that it may play a role in the development of the cerebellum and vermis.


Asunto(s)
Apraxias/genética , Ataxia Telangiectasia/genética , Ataxia/genética , Fosfatidilinositol 3-Quinasa Clase Ib/genética , Hipoalbuminemia/genética , Mutación Missense , Fosfatidilinositol 3-Quinasas/genética , Adolescente , Adulto , Animales , Apraxias/diagnóstico , Ataxia/diagnóstico , Ataxia Telangiectasia/diagnóstico , Encéfalo/patología , Ataxia Cerebelosa/congénito , Consanguinidad , ADN Helicasas , Femenino , Orden Génico , Ligamiento Genético , Homocigoto , Humanos , Hipoalbuminemia/diagnóstico , Imagen por Resonancia Magnética , Masculino , Ratones , Enzimas Multifuncionales , Linaje , Fenotipo , ARN Helicasas/genética , Relaciones entre Hermanos , Adulto Joven
11.
Mol Genet Metab ; 105(3): 479-83, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22227324

RESUMEN

Analbuminemia is a rare autosomal recessive disorder manifested by the absence or severe reduction of circulating serum albumin in homozygous or compound heterozygous subjects. It is an allelic heterogeneous defect, caused by a variety of mutations within the albumin gene. The analbuminemic condition was suspected in a Portuguese boy who presented with low albumin level (about 3.8 g/L) and a significant hypercholesterolemia, but with no clinical findings. The albumin gene was screened by single strand conformational polymorphism and heteroduplex analysis and submitted to direct DNA sequencing. The proband was found to be homozygous for a previously unreported G>A change at position c.1289+1, the first base of intron 10, which inactivates the strongly conserved GT dinucleotide at the 5' splice site consensus sequence of the intron. The effect of this mutation was evaluated by examining the cDNA obtained by RT-PCR from the albumin mRNA extracted from proband's leukocytes. The splicing defect results in the skipping of the preceding exon. The subsequent reading frame-shift in exon 11 produces a premature stop codon located 33 codons downstream the 5' end of the exon. This extensive cDNA alteration is responsible for the analbuminemic trait. Both parents were found to be heterozygous for the same mutation. DNA and cDNA sequence analysis established the diagnosis of congenital analbuminemia in the proband. The effects of the so far identified splice-site mutations in the albumin gene are discussed.


Asunto(s)
Hipoalbuminemia/genética , Empalme del ARN , Albúmina Sérica/deficiencia , Albúmina Sérica/genética , Adolescente , Secuencia de Bases , Mutación del Sistema de Lectura , Predisposición Genética a la Enfermedad , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Portugal , Análisis de Secuencia de ADN , Albúmina Sérica Humana
12.
Brain ; 134(Pt 5): 1387-99, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21486904

RESUMEN

Early onset ataxia with ocular motor apraxia and hypoalbuminaemia/ataxia-oculomotor apraxia 1 is a recessively inherited ataxia caused by mutations in the aprataxin gene. We previously reported that patients with frameshift mutations exhibit a more severe phenotype than those with missense mutations. However, reports on genotype-phenotype correlation in early onset ataxia with ocular motor apraxia and hypoalbuminaemia are controversial. To clarify this issue, we studied 58 patients from 39 Japanese families, including 40 patients homozygous for c.689_690insT and nine patients homozygous or compound heterozygous for p.Pro206Leu or p.Val263Gly mutations who were compared with regard to clinical phenotype. We performed Kaplan-Meier analysis and log-rank tests for the ages of onset of gait disturbance and the inability to walk without assistance. The cumulative rate of gait disturbance was lower among patients with p.Pro206Leu or p.Val263Gly mutations than among those homozygous for the c.689_690insT mutation (P=0.001). The cumulative rate of inability to walk without assistance was higher in patients homozygous for the c.689_690insT mutation than in those with p.Pro206Leu or p.Val263Gly mutations (P=0.004). Using a Cox proportional hazards model, we found that the homozygous c.689_690insT mutation was associated with an increased risk for onset of gait disturbance (adjusted hazard ratio: 6.60) and for the inability to walk without assistance (adjusted hazard ratio: 2.99). All patients homozygous for the c.689_690insT mutation presented ocular motor apraxia at <15 years of age. Approximately half the patients homozygous for the c.689_690insT mutation developed cognitive impairment. In contrast, in the patients with p.Pro206Leu or p.Val263Gly mutations, only ∼50% of the patients exhibited ocular motor apraxia and they never developed cognitive impairment. The stepwise multivariate regression analysis using sex, age and the number of c.689_690insT alleles as independent variables revealed that the number of c.689_690insT alleles was independently and negatively correlated with median motor nerve conduction velocities, ulnar motor nerve conduction velocities and values of serum albumin. In the patient with c.[689_690insT]+[840delT], p.[Pro206Leu]+[Pro206Leu] and p.[Pro206Leu]+[Val263Gly] mutations, aprataxin proteins were not detected by an antibody to the N-terminus of aprataxin. Furthermore Pro206Leu and Val263Gly aprataxin proteins are unstable. However, the amount of the 689_690insT aprataxin messenger RNA was also decreased, resulting in more dramatic reduction in the amount of aprataxin protein from the c.689_690insT allele. In conclusion, patients with early onset ataxia with ocular motor apraxia and hypoalbuminaemia homozygous for the c.689_690insT mutation show a more severe phenotype than those with a p.Pro206Leu or p.Val263Gly mutation.


Asunto(s)
Ataxia , Proteínas de Unión al ADN/genética , Estudios de Asociación Genética , Hipoalbuminemia , Mutación/genética , Proteínas Nucleares/genética , Trastornos de la Motilidad Ocular , Potenciales de Acción/genética , Edad de Inicio , Ataxia/complicaciones , Ataxia/etiología , Línea Celular Transformada , Proteínas de Unión al ADN/metabolismo , Salud de la Familia , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Hipoalbuminemia/complicaciones , Hipoalbuminemia/genética , Estimación de Kaplan-Meier , Conducción Nerviosa/genética , Conducción Nerviosa/fisiología , Proteínas Nucleares/metabolismo , Trastornos de la Motilidad Ocular/complicaciones , Trastornos de la Motilidad Ocular/genética , ARN Mensajero/metabolismo , Reflejo/genética , Análisis de Regresión , Tetraciclina/farmacología , Transfección/métodos
13.
Hum Mutat ; 32(4): E2118-33, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21412945

RESUMEN

APTX is the gene involved in ataxia with oculomotor apraxia type 1 (AOA1), a recessive disorder with early-onset cerebellar ataxia, oculomotor apraxia and peripheral neuropathy. The encoded protein, aprataxin, is a DNA repair protein processing the products of abortive ligations, 5'-adenylated DNA. We describe a novel nonsense mutation in APTX, c.892C>T (p.Gln298X), segregating in two AOA1 patients and leading to the loss of aprataxin protein in patient's cells. These cells, while exhibiting reduced catalase activity, are not hypersensitive to toxicity elicited by H(2)O(2) exposure at either physiologic or ice-bath temperature. On the other hand, the rate of repair of DNA single-strand-breaks (SSBs) induced in both conditions is always significantly slower in AOA1 cells. By using the alkylating agent methyl methane sulphonate (MMS) we confirmed the association of the APTX mutation with a DNA repair defect in the absence of detectable changes in susceptibility to toxicity. These results, while consistent with a role of aprataxin in the repair of SSBs induced by H(2)O(2), or MMS, demonstrate that other mechanisms may be recruited in AOA1 cells to complete the repair process, although at a slower rate. Lack of hypersensitivity to the oxidant, or MMS, also implies that delayed repair is not per se a lethal event.


Asunto(s)
Codón sin Sentido/genética , Roturas del ADN de Cadena Simple , Proteínas de Unión al ADN/genética , Mutágenos/toxicidad , Proteínas Nucleares/genética , Apraxias/genética , Ataxia Telangiectasia/genética , Secuencia de Bases , Ataxia Cerebelosa/congénito , ADN/genética , ADN/metabolismo , Daño del ADN/genética , Reparación del ADN , Humanos , Peróxido de Hidrógeno/toxicidad , Hipoalbuminemia/genética , Metilmetanosulfonato/toxicidad , Oxidantes/toxicidad , Linaje , Degeneraciones Espinocerebelosas/genética
14.
BMC Med Genet ; 12: 27, 2011 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-21324166

RESUMEN

BACKGROUND: Autosomal recessive ataxias represent a group of clinically overlapping disorders. These include ataxia with oculomotor apraxia type1 (AOA1), ataxia with oculomotor apraxia type 2 (AOA2) and ataxia-telangiectasia-like disease (ATLD). Patients are mainly characterized by cerebellar ataxia and oculomotor apraxia. Although these forms are not quite distinctive phenotypically, different genes have been linked to these disorders. Mutations in the APTX gene were reported in AOA1 patients, mutations in SETX gene were reported in patients with AOA2 and mutations in MRE11 were identified in ATLD patients. In the present study we describe in detail the clinical features and results of genetic analysis of 9 patients from 4 Saudi families with ataxia and oculomotor apraxia. METHODS: This study was conducted in the period between 2005-2010 to clinically and molecularly characterize patients with AOA phenotype. Comprehensive sequencing of all coding exons of previously reported genes related to this disorder (APTX, SETX and MRE11). RESULTS: A novel nonsense truncating mutation c.6859 C > T, R2287X in SETX gene was identified in patients from one family with AOA2. The previously reported missense mutation W210C in MRE11 gene was identified in two families with autosomal recessive ataxia and oculomotor apraxia. CONCLUSION: Mutations in APTX , SETX and MRE11 are common in patients with autosomal recessive ataxia and oculomotor apraxia. The results of the comprehensive screening of these genes in 4 Saudi families identified mutations in SETX and MRE11 genes but failed to identify mutations in APTX gene.


Asunto(s)
Proteínas de Unión al ADN/genética , Mutación , ARN Helicasas/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Apraxias/genética , Ataxia Telangiectasia/genética , Secuencia de Bases , Ataxia Cerebelosa/congénito , Mapeo Cromosómico , Codón sin Sentido , ADN/genética , ADN Helicasas , Análisis Mutacional de ADN , Femenino , Variación Genética , Humanos , Hipoalbuminemia/genética , Proteína Homóloga de MRE11 , Masculino , Enzimas Multifuncionales , Mutación Missense , Proteínas Nucleares/genética , Linaje , Arabia Saudita , Ataxias Espinocerebelosas/congénito , Degeneraciones Espinocerebelosas/genética , Adulto Joven
15.
Medicine (Baltimore) ; 100(24): e26307, 2021 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-34128868

RESUMEN

INTRODUCTION: 22q13.3 deletion syndrome is a well-known syndrome characterized by typical clinical findings including neonatal hypotonia, absent or severely delayed speech, intellectual disability, and other various features, and detection of a heterozygous deletion of chromosome 22q13.3 with the involvement of at least part of SHANK3. It is reported that 10% to 29% of patients with 22q13.3 deletion syndrome present lymphedema. Protein-losing enteropathy (PLE) has never been reported in 22q13.3 deletion syndrome. PATIENT CONCERNS: The patient presented to our institution for refractory hypoalbuminemia and chronic lymphedema in both legs. DIAGNOSIS: The patient manifested intellectual disability, absent speech, tooth grinding, dysmorphic face, and abnormal hands and toenails. Copy-number variation sequencing confirmed the maternal deletion in 22q13.31-q13.33 (chr22:46285592-51244566, hg19). The patient was genetically diagnosed with 22q13.3 deletion syndrome. INTERVENTIONS: Low-fat diets and medium-chain triglycerides supplements were prescribed. The patient was recommended to wear compression garments and elevate legs. OUTCOMES: The symptom of diarrhea was resolved, but hypoalbuminemia persisted. Lower extremities lymphedema was gradually becoming severe. CONCLUSIONS: Primary lymphedema and PLE can occur simultaneously in a patient with 22q13.3 deletion syndrome. The 2 phenotypes could share the same genetic etiology of congenital lymphatic abnormalities. CELSR1 deletion may play a role in lymphatic dysplasia. The case also provides additional proof of the pathogenic effect of CELSR1 on hereditary lymphedema.


Asunto(s)
Cadherinas/genética , Trastornos de los Cromosomas/genética , Linfedema/genética , Enteropatías Perdedoras de Proteínas/genética , Deleción Cromosómica , Cromosomas Humanos Par 22/genética , Variaciones en el Número de Copia de ADN , Femenino , Humanos , Hipoalbuminemia/genética , Discapacidad Intelectual/genética , Pierna/patología , Adulto Joven
16.
Sci Rep ; 11(1): 11290, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-34050200

RESUMEN

Hypoalbuminemia is associated with vascular endothelial dysfunction and the development of chronic cardiovascular diseases. However, the relationship between serum albumin concentration and blood pressure changes remains controversial. Community-based longitudinal cohort data collected from Korean Genome and Epidemiology Study were used in this study. Hypoalbuminemia was defined as a serum albumin concentration of ≤ 4.0 g/dL. A total of 4325 participants were categorized into control (n = 3157) and hypoalbuminemia (n = 1168) groups. Serum albumin had a non-linear relationship with the risk of hypertension development. A genome-wide association study revealed 71 susceptibility loci associated with hypoalbuminemia. Among susceptibility loci, genetic variations at rs2894536 in LOC107986598 and rs10972486 in ATP8B5P were related to elevated blood pressure. Serum albumin (HR = 0.654, 95% CI 0.521-0.820) and polymorphisms of rs2894536 (HR = 1.176, 95% CI 1.015-1.361) and rs10972486 (HR = 1.152, 95% CI 1.009-1.316) were significant predictors of hypertension development. Increased albumin concentration instrumented by 2 hypoalbuminemia-associated SNPs (rs2894536 and rs10972486) was associated with decreased HRs for hypertension development (HR = 0.762, 95% CI 0.659-0.882 and HR = 0.759, 95% CI 0.656-0.878). Our study demonstrated that genetically determined hypoalbuminemia is a significant predictor of incipient hypertension.


Asunto(s)
Hipertensión/genética , Hipoalbuminemia/genética , Albúmina Sérica/análisis , Adulto , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Humanos , Hipertensión/sangre , Hipertensión/fisiopatología , Hipoalbuminemia/sangre , Hipoalbuminemia/fisiopatología , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Pronóstico , República de Corea/epidemiología , Estudios Retrospectivos , Factores de Riesgo
17.
Adv Exp Med Biol ; 685: 21-33, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20687492

RESUMEN

DNA single-strand breaks (SSBs) are non-overlapping discontinuities in strands ofa DNA duplex. Significant attention has been given on the DNA SSB repair (SSBR) system in neurons, because the impairment of the SSBR causes human neurodegenerative disorders, including early-onset ataxia with ocular motor apraxia and hypoalbuminemia (EAOH), also known as ataxia-oculomotor apraxia Type 1 (AOA1). EAOH/AOA1 is characterized by early-onset slowly progressive ataxia, ocular motor apraxia, peripheral neuropathy and hypoalbuminemia. Neuropathological examination reveals severe loss of Purkinje cells and moderate neuronal loss in the anterior horn and dorsal root ganglia. EAOH/AOA1 is caused by the mutation in the APTX gene encoding the aprataxin (APTX) protein. APTX interacts with X-ray repair cross-complementing group 1 protein, which is a scaffold protein in SSBR. In addition, APTX-defective cells show increased sensitivity to genotoxic agents, which result in SSBs. These results indicate an important role ofAPTX in SSBR. SSBs are usually accompanied by modified or damaged 5'- and 3'-ends at the break site. Because these modified or damaged ends are not suitable for DNA ligation, they need to be restored to conventional ends prior to subsequent repair processes. APTX restores the 5'-adenylate monophosphate, 3'-phosphates and 3'-phosphoglycolate ends. The loss of function of APTX results in the accumulation of SSBs, consequently leading to neuronal cell dysfunction and death.


Asunto(s)
Apraxias , Roturas del ADN de Cadena Simple , Reparación del ADN , Proteínas de Unión al ADN , Hipoalbuminemia , Proteínas Nucleares , Degeneraciones Espinocerebelosas , Apraxias/genética , Apraxias/metabolismo , Apraxias/patología , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Hipoalbuminemia/genética , Hipoalbuminemia/metabolismo , Hipoalbuminemia/patología , Mutágenos/farmacología , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Células de Purkinje , Degeneraciones Espinocerebelosas/genética , Degeneraciones Espinocerebelosas/metabolismo , Degeneraciones Espinocerebelosas/patología , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
18.
Med Clin (Barc) ; 154(7): 275-278, 2020 04 10.
Artículo en Inglés, Español | MEDLINE | ID: mdl-31928748

RESUMEN

INTRODUCTION AND OBJECTIVES: Congenital analbuminaemia (CCA) (MIM #616000) is an autosomal recessive disorder (prevalence < 1/106) caused by defects in the ALB gene leading to absence or severe reduction of albuminaemia. This paper describes a case of CCA detected and diagnosed in our hospital. MATERIALS AND METHODS: A 42-year old woman showing hypoproteinaemia and hypoalbuminaemia of unknown aetiology. Biochemical study was performed according to routine quality controlled analytical procedures: Albuminaemia (colorimetric and nephelometric methods). Protein electrophoresis (capillary and agarose gel). Molecular study of the ALB gene: DNA extraction, PCR amplification of the 14 coding exons plus adjacent intron regions and Sanger sequencing. RESULTS: After discarding the most common causes of hypoalbuminaemia, the analbuminaemia was confirmed by nephelometry and protein electrophoresis. The proband was found to be homozygous for molecular defect in the ALB gene: variant c.1289+1G>A previously reported as Guimarães variant. CONCLUSIONS: This is the first case of CCA confirmed by molecular study in Spain. The proband shows the Guimarães variant previously described in 4 patients worldwide.


Asunto(s)
Hipoalbuminemia , Adulto , Exones , Femenino , Homocigoto , Humanos , Hipoalbuminemia/genética , Albúmina Sérica , España
19.
Artículo en Inglés | MEDLINE | ID: mdl-33101765

RESUMEN

Background: Ataxia with oculomotor apraxia (AOA1) is characterized by early-onset progressive cerebellar ataxia with peripheral neuropathy, oculomotor apraxia and hypoalbuminemia and hypercholesterolemia. Case Report: A 23-year-old previously healthy woman presented with slowly-progressive gait impairment since the age of six years. Neurological examination revealed profound areflexia, chorea, generalized dystonia and oculomotor apraxia. Brain MRI revealed mild cerebellar atrophy and needle EMG showed axonal sensorimotor neuropathy. Whole exome sequencing revealed a mutation in the aprataxin gene. Discussion: AOA1 can present with choreoathetosis mixed with dystonic features, resembling ataxia-telangiectasia. This case is instructive since mixed and complex movement disorders is not very common in AOA1. Highlights: Ataxia with oculomotor apraxia type 1 (AOA1) is characterized by early-onset ataxia and oculomotor apraxia caused by variants in the APTX gene.Ataxia is usually not the sole movement abnormality in AOA1.Hyperkinetic movement disorders, especially chorea and dystonia, may occur.Mixed and complex movement disorders is not very common in AOA1.Patients with early-onset ataxia associated with mixed movement disorders should also be investigated for AOA1.


Asunto(s)
Apraxias/fisiopatología , Ataxia Cerebelosa/congénito , Cerebelo/diagnóstico por imagen , Corea/fisiopatología , Distonía/fisiopatología , Hipoalbuminemia/fisiopatología , Reflejo Anormal/fisiología , Apraxias/diagnóstico por imagen , Apraxias/genética , Atrofia , Ataxia Cerebelosa/diagnóstico por imagen , Ataxia Cerebelosa/genética , Ataxia Cerebelosa/fisiopatología , Enfermedades Cerebelosas/fisiopatología , Cerebelo/patología , Proteínas de Unión al ADN/genética , Electromiografía , Femenino , Humanos , Hipoalbuminemia/diagnóstico por imagen , Hipoalbuminemia/genética , Proteínas Nucleares/genética , Adulto Joven
20.
Eur J Med Genet ; 62(2): 144-148, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29981851

RESUMEN

Congenital analbuminemia (OMIM # 616000) is an extremely rare autosomal recessive disorder, caused by variations in the albumin gene (ALB), which is generally thought to be a relatively benign condition in adulthood, but seems to be potentially life threatening in the pre- and peri-natal period. The subject of our study was a consanguineous family, in which we identified two analbuminemic individuals. Mutation analysis of ALB revealed that both are homozygous for a previously unreported insertion in exon 9 (c.1098dupT), causing a subsequent frame-shift with the generation of a premature stop codon, and an aberrant truncated putative protein product, p.Val367fsTer12. This variation is present in heterozygous condition in several other members of the family. The phenotype and the molecular genetics of CAA are discussed.


Asunto(s)
Hipoalbuminemia/genética , Mutación , Albúmina Sérica Humana/genética , Adulto , Anciano , Codón de Terminación , Consanguinidad , Femenino , Humanos , Hipoalbuminemia/patología , Masculino , Linaje , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA