Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Mol Genet Metab ; 140(1-2): 107678, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37574344

RESUMEN

The advancements in population screening, including newborn screening, enables the identification of disease-causing variants and timely initiation of treatment. However, screening may also identify mild variants, non-disease variants, and variants of uncertain significance (VUS). The identification of a VUS poses a challenge in terms of diagnostic uncertainty and confusion. X-linked adrenoleukodystrophy (ALD) serves as an illustrative example of this complex issue. ALD is a monogenic neurometabolic disease with a complex clinical presentation and a lack of predictive tests for clinical severity. Despite the success of ALD newborn screening, a significant proportion (62%) of missense variants identified through newborn screening exhibit uncertainty regarding their pathogenicity. Resolving this issue requires ongoing efforts to accurately classify variants and refine screening protocols. While it is undisputable that ALD newborn screening greatly benefits boys with the disease, the identification of VUS underscores the need for continuous research and collaboration in improving screening practices.


Asunto(s)
Adrenoleucodistrofia , Masculino , Recién Nacido , Humanos , Adrenoleucodistrofia/diagnóstico , Tamizaje Neonatal/métodos , Mutación Missense
2.
Genet Med ; 25(11): 100944, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37493040

RESUMEN

PURPOSE: Zellweger spectrum disorders (ZSDs) are known as autosomal recessive disorders caused by defective peroxisome biogenesis due to bi-allelic pathogenic variants in any of at least 13 different PEX genes. Here, we report 2 unrelated patients who present with an autosomal dominant ZSD. METHODS: We performed biochemical and genetic studies in blood and skin fibroblasts of the patients and demonstrated the pathogenicity of the identified PEX14 variants by functional cell studies. RESULTS: We identified 2 different single heterozygous de novo variants in the PEX14 genes of 2 patients diagnosed with ZSD. Both variants cause messenger RNA mis-splicing, leading to stable expression of similar C-terminally truncated PEX14 proteins. Functional studies indicated that the truncated PEX14 proteins lost their function in peroxisomal matrix protein import and cause increased degradation of peroxisomes, ie, pexophagy, thus exerting a dominant-negative effect on peroxisome functioning. Inhibition of pexophagy by different autophagy inhibitors or genetic knockdown of the peroxisomal autophagy receptor NBR1 resulted in restoration of peroxisomal functions in the patients' fibroblasts. CONCLUSION: Our finding of an autosomal dominant ZSD expands the genetic repertoire of ZSDs. Our study underscores that single heterozygous variants should not be ignored as possible genetic cause of diseases with an established autosomal recessive mode of inheritance.


Asunto(s)
Síndrome de Zellweger , Humanos , Alelos , Peroxisomas/genética , Peroxisomas/metabolismo , Transporte de Proteínas/fisiología , Proteínas/genética , Síndrome de Zellweger/genética
3.
J Inherit Metab Dis ; 46(1): 116-128, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36256460

RESUMEN

Males with X-linked adrenoleukodystrophy (ALD) are at high risk for developing adrenal insufficiency and/or progressive leukodystrophy (cerebral ALD) at an early age. Pathogenic variants in ABCD1 result in elevated levels of very long-chain fatty acids (VLCFA), including C26:0-lysophosphatidylcholine (C26:0-LPC). Newborn screening for ALD enables prospective monitoring and timely therapeutic intervention, thereby preventing irreversible damage and saving lives. The Dutch Health Council recommended to screen only male newborns for ALD without identifying untreatable conditions associated with elevated C26:0-LPC, like Zellweger spectrum disorders and single peroxisomal enzyme defects. Here, we present the results of the SCAN (Screening for ALD in the Netherlands) study which is the first sex-specific newborn screening program worldwide. Males with ALD are identified based on elevated C26:0-LPC levels, the presence of one X-chromosome and a variant in ABCD1, in heel prick dried bloodspots. Screening of 71 208 newborns resulted in the identification of four boys with ALD who, following referral to the pediatric neurologist and confirmation of the diagnosis, enrolled in a long-term follow-up program. The results of this pilot show the feasibility of employing a boys-only screening algorithm that identifies males with ALD without identifying untreatable conditions. This approach will be of interest to countries that are considering ALD newborn screening but are reluctant to identify girls with ALD because for girls there is no direct health benefit. We also analyzed whether gestational age, sex, birth weight and age at heel prick blood sampling affect C26:0-LPC concentrations and demonstrate that these covariates have a minimal effect.


Asunto(s)
Insuficiencia Suprarrenal , Adrenoleucodistrofia , Niño , Femenino , Humanos , Masculino , Recién Nacido , Adrenoleucodistrofia/diagnóstico , Adrenoleucodistrofia/genética , Tamizaje Neonatal/métodos , Estudios Prospectivos , Lisofosfatidilcolinas , Ácidos Grasos
4.
J Inherit Metab Dis ; 45(4): 819-831, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35403730

RESUMEN

Mitochondrial trifunctional protein (MTP) is involved in long-chain fatty acid ß-oxidation (lcFAO). Deficiency of one or more of the enzyme activities as catalyzed by MTP causes generalized MTP deficiency (MTPD), long-chain hydroxyacyl-CoA dehydrogenase deficiency (LCHADD), or long-chain ketoacyl-CoA thiolase deficiency (LCKATD). When genetic variants result in thermo-sensitive enzymes, increased body temperature (e.g. fever) can reduce enzyme activity and be a risk factor for clinical decompensation. This is the first description of five patients with a thermo-sensitive MTP deficiency. Clinical and genetic information was obtained from clinical files. Measurement of LCHAD and LCKAT activities, lcFAO-flux studies and palmitate loading tests were performed in skin fibroblasts cultured at 37°C and 40°C. In all patients (four MTPD, one LCKATD), disease manifested during childhood (manifestation age: 2-10 years) with myopathic symptoms triggered by fever or exercise. In four patients, signs of retinopathy or neuropathy were present. Plasma long-chain acylcarnitines were normal or slightly increased. HADHB variants were identified (at age: 6-18 years) by whole exome sequencing or gene panel analyses. At 37°C, LCHAD and LCKAT activities were mildly impaired and lcFAO-fluxes were normal. Remarkably, enzyme activities and lcFAO-fluxes were markedly diminished at 40°C. Preventive (dietary) measures improved symptoms for most. In conclusion, all patients with thermo-sensitive MTP deficiency had a long diagnostic trajectory and both genetic and enzymatic testing were required for diagnosis. The frequent absence of characteristic acylcarnitine abnormalities poses a risk for a diagnostic delay. Given the positive treatment effects, upfront genetic screening may be beneficial to enhance early recognition.


Asunto(s)
Errores Innatos del Metabolismo Lipídico , Miopatías Mitocondriales , Enfermedades Musculares , 3-Hidroxiacil-CoA Deshidrogenasas , Adolescente , Cardiomiopatías , Niño , Preescolar , Coenzima A , Diagnóstico Tardío , Ácidos Grasos/metabolismo , Humanos , Errores Innatos del Metabolismo Lipídico/diagnóstico , Errores Innatos del Metabolismo Lipídico/genética , Errores Innatos del Metabolismo Lipídico/metabolismo , Miopatías Mitocondriales/diagnóstico , Miopatías Mitocondriales/genética , Proteína Trifuncional Mitocondrial/deficiencia , Enfermedades Musculares/diagnóstico , Enfermedades Musculares/genética , Enfermedades del Sistema Nervioso , Rabdomiólisis
5.
Genes (Basel) ; 12(12)2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34946879

RESUMEN

Due to newborn screening for X-linked adrenoleukodystrophy (ALD), and the use of exome sequencing in clinical practice, the detection of variants of unknown significance (VUS) in the ABCD1 gene is increasing. In these cases, functional tests in fibroblasts may help to classify a variant as (likely) benign or pathogenic. We sought to establish reference ranges for these tests in ALD patients and control subjects with the aim of helping to determine the pathogenicity of VUS in ABCD1. Fibroblasts from 36 male patients with confirmed ALD, 26 healthy control subjects and 17 individuals without a family history of ALD, all with an uncertain clinical diagnosis and a VUS identified in ABCD1, were included. We performed a combination of tests: (i) a test for very-long-chain fatty acids (VLCFA) levels, (ii) a D3-C22:0 loading test to study the VLCFA metabolism and (iii) immunoblotting for ALD protein. All ALD patient fibroblasts had elevated VLCFA levels and a reduced peroxisomal ß-oxidation capacity (as measured by the D3-C16:0/D3-C22:0 ratio in the D3-C22:0 loading test) compared to the control subjects. Of the VUS cases, the VLCFA metabolism was not significantly impaired (most test results were within the reference range) in 6/17, the VLCFA metabolism was significantly impaired (most test results were within/near the ALD range) in 9/17 and a definite conclusion could not be drawn in 2/17 of the cases. Biochemical studies in fibroblasts provided clearly defined reference and disease ranges for the VLCFA metabolism. In 15/17 (88%) VUS we were able to classify the variant as being likely benign or pathogenic. This is of great clinical importance as new variants will be detected.


Asunto(s)
Miembro 1 de la Subfamilia D de Transportador de Casetes de Unión al ATP/genética , Adrenoleucodistrofia/genética , Fibroblastos/metabolismo , Mutación , Miembro 1 de la Subfamilia D de Transportador de Casetes de Unión al ATP/metabolismo , Adrenoleucodistrofia/metabolismo , Adulto , Ácidos Grasos/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Valores de Referencia
6.
JIMD Rep ; 61(1): 12-18, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34485012

RESUMEN

Early detection of congenital disorders by newborn screening (NBS) programs is essential to prevent or limit disease manifestation in affected neonates. These programs balance between the detection of the highest number of true cases and the lowest number of false-positives. In this case report, we describe four unrelated cases with a false-positive NBS result for very-long-chain acyl-CoA dehydrogenase deficiency (VLCADD). Three neonates presented with decreased but not deficient VLCAD enzyme activity and two of them carried a single heterozygous ACADVL c.1844G>A mutation. Initial biochemical investigations after positive NBS referral in these infants revealed acylcarnitine and organic acid profiles resembling those seen in multiple acyl-CoA dehydrogenase deficiency (MADD). Genetic analysis did not reveal any pathogenic mutations in the genes encoding the electron transfer flavoprotein (ETF alpha and beta subunits) nor in ETF dehydrogenase. Subsequent further diagnostics revealed decreased levels of riboflavin in the newborns and oral riboflavin administration normalized the MADD-like biochemical profiles. During pregnancy, the mothers followed a vegan, vegetarian or lactose-free diet which probably caused alimentary riboflavin deficiency in the neonates. This report demonstrates that a secondary (alimentary) maternal riboflavin deficiency in combination with reduced VLCAD activity in the newborns can result in an abnormal VLCADD/MADD acylcarnitine profile and can cause false-positive NBS. We hypothesize that maternal riboflavin deficiency contributed to the false-positive VLCADD neonatal screening results.

7.
Hum Mutat ; 40(10): 1899-1904, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31187905

RESUMEN

Primary carnitine deficiency is caused by a defect in the active cellular uptake of carnitine by Na+ -dependent organic cation transporter novel 2 (OCTN2). Genetic diagnostic yield for this metabolic disorder has been relatively low, suggesting that disease-causing variants are missed. We Sanger sequenced the 5' untranslated region (UTR) of SLC22A5 in individuals with possible primary carnitine deficiency in whom no or only one mutant allele had been found. We identified a novel 5'-UTR c.-149G>A variant which we characterized by expression studies with reporter constructs in HeLa cells and by carnitine-transport measurements in fibroblasts using a newly developed sensitive assay based on tandem mass spectrometry. This variant, which we identified in 57 of 236 individuals of our cohort, introduces a functional upstream out-of-frame translation initiation codon. We show that the codon suppresses translation from the wild-type ATG of SLC22A5, resulting in reduced OCTN2 protein levels and concomitantly lower transport activity. With an allele frequency of 24.2% the c.-149G>A variant is the most frequent cause of primary carnitine deficiency in our cohort and may explain other reported cases with an incomplete genetic diagnosis. Individuals carrying this variant should be clinically re-evaluated and monitored to determine if this variant has clinical consequences.


Asunto(s)
Regiones no Traducidas 5' , Cardiomiopatías/genética , Carnitina/deficiencia , Codón Iniciador , Predisposición Genética a la Enfermedad , Hiperamonemia/genética , Enfermedades Musculares/genética , Mutación , Miembro 5 de la Familia 22 de Transportadores de Solutos/genética , Alelos , Secuencia de Aminoácidos , Secuencia de Bases , Transporte Biológico , Cardiomiopatías/diagnóstico , Cardiomiopatías/metabolismo , Carnitina/genética , Carnitina/metabolismo , Línea Celular , Frecuencia de los Genes , Genes Reporteros , Estudios de Asociación Genética , Humanos , Hiperamonemia/diagnóstico , Hiperamonemia/metabolismo , Enfermedades Musculares/diagnóstico , Enfermedades Musculares/metabolismo , Miembro 5 de la Familia 22 de Transportadores de Solutos/metabolismo
8.
Biochim Biophys Acta Mol Basis Dis ; 1864(3): 952-958, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29287774

RESUMEN

Peroxisomal acyl-CoA oxidases catalyze the first step of beta-oxidation of a variety of substrates broken down in the peroxisome. These include the CoA-esters of very long-chain fatty acids, branched-chain fatty acids and the C27-bile acid intermediates. In rat, three peroxisomal acyl-CoA oxidases with different substrate specificities are known, whereas in humans it is believed that only two peroxisomal acyl-CoA oxidases are expressed under normal circumstances. Only three patients with ACOX2 deficiency, including two siblings, have been identified so far, showing accumulation of the C27-bile acid intermediates. Here, we performed biochemical studies in material from a novel ACOX2-deficient patient with increased levels of C27-bile acids in plasma, a complete loss of ACOX2 protein expression on immunoblot, but normal pristanic acid oxidation activity in fibroblasts. Since pristanoyl-CoA is presumed to be handled by ACOX2 specifically, these findings prompted us to re-investigate the expression of the human peroxisomal acyl-CoA oxidases. We report for the first time expression of ACOX3 in normal human tissues at the mRNA and protein level. Substrate specificity studies were done for ACOX1, 2 and 3 which revealed that ACOX1 is responsible for the oxidation of straight-chain fatty acids with different chain lengths, ACOX2 is the only human acyl-CoA oxidase involved in bile acid biosynthesis, and both ACOX2 and ACOX3 are involved in the degradation of the branched-chain fatty acids. Our studies provide new insights both into ACOX2 deficiency and into the role of the different acyl-CoA oxidases in peroxisomal metabolism.


Asunto(s)
Oxidorreductasas/genética , Oxidorreductasas/aislamiento & purificación , Acil-CoA Oxidasa , Ácidos y Sales Biliares/metabolismo , Consanguinidad , Femenino , Humanos , Recién Nacido , Hígado/metabolismo , Oxidorreductasas/deficiencia , Pakistán , Especificidad por Sustrato
9.
J. inborn errors metab. screen ; 6: e180018, 2018. graf
Artículo en Inglés | LILACS-Express | LILACS | ID: biblio-1090951

RESUMEN

Abstract The clinical as well as biochemical and genetic spectrum of peroxisomal diseases has markedly increased over the last few years, thanks to the revolutionary advances in the field of genome analysis and several -omics technologies. This has led to the recognition of novel disease phenotypes linked to mutations in previously identified peroxisomal genes as well as several hitherto unidentified peroxisomal disorders. Correct interpretation of the wealth of data especially coming from genome analysis requires functional studies at the level of metabolites (peroxisomal metabolite biomarkers), enzymes, and the metabolic pathway(s) involved. This strategy is not only required to identify the true defect in each individual patient but also to determine the extent of the deficiency as described in detail in this article.

10.
J Inherit Metab Dis ; 39(4): 531-43, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26943801

RESUMEN

Peroxisomes are dynamic organelles that play an essential role in a variety of metabolic pathways. Peroxisomal dysfunction can lead to various biochemical abnormalities and result in abnormal metabolite levels, such as increased very long-chain fatty acid or reduced plasmalogen levels. The metabolite abnormalities in peroxisomal disorders are used in the diagnostics of these disorders. In this paper we discuss in detail the different diagnostic tests available for peroxisomal disorders and focus specifically on the important role of biochemical and functional studies in cultured skin fibroblasts in reaching the right diagnosis. Several examples are shown to underline the power of such studies.


Asunto(s)
Biomarcadores/análisis , Técnicas y Procedimientos Diagnósticos , Trastorno Peroxisomal/diagnóstico , Biomarcadores/sangre , Biomarcadores/orina , Fibroblastos/patología , Humanos , Tamizaje Masivo/métodos , Redes y Vías Metabólicas/fisiología , Trastorno Peroxisomal/sangre , Trastorno Peroxisomal/metabolismo , Trastorno Peroxisomal/orina , Cultivo Primario de Células/métodos , Piel/patología
11.
Hum Mol Genet ; 24(20): 5845-54, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26220973

RESUMEN

Import of peroxisomal matrix proteins, crucial for peroxisome biogenesis, is mediated by the cytosolic receptors PEX5 and PEX7 that recognize proteins carrying peroxisomal targeting signals 1 or 2 (PTS1 or PTS2), respectively. Mutations in PEX5 or 12 other PEX genes cause peroxisome biogenesis disorders, collectively named the Zellweger spectrum disorders (ZSDs), whereas mutations in PEX7 cause rhizomelic chondrodysplasia punctata type 1 (RCDP1). Three additional RCDP types, RCDP2-3-4, are caused, respectively, by mutations in GNPAT, AGPS and FAR1, encoding enzymes involved in plasmalogen biosynthesis. Here we report a fifth type of RCDP (RCDP5) caused by a novel mutation in PEX5. In four patients with RCDP from two independent families, we identified a homozygous frame shift mutation c.722dupA (p.Val242Glyfs(∗)33) in PEX5 (GenBank: NM_001131023.1). PEX5 encodes two isoforms, PEX5L and PEX5S, and we show that the c.722dupA mutation, located in the PEX5L-specific exon 9, results in loss of PEX5L only. Both PEX5 isoforms recognize PTS1-tagged proteins, but PEX5L is also a co-receptor for PTS2-tagged proteins. Previous patients with PEX5 mutations had ZSD, mainly due to deficient import of PTS1-tagged proteins. Similarly to mutations in PEX7, loss of PEX5L results in deficient import of PTS2-tagged proteins only, thus causing RCDP instead of ZSD. We demonstrate that PEX5L expression restores the import of PTS2-tagged proteins in patient fibroblasts. Due to the biochemical overlap between RCDP1 and RCDP5, sequencing of PEX7 and exon 9 in PEX5 should be performed in patients with a selective defect in the import of PTS2-tagged proteins.


Asunto(s)
Condrodisplasia Punctata Rizomélica/genética , Mutación del Sistema de Lectura , Peroxisomas/metabolismo , Transporte de Proteínas/genética , Receptores Citoplasmáticos y Nucleares/genética , Adolescente , Adulto , Niño , Condrodisplasia Punctata Rizomélica/metabolismo , Exoma , Femenino , Humanos , Lactante , Masculino , Linaje , Receptor de la Señal 1 de Direccionamiento al Peroxisoma , Peroxisomas/genética , Isoformas de Proteínas , Receptores Citoplasmáticos y Nucleares/metabolismo , Análisis de Secuencia de ADN
12.
Orphanet J Rare Dis ; 8: 138, 2013 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-24016303

RESUMEN

BACKGROUND: Zellweger spectrum disorders (ZSDs) are multisystem genetic disorders caused by a lack of functional peroxisomes, due to mutations in one of the PEX genes, encoding proteins involved in peroxisome biogenesis. The phenotypic spectrum of ZSDs ranges from an early lethal form to much milder presentations. In cultured skin fibroblasts from mildly affected patients, peroxisome biogenesis can be partially impaired which results in a mosaic catalase immunofluorescence pattern. This peroxisomal mosaicism has been described for specific missense mutations in various PEX genes. In cell lines displaying peroxisomal mosaicism, peroxisome biogenesis can be improved when these are cultured at 30°C. This suggests that these missense mutations affect the folding and/or stability of the encoded protein. We have studied if the function of mutant PEX1, PEX6 and PEX12 can be improved by promoting protein folding using the chemical chaperone arginine. METHODS: Fibroblasts from three PEX1 patients, one PEX6 and one PEX12 patient were cultured in the presence of different concentrations of arginine. To determine the effect on peroxisome biogenesis we studied the following parameters: number of peroxisome-positive cells, levels of PEX1 protein and processed thiolase, and the capacity to ß-oxidize very long chain fatty acids and pristanic acid. RESULTS: Peroxisome biogenesis and function in fibroblasts with mild missense mutations in PEX1, 6 and 12 can be improved by arginine. CONCLUSION: Arginine may be an interesting compound to promote peroxisome function in patients with a mild peroxisome biogenesis disorder.


Asunto(s)
Arginina/farmacología , Trastorno Peroxisomal/metabolismo , Peroxisomas/efectos de los fármacos , Peroxisomas/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/genética , Proteínas de Arabidopsis/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Immunoblotting , Proteínas de la Membrana/genética , Microscopía Fluorescente , Peroxinas
13.
Biochim Biophys Acta ; 1822(9): 1430-41, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22871920

RESUMEN

Human peroxisome biogenesis disorders (PBDs) are a heterogeneous group of autosomal recessive disorders comprised of two clinically distinct subtypes: the Zellweger syndrome spectrum (ZSS) disorders and rhizomelic chondrodysplasia punctata (RCDP) type 1. PBDs are caused by defects in any of at least 14 different PEX genes, which encode proteins involved in peroxisome assembly and proliferation. Thirteen of these genes are associated with ZSS disorders. The genetic heterogeneity among PBDs and the inability to predict from the biochemical and clinical phenotype of a patient with ZSS which of the currently known 13 PEX genes is defective, has fostered the development of different strategies to identify the causative gene defects. These include PEX cDNA transfection complementation assays followed by sequencing of the thus identified PEX genes, and a PEX gene screen in which the most frequently mutated exons of the different PEX genes are analyzed. The benefits of DNA testing for PBDs include carrier testing of relatives, early prenatal testing or preimplantation genetic diagnosis in families with a recurrence risk for ZSS disorders, and insight in genotype-phenotype correlations, which may eventually assist to improve patient management. In this review we describe the current status of genetic analysis and the molecular basis of PBDs.


Asunto(s)
Trastorno Peroxisomal/genética , Peroxisomas/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/genética , Femenino , Estudios de Asociación Genética , Humanos , Proteínas de la Membrana/genética , Técnicas de Diagnóstico Molecular , Mutación , Trastorno Peroxisomal/diagnóstico , Trastorno Peroxisomal/metabolismo , Peroxisomas/fisiología , Embarazo , Diagnóstico Prenatal , Transporte de Proteínas
14.
J Med Genet ; 49(5): 307-13, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22581968

RESUMEN

BACKGROUND: Peroxisomes are organelles that proliferate continuously and play an indispensable role in human metabolism. Consequently, peroxisomal gene defects can cause multiple, often severe disorders, including the peroxisome biogenesis disorders. Currently, 13 different PEX proteins have been implicated in various stages of peroxisome assembly and protein import. Defects in any of these proteins result in a peroxisome biogenesis disorder. The authors present here a novel genetic defect specifically affecting the division of peroxisomes. METHODS: The authors have studied biochemical and microscopical peroxisomal parameters in cultured patient fibroblasts, sequenced candidate PEX genes and determined the consequence of the identified PEX11ß gene defect on peroxisome biogenesis in patient fibroblasts at different temperatures. RESULTS: The patient presented with congenital cataracts, mild intellectual disability, progressive hearing loss, sensory nerve involvement, gastrointestinal problems and recurrent migraine-like episodes. Although microscopical investigations of patient fibroblasts indicated a clear defect in peroxisome division, all biochemical parameters commonly used for diagnosing peroxisomal disorders were normal. After excluding mutations in all PEX genes previously implicated in peroxisome biogenesis disorders, it was found that the defect was caused by a homozygous non-sense mutation in the PEX11ß gene. The peroxisome division defect was exacerbated when the patient's fibroblasts were cultured at 40°C, which correlated with a marked decrease in the expression of PEX11γ. CONCLUSIONS: This novel isolated defect in peroxisome division expands the clinical and genetic spectrum of peroxisomal disorders and indicates that peroxisomal defects exist, which cannot be diagnosed by standard laboratory investigations.


Asunto(s)
Proteínas de la Membrana/genética , Mutación , Trastorno Peroxisomal/genética , Adulto , Secuencia de Bases , Estudios de Casos y Controles , Células Cultivadas , Análisis Mutacional de ADN , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Prueba de Complementación Genética , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Microscopía Fluorescente , Datos de Secuencia Molecular , Isoformas de Proteínas , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Temperatura , Transfección
15.
Hum Mutat ; 32(1): 59-69, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21031596

RESUMEN

The autosomal recessive Zellweger syndrome spectrum (ZSS) disorders comprise a main subgroup of the peroxisome biogenesis disorders and can be caused by mutations in any of 12 different currently identified PEX genes resulting in severe multisystemic disorders. To get insight into the spectrum of PEX gene defects among ZSS disorders and to investigate if additional human PEX genes are required for functional peroxisome biogenesis, we assigned over 600 ZSS fibroblast cell lines to different genetic complementation groups. These fibroblast cell lines were subjected to a complementation assay involving fusion by means of polyethylene glycol or a PEX cDNA transfection assay specifically developed for this purpose. In a majority of the cell lines we subsequently determined the underlying mutations by sequence analysis of the implicated PEX genes. The PEX cDNA transfection assay allows for the rapid identification of PEX genes defective in ZSS patients. The assignment of over 600 fibroblast cell lines to different genetic complementation groups provides the most comprehensive and representative overview of the frequency distribution of the different PEX gene defects. We did not identify any novel genetic complementation group, suggesting that all PEX gene defects resulting in peroxisome deficiency are currently known.


Asunto(s)
Mutación , Endopeptidasa Neutra Reguladora de Fosfato PHEX/genética , Síndrome de Zellweger/genética , Línea Celular , Frecuencia de los Genes , Prueba de Complementación Genética/estadística & datos numéricos , Genotipo , Humanos , Trastorno Peroxisomal/genética
16.
Ann Neurol ; 68(2): 259-63, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20695019

RESUMEN

Peroxisomal biogenesis disorders typically cause severe multisystem disease and early death. We describe a child and an adult of normal intelligence with progressive ataxia, axonal motor neuropathy, and decreased vibration sense. Both patients had marked cerebellar atrophy. Peroxisomal studies revealed a peroxisomal biogenesis disorder. Two mutations in PEX10 were found in the child, c.992G>A (novel) and c.764_765insA, and in the adult, c.2T>C (novel) and c.790C>T. Transfection with wild-type PEX10 corrected the fibroblast phenotype. Bile acid supplements and dietary restriction of phytanic acid were started. Peroxisomal biogenesis disorders should be considered in the differential diagnosis of autosomal recessive ataxia.


Asunto(s)
Trastornos de los Cromosomas/genética , Genes Recesivos/genética , Predisposición Genética a la Enfermedad/genética , Mutación/genética , Receptores Citoplasmáticos y Nucleares/genética , Degeneraciones Espinocerebelosas/genética , Células Cultivadas , Niño , Trastornos de los Cromosomas/diagnóstico , Trastornos de los Cromosomas/dietoterapia , Humanos , Masculino , Mosaicismo , Peroxinas , Degeneraciones Espinocerebelosas/diagnóstico , Degeneraciones Espinocerebelosas/dietoterapia , Adulto Joven
17.
Am J Med Genet A ; 152A(9): 2318-21, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20683989

RESUMEN

Peroxisomal biogenesis disorders (PBD) are groups of inherited neurometabolic disorders caused by defects in PEX genes. We report on a female infant, born to a consanguineous parents (first degree cousins), who presented with inactivity, poor sucking, and hypotonia early in the neonatal period. She had subtle dysmorphic features. Liver function tests were impaired with raised liver enzymes, conjugated and unconjugated hyperbilirubinemia. CT of the brain showed diffuse bilateral changes. She developed seizures with an abnormal EEG. Plasma very long chain fatty acid analysis showed high C26:0 levels and increasedC26:0/C22:0 and C24:0/C22:0 ratios, which is consistent with a PBD. Studies in fibroblasts including plasmalogen biosynthesis, peroxisomal fatty acid alfa and beta oxidation confirmed the diagnosis of PBD. Immunofluoresence microscopy revealed the absence of peroxisomes in fibroblasts. The patient was assigned to the PEX19 complementation group. Subsequent mutation analysis of the PEX19 gene revealed homozygosity for a c.320delA frameshift mutation. The patient had a stormy course with multiple admissions to the pediatric intensive care unit with pneumonia, liver impairment, sepsis, and epilepsy. At 1 year of age she developed metabolic acidosis with normal anion gap, proteinuria, aminoaciduria, and glucosuria consistent with a renal tubular defect. Abdominal ultrasound showed multiple gallstones. Other causes of gallstones like haemoglobinopathy were excluded. So far, only two siblings had been reported with mutations in the PEX19 gene. Our patient showed a previously unrecognized association of gallstones and a renal tubular defect with a PBD.


Asunto(s)
Mutación del Sistema de Lectura , Proteínas de la Membrana/genética , Trastorno Peroxisomal/genética , Femenino , Cálculos Biliares , Humanos , Lactante , Túbulos Renales/anomalías , Trastorno Peroxisomal/diagnóstico , Trastorno Peroxisomal/patología , Fenotipo
18.
J Med Genet ; 47(9): 608-15, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20647552

RESUMEN

BACKGROUND: Zellweger syndrome spectrum disorders are caused by mutations in any of at least 12 different PEX genes. This includes PEX16, which encodes an integral peroxisomal membrane protein involved in peroxisomal membrane assembly. PEX16-defective patients have been reported to have a severe clinical presentation. Fibroblasts from these patients displayed a defect in the import of peroxisomal matrix and membrane proteins, resulting in a total absence of peroxisomal remnants. OBJECTIVE: To report on six patients with an unexpected mild variant peroxisome biogenesis disorder due to mutations in the PEX16 gene. Patients presented in the preschool years with progressive spastic paraparesis and ataxia (with a characteristic pattern of progressive leucodystrophy and brain atrophy on MRI scan) and later developed cataracts and peripheral neuropathy. Surprisingly, their fibroblasts showed enlarged, import-competent peroxisomes. RESULTS: Plasma analysis revealed biochemical abnormalities suggesting a peroxisomal disorder. Biochemical variables in fibroblasts were only mildly abnormal or within the normal range. Immunofluorescence microscopy revealed the presence of import-competent peroxisomes, which were increased in size but reduced in number. Subsequent sequencing of all known PEX genes revealed five novel apparent homozygous mutations in the PEX16 gene. CONCLUSIONS: An unusual variant peroxisome biogenesis disorder caused by mutations in the PEX16 gene, with a relatively mild clinical phenotype and an unexpected phenotype in fibroblasts, was identified. Although PEX16 is involved in peroxisomal membrane assembly, PEX16 defects can present with enlarged import-competent peroxisomes in fibroblasts. This is important for future diagnostics of patients with a peroxisomal disorder.


Asunto(s)
Proteínas de la Membrana/genética , Mutación/genética , Peroxisomas/genética , Peroxisomas/patología , Adolescente , Catalasa/metabolismo , Niño , Preescolar , Análisis Mutacional de ADN , Eritrocitos/metabolismo , Femenino , Fibroblastos/enzimología , Fibroblastos/patología , Técnica del Anticuerpo Fluorescente , Prueba de Complementación Genética , Humanos , Lactante , Recién Nacido , Imagen por Resonancia Magnética , Masculino
19.
Hum Mutat ; 31(1): E1058-70, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19877282

RESUMEN

The autosomal recessive Zellweger syndrome spectrum (ZSS) disorders comprise a main subgroup of the peroxisome biogenesis disorders. The ZSS disorders can be caused by mutations in any of 12 different currently identified PEX genes resulting in severe, often lethal, multi-systemic disorders. Defects in the PEX6 gene are the second most common cause for ZSS disorders. The encoded protein PEX6 belongs to the AAA ATPase family and contains two AAA cassettes and an AAA protein family signature. The PEX6 gene consists of 17 exons and previously mutations in the PEX6 gene were found to be scattered over all exons. We developed a post-PCR high-resolution melting (HRM) curve assay to scan the PEX6 gene for potential sequence variations followed by selective sequencing to identify these. We analyzed the PEX6 genes of 75 patients assigned to the PEX6 complementation group. We identified a total of 77 different mutations of which 47 mutations have not been reported previously, and 14 polymorphic variants.


Asunto(s)
Adenosina Trifosfatasas/genética , Mutación , Reacción en Cadena de la Polimerasa/métodos , Síndrome de Zellweger/genética , ATPasas Asociadas con Actividades Celulares Diversas , Línea Celular , Células Cultivadas , Exones/genética , Fibroblastos , Humanos , Peroxisomas/metabolismo , Polimorfismo Genético , Análisis de Secuencia de ADN/métodos , Transfección , Temperatura de Transición
20.
Hum Mutat ; 30(1): 93-8, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18712838

RESUMEN

Proteins destined for the peroxisomal matrix are targeted by virtue of a peroxisomal targeting sequence type 1 (PTS1) or type 2 (PTS2). In humans, targeting of either class of proteins relies on a cytosolic receptor protein encoded by the PEX5 gene. Alternative splicing of PEX5 results in two protein variants, PEX5S and PEX5L. PEX5S is exclusively involved in PTS1 protein import, whereas PEX5L mediates the import of both PTS1 and PTS2 proteins. Genetic complementation testing with over 500 different fibroblast cell lines from patients diagnosed with a peroxisome biogenesis disorder (PBD) identified 11 cell lines with a defect in PEX5. The aim of this study was to characterize these cell lines at a biochemical and genetic level. To this end, the cultured fibroblasts were analyzed for very long chain fatty acid (VLCFA) concentrations, peroxisomal beta-and alpha-oxidation, dihydroxyacetone-phosphate acyltransferase (DHAPAT) activity, peroxisomal thiolase, and catalase immunofluorescence. Mutation analysis of the PEX5 gene revealed 11 different mutations, eight of which are novel. PTS1- and PTS2-protein import capacity was assessed by transfection of the cells with green fluorescent protein (GFP) tagged with either PTS1 or PTS2. Six cell lines showed a defect in both PTS1 and PTS2 protein import, whereas four cell lines only showed a defect in PTS1 protein import. The location of the different mutations within the PEX5 amino acid sequence correlates rather well with the peroxisomal protein import defect observed in the cell lines.


Asunto(s)
Genotipo , Trastorno Peroxisomal/genética , Peroxisomas/metabolismo , Fenotipo , Receptores Citoplasmáticos y Nucleares/genética , Secuencia de Aminoácidos , Línea Celular , Análisis Mutacional de ADN , Fibroblastos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Mutación , Trastorno Peroxisomal/metabolismo , Receptor de la Señal 1 de Direccionamiento al Peroxisoma , Transporte de Proteínas , Receptores Citoplasmáticos y Nucleares/metabolismo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...