Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Brain Dev ; 43(3): 475-481, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33234382

RESUMEN

Peroxisomal acyl-CoA oxidase (ACOX1) deficiency is a rare autosomal recessive single enzyme deficiency characterized by hypotonia, seizures, failure to thrive, developmental delay, and neurological regression starting from approximately 3 years of age. Here, we report two siblings with ACOX1 deficiency born to non-consanguineous Japanese parents. They showed mild global developmental delay from infancy and began to regress at 5 years 10 months and 5 years 6 months of age respectively. They gradually manifested with cerebellar ataxia, dysarthria, pyramidal signs, and dysphasia. Brain MRI revealed T2 high-intensity areas in the cerebellar white matter, bilateral middle cerebellar peduncle, and transverse tracts of the pons, followed by progressive atrophy of these areas. Intriguingly, the ratios of C24:0, C25:0, and C26:0 to C22:0 in plasma, which usually increase in ACOX1 deficiency were within normal ranges in both patients. On the other hand, whole exome sequencing revealed novel compound heterozygous variants in ACOX1: a frameshift variant (c.160delC:p.Leu54Serfs*18) and a missense variant (c.1259 T > C:p.Phe420Ser). The plasma concentration of individual very long chain fatty acids (C24:0, C25:0, and C26:0) was elevated, and we found that peroxisomes in fibroblasts of the patients were larger in size and fewer in number as previously reported in patients with ACOX1 deficiency. Furthermore, the C24:0 ß-oxidation activity was dramatically reduced. Our findings suggest that the elevation of individual plasma very long chain fatty acids concentration, genetic analysis including whole exome analysis, and biochemical studies on the patient's fibroblasts should be considered for the correct diagnosis of ACOX1 deficiency.


Asunto(s)
Acil-CoA Oxidasa/deficiencia , Acil-CoA Oxidasa/genética , Adrenoleucodistrofia/genética , Errores Innatos del Metabolismo Lipídico/genética , Femenino , Humanos , Masculino , Mutación , Hermanos
2.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(4): 567-576, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30312667

RESUMEN

Acyl-CoA oxidase 1 (ACOX1) deficiency is a rare and severe peroxisomal leukodystrophy associated with a very long-chain fatty acid (VLCFA) ß-oxidation defect. This neurodegenerative disease lacks relevant cell models to further decipher the pathomechanisms in order to identify novel therapeutic targets. Since peroxisomal defects in microglia appear to be a key component of peroxisomal leukodystrophies, we targeted the Acox1 gene in the murine microglial BV-2 cell line. Using CRISPR/Cas9 gene editing, we generated an Acox1-deficient cell line and validated the allelic mutations, which lead to the absence of ACOX1 protein and enzymatic activity. The activity of catalase, the enzyme degrading H2O2, was increased, likely in response to the alteration of redox homeostasis. The mutant cell line grew more slowly than control cells without obvious morphological changes. However, ultrastructural analysis revealed an increased number of peroxisomes and mitochondria associated with size reduction of mitochondria. Changes in the distribution of lipid droplets containing neutral lipids have been observed in mutant cells; lipid analysis revealed the accumulation of saturated and monounsaturated VLCFA. Besides, expression levels of genes encoding interleukin-1 beta and 6 (IL-1ß and IL-6), as well as triggering receptor expressed on myeloid cells 2 (Trem2) were found modified in the mutant cells suggesting modification of microglial polarization and phagocytosis ability. In summary, this Acox1-deficient cell line presents the main biochemical characteristics of the human disease and will serve as a promising model to further investigate the consequences of a specific microglial peroxisomal ß-oxidation defect on oxidative stress, inflammation and cellular functions.


Asunto(s)
Acil-CoA Oxidasa/deficiencia , Microglía/citología , Modelos Biológicos , Mutación , Enfermedades Neurodegenerativas/genética , Acil-CoA Oxidasa/genética , Animales , Sistemas CRISPR-Cas , Línea Celular , Proliferación Celular , Ácidos Grasos/metabolismo , Ácidos Grasos Insaturados/metabolismo , Edición Génica , Peróxido de Hidrógeno/metabolismo , Ratones , Microglía/metabolismo , Estrés Oxidativo
3.
Clin Liver Dis ; 22(4): 671-687, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30266156

RESUMEN

Inborn errors of bile acid metabolism are rare causes of neonatal cholestasis and liver disease in older children and adults. The diagnosis should be considered in the context of hyperbilirubinemia with normal serum bile acids and made by urinary liquid secondary ionization mass spectrometry or DNA testing. Cholic acid is an effective treatment of most single-enzyme defects and patients with Zellweger spectrum disorder with liver disease.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Colestasis/etiología , Hepatopatías/etiología , Errores Innatos del Metabolismo/diagnóstico , Errores Innatos del Metabolismo/genética , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/deficiencia , Acil-CoA Oxidasa/deficiencia , Hiperplasia Suprarrenal Congénita/complicaciones , Hiperplasia Suprarrenal Congénita/genética , N-Acetiltransferasa de Aminoácidos/deficiencia , Ácido Cólico/uso terapéutico , Pruebas Genéticas , Humanos , Hepatopatías/patología , Errores Innatos del Metabolismo/complicaciones , Errores Innatos del Metabolismo/tratamiento farmacológico , Racemasas y Epimerasas/deficiencia , Esteroide Hidroxilasas/deficiencia , Xantomatosis Cerebrotendinosa/complicaciones , Xantomatosis Cerebrotendinosa/genética
4.
Methods Mol Biol ; 1595: 329-342, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28409475

RESUMEN

The peroxisomal disorders (PDs) are a heterogeneous group of genetic diseases in man caused by an impairment in peroxisome biogenesis or one of the metabolic functions of peroxisomes. Thanks to the revolutionary technical developments in gene sequencing methods and their increased use in patient diagnosis, the field of genetic diseases in general and peroxisomal disorders in particular has dramatically changed in the last few years. Indeed, several novel peroxisomal disorders have been identified recently and in addition it has been realized that the phenotypic spectrum of patients affected by a PD keeps widening, which makes clinical recognition of peroxisomal patients increasingly difficult. Here, we describe these new developments and provide guidelines for the clinical and laboratory diagnosis of peroxisomal patients.


Asunto(s)
Trastorno Peroxisomal/diagnóstico , Trastorno Peroxisomal/genética , Acil-CoA Oxidasa/deficiencia , Acil-CoA Oxidasa/genética , Pruebas Genéticas , Humanos , Trastorno Peroxisomal/metabolismo , Proteína-2 Multifuncional Peroxisomal/deficiencia , Proteína-2 Multifuncional Peroxisomal/genética , Proteína-2 Multifuncional Peroxisomal/metabolismo , Peroxisomas/genética , Peroxisomas/metabolismo , Fenotipo
5.
Mol Genet Metab ; 114(4): 599-603, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25724074

RESUMEN

PURPOSE: To describe a diagnostic protocol, surveillance and treatment guidelines, genetic counseling considerations and long-term follow-up data elements developed in preparation for X-linked adrenoleukodystrophy (X-ALD) newborn screening in New York State. METHODS: A group including the director from each regional NYS inherited metabolic disorder center, personnel from the NYS Newborn Screening Program, and others prepared a follow-up plan for X-ALD NBS. Over the months preceding the start of screening, a series of conference calls took place to develop and refine a complete newborn screening system from initial positive screen results to long-term follow-up. RESULTS: A diagnostic protocol was developed to determine for each newborn with a positive screen whether the final diagnosis is X-ALD, carrier of X-ALD, Zellweger spectrum disorder, acyl CoA oxidase deficiency or D-bifunctional protein deficiency. For asymptomatic males with X-ALD, surveillance protocols were developed for use at the time of diagnosis, during childhood and during adulthood. Considerations for timing of treatment of adrenal and cerebral disease were developed. CONCLUSION: Because New York was the first newborn screening laboratory to include X-ALD on its panel, and symptoms may not develop for years, long-term follow-up is needed to evaluate the presented guidelines.


Asunto(s)
Adrenoleucodistrofia/diagnóstico , Tamizaje Neonatal , Acil-CoA Oxidasa/deficiencia , Insuficiencia Suprarrenal/diagnóstico , Algoritmos , Asesoramiento Genético , Humanos , Recién Nacido , Masculino , New York , Trastorno Peroxisomal/diagnóstico , Proteína-2 Multifuncional Peroxisomal/deficiencia , Síndrome de Zellweger/diagnóstico
6.
J Inherit Metab Dis ; 37(5): 791-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24619150

RESUMEN

OBJECTIVE: Acyl-CoA oxidase (ACOX1) deficiency is a rare disorder of peroxisomal very-long chain fatty acid oxidation. No reports detailing attempted treatment, longitudinal imaging, or neuropathology exist. We describe the natural history of clinical symptoms and brain imaging in two siblings with ACOX1 deficiency, including the younger sibling's response to allogeneic unrelated donor hematopoietic stem cell transplantation (HSCT). METHODS: We conducted retrospective chart review to obtain clinical history, neuro-imaging, and neuropathology data. ACOX1 genotyping were performed to confirm the disease. In vitro fibroblast and neural stem cell fatty acid oxidation assays were also performed. RESULTS: Both patients experienced a fatal neurodegenerative course, with late-stage cerebellar and cerebral gray matter atrophy. Serial brain magnetic resonance imaging in the younger sibling indicated demyelination began in the medulla and progressed rostrally to include the white matter of the cerebellum, pons, midbrain, and eventually subcortical white matter. The successfully engrafted younger sibling had less brain inflammation, cortical atrophy, and neuronal loss on neuro-imaging and neuropathology compared to the untreated older sister. Fibroblasts and stem cells demonstrated deficient very long chain fatty acid oxidation. INTERPRETATION: Although HSCT did not halt the course of ACOX1 deficiency, it reduced the extent of white matter inflammation in the brain. Demyelination continued because of ongoing neuronal loss, which may be due to inability of transplant to prevent progression of gray matter disease, adverse effects of chronic corticosteroid use to control graft-versus-host disease, or intervention occurring beyond a critical point for therapeutic efficacy.


Asunto(s)
Acil-CoA Oxidasa/deficiencia , Encefalopatías Metabólicas Innatas/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Encéfalo/patología , Encefalopatías Metabólicas Innatas/enzimología , Encefalopatías Metabólicas Innatas/patología , Preescolar , Resultado Fatal , Femenino , Movimientos de la Cabeza/fisiología , Humanos , Lactante , Recién Nacido , Masculino , Hipotonía Muscular/etiología , Células-Madre Neurales/trasplante , Hermanos , Resultado del Tratamiento
7.
PLoS One ; 9(1): e84853, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24465440

RESUMEN

Fatty acid-derived biofuels and biochemicals can be produced in microbes using ß-oxidation pathway engineering. In this study, the ß-oxidation pathway of Saccharomyces cerevisiae was engineered to accumulate a higher ratio of medium chain fatty acids (MCFAs) when cells were grown on fatty acid-rich feedstock. For this purpose, the haploid deletion strain Δpox1 was obtained, in which the sole acyl-CoA oxidase encoded by POX1 was deleted. Next, the POX2 gene from Yarrowia lipolytica, which encodes an acyl-CoA oxidase with a preference for long chain acyl-CoAs, was expressed in the Δpox1 strain. The resulting Δpox1 [pox2+] strain exhibited a growth defect because the ß-oxidation pathway was blocked in peroxisomes. To unblock the ß-oxidation pathway, the gene CROT, which encodes carnitine O-octanoyltransferase, was expressed in the Δpox1 [pox2+] strain to transport the accumulated medium chain acyl-coAs out of the peroxisomes. The obtained Δpox1 [pox2+, crot+] strain grew at a normal rate. The effect of these genetic modifications on fatty acid accumulation and profile was investigated when the strains were grown on oleic acids-containing medium. It was determined that the engineered strains Δpox1 [pox2+] and Δpox1 [pox2+, crot+] had increased fatty acid accumulation and an increased ratio of MCFAs. Compared to the wild-type (WT) strain, the total fatty acid production of the strains Δpox1 [pox2+] and Δpox1 [pox2+, crot+] were increased 29.5% and 15.6%, respectively. The intracellular level of MCFAs in Δpox1 [pox2+] and Δpox1 [pox2+, crot+] increased 2.26- and 1.87-fold compared to the WT strain, respectively. In addition, MCFAs in the culture medium increased 3.29-fold and 3.34-fold compared to the WT strain. These results suggested that fatty acids with an increased MCFAs ratio accumulate in the engineered strains with a modified ß-oxidation pathway. Our approach exhibits great potential for transforming low value fatty acid-rich feedstock into high value fatty acid-derived products.


Asunto(s)
Acilcoenzima A/metabolismo , Ácidos Grasos/biosíntesis , Ingeniería Metabólica , Saccharomyces cerevisiae/genética , Acil-CoA Oxidasa/deficiencia , Acil-CoA Oxidasa/genética , Biocombustibles , Carnitina Aciltransferasas/genética , Carnitina Aciltransferasas/metabolismo , Citoplasma/enzimología , Isoenzimas/genética , Isoenzimas/metabolismo , Oxidación-Reducción , Peroxisomas/enzimología , Saccharomyces cerevisiae/enzimología , Proteínas de Saccharomyces cerevisiae/genética , Transgenes , Yarrowia/química , Yarrowia/enzimología
8.
Endocrinology ; 153(6): 2568-75, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22508517

RESUMEN

Among several peroxisomal neurodegenerative disorders, the pseudoneonatal adrenoleukodystrophy (P-NALD) is characterized by the acyl-coenzyme A oxidase 1 (ACOX1) deficiency, which leads to the accumulation of very-long-chain fatty acids (VLCFA) and inflammatory demyelination. However, the components of this inflammatory process in P-NALD remain elusive. In this study, we used transcriptomic profiling and PCR array analyses to explore inflammatory gene expression in patient fibroblasts. Our results show the activation of IL-1 inflammatory pathway accompanied by the increased secretion of two IL-1 target genes, IL-6 and IL-8 cytokines. Human fibroblasts exposed to very-long-chain fatty acids exhibited increased mRNA expression of IL-1α and IL-1ß cytokines. Furthermore, expression of IL-6 and IL-8 cytokines in patient fibroblasts was down-regulated by MAPK, p38MAPK, and Jun N-terminal kinase inhibitors. Thus, the absence of acyl-coenzyme A oxidase 1 activity in P-NALD fibroblasts triggers an inflammatory process, in which the IL-1 pathway seems to be central. The use of specific kinase inhibitors may permit the modulation of the enhanced inflammatory status.


Asunto(s)
Acil-CoA Oxidasa/genética , Fibroblastos/metabolismo , Inflamación/genética , Transcriptoma , Acil-CoA Oxidasa/deficiencia , Acil-CoA Oxidasa/metabolismo , Células Cultivadas , Ácidos Grasos/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Interleucina-1/genética , Interleucina-1/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteopontina/genética , Osteopontina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
J Cell Sci ; 125(Pt 3): 589-602, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22389399

RESUMEN

Peroxisome division is regulated by several factors, termed fission factors, as well as the conditions of the cellular environment. Over the past decade, the idea of metabolic control of peroxisomal morphogenesis has been postulated, but remains largely undefined to date. In the current study, docosahexaenoic acid (DHA, C22:6n-3) was identified as an inducer of peroxisome division. In fibroblasts isolated from patients that carry defects in peroxisomal fatty acid ß-oxidation, peroxisomes are much less abundant than normal cells. Treatment of these patient fibroblasts with DHA induced the proliferation of peroxisomes to the level seen in normal fibroblasts. DHA-induced peroxisomal proliferation was abrogated by treatment with a small inhibitory RNA (siRNA) targeting dynamin-like protein 1 and with dynasore, an inhibitor of dynamin-like protein 1, which suggested that DHA stimulates peroxisome division. DHA augmented the hyper-oligomerization of Pex11pß and the formation of Pex11pß-enriched regions on elongated peroxisomes. Time-lapse imaging analysis of peroxisomal morphogenesis revealed a sequence of steps involved in peroxisome division, including elongation in one direction followed by peroxisomal fission. DHA enhanced peroxisomal division in a microtubule-independent manner. These results suggest that DHA is a crucial signal for peroxisomal elongation, a prerequisite for subsequent fission and peroxisome division.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Peroxisomas/efectos de los fármacos , 3-Hidroxiacil-CoA Deshidrogenasas/deficiencia , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Acil-CoA Oxidasa/deficiencia , Acil-CoA Oxidasa/metabolismo , Secuencia de Bases , Células Cultivadas , Ácidos Docosahexaenoicos/metabolismo , Dinaminas , Enoil-CoA Hidratasa/deficiencia , Enoil-CoA Hidratasa/metabolismo , Ácidos Grasos/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , GTP Fosfohidrolasas/antagonistas & inhibidores , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Humanos , Isomerasas/deficiencia , Isomerasas/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Modelos Biológicos , Complejos Multienzimáticos/deficiencia , Complejos Multienzimáticos/metabolismo , Oxidación-Reducción , Enzima Bifuncional Peroxisomal , Trastorno Peroxisomal/metabolismo , Trastorno Peroxisomal/patología , Peroxisomas/metabolismo , Peroxisomas/ultraestructura , Multimerización de Proteína/efectos de los fármacos , ARN Interferente Pequeño/genética , Imagen de Lapso de Tiempo
10.
Biochim Biophys Acta ; 1822(9): 1489-500, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22446031

RESUMEN

Peroxisome biogenesis and peroxisomal ß-oxidation defects are rare inherited metabolic disorders in which several organs can be affected. A panel of mouse models has been created in which genes crucial to these processes were inactivated and the ensuing pathologies studied. In mice with enzyme defects of peroxisomal ß-oxidation, the disease state strongly depends on the kind of substrates that are metabolized by the enzyme and the dietary composition. Because mice with generalized biogenesis defects seldom reach adulthood, conditional knockout models were generated to study the consequences of peroxisome deficiency in hepatocytes, different brain cell types and Sertoli cells. Although the precise relationship between the biochemical anomalies and pathologies was often not resolved, the mouse models allowed to document in detail histological abnormalities, metabolic and gene expression deregulations some of which are mediated by PPARα, and to uncover the essential role of peroxisomes in some unsuspected cell types.


Asunto(s)
Modelos Animales de Enfermedad , Metabolismo de los Lípidos , Trastorno Peroxisomal/enzimología , Peroxisomas/enzimología , Acil-CoA Oxidasa/deficiencia , Acil-CoA Oxidasa/genética , Aciltransferasas/deficiencia , Aciltransferasas/genética , Animales , Encéfalo/enzimología , Encéfalo/patología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Enoil-CoA Hidratasa/deficiencia , Enoil-CoA Hidratasa/genética , Humanos , Hígado/enzimología , Ratones , Oxidación-Reducción , Racemasas y Epimerasas/deficiencia , Racemasas y Epimerasas/genética
11.
FASEB J ; 26(2): 628-38, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22009939

RESUMEN

Obesity, a major health concern, results from an imbalance between energy intake and expenditure. Leptin-deficient ob/ob mice are paradigmatic of obesity, resulting from excess energy intake and storage. Mice lacking acyl-CoA oxidase 1 (Acox1), the first enzyme of the peroxisomal fatty acid ß-oxidation system, are characterized by increased energy expenditure and a lean body phenotype caused by sustained activation of peroxisome proliferator-activated receptor α (PPARα) by endogenous ligands in liver that remain unmetabolized in the absence of Acox1. We generated ob/ob mice deficient in Acox1 (Acox1(-/-)) to determine how the activation of PPARα by endogenous ligands might affect the obesity of ob/ob mice. In contrast to Acox1(-/-) (14.3±1.2 g at 6 mo) and the Acox1-deficient (ob/ob) double-mutant mice (23.8±4.6 g at 6 mo), the ob/ob mice are severely obese (54.3±3.2 g at 6 mo) and had significantly more (P<0.01) epididymal fat content. The resistance of Acox1(-/-)/ob/ob mice to obesity is due to increased PPARα-mediated up-regulation of genes involved in fatty acid oxidation in liver. Activation of PPARα in Acox1-deficient ob/ob mice also reduces serum glucose and insulin (P<0.05) and improves glucose tolerance and insulin sensitivity. Further, PPARα activation reduces hepatic steatosis and increases hepatocellular regenerative response in Acox1(-/-)/ob/ob mice at a more accelerated pace than in mice lacking only Acox1. However, Acox1(-/-)/ob/ob mice manifest hepatic endoplasmic reticulum (ER) stress and also develop hepatocellular carcinomas (8 of 8 mice) similar to those observed in Acox1(-/-) mice (10 of 10 mice), but unlike in ob/ob (0 of 14 mice) and OB/OB (0 of 6 mice) mice, suggesting that superimposed ER stress and PPARα activation contribute to carcinogenesis in a fatty liver. Finally, absence of Acox1 in ob/ob mice can impart resistance to high-fat diet (60% fat)-induced obesity, and their liver had significantly (P<0.01) more cell proliferation. These studies with Acox1(-/-)/ob/ob mice indicate that sustained activation of lipid-sensing nuclear receptor PPARα attenuates obesity and restores glucose homeostasis by ameliorating insulin resistance but increases the risk for liver cancer development, in part related to excess energy combustion.


Asunto(s)
Ácidos Grasos/metabolismo , Hígado/metabolismo , Obesidad/metabolismo , Obesidad/prevención & control , PPAR alfa/metabolismo , Acil-CoA Oxidasa/deficiencia , Acil-CoA Oxidasa/genética , Acil-CoA Oxidasa/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN/genética , Retículo Endoplásmico/metabolismo , Metabolismo Energético , Hígado Graso/metabolismo , Hígado Graso/patología , Hígado Graso/prevención & control , Femenino , Resistencia a la Insulina , Leptina/deficiencia , Leptina/genética , Ligandos , Neoplasias Hepáticas Experimentales/etiología , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/metabolismo , Regeneración Hepática , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/genética , Oxidación-Reducción , PPAR alfa/agonistas , Estrés Fisiológico
12.
Am J Pathol ; 179(2): 703-13, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21801867

RESUMEN

Fatty acyl-coenzyme A oxidase 1 (ACOX1) knockout (ACOX1(-/-)) mice manifest hepatic metabolic derangements that lead to the development of steatohepatitis, hepatocellular regeneration, spontaneous peroxisome proliferation, and hepatocellular carcinomas. Deficiency of ACOX1 results in unmetabolized substrates of this enzyme that function as biological ligands for peroxisome proliferator-activated receptor-α (PPARα) in liver. Here we demonstrate that sustained activation of PPARα in ACOX1(-/-) mouse liver by these ACOX1 substrates results in endoplasmic reticulum (ER) stress. Overexpression of transcriptional regulator p8 and its ER stress-related effectors such as the pseudokinase tribbles homolog 3, activating transcription factor 4, and transcription factor CCAAT/-enhancer-binding protein homologous protein as well as phosphorylation of eukaryotic translation initiation factor 2α, indicate the induction of unfolded protein response signaling in the ACOX1(-/-) mouse liver. We also show here that, in the liver, p8 is a target for all three PPAR isoforms (-α, -ß, and -γ), which interact with peroxisome proliferator response elements in p8 promoter. Sustained activation of p8 and unfolded protein response-associated ER stress in ACOX1(-/-) mouse liver contributes to hepatocyte apoptosis and liver cell proliferation culminating in the development of hepatocarcinogenesis. We also demonstrate that human ACOX1 transgene is functional in ACOX1(-/-) mice and effectively prevents metabolic dysfunctions that lead to ER stress and carcinogenic effects. Taken together, our data indicate that progressive PPARα- and p8-mediated ER stress contribute to the hepatocarcinogenesis in ACOX1(-/-) mice.


Asunto(s)
Acil-CoA Oxidasa/deficiencia , Proteínas de Unión al ADN/genética , Retículo Endoplásmico/metabolismo , Neoplasias Hepáticas/patología , Proteínas de Neoplasias/genética , PPAR alfa/genética , Acil-CoA Oxidasa/genética , Animales , Cartilla de ADN/genética , Regulación de la Expresión Génica , Genotipo , Hepatocitos/citología , Humanos , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Transgenes
13.
Am J Med Genet A ; 146A(13): 1676-81, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18536048

RESUMEN

We report on two new patients with straight-chain acyl-coenzyme A oxidase deficiency. Early onset hypotonia, seizures and psychomotor delay were observed in both cases. Plasma very-long-chain fatty acids were abnormal in both patients, whereas the plasma levels of phytanic acid, pristanic acid, the bile acid intermediates DHCA and THCA, and erythrocyte plasmalogen levels were normal. Studies in fibroblasts from the two patients revealed a deficiency of one of the two peroxisomal acyl-CoA oxidases, that is, straight-chain acyl-CoA oxidase (ACOX1). Subsequent molecular analysis of ACOX1 showed a homozygous deletion, which removes a large part of intron 3 and exons 4-14 in the first patient. Mutation analysis in the second patient revealed compound heterozygosity for two mutations, including: (1) a c.692 G > T (p.G231V) mutation and (2) skipping of exon 13 (c.1729_1935del (p.G577_E645del).


Asunto(s)
Acil-CoA Oxidasa/deficiencia , Acil-CoA Oxidasa/genética , Trastorno Peroxisomal/enzimología , Trastorno Peroxisomal/genética , Secuencia de Bases , Encéfalo/anomalías , Preescolar , Consanguinidad , ADN/genética , Femenino , Genes Recesivos , Homocigoto , Humanos , Recién Nacido , Imagen por Resonancia Magnética , Trastorno Peroxisomal/diagnóstico , Peroxisomas/enzimología , Embarazo , Diagnóstico Prenatal , Eliminación de Secuencia
14.
Neuropediatrics ; 37(2): 95-8, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16773508

RESUMEN

We present a rare case of peroxisomal acyl-CoA oxidase deficiency that was not detected by the common metabolic screening program for peroxisomal disorders. The patient presented with a typical MRI pattern showing pachygyria, perisylvian polymicrogyria, cerebral and cerebellar white matter abnormalities, and facial dysmorphia, progressive psychomotor retardation, deafness, retinopathy, peripheral neuropathy, and infantile seizures strongly indicative for a peroxisomal disorder. Yet, repetitive measurements of very long-chain fatty acids (VLCFAs) and phytanic acid in serum and plasma as well as plasmalogens in erythrocytes revealed normal values apparently excluding a peroxisomal defect (methods of measurement published by Moser and co-workers in 1980 [4 ] and 1981 [2 ]). Subsequent biochemical investigation in cultured skin fibroblasts of the patient, however, revealed elevated concentrations of VLCFAs, deficient oxidation of C26:0, but normal oxidation of both phytanic acid and pristanic acid and normal DE NOVO plasmalogen synthesis, indicative for a defect in the peroxisomal beta-oxidation system. Enzymatic studies in these fibroblasts pointed to peroxisomal acyl-CoA oxidase deficiency and subsequent molecular analyses revealed a homozygous acceptor splice site mutation IVS3-1G>A in the ACOX1 gene (MIM *609751).


Asunto(s)
Acil-CoA Oxidasa/deficiencia , Ácidos Grasos/metabolismo , Trastorno Peroxisomal/diagnóstico , Trastorno Peroxisomal/metabolismo , Acil-CoA Oxidasa/genética , Humanos , Lactante , Masculino , Trastorno Peroxisomal/genética
15.
Brain Dev ; 28(5): 287-92, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16376506

RESUMEN

Peroxisomes are ubiquitous organelles in eukaryotic cells and surrounded by a single membrane, and undergo considerable changes in size, shape and number. Peroxisomal disorders are classified into two categories: peroxisome biogenesis disorders (PBDs) and single-enzyme deficiencies (SEDs). Morphologically aberrant peroxisomes called 'peroxisomal ghosts' in PBDs are well known, however, a morphological approach to the study of peroxisomes in SEDs has been rarely reported. Here, we investigated the morphology of peroxisomes in cultured fibroblasts from patients lacking peroxisomal beta-oxidation enzymes, including acyl-CoA oxidase (AOX) or D-3-hydroxyacyl-CoA dehydratase/D-3-hydroxyacyl-CoA dehydrogenase bifunctional protein (D-BP). Morphological analysis by immunofluorescence examination using an antibody against catalase revealed a smaller number of large peroxisomes in fibroblasts from these patients. Moreover, immunoelectron microscopy using an antibody against the 70-kDa peroxisomal membrane protein (PMP70) showed large peroxisomes with various horseshoe-shaped membrane structures. These results give an important clue to elucidating the division of peroxisomes and how peroxisomes change in size, shape, number and position within cells, which are subjects for future study.


Asunto(s)
Fibroblastos/citología , Trastorno Peroxisomal/enzimología , Trastorno Peroxisomal/patología , Peroxisomas/enzimología , Peroxisomas/patología , 3-Hidroxiacil-CoA Deshidrogenasas/deficiencia , Acil-CoA Oxidasa/deficiencia , Adulto , Células Cultivadas , Preescolar , Análisis Mutacional de ADN/métodos , Enoil-CoA Hidratasa/deficiencia , Técnica del Anticuerpo Fluorescente/métodos , Humanos , Hidroliasas/deficiencia , Lactante , Isomerasas/deficiencia , Microscopía Electrónica de Transmisión/métodos , Complejos Multienzimáticos/deficiencia , Enzima Bifuncional Peroxisomal , Peroxisomas/ultraestructura
16.
Biotechnol Lett ; 27(12): 859-64, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16086248

RESUMEN

alpha,omega-Dicarboxylic acid accumulation from alkanes and alkane degradation intermediates was investigated using Yarrowia lipolytica wild type strain W29 as well as a double, a triple and a quadruple POX-deleted strains. Six genes, POX1 through POX6, encode six acyl-CoA oxidase isozymes in Y. lipolytica. All the strains accumulated dodecanedioic acid (5-20 mg ml(-1)) from the diterminal functionalised 1,12-dodecane diol and 12-hydroxdodecanoic acid. The quadruple-deleted strain was the only strain that was able to accumulate dioic acids from C16 alkanol and monocarboxylic acid as well as from C12, C14 and C16 alkanes (maximum 8 mg ml(-1) from dodecane).


Asunto(s)
Acil-CoA Oxidasa/deficiencia , Ácidos Dicarboxílicos/metabolismo , Mutación , Yarrowia/metabolismo , Acil-CoA Oxidasa/genética , Acil-CoA Oxidasa/metabolismo , Alcanos/metabolismo , Biotransformación , Eliminación de Gen , Genotipo , Isoenzimas/deficiencia , Isoenzimas/genética , Isoenzimas/metabolismo , Factores de Tiempo , Yarrowia/enzimología , Yarrowia/genética
17.
Mol Genet Metab ; 83(1-2): 16-27, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15464416

RESUMEN

Peroxisomes catalyse a large variety of different cellular functions of which most have to do with lipid metabolism. This paper deals with the role of peroxisomes in three key pathways of lipid metabolism, including: (1) etherphospholipid biosynthesis, (2) fatty acid beta-oxidation, and (3) fatty acid alpha-oxidation. Apart from a brief description of the peroxisomal enzymes involved in each of these pathways, the interaction between peroxisomes and other subcellular organelles, notably microsomes and peroxisomes, will be discussed. Finally, the current state of knowledge with respect to the different disorders of peroxisomal lipid metabolism will be described.


Asunto(s)
Metabolismo de los Lípidos , Trastorno Peroxisomal/metabolismo , Peroxisomas/fisiología , Acil-CoA Oxidasa/deficiencia , Transporte Biológico , Ácidos Grasos/metabolismo , Humanos , Mitocondrias/metabolismo , Oxidación-Reducción , Trastorno Peroxisomal/genética , Éteres Fosfolípidos/metabolismo , Racemasas y Epimerasas/deficiencia
18.
Biotechnol Lett ; 26(10): 849-54, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15269560

RESUMEN

Dodecanol (1% v/v) and dodecanoic acid (1% w/v) inhibited growth of Yarrowia lipolytica in complex media supplemented with glucose but dodecanedioic acid (1% w/v) was not toxic. Dodecanol-tolerant strains were prepared from the wild type strain H222 as well as the acyl-CoA oxidase deleted (deltaPOX2, POX3, POX5) strain MTLY35. These strains grew in rich media containing up to 10% (v/v) dodecanol. Dodecanol-tolerant strains remained dodecanol tolerant after they had been cultured in rich media without dodecanol. No significant amount of dodecanedioic acid was accumulated by the dodecanol-tolerant strains when grown on glucose in the presence of dodecanol.


Asunto(s)
Acil-CoA Oxidasa/deficiencia , Técnicas de Cultivo de Célula/métodos , Dodecanol/farmacología , Tolerancia a Medicamentos/fisiología , Ingeniería Genética/métodos , Yarrowia/efectos de los fármacos , Yarrowia/crecimiento & desarrollo , Acil-CoA Oxidasa/genética , Adaptación Fisiológica/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Especificidad de la Especie , Yarrowia/clasificación , Yarrowia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...