Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Cells ; 11(17)2022 08 24.
Article in English | MEDLINE | ID: mdl-36078043

ABSTRACT

Background: Very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency is an autosomal recessive disease that prevents the body from utilizing long-chain fatty acids for energy, most needed during stress and fasting. Symptoms can appear from infancy through childhood and adolescence or early adulthood, and include hypoglycemia, recurrent rhabdomyolysis, myopathy, hepatopathy, and cardiomyopathy. REN001 is a peroxisome-proliferator-activated receptor delta (PPARδ) agonist that modulates the expression of the genes coding for fatty acid ß-oxidation enzymes and proteins involved in oxidative phosphorylation. Here, we assessed the effect of REN001 on VLCAD-deficient patient fibroblasts. Methods: VLCAD-deficient patient and control fibroblasts were treated with REN001. Cells were harvested for gene expression analysis, protein content, VLCAD enzyme activity, cellular bioenergetics, and ATP production. Results: VLCAD-deficient cell lines responded differently to REN001 based on genotype. All cells had statistically significant increases in ACADVL gene expression. Small increases in VLCAD protein and enzyme activity were observed and were cell-line- and dose-dependent. Even with these small increases, cellular bioenergetics improved in all cell lines in the presence of REN001, as demonstrated by the oxygen consumption rate and ATP production. VLCAD-deficient cell lines containing missense mutations responded better to REN001 treatment than one containing a duplication mutation in ACADVL. Discussion: Treating VLCAD-deficient fibroblasts with the REN001 PPARδ agonist results in an increase in VLCAD protein and enzyme activity, and a decrease in cellular stress. These results establish REN001 as a potential therapy for VLCADD as enhanced expression may provide a therapeutic increase in total VLCAD activity, but suggest the need for mutation-specific treatment augmented by other treatment measures.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain , PPAR delta , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Adenosine Triphosphate/metabolism , Adolescent , Adult , Child , Congenital Bone Marrow Failure Syndromes , Energy Metabolism , Fibroblasts/metabolism , Humans , Lipid Metabolism, Inborn Errors , Mitochondrial Diseases , Muscular Diseases , PPAR delta/metabolism
2.
Orphanet J Rare Dis ; 17(1): 360, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36109795

ABSTRACT

BACKGROUND: Mitochondrial long-chain fatty acid oxidation and carnitine metabolism defects are a group of inherited metabolic diseases. We performed a retrospective cohort study to report on the phenotypic and genotypic spectrum of mitochondrial long-chain fatty acid oxidation and carnitine metabolism defects as well as their treatment outcomes. METHODS: All patients with mitochondrial long-chain fatty acid oxidation and carnitine metabolism defects were included. We divided patients into two groups to compare outcomes of those treated symptomatically (SymX) and asymptomatically (AsymX). We reviewed patient charts for clinical features, biochemical investigations, molecular genetic investigations, cardiac assessments, neuroimaging, treatments, and outcomes. RESULTS: There were 38 patients including VLCAD (n = 5), LCHAD (n = 4), CACT (n = 3), MAD (n = 1), CPT-I (n = 13), CPT-II (n = 3) deficiencies and CTD (n = 9). Fourteen patients were diagnosed symptomatically (SymX), and 24 patients were diagnosed asymptomatically (AsymX). Twenty-eight variants in seven genes were identified in 36 patients (pathogenic/likely pathogenic n = 25; variant of unknown significance n = 3). Four of those variants were novel. All patients with LCHAD deficiency had the common variant (p.Glu474Gln) in HADHA and their phenotype was similar to the patients reported in the literature for this genotype. Only one patient with VLCAD deficiency had the common p.Val283Ala in ACADVL. The different genotypes in the SymX and AsymX groups for VLCAD deficiency presented with similar phenotypes. Eight patients were treated with carnitine supplementation [CTD (n = 6), CPT-II (n = 1), and MAD (n = 1) deficiencies]. Thirteen patients were treated with a long-chain fat restricted diet and MCT supplementation. A statistically significant association was found between rhabdomyolysis, and hypoglycemia in the SymX group compared to the AsymX group. A higher number of hospital admissions, longer duration of hospital admissions and higher CK levels were observed in the SymX group, even though the symptomatic group was only 37% of the study cohort. CONCLUSION: Seven different mitochondrial long-chain fatty acid oxidation and carnitine metabolism defects were present in our study cohort. In our clinic, the prevalence of mitochondrial long-chain fatty acid oxidation and carnitine defects was 4.75%.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain , Carnitine , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/genetics , Congenital Bone Marrow Failure Syndromes , Fatty Acids/metabolism , Humans , Lipid Metabolism, Inborn Errors , Mitochondrial Diseases , Muscular Diseases , Retrospective Studies
3.
Neuromuscul Disord ; 31(6): 566-569, 2021 06.
Article in English | MEDLINE | ID: mdl-33965301

ABSTRACT

Very long-chain acyl-coenzyme A (CoA) dehydrogenase (VLCAD) deficiency is an autosomal recessive fatty acid oxidation disorder characterized by rhabdomyolysis, hypoglycemia and cardiomyopathy. The general treatment approach in adult patients is based on the prevention of catabolism. High carbohydrate, low fat diet and supplementation of medium-chain triglycerides are essential in the treatment. There is little experience with pregnancy follow-up in this patient group. We present a complicated peripartum course and successful management in a patient with VLCAD deficiency. Although high-dose glucose infusion was initiated, creatine kinase levels significantly increased in the immediate postpartum period, but the patient remained asymptomatic and rhabdomyolysis resolved rapidly after increasing the glucose infusion rate.


Subject(s)
Congenital Bone Marrow Failure Syndromes/therapy , Lipid Metabolism, Inborn Errors/therapy , Mitochondrial Diseases/therapy , Muscular Diseases/therapy , Peripartum Period , Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Adult , Female , Humans , Pregnancy , Rhabdomyolysis/therapy
4.
Mol Genet Metab ; 132(4): 227-233, 2021 04.
Article in English | MEDLINE | ID: mdl-33610471

ABSTRACT

INTRODUCTION: Triheptanoin provides long-chain fatty acid oxidation disorder (LC-FAOD) patients with an alternative to medium-even-chain triglycerides therapy. MATERIAL-METHODS: Retrospective analysis of 18 French LC-FAOD patients benefiting from early access to triheptanoin treatment. RESULTS: Eight female and 10 male patients with LC-FAOD (VLCAD, LCHAD, CACT, CPTII and MTP) were treated with triheptanoin for a median duration of 22 months (range: 9-228 months). At last consultation, triheptanoin accounted for 15-35% of their daily caloric intake. In the year following the introduction of triheptanoin, patients reported a reduction of intermittent snacking and nocturnal meals. Three patients, including 1 adult, became free of severe hypoglycaemic events. Ten of 12 paediatric patients and 4 of 6 adult patients reported reduced fatigue with reductions in the number and severity of episodes of myalgia. Of 6 patients, including 1 adult, that had required the use of a wheelchair in the year prior to triheptanoin, all but one no longer required its use. The number of emergency hospitalizations decreased, and none were recorded for paediatric patients during these 12 months. Cumulative annual days of emergency care in the home were reduced from 286 to 51 days in the year before and after initiation, respectively, and 13 patients required no such interventions. Adverse events were limited to digestive issues that dissipated over time. CONCLUSIONS: Our case-series suggests that long-term treatment of LC-FAOD paediatric and adult patients with triheptanoin is safe and leads to marked improvement of symptoms and an improved quality of life.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/genetics , Metabolic Diseases/drug therapy , Triglycerides/administration & dosage , Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Adolescent , Adult , Carnitine/genetics , Child , Child, Preschool , Female , Humans , Infant , Male , Metabolic Diseases/genetics , Metabolic Diseases/pathology , Oxidation-Reduction/drug effects , Quality of Life , Retrospective Studies , Treatment Outcome , Young Adult
5.
Sci Rep ; 10(1): 18367, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33110171

ABSTRACT

Medium-chain triglycerides (MCT), containing C8-C12 fatty acids, are used to treat several pediatric disorders and are widely consumed as a nutritional supplement. Here, we investigated the role of the sirtuin deacylase Sirt5 in MCT metabolism by feeding Sirt5 knockout mice (Sirt5KO) high-fat diets containing either C8/C10 fatty acids or coconut oil, which is rich in C12, for five weeks. Coconut oil, but not C8/C10 feeding, induced periportal macrovesicular steatosis in Sirt5KO mice. 14C-C12 degradation was significantly reduced in Sirt5KO liver. This decrease was localized to the mitochondrial ß-oxidation pathway, as Sirt5KO mice exhibited no change in peroxisomal C12 ß-oxidation. Endoplasmic reticulum ω-oxidation, a minor fatty acid degradation pathway known to be stimulated by C12 accumulation, was increased in Sirt5KO liver. Mice lacking another mitochondrial C12 oxidation enzyme, long-chain acyl-CoA dehydrogenase (LCAD), also developed periportal macrovesicular steatosis when fed coconut oil, confirming that defective mitochondrial C12 oxidation is sufficient to induce the steatosis phenotype. Sirt5KO liver exhibited normal LCAD activity but reduced mitochondrial acyl-CoA synthetase activity with C12. These studies reveal a role for Sirt5 in regulating the hepatic response to MCT and may shed light into the pathogenesis of periportal steatosis, a hallmark of human pediatric non-alcoholic fatty liver disease.


Subject(s)
Fatty Acids/metabolism , Mitochondria, Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Sirtuins/genetics , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Animals , Coconut Oil/administration & dosage , Dietary Fats/administration & dosage , Female , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Oxidation-Reduction , Triglycerides/metabolism
6.
Mol Genet Metab ; 131(1-2): 23-37, 2020.
Article in English | MEDLINE | ID: mdl-33093005

ABSTRACT

The nutrition management guideline for very-long chain acyl-CoA dehydrogenase deficiency (VLCAD) is the fourth in a series of web-based guidelines focusing on the diet treatment for inherited metabolic disorders and follows previous publication of guidelines for maple syrup urine disease (2014), phenylketonuria (2016) and propionic acidemia (2019). The purpose of this guideline is to establish harmonization in the treatment and monitoring of individuals with VLCAD of all ages in order to improve clinical outcomes. Six research questions were identified to support guideline development on: nutrition recommendations for the healthy individual, illness management, supplementation, monitoring, physical activity and management during pregnancy. This report describes the methodology used in its development including review, critical appraisal and abstraction of peer-reviewed studies and unpublished practice literature; expert input through two Delphi surveys and a nominal group process; and external review from metabolic physicians and dietitians. It includes the summary statements of the nutrition management recommendations for each research question, followed by a standardized rating based on the strength of the evidence. Online, open access of the full published guideline allows utilization by health care providers, researchers and collaborators who advise, advocate and care for individuals with VLCAD and their families and can be accessed from the Genetic Metabolic Dietitians International (https://GMDI.org) and Southeast Regional Genetics Network (https://southeastgenetics.org/ngp) websites.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/genetics , Congenital Bone Marrow Failure Syndromes/diet therapy , Lipid Metabolism, Inborn Errors/diet therapy , Mitochondrial Diseases/diet therapy , Muscular Diseases/diet therapy , Nutrition Policy , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Congenital Bone Marrow Failure Syndromes/genetics , Congenital Bone Marrow Failure Syndromes/metabolism , Congenital Bone Marrow Failure Syndromes/pathology , Female , Guidelines as Topic , Humans , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/metabolism , Lipid Metabolism, Inborn Errors/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Muscular Diseases/genetics , Muscular Diseases/metabolism , Muscular Diseases/pathology , Nutrition Therapy , Pregnancy
7.
FEBS J ; 287(16): 3511-3525, 2020 08.
Article in English | MEDLINE | ID: mdl-31971349

ABSTRACT

Very-long-chain acyl-CoA dehydrogenase deficiency (VLCAD) is the most common defect of long-chain fatty acid ß-oxidation. The recommended treatment includes the application of medium-chain triacylglycerols (MCTs). However, long-term treatment of VLCAD-/- mice resulted in the development of a sex-specific metabolic syndrome due to the selective activation of the ERK/mTORc1 signalling in females and ERK/peroxisome proliferator-activated receptor gamma pathway in males. In order to investigate a subsequent sex-specific effect of MCT on the lipid composition of the cellular membranes, we performed lipidomic analysis, SILAC-based quantitative proteomics and gene expression in fibroblasts from WT and VLCAD-/- mice of both sexes. Treatment with octanoate (C8) affected the composition of complex lipids resulting in a sex-specific signature of the molecular profile. The content of ceramides and sphingomyelins in particular differed significantly under control conditions and increased markedly in cells from mutant female mice but remained unchanged in cells from mutant males. Moreover, we observed a specific upregulation of biosynthesis of plasmalogens only in male mice, whereas in females C8 led to the accumulation of higher concentration of phosphatidylcholines and lysophosphatidylcholines. Our data on membrane lipids in VLCAD after supplementation with C8 provide evidence of a sex-specific lipid perturbation. We hypothesize a likely C8-induced pro-inflammatory response contributing to the development of a severe metabolic syndrome in female VLCAD-/- mice on long-term MCT supplementation.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/genetics , Caprylates/pharmacology , Fibroblasts/drug effects , Gene Expression/drug effects , Lipidomics/methods , Proteomics/methods , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Animals , Cells, Cultured , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Male , Mass Spectrometry , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Sex Factors
8.
J Inherit Metab Dis ; 43(6): 1232-1242, 2020 11.
Article in English | MEDLINE | ID: mdl-33448436

ABSTRACT

Anaplerotic odd-chain fatty acid supplementation has been suggested as an approach to replenish citric acid cycle intermediate (CACi) pools and facilitate adenosine triphosphate (ATP) production in subjects with long-chain fatty acid oxidation disorders, but the evidence that cellular CACi depletion exists and that repletion occurs following anaplerotic substrate supplementation is limited. We exercised very long-chain acyl-CoA dehydrogenase-deficient (VLCAD-/-) and wild-type (WT) mice to exhaustion and collected cardiac tissue for measurement of CACi by targeted metabolomics. In a second experimental group, VLCAD-/- and WT mice that had been fed chow prepared with either medium-chain triglyceride (MCT) oil or triheptanoin for 4 weeks were exercised for 60 minutes. VLCAD-/- mice exhibited lower succinate in cardiac muscle at exhaustion than WT mice suggesting lower CACi in VLCAD-/- with prolonged exercise. In mice fed either MCT or triheptanoin, succinate and malate were greater in VLCAD-/- mice fed triheptanoin compared to VLCAD-/- animals fed MCT but lower than WT mice fed triheptanoin. Long-chain odd acylcarnitines such as C19 were elevated in VLCAD-/- and WT mice fed triheptanoin suggesting some elongation of the heptanoate, but it is unknown what proportion of heptanoate was oxidized vs elongated. Prolonged exercise was associated with decreased cardiac muscle succinate in VLCAD-/- mice in comparison to WT mice. VLCAD-/- fed triheptanoin had increased succinate compared to VLCAD-/- mice fed MCT but lower than WT mice fed triheptanoin. Cardiac CACi were higher following dietary ingestion of an anaplerotic substrate, triheptanoin, in comparison to MCT.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Congenital Bone Marrow Failure Syndromes/diet therapy , Congenital Bone Marrow Failure Syndromes/metabolism , Lipid Metabolism, Inborn Errors/diet therapy , Lipid Metabolism, Inborn Errors/metabolism , Mitochondrial Diseases/diet therapy , Mitochondrial Diseases/metabolism , Muscular Diseases/diet therapy , Muscular Diseases/metabolism , Triglycerides/administration & dosage , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Animals , Citric Acid Cycle , Congenital Bone Marrow Failure Syndromes/genetics , Dietary Fats/administration & dosage , Fatty Acids/metabolism , Female , Lipid Metabolism, Inborn Errors/genetics , Liver/metabolism , Male , Mice , Mitochondrial Diseases/genetics , Muscular Diseases/genetics , Myocardium/metabolism , Oxidation-Reduction , Triglycerides/chemistry
9.
Toxicol In Vitro ; 62: 104665, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31629068

ABSTRACT

cis-5-Tetradecenoic (cis-5) and myristic (Myr) acids predominantly accumulate in patients affected by very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency. They commonly manifest myopathy with muscular pain and rhabdomyolysis, whose underlying mechanisms are poorly known. Thus, in the present study we investigated the effects of cis-5 and Myr on mitochondrial bioenergetics and Ca2+ homeostasis in rat skeletal muscle. cis-5 and Myr decreased ADP-stimulated (state 3) and CCCP-stimulated (uncoupled) respiration, especially when mitochondria were supported by NADH-linked as compared to FADH2-linked substrates. In contrast, these fatty acids increased resting respiration (state 4). Similar effects were observed in skeletal muscle fibers therefore validating the data obtained with isolated mitochondria. Furthermore, cis-5 and Myr markedly decreased mitochondrial membrane potential and Ca2+ retention capacity that were avoided by cyclosporin A plus ADP and ruthenium red, indicating that cis-5 and Myr induce mitochondrial permeability transition (MPT). Finally, docosanoic acid did not disturb mitochondrial homeostasis, indicating selective effects for Myr and cis-5. Taken together, our findings indicate that major long-chain fatty acids accumulating in VLCAD deficiency behave as metabolic inhibitors, uncouplers of oxidative phosphorylation and MPT inducers. It is presumed that these pathomechanisms contribute to the muscular symptoms and rhabdomyolysis observed in patients affected by VLCAD deficiency.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Congenital Bone Marrow Failure Syndromes/metabolism , Lipid Metabolism, Inborn Errors/metabolism , Mitochondria/drug effects , Mitochondrial Diseases/metabolism , Muscle, Skeletal/drug effects , Muscular Diseases/metabolism , Myristic Acids/toxicity , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Animals , Calcium/metabolism , Energy Metabolism/drug effects , Homeostasis/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Muscle, Skeletal/metabolism , Oxygen Consumption/drug effects , Permeability/drug effects , Rats, Wistar
10.
Article in German | MEDLINE | ID: mdl-31814088

ABSTRACT

OBJECTIVE: Exercise induced metabolic myopathy in German Hunting Terrier dogs is an autosomal-recessively inherited disorder, caused by a nonsense variant of the gene encoding for the very long-chain acyl-CoA-dehydrogenase (VLCAD) enzyme. Clinical signs include exercise- induced fatigue, muscle pain and weakness. In the present study, the long-term course of this disease was investigated over a period of 1 year in 9 affected German Hunting Terriers. The dogs were treated symptomatically with oral L-carnitine, coenzyme Q10 and a special diet characterized by a low content of long-chain fatty acids and a high proportion of carbohydrates. MATERIAL AND METHODS: In 9 affected dogs, the phenotype as well as clinical, laboratory parameters, and histopathological findings are described (time point 1) and compared to follow-up examinations 1 year later (time point 2). At both time points clinical and neurological examinations, complete blood cell count, clinical chemistry profile and the concentration of brain natriuretic peptide (NT-proBNP) were investigated. RESULTS: In the follow-up examinations, the same post-exercise clinical signs were present as in the initial presentation of the homozygous dogs. Dark-brownish discoloration of the urine, weakness, myalgia as well as stiff and tetraparetic gait were apparant. All hematological values and the concentration of NT-proBNP were within the relevant reference ranges. Plasma CK and ALT activities were compared between the first presentation and the follow- up examination and no significant differences were detected (pCK = 0.31, pALT = 0.64). Signs of myopathy remained unchanged throughout the examination period. CONCLUSION AND CLINICAL RELEVANCE: Oral supplementation with L-carnitine, coenzyme Q10 and the special dietary management did not result in any improvement of clinical signs or laboratory parameters. No progression of the disease was observed. The prognosis for affected dogs remains cautious as long-term observations of affected dogs over several years are lacking. Our findings provide further important information on inherited disorders of mitochondrial ß-oxidation in dogs, especially focused on the exercise induced metabolic myopathy in the German Hunting Terrier. This may provide new insights for novel treatment modalities in conjuntion with the development of improved breeding guidelines.


Subject(s)
Dog Diseases , Muscular Diseases , Physical Conditioning, Animal/adverse effects , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Animals , Carnitine/therapeutic use , Diet , Dog Diseases/diagnosis , Dog Diseases/genetics , Dog Diseases/therapy , Dogs , Follow-Up Studies , Muscular Diseases/diagnosis , Muscular Diseases/genetics , Muscular Diseases/therapy , Muscular Diseases/veterinary , Ubiquinone/analogs & derivatives , Ubiquinone/therapeutic use
11.
FASEB J ; 33(6): 7289-7300, 2019 06.
Article in English | MEDLINE | ID: mdl-30848932

ABSTRACT

Berberine (BBR) shows promising effects in the treatment of nonalcoholic fatty liver disease (NAFLD) by influencing various metabolic aspects. Inhibition of mitochondrial ß-oxidation (ß-OX) participates in the pathogenesis of NAFLD. Silent mating-type information regulation 2 homolog 3 (SIRT3) has been reported to regulate mitochondrial ß-OX by deacetylating its substrate, long-chain acyl-coenzyme A dehydrogenase (LCAD). This study aimed to explore whether BBR can promote mitochondrial ß-OX and the role of SIRT3 as well as the mechanisms underlying the effects of BBR on hepatic lipid metabolism in mice fed a high-fat diet (HFD). BBR can significantly improve systematic and hepatic lipid metabolism in HFD-fed mice. Metabolomics analysis revealed that ß-OX was inhibited in HFD-induced mice, as indicated by the reduced production of short and medium carbon chain acyl-carnitines, the activated form of free fatty acids, via ß-OX, which was reversed by BBR intervention. Exploration of the mechanism found that BBR intervention reversed the down-regulation of SIRT3 and decreased the LCAD hyperacetylation level in HFD-fed mice. SIRT3 knockout (KO) mice were used to identify the role of SIRT3 in the BBR's influence of ß-OX. The beneficial effects of BBR on systemic and hepatic metabolism were profoundly attenuated in KO mice. Moreover, the promotive effect of BBR on ß-OX in HFD-induced mice was partially abolished in KO mice. These results suggested that BBR alleviates HFD-induced inhibition of fatty acid ß-OX partly through SIRT3-mediated LCAD deacetylation, which may provide a novel mechanism and support BBR as a promising therapeutic for NAFLD.-Xu, X., Zhu, X.-P., Bai, J.-Y., Xia, P., Li, Y., Lu, Y., Li, X.-Y., Gao, X. Berberine alleviates nonalcoholic fatty liver induced by a high-fat diet in mice by activating SIRT3.


Subject(s)
Berberine/pharmacology , Diet, High-Fat/adverse effects , Metabolome/drug effects , Mitochondria, Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Sirtuin 3/drug effects , Acetylation , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Animals , Berberine/therapeutic use , Carnitine/analogs & derivatives , Carnitine/metabolism , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Fatty Acids/metabolism , Glucose/metabolism , Insulin Resistance , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Liver/enzymology , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Obesity/complications , Oxidation-Reduction , Protein Processing, Post-Translational , Sirtuin 3/deficiency , Sirtuin 3/physiology
12.
J Inherit Metab Dis ; 42(1): 159-168, 2019 01.
Article in English | MEDLINE | ID: mdl-30740737

ABSTRACT

BACKGROUND: Patients with very long chain acyl-CoA dehydrogenase deficiency (VLCADD), a long chain fatty acid oxidation disorder, are traditionally treated with a long chain triglyceride (LCT) restricted and medium chain triglyceride (MCT) supplemented diet. Introduction of VLCADD in newborn screening (NBS) programs has led to the identification of asymptomatic newborns with VLCADD, who may have a more attenuated phenotype and may not need dietary adjustments. OBJECTIVE: To define dietary strategies for individuals with VLCADD based on the predicted phenotype. METHOD: We evaluated long-term dietary histories of a cohort of individuals diagnosed with VLCADD identified before the introduction of VLCADD in NBS and their beta-oxidation (LC-FAO) flux score (rate of oleate oxidation) in cultured skin fibroblasts in relation to the clinical outcome. Based on these results a dietary strategy is proposed. RESULTS: Sixteen individuals with VLCADD were included. One had an LC-FAO flux score >90%, was not on a restricted diet and is asymptomatic to date. Four patients had an LC-FAO flux score <10%, and significant VLCADD related symptoms despite the use of strict diets including LCT restriction, MCT supplementation and nocturnal gastric drip feeding. Patients with an LC-FAO flux score between 10 and 90% (n = 11) showed a more heterogeneous phenotype. CONCLUSIONS: This study shows that a strict diet cannot prevent poor clinical outcome in severely affected patients and that the LC-FAO flux is a good predictor of clinical outcome in individuals with VLCADD identified before its introduction in NBS. Hereby, we propose an individualized dietary strategy based on the LC-FAO flux score.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Acyl-CoA Dehydrogenase/deficiency , Congenital Bone Marrow Failure Syndromes/drug therapy , Lipid Metabolism, Inborn Errors/drug therapy , Mitochondrial Diseases/drug therapy , Muscular Diseases/drug therapy , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Congenital Bone Marrow Failure Syndromes/metabolism , Diet , Fatty Acids/administration & dosage , Female , Humans , Infant, Newborn , Lipid Metabolism, Inborn Errors/metabolism , Male , Mitochondrial Diseases/metabolism , Muscular Diseases/metabolism , Neonatal Screening/methods , Phenotype , Triglycerides/administration & dosage
13.
J Hum Genet ; 64(2): 73-85, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30401918

ABSTRACT

Mitochondrial fatty acid oxidation disorders (FAODs) are caused by defects in ß-oxidation enzymes, including very long-chain acyl-CoA dehydrogenase (VLCAD), trifunctional protein (TFP), carnitine palmitoyltransferase-2 (CPT2), carnitine-acylcarnitine translocase (CACT) and others. During prolonged fasting, infection, or exercise, patients with FAODs present with hypoglycemia, rhabdomyolysis, cardiomyopathy, liver dysfunction, and occasionally sudden death. This article describes the diagnosis, newborn screening, and treatment of long-chain FAODs with a focus on VLCAD deficiency. VLCAD deficiency is generally classified into three phenotypes based on onset time, but the classification should be comprehensively determined based on genotype, residual enzyme activity, and clinical course, due to a lack of apparent genotype-phenotype correlation. With the expansion of newborn screening for FAODs, several issues have arisen, such as missed detection, overdiagnosis (including detection of benign/asymptomatic type), and poor prognosis of the neonatal-onset form. Meanwhile, dietary management and restriction of exercise have been unnecessary for patients with the benign/asymptomatic type of VLCAD deficiency with a high fatty acid oxidation flux score. Although L-carnitine therapy for VLCAD/TFP deficiency has been controversial, supplementation with L-carnitine may be accepted for CPT2/CACT and multiple acyl-CoA dehydrogenase deficiencies. Recently, a double-blind, randomized controlled trial of triheptanoin (seven-carbon fatty acid triglyceride) versus trioctanoin (regular medium-chain triglyceride) was conducted and demonstrated improvement of cardiac functions on triheptanoin. Additionally, although the clinical efficacy of bezafibrate remains controversial, a recent open-label clinical trial showed efficacy of this drug in improving quality of life. These drugs may be promising for the treatment of FAODs, though further studies are required.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Fatty Acids/metabolism , Hypolipidemic Agents/therapeutic use , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/drug therapy , Mass Screening , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/drug therapy , Muscular Diseases/diagnosis , Muscular Diseases/drug therapy , Congenital Bone Marrow Failure Syndromes , Disease Management , Humans , Lipid Metabolism, Inborn Errors/enzymology , Mitochondrial Diseases/enzymology , Muscular Diseases/enzymology
14.
FEBS Lett ; 592(2): 219-232, 2018 01.
Article in English | MEDLINE | ID: mdl-29237229

ABSTRACT

The white skeletal muscle of very long-chain acyl-CoA-dehydrogenase-deficient (VLCAD-/- ) mice undergoes metabolic modification to compensate for defective ß-oxidation in a progressive and time-dependent manner by upregulating glucose oxidation. This metabolic regulation seems to be accompanied by morphologic adaptation of muscle fibers toward the glycolytic fiber type II with the concomitant upregulation of mitochondrial fatty acid biosynthesis (mFASII) and lipoic acid biosynthesis. Dietary supplementation of VLCAD-/- mice with different medium-chain triglycerides over 1 year revealed that odd-chain species has no effect on muscle fiber switch, whereas even-chain species inhibit progressive metabolic adaptation. Our study shows that muscle may undergo adaptive mechanisms that are modulated by dietary supplementation. We describe for the first time a concomitant change of mFASII in this muscular adaptation process.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Fatty Acids/biosynthesis , Lipid Metabolism, Inborn Errors/metabolism , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Muscle Fibers, Fast-Twitch/physiology , Muscular Diseases/metabolism , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Animals , Cell Plasticity , Congenital Bone Marrow Failure Syndromes , Disease Models, Animal , Mice , Triglycerides/administration & dosage
15.
Mol Reprod Dev ; 85(1): 46-61, 2018 01.
Article in English | MEDLINE | ID: mdl-29219221

ABSTRACT

In vitro embryo development remains suboptimal compared to in vivo development due to the challenge from various stressors associated with in vitro culturing of oocytes. When 0.2 µM lycopene was added to oocyte in vitro maturation and embryo culture media, to assess its antioxidant effects on embryo development, we observed a significant (p < 0.05) increase in cleavage and blastocyst development rates compared to the corresponding controls (84.3 ± 0.6% vs. 73.1 ± 1.9% and 41.0 ± 1.4% vs. 33.4 ± 0.7%, respectively). Lycopene also significantly reduced (p < 0.05) intracellular reactive oxygen species concentrations in oocytes and blastocysts, whereas lipid peroxidation and mitochondrial activity increased compared to control conditions. The number of apoptotic nuclei was significantly reduced in the lycopene-treated compared to the control group (1.7 ± 0.1 vs. 4.7 ± 0.3), and the quantity of cells in the trophectoderm (207.1 ± 1.6 vs. 171.3 ± 1.0, respectively) and inner cell mass (41.9 ± 0.4 vs. 36.7 ± 0.4, respectively) was higher following treatment-although the inner cell mass-to-trophectoderm ratio was unchanged (1:3.3 vs. 1:3.4 for lycopene vs. control, respectively). Lycopene supplementation also significantly (p < 0.05) attenuated expression of IKBKB (Inhibitor of nuclear factor kappa B kinase, subunit beta) and reduced Caspase 9 and Caspase 3 protein abundance, while up-regulating GDF9 (Growth and differentiation factor 9), BMP15 (Bone morphogenetic protein 15), SOD2 (Superoxide dismutase 2), NDUFA2 (NADH dehydrogenase), ACADL (Acyl-CoA dehydrogenase, long chain), and ACSL3 (Acyl-CoA synthetase 3, long-chain membrane 3) transcription compared to control. Therefore, co-culturing with lycopene during oocyte maturation improved bovine embryo developmental potential during in vitro culture by improving embryonic resilience to stress.


Subject(s)
Antioxidants/pharmacology , Embryo Culture Techniques , Embryonic Development/drug effects , Lycopene/pharmacology , Oocytes/growth & development , Acyl-CoA Dehydrogenase, Long-Chain/biosynthesis , Animals , Blastocyst/cytology , Bone Morphogenetic Protein 15/biosynthesis , Caspase 3/analysis , Caspase 9/analysis , Cattle , Coenzyme A Ligases/biosynthesis , Growth Differentiation Factor 9/biosynthesis , I-kappa B Kinase/biosynthesis , NADH Dehydrogenase/biosynthesis , Superoxide Dismutase/biosynthesis
16.
J Inherit Metab Dis ; 40(6): 831-843, 2017 11.
Article in English | MEDLINE | ID: mdl-28871440

ABSTRACT

BACKGROUND: Observational reports suggest that supplementation that increases citric acid cycle intermediates via anaplerosis may have therapeutic advantages over traditional medium-chain triglyceride (MCT) treatment of long-chain fatty acid oxidation disorders (LC-FAODs) but controlled trials have not been reported. The goal of our study was to compare the effects of triheptanoin (C7), an anaplerotic seven-carbon fatty acid triglyceride, to trioctanoin (C8), an eight-carbon fatty acid triglyceride, in patients with LC-FAODs. METHODS: A double blinded, randomized controlled trial of 32 subjects with LC-FAODs (carnitine palmitoyltransferase-2, very long-chain acylCoA dehydrogenase, trifunctional protein or long-chain 3-hydroxy acylCoA dehydrogenase deficiencies) who were randomly assigned a diet containing 20% of their total daily energy from either C7 or C8 for 4 months was conducted. Primary outcomes included changes in total energy expenditure (TEE), cardiac function by echocardiogram, exercise tolerance, and phosphocreatine recovery following acute exercise. Secondary outcomes included body composition, blood biomarkers, and adverse events, including incidence of rhabdomyolysis. RESULTS: Patients in the C7 group increased left ventricular (LV) ejection fraction by 7.4% (p = 0.046) while experiencing a 20% (p = 0.041) decrease in LV wall mass on their resting echocardiogram. They also required a lower heart rate for the same amount of work during a moderate-intensity exercise stress test when compared to patients taking C8. There was no difference in TEE, phosphocreatine recovery, body composition, incidence of rhabdomyolysis, or any secondary outcome measures between the groups. CONCLUSIONS: C7 improved LV ejection fraction and reduced LV mass at rest, as well as lowering heart rate during exercise among patients with LC-FAODs. CLINICAL TRIAL REGISTRATION: Clinicaltrials.gov NCT01379625.


Subject(s)
Caprylates/therapeutic use , Cardiomyopathies/drug therapy , Fatty Acids/metabolism , Lipid Metabolism, Inborn Errors/drug therapy , Mitochondrial Myopathies/drug therapy , Mitochondrial Trifunctional Protein/deficiency , Nervous System Diseases/drug therapy , Rhabdomyolysis/drug therapy , Triglycerides/therapeutic use , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Adolescent , Adult , Cardiomyopathies/metabolism , Carnitine/metabolism , Child , Dietary Fats/metabolism , Double-Blind Method , Exercise/physiology , Female , Humans , Lipid Metabolism, Inborn Errors/metabolism , Male , Middle Aged , Mitochondrial Myopathies/metabolism , Mitochondrial Trifunctional Protein/metabolism , Nervous System Diseases/metabolism , Oxidation-Reduction , Rhabdomyolysis/metabolism , Young Adult
17.
J Inherit Metab Dis ; 40(3): 317-323, 2017 05.
Article in English | MEDLINE | ID: mdl-28247148

ABSTRACT

Very-long-chain-acyl-CoA-dehydrogenase deficiency is the most common disorder of mitochondrial long-chain fatty acid (LCFA) oxidation, with an incidence of 1:50,000-1:100,000 in newborns. Catabolic situations contribute to the aggravation of symptoms and induce severe metabolic derangement. Treatment for VLCAD-deficiency includes avoidance of fasting and a long-chain fat-restricted and fat-modified diet in which LCFAs are fully or partially replaced by medium-chain triglycerides (MCT). The aim of this work was to investigate the outcome and the effects of long-term treatment in a mouse model of VLCAD-deficiency. The application of a single MCT bolus in a mouse model of VLCAD-deficiency (VLCAD-/- mice) immediately prior to exercise protected the muscles from the accumulation of acylcarnitines providing the required energy and it did not affect hepatic lipid metabolism. However, when MCT was applied over the course of a year as a regular part of the diet, female VLCAD-/- mice developed a severe clinical phenotype comparable to the human metabolic syndrome. Indeed, they were characterized by massive visceral fat infiltration, hepatosteatosis, disturbed fatty acid composition, hyperlipidemia, and systemic oxidative stress. In contrast, male VLCAD-/- mice seemed to be protected and displayed only signs of insulin resistance. Besides the sex-specific response to MCT supplementation with regard to the lipid metabolism, all VLCAD-/- mice developed progressive cardiac dysfunction over time which worsened when they were treated with regular MCT resulting in severe dilated cardiomyopathy. While long term use of MCT oil in mice has adverse effects, no such effects have been demonstrated in humans, likely reflecting the differences in long chain fatty acid oxidation between the two species.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Lipid Metabolism, Inborn Errors/therapy , Mitochondrial Diseases/therapy , Muscular Diseases/therapy , Animals , Congenital Bone Marrow Failure Syndromes , Disease Models, Animal , Fatty Acids/metabolism , Humans , Lipid Metabolism/physiology , Lipid Metabolism, Inborn Errors/metabolism , Mice , Mitochondrial Diseases/metabolism , Muscular Diseases/metabolism , Triglycerides/metabolism
18.
J Lipid Res ; 58(1): 196-207, 2017 01.
Article in English | MEDLINE | ID: mdl-27884962

ABSTRACT

A rather new approach in the treatment of long-chain fatty acid oxidation disorders is represented by triheptanoin, a triglyceride with three medium-odd-chain heptanoic acids (C7), due to its anaplerotic potential. We here investigate the effects of a 1-year triheptanoin-based diet on the clinical phenotype of very long-chain-acyl-CoA-dehydrogenase-deficient (VLCAD-/-) mice. The cardiac function was assessed in VLCAD-/- mice by in vivo MRI. Metabolic adaptations were identified by the expression of genes regulating energy metabolism and anaplerotic processes using real-time PCR, and the results were correlated with the measurement of the glycolytic enzymes pyruvate dehydrogenase and pyruvate kinase. Finally, the intrahepatic lipid accumulation and oxidative stress in response to the long-term triheptanoin diet were assessed. Triheptanoin was not able to prevent the development of systolic dysfunction in VLCAD-/- mice despite an upregulation of cardiac glucose oxidation. Strikingly, the anaplerotic effects of triheptanoin were restricted to the liver. Despite this, the hepatic lipic content was increased upon triheptanoin supplementation. Our data demonstrate that the concept of anaplerosis does not apply to all tissues equally.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/genetics , Cardiomyopathies/drug therapy , Lipid Metabolism, Inborn Errors/drug therapy , Triglycerides/administration & dosage , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Energy Metabolism/drug effects , Energy Metabolism/genetics , Fatty Acids/metabolism , Heptanoic Acids/metabolism , Humans , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/metabolism , Lipid Metabolism, Inborn Errors/pathology , Liver/metabolism , Liver/pathology , Mice , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects
19.
J Inherit Metab Dis ; 39(3): 399-408, 2016 05.
Article in English | MEDLINE | ID: mdl-26907176

ABSTRACT

Blood and urine acylcarnitine profiles are commonly used to diagnose long-chain fatty acid oxidation disorders (FAOD: i.e., long-chain hydroxy-acyl-CoA dehydrogenase [LCHAD] and carnitine palmitoyltransferase 2 [CPT2] deficiency), but the global metabolic impact of long-chain FAOD has not been reported. We utilized untargeted metabolomics to characterize plasma metabolites in 12 overnight-fasted individuals with FAOD (10 LCHAD, two CPT2) and 11 healthy age-, sex-, and body mass index (BMI)-matched controls, with the caveat that individuals with FAOD consume a low-fat diet supplemented with medium-chain triglycerides (MCT) while matched controls consume a typical American diet. In plasma 832 metabolites were identified, and partial least squared-discriminant analysis (PLS-DA) identified 114 non-acylcarnitine variables that discriminated FAOD subjects and controls. FAOD individuals had significantly higher triglycerides and lower specific phosphatidylethanolamines, ceramides, and sphingomyelins. Differences in phosphatidylcholines were also found but the directionality differed by metabolite species. Further, there were few differences in non-lipid metabolites, indicating the metabolic impact of FAOD specifically on lipid pathways. This analysis provides evidence that LCHAD/CPT2 deficiency significantly alters complex lipid pathway flux. This metabolic signature may provide new clinical tools capable of confirming or diagnosing FAOD, even in subjects with a mild phenotype, and may provide clues regarding the biochemical and metabolic impact of FAOD that is relevant to the etiology of FAOD symptoms.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Carnitine O-Palmitoyltransferase/deficiency , Fatty Acids/metabolism , Lipid Metabolism, Inborn Errors/metabolism , Metabolism, Inborn Errors/metabolism , Plasma/metabolism , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Adolescent , Carnitine/analogs & derivatives , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Case-Control Studies , Ceramides/metabolism , Female , Humans , Lipid Metabolism , Male , Metabolic Networks and Pathways/physiology , Oxidation-Reduction , Phosphatidylcholines/metabolism , Phosphatidylethanolamines/metabolism , Sphingomyelins/metabolism , Triglycerides/metabolism
20.
Nutrients ; 8(1)2016 Jan 21.
Article in English | MEDLINE | ID: mdl-26805879

ABSTRACT

Korean pine nut oil (PNO) has been reported to influence weight gain and lipid metabolism. We examined whether PNO replacement in a high-fat diet (HFD) can ameliorate HFD-induced hepatic steatosis. Five-week-old male C57BL mice were fed control diets containing 10% of the energy from fat from PNO or soybean oil (SBO) (PC, SC) or HFDs with 45% of the energy from fat, with 10% from PNO or SBO and 35% from lard (PHFD, SHFD), for 12 weeks. Body weight gain and amount of white adipose tissue were lower in PHFD (10% and 18% lower, respectively) compared with SHFD. Hepatic triacylglycerol (TG) level was significantly lower in PHFD than the SHFD (26% lower). PNO consumption upregulated hepatic ACADL mRNA levels. The hepatic PPARG mRNA level was lower in the PC than in the SC. Expression of the sirtuin (SIRT) 3 protein in white adipose tissue was down-regulated in the SHFD and restored in the PHFD to the level in the lean control mice. SIRT 3 was reported to be upregulated under conditions of caloric restriction (CR) and plays a role in regulating mitochondrial function. PNO consumption resulted in lower body fat and hepatic TG accumulation in HFD-induced obesity, which seemed to be associated with the CR-mimetic response.


Subject(s)
Fatty Liver/metabolism , Lipid Metabolism/drug effects , Obesity/metabolism , Pinus/chemistry , Plant Oils/pharmacology , Triglycerides/metabolism , Acyl-CoA Dehydrogenase, Long-Chain/genetics , Adipose Tissue, White/drug effects , Animals , Diet, High-Fat/adverse effects , Dietary Fats/pharmacology , Down-Regulation/drug effects , Fatty Liver/diet therapy , Fatty Liver/etiology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Nuts/chemistry , Obesity/diet therapy , Obesity/etiology , PPAR gamma/genetics , RNA, Messenger/metabolism , Sirtuin 3/metabolism , Soybean Oil/pharmacology , Up-Regulation/drug effects , Weight Gain/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL