Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Sci Rep ; 13(1): 3465, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36859467

ABSTRACT

Augmentation of anaplerosis, or replenishment of carbon lost during intermediary metabolic transitions, is desirable in energy metabolism defects. Triheptanoin, the triglyceride of 7-carbon heptanoic acid, is anaplerotic via direct oxidation or 5-carbon ketone body generation. In this context, triheptanoin can be used to treat Glucose transporter type 1 deficiency encephalopathy (G1D). An oral triheptanoin dose of 1 g/Kg/day supplies near 35% of the total caloric intake and impacted epilepsy and cognition in G1D. This provided the motivation to establish a maximum, potentially greater dose. Using a 3 + 3 dose-finding approach useful in oncology, we studied three age groups: 4-6, 6.8-10 and 11-16 years old. This allowed us to arrive at a maximum tolerated dose of 45% of daily caloric intake for each group. Safety was ascertained via analytical blood measures. One dose-limiting toxicity, occurring in 1 of 6 subjects, was encountered in the middle age group in the context of frequently reduced gastrointestinal tolerance for all groups. Ketonemia following triheptanoin was determined in another group of G1D subjects. In them, ß-ketopentanoate and ß-hydroxypentanoate concentrations were robustly but variably increased. These results enable the rigorous clinical investigation of triheptanoin in G1D by providing dosing and initial tolerability, safety and ketonemic potential.ClinicalTrials.gov registration: NCT03041363, first registration 02/02/2017.


Subject(s)
Ketosis , Middle Aged , Humans , Child, Preschool , Glucose Transporter Type 1 , Carbon , Triglycerides
2.
Am J Vet Res ; 82(12): 955-962, 2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34727070

ABSTRACT

OBJECTIVE: To evaluate the effects of a flotation vest (FV) and water flow rate (WFR) on limb kinematics of dogs swimming against a current. ANIMALS: 7 (1 male and 6 female) healthy adult Siberian Huskies. PROCEDURES: Dogs were habituated to swim with and without an FV beside an investigator in a continuous-flow pool against WFRs up to 2.9 km/h. During each of 4 experimental sessions in a repeated-measures study, markers were wrapped around the right carpus and tarsus, and a video was recorded while each dog swam with or without an FV for about 2 minutes at each of 7 WFRs between 0 and 2.9 km/h when the WFR was incrementally decreased or increased. Motion tracking software was used to measure stroke excursion and frequency. RESULTS: Stroke excursion varied more than frequency among all dogs and in response to changes in experimental conditions. The male dog and 1 female dog were unable to complete the study. For the remaining 5 dogs across all experimental conditions, mean tarsus excursion was 30% that of the carpus. Mean total excursion (sum of the excursion-frequency products for the carpus and tarsus) decreased when an FV was worn and increased with WFR by 69% and 19% when WFR was incrementally increased and decreased, respectively. CONCLUSIONS AND CLINICAL RELEVANCE: In dogs, range of motion during swimming was greater for the carpus than tarsus, when an FV was not worn, and increased more with WFR when WFR was incrementally increased. Those factors should be considered during swimming-based rehabilitation.


Subject(s)
Extremities , Swimming , Animals , Biomechanical Phenomena , Dogs , Female , Male , Water
3.
Mol Genet Metab ; 116(4): 260-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26547562

ABSTRACT

BACKGROUND: The treatment of long-chain mitochondrial ß-oxidation disorders (LC-FOD) with a low fat-high carbohydrate diet, a diet rich in medium-even-chain triglycerides (MCT), or a combination of both has been associated with high morbidity and mortality for decades. The pathological tableau appears to be caused by energy deficiency resulting from reduced availability of citric acid cycle (CAC) intermediates required for optimal oxidation of acetyl-CoA. This hypothesis was investigated by diet therapy with carnitine and anaplerotic triheptanoin (TH). METHODS: Fifty-two documented LC-FOD patients were studied in this investigation (age range: birth to 51 years). Safety monitoring included serial quantitative measurements of routine blood chemistries, blood levels of carnitine and acylcarnitines, and urinary organic acids. RESULTS: The average frequency of serious clinical complications were reduced from ~60% with conventional diet therapy to 10% with TH and carnitine treatment and mortality decreased from ~65% with conventional diet therapy to 3.8%. Carnitine supplementation was uncomplicated. CONCLUSION: The energy deficiency in LC-FOD patients was corrected safely and more effectively with the triheptanoin diet and carnitine supplement than with conventional diet therapy. Safe intervention in neonates and infants will permit earlier intervention following pre-natal diagnosis or diagnosis by expanded newborn screening.


Subject(s)
Carnitine/therapeutic use , Fatty Acids/metabolism , Lipid Metabolism, Inborn Errors/diet therapy , Mitochondrial Diseases/diet therapy , Triglycerides/therapeutic use , Administration, Oral , Adolescent , Adult , Carnitine/analogs & derivatives , Carnitine/blood , Child , Child, Preschool , Citric Acid/urine , Female , Humans , Infant , Infant, Newborn , Lactic Acid/urine , Lipid Metabolism, Inborn Errors/blood , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/mortality , Malates/urine , Male , Middle Aged , Mitochondrial Diseases/blood , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/mortality , Oxidation-Reduction , Succinic Acid/urine , Survival Analysis , Treatment Outcome
4.
PLoS One ; 9(10): e109527, 2014.
Article in English | MEDLINE | ID: mdl-25299635

ABSTRACT

Rett syndrome (RTT) is an autism spectrum disorder (ASD) caused by mutations in the X-linked MECP2 gene that encodes methyl-CpG binding protein 2 (MeCP2). Symptoms range in severity and include psychomotor disabilities, seizures, ataxia, and intellectual disability. Symptom onset is between 6-18 months of age, a critical period of brain development that is highly energy-dependent. Notably, patients with RTT have evidence of mitochondrial dysfunction, as well as abnormal levels of the adipokines leptin and adiponectin, suggesting overall metabolic imbalance. We hypothesized that one contributor to RTT symptoms is energy deficiency due to defective nutrient substrate utilization by the TCA cycle. This energy deficit would lead to a metabolic imbalance, but would be treatable by providing anaplerotic substrates to the TCA cycle to enhance energy production. We show that dietary therapy with triheptanoin significantly increased longevity and improved motor function and social interaction in male mice hemizygous for Mecp2 knockout. Anaplerotic therapy in Mecp2 knockout mice also improved indicators of impaired substrate utilization, decreased adiposity, increased glucose tolerance and insulin sensitivity, decreased serum leptin and insulin, and improved mitochondrial morphology in skeletal muscle. Untargeted metabolomics of liver and skeletal muscle revealed increases in levels of TCA cycle intermediates with triheptanoin diet, as well as normalizations of glucose and fatty acid biochemical pathways consistent with the improved metabolic phenotype in Mecp2 knockout mice on triheptanoin. These results suggest that an approach using dietary supplementation with anaplerotic substrate is effective in improving symptoms and metabolic health in RTT.


Subject(s)
Citric Acid Cycle/drug effects , Longevity/drug effects , Methyl-CpG-Binding Protein 2/genetics , Motor Activity/drug effects , Rett Syndrome/diet therapy , Triglycerides/administration & dosage , Animals , Behavior, Animal/drug effects , Citric Acid Cycle/genetics , Diet , Disease Models, Animal , Gene Expression , Humans , Lipid Metabolism/drug effects , Male , Metabolome/drug effects , Methyl-CpG-Binding Protein 2/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Rett Syndrome/genetics , Rett Syndrome/physiopathology , Rett Syndrome/psychology
5.
JAMA Neurol ; 71(10): 1255-65, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25110966

ABSTRACT

IMPORTANCE: Disorders of brain metabolism are multiform in their mechanisms and manifestations, many of which remain insufficiently understood and are thus similarly treated. Glucose transporter type I deficiency (G1D) is commonly associated with seizures and with electrographic spike-waves. The G1D syndrome has long been attributed to energy (ie, adenosine triphosphate synthetic) failure such as that consequent to tricarboxylic acid (TCA) cycle intermediate depletion. Indeed, glucose and other substrates generate TCAs via anaplerosis. However, TCAs are preserved in murine G1D, rendering energy-failure inferences premature and suggesting a different hypothesis, also grounded on our work, that consumption of alternate TCA precursors is stimulated and may be detrimental. Second, common ketogenic diets lead to a therapeutically counterintuitive reduction in blood glucose available to the G1D brain and prove ineffective in one-third of patients. OBJECTIVE: To identify the most helpful outcomes for treatment evaluation and to uphold (rather than diminish) blood glucose concentration and stimulate the TCA cycle, including anaplerosis, in G1D using the medium-chain, food-grade triglyceride triheptanoin. DESIGN, SETTING, AND PARTICIPANTS: Unsponsored, open-label cases series conducted in an academic setting. Fourteen children and adults with G1D who were not receiving a ketogenic diet were selected on a first-come, first-enrolled basis. INTERVENTION: Supplementation of the regular diet with food-grade triheptanoin. MAIN OUTCOMES AND MEASURES: First, we show that, regardless of electroencephalographic spike-waves, most seizures are rarely visible, such that perceptions by patients or others are inadequate for treatment evaluation. Thus, we used quantitative electroencephalographic, neuropsychological, blood analytical, and magnetic resonance imaging cerebral metabolic rate measurements. RESULTS: One participant (7%) did not manifest spike-waves; however, spike-waves promptly decreased by 70% (P = .001) in the other participants after consumption of triheptanoin. In addition, the neuropsychological performance and cerebral metabolic rate increased in most patients. Eleven patients (78%) had no adverse effects after prolonged use of triheptanoin. Three patients (21%) experienced gastrointestinal symptoms, and 1 (7%) discontinued the use of triheptanoin. CONCLUSIONS AND RELEVANCE: Triheptanoin can favorably influence cardinal aspects of neural function in G1D. In addition, our outcome measures constitute an important framework for the evaluation of therapies for encephalopathies associated with impaired intermediary metabolism.


Subject(s)
Blood Glucose/metabolism , Brain/metabolism , Carbohydrate Metabolism, Inborn Errors/drug therapy , Citric Acid Cycle , Dietary Supplements , Monosaccharide Transport Proteins/deficiency , Triglycerides/therapeutic use , Adolescent , Adult , Brain/physiopathology , Carbohydrate Metabolism, Inborn Errors/metabolism , Child , Child, Preschool , Cohort Studies , Electroencephalography , Female , Glucose/metabolism , Humans , Magnetic Resonance Imaging , Male , Monosaccharide Transport Proteins/metabolism , Treatment Outcome , Young Adult
6.
Appl Microbiol Biotechnol ; 97(17): 7579-85, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23900800

ABSTRACT

Polyunsaturated fatty acids, docosahexaenoic acid (DHA, 22:6, n-3), eicosapentaenoic acid (EPA, 20:5, n-3), and arachidonic acid (ARA, 20:4 n-6), have multiple beneficial effects on human health and can be used as an important ingredient in dietary supplements, food, feed and pharmaceuticals. A variety of microorganisms has been used for commercial production of these fatty acids. The microorganisms in the Pythium family, particularly Pythium irregulare, are potential EPA producers. The aim of this work is to provide a safety assessment of P. irregulare so that the EPA derived from this species can be potentially used in various commercial applications. The genus Pythium has been widely recognized as a plant pathogen by infecting roots and colonizing the vascular tissues of various plants such as soybeans, corn and various vegetables. However, the majority of the Pythium species (including P. irregulare) have not been reported to infect mammals including humans. The only species among the Pythium family that infects mammals is P. insidiosum. There also have been no reports showing P. irregulare to contain mycotoxins or cause potentially allergenic responses in humans. Based on the safety assessment, we conclude that P. irregulare can be considered a safe source of biomass and EPA-containing oil for use as ingredients in dietary supplements, food, feed and pharmaceuticals.


Subject(s)
Dietary Supplements/analysis , Eicosapentaenoic Acid/biosynthesis , Food Additives/metabolism , Pythium/metabolism , Food Additives/analysis , Food Safety , Industrial Microbiology , Pythium/genetics , Pythium/growth & development , Safety
7.
JAMA Neurol ; 70(6): 756-63, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23609947

ABSTRACT

IMPORTANCE: The physiological relevance of acid maltase (acid α-glucosidase, an enzyme that degrades lysosomal glycogen) is well recognized in liver and muscle. In late (adult)-onset acid maltase deficiency (glycogen storage disease type II [GSD II]), glycogen accumulates inside muscular lysosomes in the context of reduced enzymatic activity present not only in muscle, but also throughout the organism. Yet, disease manifestations are commonly attributed to lysosomal disruption and autophagic vesicle buildup inside the myofiber due to a lack of obvious hepatic or broader metabolic dysfunction. However, current therapies primarily focused on reducing glycogen deposition by dietary or enzyme replacement have not been consistently beneficial, providing the motivation for a better understanding of disease mechanisms. OBJECTIVE: To provide a systematic overview of metabolism and methylation capacity using widely available analytical methods by evaluating secondary compromise of (1) the citric acid cycle, (2) methylation capacity, and (3) nutrient sensor interaction in as many as 33 patients with GSD II (ie, not all patients were available for all assessments) treated with only a low-carbohydrate/high-protein, calorie-balanced diet. DESIGN, SETTING, AND PATIENTS: Case series including clinical and analytical characterization in an academic setting involving 33 enzymatically proved adults with GSD II treated only with a low-carbohydrate/high-protein, calorie-balanced diet. MAIN OUTCOME AND MEASURE: Biochemical analysis of blood and urine samples. RESULTS: Patients exhibited evidence for disturbed energy metabolism contributing to a chronic catabolic state and those who were studied further also displayed diminished plasma methylation capacity and elevated levels of insulin-like growth factor type 1 and its carrier protein insulin-like growth factor binding protein 3 (IGFBP-3). CONCLUSIONS AND RELEVANCE: The simplest unifying interpretation of these abnormalities is nutrient sensor disturbance with secondary energy failure leading to a chronic catabolic state. Data also provide the framework for the investigation of potentially beneficial interventions, including methylation supplementation, as adjuncts specifically targeted to ameliorate the systemic metabolic abnormalities of this disorder.


Subject(s)
Diet, Carbohydrate-Restricted , Dietary Proteins/administration & dosage , Glycogen Storage Disease Type II/diet therapy , Glycogen Storage Disease Type II/metabolism , Glycogen/metabolism , Muscle, Skeletal/metabolism , Adolescent , Adult , Aged , Biomarkers/blood , Biomarkers/urine , Diet, Carbohydrate-Restricted/methods , Energy Metabolism/physiology , Female , Glycogen Storage Disease Type II/diagnosis , Humans , Male , Middle Aged , Young Adult
8.
Mol Genet Metab ; 101(2-3): 246-52, 2010.
Article in English | MEDLINE | ID: mdl-20655781

ABSTRACT

APBD is a rare disorder most often affecting adults of Ashkenazi Jewish origin due to partial deficiency of the glycogen brancher enzyme (GBE). It is characterized by progressive involvement of both the central and peripheral nervous systems and deposition of amylopectin-like polyglucosan bodies. There have been no metabolic derangements that might suggest effective therapy nor have there been any clinical improvements for control of its relentless progression. The APBD patients, in this study, experienced stabilization of disease progression, and limited functional improvement in most patients with dietary triheptanoin. Due to a plateau in clinical improvement, the reduced plasma creatinine and methionine levels prompted evaluation of other plasma methylation intermediates in this complex integrated pathway system: decreased S-adenosylmethionine (SAM) (p<0.002), increased S-adenosylhomocysteine (p<0.001), elevated creatine (p=0.001) and increased free choline (p<0.001). Plasma levels of homocysteine and guanidinoacetate were normal. Impaired metabolism of choline and creatine may relate to the progressive dysmyelination and progressive muscle weakness associated with APBD. The partial deficiency of GBE appears to produce a secondary energy deficit possibly related to inadequate reserves of normal glycogen for efficient degradation to free glucose. Dysfunctional regulation of glycogen synthase (GS) may result in continued synthesis and deposition of polyglucosan bodies. This investigation has demonstrated, for the first time, arrest of clinical deterioration with limited functional recovery with triheptanoin diet therapy and the existence of significant derangement of methylation pathways that, when corrected, may lead to even greater therapeutic benefits.


Subject(s)
1,4-alpha-Glucan Branching Enzyme/deficiency , Glucans/metabolism , Nervous System Diseases/diet therapy , Nervous System Diseases/physiopathology , Triglycerides/therapeutic use , Adult , Aged , Creatine/blood , Female , Humans , Jews/genetics , Male , Methylation , Middle Aged , S-Adenosylhomocysteine/blood , S-Adenosylmethionine/blood
9.
Mol Genet Metab ; 101(1): 9-17, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20598931

ABSTRACT

Pyruvate carboxylase (PC) is a regulated mitochondrial enzyme that catalyzes the conversion of pyruvate to oxaloacetate, a critical transition that replenishes citric acid cycle intermediates and facilitates other biosynthetic reactions that drive anabolism. Its deficiency causes multiorgan metabolic imbalance that predominantly manifests with lactic acidemia and neurological dysfunction at an early age. Three clinical forms of PC deficiency have been identified: an infantile form (Type A), a severe neonatal form (Type B), and a benign form (Type C), all of which exhibit clinical or biochemical correlates of impaired anaplerosis. There is no effective treatment for these patients and most, except those affected by the benign form, die in early life. We review the physiology of this enzyme and dissect the major clinical, biochemical, and genetic aspects of its dysfunction, emphasizing features that distinguish PC deficiency from other causes of lactic acidemia that render PC deficiency potentially treatable using novel interventions capable of enhancing anaplerosis.


Subject(s)
Pyruvate Carboxylase Deficiency Disease/metabolism , Pyruvate Carboxylase/genetics , Animals , Carbon/metabolism , Humans , Oxaloacetic Acid/metabolism , Phenotype , Pyruvate Carboxylase/metabolism , Pyruvic Acid/metabolism
10.
Am J Vet Res ; 71(3): 326-36, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20187835

ABSTRACT

OBJECTIVE: To evaluate effects of fats with odd and even numbers of carbon atoms on muscle metabolism in exercising horses with polysaccharide storage myopathy (PSSM). ANIMALS: 8 horses with PSSM (6 females and 2 males; mean +/- SD age, 6.3 +/- 3.9 years). PROCEDURES: Isocaloric diets (grain, triheptanoin, corn oil, and high-fat, low-starch [HFLS] feed) were fed for 3 weeks each; horses performed daily treadmill exercise. Grain was fed to establish an exercise target, and HFLS feed was fed as a negative control diet. Daily plasma samples were obtained. For each diet, a 15-minute exercise test was performed, and gluteus medius muscle specimens and blood samples were obtained before and after exercise. RESULTS: Feeding triheptanoin, compared with the corn oil diet, resulted in exercise intolerance; higher plasma creatine kinase (CK) activity and concentrations of C3:0- and C7:0-acylcarnitine and insulin; and lower concentrations of nonesterified fatty acids (NEFA) and C16:0-, C18:1-, and C18:2-acylcarnitine, without changes in concentrations of plasma glucose or resting muscle substrates and metabolites. Feeding grain induced higher CK activity and insulin concentrations and lower NEFA concentrations than did corn oil or HFLS feed. Feeding grain induced higher glucose concentrations than did triheptanoin and corn oil. In muscle, feeding grain resulted in lower glucose-6-phosphate, higher citrate, and higher postexercise lactate concentrations than did the other diets. CONCLUSIONS AND CLINICAL RELEVANCE: Triheptanoin had detrimental effects, reflecting decreased availability of NEFA, increased insulin stimulation of glycogen synthesis, and potential inhibition of lipid oxidation. Long-chain fats are the best dietetic for PSSM.


Subject(s)
Dietary Fats/pharmacology , Horse Diseases/physiopathology , Horses/physiology , Muscle, Skeletal/pathology , Muscular Diseases/veterinary , Physical Conditioning, Animal/adverse effects , Animal Feed , Animals , Blood Glucose/metabolism , Creatine Kinase/blood , Edible Grain , Female , Horse Diseases/blood , Horse Diseases/etiology , Horse Diseases/pathology , Horses/classification , Insulin/blood , Male , Metabolism/drug effects , Muscle, Skeletal/metabolism , Muscular Diseases/blood , Muscular Diseases/metabolism , Muscular Diseases/pathology , Running , Species Specificity , Walking
11.
Am J Physiol Endocrinol Metab ; 298(2): E362-71, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19903863

ABSTRACT

The anaplerotic odd-medium-chain triglyceride triheptanoin is used in clinical trials for the chronic dietary treatment of patients with long-chain fatty acid oxidation disorders. We previously showed (Kinman RP, Kasumov T, Jobbins KA, Thomas KR, Adams JE, Brunengraber LN, Kutz G, Brewer WU, Roe CR, Brunengraber H. Am J Physiol Endocrinol Metab 291: E860-E866, 2006) that the intravenous infusion of triheptanoin increases lipolysis traced by the turnover of glycerol. In this study, we tested whether lipolysis induced by triheptanoin infusion is accompanied by the potentially harmful release of long-chain fatty acids. Rats were infused with heptanoate +/- glycerol or triheptanoin. Intravenous infusion of triheptanoin at 40% of caloric requirement markedly increased glycerol endogenous R(a) but not oleate endogenous R(a). Thus, the activation of lipolysis was balanced by fatty acid reesterification in the same cells. The liver acyl-CoA profile showed the accumulation of intermediates of heptanoate beta-oxidation and C(5)-ketogenesis and a decrease in free CoA but no evidence of metabolic perturbation of liver metabolism such as propionyl overload. Our data suggest that triheptanoin, administered either intravenously or intraduodenally, could be used for intensive care and nutritional support of metabolically decompensated long-chain fatty acid oxidation disorders.


Subject(s)
Adipocytes/metabolism , Fatty Acids/metabolism , Lipolysis/physiology , Triglycerides/administration & dosage , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Acetyl-CoA C-Acyltransferase/metabolism , Animals , Carbon-Carbon Double Bond Isomerases/metabolism , Enoyl-CoA Hydratase/metabolism , Enteral Nutrition , Esterification/physiology , Glucose/metabolism , Heptanoates/administration & dosage , Infusions, Intravenous , Lipase/metabolism , Liver/enzymology , Male , Racemases and Epimerases/metabolism , Rats , Rats, Sprague-Dawley , Triglycerides/metabolism
12.
Am J Vet Res ; 70(8): 1043-52, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19645587

ABSTRACT

OBJECTIVE: To compare effects of corn oil or a 7-carbon fat (triheptanoin) on acylcarnitine, lipid, and carbohydrate metabolism in plasma or muscle of exercising horses. ANIMALS: 8 Thoroughbred geldings. PROCEDURES: Horses received isocaloric diets containing 650 mL of oil (triheptanoin or corn oil)/d for 18 or 25 days in a crossover design with a 26-day washout period. On day 17 or 24 of each feeding period, the respective oil (217 mL) was nasogastrically administered; 120 minutes later, horses performed a 90-minute submaximal exercise test (SET). Blood and muscle samples were obtained before oil administration and immediately before (blood only), during (blood only), immediately after, and 24 hours after SETs. RESULTS: Compared with values before oil administration, triheptanoin administration increased plasma insulin and C7:0-, C5:0- and C3:0-acylcarnitine concentrations, whereas corn oil administration increased plasma NEFA concentrations. During SETs, plasma C7:0-, C5:0-, and C3:0-acylcarnitine concentrations were higher when triheptanoin, rather than corn oil, was administered to horses. Plasma glucose, NEFA, and C2:0-, C18:1-, and C18:2-acylcarnitine concentrations increased during SETs similarly for both oils. Respiratory quotient and muscle lactate, citrate, malate, glycogen, and ATP concentrations changed similarly from before to after SETs for both oils. Compared with muscle concentrations immediately after SETs, those for glucose-6-phosphate and citrate 24 hours after SETs were lower and for glycogen were similar to values before SETs. CONCLUSIONS AND CLINICAL RELEVANCE: Fatigue was not associated with depletion of citric acid cycle intermediates for either oil. Triheptanoin induced a significantly higher insulin secretion and did not appear to enhance muscle glycogen repletion.


Subject(s)
Horses/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Physical Conditioning, Animal/physiology , Triglycerides/pharmacology , Analysis of Variance , Animals , Blood Glucose , Carbohydrate Metabolism/drug effects , Carnitine/analogs & derivatives , Carnitine/blood , Corn Oil , Cross-Over Studies , Insulin/blood , Lipid Metabolism/drug effects , Random Allocation
13.
J Biol Chem ; 284(41): 27799-27807, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19666922

ABSTRACT

We investigated the interrelations between C(4) ketogenesis (production of beta-hydroxybutyrate + acetoacetate), C(5) ketogenesis (production of beta-hydroxypentanoate + beta-ketopentanoate), and anaplerosis in isolated rat livers perfused with (13)C-labeled octanoate, heptanoate, or propionate. Mass isotopomer analysis of C(4) and C(5) ketone bodies and of related acyl-CoA esters reveal that C(4) and C(5) ketogenesis share the same pool of acetyl-CoA. Although the uptake of octanoate and heptanoate by the liver are similar, the rate of C(5) ketogenesis from heptanoate is much lower than the rate of C(4) ketogenesis from octanoate. This results from the channeling of the propionyl moiety of heptanoate into anaplerosis of the citric acid cycle. C(5) ketogenesis from propionate is virtually nil because acetoacyl-CoA thiolase does not favor the formation of beta-ketopentanoyl-CoA from propionyl-CoA and acetyl-CoA. Anaplerosis and gluconeogenesis from heptanoate are inhibited by octanoate. The data have implications for the design of diets for the treatment of long chain fatty acid oxidation disorders, such as the triheptanoin-based diet.


Subject(s)
3-Hydroxybutyric Acid/metabolism , Acetoacetates/metabolism , Ketone Bodies/biosynthesis , Liver/metabolism , Pentanoic Acids/metabolism , 3-Hydroxybutyric Acid/chemistry , Acetoacetates/chemistry , Animals , Caprylates/chemistry , Caprylates/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Gas Chromatography-Mass Spectrometry , Glucose/metabolism , Heptanoates/chemistry , Heptanoates/metabolism , Ketone Bodies/chemistry , Lipid Metabolism , Male , Oxidation-Reduction , Pentanoic Acids/chemistry , Propionates/chemistry , Propionates/metabolism , Rats , Rats, Sprague-Dawley
14.
Mol Genet Metab ; 93(2): 179-89, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18054510

ABSTRACT

We report 10 children (7 male, 3 female), 3 homozygous for c.319C>T mutation and 7 heterozygous for c.319C>T on one allele and c.625G>A variant on the other in the short-chain acyl-CoA dehydrogenase (SCAD) gene (ACADS). All were of Ashkenazi Jewish origin in which group we found a c.319C>T heterozygote frequency of 1:15 suggesting the presence of a founder mutation or selective advantage. Phenotype was variable with onset from birth to early childhood. Features included hypotonia (8/10), developmental delay (8/10), myopathy (4/10) with multicore changes in two and lipid storage in one, facial weakness (3/10), lethargy (5/10), feeding difficulties (4/10) and congenital abnormalities (3/7). One female with multiminicore myopathy had progressive external ophthalmoplegia, ptosis and cardiomyopathy with pneumonia and respiratory failure. Two brothers presented with psychosis, pyramidal signs, and multifocal white matter abnormalities on MRI brain suggesting additional genetic factors. Two other infants also had white matter changes. Elevated butyrylcarnitine (4/8), ethylmalonic aciduria (9/9), methylsuccinic aciduria (6/7), decreased butyrate oxidation in lymphoblasts (2/4) and decreased SCAD activity in fibroblasts or muscle (3/3) were shown. Expression studies of c.319C>T in mouse liver mitochondria showed it to be inactivating. c.625G>A is a common variant in ACADS that may confer disease susceptibility. Five healthy parents were heterozygous for c.319C>T and c.625G>A, suggesting reduced penetrance or broad clinical spectrum. We conclude that the c.319C>T mutation can lead to wide clinical and biochemical phenotypic variability, suggesting a complex multifactorial/polygenic condition. This should be screened for in individuals with multicore myopathy, particularly among the Ashkenazim.


Subject(s)
Butyryl-CoA Dehydrogenase/deficiency , Butyryl-CoA Dehydrogenase/genetics , Founder Effect , Jews/genetics , Point Mutation , Abnormalities, Multiple/enzymology , Abnormalities, Multiple/genetics , Adolescent , Adult , Alleles , Animals , Base Sequence , Child , Child, Preschool , DNA Primers/genetics , Female , Heterozygote , Homozygote , Humans , Infant , Infant, Newborn , Male , Metabolism, Inborn Errors/enzymology , Metabolism, Inborn Errors/genetics , Mice , Muscular Diseases/enzymology , Muscular Diseases/genetics , Phenotype , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
15.
Mol Genet Metab ; 92(4): 346-50, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17825594

ABSTRACT

Patients with mitochondrial long-chain fat oxidation deficiencies are usually treated with diets containing reduced fat and increased carbohydrate, at times via gastrostomy feeding. To ensure adequate intake of essential fatty acids, supplements are provided to their diets using commercially available oils. These oils contain large quantities of non-essential fats that are preferentially oxidized and produce disease-specific metabolites (acyl-CoA intermediates) due to the genetic defect. This study describes the concentrations of these intermediates as reflected by acylcarnitines as well as the % contribution from each of four fatty acids: palmitate, oleate, linoleate, and alpha-linolenate when incubated with fibroblasts from patients with VLCAD, LCHAD, and trifunctional protein (TFP) deficiencies. Palmitate and oleate produce the majority of disease-specific acylcarnitines with these defective cell lines (79-94%) whereas linoleate and linolenate produced less (6-21%). On average, the amount of acylcarnitines decreased with increasing unsaturation (C18:1>C18:2>C18:3:34%>11%>3%, respectively. This relationship may reflect the "gatekeeper" role of carnitine palmitoyltransferase I (CPT I). A diet comparison between Canola and a combination of Flax/Walnut oils revealed that the latter, containing the least amount of non-essential fats, reduced blood acylcarnitine levels by 33-36%. The etiology of the severe peripheral neuropathy of TFP deficiency may result from the unique metabolite, 3-keto-acyl-CoA, after conversion to a methylketone via spontaneous decarboxylation. Essential fatty acid supplementation with oils should consider these findings to decrease production of disease-specific acyl-CoA intermediates.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Carnitine/analogs & derivatives , Diet Therapy , Lipid Metabolism, Inborn Errors/diet therapy , Multienzyme Complexes/deficiency , Carnitine/metabolism , Cell Line , Diet Therapy/methods , Dietary Fats , Energy Intake , Fibroblasts/metabolism , Lipid Metabolism, Inborn Errors/etiology , Multienzyme Complexes/genetics , Oils , Oxidation-Reduction , Polyneuropathies/genetics , Polyneuropathies/metabolism
16.
Mol Genet Metab ; 2007 Jan 17.
Article in English | MEDLINE | ID: mdl-17234443

ABSTRACT

This article has been removed, consistent with Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). The Publisher apologizes for any inconvenience this may cause.

17.
Pediatr Res ; 60(3): 315-20, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16857760

ABSTRACT

The isobutyryl-CoA dehydrogenase (IBD) enzyme is involved in the degradation of valine. IBD deficiency was first reported in 1998 and subsequent genetic investigations identified acyl-CoA dehydrogenase (ACAD) 8, now IBD, as the gene responsible for IBD deficiency. Only three individuals homozygous or compound heterozygous for variations in the IBD gene have been reported. We present IBD deficiency in an additional four newborns with elevated C(4)-carnitine identified by tandem mass spectrometry (MS/MS) screening in Denmark and the United States. Three showed urinary excretions of isobutyryl-glycine, and in vitro probe analysis of fibroblasts from two newborns indicated enzymatic IBD defect. Molecular genetic analysis revealed seven new rare variations in the IBD gene (c.348C>A, c.400G>T, c.409G>A, c.455T>C, c.958G>A, c.1000C>T and c.1154G>A). Furthermore, sequence analysis of the short-chain acyl-CoA dehydrogenase (SCAD) gene revealed heterozygosity for the prevalent c.625G>A susceptibility variation in all newborns and in the first reported IBD patient. Functional studies in isolated mitochondria demonstrated that the IBD variations present in the Danish newborn (c.409G>A and c.958G>A) together with a previously published IBD variation (c.905G>A) disturbed protein folding and reduced the levels of correctly folded IBD tetramers. Accordingly, low/no IBD residual enzyme activity was detectable when the variant IBD proteins were overexpressed in Chang cells.


Subject(s)
Acyl-CoA Dehydrogenases/genetics , Carnitine/metabolism , Genetic Variation , Neonatal Screening , Female , Humans , Infant, Newborn , Male , Mass Spectrometry , Point Mutation , Protein Folding , Protein Structure, Quaternary/genetics
18.
J Inherit Metab Dis ; 29(2-3): 332-40, 2006.
Article in English | MEDLINE | ID: mdl-16763896

ABSTRACT

Beginning with phenylketonuria, dietary therapy for inborn errors has focused primarily on the restriction of the precursor to an affected catabolic pathway in an attempt to limit the production of potential toxins. Anaplerotic therapy is based on the concept that there may exist an energy deficit in these diseases that might be improved by providing alternative substrate for both the citric acid cycle (CAC) and the electron transport chain for enhanced ATP production. This article focuses on this basic problem, as it may relate to most catabolic disorders, and provides our current experience involving inherited diseases of mitochondrial fat oxidation, glycogen storage, and pyruvate metabolism using the anaplerotic compound triheptanoin. The observations have led to a realization that 'inter-organ' signalling and 'nutrient sensors' such as adenylate monophosphate mediated-protein kinase (AMPK) and mTOR (mammalian target of rapamycin) appear to play a significant role in the intermediary metabolism of these diseases. Activated AMPK turns on catabolic pathways to augment ATP production while turning off synthetic pathways that consume ATP. Information is provided regarding the inter-organ requirements for more normal metabolic function during crisis and how anaplerotic therapy using triheptanoin, as a direct source of substrate to the CAC for energy production, appears to be a more successful approach to an improved quality of life for these patients.


Subject(s)
Citric Acid Cycle , Heptanoates/pharmacology , Heptanoates/therapeutic use , Metabolism, Inborn Errors/diet therapy , Triglycerides/pharmacology , Triglycerides/therapeutic use , Animals , Fatty Acids/metabolism , Glycogen Storage Disease Type II/diet therapy , Glycogen Storage Disease Type II/metabolism , Heptanoates/metabolism , Humans , Lipid Metabolism, Inborn Errors/diet therapy , Lipid Metabolism, Inborn Errors/metabolism , Metabolism, Inborn Errors/metabolism , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Oxidation-Reduction , Pyruvate Carboxylase Deficiency Disease/diet therapy , Pyruvate Carboxylase Deficiency Disease/metabolism , Triglycerides/metabolism
19.
J Inherit Metab Dis ; 29(2-3): 327-31, 2006.
Article in English | MEDLINE | ID: mdl-16763895

ABSTRACT

This review presents the concepts of anaplerosis and cataplerosis in relation to the regulation of citric acid cycle operation. Anaplerosis is the re-filling of the catalytic intermediates of the cycle that carry acetyl-CoA as it is oxidized. The main anaplerotic substrates are pyruvate, glutamine/glutamate and precursors of propionyl-CoA (odd-chain fatty acids, specific amino acids, C(5)-ketone bodies). Cataplerosis balances anaplerosis by removing excess intermediates from the citric acid cycle. The properties of the main anaplerotic substrates are reviewed from the point of view of potential clinical applications to the treatment of some inherited and acquired conditions.


Subject(s)
Acyl Coenzyme A/metabolism , Citric Acid Cycle , Glutamic Acid/metabolism , Glutamine/metabolism , Pyruvic Acid/metabolism , Animals , Glutamic Acid/therapeutic use , Glutamine/therapeutic use , Heptanoates/metabolism , Humans , Ketone Bodies/metabolism , Metabolism, Inborn Errors/diet therapy , Metabolism, Inborn Errors/metabolism , Propionates/metabolism , Pyruvic Acid/therapeutic use , Reperfusion Injury/metabolism , Triglycerides/metabolism , Valerates/metabolism
20.
Am J Physiol Endocrinol Metab ; 291(4): E860-6, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16705058

ABSTRACT

A new chronic treatment for inherited disorders of long-chain fatty acid oxidation involves administering up to one-third of dietary calories as triheptanoin, a medium-odd-chain triglyceride (Roe CR, Sweetman L, Roe DS, David F, and Brunengraber H. J Clin Invest 110: 259-269, 2002). Heptanoate and C(5)-ketone bodies derived from its partial oxidation in liver are precursors of anaplerotic propionyl-CoA in peripheral tissues. It was hypothesized that increasing anaplerosis in peripheral tissues would boost energy production. In the present study, we tested the potential of a triheptanoin emulsion as an intravenous nutrient. Normal rats were infused with triheptanoin intravenously or intraduodenally at up to 40% of caloric requirement. The blood concentration ratio (heptanoate/C(5)-ketone bodies) was high with intravenous and low with intraduodenal triheptanoin infusion. During intravenous infusion of triheptanoin, lipolysis was stimulated but appeared compensated by fatty acid reesterification. During intraduodenal infusion of triheptanoin, lipolysis was not stimulated. Our data support the hypothesis that intravenous triheptanoin could be used to treat decompensated patients with long-chain fatty acid oxidation disorders.


Subject(s)
Heptanoates/administration & dosage , Heptanoates/pharmacokinetics , Lipid Metabolism, Inborn Errors/diet therapy , Animals , Blood Glucose/metabolism , Enteral Nutrition , Glycerol/blood , Heptanoates/blood , Infusions, Intravenous , Ketone Bodies/blood , Kinetics , Lipid Metabolism, Inborn Errors/blood , Male , Rats , Rats, Sprague-Dawley , Triglycerides/administration & dosage , Triglycerides/blood , Triglycerides/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...