Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 427
Filter
Add more filters

Complementary Medicines
Publication year range
1.
J Nutr ; 154(6): 1781-1789, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615734

ABSTRACT

BACKGROUND: Infant formulas are typically manufactured using skimmed milk, whey proteins, and vegetable oils, which excludes milk fat globule membranes (MFGM). MFGM contains polar lipids, including sphingomyelin (SM). OBJECTIVE: The objective of this study was comparison of infant plasma SM and acylcarnitine species between infants who are breastfed or receiving infant formulas with different fat sources. METHODS: In this explorative study, we focused on SM and acylcarnitine species concentrations measured in plasma samples from the TIGGA study (ACTRN12608000047392), where infants were randomly assigned to receive either a cow milk-based infant formula (CIF) with vegetable oils only or a goat milk-based infant formula (GIF) with a goat milk fat (including MFGM) and vegetable oil mixture to the age ≥4 mo. Breastfed infants were followed as a reference group. Using tandem mass spectrometry, SM species in the study formulas and SM and acylcarnitine species in plasma samples collected at the age of 4 mo were analyzed. RESULTS: Total SM concentrations (∼42 µmol/L) and patterns of SM species were similar in both formulas. The total plasma SM concentrations were not different between the formula groups but were 15 % (CIF) and 21% (GIF) lower in the formula groups than in the breastfed group. Between the formula groups, differences in SM species were statistically significant but small. Total carnitine and major (acyl) carnitine species were not different between the groups. CONCLUSIONS: The higher total SM concentration in breastfed than in formula-fed infants might be related to a higher SM content in human milk, differences in cholesterol metabolism, dietary fatty acid intake, or other factors not yet identified. SM and acylcarnitine species composition in plasma is not closely related to the formula fatty acid composition. This trial was registered at Australian New Zealand Clinical Trials Registry as ACTRN12608000047392.


Subject(s)
Carnitine , Goats , Infant Formula , Milk, Human , Milk , Sphingomyelins , Humans , Infant Formula/chemistry , Animals , Carnitine/blood , Carnitine/analogs & derivatives , Milk, Human/chemistry , Infant , Sphingomyelins/blood , Milk/chemistry , Female , Male , Cattle , Breast Feeding , Esters/blood , Infant, Newborn , Plant Oils/chemistry
2.
Lipids Health Dis ; 20(1): 151, 2021 Nov 02.
Article in English | MEDLINE | ID: mdl-34727932

ABSTRACT

BACKGROUND: Acylcarnitine is an intermediate product of fatty acid oxidation. It is reported to be closely associated with the occurrence of diabetic cardiomyopathy (DCM). However, the mechanism of acylcarnitine affecting myocardial disorders is yet to be explored. This current research explores the different chain lengths of acylcarnitines as biomarkers for the early diagnosis of DCM and the mechanism of acylcarnitines for the development of DCM in-vitro. METHODS: In a retrospective non-interventional study, 50 simple type 2 diabetes mellitus patients and 50 DCM patients were recruited. Plasma samples from both groups were analyzed by high throughput metabolomics and cluster heat map using mass spectrometry. Principal component analysis was used to compare the changes occurring in the studied 25 acylcarnitines. Multivariable binary logistic regression was used to analyze the odds ratio of each group for factors and the 95% confidence interval in DCM. Myristoylcarnitine (C14) exogenous intervention was given to H9c2 cells to verify the expression of lipid metabolism-related protein, inflammation-related protein expression, apoptosis-related protein expression, and cardiomyocyte hypertrophy and fibrosis-related protein expression. RESULTS: Factor 1 (C14, lauroylcarnitine, tetradecanoyldiacylcarnitine, 3-hydroxyl-tetradecanoylcarnitine, arachidic carnitine, octadecanoylcarnitine, 3-hydroxypalmitoleylcarnitine) and factor 4 (octanoylcarnitine, hexanoylcarnitine, decanoylcarnitine) were positively correlated with the risk of DCM. Exogenous C14 supplementation to cardiomyocytes led to increased lipid deposition in cardiomyocytes along with the obstacles in adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) signaling pathways and affecting fatty acid oxidation. This further caused myocardial lipotoxicity, ultimately leading to cardiomyocyte hypertrophy, fibrotic remodeling, and increased apoptosis. However, this effect was mitigated by the AMPK agonist acadesine. CONCLUSIONS: The increased plasma levels in medium and long-chain acylcarnitine extracted from factors 1 and 4 are closely related to the risk of DCM, indicating that these factors can be an important tool for DCM risk assessment. C14 supplementation associated lipid accumulation by inhibiting the AMPK/ACC/CPT1 signaling pathway, aggravated myocardial lipotoxicity, increased apoptosis apart from cardiomyocyte hypertrophy and fibrosis were alleviated by the acadesine.


Subject(s)
Carnitine/analogs & derivatives , Diabetes Mellitus, Type 2/complications , Diabetic Cardiomyopathies/metabolism , Lipid Metabolism , Adult , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Biomarkers/blood , Carnitine/blood , Carnitine/chemistry , Carnitine/pharmacology , Cell Line , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Lipid Metabolism/drug effects , Male , Mass Spectrometry , Middle Aged , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Myristic Acids/pharmacology , Rats , Retrospective Studies , Ribonucleosides/pharmacology , Risk Factors
3.
Sci Rep ; 11(1): 16616, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34400736

ABSTRACT

We aimed to evaluate factors associated with changes in skeletal muscle mass in hepatitis C virus (HCV)-infected patients after treatment with direct-acting antivirals (DAAs). Consecutive HCV-infected patients after treatment with DAA were recruited into the study. Patients who achieved sustained virological response (SVR); and had complete clinical information, preserved serum samples at baseline and SVR48, and skeletal muscle mass evaluations based on the psoas muscle mass index (PMI) on computed tomography at baseline and ≥ 12 months were included. Altogether, 70.7% of patients (41/58) showed increased PMI after DAA therapy, and mean relative PMI was significantly higher after DAA therapy than at baseline. There were no significant associations between baseline clinical factors routinely examined in clinical practice and increased PMI. Among factors reported to be associated with skeletal muscle loss in patients with chronic liver disease, serum zinc levels and total and free carnitine levels increased significantly after DAA therapy and only changes in serum free carnitine levels were significantly associated with an increased PMI (r = 0305, P = 0.020). In conclusion, increased skeletal muscle mass after successful HCV eradication by DAAs was significantly associated with increased serum-free carnitine levels. L-carnitine supplementation may be beneficial in patients with low skeletal muscle mass after DAA.


Subject(s)
Antiviral Agents/therapeutic use , Carnitine/blood , Hepatitis C, Chronic/drug therapy , Psoas Muscles/pathology , Adult , Aged , Aged, 80 and over , Amino Acids, Branched-Chain/blood , Carnitine/pharmacology , Carnitine/therapeutic use , Female , Hepatitis C, Chronic/blood , Hepatitis C, Chronic/pathology , Humans , Insulin-Like Growth Factor I/analysis , Male , Middle Aged , Organ Size , Sustained Virologic Response , Vitamin D/blood , Zinc/blood
4.
Am J Physiol Regul Integr Comp Physiol ; 321(3): R429-R440, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34318701

ABSTRACT

Lipopolysaccharides (LPS) challenge the metabolic integrity of high-yielding dairy cows, activating the immune system and altering energy metabolism. Fatty acid oxidation, a major energy-gaining pathway, can be improved by supplementary carnitine, facilitating the transport of fatty acids into mitochondria. The metabolic response to the LPS challenge could alter both the plasma and the milk metabolome. Plasma and milk samples collected from cows treated with (n = 27) or without (n = 27) dietary carnitine, before and after intravenous administration of LPS, were subjected to a targeted metabolomics analysis. Multivariate statistical analyses revealed that both plasma and milk metabolome changed in response to the LPS challenge in both the carnitine-supplemented and the control cows. Short-chain acylcarnitines (carbon chain length C2, C3, C4, and C5) and long-chain acylcarnitines (C14, C16, and C18) had the highest performance to indicate LPS response when testing the predictive power of single metabolites using receiver-operator characteristics (ROC) analysis. The maximum area under a ROC curve (AUC) was 0.93. Biogenic amines, including sarcosine, and amino acids such as glutamine and isoleucine had AUC > 0.80 indicating metabolic changes due to the LPS challenge. In summary, the metabolites involved in the LPS response were acylcarnitines C2 and C5, sarcosine, glutamine, and isoleucine in plasma, and acylcarnitines C4 and C5 in milk. The interrelationship of plasma and milk metabolome included correlation of acylcarnitines C2, C4, and C5 between plasma and milk.


Subject(s)
Carnitine/analogs & derivatives , Lactation/drug effects , Lipopolysaccharides/pharmacology , Milk/metabolism , Animals , Carnitine/blood , Cattle , Diet/veterinary , Dietary Supplements , Energy Metabolism/drug effects , Energy Metabolism/physiology , Fatty Acids/metabolism , Metabolomics/methods , Milk/drug effects
5.
Nutrients ; 13(6)2021 May 31.
Article in English | MEDLINE | ID: mdl-34073024

ABSTRACT

L-carnitine (LC) supplementation improves cardiac function in hemodialysis (HD) patients. However, whether reducing LC supplementation affects carnitine kinetics and cardiac function in HD patients treated with LC remains unclear. Fifty-nine HD patients previously treated with intravenous LC 1000 mg per HD session (three times weekly) were allocated to three groups: LC injection three times weekly, once weekly, and placebo, and prospectively followed up for six months. Carnitine fractions were assessed by enzyme cycling methods. Plasma and red blood cell (RBC) acylcarnitines were profiled using tandem mass spectrometry. Cardiac function was evaluated using echocardiography and plasma B-type natriuretic peptide (BNP) levels. Reducing LC administration to once weekly significantly decreased plasma carnitine fractions and RBC-free carnitine levels during the study period, which were further decreased in the placebo group (p < 0.001). Plasma BNP levels were significantly elevated in the placebo group (p = 0.03). Furthermore, changes in RBC (C16 + C18:1)/C2 acylcarnitine ratio were positively correlated with changes in plasma BNP levels (ß = 0.389, p = 0.005). Reducing LC administration for six months significantly decreased both plasma and RBC carnitine levels, while the full termination of LC increased plasma BNP levels; however, it did not influence cardiac function in HD patients.


Subject(s)
Carnitine/blood , Carnitine/pharmacokinetics , Dietary Supplements , Heart Failure/prevention & control , Heart/drug effects , Kidney Failure, Chronic/therapy , Renal Dialysis/methods , Aged , Carnitine/administration & dosage , Dose-Response Relationship, Drug , Female , Follow-Up Studies , Heart/physiopathology , Heart Failure/complications , Humans , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/complications , Male , Middle Aged , Prospective Studies , Single-Blind Method
6.
Mol Genet Metab ; 134(1-2): 37-42, 2021.
Article in English | MEDLINE | ID: mdl-34176718

ABSTRACT

Elevated citrulline and C5-OH levels are reported as part of the newborn screening of core and secondary disorders on the Recommended Uniform Screening Panel (RUSP). Additionally, some state laboratory newborn screening programs report low citrulline levels, which may be observed in proximal urea cycle disorders. We report six patients who were found on newborn screening to have low citrulline and/or elevated C5-OH levels in whom confirmatory testing showed the combination of these two abnormal analytes. Mitochondrial sequencing revealed known pathogenic variants in MT-ATP6 at high heteroplasmy levels in all cases. MT-ATP6 at these heteroplasmy levels is associated with Leigh syndrome, a progressive neurodegenerative disease. Patients were treated with supplemental citrulline and, in some cases, mitochondrial cofactor therapy. These six patients have not experienced metabolic crises or developmental regression, and early diagnosis and management may help prevent the neurological sequelae of Leigh syndrome. The affected mothers and siblings are asymptomatic or paucisymptomatic (e.g. intellectual disability, depression, migraines, obsessive-compulsive disorder, and poor balance) despite high heteroplasmy or apparent homoplasmy of the familial variant, thus expanding the clinical spectrum seen in pathogenic variants of MT-ATP6. Confirmatory plasma amino acid analysis and acylcarnitine profiling should be ordered in a patient with either low citrulline and/or elevated C5-OH, as this combination appears specific for pathogenic variants in MT-ATP6.


Subject(s)
Genetic Testing/methods , Leigh Disease/diagnosis , Leigh Disease/genetics , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Neonatal Screening/methods , Carnitine/blood , Carnitine/chemistry , Citrulline/blood , DNA, Mitochondrial/genetics , Female , Humans , Infant, Newborn , Male , Prospective Studies
7.
Ther Apher Dial ; 25(5): 595-606, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33470531

ABSTRACT

We investigated the effectiveness of monitoring serum carnitine levels in hemodialysis patients receiving L-carnitine supplementation. One-hundred forty-five hemodialysis patients were divided into three groups. Group 1 consisted of patients (n = 30) who had been receiving supplementation before this study and then discontinued at the beginning. The remaining patients were divided into Group 2 (n = 13) and Group 3 (n = 102) based on their baseline free carnitine (FC) level, <20 or ≥ 20 µmol/L. Group 2 was started on supplementation, and Groups 1 and 3 were observed without any intervention for the first 6 months. FC was measured every 6 months in all three groups up to 18 months. All patients in whom FC was less than 20 µmol/L at 6 and 12 months were prescribed supplementation. After the first 6 months, the mean ± SD of FC changed from 262.5 ± 87.5 µmol/L at baseline to 70.8 ± 33.6 µmol/L (P < 0.001) in Group 1, from 17.4 ± 1.9 to 193.9 ± 43.3 µmol/L (P < 0.001) in Group 2, and from 49.2 ± 24.6 to 44.2 ± 19.8 µmol/L (P < 0.05) in Group 3. The acyl/free carnitine changed from 0.62 to 0.59 in Group 1 (P = 0.287), from 0.76 to 0.66 in Group 2 (P = 0.054) and from 0.57 to 0.60 in Group 3 (P < 0.05). Of the 145 patients, 126 continued follow-up for the full 18 months. FC remained in the normal range (36-74 µmol/L) within the 95% CI. FC was considered a strong predictor of carnitine deficiency after 6 months (AUC: 0.9146, cut-off value: 33.8 µmol/L). FC monitoring is essential for appropriate carnitine supplementation in hemodialysis patients.


Subject(s)
Cardiomyopathies/prevention & control , Carnitine/administration & dosage , Carnitine/blood , Carnitine/deficiency , Hyperammonemia/prevention & control , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/therapy , Muscular Diseases/prevention & control , Renal Dialysis/methods , Aged , Cardiomyopathies/etiology , Dietary Supplements , Female , Humans , Hyperammonemia/etiology , Male , Muscular Diseases/etiology , Renal Dialysis/adverse effects
8.
Nutrients ; 13(2)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33513734

ABSTRACT

BACKGROUND: Research is limited in evaluating the mechanisms responsible for infant growth in response to different protein-rich foods; Methods: Targeted and untargeted metabolomics analysis were conducted on serum samples collected from an infant controlled-feeding trial that participants consumed a meat- vs. dairy-based complementary diet from 5 to 12 months of age, and followed up at 24 months. RESULTS: Isoleucine, valine, phenylalanine increased and threonine decreased over time among all participants; Although none of the individual essential amino acids had a significant impact on changes in growth Z scores from 5 to 12 months, principal component heavily weighted by BCAAs (leucine, isoleucine, valine) and phenylalanine had a positive association with changes in length-for-age Z score from 5 to 12 months. Concentrations of acylcarnitine-C4, acylcarnitine-C5 and acylcarnitine-C5:1 significantly increased over time with the dietary intervention, but none of the acylcarnitines were associated with infant growth Z scores. Quantitative trimethylamine N-oxide increased in the meat group from 5 to 12 months; Conclusions: Our findings suggest that increasing total protein intake by providing protein-rich complementary foods was associated with increased concentrations of certain essential amino acids and short-chain acyl-carnitines. The sources of protein-rich foods (e.g., meat vs. dairy) did not appear to differentially impact serum metabolites, and comprehensive mechanistic investigations are needed to identify other contributors or mediators of the diet-induced infant growth trajectories.


Subject(s)
Dairy Products , Diet , Infant Nutritional Physiological Phenomena , Meat , Metabolomics , Amino Acids, Branched-Chain/blood , Amino Acids, Essential/blood , Carnitine/analogs & derivatives , Carnitine/blood , Follow-Up Studies , Humans , Infant , Isoleucine/blood , Leucine/blood , Phenylalanine/blood , Valine/blood
9.
Am J Perinatol ; 38(8): 828-833, 2021 07.
Article in English | MEDLINE | ID: mdl-31891954

ABSTRACT

OBJECTIVE: Cystic fibrosis (CF) is a multisystemic inherited disease. The aim of this study was to determine free carnitine (FC) and acylcarnitine concentrations in CF newborns with various mutations of the CFTR gene perinatally. STUDY DESIGN: FC/acylcarnitines were determined in dried blood spots via liquid chromatography-tandem mass spectrometry (LC-MS/MS) on the third day of life of full-term normal (n = 50) and CF (n = 28) newborns. For infants with elevated immunoreactive trypsinogen values, FC/acylcarnitines were quantified again 48 hours later, followed by mutational analysis of CFTR gene via Sanger sequencing. RESULTS: Initial FC and sums of acylcarnitine concentrations were statistically significantly lower in CF patients than in controls and even lower 48 hours later. The mutations F508del and 621 + 1G > T were predominantly identified among CF patients. CONCLUSION: Low FC and acylcarnitine concentrations were measured perinatally in CF patients, for all CFTR mutations detected. Carnitine supplementation of breastfeeding mothers could be beneficial.


Subject(s)
Carnitine/analogs & derivatives , Carnitine/blood , Cystic Fibrosis/blood , Biomarkers , Carnitine/administration & dosage , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , DNA Mutational Analysis , Food, Fortified , Humans , Infant, Newborn , Milk, Human , Mutation , Neonatal Screening
10.
Acupunct Med ; 39(1): 30-40, 2021 02.
Article in English | MEDLINE | ID: mdl-32299241

ABSTRACT

OBJECTIVE: The aim of this study was to examine the effect of acupuncture on obesity-related parameters with regard to metabolomics. METHODS: This two-arm parallel, patient- and assessor-blind, sham-controlled randomized clinical trial included 120 obese participants. Participants were randomized into two groups. The treatment group received a combined intervention of manual acupuncture (MA) plus electroacupuncture (EA), while the control group received a combination of sham acupuncture interventions consisting of sham MA plus sham EA for 6 weeks. The serum lipid levels were measured as the primary outcome measure and anthropometric parameters and serum metabolic profiles including amino acids and carnitines were measured as secondary outcome measures. RESULTS: The serum lipid levels and anthropometric measurements did not significantly differ between groups, while body weight, body mass index (BMI), and waist circumference were improved within each group and the level of high-density lipoprotein cholesterol increased over time in the treatment group. There were no significant differences in amino acid levels between groups, while certain carnitine (C2, C4, C6, and l-carnitine) levels were significantly increased (p < 0.05) in the treatment group compared to the control group. CONCLUSION: In summary, 6 weeks of acupuncture treatment did not affect anthropometric and serum metabolic parameters, with the exception of certain carnitines, when compared to sham acupuncture. The observation of increased carnitine levels in the acupuncture group could result in potential long-term benefits to obese individuals. Additional studies are needed to investigate the long-term effects of acupuncture on lipid metabolism and the underlying mechanism of action. TRIAL REGISTRATION NUMBER: This study is registered at ClinicalTrials.gov (NCT02066090).


Subject(s)
Acupuncture Therapy , Obesity/therapy , Overweight/therapy , Premenopause/blood , Adult , Amino Acids/blood , Anthropometry , Carnitine/blood , Electroacupuncture , Female , Humans , Middle Aged , Obesity/blood , Overweight/blood , Prospective Studies , Treatment Outcome
11.
Ther Apher Dial ; 25(3): 304-313, 2021 Jun.
Article in English | MEDLINE | ID: mdl-32777142

ABSTRACT

Serum carnitine is decreased in hemodialysis patients, which induces muscle atrophy. Thus, we examined the different effects of l-carnitine and exercise on exercise activity and muscle status in hemodialysis patients. Twenty patients were divided into l-carnitine and cycle ergometer groups and were followed for 3 months. Muscle and fat mass, physical activities, and muscle status were evaluated by an impedance, physical function test, and magnetic resonance imaging, respectively. The l-carnitine significantly increased muscle mass (P = .023) and thigh circumference (P = .027), decreased fat mass (P = .007), and shortened chair stand-up time (P = .002) and 10-m walk test (P = .037). The fat fraction was improved by the l-carnitine (P = .047). Compared with the exercise group, l-carnitine improved the changes in 10-m walk test (P = .026), chair stand-up time (P = .014), and thigh circumference (P = .022). Baseline fibroblast growth factor-21 and myostatin levels predicted the l-carnitine-associated changes in exercise activities. l-carnitine, rather than exercise, improved physical activity and muscle status in hemodialysis patients.


Subject(s)
Carnitine/administration & dosage , Dietary Supplements , Exercise Test/methods , Exercise/physiology , Muscles/drug effects , Renal Dialysis , Carnitine/blood , Exercise Test/statistics & numerical data , Female , Humans , Japan , Magnetic Resonance Imaging/methods , Male , Middle Aged , Muscles/diagnostic imaging , Muscles/physiology , Prospective Studies
12.
Nutrients ; 13(1)2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33374696

ABSTRACT

BACKGROUND: This study aimed to identify metabolic parameters at different time points of ketogenic diet therapy (KDT) and investigate their association with response to KDT in pediatric drug-resistant epilepsy (DRE). METHODS: Prospectively, twenty-nine patients (0.67~20 years old) with DRE received classic ketogenic diet with non-fasting, gradual KD initiation protocol (GRAD-KD) for 1 year were enrolled. A total of 22 patients remaining in study received blood examinations at baseline, 3rd, 6th, 9th, and 12th months of KDT. ß-hydroxybutyrate, free carnitine, acylcarnitines, and amino acids were compared between responders (seizure reduction rate ≥ 50%) and non-responders (seizure reduction rate < 50%) to identify the effectiveness of KDT. RESULTS: The 12-month retention rate was 76%. The responders after 12 months of KDT were 59% (13/22). The free carnitine level decreased significantly at 9th months (p < 0.001) but increased toward baseline without symptoms. Propionyl carnitine (C3), Isovaleryl carnitine (C5), 3-Hydroxyisovalerylcarnitine (C5:OH) and methylmalonyl carnitine (C4-DC) decreased but 3-hydroxybutyrylcarnitine (C4:OH) increased significantly at 12th months of KDT. The glycine level was persistently higher than baseline after KDT. KDT responders had lower baseline C3 and long-chain acylcarnitines, C14 and C18, as well as lower C5, C18, and leucine/isoleucine. CONCLUSIONS: KDT should be avoided in patients with non-ketotic hyperglycemia. Routine carnitine supplementation is not recommended because hypocarnitinemia was transient and asymptomatic during KDT. Better mitochondrial ßoxidation function associates with greater KDT response.


Subject(s)
Amino Acids/blood , Carnitine/analogs & derivatives , Diet, Ketogenic , Drug Resistant Epilepsy/diet therapy , 3-Hydroxybutyric Acid/blood , Adolescent , Carnitine/blood , Case-Control Studies , Child , Child, Preschool , Diet, Ketogenic/methods , Drug Resistant Epilepsy/blood , Female , Humans , Infant , Male , Prospective Studies , Tandem Mass Spectrometry , Treatment Outcome , Young Adult
13.
Nutrients ; 12(10)2020 Sep 29.
Article in English | MEDLINE | ID: mdl-33003354

ABSTRACT

Methylmalonic acidemia (MMA) is caused by a deficiency of methyl-malonyl-CoA mutase. It is a multisystemic condition with poor clinical outcomes characterized by frequent metabolic decompensation with acidosis, hyperammonemia and encephalopathy. Restriction of intact protein and supplementation with amino acid-based formula play an important role in its management. Recently, liver transplant (LT) became a treatment option for MMA patients. However, there has been no current consensus on the post-operative nutrition management for MMA patients undergoing transplant, particularly during the initial phase of recovery period with catabolic stressors. We performed a retrospective analysis of clinical and nutritional management as well as biochemical profiles before and after LT in five patients with MMA. Through this study, we observed significant improvement of MMA-associated metabolites after LT. MMA patients were able to tolerate increased intact protein intake post-operatively. At least 1-1.5 g/kg/day of total protein during the acute phase after transplant may be tolerated without worsening of the metabolite levels. This information provides a guide in how to nutritionally manage MMA after LT.


Subject(s)
Amino Acid Metabolism, Inborn Errors/therapy , Diet, Protein-Restricted/methods , Dietary Proteins/administration & dosage , Liver Transplantation , Nutrition Therapy/methods , Adolescent , Adult , Amino Acid Metabolism, Inborn Errors/blood , Carnitine/blood , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Methylmalonic Acid/blood , Postoperative Care , Postoperative Period , Preoperative Period , Retrospective Studies , Young Adult
14.
Mol Genet Metab ; 131(3): 316-324, 2020 11.
Article in English | MEDLINE | ID: mdl-33127324

ABSTRACT

Propionic acidemia (PA) is caused by inherited deficiency of mitochondrial propionyl-CoA carboxylase (PCC) and results in significant neurodevelopmental and cardiac morbidity. However, relationships among therapeutic intervention, biochemical markers, and disease progression are poorly understood. Sixteen individuals homozygous for PCCB c.1606A > G (p.Asn536Asp) variant PA participated in a two-week suspension of therapy. Standard metabolic markers (plasma amino acids, blood spot methylcitrate, plasma/urine acylcarnitines, urine organic acids) were obtained before and after stopping treatment. These same markers were obtained in sixteen unaffected siblings. Echocardiography and electrocardiography were obtained from all subjects. We characterized the baseline biochemical phenotype of untreated PCCB c.1606A > G homozygotes and impact of treatment on PCC deficiency biomarkers. Therapeutic regimens varied widely. Suspension of therapy did not significantly alter branched chain amino acid levels, their alpha-ketoacid derivatives, or urine ketones. Carnitine supplementation significantly increased urine propionylcarnitine and its ratio to total carnitine. Methylcitrate blood spot and urine levels did not correlate with other biochemical measures or cardiac outcomes. Treatment of PCCB c.1606A > G homozygotes with protein restriction, prescription formula, and/or various dietary supplements has a limited effect on core biomarkers of PCC deficiency. These patients require further longitudinal study with standardized approaches to better understand the relationship between biomarkers and disease burden.


Subject(s)
Carbon-Carbon Ligases/genetics , Heart/physiopathology , Neurodevelopmental Disorders/genetics , Propionic Acidemia/genetics , Acids/blood , Acids/urine , Adolescent , Adult , Amino Acids/blood , Amino Acids/urine , Biomarkers/blood , Biomarkers/urine , Carbon-Carbon Ligases/blood , Carbon-Carbon Ligases/urine , Carnitine/blood , Carnitine/urine , Child , Child, Preschool , Echocardiography , Female , Heart/diagnostic imaging , Humans , Male , Mitochondria/genetics , Mitochondria/metabolism , Mutation/genetics , Neurodevelopmental Disorders/blood , Neurodevelopmental Disorders/diagnostic imaging , Neurodevelopmental Disorders/urine , Organic Chemicals/blood , Organic Chemicals/urine , Phenotype , Propionic Acidemia/blood , Propionic Acidemia/diagnostic imaging , Propionic Acidemia/urine , Young Adult
15.
PLoS One ; 15(9): e0239506, 2020.
Article in English | MEDLINE | ID: mdl-32976523

ABSTRACT

BACKGROUND: Low carnitine status may underlie the development of insulin resistance and metabolic inflexibility. Intravenous lipid infusion elevates plasma free fatty acid (FFA) concentration and is a model for simulating insulin resistance and metabolic inflexibility in healthy, insulin sensitive volunteers. Here, we hypothesized that co-infusion of L-carnitine may alleviate lipid-induced insulin resistance and metabolic inflexibility. METHODS: In a randomized crossover trial, eight young healthy volunteers underwent hyperinsulinemic-euglycemic clamps (40mU/m2/min) with simultaneous infusion of saline (CON), Intralipid (20%, 90mL/h) (LIPID), or Intralipid (20%, 90mL/h) combined with L-carnitine infusion (28mg/kg) (LIPID+CAR). Ten volunteers were randomized for the intervention arms (CON, LIPID and LIPID+CAR), but two dropped-out during the study. Therefore, eight volunteers participated in all three intervention arms and were included for analysis. RESULTS: L-carnitine infusion elevated plasma free carnitine availability and resulted in a more pronounced increase in plasma acetylcarnitine, short-, medium-, and long-chain acylcarnitines compared to lipid infusion, however no differences in skeletal muscle free carnitine or acetylcarnitine were found. Peripheral insulin sensitivity and metabolic flexibility were blunted upon lipid infusion compared to CON but L-carnitine infusion did not alleviate this. CONCLUSION: Acute L-carnitine infusion could not alleviated lipid-induced insulin resistance and metabolic inflexibility and did not alter skeletal muscle carnitine availability. Possibly, lipid-induced insulin resistance may also have affected carnitine uptake and may have blunted the insulin-induced carnitine storage in muscle. Future studies are needed to investigate this.


Subject(s)
Carnitine/administration & dosage , Fatty Acids, Nonesterified/blood , Fatty Acids, Nonesterified/metabolism , Insulin Resistance/physiology , Lipids/administration & dosage , Adult , Carnitine/analogs & derivatives , Carnitine/blood , Cross-Over Studies , Emulsions/administration & dosage , Humans , Infusion Pumps , Insulin/blood , Insulin/metabolism , Male , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Phospholipids/administration & dosage , Soybean Oil/administration & dosage , Young Adult
16.
Nutrients ; 12(10)2020 Sep 24.
Article in English | MEDLINE | ID: mdl-32987670

ABSTRACT

l-carnitine (LC) is a co-factor in fatty acid metabolism; its role with respect to aortic stiffness (AS) associated with chronic kidney disease (CKD) was unclear. Our aim was to investigate associations between serum LC levels and AS in patients with non-dialysis CKD stage 3-5. The AS patients were those with carotid-femoral pulse wave velocities (cfPWV) >10 m/s; those with cfPWV ≤10 m/s were included as controls. Serum LC was measured by liquid chromatography and mass spectrometry. Of 136 CKD patients, the 44 (32.4%) with AS were older, exhibited higher rates of diabetes, and had elevated diastolic and systolic blood pressures (SBP), elevated fasting glucose levels and lower levels of serum LC compared to controls. Multivariable logistic regression revealed that serum LC (odds ratio [OR] = 0.949, 95% confidence interval [CI] 0.911-0.988, p = 0.011) and age (OR = 1.055, 95% CI 1.013-1.099, p = 0.009) were independent predictors of AS. Multivariable stepwise linear regression revealed significant positive (age and SBP) and negative (serum LC) correlations with cfPWV. The area under the curve of serum LC as a means to predict AS in CKD patients was 0.657 (95% CI 0.571-0.736, p = 0.0009). We concluded that low serum LC is a significant predictor of AS in patients diagnosed with CKD.


Subject(s)
Carnitine/blood , Renal Insufficiency, Chronic/blood , Renal Insufficiency, Chronic/diagnosis , Vascular Stiffness , Aged , Aged, 80 and over , Blood Glucose/metabolism , Blood Pressure , Blood Urea Nitrogen , Body Height , Body Mass Index , Body Weight , Calcium/blood , Cholesterol/blood , Cross-Sectional Studies , Fasting , Female , Humans , Logistic Models , Male , Middle Aged , Phosphorus/blood , Triglycerides/blood
17.
Food Funct ; 11(9): 7707-7716, 2020 Sep 23.
Article in English | MEDLINE | ID: mdl-32915186

ABSTRACT

Previous studies suggested the potential associations of trimethylamine N-oxide (TMAO) and its metabolic precursor l-carnitine with obesity. However, existing evidence is limited and inconsistent. In the present study, we perform a cross-sectional analysis of the associations of serum levels of TMAO and l-carnitine with obesity measures, including BMI, body fat distribution and body composition in 1081 participants from the general Newfoundland population. The dietary effects of TMAO and l-carnitine in preventing high fat diet-induced obesity in both male and female mice were also evaluated. We found significant associations between higher serum l-carnitine levels and obesity (higher BMI, body fat mass and VT%) in women, but not in men after controlling multiple confounding factors. Serum TMAO levels were positively associated with age, but not obesity in both men and women. Dietary TMAO had no influence on fat accumulation in high fat diet-fed mice. However, l-carnitine supplementation prevented high fat diet-fed induced obesity in both male and female mice by up-regulating lipolysis and down-regulating lipogenesis in white adipose tissues. The present study provides further evidence for the relationships between TMAO, l-carnitine and obesity.


Subject(s)
Carnitine/blood , Methylamines/blood , Obesity/blood , Adipocytes/cytology , Adipose Tissue, White/metabolism , Adult , Animals , Body Composition , Body Weight , Carnitine/administration & dosage , Diet, High-Fat , Female , Gene Expression , Humans , Lipogenesis/genetics , Lipolysis/genetics , Liver/pathology , Male , Methylamines/administration & dosage , Mice , Mice, Inbred C57BL , Middle Aged , Obesity/genetics , Obesity/metabolism , Obesity/pathology , RNA, Messenger/genetics
18.
Nutrients ; 12(8)2020 Jul 22.
Article in English | MEDLINE | ID: mdl-32708036

ABSTRACT

L-carnitine transports fatty acids into the mitochondria for oxidation and also buffers excess acetyl-CoA away from the mitochondria. Thus, L-carnitine may play a key role in maintaining liver function, by its effect on lipid metabolism. The importance of L-carnitine in liver health is supported by the observation that patients with primary carnitine deficiency (PCD) can present with fatty liver disease, which could be due to low levels of intrahepatic and serum levels of L-carnitine. Furthermore, studies suggest that supplementation with L-carnitine may reduce liver fat and the liver enzymes alanine aminotransferase (ALT) and aspartate transaminase (AST) in patients with Non-Alcoholic Fatty Liver Disease (NAFLD). L-carnitine has also been shown to improve insulin sensitivity and elevate pyruvate dehydrogenase (PDH) flux. Studies that show reduced intrahepatic fat and reduced liver enzymes after L-carnitine supplementation suggest that L-carnitine might be a promising supplement to improve or delay the progression of NAFLD.


Subject(s)
Carnitine/blood , Carnitine/pharmacology , Fatty Acids/metabolism , Lipid Metabolism/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/prevention & control , Alanine Transaminase/blood , Aspartate Aminotransferases/blood , Cardiomyopathies/blood , Cardiomyopathies/complications , Cardiomyopathies/drug therapy , Carnitine/deficiency , Dietary Supplements , Humans , Hyperammonemia/blood , Hyperammonemia/complications , Hyperammonemia/drug therapy , Insulin Resistance , Liver/drug effects , Liver/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Muscular Diseases/blood , Muscular Diseases/complications , Muscular Diseases/drug therapy , Non-alcoholic Fatty Liver Disease/complications , Randomized Controlled Trials as Topic , Solute Carrier Family 22 Member 5/metabolism
19.
Ann Nutr Metab ; 76(4): 268-276, 2020.
Article in English | MEDLINE | ID: mdl-32683363

ABSTRACT

BACKGROUND: The detection of methylmalonic acid (MMA) by second-tier analysis has been shown to reduce the number of false positives in newborn screening (NBS) for genetically determined methylmalonic acidurias (MMAuria). In addition to genetic conditions, MMA is an indicator of vitamin B12 status, thus applicable to detect maternal vitamin B12 deficiency in the newborns screened. METHODS: Biochemical and clinical follow-up data of a 7.5-year pilot study with 1.2 million newborns screened were reviewed. RESULTS: Among 1,195,850 NBS samples, 3,595 (0.3%) fulfilled criteria for second-tier analysis of MMA. In 37 (0.003%; 1/32,000) samples, elevated concentrations of MMA were detected, resulting in diagnostic workup at a metabolic center in 21 newborns. In 6 infants (1/199,000), genetic conditions were established, 1 infant with cobalamin C deficiency (CblC) showed only a moderate elevation of MMA. The remaining 15 newborns (1/79,000) displayed significantly lower concentrations of MMA and were evaluated for maternal vitamin B12 deficiency. In 9 mothers, vitamin B12 deficiency was verified, and 6 showed no indication for vitamin B12 deficiency. Treatment with vitamin B12 normalized biochemical parameters in all 15 infants. CONCLUSIONS: Applying a 2-tier strategy measuring MMA in NBS identified genetic conditions of MMAuria. It was possible to separate severe, early-onset phenotypes from maternal vitamin B12 deficiency. However, the detection of CblC deficiency with mildly elevated MMA interferes with impaired vitamin B12 status of unknown relevance and thus burdens possibly healthy newborns. Regarding maternal vitamin B12 deficiency, testing and supplementing mothers-to-be is preferable. This might decrease straining follow-up of newborns and improve quality and overall perception of NBS.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diagnosis , Carnitine/analogs & derivatives , Dried Blood Spot Testing , Methylmalonic Acid/blood , Neonatal Screening/methods , Carnitine/blood , Diagnosis, Differential , Female , Humans , Infant, Newborn , Male , Pilot Projects , Vitamin B 12 Deficiency/diagnosis
20.
Nutr Res ; 78: 72-81, 2020 06.
Article in English | MEDLINE | ID: mdl-32544852

ABSTRACT

Diets including red meat and other animal-sourced foods may increase proteolytic fermentation and microbial-generated trimethylamine (TMA) and, subsequently, trimethylamine-N-oxide (TMAO), a metabolite associated with increased risk of cardiovascular disease and dementia. It was hypothesized that compared to usual dietary intake, a maintenance-energy high-protein diet (HPD) would increase products of proteolytic fermentation, whereas adjunctive prebiotic, probiotic, and synbiotic supplementation may mitigate these effects. An exploratory aim was to determine the association of the relative abundance of the TMA-generating taxon, Emergencia timonensis, with serum and urinary TMAO. At 5 time points (usual dietary intake, HPD diet, HPD + prebiotic, HPD + probiotic, and HPD + synbiotic), urinary (24-hour) and serum metabolites and fecal microbiota profile of healthy older women (n = 20) were measured by liquid chromatography-tandem mass spectrometry and 16S rRNA gene amplicon sequencing analyses, respectively. The HPD induced increases in serum levels of l-carnitine, indoxyl sulfate, and phenylacetylglutamine but not TMAO or p-cresyl sulfate. Urinary excretion of l-carnitine, indoxyl sulfate, phenylacetylglutamine, and TMA increased with the HPD but not TMAO or p-cresyl sulfate. Most participants had undetectable levels of E.timonensis at baseline and only 50% during the HPD interventions, suggesting other taxa are responsible for the microbial generation of TMA in these individuals. An HPD diet with or without a prebiotic, probiotic, or synbiotic elicited an increase in products of proteolytic fermentation. The urinary l-carnitine response suggests that the additional dietary l-carnitine provided was primarily bioavailable, providing little substrate for microbial conversion to TMA and subsequent TMAO formation.


Subject(s)
Diet, High-Protein , Meat , Methylamines/blood , Methylamines/urine , Aged , Carnitine/blood , Carnitine/urine , Clostridiales/isolation & purification , Cresols/blood , Cresols/urine , Cross-Over Studies , Feces/microbiology , Female , Gastrointestinal Microbiome , Glutamine/analogs & derivatives , Glutamine/urine , Humans , Indican/blood , Indican/urine , Prebiotics , Probiotics , Sulfuric Acid Esters/blood , Sulfuric Acid Esters/urine , Synbiotics
SELECTION OF CITATIONS
SEARCH DETAIL