Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
1.
Cardiovasc Pathol ; 70: 107630, 2024.
Article in English | MEDLINE | ID: mdl-38490313

ABSTRACT

A female neonate born with normal Apgar scores at 38+2 weeks of gestational age unexpectedly passed away within less than 30 hours after birth. The situation mirrored her brother's earlier demise within 24 hours post-delivery, suggesting a possible genetic disorder. Gross examination revealed widespread cyanosis and distinct yellowish changes on the cardiac ventricles. Histopathological examination disclosed lipid accumulation in the liver, heart, and kidneys. Tandem mass spectrometry detected elevated levels of 10 amino acids and 14 carnitines in cardiac blood. Trio-whole genome sequencing (Trio-WGS) identified the SLC25A20 c.199-10T>G mutation associated with carnitine-acylcarnitine translocase disease (CACTD), a type of fatty acid oxidation disorders (FAODs) with a potential for sudden death. Further validation of gene expression confirmed the functional deficiency of SLC25A20, ultimately diagnosing CACTD as the underlying cause of the neonate's demise. This case highlights the importance of prenatal metabolic and genetic screening for prospective parents and emphasizes the need for forensic doctors to integrate metabolomic and genomic investigations into autopsies for suspected inherited metabolic diseases.


Subject(s)
Carnitine Acyltransferases , Lipid Metabolism, Inborn Errors , Mutation , Humans , Infant, Newborn , Female , Carnitine Acyltransferases/deficiency , Carnitine Acyltransferases/genetics , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Lipid Metabolism, Inborn Errors/complications , Lipid Metabolism, Inborn Errors/diagnosis , Phenotype , Fatal Outcome , Genetic Predisposition to Disease , Sudden Infant Death/genetics , Sudden Infant Death/pathology , Sudden Infant Death/etiology , Autopsy , Death, Sudden, Cardiac/etiology , Death, Sudden, Cardiac/pathology , Cause of Death , Carnitine/analogs & derivatives , Carnitine/deficiency , Mitochondrial Membrane Transport Proteins/genetics , Myocardium/pathology , Myocardium/metabolism , Membrane Transport Proteins
2.
J Biol Chem ; 299(7): 104908, 2023 07.
Article in English | MEDLINE | ID: mdl-37307919

ABSTRACT

Whereas it is known that p53 broadly regulates cell metabolism, the specific activities that mediate this regulation remain partially understood. Here, we identified carnitine o-octanoyltransferase (CROT) as a p53 transactivation target that is upregulated by cellular stresses in a p53-dependent manner. CROT is a peroxisomal enzyme catalyzing very long-chain fatty acids conversion to medium chain fatty acids that can be absorbed by mitochondria during ß-oxidation. p53 induces CROT transcription through binding to consensus response elements in the 5'-UTR of CROT mRNA. Overexpression of WT but not enzymatically inactive mutant CROT promotes mitochondrial oxidative respiration, while downregulation of CROT inhibits mitochondrial oxidative respiration. Nutrient depletion induces p53-dependent CROT expression that facilitates cell growth and survival; in contrast, cells deficient in CROT have blunted cell growth and reduced survival during nutrient depletion. Together, these data are consistent with a model where p53-regulated CROT expression allows cells to be more efficiently utilizing stored very long-chain fatty acids to survive nutrient depletion stresses.


Subject(s)
Carnitine Acyltransferases , Cell Survival , Nutrients , Tumor Suppressor Protein p53 , 5' Untranslated Regions/genetics , Carnitine/metabolism , Carnitine Acyltransferases/genetics , Carnitine Acyltransferases/metabolism , Cell Growth Processes , Cell Respiration , Fatty Acids/chemistry , Fatty Acids/metabolism , Mitochondria/metabolism , Mutation , Nutrients/deficiency , Nutrients/metabolism , Oxidation-Reduction , Peroxisomes/enzymology , Response Elements/genetics , Stress, Physiological , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism
3.
J Int Med Res ; 51(4): 3000605231163811, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37115522

ABSTRACT

The current case report describes the clinical, biochemical and genetic characteristics of carnitine-acylcarnitine translocase deficiency (CACTD) in infant male and female twins that presented with symptoms shortly after elective caesarean delivery. The clinical manifestations were neonatal hypoglycaemia, arrhythmia and sudden death. The age of onset was 1.5 days and the age of the death was 1.5-3.5 days. Dried blood filter paper analysis was used for the detection of acylcarnitine. Peripheral venous blood and skin samples were used for next-generation sequencing. The twins and their parents underwent gene analysis and whole exome sequencing analyses of the solute carrier family 25 member 20 (SLC25A20; also known as carnitine-acylcarnitine translocase) gene. Both infants carried compound heterozygous variants of the SLC25A20 gene: variant M1:c.706_707insT:p.R236L fs*12 and variant M2:c.689C>G:p.P230R. The M1 variant was paternal and had not been previously reported regarding CACTD. The M2 variant was maternal. CACTD has severe clinical manifestations and a poor prognosis, which is manifested as hypoketotic hypoglycaemia, hyperammonaemia, liver function damage and elevated creatine kinase.


Subject(s)
Hypoglycemia , Lipid Metabolism, Inborn Errors , Female , Humans , Infant, Newborn , Male , Carnitine Acyltransferases/genetics , Carnitine Acyltransferases/metabolism , Hypoglycemia/genetics , Lipid Metabolism, Inborn Errors/genetics , Membrane Transport Proteins/genetics , Mutation , Twins, Dizygotic
4.
Int J Mol Sci ; 24(4)2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36835358

ABSTRACT

The Carnitine-Acylcarnitine Carrier is a member of the mitochondrial Solute Carrier Family 25 (SLC25), known as SLC25A20, involved in the electroneutral exchange of acylcarnitine and carnitine across the inner mitochondrial membrane. It acts as a master regulator of fatty acids ß-oxidation and is known to be involved in neonatal pathologies and cancer. The transport mechanism, also known as "alternating access", involves a conformational transition in which the binding site is accessible from one side of the membrane or the other. In this study, through a combination of state-of-the-art modelling techniques, molecular dynamics, and molecular docking, the structural dynamics of SLC25A20 and the early substrates recognition step have been analyzed. The results obtained demonstrated a significant asymmetry in the conformational changes leading to the transition from the c- to the m-state, confirming previous observations on other homologous transporters. Moreover, analysis of the MD simulations' trajectories of the apo-protein in the two conformational states allowed for a better understanding of the role of SLC25A20 Asp231His and Ala281Val pathogenic mutations, which are at the basis of Carnitine-Acylcarnitine Translocase Deficiency. Finally, molecular docking coupled to molecular dynamics simulations lend support to the multi-step substrates recognition and translocation mechanism already hypothesized for the ADP/ATP carrier.


Subject(s)
Carnitine Acyltransferases , Membrane Transport Proteins , Mitochondrial Membrane Transport Proteins , Humans , Infant, Newborn , Carnitine Acyltransferases/chemistry , Carnitine Acyltransferases/genetics , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/genetics , Molecular Docking Simulation , Computer Simulation
5.
J Invest Dermatol ; 143(2): 305-316.e5, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36058299

ABSTRACT

Circulating tumor cells are the key link between a primary tumor and distant metastases, but once in the bloodstream, loss of adhesion induces cell death. To identify the mechanisms relevant for melanoma circulating tumor cell survival, we performed RNA sequencing and discovered that detached melanoma cells and isolated melanoma circulating tumor cells rewire lipid metabolism by upregulating fatty acid (FA) transport and FA beta-oxidation‒related genes. In patients with melanoma, high expression of FA transporters and FA beta-oxidation enzymes significantly correlates with reduced progression-free and overall survival. Among the highest expressed regulators in melanoma circulating tumor cells were the carnitine transferases carnitine O-octanoyltransferase and carnitine acetyltransferase, which control the shuttle of peroxisome-derived medium-chain FAs toward mitochondria to fuel mitochondrial FA beta-oxidation. Knockdown of carnitine O-octanoyltransferase or carnitine acetyltransferase and short-term treatment with peroxisomal or mitochondrial FA beta-oxidation inhibitors thioridazine or ranolazine suppressed melanoma metastasis in mice. Carnitine O-octanoyltransferase and carnitine acetyltransferase depletion could be rescued by medium-chain FA supplementation, indicating that the peroxisomal supply of FAs is crucial for the survival of nonadherent melanoma cells. Our study identifies targeting the FA-based cross-talk between peroxisomes and mitochondria as a potential therapeutic opportunity to challenge melanoma progression. Moreover, the discovery of the antimetastatic activity of the Food and Drug Administration‒approved drug ranolazine carries translational potential.


Subject(s)
Melanoma , Neoplastic Cells, Circulating , Mice , Animals , Carnitine O-Acetyltransferase/genetics , Carnitine O-Acetyltransferase/metabolism , Carnitine Acyltransferases/genetics , Carnitine Acyltransferases/metabolism , Ranolazine , Oxidation-Reduction , Fatty Acids/metabolism , Melanoma/drug therapy , Carnitine/metabolism
6.
Int J Biol Macromol ; 221: 1453-1465, 2022 Nov 30.
Article in English | MEDLINE | ID: mdl-36122779

ABSTRACT

Mitochondrial carnitine/acylcarnitine carrier (CAC) is a member of the mitochondrial carrier (MC) family and imports acylcarnitine into the mitochondrial matrix in exchange for carnitine, playing a pivotal role in carnitine shuttle, crucial for fatty acid oxidation. The crystallized structure of CAC has not been solved yet, however, the availability of several in vitro/in silico studies, also based on the crystallized structures of the ADP/ATP carrier in the cytosolic-conformation and in the matrix-conformation, has made possible to confirm the hypothesis of the single-binding centered-gated pore mechanism for all the members of the MC family. In addition, our recent bioinformatics analyses allowed quantifying in silico the importance of protein residues of MC substrate binding region, of those involved in the formation of the matrix and cytosolic gates, and of those belonging to the Pro/Gly (PG) levels, proposed to be crucial for the tilting/kinking/bending of the six MC transmembrane helices, funneling the substrate translocation pathway. Here we present a combined in silico/in vitro analysis employed for investigating the role played by a group of 6 proline residues and 6 glycine residues, highly conserved in CAC, belonging to MC PG-levels. Residues of the PG-levels surround the similarly located MC common substrate binding region, and were proposed to lead conformational changes and substrate translocation, following substrate binding. For our analysis, we employed 3D molecular modeling approaches, alanine scanning site-directed mutagenesis and in vitro transport assays. Our analysis reveals that P130 (H3), G268 (H6) and G220 (H5), mutated in alanine, affect severely CAC transport activity (mutant catalytic efficiency lower than 5 % compared to the wild type CAC), most likely due to their major role in triggering CAC conformational changes, following carnitine binding. Notably, P30A (H1) and G121A (H3) CAC mutants, increase the carnitine uptake up to 217 % and 112 %, respectively, compared to the wild type CAC.


Subject(s)
Carnitine Acyltransferases , Proline , Carnitine Acyltransferases/genetics , Carnitine Acyltransferases/chemistry , Carnitine Acyltransferases/metabolism , Glycine , Carnitine , Alanine
7.
Taiwan J Obstet Gynecol ; 61(1): 153-156, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35181030

ABSTRACT

OBJECTIVE: We investigated a strategy of exome sequencing DNA from the unaffected parents and applied a set of filtering criteria to identify genes where both partners are heterozygous for a potentially pathogenic variant. CASE REPORT: We report a non-consanguineous couple who had three daughters, all spontaneous preterm birth at 36 weeks gestation and died in the first period after birth, suspected inborn errors of metabolism. Two days after birth, the first daughter presented with difficulty breathing, cyanosis and died; the second died at 33 days old; the third daughter was isolated under special care and was taken to the mother's room, developed the same symptoms and died after 5 days. Dried blood spot testing screen of 55 congenital metabolic disorders was negative. CONCLUSION: Heterogenous variant in SLC25A20 gene was found in both parents, contributing to the delineations of the neonatal phenotypes related to SLC25A20 mutation in CACTD.


Subject(s)
Carnitine Acyltransferases/deficiency , Lipid Metabolism, Inborn Errors/genetics , Membrane Transport Proteins/genetics , Premature Birth , Carnitine Acyltransferases/genetics , Female , Humans , Infant, Newborn , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/mortality , Membrane Transport Proteins/deficiency , Mutation , Pregnancy , Pregnancy Trimester, Third , Exome Sequencing
8.
Leg Med (Tokyo) ; 54: 101990, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34784499

ABSTRACT

Carnitine-acylcarnitine translocase deficiency (CACTD) is a rare and life-threatening autosomal recessive disorder of fatty acid ß-oxidation (FAO). Most patients with CACTD develop severe metabolic decompensation which deteriorates progressively and rapidly, causing death in infancy or childhood. As CACTD in some patients is asymptomatic or only with some nonspecific symptoms, the diagnosis is easy to be ignored, resulting in sudden death, which often triggers medical disputes. Herein, we report a case of neonatal sudden death with CACTD. The neonate showed a series of severe metabolic crisis, deteriorated rapidly and eventually died 3 days after delivery. Tandem mass spectrometry (MS-MS) screening of dry blood spots before death showed that the level of long-chain acylcarnitines, especially C12-C18 acylcarnitine, was increased significantly, and therefore a diagnosis of inherited metabolic disease (IMD) was suspected. Autopsy and histopathological results demonstrated that there were diffuse vacuoles in the heart and liver of the deceased. Mutation analysis revealed that the patient was a compound heterozygote with c.199-10 T > G and a novel c.1A > T mutation in the SLC25A20 gene. Pathological changes such as heart failure, arrhythmia and cardiac arrest related to mitochondrial FAO disorders are the direct cause of death, while gene mutation is the underlying cause of death.


Subject(s)
Carnitine Acyltransferases , Membrane Transport Proteins , Carnitine , Carnitine Acyltransferases/genetics , Death, Sudden/etiology , Heterozygote , Humans , Infant, Newborn , Membrane Transport Proteins/genetics , Mutation
9.
Clin Biochem ; 98: 48-53, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34626609

ABSTRACT

OBJECTIVE: Carnitine-acylcarnitine Translocase (CACT) deficiency (OMIM 212138) and carnitine palmitoyl transferase 2 (CPT2) deficiency (OMIM 60065050) are rare inherited disorders of mitochondrial long chain fatty acid oxidation. The aim of our study is to review the clinical, biochemical and molecular characteristics in children diagnosed with CACT and CPT2 deficiencies in Malaysia. DESIGN AND METHODS: This is a retrospective study. We reviewed medical records of six patients diagnosed with CACT and CPT2 deficiencies. They were identified from a selective high-risk screening of 50,579 patients from January 2010 until Jun 2020. RESULTS: All six patients had either elevation of the long chain acylcarnitines and/or an elevated (C16 + C18:1)/C2 acylcarnitine ratio. SLC25A20 gene sequencing of patient 1 and 6 showed a homozygous splice site mutation at c.199-10 T > G in intron 2. Two novel mutations at c.109C > T p. (Arg37*) in exon 2 and at c.706C > T p. (Arg236*) in exon 7 of SLC25A20 gene were found in patient 2. Patient 3 and 4 (siblings) exhibited a compound heterozygous mutation at c.638A > G p. (Asp213Gly) and novel mutation c.1073 T > G p. (Leu358Arg) in exon 4 of CPT2 gene. A significant combined prevalence at 0.01% of CACT and CPT2 deficiencies was found in the symptomatic Malaysian patients. CONCLUSIONS: The use of the (C16 + C18:1)/C2 acylcarnitine ratio in dried blood spot in our experience improves the diagnostic specificity for CACT/CPT2 deficiencies over long chain acylcarnitine (C16 and C18:1) alone. DNA sequencing for both genes aids in confirming the diagnosis.


Subject(s)
Carnitine Acyltransferases/deficiency , Carnitine O-Palmitoyltransferase/deficiency , Carnitine O-Palmitoyltransferase/genetics , Exons , Introns , Lipid Metabolism, Inborn Errors/genetics , Membrane Transport Proteins/genetics , Metabolism, Inborn Errors/genetics , Mutation , RNA Splice Sites , Carnitine Acyltransferases/blood , Carnitine Acyltransferases/genetics , Carnitine O-Palmitoyltransferase/blood , Child , Female , Humans , Lipid Metabolism, Inborn Errors/blood , Malaysia , Male , Metabolism, Inborn Errors/blood , Retrospective Studies
10.
Turk J Pediatr ; 63(4): 691-696, 2021.
Article in English | MEDLINE | ID: mdl-34449152

ABSTRACT

BACKGROUND: Carnitine-acylcarnitine translocase deficiency (CACTD) is a rare, autosomal recessive, and highly lethal fatty acid oxidation (FAO) disorder caused by defective acylcarnitine transport across the mitochondrial membrane. CACTD is characterized by severe episodes of hypoglycemia and hyperammonemia, seizures, cardiomyopathy, liver dysfunction, severe neurological damage, and muscle weakness. Herein, we described the clinical features, biochemical, and molecular findings of three patients with CACTD, presented with poor feeding, hypoglycemia, liver dysfunctions, and hyperammonemia, but died despite intensive treatment. CASES: All cases had similar signs and symptoms like poor feeding and respiratory failure associated with liver dysfunction. Urinary organic acid profiles in the presence of hypoglycemia and hyperammonemia led us to the possible diagnosis of one of fatty acid ß-oxidation defects. Results of the molecular analyses were compatible with CACTD. In addition to known mutation (c.270delC;p.Phe91Leufs*38) we detected a novel one (c.408C > A;p.Cys136*). CONCLUSIONS: All three cases died despite a very intensive therapy. Based on our experience with these three cases, it can be said that CACTD has a relatively poor prognosis, molecular studies are of most importance in suspected cases for the final diagnosis and such studies might be of help while giving genetic counselling and guidance to parents for future pregnancies.


Subject(s)
Lipid Metabolism, Inborn Errors , Muscular Diseases , Carnitine , Carnitine Acyltransferases/genetics , Female , Humans , Membrane Transport Proteins , Mutation , Pregnancy
11.
Arterioscler Thromb Vasc Biol ; 41(2): 755-768, 2021 02.
Article in English | MEDLINE | ID: mdl-33356393

ABSTRACT

OBJECTIVE: Vascular calcification is a critical pathology associated with increased cardiovascular event risk, but there are no Food and Drug Administration-approved anticalcific therapies. We hypothesized and validated that an unbiased screening approach would identify novel mediators of human vascular calcification. Approach and Results: We performed an unbiased quantitative proteomics and pathway network analysis that identified increased CROT (carnitine O-octanoyltransferase) in calcifying primary human coronary artery smooth muscle cells (SMCs). Additionally, human carotid artery atherosclerotic plaques contained increased immunoreactive CROT near calcified regions. CROT siRNA reduced fibrocalcific response in calcifying SMCs. In agreement, histidine 327 to alanine point mutation inactivated human CROT fatty acid metabolism enzymatic activity and suppressed SMC calcification. CROT siRNA suppressed type 1 collagen secretion, and restored mitochondrial proteome alterations, and suppressed mitochondrial fragmentation in calcifying SMCs. Lipidomics analysis of SMCs incubated with CROT siRNA revealed increased eicosapentaenoic acid, a vascular calcification inhibitor. CRISPR/Cas9-mediated Crot deficiency in LDL (low-density lipoprotein) receptor-deficient mice reduced aortic and carotid artery calcification without altering bone density or liver and plasma cholesterol and triglyceride concentrations. CONCLUSIONS: CROT is a novel contributing factor in vascular calcification via promoting fatty acid metabolism and mitochondrial dysfunction, as such CROT inhibition has strong potential as an antifibrocalcific therapy.


Subject(s)
Atherosclerosis/enzymology , Carnitine Acyltransferases/metabolism , Energy Metabolism , Fatty Acids/metabolism , Mitochondria/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Vascular Calcification/enzymology , Adult , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Carnitine Acyltransferases/genetics , Cells, Cultured , Disease Models, Animal , Female , Fibrosis , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mitochondria/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Osteogenesis , Proteome , Proteomics , Receptors, LDL/genetics , Receptors, LDL/metabolism , Signal Transduction , Vascular Calcification/genetics , Vascular Calcification/pathology , Vascular Calcification/prevention & control
12.
Molecules ; 25(4)2020 Feb 13.
Article in English | MEDLINE | ID: mdl-32070004

ABSTRACT

The effect of copper on the mitochondrial carnitine/acylcarnitine carrier (CAC) was studied. Transport function was assayed as [3H]carnitine/carnitine antiport in proteoliposomes reconstituted with the native protein extracted from rat liver mitochondria or with the recombinant CAC over-expressed in E. coli. Cu2+ (as well as Cu+) strongly inhibited the native transporter. The inhibition was reversed by GSH (reduced glutathione) or by DTE (dithioerythritol). Dose-response analysis of the inhibition of the native protein was performed from which an IC50 of 1.6 µM for Cu2+ was derived. The mechanism of inhibition was studied by using the recombinant WT or Cys site-directed mutants of CAC. From the dose-response curve of the effect of Cu2+ on the recombinant protein, an IC50 of 0.28 µM was derived. Inhibition kinetics revealed a non-competitive type of inhibition by Cu2+. However, a substrate protection experiment indicated that the interaction of Cu2+ with the protein occurred in the vicinity of the substrate-binding site. Dose-response analysis on Cys mutants led to much higher IC50 values for the mutants C136S or C155S. The highest value was obtained for the C136/155S double mutant, indicating the involvement of both Cys residues in the interaction with Cu2+. Computational analysis performed on the WT CAC and on Cys mutants showed a pattern of the binding energy mostly overlapping the binding affinity derived from the dose-response analysis. All the data concur with bridging of Cu2+ with the two Cys residues, which blocks the conformational changes required for transport cycle.


Subject(s)
Carnitine Acyltransferases/metabolism , Copper/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Animals , Carnitine Acyltransferases/genetics , Computational Chemistry , Kinetics , Mutagenesis, Site-Directed , Mutation/genetics , Rats , Zebrafish
13.
Molecules ; 24(18)2019 Sep 06.
Article in English | MEDLINE | ID: mdl-31500110

ABSTRACT

Carnitine plays essential roles in intermediary metabolism. In non-vegetarians, most of carnitine sources (~75%) are obtained from diet whereas endogenous synthesis accounts for around 25%. Renal carnitine reabsorption along with dietary intake and endogenous production maintain carnitine homeostasis. The precursors for carnitine biosynthesis are lysine and methionine. The biosynthetic pathway involves four enzymes: 6-N-trimethyllysine dioxygenase (TMLD), 3-hydroxy-6-N-trimethyllysine aldolase (HTMLA), 4-N-trimethylaminobutyraldehyde dehydrogenase (TMABADH), and γ-butyrobetaine dioxygenase (BBD). OCTN2 (organic cation/carnitine transporter novel type 2) transports carnitine into the cells. One of the major functions of carnitine is shuttling long-chain fatty acids across the mitochondrial membrane from the cytosol into the mitochondrial matrix for ß-oxidation. This transport is achieved by mitochondrial carnitine-acylcarnitine cycle, which consists of three enzymes: carnitine palmitoyltransferase I (CPT I), carnitine-acylcarnitine translocase (CACT), and carnitine palmitoyltransferase II (CPT II). Carnitine inborn errors of metabolism could result from defects in carnitine biosynthesis, carnitine transport, or mitochondrial carnitine-acylcarnitine cycle. The presentation of these disorders is variable but common findings include hypoketotic hypoglycemia, cardio(myopathy), and liver disease. In this review, the metabolism and homeostasis of carnitine are discussed. Then we present details of different inborn errors of carnitine metabolism, including clinical presentation, diagnosis, and treatment options. At the end, we discuss some of the causes of secondary carnitine deficiency.


Subject(s)
Cardiomyopathies/genetics , Carnitine/deficiency , Carnitine/genetics , Hyperammonemia/genetics , Metabolism, Inborn Errors/genetics , Mitochondria/enzymology , Muscular Diseases/genetics , Aldehyde Oxidoreductases/genetics , Cardiomyopathies/metabolism , Carnitine/biosynthesis , Carnitine/metabolism , Carnitine Acyltransferases/genetics , Carnitine O-Palmitoyltransferase/genetics , Humans , Hyperammonemia/metabolism , Mitochondria/genetics , Mixed Function Oxygenases/genetics , Muscular Diseases/metabolism , Oxidation-Reduction , Solute Carrier Family 22 Member 5/genetics , gamma-Butyrobetaine Dioxygenase/genetics
14.
Cells ; 8(7)2019 07 10.
Article in English | MEDLINE | ID: mdl-31295943

ABSTRACT

Aberrant activation of signaling pathways is frequently observed and reported to be associated with the progression and poor prognosis of prostate cancer (PCa). We aimed to identify key biological processes regulated by androgen receptor (AR) using gene co-expression network from single cell resolution. The bimodal index was used to evaluate whether two subpopulations exist among the single cells. Gene expression among single cells revealed averaging pitfalls and bimodality pattern. Weighted gene co-expression network analysis (WGCNA) was used to identify modules of highly correlated genes. Twenty-nine gene modules were identified and AR-regulated modules were screened by significantly overlapping reported androgen induced differentially expressed genes. The biological function "generation of precursor metabolites and energy" was significantly enriched by AR-regulated modules with bimodality, presenting differential androgen response among subpopulations. Integrating with public ChIP-seq data, two genes FECH, and CROT has AR binding sites. Public in vitro studies also show that androgen regulates FECH and CROT. After receiving androgen deprivation therapy, patients lowly express FECH and CROT. Further survival analysis indicates that FECH/CROT signature can predict PCa recurrence. We reveal the heterogeneous function of "generation of precursor metabolites and energy" upon androgen stimulation from the perspective of single cells. Inhibitors targeting this biological process will facilitate to prevent prostate cancer progression.


Subject(s)
Carnitine Acyltransferases/genetics , Ferrochelatase/genetics , Prostatic Neoplasms/genetics , Androgen Antagonists , Androgens , Carnitine Acyltransferases/metabolism , Cell Line, Tumor , Databases, Genetic , Ferrochelatase/metabolism , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks/genetics , Humans , Male , Neoplasm Recurrence, Local/genetics , Prognosis , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Signal Transduction/genetics , Single-Cell Analysis/methods , Transcriptome/genetics
15.
Biomed Pharmacother ; 118: 109130, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31306969

ABSTRACT

OBJECTIVES: The objective of this work was to determine the specific mechanisms by which resveratrol inhibits lipogenesis and stimulates lipolysis. METHODS: Twelve male mice were individually introduced into a metabolic cage for 24 h to measure basal metabolic rate, prior to intervention. They were randomly divided into two groups, resveratrol (RSV) and control (C), and administered resveratrol intraperitoneally or vehicle, respectively, for two consecutive days. After 24 h, the metabolic energy expenditure was again determined for 24 h, before mice were sacrificed. Protein and gene expression of different enzymes related to metabolism in the hepatic tissue, adipose tissue and gastrocnemius of mice were analyzed by RT-PCR, western blot or ELISA. RESULTS: We report that resveratrol lowers the respiratory quotient in old mice and that this may be due to the activation of fatty acid mobilization from white adipose tissue (because hormone-activated lipase expression is increased) and fatty acid transport into mitochondria and eventual oxidation in muscle and liver (because transport enzymes and beta oxidation enzymes are also increased). Indeed, we have observed that resveratrol in vivo causes an increase in the expression and phosphorylation of AMPKα in liver, muscle and adipose tissue and an increase in the expression of acyl-CoA synthetase, of carnitine palmitoyl transferase 1 and of carnitine acylcarnitine translocase, all enzymes involved in lipid catabolism. On the other hand, the levels of acetyl-CoA carboxylase as well as those its product, i.e. malonyl CoA, are decreased. CONCLUSIONS: We conclude that a controlled dose of resveratrol activates fatty acid mobilization and degradation and inhibits fatty acid synthesis in old mice. This is the first time that these effects of resveratrol in lipid metabolism in healthy old (non-obese) animals are reported.


Subject(s)
Aging/metabolism , Energy Metabolism/drug effects , Fatty Acids/metabolism , Lipogenesis/drug effects , Lipolysis/drug effects , Resveratrol/pharmacology , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Aging/genetics , Animals , Carnitine Acyltransferases/genetics , Carnitine Acyltransferases/metabolism , Energy Metabolism/genetics , Lipogenesis/genetics , Lipolysis/genetics , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction
16.
Biochim Biophys Acta Bioenerg ; 1860(9): 708-716, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31340138

ABSTRACT

The mitochondrial carnitine/acylcarnitine carrier (CACT) catalyzes an antiport of carnitine and acylcarnitines and also a uniport reaction with a rate of about one tenth with respect to the antiport rate. The antiport process results from the coupling of the two uniport reactions in opposite directions. In this mechanism, the transition of the carrier from the outward open conformation to the inward open one (or vice versa) is much faster for the carrier-substrate complex than for the unbound carrier. To investigate the molecular determinants that couple the binding of the substrate with the conformational transitions, site directed mutagenesis has been employed. The antiport or the uniport reaction was followed as [3H]carnitine uptake in or efflux from proteoliposomes reconstituted with the WT or Trp mutants of the rat CACT. Substitution of each the three Trp residues led to different results. Nearly no variations were observed upon substitution of W192 and/or W296 with Ala. While, substantial alteration of the transport function was observed in the mutants W224A, W224Y and W224F. Mutation of W224 led to the loss of the antiport function while the uniport function was unaltered. In these mutants impairment of the substrate affinity on the external side was also observed. The data highlights that W224 is involved in the coupling of the substrate binding with the matrix gate opening. The experimental data are in line with predictions by homology modeling of the CACT in its cytosolic (c-state) or matrix (m-state) opened conformations.


Subject(s)
Antiporters/metabolism , Carnitine Acyltransferases/metabolism , Carnitine/analogs & derivatives , Carnitine/metabolism , Tryptophan/metabolism , Amino Acid Sequence , Animals , Aspergillus nidulans , Biological Transport , Carnitine Acyltransferases/chemistry , Carnitine Acyltransferases/genetics , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Protein Conformation , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology , Tryptophan/chemistry , Tryptophan/genetics
17.
Chem Biol Interact ; 307: 179-185, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31063765

ABSTRACT

The effect of polyphenols, recognized as the principal antioxidant and beneficial molecules introduced with the diet, extracted from sweet cherry (Prunus avium L.) on the recombinant human mitochondrial carnitine/acylcarnitine transporter (CACT) has been studied in proteoliposomes. CACT transport activity, which was strongly impaired after oxidation by atmospheric O2 or H2O2, due to the formation of a disulfide bridge between cysteines 136 and 155, was restored by externally added polyphenols. CACT reduction by polyphenols was time dependent. Spectroscopic analysis of polyphenolic extracts revealed eight most represented compounds in four cultivars. Molecular docking of CACT structural omology model with the most either abundant and arguably bio-available phenolic compound (trans 3-O-feruloyl-quinic acid) of the mix, is in agreement with the experimental data since it results located in the active site close to cysteine 136 at the bottom of the translocation aqueous cavity.


Subject(s)
Carnitine Acyltransferases/metabolism , Mitochondria/metabolism , Polyphenols/metabolism , Prunus avium/chemistry , Binding Sites , Carnitine Acyltransferases/chemistry , Carnitine Acyltransferases/genetics , Humans , Hydrogen Peroxide/chemistry , Molecular Docking Simulation , Mutagenesis, Site-Directed , Oxidation-Reduction , Polyphenols/analysis , Protein Structure, Tertiary , Prunus avium/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Spectrometry, Mass, Electrospray Ionization
18.
Clin Chim Acta ; 495: 476-480, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31108048

ABSTRACT

Carnitine-acylcarnitine translocase deficiency (CACTD) is a rare autosomal recessive disorder of mitochondrial fatty acid oxidation that occurs due to mutations in the SLC25A20 gene. Severe CACTD results in neonatal or infantile sudden death. Herein, we reported six patients with CACTD diagnosed based on biochemical and molecular findings from 5 unrelated families in Guangdong from 2016 to 2017. Among them, five patients presented with hypotonia, nonketotic hypoglycemia, and arrhythmia 2 days after birth, while the other patient presented with respiratory distress, hypotonia, and arrhythmia. Five of the patients died in the neonatal period. Blood acylcarnitine concentrations determination from dried blood spots (DBS) were measured by tandem mass spectrometry (MS/MS). The SLC25A20 and CPT2 gene sequences were analyzed by direct Sanger sequencing. SLC25A20 gene analysis revealed a c.199-10T>G (IVS2-10T>G) homozygous variants in four unrelated patients and a novel mutation c.199-10T>G/c.719-8_c.719-1dupCCCACAG compound heterozygous variants in twins. This report describes the clinical characteristics, biochemical findings and molecular analysis of SLC25A20 gene of patients with CACTD in Guangdong. And our results show that the c.199-10T>G is likely the most common variant of CACTD in Guangdong population as it accounts for 83% (10/12) of the observed mutant alleles. Individuals with the c.199-10T>G genotype had a severe CACTD phenotype.


Subject(s)
Carnitine Acyltransferases/deficiency , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/genetics , Anion Transport Proteins/genetics , Carnitine Acyltransferases/genetics , China , Female , Genotype , Humans , Infant, Newborn , Male , Mitochondrial Proteins/genetics , Neonatal Screening , Organic Anion Transporters , Pedigree , Sequence Analysis, DNA
19.
Plant Sci ; 274: 432-440, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30080631

ABSTRACT

L-carnitine is present in all living kingdoms where it acts in diverse physiological processes. It is involved in lipid metabolism in animals and yeasts, notably as an essential cofactor of fatty acid intracellular trafficking. Its physiological significance is poorly understood in plants, but L-carnitine may be linked to fatty acid metabolism among other roles. Indeed, carnitine transferases activities and acylcarnitines are measured in plant tissues. Current knowledge of fatty acid trafficking in plants rules out acylcarnitines as intermediates of the peroxisomal and mitochondrial fatty acid metabolism, unlike in animals and yeasts. Instead, acylcarnitines could be involved in plastidial exportation of de novo fatty acid, or importation of fatty acids into the ER, for synthesis of specific glycerolipids. L-carnitine also contributes to cellular maintenance though antioxidant and osmolyte properties in animals and microbes. Recent data indicate similar features in plants, together with modulation of signaling pathways. The biosynthesis of L-carnitine in the plant cell shares similar precursors as in the animal and yeast cells. The elucidation of the biosynthesis pathway of L-carnitine, and the identification of the enzymes involved, is today essential to progress further in the comprehension of its biological significance in plants.


Subject(s)
Carnitine Acyltransferases/metabolism , Carnitine/analogs & derivatives , Carnitine/physiology , Fatty Acids/physiology , Plants/metabolism , Animals , Carnitine Acyltransferases/genetics , Lipid Metabolism , Mitochondria/metabolism
20.
Zhonghua Er Ke Za Zhi ; 56(7): 545-549, 2018 Jul 02.
Article in Chinese | MEDLINE | ID: mdl-29996190

ABSTRACT

Objective: To investigate the clinical, biochemical and genetic features of four carnitine-acylcarnitine translocase deficiency cases. Methods: Four cases diagnosed with carnitine-acylcarnitine translocase deficiency from Guangxi Maternal and Child Health Hospital were studied. DNA was extracted from dry blood filter for gene analysis. SLC25A20 gene analysis was performed in 1 case and the whole exon sequence analysis was performed in 3 cases. Results: Retrospective study on unrelated carnitine-acylcarnitine translocase deficiency patients, the age of onset was 1-28 d, the age of death were 1.5-30 d, main clinical features were hypoglycemia (4 cases), arrhythmia (2 cases), sudden death (2 cases). Biochemical test showed hypoglycemia (1.2-2.0 mmol/L) , elevated creatine kinase (955-8 361 U/L) and creatine kinase isozyme(199-360 U/L), normal or decreased free carnitine level (3.70-27.07 µmol/L) , elevated long-chain acylcarnitine (palmityl carnitine 1.85-14.84 µmol/L). The gene tests showed that all 4 cases carried SLC25A20 gene c.199-10T> G homozygous mutation, inherited from their parents. By analyzing the haplotype, we found that the mutation loci of C. 199-10T> G were all in the same haplotype. Conclusion: The c.199-10T> G mutation is an important molecular cause of carnitine-acylcarnitine translocase deficiency, which has relatively high frequency in Guangxi population, and is related to the founder effect.


Subject(s)
Carnitine Acyltransferases/deficiency , Lipid Metabolism, Inborn Errors , Membrane Transport Proteins , Mutation , Carnitine , Carnitine Acyltransferases/genetics , China , Founder Effect , Humans , Infant , Infant, Newborn , Lipid Metabolism, Inborn Errors/complications , Lipid Metabolism, Inborn Errors/genetics , Membrane Transport Proteins/genetics , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...