Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
BMC Cardiovasc Disord ; 24(1): 1, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38166572

ABSTRACT

BACKGROUND: Primary carnitine deficiency (PCD) denotes low carnitine levels with an autosomal recessive pattern of inheritance. Cardiomyopathy is the most common cardiac symptom in patients with PCD, and early diagnosis can prevent complications. Next-generation sequencing can identify genetic variants attributable to PCD efficiently. OBJECTIVE: We aimed to detect the genetic cause of the early manifestations of hypertrophic cardiomyopathy and metabolic abnormalities in an Iranian family. METHODS: We herein describe an 8-year-old boy with symptoms of weakness and lethargy diagnosed with PCD through clinical evaluations, lab tests, echocardiography, and cardiac magnetic resonance imaging. The candidate variant was confirmed through whole-exome sequencing, polymerase chain reaction, and direct Sanger sequencing. The binding efficacy of normal and mutant protein-ligand complexes were evaluated via structural modeling and docking studies. RESULTS: Clinical evaluations, echocardiography, and cardiac magnetic resonance imaging findings revealed hypertrophic cardiomyopathy as a clinical presentation of PCD. Whole-exome sequencing identified a new homozygous variant, SLC22A5 (NM_003060.4), c.821G > A: p.Trp274Ter, associated with carnitine transport. Docking analysis highlighted the impact of the variant on carnitine transport, further indicating its potential role in PCD development. CONCLUSIONS: The c.821G > A: p.Trp274Ter variant in SLC22A5 potentially acted as a pathogenic factor by reducing the binding affinity of organic carnitine transporter type 2 proteins for carnitine. So, the c.821G > A variant may be associated with carnitine deficiency, metabolic abnormalities, and cardiomyopathic characteristics.


Subject(s)
Cardiomyopathies , Cardiomyopathy, Hypertrophic , Hyperammonemia , Muscular Diseases , Male , Humans , Child , Muscular Diseases/diagnosis , Muscular Diseases/genetics , Carnitine/genetics , Carnitine/metabolism , Iran , Solute Carrier Family 22 Member 5/genetics , Hyperammonemia/diagnosis , Hyperammonemia/genetics , Hyperammonemia/complications , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/genetics , Cardiomyopathy, Hypertrophic/complications , Mutation
2.
Ann Noninvasive Electrocardiol ; 28(6): e13077, 2023 11.
Article in English | MEDLINE | ID: mdl-37658577

ABSTRACT

We report the case of a 13-year-old female patient presenting with presyncope and palpitations. Her electrocardiogram revealed an abbreviation of the rate-corrected QT interval with imaging showing significant left ventricular dysfunction. Carnitine levels were measured as part of her diagnostic workup, discovering a rare, reversible cause of short QT syndrome (SQTS) and associated cardiomyopathy-primary carnitine deficiency (PCD) caused by a homozygous mutation in the SLC22A5 gene, leading to an in-frame deletion mutation (NP_003051.1:p.Phe23del) affecting the organic cation transporter 2 (OCTN2) protein. Following the treatment with oral carnitine supplementation, her QT interval returned to within the normal range with significant improvement in left ventricular function.


Subject(s)
Arrhythmias, Cardiac , Cardiomyopathies , Carnitine/deficiency , Hyperammonemia , Muscular Diseases , Organic Cation Transport Proteins , Female , Humans , Adolescent , Organic Cation Transport Proteins/genetics , Solute Carrier Family 22 Member 5/genetics , Electrocardiography , Cardiomyopathies/complications , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/genetics , Mutation , Carnitine/therapeutic use , Carnitine/genetics , Syndrome
3.
Mol Autism ; 14(1): 29, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37553674

ABSTRACT

Deletion of exon 2 of the trimethyllysine hydroxylase epsilon (TMLHE) gene was identified in probands with autism spectrum disorder (ASD). TMLHE encodes the first enzyme in carnitine biosynthesis, N6-trimethyllysine dioxygenase (TMLD). Researchers have suggested that carnitine depletion could be important for the development of ASD and cognitive, locomotor and social dysfunctions, but previous findings have been inconclusive regarding the specific role of endogenous carnitine. We developed a mouse knockout model with constitutive TMLD enzyme inactivation that exhibited a significant decrease in the carnitine by more than 90% compared to wild-type (WT) mice. However, we did not observe any significant social, cognitive, or repetitive-behavior changes associated with ASD in the knockout mice; muscle strength and coordination were also not affected. In addition, the life expectancy of knockout mice was similar to that of WT mice. In conclusion, knockout of Tmlh in mice does not induce an ASD phenotype or motor dysfunction despite extremely low carnitine and gamma-butyrobetaine concentrations. Moreover, inactivation of TMLD does not induce a phenotype similar to previously described primary carnitine deficiency; indeed, our results showed that low levels of carnitine sustained adequate energy production, muscle function and social behavior in mice.


Subject(s)
Autism Spectrum Disorder , Mixed Function Oxygenases , Animals , Mice , Autism Spectrum Disorder/genetics , Carnitine/genetics , Cognition , Mice, Knockout , Phenotype , Mixed Function Oxygenases/genetics
4.
J Med Genet ; 60(12): 1177-1185, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-37487700

ABSTRACT

BACKGROUND: Newborn screening (NBS) programmes identify a wide range of disease phenotypes, which raises the question whether early identification and treatment is beneficial for all. This study aims to answer this question for primary carnitine deficiency (PCD) taking into account that NBS for PCD identifies newborns with PCD and also until then undiagnosed mothers. METHODS: We investigated clinical, genetic (variants in SLC22A5 gene) and functional (carnitine transport activity in fibroblasts) characteristics of all referred individuals through NBS (newborns and mothers) and clinically diagnosed patients with PCD (not through NBS). Disease phenotype in newborns was predicted using data from PCD mothers and cases published in literature with identical SLC22A5 variants. RESULTS: PCD was confirmed in 19/131 referred newborns, 37/82 referred mothers and 5 clinically diagnosed patients. Severe symptoms were observed in all clinically diagnosed patients, 1 newborn and none of the mothers identified by NBS. PCD was classified as severe in all 5 clinically diagnosed patients, 3/19 newborns and 1/37 mothers; as benign in 8/19 newborns and 36/37 mothers and as unknown in 8/19 newborns. Carnitine transport activity completely separated severe phenotype from benign phenotype (median (range): 4.0% (3.5-5.0)] vs 26% (9.5-42.5), respectively). CONCLUSION: The majority of mothers and a significant proportion of newborns with PCD identified through NBS are likely to remain asymptomatic without early treatment. Conversely, a small proportion of newborns with predicted severe PCD could greatly benefit from early treatment. Genetic variants and carnitine transport activity can be used to distinguish between these groups.


Subject(s)
Carnitine , Neonatal Screening , Female , Humans , Infant, Newborn , Retrospective Studies , Solute Carrier Family 22 Member 5/genetics , Mutation , Carnitine/genetics
5.
Proc Natl Acad Sci U S A ; 119(46): e2210247119, 2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36343260

ABSTRACT

Genetic variants in SLC22A5, encoding the membrane carnitine transporter OCTN2, cause the rare metabolic disorder Carnitine Transporter Deficiency (CTD). CTD is potentially lethal but actionable if detected early, with confirmatory diagnosis involving sequencing of SLC22A5. Interpretation of missense variants of uncertain significance (VUSs) is a major challenge. In this study, we sought to characterize the largest set to date (n = 150) of OCTN2 variants identified in diverse ancestral populations, with the goals of furthering our understanding of the mechanisms leading to OCTN2 loss-of-function (LOF) and creating a protein-specific variant effect prediction model for OCTN2 function. Uptake assays with 14C-carnitine revealed that 105 variants (70%) significantly reduced transport of carnitine compared to wild-type OCTN2, and 37 variants (25%) severely reduced function to less than 20%. All ancestral populations harbored LOF variants; 62% of green fluorescent protein (GFP)-tagged variants impaired OCTN2 localization to the plasma membrane of human embryonic kidney (HEK293T) cells, and subcellular localization significantly associated with function, revealing a major LOF mechanism of interest for CTD. With these data, we trained a model to classify variants as functional (>20% function) or LOF (<20% function). Our model outperformed existing state-of-the-art methods as evaluated by multiple performance metrics, with mean area under the receiver operating characteristic curve (AUROC) of 0.895 ± 0.025. In summary, in this study we generated a rich dataset of OCTN2 variant function and localization, revealed important disease-causing mechanisms, and improved upon machine learning-based prediction of OCTN2 variant function to aid in variant interpretation in the diagnosis and treatment of CTD.


Subject(s)
Carnitine , Organic Cation Transport Proteins , Humans , Solute Carrier Family 22 Member 5/genetics , Solute Carrier Family 22 Member 5/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , HEK293 Cells , Carnitine/genetics , Carnitine/metabolism , Genomics
7.
J Biol Chem ; 297(5): 101327, 2021 11.
Article in English | MEDLINE | ID: mdl-34688665

ABSTRACT

The production of trimethylamine (TMA) from quaternary amines such as l-carnitine or γ-butyrobetaine (4-(trimethylammonio)butanoate) by gut microbial enzymes has been linked to heart disease. This has led to interest in enzymes of the gut microbiome that might ameliorate net TMA production, such as members of the MttB superfamily of proteins, which can demethylate TMA (e.g., MttB) or l-carnitine (e.g., MtcB). Here, we show that the human gut acetogen Eubacterium limosum demethylates γ-butyrobetaine and produces MtyB, a previously uncharacterized MttB superfamily member catalyzing the demethylation of γ-butyrobetaine. Proteomic analyses of E. limosum grown on either γ-butyrobetaine or dl-lactate were employed to identify candidate proteins underlying catabolic demethylation of the growth substrate. Three proteins were significantly elevated in abundance in γ-butyrobetaine-grown cells: MtyB, MtqC (a corrinoid-binding protein), and MtqA (a corrinoid:tetrahydrofolate methyltransferase). Together, these proteins act as a γ-butyrobetaine:tetrahydrofolate methyltransferase system, forming a key intermediate of acetogenesis. Recombinant MtyB acts as a γ-butyrobetaine:MtqC methyltransferase but cannot methylate free cobalamin cofactor. MtyB is very similar to MtcB, the carnitine methyltransferase, but neither was detectable in cells grown on carnitine nor was detectable in cells grown with γ-butyrobetaine. Both quaternary amines are substrates for either enzyme, but kinetic analysis revealed that, in comparison to MtcB, MtyB has a lower apparent Km for γ-butyrobetaine and higher apparent Vmax, providing a rationale for MtyB abundance in γ-butyrobetaine-grown cells. As TMA is readily produced from γ-butyrobetaine, organisms with MtyB-like proteins may provide a means to lower levels of TMA and proatherogenic TMA-N-oxide via precursor competition.


Subject(s)
Bacterial Proteins/chemistry , Betaine/analogs & derivatives , Carnitine/chemistry , Eubacterium/enzymology , Methyltransferases/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Betaine/chemistry , Betaine/metabolism , Carnitine/genetics , Carnitine/metabolism , Eubacterium/genetics , Gastrointestinal Microbiome , Humans , Methyltransferases/genetics , Methyltransferases/metabolism , Symbiosis
8.
Crit Rev Biochem Mol Biol ; 56(4): 360-372, 2021 08.
Article in English | MEDLINE | ID: mdl-33823724

ABSTRACT

Electron transfer flavoprotein dehydrogenase, also called ETF-ubiquinone oxidoreductase (ETF-QO), is a protein localized in the inner membrane of mitochondria, playing a central role in the electron-transfer system. Indeed, ETF-QO mediates electron transport from flavoprotein dehydrogenases to the ubiquinone pool. ETF-QO mutations are often associated with riboflavin-responsive multiple acyl-CoA dehydrogenase deficiency (RR-MADD, OMIM#231680), a multisystem genetic disease characterized by various clinical manifestations with different degrees of severity. In this review, we outline the clinical features correlated with ETF-QO deficiency and the benefits obtained from different treatments, such as riboflavin, L-carnitine and/or coenzyme Q10 supplementation, and a diet poor in fat and protein. Moreover, we provide a detailed summary of molecular and bioinformatic investigations, describing the mutations identified in ETFDH gene and highlighting their predicted impact on enzymatic structure and activity. In addition, we report biochemical and functional analysis, performed in HEK293 cells and patient fibroblasts and muscle cells, to show the relationship between the nature of ETFDH mutations, the variable impairment of enzyme function, and the different degrees of RR-MADD severity. Finally, we describe in detail 5 RR-MADD patients carrying different ETFDH mutations and presenting variable degrees of clinical symptom severity.


Subject(s)
Electron-Transferring Flavoproteins , Iron-Sulfur Proteins , Mitochondria , Multiple Acyl Coenzyme A Dehydrogenase Deficiency , Mutation , Oxidoreductases Acting on CH-NH Group Donors , Animals , Carnitine/genetics , Carnitine/metabolism , Electron-Transferring Flavoproteins/genetics , Electron-Transferring Flavoproteins/metabolism , Humans , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Mitochondria/enzymology , Mitochondria/genetics , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/enzymology , Multiple Acyl Coenzyme A Dehydrogenase Deficiency/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Oxidoreductases Acting on CH-NH Group Donors/metabolism , Ubiquinone/analogs & derivatives , Ubiquinone/genetics , Ubiquinone/metabolism
9.
Mol Genet Metab ; 132(4): 227-233, 2021 04.
Article in English | MEDLINE | ID: mdl-33610471

ABSTRACT

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


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/genetics , Metabolic Diseases/drug therapy , Triglycerides/administration & dosage , Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Adolescent , Adult , Carnitine/genetics , Child , Child, Preschool , Female , Humans , Infant , Male , Metabolic Diseases/genetics , Metabolic Diseases/pathology , Oxidation-Reduction/drug effects , Quality of Life , Retrospective Studies , Treatment Outcome , Young Adult
10.
Mol Genet Genomic Med ; 9(2): e1583, 2021 02.
Article in English | MEDLINE | ID: mdl-33560599

ABSTRACT

BACKGROUND: Rare studies focused on the tandem mass spectrometry (MS/MS) findings for the primary carnitine deficiency (PCD) in the neonates in China mainland. In this study, we aim to analyze the gene mutation spectrum of PCD in Fujian Province in China mainland. METHODS: Primary carnitine deficiency (PCD) samples used in this study were selected from 95,453 cases underwent neonatal screening between May 2015 and February 2020. SLC22A5 gene sequencing was performed on the neonates and their parents with C0 level of less than 8.8 µmol/L. RESULTS: Ten patients (male: 7; female: 3) were finally included in this study. Among these patients, nine were neonates, and one was maternal decline of C0 of less than 8.8 µmol/L. The maternal case showed two types of mutations of SLC22A5 including c.760C>T(p.R254*) and c.1400C>G(p.S467C). The other nine neonates showed compound mutations involving nine types in 18 sites, among which two mutations [i.e., c.37G>T(p.E13*) and c.694A>G(p.T232A)] were novel that had never been reported before. Bioinformatic analysis indicated that c.37G>T(p.E13*) was a pathogenic mutation, while the c.694A>G (p.T232A) was considered to be likely pathogenic. CONCLUSION: MS/MS screening on PCD contributed to the early diagnosis and screening. In addition, SLC22A5 gene mutation analysis contributed to the PCD screening.


Subject(s)
Cardiomyopathies/genetics , Carnitine/deficiency , Hyperammonemia/genetics , Muscular Diseases/genetics , Phenotype , Adult , Cardiomyopathies/blood , Cardiomyopathies/diagnosis , Carnitine/analogs & derivatives , Carnitine/blood , Carnitine/genetics , Female , Gene Frequency , Humans , Hyperammonemia/blood , Hyperammonemia/diagnosis , Infant , Male , Muscular Diseases/blood , Muscular Diseases/diagnosis , Mutation , Solute Carrier Family 22 Member 5/genetics
11.
Mol Genet Metab ; 131(1-2): 90-97, 2020.
Article in English | MEDLINE | ID: mdl-32928639

ABSTRACT

BACKGROUND: The plasma acylcarnitine profile is frequently used as a biochemical assessment for follow-up in diagnosed patients with fatty acid oxidation disorders (FAODs). Disease specific acylcarnitine species are elevated during metabolic decompensation but there is clinical and biochemical heterogeneity among patients and limited data on the utility of an acylcarnitine profile for routine clinical monitoring. METHODS: We evaluated plasma acylcarnitine profiles from 30 diagnosed patients with long-chain FAODs (carnitine palmitoyltransferase-2 (CPT2), very long-chain acyl-CoA dehydrogenase (VLCAD), and long-chain 3-hydroxy acyl-CoA dehydrogenase or mitochondrial trifunctional protein (LCHAD/TFP) deficiencies) collected after an overnight fast, after feeding a controlled low-fat diet, and before and after moderate exercise. Our purpose was to describe the variability in this biomarker and how various physiologic states effect the acylcarnitine concentrations in circulation. RESULTS: Disease specific acylcarnitine species were higher after an overnight fast and decreased by approximately 60% two hours after a controlled breakfast meal. Moderate-intensity exercise increased the acylcarnitine species but it varied by diagnosis. When analyzed for a genotype/phenotype correlation, the presence of the common LCHADD mutation (c.1528G > C) was associated with higher levels of 3-hydroxyacylcarnitines than in patients with other mutations. CONCLUSIONS: We found that feeding consistently suppressed and that moderate intensity exercise increased disease specific acylcarnitine species, but the response to exercise was highly variable across subjects and diagnoses. The clinical utility of routine plasma acylcarnitine analysis for outpatient treatment monitoring remains questionable; however, if acylcarnitine profiles are measured in the clinical setting, standardized procedures are required for sample collection to be of value.


Subject(s)
Cardiomyopathies/blood , Carnitine O-Palmitoyltransferase/deficiency , Carnitine/analogs & derivatives , Congenital Bone Marrow Failure Syndromes/blood , Lipid Metabolism, Inborn Errors/blood , Metabolism, Inborn Errors/blood , Mitochondrial Diseases/blood , Mitochondrial Myopathies/blood , Mitochondrial Trifunctional Protein/deficiency , Muscular Diseases/blood , Nervous System Diseases/blood , Rhabdomyolysis/blood , 3-Hydroxyacyl CoA Dehydrogenases/genetics , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Acetyl-CoA C-Acyltransferase/genetics , Acetyl-CoA C-Acyltransferase/metabolism , Acyl-CoA Dehydrogenase, Long-Chain/blood , Carbon-Carbon Double Bond Isomerases/genetics , Carbon-Carbon Double Bond Isomerases/metabolism , Cardiomyopathies/diet therapy , Cardiomyopathies/pathology , Cardiomyopathies/therapy , Carnitine/blood , Carnitine/genetics , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/blood , Congenital Bone Marrow Failure Syndromes/diet therapy , Congenital Bone Marrow Failure Syndromes/pathology , Congenital Bone Marrow Failure Syndromes/therapy , Enoyl-CoA Hydratase/genetics , Enoyl-CoA Hydratase/metabolism , Exercise Therapy , Fasting , Female , Humans , Lipid Metabolism, Inborn Errors/diet therapy , Lipid Metabolism, Inborn Errors/pathology , Lipid Metabolism, Inborn Errors/therapy , Long-Chain-3-Hydroxyacyl-CoA Dehydrogenase/blood , Male , Metabolism, Inborn Errors/diet therapy , Metabolism, Inborn Errors/pathology , Metabolism, Inborn Errors/therapy , Mitochondrial Diseases/diet therapy , Mitochondrial Diseases/pathology , Mitochondrial Diseases/therapy , Mitochondrial Myopathies/diet therapy , Mitochondrial Myopathies/pathology , Mitochondrial Myopathies/therapy , Mitochondrial Trifunctional Protein/blood , Muscular Diseases/diet therapy , Muscular Diseases/pathology , Muscular Diseases/therapy , Nervous System Diseases/diet therapy , Nervous System Diseases/pathology , Nervous System Diseases/therapy , Racemases and Epimerases/genetics , Racemases and Epimerases/metabolism , Rhabdomyolysis/diet therapy , Rhabdomyolysis/pathology , Rhabdomyolysis/therapy
12.
J Biosci ; 452020.
Article in English | MEDLINE | ID: mdl-32713855

ABSTRACT

The idea of regenerating lost myocardium via cell-based therapies remains as highly considerable. C-kit? stem/ progenitor cells are represented to be suitable candidates for cardiac regeneration compared to other stem cells. A multitude of cytokines from these cells are known to give such multifunctional properties; however, the associated mechanisms of these factors are yet to be totally understood. The aim of the present study was to investigate the in vitro effect of L-carnitine (LC) on cardiac differentiation of c-kit+ cells using a cytokines secretion assay. For this purpose, bone-marrow-resident-c-kit+ cells were enriched by MACS method, and were differentiated to cardiac cells using cardiomyocyte differentiation medium in the absence (control group) and presence of LC (experimental group). Also, characterization of enriched c-kit+ cells was performed using flow cytometry and immunocytochemistry. In the following, the cells were subjected to real-time PCR and Western blotting assay for gene and protein assessment, respectively. Afterward, culture medium was collected from both control (-LC) and experimental groups (+ LC) for cytokine measurement. It was found that 0.2 mM LC significantly increased the mRNA and protein expression of cardiac markers of Ang-1, Ang-2, C-TnI, VEGF, vWF, and SMA in c-kit+-cardiomyogenic-differentiated cells. Also, the significant presence of IL-6, IGF-1, TGF- ß and VEGF were obvious in the cultured media from the experimental group compared with the control group. It can be concluded that the mentioned in vitro effects of LC on cardiac differentiation of c-kit+ cells could have resulted from the secreted cytokines IL-6, IGF-1, TGF- ß and VEGF.


Subject(s)
Heart/growth & development , Insulin-Like Growth Factor I/genetics , Interleukin-6/genetics , Proto-Oncogene Proteins c-kit/genetics , Vascular Endothelial Growth Factor A/genetics , Animals , Bone Marrow Cells/cytology , Carnitine/genetics , Cell- and Tissue-Based Therapy , Flow Cytometry , Heart/physiopathology , Humans , Mesenchymal Stem Cells/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Regeneration/genetics , Transforming Growth Factor beta/genetics
13.
Zhonghua Er Ke Za Zhi ; 58(6): 476-481, 2020 Jun 02.
Article in Chinese | MEDLINE | ID: mdl-32521959

ABSTRACT

Objective: To evaluate and improve the performance of the newborn screening program for primary carnitine deficiency (PCD) based on tandem mass spectrometry and to investigate the incidence of PCD and molecular characteristics of SLC22A5 gene in Guangzhou. Methods: A total of 200 180 neonates born in Guangzhou from 2015 to 2019 were enrolled into the newborn screening program for PCD by tandem mass spectrometry at Guangzhou Newborn Screening Center. The positive results of screening for PCD was defined as free carnitine (C0) less than 10 µmol/L with decreased acylcarnitine species in dried blood spots of three to seven days after birth. Screen-positive newborns and their mothers were recalled for another blood spot sample. The diagnosis was confirmed based on decreased levels of C0 and acylcarnitine species in recalled blood spots and genetic analysis in SLC22A5 gene sequencing. The utility of using the sum of propionylcarnitine and palmitoylcarnitine (C3+C16) as a biomarker for acylcarnitine species in newborn screening was retrospectively evaluated. The levels of C0 and (C3+C16) at first screening were compared between newborns with PCD and newborns born to mothers with PCD by independent t test. The variant spectrum and known pathogenic variants carrier rate of SLC22A5 in 2 395 healthy children in Guangzhou Women and Children's Medical Center through whole exon sequencing were analyzed. Results: Among 200 180 neonates, 239 (0.12%) cases were screen-positive for PCD. A total of 37 patients including 15 newborns and 22 mothers had confirmed PCD. The incidence of PCD was 1/13 345 in newborns and 1/9 099 in mothers, respectively. The positive predictive value of this program was 15.5%. Taking cutoff values of C0<8.5 µmol/L or C0 8.5~9.9 µmol/L with (C3+C16)<2 µmol/L, the number of screen-positive cases would be reduced from 810 to 224 without additional false negative case, when compared with cutoff value C0<10 µmol/L only. Both levels of C0 and (C3+C16) at first screening were not significant difference between newborns with PCD and newborns born to mothers with PCD ((6.2±2.4) vs. (5.0±1.8) µmol/L, (1.4±0.4) vs. (1.2±0.5) µmol/L, t=3.826, 0.326; P=0.058, 0.572). Seven PCD mothers experienced moderate fatigue and dizziness in the morning. One of them presented with cardiomyopathy in pregnancy. Genetic analysis of the SLC22A5 gene showed that p.S467C, p.F17L, p.R254X were the three most common variants in newborns with PCD. In PCD mothers and healthy children, the p.S467C, p.F17L and R399W were the three most common whereas the severe variant p.R254X was rare. The population carrier rate for pathogenic variants was 1 in 65 and the estimated incidence of PCD was about 1/16 500. Conclusions: Newborn screening can detect PCD both in newborns and mothers. Adding a quantitative biomarker (C3+C16) <2 µmol/L into the newborn screening program can improve the PCD screen performance. The severe variant p.R253X was common in PCD newborns but rare in PCD mothers and healthy children, indicating that the current screening program maybe failed to detect all PCD newborns and under-estimated the incidence rate of PCD in Guangzhou.


Subject(s)
Cardiomyopathies/genetics , Carnitine/blood , Carnitine/deficiency , Hyperammonemia/diagnosis , Muscular Diseases/diagnosis , Neonatal Screening/methods , Solute Carrier Family 22 Member 5/genetics , Cardiomyopathies/diagnosis , Cardiomyopathies/metabolism , Carnitine/genetics , Child , Female , Genetic Predisposition to Disease , Humans , Hyperammonemia/genetics , Infant, Newborn , Muscular Diseases/genetics , Pregnancy , Retrospective Studies , Tandem Mass Spectrometry
14.
Clin Chim Acta ; 507: 199-204, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32371215

ABSTRACT

Testing for primary carnitine deficiency (PCD) has been implemented in many newborn screening (NBS) programs, but few large-scale studies on NBS for PCD have been reported in China. This study aimed to assess the incidence and biochemical, clinical, and genetic characteristics of PCD discovered by NBS. Dried blood spots from newborns were analyzed by tandem mass spectrometry (MS/MS) and suspected positive patients were further tested using molecular genetic analysis. Infants who carried two variants in SLC22A5 or those with extremely low free carnitine levels during recall were referred for follow-up and treatment. Over 3.4 million newborns were screened and 113 newborns were diagnosed with PCD, yielding a positive predictive value of 1.93%. In addition, 63 mothers with PCD were identified. The incidence of PCD in newborns and mothers in Zhejiang was 1:30,182 and 1:54,137, respectively. Thirty-seven distinct variants were identified in SLC22A5 of which 10 were novel. c.1400C > G (p.S467C) was the most prevalent variant in both newborns and mothers with PCD, while c.760C > T (p.R254*), which is reportedly common in other Chinese regions, was rarely detected in maternal PCD patients. This study reports the largest series of patients with PCD detected by NBS and identifies 10 novel variants, expanding the variant spectrum of SLC22A5.


Subject(s)
Cardiomyopathies/blood , Carnitine/deficiency , Hyperammonemia/blood , Muscular Diseases/blood , Neonatal Screening , Cardiomyopathies/genetics , Carnitine/blood , Carnitine/genetics , China , Humans , Hyperammonemia/genetics , Infant, Newborn , Muscular Diseases/genetics , Solute Carrier Family 22 Member 5/blood , Solute Carrier Family 22 Member 5/genetics , Tandem Mass Spectrometry
15.
J Pediatr Endocrinol Metab ; 33(6): 683-690, 2020 May 24.
Article in English | MEDLINE | ID: mdl-32447334

ABSTRACT

Background Fatty acid ß-oxidation disorders (FAODs) include more than 15 distinct disorders and have a wide variety of symptoms, usually not evident between episodes of acute decompensation. After the introduction of newborn screening (NBS) using tandem mass spectrometry (MS/MS), early identification of FAODs has become feasible. We analyzed the MS/MS results in Tianjin, China during a six-year period to evaluate the incidence, disease spectrum, and genetic characteristics of FAODs. Methods We analyzed the MS/MS results for screening FAODs from May 2013 to December 2018 in Tianjin, China. Infants with positive screening results were confirmed through next-generation sequencing and validated by Sanger sequencing. Results A total of 220,443 infants were screened and 25 FAODs patients were identified (1:8,817). Primary carnitine deficiency (PCD) with an incidence rate up to 1:20,040 was the most common disorder among all FAODs. Recurrent mutations of relatively common diseases, like PCD and short-chain acyl-CoA dehydrogenase deficiency (SCADD), were identified. During the follow-up, two patients suffered from sudden death due to carnitine palmitoyl transferase-Ⅱ deficiency (CPT Ⅱ) and very-long-chain acyl-CoA dehydrogenase deficiency (VLCAD). Conclusion Our data indicated that FAODs are relatively common in Tianjin and may even cause infant death in certain cases. The elucidated disease spectrum and genetic backgrounds elucidated in this study may contribute to the treatment and prenatal genetic counseling of FAODs.


Subject(s)
Fatty Acids/metabolism , Lipid Metabolism Disorders/diagnosis , Lipid Metabolism Disorders/epidemiology , Lipid Metabolism Disorders/genetics , Cardiomyopathies/diagnosis , Cardiomyopathies/epidemiology , Cardiomyopathies/genetics , Carnitine/deficiency , Carnitine/genetics , Carnitine O-Palmitoyltransferase/deficiency , Carnitine O-Palmitoyltransferase/genetics , China/epidemiology , Congenital Bone Marrow Failure Syndromes/diagnosis , Congenital Bone Marrow Failure Syndromes/epidemiology , Congenital Bone Marrow Failure Syndromes/genetics , Female , Follow-Up Studies , Humans , Hyperammonemia/diagnosis , Hyperammonemia/epidemiology , Hyperammonemia/genetics , Infant, Newborn , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/epidemiology , Lipid Metabolism, Inborn Errors/genetics , Male , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/epidemiology , Metabolism, Inborn Errors/genetics , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/epidemiology , Mitochondrial Diseases/genetics , Muscular Diseases/diagnosis , Muscular Diseases/epidemiology , Muscular Diseases/genetics , Neonatal Screening/methods , Oxidation-Reduction , Tandem Mass Spectrometry
16.
J Appl Toxicol ; 40(7): 1004-1013, 2020 07.
Article in English | MEDLINE | ID: mdl-32084307

ABSTRACT

Amiodarone is known to induce hepatic injury in some recipients. We applied an untargeted metabolomics approach to identify endogenous metabolites with potential as biomarkers for amiodarone-induced liver injury. Oral amiodarone administration for 1 week in rats resulted in significant elevation of acylcarnitines and phospholipids in the liver. Hepatic short- and medium-chain acylcarnitines were dramatically increased in a dose-dependent manner, while the serum levels of these acylcarnitines did not change substantially. In addition, glucose levels were significantly increased in both the serum and liver. Gene expression profiling showed that the hepatic mRNA levels of Cpt1, Cpt2, and Acat1 were significantly suppressed, whereas those of Acot1, Acly, Acss2, and Acsl3 were increased. These results suggest that hepatic acylcarnitines and glucose levels might be increased due to disruption of mitochondrial function and suppression of glucose metabolism. Perturbation of energy metabolism might be associated with amiodarone-induced hepatotoxicity.


Subject(s)
Amiodarone/toxicity , Biomarkers/metabolism , Carnitine/blood , Carnitine/genetics , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Liver/metabolism , RNA, Messenger , Administration, Oral , Amiodarone/administration & dosage , Animals , Genetic Variation , Male , Metabolomics , Rats , Rats, Sprague-Dawley
17.
FASEB J ; 34(1): 1546-1557, 2020 01.
Article in English | MEDLINE | ID: mdl-31914600

ABSTRACT

G protein-coupled receptors (GPCRs) comprise the largest group of membrane receptors in eukaryotic genomes and collectively they regulate nearly all cellular processes. Despite the widely recognized importance of this class of proteins, many GPCRs remain understudied. G protein-coupled receptor 27 (Gpr27) is an orphan GPCR that displays high conservation during vertebrate evolution. Although, GPR27 is known to be expressed in tissues that regulate metabolism including the pancreas, skeletal muscle, and adipose tissue, its functions are poorly characterized. Therefore, to investigate the potential roles of Gpr27 in energy metabolism, we generated a whole body gpr27 knockout zebrafish line. Loss of gpr27 potentiated the elevation in glucose levels induced by pharmacological or nutritional perturbations. We next leveraged a mass spectrometry metabolite profiling platform to identify other potential metabolic functions of Gpr27. Notably, genetic deletion of gpr27 elevated medium-chain acylcarnitines, in particular C6-hexanoylcarnitine, C8-octanoylcarnitine, C9-nonanoylcarnitine, and C10-decanoylcarnitine, lipid species known to be associated with insulin resistance in humans. Concordantly, gpr27 deletion in zebrafish abrogated insulin-dependent Akt phosphorylation and glucose utilization. Finally, loss of gpr27 increased the expression of key enzymes in carnitine shuttle complex, in particular the homolog to the brain-specific isoform of CPT1C which functions as a hypothalamic energy senor. In summary, our findings shed light on the biochemical functions of Gpr27 by illuminating its role in lipid metabolism, insulin signaling, and glucose homeostasis.


Subject(s)
Carnitine/analogs & derivatives , Glucose/metabolism , Homeostasis/genetics , Insulin Resistance/genetics , Receptors, G-Protein-Coupled/genetics , Zebrafish/genetics , Animals , Carnitine/genetics , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Gene Deletion , Glucose/genetics , Insulin/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Zebrafish/metabolism
18.
J Inherit Metab Dis ; 43(2): 290-296, 2020 03.
Article in English | MEDLINE | ID: mdl-31373028

ABSTRACT

Primary carnitine deficiency (PCD) affects fatty acid oxidation and is associated with cardiomyopathy and cardiac arrhythmia, but the risk of sudden death in PCD is unknown. The Faroe Islands have a high prevalence of PCD, 1:300. This study systematically investigated a possible association between untreated PCD and sudden death in young Faroese subjects. We investigated all medico-legal cases of sudden death between 1979 and 2012 among subjects below the age of 45. Stored biomaterial was examined with molecular genetic analysis to reveal PCD. We compared the prevalence of PCD among sudden death cases with that of the background population (0.23%) to calculate the odds ratio (OR) for sudden death with PCD. Biomaterial was available and genetically analyzed from 53 of 65 sudden death cases (82%) in the Faroe Islands. Six (one male and five females) of the 53 cases were homozygous for the PCD related c.95A>G mutation-a prevalence of 11.3% (95% CI 5%-23%) and an OR of 54.3 (95% CI 21-138, P < .0001) for the association between sudden death and untreated PCD. Only 11 of the 53 sudden death cases were women-of whom five were homozygous for the c.95A>G mutation (45.5%) yielding an OR of 348.8 (95% CI 94-1287, P < .0001) for the association between sudden death and untreated PCD in females. This study showed a strong association between sudden death and untreated PCD, especially in females.


Subject(s)
Arrhythmias, Cardiac/etiology , Cardiomyopathies/complications , Carnitine/deficiency , Death, Sudden, Cardiac/etiology , Hyperammonemia/complications , Muscular Diseases/complications , Adolescent , Adult , Cardiomyopathies/genetics , Carnitine/genetics , Child , Child, Preschool , Denmark , Female , Homozygote , Humans , Hyperammonemia/genetics , Infant , Infant, Newborn , Linear Models , Male , Middle Aged , Muscular Diseases/genetics , Mutation , Sex Factors , Young Adult
19.
Mol Genet Metab ; 129(3): 213-218, 2020 03.
Article in English | MEDLINE | ID: mdl-31864849

ABSTRACT

Carnitine Uptake Defect (CUD) is an autosomal recessive disorder due to mutations in the SLC22A5 gene. Classically patients present in infancy with profound muscle weakness and cardiomyopathy with characteristic EKG findings. Later presentations include recurrent hypoketotic hypoglycemia, proximal limb girdle myopathy,and/or recurrent muscle pain. Newborn screening detects most of these clinical variants but in addition has identified maternal CUD often in asymptomatic women. We describe a family ascertained through 3 newborn screening (NBS) positive infants found to be unaffected themselves but in whom the mothers (sisters) were affected. There were also two affected children born to an affected male and his heterozygous wife who were false negatives on NBS but had increased fractional excretion of free carnitine in the urine. Analysis on a Next Generation Sequencing panel specifically designed to fully cover newborn screening disease targets showed a homozygous change in the five probands (SLC22A5; NM_003060:c.-149G > A; p.?). The mutation segregates with the CUD within the family. It is in the 5' UTR and has a frequency within the gnomAd database of 0.001198. Plasma carnitine was decreased and fractional excretion of free carnitine was increased in all affected individuals. Functional carnitine uptake studies in cultured skin fibroblasts of one proband showed carnitine uptake at the 5 µM concentration to be 6% of controls. Relative expression of OCTN2 mRNA to beta-actin mRNA by qRT-PCR was increased in a proband relative to controls by a factor of 465-fold. Western blotting revealed a 120 kDa protein band, as well as a weaker 240 kDa band in the proband, the significance of which is unknown at this time.


Subject(s)
5' Untranslated Regions/genetics , Cardiomyopathies/diagnosis , Cardiomyopathies/genetics , Carnitine/blood , Carnitine/deficiency , Hyperammonemia/diagnosis , Hyperammonemia/genetics , Muscular Diseases/diagnosis , Muscular Diseases/genetics , Solute Carrier Family 22 Member 5/genetics , Actins/genetics , Actins/metabolism , Biological Transport, Active/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Carnitine/genetics , Carnitine/metabolism , Cells, Cultured , Child , Child, Preschool , Female , Fibroblasts/metabolism , Heterozygote , High-Throughput Nucleotide Sequencing , Homozygote , Humans , Hyperammonemia/metabolism , Hyperammonemia/physiopathology , Infant , Infant, Newborn , Male , Muscular Diseases/metabolism , Muscular Diseases/physiopathology , Mutation , Neonatal Screening , Pedigree , Skin/cytology , Skin/metabolism , Solute Carrier Family 22 Member 5/metabolism , Exome Sequencing
20.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 36(12): 1167-1170, 2019 Dec 10.
Article in Chinese | MEDLINE | ID: mdl-31813139

ABSTRACT

OBJECTIVE: To study the prevalence, clinical and genetic characteristics of primary carnitine deficiency (PCD). METHODS: From January 2013 to December 2017, 720 667 newborns and their mothers were tested for PCD by tandem mass spectrometry. Potential mutations of carnitine transporter gene SLC22A5 among suspected PCD patients were analyzed. Dietary guidance and L-carnitine supplementation were provided to the parents. Growth and intelligence development were surveyed during follow-up. RESULTS: In total 21 neonates and 6 mothers were diagnosed with PCD, which yielded an incidence of 1 in 34 317. Eighteen SLC22A5 mutations were detected, which included 4 novel mutations, namely c.1484T>C, c.394-1G>T, c.431T>C and c.265-266insGGCTCGCCACC. Eighteen patients were found to carry compound heterozygous mutations and 3 have carried homozygous SLC22A5 mutations. Three mothers carried compound heterozygous mutations and 2 carried homozygous mutations. Common mutations included c.1400C>G (42.3%), c.760C>T (11.5%) and c.51C>G (7.7%). During the 8-42 month follow-up, neonates with PCD showed no clinical symptoms but normal growth. Blood level of free carnitine was raised in all mothers after the treatment. CONCLUSION: The incidence of neonatal PCD in Henan is 1 in 34 317, with the most common mutation being c.1400C>G. Above finding has enriched the spectrum of SLC22A5 gene mutations.


Subject(s)
Cardiomyopathies/genetics , Carnitine/deficiency , Hyperammonemia/genetics , Muscular Diseases/genetics , Solute Carrier Family 22 Member 5/genetics , Cardiomyopathies/epidemiology , Carnitine/administration & dosage , Carnitine/genetics , China , Female , Humans , Hyperammonemia/epidemiology , Infant, Newborn , Muscular Diseases/epidemiology , Mutation , Neonatal Screening
SELECTION OF CITATIONS
SEARCH DETAIL
...