Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Cardiovasc Disord ; 24(1): 1, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38166572

RESUMEN

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.


Asunto(s)
Cardiomiopatías , Cardiomiopatía Hipertrófica , Hiperamonemia , Enfermedades Musculares , Masculino , Humanos , Niño , Enfermedades Musculares/diagnóstico , Enfermedades Musculares/genética , Carnitina/genética , Carnitina/metabolismo , Irán , Miembro 5 de la Familia 22 de Transportadores de Solutos/genética , Hiperamonemia/diagnóstico , Hiperamonemia/genética , Hiperamonemia/complicaciones , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/genética , Cardiomiopatía Hipertrófica/complicaciones , Mutación
2.
Mol Genet Metab ; 140(4): 107733, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37979236

RESUMEN

BACKGROUND: Systemic primary carnitine deficiency (PCD) is characterized by cardiomyopathy and arrhythmia. Without carnitine supplementation, progression is usually towards fatal cardiac decompensation. While the cardiomyopathy is most likely secondary to energy deficiency, the mechanism of arrhythmia is unclear, and may be related to a short QT interval. OBJECTIVE: We aim to describe rhythmic manifestations at diagnosis and with carnitine supplementation. METHODS: French patients diagnosed for PCD were retrospectively included. Clinical and para clinical data at diagnosis and during follow-up were collected. Electrocardiograms with QT interval measurements were blinded reviewed by two paediatric cardiologists. RESULTS: Nineteen patients (median age at diagnosis 2.3 years (extremes 0.3-28.9)) followed in 8 French centres were included. At diagnosis, 21% of patients (4/19) had arrhythmia (2 ventricular fibrillations, 1 ventricular tachycardia and 1 sudden death), and 84% (16/19) had cardiomyopathy. Six electrocardiograms before treatment out of 11 available displayed a short QT (QTc < 340 ms). Median corrected QTc after carnitine supplementation was 404 ms (extremes 341-447) versus 350 ms (extremes 282-421) before treatment (p < 0.001). The whole QTc was prolonged, and no patient reached the criterion of short QT syndrome with carnitine supplementation. Three patients died, probably from rhythmic cause without carnitine supplementation (two extra-hospital sudden deaths and one non-recoverable rhythmic storm before carnitine supplementation), whereas no rhythmic complication occurred in patients with carnitine supplementation. CONCLUSION: PCD is associated with shortening of the QT interval inducing severe arrhythmia. A potential explanation would be a toxic effect of accumulated fatty acid and metabolites on ionic channels embedded in the cell membrane. Carnitine supplementation normalizes the QTc and prevents arrhythmia. Newborn screening of primary carnitine deficiency would prevent avoidable deaths.


Asunto(s)
Cardiomiopatías , Síndrome de QT Prolongado , Recién Nacido , Niño , Humanos , Preescolar , Muerte Súbita Cardíaca/etiología , Muerte Súbita Cardíaca/prevención & control , Estudios Retrospectivos , Arritmias Cardíacas/complicaciones , Cardiomiopatías/complicaciones , Carnitina/metabolismo , Electrocardiografía/efectos adversos
3.
Clin Genet ; 103(6): 644-654, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36840705

RESUMEN

Biallelic variants in the ACADM gene cause medium-chain acyl-CoA dehydrogenase deficiency (MCADD). This study reports on differences in the occurrence of secondary free carnitine (C0) deficiency and different biochemical phenotypes related to genotype and age in 109 MCADD patients followed-up at a single tertiary care center during 22 years. C0 deficiency occurred earlier and more frequently in c.985A>G homozygotes (genotype A) compared to c.985A>G compound heterozygotes (genotype B) and individuals carrying variants other than c.985A>G and c.199C>T (genotype D) (median age 4.2 vs. 6.6 years; p < 0.001). No patient carrying c.199C>T (genotype C) developed C0 deficiency. A daily dosage of 20-40 mg/kg carnitine was sufficient to maintain normal C0 concentrations. Compared to genotype A as reference group, octanoylcarnitine (C8) was significantly lower in genotypes B and C, whereas C0 was significantly higher by 8.28 µmol/L in genotype C (p < 0.05). In conclusion, C0 deficiency is mainly found in patients with pathogenic genotypes associated with high concentrations of presumably toxic acylcarnitines, while individuals carrying the variant c.199C>T are spared and show consistently mild biochemical phenotypes into adulthood. Low-dose carnitine supplementation maintains normal C0 concentrations. However, future studies need to evaluate clinical benefits on acute and chronic manifestations of MCADD.


Asunto(s)
Errores Innatos del Metabolismo Lipídico , Tamizaje Neonatal , Humanos , Recién Nacido , Genotipo , Errores Innatos del Metabolismo Lipídico/genética , Carnitina , Aminoácidos , Estudios de Asociación Genética , Acil-CoA Deshidrogenasa/química , Acil-CoA Deshidrogenasa/genética
4.
Ann Noninvasive Electrocardiol ; 28(6): e13077, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37658577

RESUMEN

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.


Asunto(s)
Arritmias Cardíacas , Cardiomiopatías , Carnitina/deficiencia , Hiperamonemia , Enfermedades Musculares , Proteínas de Transporte de Catión Orgánico , Femenino , Humanos , Adolescente , Proteínas de Transporte de Catión Orgánico/genética , Miembro 5 de la Familia 22 de Transportadores de Solutos/genética , Electrocardiografía , Cardiomiopatías/complicaciones , Cardiomiopatías/diagnóstico por imagen , Cardiomiopatías/genética , Mutación , Carnitina/uso terapéutico , Carnitina/genética , Síndrome
5.
Pediatr Int ; 65(1): e15404, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36321377

RESUMEN

BACKGROUND: Primary carnitine deficiency (PCD) is screened by expanded newborn screening (NBS) using tandem mass spectrometry (MS/MS) that can detect both affected neonates and mothers. This study aimed to delineate the clinical, biochemical, and molecular findings of Thai PCD patients. METHODS: Expanded NBS using MS/MS was implemented in Bangkok and 146,757 neonates were screened between 2014 and 2018. PCD was screened by low free carnitine (C0) levels in dried blood spots. Plasma C0 levels and C0 clearance values were measured in neonates and their mothers with positive screening results. Clinically diagnosed cases were described. The coding regions and intron-exon boundaries of the SLC22A5 gene were sequenced in all cases with low plasma C0 levels. RESULTS: There were 14 cases with confirmed PCD: two clinically diagnosed cases, and 12 cases identified through NBS including five newborns, six mothers, and one older sibling. Thus, the incidence of PCD in neonates was 1:29,351. All affected neonates and mothers were asymptomatic except one mother with dilated cardiomyopathy. SLC22A5 gene sequencing identified biallelic causative variants in all cases, comprising 10 different variants of which four were novel. c.51C > G (p.Phe17Leu) and c.760C > T (p.Arg254Ter) were the most prevalent variants in this study. Cases with significant clinical features tended to have higher C0 clearance values. CONCLUSIONS: Primary carnitine deficiency is a common inherited metabolic disorder (IMD) in Thailand. Our findings broaden the spectrum of SLC22A5 variants. The future national NBS program will shed more light on PCD and other IMDs in Thailand.


Asunto(s)
Cardiomiopatías , Miembro 5 de la Familia 22 de Transportadores de Solutos , Espectrometría de Masas en Tándem , Femenino , Humanos , Recién Nacido , Cardiomiopatías/diagnóstico , Cardiomiopatías/genética , Carnitina/metabolismo , Mutación , Tamizaje Neonatal/métodos , Miembro 5 de la Familia 22 de Transportadores de Solutos/genética , Pueblos del Sudeste Asiático/genética , Tailandia/epidemiología
6.
J Inherit Metab Dis ; 45(6): 1118-1129, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35778950

RESUMEN

Our aim was to study the effect of secondary carnitine deficiency (SCD) and carnitine supplementation on important outcome measures for persons with medium-chain Acyl-CoA dehydrogenase deficiency (MCADD). We performed a large retrospective observational study using all recorded visits of persons with MCADD in the University Medical Center Groningen, the Netherlands, between October 1994 and October 2019. Frequency and duration of acute unscheduled preventive hospital visits, exercise tolerance, fatigue, and muscle pain were considered important clinical outcomes and were studied in relation to (acyl)carnitine profile and carnitine supplementation status. The study encompassed 1228 visits of 93 persons with MCADD. >60% had SCD during follow-up. This included only persons with severe MCADD. Carnitine supplementation and SCD were unrelated to the frequency and duration of the acute unscheduled preventive hospital visits (P > 0.05). The relative risk for fatigue, muscle ache, or exercise intolerance was equal between persons with and without SCD (RR 1.6, 95% CI 0.48-5.10, P = 0.4662). No episodes of metabolic crisis were recorded in non-carnitine-supplemented persons with MCADD and SCD. In some persons with MCADD, SCD resolved without carnitine supplementation. There is absence of real-world evidence in favor of routine carnitine analysis and carnitine supplementation in the follow-up of persons with MCADD.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos , Errores Innatos del Metabolismo Lipídico , Humanos , Acil-CoA Deshidrogenasa , Errores Innatos del Metabolismo Lipídico/metabolismo , Estudios Retrospectivos
7.
J Inherit Metab Dis ; 45(3): 386-405, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34997761

RESUMEN

A broad spectrum of signs and symptoms has been attributed to primary carnitine deficiency (PCD) since its first description in 1973. Advances in diagnostic procedures have improved diagnostic accuracy and the introduction of PCD in newborn screening (NBS) programs has led to the identification of an increasing number of PCD patients, including mothers of screened newborns, who may show a different phenotype compared to clinically diagnosed patients. To elucidate the spectrum of signs and symptoms in PCD patients, we performed a structured literature review. Using a case-by-case approach, clinical characteristics, diagnostic data, and mode of patient identification were recorded. Signs and symptoms were categorized by organ involvement. In total, 166 articles were included, reporting data on 757 individual patients. In almost 20% (N = 136) of the cases, the diagnosis was based solely on low carnitine concentration which we considered an uncertain diagnosis of PCD. The remaining 621 cases had a diagnosis based on genetic and/or functional (ie, carnitine transporter activity) test results. In these 621 cases, cardiac symptoms (predominantly cardiomyopathy) were the most prevalent (23.8%). Neurological (7.1%), hepatic (8.4%), and metabolic (9.2%) symptoms occurred mainly in early childhood. Adult onset of symptoms occurred in 16 of 194 adult patients, of whom 6 (3.1%) patients suffered a severe event without any preceding symptom (five cardiac events and one coma). In conclusion, symptoms in PCD predominantly develop in early childhood. Most newborns and mothers of newborns detected through NBS remain asymptomatic. However, though rarely, severe complications do occur in both groups.


Asunto(s)
Cardiomiopatías , Hiperamonemia , Enfermedades Musculares , Cardiomiopatías/complicaciones , Cardiomiopatías/diagnóstico , Cardiomiopatías/genética , Carnitina/deficiencia , Carnitina/metabolismo , Preescolar , Humanos , Hiperamonemia/complicaciones , Hiperamonemia/diagnóstico , Hiperamonemia/genética , Recién Nacido , Enfermedades Musculares/complicaciones , Enfermedades Musculares/diagnóstico , Enfermedades Musculares/genética , Tamizaje Neonatal/métodos , Miembro 5 de la Familia 22 de Transportadores de Solutos/genética
8.
Chemotherapy ; 67(2): 96-101, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34839292

RESUMEN

BACKGROUND: Some chemotherapeutic agents cause carnitine deficiency, which causes general fatigue. However, there is no study on carnitine deficiency in patients with chronic myeloid leukemia (CML) during tyrosine kinase inhibitor (TKI) therapy. OBJECTIVE: In this study, we investigated carnitine concentrations in patients with CML receiving TKI therapy. METHOD: This study included patients with well-controlled CML. Total carnitine and free carnitine concentrations were evaluated using the enzyme cycling method. The brief fatigue inventory (BFI) and cancer fatigue scale (CFS) were used to assess general fatigue developed during TKI therapy. RESULTS: Fifty-five patients on TKI therapy were included. Of these, 12 (21.8%) patients had low free carnitine concentrations. Free carnitine concentrations were higher in men than in women. Younger age was closely associated with lower free carnitine concentrations. TKI type, TKI dose, treatment response, or therapy duration were not associated with free carnitine concentrations. None of the scores (the global fatigue score with the BFI and CFS score) correlated with carnitine concentrations. Concentrations of free carnitine in patients in the treatment-free remission group were slightly higher than those in the TKI group, with only 9.1% having a low concentration of free carnitine. CONCLUSION: Carnitine deficiency is probably not a major cause of general fatigue but may occur in patients with CML receiving TKI therapy.


Asunto(s)
Leucemia Mielógena Crónica BCR-ABL Positiva , Femenino , Humanos , Masculino , Cardiomiopatías , Carnitina/deficiencia , Fatiga/etiología , Hiperamonemia , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Enfermedades Musculares , Inhibidores de Proteínas Quinasas/efectos adversos
9.
Turk J Med Sci ; 52(3): 724-729, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36326333

RESUMEN

BACKGROUND: Juvenile idiopathic arthritis (JIA) is the most common rheumatic disease in childhood and manifests mainly as autoinflammation of the joints and other tissues. Several treatment options such as nonsteroidal antiinflammatory drugs, methotrexate, and intra-articular steroids are widely used to relieve and improve this inflammation. Secondary carnitine deficiency can be detected in chronic diseases by either renal loss or increased demand. While carnitine status can be associated with several conditions, in the present study our aim is to determine the levels of free carnitine and acyl-carnitine in Turkish JIA patients. METHODS: One hundred and fourteen patients diagnosed with juvenile idiopathic arthritis and 50 healthy individuals who served as the control group were included in the study. A fasting blood sample was collected from the children in both groups to determine free carnitine and acylcarnitine ester by quadripole electrospray tandem mass spectrometry (ESI-MS/ MS). RESULTS: Screening of acyl-carnitine profile revealed free carnitine, C14, C14:2, C16, C16-OH, and C18 carnitine levels were higher (p < 0.0001, p < 0.0001, p < 0.001, p < 0.001, and p = 0.011, respectively), while C2, C3, C4, C6, C8, C10, C10:1, C10:2, C3DC, C4DC, C5DC, C4-OH, and C18:1-OH carnitine levels were lower (p < 0.0001) in JIA patients in comparison to the control group. Total acyl-carnitine levels (p < 0.001) and acyl-carnitine to free carnitine ratio (p < 0.001) were also lower in JIA patients than the control group. Free carnitine levels were significantly higher (48.05 ± 13.36 µmol/L) in patients under antiinflammatory drug therapy than those who did not receive any treatment (43.18 ± 7.96 µmol/L) (p = 0.004). DISCUSSION: In the present study we were not able to define secondary carnitine deficiency in JIA patients, although free carnitine and acyl-carnitine variations were detected in JIA patients. In conclusion, routine carnitine supplementation is not recommended in all patients with JIA.


Asunto(s)
Artritis Juvenil , Niño , Humanos , Artritis Juvenil/tratamiento farmacológico , Carnitina , Espectrometría de Masas en Tándem , Ayuno , Aminoácidos
10.
Mol Genet Metab ; 129(3): 213-218, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31864849

RESUMEN

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.


Asunto(s)
Regiones no Traducidas 5'/genética , Cardiomiopatías/diagnóstico , Cardiomiopatías/genética , Carnitina/sangre , Carnitina/deficiencia , Hiperamonemia/diagnóstico , Hiperamonemia/genética , Enfermedades Musculares/diagnóstico , Enfermedades Musculares/genética , Miembro 5 de la Familia 22 de Transportadores de Solutos/genética , Actinas/genética , Actinas/metabolismo , Transporte Biológico Activo/genética , Cardiomiopatías/metabolismo , Cardiomiopatías/fisiopatología , Carnitina/genética , Carnitina/metabolismo , Células Cultivadas , Niño , Preescolar , Femenino , Fibroblastos/metabolismo , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Hiperamonemia/metabolismo , Hiperamonemia/fisiopatología , Lactante , Recién Nacido , Masculino , Enfermedades Musculares/metabolismo , Enfermedades Musculares/fisiopatología , Mutación , Tamizaje Neonatal , Linaje , Piel/citología , Piel/metabolismo , Miembro 5 de la Familia 22 de Transportadores de Solutos/metabolismo , Secuenciación del Exoma
11.
J Inherit Metab Dis ; 43(2): 290-296, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31373028

RESUMEN

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.


Asunto(s)
Arritmias Cardíacas/etiología , Cardiomiopatías/complicaciones , Carnitina/deficiencia , Muerte Súbita Cardíaca/etiología , Hiperamonemia/complicaciones , Enfermedades Musculares/complicaciones , Adolescente , Adulto , Cardiomiopatías/genética , Carnitina/genética , Niño , Preescolar , Dinamarca , Femenino , Homocigoto , Humanos , Hiperamonemia/genética , Lactante , Recién Nacido , Modelos Lineales , Masculino , Persona de Mediana Edad , Enfermedades Musculares/genética , Mutación , Factores Sexuales , Adulto Joven
12.
BMC Pediatr ; 20(1): 478, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33050909

RESUMEN

BACKGROUND: Primary carnitine deficiency (PCD) is an autosomal recessive disorder affecting the carnitine cycle and resulting in defective fatty acid oxidation. Neonatal intrahepatic cholestasis caused by citrin deficiency (NICCD) is an autosomal recessive disorder and one of the main causes of inherited neonatal cholestasis. Both PCD and NICCD are included in the current expanded newborn screening (NBS) targets. CASE PRESENTATION: Targeted exome sequencing was performed on a Chinese proband, and Sanger sequencing was utilised to validate the detected mutations. The patient who was initially suspected to have PCD based on the NBS results presented with neonatal intrahepatic cholestasis and ventricular septal defect. Further investigations not only confirmed PCD but also revealed the presence of NICCD. Four distinct mutations were detected, including c.51C > G (p.F17L) and c.760C > T (p.R254X) in SLC22A5 as well as c.615 + 5G > A and IVS16ins3kb in SLC25A13. CONCLUSIONS: This is the first reported case of PCD and NICCD occurring in the same patient. The dual disorders in a newborn broaden our understanding of inherited metabolic diseases. Thus, this study highlighted the importance of further genetic testing in patients presenting with unusual metabolic screening findings.


Asunto(s)
Carnitina , Colestasis Intrahepática , Citrulinemia , Cardiomiopatías , Carnitina/deficiencia , China , Colestasis Intrahepática/etiología , Colestasis Intrahepática/genética , Citrulinemia/complicaciones , Humanos , Hiperamonemia , Recién Nacido , Proteínas de Transporte de Membrana Mitocondrial/genética , Enfermedades Musculares , Mutación , Miembro 5 de la Familia 22 de Transportadores de Solutos
13.
Ren Fail ; 42(1): 146-153, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32003308

RESUMEN

Carnitine deficiency contributes to developing various pathological conditions, such as cardiac dysfunction, muscle weakness, and erythropoietin-resistant anemia in patients undergoing hemodialysis. However, a conclusion has not been reached concerning the prevalence and the effect of carnitine deficiency in patients undergoing peritoneal dialysis (PD). In this study, the prevalence of carnitine deficiency and the clinical factors associated with carnitine deficiency were investigated in 60 patients undergoing PD. The median age of the patients was 62.5 years (52.5-72.5 years), the proportion of male sex was 44/60 (73.3%), and the median PD period was 24 months (12-45 months). Carnitine deficiency (acyl carnitine/free carnitine ratio >0.4) was detected in 56/60 (93%) patients. Multiple regression analysis showed that the erythropoietin resistance index was independently associated with carnitine deficiency (ß = 0.283, p = 0.04). These results suggest that carnitine plays pivotal roles in hematogenesis in patients undergoing PD.


Asunto(s)
Carnitina/deficiencia , Resistencia a Medicamentos , Eritropoyetina/uso terapéutico , Fallo Renal Crónico/terapia , Diálisis Peritoneal , Anciano , Anemia/etiología , Carnitina/sangre , Estudios Transversales , Eritropoyetina/administración & dosificación , Femenino , Humanos , Japón , Fallo Renal Crónico/sangre , Fallo Renal Crónico/complicaciones , Masculino , Persona de Mediana Edad , Análisis de Regresión , Estudios Retrospectivos
14.
J Emerg Med ; 59(1): e17-e20, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32448729

RESUMEN

BACKGROUND: Primary carnitine deficiency (PCD) is a rare but potentially life-threatening genetic disorder if left untreated. Although some patients remain asymptomatic lifelong, a few patients present with hepatic encephalopathy, hypoglycemia, cardiomyopathy, dysrhythmia, and even sudden death. CASE REPORT: A 25-year-old woman with PCD collapsed suddenly while eating lunch. Bystander cardiopulmonary resuscitation (CPR) was performed for 8 min, with automated external defibrillation once before admission. Upon arrival at our emergency department (ED), she was unresponsive without a pulse or spontaneous breathing. The initial heart rhythm on the electrocardiogram monitor was ventricular fibrillation (VF). The medical staff continued CPR with defibrillation for sustained VF. Return of spontaneous circulation (ROSC) was achieved after a total resuscitation time of 14 min, with defibrillation twice after cardiac arrest. The heart rhythm after ROSC was atrial fibrillation, with a rapid ventricular rate initially and subsequent progression to sinus tachycardia with diffuse ST segment depression and a prolonged QT interval. Her low carnitine level was consistent with her underlying disease. Cardiac magnetic resonance imaging and sonography for detection of cardiomyopathy showed no significant findings. With carnitine supplementation for a few days, her plasma carnitine level returned to 30 µM, with no recurrence of ventricular dysrhythmia. WHY SHOULD AN EMERGENCY PHYSICIAN BE AWARE OF THIS?: PCD is rare but could be life-threatening, and compiling detailed histories may help emergency physicians to determine the cause of sudden cardiac death after resuscitation. This information may be used to correct potential underlying problems and prevent recurrence of the condition after treatment.


Asunto(s)
Cardiomiopatías , Reanimación Cardiopulmonar , Hiperamonemia , Adulto , Cardiomiopatías/complicaciones , Cardiomiopatías/diagnóstico , Carnitina/deficiencia , Carnitina/uso terapéutico , Cardioversión Eléctrica , Femenino , Humanos , Enfermedades Musculares , Fibrilación Ventricular/complicaciones
15.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 45(10): 1164-1171, 2020 Oct 28.
Artículo en Inglés, Zh | MEDLINE | ID: mdl-33268576

RESUMEN

OBJECTIVES: Primary carnitine deficiency (PCD) is a rare fatty acid metabolism disorder that can cause neonatal death. This study aims to analyze carnitine levels and detect SLC22A5 gene in newborns with carnitine deficiency, to provide a basis for early diagnosis of PCD, and to explore the relationship between carnitine in blood and SLC22A5 genotype. METHODS: A total of 40 neonates with low free carnitine (C0<10 µmol/L) in blood were the subjects of the study. SLC22A5 gene was detected by Sanger sequencing to analyze the value of carnitine, the results of gene test and their relationship. RESULTS: A total of 15 variants of SLC22A5 gene were detected, including 11 pathogenic or likely pathogenic variants and 4 variants of uncertain significance. There were 5 new mutations: c.288delG (p.G96fsX33), c.744_745insTCG (p.M258_L259insS), c.752A>G (p.Y251C), c.495 C>A (p.R165E), and c.1298T>C (p.M433T). We found 14 PCD patients including 2 homozygous mutations and 12 heterozygous mutations, 14 with 1 mutation, and 12 with no mutation among 40 children. The C0 concentration of children with SLC22A5 gene homozygous or complex heterozygous mutations was (4.95±1.62) µmol/L in the initial screening, and (3.90±1.33) µmol/L in the second screening. The C0 concentration of children with no mutation was (7.04±2.05) µmol/L in the initial screening, and (8.02±2.87) µmol/L in the second screening. There were significant differences between children with homozygous or compound heterozygous mutations and with no mutation in C0 concentration of the initial and the second screening (both P<0.05), as well as between children with truncated mutation and with untruncated mutation in C0 concentration of the initial screening (P=0.022). CONCLUSIONS: There are 5 new mutations which enriched the mutation spectrum of SLC22A5 gene. C0<5 µmol/L is highly correlated with SLC22A5 gene homozygous or compound heterozygous mutations. Children with truncated mutation may have lower C0 concentration than that with untruncated mutation in the initial screening.


Asunto(s)
Hiperamonemia , Enfermedades Musculares , Cardiomiopatías , Carnitina/deficiencia , Niño , Humanos , Hiperamonemia/genética , Recién Nacido , Enfermedades Musculares/genética , Mutación , Miembro 5 de la Familia 22 de Transportadores de Solutos/genética
16.
Hum Mutat ; 40(10): 1899-1904, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31187905

RESUMEN

Primary carnitine deficiency is caused by a defect in the active cellular uptake of carnitine by Na+ -dependent organic cation transporter novel 2 (OCTN2). Genetic diagnostic yield for this metabolic disorder has been relatively low, suggesting that disease-causing variants are missed. We Sanger sequenced the 5' untranslated region (UTR) of SLC22A5 in individuals with possible primary carnitine deficiency in whom no or only one mutant allele had been found. We identified a novel 5'-UTR c.-149G>A variant which we characterized by expression studies with reporter constructs in HeLa cells and by carnitine-transport measurements in fibroblasts using a newly developed sensitive assay based on tandem mass spectrometry. This variant, which we identified in 57 of 236 individuals of our cohort, introduces a functional upstream out-of-frame translation initiation codon. We show that the codon suppresses translation from the wild-type ATG of SLC22A5, resulting in reduced OCTN2 protein levels and concomitantly lower transport activity. With an allele frequency of 24.2% the c.-149G>A variant is the most frequent cause of primary carnitine deficiency in our cohort and may explain other reported cases with an incomplete genetic diagnosis. Individuals carrying this variant should be clinically re-evaluated and monitored to determine if this variant has clinical consequences.


Asunto(s)
Regiones no Traducidas 5' , Cardiomiopatías/genética , Carnitina/deficiencia , Codón Iniciador , Predisposición Genética a la Enfermedad , Hiperamonemia/genética , Enfermedades Musculares/genética , Mutación , Miembro 5 de la Familia 22 de Transportadores de Solutos/genética , Alelos , Secuencia de Aminoácidos , Secuencia de Bases , Transporte Biológico , Cardiomiopatías/diagnóstico , Cardiomiopatías/metabolismo , Carnitina/genética , Carnitina/metabolismo , Línea Celular , Frecuencia de los Genes , Genes Reporteros , Estudios de Asociación Genética , Humanos , Hiperamonemia/diagnóstico , Hiperamonemia/metabolismo , Enfermedades Musculares/diagnóstico , Enfermedades Musculares/metabolismo , Miembro 5 de la Familia 22 de Transportadores de Solutos/metabolismo
17.
Toxicol Appl Pharmacol ; 368: 18-25, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30735677

RESUMEN

In spite of remarkable reduction in the number of children born with HIV due to antiretroviral therapy, concerns remain on the short- and long-term effects of antiretroviral drugs at the feto-placental unit. Cardio- and skeletal myopathies have been reported in children exposed to antiretroviral drugs prenatally. These conditions have also been described in perturbed placental transfer of l-carnitine, an essential co-factor in fatty acid oxidation. Due to limited fetal and placental synthesis, carnitine supply is maintained through the placental carnitine uptake from maternal blood by the organic cation/carnitine transporters OCTN1 and OCTN2 (SLC22A4 and SLC22A5, respectively). The aim of our study was to investigate potential inhibition of placental carnitine uptake by a broad range of antiretroviral drugs comprising nucleoside/nucleotide reverse transcriptase inhibitors (lamivudine, zidovudine, abacavir, tenofovir disoproxil fumarate), non-nucleoside reverse transcriptase inhibitors (rilpivirine, efavirenz, etravirine), protease inhibitors (ritonavir, lopinavir, atazanavir, saquinavir, tipranavir), integrase inhibitors (raltegravir, dolutegravir, elvitegravir) and viral entry inhibitor, maraviroc. Studies in choriocarcinoma BeWo cells and human placenta-derived models confirmed predominant expression and function of OCTN2 above OCTN1 in l-carnitine transport. Subsequent screenings in BeWo cells and isolated MVM vesicles revealed seven antiretroviral drugs as inhibitors of the Na+-dependent l-carnitine uptake, corresponding to OCTN2. Ritonavir, saquinavir and elvitegravir showed the highest inhibitory potential which was further confirmed for ritonavir and saquinavir in placental fresh villous fragments. Our data indicate possible impairment in placental and fetal supply of l-carnitine with ritonavir and saquinavir, while suggesting retained placental carnitine transport with the other antiretroviral drugs.


Asunto(s)
Antirretrovirales/toxicidad , Carnitina/metabolismo , Placenta/efectos de los fármacos , Miembro 5 de la Familia 22 de Transportadores de Solutos/antagonistas & inhibidores , Transporte Biológico , Línea Celular Tumoral , Femenino , Humanos , Exposición Materna/efectos adversos , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Proteínas de Transporte de Catión Orgánico/metabolismo , Placenta/metabolismo , Embarazo , Medición de Riesgo , Miembro 5 de la Familia 22 de Transportadores de Solutos/metabolismo , Simportadores
18.
J Infect Chemother ; 25(11): 913-916, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31189503

RESUMEN

Rotavirus infection is a major cause of gastroenteritis, which occurs mainly in children. Liver dysfunction due to rotavirus gastroenteritis has been reported; however, acute hepatitis due to this disease is very rare. We present a rare case in which rotavirus gastroenteritis led to sequential diagnosis of acute hepatitis and systemic primary carnitine deficiency (CDSP) in a 1-year-old girl. The patient's symptoms (hypoglycemia, hepatomegaly, and elevated levels of serum transaminases and creatinine kinase) suggested a steatosis causing liver dysfunction. She was initially considered to have a beta oxygenation defect or secondary carnitine deficiency caused by pivalic acid-containing antibiotics; however, repetitive carnitine analysis and free carnitine clearance measurement confirmed primary carnitine deficiency (carnitine transporter deficiency). Children with severe liver dysfunction due to rotavirus infection and presenting with liver steatosis should undergo blood acyl carnitine analysis to detect potential carnitine or other beta oxidation deficiencies, especially if newborn screening for these diseases is not available.


Asunto(s)
Cardiomiopatías/etiología , Carnitina/deficiencia , Gastroenteritis/complicaciones , Gastroenteritis/etiología , Hepatitis/etiología , Hiperamonemia/etiología , Enfermedades Musculares/etiología , Infecciones por Rotavirus/complicaciones , Enfermedad Aguda , Cardiomiopatías/virología , Femenino , Gastroenteritis/virología , Hepatitis/virología , Humanos , Hiperamonemia/virología , Lactante , Enfermedades Musculares/virología , Rotavirus/patogenicidad
19.
BMC Pediatr ; 19(1): 79, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30885166

RESUMEN

BACKGROUND: Maternofetal carnitine transport through the placenta is the main route of fetal carnitine uptake. Decreased free carnitine levels discovered by newborn screening has identified many asymptomatic adult women with systemic primary carnitine deficiency (PCD). Here, we presented amplitude integrated electroencephalogram (aEEG) and magnetic resonance imaging (MRI) findings from a neonate with epilepsy whose mother was carnitine deficient. CASE PRESENTATION: A one-day-old female newborn was admitted after experiencing seizures for half a day; status epilepticus was found on the continuous normal voltage background pattern with immature sleep-wake cycling during aEEG monitoring. On T1-weighted, T2-weighted, FLAIR, and DWI head MRI, there were various degrees of hyperintense signals and diffusion restrictions in the deep white matter of the right hemisphere. Tandem mass spectrometry discovered carnitine deficiency on the second day, which elevated to normal by the 9th day before L-carnitine supplementation was started. The patient was treated with phenobarbital after admission. No further seizures were noted by day 5. It was confirmed that the patient's mother had a low level of serum-free carnitine. Gene analyses revealed that the newborn had heterozygote mutations on c.1400C > G of the SLC22A5 gene, and her mother had homozygous mutations on c.1400C > G. The patient had a good outcome at the 8-month follow up. CONCLUSIONS: Maternal carnitine deficiency that occurs during the perinatal period may manifest as secondary epilepsy with cerebral injury in neonates. The short-term neurodevelopmental outcomes were good. Early diagnosis of asymptomatic PCD in female patients can provide guidance for future pregnancies.


Asunto(s)
Cardiomiopatías/complicaciones , Carnitina/deficiencia , Hiperamonemia/complicaciones , Enfermedades Musculares/complicaciones , Convulsiones/etiología , Encéfalo/diagnóstico por imagen , Cardiomiopatías/diagnóstico , Cardiomiopatías/genética , Carnitina/sangre , Carnitina/genética , Electroencefalografía , Femenino , Enfermedades Fetales/etiología , Humanos , Hiperamonemia/diagnóstico , Hiperamonemia/genética , Recién Nacido , Enfermedades del Recién Nacido/diagnóstico , Imagen por Resonancia Magnética , Madres , Enfermedades Musculares/diagnóstico , Enfermedades Musculares/genética , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA