Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Mol Genet Metab ; 134(3): 257-266, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34635437

RESUMEN

Propionic acidemia (PA) is an autosomal recessive metabolic disorder after gene encoding propionyl-CoA carboxylase, Pcca or Pccb, is mutated. This genetic disorder could develop various complications which are ascribed to dysregulated propionyl-CoA metabolism in organs. However, the effect of attenuated PCC on propionyl-CoA metabolism in different organs remains to be fully understood. We investigated metabolic perturbations in organs of Pcca-/-(A138T) mice (a mouse model of PA) under chow diet and acute administration of [13C3]propionate to gain insight into pathological mechanisms of PA. With chow diet, the metabolic alteration is organ dependent. l-Carnitine reduction induced by propionylcarnitine accumulation only occurs in lung and liver of Pcca-/- (A138T) mice. [13C3]Propionate tracing data demonstrated that PCC activity was dramatically reduced in Pcca-/-(A138T) brain, lung, liver, kidney, and adipose tissues, but not significantly changed in Pcca-/-(A138T) muscles (heart and skeletal muscles) and pancreas, which was largely supported by PCCA expression data. The largest expansion of propionylcarnitine in Pcca-/-(A138T) heart after acute administration of propionate indicated the vulnerability of heart to high circulating propionate. The overwhelming propionate in blood also stimulated ketone production from the increased fatty acid oxidation in Pcca-/-(A138T) liver by lowering malonyl-CoA, which has been observed in cases where metabolic decompensation occurs in PA patients. This work shed light on organ-specific metabolic alternations under varying severities of PA.


Asunto(s)
Acilcoenzima A/análisis , Propionatos/metabolismo , Acidemia Propiónica/fisiopatología , Alimentación Animal , Animales , Modelos Animales de Enfermedad , Corazón/fisiopatología , Hígado/química , Hígado/fisiopatología , Pulmón/química , Pulmón/fisiopatología , Masculino , Análisis de Flujos Metabólicos , Metabolómica , Metilmalonil-CoA Descarboxilasa/genética , Ratones , Propionatos/sangre
2.
Mol Genet Metab ; 130(1): 41-48, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32067920

RESUMEN

BACKGROUND: Propionic acidemia (PA) is an organic aciduria caused by inherited deficiency of propionyl-CoA carboxylase. Left ventricular dysfunction and QT prolongation may lead to life-threatening complications. Systematic analyses of cardiac phenotypes, in particular effects of specific cardiac therapies, are scarce. METHODS: In this longitudinal observational monocentric study (data from 1989 to 2017) all PA patients treated at our center were included. Echocardiographic parameters (left ventricular end-diastolic diameter: LVEDD, left ventricular shortening fraction, mitral valve Doppler inflow pattern) and 12­lead electrocardiogram recordings (corrected QT interval: QTc) were analyzed. Symptomatic patients were dichotomized to the group "early-onset" (symptoms within 28 days of life) and "late-onset" (symptoms after 28 days). Associations between cardiac function, LVEDD, QTc and clinical parameters (age at onset, beta-blocker or Angiotensin-converting enzyme inhibitor = ACE-I therapy) were analyzed. RESULTS: 18 patients with PA were enrolled, 17 of them were symptomatic and one asymptomatic, with a median age at diagnosis of 6 days. 14/17 (82%) had early onset disease manifestation. Systolic left ventricular dysfunction (i.e. hypokinetic phenotype of cardiomyopathy) was diagnosed in 7/18 (39%) patients at a median age of 14.4 years, all had early onset. Two patients had a dilated left ventricle and systolic left ventricular dysfunction (i.e. dilated hypokinetic phenotype - dilated cardiomyopathy). Diastolic left ventricular dysfunction was found in 11/18 (61%) individuals, typically preceding systolic left ventricular dysfunction. ACE-I therapy did not improve systolic left ventricular function. Mean QTc was 445 ms (+/- 18.11 ms). Longer QTc was associated with larger LVEDD. CONCLUSIONS: Systolic left ventricular dysfunction was found in 39% of patients, reflecting high disease severity. Two thirds of all individuals showed signs of diastolic left ventricular dysfunction usually preceding systolic left ventricular dysfunction; it therefore may be considered as an indicator for early cardiac disease manifestation, possibly allowing earlier treatment modification. Unresponsiveness to routine cardiac therapy highlights the need to evaluate further strategies, such as liver transplantation.


Asunto(s)
Cardiomiopatías/complicaciones , Síndrome de QT Prolongado/complicaciones , Acidemia Propiónica/complicaciones , Disfunción Ventricular Izquierda/complicaciones , Adolescente , Antagonistas Adrenérgicos beta/uso terapéutico , Adulto , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Cardiomiopatías/tratamiento farmacológico , Cardiomiopatías/fisiopatología , Niño , Preescolar , Estudios de Cohortes , Ecocardiografía , Electrocardiografía , Femenino , Estudios de Seguimiento , Humanos , Lactante , Síndrome de QT Prolongado/tratamiento farmacológico , Síndrome de QT Prolongado/fisiopatología , Estudios Longitudinales , Masculino , Acidemia Propiónica/fisiopatología , Disfunción Ventricular Izquierda/tratamiento farmacológico , Disfunción Ventricular Izquierda/fisiopatología , Adulto Joven
3.
Transl Res ; 218: 43-56, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31951825

RESUMEN

Cardiac alterations (hypertrophic/dilated cardiomyopathy, and rhythm alterations) are one of the major causes of mortality and morbidity in propionic acidemia (PA), caused by the deficiency of the mitochondrial enzyme propionyl-CoA carboxylase (PCC), involved in the catabolism of branched-chain amino acids, cholesterol, and odd-chain fatty acids. Impaired mitochondrial oxidative phosphorylation has been documented in heart biopsies of PA patients, as well as in the hypomorphic Pcca-/-(A138T) mouse model, in the latter correlating with increased oxidative damage and elevated expression of cardiac dysfunction biomarkers atrial and brain natriuretic peptides (ANP and BNP) and beta-myosin heavy chain (ß-MHC). Here we characterize the cardiac phenotype in the PA mouse model by histological and echocardiography studies and identify a series of upregulated cardiac-enriched microRNAs (miRNAs) in the PA mouse heart, some of them also altered as circulating miRNAs in PA patients' plasma samples. In PA mice hearts, we show alterations in signaling pathways regulated by the identified miRNAs, which could be contributing to cardiac remodeling and dysfunction; notably, an activation of the mammalian target of rapamycin (mTOR) pathway and a decrease in autophagy, which are reverted by rapamycin treatment. In vitro studies in HL-1 cardiomyocytes indicate that propionate, the major toxic metabolite accumulating in the disease, triggers the increase in expression levels of miRNAs, BNP, and ß-MHC, concomitant with an increase in reactive oxygen species. Our results highlight miRNAs and signaling alterations in the PCC-deficient heart which may contribute to the development of PA-associated cardiomyopathy and provide a basis to identify new targets for therapeutic intervention.


Asunto(s)
Cardiomiopatías/genética , MicroARNs/genética , Acidemia Propiónica/genética , Animales , Cardiomiopatías/complicaciones , Cardiomiopatías/fisiopatología , Línea Celular , Ecocardiografía , Humanos , Ratones , Acidemia Propiónica/complicaciones , Acidemia Propiónica/fisiopatología , Transducción de Señal
4.
J Inherit Metab Dis ; 43(3): 424-437, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31828787

RESUMEN

Evidence for effectiveness of newborn screening (NBS) for propionic acidemia (PA) and isolated methylmalonic acidemia (MMA) is scarce. Prior to implementation in the Netherlands, we aim to estimate the expected health gain of NBS for PA and MMA. In this national retrospective cohort study, the clinical course of 76/83 Dutch PA and MMA patients, diagnosed between January 1979 and July 2019, was evaluated. Five clinical outcome parameters were defined: adverse outcome of the first symptomatic phase, frequency of acute metabolic decompensations (AMD), cognitive function, mitochondrial complications, and treatment-related complications. Outcomes of patients identified by family testing were compared with the outcomes of their index siblings. An adverse outcome due to the first symptomatic phase was recorded in 46% of the clinically diagnosed patients. Outcome of the first symptomatic phase was similar in 5/9 sibling pairs and better in 4/9 pairs. Based on the day of diagnosis of the clinically diagnosed patients and sibling pair analysis, a preliminary estimated reduction of adverse outcome due to the first symptomatic phase from 46% to 36%-38% was calculated. Among the sibling pairs, AMD frequency, cognitive function, mitochondrial, and treatment-related complications were comparable. These results suggest that the health gain of NBS for PA and MMA in overall outcome may be limited, as only a modest decrease of adverse outcomes due to the first symptomatic phase is expected. With current clinical practice, no reduced AMD frequency, improved cognitive function, or reduced frequency of mitochondrial or treatment-related complications can be expected.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Enfermedades Mitocondriales/complicaciones , Acidemia Propiónica/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Cognición , Femenino , Humanos , Recién Nacido , Estimación de Kaplan-Meier , Masculino , Ácido Metilmalónico , Enfermedades Mitocondriales/fisiopatología , Tamizaje Neonatal , Países Bajos , Acidemia Propiónica/fisiopatología , Acidemia Propiónica/terapia , Estudios Retrospectivos , Hermanos
5.
J Inherit Metab Dis ; 42(5): 745-761, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31119742

RESUMEN

Despite realizing increased survival rates for propionic acidemia (PA) and methylmalonic acidemia (MMA) patients, the current therapeutic regimen is inadequate for preventing or treating the devastating complications that still can occur. The elucidation of pathophysiology of these complications allows us to evaluate and rethink treatment strategies. In this review we display and discuss potential therapy targets and we give a systematic overview on current, experimental and unexplored treatment strategies in order to provide insight in what we have to offer PA and MMA patients, now and in the future. Evidence on the effectiveness of treatment strategies is often scarce, since none were tested in randomized clinical trials. This raises concerns, since even the current consensus on best practice treatment for PA and MMA is not without controversy. To attain substantial improvements in overall outcome, gene, mRNA or enzyme replacement therapy is most promising since permanent reduction of toxic metabolites allows for a less strict therapeutic regime. Hereby, both mitochondrial-associated and therapy induced complications can theoretically be prevented. However, the road from bench to bedside is long, as it is challenging to design a drug that is delivered to the mitochondria of all tissues that require enzymatic activity, including the brain, without inducing any off-target effects. To improve survival rate and quality of life of PA and MMA patients, there is a need for systematic (re-)evaluation of accepted and potential treatment strategies, so that we can better determine who will benefit when and how from which treatment strategy.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/terapia , Enfermedades Mitocondriales/terapia , Acidemia Propiónica/terapia , Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Animales , Encéfalo/metabolismo , Manejo de la Enfermedad , Humanos , Mitocondrias/metabolismo , Enfermedades Mitocondriales/fisiopatología , Acidemia Propiónica/fisiopatología
6.
J Inherit Metab Dis ; 42(5): 730-744, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31119747

RESUMEN

Over the last decades, advances in clinical care for patients suffering from propionic acidemia (PA) and isolated methylmalonic acidemia (MMA) have resulted in improved survival. These advances were possible thanks to new pathophysiological insights. However, patients may still suffer from devastating complications which largely determine the unsatisfying overall outcome. To optimize our treatment strategies, better insight in the pathophysiology of complications is needed. Here, we perform a systematic data-analysis of cohort studies and case-reports on PA and MMA. For each of the prevalent and rare complications, we summarize the current hypotheses and evidence for the underlying pathophysiology of that complication. A common hypothesis on pathophysiology of many of these complications is that mitochondrial impairment plays a major role. Assuming that complications in which mitochondrial impairment may play a role are overrepresented in monogenic mitochondrial diseases and, conversely, that complications in which mitochondrial impairment does not play a role are underrepresented in mitochondrial disease, we studied the occurrence of the complications in PA and MMA in mitochondrial and other monogenic diseases, using data provided by the Human Phenotype Ontology. Lastly, we combined this with evidence from literature to draw conclusions on the possible role of mitochondrial impairment in each complication. Altogether, this review provides a comprehensive overview on what we, to date, do and do not understand about pathophysiology of complications occurring in PA and MMA and about the role of mitochondrial impairment herein.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/complicaciones , Mitocondrias/patología , Enfermedades Mitocondriales/etiología , Acidemia Propiónica/complicaciones , Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Animales , Humanos , Enfermedades Mitocondriales/fisiopatología , Acidemia Propiónica/fisiopatología
7.
Arch Argent Pediatr ; 117(3): e288-e291, 2019 06 01.
Artículo en Español | MEDLINE | ID: mdl-31063319

RESUMEN

Propionic acidemia is an infrequent disorder with an autosomal recessive inheritance pattern caused by the deficiency of the mitochondrial enzyme propionyl-CoA carboxylase that converts propionyl-CoA to D-methylmalonyl-CoA. We present the case of a male newborn who showed signs of respiratory distress, vomiting and tiredness during feeding. He presented metabolic acidosis, positive serum and urine ketone bodies, hyperammonemia, anemia, thrombocytopenia and hypoproteinemia. The biochemical study by gas chromatography coupled to mass spectrometry in a urine sample was suggestive of propionic acidemia. The molecular study in the PCCA gene found the mutations c.893A>G (p.K298R) in the father and c.937C> T (p.R313X) in the mother. There is a need to establish the diagnosis of this infrequent entity to implement the therapeutic measures available and provide the appropriate genetic counseling.


La acidemia propiónica es un trastorno infrecuente con patrón de herencia autosómico recesivo causado por la deficiencia de la enzima mitocondrial propionil-CoA carboxilasa, que convierte el propionil-CoA a D-metilmalonil-CoA. Se expone el caso de un recién nacido masculino con signos de dificultad respiratoria, vómitos y cansancio durante la alimentación. Presentó acidosis metabólica, cuerpos cetónicos en el suero y la orina positivos, hiperamonemia, anemia, trombocitopenia e hipoproteinemia. El estudio bioquímico por cromatografía de gases acoplada a espectrometría de masas en la muestra de orina fue sugestivo de acidemia propiónica. El estudio molecular en el gen PCCA encontró las mutaciones c.893A>G (p.K298R) en el padre y c.937C>T (p.R313X) en la madre. Existe la necesidad de establecer el diagnóstico de esta entidad infrecuente para implementar las medidas terapéuticas disponibles y aportar el oportuno asesoramiento genético.


Asunto(s)
Metilmalonil-CoA Descarboxilasa/genética , Acidemia Propiónica/diagnóstico , Cromatografía de Gases y Espectrometría de Masas , Humanos , Recién Nacido , Masculino , Mutación , Acidemia Propiónica/genética , Acidemia Propiónica/fisiopatología
8.
Orphanet J Rare Dis ; 14(1): 73, 2019 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-30940196

RESUMEN

BACKGROUND: Most patients with isolated methylmalonic acidemia (MMA) /propionic acidemia (PA) presenting during the neonatal period with acute metabolic distress are at risk for death and significant neurodevelopmental disability. The nationwide newborn screening for MMA/PA has been in place in Taiwan from January, 2000 and data was collected until December, 2016. RESULTS: During the study period, 3,155,263 newborns were screened. The overall incidence of MMA mutase type cases was 1/121,356 (n = 26), 1 cobalamin B was detected and that for PA cases (n = 4) was 1/788,816. The time of referral is 8.8 days for MMA patients, and 7.5 days for PA patients. The MMA mutase type patients have higher AST, ALT, and NH3 values as well as a lower pH value (p < 0.05). The mean age for liver transplantation (LT) is 402 days (range from 0.6-6.7 yr) with 16 out of 20 cases (80.0%) using living donors. The mean admission length shortened from 90.6 days/year (pre-LT) to 5.3 days/year (at 3rd year post-LT) (p < 0.0005). Similarly, the tube feeding ratio decreased from 67.8 to 0.50% (p < 0.00005). The anxiety level of the caregiver was reduced from 33.4 to 27.2 after LT (p = 0.001) and the DQ/IQ performance of the patients was improved after LT from 50 to 60.1 (p = 0.07). CONCLUSION: MMA/PA patients with LT do survive and have reduced admission time, reduced tube feeding and the caregiver is less anxious.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Trasplante de Hígado/normas , Acidemia Propiónica/fisiopatología , Acidemia Propiónica/terapia , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/mortalidad , Cuidadores/psicología , Cuidadores/estadística & datos numéricos , Nutrición Enteral/estadística & datos numéricos , Femenino , Hospitalización/estadística & datos numéricos , Humanos , Recién Nacido , Trasplante de Hígado/mortalidad , Masculino , Mutación , Tamizaje Neonatal , Acidemia Propiónica/genética , Acidemia Propiónica/mortalidad , Taiwán , Resultado del Tratamiento
9.
Clin Biochem ; 66: 100-102, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30753843

RESUMEN

OBJECTIVES: Elevated levels of metabolites such as ammonia and propionylcarnitine in propionic acidemia (PA) lead to an increased reactive oxygen species (ROS) production which could activate and stabilize the epigenetic regulated hypoxia-inducible factor-1α (HIF-1α). In order to evaluate the DNA methylation status of the HIF-1α binding site in PA, we investigated the antioxidant gluthatione peroxidase 3 gene (GPX3) promoter region. DESIGN AND METHODS: Using leukocyte DNA extracted from bloodspots collected 2-4 days after birth from diet free newborns, the cytosine phosphodiester bond guanine (CpG) dinucleotides of a HIF-1α binding site (CGTTTTTTACG) in the promoter region of GPX3 was retrospectively analysed. Patients included 7 PA. and 7 healthy controls (KO) respectively. RESULTS: A demethylated TGTTTTTTATG allele was detected in 3 PA patients with blood ammonia (NH3) concentrations of 500, 595, and 987 umol/L respectively; a demethylated/partial methylated TGTTTTTTAC/TG allele in 4 PA patients (2 PA with blood NH3 = 213, 271 umol/L respectively); a partial methylated C/TGTTTTTTAC/TG allele in 5 healthy controls respectively; a partial methylated/methylated C/TGTTTTTTACG allele in 2 healthy controls. CONCLUSION: Our results suggest that at excess NH3, the DNA methylation status of the HIF-1α binding site of GPX3 in newborns with PA is demethylated (TGTTTTTTATG allele). However, the demethylated allele has to be confirmed as a statistically significant change in more patients.


Asunto(s)
Amoníaco/metabolismo , Metilación de ADN/fisiología , Glutatión Peroxidasa/genética , Hiperamonemia/fisiopatología , Acidemia Propiónica/fisiopatología , Sitios de Unión/fisiología , Desmetilación , Humanos , Recién Nacido , Regiones Promotoras Genéticas/fisiología
10.
J Med Case Rep ; 12(1): 103, 2018 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-29679984

RESUMEN

BACKGROUND: Propionic acidemia is a rare autosomal recessive inherited metabolic disorder that can inhibit the synthesis of N-acetylglutamate, the obligatory activator in urea synthesis, leading to hyperammonemia. N-carbamylglutamate ameliorates hyperammonemia in decompensated propionic acidemia. The effects of long-term continuous N-acetylglutamate administration in such patients are unknown. We report our clinical experience with continuous administration of N-acetylglutamate for 6 years in a patient with propionic acidemia frequently presenting with hyperammonemia. CASE PRESENTATION: A male Caucasian patient with frequently decompensated propionic acidemia and hyperammonemia was admitted 78 times for acute attacks during the first 9 years of his life. Continuous daily treatment with oral N-carbamylglutamate 100 mg/kg (50 mg/kg after 6 months) was initiated. During 6 years of treatment, he had a significant decrease in his mean plasma ammonia levels (75.7 µmol/L vs. 140.3 µmol/L before N-carbamylglutamate therapy, p < 0.005 [normal range 50-80 µmol/L]) and fewer acute episodes (two in 6 years). CONCLUSION: Our results suggest a benefit of N-acetylglutamate administration outside the emergency setting. If this observation is confirmed, future studies should aim to optimize the dosage and explore effects of the dosage requirements on other drugs and on protein tolerance.


Asunto(s)
Glutamatos/administración & dosificación , Hiperamonemia/sangre , Acidemia Propiónica/tratamiento farmacológico , Administración Oral , Adolescente , N-Acetiltransferasa de Aminoácidos/sangre , N-Acetiltransferasa de Aminoácidos/efectos de los fármacos , Biomarcadores/sangre , Enfermedad Crónica , Discapacidades del Desarrollo/complicaciones , Relación Dosis-Respuesta a Droga , Humanos , Hiperamonemia/etiología , Masculino , Acidemia Propiónica/dietoterapia , Acidemia Propiónica/fisiopatología , Trastornos Innatos del Ciclo de la Urea/sangre
11.
Biochem J ; 475(4): 749-758, 2018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29339464

RESUMEN

Propionic acidemia is the accumulation of propionate in blood due to dysfunction of propionyl-CoA carboxylase. The condition causes lethargy and striatal degeneration with motor impairment in humans. How propionate exerts its toxic effect is unclear. Here, we show that intravenous administration of propionate causes dose-dependent propionate accumulation in the brain and transient lethargy in mice. Propionate, an inhibitor of histone deacetylase, entered GABAergic neurons, as could be seen from increased neuronal histone H4 acetylation in the striatum and neocortex. Propionate caused an increase in GABA (γ-amino butyric acid) levels in the brain, suggesting inhibition of GABA breakdown. In vitro propionate inhibited GABA transaminase with a Ki of ∼1 mmol/l. In isolated nerve endings, propionate caused increased release of GABA to the extracellular fluid. In vivo, propionate reduced cerebral glucose metabolism in both striatum and neocortex. We conclude that propionate-induced inhibition of GABA transaminase causes accumulation of GABA in the brain, leading to increased extracellular GABA concentration, which inhibits neuronal activity and causes lethargy. Propionate-mediated inhibition of neuronal GABA transaminase, an enzyme of the inner mitochondrial membrane, indicates entry of propionate into neuronal mitochondria. However, previous work has shown that neurons are unable to metabolize propionate oxidatively, leading us to conclude that propionyl-CoA synthetase is probably absent from neuronal mitochondria. Propionate-induced inhibition of energy metabolism in GABAergic neurons may render the striatum, in which >90% of the neurons are GABAergic, particularly vulnerable to degeneration in propionic acidemia.


Asunto(s)
4-Aminobutirato Transaminasa/antagonistas & inhibidores , Neuronas GABAérgicas/efectos de los fármacos , Letargia/metabolismo , Propionatos/administración & dosificación , Acidemia Propiónica/metabolismo , 4-Aminobutirato Transaminasa/metabolismo , Animales , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Neuronas GABAérgicas/metabolismo , Glucosa/metabolismo , Inhibidores de Histona Desacetilasas/administración & dosificación , Histona Desacetilasas , Humanos , Letargia/inducido químicamente , Letargia/fisiopatología , Metilmalonil-CoA Descarboxilasa/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neocórtex/efectos de los fármacos , Neocórtex/metabolismo , Neocórtex/patología , Acidemia Propiónica/inducido químicamente , Acidemia Propiónica/fisiopatología , Ácido gamma-Aminobutírico/metabolismo
12.
Mol Genet Metab ; 122(4): 145-152, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29033250

RESUMEN

Propionyl-CoA carboxylase (PCC) is the enzyme which catalyzes the carboxylation of propionyl-CoA to methylmalonyl-CoA and is encoded by the genes PCCA and PCCB to form a hetero-dodecamer. Dysfunction of PCC leads to the inherited metabolic disorder propionic acidemia, which can result in an affected individual presenting with metabolic acidosis, hyperammonemia, lethargy, vomiting and sometimes coma and death if not treated. Individuals with propionic acidemia also have a number of long term complications resulting from the dysfunction of the PCC enzyme. Here we present an overview of the current knowledge about the structure and function of PCC. We review an updated list of human variants which are published and provide an overview of the disease.


Asunto(s)
Ligasas de Carbono-Carbono/química , Ligasas de Carbono-Carbono/metabolismo , Acidemia Propiónica/enzimología , Ligasas de Carbono-Carbono/genética , Humanos , Hiperamonemia/complicaciones , Ácido Láctico/análogos & derivados , Ácido Láctico/metabolismo , Metilmalonil-CoA Descarboxilasa/genética , Mutación , Acidemia Propiónica/complicaciones , Acidemia Propiónica/genética , Acidemia Propiónica/fisiopatología , Relación Estructura-Actividad
13.
Mol Genet Metab ; 122(1-2): 43-50, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28774709

RESUMEN

Oxidative stress contributes to the pathogenesis of propionic acidemia (PA), a life threatening disease caused by the deficiency of propionyl CoA-carboxylase, in the catabolic pathway of branched-chain amino acids, odd-number chain fatty acids and cholesterol. Patients develop multisystemic complications including seizures, extrapyramidal symptoms, basal ganglia deterioration, pancreatitis and cardiomyopathy. The accumulation of toxic metabolites results in mitochondrial dysfunction, increased reactive oxygen species and oxidative damage, all of which have been documented in patients' samples and in a hypomorphic mouse model. Here we set out to investigate whether treatment with a mitochondria-targeted antioxidant, MitoQ, or with the natural polyphenol resveratrol, which is reported to have antioxidant and mitochondrial activation properties, could ameliorate the altered redox status and its functional consequences in the PA mouse model. The results show that oral treatment with MitoQ or resveratrol decreases lipid peroxidation and the expression levels of DNA repair enzyme OGG1 in PA mouse liver, as well as inducing tissue-specific changes in the expression of antioxidant enzymes. Notably, treatment decreased the cardiac hypertrophy marker BNP that is found upregulated in the PA mouse heart. Overall, the results provide in vivo evidence to justify more in depth investigations of antioxidants as adjuvant therapy in PA.


Asunto(s)
Antioxidantes/uso terapéutico , Compuestos Organofosforados/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Acidemia Propiónica/tratamiento farmacológico , Estilbenos/uso terapéutico , Ubiquinona/análogos & derivados , Administración Oral , Aminoácidos de Cadena Ramificada , Animales , Antioxidantes/administración & dosificación , Modelos Animales de Enfermedad , Corazón/efectos de los fármacos , Humanos , Peroxidación de Lípido/efectos de los fármacos , Ratones , Compuestos Organofosforados/administración & dosificación , Acidemia Propiónica/fisiopatología , Resveratrol , Estilbenos/administración & dosificación , Ubiquinona/administración & dosificación , Ubiquinona/uso terapéutico
14.
Mol Genet Metab ; 122(1-2): 51-59, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28712602

RESUMEN

BACKGROUND: Propionic acidemia is a rare metabolic disorder caused by a deficiency of propionyl- CoA carboxylase, the enzyme converting propionyl-CoA to methylmalonyl-CoA that subsequently enters the citric acid cycle as succinyl-CoA. Patients with propionic acidemia cannot metabolize propionic acid, which combines with oxaloacetate to form methylcitric acid. This, with the defective supply of succinyl-CoA, may lead to a deficiency in citric acid cycle intermediates. PURPOSE: The objective of this study was to determine whether supplements with glutamine (400mg/kg per day), citrate (7.5mEq/kg per day), or ornithine α-ketoglutarate (400mg/kg per day) (anaplerotic agents that could fill up the citric acid cycle) would affect plasma levels of glutamine and ammonia, the urinary excretion of Krebs cycle intermediates, and the clinical outcome in 3 patients with propionic acidemia. METHODS: Each supplement was administered daily for four weeks with a two week washout period between supplements. The supplement that produced the most favorable changes was supplemented for 30 weeks following the initial study period and then for a 2 year extension. RESULTS: The urinary excretion of the Krebs cycle intermediates, α-ketoglutarate, succinate, and fumarate increased significantly compared to baseline during citrate supplementation, but not with the other two supplements. For this reason, citrate supplements were continued in the second part of the study. The urinary excretion of methylcitric acid and 3-hydroxypropionic acid did not change with any intervention. No significant changes in ammonia or glutamine levels were observed with any supplement. However, supplementation with any anaplerotic agents normalized the physiological buffering of ammonia by glutamate, with plasma glutamate and alanine levels significantly increasing, rather than decreasing with increasing ammonia levels. No significant side effects were observed with any therapy and safety labs (blood counts, chemistry and thyroid profile) remained unchanged. Motor and cognitive development was severely delayed before the trial and did not change significantly with therapy. Hospitalizations per year did not change during the trial period, but decreased significantly (p<0.05) in the 2years following the study (when citrate was continued) compared to the 2years before and during the study. CONCLUSIONS: These results indicate that citrate entered the Krebs cycle providing successful anaplerotic therapy by increasing levels of the downstream intermediates of the Krebs cycle: α-ketoglutarate, succinate and fumarate. Citrate supplements were safe and might have contributed to reduce hospitalizations in patients with propionic acidemia.


Asunto(s)
Ciclo del Ácido Cítrico/efectos de los fármacos , Ácido Cítrico/administración & dosificación , Suplementos Dietéticos , Glutamina/administración & dosificación , Ornitina/análogos & derivados , Acidemia Propiónica/dietoterapia , Aminoácidos/sangre , Amoníaco/sangre , Ligasas de Carbono-Carbono/metabolismo , Niño , Preescolar , Citratos/orina , Ácido Cítrico/efectos adversos , Suplementos Dietéticos/efectos adversos , Femenino , Glutamina/efectos adversos , Glutamina/sangre , Humanos , Ácido Láctico/análogos & derivados , Ácido Láctico/orina , Masculino , Ornitina/administración & dosificación , Acidemia Propiónica/metabolismo , Acidemia Propiónica/fisiopatología , Resultado del Tratamiento
15.
Sci Rep ; 7(1): 5727, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28720782

RESUMEN

miRNome expression profiling was performed in a mouse model of propionic acidemia (PA) and in patients' plasma samples to investigate the role of miRNAs in the pathophysiology of the disease and to identify novel biomarkers and therapeutic targets. PA is a potentially lethal neurometabolic disease with patients developing neurological deficits and cardiomyopathy in the long-term, among other complications. In the PA mouse liver we identified 14 significantly dysregulated miRNAs. Three selected miRNAs, miR-34a-5p, miR-338-3p and miR-350, were found upregulated in brain and heart tissues. Predicted targets involved in apoptosis, stress-signaling and mitochondrial function, were inversely found down-regulated. Functional analysis with miRNA mimics in cellular models confirmed these findings. miRNA profiling in plasma samples from neonatal PA patients and age-matched control individuals identified a set of differentially expressed miRNAs, several were coincident with those identified in the PA mouse, among them miR-34a-5p and miR-338-3p. These two miRNAs were also found dysregulated in childhood and adult PA patients' cohorts. Taken together, the results reveal miRNA signatures in PA useful to identify potential biomarkers, to refine the understanding of the molecular mechanisms of this rare disease and, eventually, to improve the management of patients.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/análisis , MicroARNs/sangre , Acidemia Propiónica/patología , Acidemia Propiónica/fisiopatología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Recién Nacido , Hígado/patología , Ratones , Miocardio/patología , Plasma/química
16.
Mol Genet Metab ; 119(4): 317-321, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27825584

RESUMEN

Certain inborn errors of metabolism have been suggested to increase the risk of autistic behavior. In an animal model, propionic acid ingestion triggered abnormal behavior resembling autism. So far only a few cases were reported with propionic acidemia and autistic features. From a series of twelve consecutively diagnosed cases with propionic acidemia, we report on eight patients with autistic features. The patients were followed 2-4 times a year and underwent regular clinical, dietary and laboratory investigations. Psychological evaluation was performed every second to fourth year. All patients were compliant with the standard diet and carnitine supplementation. None of the patients had frequent metabolic decompensations. From the metabolic factors known to impact neuropsychological outcome we detected chronically decreased valine levels and altered valine to leucine ratios in five out of the eight patients. Recurrent lactic acid elevations were present in six out of the eight patients. Five of the eight patients were diagnosed with Autism Spectrum Disorder, four of them had pathogenic variants in PCCB. Disorder according to DSM-IV and/or DSM-5 criteria. One of the patients diagnosed with propionic acidemia by newborn screening had the most significant behavioral features and another was diagnosed with Autism Spectrum Disorder prior to propionic acidemia. We hypothesize that chronic suboptimal intracellular metabolic balance may be responsible for the increased risk for autistic features in propionic acidemia. We propose that patients diagnosed with propionic acidemia should be screened for Autism Spectrum Disorder.


Asunto(s)
Trastorno del Espectro Autista/genética , Metilmalonil-CoA Descarboxilasa/genética , Tamizaje Neonatal , Acidemia Propiónica/genética , Adolescente , Adulto , Animales , Trastorno del Espectro Autista/complicaciones , Trastorno del Espectro Autista/diagnóstico , Trastorno del Espectro Autista/fisiopatología , Niño , Femenino , Humanos , Recién Nacido , Leucina/metabolismo , Masculino , Propionatos/metabolismo , Acidemia Propiónica/complicaciones , Acidemia Propiónica/diagnóstico , Acidemia Propiónica/fisiopatología , Adulto Joven
17.
Transl Psychiatry ; 6(10): e927, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27779624

RESUMEN

Propionic acid (PPA) is a ubiquitous short-chain fatty acid, which is a major fermentation product of the enteric microbiome. PPA is a normal intermediate of metabolism and is found in foods, either naturally or as a preservative. PPA and its derivatives have been implicated in both health and disease. Whereas PPA is an energy substrate and has many proposed beneficial effects, it is also associated with human disorders involving mitochondrial dysfunction, including propionic acidemia and autism spectrum disorders (ASDs). We aimed to investigate the dichotomy between the health and disease effects of PPA by measuring mitochondrial function in ASD and age- and gender-matched control lymphoblastoid cell lines (LCLs) following incubation with PPA at several concentrations and durations both with and without an in vitro increase in reactive oxygen species (ROS). Mitochondrial function was optimally increased at particular exposure durations and concentrations of PPA with ASD LCLs, demonstrating a greater enhancement. In contrast, increasing ROS negated the positive PPA effect with the ASD LCLs, showing a greater detriment. These data demonstrate that enteric microbiome metabolites such as PPA can have both beneficial and toxic effects on mitochondrial function, depending on concentration, exposure duration and microenvironment redox state with these effects amplified in LCLs derived from individuals with ASD. As PPA, as well as enteric bacteria, which produce PPA, have been implicated in a wide variety of diseases, including ASD, diabetes, obesity and inflammatory diseases, insight into this metabolic modulator from the host microbiome may have wide applications for both health and disease.


Asunto(s)
Trastorno del Espectro Autista/fisiopatología , Microbioma Gastrointestinal/fisiología , Mitocondrias/fisiología , Enfermedades Mitocondriales/fisiopatología , Propionatos/metabolismo , Acidemia Propiónica/fisiopatología , Estudios de Casos y Controles , Línea Celular , Niño , Humanos , Masculino
19.
J Pediatr Endocrinol Metab ; 29(7): 863-6, 2016 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-27089410

RESUMEN

Although propionic acidemia (PA) is a devastating inherited metabolic disease, with early diagnosis and advanced treatment strategies prognosis gets better and girls can survive to child-bearing ages. We share the detailed follow-up data of the pregnancy, successful labor with cesarean section and a healthy baby of a Turkish patient with PA.


Asunto(s)
Lactancia , Metilmalonil-CoA Descarboxilasa/genética , Mutación , Complicaciones del Embarazo/genética , Acidemia Propiónica/genética , Adulto , Cesárea , Desarrollo Infantil , Dieta Vegetariana , Femenino , Homocigoto , Humanos , Recién Nacido de Bajo Peso , Recién Nacido , Embarazo , Complicaciones del Embarazo/dietoterapia , Complicaciones del Embarazo/fisiopatología , Acidemia Propiónica/dietoterapia , Acidemia Propiónica/fisiopatología , Nacimiento a Término , Turquía
20.
Heart Rhythm ; 13(6): 1335-45, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26854997

RESUMEN

BACKGROUND: Propionic acidemia (PROP) is a rare metabolic disorder caused by deficiency of propionyl-CoA carboxylase. PROP patients demonstrate QT prolongations associated with ventricular tachycardia and syncopes. Mechanisms responsible for this acquired long QT syndrome (acqLQTS) are unknown. OBJECTIVE: The aim of the study was to investigate acute and chronic effects of metabolites accumulating in PROP patients on major repolarizing potassium currents (IKs and IKr) and their channel subunits. METHODS: Voltage clamp studies were performed in CHO-KCNQ1/KCNE1 or HEK-KCNH2 cells to determine effects of propionic acid (PA; 1-10 mM), propionylcarnitine (PC; 25 µM-10 mM), methylcitrate (MC; 25 µM-10 mM), 0.2 M phosphate buffer (PB), or patient serum on IKs and IKr currents. Metabolite effects on action potentials were recorded in current clamp mode in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM). Protein expression of α- and ß-subunits of IKs (KCNQ1/KCNE1) and IKr (KCNH2) was evaluated with Western blots. RESULTS: Acute application of PA, PC, MC, and patient serum had no direct effect on net IKr densities (and KCNH2 expression), although it changed IKr gating kinetics. In contrast, PA, PC, MC, and patient serum all reduced IKs-tail (-67% ± 4.2%, -27% ± 6.7%, -16% ± 6.3%, -42.8% ± 5.15; P < .001) and IKs-end pulse currents. PA significantly prolonged action potential duration (APD) in hiPSC-CM and QT interval in wild-type but not in LQT1 rabbits lacking IKs. Moreover, PC and MC (1 mM) decreased KCNQ1 protein expression (relative density: 0.58 ± 0.08 and 0.16 ± 0.05; P < .01). Chronic exposure to 10 mM PA, in contrast, increased KCNQ1 5.4-fold (P < .001) owing to decreased protein degradation. CONCLUSION: Acute reduction of IKs by PROP metabolites may be responsible for APD prolongation and acqLQTS observed in PROP patients.


Asunto(s)
Metilmalonil-CoA Descarboxilasa/metabolismo , Acidemia Propiónica , Animales , Sistema de Conducción Cardíaco/metabolismo , Sistema de Conducción Cardíaco/fisiopatología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/etiología , Síndrome de QT Prolongado/fisiopatología , Canales de Potasio con Entrada de Voltaje/metabolismo , Acidemia Propiónica/complicaciones , Acidemia Propiónica/metabolismo , Acidemia Propiónica/fisiopatología , Conejos , Taquicardia Ventricular/etiología , Taquicardia Ventricular/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...