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

Métodos Terapéuticos y Terapias MTCI
Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Orphanet J Rare Dis ; 19(1): 17, 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38238766

RESUMEN

Aromatic L-amino acid decarboxylase (AADC) deficiency is a rare genetic disorder of monoamine neurotransmitter synthesis that presents with a range of symptoms, including motor dysfunction and limited attainment of developmental motor milestones. The approval of eladocagene exuparvovec, a gene therapy for AADC deficiency with demonstrated efficacy for motor improvements, now expands the range of motor outcomes possible for patients with this disorder. However, recommendations and guidelines for therapy following treatment with gene therapy are lacking. To ensure patients can reach their full potential following treatment with gene therapy, it is essential they receive rehabilitation therapies designed specifically with their impairments and goals in mind. Therefore, we highlight specific rehabilitative needs of patients following gene therapy and propose a set of recommendations for the post-treatment period based on collective experiences of therapists, physicians, and caregivers treating and caring for patients with AADC deficiency who have been treated with gene therapy. These recommendations include a focus on periods of intensive therapy, facilitating active movements, training for functional abilities, cognitive and communication training, parent/caregiver empowerment, collaboration between therapists and caregivers to develop in-home programs, and the incorporation of supplemental forms of therapy that patients and their families may find more enjoyable and engaging. Many of these rehabilitative strategies may be employed prior to gene therapy. However, these recommendations will be valuable for therapists, caregivers, and wider treatment teams as they prepare for the post-treatment journey with these patients. Furthermore, the considerations and recommendations presented here may prove beneficial outside the AADC deficiency community as gene therapies and other treatments are developed and approved for other rare diseases.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos , Humanos , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Descarboxilasas de Aminoácido-L-Aromático/genética , Terapia Genética , Aminoácidos
2.
Nutrients ; 15(3)2023 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-36771238

RESUMEN

3-Hydroxy-3-Methylglutaryl-CoA Lyase (HMGCL) deficiency can be a very severe disorder that typically presents with acute metabolic decompensation with features of hypoketotic hypoglycemia, hyperammonemia, and metabolic acidosis. A retrospective chart and literature review of Australian patients over their lifespan, incorporating acute and long-term dietary management, was performed. Data from 10 patients contributed to this study. The index case of this disorder was lost to follow-up, but there is 100% survival in the remainder of the cases despite several having experienced life-threatening episodes. In the acute setting, five of nine patients have used 900 mg/kg/day of sodium D,L 3-hydroxybutyrate in combination with intravenous dextrose-containing fluids (delivering glucose above estimated basal utilization requirements). All patients have been on long-term protein restriction, and those diagnosed more recently have had additional fat restriction. Most patients take L-carnitine. Three children and none of the adults take nocturnal uncooked cornstarch. Of the cohort, there were two patients that presented atypically-one with fulminant liver failure and the other with isolated developmental delay. Dietary management in patients with HMGCL deficiency is well tolerated, and rapid institution of acute supportive metabolic treatment is imperative to optimizing survival and improve outcomes in this disorder.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos , Hiperamonemia , Niño , Adulto , Humanos , Estudios Retrospectivos , Australia , Errores Innatos del Metabolismo de los Aminoácidos/terapia
3.
J Inherit Metab Dis ; 46(3): 482-519, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36221165

RESUMEN

Glutaric aciduria type 1 is a rare inherited neurometabolic disorder of lysine metabolism caused by pathogenic gene variations in GCDH (cytogenic location: 19p13.13), resulting in deficiency of mitochondrial glutaryl-CoA dehydrogenase (GCDH) and, consequently, accumulation of glutaric acid, 3-hydroxyglutaric acid, glutaconic acid and glutarylcarnitine detectable by gas chromatography/mass spectrometry (organic acids) and tandem mass spectrometry (acylcarnitines). Depending on residual GCDH activity, biochemical high and low excreting phenotypes have been defined. Most untreated individuals present with acute onset of striatal damage before age 3 (to 6) years, precipitated by infectious diseases, fever or surgery, resulting in irreversible, mostly dystonic movement disorder with limited life expectancy. In some patients, striatal damage develops insidiously. In recent years, the clinical phenotype has been extended by the finding of extrastriatal abnormalities and cognitive dysfunction, preferably in the high excreter group, as well as chronic kidney failure. Newborn screening is the prerequisite for pre-symptomatic start of metabolic treatment with low lysine diet, carnitine supplementation and intensified emergency treatment during catabolic episodes, which, in combination, have substantially improved neurologic outcome. In contrast, start of treatment after onset of symptoms cannot reverse existing motor dysfunction caused by striatal damage. Dietary treatment can be relaxed after the vulnerable period for striatal damage, that is, age 6 years. However, impact of dietary relaxation on long-term outcomes is still unclear. This third revision of evidence-based recommendations aims to re-evaluate previous recommendations (Boy et al., J Inherit Metab Dis, 2017;40(1):75-101; Kolker et al., J Inherit Metab Dis 2011;34(3):677-694; Kolker et al., J Inherit Metab Dis, 2007;30(1):5-22) and to implement new research findings on the evolving phenotypic diversity as well as the impact of non-interventional variables and treatment quality on clinical outcomes.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos , Encefalopatías Metabólicas , Humanos , Glutaril-CoA Deshidrogenasa , Lisina/metabolismo , Encefalopatías Metabólicas/diagnóstico , Encefalopatías Metabólicas/genética , Encefalopatías Metabólicas/terapia , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Glutaratos/metabolismo
4.
J Inherit Metab Dis ; 45(5): 937-951, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35618652

RESUMEN

OBJECTIVES: To present the very long-term follow up of patients with cobalamin A (cblA) deficiency. METHODS: A retrospective case series of adult (>16 years) patients with molecular or enzymatic diagnosis of cblA deficiency. RESULTS: We included 23 patients (mean age: 27 ± 7.6 years; mean follow-up: 24.9 ± 7.6 years). Disease onset was mostly pediatric (78% < 1 year, median = 4 months) with acute neurologic deterioration (65%). Eight patients presented with chronic symptoms, and one had an adult-onset mild cblA deficiency. Most of the patients (61%) were initially classified as vitamin B12-unresponsive methylmalonic aciduria (MMA); in vitro B12 responsiveness was subsequently found in all the tested patients (n = 13). Initial management consisted of protein restriction (57%), B12 (17%), or both (26%). The main long-term problems were intellectual disability (39%) and renal failure (30%). However, 56.5% of the patients were living independently. Intellectual disability was equally distributed among the initial treatment groups, while renal failure (moderate and beginning at the age of 38 years) was present in only one out of seven patients initially treated with B12. CONCLUSIONS: We provide a detailed picture of the long-term outcome of a series of adult cblA patients, mostly diagnosed before the enzymatic and molecular era. We confirm that about 35% of the patients do not present acutely, underlining the importance of measuring MMA in any case of unexplained chronic renal failure, intellectual disability, or growth delay. In addition, we describe a patient with a milder adult-onset form. Early B12 supplementation seems to protect from severe renal insufficiency.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos , Discapacidad Intelectual , Fallo Renal Crónico , Deficiencia de Vitamina B 12 , Adulto , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Niño , Humanos , Ácido Metilmalónico , Estudios Retrospectivos , Vitamina B 12/uso terapéutico , Deficiencia de Vitamina B 12/complicaciones , Deficiencia de Vitamina B 12/diagnóstico , Deficiencia de Vitamina B 12/tratamiento farmacológico , Adulto Joven
5.
J Nutr ; 150(Suppl 1): 2556S-2560S, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33000154

RESUMEN

Lysine is an essential amino acid, and inherited diseases of its metabolism therefore represent defects of lysine catabolism. Although some of these enzyme defects are not well described yet, glutaric aciduria type I (GA1) and antiquitin (2-aminoadipic-6-semialdehyde dehydrogenase) deficiency represent the most well-characterized diseases. GA1 is an autosomal recessive disorder due to a deficiency of glutaryl-CoA dehydrogenase. Untreated patients exhibit early onset macrocephaly and may present a neurological deterioration with regression and movement disorder at the time of a presumably "benign" infection most often during the first year of life. This is associated with a characteristic neuroimaging pattern with frontotemporal atrophy and striatal injuries. Diagnosis relies on the identification of glutaric and 3-hydroxyglutaric acid in urine along with plasma glutarylcarnitine. Treatment consists of a low-lysine diet aiming at reducing the putatively neurotoxic glutaric and 3-hydroxyglutaric acids. Additional therapeutic measures include administration of l-carnitine associated with emergency measures at the time of intercurrent illnesses aiming at preventing brain injury. Early treated (ideally through newborn screening) patients exhibit a favorable long-term neurocognitive outcome, whereas late-treated or untreated patients may present severe neurocognitive irreversible disabilities. Antiquitin deficiency is the most common form of pyridoxine-dependent epilepsy. α-Aminoadipic acid semialdehyde (AASA) and Δ-1-piperideine-6-carboxylate (P6C) accumulate proximal to the enzymatic block. P6C forms a complex with pyridoxal phosphate (PLP), a key vitamer of pyridoxine, thereby reducing PLP bioavailability and subsequently causing epilepsy. Urinary AASA is a biomarker of antiquitin deficiency. Despite seizure control, only 25% of the pyridoxine-treated patients show normal neurodevelopment. Low-lysine diet and arginine supplementation are proposed in some patients with decrease of AASA, but the impact on neurodevelopment is unclear. In summary, GA1 and antiquitin deficiency are the 2 main human defects of lysine catabolism. Both include neurological impairment. Lysine dietary restriction is a key therapy for GA1, whereas its benefits in antiquitin deficiency appear less clear.


Asunto(s)
Aldehído Deshidrogenasa/deficiencia , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Encefalopatías Metabólicas Innatas/metabolismo , Encefalopatías Metabólicas/metabolismo , Encéfalo/metabolismo , Epilepsia/metabolismo , Glutaril-CoA Deshidrogenasa/deficiencia , Lisina/metabolismo , Ácido 2-Aminoadípico/análogos & derivados , Ácido 2-Aminoadípico/metabolismo , Aldehído Deshidrogenasa/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Arginina/uso terapéutico , Encéfalo/patología , Encefalopatías Metabólicas/terapia , Encefalopatías Metabólicas Innatas/terapia , Carnitina/análogos & derivados , Carnitina/metabolismo , Carnitina/uso terapéutico , Epilepsia/terapia , Glutaratos/metabolismo , Glutaril-CoA Deshidrogenasa/metabolismo , Humanos , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/terapia , Fosfato de Piridoxal/metabolismo , Piridoxina/metabolismo , Piridoxina/uso terapéutico
6.
Nutrients ; 12(10)2020 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-33003354

RESUMEN

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


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/terapia , Dieta con Restricción de Proteínas/métodos , Proteínas en la Dieta/administración & dosificación , Trasplante de Hígado , Terapia Nutricional/métodos , Adolescente , Adulto , Errores Innatos del Metabolismo de los Aminoácidos/sangre , Carnitina/sangre , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Ácido Metilmalónico/sangre , Cuidados Posoperatorios , Periodo Posoperatorio , Periodo Preoperatorio , Estudios Retrospectivos , Adulto Joven
7.
Nutrients ; 12(2)2020 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-32069872

RESUMEN

BACKGROUND: Methylmalonic acidemia (MMA) is an autosomal recessive disorder treated with precursor-free medical food while limiting natural protein. This retrospective chart review was to determine if there was a relationship between medical food, valine (VAL) and/or isoleucine (ILE) supplementation, total protein intake, and plasma amino acid profiles. Methods: A chart review, of patients aged 31 days or older with MMA treated with dietary intervention and supplementation of VAL and/or ILE and followed at the Children's Hospital Colorado Inherited Metabolic Diseases Clinic. Dietary prescriptions and plasma amino acid concentrations were obtained at multiple time points. RESULTS: Baseline mean total protein intake for five patients was 198% of Recommended Dietary Allowance (RDA) with 107% natural protein and 91% medical food. Following intervention, total protein intake (p = 0.0357), protein from medical food (p = 0.0142), and leucine (LEU) from medical food (p = 0.0276) were lower, with no significant change in natural protein intake (p = 0.2036). At baseline, 80% of patients received VAL supplementation and 100% received ILE supplementation. After intervention, only one of the cohort remained on supplementation. There was no statistically significant difference in plasma propiogenic amino acid concentrations. CONCLUSIONS: Decreased intake of LEU from medical food allowed for discontinuation of amino acid supplementation, while meeting the RDA for protein.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/terapia , Proteínas en la Dieta/administración & dosificación , Suplementos Dietéticos , Isoleucina/administración & dosificación , Valina/administración & dosificación , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Nutrición Enteral/métodos , Femenino , Humanos , Lactante , Masculino , Estudios Retrospectivos , Resultado del Tratamiento
8.
J Cell Mol Med ; 22(3): 1601-1613, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29265583

RESUMEN

Methylmalonic aciduria (MMA) is a disorder of organic acid metabolism resulting from a functional defect of the mitochondrial enzyme, methylmalonyl-CoA mutase (MCM). The main treatments for MMA patients are dietary restriction of propiogenic amino acids and carnitine supplementation. Liver or combined liver/kidney transplantation has been used to treat those with the most severe clinical manifestations. Thus, therapies are necessary to help improve quality of life and prevent liver, renal and neurological complications. Previously, we successfully used the TAT-MTS-Protein approach for replacing a number of mitochondrial-mutated proteins. In this targeted system, TAT, an 11 a.a peptide, which rapidly and efficiently can cross biological membranes, is fused to a mitochondrial targeting sequence (MTS), followed by the mitochondrial mature protein which sends the protein into the mitochondria. In the mitochondria, the TAT-MTS is cleaved off and the native protein integrates into its natural complexes and is fully functional. In this study, we used heterologous MTSs of human, nuclear-encoded mitochondrial proteins, to target the human MCM protein into the mitochondria. All fusion proteins reached the mitochondria and successfully underwent processing. Treatment of MMA patient fibroblasts with these fusion proteins restored mitochondrial activity such as ATP production, mitochondrial membrane potential and oxygen consumption, indicating the importance of mitochondrial function in this disease. Treatment with the fusion proteins enhanced cell viability and most importantly reduced MMA levels. Treatment also enhanced albumin and urea secretion in a CRISPR/Cas9-engineered HepG2 MUT (-/-) liver cell line. Therefore, we suggest using this TAT-MTS-Protein approach for the treatment of MMA.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Fibroblastos/enzimología , Productos del Gen tat/genética , Metilmalonil-CoA Mutasa/genética , Mitocondrias/enzimología , Proteínas Recombinantes de Fusión/genética , Errores Innatos del Metabolismo de los Aminoácidos/enzimología , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/patología , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Sistemas CRISPR-Cas , Escherichia coli/genética , Escherichia coli/metabolismo , Fibroblastos/patología , Expresión Génica , Productos del Gen tat/metabolismo , Terapia Genética/métodos , Células Hep G2 , Humanos , Hígado/enzimología , Hígado/patología , Potencial de la Membrana Mitocondrial , Ácido Metilmalónico/metabolismo , Metilmalonil-CoA Mutasa/metabolismo , Mitocondrias/patología , Enfermedades Mitocondriales/enzimología , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Enfermedades Mitocondriales/terapia , Plásmidos/química , Plásmidos/metabolismo , Cultivo Primario de Células , Ingeniería de Proteínas/métodos , Señales de Clasificación de Proteína/genética , Proteínas Recombinantes de Fusión/metabolismo , Transfección
9.
Liver Transpl ; 21(9): 1208-18, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25990417

RESUMEN

Propionic acidemia (PA) and classical methylmalonic acidemia (MMA) are rare inborn errors of metabolism that can cause early mortality and significant morbidity. The mainstay of disease management is lifelong protein restriction. As an alternative, liver transplantation (LT) may improve survival, quality of life, and prevent further neurological deterioration. The aim of our study was to estimate the incremental costs and outcomes of LT versus nutritional support in patients with early-onset MMA or PA. We constructed a Markov model to simulate and compare life expectancies, quality-adjusted life years (QALYs), and lifetime direct and indirect costs for a cohort of newborns with MMA or PA who could either receive LT or be maintained on conventional nutritional support. We conducted a series of 1-way and probabilistic sensitivity analyses. In the base case, LT on average resulted in 1.5 more life years lived, 7.9 more QALYs, and a savings of $582,369 for lifetime societal cost per individual compared to nutritional support. LT remained more effective and less costly in all 1-way sensitivity analyses. In the probabilistic sensitivity analysis, LT was cost-effective at the $100,000/QALY threshold in more than 90% of the simulations and cost-saving in over half of the simulations. LT is likely a dominant treatment strategy compared to nutritional support in newborns with classical MMA or PA.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/economía , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Dieta con Restricción de Proteínas/economía , Trasplante de Hígado/economía , Apoyo Nutricional/economía , Acidemia Propiónica/economía , Acidemia Propiónica/terapia , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/mortalidad , Análisis Costo-Beneficio , Árboles de Decisión , Dieta con Restricción de Proteínas/efectos adversos , Costos de la Atención en Salud , Humanos , Recién Nacido , Trasplante de Hígado/efectos adversos , Trasplante de Hígado/mortalidad , Cadenas de Markov , Modelos Económicos , Apoyo Nutricional/efectos adversos , Acidemia Propiónica/diagnóstico , Acidemia Propiónica/mortalidad , Calidad de Vida , Años de Vida Ajustados por Calidad de Vida , Factores de Riesgo , Sobrevivientes , Factores de Tiempo , Resultado del Tratamiento
10.
BMJ Case Rep ; 20142014 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-24686805

RESUMEN

A 27-year-old woman with vitamin B12 responsive form of methylmalonic acidaemia (MMA) was pregnant with her first child. Treatment was unaltered during pregnancy: a low-protein diet and supplements. Her pregnancy was uncomplicated. She had a spontaneous delivery of a healthy girl with no MMA. The postpartum period was uneventful. MMA is a rare autosomal recessive metabolic disorder caused by a deficiency of methylmalonyl coenzyme A mutase or its vitamin B12-dependent cofactor, leading to a toxic accumulation of methylmalonyl acid in plasma and urine. Clinical presentation involves otherwise unexplained deterioration and neurological dysfunction, recurrent vomiting, dehydration, lethargy, respiratory distress and muscular hypotonia. Long-term sequelae are neurological problems, renal failure, pancreatitis and cardiomyopathy. This is the 11th reported case of pregnancy in a woman with MMA.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/terapia , Dieta con Restricción de Proteínas , Suplementos Dietéticos , Complicaciones del Embarazo/terapia , Adulto , Carbonato de Calcio/administración & dosificación , Carnitina/administración & dosificación , Femenino , Humanos , Embarazo , Vitamina B 12/administración & dosificación , Complejo Vitamínico B/administración & dosificación , Sulfato de Zinc/administración & dosificación
11.
Zhonghua Er Ke Za Zhi ; 51(3): 194-8, 2013 Mar.
Artículo en Chino | MEDLINE | ID: mdl-23751581

RESUMEN

OBJECTIVE: Combined methylmalonic acidemia with homocystinuria is a common form of methylmalonic acidemia in China. Patients with this disease can progress to death without timely and effective treatment. This study aimed to analyze the treatment outcomes of patients with combined methylmalonic acidemia and homocystinuria. METHOD: From September 2004 to April 2012, 58 patients with combined methylmalonic acidemia and homocystinuria (34 males and 24 females) were diagnosed and treated in our hospital. Fifty cases were from clinical patients including 42 early-onset cases and 8 late-onset cases. Their age when they were diagnosed ranged from 18 days to 30.8 years. The other 8 cases were from newborn screening. All the patients were treated with vitamin B12, betaine, folic acid, vitamin B6, and L-carnitine. The physical and neuropsychological development, general laboratory tests, the levels of amino acids, acylcarnitines, and homocysteine in blood, and organic acids in urine were followed up. RESULT: The follow-up period ranged from 1 month to 7.1 years. Three cases died (all were early-onset cases). In the other patients after treatment, the symptoms such as recurrent vomiting, seizures, lethargy, and poor feeding disappeared, muscle strength and muscle tension were improved, and general biochemical abnormalities such as anemia and metabolic acidosis were corrected. Among the surviving 55 cases, 49 had neurological impairments such as developmental delay and mental retardation. The median levels of blood propionylcarnitine and its ratio with acetylcarnitine, serum homocysteine, and urine methylmalonic acid were significantly decreased (P < 0.01), from 7.73 µmol/L (ranged from 1.5 to 18.61 µmol/L), 0.74 (ranged from 0.29 to 2.06), 97.3 µmol/L (ranged from 25.1 to 250 µmol/L) and 168.55 (ranged from 3.66 to 1032.82) before treatment to 2.74 µmol/L (ranged from 0.47 to 12.09 µmol/L), 0.16 (ranged from 0.03 to 0.62), 43.8 µmol/L (ranged from 17 to 97.8 µmol/L) and 6.81 (ranged from 0 to 95.43) after treatment, respectively. CONCLUSION: Patients with combined methylmalonic acidemia and homocystinuria respond to a combined treatment consisting of supplementation of hydroxycobalamin, betaine, folic acid, vitamin B6 and L-carnitine with clinical and biochemical improvement. But the long-term outcomes are unsatisfactory, with neurological sequelae in most patients.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/terapia , Homocistinuria/terapia , Hidroxocobalamina/uso terapéutico , Vitamina B 12/uso terapéutico , Adolescente , Adulto , Errores Innatos del Metabolismo de los Aminoácidos/sangre , Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Betaína/administración & dosificación , Betaína/uso terapéutico , Carnitina/análogos & derivados , Carnitina/sangre , Niño , Preescolar , Femenino , Estudios de Seguimiento , Homocistina/sangre , Homocistinuria/sangre , Homocistinuria/diagnóstico , Humanos , Hidroxocobalamina/administración & dosificación , Lactante , Recién Nacido , Masculino , Ácido Metilmalónico/orina , Tamizaje Neonatal , Resultado del Tratamiento , Vitamina B 12/administración & dosificación , Deficiencia de Vitamina B 12/congénito , Adulto Joven
12.
J Clin Anesth ; 25(2): 141-5, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23352788

RESUMEN

The management of cesarean delivery for a parturient with placenta previa at 36 weeks' gestation and glutaric aciduria type 1 is presented. The management goal was to prevent encephalopathic crisis by ensuring adequate caloric intake with dextrose infusion and to provide carnitine supplementation and adequate anesthesia.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/terapia , Encefalopatías Metabólicas/terapia , Cesárea , Complicaciones del Embarazo/terapia , Atención Prenatal/métodos , Anestesia Obstétrica/métodos , Anestesia Raquidea/métodos , Carnitina/uso terapéutico , Femenino , Glucosa/administración & dosificación , Glutaril-CoA Deshidrogenasa/deficiencia , Humanos , Infusiones Intravenosas , Atención Perioperativa/métodos , Placenta Previa/cirugía , Embarazo , Complejo Vitamínico B/uso terapéutico , Adulto Joven
13.
Pediatr Nephrol ; 28(2): 227-35, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22814947

RESUMEN

Methylmalonic acidurias are a heterogeneous group of inborn errors of branched-chain amino acid metabolism. Depending on the underlying etiology, acute or chronic renal disease constitutes major (long-term) complications. In recent decades, overall survival has improved due to optimized treatment strategies based on the use of standardized emergency protocols and dialysis techniques. The majority of these patients, especially those having mut°, cblB, and cblA deficiency, are at increased risk of developing chronic kidney disease secondary to tubulointerstitial nephritis to require hemo- or peritoneal dialysis. Kidney and/or liver transplantation, as organ replacement, or even gene therapy on a limited scale, are controversially discussed treatment options in methylmalonic acidurias. The pathophysiological basis of renal disease has not been clarified in detail until now, but a severe mitochondrial dysfunction and an impairment of tubular dicarboxylic acid transport due to accumulated toxic metabolic compounds has been recently proposed. Another severe renal complication of methylmalonic acidurias is the occurrence of cblC-associated infantile atypical hemolytic syndrome, which can result in acute kidney injury. Close collaboration between (pediatric) nephrologists and metabolic specialists is required for the long-term management of these patients.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/complicaciones , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Ácido Metilmalónico/metabolismo , Insuficiencia Renal Crónica/etiología , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Dieta , Suplementos Dietéticos , Genotipo , Humanos , Trasplante de Riñón , Mitocondrias/metabolismo , Fenotipo , Terapia de Reemplazo Renal
14.
Orphanet J Rare Dis ; 7: 48, 2012 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-22830360

RESUMEN

Glutamine synthetase (GS) is ubiquitously expressed in mammalian organisms and is a key enzyme in nitrogen metabolism. It is the only known enzyme capable of synthesising glutamine, an amino acid with many critical roles in the human organism. A defect in GLUL, encoding for GS, leads to congenital systemic glutamine deficiency and has been described in three patients with epileptic encephalopathy. There is no established treatment for this condition.Here, we describe a therapeutic trial consisting of enteral and parenteral glutamine supplementation in a four year old patient with GS deficiency. The patient received increasing doses of glutamine up to 1020 mg/kg/day. The effect of this glutamine supplementation was monitored clinically, biochemically, and by studies of the electroencephalogram (EEG) as well as by brain magnetic resonance imaging and spectroscopy.Treatment was well tolerated and clinical monitoring showed improved alertness. Concentrations of plasma glutamine normalized while levels in cerebrospinal fluid increased but remained below the lower reference range. The EEG showed clear improvement and spectroscopy revealed increasing concentrations of glutamine and glutamate in brain tissue. Concomitantly, there was no worsening of pre-existing chronic hyperammonemia.In conclusion, supplementation of glutamine is a safe therapeutic option for inherited GS deficiency since it corrects the peripheral biochemical phenotype and partially also improves the central biochemical phenotype. There was some clinical improvement but the patient had a long standing severe encephalopathy. Earlier supplementation with glutamine might have prevented some of the neuronal damage.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/terapia , Aminoácidos/metabolismo , Glutamato-Amoníaco Ligasa/deficiencia , Glutamina/administración & dosificación , Encéfalo/patología , Niño , Humanos , Imagen por Resonancia Magnética
15.
J Inherit Metab Dis ; 35(5): 761-76, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22170564

RESUMEN

Δ(1)-Pyrroline-5-carboxylate synthetase (P5CS) catalyzes the first two steps of ornithine/proline biosynthesis. P5CS deficiency has been reported in three families, with patients presenting with cutis/joint laxity, cataracts, and neurodevelopmental delay. Only one family exhibited metabolic changes consistent with P5CS deficiency (low proline/ornithine/citrulline/arginine; fasting hyperammonemia). Here we report a new P5CS-deficient patient presenting the complete clinical/metabolic phenotype and carrying p.G93R and p.T299I substitutions in the γ-glutamyl kinase (γGK) component of P5CS. The effects of these substitutions are (1) tested in mutagenesis/functional studies with E.coli γGK, (2) rationalized by structural modelling, and (3) reflected in decreased P5CS protein in patient fibroblasts (shown by immunofluorescence). Using optical/electron microscopy on skin biopsy, we show collagen/elastin fiber alterations that may contribute to connective tissue laxity and are compatible with our angio-MRI finding of kinky brain vessels in the patient. MR spectroscopy revealed decreased brain creatine, which normalized after sustained arginine supplementation, with improvement of neurodevelopmental and metabolic parameters, suggesting a pathogenic role of brain creatine decrease and the value of arginine therapy. Morphological and functional studies of fibroblast mitochondria show that P5CS deficiency is not associated with the mitochondrial alterations observed in Δ(1)-pyrroline-5-carboxylate reductase deficiency (another proline biosynthesis defect presenting cutis laxa and neurological alterations).


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Arginina/uso terapéutico , Ornitina-Oxo-Ácido Transaminasa/deficiencia , Errores Innatos del Metabolismo de los Aminoácidos/enzimología , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Secuencia de Aminoácidos , Humanos , Lactante , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Ornitina-Oxo-Ácido Transaminasa/metabolismo , Fenotipo
16.
Orphanet J Rare Dis ; 4: 29, 2009 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-20030831

RESUMEN

BACKGROUND: Pulmonary alveolar proteinosis (PAP) is a syndrome with multiple etiologies and is often deadly in lysinuric protein intolerance (LPI). At present, PAP is treated by whole lung lavage or with granulocyte/monocyte colony stimulating factor (GM-CSF); however, the effectiveness of GM-CSF in treating LPI associated PAP is uncertain. We hypothesized that GM-CSF and surfactant protein D (SP-D) would enhance the clearance of proteins and dying cells that are typically present in the airways of PAP lungs. METHODS: Cells and cell-free supernatant of therapeutic bronchoalveolar lavage fluid (BALF) of a two-year-old patient with LPI were isolated on multiple occasions. Diagnostic BALF samples from an age-matched patient with bronchitis or adult PAP patients were used as controls. SP-D and total protein content of the supernatants were determined by BCA assays and Western blots, respectively. Cholesterol content was determined by a calorimetic assay or Oil Red O staining of cytospin preparations. The cells and surfactant lipids were also analyzed by transmission electron microscopy. Uptake of Alexa-647 conjugated BSA and DiI-labelled apoptotic Jurkat T-cells by BAL cells were studied separately in the presence or absence of SP-D (1 microg/ml) and/or GM-CSF (10 ng/ml), ex vivo. Specimens were analyzed by light and fluorescence microscopy. RESULTS: Here we show that large amounts of cholesterol, and large numbers of cholesterol crystals, dying cells, and lipid-laden foamy alveolar macrophages were present in the airways of the LPI patient. Although SP-D is present, its bioavailability is low in the airways. SP-D was partially degraded and entrapped in the unusual surfactant lipid tubules with circular lattice, in vivo. We also show that supplementing SP-D and GM-CSF increases the uptake of protein and dying cells by healthy LPI alveolar macrophages, ex vivo. Serendipitously, we found that these cells spontaneously generated granulomas, ex vivo, and GM-CSF treatment drastically increased the number of granulomas whereas SP-D treatment counteracted the adverse effect of GM-CSF. CONCLUSIONS: We propose that increased GM-CSF and decreased bioavailability of SP-D may promote granuloma formation in LPI, and GM-CSF may not be suitable for treating PAP in LPI. To improve the lung condition of LPI patients with PAP, it would be useful to explore alternative therapies for increasing dead cell clearance while decreasing cholesterol content in the airways.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/terapia , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Granuloma/metabolismo , Lisina/metabolismo , Macrófagos Alveolares/fisiología , Proteína D Asociada a Surfactante Pulmonar , Adulto , Errores Innatos del Metabolismo de los Aminoácidos/inmunología , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/citología , Preescolar , Colesterol/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Pulmón/citología , Pulmón/metabolismo , Proteinosis Alveolar Pulmonar/inmunología , Proteinosis Alveolar Pulmonar/metabolismo , Proteinosis Alveolar Pulmonar/fisiopatología , Proteinosis Alveolar Pulmonar/terapia , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/fisiología , Proteína D Asociada a Surfactante Pulmonar/metabolismo , Proteína D Asociada a Surfactante Pulmonar/uso terapéutico , Resultado del Tratamiento
17.
Metabolism ; 57(4): 549-54, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18328359

RESUMEN

The aim of the study was to investigate the prevalence and mechanisms of development of carnitine deficiency in patients with lysinuric protein intolerance (LPI). In our cohort of 37 Finnish patients with LPI, 8 (8-52 years of age) have been diagnosed with hypocarnitinemia. Their free and total serum carnitine levels, acyl carnitine profiles, renal function, diet, and medication were compared with the data from 8 age- and sex-matched patients with LPI not treated with carnitine supplementation. In patients with LPI, hypocarnitinemia was strongly associated with female sex, renal insufficiency, and the use of ammonia-scavenging drugs. Of the 8 hypocarnitinemic patients, 3 complained of muscle weakness, and their symptoms disappeared during carnitine supplementation. Oral lysine supplementation did not correct hypocarnitinemia in our patients. The patients with LPI are at considerable risk for carnitine deficiency. Supplementation of hypocarnitinemic LPI patients with oral L-carnitine improved serum total carnitine values, but the ratio of free and total carnitine remained subnormal in all supplemented patients except one. Furthermore, decreased ratio of free and total serum carnitine was common even in LPI patients with normal total serum carnitine concentration.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Carnitina/deficiencia , Lisina/orina , Adolescente , Adulto , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Niño , Suplementos Dietéticos , Femenino , Humanos , Masculino , Persona de Mediana Edad
18.
J Inherit Metab Dis ; 29(2-3): 288-98, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16763890

RESUMEN

In a series of 137 patients with methylmalonic acidaemia (MMA) and propionic acidaemia (PA) diagnosed since the early 1970s, we report in more detail 81 patients (51 MMA and 30 PA) diagnosed between 1988 and 2005. In this series, 14% of patients died at initial access revealing the disease before or despite treatment, 18% died later, and the remainder (68%) are still alive. All patients were treated with the same protocol of enteral feeds with a low-protein diet adjusted to individual tolerance, carnitine, antibiotics, and only occasional use of an amino acid (AA) mixture. There was intensive follow-up and monitoring using measurements of urinary urea. Thirty-nine patients with severe forms, followed for more than 3 years, are analysed in particular detail. Of the 17 PA patients, 6 had moderate disability (all neonatal-onset forms), whereas 11 were normal or slightly delayed in their mental development. Four presented with cardiomyopathy, of whom 2 died. Of the 22 MMA patients, 13 presented in the neonatal period, of whom 3 died later, 2 are in renal failure and only 5 are still alive and have a normal or slightly delayed mental development. In the 9 patients with late-onset forms, there were no deaths and all patients but one have normal mental development. Among the 39 patients, only 40% were given an AA supplement at 3 years, and 50% between 6 and 11 years. The actual intake of natural protein was 0.92, 0.78 and 0.77 g/kg per day at 3, 6 and 11 years, respectively, in patients without AA supplementation, whereas it was 0.75, 0.74 and 0.54 g/kg per day in the group who received small quantities of AA (0.4-0.6 g/kg per day). In both groups, feeding disorders were frequent: 55% at 3 years, 35% at 6 years and 12% at 11 years. Many patients were given a food supplement by tube overnight or were even exclusively tube fed: 60% at 3 years, 48% at 6 years and still 27% at 11 years. Growth velocity was near the normal values. Plasma valine and isoleucine were low to very low, as were leucine and phenylalanine but to a lesser extent. Albumin, vitamins, trace elements and markers of bone metabolism were within the normal values. IGF1, 24-hour urine calcium and body mass density were low. Body composition showed a normal to low lean mass and a normal to high fat mass.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/terapia , Aminoácidos/uso terapéutico , Dieta con Restricción de Proteínas , Suplementos Dietéticos , Nutrición Enteral , Ácido Metilmalónico/orina , Propionatos/orina , Errores Innatos del Metabolismo de los Aminoácidos/dietoterapia , Errores Innatos del Metabolismo de los Aminoácidos/tratamiento farmacológico , Errores Innatos del Metabolismo de los Aminoácidos/orina , Aminoácidos/sangre , Estatura , Peso Corporal , Química Farmacéutica , Niño , Preescolar , Proteínas en la Dieta/metabolismo , Ingestión de Alimentos , Femenino , Estudios de Seguimiento , Hospitalización , Humanos , Ácido Láctico/análogos & derivados , Ácido Láctico/orina , Masculino , Evaluación Nutricional , Resultado del Tratamiento
19.
J Inherit Metab Dis ; 29(2-3): 281-7, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16763889

RESUMEN

Failure to thrive has been described in patients with organic acidaemias due to a variety of causes, both organic and inorganic. Failure to thrive in patients with methylmalonic acidaemia (MMA) and propionic acidaemia (PA) may be related to inadequate protein and energy intake rather than pathology of disease. Inadequate protein intake can also result in decreased resting energy expenditure, clinical signs and symptoms of amino acid deficiency, increased risk of infection, and developmental delay. Amino acid-based formulas (also referred to as 'medical foods') provide a key source of nitrogen, energy, vitamins and minerals which, when prescribed appropriately, can promote anabolism and growth. Although protein requirements in patients with organic acidaemias have not been elucidated, providing an adequate balance of protein, energy and other nutrients will help promote growth.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/terapia , Aminoácidos/uso terapéutico , Suplementos Dietéticos , Ácido Metilmalónico/sangre , Apoyo Nutricional , Propionatos/sangre , Errores Innatos del Metabolismo de los Aminoácidos/sangre , Errores Innatos del Metabolismo de los Aminoácidos/tratamiento farmacológico , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Aminoácidos/metabolismo , Química Farmacéutica , Metabolismo Energético , Humanos , Ácido Metilmalónico/metabolismo , Propionatos/metabolismo , Proteínas/metabolismo , Resultado del Tratamiento
20.
Mol Genet Metab ; 89(3): 274-6, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16580861

RESUMEN

Cerebral creatine and guanidinoacetate and blood and urine metabolites were studied in four patients with argininosuccinate synthetase (ASS) or argininosuccinate lyase (ASL) deficiency receiving large doses of arginine. Urine and blood metabolites varied largely. Cerebral guanidinoacetate was increased in all patients, while cerebral creatine was low in ASS and high in ASL deficiency. Because high cerebral guanidinoacetate might be toxic, lowering the arginine supplementation with additional creatine supplementation might be important.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/terapia , Argininosuccinato Sintasa/deficiencia , Aciduria Argininosuccínica , Encéfalo/metabolismo , Creatina/metabolismo , Glicina/análogos & derivados , Arginina/sangre , Niño , Preescolar , Creatina/sangre , Creatina/orina , Femenino , Glicina/sangre , Glicina/metabolismo , Glicina/orina , Humanos , Recién Nacido , Masculino , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA