Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Mol Genet Metab Rep ; 39: 101084, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38694233

RESUMEN

Phenylketonuria (PKU) is a genetic disorder caused by deficiency of the enzyme phenylalanine hydroxylase (PAH), which results in phenylalanine (Phe) accumulation in the blood and brain, and requires lifelong treatment to keep blood Phe in a safe range. Pegvaliase is an enzyme-substitution therapy approved for individuals with PKU and uncontrolled blood Phe concentrations (>600 µmol/L) despite prior management. Aggregated results from the PRISM clinical trials demonstrated substantial and sustained reductions in blood Phe with a manageable safety profile, but also noted individual variation in time to and dose needed for a first response. This analysis reports longer-term aggregate findings and characterizes individual participant responses to pegvaliase using final data from the randomized trials PRISM-1 (NCT01819727) and PRISM-2 (NCT01889862), and the open-label extension study 165-304 (NCT03694353). In 261 adult participants with a mean of 36.6 months of pegvaliase treatment, 71.3%, 65.1%, and 59.4% achieved clinically significant blood Phe levels of ≤600, ≤360, and ≤ 120 µmol/L, respectively. Some participants achieved blood Phe reductions with <20 mg/day pegvaliase, although most required higher doses. Based on Kaplan-Meier analysis, median (minimum, maximum) time to first achievement of a blood Phe threshold of ≤600, ≤360, or ≤ 120 µmol/L was 4.4 (0.0, 54.0), 8.0 (0.0, 57.0), and 11.6 (0.0, 66.0) months, respectively. Once achieved, blood Phe levels remained below clinical threshold in most participants. Sustained Phe response (SPR), a new method described within for measuring durability of blood Phe response, was achieved by 85.5%, 84.7%, and 78.1% of blood Phe responders at blood Phe thresholds of ≤600, ≤360, or ≤ 120 µmol/L, respectively. Longer-term safety data were consistent with previous reports, with the most common adverse events (AEs) being arthralgia, injection site reactions, headache, and injection site erythema. The incidence of most AEs, including hypersensitivity AEs, was higher during the early treatment phase (≤6 months) than later during treatment. In conclusion, using data from three key pegvaliase clinical trials, participants treated with pegvaliase were able to reach clinically significant blood Phe reductions to clinical thresholds of ≤600, ≤360, or ≤ 120 µmol/L during early treatment, with safety profiles improving from early to sustained treatment. This study also supports the use of participant-level data and new ways of looking at durable blood Phe responses to better characterize patients' individual PKU treatment journeys.

2.
Nat Metab ; 5(10): 1685-1690, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37770764

RESUMEN

Despite available treatment options, many patients with phenylketonuria (PKU) cannot achieve target plasma phenylalanine (Phe) levels1. We previously modified Escherichia coli Nissle 1917 to metabolize Phe in the gut after oral administration (SYNB1618) and designed a second strain (SYNB1934) with enhanced activity of phenylalanine ammonia lyase2,3. In a 14-day open-label dose-escalation study (Synpheny-1, NCT04534842 ), we test a primary endpoint of change from baseline in labeled Phe (D5-Phe AUC0-24; D5-Phe area under the curve (AUC) over 24 hours after D5-Phe administration) in plasma after D5-Phe challenge in adult participants with screening Phe of greater than 600 µM. Secondary endpoints were the change from baseline in fasting plasma Phe and the incidence of treatment-emergent adverse events. A total of 20 participants (ten male and ten female) were enrolled and 15 completed the study treatment. Here, we show that both strains lower Phe levels in participants with PKU: D5-Phe AUC0-24 was reduced by 43% from baseline with SYNB1934 and by 34% from baseline with SYNB1618. SYNB1934 led to a decrease in fasting plasma Phe of 40% (95% CI, -52, -24). There were no serious adverse events or infections. Four participants discontinued because of adverse events, and one withdrew during the baseline period. We show that synthetic biotics can metabolize Phe in the gut, lower post-prandial plasma Phe levels and lower fasting plasma Phe in patients with PKU.


Asunto(s)
Fenilalanina , Fenilcetonurias , Adulto , Humanos , Masculino , Femenino , Fenilalanina/uso terapéutico , Fenilcetonurias/tratamiento farmacológico , Fenilanina Amoníaco-Liasa/uso terapéutico , Administración Oral , Escherichia coli
3.
Mol Genet Metab ; 133(3): 231-241, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33985889

RESUMEN

One of the most vital elements of management for patients with inborn errors of intermediary metabolism is the promotion of anabolism, the state in which the body builds new components, and avoidance of catabolism, the state in which the body breaks down its own stores for energy. Anabolism is maintained through the provision of a sufficient supply of substrates for energy, as well as critical building blocks of essential amino acids, essential fatty acids, and vitamins for synthetic function and growth. Patients with metabolic diseases are at risk for decompensation during prolonged fasting, which often occurs during illnesses in which enteral intake is compromised. During these times, intravenous nutrition must be supplied to fully meet the specific nutritional needs of the patient. We detail our approach to intravenous management for metabolic patients and its underlying rationale. This generally entails a combination of intravenous glucose and lipid as well as early introduction of protein and essential vitamins. We exemplify the utility of our approach in case studies, as well as scenarios and specific disorders which require a more careful administration of nutritional substrates or a modification of macronutrient ratios.


Asunto(s)
Errores Innatos del Metabolismo/complicaciones , Errores Innatos del Metabolismo/terapia , Metabolismo , Administración Intravenosa , Niño , Dieta Cetogénica , Glucosa/administración & dosificación , Humanos , Lípidos/administración & dosificación , Estado Nutricional , Vitaminas/administración & dosificación
4.
Pediatr Dev Pathol ; 23(3): 189-196, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31542992

RESUMEN

The central nervous system (CNS) is a highly complex and energy-dependent organ that is subject to a wide variety of metabolic, hypoxic-ischemic, and infectious insults that result in cystic changes. Diagnosis of metabolic defects causing extensive cystic changes is particularly challenging for the pediatric pathologist, due to the rarity of these conditions. Pyruvate dehydrogenase (PDH) deficiency is one of the most common etiologies of congenital lactic acidosis, caused by mutations in subunits of the large mitochondrial matrix complex, and characterized by periventricular cysts, although few detailed reports focusing on neuropathologic findings exist. In addition, rare defects in other mitochondrial enzymes such as short-chain enoyl-CoA hydratase (SCEH, encoded by ECHS1 gene) can cause secondary PDH deficiency and present with neonatal lactic acidosis, but neuropathological findings have never been reported. Nonmetabolic conditions can also produce CNS cystic lesions, primarily in newborns. The pathologist must therefore distinguish between these etiologically disparate conditions which can produce CNS cavitary lesions. Here, we compare and contrast the gross and microscopic findings of cysts associated with cases of PDH and SCEH deficiencies with other neonatal cystic brain diseases including periventricular leukomalacia, neonatal Alexander disease, Canavan disease, and a case of cysts associated with a vascular abnormality. Our studies show that PDH and SCEH deficiencies are not grossly or histologically distinguishable from each other and both are associated with smooth-walled cysts largely limited to the telencephalic germinal matrix. Both show an absence of prominent hemosiderin deposits, Rosenthal fibers, vacuolization of the white matter, and gliosis or axonal damage in the surrounding parenchyma. These features can help distinguish PDH/SCEH deficiency from other pediatric/neonatal cystic CNS disorders, especially those produced by hypoxic ischemic conditions. Cysts, usually bilateral, confined to the telencephalic germinal matrix should elicit metabolic and genetic testing to appropriately diagnose PDH and SCEH and distinguish them from each other.


Asunto(s)
Encefalopatías/etiología , Quistes del Sistema Nervioso Central/etiología , Quistes del Sistema Nervioso Central/patología , Enoil-CoA Hidratasa/deficiencia , Enfermedad por Deficiencia del Complejo Piruvato Deshidrogenasa/patología , Encéfalo/patología , Encefalopatías/diagnóstico , Encefalopatías/patología , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Enfermedad por Deficiencia del Complejo Piruvato Deshidrogenasa/etiología
5.
J Inherit Metab Dis ; 42(3): 424-437, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30873612

RESUMEN

STUDY OBJECTIVE: A phase 1/2 clinical trial was performed in individuals with cystathionine ß synthase (CBS) deficient homocystinuria with aims to: (a) assess pharmacokinetics and safety of taurine therapy, (b) evaluate oxidative stress, inflammation, and vascular function in CBS deficiency, and (c) evaluate the impact of short-term taurine treatment. METHODS: Individuals with pyridoxine-nonresponsive CBS deficiency with homocysteine >50 µM, without inflammatory disorder or on antioxidant therapy were enrolled. Biomarkers of oxidative stress and inflammation, endothelial function (brachial artery flow-mediated dilation [FMD]), and disease-related metabolites obtained at baseline were compared to normal values. While maintaining current treatment, patients were treated with 75 mg/kg taurine twice daily, and treatment response assessed after 4 hours and 4 days. RESULTS: Fourteen patients (8-35 years; 8 males, 6 females) were enrolled with baseline homocysteine levels 161 ± 67 µM. The study found high-dose taurine to be safe when excluding preexisting hypertriglyceridemia. Taurine pharmacokinetics showed a rapid peak level returning to near normal levels at 12 hours, but had slow accumulation and elevated predosing levels after 4 days of treatment. Only a single parameter of oxidative stress, 2,3-dinor-8-isoprostaglandin-F2α, was elevated at baseline, with no elevated inflammatory parameters, and no change in FMD values overall. Taurine had no effect on any of these parameters. However, the effect of taurine was strongly related to pretreatment FMD values; and taurine significantly improved FMD in the subset of individuals with pretreatment FMD values <10% and in individuals with homocysteine levels >125 µM, pertinent to endothelial function. CONCLUSION: Taurine improves endothelial function in CBS-deficient homocystinuria in patients with preexisting reduced function.


Asunto(s)
Biomarcadores/metabolismo , Cistationina betasintasa/metabolismo , Homocistinuria/tratamiento farmacológico , Taurina/farmacocinética , Taurina/uso terapéutico , Adolescente , Adulto , Arteria Braquial/efectos de los fármacos , Niño , Cistationina betasintasa/deficiencia , Femenino , Homocisteína/metabolismo , Homocistinuria/genética , Humanos , Inflamación/tratamiento farmacológico , Masculino , Estrés Oxidativo/efectos de los fármacos , Estados Unidos , Adulto Joven
6.
Mol Genet Metab ; 125(3): 217-227, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30146451

RESUMEN

BACKGROUND: Phenylketonuria (PKU) is caused by a deficiency in phenylalanine hydroxylase enzyme activity that leads to phenylalanine (Phe) accumulation in the blood and brain. Elevated blood Phe levels are associated with complications in adults, including neurological, psychiatric, and cognitive issues. Even with nutrition and pharmacological management, the majority of adults with PKU do not maintain blood Phe levels at or below guideline recommended levels. Pegvaliase, PEGylated recombinant Anabaena variabilis phenylalanine ammonia lyase (PAL), converts Phe to trans-cinnamic acid and ammonia, and is an investigational enzyme substitution therapy to lower blood Phe in adults with PKU. METHODS: Pegvaliase was administered using an induction, titration, and maintenance dosing regimen in adults with PKU naïve to pegvaliase treatment. Doses were gradually increased until blood Phe ≤ 600 µmol/L was achieved. The maintenance dose was the dose at which participants achieved and sustained blood Phe ≤ 600 µmol/L for at least 4 weeks without dose modification. Analyses were performed for participants who achieved (Group A, n = 11) and did not achieve (Group B, n = 13) maintenance dose during the first 24 weeks of study treatment. RESULTS: Baseline mean blood Phe for Group A and Group B were 1135 µmol/L and 1198 µmol/L, respectively. Mean blood Phe ≤ 600 µmol/L was achieved for Group A by Week 11 (mean blood Phe of 508 ±â€¯483 µmol/L) and for Group B by Week 48 (mean blood Phe of 557 ±â€¯389 µmol/L). The most common adverse events involved hypersensitivity reactions, which were mostly mild to moderate in severity and decreased over time. One participant in Group B had four acute systemic hypersensitivity events of anaphylaxis consistent with clinical National Institute of Allergy and Infectious Disease/Food Allergy and Anaphylaxis Network criteria; all events were non-IgE mediated and resolved without sequelae, with pegvaliase dosing discontinued after the fourth event. The incidence and titers of anti-drug antibodies were generally lower in Group A compared to Group B. CONCLUSIONS: Pegvaliase administered with an induction, titration, and maintenance dosing regimen demonstrated substantial efficacy at reducing blood Phe in both Group A and Group B by Week 48, with a manageable safety profile in most participants. Blood Phe reduction due to pegvaliase appears to be related to dose, treatment duration, and individual immune response; given additional time on treatment and dose titration, later Phe responders (Group B) achieved benefit similar to early Phe responders (Group A), with similar long-term safety profiles.


Asunto(s)
Fenilanina Amoníaco-Liasa/administración & dosificación , Fenilalanina/sangre , Fenilcetonurias/tratamiento farmacológico , Proteínas Recombinantes/administración & dosificación , Adolescente , Adulto , Anciano , Anticuerpos/sangre , Pruebas Diagnósticas de Rutina , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fenilanina Amoníaco-Liasa/química , Fenilcetonurias/sangre , Fenilcetonurias/patología , Proteínas Recombinantes/química , Adulto Joven
7.
Orphanet J Rare Dis ; 13(1): 108, 2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29973227

RESUMEN

BACKGROUND: Deficiency of phenylalanine hydroxylase causes phenylketonuria (PKU) with elevated phenylalanine (Phe) levels and associated neuropsychiatric and neurocognitive symptoms. Pegvaliase (PEGylated phenylalanine ammonia lyase) is an investigational agent to lower plasma Phe in adults with PKU. This study aimed to characterize the long-term efficacy, safety, and immunogenicity of pegvaliase in adults with PKU. METHODS: PAL-003 is an ongoing, open-label, long-term extension study of the pegvaliase dose-finding parent phase 2 studies. Participants continued the dose of pegvaliase from one of three parent studies, with dose adjustments to achieve a plasma Phe concentration between 60 and 600 µmol/L. RESULTS: Mean (standard deviation [SD]) plasma Phe at treatment-naïve baseline for 80 participants in the parent studies was 1302.4 (351.5) µmol/L. In the 68 participants who entered the extension study, plasma Phe decreased 58.9 (39)% from baseline, to 541.6 (515.5) µmol/L at Week 48 of treatment. Plasma Phe concentrations ≤120 µmol/L, ≤360 µmol/L, and ≤ 600 µmol/L were achieved by 78.7, 80.0, and 82.5% of participants, respectively. Mean (SD) protein intake at baseline was 69.4 (40.4) g/day (similar to the recommended intake for the unaffected population) and remained stable throughout the study. All participants experienced adverse events (AEs), which were limited to mild or moderate severity in most (88.8%); the most common AEs were injection-site reaction (72.5%), injection-site erythema (67.5%), headache (67.5%), and arthralgia (65.0%). The AE rate decreased from 58.3 events per person-year in the parent studies to 18.6 events per person-year in the extension study. CONCLUSIONS: Pegvaliase treatment in adults with PKU produced meaningful and persistent reductions in mean plasma Phe concentration with a manageable safety profile for most subjects that continued with long-term treatment. TRIAL REGISTRATION: ClinicalTrials.gov , NCT00924703. Registered June 18, 2009, https://clinicaltrials.gov/ct2/show/NCT00924703.


Asunto(s)
Fenilanina Amoníaco-Liasa/uso terapéutico , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/metabolismo , Proteínas Recombinantes/uso terapéutico , Humanos , Fenilalanina/sangre , Fenilanina Amoníaco-Liasa/metabolismo , Fenilcetonurias/sangre
8.
J Inherit Metab Dis ; 2018 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-29736632
10.
Mitochondrion ; 21: 1-10, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25575635

RESUMEN

Muscle, heart and liver were analyzed in a male subject who succumbed to HSD10 disease. Respiratory chain enzyme analysis and BN-PAGE showed reduced activities and assembly of complexes I, III, IV, and V. The mRNAs of all RNase P subunits were preserved in heart and overexpressed in muscle, but MRPP2 protein was severely decreased. RNase P upregulation correlated with increased expression of mitochondrial biogenesis factors and preserved mitochondrial enzymes in muscle, but not in heart where this compensatory mechanism was incomplete. We demonstrate elevated amounts of unprocessed pre-tRNAs and mRNA transcripts encoding mitochondrial subunits indicating deficient RNase P activity. This study provides evidence of abnormal mitochondrial RNA processing causing mitochondrial energy failure in HSD10 disease.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Acetil-CoA C-Acetiltransferasa/deficiencia , ADN Mitocondrial/metabolismo , Metabolismo Energético , Errores Innatos del Metabolismo Lipídico/fisiopatología , Mitocondrias/fisiología , Transcripción Genética , 3-Hidroxiacil-CoA Deshidrogenasas/genética , Respiración de la Célula , Discinesias , Transporte de Electrón , Expresión Génica , Humanos , Lactante , Recién Nacido , Hígado/patología , Masculino , Discapacidad Intelectual Ligada al Cromosoma X , Mitocondrias/genética , Músculos/patología , Miocardio/patología , Procesamiento Postranscripcional del ARN
11.
Mol Genet Metab ; 108(1): 13-7, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23177662

RESUMEN

OBJECTIVE: To study the impact of geographic access to care on metabolic control and compliance in phenylketonuria (PKU). STUDY DESIGN: Phenylalanine (Phe) levels and number of samples obtained were abstracted from a data base of 76 patients age <21 years and compared for age, sex, and distance to clinic. Levels and number of samples were compared to the clinic guidelines for age. RESULTS: There was a strong positive correlation between age and Phe levels in adolescents and young adults while age and number of samples submitted were negatively correlated. There was not a significant correlation between Phe levels and distance to clinic, nor was there a significant difference in the Phe levels by distant categories (Denver metro, Front Range, distant area). However, there was a decrease in number of samples sent compared to clinic guidelines by distance, with patients residing in distant areas (>100 miles) sending significantly less samples. CONCLUSION: Geographic access to care does not impact control of Phe levels, but it does affect the number of monitoring samples sent to the clinic. Age groups of adolescents and young adults have a strong impact on both control of Phe levels and number of monitoring samples compared to clinic guidelines.


Asunto(s)
Accesibilidad a los Servicios de Salud , Cooperación del Paciente , Fenilcetonurias/terapia , Adolescente , Adulto , Niño , Femenino , Geografía , Humanos , Masculino , Fenilcetonurias/metabolismo , Adulto Joven
12.
Pediatr Radiol ; 43(7): 882-5, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23250031

RESUMEN

We present a neonate with molybdenum cofactor deficiency imaged at presentation during the first month of life and at 5 months with diffusion-weighted brain MRI. While the imaging features of this disease have previously been reported, this case highlights a distinctive initial pattern of widespread restricted diffusion involving cortex at the depths of sulci. Other case series have published diffusion-weighted images (DWI) with this pattern but never specifically commented on this finding. This distinct DWI pattern also accounts for the configuration of ulegyria frequently described on later imaging. Early recognition of this unique initial DWI pattern could avoid misdiagnosis and better direct counseling and management.


Asunto(s)
Corteza Cerebral/patología , Imagen de Difusión por Resonancia Magnética/métodos , Errores Innatos del Metabolismo de los Metales/patología , Femenino , Humanos , Recién Nacido , Molibdoferredoxina
13.
Mol Genet Metab ; 105(4): 672-6, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22310224

RESUMEN

PURPOSE: To understand current patient selection, dosing, and response criteria used for sapropterin dihydrochloride (sapropterin, Kuvan®) to treat phenylketonuria (PKU). METHODS: Results of a 2010 survey of twenty-nine academic medical centers are reported to describe practice patterns in comparison to results of a survey done in 2008 and to what is reported in the literature. RESULTS/CONCLUSIONS: In addition to reduction in blood phenylalanine (Phe) levels, clinicians report using broader disease-management approaches when evaluating clinical benefit of sapropterin, including consideration of increased Phe tolerance and behavioral changes. Similar approaches are reported in the literature.


Asunto(s)
Biopterinas/análogos & derivados , Selección de Paciente , Fenilalanina/sangre , Fenilcetonurias/diagnóstico , Fenilcetonurias/tratamiento farmacológico , Biopterinas/uso terapéutico , Preescolar , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Lactante , Recién Nacido , Óxido Nítrico/metabolismo , Fenilcetonurias/sangre , Guías de Práctica Clínica como Asunto , Pronóstico
14.
Curr Opin Pediatr ; 23(6): 693-9, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21926623

RESUMEN

Great strides in the identification, etiologic understanding, and treatment of metabolic and genetic disorders associated with infantile seizures have occurred in recent years. We explain the cause, pathogenesis, diagnosis, presentation, and treatment of certain metabolic disorders that now have defined interventions that improve and optimize neurologic outcome. A systematic approach to infantile seizures will allow the primary practitioner to more effectively create a differential diagnosis with close attention to the disorders that have specific treatment applications.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Electroencefalografía/métodos , Espasmos Infantiles/diagnóstico , Diagnóstico Diferencial , Estudios de Seguimiento , Humanos , Lactante , Masculino , Espasmos Infantiles/tratamiento farmacológico
15.
Mol Genet Metab ; 103(4): 330-7, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21601502

RESUMEN

Cystathionine beta-synthase (CBS) deficient homocystinuria (HCU) is an inherited metabolic defect that if untreated, typically results in cognitive impairment, connective tissue disturbances, atherosclerosis and thromboembolic disease. In recent years, chronic inappropriate expression of the inflammatory response has emerged as a major driving force of both thrombosis and atherosclerotic lesion development. We report here a characterization of the abnormalities in cytokine expression induced in both a mouse model of HCU and human subjects with the disease in the presence and absence of homocysteine lowering therapy. HCU mice exhibited highly significant induction of the pro-inflammatory cytokines Il-1alpha, Il-1beta and TNF-alpha. Similarly, in untreated/poorly compliant human subjects with HCU we observed constitutive induction of multiple pro-inflammatory cytokines (IL-1alpha, IL-6, TNF-alpha, Il-17 and IL-12(p70)) and chemotactic chemokines (fractalkine, MIP-1alpha and MIP-1beta) compared to normal controls. These HCU patients also exhibited significant induction of IL-9, TGF-alpha and G-CSF. The expression levels of anti-inflammatory cytokines were unaffected in both HCU mice and human subjects with the disease. In the human subjects, homocysteine lowering therapy was associated with either normalization or significant reduction of all of the pro-inflammatory cytokines and chemokines investigated. We conclude that HCU is a disease of chronic inflammation and that aberrant cytokine expression has the potential to contribute to multiple aspects of pathogenesis. Our findings indicate that anti-inflammatory strategies could serve as a useful adjuvant therapy for this disease.


Asunto(s)
Quimiocinas/metabolismo , Cistationina betasintasa/genética , Homocistinuria/metabolismo , Adolescente , Adulto , Animales , Betaína/farmacología , Quimiocina CCL4/metabolismo , Niño , Preescolar , Cistationina betasintasa/deficiencia , Cistationina betasintasa/metabolismo , Femenino , Homocistinuria/terapia , Humanos , Interleucina-17/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Transgénicos , Factor de Necrosis Tumoral alfa/metabolismo
16.
J Inherit Metab Dis ; 33(4): 421-7, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20532982

RESUMEN

Scheie syndrome is the most attenuated and rarest form of mucopolysaccharidosis type I (MPS I), an inherited lysosomal storage disorder. Only small patient series have previously been reported. Using natural history data from the uniquely large population of 78 Scheie patients enrolled in the MPS I Registry, we characterized the onset and prevalence of clinical manifestations and explored reasons for delayed diagnosis of the disease. Median patient age was 17.5 years; 46% of the patients were male, and 88% were Caucasian. Of 25 MPS I-related clinical features, cardiac valve abnormalities, joint contractures, and corneal clouding were each reported by >80% and all three by 53% of patients. Carpal tunnel syndrome, hernia, coarse facial features, and hepatomegaly were each reported by >50% of patients. Age at onset of the clinical features varied widely between individuals, but the median age at onset was 3 years for hernia and between 5 and 12 years for most features, including coarse facial features, hepatomegaly, joint contractures, bone deformities, cardiac valve abnormalities, cognitive impairment, and corneal clouding. Carpal tunnel syndrome, cardiomyopathy, and myelopathy arose more commonly during adolescence or adulthood. Delays up to 47 years intervened between symptom onset and disease diagnosis, and the longest delays were associated with later age at symptom onset and symptom onset before 1980. In summary, Scheie syndrome usually emerges during childhood, and recognition of attenuated MPS I requires awareness of the multisystemic disease manifestations and their diverse presentation. Given the availability of etiologic treatment, prompt diagnosis is important.


Asunto(s)
Edad de Inicio , Mucopolisacaridosis I/epidemiología , Mucopolisacaridosis I/fisiopatología , Adolescente , Adulto , Distribución por Edad , Cardiomiopatías/epidemiología , Síndrome del Túnel Carpiano/epidemiología , Niño , Preescolar , Progresión de la Enfermedad , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Mucopolisacaridosis I/diagnóstico , Prevalencia , Sistema de Registros/estadística & datos numéricos , Enfermedades de la Médula Espinal/epidemiología , Adulto Joven
17.
Pediatr Res ; 68(2): 159-64, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20453710

RESUMEN

This patient presented on the first day of life with pronounced lactic acidosis with an elevated lactate/pyruvate ratio. Urine organic acids showed Krebs cycle metabolites and mildly elevated methylmalonate and methylcitrate. The acylcarnitine profile showed elevated propionylcarnitine and succinylcarnitine. Amino acids showed elevated glutamic acid, glutamine, proline, and alanine. From the age 2 of mo on, she had elevated transaminases and intermittent episodes of liver failure. Liver biopsy showed steatosis and a decrease of mitochondrial DNA to 50% of control. She had bilateral sensorineural hearing loss. Over the course of the first 2 y of life, she developed a progressively severe myopathy with pronounced muscle weakness eventually leading to respiratory failure, Leigh disease, and recurrent hepatic failure. The hepatic symptoms and the metabolic parameters temporarily improved on treatment with aspartate, but neither muscle symptoms nor brain lesions improved. Laboratory testing revealed a deficiency of succinyl-CoA ligase enzyme activity and protein in fibroblasts because of a novel homozygous mutation in the SUCLG1 gene: c.40A>T (p.M14L). Functional analysis suggests that this methionine is more likely to function as the translation initiator methionine, explaining the pathogenic nature of the mutation. Succinyl-CoA ligase deficiency due to an SUCLG1 mutation is a new cause for mitochondrial hepatoencephalomyopathy.


Asunto(s)
Encefalopatías Metabólicas , Hepatopatías , Enfermedades Mitocondriales , Succinato-CoA Ligasas/deficiencia , Secuencia de Aminoácidos , Secuencia de Bases , Encéfalo/metabolismo , Encéfalo/patología , Encefalopatías Metabólicas/enzimología , Encefalopatías Metabólicas/genética , Encefalopatías Metabólicas/patología , Análisis Mutacional de ADN , Resultado Fatal , Femenino , Humanos , Lactante , Recién Nacido , Enfermedad de Leigh/enzimología , Enfermedad de Leigh/genética , Enfermedad de Leigh/patología , Hepatopatías/enzimología , Hepatopatías/genética , Hepatopatías/patología , Imagen por Resonancia Magnética , Enfermedades Mitocondriales/enzimología , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/patología , Datos de Secuencia Molecular , Mutación , Succinato-CoA Ligasas/genética
18.
Mol Genet Metab ; 95(4): 201-5, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18974016

RESUMEN

We investigated in a patient with holocarboxylase synthetase deficiency, the relation between the biochemical and genetic factors of the mutant protein with the pharmacokinetic factors of successful biotin treatment. A girl exhibited abnormal skin at birth, and developed in the first days of life neonatal respiratory distress syndrome and metabolic abnormalities diagnostic of multiple carboxylase deficiency. Enzyme assays showed low carboxylase activities. Fibroblast analysis showed poor incorporation of biotin into the carboxylases, and low transfer of biotin by the holocarboxylase synthetase enzyme. Kinetic studies identified an increased Km but a preserved Vmax. Mutation analysis showed the child to be a compound heterozygote for a new nonsense mutation Q379X and for a novel missense mutation Y663H. This mutation affects a conserved amino acid, which is located the most 3' of all recorded missense mutations thus far described, and extends the region of functional biotin interaction. Treatment with biotin 100mg/day gradually improved the biochemical abnormalities in blood and in cerebrospinal fluid (CSF), corrected the carboxylase enzyme activities, and provided clinical stability and a normal neurodevelopmental outcome. Plasma concentrations of biotin were increased to more than 500 nM, thus exceeding the increased Km of the mutant enzyme. At these pharmacological concentrations, the CSF biotin concentration was half the concentration in blood. Measuring these pharmacokinetic variables can aid in optimizing treatment, as individual tailoring of dosing to the needs of the mutation may be required.


Asunto(s)
Biotina/administración & dosificación , Ligasas de Carbono-Nitrógeno/deficiencia , Deficiencia de Holocarboxilasa Sintetasa/tratamiento farmacológico , Secuencia de Aminoácidos , Biotina/metabolismo , Ligasas de Carbono-Nitrógeno/química , Ligasas de Carbono-Nitrógeno/genética , Células Cultivadas , Femenino , Deficiencia de Holocarboxilasa Sintetasa/genética , Deficiencia de Holocarboxilasa Sintetasa/metabolismo , Humanos , Recién Nacido , Cinética , Datos de Secuencia Molecular , Mutación , Alineación de Secuencia
19.
Mol Genet Metab ; 94(4): 485-490, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18495510

RESUMEN

The mitochondrial 13513G>A (D393N) mutation in the ND5 subunit of the respiratory chain complex I was initially described in association with MELAS syndrome. Recent observations have linked this mutation to Leigh disease. We screened for the 13513G>A mutation in a cohort of 265 patients with Leigh and Leigh-like disease. The mutation was found in a total of 5 patients. An additional patient who had clinical presentation consistent with a Leigh-like phenotype but with a normal brain MRI was added to the cohort. None of an additional 88 patients meeting MELAS disease criteria, nor 56 patients with respiratory chain deficiency screened for the 13513G>A were found positive for the mutation. The most frequent clinical manifestations in our patients were hypotonia, ocular and cerebellar involvement. Low mutation heteroplasmy in the range of 20-40% was observed in all 6 patients. This observation is consistent with the previously reported low heteroplasmy of this mutation in some patients with the 13513G>A mutation and complex I deficiency. However, normal complex I activity was observed in two patients in our cohort. As most patients with Leigh-like disease and the 13513G>A mutation have been described with complex I deficiency, this report adds to the previously reported subset of patients with normal respiratory complex function. We conclude that in any patient with Leigh or Leigh-like disease, testing for the 13513G>A mutation is clinically relevant and low mutant loads in blood or muscle may be considered pathogenic, in the presence of normal respiratory chain enzyme activities.


Asunto(s)
Complejo I de Transporte de Electrón/deficiencia , Enfermedad de Leigh/enzimología , Enfermedad de Leigh/genética , Mitocondrias/genética , Proteínas Mitocondriales/deficiencia , Músculos/enzimología , Mutación Puntual , Secuencia de Aminoácidos , Preescolar , Estudios de Cohortes , Análisis Mutacional de ADN , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/genética , Femenino , Humanos , Lactante , Masculino , Mitocondrias/química , Mitocondrias/enzimología , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Datos de Secuencia Molecular , Músculos/química , Mutación Missense , Fenotipo , Alineación de Secuencia
20.
Eur J Pediatr ; 167(7): 771-6, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17891417

RESUMEN

A 6-week-old child presented with hypotonia, myopathy, and a rapidly worsening dilated cardiomyopathy with severe atrial and ventricular arrhythmias and pulmonary hypertension, which proved fatal at age 3 months. Biochemical analysis showed a combined deficiency of the enzymatic activities of complexes I and IV and molecular studies identified a T14709C mutation in the mitochondrial tRNA glutamic acid gene. A review of symptomatology in patients with this mutation shows that it mainly presents in childhood or young adults with mild myopathy and diabetes mellitus. Infants with a high, nearly homoplasmic mutant load can present with more severe symptoms including cardiomyopathy. Families with this mitochondrial DNA mutation should be aware that increased mutant load in a subsequent generation may result in severe and often fatal cardiac symptoms.


Asunto(s)
Cardiomiopatías/etiología , Enfermedades Mitocondriales/genética , ARN de Transferencia de Ácido Glutámico/genética , Cardiomiopatías/fisiopatología , Resultado Fatal , Femenino , Humanos , Lactante , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...