Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Int Rev Cell Mol Biol ; 372: 159-174, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36064263

RESUMEN

Phenylketonuria (PKU) is a metabolic rare disease characterized by a failure of the body to clear out the high levels of Phenylalanine (Phe), leading to devastating neurological defects and growth retardation. PKU was discovered in 1934 by AsbjrØrn FØlling, and even though there have been continuous efforts from the scientific community to find therapeutic approaches to modulate the high levels of phenylalanine found in the body of the PKU patients, an efficient therapy still needs to be developed. Current standard of care includes low phenylalanine diets, but the strict restrictions for patients and families makes it very difficult to adequately being implemented. FDA has approved two drugs to help reduce Phe levels in PKU patients: an enzyme substitution therapy, Palynziq® (PEGylated recombinant phenylalanine ammonia lyase), and Kuvan®, a supplemental tetrahydrobiopterin (BH4) cofactor that enhances residual enzyme activity. Both treatments are restricted to certain PKU patients' population, and, therefore, there are still high unmet needs for most of the patients. The present review will focus on current advancements in lipid nanoparticles (LNP)-mRNA technologies and their potential in treating the root cause of PKU, a therapeutic approach that will be analyzed in the context of other promising therapeutic approaches that are been developed for PKU.


Asunto(s)
Fenilcetonurias , Humanos , Liposomas , Nanopartículas , Fenilalanina/metabolismo , Fenilalanina/uso terapéutico , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/genética , ARN Mensajero/uso terapéutico , Tecnología
2.
Hum Genomics ; 16(1): 22, 2022 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-35854334

RESUMEN

This review discusses the epidemiology, pathophysiology, genetic etiology, and management of phenylketonuria (PKU). PKU, an autosomal recessive disease, is an inborn error of phenylalanine (Phe) metabolism caused by pathogenic variants in the phenylalanine hydroxylase (PAH) gene. The prevalence of PKU varies widely among ethnicities and geographic regions, affecting approximately 1 in 24,000 individuals worldwide. Deficiency in the PAH enzyme or, in rare cases, the cofactor tetrahydrobiopterin results in high blood Phe concentrations, causing brain dysfunction. Untreated PKU, also known as PAH deficiency, results in severe and irreversible intellectual disability, epilepsy, behavioral disorders, and clinical features such as acquired microcephaly, seizures, psychological signs, and generalized hypopigmentation of skin (including hair and eyes). Severe phenotypes are classic PKU, and less severe forms of PAH deficiency are moderate PKU, mild PKU, mild hyperphenylalaninaemia (HPA), or benign HPA. Early diagnosis and intervention must start shortly after birth to prevent major cognitive and neurological effects. Dietary treatment, including natural protein restriction and Phe-free supplements, must be used to maintain blood Phe concentrations of 120-360 µmol/L throughout the life span. Additional treatments include the casein glycomacropeptide (GMP), which contains very limited aromatic amino acids and may improve immunological function, and large neutral amino acid (LNAA) supplementation to prevent plasma Phe transport into the brain. The synthetic BH4 analog, sapropterin hydrochloride (i.e., Kuvan®, BioMarin), is another potential treatment that activates residual PAH, thus decreasing Phe concentrations in the blood of PKU patients. Moreover, daily subcutaneous injection of pegylated Phe ammonia-lyase (i.e., pegvaliase; PALYNZIQ®, BioMarin) has promised gene therapy in recent clinical trials, and mRNA approaches are also being studied.


Asunto(s)
Fenilalanina Hidroxilasa , Fenilcetonurias , Humanos , Fenilalanina/metabolismo , Fenilalanina/uso terapéutico , Fenilalanina Hidroxilasa/genética , Fenilalanina Hidroxilasa/metabolismo , Fenilalanina Hidroxilasa/uso terapéutico , Fenilcetonurias/genética , Fenilcetonurias/terapia
3.
J Med Food ; 25(5): 487-494, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35325557

RESUMEN

Phenylketonuria (PKU) is an autosomal recessive disease caused by variants in the gene that encodes phenylalanine hydroxylase (PAH), limiting the metabolism of phenylalanine (Phe). When PAH activity is absent or hindered, Phe is not converted to tyrosine, leading to an accumulation of Phe in the blood, which can cause serious neurological complications. Once PKU is diagnosed, treatment should be started immediately, and the basis for this is dietary restriction of foods with high levels of Phe, associated with the use of protein substitutes and intake of foods with low protein content. This restriction accompanies patients throughout their lives, making their diets unpalatable and monotonous, which represents a major challenge for health professionals and patients, considering that these factors favor food transgression. In this context, the objective of this work was to carry out an integrative review based on evidence regarding the intake of fruits and vegetables, by phenylketonurics, taking into account the greater or lesser tolerance to Phe. Since, some researchers have dedicated themselves to evaluating the biochemical effect of unrestricted consumption of fruits and vegetables at PKU, unifying the information in this regard. It was observed that the intake of vegetable protein by patients with PKU has shown to be promising since the studies indicate that the intake of these proteins does not present adverse effects to the metabolic control of the Phe.


Asunto(s)
Fenilalanina , Fenilcetonurias , Dieta con Restricción de Proteínas , Frutas/metabolismo , Humanos , Fenilcetonurias/diagnóstico , Fenilcetonurias/genética , Verduras
4.
Metab Brain Dis ; 37(3): 743-760, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34997870

RESUMEN

BACKGROUND: Disorders of tetrahydrobiopterin metabolism represent a rare group of inherited neurotransmitter disorders that manifests mainly in infancy or childhood with developmental delay, neuroregression, epilepsy, movement disorders, and autonomic symptoms. METHODOLOGY: A retrospective review of genetically confirmed cases of disorders of tetrahydrobiopterin metabolism over a period of three years (Jan 2018 to Jan 2021) was performed across two paediatric neurology centres from South India. RESULTS: A total of nine patients(M:F=4:5) fulfilled the eligibility criteria. The genetic variants detected include homozygous mutations in the QDPR(n=6), GCH1(n=2), and PTS(n=1) genes. The median age at onset of symptoms was 6-months(range 3-78 months), while that at diagnosis was 15-months (8-120 months), resulting in a median delay in diagnosis of 9-months. The main clinical manifestations included neuroregression (89%), developmental delay(78%), dystonia(78%) and seizures(55%). Management strategies included a phenylalanine restricted diet, levodopa/carbidopa, 5-Hydroxytryphtophan, and folinic acid. Only, Patient-2 afforded and received BH4 supplementation at a sub-optimal dose later in the disease course. We had a median duration of follow up of 15 months (range 2-48 months). Though the biochemical response has been marked; except for patients with GTPCH deficiency, only mild clinical improvement was noted with regards to developmental milestones, seizures, or dystonia in others. CONCLUSION: Tetrahydrobiopterin deficiencies represent a rare yet potentially treatable cause for non-phenylketonuria hyperphenylalaninemia with better outcomes when treated early in life. Screening for disorders of biopterin metabolism in patients with hyperphenylalaninemia prevents delayed diagnosis. This study expands the genotype-phenotype spectrum of patients with disorders of tetrahydrobiopterin metabolism from South India.


Asunto(s)
Distonía , Fenilcetonurias , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Biopterinas/uso terapéutico , Niño , Preescolar , Distonía/genética , Femenino , Humanos , Lactante , Masculino , Fenilalanina , Fenilcetonurias/diagnóstico , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/genética
5.
Nat Metab ; 3(8): 1125-1132, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34294923

RESUMEN

Phenylketonuria (PKU) is a rare disease caused by biallelic mutations in the PAH gene that result in an inability to convert phenylalanine (Phe) to tyrosine, elevated blood Phe levels and severe neurological complications if untreated. Most patients are unable to adhere to the protein-restricted diet, and thus do not achieve target blood Phe levels. We engineered a strain of E. coli Nissle 1917, designated SYNB1618, through insertion of the genes encoding phenylalanine ammonia lyase and L-amino acid deaminase into the genome, which allow for bacterial consumption of Phe within the gastrointestinal tract. SYNB1618 was studied in a phase 1/2a randomized, placebo-controlled, double-blind, multi-centre, in-patient study ( NCT03516487 ) in adult healthy volunteers (n = 56) and patients with PKU and blood Phe level ≥600 mmol l-1 (n = 14). Participants were randomized to receive a single dose of SYNB1618 or placebo (part 1) or up to three times per day for up to 7 days (part 2). The primary outcome of this study was safety and tolerability, and the secondary outcome was microbial kinetics. A D5-Phe tracer (15 mg kg-1) was used to study exploratory pharmacodynamic effects. SYNB1618 was safe and well tolerated with a maximum tolerated dose of 2 × 1011 colony-forming units. Adverse events were mostly gastrointestinal and of mild to moderate severity. All participants cleared the bacteria within 4 days of the last dose. Dose-responsive increases in strain-specific Phe metabolites in plasma (trans-cinnamic acid) and urine (hippuric acid) were observed, providing a proof of mechanism for the potential to use engineered bacteria in the treatment of rare metabolic disorders.


Asunto(s)
Terapia Biológica/métodos , Escherichia coli , Fenilcetonurias/terapia , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Terapia Biológica/efectos adversos , Escherichia coli/enzimología , Escherichia coli/genética , Ingeniería Genética , Humanos , Fenilanina Amoníaco-Liasa/genética , Fenilanina Amoníaco-Liasa/metabolismo , Fenilcetonurias/sangre , Fenilcetonurias/genética , Resultado del Tratamiento
6.
J Food Biochem ; 44(3): e13151, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31960483

RESUMEN

Phenylketonuria (PKU) is an inborn disease caused by defective phenylalanine hydroxylase, which consequently results in the accumulation of phenylalanine in the brain leading to further complications. One of the promising approaches in dietary treatment is the supplementation of large neutral amino acid (LNAA). The LNAA compete with phenylalanine for the common L-type LNAA transporter across the blood-brain barrier, and decrease phenylalanine levels in the brain. In this study, the earlier LNAA-enriched protein model was improved (Protein Model-66) and validated in silico. The reverse translated and codon-optimized synthetic LNAA66 gene was cloned into pPICZαC and expressed in Pichia pastoris. The expressed protein was purified by His Select affinity chromatography. SDS-PAGE and Western blotting analysis showed a band at an expected molecular weight of 12 kDa, confirming the expression of the modeled protein. To our knowledge, this is the first report showing the cloning and expression of an in silico designed LNAA-enriched protein. PRACTICAL APPLICATIONS: One of the promising dietary treatment of phenylketonuria (PKU) is the supplementation of large neutral amino acid (LNAA), wherein high levels of LNAA compete with phenylalanine for the same L-type LNAA transporter, and consequently decrease phenylalanine accumulation in the brain, thereby decreasing neurological complications. For the first time, here, we are showing that an in silico designed and validated Protein Model-66, rich in LNAA, can be successfully cloned and expressed in Pichia pastoris. The complete biochemical and structural characterization of this protein will give a clear insight into its potential application for PKU treatment. The protein can be potentially used as a supplement to treat PKU to those who are non-adherent to the restricted, non-palatable, and expensive diet. Furthermore, this novel and effective strategy of in silico designing, cloning and expression can be exploited to develop proteins for various applications of industrial, food, medical, and academic relevance.


Asunto(s)
Aminoácidos Neutros , Fenilcetonurias , Clonación Molecular , Simulación por Computador , Humanos , Fenilcetonurias/genética , Saccharomycetales
8.
Nutrients ; 10(12)2018 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-30544518

RESUMEN

Children with phenylketonuria (PKU) follow a protein restricted diet with negligible amounts of docosahexaenoic acid (DHA). Low DHA intakes might explain subtle neurological deficits in PKU. We studied whether a DHA supply modified plasma DHA and neurological and intellectual functioning in PKU. In a double-blind multicentric trial, 109 PKU patients were randomized to DHA doses from 0 to 7 mg/kg&day for six months. Before and after supplementation, we determined plasma fatty acid concentrations, latencies of visually evoked potentials, fine and gross motor behavior, and IQ. Fatty acid desaturase genotypes were also determined. DHA supplementation increased plasma glycerophospholipid DHA proportional to dose by 0.4% DHA per 1 mg intake/kg bodyweight. Functional outcomes were not associated with DHA status before and after intervention and remained unchanged by supplementation. Genotypes were associated with plasma arachidonic acid levels and, if considered together with the levels of the precursor alpha-linolenic acid, also with DHA. Functional outcomes and supplementation effects were not significantly associated with genotype. DHA intakes up to 7 mg/kg did not improve neurological functions in PKU children. Nervous tissues may be less prone to low DHA levels after infancy, or higher doses might be required to impact neurological functions. In situations of minimal dietary DHA, endogenous synthesis of DHA from alpha-linolenic acid could relevantly contribute to DHA status.


Asunto(s)
Cognición/efectos de los fármacos , Ácidos Docosahexaenoicos/sangre , Ácidos Docosahexaenoicos/uso terapéutico , Destreza Motora/efectos de los fármacos , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/fisiopatología , Adolescente , Niño , Ácido Graso Desaturasas/genética , Femenino , Humanos , Masculino , Fenilcetonurias/epidemiología , Fenilcetonurias/genética
9.
J Inherit Metab Dis ; 41(4): 709-718, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29520738

RESUMEN

Hyperphenylalaninemia (HPA) caused by hepatic phenylalanine hydroxylase (PAH) deficiency has severe consequences on brain monoamine neurotransmitter metabolism. We have studied monoamine neurotransmitter status and the effect of tetrahydrobiopterin (BH4) treatment in Pahenu1/enu2 (ENU1/2) mice, a model of partial PAH deficiency. These mice exhibit elevated blood L-phenylalanine (L-Phe) concentrations similar to that of mild hyperphenylalaninemia (HPA), but brain levels of L-Phe are still ~5-fold elevated compared to wild-type. We found that brain L-tyrosine, L-tryptophan, BH4 cofactor and catecholamine concentrations, and brain tyrosine hydroxylase (TH) activity were normal in these mice but that brain serotonin, 5-hydroxyindolacetic acid (5HIAA) and 3-methoxy-4-hydroxyphenylglycol (MHPG) content, and brain TH protein, as well as tryptophan hydroxylase type 2 (TPH2) protein levels and activity were reduced in comparison to wild-type mice. Parenteral L-Phe loading conditions did not lead to significant changes in brain neurometabolite concentrations. Remarkably, enteral BH4 treatment, which normalized brain L-Phe levels in ENU1/2 mice, lead to only partial recovery of brain serotonin and 5HIAA concentrations. Furthermore, indirect evidence indicated that the GTP cyclohydrolase I (GTPCH) feedback regulatory protein (GFRP) complex may be a sensor for brain L-Phe elevation to ameliorate the toxic effects of HPA. We conclude that BH4 treatment of HPA toward systemic L-Phe lowering reverses elevated brain L-Phe content but the recovery of TPH2 protein and activity as well as serotonin levels is suboptimal, indicating that patients with mild HPA and mood problems (depression or anxiety) treated with the current diet may benefit from supplementation with BH4 and 5-OH-tryptophan.


Asunto(s)
Biopterinas/análogos & derivados , Encéfalo/metabolismo , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/metabolismo , Serotonina/metabolismo , Animales , Biopterinas/farmacología , Modelos Animales de Enfermedad , Dopamina/metabolismo , Humanos , Ratones , Ratones Mutantes , Neurotransmisores/metabolismo , Fenilalanina/sangre , Fenilalanina/metabolismo , Fenilalanina Hidroxilasa/metabolismo , Fenilcetonurias/genética , Triptófano Hidroxilasa/metabolismo , Tirosina 3-Monooxigenasa/metabolismo
10.
Mol Genet Metab ; 123(1): 1-5, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29174366

RESUMEN

Patients with hyperphenylalaninemia (HPA) are detected through newborn screening for phenylketonuria (PKU). HPA is known to be caused by deficiencies of the enzyme phenylalanine hydroxylase (PAH) or its cofactor tetrahydrobiopterin (BH4). Current guidelines for the differential diagnosis of HPA would, however, miss a recently described DNAJC12 deficiency. The co-chaperone DNAJC12 is, together with the 70kDa heat shock protein (HSP70), responsible for the proper folding of PAH. All DNAJC12-deficient patients investigated to date responded to a challenge with BH4 by lowering their blood phenylalanine levels. In addition, the patients presented with low levels of biogenic amine in CSF and responded to supplementation with BH4, L-dopa/carbidopa and 5-hydroxytryptophan. The phenotypic spectrum ranged from mild autistic features or hyperactivity to severe intellectual disability, dystonia and parkinsonism. Late diagnosis result in permanent neurological disability, while early diagnosed and treated patients develop normally. Molecular diagnostics for DNAJC12 variants are thus mandatory in all patients in which deficiencies of PAH and BH4 are genetically excluded.


Asunto(s)
Fenilalanina Hidroxilasa/genética , Fenilalanina/genética , Fenilcetonurias/genética , Proteínas Represoras/genética , Aminas Biogénicas/líquido cefalorraquídeo , Humanos , Recién Nacido , Levodopa/genética , Levodopa/metabolismo , Tamizaje Neonatal , Patología Molecular , Fenilalanina/metabolismo , Fenilalanina Hidroxilasa/metabolismo , Fenilcetonurias/líquido cefalorraquídeo , Fenilcetonurias/diagnóstico , Fenilcetonurias/patología , Pliegue de Proteína , Proteínas Represoras/deficiencia
11.
Orphanet J Rare Dis ; 12(1): 132, 2017 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-28724394

RESUMEN

BACKGROUND: Newborn Screening Connect (NBS Connect) is a web-based self-reported patient registry and resource for individuals and families affected by disorders included in the newborn screening panel. NBS Connect was launched in 2012 by Emory University after years of planning and grassroots work by professionals, consumers, and industry. Individuals with phenylketonuria (PKU), maple syrup urine disease (MSUD) or tyrosinemia (TYR) have been recruited through distribution of outreach materials, presentations at parent organization meetings and direct recruitment at clinic appointments. Participants complete online profiles generating data on diagnosis, treatment, symptoms, outcomes, barriers to care, and quality of life. Resources such as education materials, information on the latest research and clinical trials, recipes, interactive health tracking systems, and professional support tools are described. In addition, to examine the ability of NBS Connect to generate data that guides hypothesis-driven research, data pertaining to age at diagnosis, bone health, and skin conditions in individuals with PKU were assessed. The objective of this paper is to describe the development of NBS Connect and highlight its data, resources and research contributions. RESULTS: In September 2016, NBS Connect had 442 registered participants: 314 (71%) individuals with PKU, 68 (15%) with MSUD, 20 (5%) with TYR, and 40 (9%) with other disorders on the NBS panel. Age at diagnosis was less than 4 weeks in 285 (89%) of 319 respondents to this question and between 1 month and 14 years in 29 (9%) individuals. Of 216 respondents with PKU, 33 (15%) had a DXA scan in the past year. Of 217 respondents with PKU, 99 (46%) reported at least one skin condition. CONCLUSIONS: NBS Connect was built and refined with feedback from all stakeholders, including individuals with inherited metabolic disorders. Based on patient-reported data, future studies can be initiated to test hypotheses such as the relationship between PKU and skin conditions. Patient registries like NBS Connect can inform hypothesis-driven research, contributing to knowledge generation and following the current trend in moving from traditional medicine towards evidence-based practice. NBS Connect will help clinicians understand long-term outcomes of rare disorders, contributing to better patient care and quality of life.


Asunto(s)
Enfermedad de la Orina de Jarabe de Arce/genética , Tamizaje Neonatal , Fenilcetonurias/genética , Enfermedades Raras , Sistema de Registros , Tirosinemias/genética , Humanos , Recién Nacido , Internet
12.
Mol Genet Metab ; 121(2): 104-110, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28465125

RESUMEN

OBJECTIVE: Determine whether body composition as it relates to dietary protein in patients with phenylalanine hydroxylase (PAH) deficiency is associated with genotype, dietary factors, and lifestyle choices. METHODS: We examined associations between protein intake (intact and medical foods: MF) and body composition in PAH-deficient patients along with, physical activity, and genotype. Protein intakes (total, intact, and MF) were analysed from three-day food records with Nutrition Data System for Research (NDSR) in 59 children and 27 adults (N=86, median age=16.0years). The severity of PAH deficiency was classified using the genotype assigned value method (AV sum). Physical activity was assessed using a study-developed question (light vs. intense activity). Body composition was measured by DXA, including android:gynoid ratio (A:G), fat-free mass index (FFMI), fat mass index (FMI), and FMI:FFMI ratio. RESULTS: High intact protein intake was associated with high FFMI (rs=0.75, p=0.008) and low FMI:FFMI (rs=-0.59, p=0.04) in adults. Only in children, MF protein (rs=0.38, p=0.04) was directly proportional to FFMI. Median intact protein intakes of adults (25.1 vs. 9.9g/d, p<0.001) and children (11 vs. 6g/d, p<0.001) were higher than prescribed. Only in adults, the actual median MF protein intake was lower than prescribed (53 vs. 60g/d, p=0.03). In adults and children, light activity was associated with higher fat mass indices compared to intense activity (adults: FMI:FFMI: ß=1.1, p=0.001, children: FMI:FFMI: ß=1.1, p=0.007; FMI ß=2.1, p=0.01; A:G ß=1.1, p=0.04). All associations remained significant after covariate adjustment. Genotype was not associated with body composition. CONCLUSIONS: Although fat-free mass in adults was positively associated with intact protein intake, it should be consumed as prescribed per individual tolerance to maintain plasma Phe concentrations within treatment range. In children, total protein maximized with MF should be encouraged to promote lean mass. Nutrition counselling could be complemented with physical activity recommendations for optimal clinical outcomes.


Asunto(s)
Composición Corporal , Proteínas en la Dieta/administración & dosificación , Ejercicio Físico , Fenilcetonurias/genética , Fenilcetonurias/fisiopatología , Adolescente , Adulto , Índice de Masa Corporal , Niño , Preescolar , Proteínas en la Dieta/análisis , Femenino , Genotipo , Humanos , Estilo de Vida , Masculino , Persona de Mediana Edad , Terapia Nutricional , Obesidad , Fenilcetonurias/metabolismo , Adulto Joven
13.
J Mol Cell Cardiol ; 87: 152-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26254182

RESUMEN

Rupture of abdominal aortic aneurysm (AAA) is a lethal event. No oral medicine has been available to prevent or treat AAA. We have recently identified a novel mechanism of eNOS uncoupling by which AAA develops, in angiotensin II (Ang II) infused hyperphenylalaninemia 1 (hph-1) mice. Using this unique model we investigated effects on AAA formation of the L-type calcium channel blocker nifedipine, in view of the unclear relationship between hypertension and AAA, and unclear mechanisms of aneurysm protective effects of some blood pressure lowering drugs. Six-month old hph-1 mice were infused with Ang II (0.7 mg/kg/day) for 2 weeks, and fed nifedipine chow at two different doses (5 and 20 mg/kg/day). While the high dose of nifedipine reduced blood pressure, the lower dose had no effect. Interestingly, the incidence rate of AAA dropped from 71% to 7 and 12.5% for low and high dose nifedipine, respectively. Expansion of abdominal aorta, determined by ultrasound imaging, was abolished by both doses of nifedipine, which recoupled eNOS completely to improve NO bioavailability. Both also abrogated aortic superoxide production. Of note, Ang II activation of NADPH oxidase in vascular smooth muscle cells and endothelial cells, known to uncouple eNOS, was also attenuated by nifedipine. Although low dose was a sub-pressor while the high dose reduced blood pressure via inhibition of calcium channels, both doses were highly effective in preventing AAA by preserving eNOS coupling activity to eliminate sustained oxidative stress from uncoupled eNOS. These data demonstrate that oral treatment of nifedipine is highly effective in preserving eNOS function to attenuate AAA formation. Nifedipine may be used for AAA prevention either at low dose in AAA risk group, or at high dose in patients with co-existing hypertension.


Asunto(s)
Antihipertensivos/administración & dosificación , Aneurisma de la Aorta Abdominal/tratamiento farmacológico , Nifedipino/administración & dosificación , Óxido Nítrico Sintasa de Tipo III/metabolismo , Angiotensina II/genética , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/patología , Canales de Calcio Tipo L/efectos de los fármacos , Humanos , Hipertensión/tratamiento farmacológico , Hipertensión/patología , Ratones , Ratones Transgénicos , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Estrés Oxidativo/efectos de los fármacos , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/genética , Fenilcetonurias/patología
14.
Sci Rep ; 4: 6941, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25373594

RESUMEN

Although it is recognized that the abnormal accumulation of amino acid is a cause of the symptoms in metabolic disease such as phenylketonuria (PKU), the relationship between disease severity and serum amino acid levels is not well understood due to the lack of experimental model. Here, we present a novel in vitro cellular model using K562-D cells that proliferate slowly in the presence of excessive amount of phenylalanine within the clinically observed range, but not phenylpyruvate. The increased expression of the L-type amino acid transporter (LAT2) and its adapter protein 4F2 heavy chain appeared to be responsible for the higher sensitivity to phenylalanine in K562-D cells. Supplementation with valine over phenylalanine effectively restored cell proliferation, although other amino acids did not improve K562-D cell proliferation over phenylalanine. Biochemical analysis revealed mammalian target of rapamycin complex (mTORC) as a terminal target of phenylalanine in K562-D cell proliferation, and supplementation of valine restored mTORC1 activity. Our results show that K562-D cell can be a potent tool for the investigation of PKU at the molecular level and to explore new therapeutic approaches to the disease.


Asunto(s)
Modelos Biológicos , Fenilalanina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Transporte Biológico , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteína-1 Reguladora de Fusión/genética , Proteína-1 Reguladora de Fusión/metabolismo , Regulación de la Expresión Génica , Hemina/farmacología , Humanos , Células K562 , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Fenilalanina/farmacología , Fenilcetonurias/genética , Fenilcetonurias/metabolismo , Fenilcetonurias/patología , Ácidos Fenilpirúvicos/metabolismo , Ácidos Fenilpirúvicos/farmacología , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Valina/metabolismo , Valina/farmacología
15.
J Inherit Metab Dis ; 37(5): 735-43, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24487571

RESUMEN

Monoamine neurotransmitter deficiency has been implicated in the etiology of neuropsychiatric symptoms associated with chronic hyperphenylalaninemia in phenylketonuria (PKU). Two proposed explanations for neurotransmitter deficiency in PKU include first, that chronically elevated blood L-phenylalanine (Phe) inhibits the transport of L-tyrosine (Tyr) and L-tryptophan (Trp), the substrates for dopamine and serotonin synthesis respectively, into brain. In the second hypothesis, elevated Phe competitively inhibits brain tyrosine hydroxylase (TH) and tryptophan hydroxylase (TPH) activities, the rate limiting steps in dopamine and serotonin synthesis. Dietary supplementation with large neutral amino acids (LNAA) including Tyr and Trp has been recommended for individuals with chronically elevated blood Phe in an attempt to restore amino acid and monoamine homeostasis in brain. As a potential alternative treatment approach, we demonstrate that pharmacologic inhibition of Tyr degradation through oral administration of nitisinone (NTBC) yielded sustained increases in blood and brain Tyr, decreased blood and brain Phe, and consequently increased dopamine synthesis in a murine model of PKU. Our results suggest that Phe-mediated inhibition of TH activity is the likely mechanism of impaired dopamine synthesis in PKU. Pharmacologic inhibition of Tyr degradation may be a promising adjunct therapy for CNS monoamine neurotransmitter deficiency in hyperphenylalaninemic individuals with PKU.


Asunto(s)
Química Encefálica/efectos de los fármacos , Ciclohexanonas/uso terapéutico , Dopamina/deficiencia , Inhibidores Enzimáticos/uso terapéutico , Nitrobenzoatos/uso terapéutico , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/metabolismo , Tirosina/metabolismo , Aminoácidos/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neurotransmisores/deficiencia , Fenilcetonurias/genética
16.
Brain Dev ; 36(3): 268-71, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23660475

RESUMEN

Autosomal recessive guanosine triphosphate cyclohydrolase I (GTPCH) deficiency is an inborn error of tetrahydrobiopterin (BH4) synthesis from GTP. GTPCH deficiency causes severe reduction of BH4, resulting in hyperphenylalaninemia (HPA) and decreased dopamine and serotonin synthesis. Without treatment, a patient with GTPCH deficiency develops complex neurological dysfunctions, including dystonia and developmental delays. The first Japanese patient with GTPCH deficiency was discovered by HPA during asymptomatic newborn screening. The phenylalanine level at the age of 5days was 1273µmol/L (cutoff value, 180.0µmol/L). The high serum phenylalanine level was decreased to normal after adequate BH4 oral supplementation. Serum and urinary pteridine examination revealed very low levels of neopterin and biopterin. Sequence analysis of GCH1 revealed compound heterozygous point mutations, including a novel point mutation (p.R235W). Replacement therapy with BH4 and L-dopa/carbidopa were started at the age of 1month, and 5-hydroxytryptophan (5-HTP) was started at the age of 5months. At 10months of age, the patient showed slight dystonia but no obvious developmental delay. Cerebrospinal fluid should be examined to determine the appropriate dosage of supplement drugs. In conclusion, it is important to control the serum phenylalanine level and perform early replacement of neurotransmitters to prevent neurological dysfunction.


Asunto(s)
Biopterinas/análogos & derivados , GTP Ciclohidrolasa/genética , Fenilcetonurias/genética , Fenilcetonurias/terapia , Biopterinas/uso terapéutico , Análisis Mutacional de ADN , Humanos , Recién Nacido , Japón , Masculino , Tamizaje Neonatal , Fenilalanina/sangre , Fenilcetonurias/sangre , Fenilcetonurias/diagnóstico , Mutación Puntual , Factores de Tiempo , Resultado del Tratamiento
17.
Acta Biochim Pol ; 60(4): 613-6, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24350308

RESUMEN

Tetrahydrobiopterin (BH4) has been recently approved as a treatment of patients with phenylketonuria. However, as a confirmation of BH4-responsiveness, it might require a very expensive trial treatment with BH4 or prolonged BH4-loading procedures. The selection of patients eligible for BH4-therapy by means of genotyping of the PAH gene mutations may be recommended as a complementary approach. A population-wide genotyping study was carried out in 1286 Polish phenyloketonuria-patients. The aim was to estimate the BH4 demand and to cover prospectively the treatment by a National Health Fund. A total of 95 types of mutations were identified. Genetic variants corresponding with probable BH4-responsiveness were found in 28.2% of cases. However, patients with mild or classical phenylketonuria who require continuous treatment accounted for 11.4% of the studied population only. Analysis of the published data shows similar percentage of the "BH4-responsive" variants of a PAH gene in patients from other countries of Eastern Europe. Therefore, it can be concluded, that the proportion of phenylketonuria-patients who could benefit from the use of BH4 reaches approximately 10% in the entire region.


Asunto(s)
Biopterinas/análogos & derivados , Mutación/genética , Fenilalanina Hidroxilasa/genética , Fenilcetonurias/tratamiento farmacológico , Biomarcadores Farmacológicos , Biopterinas/administración & dosificación , Genotipo , Humanos , Fenilalanina/deficiencia , Fenilalanina/genética , Fenilcetonurias/genética , Polonia
18.
J Inherit Metab Dis ; 36(6): 955-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23532445

RESUMEN

Phenylketonuria (PKU, OMIM 261600) is an autosomal recessive inborn error of phenylalanine metabolism, predominantly caused by mutations in the phenylalanine hydroxylase (PAH) gene. Approximately 10% of patients carry a nonsense mutation, which results in an inactive or unstable truncated protein. In some genetic disorders, including cystic fibrosis and Duchenne muscular dystrophy, restoration of full-length protein has been achieved by aminoglycoside antibiotics, such as gentamicin and G-418 (Geneticin). More recently, nonsense read-through has been induced at greater rates using a non-aminoglycoside drug, PTC124 (Ataluren), which has the advantage of being non-toxic in contrast to the antibiotics. The efficacy of read-through induced by three compounds, aminoglycosides G418 and gentamicin, and PTC124 were evaluated for four nonsense mutations of PAH in an in vitro expression system in two mammalian cell lines (COS-7 and HEK293). The production of full-length PAH was investigated using western blotting and the functionality confirmed by enzyme activity. Gentamicin and G-418 induced read-through of nonsense PAH mutations in HEK293 cells. The read-through product partially restored enzymatic activity, which was significantly less than that of wild-type, but comparable to a missense mutation of PAH associated with less severe forms of PKU. Treatment with PTC124 up to 100 µM did not result in full-length PAH polypeptide. Nonsense read-through drugs are a potential form of treatment for PKU, although the high dosage of aminoglycosides used is not appropriate in a clinical setting. In vitro studies with new non-toxic read-through agents as well as in vivo studies would also be essential to determine the extent of read-through required to restore normal phenylalanine levels.


Asunto(s)
Aminoglicósidos/uso terapéutico , Codón sin Sentido , Gentamicinas/uso terapéutico , Oxadiazoles/uso terapéutico , Fenilalanina Hidroxilasa/genética , Fenilcetonurias/tratamiento farmacológico , Aminoglicósidos/genética , Animales , Células COS , Chlorocebus aethiops , Codón sin Sentido/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Terapia Genética/métodos , Células HEK293 , Humanos , Fenilcetonurias/genética , Fenilcetonurias/patología
19.
IUBMB Life ; 65(4): 341-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23457044

RESUMEN

Mammalian phenylalanine hydroxylase (PAH) catalyzes the rate-limiting step in the phenylalanine catabolism, consuming about 75% of the phenylalanine input from the diet and protein catabolism under physiological conditions. In humans, mutations in the PAH gene lead to phenylketonuria (PKU), and most mutations are mainly associated with PAH misfolding and instability. The established treatment for PKU is a phenylalanine-restricted diet and, recently, supplementation with preparations of the natural tetrahydrobiopterin cofactor also shows effectiveness for some patients. Since 1997 there has been a significant increase in the understanding of the structure, catalytic mechanism, and regulation of PAH by its substrate and cofactor, in addition to improved correlations between genotype and phenotype in PKU. Importantly, there has also been an increased number of studies on the structure and function of PAH from bacteria and lower eukaryote organisms, revealing an additional anabolic role of the enzyme in the synthesis of melanin-like pigments. In this review, we discuss these recent studies, which contribute to define the evolutionary adaptation of the PAH structure and function leading to sophisticated regulation for effective catabolic processing of phenylalanine in mammalian organisms.


Asunto(s)
Fenilalanina Hidroxilasa/química , Fenilalanina/metabolismo , Fenilcetonurias/genética , Animales , Biopterinas/análogos & derivados , Biopterinas/química , Humanos , Cinética , Melaninas/biosíntesis , Melaninas/química , Mutación , Fenilalanina/química , Fenilalanina Hidroxilasa/genética , Fenilalanina Hidroxilasa/metabolismo , Conformación Proteica , Pliegue de Proteína
20.
J Inherit Metab Dis ; 36(3): 513-23, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22976763

RESUMEN

Transport of large neutral amino acids (LNAA) across the blood brain barrier (BBB) is facilitated by the L-type amino acid transporter, LAT1. Peripheral accumulation of one LNAA (e.g., phenylalanine (phe) in PKU) is predicted to increase uptake of the offending amino acid to the detriment of others, resulting in disruption of brain amino acid homeostasis. We hypothesized that selected non-physiological amino acids (NPAAs) such as DL-norleucine (NL), 2-aminonorbornane (NB; 2-aminobicyclo-(2,1,1)-heptane-2-carboxylic acid), 2-aminoisobutyrate (AIB), and N-methyl-aminoisobutyrate (MAIB), acting as competitive inhibitors of various brain amino acid transporters, could reduce brain phe in Pah (enu2) mice, a relevant murine model of PKU. Oral feeding of 5 % NL, 5 % AIB, 0.5 % NB and 3 % MAIB reduced brain phe by 56 % (p < 0.01), -1 % (p = NS), 27 % (p < 0.05) and 14 % (p < 0.01), respectively, compared to untreated subjects. Significant effects on other LNAAs (tyrosine, methionine, branched chain amino acids) were also observed, however, with MAIB displaying the mildest effects. Of interest, MAIB represents an inhibitor of the system A (alanine) transporter that primarily traffics small amino acids and not LNAAs. Our studies represent the first in vivo use of these NPAAs in Pah (enu2) mice, and provide proof-of-principle for their further preclinical development, with the long-term objective of identifying NPAA combinations and concentrations that selectively restrict brain phe transport while minimally impacting other LNAAs and downstream intermediates.


Asunto(s)
Aminoácidos/uso terapéutico , Encéfalo/metabolismo , Fenilalanina/metabolismo , Fenilcetonurias/tratamiento farmacológico , Aminoácidos Cíclicos/uso terapéutico , Ácidos Aminoisobutíricos/uso terapéutico , Animales , Encéfalo/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Terapia Molecular Dirigida , Norleucina/uso terapéutico , Fenilcetonurias/genética , Fenilcetonurias/metabolismo , Fenilcetonurias/patología , Proyectos Piloto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA