Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros

Medicinas Tradicionales
Medicinas Complementárias
Intervalo de año de publicación
1.
J Inherit Metab Dis ; 46(4): 573-585, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36880400

RESUMEN

The inborn error of metabolism phenylketonuria (PKU, OMIM 261600) is most often due to inactivation of phenylalanine hydroxylase (PAH), which converts phenylalanine (Phe) into tyrosine (Tyr). The reduced PAH activity increases blood concentration of phenylalanine and urine levels of phenylpyruvate. Flux balance analysis (FBA) of a single-compartment model of PKU predicts that maximum growth rate should be reduced unless Tyr is supplemented. However, the PKU phenotype is lack of development of brain function specifically, and Phe reduction rather than Tyr supplementation cures the disease. Phe and Tyr cross the blood-brain barrier (BBB) through the aromatic amino acid transporter implying that the two transport reactions interact. However, FBA does not accommodate such competitive interactions. We here report on an extension to FBA that enables it to deal with such interactions. We built a three-compartment model, made the common transport across the BBB explicit, and included dopamine and serotonin synthesis as parts of the brain function to be delivered by FBA. With these ramifications, FBA of the genome-scale metabolic model extended to three compartments does explain that (i) the disease is brain specific, (ii) phenylpyruvate in urine is a biomarker, (iii) excess of blood-phenylalanine rather than shortage of blood-tyrosine causes brain pathology, and (iv) Phe deprivation is the better therapy. The new approach also suggests (v) explanations for differences in pathology between individuals with the same PAH inactivation, and (vi) interference of disease and therapy with the functioning of other neurotransmitters.


Asunto(s)
Fenilalanina Hidroxilasa , Fenilcetonurias , Humanos , Fenilcetonurias/metabolismo , Ácidos Fenilpirúvicos , Fenilalanina Hidroxilasa/genética , Fenilalanina , Tirosina/metabolismo
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.
PLoS One ; 17(1): e0261150, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35015767

RESUMEN

INTRODUCTION: Management of phenylketonuria (PKU) is mainly achieved through dietary control with limited intake of phenylalanine (Phe) from food, supplemented with low protein (LP) food and a mixture of free synthetic (FS) amino acids (AA) (FSAA). Casein glycomacropeptide (CGMP) is a natural peptide released in whey during cheese making by the action of the enzyme chymosin. Because CGMP in its pure form does not contain Phe, it is nutritionally suitable as a supplement in the diet for PKU when enriched with specific AAs. Lacprodan® CGMP-20 (= CGMP) used in this study contained only trace amounts of Phe due to minor presence of other proteins/peptides. OBJECTIVE: The aims were to address the following questions in a classical PKU mouse model: Study 1, off diet: Can pure CGMP or CGMP supplemented with Large Neutral Amino Acids (LNAA) as a supplement to normal diet significantly lower the content of Phe in the brain compared to a control group on normal diet, and does supplementation of selected LNAA results in significant lower brain Phe level?. Study 2, on diet: Does a combination of CGMP, essential (non-Phe) EAAs and LP diet, provide similar plasma and brain Phe levels, growth and behavioral skills as a formula which alone consist of FSAA, with a similar composition?. MATERIAL AND METHODS: 45 female mice homozygous for the Pahenu2 mutation were treated for 12 weeks in five different groups; G1(N-CGMP), fed on Normal (N) casein diet (75%) in combination with CGMP (25%); G2 (N-CGMP-LNAA), fed on Normal (N) casein diet (75%) in combination with CGMP (19,7%) and selected LNAA (5,3% Leu, Tyr and Trp); G3 (N), fed on normal casein diet (100%); G4 (CGMP-EAA-LP), fed on CGMP (70,4%) in combination with essential AA (19,6%) and LP diet; G5 (FSAA-LP), fed on FSAA (100%) and LP diet. The following parameters were measured during the treatment period: Plasma AA profiles including Phe and Tyr, growth, food and water intake and number of teeth cut. At the end of the treatment period, a body scan (fat and lean body mass) and a behavioral test (Barnes Maze) were performed. Finally, the brains were examined for content of Phe, Tyr, Trp, dopamine (DA), 3,4-dihydroxyphenylacetic acid (DOPAC), serotonin (5-HT) and 5-hydroxyindole-acetic acid (5-HIAA), and the bone density and bone mineral content were determined by dual-energy x-ray absorptiometry. RESULTS: Study 1: Mice off diet supplemented with CGMP (G1 (N-CGMP)) or supplemented with CGMP in combination with LNAA (G2 (N-CGMP-LNAA)) had significantly lower Phe in plasma and in the brain compared to mice fed only casein (G3 (N)). Extra LNAA (Tyr, Trp and Leu) to CGMP did not have any significant impact on Phe levels in the plasma and brain, but an increase in serotonin was measured in the brain of G2 mice compared to G1. Study 2: PKU mice fed with mixture of CGMP and EAA as supplement to LP diet (G4 (CGMP-EAA-LP)) demonstrated lower plasma-Phe levels but similar brain- Phe levels and growth as mice fed on an almost identical combination of FSAA (G5 (FSAA-LP)). CONCLUSION: CGMP can be a relevant supplement for the treatment of PKU.


Asunto(s)
Aminoácidos/uso terapéutico , Caseínas/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Fenilcetonurias/dietoterapia , Aminoácidos/sangre , Aminoácidos/síntesis química , Animales , Densidad Ósea , Huesos/diagnóstico por imagen , Huesos/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Suplementos Dietéticos , Modelos Animales de Enfermedad , Femenino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Fenilalanina/análisis , Fenilalanina/sangre , Fenilalanina Hidroxilasa/deficiencia , Fenilalanina Hidroxilasa/genética , Serotonina/sangre , Tirosina/sangre
4.
J Acad Nutr Diet ; 122(7): 1283-1295.e3, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34968752

RESUMEN

BACKGROUND: Phenylketonuria (PKU) is an inborn error of metabolism that impairs the function of the enzyme phenylalanine hydroxylase. Historical treatment includes limiting dietary phenylalanine (Phe) consumption while supplementing with medical food; however, this treatment has been associated with complications, such as nutritional deficiencies and disruptions in the gut microbiota. OBJECTIVE: The study aim was to compare dietary and gut microbiome differences between adult patients on a traditional PKU diet with those receiving the enzyme substitution therapy Palynziq on a liberalized diet while controlling blood Phe levels to <600 µmol/L (to convert to mg/dL divide by 60.5). DESIGN: A cross-sectional study was conducted comparing patients on a traditional Phe-restricted diet with patients receiving Palynziq eating a liberalized diet. PARTICIPANTS/SETTING: Six patients eating a traditional Phe-restricted diet with medical food and 6 patients on Palynziq eating a liberalized diet without medical food intake for more than 3 years were selected from the University of Kentucky Metabolic Clinic from August to December 2019. MAIN OUTCOME MEASURES: Nutrient intake from 3-day diet records and fecal microbiome taxonomic abundances were analyzed. STATISTICAL ANALYSIS: Mann-Whitney U tests were used for dietary data analysis. Differential abundance analysis for microbiome taxa and pathway data was done using DESeq2 analysis. RESULTS: Dietary data showed patients receiving Palynziq consumed a lower percent of kilocalories from total protein and lower amounts of most micronutrients, but consumed greater amounts of intact protein and cholesterol (P < .05). Microbiome data revealed a greater abundance of the phylum Verrucomicrobia and genus Lachnobacterium in the Traditional group and a greater abundance of the genus Prevotella in the Palynziq group (P < .05). Pathway analysis depicted greater enrichment in carotenoid and amino acid metabolism pathways in the Traditional group (P < .05). Protein (% kcal), dietary fiber (g), fat (% kcal), linolenic acid (% Dietary Reference Intakes), and age were correlated with the underlying microbial community structure for both groups combined. CONCLUSIONS: Patients with PKU treated with Palynziq on a liberalized diet manifest significant differences in diet composition compared with those treated with traditional Phe-restricted diets. Several of these dietary differences may affect the microbiome architecture.


Asunto(s)
Dieta , Terapia de Reemplazo Enzimático , Microbioma Gastrointestinal , Fenilanina Amoníaco-Liasa , Fenilcetonurias , Adulto , Estudios Transversales , Humanos , Fenilalanina , Fenilanina Amoníaco-Liasa/uso terapéutico , Fenilalanina Hidroxilasa , Fenilcetonurias/terapia , Proteínas Recombinantes/uso terapéutico
5.
Int J Mol Sci ; 22(15)2021 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-34360752

RESUMEN

Polymeric-based nano drug delivery systems have been widely exploited to overcome protein instability during formulation. Presently, a diverse range of polymeric agents can be used, among which polysaccharides, such as chitosan (CS), hyaluronic acid (HA) and cyclodextrins (CDs), are included. Due to its unique biological and physicochemical properties, CS is one of the most used polysaccharides for development of protein delivery systems. However, CS has been described as potentially immunogenic. By envisaging a biosafe cytocompatible and haemocompatible profile, this paper reports the systematic development of a delivery system based on CS and derived with HA and CDs to nanoencapsulate the model human phenylalanine hydroxylase (hPAH) through ionotropic gelation with tripolyphosphate (TPP), while maintaining protein stability and enzyme activity. By merging the combined set of biopolymers, we were able to effectively entrap hPAH within CS nanoparticles with improvements in hPAH stability and the maintenance of functional activity, while simultaneously achieving strict control of the formulation process. Detailed characterization of the developed nanoparticulate systems showed that the lead formulations were internalized by hepatocytes (HepG2 cell line), did not reveal cell toxicity and presented a safe haemocompatible profile.


Asunto(s)
Quitosano , Enzimas Inmovilizadas , Ensayo de Materiales , Nanopartículas/química , Fenilalanina Hidroxilasa , Quitosano/química , Quitosano/farmacología , Evaluación Preclínica de Medicamentos , Estabilidad de Enzimas , Enzimas Inmovilizadas/química , Enzimas Inmovilizadas/farmacología , Células HEK293 , Células Hep G2 , Humanos , Fenilalanina Hidroxilasa/química , Fenilalanina Hidroxilasa/farmacología
6.
Orphanet J Rare Dis ; 15(1): 171, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32605583

RESUMEN

BACKGROUND: Phenylketonuria (PKU) is an autosomal recessive inborn error of phenylalanine metabolism caused by deficiency in the enzyme phenylalanine hydroxylase that converts phenylalanine into tyrosine. MAIN BODY: In 2017 the first European PKU Guidelines were published. These guidelines contained evidence based and/or expert opinion recommendations regarding diagnosis, treatment and care for patients with PKU of all ages. This manuscript is a supplement containing the practical application of the dietary treatment. CONCLUSION: This handbook can support dietitians, nutritionists and physicians in starting, adjusting and maintaining dietary treatment.


Asunto(s)
Fenilalanina Hidroxilasa , Fenilcetonurias , Dieta , Humanos , Fenilalanina , Tirosina
7.
J Pediatr Endocrinol Metab ; 33(12): 1519-1523, 2020 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-33581706

RESUMEN

OBJECTIVES: Phenylalanine (Phe) hydroxylase (PAH) deficiency leads to hyperphenylalaninemia (HPA) and tyrosine (Tyr) depletion. We investigated Tyr homeostasis in patients with PAH deficiency and the effect of a slow-release amino acids therapy in phenylketonuria (PKU). METHODS: We performed four complementary investigations: (1) Tyr concentrations were monitored in 114 patients (10.6 ± 11.9 years) with PKU on dietary treatment supplemented with traditional amino acid formulations (n=52, 1175 samples) or non-PKU HPA on a free diet (n=62, 430 samples); (2) Tyr metabolism in PKU was quantitatively evaluated in three patients by a simple Tyr oral loading test (100 mg/kg); (3) diurnal and (4) long-term Tyr concentrations were evaluated in 5 and 13 patients with PKU, respectively, who switched from traditional to slow-release amino acids therapy. RESULTS: 1) Tyr concentrations in the PKU population were subnormal and significantly lower than in non-PKU HPA (p<0.01); (2) the response to a Tyr loading test in PKU was normal, with basal Tyr concentrations reached within 12 h; (3) the diurnal metabolic profile in patients on slow-release amino acids therapy revealed higher morning fasting and nocturnal Tyr concentrations with respect to traditional therapy (p<0.01); (4) this picture was confirmed at follow-up, with normalization of morning fasting Tyr concentrations in patients on slow-release amino acids therapy (p<0.01) and unchanged Phe control (p=0.19). CONCLUSIONS: Slow-release amino acids therapy can improve Tyr homeostasis in PKU. If associated to optimized Phe control, such a metabolic goal may allow long-term clinical benefits in patients with PKU.


Asunto(s)
Aminoácidos/administración & dosificación , Suplementos Dietéticos , Homeostasis , Fenilalanina Hidroxilasa/deficiencia , Fenilcetonurias/tratamiento farmacológico , Tirosina/metabolismo , Adolescente , Adulto , Estudios de Casos y Controles , Niño , Femenino , Estudios de Seguimiento , Humanos , Estudios Longitudinales , Masculino , Fenilcetonurias/metabolismo , Fenilcetonurias/patología , Pronóstico , Adulto Joven
9.
PLoS One ; 14(3): e0213391, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30875376

RESUMEN

INTRODUCTION: In phenylketonuria (PKU), a gene mutation in the phenylalanine metabolic pathway causes accumulation of phenylalanine (Phe) in blood and brain. Although early introduction of a Phe-restricted diet can prevent severe symptoms from developing, patients who are diagnosed and treated early still experience deficits in cognitive functioning indicating shortcomings of current treatment. In the search for new and/or additional treatment strategies, a specific nutrient combination (SNC) was postulated to improve brain function in PKU. In this study, a long-term dietary intervention with a low-Phe diet, a specific combination of nutrients designed to improve brain function, or both concepts together was investigated in male and female BTBR PKU and WT mice. MATERIAL & METHODS: 48 homozygous wild-types (WT, +/+) and 96 PKU BTBRPah2 (-/-) male and female mice received dietary interventions from postnatal day 31 till 10 months of age and were distributed in the following six groups: high Phe diet (WT C-HP, PKU C-HP), high Phe plus specific nutrient combination (WT SNC-HP, PKU SNC-HP), PKU low-Phe diet (PKU C-LP), and PKU low-Phe diet plus specific nutrient combination (PKU SNC- LP). Memory and motor function were tested at time points 3, 6, and 9 months after treatment initiation in the open field (OF), novel object recognition test (NOR), spatial object recognition test (SOR), and the balance beam (BB). At the end of the experiments, brain neurotransmitter concentrations were determined. RESULTS: In the NOR, we found that PKU mice, despite being subjected to high Phe conditions, could master the task on all three time points when supplemented with SNC. Under low Phe conditions, PKU mice on control diet could master the NOR at all three time points, while PKU mice on the SNC supplemented diet could master the task at time points 6 and 9 months. SNC supplementation did not consistently influence the performance in the OF, SOR or BB in PKU mice. The low Phe diet was able to normalize concentrations of norepinephrine and serotonin; however, these neurotransmitters were not influenced by SNC supplementation. CONCLUSION: This study demonstrates that both a long-lasting low Phe diet, the diet enriched with SNC, as well as the combined diet was able to ameliorate some, but not all of these PKU-induced abnormalities. Specifically, this study is the first long-term intervention study in BTBR PKU mice that shows that SNC supplementation can specifically improve novel object recognition.


Asunto(s)
Encéfalo/fisiopatología , Nutrientes/administración & dosificación , Fenilalanina/administración & dosificación , Fenilcetonurias/dietoterapia , Fenilcetonurias/fisiopatología , Factores de Edad , Animales , Conducta Animal/fisiología , Modelos Animales de Enfermedad , Femenino , Aprendizaje/fisiología , Masculino , Memoria/fisiología , Ratones , Ratones Noqueados , Neurotransmisores/metabolismo , Fenilalanina Hidroxilasa/deficiencia , Fenilalanina Hidroxilasa/genética , Fenilcetonurias/psicología
10.
Nutrition ; 59: 180-181, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30504004

RESUMEN

Phenylketonuria (PKU) is an autosomal recessive inborn error of phenylalanine (phe) metabolism caused by a deficiency in the enzyme phenylalanine hydroxylase that converts phe into tyrosine. If left untreated, PKU results in increased phe concentrations in the blood and in the brain, which cause severe intellectual disability, epilepsy, and behavioral problems. These disorders can be prevented if a diet low in phe is introduced. This report focuses on a preterm newborn (gestational age 29 wk) with adequate weight (1290 g) and no family history of PKU. His parents had not received metabolic neonatal screening. A blood sample at 16 d of age and a weight of 1430 g showed phe 420 µmol/L, compatible with mild PKU. Mixed feeding was initiated with a formula free of phe (X-Phe), and breastfeeding was fortified with a contribution of 3.5 g/kg daily (2.5 g X-Phe and 1 g of high-value biological proteins). The next measurements of amino acid levels in the blood and urine were normal, and the progenitors study for PKU was negative. Normal feeding was reintroduced with normal neurologic and metabolic later evolution. The disorders of the metabolism of phe, in most cases, are due to a genetic condition. However, there are infrequent cases of transient hyperphenylalaninemia secondary to delayed maturation of the hydroxylation enzyme system. They are especially significant in premature infants. Although these forms have not been shown to cause sequelae, in view of high levels of phe in the blood, phe consumption must be restricted.


Asunto(s)
Fenilalanina Hidroxilasa/metabolismo , Fenilcetonurias/enzimología , Lactancia Materna , Alimentos Fortificados , Humanos , Hidroxilación , Recién Nacido , Recien Nacido Prematuro , Masculino , Fenilalanina/sangre , Fenilcetonurias/sangre , Fenilcetonurias/dietoterapia
12.
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
13.
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
14.
Curr Drug Targets ; 17(13): 1515-26, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26953246

RESUMEN

The aromatic amino acid hydroxylase (AAAH) enzyme family includes phenylalanine hydroxylase (PAH), tyrosine hydroxylase (TH) and the tryptophan hydroxylases (TPH1 and TPH2). All four members of the AAAH family require iron, dioxygen and the cofactor (6R)-L-erythro-5,6,7,8-tetrahydrobiopterin (BH4) to hydroxylate their respective substrates. The AAAHs are involved in severe diseases; whereas polymorphisms and variants in the TPH genes are associated to neuropsychiatric disorders, mutations in PAH and TH are responsible for the autosomal recessive disorders phenylketonuria (PKU) and TH deficiency (THD), respectively. A large number of PKU and THD-causing mutations give rise to unstable, misfolded proteins. The degree of conformational instability correlates well with the severity of the patient phenotypes, underlying the relevance of searching for stabilizing compounds that may protect from loss of protein and activity in vivo. Supplementation with the cofactor BH4 exerts a multifactorial response in PAH, where one of the main mechanisms for the induced increase in PAH activity in BH4- responsive PKU patients appears to be a pharmacological chaperone effect. For TH the stabilizing effect of BH4 is less established. On the other hand, a number of compounds with pharmacological chaperone potential for PKU and THD mutants have been discovered. The stabilizing effect of these compounds has been established in vitro, in cells and in animal models. A recent study with TH has revealed different mechanisms for the action of pharmacological chaperones and identifies a subtype of compounds that preserve TH activity by weak binding to the catalytic iron. It is expected that synergistic combinations of different pharmacological chaperones could provide patient-tailored therapeutic options.


Asunto(s)
Biopterinas/análogos & derivados , Diseño de Fármacos , Chaperonas Moleculares/farmacología , Animales , Biopterinas/metabolismo , Humanos , Mutación , Fenilalanina Hidroxilasa/genética , Fenilalanina Hidroxilasa/metabolismo , Pliegue de Proteína , Triptófano Hidroxilasa/genética , Triptófano Hidroxilasa/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
15.
J Sci Food Agric ; 96(1): 140-9, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25564785

RESUMEN

BACKGROUND: Seedling roots of anthocyanin-rich corn (Zea mays) cultivars contain high levels of phenylalanine ammonia lyase (PAL) activity. The development of a natural dietary supplement containing corn roots could provide the means to improve the restrictive diet of phenylketonuria (PKU) patients by increasing their tolerance to dietary phenylalanine (Phe). Therefore this research was undertaken to explore the sensory characteristics of roots of four corn cultivars as well as to develop and evaluate food products (cereal bar, beverage, jam-like spread) to which roots had been added. RESULTS: Sensory profiles of corn roots were investigated using ten trained judges. Roots of Japanese Striped corn seedlings were more bitter, pungent and astringent than those of white and yellow cultivars, while roots from the Blue Jade cultivar had a more pronounced earthy/mushroom aroma. Consumer research using 24 untrained panelists provided hedonic (degree-of-liking) assessments for products with and without roots (controls). The former had lower mean scores than the controls; however, the cereal bar had scores above 5 on the nine-point scale for all hedonic assessments compared with the other treated products. CONCLUSION: By evaluating low-Phe food products containing corn roots, this research ascertained that the root-containing low-Phe cereal bar was an acceptable 'natural' dietary supplement for PKU-affected individuals.


Asunto(s)
Suplementos Dietéticos , Alimentos Formulados/análisis , Fenilanina Amoníaco-Liasa/farmacología , Fenilalanina/metabolismo , Fenilcetonurias/dietoterapia , Raíces de Plantas/química , Zea mays/química , Antocianinas/metabolismo , Grano Comestible , Humanos , Odorantes , Fenilanina Amoníaco-Liasa/administración & dosificación , Fenilanina Amoníaco-Liasa/metabolismo , Fenilalanina Hidroxilasa/metabolismo , Raíces de Plantas/enzimología , Raíces de Plantas/metabolismo , Plantones/química , Plantones/enzimología , Plantones/metabolismo , Gusto , Zea mays/enzimología , Zea mays/metabolismo
16.
Mol Genet Metab ; 115(2-3): 78-83, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25943030

RESUMEN

BACKGROUND: Phenylketonuria (PKU) is an autosomal recessive disorder caused by deficiency of hepatic phenylalanine hydroxylase (PAH) leading to increased levels of phenylalanine in the plasma. Phenylalanine levels and phenylalanine hydroxylase (PAH) activity monitoring are currently limited to conventional blood dot testing. 1-(13)C-phenylalanine, a stable isotope can be used to examine phenylalanine metabolism, as the conversion of phenylalanine to tyrosine occurs in vivo via PAH and subsequently releases the carboxyl labeled (13)C as (13)CO2 in breath. OBJECTIVE: Our objective was to examine phenylalanine metabolism in children with PKU using a minimally-invasive 1-(13)C-phenylalanine breath test ((13)C-PBT). DESIGN: Nine children (7 M: 2 F, mean age 12.5 ± 2.87 y) with PKU participated in the study twice: once before and once after sapropterin supplementation. Children were provided 6 mg/kg oral dose of 1-(13)C-phenylalanine and breath samples were collected at 20 min intervals for a period of 2h. Rate of CO2 production was measured at 60 min post-oral dose using indirect calorimetry. The percentage of 1-(13)C-phenylalanine exhaled as (13)CO2 was measured over a 2h period. Prior to studying children with PKU, we tested the study protocol in healthy children (n = 6; 4M: 2F, mean age 10.2 ± 2.48 y) as proof of principle. RESULTS: Production of a peak enrichment (Cmax) of (13)CO2 (% of dose) in all healthy children occurred at 20 min ranging from 17-29% of dose, with a subsequent return to ~5% by the end of 2h. Production of (13)CO2 from 1-(13)C-phenylalanine in all children with PKU prior to sapropterin treatment remained low. Following sapropterin supplementation for a week, production of (13)CO2 significantly increased in five children with a subsequent decline in blood phenylalanine levels, suggesting improved PAH activity. Sapropterin treatment was not effective in three children whose (13)CO2 production remained unchanged, and did not show a reduction in blood phenylalanine levels and improvement in dietary phenylalanine tolerance. CONCLUSIONS: Our study shows that the (13)C-PBT can be a minimally invasive, safe and reliable measure to examine phenylalanine metabolism in children with phenylketonuria. The breath data are corroborated by blood phenylalanine levels in children who had increased responses in (13)CO2 production, as reviewed post-hoc from clinical charts.


Asunto(s)
Pruebas Respiratorias/métodos , Fenilalanina/metabolismo , Fenilcetonurias/metabolismo , Adolescente , Biopterinas/análogos & derivados , Biopterinas/uso terapéutico , Dióxido de Carbono/química , Dióxido de Carbono/metabolismo , Isótopos de Carbono/química , Isótopos de Carbono/metabolismo , Niño , Femenino , Humanos , Hígado/metabolismo , Masculino , Chaperonas Moleculares/uso terapéutico , Fenilalanina/química , Fenilalanina Hidroxilasa/química , Fenilalanina Hidroxilasa/metabolismo , Fenilcetonurias/tratamiento farmacológico , Fenilcetonurias/enzimología
17.
Mol Genet Metab ; 115(2-3): 72-7, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25990862

RESUMEN

While phenylalanine (PHE) is the toxic insult in phenylketonuria (PKU), mechanisms underlying PHE toxicity remain ill-defined. Altered DNA methylation in response to toxic exposures is well-recognized. DNA methylation patterns were assessed in blood and brain from PKU patients to determine if PHE toxicity impacts methylation. Methylome assessment, utilizing methylated DNA immunoprecipitation and paired-end sequencing, was performed in DNA obtained from brain tissue of classical PKU patients, leukocytes from poorly controlled PKU patients, leukocytes from well controlled PKU patients, and appropriate control tissues. In PKU brain tissue, expression analysis determined the impact of methylation on gene function. Differential methylation was observed in brain tissue of PKU patients and expression studies identified downstream impact on gene expression. Altered patterns of methylation were observed in leukocytes of well controlled and poorly controlled patients with more extensive methylation in patients with high PHE exposure. Differential methylation of noncoding RNA genes was extensive in patients with high PHE exposure but minimal in well controlled patients. Methylome repatterning leading to altered gene expression was present in brain tissue of PKU patients, suggesting a role in neuropathology. Aberrant methylation is observed in leukocytes of PKU patients and is influenced by PHE exposure. DNA methylation may provide a biomarker relating to historic PHE exposure.


Asunto(s)
Encefalopatías/metabolismo , Metilación de ADN , Fenilalanina Hidroxilasa/genética , Fenilcetonurias/metabolismo , Anciano , Femenino , Lóbulo Frontal/metabolismo , Lóbulo Frontal/patología , Humanos , Leucocitos , Persona de Mediana Edad , Fenilalanina/sangre
18.
Curr Opin Psychiatry ; 28(3): 201-6, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25768084

RESUMEN

PURPOSE OF REVIEW: Since decades immunological aberrancies have been reported in schizophrenia patients. As schizophrenia represents a heterogenous disorder with a variety of clinical manifestations, complex interactions between the immune system in the brain might have important etiological implications. RECENT FINDINGS: Recent findings of altered expression of immune-related genes, changes of peripheral and central cytokines, antibodies and immune cells point toward dysbalanced immune response processes in schizophrenia. SUMMARY: Based on immunogenetic factors, immune dysfunctions caused by infections, increased autoimmune reactivity and low-grade inflammatory processes in the periphery as well as in central nervous system may affect neurobiological circuits including changed neurotransmitter metabolisms contributing to pathophysiological alterations in schizophrenia. These immunological abnormalities might provide tools for better diagnostic characterization of this heterogenous disease and on the other side, they may also support the development of immune-related therapeutic strategies.


Asunto(s)
Encéfalo/metabolismo , Citocinas/inmunología , Inflamación/inmunología , Esquizofrenia/metabolismo , Autoinmunidad , Encéfalo/inmunología , Humanos , Neurotransmisores/inmunología , Fenilalanina Hidroxilasa/metabolismo , Psiconeuroinmunología , Pteridinas/metabolismo , Esquizofrenia/inmunología
19.
J Ind Microbiol Biotechnol ; 42(4): 655-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25645094

RESUMEN

Tyrosine is a proteinogenic aromatic amino acid that is often used as a supplement of food and animal feed, as well as a (bio-)synthetic precursor to various pharmaceutically or industrially important molecules. Extensive metabolic engineering efforts have been made towards the efficient and cost-effective microbial production of tyrosine. Conventional strategies usually focus on eliminating intrinsic feedback inhibition and redirecting carbon flux into the shikimate pathway. In this study, we found that continuous conversion of phenylalanine into tyrosine by the action of tetrahydromonapterin (MH4)-utilizing phenylalanine 4-hydroxylase (P4H) can bypass the feedback inhibition in Escherichia coli, leading to tyrosine accumulation in the cultures. First, expression of the P4H from Xanthomonas campestris in combination with an MH4 recycling system in wild-type E. coli allowed the strain to accumulate tyrosine at 262 mg/L. On this basis, enhanced expression of the key enzymes associated with the shikimate pathway and the MH4 biosynthetic pathway resulted in the elevation of tyrosine production up to 401 mg/L in shake flasks. This work demonstrated a novel approach to tyrosine production and verified the possibility to alleviate feedback inhibition by creating a phenylalanine sink.


Asunto(s)
Escherichia coli/genética , Escherichia coli/metabolismo , Retroalimentación Fisiológica , Ingeniería Metabólica , Fenilalanina/metabolismo , Tirosina/biosíntesis , Vías Biosintéticas , Hidroxilación , Neopterin/análogos & derivados , Neopterin/metabolismo , Fenilalanina Hidroxilasa/genética , Fenilalanina Hidroxilasa/metabolismo , Ácido Shikímico/metabolismo , Xanthomonas campestris/enzimología , Xanthomonas campestris/genética
20.
J Microbiol Biotechnol ; 25(6): 782-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25563416

RESUMEN

In this study, we developed an assay system for missense mutations in human phenylalanine hydroxylases (hPAHs). To demonstrate the reliability of the system, eight mutant proteins (F39L, K42I, L48S, I65T, R252Q, L255V, S349L, and R408W) were expressed in a mutant strain (pah(-)) of Dictyostelium discoideum Ax2 disrupted in the indigenous gene encoding PAH. The transformed pah- cells grown in FM minimal medium were measured for growth rate and PAH activity to reveal a positive correlation between them. The protein level of hPAH was also determined by western blotting to show the impact of each mutation on protein stability and catalytic activity. The result was highly compatible with the previous ones obtained from other expression systems, suggesting that Dictyostelium is a dependable alternative to other expression systems. Furthermore, we found that both the protein level and activity of S349L and R408W, which were impaired severely in protein stability, were rescued in HL5 nutrient medium. Although the responsible component(s) remains unidentified, this unexpected finding showed an important advantage of our expression system for studying unstable proteins. As an economic and stable cell-based expression system, our development will contribute to mass-screening of pharmacological chaperones for missense PAH mutations as well as to the in-depth characterization of individual mutations.


Asunto(s)
Medios de Cultivo/química , Dictyostelium/efectos de los fármacos , Dictyostelium/crecimiento & desarrollo , Estabilidad de Enzimas/efectos de los fármacos , Fenilalanina Hidroxilasa/genética , Fenilalanina Hidroxilasa/metabolismo , Dictyostelium/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Ensayos Analíticos de Alto Rendimiento , Humanos , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fenilalanina Hidroxilasa/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA