Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 750
Filter
1.
Curr Drug Saf ; 19(2): 208-217, 2024.
Article in English | MEDLINE | ID: mdl-36999718

ABSTRACT

Phenylalanine, an essential amino acid, is the "building block" of protein. It has a tremendous role in different aspects of metabolic events. The tyrosine pathway is the prime one and is typically used to degrade dietary phenylalanine. Phenylalanine exceeds its limit in bodily fluids and the brain when the enzyme, phenylalanine decarboxylase, phenylalanine transaminase, phenylalanine hydroxylase (PAH) or its cofactor tetrahydrobiopterin (BH4) is deficient causes phenylketonuria, schizophrenia, attentiondeficit/ hyperactivity disorder and another neuronal effect. Tyrosine, an amino acid necessary for synthesizing the pigments in melanin, is produced by its primary metabolic pathway. Deficiency/abnormality in metabolic enzymes responsible for the catabolism pathway of Phenylalanine causes an accumulation of the active intermediate metabolite, resulting in several abnormalities, such as developmental delay, tyrosinemias, alkaptonuria, albinism, hypotension and several other undesirable conditions. Dietary restriction of the amino acid(s) can be a therapeutic approach to avoid such undesirable conditions when the level of metabolic enzyme is unpredictable. After properly identifying the enzymatic level, specific pathophysiological conditions can be managed more efficiently.


Subject(s)
Phenylalanine Hydroxylase , Phenylketonurias , Humans , Phenylalanine/metabolism , Phenylketonurias/metabolism , Phenylalanine Hydroxylase/chemistry , Phenylalanine Hydroxylase/metabolism , Amino Acids , Tyrosine/metabolism
2.
Discov Med ; 35(177): 533-538, 2023 08.
Article in English | MEDLINE | ID: mdl-37553307

ABSTRACT

BACKGROUND: Phenylalanine hydroxylase deficiency (PAHD) is an autosomal recessive disorder affecting phenylalanine (Phe) metabolism caused by mutations in the phenylalanine hydroxylase (PAH) gene. It has a complex phenotype with many variants and genotypes in various populations. This study sets out to analyze the screening results of children with phenylketonuria (PKU) in Yinchuan City and characterize the mutation variants of the PAH gene. METHODS: Phenylketonuria screening results were retrospectively analyzed in 398,605 neonates (207,361 males and 191,244 females) born in different maternity hospitals in Yinchuan City between January 2017 and December 2021. Screening for genetic metabolic diseases was performed with parental consent at their own expense. A comprehensive diagnosis was performed by integrating tandem mass spectrometry (MS/MS) findings with clinical presentations. High-throughput sequencing (HTS) was used to detect genetic and metabolic disease-associated genes in children with PKU who were clinically diagnosed and voluntarily tested. The identified loci were validated through Sanger sequencing and parental verification. RESULTS: Among the screened newborns, 45 (11.3/100,000) PKU cases were diagnosed. In the 38 cases that underwent self-financed PAH sequencing, 56 mutations were detected in 76 chromosomes, with an overall detection rate of 73.7%. All patients harbored mutant genes, and the 56 mutations detected identified represented 14 variants, including 8 missense mutations, 2 splicing mutations, 2 nonsense mutations, and 2 silent mutations. The mutations were primarily distributed in exons 2, 3, 6, 7, 9, 11, and intron 4, with the highest frequency observed in exon 7 (25 [44.7%]), followed by exon 11 (15 [26.7%]). The most prevalent mutations were exon 7-p.R252W (10 [17.9%]) and exon 7-p.R261Q (8 [14.3%]). CONCLUSIONS: The PAH gene mutations in children with PKU in Yinchuan City are predominantly concentrated in exons 6, 7, and 11, with the highest detection rates observed for p.R252W and p.R261Q mutations.


Subject(s)
Phenylalanine Hydroxylase , Phenylketonurias , Pregnancy , Male , Child , Female , Humans , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Retrospective Studies , Tandem Mass Spectrometry , Phenylketonurias/epidemiology , Phenylketonurias/genetics , Phenylketonurias/diagnosis , Mutation , Genotype
3.
Nat Commun ; 14(1): 3451, 2023 06 10.
Article in English | MEDLINE | ID: mdl-37301931

ABSTRACT

Phenylketonuria (PKU), an autosomal recessive disorder caused by pathogenic variants in the phenylalanine hydroxylase (PAH) gene, results in the accumulation of blood phenylalanine (Phe) to neurotoxic levels. Current dietary and medical treatments are chronic and reduce, rather than normalize, blood Phe levels. Among the most frequently occurring PAH variants in PKU patients is the P281L (c.842C>T) variant. Using a CRISPR prime-edited hepatocyte cell line and a humanized PKU mouse model, we demonstrate efficient in vitro and in vivo correction of the P281L variant with adenine base editing. With the delivery of ABE8.8 mRNA and either of two guide RNAs in vivo using lipid nanoparticles (LNPs) in humanized PKU mice, we observe complete and durable normalization of blood Phe levels within 48 h of treatment, resulting from corrective PAH editing in the liver. These studies nominate a drug candidate for further development as a definitive treatment for a subset of PKU patients.


Subject(s)
Phenylalanine Hydroxylase , Phenylketonurias , Mice , Animals , Phenylketonurias/genetics , Phenylketonurias/therapy , Phenylketonurias/metabolism , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Liver/metabolism , Hepatocytes/metabolism , Disease Models, Animal
4.
Hum Genomics ; 17(1): 36, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37098607

ABSTRACT

BACKGROUND: Phenylketonuria (PKU) is a common, congenital, autosomal recessive, metabolic disorder caused by Phenylalanine hydroxylase (PAH) variants. METHODS: 967 PKU patients from Gansu, China were genotyped by Sanger sequencing, multiplex ligation-dependent probe amplification, and whole exome sequencing. We analyzed the variants of PAH exons, their flanking sequences, and introns. RESULTS: The detection of deep intronic variants in PAH gene can significantly improve the genetic diagnostic rate of PKU. The distribution of PAH variants among PKU subtypes may be related to the unique genetic background in Gansu, China. CONCLUSION: The identification of PAH hotspot variants will aid the development of large-scale neonatal genetic screening for PKU. The five new PAH variants found in this study further expand the spectrum of PAH variants. Genotype-phenotype correlation analysis may help predict the prognosis of PKU patients and enable precise treatment regimens to be developed.


Subject(s)
Phenylalanine Hydroxylase , Phenylketonurias , Humans , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Phenylketonurias/genetics , Phenylketonurias/diagnosis , Mutation , Genotype , Genetic Association Studies , China , Phenotype
5.
J Mol Diagn ; 25(5): 284-294, 2023 05.
Article in English | MEDLINE | ID: mdl-36849017

ABSTRACT

Phenylalanine hydroxylase (PAH) deficiency or phenylketonuria (PKU) is the most common cause of hyperphenylalaninemia (HPA), and approximately 5% of patients remain genetically unsolved. Identifying deep intronic PAH variants may help improve their molecular diagnostic rate. Next-generation sequencing was utilized to detect the whole PAH gene in 96 patients with genetically unsolved HPA from 2013 to 2022. The effects of deep intronic variants on pre-mRNA splicing were investigated by minigene-based assay. The allelic phenotype values of recurrent deep intronic variants were calculated. Twelve deep intronic PAH variants, located in intron 5 (c.509+434C>T), intron 6 (c.706+288T>G, c.706+519T>C, c.706+531T>C, c.706+535G>T, c.706+600A>C, c.706+603T>G, and c.706+608A>C), intron 10 (c.1065+241C>A and c.1065+258C>A), and intron 11 (c.1199+502A>T and c.1199+745T>A) were identified in 80.2% (77/96) patients. Ten of the 12 variants were novel, and they all generated pseudoexons in mRNA, leading to frameshift or lengthened proteins. The most prevalent deep intronic variant was c.1199+502A>T, followed by c.1065+241C>A, c.1065+258C>A, and c.706+531T>C. The metabolic phenotypes of the four variants were assigned as classic PKU, mild HPA, mild HPA, and mild PKU, respectively. The results suggest that deep intronic PAH variants improved the diagnostic rate from 95.3% to 99.3% in the overall patients with HPA. Our data demonstrate the importance of assessing noncoding variants in genetic diseases. Pseudoexon inclusion caused by deep intronic variants may represent a recurrent mechanism.


Subject(s)
Phenylalanine Hydroxylase , Phenylketonurias , Humans , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Genotype , Introns/genetics , Phenylketonurias/diagnosis , Phenylketonurias/genetics , Mutation , Phenotype
6.
Mol Genet Metab ; 138(3): 107509, 2023 03.
Article in English | MEDLINE | ID: mdl-36791482

ABSTRACT

Phenylketonuria (PKU, MIM #261600) is one of the most common inborn errors of metabolism (IEM) with an incidence of 1:10000 in the European population. PKU is caused by autosomal recessive mutations in phenylalanine hydroxylase (PAH) and manifests with elevation of phenylalanine (Phe) in plasma and urine. Untreated PKU manifests with intellectual disability including seizures, microcephaly and behavioral abnormalities. Early treatment and good compliance result in a normal intellectual outcome in many but not in all patients. This study examined plasma metabolites in patients with PKU (n = 27), hyperphenylalaninemia (HPA, n = 1) and healthy controls (n = 32) by LC- MS/MS. We hypothesized that PKU patients would exhibit a distinct "submetabolome" compared to that of healthy controls. We further hypothesized that the submetabolome of PKU patients with good metabolic control would resemble that of healthy controls. Results from this study show: (i) Distinct clustering of healthy controls and PKU patients based on polar metabolite profiling, (ii) Increased and decreased concentrations of metabolites within and afar from the Phe pathway in treated patients, and (iii) A specific PKU-submetabolome independently of metabolic control assessed by Phe in plasma. We examined the relationship between PKU metabolic control and extended metabolite profiles in plasma. The PKU submetabolome characterized in this study represents the combined effects of dietary adherence, adjustments in metabolic pathways to compensate for defective Phe processing, as well as metabolic derangements that could not be corrected with dietary management even in patients classified as having good metabolic control. New therapeutic targets may be uncovered to approximate the PKU submetabolome to that of healthy controls and prevent long-term organ damage.


Subject(s)
Phenylalanine Hydroxylase , Phenylketonurias , Humans , Disease Hotspot , Tandem Mass Spectrometry , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Phenylalanine , Cluster Analysis
7.
Arch Biochem Biophys ; 735: 109518, 2023 02.
Article in English | MEDLINE | ID: mdl-36639008

ABSTRACT

The aromatic amino acid hydroxylases phenylalanine hydroxylase, tyrosine hydroxylase, and tryptophan hydroxylase are non-heme iron enzymes that catalyze key physiological reactions. This review discusses the present understanding of the common catalytic mechanism of these enzymes and recent advances in understanding the relationship between their structures and their regulation.


Subject(s)
Mixed Function Oxygenases , Phenylalanine Hydroxylase , Mixed Function Oxygenases/chemistry , Tryptophan Hydroxylase/chemistry , Tryptophan Hydroxylase/metabolism , Tyrosine 3-Monooxygenase/chemistry , Tyrosine 3-Monooxygenase/metabolism , Phenylalanine Hydroxylase/chemistry , Phenylalanine Hydroxylase/metabolism , Amino Acids, Aromatic , Catalysis
8.
Cell Biol Toxicol ; 39(5): 2295-2310, 2023 10.
Article in English | MEDLINE | ID: mdl-35449354

ABSTRACT

Phenylalanine hydroxylase (PAH) is the key enzyme in phenylalanine metabolism, deficiency of which is associated with the most common metabolic phenotype of phenylketonuria (PKU) and hyperphenylalaninemia (HPA). A bulk of PKU disease-associated missense mutations in the PAH gene have been studied, and the consequence of each PAH variant vary immensely. Prior research established that PKU-associated variants possess defects in protein folding with reduced cellular stability leading to rapid degradation. However, recent evidence revealed that PAH tetramers exist as a mixture of resting state and activated state whose transition depends upon the phenylalanine concentration and certain PAH variants that fail to modulate the structural equilibrium are associated with PKU disease. Collectively, these findings framed our understanding of the complex genotype-phenotype correlation in PKU. In the current study, we substantiate a link between PAH protein stability and its degradation by the ubiquitin-mediated proteasomal degradation system. Here, we provide an evidence that PAH protein undergoes ubiquitination and proteasomal degradation, which can be reversed by deubiquitinating enzymes (DUBs). We identified USP19 as a novel DUB that regulates PAH protein stability. We found that ectopic expression of USP19 increased PAH protein level, whereas depletion of USP19 promoted PAH protein degradation. Our study indicates that USP19 interacts with PAH and prevents polyubiquitination of PAH subsequently extending the half-life of PAH protein. Finally, the increase in the level of PAH protein by the deubiquitinating activity of USP19 resulted in enhanced metabolic function of PAH. In summary, our study identifies the role of USP19 in regulating PAH protein stability and promotes its metabolic activity. Graphical highlights 1. E3 ligase Cdh1 promotes PAH protein degradation leading to insufficient cellular amount of PAH causing PKU. 2. A balance between E3 ligase and DUB is important to regulate the proteostasis of PAH. 3. USP19 deubiquitinates and stabilizes PAH further protecting it from rapid degradation. 4. USP19 increases the enzymatic activity of PAH, thus maintaining normal Phe levels.


Subject(s)
Phenylalanine Hydroxylase , Phenylketonurias , Humans , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/chemistry , Phenylalanine Hydroxylase/metabolism , Phenylketonurias/genetics , Ubiquitin-Protein Ligases/metabolism , Protein Stability , Phenylalanine/metabolism , Deubiquitinating Enzymes/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism
9.
Arch Biochem Biophys ; 729: 109378, 2022 Oct 30.
Article in English | MEDLINE | ID: mdl-35995215

ABSTRACT

Phenylalanine hydroxylase (PheH) is a pterin-dependent, mononuclear nonheme iron(II) oxygenase that uses the oxidative power of O2 to hydroxylate phenylalanine to form tyrosine. PheH is a member of a superfamily of O2-activating enzymes that utilizes a common metal binding motif: the 2-His-1-carboxylate facial triad. Like most members of this superfamily, binding of substrates to PheH results in a reorganization of its active site to allow O2 activation. Exploring the energetics of each step before O2 activation can provide mechanistic insight into the initial steps that support the highly specific O2 activation pathway carried out by this metalloenzyme. Here the thermal stability of PheH and its substrate complexes were investigated under an anaerobic environment by using differential scanning calorimetry. In context with known binding constants for PheH, a thermodynamic cycle associated with iron(II), tetrahydrobiopterin (BH4), and phenylalanine binding to the active site was generated, showing a distinctive cooperativity between the binding of BH4 and Phe. The addition of phenylalanine and BH4 to PheH·Fe increased the stability of this enzyme (ΔTm of 8.5 (±0.7) °C with an associated δΔH of 43.0 (±2.9) kcal/mol). The thermodynamic data presented here gives insight into the complicated interactions between metal center, cofactor, and substrate, and how this interplay sets the stage for highly specific, oxidative C-H activation in this enzyme.


Subject(s)
Metalloproteins , Phenylalanine Hydroxylase , Biopterins/analogs & derivatives , Chromobacterium , Ferrous Compounds , Iron/metabolism , Kinetics , Metalloproteins/metabolism , Phenylalanine/metabolism , Phenylalanine Hydroxylase/chemistry , Phenylalanine Hydroxylase/metabolism , Pterins/chemistry , Pterins/metabolism , Thermodynamics , Tyrosine
10.
Hum Genomics ; 16(1): 22, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35854334

ABSTRACT

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.


Subject(s)
Phenylalanine Hydroxylase , Phenylketonurias , Humans , Phenylalanine/metabolism , Phenylalanine/therapeutic use , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Phenylalanine Hydroxylase/therapeutic use , Phenylketonurias/genetics , Phenylketonurias/therapy
11.
Nutr Rev ; 80(10): 2100-2112, 2022 09 05.
Article in English | MEDLINE | ID: mdl-35551410

ABSTRACT

Phenylketonuria (PKU) is a rare genetic disease that causes brain toxicity due to the inability of the body to convert dietary phenylalanine to tyrosine by the action of phenylalanine hydroxylase. The only treatment for PKU so far is lifelong dietary intervention to ensure normal human growth and neurodevelopment. However, in adults, low long-term adherence to this type of dietary intervention has been observed. Given the important role of the intestinal microbiota in the process of digestion and disease prevention, probiotics could be a therapeutic strategy to help degrade dietary phenylalanine, reducing its levels before ingestion. Genetically modified probiotics designed as live biotherapeutic agents for the treatment of specific diseases are sophisticated alternative therapeutic strategies. In this review, the focus is on demonstrating what has been elucidated so far about the use of next-generation probiotics as a therapeutic strategy in the treatment of individuals with PKU. The results described in the literature are encouraging and use genetically modified engineered probiotics showing efficacy both in vitro and in vivo. These probiotics appear to be suitable for meeting the unmet need for new drugs for PKU.


Subject(s)
Phenylalanine Hydroxylase , Phenylketonurias , Probiotics , Adult , Diet , Humans , Phenylalanine/metabolism , Phenylalanine/therapeutic use , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Phenylalanine Hydroxylase/therapeutic use , Phenylketonurias/drug therapy , Phenylketonurias/genetics , Probiotics/therapeutic use
12.
Acta Crystallogr D Struct Biol ; 78(Pt 5): 586-598, 2022 May 01.
Article in English | MEDLINE | ID: mdl-35503207

ABSTRACT

Phenylalanine hydroxylase (PAH), which belongs to the aromatic amino-acid hydroxylase family, is involved in protein synthesis and pyomelanine production through the hydroxylation of phenylalanine to tyrosine. In this study, the crystal structure of PAH from Bacillus cereus ATCC 14579 (BcPAH) with an additional 280 amino acids in the C-terminal region was determined. The structure of BcPAH consists of three distinct domains: a core domain with two additional inserted α-helices and two novel auxiliary domains: BcPAH-AD1 and BcPAH-AD2. Structural homologues of BcPAH-AD1 and BcPAH-AD2 are known to be involved in mRNA regulation and protein-protein interactions, and thus it was speculated that BcPAH might utilize the auxiliary domains for interaction with its partner proteins. Furthermore, phylogenetic tree analysis revealed that the three-domain PAHs, including BcPAH, are completely distinctive from both conventional prokaryotic PAHs and eukaryotic PAHs. Finally, biochemical studies of BcPAH showed that BcPAH-AD1 might be important for the structural integrity of the enzyme and that BcPAH-AD2 is related to enzyme stability and/or activity. Investigations into the intracellular functions of the two auxiliary domains and the relationship between these functions and the activity of PAH are required.


Subject(s)
Phenylalanine Hydroxylase , Bacillus cereus/metabolism , Enzyme Stability , Mixed Function Oxygenases , Phenylalanine Hydroxylase/chemistry , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Phylogeny
13.
J Pediatr Endocrinol Metab ; 35(3): 387-391, 2022 Mar 28.
Article in English | MEDLINE | ID: mdl-35026061

ABSTRACT

OBJECTIVES: Phenylketonuria (PKU) is an inherited autosomal recessive disorder of phenylalanine metabolism. It is mainly caused by a deficiency in phenylalanine hydroxylase (PAH) and frequently diagnosed with Sanger sequencing. To some extent, allelic dropout can explain the inconsistency in genotype and phenotype. METHODS: Three families were evaluated through DNA sequence analysis, multiplex ligation-dependent probe amplification (MLPA) and prenatal diagnosis technologies. The possibility of inconsistency in phenotype and genotype with c.331C>T variant was analysed. RESULTS: Through pedigree analysis, three mothers carried a homozygous c.331C>T variant, which was a false-positive result. New primers were used, and this error was caused by allelic dropout. In this case, c.158G>A was likely a benign variant. CONCLUSIONS: Sequence variants in primer-binding regions could cause allelic dropout, creating unpredictable errors in genotyping. Our results emphasised the need for careful measures to treat genotype-phenotype inconsistencies.


Subject(s)
Phenylalanine Hydroxylase , Phenylketonurias , Alleles , Female , Genotype , Humans , Mutation , Phenotype , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Phenylketonurias/diagnosis , Phenylketonurias/genetics , Pregnancy
14.
Sci China Life Sci ; 65(4): 718-730, 2022 04.
Article in English | MEDLINE | ID: mdl-32815069

ABSTRACT

Genome editing through adeno-associated viral (AAV) vectors is a promising gene therapy strategy for various diseases, especially genetic disorders. However, homologous recombination (HR) efficiency is extremely low in adult animal models. We assumed that increasing AAV transduction efficiency could increase genome editing activity, especially HR efficiency, for in vivo gene therapy. Firstly, a mouse phenylketonuria (PKU) model carrying a pathogenic R408W mutation in phenylalanine hydroxylase (Pah) was generated. Through co-delivery of the general AAV receptor (AAVR), we found that AAVR could dramatically increase AAV transduction efficiency in vitro and in vivo. Furthermore, co-delivery of SaCas9/sgRNA/donor templates with AAVR via AAV8 vectors increased indel rate over 2-fold and HR rate over 15-fold for the correction of the single mutation in PahR408W mice. Moreover, AAVR co-injection successfully increased the site-specific insertion rate of a 1.4 kb Pah cDNA by 11-fold, bringing the HR rate up to 7.3% without detectable global off-target effects. Insertion of Pah cDNA significantly decreased the Phe level and ameliorated PKU symptoms. This study demonstrates a novel strategy to dramatically increase AAV transduction which substantially enhanced in vivo genome editing efficiency in adult animal models, showing clinical potential for both conventional and genome editing-based gene therapy.


Subject(s)
Liver Diseases , Phenylalanine Hydroxylase , Phenylketonurias , Animals , DNA, Complementary , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Gene Editing , Genetic Vectors/genetics , Mice , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/metabolism , Phenylketonurias/genetics , Phenylketonurias/therapy
15.
J Biomol Struct Dyn ; 40(19): 9018-9029, 2022.
Article in English | MEDLINE | ID: mdl-33970801

ABSTRACT

Phenylketonuria is an autosomal recessive disorder caused by mutations in the phenylalanine hydroxylase gene. In phenylketonuria causes various symptoms including severe mental retardation. PAH gene of a classical Phenylketonuria patient was sequenced, and two novel heterozygous mutations, p.Y198N and p.Y204F, were found. This study aimed to reveal the impacts of these variants on the structural stability of the PAH enzyme. In-silico analyses using prediction tools and molecular dynamics simulations were performed. Mutations were introduced to the wild type catalytic monomer and full length tetramer crystal structures. Variant pathogenicity analyses predicted p.Y198N to be damaging, and p.Y204F to be benign by some prediction tools and damaging by others. Simulations suggested p.Y198N mutation cause significant fluctuations in the spatial organization of two catalytic residues in the temperature accelerated MD simulations with the monomer and increased root-mean-square deviations in the tetramer structure. p.Y204F causes noticeable changes in the spatial positioning of T278 suggesting a possible segregation from the catalytic site in temperature accelerated MD simulations with the monomer. This mutation also leads to increased root-mean-square fluctuations in the regulatory domain which may lead to conformational change resulting in inhibition of dimerization and enzyme activation. Our study reports two novel mutations in the PAH gene and gives insight to their effects on the PAH activity. MD simulations did not yield conclusive results that explains the phenotype but gave plausible insight to possible effects which should be investigated further with in-silico and in-vitro studies to assess the roles of these mutations in etiology of PKU. Communicated by Ramaswamy H. Sarma.


Subject(s)
Phenylalanine Hydroxylase , Phenylketonurias , Humans , Molecular Dynamics Simulation , Phenylalanine Hydroxylase/genetics , Phenylalanine Hydroxylase/chemistry , Phenylalanine Hydroxylase/metabolism , Phenylketonurias/genetics , Mutation , Catalytic Domain
16.
Sci Rep ; 11(1): 22886, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34819582

ABSTRACT

Phenylketonuria (PKU) is a genetic deficiency of phenylalanine hydroxylase (PAH) in liver resulting in blood phenylalanine (Phe) elevation and neurotoxicity. A pegylated phenylalanine ammonia lyase (PEG-PAL) metabolizing Phe into cinnamic acid was recently approved as treatment for PKU patients. A potentially one-time rAAV-based delivery of PAH gene into liver to convert Phe into tyrosine (Tyr), a normal way of Phe metabolism, has now also entered the clinic. To understand differences between these two Phe lowering strategies, we evaluated PAH and PAL expression in livers of PAHenu2 mice on brain and liver functions. Both lowered brain Phe and increased neurotransmitter levels and corrected animal behavior. However, PAL delivery required dose optimization, did not elevate brain Tyr levels and resulted in an immune response. The effect of hyperphenylalanemia on liver functions in PKU mice was assessed by transcriptome and proteomic analyses. We observed an elevation in Cyp4a10/14 proteins involved in lipid metabolism and upregulation of genes involved in cholesterol biosynthesis. Majority of the gene expression changes were corrected by PAH and PAL delivery though the role of these changes in PKU pathology is currently unclear. Taken together, here we show that blood Phe lowering strategy using PAH or PAL corrects both brain pathology as well as previously unknown lipid metabolism associated pathway changes in liver.


Subject(s)
Genetic Therapy , Liver/enzymology , Phenylalanine Ammonia-Lyase/metabolism , Phenylalanine Hydroxylase/metabolism , Phenylalanine/blood , Phenylketonurias/therapy , Transcriptome , Animals , Biomarkers/blood , Brain/metabolism , Brain/pathology , Disease Models, Animal , Down-Regulation , Gene Expression Profiling , Male , Mice, Knockout , Phenylalanine Ammonia-Lyase/genetics , Phenylalanine Hydroxylase/genetics , Phenylketonurias/blood , Phenylketonurias/genetics , Phenylketonurias/pathology , Proteome , Proteomics
17.
Science ; 373(6555): 662-673, 2021 08 06.
Article in English | MEDLINE | ID: mdl-34353949

ABSTRACT

The functional role of long noncoding RNAs (lncRNAs) in inherited metabolic disorders, including phenylketonuria (PKU), is unknown. Here, we demonstrate that the mouse lncRNA Pair and human HULC associate with phenylalanine hydroxylase (PAH). Pair-knockout mice exhibited excessive blood phenylalanine (Phe), musty odor, hypopigmentation, growth retardation, and progressive neurological symptoms including seizures, which faithfully models human PKU. HULC depletion led to reduced PAH enzymatic activities in human induced pluripotent stem cell-differentiated hepatocytes. Mechanistically, HULC modulated the enzymatic activities of PAH by facilitating PAH-substrate and PAH-cofactor interactions. To develop a therapeutic strategy for restoring liver lncRNAs, we designed GalNAc-tagged lncRNA mimics that exhibit liver enrichment. Treatment with GalNAc-HULC mimics reduced excessive Phe in Pair -/- and Pah R408W/R408W mice and improved the Phe tolerance of these mice.


Subject(s)
Phenylalanine Hydroxylase/metabolism , Phenylalanine/metabolism , Phenylketonurias/genetics , RNA, Long Noncoding/genetics , Acetylgalactosamine , Animals , Biopterins/analogs & derivatives , Biopterins/metabolism , Biopterins/therapeutic use , Diet , Disease Models, Animal , Female , Hepatocytes/metabolism , Humans , Liver/embryology , Liver/metabolism , Male , Mice , Mice, Knockout , Nucleic Acid Conformation , Phenylalanine/administration & dosage , Phenylalanine Hydroxylase/deficiency , Phenylalanine Hydroxylase/genetics , Phenylketonurias/drug therapy , Phenylketonurias/metabolism , Protein Binding , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/metabolism , RNA, Long Noncoding/therapeutic use
18.
Life Sci Alliance ; 4(8)2021 08.
Article in English | MEDLINE | ID: mdl-34145024

ABSTRACT

Members of the protein kinase D (PKD) family (PKD1, 2, and 3) integrate hormonal and nutritional inputs to regulate complex cellular metabolism. Despite the fact that a number of functions have been annotated to particular PKDs, their molecular targets are relatively poorly explored. PKD3 promotes insulin sensitivity and suppresses lipogenesis in the liver of animals fed a high-fat diet. However, its substrates are largely unknown. Here we applied proteomic approaches to determine PKD3 targets. We identified more than 300 putative targets of PKD3. Furthermore, biochemical analysis revealed that PKD3 regulates cAMP-dependent PKA activity, a master regulator of the hepatic response to glucagon and fasting. PKA regulates glucose, lipid, and amino acid metabolism in the liver, by targeting key enzymes in the respective processes. Among them the PKA targets phenylalanine hydroxylase (PAH) catalyzes the conversion of phenylalanine to tyrosine. Consistently, we showed that PKD3 is activated by glucagon and promotes glucose and tyrosine levels in hepatocytes. Therefore, our data indicate that PKD3 might play a role in the hepatic response to glucagon.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Glucagon/pharmacology , Hepatocytes/cytology , Protein Kinase C/metabolism , Proteomics/methods , Animals , Cells, Cultured , Fasting , Glucose/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Mice , Phenylalanine Hydroxylase/metabolism , Phosphorylation , Primary Cell Culture , Protein Interaction Maps , Tyrosine/metabolism
19.
J Inherit Metab Dis ; 44(6): 1369-1381, 2021 11.
Article in English | MEDLINE | ID: mdl-33896013

ABSTRACT

Phenylketonuria (PKU) is the most common inborn error of metabolism of the liver, and results from mutations of both alleles of the phenylalanine hydroxylase gene (PAH). As such, it is a suitable target for gene therapy via gene delivery with a recombinant adeno-associated virus (AAV) vector. Here we use the synthetic AAV vector Anc80 via systemic administration to deliver a functional copy of a codon-optimized human PAH gene, with or without an intron spacer, to the Pahenu2 mouse model of PKU. Dose-dependent transduction of the liver and expression of PAH mRNA were present with both vectors, resulting in significant and durable reduction of circulating phenylalanine, reaching near control levels in males. Coat color of treated Pahenu2 mice reflected an increase in pigmentation from brown to the black color of control animals, further indicating functional restoration of phenylalanine metabolism and its byproduct melanin. There were no adverse effects associated with administration of AAV up to 5 × 1012 VG/kg, the highest dose tested. Only minor and/or transient variations in some liver enzymes were observed in some of the AAV-dosed animals which were not associated with pathology findings in the liver. Finally, there was no impact on cell turnover or apoptosis as evaluated by Ki-67 and TUNEL staining, further supporting the safety of this approach. This study demonstrates the therapeutic potential of AAV Anc80 to safely and durably cure PKU in a mouse model, supporting development for clinical consideration.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Phenylalanine Hydroxylase/genetics , Phenylketonurias/therapy , Animals , Cell Line , DNA, Recombinant/administration & dosage , Disease Models, Animal , Female , Genetic Vectors/genetics , Hair Color , Humans , Injections, Intravenous , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , Phenylalanine/blood , Phenylalanine Hydroxylase/immunology , Phenylalanine Hydroxylase/metabolism , Transduction, Genetic/methods
20.
Biomolecules ; 11(3)2021 03 19.
Article in English | MEDLINE | ID: mdl-33808760

ABSTRACT

Phenylketonuria (PKU) is a genetic disease caused by deficient activity of human phenylalanine hydroxylase (hPAH) that, when untreated, can lead to severe psychomotor impairment. Protein misfolding is recognized as the main underlying pathogenic mechanism of PKU. Therefore, the use of stabilizers of protein structure and/or activity is an attractive therapeutic strategy for this condition. Here, we report that 3-hydroxyquinolin-2(1H)-one derivatives can act as protectors of hPAH enzyme activity. Electron paramagnetic resonance spectroscopy demonstrated that the 3-hydroxyquinolin-2(1H)-one compounds affect the coordination of the non-heme ferric center at the enzyme active-site. Moreover, surface plasmon resonance studies showed that these stabilizing compounds can be outcompeted by the natural substrate l-phenylalanine. Two of the designed compounds functionally stabilized hPAH by maintaining protein activity. This effect was observed on the recombinant purified protein and in a cellular model. Besides interacting with the catalytic iron, one of the compounds also binds to the N-terminal regulatory domain, although to a different location from the allosteric l-Phe binding site, as supported by the solution structures obtained by small-angle X-ray scattering.


Subject(s)
Phenylalanine Hydroxylase/metabolism , Quinolones/chemistry , Quinolones/pharmacology , Catalytic Domain , Electron Spin Resonance Spectroscopy , Fluorometry , HEK293 Cells , Humans , Metabolic Diseases/metabolism , Models, Molecular , Phenylalanine/metabolism , Phenylketonurias/metabolism , Surface Plasmon Resonance , Trypsin
SELECTION OF CITATIONS
SEARCH DETAIL
...