Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
Mol Genet Genomic Med ; 12(1): e2294, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37818795

ABSTRACT

BACKGROUND: Hyperphenylalaninemia (HPA) is a metabolic disorder classified into phenylalanine-4-hydroxylase (PAH) and non-PAH deficiency. The latter is produced by mutations in genes involved in the tetrahydrobiopterin (BH4) biosynthesis pathway and DNAJC12 pathogenetic variants. The BH4 metabolism, including de novo biosynthesis involved genes (i.e., guanosine 5'-triphosphate cyclohydrolase I (GTPCH/GCH1), sepiapterin reductase (SR/SPR), 6-pyruvoyl-tetrahydropterin synthase (PTPS/PTS)), and two genes that play roles in cofactor regeneration pathway (i.e., dihydropteridine reductase (DHPR/QDPR) and pterin-4α-carbinolamine dehydratase (PCD/PCBD1)). The subsequent systemic hyperphenylalaninemia and monoamine neurotransmitter deficiency lead to neurological consequences. The high rate of consanguineous marriages in Iran substantially increases the incidence of BH4 deficiency. METHODS: We utilized the Sanger sequencing technique in this study to investigate 14 Iranian patients with non-PAH deficiency. All affected subjects in this study had HPA and no mutation was detected in their PAH gene. RESULTS: We successfully identified six mutant alleles in BH4-deficiency-associated genes, including three novel mutations: one in QDPR, one in PTS, and one in the PCBD1 gene, thus giving a definite diagnosis to these patients. CONCLUSION: In this light, appropriate patient management may follow. The clinical effect of reported variants is essential for genetic counseling and prenatal diagnosis in the patients' families and significant for the improvement of precision medicine.


Subject(s)
Phenylalanine Hydroxylase , Phenylketonurias , Pregnancy , Female , Humans , Iran , Phenylketonurias/genetics , Phenylketonurias/epidemiology , Biopterins , Dihydropteridine Reductase/genetics , Phenylalanine Hydroxylase/genetics
2.
Cell Oncol (Dordr) ; 46(5): 1333-1350, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37099250

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) are noncoding RNAs. Accumulating evidence suggests that circRNAs play a critical role in human biological processes, especially tumorigenesis, and development. However, the exact mechanisms of action of circRNAs in hepatocellular carcinoma (HCC) remain unclear. METHODS: Bioinformatic tools and RT-qPCR were used to identify the role of circDHPR, a circRNA derived from the dihydropteridine reductase (DHPR) locus, in HCC and para-carcinoma tissues. Kaplan-Meier analysis and the Cox proportional hazard model were used to analyze the correlation between circDHPR expression and patient prognosis. Lentiviral vectors were used to establish stable circDHPR-overexpressing cells. In vitro and in vivo studies have shown that tumor proliferation and metastasis are affected by circDHPR. Mechanistic assays, including Western blotting, immunohistochemistry, dual-luciferase reporter assays, fluorescence in situ hybridization, and RNA immunoprecipitation, have demonstrated the molecular mechanism underlying circDHPR. RESULTS: CircDHPR was downregulated in HCC, and low circDHPR expression was associated with poor overall survival and disease-free survival rates. CircDHPR overexpression inhibits tumor growth and metastasis in vitro and in vivo. Further systematic studies revealed that circDHPR binds to miR-3194-5p, an upstream regulator of RASGEF1B. This endogenous competition suppresses the silencing effect of miR-3194-5p. We confirmed that circDHPR overexpression inhibited HCC growth and metastasis by sponging miR-3194-5p to upregulate the expression of RASGEF1B, which is regarded as a suppressor of the Ras/MAPK signaling pathway. CONCLUSIONS: Aberrant circDHPR expression leads to uncontrolled cell proliferation, tumorigenesis, and metastasis. CircDHPR may serve as a biomarker and therapeutic target for HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , Humans , Carcinoma, Hepatocellular/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Liver Neoplasms/metabolism , Dihydropteridine Reductase/genetics , Dihydropteridine Reductase/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , In Situ Hybridization, Fluorescence , Cell Line, Tumor , Cell Proliferation/genetics , Carcinogenesis/pathology , Gene Expression Regulation, Neoplastic
3.
J Pharmacol Sci ; 150(3): 173-179, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36184122

ABSTRACT

Quinonoid dihydropteridine reductase (QDPR) regenerates tetrahydrobiopterin (BH4), which is an essential cofactor for catecholamine and serotonin (5-hydroxytryptamine, 5-HT) biosynthesis. Serotonin is known as an important platelet agonist, but its role under BH4-synthesizing or recycling enzymes deficiency is unknown. In the present study, we evaluated the effect of Qdpr gene disruption on platelet aggregation using knockout (Qdpr-/-) mice. Platelet aggregation was monitored by light transmission aggregometry using adenosine diphosphate (ADP) and collagen as agonists. We also assessed how platelet aggregation was modified by 5-HT recovery through supplementation with 5-hydroxytryptophan (5-HTP), a 5-HT precursor, or by blocking the serotonin 5-HT2A receptor. Platelet aggregation in the Qdpr-/- mice was significantly suppressed in comparison with that in wild-type (Qdpr+/+) mice, particularly at the maintenance phase of aggregation. 5-HT storage was decreased in Qdpr-/- platelets, and 5-HTP supplementation recovered not only the intraplatelet 5-HT levels but also platelet aggregation. In addition, 5-HT signal blockade using sarpogrelate suppressed platelet aggregation in Qdpr+/+ mice, and platelets in Qdpr-/- mice were hardly affected. Our results indicate that QDPR deficiency suppresses platelet aggregation by impairing 5-HT biosynthesis in mice.


Subject(s)
Dihydropteridine Reductase , Platelet Aggregation , 5-Hydroxytryptophan/pharmacology , Adenosine Diphosphate/pharmacology , Animals , Biopterins/analogs & derivatives , Catecholamines , Collagen , Dihydropteridine Reductase/genetics , Dihydropteridine Reductase/pharmacology , Mice , Receptor, Serotonin, 5-HT2A , Serotonin/pharmacology
4.
Mol Genet Metab ; 133(2): 123-136, 2021 06.
Article in English | MEDLINE | ID: mdl-33903016

ABSTRACT

Tetrahydrobiopterin (BH4) deficiency is caused by genetic variants in the three genes involved in de novo cofactor biosynthesis, GTP cyclohydrolase I (GTPCH/GCH1), 6-pyruvoyl-tetrahydropterin synthase (PTPS/PTS), sepiapterin reductase (SR/SPR), and the two genes involved in cofactor recycling, carbinolamine-4α-dehydratase (PCD/PCBD1) and dihydropteridine reductase (DHPR/QDPR). Dysfunction in BH4 metabolism leads to reduced cofactor levels and may result in systemic hyperphenylalaninemia and/or neurological sequelae due to secondary deficiency in monoamine neurotransmitters in the central nervous system. More than 1100 patients with BH4 deficiency and 800 different allelic variants distributed throughout the individual genes are tabulated in database of pediatric neurotransmitter disorders PNDdb. Here we provide an update on the molecular-genetic analysis and structural considerations of these variants, including the clinical courses of the genotypes. From a total of 324 alleles, 11 are associated with the autosomal recessive form of GTPCH deficiency presenting with hyperphenylalaninemia (HPA) and neurotransmitter deficiency, 295 GCH1 variant alleles are detected in the dominant form of L-dopa-responsive dystonia (DRD or Segawa disease) while phenotypes of 18 alleles remained undefined. Autosomal recessive variants observed in the PTS (199 variants), PCBD1 (32 variants), and QDPR (141 variants) genes lead to HPA concomitant with central monoamine neurotransmitter deficiency, while SPR deficiency (104 variants) presents without hyperphenylalaninemia. The clinical impact of reported variants is essential for genetic counseling and important for development of precision medicine.


Subject(s)
Alcohol Oxidoreductases/genetics , GTP Cyclohydrolase/genetics , Phenylketonurias/genetics , Phosphorus-Oxygen Lyases/genetics , Biopterins/analogs & derivatives , Biopterins/genetics , Biopterins/metabolism , Dihydropteridine Reductase/genetics , Dystonia/genetics , Dystonia/metabolism , Dystonia/pathology , Genetic Predisposition to Disease , Humans , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/metabolism , Metabolism, Inborn Errors/pathology , Microtubule-Associated Proteins/genetics , Phenylketonurias/classification , Phenylketonurias/metabolism , Phenylketonurias/pathology , Psychomotor Disorders/genetics , Psychomotor Disorders/metabolism , Psychomotor Disorders/pathology
5.
Sci Adv ; 5(9): eaax0365, 2019 09.
Article in English | MEDLINE | ID: mdl-31555733

ABSTRACT

Mitochondria are highly dynamic organelles. Through a large-scale in vivo RNA interference (RNAi) screen that covered around a quarter of the Drosophila melanogaster genes (4000 genes), we identified 578 genes whose knockdown led to aberrant shapes or distributions of mitochondria. The complex analysis revealed that knockdown of the subunits of proteasomes, spliceosomes, and the electron transport chain complexes could severely affect mitochondrial morphology. The loss of Dhpr, a gene encoding an enzyme catalyzing tetrahydrobiopterin regeneration, leads to a reduction in the numbers of tyrosine hydroxylase neurons, shorter lifespan, and gradual loss of muscle integrity and climbing ability. The affected mitochondria in Dhpr mutants are swollen and have fewer cristae, probably due to lower levels of Drp1 S-nitrosylation. Overexpression of Drp1, but not of S-nitrosylation-defective Drp1, rescued Dhpr RNAi-induced mitochondrial defects. We propose that Dhpr regulates mitochondrial morphology and tissue homeostasis by modulating S-nitrosylation of Drp1.


Subject(s)
Dihydropteridine Reductase , Drosophila Proteins , Mitochondria , Mitochondrial Proteins , Animals , Dihydropteridine Reductase/genetics , Dihydropteridine Reductase/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , RNA Interference
6.
PLoS One ; 14(4): e0215162, 2019.
Article in English | MEDLINE | ID: mdl-30995231

ABSTRACT

Dihydropteridine reductase (QDPR) catalyzes the recycling of tetrahydrobiopterin (BH4), a cofactor in dopamine, serotonin, and phenylalanine metabolism. QDPR-deficient patients develop neurological symptoms including hypokinesia, truncal hypotonia, intellectual disability and seizures. The underlying pathomechanisms are poorly understood. We established a zebrafish model for QDPR deficiency and analyzed the expression as well as function of all zebrafish QDPR homologues during embryonic development. The homologues qdpra is essential for pigmentation and phenylalanine metabolism. Qdprb1 is expressed in the proliferative zones of the optic tectum and eye. Knockdown of qdprb1 leads to up-regulation of pro-proliferative genes and increased number of phospho-histone3 positive mitotic cells. Expression of neuronal and astroglial marker genes is concomitantly decreased. Qdprb1 hypomorphic embryos develop microcephaly and reduced eye size indicating a role for qdprb1 in the transition from cell proliferation to differentiation. Glutamine accumulation biochemically accompanies the developmental changes. Our findings provide novel insights into the neuropathogenesis of QDPR deficiency.


Subject(s)
Cell Proliferation/genetics , Glutamine , Melanins , Neuroglia/metabolism , Phenylketonurias , Zebrafish , Animals , Dihydropteridine Reductase/genetics , Disease Models, Animal , Gene Knockdown Techniques , Glutamine/genetics , Glutamine/metabolism , Humans , Melanins/biosynthesis , Melanins/genetics , Phenylketonurias/genetics , Phenylketonurias/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
7.
J Pediatr Endocrinol Metab ; 31(8): 911-916, 2018 Aug 28.
Article in English | MEDLINE | ID: mdl-30001213

ABSTRACT

Background The overall incidence of hyperphenylalaninemia (HPA) in China is 1:11,763, with tetrahydrobiopterin (BH4) deficiency accounting for 8.55% of patients with HPA in the mainland. Much progress has been made in the diagnosis and treatment of BH4 deficiency with the introduction of neonatal screening in China. However, the screening rate is still low and screening is not universally available. Methods A total of 44 BH4-deficient patients were enrolled in this study, of which 39 were diagnosed with BH4 deficiency, while the remaining five showed typical characteristics of BH4 deficiency at a later period. The entire coding regions and adjacent intronic regions of GCH1, PTS, PCBD1 and QDPR genes were analyzed using target sequencing. Results Nineteen (n=19) different mutations in the PTS gene including four novel mutations and one mutation in QDPR were identified. p.P87S, p.D96N, IVS1-291A>G, p.N52S, p.K91R, p.V56M, p.T106M and p.F40GfsX53 in PTS were the prevalent mutations with ≥3% relative frequency. The mutation p.R221X in the QDPR gene was found with relatively lower frequencies (2.27%). The remaining 12 mutations in PTS were found at relative frequencies of 1.14%. Conclusions The results could be of value for genetic counseling and prenatal diagnosis in the patients' families and for the molecular diagnosis of BH4 deficiencies. Furthermore, four novel mutations expand and improve the PTS mutation database.


Subject(s)
Asian People/genetics , Biomarkers/analysis , Mutation , Neonatal Screening , Phenylketonurias/diagnosis , Phenylketonurias/genetics , Prenatal Diagnosis , Child, Preschool , China/epidemiology , Dihydropteridine Reductase/genetics , Female , Follow-Up Studies , GTP Cyclohydrolase/genetics , Humans , Hydro-Lyases/genetics , Infant , Infant, Newborn , Male , Peroxisomal Targeting Signals/genetics , Phenylketonurias/epidemiology , Prognosis
8.
Int J Biol Macromol ; 110: 567-572, 2018 Apr 15.
Article in English | MEDLINE | ID: mdl-29355631

ABSTRACT

Quinine dihydropteridinereductase (QDPR) is involved in the synthesis of tetradihydrobiopteridine (BH4) that serve as cofactor for many aromatic hydroxylases including induced nitric oxide synthase (NOS) leading to NO production. Increased activity of QDPR has been associated with decrease levels of TGF-ß, a cytokine that regulates the immune response and that elevated levels of NO has been associated with neurodegenerative diseases. Thus, expression of QDPR in astrocytes is essential to study the pathological changes observed in many neurodegenerative disorders. We have expressed QDPR in astrocytes and generated stably expressing clones that overexpresses QDPR. We further verified the specificity of QDPR expression using immunofluorescence and immunoblotting. To further confirm, we purified QDPR using Ni-NTA column and subjected the purified fraction to immunoblotting using anti-QDPR antibody and identified two major protein products of QDPR resolving at 25 and 17 kDa as reported in the literature. In order to further assess the significance of QDPR expression, we verified the expression of iNOS in QDPR over expressing cells. We show for the first time statistically significant up regulation of iNOS in QDPR overexpressing astrocytes. Increased expression of iNOS associated with astrocyte pathology seen in many neurodegenerative disorders may have implications in autoimmune neurodegenerative disorders.


Subject(s)
Astrocytes/enzymology , Dihydropteridine Reductase , Gene Expression Regulation, Enzymologic , Neurodegenerative Diseases/enzymology , Astrocytes/pathology , Dihydropteridine Reductase/biosynthesis , Dihydropteridine Reductase/chemistry , Dihydropteridine Reductase/genetics , Dihydropteridine Reductase/isolation & purification , Humans , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics
9.
Acta Biochim Biophys Sin (Shanghai) ; 49(8): 706-712, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28633336

ABSTRACT

Dihydropteridine reductase (QDPR) plays an important role in the recycling of BH4 and is closely related to oxidative stress. We have previously reported that the overexpression of QDPR in human kidney HEK293T cells significantly protected against oxidative stress, and these beneficial effects were abolished by A278C mutation. To evaluate the effect of wild-type and mutant QDPR on autophagy and its mechanism in HEK293T cells, we constructed the wild-type and mutant QDPR expression plasmids and transfected them into HEK293T cells. Three days later, cells were collected to observe the expression of fusion protein and the intracellular production of reactive oxygen species (ROS). Western blot analysis was employed to evaluate the change of mTOR and ribosomal protein S6 kinase B1 (S6K1) signaling and the expression of LC-I, LC-II, Bcl-1, Bcl-2, p62, and p53. The results showed that the exogenous wild-type QDPR significantly decreased the expression of mTOR and phosphorylation of the mTOR and S6K1. Mutation of QDPR inhibited the regulation of mTOR, suggesting that QDPR is a positive regulator of autophagy via suppressing mTOR signaling. The expressions of p62, LC3-II and Beclin 1 were dramatically enhanced in wild-type QDPR group, which were reversed after QDPR mutation. Additionally, mutation of QDPR altered the upregulation of QDPR on Beclin 2. It is therefore concluded that QDPR appears to play an important role in enhancing autophagy, and its mutation contributes to dysregulation of autophagy.


Subject(s)
Autophagy/genetics , Dihydropteridine Reductase/genetics , Mutation , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , Dihydropteridine Reductase/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Oxidative Stress , Phosphorylation , Reactive Oxygen Species/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism
10.
Mol Genet Metab ; 121(2): 83-90, 2017 06.
Article in English | MEDLINE | ID: mdl-28412083

ABSTRACT

We sought to determine the molecular composition of human cerebrospinal fluid (CSF) and identify the biochemical pathways represented in CSF to understand the potential for untargeted screening of inborn errors of metabolism (IEMs). Biochemical profiles for each sample were obtained using an integrated metabolomics workflow comprised of four chromatographic techniques followed by mass spectrometry. Secondarily, we wanted to compare the biochemical profile of CSF with those of plasma and urine within the integrated mass spectrometric-based metabolomic workflow. Three sample types, CSF (N=30), urine (N=40) and EDTA plasma (N=31), were analyzed from retrospectively collected pediatric cohorts of equivalent age and gender characteristics. We identified 435 biochemicals in CSF representing numerous biological and chemical/structural families. Sixty-three percent (273 of 435) of the biochemicals detected in CSF also were detected in urine and plasma, another 32% (140 of 435) were detected in either plasma or urine, and 5% (22 of 435) were detected only in CSF. Analyses of several metabolites showed agreement between clinically useful assays and the metabolomics approach. An additional set of CSF and plasma samples collected from the same patient revealed correlation between several biochemicals detected in paired samples. Finally, analysis of CSF from a pediatric case with dihydropteridine reductase (DHPR) deficiency demonstrated the utility of untargeted global metabolic phenotyping as a broad assessment to screen samples from patients with undifferentiated phenotypes. The results indicate a single CSF sample processed with an integrated metabolomics workflow can be used to identify a large breadth of biochemicals that could be useful for identifying disrupted metabolic patterns associated with IEMs.


Subject(s)
Cerebrospinal Fluid Proteins/genetics , Cerebrospinal Fluid Proteins/metabolism , Cerebrospinal Fluid/chemistry , Cerebrospinal Fluid/metabolism , Metabolome , Metabolomics/methods , Adolescent , Biomarkers/blood , Biomarkers/urine , Cerebrospinal Fluid Proteins/analysis , Cerebrospinal Fluid Proteins/chemistry , Child , Child, Preschool , Dihydropteridine Reductase/blood , Dihydropteridine Reductase/genetics , Dihydropteridine Reductase/metabolism , Dihydropteridine Reductase/urine , Female , Humans , Infant , Male , Mass Spectrometry/methods , Metabolism, Inborn Errors/diagnosis , Phenotype , Retrospective Studies , Young Adult
11.
Sci Rep ; 6: 35285, 2016 10 21.
Article in English | MEDLINE | ID: mdl-27767067

ABSTRACT

Nitric oxide (NO) is a toxic free radical produced by neutrophils and macrophages in response to infection. Uropathogenic Escherichia coli (UPEC) induces a variety of defence mechanisms in response to NO, including direct NO detoxification (Hmp, NorVW, NrfA), iron-sulphur cluster repair (YtfE), and the expression of the NO-tolerant cytochrome bd-I respiratory oxidase (CydAB). The current study quantifies the relative contribution of these systems to UPEC growth and survival during infection. Loss of the flavohemoglobin Hmp and cytochrome bd-I elicit the greatest sensitivity to NO-mediated growth inhibition, whereas all but the periplasmic nitrite reductase NrfA provide protection against neutrophil killing and promote survival within activated macrophages. Intriguingly, the cytochrome bd-I respiratory oxidase was the only system that augmented UPEC survival in a mouse model after 2 days, suggesting that maintaining aerobic respiration under conditions of nitrosative stress is a key factor for host colonisation. These findings suggest that while UPEC have acquired a host of specialized mechanisms to evade nitrosative stresses, the cytochrome bd-I respiratory oxidase is the main contributor to NO tolerance and host colonisation under microaerobic conditions. This respiratory complex is therefore of major importance for the accumulation of high bacterial loads during infection of the urinary tract.


Subject(s)
Cytochromes/genetics , Dihydropteridine Reductase/genetics , Electron Transport Chain Complex Proteins/genetics , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Hemeproteins/genetics , Host-Pathogen Interactions , NADH, NADPH Oxidoreductases/genetics , Oxidoreductases/genetics , Uropathogenic Escherichia coli/genetics , Animals , Cytochrome b Group , Cytochrome c Group/deficiency , Cytochrome c Group/genetics , Cytochromes/deficiency , Disease Models, Animal , Drug Resistance, Multiple, Bacterial/genetics , Electron Transport Chain Complex Proteins/deficiency , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Hemeproteins/deficiency , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbial Viability , NADH, NADPH Oxidoreductases/deficiency , Neutrophils/immunology , Neutrophils/microbiology , Nitric Oxide/metabolism , Oxidoreductases/deficiency , Urinary Tract Infections/microbiology , Uropathogenic Escherichia coli/growth & development
12.
BMC Med Genet ; 17(1): 65, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27613114

ABSTRACT

BACKGROUND: Mutations in Leucine-rich repeat kinase 2 NM_198578 (LRRK2 c.6055G > A (p.G2019S), LRRK2 c.4321C > G (p.R1441G)) and alpha-synuclein NM_000345 (SNCA c.209G > A (p.A53T)) genes causing Parkinson's disease (PD) are common in Mediterranean populations. Variants in the Quinoid Dihydropteridine Reductase NM_000320 (QDPR c.68G > A (p.G23D)), Sepiapterin Reductase NM_003124 (SPR c.596-2A > G) and Methylenetetrahydrofolate Reductase NM_005957 (MTHFR c.677C > T and c.1298A > C) genes are frequent in Malta and potential candidates for PD. METHODS: 178 cases and 402 control samples from Malta collected as part of the Geoparkinson project were genotyped for MTHFR polymorphisms, QDPR and SPR mutations. Only PD and parkinsonism cases were tested for SNCA and LRRK2 mutations. RESULTS: LRRK2 c.4321C > G and SNCA c.209G > A were not detected. The LRRK2 c.6055G > A mutation was found in 3.1 % of Maltese PD cases. The QDPR mutation was found in both cases and controls and did not increase risk for PD. The SPR mutation was found in controls only. The odds ratios for MTHFR polymorphisms were not elevated. CONCLUSIONS: The LRRK2 c.6055G > A is a cause of PD in the Maltese, whilst QDPR c.68G > A, SPR c.596-2A > G and MTHFR c.677C > T and c.1298A > C are not important determinants of PD.


Subject(s)
Alcohol Oxidoreductases/genetics , Dihydropteridine Reductase/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Parkinson Disease/genetics , White People/genetics , Adult , Aged , Aged, 80 and over , Alleles , Case-Control Studies , Female , Gene Frequency , Genotype , Humans , Male , Malta , Middle Aged , Odds Ratio , Parkinson Disease/pathology , Polymorphism, Single Nucleotide
13.
Mol Biol Evol ; 33(8): 1979-87, 2016 08.
Article in English | MEDLINE | ID: mdl-27189567

ABSTRACT

Due to their functional independence, proteins that comprise standalone metabolic units, which we name single-protein metabolic modules, may be particularly prone to gene duplication (GD) and horizontal gene transfer (HGT). Flavohemoglobins (flavoHbs) are prime examples of single-protein metabolic modules, detoxifying nitric oxide (NO), a ubiquitous toxin whose antimicrobial properties many life forms exploit, to nitrate, a common source of nitrogen for organisms. FlavoHbs appear widespread in bacteria and have been identified in a handful of microbial eukaryotes, but how the distribution of this ecologically and biomedically important protein family evolved remains unknown. Reconstruction of the evolutionary history of 3,318 flavoHb protein sequences covering the family's known diversity showed evidence of recurrent HGT at multiple evolutionary scales including intrabacterial HGT, as well as HGT from bacteria to eukaryotes. One of the most striking examples of HGT is the acquisition of a flavoHb by the dandruff- and eczema-causing fungus Malassezia from Corynebacterium Actinobacteria, a transfer that growth experiments show is capable of mediating NO resistance in fungi. Other flavoHbs arose via GD; for example, many filamentous fungi possess two flavoHbs that are differentially targeted to the cytosol and mitochondria, likely conferring protection against external and internal sources of NO, respectively. Because single-protein metabolic modules such as flavoHb function independently, readily undergo GD and HGT, and are frequently involved in organismal defense and competition, we suggest that they represent "plug-and-play" proteins for ecological arms races.


Subject(s)
Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Eukaryota/genetics , Eukaryota/metabolism , Hemeproteins/genetics , Hemeproteins/metabolism , Nitric Oxide/metabolism , Adaptation, Biological/genetics , Amino Acid Sequence , Biological Evolution , Computational Biology , Databases, Nucleic Acid , Dihydropteridine Reductase/genetics , Dihydropteridine Reductase/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Evolution, Molecular , Fungi/genetics , Gene Duplication , Gene Transfer, Horizontal , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , Phylogeny
14.
Zhonghua Fu Chan Ke Za Zhi ; 51(12): 890-894, 2016 Dec 25.
Article in Chinese | MEDLINE | ID: mdl-28057123

ABSTRACT

Objective: To analyze the variations of PTPS gene in patients with suspected 6-pyruvoyl-tetra hydropterin synthase deficiency (PTPSD) and to make prenatal diagnosis in high-risk families. Methods: Chemiluminescence was used for phenylalanine detection in blood or dried blood spots.Patients with phenylalanine concentration over 120 µmol/L were detected by urine pterin analysis, and the activity of dihydropteridine reductase (DHPR) was detected. tetrahydrobiopterin loading tests were performed in suspected patients with abnormal urinary pterin profiles. PTPS gene variation analysis was performed by direct Sanger sequencing based on PCR amplification. Prenatal diagnosis in 7 high-risk families was performed by chorionic villus sampling when the genotype was identified. Results: In 656 patients with hyperphenylalanine, 22 cases were diagnosed as PTPSD clinically. 16 variations were detected in the 22 PTPSD cases. The 5 variations, p.Lys77Arg, p.Ile84Phe, c.315-2A>G, c.244-2A>T, c.187-1G>T, were identified as novel variations. Two fetuses carried the same mutation with the proband and therefore were thought to be PTPSD fetuses. Three fetuses carried only one mutant allele and thus were thought to be PTPSD carriers. The other 2 fetuses carried no mutations and were presumed normal. Conclusions: PTPS gene variation analysis is necessary to confirm the diagnosis. Prenatal diagnosis could help avoiding the defect birth in PTPSD families.


Subject(s)
Dihydropteridine Reductase/genetics , Mutation/genetics , Phenylketonurias/genetics , Phosphorus-Oxygen Lyases/deficiency , Prenatal Diagnosis , Alleles , Biopterins/analogs & derivatives , Chorionic Villi Sampling , Female , Fetus , Genetic Testing , Genotype , Heterozygote , Humans , Luminescence , Nitric Oxide Synthase , Phenylalanine/blood , Phenylketonurias/diagnosis , Phosphorus-Oxygen Lyases/genetics , Polymerase Chain Reaction , Pregnancy
15.
Metab Eng ; 31: 22-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26112956

ABSTRACT

The importance of NO(∙) to immunity is highlighted by the diversity of pathogens that require NO(∙)-defensive systems to establish infections. Proteases have been identified to aid pathogens in surviving macrophage attack, inspiring us to investigate their role during NO(∙) stress in Escherichia coli. We discovered that the elimination of ClpP largely impaired NO(∙) detoxification by E. coli. Using a quantitative model of NO(∙) stress, we employed an ensemble-guided approach to identify the underlying mechanism. Iterations of in silico analyses and corresponding experiments identified the defect to result from deficient transcript levels of hmp, which encodes NO(∙) dioxygenase. Interestingly, the defect was not confined to hmp, as ΔclpP imparted widespread perturbations to the expression of NO(∙)-responsive genes. This work identified a target for anti-infective therapies based on disabling NO(∙) defenses, and demonstrated the utility of model-based approaches for exploring the complex, systems-level stress exerted by NO(∙).


Subject(s)
Dihydropteridine Reductase/genetics , Endopeptidase Clp/physiology , Escherichia coli Proteins/genetics , Escherichia coli Proteins/physiology , Escherichia coli/genetics , Hemeproteins/genetics , NADH, NADPH Oxidoreductases/genetics , Nitric Oxide/metabolism , RNA, Messenger/analysis , Biocatalysis , Escherichia coli/metabolism , Promoter Regions, Genetic , Sequence Analysis, RNA , Stress, Physiological , Transcription, Genetic
16.
Article in English | MEDLINE | ID: mdl-25899859

ABSTRACT

Tetrahydrobiopterin (BH4) is an essential cofactor of aromatic amino acid hydroxylases and nitric oxide synthase so that BH4 plays a key role in many biological processes. BH4 deficiency is associated with numerous metabolic syndromes and neuropsychological disorders. BH4 concentration in mammals is maintained through a de novo synthesis pathway and a regeneration pathway. Previous studies showed that the de novo pathway of BH4 is similar between insects and mammals. However, knowledge about the regeneration pathway of BH4 (RPB) is very limited in insects. Several mutants in the silkworm Bombyx mori have been approved to be associated with BH4 deficiency, which are good models to research on the RPB in insects. In this study, homologous genes encoding two enzymes, pterin-4a-carbinolamine dehydratase (PCD) and dihydropteridine reductase (DHPR) involving in RPB have been cloned and identified from B. mori. Enzymatic activity of DHPR was found in the fat body of wild type silkworm larvae. Together with the transcription profiles, it was indicated that BmPcd and BmDhpr might normally act in the RPB of B. mori and the expression of BmDhpr was activated in the brain and sexual glands while BmPcd was expressed in a wider special pattern when the de novo pathway of BH4 was lacked in lemon. Biochemical analyses showed that the recombinant BmDHPR exhibited high enzymatic activity and more suitable parameters to the coenzyme of NADH in vitro. The results in this report give new information about the RPB in B. mori and help in better understanding insect BH4 biosynthetic networks.


Subject(s)
Biopterins/analogs & derivatives , Bombyx/enzymology , Bombyx/physiology , Dihydropteridine Reductase/metabolism , Hydro-Lyases/metabolism , Larva/physiology , Amino Acid Sequence , Animals , Biopterins/metabolism , Bombyx/chemistry , Dihydropteridine Reductase/chemistry , Dihydropteridine Reductase/genetics , Enzyme Activation , Hydro-Lyases/chemistry , Hydro-Lyases/genetics , Metabolic Networks and Pathways , Molecular Sequence Data , RNA, Messenger/metabolism , Regeneration
17.
Ir Med J ; 108(10): 312-4, 2015.
Article in English | MEDLINE | ID: mdl-26817292

ABSTRACT

DHPR deficiency is a rare autosomal recessively inherited metabolic disorder of tetrahydrobiopterin (BH4) regeneration. Clinical symptoms may comprise microcephaly, developmental delay, ataxia and seizures. BH4 is the cofactor for the enzyme phenylalanine (Phe)hydroxylase (PAH), and for tryptophan and tyrosine hydroxylases, both of which are essential for serotonin and dopamine biosynthesis. We present four patients in two families who are being treated at the National Centre for Inherited Metabolic Disorders (NCIMD). All are members of the Irish Traveller population. We have identified a homozygous mutation, c.353C>T, in the DHPR (QDPR) gene which, to the best of our knowledge, has not been previously described. The mainstay of treatment is a life-long Phe-restricted diet together with supplementation of L-dopa and 5-hydroxy tryptophan (5-HT) and folinic acid. In Ireland, there is neurological comorbidity in our adult DHPR patients, although the overall outcome is satisfactory and one affected female has three healthy children.


Subject(s)
Dihydropteridine Reductase/genetics , Phenylketonurias/genetics , Adult , Female , Humans , Infant , Male
18.
World J Pediatr ; 10(3): 219-26, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25124972

ABSTRACT

BACKGROUND: This study aimed to investigate the mutation spectrum of the QDPR gene, to determine the effect of mutations on dihydropteridine reductase (DHPR) structure/function, to discuss the potential genotypephenotype correlation, and to evaluate the clinical outcome of Chinese patients after treatment. METHODS: Nine DHPR-deficient patients were enrolled in this study and seven of them underwent neonatal screening. QDPR gene mutations were analyzed and confirmed by routine methods. The potential pathogenicity of missense variants was analyzed using Clustal X, PolyPhen program and Swiss-PDB Viewer 4.04_OSX software, respectively. The clinical outcomes of the patients were evaluated after long-term treatment. RESULTS: In 10 mutations of the 9 patients, 4 were novel mutations (G20V, V86D, G130S and A175R), 4 were reported by us previously, and 2 known mutations were identified. R221X was a hotspot mutation (27.7%) in our patients. Eight missense mutations probably had damage to protein. Six patients in this series were treated with a good control of phenylalanine level. The height and weight of the patients were normal at the age of 4 months to 7.5 years. Four patients, who underwent a neonatal screening and were treated early, showed a normal mental development. In 2 patients diagnosed late, neurological symptoms were significantly improved. CONCLUSIONS: The mutation spectrum of the QDPR gene is different in the Chinese population. Most mutations are related to severe phenotype. The determination of DHPR activity should be performed in patients with hyperphenylalaninemia. DHPR-deficient patients who were treated below the age of 2 months may have a near normal mental development.


Subject(s)
Asian People/genetics , Dihydropteridine Reductase/genetics , Mutation , Phenylketonurias/genetics , Biomarkers/blood , Female , Follow-Up Studies , Genotype , Humans , Infant, Newborn , Male , Mutation, Missense , Neonatal Screening , Phenotype , Phenylketonurias/diagnosis , Phenylketonurias/drug therapy , Point Mutation , Treatment Outcome
19.
Arch Microbiol ; 195(8): 559-69, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23824318

ABSTRACT

The csgD gene codes for the regulatory protein CsgD. CsgD upregulates the synthesis of the adhesive fimbriae, curli, that are important for biofilm formation and downregulates flagellar synthesis. We compared the expression of genes involved in folate metabolism and a gene (hmp) in strains with an intact csgD gene and with a deletion in csgD. The hmp gene codes a flavohemoglobin that inactivates nitric oxide. Expression was monitored by measuring light production from single copy lux operon fusions. At late times of growth, expression of genes responsible for methylene tetrahydrofolate synthesis (glyA and gcvTHP) and formyltetrahydrofolate recycling (purU) was higher in cells with CsgD than those without. In contrast, expression of hmp was lower in the presence of CsgD throughout the period monitored. We used a novel defined medium which should assist in defining nutritional factors that contribute to curli formation.


Subject(s)
Dihydropteridine Reductase/genetics , Escherichia coli K12/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Folic Acid/metabolism , Gene Expression Regulation, Bacterial , Hemeproteins/genetics , NADH, NADPH Oxidoreductases/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Dihydropteridine Reductase/metabolism , Escherichia coli K12/metabolism , Hemeproteins/metabolism , Mutation , NADH, NADPH Oxidoreductases/metabolism , Operon , Promoter Regions, Genetic
20.
J Inherit Metab Dis ; 35(6): 963-73, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22729819

ABSTRACT

OBJECTIVES: The present study summarizes clinical and biochemical findings, current treatment strategies and follow-up in patients with tetrahydrobiopterin (BH(4)) deficiencies. METHODS: We analyzed the clinical, biochemical and treatment data of 626 patients with BH(4) deficiencies [355 with 6-pyruvoyl-tetrahydropterin synthase (PTPS), 217 with dihydropteridine reductase (DHPR), 31 with autosomal recessive GTP cyclohydrolase I (GTPCH), and 23 with pterin-4a-carbinolamine dehydratase (PCD) deficiencies] from the BIODEF Database. Patients with autosomal dominant GTPCH and SR deficiencies will not be discussed in detail. RESULTS: Up to 57 % of neonates with BH(4) deficiencies are already clinically symptomatic. During infancy and childhood, the predominant symptoms are muscular hypotonia, mental retardation and age-dependent movement disorders, including dystonia. The laboratory diagnosis of BH(4) deficiency is based on a positive newborn screening (NBS) for phenylketonuria (PKU), characteristic profiles of urinary or dried blood spot pterins (biopterin, neopterin, and primapterin), and the measurement of DHPR activity in blood. Some patients with autosomal recessive GTPCH deficiency and all with sepiapterin reductase deficiency may be diagnosed late due to normal blood phenylalanine in NBS. L-dopa, 5-hydroxytryptophan, and BH(4) are supplemented in PTPS and GTPCH-deficient patients, whereas L-dopa, 5-hydroxytryptophan, folinic acid and diet are used in DHPR-deficient patients. Medication doses vary widely among patients, and our understanding of the effects of dopamine agonists and monoamine catabolism inhibitors are limited. CONCLUSIONS: BH(4) deficiencies are a group of treatable pediatric neurotransmitter disorders that are characterized by motor dysfunction, mental retardation, impaired muscle tone, movement disorders and epileptic seizures. Although the outcomes of BH(4) deficiencies are highly variable, early diagnosis and treatment result in improved outcomes.


Subject(s)
Biopterins/analogs & derivatives , Phenylketonurias/metabolism , Biopterins/deficiency , Data Collection , Databases, Factual , Dihydropteridine Reductase/genetics , Female , Humans , Hydro-Lyases/deficiency , Hydro-Lyases/genetics , Infant , Infant, Newborn , Internationality , Male , Phenylketonurias/diagnosis , Phenylketonurias/genetics , Phenylketonurias/therapy , Phosphorus-Oxygen Lyases/deficiency , Phosphorus-Oxygen Lyases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...