Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
1.
Biochem Biophys Res Commun ; 717: 150059, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38723517

ABSTRACT

Tetrahydrobiopterin (BH4) is an essential cofactor for dopamine and serotonin synthesis in monoaminergic neurons, phenylalanine metabolism in hepatocytes, and nitric oxide synthesis in endothelial and immune cells. BH4 is consumed as a cofactor or is readily oxidized by autooxidation. Quinonoid dihydropteridine reductase (QDPR) is an enzyme that reduces quinonoid dihydrobiopterin (qBH2) back to BH4, and we have previously demonstrated the significance of QDPR in maintaining BH4 in vivo using Qdpr-KO mice. In addition to the levels of BH4 in the cells, the ratios of oxidized to reduced forms of BH4 are supposed to be important for regulating nitric oxide synthase (NOS) via the so-called uncoupling of NOS. However, previous studies were limited due to the absence of specific and high-affinity inhibitors against QDPR. Here, we performed a high-throughput screening for a QDPR inhibitor and identified Compound 9b with an IC50 of 0.72 µM. To understand the inhibition mechanism, we performed kinetic analyses and molecular dynamics simulations. Treatment with 9b combined with methotrexate (MTX), an inhibitor of another BH4-reducing enzyme, dihydrofolate reductase (DHFR), significantly oxidized intracellular redox states in HepG2, Jurkat, SH-SY5Y, and PC12D cells. Collectively, these findings suggest that 9b may enhance the anticancer and anti-autoimmune effects of MTX.


Subject(s)
Biopterins , Dihydropteridine Reductase , Drug Synergism , Methotrexate , Methotrexate/pharmacology , Biopterins/analogs & derivatives , Biopterins/metabolism , Humans , Dihydropteridine Reductase/metabolism , Enzyme Inhibitors/pharmacology , Oxidation-Reduction/drug effects , Animals , Molecular Dynamics Simulation
2.
J Biochem ; 174(5): 441-450, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37540845

ABSTRACT

Quinonoid dihydropteridine reductase (QDPR) catalyses the reduction of quinonoid-form dihydrobiopterin (qBH2) to tetrahydrobiopterin (BH4). BH4 metabolism is a drug target for neglected tropical disorders because trypanosomatid protozoans, including Leishmania and Trypanosoma, require exogenous sources of biopterin for growth. Although QDPR is a key enzyme for maintaining intracellular BH4 levels, the precise catalytic properties and reaction mechanisms of QDPR are poorly understood due to the instability of quinonoid-form substrates. In this study, we analysed the binding profile of qBH2 to human QDPR in combination with in silico and in vitro methods. First, we performed docking simulation of qBH2 to QDPR to obtain possible binding modes of qBH2 at the active site of QDPR. Then, among them, we determined the most plausible binding mode using molecular dynamics simulations revealing its atomic-level interactions and confirmed it with the in vitro assay of mutant enzymes. Moreover, it was found that not only qBH2 but also quinonoid-form dihydrofolate (qDHF) could be potential physiological substrates for QDPR, suggesting that QDPR may be a bifunctional enzyme. These findings in this study provide important insights into biopterin and folate metabolism and would be useful for developing drugs for neglected tropical diseases.


Subject(s)
Biopterins , Dihydropteridine Reductase , Humans , Dihydropteridine Reductase/metabolism
3.
Free Radic Biol Med ; 205: 90-99, 2023 08 20.
Article in English | MEDLINE | ID: mdl-37253411

ABSTRACT

Nitric oxide (·NO) is a prevalent antimicrobial that is known to damage iron-containing enzymes in amino acid (AA) biosynthesis pathways. With Escherichia coli, ·NO is detoxified in aerobic environments by Hmp, which is an enzyme that is synthesized de novo in response to ·NO. With this knowledgebase, it is expected that the availability of AAs in the extracellular environment would enhance ·NO detoxification, because AAs would foster translation of Hmp. However, we observed that ·NO detoxification by E. coli was far slower in populations grown and treated in the presence of AAs (AA+) in comparison to those grown and stressed in the absence of AAs (AA-). Further experiments revealed that AA+ populations had difficulty translating proteins under ·NO stress, and that ·NO activated the stringent response in AA+ populations. Additional work revealed significant ATP depletion in ·NO-stressed AA+ cultures that far exceeded that of ·NO-stressed AA- populations. Transcription, translation, and RelA were not found to be significant contributors to the ATP depletion observed, whereas AA import was implicated as a significant ATP consumption pathway. Alleviating ATP depletion while maintaining access to AAs partially restored ·NO detoxification, which suggested that ATP depletion contributed to the translational difficulties observed in ·NO-stressed AA+ populations. These data reveal an unexpected interaction within the ·NO response network of E. coli that stimulates a stringent response by RelA in conditions where AAs are plentiful.


Subject(s)
Escherichia coli Proteins , Hemeproteins , Escherichia coli/genetics , Escherichia coli/metabolism , Nitric Oxide/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Amino Acids/metabolism , NADH, NADPH Oxidoreductases/metabolism , Hemeproteins/metabolism , Dihydropteridine Reductase/metabolism , Adenosine Triphosphate/metabolism
4.
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
5.
J Biol Chem ; 296: 100186, 2021.
Article in English | MEDLINE | ID: mdl-33310705

ABSTRACT

The substrates O2 and NO cooperatively activate the NO dioxygenase function of Escherichia coli flavohemoglobin. Steady-state and transient kinetic measurements support a structure-based mechanistic model in which O2 and NO movements and conserved amino acids at the E11, G8, E2, E7, B10, and F7 positions within the globin domain control activation. In the cooperative and allosteric mechanism, O2 migrates to the catalytic heme site via a long hydrophobic tunnel and displaces LeuE11 away from the ferric iron, which forces open a short tunnel to the catalytic site gated by the ValG8/IleE15 pair and LeuE11. NO permeates this tunnel and leverages upon the gating side chains triggering the CD loop to furl, which moves the E and F-helices and switches an electron transfer gate formed by LysF7, GlnE7, and water. This allows FADH2 to reduce the ferric iron, which forms the stable ferric-superoxide-TyrB10/GlnE7 complex. This complex reacts with internalized NO with a bimolecular rate constant of 1010 M-1 s-1 forming nitrate, which migrates to the CD loop and unfurls the spring-like structure. To restart the cycle, LeuE11 toggles back to the ferric iron. Actuating electron transfer with O2 and NO movements averts irreversible NO poisoning and reductive inactivation of the enzyme. Together, structure snapshots and kinetic constants provide glimpses of intermediate conformational states, time scales for motion, and associated energies.


Subject(s)
Dihydropteridine Reductase/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , NADH, NADPH Oxidoreductases/metabolism , Oxygenases/metabolism , Allosteric Regulation , Dihydropteridine Reductase/chemistry , Escherichia coli/chemistry , Escherichia coli Infections/microbiology , Escherichia coli Proteins/chemistry , Humans , Models, Molecular , NADH, NADPH Oxidoreductases/chemistry , Nitric Oxide/metabolism , Oxygenases/chemistry , Protein Conformation
6.
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
7.
Article in English | MEDLINE | ID: mdl-29263060

ABSTRACT

The biological signal molecule nitric oxide (NO) was found to induce biofilm dispersal across a range of bacterial species, which led to its consideration for therapeutic strategies to treat biofilms and biofilm-related infections. However, biofilms are often not completely dispersed after exposure to NO. To better understand this phenomenon, we investigated the response of Pseudomonas aeruginosa biofilm cells to successive NO treatments. When biofilms were first pretreated with a low, noneffective dose of NO, a second dose of the signal molecule at a concentration usually capable of inducing dispersal did not have any effect. Amperometric analysis revealed that pretreated P. aeruginosa cells had enhanced NO-scavenging activity, and this effect was associated with the production of the flavohemoglobin Fhp. Further, quantitative real-time reverse transcription-PCR (qRT-PCR) analysis showed that fhp expression increased by over 100-fold in NO-pretreated biofilms compared to untreated biofilms. Biofilms of mutant strains harboring mutations in fhp or fhpR, encoding a NO-responsive regulator of fhp, were not affected in their dispersal response after the initial pretreatment with NO. Overall, these results suggest that FhpR can sense NO to trigger production of the flavohemoglobin Fhp and inhibit subsequent dispersal responses to NO. Finally, the addition of imidazole, which can inhibit the NO dioxygenase activity of flavohemoglobin, attenuated the prevention of dispersal after NO pretreatment and improved the dispersal response in older, starved biofilms. This study clarifies the underlying mechanisms of impaired dispersal induced by repeated NO treatments and offers a new perspective for improving the use of NO in biofilm control strategies.


Subject(s)
Biofilms/drug effects , Imidazoles/pharmacology , Nitric Oxide/metabolism , Pseudomonas aeruginosa/metabolism , Bacterial Proteins/metabolism , Dihydropteridine Reductase/metabolism , Gene Expression Regulation, Bacterial/drug effects , Pseudomonas aeruginosa/drug effects
8.
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
9.
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
10.
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
11.
Biochem Biophys Res Commun ; 476(1): 29-34, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27207837

ABSTRACT

Nitric oxide (NO) detoxification enzymes, such as NO dioxygenase (NOD) and NO reductase (NOR), are important to the virulence of numerous bacteria. Pathogens use these defense systems to ward off immune-generated NO, and they do so in environments that contain additional stressors, such as reactive oxygen species, nutrient deprivation, and acid stress. NOD and NOR both use reducing equivalents to metabolically deactivate NO, which suggests that nutrient deprivation could negatively impact their functionality. To explore the relationship between NO detoxification and nutrient deprivation, we examined the ability of Escherichia coli to detoxify NO under different levels of carbon source availability in aerobic cultures. We observed failure of NO detoxification under both carbon source limitation and starvation, and those failures could have arisen from inabilities to synthesize Hmp (NOD of E. coli) and/or supply it with sufficient NADH (preferred electron donor). We found that when limited quantities of carbon source were provided, NO detoxification failed due to insufficient NADH, whereas starvation prevented Hmp synthesis, which enabled cells to maintain their NADH levels. This maintenance of NADH levels under starvation was confirmed to be dependent on the absence of Hmp. Intriguingly, these data show that under NO stress, carbon-starved E. coli are better positioned with regard to reducing power to cope with other stresses than cells that had consumed an exhaustible amount of carbon.


Subject(s)
Carbon/metabolism , Dihydropteridine Reductase/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Hemeproteins/metabolism , NADH, NADPH Oxidoreductases/metabolism , Nitric Oxide/metabolism , Oxygenases/metabolism , Escherichia coli/cytology , NADP/metabolism , Oxidation-Reduction , Stress, Physiological
12.
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
13.
J Biol Chem ; 289(43): 29471-82, 2014 Oct 24.
Article in English | MEDLINE | ID: mdl-25193663

ABSTRACT

CO and NO are small toxic gaseous molecules that play pivotal roles in biology as gasotransmitters. During bacterial infection, NO, produced by the host via the inducible NO synthase, exerts critical antibacterial effects while CO, generated by heme oxygenases, enhances phagocytosis of macrophages. In Escherichia coli, other bacteria and fungi, the flavohemoglobin Hmp is the most important detoxification mechanism converting NO and O2 to the ion nitrate (NO3(-)). The protoheme of Hmp binds not only O2 and NO, but also CO so that this ligand is expected to be an inhibitor of NO detoxification in vivo and in vitro. CORM-3 (Ru(CO)(3)Cl(glycinate)) is a metal carbonyl compound extensively used and recently shown to have potent antibacterial properties. In this study, attenuation of the NO resistance of E. coli by CORM-3 is demonstrated in vivo. However, polarographic measurements showed that CO gas, but not CORM-3, produced inhibition of the NO detoxification activity of Hmp in vitro. Nevertheless, CO release from CORM-3 in the presence of soluble cellular compounds is demonstrated by formation of carboxy-Hmp. We show that the inability of CORM-3 to inhibit the activity of purified Hmp is due to slow release of CO in protein solutions alone i.e. when sodium dithionite, widely used in previous studies of CO release from CORM-3, is excluded. Finally, we measure intracellular CO released from CORM-3 by following the formation of carboxy-Hmp in respiring cells. CORM-3 is a tool to explore the concerted effects of CO and NO in vivo.


Subject(s)
Carbon Monoxide/metabolism , Dihydropteridine Reductase/metabolism , Escherichia coli Proteins/metabolism , Hemeproteins/metabolism , NADH, NADPH Oxidoreductases/metabolism , Nitric Oxide/metabolism , Organometallic Compounds/metabolism , Anaerobiosis/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Intracellular Space/metabolism , Iron/metabolism , Solubility , Sulfates/pharmacology , Suspensions
14.
BMC Microbiol ; 14: 65, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24629000

ABSTRACT

BACKGROUND: Nitric oxide (NO) is produced as part of the host immune response to bacterial infections, including urinary tract infections. The enzyme flavohemoglobin, coded by the hmp gene, is involved in protecting bacterial cells from the toxic effects of NO and represents a potentially interesting target for development of novel treatment concepts against resistant uropathogenic bacteria. The aim of the present study was to investigate if the in vitro antibacterial effects of NO can be enhanced by pharmacological modulation of the enzyme flavohemoglobin. RESULTS: Four clinical isolates of multidrug-resistant extended-spectrum ß-lactamase (ESBL)-producing uropathogenic E. coli were included in the study. It was shown that the NO-donor substance DETA/NO, but not inactivated DETA/NO, caused an initial growth inhibition with regrowth noted after 8 h of exposure. An hmp-deficient strain showed a prolonged growth inhibition in response to DETA/NO compared to the wild type. The imidazole antibiotic miconazole, that has been shown to inhibit bacterial flavohemoglobin activity, prolonged the DETA/NO-evoked growth inhibition. When miconazole was combined with polymyxin B nonapeptide (PMBN), in order to increase the bacterial wall permeability, DETA/NO caused a prolonged bacteriostatic response that lasted for up to 24 h. CONCLUSION: An NO-donor in combination with miconazole and PMBN showed enhanced antimicrobial effects and proved effective against multidrug-resistant ESBL-producing uropathogenic E. coli.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dihydropteridine Reductase/metabolism , Escherichia coli Proteins/metabolism , Hemeproteins/metabolism , Miconazole/pharmacology , NADH, NADPH Oxidoreductases/metabolism , Nitric Oxide/pharmacology , Polymyxin B/analogs & derivatives , Uropathogenic Escherichia coli/drug effects , Drug Synergism , Humans , Microbial Sensitivity Tests , Nanoparticles/metabolism , Polymyxin B/pharmacology , Uropathogenic Escherichia coli/enzymology , Uropathogenic Escherichia coli/growth & development , beta-Lactamases/metabolism
15.
Vestn Ross Akad Med Nauk ; (7-8): 69-77, 2014.
Article in Russian | MEDLINE | ID: mdl-25563006

ABSTRACT

BACKGROUND: Phenylketonuria (PKU) is an autosomal recessive inherited disease associated with impaired metabolism of the amino acids phenylalanine (Phe) and tyrosine. The main criterion for diagnosis of PKU is high blood Phe level determined during neonatal screening. In case where PKU patient is responsive to tetrahydrobiopterin treatment, sapropterin restores the impaired activity of the enzyme phenylalanine hydroxylase, resulting in the stimulation of normal Phe metabolism and thereby enhancing patient tolerance to natural products. AIM: The present open, non-comparative clinical study was initiated to assess the degree and frequency of response after 8-day sapropterin administration and assess the safety of 6-week sapropterin treatment in patients with PKU and hyperphenylalaninemia. PATIENTS AND METHODS: The study enrolled 90 patients with PKU. The criterion of response to 8-day sapropterin therapy was the reduction of Phe blood levels ≥ 30% compared with the baseline value. RESULTS: Positive response to treatment was observed in 30 (33.3%) patients (95% CI 23.7-44.1). The mean percentage change in Phe blood levels after the 8-day response test period compared to Phe levels prior to dosing was 14.1 ± 28.4% in the overall subject population (95% CI 8.2-20.1) and 44.3 ± 15.1% in the subpopulation of patients with a positive response (95% CI 38.6-49.9). During the study, adverse events were reported in 24 (26.7%) patients in the overall population in 16 (53.3%) patients in the subpopulation who had a response. CONCLUSION: The study results confirmed the efficacy and safety of sapropterin therapy in patients with PKU, which is consistent with international clinical trials data.


Subject(s)
Biopterins/analogs & derivatives , Phenylalanine/blood , Phenylketonurias , Adolescent , Biopterins/administration & dosage , Biopterins/adverse effects , Child , Child, Preschool , Coenzymes/administration & dosage , Coenzymes/adverse effects , Dihydropteridine Reductase/metabolism , Drug Monitoring/methods , Female , Humans , Infant, Newborn , Male , Neonatal Screening/methods , Phenylalanine Hydroxylase/metabolism , Phenylketonurias/blood , Phenylketonurias/drug therapy , Phenylketonurias/physiopathology , Severity of Illness Index , Treatment Outcome
16.
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
17.
Nitric Oxide ; 34: 65-75, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23764490

ABSTRACT

BACKGROUND: During infection and pathogenesis, Campylobacter, the leading cause of gastroenteritis, encounters NO and reactive nitrogen species (RNS) derived from the host. To combat these species, Campylobacter jejuni expresses two haemoglobins: the single domain haemoglobin (Cgb) detoxifies NO but the role of the truncated globin (Ctb) is unclear. Confirmation of Cgb activity and more extensive exploration of Ctb function(s) in vivo are restricted due to difficulties in expressing proteins in Campylobacter and our lack of understanding of how the globin haems are re-reduced after ligand reactions. METHODS: The cgb and ctb genes were cloned under the control of arabinose-inducible promoters and the globins expressed in an Escherichia coli mutant lacking the main NO detoxification mechanisms (Hmp and the Nor system comprising the transcription regulator NorR, the flavorubredoxin and its reductase (NorVW)); cellular responses under oxidative and nitrosative stress conditions were assessed. Spectroscopic changes of the Cgb and Ctb haems in soluble fractions after oxidation by NO were evaluated. Construction of E. coli nor mutants and a ubiquinone-defective strain allowed the exploration of the flavorubredoxin reductase and the aerobic respiratory chain as candidates for Cgb electron donors in E. coli mutants. RESULTS: Cgb, but not Ctb, complements the NO- and RNS-sensitive phenotype of an E. coli hmp mutant in aerobic conditions; however, Cgb fails to protect an hmp norR mutant in the absence of oxygen. Reduction of Cgb and Ctb in E. coli and C. jejuni soluble extracts and turnover after NO oxidation is demonstrated. Finally, we report a minor role for NorW as a Cgb reductase partner in E. coli but no role for respiratory electron flux in globin redox cycling. CONCLUSIONS: The NO detoxification capacity of Cgb is confirmed by heterologous expression in E. coli. The reducibility of Cgb and Ctb in E. coli and C. jejuni extracts and the lack of dependence of reduction upon flavorubredoxin reductase and the respiratory chain in E. coli argue in favor of a non-specific reductase system. GENERAL SIGNIFICANCE: We present the most persuasive evidence to date that Cgb, but not Ctb, confers tolerance to NO and RNS by reaction with NO. Since certain hypotheses for the mechanism of haem re-reduction in E. coli following the reaction with NO are not proven, the mechanisms of reduction in C. jejuni now require challenging experimental evaluation.


Subject(s)
Bacterial Proteins/metabolism , Campylobacter jejuni/metabolism , Nitric Oxide/metabolism , Nitrosation/physiology , Stress, Physiological/physiology , Truncated Hemoglobins/metabolism , Anaerobiosis , Bacterial Proteins/genetics , Dihydropteridine Reductase/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Hemeproteins/metabolism , NAD/metabolism , NADH, NADPH Oxidoreductases/metabolism , Oxidation-Reduction , S-Nitrosoglutathione/metabolism , Trans-Activators/metabolism , Truncated Hemoglobins/genetics
18.
Biochem Cell Biol ; 91(3): 187-93, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23668792

ABSTRACT

Quinoid dihydropteridine reductase (QDPR) is an enzyme involved in the metabolic pathway of tetrahydrobiopterin (BH4). BH4 is an essential cofactor of nitric oxide synthase (NOS) and can catalyze arginine to citrulline to release nitric oxide. Point mutations of QDPR have been found in the renal cortex of spontaneous Otsuka Long Evans Tokushima Fatty (OLETF) diabetic rats. However, the role of QDPR in DN is not clear. This study investigates the effects of QDPR overexpression and knockdown on gene expression in the kidney. Rat QDPR cDNA was cloned into pcDNA3.1 vector and transfected in human kidney cells (293T). The expression of NOS, transforming growth factor beta 1 (TGF-ß1), Smad3, and NADPH oxidase were examined by RT-PCR and Western blot analyses. BH4 was assayed by using ELISA. Expression of QDPR was significantly decreased and TGF-ß1 and Smad3 were increased in the renal cortex of diabetic rats. Transfection of QDPR into 293T cells increased the abundance of QDPR in cytoplasm and significantly reduced the expression of TGF-ß1, Smad3, and the NADPH oxidases NOX1 and NOX4. Moreover, abundance of neuronal NOS (nNOS) mRNA and BH4 content were significantly increased. Furthermore, inhibition of QDPR resulted in a significant increase in TGF-ß1 expression. In conclusion, QDPR might be an important factor mediating diabetic nephropathy through its regulation of TGF-ß1/Smad3 signaling and NADPH oxidase.


Subject(s)
Dihydropteridine Reductase/metabolism , Gene Expression , Kidney/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Base Sequence , Cell Line , DNA Primers , Humans , Kidney/cytology , Rats , Rats, Wistar
19.
Mol Genet Metab ; 105(4): 582-4, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22325981

ABSTRACT

The traditional treatment of severe disorders of tetrahydrobiopterin (BH4) metabolism is based on the replacement therapy with BH4, 5-hydroxytryptophan, and L-dopa. Major problems are encountered with L-dopa therapy, especially with increasing age when higher doses are necessary, because of its short half-life and adverse effects. Consequently, different L-dopa-sparing strategies have been successively introduced, with partial reduction of L-dopa dosage and amelioration of the clinical outcome. Recently, we demonstrated that the dopamine agonist pramipexole improves the therapeutic effect of L-dopa in 6-pyruvoyl tetrahydropterin synthase (PTPS) deficiency, the most common disorder of BH4 metabolism. Here we report its effectiveness in two patients (males, 7 and 22 years) with dihydropteridine reductase (DHPR) deficiency, the second most frequent cause of BH4 deficiency. Both patients experienced residual symptoms of dopamine deficiency, movement and behavioral disability, and complications of L-dopa therapy, associated with fluctuating hyperprolactinemia. They had full clinical and biochemical assessment, by an adapted Unified Parkinson's Disease Rating Scale (UPDRS) and measurement of diurnal plasma prolactin (PRL) profile before and after a trial with pramipexole. Besides allowing the reduction of L-dopa daily dosage (-58%) and administrations (from three to two) in one patient and to stop L-dopa therapy in the other, the introduction of pramipexole markedly improved and stabilized clinical and biochemical picture in both patients, as revealed by reduction of UPDRS scores and normalization of diurnal plasma prolactin profiles. Dopamine agonists can improve or even replace L-dopa therapy in disorders of synthesis and regeneration of BH4.


Subject(s)
Benzothiazoles/therapeutic use , Dopamine Agonists/therapeutic use , Levodopa/therapeutic use , Phenylketonurias/drug therapy , Adult , Behavior/drug effects , Biopterins/analogs & derivatives , Biopterins/metabolism , Child , Dihydropteridine Reductase/metabolism , Dopamine/metabolism , Humans , Locomotion/drug effects , Male , Phosphorus-Oxygen Lyases/deficiency , Phosphorus-Oxygen Lyases/drug effects , Pramipexole , Prolactin/blood , Young Adult
20.
FEBS Lett ; 585(19): 3047-51, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21871890

ABSTRACT

Dictyostelium discoideum Ax2 produces both L-erythro-tetrahydrobiopterin (BH4) and its stereoisomer D-threo-BH4 (DH4). The putative cofactor function of them for phenylalanine hydroxylase (PAH) was investigated through genetic manipulation and quantitative determination of pteridines. In addition to establishing that dihydropteridine reductase (DHPR) and dihydrofolate reductase (DHFR) constitute the regeneration pathway of both BH4 and DH4, the results suggested that BH4 is a preferential cofactor for PAH in vivo, not a secondary product of DH4, which functions mainly as an antioxidant. Our result also demonstrated that PAH may be essential for Dictyostelium growth in nature, and thus it appears that the organism has evolved a strategy to maintain BH4 level via regeneration pathway at the expense of DH4 under oxidative stress conditions.


Subject(s)
Biopterins/analogs & derivatives , Dictyostelium/metabolism , Biopterins/chemistry , Biopterins/metabolism , Dictyostelium/chemistry , Dictyostelium/genetics , Dihydropteridine Reductase/genetics , Dihydropteridine Reductase/metabolism , Gene Knockdown Techniques , Oxidative Stress , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Stereoisomerism , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...