Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 374
Filtrar
1.
Biochem Biophys Res Commun ; 717: 150059, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-38723517

RESUMO

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.


Assuntos
Biopterinas , Di-Hidropteridina Redutase , Sinergismo Farmacológico , Metotrexato , Metotrexato/farmacologia , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Humanos , Di-Hidropteridina Redutase/metabolismo , Inibidores Enzimáticos/farmacologia , Oxirredução/efeitos dos fármacos , Animais , Simulação de Dinâmica Molecular
2.
Mol Cell Proteomics ; 20: 100036, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33545363

RESUMO

To identify novel autoantibodies of Takayasu arteritis (TAK) using HuProt array-based approach, a two-phase approach was adopted. In Phase I, serum samples collected from 40 TAK patients, 15 autoimmune disease patients, and 20 healthy subjects were screened to identify TAK-specific autoantibodies using human protein (HuProt) arrays. In phase II, the identified candidate autoantibodies were validated with TAK-focused arrays using an additional cohort comprised of 109 TAK patients, 110 autoimmune disease patients, and 96 healthy subjects. Subsequently, the TAK-specific autoantibodies validated in phase II were further confirmed using western blot analysis. We identified and validated eight autoantibodies as potential TAK-specific diagnostic biomarkers, including anti-SPATA7, -QDPR, -SLC25A2, -PRH2, -DIXDC1, -IL17RB, -ZFAND4, and -NOLC1 antibodies, with AUC of 0.803, 0.801, 0.780, 0.696, 0.695, 0.678, 0.635, and 0.613, respectively. SPATA7 could distinguish TAK from healthy and disease controls with 73.4% sensitivity at 85.4% specificity, while QDPR showed 71.6% sensitivity at 86.4% specificity. SLC25A22 showed the highest sensitivity of 80.7%, but at lower specificity of 67.0%. In addition, PRH2, IL17RB, and NOLC1 showed good specificities of 88.3%, 85.9%, and 86.9%, respectively, but at lower sensitivities (<50%). Finally, DIXDC1 and ZFAND4 showed moderate performance as compared with the other autoantibodies. Using a decision tree model, we could reach a specificity of 94.2% with AUC of 0.843, a significantly improved performance as compared with that by each individual biomarker. The performances of three autoantibodies, namely anti-SPATA7, -QDPR, and -PRH2, were successfully confirmed with western blot analysis. Using this two-phase strategy, we identified and validated eight novel autoantibodies as TAK-specific biomarker candidates, three of which could be readily adopted in a clinical setting.


Assuntos
Autoanticorpos/sangue , Arterite de Takayasu/sangue , Adulto , Autoantígenos/imunologia , Biomarcadores/sangue , Proteínas de Ligação a DNA/imunologia , Árvores de Decisões , Di-Hidropteridina Redutase/imunologia , Feminino , Humanos , Masculino , Análise Serial de Proteínas , Proteínas Salivares Ricas em Prolina/imunologia , Arterite de Takayasu/imunologia , Adulto Jovem
3.
J Biol Chem ; 296: 100186, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33310705

RESUMO

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.


Assuntos
Di-Hidropteridina Redutase/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , NADH NADPH Oxirredutases/metabolismo , Oxigenases/metabolismo , Regulação Alostérica , Di-Hidropteridina Redutase/química , Escherichia coli/química , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/química , Humanos , Modelos Moleculares , NADH NADPH Oxirredutases/química , Óxido Nítrico/metabolismo , Oxigenases/química , Conformação Proteica
4.
J Neurochem ; 161(2): 129-145, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35233765

RESUMO

Increasing evidence suggests the involvement of peripheral amino acid metabolism in the pathophysiology of neuropsychiatric disorders, whereas the molecular mechanisms are largely unknown. Tetrahydrobiopterin (BH4) is a cofactor for enzymes that catalyze phenylalanine metabolism, monoamine synthesis, nitric oxide production, and lipid metabolism. BH4 is synthesized from guanosine triphosphate and regenerated by quinonoid dihydropteridine reductase (QDPR), which catalyzes the reduction of quinonoid dihydrobiopterin. We analyzed Qdpr-/- mice to elucidate the physiological significance of the regeneration of BH4. We found that the Qdpr-/- mice exhibited mild hyperphenylalaninemia and monoamine deficiency in the brain, despite the presence of substantial amounts of BH4 in the liver and brain. Hyperphenylalaninemia was ameliorated by exogenously administered BH4, and dietary phenylalanine restriction was effective for restoring the decreased monoamine contents in the brain of the Qdpr-/- mice, suggesting that monoamine deficiency was caused by the secondary effect of hyperphenylalaninemia. Immunohistochemical analysis showed that QDPR was primarily distributed in oligodendrocytes but hardly detectable in monoaminergic neurons in the brain. Finally, we performed a behavioral assessment using a test battery. The Qdpr-/- mice exhibited enhanced fear responses after electrical foot shock. Taken together, our data suggest that the perturbation of BH4 metabolism should affect brain monoamine levels through alterations in peripheral amino acid metabolism, and might contribute to the development of anxiety-related psychiatric disorders. Cover Image for this issue: https://doi.org/10.1111/jnc.15398.


Assuntos
Biopterinas , Fenilcetonúrias , Animais , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Di-Hidropteridina Redutase , Medo , Humanos , Camundongos , Fenilalanina , Fenilcetonúrias/genética , Fenilcetonúrias/metabolismo
5.
J Pharmacol Sci ; 150(3): 173-179, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36184122

RESUMO

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.


Assuntos
Di-Hidropteridina Redutase , Agregação Plaquetária , 5-Hidroxitriptofano/farmacologia , Difosfato de Adenosina/farmacologia , Animais , Biopterinas/análogos & derivados , Catecolaminas , Colágeno , Di-Hidropteridina Redutase/genética , Di-Hidropteridina Redutase/farmacologia , Camundongos , Receptor 5-HT2A de Serotonina , Serotonina/farmacologia
6.
Mol Genet Metab ; 133(2): 123-136, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33903016

RESUMO

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.


Assuntos
Oxirredutases do Álcool/genética , GTP Cicloidrolase/genética , Fenilcetonúrias/genética , Fósforo-Oxigênio Liases/genética , Biopterinas/análogos & derivados , Biopterinas/genética , Biopterinas/metabolismo , Di-Hidropteridina Redutase/genética , Distonia/genética , Distonia/metabolismo , Distonia/patologia , Predisposição Genética para Doença , Humanos , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/metabolismo , Erros Inatos do Metabolismo/patologia , Proteínas Associadas aos Microtúbulos/genética , Fenilcetonúrias/classificação , Fenilcetonúrias/metabolismo , Fenilcetonúrias/patologia , Transtornos Psicomotores/genética , Transtornos Psicomotores/metabolismo , Transtornos Psicomotores/patologia
7.
Proc Natl Acad Sci U S A ; 113(12): E1757-66, 2016 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-26951670

RESUMO

The virulence of many pathogens depends upon their ability to cope with immune-generated nitric oxide (NO·). In Escherichia coli, the major NO· detoxification systems are Hmp, an NO· dioxygenase (NOD), and NorV, an NO· reductase (NOR). It is well established that Hmp is the dominant system under aerobic conditions, whereas NorV dominates anaerobic conditions; however, the quantitative contributions of these systems under the physiologically relevant microaerobic regime remain ill defined. Here, we investigated NO· detoxification in environments ranging from 0 to 50 µM O2, and discovered a regime in which E. coli NO· defenses were severely compromised, as well as conditions that exhibited oscillations in the concentration of NO·. Using an integrated computational and experimental approach, E. coli NO· detoxification was found to be extremely impaired at low O2 due to a combination of its inhibitory effects on NorV, Hmp, and translational activities, whereas oscillations were found to result from a kinetic competition for O2 between Hmp and respiratory cytochromes. Because at least 777 different bacterial species contain the genetic requirements of this stress response oscillator, we hypothesize that such oscillatory behavior could be a widespread phenomenon. In support of this hypothesis,Pseudomonas aeruginosa, whose respiratory and NO· response networks differ considerably from those of E. coli, was found to exhibit analogous oscillations in low O2 environments. This work provides insight into how bacterial NO· defenses function under the low O2 conditions that are likely to be encountered within host environments.


Assuntos
Escherichia coli/metabolismo , Óxido Nítrico/metabolismo , Aerobiose , Simulação por Computador , Di-Hidropteridina Redutase/fisiologia , Proteínas de Escherichia coli/fisiologia , Hemeproteínas/fisiologia , Interações Hospedeiro-Patógeno , Modelos Biológicos , NADH NADPH Oxirredutases/fisiologia , Oxirredutases/fisiologia , Oxigênio/farmacologia , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/metabolismo , Especificidade da Espécie
8.
Artigo em Inglês | MEDLINE | ID: mdl-29263060

RESUMO

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.


Assuntos
Biofilmes/efeitos dos fármacos , Imidazóis/farmacologia , Óxido Nítrico/metabolismo , Pseudomonas aeruginosa/metabolismo , Proteínas de Bactérias/metabolismo , Di-Hidropteridina Redutase/metabolismo , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Pseudomonas aeruginosa/efeitos dos fármacos
9.
Mol Biol Evol ; 33(8): 1979-87, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27189567

RESUMO

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.


Assuntos
Bactérias/genética , Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Eucariotos/genética , Eucariotos/metabolismo , Hemeproteínas/genética , Hemeproteínas/metabolismo , Óxido Nítrico/metabolismo , Adaptação Biológica/genética , Sequência de Aminoácidos , Evolução Biológica , Biologia Computacional , Bases de Dados de Ácidos Nucleicos , Di-Hidropteridina Redutase/genética , Di-Hidropteridina Redutase/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Evolução Molecular , Fungos/genética , Duplicação Gênica , Transferência Genética Horizontal , NADH NADPH Oxirredutases/genética , NADH NADPH Oxirredutases/metabolismo , Filogenia
10.
Mol Genet Metab ; 121(2): 83-90, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28412083

RESUMO

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.


Assuntos
Proteínas do Líquido Cefalorraquidiano/genética , Proteínas do Líquido Cefalorraquidiano/metabolismo , Líquido Cefalorraquidiano/química , Líquido Cefalorraquidiano/metabolismo , Metaboloma , Metabolômica/métodos , Adolescente , Biomarcadores/sangue , Biomarcadores/urina , Proteínas do Líquido Cefalorraquidiano/análise , Proteínas do Líquido Cefalorraquidiano/química , Criança , Pré-Escolar , Di-Hidropteridina Redutase/sangue , Di-Hidropteridina Redutase/genética , Di-Hidropteridina Redutase/metabolismo , Di-Hidropteridina Redutase/urina , Feminino , Humanos , Lactente , Masculino , Espectrometria de Massas/métodos , Erros Inatos do Metabolismo/diagnóstico , Fenótipo , Estudos Retrospectivos , Adulto Jovem
11.
Acta Biochim Biophys Sin (Shanghai) ; 49(8): 706-712, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28633336

RESUMO

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.


Assuntos
Autofagia/genética , Di-Hidropteridina Redutase/genética , Mutação , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética , Di-Hidropteridina Redutase/metabolismo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Estresse Oxidativo , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Serina-Treonina Quinases TOR/metabolismo
12.
Biochem Biophys Res Commun ; 476(1): 29-34, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27207837

RESUMO

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.


Assuntos
Carbono/metabolismo , Di-Hidropteridina Redutase/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Hemeproteínas/metabolismo , NADH NADPH Oxirredutases/metabolismo , Óxido Nítrico/metabolismo , Oxigenases/metabolismo , Escherichia coli/citologia , NADP/metabolismo , Oxirredução , Estresse Fisiológico
13.
BMC Med Genet ; 17(1): 65, 2016 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-27613114

RESUMO

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.


Assuntos
Oxirredutases do Álcool/genética , Di-Hidropteridina Redutase/genética , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Doença de Parkinson/genética , População Branca/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Estudos de Casos e Controles , Feminino , Frequência do Gene , Genótipo , Humanos , Masculino , Malta , Pessoa de Meia-Idade , Razão de Chances , Doença de Parkinson/patologia , Polimorfismo de Nucleotídeo Único
14.
Zhonghua Fu Chan Ke Za Zhi ; 51(12): 890-894, 2016 Dec 25.
Artigo em Zh | MEDLINE | ID: mdl-28057123

RESUMO

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.


Assuntos
Di-Hidropteridina Redutase/genética , Mutação/genética , Fenilcetonúrias/genética , Fósforo-Oxigênio Liases/deficiência , Diagnóstico Pré-Natal , Alelos , Biopterinas/análogos & derivados , Amostra da Vilosidade Coriônica , Feminino , Feto , Testes Genéticos , Genótipo , Heterozigoto , Humanos , Luminescência , Óxido Nítrico Sintase , Fenilalanina/sangue , Fenilcetonúrias/diagnóstico , Fósforo-Oxigênio Liases/genética , Reação em Cadeia da Polimerase , Gravidez
15.
J Biol Chem ; 289(43): 29471-82, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25193663

RESUMO

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.


Assuntos
Monóxido de Carbono/metabolismo , Di-Hidropteridina Redutase/metabolismo , Proteínas de Escherichia coli/metabolismo , Hemeproteínas/metabolismo , NADH NADPH Oxirredutases/metabolismo , Óxido Nítrico/metabolismo , Compostos Organometálicos/metabolismo , Anaerobiose/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Espaço Intracelular/metabolismo , Ferro/metabolismo , Solubilidade , Sulfatos/farmacologia , Suspensões
16.
Metab Eng ; 31: 22-34, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26112956

RESUMO

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(∙).


Assuntos
Di-Hidropteridina Redutase/genética , Endopeptidase Clp/fisiologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/fisiologia , Escherichia coli/genética , Hemeproteínas/genética , NADH NADPH Oxirredutases/genética , Óxido Nítrico/metabolismo , RNA Mensageiro/análise , Biocatálise , Escherichia coli/metabolismo , Regiões Promotoras Genéticas , Análise de Sequência de RNA , Estresse Fisiológico , Transcrição Gênica
17.
Ir Med J ; 108(10): 312-4, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26817292

RESUMO

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.


Assuntos
Di-Hidropteridina Redutase/genética , Fenilcetonúrias/genética , Adulto , Feminino , Humanos , Lactente , Masculino
18.
J Bacteriol ; 196(1): 60-9, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24142248

RESUMO

Corynebacterium glutamicum ArnR is a novel transcriptional regulator that represses expression of the nitrate reductase operon narKGHJI and the nitric oxide (NO)-detoxifying flavohemoglobin gene hmp under aerobic conditions. In a previous study, we showed that ArnR-mediated repression is relieved during anaerobic nitrate respiration, but we could not pinpoint the specific signal that ArnR senses. In this study, we show that in the absence of nitrate, ArnR-mediated repression is maintained under anaerobic conditions. The derepression in response to nitrate is eliminated by disruption of narG, suggesting that ArnR senses nitrate derivatives generated during nitrate respiration. Specifically, the hmp gene is upregulated in the presence of nitrite or nitric oxide (NO) in an ArnR-dependent manner, although the response of narK appears to be greatly affected by ArnR-independent regulation. In vitro binding of ArnR to the narK and hmp promoter regions is more strongly inhibited by NO than by nitrite. We previously showed that the UV-visible spectrum of ArnR is typical of a Fe-S cluster-containing protein. Site-directed mutagenesis of each of three cysteine residues, which are possibly involved in coordination of the cofactor in the ArnR protein, results in loss of the binding of this protein to its target promoters in vitro and eliminates the repression of the target genes in vivo under aerobic conditions. These observations suggest that the cofactor coordinated by these three cysteine residues in the ArnR protein plays a critical role in the NO-responsive expression of the narKGHJI operon and the hmp gene.


Assuntos
Corynebacterium glutamicum/genética , Corynebacterium glutamicum/metabolismo , Di-Hidropteridina Redutase/biossíntese , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Nitrato Redutase/biossíntese , Óxido Nítrico/metabolismo , Proteínas Repressoras/metabolismo , Aerobiose , Anaerobiose , Análise Mutacional de DNA , Mutagênese Sítio-Dirigida , Nitritos/metabolismo , Óperon , Proteínas Repressoras/genética
19.
BMC Microbiol ; 14: 65, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24629000

RESUMO

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.


Assuntos
Antibacterianos/farmacologia , Di-Hidropteridina Redutase/metabolismo , Proteínas de Escherichia coli/metabolismo , Hemeproteínas/metabolismo , Miconazol/farmacologia , NADH NADPH Oxirredutases/metabolismo , Óxido Nítrico/farmacologia , Polimixina B/análogos & derivados , Escherichia coli Uropatogênica/efeitos dos fármacos , Sinergismo Farmacológico , Humanos , Testes de Sensibilidade Microbiana , Nanopartículas/metabolismo , Polimixina B/farmacologia , Escherichia coli Uropatogênica/enzimologia , Escherichia coli Uropatogênica/crescimento & desenvolvimento , beta-Lactamases/metabolismo
20.
Vestn Ross Akad Med Nauk ; (7-8): 69-77, 2014.
Artigo em Russo | MEDLINE | ID: mdl-25563006

RESUMO

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.


Assuntos
Biopterinas/análogos & derivados , Fenilalanina/sangue , Fenilcetonúrias , Adolescente , Biopterinas/administração & dosagem , Biopterinas/efeitos adversos , Criança , Pré-Escolar , Coenzimas/administração & dosagem , Coenzimas/efeitos adversos , Di-Hidropteridina Redutase/metabolismo , Monitoramento de Medicamentos/métodos , Feminino , Humanos , Recém-Nascido , Masculino , Triagem Neonatal/métodos , Fenilalanina Hidroxilase/metabolismo , Fenilcetonúrias/sangue , Fenilcetonúrias/tratamento farmacológico , Fenilcetonúrias/fisiopatologia , Índice de Gravidade de Doença , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA