Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 967
Filter
1.
Nitric Oxide ; 146: 19-23, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38521487

ABSTRACT

The mammalian brain is exquisitely vulnerable to lack of oxygen. However, the mechanism underlying the brain's sensitivity to hypoxia is incompletely understood. In this narrative review, we present a case for sulfide catabolism as a key defense mechanism of the brain against acute oxygen shortage. We will examine literature on the role of sulfide in hypoxia/ischemia, deep hibernation, and leigh syndrome patients, and present our recent data that support the neuroprotective effects of sulfide catabolism and persulfide production. When oxygen levels become low, hydrogen sulfide (H2S) accumulates in brain cells and impairs the ability of these cells to use the remaining, available oxygen to produce energy. In recent studies, we found that hibernating ground squirrels, which can withstand very low levels of oxygen, have high levels of sulfide:quinone oxidoreductase (SQOR) and the capacity to catabolize hydrogen sulfide in the brain. Silencing SQOR increased the sensitivity of the brain of squirrels and mice to hypoxia, whereas neuron-specific SQOR expression prevented hypoxia-induced sulfide accumulation, bioenergetic failure, and ischemic brain injury in mice. Excluding SQOR from mitochondria increased sensitivity to hypoxia not only in the brain but also in heart and liver. Pharmacological agents that scavenge sulfide and/or increase persulfide maintained mitochondrial respiration in hypoxic neurons and made mice resistant to ischemic injury to the brain or spinal cord. Drugs that oxidize hydrogen sulfide and/or increase persulfide may prove to be an effective approach to the treatment of patients experiencing brain injury caused by oxygen deprivation or mitochondrial dysfunction.


Subject(s)
Hibernation , Neuroprotection , Hibernation/physiology , Animals , Humans , Sulfides/metabolism , Sulfides/pharmacology , Hydrogen Sulfide/metabolism , Brain/metabolism , Mice , Sciuridae/metabolism , Leigh Disease/metabolism , Quinone Reductases/metabolism
2.
Biochem Biophys Res Commun ; 690: 149096, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37988924

ABSTRACT

Electron-driven process helps the living organism in the generations of energy, biomass production and detoxification of synthetic compounds. Soluble quinone oxidoreductases (QORs) mediate the transfer of an electron from NADPH to various quinone and other compounds, helping in the detoxification of quinones. QORs play a crucial role in cellular metabolism and are thus potential targets for drug development. Here we report the crystal structure of the NADPH-dependent QOR from Leishmania donovani (LdQOR) at 2.05 Å. The enzyme exists as a homo-dimer, with each protomer consisting of two domains, responsible for binding NADPH cofactor and the substrate. Interestingly, the human QOR exists as a tetramer. Comparative analysis of the oligomeric interfaces of LdQOR with HsQOR shows no significant differences in the protomer/dimer assembly. The tetrameric interface of HsQOR is stabilized by salt bridges formed between Arg 169 and Glu 271 which is non-existent in LdQOR, with an Alanine replacing the glutamate. This distinct feature is conserved across other dimeric QORs, indicating the importance of this interaction for tetramer association. Among the homologs, the sequences of the loop region involved in the stabilization and binding of the adenine ring of the NADPH shows significant differences except for an Arginine & glycine residues. In dimer QORs, this Arginine acts as a gate to the co-factor, while the NADPH binding mode in the human homolog is distinct, stabilized by His 200 and Asn 229, which are not conserved in LdQOR. These distinct features have the potential to be utilized for therapeutic interventions.


Subject(s)
NAD(P)H Dehydrogenase (Quinone) , Quinone Reductases , Humans , NADP/metabolism , Protein Subunits , NAD(P)H Dehydrogenase (Quinone)/metabolism , Quinone Reductases/chemistry , Quinone Reductases/metabolism , Quinones , Arginine , Binding Sites , Crystallography, X-Ray
3.
Sci Rep ; 13(1): 21624, 2023 12 07.
Article in English | MEDLINE | ID: mdl-38062122

ABSTRACT

Dopaminergic degeneration is a central feature of Parkinson's disease (PD), but glial dysfunction may accelerate or trigger neuronal death. In fact, astrocytes play a key role in the maintenance of the blood-brain barrier and detoxification. 6-hydroxydopamine (6OHDA) is used to induce PD in rodent models due to its specific toxicity to dopaminergic neurons, but its effect on astrocytes has been poorly investigated. Here, we show that 6OHDA dose-dependently impairs autophagy in human U373 cells and primary murine astrocytes in the absence of cell death. LC3II downregulation was observed 6 to 48 h after treatment. Interestingly, 6OHDA enhanced NRH:quinone oxidoreductase 2 (NQO2) expression and activity in U373 cells, even if 6OHDA turned out not to be its substrate. Autophagic flux was restored by inhibition of NQO2 with S29434, which correlated with a partial reduction in oxidative stress in response to 6OHDA in human and murine astrocytes. NQO2 inhibition also increased the neuroprotective capability of U373 cells, since S29434 protected dopaminergic SHSY5Y cells from 6OHDA-induced cell death when cocultured with astrocytes. The toxic effects of 6OHDA on autophagy were attenuated by silencing NQO2 in human cells and primary astrocytes from NQO2-/- mice. Finally, the analysis of Gene Expression Omnibus datasets showed elevated NQO2 gene expression in the blood cells of early-stage PD patients. These data support a toxifying function of NQO2 in dopaminergic degeneration via negative regulation of autophagy and neuroprotection in astrocytes, suggesting a potential pharmacological target in PD.


Subject(s)
Parkinson Disease , Quinone Reductases , Humans , Mice , Animals , Oxidopamine/pharmacology , Neuroprotection , Astrocytes/metabolism , Parkinson Disease/genetics , Quinone Reductases/metabolism , Autophagy , Dopaminergic Neurons/metabolism
4.
J Clin Invest ; 133(19)2023 10 02.
Article in English | MEDLINE | ID: mdl-37561584

ABSTRACT

Biological aging can be described as accumulative, prolonged metabolic stress and is the major risk factor for cognitive decline and Alzheimer's disease (AD). Recently, we identified and described a quinone reductase 2 (QR2) pathway in the brain, in which QR2 acts as a removable memory constraint and metabolic buffer within neurons. QR2 becomes overexpressed with age, and it is possibly a novel contributing factor to age-related metabolic stress and cognitive deficit. We found that, in human cells, genetic removal of QR2 produced a shift in the proteome opposing that found in AD brains while simultaneously reducing oxidative stress. We therefore created highly specific QR2 inhibitors (QR2is) to enable evaluation of chronic QR2 inhibition as a means to reduce biological age-related metabolic stress and cognitive decline. QR2is replicated results obtained by genetic removal of QR2, while local QR2i microinjection improved hippocampal and cortical-dependent learning in rats and mice. Continuous consumption of QR2is in drinking water improved cognition and reduced pathology in the brains of AD-model mice (5xFAD), with a noticeable between-sex effect on treatment duration. These results demonstrate the importance of QR2 activity and pathway function in the healthy and neurodegenerative brain and what we believe to be the great therapeutic potential of QR2is as first-in-class drugs.


Subject(s)
Alzheimer Disease , Quinone Reductases , Animals , Humans , Mice , Rats , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Hippocampus/metabolism , Oxidative Stress , Quinone Reductases/antagonists & inhibitors , Quinone Reductases/genetics , Quinone Reductases/metabolism , Stress, Physiological
5.
Sci Rep ; 13(1): 12226, 2023 07 28.
Article in English | MEDLINE | ID: mdl-37507428

ABSTRACT

Bacterial energy metabolism has become a promising target for next-generation tuberculosis chemotherapy. One strategy to hamper ATP production is to inhibit the respiratory oxidases. The respiratory chain of Mycobacterium tuberculosis comprises a cytochrome bcc:aa3 and a cytochrome bd ubiquinol oxidase that require a combined approach to block their activity. A quinazoline-type compound called ND-011992 has previously been reported to ineffectively inhibit bd oxidases, but to act bactericidal in combination with inhibitors of cytochrome bcc:aa3 oxidase. Due to the structural similarity of ND-011992 to quinazoline-type inhibitors of respiratory complex I, we suspected that this compound is also capable of blocking other respiratory chain complexes. Here, we synthesized ND-011992 and a bromine derivative to study their effect on the respiratory chain complexes of Escherichia coli. And indeed, ND-011992 was found to inhibit respiratory complex I and bo3 oxidase in addition to bd-I and bd-II oxidases. The IC50 values are all in the low micromolar range, with inhibition of complex I providing the lowest value with an IC50 of 0.12 µM. Thus, ND-011992 acts on both, quinone reductases and quinol oxidases and could be very well suited to regulate the activity of the entire respiratory chain.


Subject(s)
Escherichia coli Proteins , Quinone Reductases , Hydroquinones/pharmacology , Hydroquinones/metabolism , Electron Transport Complex I/metabolism , Quinone Reductases/metabolism , Oxidoreductases/metabolism , Electron Transport Complex IV/metabolism , Cytochromes/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Cytochrome b Group/metabolism
6.
Biochim Biophys Acta Bioenerg ; 1864(2): 148958, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36758662

ABSTRACT

Pyruvate:quinone oxidoreductases (PQOs) catalyse the oxidative decarboxylation of pyruvate to acetate and concomitant reduction of quinone to quinol with the release of CO2. They are thiamine pyrophosphate (TPP) and flavin-adenine dinucleotide (FAD) containing enzymes, which interact with the membrane in a monotopic way. PQOs are considered as part of alternatives to most recognized pyruvate catabolizing pathways, and little is known about their taxonomic distribution and structural/functional relationship. In this bioinformatics work we tackled these gaps in PQO knowledge. We used the KEGG database to identify PQO coding genes, performed a multiple sequence analysis which allowed us to study the amino acid conservation on these enzymes, and looked at their possible cellular function. We observed that PQOS are enzymes exclusively present in prokaryotes with most of the sequences identified in bacteria. Regarding the amino acid sequence conservation, we found that 75 amino acid residues (out of 570, on average) have a conservation over 90 %, and that the most conserved regions in the protein are observed around the TPP and FAD binding sites. We systematized the presence of conserved features involved in Mg2+, TPP and FAD binding, as well as residues directly linked to the catalytic mechanism. We also established the presence of a new motif named "HEH lock", possibly involved in the dimerization process. The results here obtained for the PQO protein family contribute to a better understanding of the biochemistry of these respiratory enzymes.


Subject(s)
Pyruvic Acid , Quinone Reductases , Amino Acid Sequence , Flavin-Adenine Dinucleotide/metabolism , Proteins , Quinone Reductases/metabolism , Amino Acids , NAD(P)H Dehydrogenase (Quinone)/metabolism , Quinones
7.
CNS Neurosci Ther ; 29(4): 1129-1141, 2023 04.
Article in English | MEDLINE | ID: mdl-36650666

ABSTRACT

INTRODUCTION: For investigating the mechanism of brain injury caused by chronic fluorosis, this study was designed to determine whether NRH:quinone oxidoreductase 2 (NQO2) can influence autophagic disruption and oxidative stress induced in the central nervous system exposed to a high level of fluoride. METHODS: Sprague-Dawley rats drank tap water containing different concentrations of fluoride for 3 or 6 months. SH-SY5Y cells were either transfected with NQO2 RNA interference or treated with NQO2 inhibitor or activator and at the same time exposed to fluoride. The enrichment of gene signaling pathways related to autophagy was evaluated by Gene Set Enrichment Analysis; expressions of NQO2 and autophagy-related protein 5 (ATG5), LC3-II and p62, and mammalian target of rapamycin (mTOR) were quantified by Western-blotting or fluorescent staining; and the levels of malondialdehyde (MDA) and superoxide dismutase (SOD) assayed biochemically and reactive oxygen species (ROS) detected by flow cytometry. RESULTS: In the hippocampal CA3 region of rats exposed to high fluoride, the morphological characteristics of neurons were altered; the numbers of autophagosomes in the cytoplasm and the levels of NQO2 increased; the level of p-mTOR was decreased, and the levels of ATG5, LC3-II and p62 were elevated; and genes related to autophagy enriched. In vitro, in addition to similar changes in NQO2, p-mTOR, ATG5, LC3 II, and p62, exposure of SH-SY5Y cells to fluoride enhanced MDA and ROS contents and reduced SOD activity. Inhibition of NQO2 with RNAi or an inhibitor attenuated the disturbance of the autophagic flux and enhanced oxidative stress in these cells exposed to high fluoride. CONCLUSION: Our findings indicate that NQO2 may be involved in regulating autophagy and oxidative stress and thereby exerts an impact on brain injury caused by chronic fluorosis.


Subject(s)
Brain Injuries , Neuroblastoma , Quinone Reductases , Rats , Humans , Animals , Fluorides/pharmacology , Reactive Oxygen Species/metabolism , Rats, Sprague-Dawley , Quinone Reductases/metabolism , Oxidative Stress , Autophagy , TOR Serine-Threonine Kinases/metabolism , Hippocampus/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Mammals/metabolism
8.
Biochim Biophys Acta Bioenerg ; 1864(2): 148948, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36481274

ABSTRACT

Staphylococcus aureus is an opportunistic pathogen and one of the most frequent causes for community acquired and nosocomial bacterial infections. Even so, its energy metabolism is still under explored and its respiratory enzymes have been vastly overlooked. In this work, we unveil the dihydroorotate:quinone oxidoreductase (DHOQO) from S. aureus, the first example of a DHOQO from a Gram-positive organism. This protein was shown to be a FMN containing menaquinone reducing enzyme, presenting a Michaelis-Menten behaviour towards the two substrates, which was inhibited by Brequinar, Leflunomide, Lapachol, HQNO, Atovaquone and TFFA with different degrees of effectiveness. Deletion of the DHOQO coding gene (Δdhoqo) led to lower bacterial growth rates, and effected in cell morphology and metabolism, most importantly in the pyrimidine biosynthesis, here systematized for S. aureus MW2 for the first time. This work unveils the existence of a functional DHOQO in the respiratory chain of the pathogenic bacterium S. aureus, enlarging the understanding of its energy metabolism.


Subject(s)
Quinones , Staphylococcus aureus , Atovaquone , Electron Transport , Quinones/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Quinone Reductases/metabolism
9.
Appl Microbiol Biotechnol ; 106(22): 7505-7517, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36219222

ABSTRACT

Monotopic membrane-bound flavoproteins, sulfide:quinone oxidoreductases (SQRs), have a variety of physiological functions, including sulfide detoxification. SQR enzymes are classified into six groups. SQRs use the flavin adenine dinucleotide (FAD) cofactor to transfer electrons from sulfide to quinone. A type VI SQR of the photosynthetic purple sulfur bacterium, Thiocapsa roseopersicina (TrSqrF), has been previously characterized, and the mechanism of sulfide oxidation has been proposed. This paper reports the characterization of quinone binding site (QBS) of TrSqrF composed of conserved aromatic and apolar amino acids. Val331, Ile333, and Phe366 were identified near the benzoquinone ring of enzyme-bound decylubiquinone (dUQ) using the TrSqrF homology model. In silico analysis revealed that Val331 and Ile333 alternately connected with the quinone head group via hydrogen bonds, and Phe366 and Trp369 bound the quinones via hydrophobic interactions. TrSqrF variants containing alanine (V331A, I333A, F366A) and aromatic amino acid (V331F, I333F, F366Y), as well as a C-terminal α-helix deletion (CTD) mutant were generated. These amino acids are critical for quinone binding and, thus, catalysis. Spectroscopic analyses proved that all mutants contained FAD. I333F replacement resulted in the lack of the charge transfer complex. In summary, the interactions described above maintain the quinone molecule's head in an optimal position for direct electron transfer from FAD. Surprisingly, the CTD mutant retained a relatively high level of specific activity while remaining membrane-anchored. This is a unique study because it focuses on the QBS and the oxidative stage of a type VI sulfide-dependent quinone reduction. KEY POINTS: • V331, I333, F366, and W369 were shown to interact with decylubiquinone in T. roseopersicina SqrF • These amino acids are involved in proper positioning of quinones next to FAD • I333 is essential in formation of a charge transfer complex from FAD to quinone.


Subject(s)
Flavin-Adenine Dinucleotide , Quinone Reductases , Quinone Reductases/genetics , Quinone Reductases/metabolism , Sulfides/metabolism , Benzoquinones , Binding Sites , Oxidation-Reduction , Amino Acids/metabolism
10.
Int J Mol Sci ; 23(18)2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36142607

ABSTRACT

Long non-coding RNAs (lncRNAs) play critical roles in human cancers. HOXA11 anti-sense RNA (HOXA11-AS) is an lncRNA belonging to the homeobox (HOX) gene cluster that promotes liver metastasis in human colon cancer. However, its role and mechanism of action in human oral squamous cell carcinoma (OSCC) are unclear. In this study, we investigated HOXA11-AS expression and function in human OSCC tissues and cell lines, as well as a mouse model of OSCC. Our analyses showed that HOXA11-AS expression in human OSCC cases correlates with lymph node metastasis, nicotinamide adenine dinucleotide (NAD)(P)H: quinone oxidoreductase 1 (NQO1) upregulation, and dihydronicotinamide riboside (NRH): quinone oxidoreductase 2 (NQO2) downregulation. Using the human OSCC cell lines HSC3 and HSC4, we demonstrate that HOXA11-AS promotes NQO1 expression by sponging microRNA-494. In contrast, HOXA11-AS recruits zeste homolog 2 (EZH2) to the NQO2 promoter to suppress its expression via the trimethylation of H3K27. The upregulation of NQO1 enzymatic activity by HOXA11-AS results in the consumption of flavin adenine dinucleotide (FAD), which reduces FAD-requiring glyceraldehyde-3-phosphate dehydrogenase (GAPDH) activity and suppresses glycolysis. However, our analyses show that lactic acid fermentation levels are preserved by glutaminolysis due to increased malic enzyme-1 expression, promoting enhanced proliferation, invasion, survival, and drug resistance. In contrast, suppression of NQO2 expression reduces the consumption of NRH via NQO2 enzymatic activity and increases NAD levels, which promotes enhanced stemness and metastatic potential. In mouse tumor models, knockdown of HOXA11-AS markedly suppressed tumor growth and lung metastasis. From these findings, targeting HOXA11-AS may strongly suppress high-grade OSCC by regulating both NQO1 and NQO2.


Subject(s)
Carcinoma, Squamous Cell , Homeodomain Proteins/metabolism , MicroRNAs , Mouth Neoplasms , NAD(P)H Dehydrogenase (Quinone)/metabolism , Quinone Reductases/metabolism , RNA, Long Noncoding , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , Flavin-Adenine Dinucleotide/genetics , Genes, Homeobox , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Humans , Lactic Acid , Mice , MicroRNAs/genetics , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , NAD/genetics , Quinones , RNA, Antisense , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics
11.
Methods Mol Biol ; 2550: 283-289, 2022.
Article in English | MEDLINE | ID: mdl-36180699

ABSTRACT

Melatonin, (N-acetyl-5-methoxytryptamine), is a neurohormone which possesses a wide range of biological effects. The effects mediated by melatonin are in part attributed to the antioxidant properties of the molecule. For a long time, melatonin had been described as a ligand of a putative "receptor" present in mammalian brains named MT3. Several studies were thus carried out with the goal of clarifying the nature of this melatonin "receptor." The experimental setup of the binding measurements is unusual. The present chapter aims at describing this technique. This binding site was confirmed independently by several groups, and it was eventually demonstrated that MT3 was the enzyme quinone reductase 2 (NQO2).


Subject(s)
Melatonin , Quinone Reductases , 5-Methoxytryptamine , Animals , Antioxidants , Binding Sites , Ligands , Mammals/metabolism , Melatonin/metabolism , Quinone Reductases/metabolism , Receptors, Melatonin/metabolism
12.
Methods Mol Biol ; 2550: 305-314, 2022.
Article in English | MEDLINE | ID: mdl-36180701

ABSTRACT

To ensure the physical interaction between a protein and its ligand, many techniques can be applied. One of them, isothermal titration calorimetry (ITC), measures the heat exchange between a forming molecular complex and its milieu. From this heat exchange, it is possible to acquire the thermodynamic parameters, the binding stoichiometry and the affinity constant (Ka) between the two interacting binding partners, which can then be used to determine the dissociation constant (Kd). We made use of ITC to determine the true Kd of melatonin for its putative receptor MT3, also known as the enzyme quinone reductase 2 (NQO2). In this chapter, we describe the step-by-step procedure for performing this experiment and extend it to 2-iodomelatonin, a melatonin derivative that was used in the initial identification and characterization of MT3. The dissociation constants of melatonin and 2-iodomelatonin toward NQO2 derived from these experiments are in line with data reported previously, albeit using alternative techniques.


Subject(s)
Melatonin , Quinone Reductases , Calorimetry/methods , Humans , Ligands , Melatonin/metabolism , Protein Binding , Quinone Reductases/metabolism , Thermodynamics
13.
Methods Mol Biol ; 2550: 291-304, 2022.
Article in English | MEDLINE | ID: mdl-36180700

ABSTRACT

Melatonin (N-acetyl-5-methoxytryptamine) is a neurohormone which possesses a wide range of biological effects. The effects mediated by melatonin are in part attributed to the antioxidant properties of the molecule, which may act as scavenger of free radicals, and also to the binding of melatonin to its protein targets. For a long time, melatonin had been described as a ligand of a putative "receptor" present in the mammalian brain. Several studies were thus carried out with the goal of clarifying the nature of this melatonin "receptor," which led to the discovery of MT3 as the third melatonin binding site. This binding site was confirmed independently by several groups, and it was eventually demonstrated that MT3 was the enzyme quinone reductase 2 (NQO2). Among the different approaches used to validate that MT3 was indeed NQO2, the co-crystallization of NQO2 with melatonin was key in demonstrating the exact binding site and mode of melatonin to the enzyme and led to a clear understanding of the residues important for protein binding and inhibition. In this chapter, we described the details for the cloning, expression, and purification of the human enzyme NQO2. We also describe a detailed protocol for the crystallization of melatonin with this protein.


Subject(s)
Melatonin , Quinone Reductases , 5-Methoxytryptamine , Animals , Antioxidants , Cloning, Molecular , Crystallization , Humans , Ligands , Mammals/metabolism , Melatonin/metabolism , Quinone Reductases/genetics , Quinone Reductases/metabolism , Receptors, Melatonin/metabolism , X-Rays
14.
Methods Mol Biol ; 2550: 315-321, 2022.
Article in English | MEDLINE | ID: mdl-36180702

ABSTRACT

The third melatonin binding site MT3 turned out to be an enzyme, NQO2 (E.C. 1.6.99.2). Its catalytic activity is inhibited by melatonin with an IC50 in the 50-100 µM range. Some of the functions of melatonin at pharmacological concentrations (1 µM and above) might be explained by this inhibition capacity of melatonin at NQO2. In order to determine precisely these parameters, it is required to comprehend the basic enzymology of this enzyme. In the following chapter, we present the basic conditions of measuring NQO2 catalytic activities and inhibition.


Subject(s)
Melatonin , Quinone Reductases , Binding Sites , Melatonin/metabolism , Melatonin/pharmacology , Quinone Reductases/chemistry , Quinone Reductases/metabolism
15.
Methods Mol Biol ; 2550: 323-328, 2022.
Article in English | MEDLINE | ID: mdl-36180703

ABSTRACT

Melatonin exerts its effects through a series of target proteins/receptors and enzymes. Its antioxidant capacity might be due to its capacity to inhibit a quinone reductase (NQO2) at high concentration (50 µM). Demonstrating the existence of a complex between a compound and a protein is often not easy. It requires either that the compound is an inhibitor-and the complex translates by an inhibition of the catalytic activity-or the compound is radiolabeled-and the complex translates in standard binding approaches, such as in receptology. Outside these two cases, the detection of the protein:small molecule complexes by mass spectrometry has recently been made possible, thanks to the development of so-called native mass spectrometry. Using this approach, one can measure masses corresponding to an intact noncovalent complex between a compound and its target, usually after titration or competition experiments. In the present chapter, we detail the characterization of NQO2:melatonin interaction using native mass spectrometry.


Subject(s)
Melatonin , Quinone Reductases , Antioxidants , Quinone Reductases/chemistry , Quinone Reductases/metabolism , Spectrometry, Mass, Electrospray Ionization
16.
Phytochemistry ; 203: 113396, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35998831

ABSTRACT

Prenylated and hydroxyprenylated piceatannol, resveratrol and pinosylvin derivatives were isolated from resin produced by three Australian Lepidosperma Labill. Species (Cyperaceae). From L. congestum R.Br. one known compound, 3',5'-bis-prenyl-E-resveratrol, and five undescribed compounds were isolated, 3'-O-prenyl-5'-prenyl-E-piceatannol, 5',6'-bis-prenyl-E-piceatannol, 5'-prenyl-E-piceatannol, 3',5'-bis(3-hydroxy-3-methylbutyl)-E-resveratrol and 3',5'-bis-E-hydroxyprenyl-E-resveratrol. From L. gunnii Boeckeler one undescribed compound was isolated, 3'-E-hydroxyprenyl-5'-Z-hydroxyprenyl-E-resveratrol. From L. laterale R.Br. six undescribed compounds were isolated, 3-O-prenyl-E-pinosylvin, 3-O-Z-hydroxyprenyl-E-pinosylvin, 3'-Z-hydroxyprenyl-E-resveratrol, 3-O-Z-hydroxyprenyl-E-resveratrol, 3-O-Z-hydroxyprenyl-4'-O-methyl-E-resveratrol, and 3-O-prenyl-3'-δ,δ'-dihydroxyprenyl-E-resveratrol. Compounds, including a reference compound 3-O-prenyl-3'-O-methyl-E-piceatannol, were screened in an assay for melatoninergic binding to MT1 and MT2 receptors and binding to QR2/MT3 enzyme, and for inhibition of QR2/MT3 in a functional assay. Strong binding was observed for 3-O-Z-hydroxyprenyl-E-resveratrol with a Ki of 0.022 nM and the strongest inhibition of QR2/MT3 observed was for the reference compound, 3-O-prenyl-3'-O-methyl-E-piceatannol, with an inhibition of 61% at 1 µM and 95% at 10 µM. The three most active binders and inhibitors of QR2/MT3 were found to have a common substructure corresponding to 3-O-prenylresveratrol.


Subject(s)
Cyperaceae , Quinone Reductases , Stilbenes , Australia , Neoprene , Quinone Reductases/metabolism , Resveratrol , Stilbenes/chemistry , Stilbenes/pharmacology
17.
Nat Commun ; 13(1): 4082, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35882843

ABSTRACT

The Na+-pumping NADH-ubiquinone oxidoreductase (Na+-NQR) couples electron transfer from NADH to ubiquinone with Na+-pumping, generating an electrochemical Na+ gradient that is essential for energy-consuming reactions in bacteria. Since Na+-NQR is exclusively found in prokaryotes, it is a promising target for highly selective antibiotics. However, the molecular mechanism of inhibition is not well-understood for lack of the atomic structural information about an inhibitor-bound state. Here we present cryo-electron microscopy structures of Na+-NQR from Vibrio cholerae with or without a bound inhibitor at 2.5- to 3.1-Å resolution. The structures reveal the arrangement of all six redox cofactors including a herein identified 2Fe-2S cluster located between the NqrD and NqrE subunits. A large part of the hydrophilic NqrF is barely visible in the density map, suggesting a high degree of flexibility. This flexibility may be responsible to reducing the long distance between the 2Fe-2S centers in NqrF and NqrD/E. Two different types of specific inhibitors bind to the N-terminal region of NqrB, which is disordered in the absence of inhibitors. The present study provides a foundation for understanding the function of Na+-NQR and the binding manner of specific inhibitors.


Subject(s)
Quinone Reductases , Vibrio cholerae , Bacterial Proteins/metabolism , Cryoelectron Microscopy , Electron Transport Complex I/metabolism , Oxidation-Reduction , Quinone Reductases/metabolism , Sodium/metabolism , Vibrio cholerae/metabolism
18.
Redox Biol ; 53: 102345, 2022 07.
Article in English | MEDLINE | ID: mdl-35653932

ABSTRACT

Heterotrophic bacteria and human mitochondria often use sulfide: quinone oxidoreductase (SQR) and persulfide dioxygenase (PDO) to oxidize sulfide to sulfite and thiosulfate. Bioinformatic analysis showed that the genes encoding RHOD domains were widely presented in annotated sqr-pdo operons and grouped into three types: fused with an SQR domain, fused with a PDO domain, and dissociated proteins. Biochemical evidence suggests that RHODs facilitate the formation of thiosulfate and promote the reaction between inorganic polysulfide and glutathione to produce glutathione polysulfide. However, the physiological roles of RHODs during sulfide oxidation by SQR and PDO could only be tested in an RHOD-free host. To test this, 8 genes encoding RHOD domains in Escherichia coli MG1655 were deleted to produce E. coli RHOD-8K. The sqrCp and pdoCp genes from Cupriavidus pinatubonensis JMP134 were cloned into E. coli RHOD-8K. SQRCp contains a fused RHOD domain at the N-terminus. When the fused RHOD domain of SQRCp was inactivated, the cells oxidized sulfide into increased thiosulfate with the accumulation of cellular sulfane sulfur in comparison with cells containing the intact sqrCp and pdoCp. The complementation of dissociated DUF442 minimized the accumulation of cellular sulfane sulfur and reduced the production of thiosulfate. Further analysis showed that the fused DUF442 domain modulated the activity of SQRCp and prevented it from directly passing the produced sulfane sulfur to GSH. Whereas, the dissociated DUF442 enhanced the PDOCp activity by several folds. Both DUF442 forms minimized the accumulation of cellular sulfane sulfur, which spontaneously reacted with GSH to produce GSSG, causing disulfide stress during sulfide oxidation. Thus, RHODs may play multiple roles during sulfide oxidation.


Subject(s)
Hydrogen Sulfide , Quinone Reductases , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Disulfides/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Glutathione/metabolism , Humans , Hydrogen Sulfide/metabolism , Oxidation-Reduction , Quinone Reductases/chemistry , Quinone Reductases/genetics , Quinone Reductases/metabolism , Sulfides/metabolism , Sulfur/metabolism , Thiosulfate Sulfurtransferase/genetics , Thiosulfate Sulfurtransferase/metabolism , Thiosulfates/metabolism
19.
Biochem J ; 479(1): 111-127, 2022 01 14.
Article in English | MEDLINE | ID: mdl-34981811

ABSTRACT

The cytochrome b6f complex (b6f) has been initially considered as the ferredoxin-plastoquinone reductase (FQR) during cyclic electron flow (CEF) with photosystem I that is inhibited by antimycin A (AA). The binding of AA to the b6f Qi-site is aggravated by heme-ci, which challenged the FQR function of b6f during CEF. Alternative models suggest that PROTON GRADIENT REGULATION5 (PGR5) is involved in a b6f-independent, AA-sensitive FQR. Here, we show in Chlamydomonas reinhardtii that the b6f is conditionally inhibited by AA in vivo and that the inhibition did not require PGR5. Instead, activation of the STT7 kinase upon anaerobic treatment induced the AA sensitivity of b6f which was absent from stt7-1. However, a lock in State 2 due to persisting phosphorylation in the phosphatase double mutant pph1;pbcp did not increase AA sensitivity of electron transfer. The latter required a redox poise, supporting the view that state transitions and CEF are not coercively coupled. This suggests that the b6f-interacting kinase is required for structure-function modulation of the Qi-site under CEF favoring conditions. We propose that PGR5 and STT7 independently sustain AA-sensitive FQR activity of the b6f. Accordingly, PGR5-mediated electron injection into an STT7-modulated Qi-site drives a Mitchellian Q cycle in CEF conditions.


Subject(s)
Antimycin A/pharmacology , Chlamydomonas reinhardtii/enzymology , Cytochrome b6f Complex/metabolism , Electrons , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Thylakoids/enzymology , Antimycin A/metabolism , Cytochrome b6f Complex/antagonists & inhibitors , Electron Transport/drug effects , Enzyme Activation , Ferredoxins/metabolism , Light-Harvesting Protein Complexes/metabolism , Oxidation-Reduction , Oxidoreductases Acting on Sulfur Group Donors/metabolism , Phosphorylation/drug effects , Photosynthesis/physiology , Photosystem I Protein Complex/metabolism , Plastoquinone/metabolism , Quinone Reductases/metabolism
20.
J Biol Chem ; 298(1): 101435, 2022 01.
Article in English | MEDLINE | ID: mdl-34808207

ABSTRACT

The dual roles of H2S as an endogenously synthesized respiratory substrate and as a toxin raise questions as to how it is cleared when the electron transport chain is inhibited. Sulfide quinone oxidoreductase (SQOR) catalyzes the first step in the mitochondrial H2S oxidation pathway, using CoQ as an electron acceptor, and connects to the electron transport chain at the level of complex III. We have discovered that at high H2S concentrations, which are known to inhibit complex IV, a new redox cycle is established between SQOR and complex II, operating in reverse. Under these conditions, the purine nucleotide cycle and the malate aspartate shuttle furnish fumarate, which supports complex II reversal and leads to succinate accumulation. Complex II knockdown in colonocytes decreases the efficiency of H2S clearance while targeted knockout of complex II in intestinal epithelial cells significantly decreases the levels of thiosulfate, a biomarker of H2S oxidation, to approximately one-third of the values seen in serum and urine samples from control mice. These data establish the physiological relevance of this newly discovered redox circuitry between SQOR and complex II for prioritizing H2S oxidation and reveal the quantitatively significant contribution of intestinal epithelial cells to systemic H2S metabolism.


Subject(s)
Hydrogen Sulfide , Quinone Reductases , Animals , Electron Transport Complex IV/antagonists & inhibitors , Electron Transport Complex IV/metabolism , Hydrogen Sulfide/metabolism , Mice , Oxidation-Reduction , Quinone Reductases/genetics , Quinone Reductases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...