Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.166
Filter
1.
ACS Appl Mater Interfaces ; 16(29): 37521-37529, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-38985575

ABSTRACT

Sodium ions and protons regulate various fundamental processes at the cell and tissue levels across all biological kingdoms. It is therefore pivotal for bioelectronic devices, such as biosensors and biotransducers, to control the transport of these ions through biological membranes. Our study explores the regulation of proton and sodium concentrations by integrating an Na+-type ATP synthase, a glucose dehydrogenase (GDH), and a urease into a multienzyme logic system. This system is designed to operate using various chemical control input signals, while the output current corresponds to the local change in proton or sodium concentrations. Therein, a H+ and Na+ biotransducer was integrated to fulfill the roles of signal transducers for the monitoring and simultaneous control of Na+ and H+ levels, respectively. To increase the proton concentration at the output, we utilized GDH driven by the inputs of glucose and nicotinamide adenine dinucleotide (NAD+), while recorded the signal change from the biotransducer, together acting as an AND enzyme logic gate. On the contrary, we introduced urease enzyme which hydrolyzed urea to control the decrease in proton concentration, serving as a NOT gate and reset. By integrating these two enzyme logic gates we formed a simple multienzyme logic system for the control of proton concentrations. Furthermore, we also demonstrate a more complex, Na+-type ATP synthase-urease multienzyme logic system, controlled by the two different inputs of ADP and urea. By monitoring the voltage of the peak current as the output signal, this logic system acts as an AND enzyme logic gate. This study explores how multienzyme logic systems can modulate biologically important ion concentrations, opening the door toward advanced biological on-demand control of a variety of bioelectronic enzyme-based devices, such as biosensors and biotransducers.


Subject(s)
Glucose 1-Dehydrogenase , Sodium , Sodium/metabolism , Sodium/chemistry , Glucose 1-Dehydrogenase/metabolism , Glucose 1-Dehydrogenase/chemistry , Urease/metabolism , Urease/chemistry , Protons , Glucose/metabolism , Biosensing Techniques/methods , NAD/metabolism , NAD/chemistry
2.
J Am Chem Soc ; 146(28): 18817-18822, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38968608

ABSTRACT

NAD(H)-dependent enzymes play a crucial role in the biosynthesis of pharmaceuticals and fine chemicals, but the limited recyclability of the NAD(H) cofactor hinders its more general application. Here, we report the generation of mechano-responsive PEI-modified Cry3Aa protein crystals and their use for NADH recycling over multiple reaction cycles. For demonstration of its practical utility, a complementary Cry3Aa protein particle containing genetically encoded and co-immobilized formate dehydrogenase for NADH regeneration and leucine dehydrogenase for catalyzing the NADH-dependent l-tert-leucine (l-tert-Leu) biosynthesis has been produced. When combined with the PEI-modified Cry3Aa crystal, the resultant reaction system could be used for the efficient biosynthesis of l-tert-Leu for up to 21 days with a 10.5-fold improvement in the NADH turnover number.


Subject(s)
Formate Dehydrogenases , NAD , NAD/metabolism , NAD/chemistry , Formate Dehydrogenases/metabolism , Formate Dehydrogenases/chemistry , Leucine Dehydrogenase/metabolism , Leucine Dehydrogenase/chemistry , Crystallization , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Models, Molecular
3.
Anal Chem ; 96(32): 13308-13316, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39078110

ABSTRACT

NAD(P)H: quinone oxidoreductase-1 (NQO1) plays critical roles in antioxidation and abnormally overexpresses in tumors. Developing a fast and sensitive method of monitoring NQO1 will greatly promote cancer diagnosis in clinical practice. This study introduces a transformative colorimetric detection strategy for NQO1, harnessing an innovative competitive substrate mechanism between NQO1 and a new NADH oxidase (NOX) mimic, cobalt-nitrogen-doped carbon nanozyme (CoNC). This method ingeniously exploits the differential consumption of NADH in the presence of NQO1 to modulate the generation of H2O2 from CoNC catalysis, which is then quantified through a secondary, peroxidase-mimetic cascade reaction involving Prussian blue (PB) nanoparticles. This dual-stage reaction framework not only enhances the sensitivity of NQO1 detection, achieving a limit of detection as low as 0.67 µg mL-1, but also enables the differentiation between cancerous and noncancerous cells by their enzymatic activity profiles. Moreover, CoNC exhibits exceptional catalytic efficiency, with a specific activity reaching 5.2 U mg-1, significantly outperforming existing NOX mimics. Beyond mere detection, CoNC serves a dual role, acting as both a robust mimic of cytochrome c reductase (Cyt c) and a cornerstone for enzymatic regeneration, thereby broadening the scope of its biological applications. This study not only marks a significant step forward in the bioanalytical application of nanozymes but also sets the stage for their expanded use in clinical diagnostics and therapeutic monitoring.


Subject(s)
Colorimetry , NAD(P)H Dehydrogenase (Quinone) , NADH, NADPH Oxidoreductases , NAD(P)H Dehydrogenase (Quinone)/metabolism , NAD(P)H Dehydrogenase (Quinone)/chemistry , Humans , NADH, NADPH Oxidoreductases/metabolism , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Multienzyme Complexes/metabolism , Multienzyme Complexes/chemistry , Cobalt/chemistry , Carbon/chemistry , Biomimetics , Limit of Detection , Nitrogen/chemistry , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Ferrocyanides/chemistry , NAD/metabolism , NAD/chemistry
4.
Dalton Trans ; 53(32): 13591-13601, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39078263

ABSTRACT

Here, we have synthesized and characterized three visible light responsive terpyridine based-Re(I)-tricarbonyl complexes; [Re(CO)3(ph-tpy)Cl] (Retp1), [Re(CO)3(an-tpy)Cl] (Retp2), and [Re(CO)3(py-tpy)Cl] (Retp3) where ph-tpy = 4'-phenyl-2,2':6',2″-terpyridine; an-tpy = 4'-anthracenyl-2,2':6',2″-terpyridine, py-tpy = 4'-pyrenyl-2,2':6',2″-terpyridine. The structures of Retp1 and Retp2 were confirmed from the SC-XRD data, indicating distorted octahedral structures. Unlike traditional PDT agents, these complexes generated reactive oxygen species (ROS) via type I and type II pathways and oxidized redox crucial NADH (reduced nicotinamide adenine dinucleotide) upon visible light exposure. Retp3 showed significant mitochondrial localization and demonstrated photoactivated anticancer activity (IC50 ∼ 2 µM) by inducing ROS-mediated cell death in cancer cells selectively (photocytotoxicity Index, PI > 28) upon compromising mitochondrial function in A549 cells. Their diagnostic capabilities were ultimately assessed using clinically relevant 3D multicellular tumor spheroids (MCTs).


Subject(s)
Antineoplastic Agents , Coordination Complexes , NAD , Oxidation-Reduction , Pyridines , Reactive Oxygen Species , Rhenium , Humans , Reactive Oxygen Species/metabolism , NAD/chemistry , NAD/metabolism , Pyridines/chemistry , Pyridines/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/pharmacology , Coordination Complexes/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Rhenium/chemistry , Rhenium/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemical synthesis , Light , Drug Screening Assays, Antitumor , Photochemotherapy , Molecular Structure , Cell Proliferation/drug effects , Cell Survival/drug effects , A549 Cells , Cell Line, Tumor
5.
J Am Chem Soc ; 146(30): 21025-21033, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39025790

ABSTRACT

Artificial photosynthesis represents a sustainable strategy for accessing high-value chemicals; however, the conversion efficiency is significantly limited by its difficulty in the cycle of coenzymes such as NADH. In this study, we report a series of isostructural triazine covalent organic frameworks (COFs) and explore their N-substituted microenvironment-dependent photocatalytic activity for NADH regeneration. We discovered that the rational alteration of N-heterocyclic species, which are linked to the triazine center through an imine linkage, can significantly regulate both the electron band structure and planarity of a COF layer. This results in different separation efficiencies of the photoinduced electron-hole pairs and electron transfer behavior within and between individual layers. The optimal COF catalyst herein achieves an NADH regeneration capacity of 89% within 20 min, outperforming most of the reported nanomaterial photocatalysts. Based on this, an artificial photosynthesis system is constructed for the green synthesis of a high-value compound, L-glutamate, and its conversion efficiency significantly surpasses the enzymatic approach without the NADH photocatalytic cycle. This work offers new insights into the coenzyme regeneration by means of regulating the distal heterocyclic microenvironment of a COF skeleton, holding great potential for the green photosynthesis of important chemicals.


Subject(s)
Metal-Organic Frameworks , Triazines , Triazines/chemistry , Catalysis , Metal-Organic Frameworks/chemistry , NAD/chemistry , NAD/metabolism , Photochemical Processes , Molecular Structure , Coenzymes/chemistry , Coenzymes/metabolism , Photosynthesis
6.
Anal Chem ; 96(28): 11549-11556, 2024 07 16.
Article in English | MEDLINE | ID: mdl-38958207

ABSTRACT

Human-borne acetone is a potent marker of lipid metabolism. Here, an enzyme immobilization method for secondary alcohol dehydrogenase (S-ADH), which is suitable for highly sensitive and selective biosensing of acetone, was developed, and then its applicability was demonstrated for spatiotemporal imaging of concentration distribution. After various investigations, S-ADH-immobilized meshes could be prepared with less than 5% variation by cross-linking S-ADH with glutaraldehyde on a cotton mesh at 40 °C for 15 min. Furthermore, high activity was obtained by adjusting the concentration of the coenzyme nicotinamide adenine dinucleotide (NADH) solution added to the S-ADH-immobilized mesh to 500 µM and the solvent to a potassium phosphate buffer solution at pH 6.5. The gas imaging system using the S-ADH-immobilized mesh was able to image the decrease in NADH fluorescence (ex 340 nm, fl 490 nm) caused by the catalytic reaction of S-ADH and the acetone distribution in the concentration range of 0.1-10 ppm-v, including the breath concentration of healthy people at rest. The exhaled breath of two healthy subjects at 6 h of fasting was quantified as 377 and 673 ppb-v, which were consistent with the values quantified by gas chromatography-mass spectrometry.


Subject(s)
Acetone , Breath Tests , Enzymes, Immobilized , Acetone/analysis , Acetone/chemistry , Humans , Enzymes, Immobilized/chemistry , Enzymes, Immobilized/metabolism , Biosensing Techniques , Alcohol Oxidoreductases/chemistry , Alcohol Oxidoreductases/metabolism , Gases/chemistry , Gases/analysis , Exhalation , NAD/analysis , NAD/chemistry , NAD/metabolism
7.
Anal Chem ; 96(29): 12120-12128, 2024 07 23.
Article in English | MEDLINE | ID: mdl-38990044

ABSTRACT

Dihydro-nicotinamide adenine dinucleotide (NADH) detection is crucial since it is a vital coenzyme in organism metabolism. Compared to the traditional method based on natural NADH oxidase (NOX), nanozymes with multienzyme-like activity can catalyze multistage reactions in a singular setup, simplifying detection processes and enhancing sensitivity. In this study, an innovative NADH detection method was developed using iron-doped carbon (Fe@C) nanozyme synthesized from metal-organic frameworks with in situ reduced Pt clusters. This nanozyme composite (Pt/Fe@C) demonstrated dual NOX and peroxidase-like characteristics, significantly enhancing the catalytic efficiency and enabling NADH conversion to NAD+ and H2O2 with subsequent detection. The collaborative research involving both experimental and theoretical simulations has uncovered the catalytic process and the cooperative effect of Fe and Pt atoms, leading to enhanced oxygen adsorption and activation, as well as a decrease in the energy barrier of the key step in the H2O2 decomposition process. These findings indicate that the catalytic performance of Pt/Fe@C in NOX-like and POD-like reactions can be significantly improved. The colorimetric sensor detects NADH with a limit of detection as low as 0.4 nM, signifying a breakthrough in enzyme-mimicking nanozyme technology for precise NADH measurement.


Subject(s)
Carbon , Metal-Organic Frameworks , NAD , Platinum , NAD/chemistry , Metal-Organic Frameworks/chemistry , Platinum/chemistry , Carbon/chemistry , Iron/chemistry , Hydrogen Peroxide/chemistry , Colorimetry/methods , Humans , Catalysis , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Biomimetic Materials/chemistry , Limit of Detection , NADH, NADPH Oxidoreductases
8.
Bioorg Chem ; 149: 107509, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38824699

ABSTRACT

In mammals, nicotinamide phosphoribosyltransferase (NAMPT) is a crucial enzyme in the nicotinamide adenine dinucleotide (NAD+) synthesis pathway catalyzing the condensation of nicotinamide (NAM) with 5-phosphoribosyl-1-pyrophosphate (PRPP) to produce nicotinamide mononucleotide (NMN). Given the pivotal role of NAD+ in a range of cellular functions, including DNA synthesis, redox reactions, cytokine generation, metabolism, and aging, NAMPT has become a promising target for many diseases, notably cancer. Therefore, various NAMPT inhibitors have been reported and classified as first and second-generation based on their chemical structures and design strategies, dual-targeted being one. However, most NAMPT inhibitors suffer from several limitations, such as dose-dependent toxicity and poor pharmacokinetic properties. Consequently, there is no clinically approved NAMPT inhibitor. Hence, research on discovering more effective and less toxic dual-targeted NAMPT inhibitors with desirable pharmacokinetic properties has drawn attention recently. This review summarizes the previously reported dual-targeted NAMPT inhibitors, focusing on their design strategies and advantages over the single-targeted therapies.


Subject(s)
Antineoplastic Agents , Enzyme Inhibitors , Neoplasms , Nicotinamide Phosphoribosyltransferase , Animals , Humans , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemical synthesis , Molecular Structure , Neoplasms/drug therapy , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Structure-Activity Relationship , NAD/chemistry , NAD/metabolism , Niacinamide/chemistry
9.
Nat Commun ; 15(1): 4900, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851775

ABSTRACT

Excitation energy transfer (EET) is a key photoinduced process in biological chromophoric assemblies. Here we investigate the factors which can drive EET into efficient ultrafast sub-ps regimes. We demonstrate how a coherent transport of electronic population could facilitate this in water solvated NADH coenzyme and uncover the role of an intermediate dark charge-transfer state. High temporal resolution ultrafast optical spectroscopy gives a 54±11 fs time constant for the EET process. Nonadiabatic quantum dynamical simulations computed through the time-evolution of multidimensional wavepackets suggest that the population transfer is mediated by photoexcited molecular vibrations due to strong coupling between the electronic states. The polar aqueous solvent environment leads to the active participation of a dark charge transfer state, accelerating the vibronically coherent EET process in favorably stacked conformers and solvent cavities. Our work demonstrates how the interplay of structural and environmental factors leads to diverse pathways for the EET process in flexible heterodimers and provides general insights relevant for coherent EET processes in stacked multichromophoric aggregates like DNA strands.


Subject(s)
Energy Transfer , NAD , NAD/chemistry , NAD/metabolism , Quantum Theory , Water/chemistry
10.
Langmuir ; 40(25): 13144-13154, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38869442

ABSTRACT

Reduced nicotinamide adenine dinucleotide (NADH)-detecting electrochemical sensors are attractive in monitoring and diagnosing various physiological disorders of NADH abnormalities. The NADH detection methods using conventional electrodes are challenging due to slow electron transfer and fouling effect. Interestingly, paper-based flexible and disposable electrodes (PE) are superior for sensing biomolecules through simple detection procedures with excellent sensitivity and selectivity. Herein, to construct a conducting polypeptide-modified paper electrode, initially, polytyrosine (PTyr) is synthesized from l-tyrosine N-carboxy anhydride through ring-opening polymerization, and PTyr is drop-coated on the PE. The PTyr-modified paper electrode (PMPE) demonstrated excellent electrochemical properties and facilitated the electrooxidation of NADH at a lower potential of 576 mV. The PMPE displayed a linear detection between 25 and 145 µM of NADH concentration, with a lower detection limit of 0.340 µM. Under ideal circumstances, the sensor developed displayed an excellent NADH detection capability without interference with the most common electroactive species, ascorbic acid. The PMPE facilitates good electrocatalytic activity toward NADH, which can also be employed as a substrate material for biofuel cells.


Subject(s)
Electrodes , NAD , Paper , NAD/analysis , NAD/chemistry , Peptides/chemistry , Electrochemical Techniques/methods , Electrochemical Techniques/instrumentation , Oxidation-Reduction , Limit of Detection , Biosensing Techniques/methods
11.
Bioorg Chem ; 147: 107418, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38703441

ABSTRACT

A key approach in developing green chemistry involves converting solar energy into chemical energy of biomolecules through photocatalysis. Photocatalysis can facilitate the regeneration of nicotinamide cofactors during redox processes. Nicotinamide cofactor biomimetics (NCBs) are economical substitutes for natural cofactors. Here, photocatalytic regeneration of NADH and reduced NCBs (NCBsred) using graphitic carbon nitride (g-C3N4) was developed. The process involves g-C3N4 as the photocatalyst, Cp*Rh(bpy)H2O2+ as the electron mediator, and Triethanolamine as the electron donor, facilitating the reduction of NAD+ and various oxidative NCBs (NCBsox) under light irradiation. Notably, the highest reduction yield of 48.32 % was achieved with BANA+, outperforming the natural cofactor NAD+. Electrochemical analysis reveals that the reduction efficiency and capacity of cofactors relies on their redox potentials. Additionally, a coupled photo-enzymatic catalysis system was explored for the reduction of 4-Ketoisophorone by Old Yellow Enzyme XenA. Among all the NCBsox and NAD+, the highest conversion ratio of over 99 % was obtained with BANA+. After recycled for 8 times, g-C3N4 maintained over 93.6 % catalytic efficiency. The photocatalytic cofactor regeneration showcases its outstanding performance with NAD+ as well as NCBsox. This work significantly advances the development of photocatalytic cofactor regeneration for artificial cofactors and its potential application.


Subject(s)
Biocatalysis , Oxidation-Reduction , Photochemical Processes , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Molecular Structure , NAD/chemistry , NAD/metabolism , Biomimetics , Niacinamide/chemistry , Niacinamide/metabolism , Nitrogen Compounds/chemistry , Graphite
12.
Chem Commun (Camb) ; 60(46): 5932-5935, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38757567

ABSTRACT

A novel NIR fluorescent probe based on quinoline-conjugated benzo[cd]indol dual-salt for NADH was developed. This probe swiftly detects and responds sensitively to both endogenous and exogenous NADH alterations, enabling imaging of NADH fluctuations in type II diabetic and AD model cells.


Subject(s)
Fluorescent Dyes , Mitochondria , NAD , Fluorescent Dyes/chemistry , Fluorescent Dyes/chemical synthesis , NAD/analysis , NAD/chemistry , Mitochondria/metabolism , Mitochondria/chemistry , Humans , Quinolines/chemistry , Infrared Rays , Optical Imaging , Animals , Diabetes Mellitus, Type 2
13.
Adv Mater ; 36(30): e2310731, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38805174

ABSTRACT

As a central metabolic molecule, nicotinamide adenine dinucleotide (NAD+) can potentially treat acute kidney injury (AKI) and chronic kidney disease (CKD); however, its bioavailability is poor due to short half-life, instability, the deficiency of targeting, and difficulties in transmembrane transport. Here a physiologically adaptive gallic acid-NAD+ nanoparticle is designed, which has ultrasmall size and pH-responsiveness, passes through the glomerular filtration membrane to reach injured renal tubules, and efficiently delivers NAD+ into the kidneys. With an effective accumulation in the kidneys, it restores renal function, immune microenvironment homeostasis, and mitochondrial homeostasis of AKI mice via the NAD+-Sirtuin-1 axis, and exerts strong antifibrotic effects on the AKI-to-CKD transition by inhibiting TGF-ß signaling. It also exhibits excellent stability, biodegradable, and biocompatible properties, ensuring its long-term safety, practicality, and clinical translational feasibility. The present study shows a potential modality of mitochondrial repair and immunomodulation through nanoagents for the efficient and safe treatment of AKI and CKD.


Subject(s)
Acute Kidney Injury , Anti-Inflammatory Agents , Kidney , Mitochondria , NAD , Nanoparticles , Polyphenols , Renal Insufficiency, Chronic , Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Animals , NAD/metabolism , NAD/chemistry , Mice , Mitochondria/metabolism , Mitochondria/drug effects , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/pathology , Polyphenols/chemistry , Polyphenols/pharmacology , Nanoparticles/chemistry , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Kidney/metabolism , Kidney/pathology , Disease Progression , Gallic Acid/chemistry , Gallic Acid/pharmacology , Humans
14.
Biochemistry ; 63(10): 1347-1358, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38691339

ABSTRACT

The physiological role of dihydroorotate dehydrogenase (DHOD) enzymes is to catalyze the oxidation of dihydroorotate to orotate in pyrimidine biosynthesis. DHOD enzymes are structurally diverse existing as both soluble and membrane-associated forms. The Family 1 enzymes are soluble and act either as conventional single subunit flavin-dependent dehydrogenases known as Class 1A (DHODA) or as unusual heterodimeric enzymes known as Class 1B (DHODB). DHODBs possess two active sites separated by ∼20 Å, each with a noncovalently bound flavin cofactor. NAD is thought to interact at the FAD containing site, and the pyrimidine substrate is known to bind at the FMN containing site. At the approximate center of the protein is a single Fe2S2 center that is assumed to act as a conduit, facilitating one-electron transfers between the flavins. We present anaerobic transient state analysis of a DHODB enzyme from Lactoccocus lactis. The data presented primarily report the exothermic reaction that reduces orotate to dihydroorotate. The reductive half reaction reveals rapid two-electron reduction that is followed by the accumulation of a four-electron reduced state when NADH is added in excess, suggesting that the initial two electrons acquired reside on the FMN cofactor. Concomitant with the first reduction is the accumulation of a long-wavelength absorption feature consistent with the blue form of a flavin semiquinone. Spectral deconvolution and fitting to a model that includes reversibility for the second electron transfer reveals equilibrium accumulation of a flavin bisemiquinone state that has features of both red and blue semiquinones. Single turnover reactions with limiting NADH and excess orotate reveal that the flavin bisemiquinone accumulates with reduction of the enzyme by NADH and decays with reduction of the pyrimidine substrate, establishing the bisemiquinone as a fractional state of the two-electron reduced intermediate observed.


Subject(s)
Dihydroorotate Dehydrogenase , Lactococcus lactis , Biocatalysis , Catalysis , Catalytic Domain , Flavin Mononucleotide/metabolism , Flavin Mononucleotide/chemistry , Flavin-Adenine Dinucleotide/metabolism , Flavin-Adenine Dinucleotide/chemistry , Kinetics , Lactococcus lactis/enzymology , Lactococcus lactis/metabolism , NAD/metabolism , NAD/chemistry , Oxidation-Reduction
15.
J Biol Inorg Chem ; 29(4): 455-475, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38780762

ABSTRACT

Ferritins are multimeric nanocage proteins that sequester/concentrate excess of free iron and catalytically synthesize a hydrated ferric oxyhydroxide bio-mineral. Besides functioning as the primary intracellular iron storehouses, these supramolecular assemblies also oversee the controlled release of iron to meet physiologic demands. By virtue of the reducing nature of the cytosol, reductive dissolution of ferritin-iron bio-mineral by physiologic reducing agents might be a probable pathway operating in vivo. Herein, to explore this reductive iron-release pathway, a series of quinone analogs differing in size, position/nature of substituents and redox potentials were employed to relay electrons from physiologic reducing agent, NADH, to the ferritin core. Quinones are well known natural electron/proton mediators capable of facilitating both 1/2 electron transfer processes and have been implicated in iron/nutrient acquisition in plants and energy transduction. Our findings on the structure-reactivity of quinone mediators highlight that iron release from ferritin is dictated by electron-relay capability (dependent on E1/2 values) of quinones, their molecular structure (i.e., the presence of iron-chelation sites and the propensity for H-bonding) and the type/amount of reactive oxygen species (ROS) they generate in situ. Juglone/Plumbagin released maximum iron due to their intermediate E1/2 values, presence of iron chelation sites, the ability to inhibit in situ generation of H2O2 and form intramolecular H-bonding (possibly promotes semiquinone formation). This study may strengthen our understanding of the ferritin-iron-release process and their significance in bioenergetics/O2-based cellular metabolism/toxicity while providing insights on microbial/plant iron acquisition and the dynamic host-pathogen interactions.


Subject(s)
Ferritins , Iron , NAD , Oxidation-Reduction , Quinones , Reactive Oxygen Species , Ferritins/chemistry , Ferritins/metabolism , Iron/metabolism , Iron/chemistry , NAD/metabolism , NAD/chemistry , Oxygen/metabolism , Oxygen/chemistry , Quinones/chemistry , Quinones/metabolism , Reactive Oxygen Species/metabolism , Mycobacterium
16.
Colloids Surf B Biointerfaces ; 239: 113903, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38599036

ABSTRACT

Nicotinamide mononucleotide (NMN) is being investigated for its ability to address the decline in NAD+ level during aging. This study aimed to construct a delivery system based on ovalbumin and fucoidan nanoparticles to ameliorate the bioaccessibility of NMN by increasing NAD+ level in aging mouse model. The NMN-loaded ovalbumin and fucoidan nanoparticles (OFNPs) were about 177 nm formed by the interplay of hydrogen bonds between ovalbumin and fucoidan. Compared with free NMN, NMN-loaded OFNPs intervention could obviously improve the antioxidant enzyme activity of senescent cell induced by D-galactose. The NMN-loaded OFNPs treatment could ameliorate the loss of weight and organ index induced by senescence, and maintain the water content for the aging mice. The Morris maze test indicated that hitting blind side frequency and escape time of NMN-loaded OFNPs group decreased by 13% and 35% compared with that of free NMN group. Furthermore, the NMN-loaded OFNPs significantly alleviated the age-related oxidative stress and increased the generation of NAD+ 1.34 times by improving the bioaccessibility of NMN. Our data in this study supplied a strategy to enhance the bioavailability of NMN in senescence treatment.


Subject(s)
Aging , Galactose , NAD , Nanoparticles , Nicotinamide Mononucleotide , Polysaccharides , Animals , Galactose/chemistry , Nanoparticles/chemistry , Mice , Aging/drug effects , Polysaccharides/chemistry , Polysaccharides/pharmacology , NAD/metabolism , NAD/chemistry , Nicotinamide Mononucleotide/chemistry , Nicotinamide Mononucleotide/pharmacology , Particle Size , Male , Oxidative Stress/drug effects , Ovalbumin
17.
J Mol Graph Model ; 130: 108778, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38652998

ABSTRACT

SIRT6 is of interest for its promising effect in the treatment of aging-related diseases. Studies have shown quercetin (QUE) and its derivatives have varying degrees of effect on the catalytic effect of SIRT6. In the research, the effect of QUE on the protein-substrate interaction in the SIRT6-mediated mono-ADP ribosylation system was investigated by conventional molecular dynamics (MD) simulations combined with MM/PBSA binding free energy calculations. The results show that QUE can bind stably to SIRT6 with the binding energy of -22.8 kcal/mol and further affect the atomic interaction between SIRT6 and NAD+ (or H3K9), resulting in an increased affinity between SIRT6-NAD+ and decreased SIRT6-H3K9 binding capacity. At the same time, the binding of QUE can also alter some structural characteristics of the protein, with large shifts occurring in the residue regions involving the N-terminal (residues 1-27), Rossmann fold regions (residues 55-92), and ZBD (residues 164-179). Thus, QUE shows great potential as a scaffold for the design of novel potent SIRT6 modulators.


Subject(s)
Molecular Dynamics Simulation , Protein Binding , Quercetin , Sirtuins , Quercetin/chemistry , Quercetin/pharmacology , Sirtuins/chemistry , Sirtuins/metabolism , Humans , Binding Sites , NAD/chemistry , NAD/metabolism , Thermodynamics , Molecular Docking Simulation , Substrate Specificity , Hydrogen Bonding , Protein Conformation
18.
ChemistryOpen ; 13(8): e202400064, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38607952

ABSTRACT

The direct electrochemical reduction of nicotinamide adenine dinucleotide (NAD+) results in various products, complicating the regeneration of the crucial 1,4-NADH cofactor for enzymatic reactions. Previous research primarily focused on steady-state polarization to examine potential impacts on product selectivity. However, this study explores the influence of dynamic conditions on the selectivity of NAD+ reduction products by comparing two dynamic profiles with steady-state conditions. Our findings reveal that the main products, including 1,4-NADH, several dimers, and ADP-ribose, remained consistent across all conditions. A minor by-product, 1,6-NADH, was also identified. The product distribution varied depending on the experimental conditions (steady state vs. dynamic) and the concentration of NAD+, with higher concentrations and overpotentials promoting dimerization. The optimal yield of 1,4-NADH was achieved under steady-state conditions with low overpotential and NAD+ concentrations. While dynamic conditions enhanced the 1,4-NADH yield at shorter reaction times, they also resulted in a significant amount of unidentified products. Furthermore, this study assessed the potential of using pulsed electrochemical regeneration of 1,4-NADH with enoate reductase (XenB) for cyclohexenone reduction.


Subject(s)
Electrochemical Techniques , NAD , Oxidation-Reduction , NAD/chemistry , NAD/metabolism
19.
Chemistry ; 30(35): e202400411, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38640109

ABSTRACT

Nicotinamide adenine dinucleotide (NAD+) is a redox active molecule that is universally found in biology. Despite the importance and simplicity of this molecule, few reports exist that investigate which molecular features are important for the activity of this ribodinucleotide. By exploiting the nonenzymatic reduction and oxidation of NAD+ by pyruvate and methylene blue, respectively, we were able to identify key molecular features necessary for the intrinsic activity of NAD+ through kinetic analysis. Such features may explain how NAD+ could have been selected early during the emergence of life. Simpler molecules, such as nicotinamide, that lack an anomeric carbon are incapable of accepting electrons from pyruvate. The phosphate moiety inhibits activity in the absence of metal ions but facilitates activity at physiological pH and model prebiotic conditions by recruiting catalytic Mg2+. Reduction proceeds through consecutive single electron transfer events. Of the derivatives tested, including nicotinamide mononucleotide, nicotinamide riboside, 3-(aminocarbonyl)-1-(2,3-dihydroxypropyl)pyridinium, 1-methylnicotinamide, and nicotinamide, only NAD+ and nicotinamide mononucleotide would be capable of efficiently accepting and donating electrons within a nonenzymatic electron transport chain. The data are consistent with early metabolic chemistry exploiting NAD+ or nicotinamide mononucleotide and not simpler molecules.


Subject(s)
Magnesium , NAD , Niacinamide , Oxidation-Reduction , NAD/chemistry , NAD/metabolism , Magnesium/chemistry , Niacinamide/chemistry , Niacinamide/analogs & derivatives , Binding Sites , Kinetics , Nicotinamide Mononucleotide/chemistry , Nicotinamide Mononucleotide/metabolism , Pyridinium Compounds/chemistry , Methylene Blue/chemistry , Pyruvic Acid/chemistry , Pyruvic Acid/metabolism , Electron Transport
20.
Angew Chem Int Ed Engl ; 63(27): e202404024, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38641561

ABSTRACT

Here we demonstrate the preparation of enzyme-metal biohybrids of NAD+ reductase with biocatalytically-synthesised small gold nanoparticles (NPs, <10 nm) and core-shell gold-platinum NPs for tandem catalysis. Despite the variety of methods available for NP synthesis, there remains a need for more sustainable strategies which also give precise control over the shape and size of the metal NPs for applications in catalysis, biomedical devices, and electronics. We demonstrate facile biosynthesis of spherical, highly uniform, gold NPs under mild conditions using an isolated enzyme moiety, an NAD+ reductase, to reduce metal salts while oxidising a nicotinamide-containing cofactor. By subsequently introducing platinum salts, we show that core-shell Au@Pt NPs can then be formed. Catalytic function of these enzyme-Au@Pt NP hybrids was demonstrated for H2-driven NADH recycling to support enantioselective ketone reduction by an NADH-dependent alcohol dehydrogenase.


Subject(s)
Biocatalysis , Gold , Metal Nanoparticles , NAD , Platinum , Metal Nanoparticles/chemistry , NAD/chemistry , NAD/metabolism , Gold/chemistry , Platinum/chemistry , Hydrogen/chemistry , Hydrogen/metabolism , Alcohol Dehydrogenase/metabolism , Alcohol Dehydrogenase/chemistry , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL