Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.703
Filtrar
1.
Int J Mol Sci ; 25(18)2024 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-39337629

RESUMO

Different types of macrophages (Mφ) are involved in atherogenesis, including inflammatory Mφ and foamy Mφ (FM). Our previous study demonstrated that two-photon excited fluorescence (TPEF) imaging of NADH and FAD autofluorescence (AF) could distinguish experimental models that mimic the different atherosclerotic Mφ types. The present study assessed whether optical differences correlated with phenotypic and functional differences, potentially guiding diagnostic and therapeutic strategies. Phenotypic differences were investigated using three-dimensional principal component analysis and multi-color flow cytometry. Functional analyses focused on cytokine production, metabolic profiles, and cellular oxidative stress, in LDL dose-dependent assays, to understand the origin of AF in the FAD spectrum and assess FM ability to transition toward an immunoregulatory phenotype and function. Phenotypic studies revealed that FM models generated with acetylated LDL (Mac) were closer to immunoregulatory Mφ, while those generated with oxidized LDL (Mox) more closely resembled inflammatory Mφ. The metabolic analysis confirmed that inflammatory Mφ primarily used glycolysis, while immunoregulatory Mφ mainly depended on mitochondrial respiration. FM models employed both pathways; however, FM models generated with high doses of modified LDL showed reduced mitochondrial respiration, particularly Mox FM. Thus, the high AF in the FAD spectrum in Mox was not linked to increased mitochondrial respiration, but correlated with the dose of oxidized LDL, leading to increased production of reactive oxygen species (ROS) and lysosomal ceroid accumulation. High FAD-like AF, ROS, and ceroid accumulation were reduced by incubation with α-tocopherol. The cytokine profiles supported the phenotypic analysis, indicating that Mox FM exhibited greater inflammatory activity than Mac FM, although both could be redirected toward immunoregulatory functions, albeit to different degrees. In conclusion, in the context of immunoregulatory therapies for atherosclerosis, it is crucial to consider FM, given their prevalence in plaques and our results, as potential targets, regardless of their inflammatory status, alongside non-foamy inflammatory Mφ.


Assuntos
Aterosclerose , Macrófagos , Fenótipo , Aterosclerose/metabolismo , Aterosclerose/imunologia , Aterosclerose/patologia , Macrófagos/metabolismo , Macrófagos/imunologia , Humanos , Animais , Estresse Oxidativo , Lipoproteínas LDL/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Citocinas/metabolismo , Células Espumosas/metabolismo , Mitocôndrias/metabolismo , Camundongos , Flavina-Adenina Dinucleotídeo/metabolismo
2.
Biofouling ; 40(9): 617-631, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39291398

RESUMO

The impact of Flavin adenine dinucleotide (FAD) on sulfate-reducing bacteria (SRB) corrosion of a pipeline welded joint (WJ) was investigated under anaerobic condition in this paper. The results showed that the thickness of the corrosion product on heat affected zone (HAZ) was lower than that on base metal (BM) and welded zone (WZ), and the FAD addition enhanced the development of the protruding microbial tubercles on the WJ. The local corrosion degrees of the BM and WZ coupons were significantly higher than that of the HAZ coupon. Besides, the FAD addition simultaneously promoted local corrosion of all three zones of the WJ in the SRB inoculated environment, and the promotion role was much more pronounced on the WZ coupons. The selective promotion effect of FAD on SRB corrosion in the WJ was attributed to the special structure of the WZ, the selected SRB attachment and the FAD/FADH2 redox feedback cycle.


Assuntos
Desulfovibrio desulfuricans , Flavina-Adenina Dinucleotídeo , Corrosão , Flavina-Adenina Dinucleotídeo/metabolismo , Flavina-Adenina Dinucleotídeo/química , Desulfovibrio desulfuricans/metabolismo , Oxirredução , Biofilmes
3.
ACS Appl Mater Interfaces ; 16(33): 44004-44017, 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39132979

RESUMO

Enzyme-mediator bioconjugation is emerging as a building block for designing electrode platforms for the construction of biosensors and biofuel cells. Here, we report a one-pot bioconjugation technique for flavin adenine dinucleotide-dependent glucose dehydrogenase (FAD-GDH) and thionine (TH) using a series of cross-linkers, including epoxy, N-hydroxysuccinimide (NHS), and aldehydes. In this technique, FAD-GDH and thionine are conjugated through an amine cross-linking reaction to generate a redox network, which has been successfully employed for the oxidation of glucose. The bioconjugation chemistry of cross-linkers with the amino groups on FAD-GDH and thionine plays a vital role in generating distinct network structures. The epoxy-type cross-linker reacts with the primary and secondary amines of thionine at room temperature, thereby producing an FAD-GDH-TH-FAD-GDH hyperbranched bioconjugate network, the aldehyde undergoes a rapid cross-linking reaction to produce a network of FAD-GDH-FAD-GDH, while the NHS-based cross-linker can react with the primary amines of both FAD-GDH and thionine, forming an FAD-GDH-cross-linker-TH polymeric network. This reaction has the potential to enable the conjugation of a redox mediator with a FAD-GDH network, which is particularly essential when designing an enzyme electrode platform. The data demonstrated that the polymeric cross-linked network based on the NHS cross-linker exhibited a considerable increase in electron transport while producing a catalytic current of 830 µA cm-2. The cross-linker spacer arm length also affects the overall electrochemical function of the network and its performance; an adequate spacer length containing a cross-linker is required, resulting in a faster electron transfer. Finally, a leaching test confirmed that the stability of the enzyme electrode was improved when the electrode was tested using the redox probe. This study elucidates the relationship between cross-linking chemistry and redox network structure and enhances the high performance of enzyme electrode platforms for the oxidation of glucose.


Assuntos
Técnicas Biossensoriais , Reagentes de Ligações Cruzadas , Glucose 1-Desidrogenase , Oxirredução , Fenotiazinas , Fenotiazinas/química , Glucose 1-Desidrogenase/química , Glucose 1-Desidrogenase/metabolismo , Reagentes de Ligações Cruzadas/química , Técnicas Biossensoriais/métodos , Glucose/química , Flavina-Adenina Dinucleotídeo/química , Flavina-Adenina Dinucleotídeo/metabolismo , Eletrodos , Técnicas Eletroquímicas , Enzimas Imobilizadas/química , Enzimas Imobilizadas/metabolismo , Biocatálise
4.
Biochem Biophys Res Commun ; 733: 150575, 2024 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-39197199

RESUMO

Flavin monooxygenases (FMOs) have been widely used in the biosynthesis of natural compounds due to their excellent stereoselectivity, regioselectivity and chemoselectivity. Stenotrophomonas maltophilia flavin monooxygenase (SmFMO) has been reported to catalyze the oxidation of various thiols to corresponding sulfoxides, but its activity is relatively low. Herein, we obtained a mutant SmFMOF52G which showed 4.35-fold increase in kcat/Km (4.96 mM-1s-1) and 6.84-fold increase in enzyme activity (81.76 U/g) compared to the SmFMOWT (1.14 mM-1s-1 and 11.95 U/g) through semi-rational design guided by structural analysis and catalytic mechanism combined with high-throughput screening. By forming hydrogen bond with O4 atom of FAD isoalloxazine ring and reducing steric hindrance, the conformation of FAD isoalloxazine ring in SmFMOF52G is more stable, and NADPH and substrate are closer to FAD isoalloxazine ring, shortening the distances of hydrogen transfer and substrate oxygenation, thereby increasing the rate of reduction and oxidation reactions and enhancing enzyme activity. Additionally, the overall structural stability and substrate binding capacity of the SmFMOF52G have significant improved than that of SmFMOWT. The strategy used in this study to improve the enzyme activity of FMOs may have generality, providing important references for the rational and semi-rational engineering of FMOs.


Assuntos
Flavina-Adenina Dinucleotídeo , Flavinas , Oxigenases , Flavina-Adenina Dinucleotídeo/metabolismo , Flavina-Adenina Dinucleotídeo/química , Flavinas/metabolismo , Flavinas/química , Oxigenases/metabolismo , Oxigenases/química , Oxigenases/genética , Stenotrophomonas maltophilia/enzimologia , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Modelos Moleculares , Oxirredução , Especificidade por Substrato , Cinética
5.
Biochemistry ; 63(18): 2380-2389, 2024 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-39213510

RESUMO

Flavin cofactors offer a wide range of chemical mechanisms to support a great diversity in catalytic function. As a corollary, such diversity necessitates careful control within each flavoprotein to limit its function to an appropriate subset of possible reactions and substrates. This task falls to the protein environment surrounding the flavin in most enzymes. For iodotyrosine deiodinase that catalyzes a reductive dehalogenation of halotyrosines, substrates can dictate the chemistry available to the flavin. Their ability to stabilize the necessary one-electron reduced semiquinone form of flavin strictly depends on a direct coordination between the flavin and α-ammonium and carboxylate groups of its substrates. While perturbations to the carboxylate group do not significantly affect binding to the resting oxidized form of the deiodinase, dehalogenation (kcat/Km) is suppressed by over 2000-fold. Lack of the α-ammonium group abolishes detectable binding and dehalogenation. Substitution of the ammonium group with a hydroxyl group does not restore measurable binding but does support dehalogenation with an efficiency greater than those of the carboxylate derivatives. Consistent with these observations, the flavin semiquinone does not accumulate during redox titration in the presence of inert substrate analogues lacking either the α-ammonium or carboxylate groups. As a complement, a nitroreductase activity based on hydride transfer is revealed for the appropriate substrates with perturbations to their zwitterion.


Assuntos
Iodeto Peroxidase , Iodeto Peroxidase/metabolismo , Iodeto Peroxidase/química , Flavinas/metabolismo , Flavinas/química , Especificidade por Substrato , Oxirredução , Cinética , Flavina-Adenina Dinucleotídeo/metabolismo , Flavina-Adenina Dinucleotídeo/química , Flavina-Adenina Dinucleotídeo/análogos & derivados , Dinitrocresóis/metabolismo , Dinitrocresóis/química , Halogenação
6.
J Biophotonics ; 17(10): e202400294, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39198025

RESUMO

The fluorescence intensities of the cellular respiratory cofactors NADH (reduced nicotinamide adenine dinucleotide) and FAD++ (oxidized flavin adenine dinucleotide) reflect energy metabolism in skin and other tissues and can be quantified in vivo by fluorescence spectroscopy (FS). However, the variability of physiological parameters largely determines the reproducibility of measurement results and the reliability of the diagnostic test. In this prospective study, we evaluated the interday reproducibility of NADH and FAD++ fluorescence intensity measurements in the skin of 51 healthy volunteers assessed by the FS at baseline, after local cooling (10°C) and heating of the skin (35°C). Results showed that the fluorescence amplitude of NADH (AFNADH) in forearm skin was the most reproducible of the FS parameters studied. Assessment of AFNADH in the dorsal forearm in combination with a thermal functional test is the most promising method for clinical use for assessing energy metabolism in the skin.


Assuntos
Metabolismo Energético , NAD , Espectrometria de Fluorescência , Temperatura , Humanos , Masculino , Adulto , Reprodutibilidade dos Testes , Feminino , NAD/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Pele/metabolismo , Espaço Intracelular/metabolismo , Adulto Jovem , Biomarcadores/metabolismo , Pessoa de Meia-Idade
7.
mSphere ; 9(9): e0034724, 2024 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-39189775

RESUMO

Flavin adenine dinucleotide (FAD) is an essential cofactor for numerous flavoenzymes present in all living organisms. The biosynthesis of FAD from riboflavin involves two sequential reactions catalyzed by riboflavin kinase and flavin adenine dinucleotide synthase (FADS). Entamoeba histolytica, the protozoan parasite responsible for amebiasis, apparently lacks a gene encoding FADS that share similarity with bacterial and eukaryotic canonical FADS, yet it can synthesize FAD. In this study, we have identified the gene responsible for FADS and thoroughly characterized physiological and biochemical properties of FADS from E. histolytica. Phylogenetic analysis revealed that the gene was likely laterally transferred from archaea. The kinetic properties of recombinant EhFADS were consistent with the notion that EhFADS is of archaeal origin, exhibiting KM and kcat values similar to those of the arachaeal enzyme while significantly differing from the human counterpart. Repression of gene expression of EhFADS by epigenetic gene silencing caused substantial reduction in FAD levels and parasite growth, underscoring the importance of EhFADS for the parasite. Furthermore, we demonstrated that EhFADS gene silencing reduced thioredoxin reductase activity, which requires FAD as a cofactor and makes the ameba more susceptible to metronidazole. In summary, this study unveils unique evolutionary and biochemical features of EhFADS and underscores its significance as a promising drug target in combating human amebiasis.IMPORTANCEFAD is important for all forms of life, yet its role and metabolism are still poorly studied in E. histolytica, the protozoan parasite causing human amebiasis. Our study uncovers the evolutionary unique key enzyme, archaeal-type FADS for FAD biosynthesis from E. histolytica for the first time. Additionally, we showed the essentiality of this enzyme for parasite survival, highlighting its potential as target for drug development against E. histolytica infections.


Assuntos
Archaea , Entamoeba histolytica , Flavina-Adenina Dinucleotídeo , Filogenia , Entamoeba histolytica/genética , Entamoeba histolytica/enzimologia , Entamoeba histolytica/efeitos dos fármacos , Flavina-Adenina Dinucleotídeo/metabolismo , Archaea/genética , Archaea/enzimologia , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Cinética , Antiprotozoários/farmacologia , Humanos , Nucleotidiltransferases
8.
Biochim Biophys Acta Rev Cancer ; 1879(5): 189149, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38971209

RESUMO

Flavins and their associated proteins have recently emerged as compelling players in the landscape of cancer biology. Flavins, encompassing flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), serve as coenzymes in a multitude of cellular processes, such as metabolism, apoptosis, and cell proliferation. Their involvement in oxidative phosphorylation, redox homeostasis, and enzymatic reactions has long been recognized. However, recent research has unveiled an extended role for flavins in the context of cancer. In parallel, riboflavin transporters (RFVTs), FAD synthase (FADS), and riboflavin kinase (RFK) have gained prominence in cancer research. These proteins, responsible for riboflavin uptake, FAD biosynthesis, and FMN generation, are integral components of the cellular machinery that governs flavin homeostasis. Dysregulation in the expression/function of these proteins has been associated with various cancers, underscoring their potential as diagnostic markers, therapeutic targets, and key determinants of cancer cell behavior. This review embarks on a comprehensive exploration of the multifaceted role of flavins and of the flavoproteins involved in nucleus-mitochondria crosstalk in cancer. We journey through the influence of flavins on cancer cell energetics, the modulation of RFVTs in malignant transformation, the diagnostic and prognostic significance of FADS, and the implications of RFK in drug resistance and apoptosis. This review also underscores the potential of these molecules and processes as targets for novel diagnostic and therapeutic strategies, offering new avenues for the battle against this relentless disease.


Assuntos
Flavinas , Homeostase , Neoplasias , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Flavinas/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Animais , Apoptose , Fosfotransferases (Aceptor do Grupo Álcool)
9.
Mol Metab ; 87: 101981, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38971403

RESUMO

OBJECTIVE: The metabolism of different cells within the same microenvironment can differ and dictate physiological or pathological adaptions. Current single-cell analysis methods of metabolism are not label-free. METHODS: The study introduces a label-free, live-cell analysis method assessing endogenous fluorescence of NAD(P)H and FAD in surface-stained cells by flow cytometry. RESULTS: OxPhos inhibition, mitochondrial uncoupling, glucose exposure, genetic inactivation of glucose uptake and mitochondrial respiration alter the optical redox ratios of FAD and NAD(P)H as measured by flow cytometry. Those alterations correlate strongly with measurements obtained by extracellular flux analysis. Consequently, metabolically distinct live B-cell populations can be resolved, showing that human memory B-cells from peripheral blood exhibit a higher glycolytic flexibility than naïve B cells. Moreover, the comparison of blood-derived B- and T-lymphocytes from healthy donors and rheumatoid arthritis patients unleashes rheumatoid arthritis-associated metabolic traits in human naïve and memory B-lymphocytes. CONCLUSIONS: Taken together, these data show that the optical redox ratio can depict metabolic differences in distinct cell populations by flow cytometry.


Assuntos
Linfócitos B , Flavina-Adenina Dinucleotídeo , Citometria de Fluxo , NAD , Análise de Célula Única , Humanos , Citometria de Fluxo/métodos , NAD/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Análise de Célula Única/métodos , Linfócitos B/metabolismo , Mitocôndrias/metabolismo , Linfócitos T/metabolismo , Oxirredução , Fluorescência , Artrite Reumatoide/metabolismo , Glicólise , Fosforilação Oxidativa , Feminino , Masculino , Glucose/metabolismo
10.
Int J Biol Macromol ; 275(Pt 2): 133721, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38986972

RESUMO

Flavin reductases play a vital role in catalyzing the reduction of flavin through NADH or NADPH oxidation. The gene encoding flavin reductase from the thermophilic bacterium Geobacillus mahadii Geo-05 (GMHpaC) was cloned, overexpressed in Escherichia coli BL21 (DE3) pLysS, and purified to homogeneity. The purified recombinant GMHpaC (Class II) contains chromogenic cofactors, evidenced by maximal absorbance peaks at 370 nm and 460 nm. GMHpaC stands out as the most thermostable and pH-tolerant flavin reductase reported to date, retaining up to 95 % catalytic activity after incubation at 70 °C for 30 min and maintaining over 80 % activity within a pH range of 2-12 for 30 min. Furthermore, GMHpaC's catalytic activity increases by 52 % with FMN as a co-factor compared to FAD and riboflavin. GMHpaC, coupled with 4-hydroxyphenylacetate-3-monooxygenase (GMHpaB) from G. mahadii Geo-05, enhances the hydroxylation of 4-hydroxyphenylacetate (HPA) by 85 %. The modeled structure of GMHpaC reveals relatively conserved flavin and NADH binding sites. Modeling and docking studies shed light on structural features and amino acid substitutions that determine GMHpaC's co-factor specificity. The remarkable thermostability, high catalytic activity, and general stability exhibited by GMHpaC position it as a promising enzyme candidate for various industrial applications.


Assuntos
Estabilidade Enzimática , FMN Redutase , Geobacillus , Geobacillus/enzimologia , Geobacillus/genética , FMN Redutase/genética , FMN Redutase/metabolismo , FMN Redutase/química , Clonagem Molecular , Concentração de Íons de Hidrogênio , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/isolamento & purificação , Sequência de Aminoácidos , Cinética , Simulação de Acoplamento Molecular , Temperatura , Mononucleotídeo de Flavina/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Modelos Moleculares , Sítios de Ligação , Escherichia coli/genética , Oxigenases de Função Mista
11.
Opt Lett ; 49(14): 4054-4057, 2024 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-39008774

RESUMO

Two-photon autofluorescence (TPAF) imaging is able to offer precise cellular metabolic information with high spatiotemporal resolution, making it a promising biopsy tool. The technique is greatly hampered by the complexity of either the optical system or data processing. Here, the excitation wavelength was optimized to simultaneously excite both flavin adenine dinucleotide and nicotinamide adenine dinucleotide and eliminate the unexpected TPAF. The optical redox ratio (ORR) images were robustly achieved without additional calibration under the optimized single-wavelength excitation. The in vitro, ex vivo, and in vivo biopsy by the TPAF method were systematically studied and compared using hepato-cellular carcinoma and metastasis as examples. It was demonstrated that the proposed TPAF method simplified the optical system, improved the robustness of ORR, and enabled early-stage cancer diagnosis, showing distinguished advantages as compared with previous methods.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Imagem Óptica , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/patologia , Carcinoma Hepatocelular/diagnóstico por imagem , Carcinoma Hepatocelular/patologia , Imagem Óptica/métodos , Humanos , Animais , Metástase Neoplásica , Biópsia , Camundongos , NAD/metabolismo , Fótons , Flavina-Adenina Dinucleotídeo/metabolismo , Linhagem Celular Tumoral
12.
Microbiol Spectr ; 12(8): e0320723, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-38916330

RESUMO

Riboflavin (vitamin B2) is the precursor of the flavin coenzymes, FAD and FMN, which play a central role in cellular redox metabolism. While humans must obtain riboflavin from dietary sources, certain microbes, including Mycobacterium tuberculosis (Mtb), can biosynthesize riboflavin de novo. Riboflavin precursors have also been implicated in the activation of mucosal-associated invariant T (MAIT) cells which recognize metabolites derived from the riboflavin biosynthesis pathway complexed to the MHC-I-like molecule, MR1. To investigate the biosynthesis and function of riboflavin and its pathway intermediates in mycobacterial metabolism and physiology, we constructed conditional knockdowns (hypomorphs) in riboflavin biosynthesis and utilization genes in Mycobacterium smegmatis (Msm) and Mtb by inducible CRISPR interference. Using this comprehensive panel of hypomorphs, we analyzed the impact of gene silencing on viability, on the transcription of (other) riboflavin pathway genes, on the levels of the pathway proteins, and on riboflavin itself. Our results revealed that (i) despite lacking a canonical transporter, both Msm and Mtb assimilate exogenous riboflavin when supplied at high concentration; (ii) there is functional redundancy in lumazine synthase activity in Msm; (iii) silencing of ribA2 or ribF is profoundly bactericidal in Mtb; and (iv) in Msm, ribA2 silencing results in concomitant knockdown of other pathway genes coupled with RibA2 and riboflavin depletion and is also bactericidal. In addition to their use in genetic validation of potential drug targets for tuberculosis, this collection of hypomorphs provides a useful resource for future studies investigating the role of pathway intermediates in MAIT cell recognition of mycobacteria. IMPORTANCE: The pathway for biosynthesis and utilization of riboflavin, precursor of the essential coenzymes, FMN and FAD, is of particular interest in the flavin-rich pathogen, Mycobacterium tuberculosis (Mtb), for two important reasons: (i) the pathway includes potential tuberculosis (TB) drug targets and (ii) intermediates from the riboflavin biosynthesis pathway provide ligands for mucosal-associated invariant T (MAIT) cells, which have been implicated in TB pathogenesis. However, the riboflavin pathway is poorly understood in mycobacteria, which lack canonical mechanisms to transport this vitamin and to regulate flavin coenzyme homeostasis. By conditionally disrupting each step of the pathway and assessing the impact on mycobacterial viability and on the levels of the pathway proteins as well as riboflavin, our work provides genetic validation of the riboflavin pathway as a target for TB drug discovery and offers a resource for further exploring the association between riboflavin biosynthesis, MAIT cell activation, and TB infection and disease.


Assuntos
Mycobacterium smegmatis , Mycobacterium tuberculosis , Riboflavina , Riboflavina/biossíntese , Riboflavina/metabolismo , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium smegmatis/metabolismo , Mycobacterium smegmatis/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Humanos , Flavina-Adenina Dinucleotídeo/metabolismo , Vias Biossintéticas/genética , Técnicas de Silenciamento de Genes , Células T Invariantes Associadas à Mucosa/metabolismo , Regulação Bacteriana da Expressão Gênica
13.
J Chem Inf Model ; 64(12): 4773-4780, 2024 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-38837697

RESUMO

Lysine-specific demethylase 1 (LSD1), a highly sophisticated epigenetic regulator, orchestrates a range of critical cellular processes, holding promising therapeutic potential for treating diverse diseases. However, the clinical research progress targeting LSD1 is very slow. After 20 years of research, only one small-molecule drug, BEA-17, targeting the degradation of LSD1 and CoREST has been approved by the U.S. Food and Drug Administration. The primary reason for this may be the lack of abundant structural data regarding its intricate functions. To gain a deeper understanding of its conformational dynamics and guide the drug design process, we conducted molecular dynamics simulations to explore the conformational states of LSD1 in the apo state and under the influence of cofactors of flavin adenine dinucleotide (FAD) and CoREST. Our results showed that, across all states, the substrate binding pocket exhibited high flexibility, whereas the FAD binding pocket remained more stable. These distinct dynamical properties are essential for LSD1's ability to bind various substrates while maintaining efficient demethylation activity. Both pockets can be enlarged by merging with adjacent pockets, although only the substrate binding pocket can shrink into smaller pockets. These new pocket shapes can inform inhibitor design, particularly for selectively FAD-competitive inhibitors of LSD1, given the presence of numerous FAD-dependent enzymes in the human body. More interestingly, in the absence of FAD binding, the united substrate and FAD binding pocket are partitioned by the conserved residue of Tyr761, offering valuable insights for the design of inhibitors that disrupt the crucial steric role of Tyr761 and the redox role of FAD. Additionally, we identified pockets that positively or negatively correlate with the substrate and FAD binding pockets, which can be exploited for the design of allosteric or concurrent inhibitors. Our results reveal the intricate dynamical properties of LSD1 as well as multiple novel conformational states, which deepen our understanding of its sophisticated functions and aid in the rational design of new inhibitors.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos , Flavina-Adenina Dinucleotídeo , Histona Desmetilases , Simulação de Dinâmica Molecular , Histona Desmetilases/antagonistas & inibidores , Histona Desmetilases/metabolismo , Histona Desmetilases/química , Flavina-Adenina Dinucleotídeo/metabolismo , Flavina-Adenina Dinucleotídeo/química , Sítios de Ligação , Humanos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Especificidade por Substrato , Conformação Proteica , Ligação Proteica
14.
Nat Commun ; 15(1): 5167, 2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38886362

RESUMO

Methylenetetrahydrofolate reductase (MTHFR) is a pivotal flavoprotein connecting the folate and methionine methyl cycles, catalyzing the conversion of methylenetetrahydrofolate to methyltetrahydrofolate. Human MTHFR (hMTHFR) undergoes elaborate allosteric regulation involving protein phosphorylation and S-adenosylmethionine (AdoMet)-dependent inhibition, though other factors such as subunit orientation and FAD status remain understudied due to the lack of a functional structural model. Here, we report crystal structures of Chaetomium thermophilum MTHFR (cMTHFR) in both active (R) and inhibited (T) states. We reveal FAD occlusion by Tyr361 in the T-state, which prevents substrate interaction. Remarkably, the inhibited form of cMTHFR accommodates two AdoMet molecules per subunit. In addition, we conducted a detailed investigation of the phosphorylation sites in hMTHFR, three of which were previously unidentified. Based on the structural framework provided by our cMTHFR model, we propose a possible mechanism to explain the allosteric structural transition of MTHFR, including the impact of phosphorylation on AdoMet-dependent inhibition.


Assuntos
Chaetomium , Metilenotetra-Hidrofolato Redutase (NADPH2) , S-Adenosilmetionina , Metilenotetra-Hidrofolato Redutase (NADPH2)/metabolismo , Metilenotetra-Hidrofolato Redutase (NADPH2)/química , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , S-Adenosilmetionina/metabolismo , S-Adenosilmetionina/química , Regulação Alostérica , Chaetomium/enzimologia , Chaetomium/metabolismo , Chaetomium/genética , Fosforilação , Humanos , Cristalografia por Raios X , Modelos Moleculares , Flavina-Adenina Dinucleotídeo/metabolismo , Flavina-Adenina Dinucleotídeo/química
15.
BMC Infect Dis ; 24(1): 636, 2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-38918706

RESUMO

BACKGROUND: Schistosomiasis is a parasitic disease caused by trematodes of the genus Schistosoma. The intravascular worms acquire the nutrients necessary for their survival from host blood. Since all animals are auxotrophic for riboflavin (vitamin B2), schistosomes too must import it to survive. Riboflavin is an essential component of the coenzymes flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD); these support key functions of dozens of flavoenzymes. METHODS: Here, using a combination of metabolomics, enzyme kinetics and in silico molecular analysis, we focus on the biochemistry of riboflavin and its metabolites in Schistosoma mansoni (Sm). RESULTS: We show that when schistosomes are incubated in murine plasma, levels of FAD decrease over time while levels of FMN increase. We show that live schistosomes cleave exogenous FAD to generate FMN and this ability is significantly blocked when expression of the surface nucleotide pyrophosphatase/phosphodiesterase ectoenzyme SmNPP5 is suppressed using RNAi. Recombinant SmNPP5 cleaves FAD with a Km of 178 ± 5.9 µM and Kcat/Km of 324,734 ± 36,347 M- 1.S- 1. The FAD-dependent enzyme IL-4I1 drives the oxidative deamination of phenylalanine to produce phenylpyruvate and H2O2. Since schistosomes are damaged by H2O2, we determined if SmNPP5 could impede H2O2 production by blocking IL-4I1 action in vitro. We found that this was not the case; covalently bound FAD on IL-4I1 appears inaccessible to SmNPP5. We also report that live schistosomes can cleave exogenous FMN to generate riboflavin and this ability is significantly impeded when expression of a second surface ectoenzyme (alkaline phosphatase, SmAP) is suppressed. Recombinant SmAP cleaves FMN with a Km of 3.82 ± 0.58 mM and Kcat/Km of 1393 ± 347 M- 1.S- 1. CONCLUSIONS: The sequential hydrolysis of FAD by tegumental ecto-enzymes SmNPP5 and SmAP can generate free vitamin B2 around the worms from where it can be conveniently imported by the recently described schistosome riboflavin transporter SmaRT. Finally, we identified in silico schistosome homologs of enzymes that are involved in intracellular vitamin B2 metabolism. These are riboflavin kinase (SmRFK) as well as FAD synthase (SmFADS); cDNAs encoding these two enzymes were cloned and sequenced. SmRFK is predicted to convert riboflavin to FMN while SmFADS could further act on FMN to regenerate FAD in order to facilitate robust vitamin B2-dependent metabolism in schistosomes.


Assuntos
Mononucleotídeo de Flavina , Flavina-Adenina Dinucleotídeo , Riboflavina , Schistosoma mansoni , Riboflavina/metabolismo , Mononucleotídeo de Flavina/metabolismo , Animais , Flavina-Adenina Dinucleotídeo/metabolismo , Schistosoma mansoni/metabolismo , Schistosoma mansoni/genética , Camundongos , Humanos , Esquistossomose mansoni/parasitologia , Esquistossomose mansoni/metabolismo
16.
J Phys Chem B ; 128(26): 6283-6290, 2024 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-38913544

RESUMO

Cryptochromes are essential flavoproteins for circadian rhythms and avian magnetoreception. Flavin adenine dinucleotide (FAD), a chromophore within cryptochromes, absorbs blue light, initiating electron transfer processes that lead to a biological signaling cascade. A key step in this cascade is the formation of the FAD semiquinone radical (FADH•), characterized through a specific red-light absorption. The absorption spectra of FADH• in cryptochromes are, however, significantly different from those recorded for the cofactor in solution, primarily due to protein-induced shifts in the absorption peaks. This study employs a multiscale approach, combining molecular dynamics (MD) simulations with quantum mechanical/molecular mechanical (QM/MM) methodologies, to investigate the influence of protein dynamics on embedded FADH• absorption. We emphasize the role of the protein's polarizable environment in the shaping of the absorption spectrum, crucial for accurate spectral predictions in cryptochromes. Our findings provide valuable insights into the absorption process, advancing our understanding of cryptochrome functioning.


Assuntos
Arabidopsis , Criptocromos , Flavina-Adenina Dinucleotídeo , Simulação de Dinâmica Molecular , Teoria Quântica , Criptocromos/química , Criptocromos/metabolismo , Arabidopsis/metabolismo , Arabidopsis/química , Flavina-Adenina Dinucleotídeo/química , Flavina-Adenina Dinucleotídeo/metabolismo , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/metabolismo
17.
Chemistry ; 30(48): e202402055, 2024 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-38884181

RESUMO

Enzymes play a fundamental role in cellular metabolism. A wide range of enzymes require the presence of complementary coenzymes and cofactors to function properly. While coenzymes are believed to have been part of the last universal ancestor (LUCA) or have been present even earlier, the syntheses of crucial coenzymes like the redox-active coenzymes flavin adenine dinucleotide (FAD) or nicotinamide adenine dinucleotide (NAD+) remain challenging. Here, we present a pathway to NAD+ under prebiotic conditions starting with ammonia, cyanoacetaldehyde, prop-2-ynal and sugar-forming precursors, yielding in situ the nicotinamide riboside. Regioselective phosphorylation and water stable light activated adenosine monophosphate derivatives allow for topographically and irradiation-controlled formation of NAD+. Our findings indicate that NAD+, a coenzyme vital to life, can be formed non-enzymatically from simple organic feedstock molecules via photocatalytic activation under prebiotically plausible early Earth conditions in a continuous process under aqueous conditions.


Assuntos
NAD , NAD/química , NAD/metabolismo , Amônia/química , Niacinamida/química , Niacinamida/análogos & derivados , Fosforilação , Prebióticos , Monofosfato de Adenosina/química , Catálise , Acetaldeído/química , Oxirredução , Água/química , Compostos de Piridínio/química , Flavina-Adenina Dinucleotídeo/química , Flavina-Adenina Dinucleotídeo/metabolismo
18.
J Biomed Opt ; 29(Suppl 2): S22709, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38881557

RESUMO

Significance: To enable non-destructive longitudinal assessment of drug agents in intact tumor tissue without the use of disruptive probes, we have designed a label-free method to quantify the health of individual tumor cells in excised tumor tissue using multiphoton fluorescence lifetime imaging microscopy (MP-FLIM). Aim: Using murine tumor fragments which preserve the native tumor microenvironment, we seek to demonstrate signals generated by the intrinsically fluorescent metabolic co-factors nicotinamide adenine dinucleotide phosphate [NAD(P)H] and flavin adenine dinucleotide (FAD) correlate with irreversible cascades leading to cell death. Approach: We use MP-FLIM of NAD(P)H and FAD on tissues and confirm viability using standard apoptosis and live/dead (Caspase 3/7 and propidium iodide, respectively) assays. Results: Through a statistical approach, reproducible shifts in FLIM data, determined through phasor analysis, are shown to correlate with loss of cell viability. With this, we demonstrate that cell death achieved through either apoptosis/necrosis or necroptosis can be discriminated. In addition, specific responses to common chemotherapeutic treatment inducing cell death were detected. Conclusions: These data demonstrate that MP-FLIM can detect and quantify cell viability without the use of potentially toxic dyes, thus enabling longitudinal multi-day studies assessing the effects of therapeutic agents on tumor fragments.


Assuntos
Sobrevivência Celular , Microscopia de Fluorescência por Excitação Multifotônica , Animais , Camundongos , Sobrevivência Celular/efeitos dos fármacos , Microscopia de Fluorescência por Excitação Multifotônica/métodos , Apoptose , Flavina-Adenina Dinucleotídeo/química , NADP/metabolismo , Linhagem Celular Tumoral , Imagem Óptica/métodos
19.
Biochemistry ; 63(10): 1347-1358, 2024 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-38691339

RESUMO

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.


Assuntos
Di-Hidro-Orotato Desidrogenase , Lactococcus lactis , Biocatálise , Catálise , Domínio Catalítico , Mononucleotídeo de Flavina/metabolismo , Mononucleotídeo de Flavina/química , Flavina-Adenina Dinucleotídeo/metabolismo , Flavina-Adenina Dinucleotídeo/química , Cinética , Lactococcus lactis/enzimologia , Lactococcus lactis/metabolismo , NAD/metabolismo , NAD/química , Oxirredução
20.
Nat Commun ; 15(1): 3994, 2024 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-38734761

RESUMO

NADPH oxidase 5 (NOX5) catalyzes the production of superoxide free radicals and regulates physiological processes from sperm motility to cardiac rhythm. Overexpression of NOX5 leads to cancers, diabetes, and cardiovascular diseases. NOX5 is activated by intracellular calcium signaling, but the underlying molecular mechanism of which - in particular, how calcium triggers electron transfer from NADPH to FAD - is still unclear. Here we capture motions of full-length human NOX5 upon calcium binding using single-particle cryogenic electron microscopy (cryo-EM). By combining biochemistry, mutagenesis analyses, and molecular dynamics (MD) simulations, we decode the molecular basis of NOX5 activation and electron transfer. We find that calcium binding to the EF-hand domain increases NADPH dynamics, permitting electron transfer between NADPH and FAD and superoxide production. Our structural findings also uncover a zinc-binding motif that is important for NOX5 stability and enzymatic activity, revealing modulation mechanisms of reactive oxygen species (ROS) production.


Assuntos
Cálcio , NADPH Oxidase 5 , NADP , Humanos , Sítios de Ligação , Cálcio/metabolismo , Microscopia Crioeletrônica , Transporte de Elétrons , Ativação Enzimática , Flavina-Adenina Dinucleotídeo/metabolismo , Simulação de Dinâmica Molecular , NADP/metabolismo , NADPH Oxidase 5/metabolismo , NADPH Oxidase 5/genética , NADPH Oxidase 5/química , Ligação Proteica , Espécies Reativas de Oxigênio/metabolismo , Superóxidos/metabolismo , Zinco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA