Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Immunol ; 24(1): 34, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37752417

RESUMEN

BACKGROUND: Rapid and accurate diagnosis of individuals with SARS-CoV-2 infection is an effective way to prevent and control the spread of COVID-19. Although the detection of SARS-CoV-2 viral RNA by RT-qPCR is the gold standard for COVID-19 testing, the use of antigen-detecting rapid diagnostic tests (Ag-RDTs) is emerging as a complementary surveillance tool as Omicron case numbers skyrocket worldwide. However, the results from Ag-RDTs are less accurate in individuals with low viral loads. RESULTS: To develop a highly sensitive and accurate Ag-RDT, 90 monoclonal antibodies were raised from guinea pigs immunized with SARS CoV-2 nucleocapsid protein (CoV-2-NP). By applying a capture antibody recognizing the structural epitope of the N-terminal domain of CoV-2-NP and a detection antibody recognizing the C-terminal tail of CoV-2-NP to an automated chemiluminescence flow-through membrane immunoassay device, we developed a novel Ag-RDT, CoV-2-POCube. The CoV-2-POCube exclusively recognizes CoV-2-NP variants but not the nucleocapsid proteins of other human coronaviruses. The CoV-2-POCube achieved a limit of detection sensitivity of 0.20 ~ 0.66 pg/mL of CoV-2-NPs, demonstrating more than 100 times greater sensitivity than commercially available SARS-CoV-2 Ag-RDTs. CONCLUSIONS: CoV-2-POCube has high analytical sensitivity and can detect SARS-CoV-2 variants in 15 min without observing the high-dose hook effect, thus meeting the need for early SARS-CoV-2 diagnosis with lower viral load. CoV-2-POCube is a promising alternative to currently available diagnostic devices for faster clinical decision making in individuals with suspected COVID-19 in resource-limited settings.


Asunto(s)
Anticuerpos Monoclonales , COVID-19 , Humanos , Animales , Cobayas , SARS-CoV-2 , Prueba de COVID-19 , COVID-19/diagnóstico , Sensibilidad y Especificidad , Inmunoensayo
2.
J Biol Chem ; 299(8): 105001, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37394006

RESUMEN

NADH-ubiquinone (UQ) oxidoreductase (complex I) couples electron transfer from NADH to UQ with proton translocation in its membrane part. The UQ reduction step is key to triggering proton translocation. Structural studies have identified a long, narrow, tunnel-like cavity within complex I, through which UQ may access a deep reaction site. To elucidate the physiological relevance of this UQ-accessing tunnel, we previously investigated whether a series of oversized UQs (OS-UQs), whose tail moiety is too large to enter and transit the narrow tunnel, can be catalytically reduced by complex I using the native enzyme in bovine heart submitochondrial particles (SMPs) and the isolated enzyme reconstituted into liposomes. Nevertheless, the physiological relevance remained unclear because some amphiphilic OS-UQs were reduced in SMPs but not in proteoliposomes, and investigation of extremely hydrophobic OS-UQs was not possible in SMPs. To uniformly assess the electron transfer activities of all OS-UQs with the native complex I, here we present a new assay system using SMPs, which were fused with liposomes incorporating OS-UQ and supplemented with a parasitic quinol oxidase to recycle reduced OS-UQ. In this system, all OS-UQs tested were reduced by the native enzyme, and the reduction was coupled with proton translocation. This finding does not support the canonical tunnel model. We propose that the UQ reaction cavity is flexibly open in the native enzyme to allow OS-UQs to access the reaction site, but their access is obstructed in the isolated enzyme as the cavity is altered by detergent-solubilizing from the mitochondrial membrane.


Asunto(s)
Complejo I de Transporte de Electrón , Ubiquinona , Animales , Bovinos , Ubiquinona/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Membranas Mitocondriales/metabolismo , NAD/metabolismo , Protones , Liposomas
3.
ACS Med Chem Lett ; 14(2): 211-216, 2023 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-36793437

RESUMEN

Mitochondrial oxidative phosphorylation (OXPHOS) is an essential cellular metabolic process that generates ATP. The enzymes involved in OXPHOS are considered to be promising druggable targets. Through screening of an in-house synthetic library with bovine heart submitochondrial particles, we identified a unique symmetric bis-sulfonamide, KPYC01112 (1) as an inhibitor targeting NADH-quinone oxidoreductase (complex I). Structural modifications of KPYC01112 (1) led to the discovery of the more potent inhibitors 32 and 35 possessing long alkyl chains (IC50 = 0.017 and 0.014 µM, respectively). A photoaffinity labeling experiment using a newly synthesized photoreactive bis-sulfonamide ([125I]-43) revealed that it binds to the 49-kDa, PSST, and ND1 subunits which make up the quinone-accessing cavity of complex I.

4.
Nat Commun ; 13(1): 4082, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35882843

RESUMEN

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.


Asunto(s)
Quinona Reductasas , Vibrio cholerae , Proteínas Bacterianas/metabolismo , Microscopía por Crioelectrón , Complejo I de Transporte de Electrón/metabolismo , Oxidación-Reducción , Quinona Reductasas/metabolismo , Sodio/metabolismo , Vibrio cholerae/metabolismo
5.
J Biol Chem ; 298(7): 102075, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35643318

RESUMEN

The ubiquinone (UQ) reduction step catalyzed by NADH-UQ oxidoreductase (mitochondrial respiratory complex I) is key to triggering proton translocation across the inner mitochondrial membrane. Structural studies have identified a long, narrow, UQ-accessing tunnel within the enzyme. We previously demonstrated that synthetic oversized UQs, which are unlikely to transit this narrow tunnel, are catalytically reduced by native complex I embedded in submitochondrial particles but not by the isolated enzyme. To explain this contradiction, we hypothesized that access of oversized UQs to the reaction site is obstructed in the isolated enzyme because their access route is altered following detergent solubilization from the inner mitochondrial membrane. In the present study, we investigated this using two pairs of photoreactive UQs (pUQm-1/pUQp-1 and pUQm-2/pUQp-2), with each pair having the same chemical properties except for a ∼1.0 Å difference in side-chain widths. Despite this subtle difference, reduction of the wider pUQs by the isolated complex was significantly slower than of the narrower pUQs, but both were similarly reduced by the native enzyme. In addition, photoaffinity-labeling experiments using the four [125I]pUQs demonstrated that their side chains predominantly label the ND1 subunit with both enzymes but at different regions around the tunnel. Finally, we show that the suppressive effects of different types of inhibitors on the labeling significantly changed depending on [125I]pUQs used, indicating that [125I]pUQs and these inhibitors do not necessarily share a common binding cavity. Altogether, we conclude that the reaction behaviors of pUQs cannot be simply explained by the canonical UQ tunnel model.


Asunto(s)
Complejo I de Transporte de Electrón , Ubiquinona , Sitios de Unión , Complejo I de Transporte de Electrón/metabolismo , Mitocondrias/metabolismo , Partículas Submitocóndricas/metabolismo , Ubiquinona/metabolismo
6.
Biochim Biophys Acta Bioenerg ; 1863(5): 148547, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35337841

RESUMEN

The Na+-pumping NADH-ubiquinone (UQ) oxidoreductase (Na+-NQR) is an essential bacterial respiratory enzyme that generates a Na+ gradient across the cell membrane. However, the mechanism that couples the redox reactions to Na+ translocation remains unknown. To address this, we examined the relation between reduction of UQ and Na+ translocation using a series of synthetic UQs with Vibrio cholerae Na+-NQR reconstituted into liposomes. UQ0 that has no side chain and UQCH3 and UQC2H5, which have methyl and ethyl side chains, respectively, were catalytically reduced by Na+-NQR, but their reduction generated no membrane potential, indicating that the overall electron transfer and Na+ translocation are not coupled. While these UQs were partly reduced by electron leak from the cofactor(s) located upstream of riboflavin, this complete loss of Na+ translocation cannot be explained by the electron leak. Lengthening the UQ side chain to n-propyl (C3H7) or longer significantly restored Na+ translocation. It has been considered that Na+ translocation is completed when riboflavin, a terminal redox cofactor residing within the membrane, is reduced. In this view, the role of UQ is simply to accept electrons from the reduced riboflavin to regenerate the stable neutral riboflavin radical and reset the catalytic cycle. However, the present study revealed that the final UQ reduction via reduced riboflavin makes an important contribution to Na+ translocation through a critical role of its side chain. Based on the results, we discuss the critical role of the UQ side chain in Na+ translocation.


Asunto(s)
Vibrio cholerae , Complejo I de Transporte de Electrón/metabolismo , Riboflavina/metabolismo , Sodio/metabolismo , Ubiquinona/metabolismo
7.
Biochim Biophys Acta Bioenerg ; 1863(2): 148520, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34896079

RESUMEN

Tamoxifen has been widely used in the treatment of estrogen receptor (ER)-positive breast cancer, whereas it also exhibits ER-independent anticancer effects in various cancer cell types. As one of the convincing mechanisms underlying the ER-independent effects, induction of apoptosis through mitochondrial dysfunction has been advocated. However, the mechanism of action of tamoxifen even at the isolated mitochondrial level is not fully understood and remains controversial. Here, we attempted to comprehensively understand tamoxifen's multiple actions in isolated rat liver mitochondria through not only revisiting the actions hitherto reported but also conducting originally designed experiments. Using submitochondrial particles, we found that tamoxifen has potential as an inhibitor of both respiratory complex I and ATP synthase. However, these inhibitory effects were not elicited in intact mitochondria, likely because penetration of tamoxifen across the inner mitochondrial membrane is highly restricted owing to its localized positive charge (-N+H(CH3)2). This restricted penetration may also explain why tamoxifen is unable to function as a protonophore-type uncoupler in mitochondria. Moreover, tamoxifen suppressed opening of the mitochondrial permeability transition pore induced by Ca2+ overload through enhancing phosphate uptake into the matrix. The photoaffinity labeling experiments using a photolabile tamoxifen derivative (pTAM1) indicated that pTAM1 specifically binds to voltage-dependent anion channels (VDACs) 1 and 3, which regulate transport of various substances into mitochondria. The binding of tamoxifen to VDAC1 and/or VDAC3 could be responsible for the enhancement of phosphate uptake. Taking all the results together, we consider the principal impairment of mitochondrial functions caused by tamoxifen.


Asunto(s)
Tamoxifeno
8.
Biosci Biotechnol Biochem ; 85(12): 2368-2377, 2021 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-34625801

RESUMEN

The mitochondrial machineries presiding over ATP synthesis via oxidative phosphorylation are promising druggable targets. Fusaramin, a 3-acyl tetramic acid isolated from Fusarium concentricum FKI-7550, is an inhibitor of oxidative phosphorylation in Saccharomyces cerevisiae mitochondria, although its target has yet to be identified. Fusaramin significantly interfered with [3H]ADP uptake by yeast mitochondria at the concentration range inhibiting oxidative phosphorylation. A photoreactive fusaramin derivative (pFS-5) specifically labeled voltage-dependent anion channel 1 (VDAC1), which facilitates trafficking of ADP/ATP across the outer mitochondrial membrane. These results strongly suggest that the inhibition of oxidative phosphorylation by fusaramin is predominantly attributable to the impairment of VDAC1 functions. Fusaramin also inhibited FoF1-ATP synthase and ubiquinol-cytochrome c oxidoreductase (complex III) at concentrations higher than those required for the VDAC inhibition. Considering that other tetramic acid derivatives are reported to inhibit FoF1-ATP synthase and complex III, natural tetramic acids were found to elicit multiple inhibitory actions against mitochondrial machineries.


Asunto(s)
Fosforilación Oxidativa
9.
J Biol Chem ; 297(5): 101173, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34499926

RESUMEN

Bisphenol A and its derivatives are recognized as endocrine disruptors based on their complex effects on estrogen receptor (ER) signaling. While the effects of bisphenol derivatives on ERα have been thoroughly evaluated, how these chemicals affect ERß signaling is less well understood. Herein, we sought to identify novel ERß ligands using a radioligand competitive binding assay to screen a chemical library of bisphenol derivatives. Many of the compounds identified showed intriguing dual activities as both ERα agonists and ERß antagonists. Docking simulations of these compounds and ERß suggested that they bound not only to the canonical binding site of ERß but also to the coactivator binding site located on the surface of the receptor, suggesting that they act as coactivator-binding inhibitors (CBIs). Receptor-ligand binding experiments using WT and mutated ERß support the presence of a second ligand-interaction position at the coactivator-binding site in ERß, and direct binding experiments of ERß and a coactivator peptide confirmed that these compounds act as CBIs. Our study is the first to propose that bisphenol derivatives act as CBIs, presenting critical insight for the future development of ER signaling-based drugs and their potential to function as endocrine disruptors.


Asunto(s)
Compuestos de Bencidrilo , Receptor beta de Estrógeno , Fenoles , Transducción de Señal/efectos de los fármacos , Compuestos de Bencidrilo/química , Compuestos de Bencidrilo/farmacología , Receptor beta de Estrógeno/química , Receptor beta de Estrógeno/genética , Receptor beta de Estrógeno/metabolismo , Células HeLa , Humanos , Mutación , Fenoles/química , Fenoles/farmacología , Unión Proteica , Transducción de Señal/genética
10.
Biochim Biophys Acta Bioenerg ; 1862(8): 148432, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33932367

RESUMEN

The Na+-pumping NADH-ubiquinone oxidoreductase (Na+-NQR) is a main ion transporter in many pathogenic bacteria. We previously proposed that N-terminal stretch of the NqrB subunit plays an important role in regulating the ubiquinone reaction at the adjacent NqrA subunit in Vibrio cholerae Na+-NQR. However, since approximately three quarters of the stretch (NqrB-Met1-Pro37) was not modeled in an earlier crystallographic study, its structure and function remain unknown. If we can develop a method that enables pinpoint modification of this stretch by functional chemicals (such as spin probes), it could lead to new ways to investigate the unsettled issues. As the first step to this end, we undertook to specifically attach an alkyne group to a lysine located in the stretch via protein-ligand affinity-driven substitution using synthetic ligands NAS-K1 and NAS-K2. The alkyne, once attached, can serve as an "anchor" for connecting functional chemicals via convenient click chemistry. After a short incubation of isolated Na+-NQR with these ligands, alkyne was predominantly incorporated into NqrB. Proteomic analyses in combination with mutagenesis of predicted target lysines revealed that alkyne attaches to NqrB-Lys22 located at the nonmodeled region of the stretch. This study not only achieved the specific modification initially aimed for but also provided valuable information about positioning of the nonmodeled region. For example, the fact that hydrophobic NAS-Ks come into contact with NqrB-Lys22 suggests that the nonmodeled region may orient toward the membrane phase rather than protruding into cytoplasmic medium. This conformation may be essential for regulating the ubiquinone reaction in the adjacent NqrA.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Lisina/química , Proteoma/análisis , Vibrio cholerae/enzimología , Sitios de Unión , Transporte Iónico , Conformación Proteica , Subunidades de Proteína , Sodio/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo
11.
Biochemistry ; 60(10): 813-824, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33650850

RESUMEN

The ubiquinone reduction step in NADH-ubiquinone oxidoreductase (complex I) is the key to triggering proton translocation in its membrane part. Although the existence of a long and narrow quinone-access channel has been identified, it remains debatable whether the channel model can account for binding of various ligands (ubiquinones and inhibitors) to the enzyme. We previously proposed that the matrix-side interfacial region of the 49 kDa, ND1, PSST, and 39 kDa subunits, which is covered by a loop connecting transmembrane helices (TMHs) 1 and 2 of ND3, may be the area for entry of some bulky ligands into the quinone reaction cavity. However, this proposition lacks direct evidence that the cavity is accessible from the putative matrix-side region, which allows ligands to pass. To address this, we examined whether Cys39 of ND3 and Asp160 of 49 kDa can be specifically cross-linked by bifunctional cross-linkers (tetrazine-maleimide hybrid, named TMBC). On the basis of the structural models of complex I, such dual cross-linking is unexpected because ND3 Cys39 and 49 kDa Asp160 are located on the TMH1-2 loop and deep inside the channel, respectively, and hence, they are physically separated by peptide chains forming the channel wall. However, three TMBCs with different spacer lengths did cross-link the two residues, resulting in the formation of new cross-linked ND3/49 kDa subunits. Chemical modification of either ND3 Cys39 or 49 kDa Asp160 blocked the dual cross-linking, ensuring the specificity of the cross-linking. Altogether, this study provides direct evidence that the quinone reaction cavity is indeed accessible from the proposed matrix-side region covered by the ND3 TMH1-2 loop.


Asunto(s)
Reactivos de Enlaces Cruzados/química , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Mitocondrias Cardíacas/metabolismo , Ubiquinona/metabolismo , Animales , Sitios de Unión , Dominio Catalítico , Bovinos , Transporte de Electrón , Ligandos , Conformación Proteica , Subunidades de Proteína , Protones
12.
J Antibiot (Tokyo) ; 73(10): 721-728, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32759961

RESUMEN

Basidiomycetes are known to biosynthesize many biologically interesting compounds, including terpenoids. However, they are notoriously difficult to manipulate. Previously, we identified the gene cluster encoding enzymes responsible for the biosynthesis of lagopodins, cuparene-type sesquiterpenoid quinone natural products in Coprinopsis cinerea. In this study, we attempted to increase the productivity of lagopodin B (1) and related pathway products by overexpressing the terpene cyclase gene cop6 in C. cinerea to determine the details of the complex lagopodin and hitoyol biosynthetic pathway. Random integration of the cop6 into the genome of the ku70-deficient C. cinerea strain resulted in an ~2.4-fold increase in the production of 1. However, integration of cop6 into a highly transcribed position within the chromosome we designated as an expression boost area (EBA) resulted in an ~14-fold greater production of 1. Furthermore, the EBA-integration strain allowed us to isolate a previously undetected product 2, which we determined to be the known compound, hydroxylagopodin B. This finding expanded our understanding of the lagopodin-hitoyol biosynthetic pathway and allowed us to hypothesize a possible mechanism for the biosynthesis of a related homodimeric compound, lagopodin C. Our results demonstrate the potential of targeting EBA to integrate key biosynthetic genes into the genome for enhancing the production of difficult-to-obtain compounds for studying the biosynthesis of complex secondary metabolites in basidiomycetes and other complex eukaryotic organisms.


Asunto(s)
Agaricales/metabolismo , Sesquiterpenos/metabolismo , Agaricales/genética , Regulación Fúngica de la Expresión Génica/genética , Genes Fúngicos/genética , Ingeniería Genética/métodos , Redes y Vías Metabólicas/genética , Reacción en Cadena de la Polimerasa , ARN de Hongos/genética , Análisis de Secuencia de ARN
13.
J Biol Chem ; 295(36): 12739-12754, 2020 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32690607

RESUMEN

The Na+-pumping NADH-ubiquinone (UQ) oxidoreductase (Na+-NQR) is present in the respiratory chain of many pathogenic bacteria and is thought to be a promising antibiotic target. Whereas many details of Na+-NQR structure and function are known, the mechanisms of action of potent inhibitors is not well-understood; elucidating the mechanisms would not only advance drug design strategies but might also provide insights on a terminal electron transfer from riboflavin to UQ. To this end, we performed photoaffinity labeling experiments using photoreactive derivatives of two known inhibitors, aurachin and korormicin, on isolated Vibrio cholerae Na+-NQR. The inhibitors labeled the cytoplasmic surface domain of the NqrB subunit including a protruding N-terminal stretch, which may be critical to regulate the UQ reaction in the adjacent NqrA subunit. The labeling was blocked by short-chain UQs such as ubiquinone-2. The photolabile group (2-aryl-5-carboxytetrazole (ACT)) of these inhibitors reacts with nucleophilic amino acids, so we tested mutations of nucleophilic residues in the labeled region of NqrB, such as Asp49 and Asp52 (to Ala), and observed moderate decreases in labeling yields, suggesting that these residues are involved in the interaction with ACT. We conclude that the inhibitors interfere with the UQ reaction in two ways: the first is blocking structural rearrangements at the cytoplasmic interface between NqrA and NqrB, and the second is the direct obstruction of UQ binding at this interfacial area. Unusual competitive behavior between the photoreactive inhibitors and various competitors corroborates our previous proposition that there may be two inhibitor binding sites in Na+-NQR.


Asunto(s)
Proteínas Bacterianas/metabolismo , NADH NADPH Oxidorreductasas/metabolismo , Ubiquinona/metabolismo , Vibrio cholerae/metabolismo , Proteínas Bacterianas/genética , NADH NADPH Oxidorreductasas/genética , Ubiquinona/genética , Vibrio cholerae/genética
14.
J Biol Chem ; 295(21): 7481-7491, 2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32295842

RESUMEN

The small molecule IACS-010759 has been reported to potently inhibit the proliferation of glycolysis-deficient hypoxic tumor cells by interfering with the functions of mitochondrial NADH-ubiquinone oxidoreductase (complex I) without exhibiting cytotoxicity at tolerated doses in normal cells. Considering the significant cytotoxicity of conventional quinone-site inhibitors of complex I, such as piericidin and acetogenin families, we hypothesized that the mechanism of action of IACS-010759 on complex I differs from that of other known quinone-site inhibitors. To test this possibility, here we investigated IACS-010759's mechanism in bovine heart submitochondrial particles. We found that IACS-010759, like known quinone-site inhibitors, suppresses chemical modification by the tosyl reagent AL1 of Asp160 in the 49-kDa subunit, located deep in the interior of a previously proposed quinone-access channel. However, contrary to the other inhibitors, IACS-010759 direction-dependently inhibited forward and reverse electron transfer and did not suppress binding of the quinazoline-type inhibitor [125I]AzQ to the N terminus of the 49-kDa subunit. Photoaffinity labeling experiments revealed that the photoreactive derivative [125I]IACS-010759-PD1 binds to the middle of the membrane subunit ND1 and that inhibitors that bind to the 49-kDa or PSST subunit cannot suppress the binding. We conclude that IACS-010759's binding location in complex I differs from that of any other known inhibitor of the enzyme. Our findings, along with those from previous study, reveal that the mechanisms of action of complex I inhibitors with widely different chemical properties are more diverse than can be accounted for by the quinone-access channel model proposed by structural biology studies.


Asunto(s)
Complejo I de Transporte de Electrón/antagonistas & inhibidores , Glucólisis/efectos de los fármacos , Mitocondrias Cardíacas/enzimología , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/metabolismo , Oxadiazoles/farmacología , Piperidinas/farmacología , Animales , Bovinos , Hipoxia de la Célula/efectos de los fármacos , Complejo I de Transporte de Electrón/metabolismo , Humanos , Mitocondrias Cardíacas/patología , Proteínas de Neoplasias/metabolismo , Neoplasias/patología
15.
J Biol Chem ; 295(8): 2449-2463, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-31953326

RESUMEN

NADH-quinone oxidoreductase (complex I) couples electron transfer from NADH to quinone with proton translocation across the membrane. Quinone reduction is a key step for energy transmission from the site of quinone reduction to the remotely located proton-pumping machinery of the enzyme. Although structural biology studies have proposed the existence of a long and narrow quinone-access channel, the physiological relevance of this channel remains debatable. We investigated here whether complex I in bovine heart submitochondrial particles (SMPs) can catalytically reduce a series of oversized ubiquinones (OS-UQs), which are highly unlikely to transit the narrow channel because their side chain includes a bulky "block" that is ∼13 Šacross. We found that some OS-UQs function as efficient electron acceptors from complex I, accepting electrons with an efficiency comparable with ubiquinone-2. The catalytic reduction and proton translocation coupled with this reduction were completely inhibited by different quinone-site inhibitors, indicating that the reduction of OS-UQs takes place at the physiological reaction site for ubiquinone. Notably, the proton-translocating efficiencies of OS-UQs significantly varied depending on their side-chain structures, suggesting that the reaction characteristics of OS-UQs affect the predicted structural changes of the quinone reaction site required for triggering proton translocation. These results are difficult to reconcile with the current channel model; rather, the access path for ubiquinone may be open to allow OS-UQs to access the reaction site. Nevertheless, contrary to the observations in SMPs, OS-UQs were not catalytically reduced by isolated complex I reconstituted into liposomes. We discuss possible reasons for these contradictory results.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Mitocondrias Cardíacas/metabolismo , Sondas Moleculares/metabolismo , Ubiquinona/química , Ubiquinona/metabolismo , Alquinos/metabolismo , Animales , Bovinos , Simulación por Computador , Transporte de Electrón , Potencial de la Membrana Mitocondrial , Proteínas Mitocondriales/metabolismo , Modelos Moleculares , NAD/metabolismo , Oxidorreductasas/metabolismo , Proteínas de Plantas/metabolismo , Subunidades de Proteína/metabolismo , Proteolípidos/metabolismo , Protones , Partículas Submitocóndricas/metabolismo
16.
Org Lett ; 21(18): 7582-7586, 2019 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-31496254

RESUMEN

LaeA is a positive global regulator of secondary metabolism in Ascomycetes, but its role in Basidiomycetes, including medicinal mushrooms, remains uncharacterized. Here, knockout of laeA in the model mushroom Coprinopsis cinerea unexpectedly upregulated the biosynthesis of a novel siderophore, coprinoferrin. Furthermore, knockout of the nonribosomal peptide synthetase-encoding cpf1 responsible for coprinoferrin biosynthesis resulted in growth defect and loss of fruiting body formation, indicating the complex role that this natural product plays in fungal cell development.


Asunto(s)
Agaricales/química , Ascomicetos/química , Cuerpos Fructíferos de los Hongos/química , Sideróforos/química , Agaricales/genética , Ascomicetos/metabolismo , Cuerpos Fructíferos de los Hongos/metabolismo , Estructura Molecular , Sideróforos/metabolismo
17.
Sci Rep ; 9(1): 9954, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31289329

RESUMEN

Bisphenol A (BPA) is used as an industrial raw material for polycarbonate plastics and epoxy resins; however, various concerns have been reported regarding its status as an endocrine-disrupting chemical. BPA interacts not only with oestrogen receptors (ERs) but constitutive androstane receptor, pregnane X receptor, and oestrogen-related receptor γ (ERRγ); therefore, the bisphenol structure represents a privileged structure for the nuclear-receptor superfamily. Here, we screen 127 BPA-related compounds by competitive-binding assay using [3H]oestradiol and find that 20 compounds bind to ERα with high affinity. We confirm most of these as ERα agonists; however, four compounds, including bisphenol M and bisphenol P act as novel antagonists. These structures harbour three benzene rings in tandem with terminal hydroxy groups at para-positions, with this tandem tri-ring bisphenol structure representing a novel privileged structure for an ERα antagonist. Additionally, we perform an ab initio calculation and develop a new clipping method for halogen bonding or non-covalent interaction using DV-Xα evaluation for biomolecules.


Asunto(s)
Antineoplásicos/farmacología , Compuestos de Bencidrilo/metabolismo , Receptor alfa de Estrógeno/agonistas , Receptor alfa de Estrógeno/antagonistas & inhibidores , Estrógenos no Esteroides/metabolismo , Fenoles/metabolismo , Antineoplásicos/química , Compuestos de Bencidrilo/química , Unión Competitiva , Descubrimiento de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Receptor alfa de Estrógeno/metabolismo , Estrógenos no Esteroides/química , Glutatión Transferasa/metabolismo , Células HeLa , Humanos , Luciferasas/metabolismo , Estructura Molecular , Fenoles/química
18.
Org Biomol Chem ; 17(2): 234-239, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30556075

RESUMEN

Use of the ku70-deficient strain of Coprinopsis cinerea enabled confirmation within the native context of the central role the sesquiterpene synthase Cop6 plays in lagopodin biosynthesis. Furthermore, yeast in vivo bioconversion and in vitro assays of two cytochrome P450 monooxygenases Cox1 and Cox2 allowed elucidation of the network of oxidation steps that build structural complexity onto the α-cuprenene framework during the biosynthesis of lagopodins. Three new compounds were identified as intermediates formed by the redox enzymes.


Asunto(s)
Coprinus/enzimología , Coprinus/metabolismo , Sesquiterpenos/metabolismo , Vías Biosintéticas , Coprinus/química , Sistema Enzimático del Citocromo P-450/metabolismo , Proteínas Fúngicas/metabolismo , Ligasas/metabolismo , Oxidación-Reducción , Quinonas/química , Quinonas/metabolismo , Sesquiterpenos/química
19.
Biochemistry ; 56(32): 4279-4287, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28707880

RESUMEN

We previously showed that a bulky ring-strained cycloalkyne possessing a rhodamine fluorophore directly reacts (via strain-promoted click chemistry) with the azido group incorporated (via ligand-directed tosyl chemistry) into Asp160 in the 49 kDa subunit of complex I in bovine heart submitochondrial particles [Masuya, T., et al. (2014) Biochemistry 53, 7816-7823]. This two-step conjugation may be a promising technique for specific chemical modifications of the quinone-access channel in complex I by various molecular probes, which would lead to new methodologies for studying the enzyme. However, because the reactivities of ring-strained cycloalkynes are generally high, they also react with other nucleophilic amino acids in mitochondrial proteins, resulting in significant undesired side reactions. To minimize side reactions and achieve precise pinpoint chemical modification of 49 kDa Asp160, we investigated an optimal pair of chemical tags for the two-step conjugation reaction. We found that instead of strain-promoted click chemistry, Diels-Alder cycloaddition of a pair of cyclopropene incorporated into 49 kDa Asp160 (via ligand-directed tosyl chemistry) and externally added tetrazine is more efficient for the pinpoint modification. An excess of quinone-site inhibitors did not interfere with Diels-Alder cycloaddition between the cyclopropene and tetrazine. These results along with the previous findings (cited above) strongly suggest that in contrast to the predicted quinone-access channel modeled by X-ray crystallographic and single-particle cryo-electron microscopic studies, the channel is open or undergoes large structural rearrangements to allow bulky ligands into the proximity of 49 kDa Asp160.


Asunto(s)
Complejo I de Transporte de Electrón/química , Mitocondrias Cardíacas/enzimología , Modelos Moleculares , Sondas Moleculares/química , Animales , Bovinos , Química Clic/métodos , Ciclopropanos/química
20.
Biosci Biotechnol Biochem ; 80(8): 1464-9, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27140857

RESUMEN

We previously produced the unique ubiquinone QT ("decoupling" quinone), the catalytic reduction of which in NADH-quinone oxidoreduction with bovine heart mitochondrial NADH-ubiquinone oxidoreductase (complex I) is completely decoupled from proton translocation across the membrane domain. This feature is markedly distinct from those of typical short-chain quinones such as ubiquinone-1. To further characterize the features of the QT reaction with complex I, we herein synthesized three QT analogs, QT2-QT4, and characterized their electron transfer reactions. We found that all aspects of electron transfer (e.g. electron-accepting activity and membrane potential formation) vary significantly among these analogs. The features of QT2 as decoupling quinone were slightly superior to those of original QT. Based on these results, we conclude that the bound positions of QTs within the quinone binding cavity susceptibly change depending on their side-chain structures, and the positions, in turn, govern the behavior of QTs as electron acceptors.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Mitocondrias Cardíacas/metabolismo , NAD/metabolismo , Protones , Quinonas/metabolismo , Ubiquinona/metabolismo , Animales , Sitios de Unión , Bovinos , Fraccionamiento Celular , Transporte de Electrón , Complejo I de Transporte de Electrón/química , Cinética , Potencial de la Membrana Mitocondrial , Mitocondrias Cardíacas/química , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , NAD/química , Oxidación-Reducción , Quinonas/química , Relación Estructura-Actividad , Ubiquinona/análogos & derivados , Ubiquinona/síntesis química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...