Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.534
Filtrar
1.
Microb Cell Fact ; 23(1): 234, 2024 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-39182107

RESUMEN

BACKGROUND: Several two-component systems of Streptomyces coelicolor, a model organism used for studying antibiotic production in Streptomyces, affect the expression of the bfr (SCO2113) gene that encodes a bacterioferritin, a protein involved in iron storage. In this work, we have studied the effect of the deletion mutant ∆bfr in S. coelicolor. RESULTS: The ∆bfr mutant exhibits a delay in morphological differentiation and produces a lesser amount of the two pigmented antibiotics (actinorhodin and undecylprodigiosin) compared to the wild type on complex media. The effect of iron in minimal medium was tested in the wild type and ∆bfr mutant. Consequently, we also observed different levels of production of the two pigmented antibiotics between the two strains, depending on the iron concentration and the medium (solid or liquid) used. Contrary to expectations, no differences in intracellular iron concentration were detected between the wild type and ∆bfr mutant. However, a higher level of reactive oxygen species in the ∆bfr mutant and a higher tolerance to oxidative stress were observed. Proteomic analysis showed no variation in iron response proteins, but there was a lower abundance of proteins related to actinorhodin and ribosomal proteins, as well as others related to secondary metabolite production and differentiation. Additionally, a higher abundance of proteins related to various types of stress, such as respiration and hypoxia among others, was also revealed. Data are available via ProteomeXchange with identifier PXD050869. CONCLUSION: This bacterioferritin in S. coelicolor (Bfr) is a new element in the complex regulation of secondary metabolism in S. coelicolor and, additionally, iron acts as a signal to modulate the biosynthesis of active molecules. Our model proposes an interaction between Bfr and iron-containing regulatory proteins. Thus, identifying these interactions would provide new information for improving antibiotic production in Streptomyces.


Asunto(s)
Antraquinonas , Antibacterianos , Proteínas Bacterianas , Ferritinas , Hierro , Streptomyces coelicolor , Streptomyces coelicolor/metabolismo , Streptomyces coelicolor/genética , Streptomyces coelicolor/crecimiento & desarrollo , Antibacterianos/biosíntesis , Antibacterianos/metabolismo , Ferritinas/metabolismo , Ferritinas/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Hierro/metabolismo , Antraquinonas/metabolismo , Grupo Citocromo b/metabolismo , Grupo Citocromo b/genética , Regulación Bacteriana de la Expresión Génica , Prodigiosina/metabolismo , Prodigiosina/análogos & derivados , Prodigiosina/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Proteómica , Benzoisocromanquinonas
2.
Sci Rep ; 14(1): 18242, 2024 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-39107474

RESUMEN

Iron storage proteins, e.g., vertebrate ferritin, and the ferritin-like bacterioferritin (Bfr) and bacterial ferritin (Ftn), are spherical, hollow proteins that catalyze the oxidation of Fe2+ at binuclear iron ferroxidase centers (FOC) and store the Fe3+ in their interior, thus protecting cells from unwanted Fe3+/Fe2+ redox cycling and storing iron at concentrations far above the solubility of Fe3+. Vertebrate ferritins are heteropolymers of H and L subunits with only the H subunits having FOC. Bfr and Ftn were thought to coexist in bacteria as homopolymers, but recent evidence indicates these molecules are heteropolymers assembled from Bfr and Ftn subunits. Despite the heteropolymeric nature of vertebrate and bacterial ferritins, structures have been determined only for recombinant proteins constituted by a single subunit type. Herein we report the structure of Acinetobacter baumannii bacterioferritin, the first structural example of a heteropolymeric ferritin or ferritin-like molecule, assembled from completely overlapping Ftn homodimers harboring FOC and Bfr homodimers devoid of FOC but binding heme. The Ftn homodimers function by catalyzing the oxidation of Fe2+ to Fe3+, while the Bfr homodimers bind a cognate ferredoxin (Bfd) which reduces the stored Fe3+ by transferring electrons via the heme, enabling Fe2+ mobilization to the cytosol for incorporation in metabolism.


Asunto(s)
Acinetobacter baumannii , Proteínas Bacterianas , Grupo Citocromo b , Ferritinas , Ferritinas/química , Ferritinas/metabolismo , Acinetobacter baumannii/metabolismo , Grupo Citocromo b/química , Grupo Citocromo b/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Modelos Moleculares , Multimerización de Proteína , Hierro/metabolismo , Hierro/química , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Oxidación-Reducción , Conformación Proteica
3.
J Infect Dis ; 230(1): e149-e158, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39052707

RESUMEN

BACKGROUND: Cytochrome bd complexes are respiratory oxidases found exclusively in prokaryotes that are important during infection for numerous bacterial pathogens. METHODS: In silico docking was employed to screen approved drugs for their ability to bind to the quinol site of Escherichia coli cytochrome bd-I. Respiratory inhibition was assessed with oxygen electrodes using membranes isolated from E. coli and methicillin-resistant Staphylococcus aureus strains expressing single respiratory oxidases (ie, cytochromes bd, bo', or aa3). Growth/viability assays were used to measure bacteriostatic and bactericidal effects. RESULTS: The steroid drugs ethinylestradiol and quinestrol inhibited E. coli bd-I activity with median inhibitory concentration (IC50) values of 47 ± 28.9 µg/mL (158 ± 97.2 µM) and 0.2 ± 0.04 µg/mL (0.5 ± 0.1 µM), respectively. Quinestrol inhibited growth of an E. coli "bd-I only" strain with an IC50 of 0.06 ± 0.02 µg/mL (0.2 ± 0.07 µM). Growth of an S. aureus "bd only" strain was inhibited by quinestrol with an IC50 of 2.2 ± 0.43 µg/mL (6.0 ± 1.2 µM). Quinestrol exhibited potent bactericidal effects against S. aureus but not E. coli. CONCLUSIONS: Quinestrol inhibits cytochrome bd in E. coli and S. aureus membranes and inhibits the growth of both species, yet is only bactericidal toward S. aureus.


Asunto(s)
Antibacterianos , Escherichia coli , Staphylococcus aureus Resistente a Meticilina , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Escherichia coli/efectos de los fármacos , Escherichia coli/enzimología , Antibacterianos/farmacología , Simulación del Acoplamiento Molecular , Oxidorreductasas/antagonistas & inhibidores , Oxidorreductasas/metabolismo , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/metabolismo , Pruebas de Sensibilidad Microbiana , Esteroides/farmacología , Esteroides/química , Proteínas del Complejo de Cadena de Transporte de Electrón/antagonistas & inhibidores , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Grupo Citocromo b , Citocromos/antagonistas & inhibidores , Citocromos/metabolismo
4.
J Bacteriol ; 206(8): e0013024, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-38995039

RESUMEN

c-di-AMP is an essential second messenger that binds and regulates several proteins of different functions within bacterial cells. Among those, PstA is a structurally conserved c-di-AMP-binding protein, but its function is largely unknown. PstA is structurally similar to PII signal transduction proteins, although it specifically binds c-di-AMP rather than other PII ligands such as ATP and α-ketoglutarate. In Listeria monocytogenes, we found that PstA increases ß-lactam susceptibility at normal and low c-di-AMP levels, but increases ß-lactam resistance upon c-di-AMP accumulation. Examining a PstA mutant defective for c-di-AMP binding, we found the apo form of PstA to be toxic for ß-lactam resistance, and the c-di-AMP-bound form to be beneficial. Intriguingly, a role for PstA in ß-lactam resistance is only prominent in aerobic cultures, and largely diminished under hypoxic conditions, suggesting that PstA function is linked to aerobic metabolism. However, PstA does not control aerobic growth rate, and has a modest influence on the tricarboxylic acid cycle and membrane potential-an indicator of cellular respiration. The regulatory role of PstA in ß-lactam resistance is unrelated to reactive oxygen species or oxidative stress. Interestingly, during aerobic growth, PstA function requires the cytochrome bd oxidase (CydAB), a component of the respiratory electron transport chain. The requirement for CydAB might be related to its function in maintaining a membrane potential, or redox stress response activities. Altogether, we propose a model in which apo-PstA diminishes ß-lactam resistance by interacting with an effector protein, and this activity can be countered by c-di-AMP binding or a by-product of redox stress. IMPORTANCE: PstA is a structurally conserved c-di-AMP-binding protein that is broadly present among Firmicutes bacteria. Furthermore, PstA binds c-di-AMP at high affinity and specificity, indicating an important role in the c-di-AMP signaling network. However, the molecular function of PstA remains elusive. Our findings reveal contrasting roles of PstA in ß-lactam resistance depending on c-di-AMP-binding status. We also define physiological conditions for PstA function during aerobic growth. Future efforts can exploit these conditions to identify PstA interaction partners under ß-lactam stress.


Asunto(s)
Proteínas Bacterianas , Listeria monocytogenes , Resistencia betalactámica , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/genética , Listeria monocytogenes/enzimología , Listeria monocytogenes/metabolismo , Listeria monocytogenes/crecimiento & desarrollo , Resistencia betalactámica/genética , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Grupo Citocromo b/metabolismo , Grupo Citocromo b/genética , beta-Lactamas/farmacología , Antibacterianos/farmacología , Regulación Bacteriana de la Expresión Génica
5.
J Inorg Biochem ; 259: 112653, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38943845

RESUMEN

Cytochrome bd-I from Escherichia coli belongs to the superfamily of prokaryotic bd-type oxygen reductases. It contains three hemes, b558, b595 and d, and couples oxidation of quinol by dioxygen with the generation of a proton-motive force. The enzyme exhibits resistance to various stressors and is considered as a target protein for next-generation antimicrobials. By using electronic absorption and MCD spectroscopy, this work shows that cyanide binds to heme d2+ in the isolated fully reduced cytochrome bd-I. Cyanide-induced difference absorption spectra display changes near the heme d2+ α-band, a minimum at 633 nm and a maximum around 600 nm, and a W-shaped response in the Soret region. Apparent dissociation constant (Kd) of the cyanide complex of heme d2+ is ∼0.052 M. Kinetics of cyanide binding is monophasic, indicating the presence of a single ligand binding site in the enzyme. Consistently, MCD data show that cyanide binds to heme d2+ but not to b5582+ or b5952+. This agrees with the published structural data that the enzyme's active site is not a di-heme site. The observed rate of binding (kobs) increases as the concentration of cyanide is increased, giving a second-order rate constant (kon) of ∼0.1 M-1 s-1.


Asunto(s)
Cianuros , Proteínas de Escherichia coli , Escherichia coli , Hemo , Oxidorreductasas , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Escherichia coli/enzimología , Cianuros/metabolismo , Cianuros/química , Hemo/metabolismo , Hemo/química , Oxidorreductasas/metabolismo , Oxidorreductasas/química , Oxidación-Reducción , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/química , Grupo Citocromo b/metabolismo , Grupo Citocromo b/química , Cinética , Citocromos/metabolismo , Citocromos/química , Sitios de Unión , Unión Proteica
6.
Nat Commun ; 15(1): 5211, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890314

RESUMEN

Photosystem II (PSII) catalyzes water oxidation and plastoquinone reduction by utilizing light energy. It is highly susceptible to photodamage under high-light conditions and the damaged PSII needs to be restored through a process known as the PSII repair cycle. The detailed molecular mechanism underlying the PSII repair process remains mostly elusive. Here, we report biochemical and structural features of a PSII-repair intermediate complex, likely arrested at an early stage of the PSII repair process in the green alga Chlamydomonas reinhardtii. The complex contains three protein factors associated with a damaged PSII core, namely Thylakoid Enriched Factor 14 (TEF14), Photosystem II Repair Factor 1 (PRF1), and Photosystem II Repair Factor 2 (PRF2). TEF14, PRF1 and PRF2 may facilitate the release of the manganese-stabilizing protein PsbO, disassembly of peripheral light-harvesting complexes from PSII and blockage of the QB site, respectively. Moreover, an α-tocopherol quinone molecule is located adjacent to the heme group of cytochrome b559, potentially fulfilling a photoprotective role by preventing the generation of reactive oxygen species.


Asunto(s)
Chlamydomonas reinhardtii , Complejo de Proteína del Fotosistema II , Complejo de Proteína del Fotosistema II/metabolismo , Chlamydomonas reinhardtii/metabolismo , Chlamydomonas reinhardtii/genética , Tilacoides/metabolismo , Complejos de Proteína Captadores de Luz/metabolismo , Complejos de Proteína Captadores de Luz/genética , Proteínas de Plantas/metabolismo , Proteínas de Plantas/genética , Grupo Citocromo b/metabolismo , Grupo Citocromo b/genética , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Luz
7.
Biochim Biophys Acta Bioenerg ; 1865(3): 149045, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38614453

RESUMEN

Cytochrome bo3 quinol oxidase belongs to the heme­copper-oxidoreductase (HCO) superfamily, which is part of the respiratory chain and essential for cell survival. While the reaction mechanism of cyt bo3 has been studied extensively over the last decades, specific details about its substrate binding and product release have remained unelucidated due to the lack of structural information. Here, we report a 2.8 Å cryo-electron microscopy structure of cyt bo3 from Escherichia coli assembled in peptidiscs. Our structural model shows a conformation for amino acids 1-41 of subunit I different from all previously published structures while the remaining parts of this enzyme are similar. Our new conformation shows a "U-shape" assembly in contrast to the transmembrane helix, named "TM0", in other reported structural models. However, TM0 blocks ubiquinone-8 (reaction product) release, suggesting that other cyt bo3 conformations should exist. Our structural model presents experimental evidence for an "open" conformation to facilitate substrate/product exchange. This work helps further understand the reaction cycle of this oxidase, which could be a benefit for potential drug/antibiotic design for health science.


Asunto(s)
Microscopía por Crioelectrón , Grupo Citocromo b , Proteínas de Escherichia coli , Escherichia coli , Ubiquinona , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo , Ubiquinona/química , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/ultraestructura , Escherichia coli/enzimología , Grupo Citocromo b/química , Grupo Citocromo b/metabolismo , Conformación Proteica , Modelos Moleculares , Citocromos/química , Citocromos/metabolismo
8.
Angew Chem Int Ed Engl ; 63(16): e202401379, 2024 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-38407997

RESUMEN

Ferritins are multimeric cage-forming proteins that play a crucial role in cellular iron homeostasis. All H-chain-type ferritins harbour a diiron site, the ferroxidase centre, at the centre of a 4 α-helical bundle, but bacterioferritins are unique in also binding 12 hemes per 24 meric assembly. The ferroxidase centre is known to be required for the rapid oxidation of Fe2+ during deposition of an immobilised ferric mineral core within the protein's hollow interior. In contrast, the heme of bacterioferritin is required for the efficient reduction of the mineral core during iron release, but has little effect on the rate of either oxidation or mineralisation of iron. Thus, the current view is that these two cofactors function in iron uptake and release, respectively, with no functional overlap. However, rapid electron transfer between the heme and ferroxidase centre of bacterioferritin from Escherichia coli was recently demonstrated, suggesting that the two cofactors may be functionally connected. Here we report absorbance and (magnetic) circular dichroism spectroscopies, together with in vitro assays of iron-release kinetics, which demonstrate that the ferroxidase centre plays an important role in the reductive mobilisation of the bacterioferritin mineral core, which is dependent on the heme-ferroxidase centre electron transfer pathway.


Asunto(s)
Ceruloplasmina , Hierro , Hierro/química , Ceruloplasmina/química , Escherichia coli/metabolismo , Ferritinas/química , Proteínas Bacterianas/metabolismo , Grupo Citocromo b/química , Minerales , Oxidación-Reducción , Hemo/metabolismo
9.
Int J Mol Sci ; 25(2)2024 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-38279276

RESUMEN

The terminal oxidases of bacterial aerobic respiratory chains are redox-active electrogenic enzymes that catalyze the four-electron reduction of O2 to 2H2O taking out electrons from quinol or cytochrome c. Living bacteria often deal with carbon monoxide (CO) which can act as both a signaling molecule and a poison. Bacterial terminal oxidases contain hemes; therefore, they are potential targets for CO. However, our knowledge of this issue is limited and contradictory. Here, we investigated the effect of CO on the cell growth and aerobic respiration of three different Escherichia coli mutants, each expressing only one terminal quinol oxidase: cytochrome bd-I, cytochrome bd-II, or cytochrome bo3. We found that following the addition of CO to bd-I-only cells, a minimal effect on growth was observed, whereas the growth of both bd-II-only and bo3-only strains was severely impaired. Consistently, the degree of resistance of aerobic respiration of bd-I-only cells to CO is high, as opposed to high CO sensitivity displayed by bd-II-only and bo3-only cells consuming O2. Such a difference between the oxidases in sensitivity to CO was also observed with isolated membranes of the mutants. Accordingly, O2 consumption of wild-type cells showed relatively low CO sensitivity under conditions favoring the expression of a bd-type oxidase.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Monóxido de Carbono/farmacología , Monóxido de Carbono/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Grupo Citocromo b/genética , Grupo Citocromo b/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Citocromos/genética , Citocromos/metabolismo , Oxidación-Reducción , Oxidorreductasas/genética , Oxidorreductasas/metabolismo , Respiración
10.
PLoS One ; 18(12): e0291564, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38039324

RESUMEN

Cytochrome b561 (cytb561) proteins comprise a family of transmembrane oxidoreductases that transfer single electrons across a membrane. Most eukaryotic species, including insects, possess multiple cytb561 homologs. To learn more about this protein family in insects, we carried out a bioinformatics-based investigation of cytb561 family members from nine species representing eight insect orders. We performed a phylogenetic analysis to classify insect cytb561 ortholog groups. We then conducted sequence analyses and analyzed protein models to predict structural elements that may impact the biological functions and localization of these proteins, with a focus on possible ferric reductase activity. Our study revealed three ortholog groups, designated CG1275, Nemy, and CG8399, and a fourth group of less-conserved genes. We found that CG1275 and Nemy proteins are similar to a human ferric reductase, duodenal cytochrome b561 (Dcytb), and have many conserved amino acid residues that function in substrate binding in Dcytb. Notably, CG1275 and Nemy proteins contain a conserved histidine and other residues that play a role in ferric ion reduction by Dcytb. Nemy proteins were distinguished by a novel cysteine-rich cytoplasmic loop sequence. CG8399 orthologs are similar to a putative ferric reductase in humans, stromal cell-derived receptor 2. Like other members of the CYBDOM class of cytb561 proteins, these proteins contain reeler, DOMON, and cytb561 domains. Drosophila melanogaster CG8399 is the only insect cytb561 with known ferric reductase activity. Our investigation of the DOMON domain in CG8399 proteins revealed a probable heme-binding site and a possible site for ferric reduction. The fourth group includes a subgroup of proteins with a conserved "KXXXXKXH" non-cytoplasmic loop motif that may be a substrate binding site and is present in a potential ferric reductase, human tumor suppressor cytochrome b561. This study provides a foundation for future investigations of the biological functions of cytb561 genes in insects.


Asunto(s)
Drosophila melanogaster , Oxidorreductasas , Animales , Humanos , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Filogenia , Oxidorreductasas/metabolismo , Grupo Citocromo b/genética , Grupo Citocromo b/metabolismo , Hierro/metabolismo
11.
Biochemistry (Mosc) ; 88(10): 1504-1512, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38105020

RESUMEN

An overview of current notions on the mechanism of generation of a transmembrane electric potential difference (Δψ) during the catalytic cycle of a bd-type triheme terminal quinol oxidase is presented in this work. It is suggested that the main contribution to Δψ formation is made by the movement of H+ across the membrane along the intra-protein hydrophilic proton-conducting pathway from the cytoplasm to the active site for oxygen reduction of this bacterial enzyme.


Asunto(s)
Grupo Citocromo b , Proteínas de Escherichia coli , Potenciales de la Membrana , Grupo Citocromo b/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Citocromos/metabolismo , Oxidación-Reducción
12.
J Inorg Biochem ; 247: 112341, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37515940

RESUMEN

Carbon monoxide (CO) plays a multifaceted role in the physiology of organisms, from poison to signaling molecule. Heme proteins, including terminal oxidases, are plausible CO targets. Three quinol oxidases terminate the branched aerobic respiratory chain of Escherichia coli. These are the heme­copper cytochrome bo3 and two copper-lacking bd-type cytochromes, bd-I and bd-II. All three enzymes generate a proton motive force during the four-electron oxygen reduction reaction that is used for ATP production. The bd-type oxidases also contribute to mechanisms of bacterial defense against various types of stresses. Here we report that in E. coli cells, at the enzyme concentrations tested, cytochrome bd-I is much more resistant to inhibition by CO than cytochrome bd-II and cytochrome bo3. The apparent half-maximal inhibitory concentration values, IC50, for inhibition of O2 consumption of the membrane-bound bd-II and bo3 oxidases by CO at ~150 µM O2 were estimated to be 187.1 ± 11.1 and 183.3 ± 13.5 µM CO, respectively. Under the same conditions, the maximum inhibition observed with the membrane-bound cytochrome bd-I was 20 ± 10% at ~200 µM CO.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Escherichia coli/metabolismo , Monóxido de Carbono/farmacología , Monóxido de Carbono/metabolismo , Cobre/metabolismo , Proteínas de Escherichia coli/metabolismo , Grupo Citocromo b , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Citocromos/metabolismo , Oxidorreductasas/metabolismo , Oxidación-Reducción
13.
Sci Rep ; 13(1): 12226, 2023 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-37507428

RESUMEN

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


Asunto(s)
Proteínas de Escherichia coli , Quinona Reductasas , Hidroquinonas/farmacología , Hidroquinonas/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Quinona Reductasas/metabolismo , Oxidorreductasas/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Citocromos/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Grupo Citocromo b/metabolismo
14.
Inorg Chem ; 62(10): 4066-4075, 2023 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-36857027

RESUMEN

The cytochrome bd oxygen reductase catalyzes the four-electron reduction of dioxygen to two water molecules. The structure of this enzyme reveals three heme molecules in the active site, which differs from that of heme-copper cytochrome c oxidase. The quantum chemical cluster approach was used to uncover the reaction mechanism of this intriguing metalloenzyme. The calculations suggested that a proton-coupled electron transfer reduction occurs first to generate a ferrous heme b595. This is followed by the dioxygen binding at the heme d center coupled with an outer-sphere electron transfer from the ferrous heme b595 to the dioxygen moiety, affording a ferric ion superoxide intermediate. A second proton-coupled electron transfer produces a heme d ferric hydroperoxide, which undergoes efficient O-O bond cleavage facilitated by an outer-sphere electron transfer from the ferrous heme b595 to the O-O σ* orbital and an inner-sphere proton transfer from the heme d hydroxyl group to the leaving hydroxide. The synergistic benefits of the two types of hemes rationalize the highly efficient oxygen reduction repertoire for the multi-heme-dependent cytochrome bd oxygen reductase family.


Asunto(s)
Proteínas de Escherichia coli , Oxidorreductasas , Oxidorreductasas/química , Oxígeno/química , Protones , Electrones , Grupo Citocromo b/metabolismo , Proteínas de Escherichia coli/química , Proteínas del Complejo de Cadena de Transporte de Electrón/química , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Citocromos/química , Oxidación-Reducción , Hemo/química , Hierro
15.
J Biol Chem ; 299(3): 102968, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36736898

RESUMEN

Photosystem II (PSII), the water:plastoquinone oxidoreductase of oxygenic photosynthesis, contains a heme b559 iron whose axial ligands are provided by histidine residues from the α (PsbE) and ß (PsbF) subunits. PSII assembly depends on accessory proteins that facilitate the step-wise association of its protein and pigment components into a functional complex, a process that is challenging to study due to the low accumulation of assembly intermediates. Here, we examined the putative role of the iron[1Fe-0S]-containing protein rubredoxin 1 (RBD1) as an assembly factor for cytochrome b559, using the RBD1-lacking 2pac mutant from Chlamydomonas reinhardtii, in which the accumulation of PSII was rescued by the inactivation of the thylakoid membrane FtsH protease. To this end, we constructed the double mutant 2pac ftsh1-1, which harbored PSII dimers that sustained its photoautotrophic growth. We purified PSII from the 2pac ftsh1-1 background and found that α and ß cytochrome b559 subunits are still present and coordinate heme b559 as in the WT. Interestingly, immunoblot analysis of dark- and low light-grown 2pac ftsh1-1 showed the accumulation of a 23-kDa fragment of the D1 protein, a marker typically associated with structural changes resulting from photodamage of PSII. Its cleavage occurs in the vicinity of a nonheme iron which binds to PSII on its electron acceptor side. Altogether, our findings demonstrate that RBD1 is not required for heme b559 assembly and point to a role for RBD1 in promoting the proper folding of D1, possibly via delivery or reduction of the nonheme iron during PSII assembly.


Asunto(s)
Chlamydomonas reinhardtii , Grupo Citocromo b , Complejo de Proteína del Fotosistema II , Rubredoxinas , Grupo Citocromo b/genética , Grupo Citocromo b/metabolismo , Hemo/metabolismo , Hierro/metabolismo , Complejo de Proteína del Fotosistema II/genética , Complejo de Proteína del Fotosistema II/metabolismo , Rubredoxinas/metabolismo , Chlamydomonas reinhardtii/genética , Chlamydomonas reinhardtii/metabolismo
16.
Cell Death Dis ; 14(1): 1, 2023 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-36593242

RESUMEN

DEAD box helicase 17 (DDX17) has been reported to be involved in the initiation and development of several cancers. However, the functional role and mechanisms of DDX17 in colorectal cancer (CRC) malignant progression and metastasis remain unclear. Here, we reported that DDX17 expression was increased in CRC tissues compared with noncancerous mucosa tissues and further upregulated in CRC liver metastasis compared with patient-paired primary tumors. High levels of DDX17 were significantly correlated with aggressive phenotypes and worse clinical outcomes in CRC patients. Ectopic expression of DDX17 promoted cell migration and invasion in vitro and in vivo, while the opposite results were obtained in DDX17-deficient CRC cells. We identified miR-149-3p as a potential downstream miRNA of DDX17 through RNA sequencing analysis, and miR-149-3p displayed a suppressive effect on the metastatic potential of CRC cells. We demonstrated that CYBRD1 (a ferric reductase that contributes to dietary iron absorption) was a direct target of miR-149-3p and that miR-149-3p was required for DDX17-mediated regulation of CYBRD1 expression. Moreover, DDX17 contributed to the metastasis and epithelial to mesenchymal transition (EMT) of CRC cells via downregulation of miR-149-3p, which resulted in increased CYBRD1 expression. In conclusion, our findings not only highlight the significance of DDX17 in the aggressive development and prognosis of CRC patients, but also reveal a novel mechanism underlying DDX17-mediated CRC cell metastasis and EMT progression through manipulation of the miR-149-3p/CYBRD1 pathway.


Asunto(s)
Neoplasias Colorrectales , Grupo Citocromo b , ARN Helicasas DEAD-box , MicroARNs , Humanos , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Colorrectales/patología , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Regulación hacia Abajo , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , Metástasis de la Neoplasia , Grupo Citocromo b/genética , Grupo Citocromo b/metabolismo
17.
Biochim Biophys Acta Bioenerg ; 1864(2): 148952, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36535430

RESUMEN

Escherichia coli contains two cytochrome bd oxidases, bd-I and bd-II. The structure of both enzymes is highly similar, but they exhibit subtle differences such as the accessibility of the active site through a putative proton channel. Here, we demonstrate that the duroquinol:dioxygen oxidoreductase activity of bd-I increased with alkaline pH, whereas bd-II showed a broad activity maximum around pH 7. Likewise, the pH dependence of NO release from the reduced active site, an essential property of bd oxidases, differed between the two oxidases as detected by UV/vis spectroscopy. Both findings may be attributed to differences in the proton channel leading to the active site heme d. The channel comprises a titratable residue (Asp58B in bd-I and Glu58B in bd-II). Conservative mutations at this position drastically altered NO release demonstrating its contribution to the process.


Asunto(s)
Proteínas de Escherichia coli , Oxidorreductasas , Oxidorreductasas/metabolismo , Escherichia coli , Citocromos/química , Protones , Proteínas de Escherichia coli/metabolismo , Grupo Citocromo b/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Complejo IV de Transporte de Electrones , Concentración de Iones de Hidrógeno
18.
Eur J Med Chem ; 245(Pt 1): 114896, 2023 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-36370551

RESUMEN

The cytochrome bcc-aa3 oxidase (Cyt-bcc) of Mycobacterium tuberculosis (Mtb) is a promising anti-tuberculosis target. However, when Cyt-bcc is inhibited, cytochrome bd terminal oxidase (Cyt-bd) can still maintain the activity of the respiratory chain and drive ATP synthesis. Through virtual screening and biological validation, we discovered two FDA-approved drugs, ivacaftor and roquinimex, exhibited moderate binding affinity to Cyt-bd. Structural modifications of them led to 1-hydroxy-2-methylquinolin-4(1H)-one derivatives as potent new Cyt-bd inhibitors. Compound 8d binds to Cyt-bd with a Kd value of 4.17 µM and inhibits the growth of the Cyt-bcc knock-out strain (ΔqcrCAB, Cyt-bd+) with a MIC value of 6.25 µM. The combination of 8d with the Cyt-bcc inhibitor Q203 completely inhibited oxygen consumption of the wild-type strain and the inverted-membrane vesicles expressing M. tuberculosis Cyt-bd (ΔcydAB::MtbCydAB+). Our study provides a promising starting point for the development of novel dual chemotherapies for tuberculosis.


Asunto(s)
Antituberculosos , Grupo Citocromo b , Grupo Citocromo d , Mycobacterium tuberculosis , Oxidorreductasas , Humanos , Antituberculosos/química , Antituberculosos/farmacología , Mycobacterium tuberculosis/efectos de los fármacos , Mycobacterium tuberculosis/enzimología , Oxidorreductasas/antagonistas & inhibidores , Tuberculosis/tratamiento farmacológico , Grupo Citocromo b/antagonistas & inhibidores , Grupo Citocromo d/antagonistas & inhibidores
19.
FEBS Lett ; 597(4): 547-556, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36460943

RESUMEN

Cytochrome bd-I from Escherichia coli is a terminal oxidase in the respiratory chain that plays an important role under stress conditions. Cytochrome bd-I was thought to consist of the major subunits CydA and CydB plus the small CydX subunit. Recent high-resolution structures of cytochrome bd-I demonstrated the presence of an additional subunit, CydH/CydY (called CydH here), the function of which is unclear. In this report, we show that in the absence of CydH, cytochrome bd-I is catalytically active, can sustain bacterial growth and displays haem spectra and susceptibility for haem-binding inhibitors comparable to the wild-type enzyme. Removal of CydH did not elicit catalase activity of cytochrome bd-I in our experimental system. Taken together, in the absence of the CydH subunit cytochrome bd-I retained key enzymatic properties.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Grupo Citocromo b/genética , Grupo Citocromo b/química , Citocromos/genética , Citocromos/química , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/química , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Hemo
20.
Biotechnol Adv ; 61: 108057, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36328189

RESUMEN

Bacterioferritin (Bfr) is a subfamily of ferritin protein family. Bfrs are composed of 24 identical subunits and self-assemble into 4-3-2-fold symmetric cage-like structure with the incorporation of 12 heme groups into twelve 2-fold symmetric binding sites between subunits. Bfr protein cage has an outer diameter of ∼12 nm and interior cavity diameter of ∼8 nm with a total of 62 pores to connect the interior cavity with the bulk solution outside the protein nanocage. In vivo, the interior cavity of Bfr can store up to ∼2700 iron atoms in the ferrihydrite-like mineral. Recent years, more and more Bfr structures have been solved, which elucidated more details about the ferroxidase center, the catalytic mechanism, the possible channels used by iron ions to access the interior cavity, the electron transfer pathway involved in the iron redox cycle, and the molecular function of the heme group. The preliminary applications of both mammalian and bacterial ferritins in drug delivery, imaging diagnosis, and nanoparticle vaccine make Bfr exploration uniquely attractive for researchers from a broad range of research fields because Bfr has advantages over ferritins in controlling the self-assembly and redesigning the subunit. In this article, we outline the structure of Bfr, review the recent progress in the molecular mechanism of Bfr to store and release iron, and focus on the self-assembly and genetic modification of Bfr nanocage. Based on the comparison between Bfr and other ferritin family members, we further discuss the potential applications of Bfr. We expect that both fundamental and applied researches on Bfr will attract broad interest in protein nanocage design, nanomedicine, precise therapy, nanoparticle vaccine, bionanotechnology, bionanoelectronics, and so on.


Asunto(s)
Grupo Citocromo b , Ferritinas , Animales , Hierro , Hemo , Mamíferos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA