Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.650
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Biochemistry (Mosc) ; 87(8): 720-730, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36171653

RESUMEN

Cytochrome bd-II is one of the three terminal quinol oxidases of the aerobic respiratory chain of Escherichia coli. Preparations of the detergent-solubilized untagged bd-II oxidase isolated from the bacterium were shown to scavenge hydrogen peroxide (H2O2) with high rate producing molecular oxygen (O2). Addition of H2O2 to the same buffer that does not contain enzyme or contains thermally denatured cytochrome bd-II does not lead to any O2 production. The latter observation rules out involvement of adventitious transition metals bound to the protein. The H2O2-induced O2 production is not susceptible to inhibition by N-ethylmaleimide (the sulfhydryl binding compound), antimycin A (the compound that binds specifically to a quinol binding site), and CO (diatomic gas that binds specifically to the reduced heme d). However, O2 formation is inhibited by cyanide (IC50 = 4.5 ± 0.5 µM) and azide. Addition of H2O2 in the presence of dithiothreitol and ubiquinone-1 does not inactivate cytochrome bd-II and apparently does not affect the O2 reductase activity of the enzyme. The ability of cytochrome bd-II to detoxify H2O2 could play a role in bacterial physiology by conferring resistance to the peroxide-mediated stress.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa , Proteínas de Escherichia coli , Escherichia coli , Antimicina A/metabolismo , Azidas/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Cianuros/metabolismo , Grupo Citocromo b/metabolismo , Citocromos/metabolismo , Detergentes , Ditiotreitol/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Etilmaleimida/metabolismo , Peróxido de Hidrógeno/metabolismo , Hidroquinonas/metabolismo , Oxidación-Reducción , Oxidorreductasas/metabolismo , Oxígeno/metabolismo , Ubiquinona/metabolismo
9.
Nanoscale ; 14(34): 12322-12331, 2022 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-35969005

RESUMEN

The thermal and chemical stability of 24mer ferritins has led to attempts to exploit their naturally occurring nanoscale (8 nm) internal cavities for biotechnological applications. An area of increasing interest is the encapsulation of molecules either for medical or biocatalysis applications. Encapsulation requires ferritin dissociation, typically induced using high temperature or acidic conditions (pH ≥ 2), which generally precludes the inclusion of fragile cargo such as proteins or peptide fragments. Here we demonstrate that minimizing salt concentration combined with adjusting the pH to ≤8.5 (i.e. low proton/metal ion concentration) reversibly shifts the naturally occurring equilibrium between dimeric and 24meric assemblies of Escherichia coli bacterioferritin (Bfr) in favour of the disassembled form. Interconversion between the different oligomeric forms of Bfr is sufficiently slow under these conditions to allow the use of size exclusion chromatography to obtain wild type protein in the purely dimeric and 24meric forms. This control over association state was exploited to bind heme at natural sites that are not accessible in the assembled protein. The potential for biotechnological applications was demonstrated by the encapsulation of a small, acidic [3Fe-4S] cluster-containing ferredoxin within the Bfr internal cavity. The capture of ∼4-6 negatively charged ferredoxin molecules per cage indicates that charge complementarity with the inner protein surface is not an essential determinant of successful encapsulation.


Asunto(s)
Grupo Citocromo b , Ferredoxinas , Proteínas Bacterianas/química , Grupo Citocromo b/química , Grupo Citocromo b/metabolismo , Escherichia coli/metabolismo , Ferredoxinas/metabolismo , Ferritinas/química
10.
FEBS Lett ; 596(18): 2418-2424, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36029102

RESUMEN

The reduction of oxygen to water is crucial to life and a central metabolic process. To fulfil this task, prokaryotes use among other enzymes cytochrome bd oxidases (Cyt bds) that also play an important role in bacterial virulence and antibiotic resistance. To fight microbial infections by pathogens, an in-depth understanding of the enzyme mechanism is required. Here, we combine bioinformatics, mutagenesis, enzyme kinetics and FTIR spectroscopy to demonstrate that proton delivery to the active site contributes to the rate limiting steps in Cyt bd-I and involves Asp58 of subunit CydB. Our findings reveal a previously unknown catalytic function of subunit CydB in the reaction of Cyt bd-I.


Asunto(s)
Proteínas de Escherichia coli , Escherichia coli , Grupo Citocromo b/genética , Grupo Citocromo b/metabolismo , Citocromos/química , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Oxidorreductasas/metabolismo , Oxígeno/metabolismo , Protones , Agua/metabolismo
11.
New Phytol ; 233(2): 766-780, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34625967

RESUMEN

Cytochrome (Cyt) b559 is a key component of the photosystem II complex (PSII) that is essential for its proper functioning and assembly. Site-directed mutants of the model cyanobacterium Synechocystis sp. PCC6803 with mutated heme axial ligands of Cyt b559 have little PSII and are therefore unable to grow photoautotrophically. Here we describe two types of Synechocystis autotrophic transformants that retained the same mutations in Cyt b559 but are able to accumulate PSII and grow photoautotrophically. Whole-genome sequencing revealed that all of these autotrophic transformants carried a variable number of tandem repeats (from 5 to 15) of chromosomal segments containing the psbEFLJ operon. RNA-seq analysis showed greatly increased transcript levels of the psbEFLJ operon in these autotrophic transformants. Multiple copies of the psbEFLJ operon in these transformants were only maintained during autotrophic growth, while its copy numbers gradually decreased under photoheterotrophic conditions. Two-dimensional PAGE analysis of membrane proteins revealed a strong deficiency in PSII complexes in the Cyt b559 mutants that was reversed in the autotrophic transformants. These results illustrate how tandem gene amplification restores PSII accumulation and photoautotrophic growth in Cyt b559 mutants of cyanobacteria, and may serve as an important adaptive mechanism for cyanobacterial survival.


Asunto(s)
Complejo de Proteína del Fotosistema II , Synechocystis , Grupo Citocromo b/genética , Grupo Citocromo b/metabolismo , Citocromos b/genética , Citocromos b/metabolismo , Amplificación de Genes , Complejo de Proteína del Fotosistema II/genética , Complejo de Proteína del Fotosistema II/metabolismo , Synechocystis/metabolismo
12.
Proc Natl Acad Sci U S A ; 118(50)2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34873041

RESUMEN

The treatment of infectious diseases caused by multidrug-resistant pathogens is a major clinical challenge of the 21st century. The membrane-embedded respiratory cytochrome bd-type oxygen reductase is a critical survival factor utilized by pathogenic bacteria during infection, proliferation and the transition from acute to chronic states. Escherichia coli encodes for two cytochrome bd isoforms that are both involved in respiration under oxygen limited conditions. Mechanistic and structural differences between cydABX (Ecbd-I) and appCBX (Ecbd-II) operon encoded cytochrome bd variants have remained elusive in the past. Here, we demonstrate that cytochrome bd-II catalyzes oxidation of benzoquinols while possessing additional specificity for naphthoquinones. Our data show that although menaquinol-1 (MK1) is not able to directly transfer electrons onto cytochrome bd-II from E. coli, it has a stimulatory effect on its oxygen reduction rate in the presence of ubiquinol-1. We further determined cryo-EM structures of cytochrome bd-II to high resolution of 2.1 Å. Our structural insights confirm that the general architecture and substrate accessible pathways are conserved between the two bd oxidase isoforms, but two notable differences are apparent upon inspection: (i) Ecbd-II does not contain a CydH-like subunit, thereby exposing heme b595 to the membrane environment and (ii) the AppB subunit harbors a structural demethylmenaquinone-8 molecule instead of ubiquinone-8 as found in CydB of Ecbd-I Our work completes the structural landscape of terminal respiratory oxygen reductases of E. coli and suggests that structural and functional properties of the respective oxidases are linked to quinol-pool dependent metabolic adaptations in E. coli.


Asunto(s)
Grupo Citocromo b/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Oxidorreductasas/metabolismo , Grupo Citocromo b/genética , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Modelos Moleculares , Oxidorreductasas/genética , Conformación Proteica , Isoformas de Proteínas
13.
Inorg Chem ; 60(22): 16937-16952, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34695354

RESUMEN

The uptake and utilization of iron remains critical for the survival/virulence of the host/pathogens in spite of the limitations (low bioavailability/high toxicity) associated with this nutrient. Both the host and pathogens manage to overcome these problems by utilizing the iron repository protein nanocages, ferritins, which not only sequester and detoxify the free Fe(II) ions but also decrease the iron solubility gap by synthesizing/encapsulating the Fe(III)-oxyhydroxide biomineral in its central hollow nanocavity. Bacterial pathogens including Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, encode a distinct subclass of ferritins called bacterioferritin (BfrA), which binds heme, the versatile redox cofactor, via coaxial, conserved methionine (M52) residues at its subunit-dimer interfaces. However, the exact role of heme in Mtb BfrA remains yet to be established. Therefore, its coaxial ligands were altered via site-directed mutagenesis, which resulted in both heme-bound (M52C; ∼1 heme per cage) and heme-free (M52H and M52L) variants, indicating the importance of M52 residues as preferential heme binding axial ligands in Mtb BfrA. All these variants formed intact nanocages of similar size and iron-loading ability as that of wild-type (WT) Mtb BfrA. However, the as-isolated heme-bound variants (WT and M52C) exhibited enhanced protein stability and reductive iron mobilization as compared to their heme-free analogues (M52H and M52L). Further, increasing the heme content in BfrA variants by reconstitution not only enhanced the cage stability but also facilitated the iron mobilization, suggesting the role of heme. In contrary, heme altered the ferroxidase activity to a lesser extent despite facilitating the accumulation of the reactive intermediates formed during the course of the reaction. The current study suggests that heme in Mtb BfrA enhances the overall stability of the protein and possibly acts as an intrinsic electron relay station to influence the iron mineral dissolution and thus may be associated with Mtb's pathogenicity.


Asunto(s)
Proteínas Bacterianas/metabolismo , Grupo Citocromo b/metabolismo , Ferritinas/metabolismo , Hemo/metabolismo , Mycobacterium tuberculosis/química , Proteínas Bacterianas/química , Grupo Citocromo b/química , Ferritinas/química , Hemo/química , Ligandos , Estructura Molecular , Mycobacterium tuberculosis/metabolismo
14.
J Clin Lab Anal ; 35(12): e24044, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34714955

RESUMEN

BACKGROUND: Lung adenocarcinoma (LUAD) is a malignant tumor with a high fatality rate and poor overall survival, while molecular targets diagnosing and alleviating lung cancer remain inadequate. METHODS: In this article, we highlighted the upregulation of microRNA-423-3p (miR-423-3p) in LUAD, especially in smokers aged over 40, and revealed that the high expression of miR-423-3p was significantly associated with smoker, age, and pathologic stage of LUAD patients. RESULTS: Moreover, overexpressing miR-423-3p could facilitate LUAD cell proliferation, invasion, adhesion, and epithelial-mesenchymal transition (EMT) process, while depleted miR-423-3p caused repressive influence upon it. Mechanically, we identified that miR-423-3p could activate FAK signaling pathway through binding to the 3'-UTR of cytochrome B reductase 1 (CYBRD1). Furthermore, we demonstrated that CYBRD1 was lowly expressed in LUAD, and miR-423-3p overexpression could rescue the impairment of LUAD cell proliferation, invasion, adhesion, and EMT caused by CYBRD1 depletion. Noticeably, miR-423-3p depletion efficiently hindered LUAD tumor growth in vivo. CONCLUSION: Collectively, our findings demonstrated that miR-423-3p/CYBRD1 axis could be regarded as a promising biomarker to alleviate the poor LUAD prognosis.


Asunto(s)
Adenocarcinoma del Pulmón/patología , Grupo Citocromo b/genética , Quinasa 1 de Adhesión Focal/metabolismo , Neoplasias Pulmonares/patología , MicroARNs/genética , Oxidorreductasas/genética , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/mortalidad , Adulto , Anciano , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Grupo Citocromo b/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Quinasa 1 de Adhesión Focal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Ratones Endogámicos BALB C , Persona de Mediana Edad , Oxidorreductasas/metabolismo , Transducción de Señal/genética , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Microb Cell Fact ; 20(1): 157, 2021 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-34391414

RESUMEN

BACKGROUND: Butenyl-spinosyn, produced by Saccharopolyspora pogona, is a promising biopesticide due to excellent insecticidal activity and broad pesticidal spectrum. Bacterioferritin (Bfr, encoded by bfr) regulates the storage and utilization of iron, which is essential for the growth and metabolism of microorganisms. However, the effect of Bfr on the growth and butenyl-spinosyn biosynthesis in S. pogona has not been explored. RESULTS: Here, we found that the storage of intracellular iron influenced butenyl-spinosyn biosynthesis and the stress resistance of S. pogona, which was regulated by Bfr. The overexpression of bfr increased the production of butenyl-spinosyn by 3.14-fold and enhanced the tolerance of S. pogona to iron toxicity and oxidative damage, while the knockout of bfr had the opposite effects. Based on the quantitative proteomics analysis and experimental verification, the inner mechanism of these phenomena was explored. Overexpression of bfr enhanced the iron storage capacity of the strain, which activated polyketide synthase genes and enhanced the supply of acyl-CoA precursors to improve butenyl-spinosyn biosynthesis. In addition, it induced the oxidative stress response to improve the stress resistance of S. pogona. CONCLUSION: Our work reveals the role of Bfr in increasing the yield of butenyl-spinosyn and enhancing the stress resistance of S. pogona, and provides insights into its enhancement on secondary metabolism, which provides a reference for optimizing the production of secondary metabolites in actinomycetes.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Grupo Citocromo b/genética , Grupo Citocromo b/metabolismo , Ferritinas/genética , Ferritinas/metabolismo , Insecticidas/metabolismo , Hierro/metabolismo , Macrólidos/metabolismo , Saccharopolyspora/metabolismo , Proteínas Bacterianas/farmacología , Grupo Citocromo b/farmacología , Ferritinas/farmacología , Ingeniería Genética , Macrólidos/clasificación , Proteómica , Saccharopolyspora/efectos de los fármacos , Saccharopolyspora/genética , Saccharopolyspora/crecimiento & desarrollo
16.
Proc Natl Acad Sci U S A ; 118(33)2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34389670

RESUMEN

Hemes are common elements of biological redox cofactor chains involved in rapid electron transfer. While the redox properties of hemes and the stability of the spin state are recognized as key determinants of their function, understanding the molecular basis of control of these properties is challenging. Here, benefiting from the effects of one mitochondrial disease-related point mutation in cytochrome b, we identify a dual role of hydrogen bonding (H-bond) to the propionate group of heme bH of cytochrome bc1, a common component of energy-conserving systems. We found that replacing conserved glycine with serine in the vicinity of heme bH caused stabilization of this bond, which not only increased the redox potential of the heme but also induced structural and energetic changes in interactions between Fe ion and axial histidine ligands. The latter led to a reversible spin conversion of the oxidized Fe from 1/2 to 5/2, an effect that potentially reduces the electron transfer rate between the heme and its redox partners. We thus propose that H-bond to the propionate group and heme-protein packing contribute to the fine-tuning of the redox potential of heme and maintaining its proper spin state. A subtle balance is needed between these two contributions: While increasing the H-bond stability raises the heme potential, the extent of increase must be limited to maintain the low spin and diamagnetic form of heme. This principle might apply to other native heme proteins and can be exploited in engineering of artificial heme-containing protein maquettes.


Asunto(s)
Grupo Citocromo b/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Mitocondrias/metabolismo , Rhodobacter capsulatus/metabolismo , Antimicina A/análogos & derivados , Grupo Citocromo b/genética , Espectroscopía de Resonancia por Spin del Electrón , Complejo III de Transporte de Electrones/genética , Enlace de Hidrógeno , Modelos Moleculares , Mutación , Oxidación-Reducción , Conformación Proteica , Análisis Espectral/métodos
17.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34417297

RESUMEN

Two independent structures of the proton-pumping, respiratory cytochrome bo3 ubiquinol oxidase (cyt bo3 ) have been determined by cryogenic electron microscopy (cryo-EM) in styrene-maleic acid (SMA) copolymer nanodiscs and in membrane scaffold protein (MSP) nanodiscs to 2.55- and 2.19-Å resolution, respectively. The structures include the metal redox centers (heme b, heme o3 , and CuB), the redox-active cross-linked histidine-tyrosine cofactor, and the internal water molecules in the proton-conducting D channel. Each structure also contains one equivalent of ubiquinone-8 (UQ8) in the substrate binding site as well as several phospholipid molecules. The isoprene side chain of UQ8 is clamped within a hydrophobic groove in subunit I by transmembrane helix TM0, which is only present in quinol oxidases and not in the closely related cytochrome c oxidases. Both structures show carbonyl O1 of the UQ8 headgroup hydrogen bonded to D75I and R71I In both structures, residue H98I occupies two conformations. In conformation 1, H98I forms a hydrogen bond with carbonyl O4 of the UQ8 headgroup, but in conformation 2, the imidazole side chain of H98I has flipped to form a hydrogen bond with E14I at the N-terminal end of TM0. We propose that H98I dynamics facilitate proton transfer from ubiquinol to the periplasmic aqueous phase during oxidation of the substrate. Computational studies show that TM0 creates a channel, allowing access of water to the ubiquinol headgroup and to H98I.


Asunto(s)
Grupo Citocromo b/química , Grupo Citocromo b/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Hemo/metabolismo , Fosfolípidos/metabolismo , Bombas de Protones , Ubiquinona/metabolismo , Sitios de Unión , Microscopía por Crioelectrón , Hemo/química , Oxidación-Reducción , Conformación Proteica
18.
Nat Commun ; 12(1): 4621, 2021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34330928

RESUMEN

Cytochromes bd are ubiquitous amongst prokaryotes including many human-pathogenic bacteria. Such complexes are targets for the development of antimicrobial drugs. However, an understanding of the relationship between the structure and functional mechanisms of these oxidases is incomplete. Here, we have determined the 2.8 Å structure of Mycobacterium smegmatis cytochrome bd by single-particle cryo-electron microscopy. This bd oxidase consists of two subunits CydA and CydB, that adopt a pseudo two-fold symmetrical arrangement. The structural topology of its Q-loop domain, whose function is to bind the substrate, quinol, is significantly different compared to the C-terminal region reported for cytochromes bd from Geobacillus thermodenitrificans (G. th) and Escherichia coli (E. coli). In addition, we have identified two potential oxygen access channels in the structure and shown that similar tunnels also exist in G. th and E. coli cytochromes bd. This study provides insights to develop a framework for the rational design of antituberculosis compounds that block the oxygen access channels of this oxidase.


Asunto(s)
Proteínas Bacterianas/ultraestructura , Microscopía por Crioelectrón/métodos , Grupo Citocromo b/ultraestructura , Proteínas del Complejo de Cadena de Transporte de Electrón/ultraestructura , Mycobacterium smegmatis/enzimología , Oxidorreductasas/ultraestructura , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Grupo Citocromo b/química , Grupo Citocromo b/metabolismo , Transporte de Electrón , Proteínas del Complejo de Cadena de Transporte de Electrón/química , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Hemo/química , Hemo/metabolismo , Modelos Moleculares , Mycobacterium smegmatis/genética , Oxidorreductasas/química , Oxidorreductasas/metabolismo , Oxígeno/metabolismo , Conformación Proteica , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura , Especificidad por Sustrato
19.
Biochim Biophys Acta Bioenerg ; 1862(9): 148450, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34022199

RESUMEN

Cytochrome ba3 from Thermus thermophilus belongs to the B family of heme-copper oxidases and pumps protons across the membrane with an as yet unknown mechanism. The K channel of the A family heme-copper oxidases provides delivery of a substrate proton from the internal water phase to the binuclear heme-copper center (BNC) during the reductive phase of the catalytic cycle, while the D channel is responsible for transferring both substrate and pumped protons. By contrast, in the B family oxidases there is no D-channel and the structural equivalent of the K channel seems to be responsible for the transfer of both categories of protons. Here we have studied the effect of the T315V substitution in the K channel on the kinetics of membrane potential generation coupled to the oxidative half-reaction of the catalytic cycle of cytochrome ba3. The results suggest that the mutated enzyme does not pump protons during the reaction of the fully reduced form with molecular oxygen in a single turnover. Specific inhibition of proton pumping in the T315V mutant appears to be a consequence of inability to provide rapid (τ ~ 100 µs) reprotonation of the internal transient proton donor(s) of the K channel. In contrast to the A family, the K channel of the B-type oxidases is necessary for the electrogenic transfer of both pumped and substrate protons during the oxidative half-reaction of the catalytic cycle.


Asunto(s)
Grupo Citocromo b/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Proteínas Mutantes/metabolismo , Canales de Potasio/metabolismo , Bombas de Protones/metabolismo , Thermus thermophilus/metabolismo , Hemo/metabolismo , Modelos Moleculares , Mutación , Oxidación-Reducción , Oxidorreductasas/metabolismo , Oxígeno/metabolismo , Unión Proteica , Conformación Proteica
20.
Biochim Biophys Acta Bioenerg ; 1862(8): 148436, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-33940039

RESUMEN

Cytochrome bd oxidase is a bacterial terminal oxygen reductase that was suggested to enable adaptation to different environments and to confer resistance to stress conditions. An electrocatalytic study of the cyt bd oxidases from Escherichia coli, Corynebacterium glutamicum and Geobacillus thermodenitrificans gives evidence for a different reactivity towards oxygen. An inversion of the redox potential values of the three hemes is found when comparing the enzymes from different bacteria. This inversion can be correlated with different protonated glutamic acids as evidenced by reaction induced FTIR spectroscopy. The influence of the microenvironment of the hemes on the reactivity towards oxygen is discussed.


Asunto(s)
Corynebacterium glutamicum/enzimología , Grupo Citocromo b/metabolismo , Electrodos , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/enzimología , Geobacillus/enzimología , Oxidorreductasas/metabolismo , Oxígeno/metabolismo , Catálisis , Oxígeno/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...