Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.830
Filter
1.
Int J Mol Sci ; 25(2)2024 Jan 20.
Article in English | MEDLINE | ID: mdl-38279276

ABSTRACT

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.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Carbon Monoxide/pharmacology , Carbon Monoxide/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Cytochrome b Group/genetics , Cytochrome b Group/metabolism , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , Cytochromes/genetics , Cytochromes/metabolism , Oxidation-Reduction , Oxidoreductases/genetics , Oxidoreductases/metabolism , Respiration
2.
PLoS One ; 18(12): e0291564, 2023.
Article in English | MEDLINE | ID: mdl-38039324

ABSTRACT

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.


Subject(s)
Drosophila melanogaster , Oxidoreductases , Animals , Humans , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Phylogeny , Oxidoreductases/metabolism , Cytochrome b Group/genetics , Cytochrome b Group/metabolism , Iron/metabolism
3.
J Biol Chem ; 299(3): 102968, 2023 03.
Article in English | MEDLINE | ID: mdl-36736898

ABSTRACT

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.


Subject(s)
Chlamydomonas reinhardtii , Cytochrome b Group , Photosystem II Protein Complex , Rubredoxins , Cytochrome b Group/genetics , Cytochrome b Group/metabolism , Heme/metabolism , Iron/metabolism , Photosystem II Protein Complex/genetics , Photosystem II Protein Complex/metabolism , Rubredoxins/metabolism , Chlamydomonas reinhardtii/genetics , Chlamydomonas reinhardtii/metabolism
4.
Cell Death Dis ; 14(1): 1, 2023 01 02.
Article in English | MEDLINE | ID: mdl-36593242

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms , Cytochrome b Group , DEAD-box RNA Helicases , MicroRNAs , Humans , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/pathology , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Down-Regulation , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Metastasis , Cytochrome b Group/genetics , Cytochrome b Group/metabolism
5.
Biochim Biophys Acta Bioenerg ; 1864(2): 148952, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36535430

ABSTRACT

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.


Subject(s)
Escherichia coli Proteins , Oxidoreductases , Oxidoreductases/metabolism , Escherichia coli , Cytochromes/chemistry , Protons , Escherichia coli Proteins/metabolism , Cytochrome b Group/genetics , Electron Transport Chain Complex Proteins/metabolism , Electron Transport Complex IV , Hydrogen-Ion Concentration
6.
FEBS Lett ; 597(4): 547-556, 2023 02.
Article in English | MEDLINE | ID: mdl-36460943

ABSTRACT

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.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Cytochrome b Group/genetics , Cytochrome b Group/chemistry , Cytochromes/genetics , Cytochromes/chemistry , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Heme
7.
FEBS Lett ; 596(18): 2418-2424, 2022 09.
Article in English | MEDLINE | ID: mdl-36029102

ABSTRACT

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.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Cytochrome b Group/genetics , Cytochrome b Group/metabolism , Cytochromes/chemistry , Electron Transport Chain Complex Proteins/metabolism , Electron Transport Complex IV/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Oxidoreductases/metabolism , Oxygen/metabolism , Protons , Water/metabolism
8.
Int J Mol Sci ; 23(3)2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35162964

ABSTRACT

The branched aerobic respiratory chain in Bacillus cereus comprises three terminal oxidases: cytochromes aa3, caa3, and bd. Cytochrome caa3 requires heme A for activity, which is produced from heme O by heme A synthase (CtaA). In this study, we deleted the ctaA gene in B. cereus AH187 strain, this deletion resulted in loss of cytochrome caa3 activity. Proteomics data indicated that B. cereus grown in glucose-containing medium compensates for the loss of cytochrome caa3 activity by remodeling its respiratory metabolism. This remodeling involves up-regulation of cytochrome aa3 and several proteins involved in redox stress response-to circumvent sub-optimal respiratory metabolism. CtaA deletion changed the surface-composition of B. cereus, affecting its motility, autoaggregation phenotype, and the kinetics of biofilm formation. Strikingly, proteome remodeling made the ctaA mutant more resistant to cold and exogenous oxidative stresses compared to its parent strain. Consequently, we hypothesized that ctaA inactivation could improve B. cereus fitness in a nutrient-limited environment.


Subject(s)
Bacillus cereus/growth & development , Bacterial Proteins/genetics , Cytochrome b Group/genetics , Cytochrome c Group/metabolism , Cytochromes a3/metabolism , Cytochromes a/metabolism , Gene Deletion , Membrane Proteins/genetics , Bacillus cereus/genetics , Bacillus cereus/metabolism , Bacterial Proteins/metabolism , Biofilms/growth & development , Electron Transport Complex IV/metabolism , Heme/analogs & derivatives , Heme/metabolism , Oxidative Stress , Phenotype , Proteomics , Signal Transduction
9.
New Phytol ; 233(2): 766-780, 2022 01.
Article in English | MEDLINE | ID: mdl-34625967

ABSTRACT

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.


Subject(s)
Photosystem II Protein Complex , Synechocystis , Cytochrome b Group/genetics , Cytochrome b Group/metabolism , Cytochromes b/genetics , Cytochromes b/metabolism , Gene Amplification , Photosystem II Protein Complex/genetics , Photosystem II Protein Complex/metabolism , Synechocystis/metabolism
10.
Proc Natl Acad Sci U S A ; 118(50)2021 12 14.
Article in English | MEDLINE | ID: mdl-34873041

ABSTRACT

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.


Subject(s)
Cytochrome b Group/metabolism , Electron Transport Chain Complex Proteins/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/physiology , Oxidoreductases/metabolism , Cytochrome b Group/genetics , Electron Transport Chain Complex Proteins/genetics , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Models, Molecular , Oxidoreductases/genetics , Protein Conformation , Protein Isoforms
11.
J Clin Lab Anal ; 35(12): e24044, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34714955

ABSTRACT

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.


Subject(s)
Adenocarcinoma of Lung/pathology , Cytochrome b Group/genetics , Focal Adhesion Kinase 1/metabolism , Lung Neoplasms/pathology , MicroRNAs/genetics , Oxidoreductases/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/mortality , Adult , Aged , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cytochrome b Group/metabolism , Epithelial-Mesenchymal Transition/physiology , Focal Adhesion Kinase 1/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Mice, Inbred BALB C , Middle Aged , Oxidoreductases/metabolism , Signal Transduction/genetics , Xenograft Model Antitumor Assays
12.
Proc Natl Acad Sci U S A ; 118(33)2021 08 17.
Article in English | MEDLINE | ID: mdl-34389670

ABSTRACT

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.


Subject(s)
Cytochrome b Group/metabolism , Electron Transport Complex III/metabolism , Mitochondria/metabolism , Rhodobacter capsulatus/metabolism , Antimycin A/analogs & derivatives , Cytochrome b Group/genetics , Electron Spin Resonance Spectroscopy , Electron Transport Complex III/genetics , Hydrogen Bonding , Models, Molecular , Mutation , Oxidation-Reduction , Protein Conformation , Spectrum Analysis/methods
13.
Microb Cell Fact ; 20(1): 157, 2021 Aug 14.
Article in English | MEDLINE | ID: mdl-34391414

ABSTRACT

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.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cytochrome b Group/genetics , Cytochrome b Group/metabolism , Ferritins/genetics , Ferritins/metabolism , Insecticides/metabolism , Iron/metabolism , Macrolides/metabolism , Saccharopolyspora/metabolism , Bacterial Proteins/pharmacology , Cytochrome b Group/pharmacology , Ferritins/pharmacology , Genetic Engineering , Macrolides/classification , Proteomics , Saccharopolyspora/drug effects , Saccharopolyspora/genetics , Saccharopolyspora/growth & development
14.
Int J Mol Sci ; 22(16)2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34445407

ABSTRACT

Haematopoiesis is a paradigm of cell differentiation because of the wide variety and overwhelming number of mature blood cells produced daily. Under stress conditions, the organism must adapt to a boosted demand for blood cells. Chronic granulomatous disease (CGD) is a genetic disease caused by inactivating mutations that affect the phagocyte oxidase. Besides a defective innate immune system, CGD patients suffer from recurrent hyper-inflammation episodes, circumstances upon which they must face emergency haematopoiesis. The targeting of Cybb and Ncf1 genes have produced CGD animal models that are a useful surrogate when studying the pathophysiology and treatment of this disease. Here, we show that Cyba-/- mice spontaneously develop granuloma and, therefore, constitute a CGD animal model to complement the existing Cybb-/- and Ncf1-/- models. More importantly, we have analysed haematopoiesis in granuloma-bearing Cyba-/- mice. These animals showed a significant loss of weight, developed remarkable splenomegaly, bone marrow myeloid hyperplasia, and signs of anaemia. Haematological analyses showed a sharped decrease of B-cells and a striking development of myeloid cells in all compartments. Collectively, our results show that granuloma inflammatory lesions dramatically change haematopoiesis homeostasis. Consequently, we suggest that besides their defective innate immunity, the alteration of haematopoiesis homeostasis upon granuloma may contribute to the dismal outcome of CGD.


Subject(s)
B-Lymphocytes/metabolism , Cytochrome b Group/genetics , Granulomatous Disease, Chronic/pathology , Myeloid Cells/pathology , NADPH Oxidases/genetics , Animals , CRISPR-Cas Systems , Cell Lineage , Disease Models, Animal , Female , Gene Knockdown Techniques , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/immunology , Humans , Hyperplasia , Male , Mice , Myeloid Cells/immunology
15.
Int J Mol Sci ; 22(11)2021 Jun 05.
Article in English | MEDLINE | ID: mdl-34198827

ABSTRACT

The objective of this study was to investigate molecular mechanisms underlying the ability of carnosic acid to attenuate an early increase in reactive oxygen species (ROS) levels during MDI-induced adipocyte differentiation. The levels of superoxide anion and ROS were determined using dihydroethidium (DHE) and 2'-7'-dichlorofluorescin diacetate (DCFH-DA), respectively. Both superoxide anion and ROS levels peaked on the second day of differentiation. They were suppressed by carnosic acid. Carnosic acid attenuates the translation of NADPH (nicotinamide adenine dinucleotide phosphate) oxidase 4 (Nox4), p47phox, and p22phox, and the phosphorylation of nuclear factor-kappa B (NF-κB) and NF-κB inhibitor (IkBa). The translocation of NF-κB into the nucleus was also decreased by carnosic acid. In addition, carnosic acid increased the translation of heme oxygenase-1 (HO-1), γ-glutamylcysteine synthetase (γ-GCSc), and glutathione S-transferase (GST) and both the translation and nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2). Taken together, these results indicate that carnosic acid could down-regulate ROS level in an early stage of MPI-induced adipocyte differentiation by attenuating ROS generation through suppression of NF-κB-mediated translation of Nox4 enzyme and increasing ROS neutralization through induction of Nrf2-mediated translation of phase II antioxidant enzymes such as HO-1, γ-GCS, and GST, leading to its anti-adipogenetic effect.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , Abietanes/pharmacology , DNA Helicases/genetics , Heme Oxygenase-1/genetics , Membrane Proteins/genetics , NADPH Oxidase 4/genetics , NF-KappaB Inhibitor alpha/genetics , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Antioxidants/pharmacology , Cell Differentiation/drug effects , Cytochrome b Group/genetics , Ethidium/analogs & derivatives , Ethidium/pharmacology , Fluoresceins/pharmacology , Glutathione Transferase/genetics , Mice , NADPH Oxidases/genetics , Protein Biosynthesis/drug effects , Reactive Oxygen Species/metabolism
16.
ISME J ; 15(12): 3534-3548, 2021 12.
Article in English | MEDLINE | ID: mdl-34145390

ABSTRACT

Cytochrome bd-type oxygen reductases (cytbd) belong to one of three enzyme superfamilies that catalyze oxygen reduction to water. They are widely distributed in Bacteria and Archaea, but the full extent of their biochemical diversity is unknown. Here we used phylogenomics to identify three families and several subfamilies within the cytbd superfamily. The core architecture shared by all members of the superfamily consists of four transmembrane helices that bind two active site hemes, which are responsible for oxygen reduction. While previously characterized cytochrome bd-type oxygen reductases use quinol as an electron donor to reduce oxygen, sequence analysis shows that only one of the identified families has a conserved quinol binding site. The other families are missing this feature, suggesting that they use an alternative electron donor. Multiple gene duplication events were identified within the superfamily, resulting in significant evolutionary and structural diversity. The CydAA' cytbd, found exclusively in Archaea, is formed by the co-association of two superfamily paralogs. We heterologously expressed CydAA' from Caldivirga maquilingensis and demonstrated that it performs oxygen reduction with quinol as an electron donor. Strikingly, CydAA' is the first isoform of cytbd containing only b-type hemes shown to be active when isolated from membranes, demonstrating that oxygen reductase activity in this superfamily is not dependent on heme d.


Subject(s)
Archaea/genetics , Archaeal Proteins/genetics , Cytochrome b Group/genetics , Electron Transport Chain Complex Proteins/genetics , Oxidoreductases , Archaea/enzymology , Evolution, Molecular , Oxidation-Reduction , Oxidoreductases/genetics , Oxygen
17.
NPJ Biofilms Microbiomes ; 7(1): 35, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33863914

ABSTRACT

Nutrient gradients in biofilms cause bacteria to organize into metabolically versatile communities capable of withstanding threats from external agents including bacteriophages, phagocytes, and antibiotics. We previously determined that oxygen availability spatially organizes respiration in uropathogenic Escherichia coli biofilms, and that the high-affinity respiratory quinol oxidase cytochrome bd is necessary for extracellular matrix production and biofilm development. In this study we investigate the physiologic consequences of cytochrome bd deficiency in biofilms and determine that loss of cytochrome bd induces a biofilm-specific increase in expression of general diffusion porins, leading to elevated outer membrane permeability. In addition, loss of cytochrome bd impedes the proton mediated efflux of noxious chemicals by diminishing respiratory flux. As a result, loss of cytochrome bd enhances cellular accumulation of noxious chemicals and increases biofilm susceptibility to antibiotics. These results identify an undescribed link between E. coli biofilm respiration and stress tolerance, while suggesting the possibility of inhibiting cytochrome bd as an antibiofilm therapeutic approach.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Cytochrome b Group/genetics , Drug Resistance, Bacterial , Electron Transport Chain Complex Proteins/genetics , Escherichia coli Proteins/genetics , Oxidoreductases/genetics , Uropathogenic Escherichia coli/physiology , Alleles , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Biofilms/growth & development , Cytochrome b Group/metabolism , Dose-Response Relationship, Drug , Electron Transport Chain Complex Proteins/metabolism , Escherichia coli Proteins/metabolism , Gene Knockdown Techniques , Microbial Sensitivity Tests , Oxidoreductases/metabolism , Plankton/drug effects , Plankton/genetics , Uropathogenic Escherichia coli/drug effects
18.
Nutrients ; 13(3)2021 Feb 27.
Article in English | MEDLINE | ID: mdl-33673530

ABSTRACT

Anemia is a frequent finding in children with celiac disease but the detailed pathophysiological mechanisms in the intestine remain obscure. One possible explanation could be an abnormal expression of duodenal iron transport proteins. However, the results have so far been inconsistent. We investigated this issue by comparing immunohistochemical stainings of duodenal cytochrome B (DCYTB), divalent metal transporter 1 (DMT1), ferroportin, hephaestin and transferrin receptor 1 (TfR1) in duodenal biopsies between 27 children with celiac disease and duodenal atrophy, 10 celiac autoantibody-positive children with potential celiac disease and six autoantibody-negative control children. Twenty out of these 43 subjects had anemia. The expressions of the iron proteins were investigated with regard to saturation and the percentage of the stained area or stained membrane length of the enterocytes. The results showed the stained area of ferroportin to be increased and the saturation of hephaestin to be decreased in celiac disease patients compared with controls. There were no differences in the transporter protein expressions between anemic and non-anemic patients. The present results suggest an iron status-independent alteration of ferroportin and hephaestin proteins in children with histologically confirmed celiac disease.


Subject(s)
Antigens, CD/metabolism , Cation Transport Proteins/metabolism , Celiac Disease/metabolism , Cytochrome b Group/metabolism , Membrane Proteins/metabolism , Oxidoreductases/metabolism , Receptors, Transferrin/metabolism , Transcription Factors/metabolism , Adolescent , Anemia, Iron-Deficiency/complications , Anemia, Iron-Deficiency/metabolism , Antigens, CD/genetics , Cation Transport Proteins/genetics , Celiac Disease/complications , Child , Child, Preschool , Cytochrome b Group/genetics , Female , Gene Expression Regulation/physiology , Humans , Male , Membrane Proteins/genetics , Oxidoreductases/genetics , Receptors, Transferrin/genetics , Transcription Factors/genetics
19.
mBio ; 12(1)2021 01 19.
Article in English | MEDLINE | ID: mdl-33468700

ABSTRACT

5-Aminosalicylic acid (5-ASA), a peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist, is a widely used first-line medication for the treatment of ulcerative colitis, but its anti-inflammatory mechanism is not fully resolved. Here, we show that 5-ASA ameliorates colitis in dextran sulfate sodium (DSS)-treated mice by activating PPAR-γ signaling in the intestinal epithelium. DSS-induced colitis was associated with a loss of epithelial hypoxia and a respiration-dependent luminal expansion of Escherichia coli, which could be ameliorated by treatment with 5-ASA. However, 5-ASA was no longer able to reduce inflammation, restore epithelial hypoxia, or blunt an expansion of E. coli in DSS-treated mice that lacked Pparg expression specifically in the intestinal epithelium. These data suggest that the anti-inflammatory activity of 5-ASA requires activation of epithelial PPAR-γ signaling, thus pointing to the intestinal epithelium as a potential target for therapeutic intervention in ulcerative colitis.IMPORTANCE An expansion of Enterobacterales in the fecal microbiota is a microbial signature of dysbiosis that is linked to many noncommunicable diseases, including ulcerative colitis. Here, we used Escherichia coli, a representative of the Enterobacterales, to show that its dysbiotic expansion during colitis can be remediated by modulating host epithelial metabolism. Dextran sulfate sodium (DSS)-induced colitis reduced mitochondrial activity in the colonic epithelium, thereby increasing the amount of oxygen available to fuel an E. coli expansion through aerobic respiration. Activation of epithelial peroxisome proliferator-activated receptor gamma (PPAR-γ) signaling with 5-aminosalicylic acid (5-ASA) was sufficient to restore mitochondrial activity and blunt a dysbiotic E. coli expansion. These data identify the host's epithelial metabolism as a potential treatment target to remediate microbial signatures of dysbiosis, such as a dysbiotic E. coli expansion in the fecal microbiota.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Colitis/drug therapy , Dysbiosis/drug therapy , Escherichia coli/drug effects , Mesalamine/pharmacology , PPAR gamma/genetics , Animals , Colitis/genetics , Colitis/microbiology , Colitis/pathology , Colon/drug effects , Colon/microbiology , Colon/pathology , Cytochrome b Group/genetics , Cytochrome b Group/metabolism , Dextran Sulfate/administration & dosage , Dysbiosis/genetics , Dysbiosis/microbiology , Dysbiosis/pathology , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/pathogenicity , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Female , Gene Expression Regulation , Inflammation , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Nitrate Reductase/genetics , Nitrate Reductase/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , PPAR gamma/agonists , PPAR gamma/metabolism , Treatment Outcome
20.
Antioxid Redox Signal ; 34(16): 1280-1318, 2021 06 01.
Article in English | MEDLINE | ID: mdl-32924537

ABSTRACT

Significance: Cytochrome bd is a ubiquinol:oxygen oxidoreductase of many prokaryotic respiratory chains with a unique structure and functional characteristics. Its primary role is to couple the reduction of molecular oxygen, even at submicromolar concentrations, to water with the generation of a proton motive force used for adenosine triphosphate production. Cytochrome bd is found in many bacterial pathogens and, surprisingly, in bacteria formally denoted as anaerobes. It endows bacteria with resistance to various stressors and is a potential drug target. Recent Advances: We summarize recent advances in the biochemistry, structure, and physiological functions of cytochrome bd in the light of exciting new three-dimensional structures of the oxidase. The newly discovered roles of cytochrome bd in contributing to bacterial protection against hydrogen peroxide, nitric oxide, peroxynitrite, and hydrogen sulfide are assessed. Critical Issues: Fundamental questions remain regarding the precise delineation of electron flow within this multihaem oxidase and how the extraordinarily high affinity for oxygen is accomplished, while endowing bacteria with resistance to other small ligands. Future Directions: It is clear that cytochrome bd is unique in its ability to confer resistance to toxic small molecules, a property that is significant for understanding the propensity of pathogens to possess this oxidase. Since cytochrome bd is a uniquely bacterial enzyme, future research should focus on harnessing fundamental knowledge of its structure and function to the development of novel and effective antibacterial agents.


Subject(s)
Bacteria/growth & development , Cytochrome b Group/chemistry , Cytochrome b Group/metabolism , Cytochrome d Group/chemistry , Cytochrome d Group/metabolism , Bacteria/enzymology , Bacteria/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cytochrome b Group/genetics , Cytochrome d Group/genetics , Drug Resistance, Bacterial , Gene Expression Regulation, Bacterial , Models, Molecular , Multigene Family , Protein Conformation , Stress, Physiological
SELECTION OF CITATIONS
SEARCH DETAIL
...