Regulation of Eukaryotic Cytochrome c Oxidase

A topical collection in Cells (ISSN 2073-4409).

Viewed by 35072

Editors


E-Mail Website
Collection Editor
Department of Chemistry / Biochemistry, Philipps-University Marburg, Hans-Meerwein-Strasse, D-35032 Marburg, Germany
Interests: respiration; mitochondria; cytochrome c oxidase; calcium signaling

E-Mail Website
Collection Editor
1. Cardiovascular Research Lab, Biochemical Pharmacological Center, Philipps-University Marburg, Karl-von-Frisch-Strasse 2, D-35043 Marburg, Germany
2. Department of Heart Surgery, The University Hospital of Giessen and Marburg, Location Marburg, Baldinger Strasse 1, D-35043 Marburg, Germany
Interests: physiological and pathophysiological role of mitochondria in the heart; function and dysfunction of cytochrome c oxidase; pharmaco-bioenergetics; structural and functional organization of mitochondrial respiratory chain

E-Mail Website
Collection Editor
1. Cardiovascular Research Lab, Biochemical Pharmacological Center, Philipps-University Marburg, Karl-von-Frisch-Strasse 2, D-35043 Marburg, Germany
2. Department of Heart Surgery, The University Hospital of Giessen and Marburg, Location Marburg, Baldinger Strasse 1, D-35043 Marburg, Germany
Interests: metabolism in the ischemic heart; mitochondrial respiration; cytochrome c oxidase in stress; myocardprotection during heart surgery; pharmaco-bioenergetics

Topical Collection Information

Dear Colleagues,

A Topical Collection on “Regulation of Eucaryotic Cytochrome c oxidase” is planned to be published by the journal “Cells” this year. In this issue we intend to compile papers on the regulation of eucaryotic cytochrome c oxidase (CytOx) and mitochondrial respiration. Hence, we prefer to include papers presenting the regulation of CytOx activity having physiological significance. The detailed mechanisms of electron and proton transfer within the mitochondrially-encoded catalytic subunits I–III, which are similar to bacterial CytOx subunits I–III, will not be covered. Data on the regulation of CytOx activity by interaction with low molecular weight compounds (e.g. adenine nucleotides, calcium, zinc, di-iodothyronine) and with proteins (r.g., NDUFA4, Higda-1), regulation by expression of subunit isozymes, by phosphorylation and other chemical modifications, by supercomplex formation (e.g. respirasome) are welcome. In addition influence of subunit overexpression during cancerogenesis are wellcome.

Prof. Dr. Bernhard Kadenbach
Dr. Rabia Ramzan
Prof. Dr. Sebastian Vogt
Collection Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the collection website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • phosphorylation of CytOx
  • regulation of mitochondrial respiration
  • expression of CytOx isozymes
  • overexpression of CytOx subunits during cancerogenesis
  • CytOx regulation in supercomplexes
  • CytOx dimerization/monomerization
  • allosteric ATP-inhibition of CytOx

Published Papers (11 papers)

2022

Jump to: 2021, 2020

15 pages, 2602 KiB  
Article
Cytochrome c Oxidase Inhibition by ATP Decreases Mitochondrial ROS Production
by Rabia Ramzan, Amalia M. Dolga, Susanne Michels, Petra Weber, Carsten Culmsee, Ardawan J. Rastan and Sebastian Vogt
Cells 2022, 11(6), 992; https://doi.org/10.3390/cells11060992 - 14 Mar 2022
Cited by 9 | Viewed by 3255
Abstract
This study addresses the eventual consequence of cytochrome c oxidase (CytOx) inhibition by ATP at high ATP/ADP ratio in isolated rat heart mitochondria. Earlier, it has been demonstrated that the mechanism of allosteric ATP inhibition of CytOx is one of the key regulations [...] Read more.
This study addresses the eventual consequence of cytochrome c oxidase (CytOx) inhibition by ATP at high ATP/ADP ratio in isolated rat heart mitochondria. Earlier, it has been demonstrated that the mechanism of allosteric ATP inhibition of CytOx is one of the key regulations of mitochondrial functions. It is relevant that aiming to maintain a high ATP/ADP ratio for the measurement of CytOx activity effectuating the enzymatic inhibition as well as mitochondrial respiration, optimal concentration of mitochondria is critically important. Likewise, only at this concentration, were the differences in ΔΨm and ROS concentrations measured under various conditions significant. Moreover, when CytOx activity was inhibited in the presence of ATP, mitochondrial respiration and ΔΨm both remained static, while the ROS production was markedly decreased. Consubstantial results were found when the electron transport chain was inhibited by antimycin A, letting only CytOx remain functional to support the energy production. This seems to corroborate that the decrease in mitochondrial ROS production is solely the effect of ATP binding to CytOx which results in static respiration as well as membrane potential. Full article
Show Figures

Figure 1

2021

Jump to: 2022, 2020

14 pages, 1986 KiB  
Article
Cholate Disrupts Regulatory Functions of Cytochrome c Oxidase
by Rabia Ramzan, Jörg Napiwotzki, Petra Weber, Bernhard Kadenbach and Sebastian Vogt
Cells 2021, 10(7), 1579; https://doi.org/10.3390/cells10071579 - 23 Jun 2021
Cited by 4 | Viewed by 2044
Abstract
Cytochrome c oxidase (CytOx), the oxygen-accepting and rate-limiting enzyme of mitochondrial respiration, binds with 10 molecules of ADP, 7 of which are exchanged by ATP at high ATP/ADP-ratios. These bound ATP and ADP can be exchanged by cholate, which is generally used for [...] Read more.
Cytochrome c oxidase (CytOx), the oxygen-accepting and rate-limiting enzyme of mitochondrial respiration, binds with 10 molecules of ADP, 7 of which are exchanged by ATP at high ATP/ADP-ratios. These bound ATP and ADP can be exchanged by cholate, which is generally used for the purification of CytOx. Many crystal structures of isolated CytOx were performed with the enzyme isolated from mitochondria using sodium cholate as a detergent. Cholate, however, dimerizes the enzyme isolated in non-ionic detergents and induces a structural change as evident from a spectral change. Consequently, it turns off the “allosteric ATP-inhibition of CytOx”, which is reversibly switched on under relaxed conditions via cAMP-dependent phosphorylation and keeps the membrane potential and ROS formation in mitochondria at low levels. This cholate effect gives an insight into the structural-functional relationship of the enzyme with respect to ATP inhibition and its role in mitochondrial respiration and energy production. Full article
Show Figures

Figure 1

22 pages, 809 KiB  
Review
Regulation of Cytochrome c Oxidase by Natural Compounds Resveratrol, (–)-Epicatechin, and Betaine
by Icksoo Lee
Cells 2021, 10(6), 1346; https://doi.org/10.3390/cells10061346 - 29 May 2021
Cited by 9 | Viewed by 3442
Abstract
Numerous naturally occurring molecules have been studied for their beneficial health effects. Many compounds have received considerable attention for their potential medical uses. Among them, several substances have been found to improve mitochondrial function. This review focuses on resveratrol, (–)-epicatechin, and betaine and [...] Read more.
Numerous naturally occurring molecules have been studied for their beneficial health effects. Many compounds have received considerable attention for their potential medical uses. Among them, several substances have been found to improve mitochondrial function. This review focuses on resveratrol, (–)-epicatechin, and betaine and summarizes the published data pertaining to their effects on cytochrome c oxidase (COX) which is the terminal enzyme of the mitochondrial electron transport chain and is considered to play an important role in the regulation of mitochondrial respiration. In a variety of experimental model systems, these compounds have been shown to improve mitochondrial biogenesis in addition to increased COX amount and/or its enzymatic activity. Given that they are inexpensive, safe in a wide range of concentrations, and effectively improve mitochondrial and COX function, these compounds could be attractive enough for possible therapeutic or health improvement strategies. Full article
Show Figures

Figure 1

13 pages, 1163 KiB  
Review
Multiple Mechanisms Regulate Eukaryotic Cytochrome C Oxidase
by Rabia Ramzan, Bernhard Kadenbach and Sebastian Vogt
Cells 2021, 10(3), 514; https://doi.org/10.3390/cells10030514 - 28 Feb 2021
Cited by 22 | Viewed by 4159
Abstract
Cytochrome c oxidase (COX), the rate-limiting enzyme of mitochondrial respiration, is regulated by various mechanisms. Its regulation by ATP (adenosine triphosphate) appears of particular importance, since it evolved early during evolution and is still found in cyanobacteria, but not in other bacteria. Therefore [...] Read more.
Cytochrome c oxidase (COX), the rate-limiting enzyme of mitochondrial respiration, is regulated by various mechanisms. Its regulation by ATP (adenosine triphosphate) appears of particular importance, since it evolved early during evolution and is still found in cyanobacteria, but not in other bacteria. Therefore the “allosteric ATP inhibition of COX” is described here in more detail. Most regulatory properties of COX are related to “supernumerary” subunits, which are largely absent in bacterial COX. The “allosteric ATP inhibition of COX” was also recently described in intact isolated rat heart mitochondria. Full article
Show Figures

Figure 1

25 pages, 2107 KiB  
Review
Cytochrome c Oxidase at Full Thrust: Regulation and Biological Consequences to Flying Insects
by Rafael D. Mesquita, Alessandro Gaviraghi, Renata L.S. Gonçalves, Marcos A. Vannier-Santos, Julio A. Mignaco, Carlos Frederico L. Fontes, Luciana E.S.F. Machado and Marcus F. Oliveira
Cells 2021, 10(2), 470; https://doi.org/10.3390/cells10020470 - 22 Feb 2021
Cited by 3 | Viewed by 3893
Abstract
Flight dispersal represents a key aspect of the evolutionary and ecological success of insects, allowing escape from predators, mating, and colonization of new niches. The huge energy demand posed by flight activity is essentially met by oxidative phosphorylation (OXPHOS) in flight muscle mitochondria. [...] Read more.
Flight dispersal represents a key aspect of the evolutionary and ecological success of insects, allowing escape from predators, mating, and colonization of new niches. The huge energy demand posed by flight activity is essentially met by oxidative phosphorylation (OXPHOS) in flight muscle mitochondria. In insects, mitochondrial ATP supply and oxidant production are regulated by several factors, including the energy demand exerted by changes in adenylate balance. Indeed, adenylate directly regulates OXPHOS by targeting both chemiosmotic ATP production and the activities of specific mitochondrial enzymes. In several organisms, cytochrome c oxidase (COX) is regulated at transcriptional, post-translational, and allosteric levels, impacting mitochondrial energy metabolism, and redox balance. This review will present the concepts on how COX function contributes to flying insect biology, focusing on the existing examples in the literature where its structure and activity are regulated not only by physiological and environmental factors but also how changes in its activity impacts insect biology. We also performed in silico sequence analyses and determined the structure models of three COX subunits (IV, VIa, and VIc) from different insect species to compare with mammalian orthologs. We observed that the sequences and structure models of COXIV, COXVIa, and COXVIc were quite similar to their mammalian counterparts. Remarkably, specific substitutions to phosphomimetic amino acids at critical phosphorylation sites emerge as hallmarks on insect COX sequences, suggesting a new regulatory mechanism of COX activity. Therefore, by providing a physiological and bioenergetic framework of COX regulation in such metabolically extreme models, we hope to expand the knowledge of this critical enzyme complex and the potential consequences for insect dispersal. Full article
Show Figures

Graphical abstract

19 pages, 3867 KiB  
Article
Loss of COX4I1 Leads to Combined Respiratory Chain Deficiency and Impaired Mitochondrial Protein Synthesis
by Kristýna Čunátová, David Pajuelo Reguera, Marek Vrbacký, Erika Fernández-Vizarra, Shujing Ding, Ian M. Fearnley, Massimo Zeviani, Josef Houštěk, Tomáš Mráček and Petr Pecina
Cells 2021, 10(2), 369; https://doi.org/10.3390/cells10020369 - 10 Feb 2021
Cited by 20 | Viewed by 3788
Abstract
The oxidative phosphorylation (OXPHOS) system localized in the inner mitochondrial membrane secures production of the majority of ATP in mammalian organisms. Individual OXPHOS complexes form supramolecular assemblies termed supercomplexes. The complexes are linked not only by their function but also by interdependency of [...] Read more.
The oxidative phosphorylation (OXPHOS) system localized in the inner mitochondrial membrane secures production of the majority of ATP in mammalian organisms. Individual OXPHOS complexes form supramolecular assemblies termed supercomplexes. The complexes are linked not only by their function but also by interdependency of individual complex biogenesis or maintenance. For instance, cytochrome c oxidase (cIV) or cytochrome bc1 complex (cIII) deficiencies affect the level of fully assembled NADH dehydrogenase (cI) in monomeric as well as supercomplex forms. It was hypothesized that cI is affected at the level of enzyme assembly as well as at the level of cI stability and maintenance. However, the true nature of interdependency between cI and cIV is not fully understood yet. We used a HEK293 cellular model where the COX4 subunit was completely knocked out, serving as an ideal system to study interdependency of cI and cIV, as early phases of cIV assembly process were disrupted. Total absence of cIV was accompanied by profound deficiency of cI, documented by decrease in the levels of cI subunits and significantly reduced amount of assembled cI. Supercomplexes assembled from cI, cIII, and cIV were missing in COX4I1 knock-out (KO) due to loss of cIV and decrease in cI amount. Pulse-chase metabolic labeling of mitochondrial DNA (mtDNA)-encoded proteins uncovered a decrease in the translation of cIV and cI subunits. Moreover, partial impairment of mitochondrial protein synthesis correlated with decreased content of mitochondrial ribosomal proteins. In addition, complexome profiling revealed accumulation of cI assembly intermediates, indicating that cI biogenesis, rather than stability, was affected. We propose that attenuation of mitochondrial protein synthesis caused by cIV deficiency represents one of the mechanisms, which may impair biogenesis of cI. Full article
Show Figures

Figure 1

27 pages, 4456 KiB  
Review
Regulation of COX Assembly and Function by Twin CX9C Proteins—Implications for Human Disease
by Stephanie Gladyck, Siddhesh Aras, Maik Hüttemann and Lawrence I. Grossman
Cells 2021, 10(2), 197; https://doi.org/10.3390/cells10020197 - 20 Jan 2021
Cited by 10 | Viewed by 2805
Abstract
Oxidative phosphorylation is a tightly regulated process in mammals that takes place in and across the inner mitochondrial membrane and consists of the electron transport chain and ATP synthase. Complex IV, or cytochrome c oxidase (COX), is the terminal enzyme of the electron [...] Read more.
Oxidative phosphorylation is a tightly regulated process in mammals that takes place in and across the inner mitochondrial membrane and consists of the electron transport chain and ATP synthase. Complex IV, or cytochrome c oxidase (COX), is the terminal enzyme of the electron transport chain, responsible for accepting electrons from cytochrome c, pumping protons to contribute to the gradient utilized by ATP synthase to produce ATP, and reducing oxygen to water. As such, COX is tightly regulated through numerous mechanisms including protein–protein interactions. The twin CX9C family of proteins has recently been shown to be involved in COX regulation by assisting with complex assembly, biogenesis, and activity. The twin CX9C motif allows for the import of these proteins into the intermembrane space of the mitochondria using the redox import machinery of Mia40/CHCHD4. Studies have shown that knockdown of the proteins discussed in this review results in decreased or completely deficient aerobic respiration in experimental models ranging from yeast to human cells, as the proteins are conserved across species. This article highlights and discusses the importance of COX regulation by twin CX9C proteins in the mitochondria via COX assembly and control of its activity through protein–protein interactions, which is further modulated by cell signaling pathways. Interestingly, select members of the CX9C protein family, including MNRR1 and CHCHD10, show a novel feature in that they not only localize to the mitochondria but also to the nucleus, where they mediate oxygen- and stress-induced transcriptional regulation, opening a new view of mitochondrial-nuclear crosstalk and its involvement in human disease. Full article
Show Figures

Graphical abstract

2020

Jump to: 2022, 2021

20 pages, 2224 KiB  
Review
HIGD-Driven Regulation of Cytochrome c Oxidase Biogenesis and Function
by Alba Timón-Gómez, Emma L. Bartley-Dier, Flavia Fontanesi and Antoni Barrientos
Cells 2020, 9(12), 2620; https://doi.org/10.3390/cells9122620 - 06 Dec 2020
Cited by 22 | Viewed by 3332
Abstract
The biogenesis and function of eukaryotic cytochrome c oxidase or mitochondrial respiratory chain complex IV (CIV) undergo several levels of regulation to adapt to changing environmental conditions. Adaptation to hypoxia and oxidative stress involves CIV subunit isoform switch, changes in phosphorylation status, and [...] Read more.
The biogenesis and function of eukaryotic cytochrome c oxidase or mitochondrial respiratory chain complex IV (CIV) undergo several levels of regulation to adapt to changing environmental conditions. Adaptation to hypoxia and oxidative stress involves CIV subunit isoform switch, changes in phosphorylation status, and modulation of CIV assembly and enzymatic activity by interacting factors. The latter include the Hypoxia Inducible Gene Domain (HIGD) family yeast respiratory supercomplex factors 1 and 2 (Rcf1 and Rcf2) and two mammalian homologs of Rcf1, the proteins HIGD1A and HIGD2A. Whereas Rcf1 and Rcf2 are expressed constitutively, expression of HIGD1A and HIGD2A is induced under stress conditions, such as hypoxia and/or low glucose levels. In both systems, the HIGD proteins localize in the mitochondrial inner membrane and play a role in the biogenesis of CIV as a free unit or as part as respiratory supercomplexes. Notably, they remain bound to assembled CIV and, by modulating its activity, regulate cellular respiration. Here, we will describe the current knowledge regarding the specific and overlapping roles of the several HIGD proteins in physiological and stress conditions. Full article
Show Figures

Figure 1

16 pages, 2384 KiB  
Review
The Interplay among Subunit Composition, Cardiolipin Content, and Aggregation State of Bovine Heart Cytochrome c Oxidase
by Erik Sedlák, Tibor Kožár and Andrey Musatov
Cells 2020, 9(12), 2588; https://doi.org/10.3390/cells9122588 - 03 Dec 2020
Cited by 4 | Viewed by 2313
Abstract
Mitochondrial cytochrome c oxidase (CcO) is a multisubunit integral membrane complex consisting of 13 dissimilar subunits, as well as three to four tightly bound molecules of cardiolipin (CL). The monomeric unit of CcO is able to form a dimer and participate in the [...] Read more.
Mitochondrial cytochrome c oxidase (CcO) is a multisubunit integral membrane complex consisting of 13 dissimilar subunits, as well as three to four tightly bound molecules of cardiolipin (CL). The monomeric unit of CcO is able to form a dimer and participate in the formation of supercomplexes in the inner mitochondrial membrane. The structural and functional integrity of the enzyme is crucially dependent on the full subunit complement and the presence of unperturbed bound CL. A direct consequence of subunit loss, CL removal, or its oxidative modification is the destabilization of the quaternary structure, loss of the activity, and the inability to dimerize. Thus, the intimate interplay between individual components of the complex is imperative for regulation of the CcO aggregation state. While it appears that the aggregation state of CcO might affect its conformational stability, the functional role of the aggregation remains unclear as both monomeric and dimeric forms of CcO seem to be fully active. Here, we review the current status of our knowledge with regard to the role of dimerization in the function and stability of CcO and factors, such as subunit composition, amphiphilic environment represented by phospholipids/detergents, and posttranslational modifications that play a role in the regulation of the CcO aggregation state. Full article
Show Figures

Figure 1

16 pages, 2713 KiB  
Article
Interaction of Cytochrome C Oxidase with Steroid Hormones
by Ilya P. Oleynikov, Natalia V. Azarkina, Tatiana V. Vygodina and Alexander A. Konstantinov
Cells 2020, 9(10), 2211; https://doi.org/10.3390/cells9102211 - 29 Sep 2020
Cited by 7 | Viewed by 2150
Abstract
Estradiol, testosterone and other steroid hormones inhibit cytochrome c oxidase (CcO) purified from bovine heart. The inhibition is strongly dependent on concentration of dodecyl-maltoside (DM) in the assay. The plots of Ki vs [DM] are linear for both estradiol and testosterone which [...] Read more.
Estradiol, testosterone and other steroid hormones inhibit cytochrome c oxidase (CcO) purified from bovine heart. The inhibition is strongly dependent on concentration of dodecyl-maltoside (DM) in the assay. The plots of Ki vs [DM] are linear for both estradiol and testosterone which may indicate an 1:1 stoichiometry competition between the hormones and the detergent. Binding of estradiol, but not of testosterone, brings about spectral shift of the oxidized CcO consistent with an effect on heme a33+. We presume that the hormones bind to CcO at the bile acid binding site described by Ferguson-Miller and collaborators. Estradiol is shown to inhibit intraprotein electron transfer between hemes a and a3. Notably, neither estradiol nor testosterone suppresses the peroxidase activity of CcO. Such a specific mode of action indicates that inhibition of CcO activity by the hormones is associated with impairing proton transfer via the K-proton channel. Full article
Show Figures

Figure 1

18 pages, 3519 KiB  
Article
Brain-Specific Serine-47 Modification of Cytochrome c Regulates Cytochrome c Oxidase Activity Attenuating ROS Production and Cell Death: Implications for Ischemia/Reperfusion Injury and Akt Signaling
by Hasini A. Kalpage, Junmei Wan, Paul T. Morse, Icksoo Lee and Maik Hüttemann
Cells 2020, 9(8), 1843; https://doi.org/10.3390/cells9081843 - 06 Aug 2020
Cited by 16 | Viewed by 2738
Abstract
We previously reported that serine-47 (S47) phosphorylation of cytochrome c (Cytc) in the brain results in lower cytochrome c oxidase (COX) activity and caspase-3 activity in vitro. We here analyze the effect of S47 modification in fibroblast cell lines stably expressing [...] Read more.
We previously reported that serine-47 (S47) phosphorylation of cytochrome c (Cytc) in the brain results in lower cytochrome c oxidase (COX) activity and caspase-3 activity in vitro. We here analyze the effect of S47 modification in fibroblast cell lines stably expressing S47E phosphomimetic Cytc, unphosphorylated WT, or S47A Cytc. Our results show that S47E Cytc results in partial inhibition of mitochondrial respiration corresponding with lower mitochondrial membrane potentials (ΔΨm) and reduced reactive oxygen species (ROS) production. When exposed to an oxygen-glucose deprivation/reoxygenation (OGD/R) model simulating ischemia/reperfusion injury, the Cytc S47E phosphomimetic cell line showed minimal ROS generation compared to the unphosphorylated WT Cytc cell line that generated high levels of ROS upon reoxygenation. Consequently, the S47E Cytc cell line also resulted in significantly lower cell death upon exposure to OGD/R, confirming the cytoprotective role of S47 phosphorylation of Cytc. S47E Cytc also resulted in lower cell death upon H2O2 treatment. Finally, we propose that pro-survival kinase Akt (protein kinase B) is a likely mediator of the S47 phosphorylation of Cytc in the brain. Akt inhibitor wortmannin abolished S47 phosphorylation of Cytc, while the Akt activator SC79 maintained S47 phosphorylation of Cytc. Overall, our results suggest that loss of S47 phosphorylation of Cytc during brain ischemia drives reperfusion injury through maximal electron transport chain flux, ΔΨm hyperpolarization, and ROS-triggered cell death. Full article
Show Figures

Graphical abstract

Back to TopTop