Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cells ; 10(7)2021 06 23.
Article in English | MEDLINE | ID: mdl-34201437

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 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.


Subject(s)
Cholates/pharmacology , Electron Transport Complex IV/metabolism , Adenosine Diphosphate/pharmacology , Animals , Cattle , Mitochondria, Heart/genetics , Mitochondria, Heart/metabolism , Rats , Spectrophotometry, Ultraviolet
2.
Cells ; 10(3)2021 02 28.
Article in English | MEDLINE | ID: mdl-33671025

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 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.


Subject(s)
Electron Transport Complex IV/metabolism , Eukaryota/metabolism , Animals , Rats
3.
Mitochondrion ; 58: 296-302, 2021 05.
Article in English | MEDLINE | ID: mdl-33069909

ABSTRACT

ATP, the universal energy currency in all living cells, is mainly synthesized in mitochondria by oxidative phosphorylation (OXPHOS). The final and rate limiting step of the respiratory chain is cytochrome c oxidase (COX) which represents the regulatory center of OXPHOS. COX is regulated through binding of various effectors to its "supernumerary" subunits, by reversible phosphorylation, and by expression of subunit isoforms. Of particular interest is its feedback inhibition by ATP, the final product of OXPHOS. This "allosteric ATP-inhibition" of phosphorylated and dimeric COX maintains a low and healthy mitochondrial membrane potential (relaxed state), and prevents the formation of ROS (reactive oxygen species) which are known to cause numerous diseases. Excessive work and stress abolish this feedback inhibition of COX by Ca2+-activated dephosphorylation which leads to monomerization and movement of NDUFA4 from complex I to COX with higher rates of COX activity and ATP synthesis (active state) but increased ROS formation and decreased efficiency.


Subject(s)
Electron Transport Complex IV/metabolism , Mitochondria/metabolism , Allosteric Regulation , Oxidative Phosphorylation
4.
World J Biol Chem ; 11(2): 52-61, 2020 Sep 27.
Article in English | MEDLINE | ID: mdl-33024517

ABSTRACT

The generation of cellular energy in the form of ATP occurs mainly in mitochondria by oxidative phosphorylation. Cytochrome c oxidase (CytOx), the oxygen accepting and rate-limiting step of the respiratory chain, regulates the supply of variable ATP demands in cells by "allosteric ATP-inhibition of CytOx." This mechanism is based on inhibition of oxygen uptake of CytOx at high ATP/ADP ratios and low ferrocytochrome c concentrations in the mitochondrial matrix via cooperative interaction of the two substrate binding sites in dimeric CytOx. The mechanism keeps mitochondrial membrane potential ΔΨm and reactive oxygen species (ROS) formation at low healthy values. Stress signals increase cytosolic calcium leading to Ca2+-dependent dephosphorylation of CytOx subunit I at the cytosolic side accompanied by switching off the allosteric ATP-inhibition and monomerization of CytOx. This is followed by increase of ΔΨm and formation of ROS. A hypothesis is presented suggesting a dynamic change of binding of NDUFA4, originally identified as a subunit of complex I, between monomeric CytOx (active state with high ΔΨm, high ROS and low efficiency) and complex I (resting state with low ΔΨm, low ROS and high efficiency).

5.
J Mol Med (Berl) ; 98(5): 651-657, 2020 05.
Article in English | MEDLINE | ID: mdl-32313986

ABSTRACT

Psychosocial stress is known to cause an increased incidence of coronary heart disease. In addition, multiple other diseases like cancer and diabetes mellitus have been related to stress and are mainly based on excessive formation of reactive oxygen species (ROS) in mitochondria. The molecular interactions between stress and ROS, however, are still unknown. Here we describe the missing molecular link between stress and an increased cellular ROS, based on the regulation of cytochrome c oxidase (COX). In normal healthy cells, the "allosteric ATP inhibition of COX" decreases the oxygen uptake of mitochondria at high ATP/ADP ratios and keeps the mitochondrial membrane potential (ΔΨm) low. Above ΔΨm values of 140 mV, the production of ROS in mitochondria increases exponentially. Stress signals like hypoxia, stress hormones, and high glutamate or glucose in neurons increase the cytosolic Ca2+ concentration which activates a mitochondrial phosphatase that dephosphorylates COX. This dephosphorylated COX exhibits no allosteric ATP inhibition; consequently, an increase of ΔΨm and ROS formation takes place. The excess production of mitochondrial ROS causes apoptosis or multiple diseases.


Subject(s)
Electron Transport Complex IV/metabolism , Reactive Oxygen Species/metabolism , Stress, Physiological , Stress, Psychological , Adenosine Triphosphate/metabolism , Animals , Biomarkers , Calcium/metabolism , Humans , Membrane Potential, Mitochondrial , Mitochondria/genetics , Mitochondria/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Signal Transduction
6.
Mitochondrion ; 49: 149-155, 2019 11.
Article in English | MEDLINE | ID: mdl-31419492

ABSTRACT

Almost all energy consumed by higher organisms, either in the form of ATP or heat, is produced in mitochondria by respiration and oxidative phosphorylation through five protein complexes in the inner membrane. High-resolution x-ray analysis of crystallized cytochrome c oxidase (CytOx), the final oxygen-accepting complex of the respiratory chain, isolated by using cholate as detergent, revealed a dimeric structure with 13 subunits in each monomer. In contrast, CytOx isolated with non-ionic detergents appeared to be monomeric. Our data indicate in vivo a continuous transition between CytOx monomers and dimers via reversible phosphorylation. Increased intracellular calcium, as a consequence of stress, dephosphorylates and monomerises CytOx, whereas cAMP rephosphorylates and dimerises it. Only dimeric CytOx exhibits an "allosteric ATP-inhibition" which inhibits respiration at high cellular ATP/ADP-ratios and could prevent oxygen radical formation and the generation of diseases.


Subject(s)
Electron Transport Complex IV/chemistry , Mitochondrial Proteins/chemistry , Oxygen Consumption , Protein Multimerization , Allosteric Regulation , Animals , Crystallography, X-Ray , Electron Transport Complex IV/metabolism , Male , Mitochondrial Proteins/metabolism , Rats , Rats, Wistar
7.
Trends Endocrinol Metab ; 28(11): 761-770, 2017 11.
Article in English | MEDLINE | ID: mdl-28988874

ABSTRACT

Cytochrome c oxidase (CcO) is the final oxygen accepting enzyme complex (complex IV) of the mitochondrial respiratory chain. In contrast to the other complexes (I, II, and III), CcO is highly regulated via isoforms for six of its ten nuclear-coded subunits, which are differentially expressed in species, tissues, developmental stages, and cellular oxygen concentrations. Recent publications have claimed that NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 4 (NDUFA4), originally identified as subunit of complex I, represents a 14th subunit of CcO. Results on CcO composition in tissues from adult animals and the review of data from recent literature strongly suggest that NDUFA4 is not a 14th subunit of CcO but may represent an assembly factor for CcO or supercomplexes (respirasomes) in mitochondria of growing cells and cancer tissues.


Subject(s)
Electron Transport Complex IV/metabolism , Animals , Electron Transport Complex IV/physiology , Humans , Mammals , Protein Binding , Protein Isoforms , Protein Subunits
8.
Mitochondrion ; 24: 64-76, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26190566

ABSTRACT

Cytochrome c oxidase (COX) from mammals and birds is composed of 13 subunits. The three catalytic subunits I-III are encoded by mitochondrial DNA, the ten nuclear-coded subunits (IV, Va, Vb, VIa, VIb, VIc, VIIa, VIIb, VIIc, VIII) by nuclear DNA. The nuclear-coded subunits are essentially involved in the regulation of oxygen consumption and proton translocation by COX, since their removal or modification changes the activity and their mutation causes mitochondrial diseases. Respiration, the basis for ATP synthesis in mitochondria, is differently regulated in organs and species by expression of tissue-, developmental-, and species-specific isoforms for COX subunits IV, VIa, VIb, VIIa, VIIb, and VIII, but the holoenzyme in mammals is always composed of 13 subunits. Various proteins and enzymes were shown, e.g., by co-immunoprecipitation, to bind to specific COX subunits and modify its activity, but these interactions are reversible, in contrast to the tightly bound 13 subunits. In addition, the formation of supercomplexes with other oxidative phosphorylation complexes has been shown to be largely variable. The regulatory complexity of COX is increased by protein phosphorylation. Up to now 18 phosphorylation sites have been identified under in vivo conditions in mammals. However, only for a few phosphorylation sites and four nuclear-coded subunits could a specific function be identified. Research on the signaling pathways leading to specific COX phosphorylations remains a great challenge for understanding the regulation of respiration and ATP synthesis in mammalian organisms. This article reviews the function of the individual COX subunits and their isoforms, as well as proteins and small molecules interacting and regulating the enzyme.


Subject(s)
Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Mitochondria/enzymology , Mitochondria/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Adenosine Triphosphate/biosynthesis , Animals , Gene Expression Regulation , Humans , Mammals , Oxidative Phosphorylation , Oxygen Consumption , Phosphorylation , Protein Processing, Post-Translational
9.
Mitochondrion ; 13(1): 1-6, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23178790

ABSTRACT

Degenerative diseases are in part based on elevated production of ROS (reactive oxygen species) in mitochondria, mainly during stress and excessive work under stress (strenuous exercise). The production of ROS increases with increasing mitochondrial membrane potential (ΔΨ(m)). A mechanism is described which is suggested to keep ΔΨ(m) at low values under normal conditions thus preventing ROS formation, but is switched off under stress and excessive work to maximize the rate of ATP synthesis, accompanied by decreased efficiency. Low ΔΨ(m) and low ROS production are suggested to occur by inhibition of respiration at high [ATP]/[ADP] ratios. The nucleotides interact with phosphorylated cytochrome c oxidase (COX), representing the step with the highest flux-control coefficient of mitochondrial respiration. At stress and excessive work neural signals are suggested to dephosphorylate the enzyme and abolish the control of COX activity (respiration) by the [ATP]/[ADP] ratio with consequent increase of ΔΨ(m) and ROS production. The control of COX by the [ATP]/[ADP] ratio, in addition, is proposed to increase the efficiency of ATP production via a third proton pumping pathway, identified in eukaryotic but not in prokaryotic COX. We conclude that 'oxidative stress' occurs when the control of COX activity by the [ATP]/[ADP] ratio is switched off via neural signals.


Subject(s)
Eukaryota/physiology , Oxidative Stress , Stress, Physiological , Adenosine Triphosphate/metabolism , Energy Metabolism , Metabolic Networks and Pathways , Models, Biological , Reactive Oxygen Species/metabolism
10.
Methods Mol Biol ; 893: 345-58, 2012.
Article in English | MEDLINE | ID: mdl-22665310

ABSTRACT

Many cellular processes are regulated by reversible phosphorylation to change the activity state of proteins. One example is cytochrome c oxidase (COX) with its important function for energy metabolism in the mitochondria. The phosphorylation of this enzyme is a prerequisite for the allosteric ATP-inhibition and therefore necessary to adapt energy production to ATP demand of the cell. Its hydrophobic nature hampers the recognition of phosphorylated amino acids in most subunits of this complex, and as a consequence, only a few phosphorylation sites were identified by mass spectrometry. We describe here a method that enables the analysis of integral membrane proteins by chemical cleavage with cyanogen bromide (BrCN), a method that improves the mass spectrometric detection of hydrophobic proteins. The low abundance of phosphopeptides requires efficient enrichment techniques, such as TiO(2)-based methods. However, this strategy failed in our hands when just BrCN-cleaved peptides were used. Only an additional size-reduction with trypsin produced peptides with optimal properties for enrichment and MS-identification. Another bottleneck was the correct assignment of phosphoserine and phosphothreonine because peptide-ion fragmentation by collision induced dissociation (CID) often results in neutral loss of HPO(3) or H(2)PO(4) from the precursor, decreasing fragmentations that define the peptide sequence and the phosphorylation site. The additional usage of electron transfer dissociation (ETD) as an alternative fragmentation method enabled the precise assignment of the phosphorylated amino acids. In a total of six, new phosphorylation sites of four COX-subunits were identified by this strategy.


Subject(s)
Electron Transport Complex IV/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Protein Processing, Post-Translational , Animals , Cattle , Chromatography, Reverse-Phase , Electron Transport Complex IV/chemistry , Electron Transport Complex IV/isolation & purification , Electrophoresis, Polyacrylamide Gel , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/chemistry , Membrane Proteins/isolation & purification , Mitochondria, Heart/enzymology , Myocardium/enzymology , Peptide Fragments/chemistry , Phosphoproteins/chemistry , Phosphoproteins/isolation & purification , Phosphorylation , Proteolysis , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Trypsin/chemistry
11.
Adv Exp Med Biol ; 748: 1-11, 2012.
Article in English | MEDLINE | ID: mdl-22729852

ABSTRACT

The basic mechanism of ATP synthesis in the mitochondria by oxidative phosphorylation (OxPhos) was revealed in the second half of the twentieth century. The OxPhos complexes I-V have been analyzed concerning their subunit composition, genes, and X-ray structures. This book presents new developments regarding the morphology, biogenesis, gene evolution, heat, and reactive oxygen species (ROS) generation in mitochondria, as well as the structure and supercomplex formation of OxPhos complexes. In addition, multiple mitochondrial diseases based on mutations of nuclear-encoded genes have been identified. Little is known, however, of the regulation of OxPhos according to the variable cellular demands of ATP. In particular, the functions of the supernumerary (nuclear-encoded) subunits of mitochondrial OxPhos complexes, which are mostly absent in bacteria, remain largely unknown, although the corresponding and conserved core subunits exhibit the same catalytic activity. Identification of regulatory pathways modulating OxPhos activity, by subunit isoform expression, by allosteric interaction with ATP/ADP, by reversible phosphorylation of protein subunits, or by supercomplex formation, will help to understand the role of mitochondria in the many degenerative diseases, mostly based on ROS formation in mitochondria and/or insufficient energy production.


Subject(s)
Mitochondria/metabolism , Oxidative Phosphorylation , Adenosine Triphosphate/metabolism , Animals , Humans , Proton-Translocating ATPases/chemistry , Reactive Oxygen Species/metabolism
12.
Adv Exp Med Biol ; 748: 265-81, 2012.
Article in English | MEDLINE | ID: mdl-22729862

ABSTRACT

During evolution from prokaryotes to eukaryotes, the main function of cytochrome c oxidase (COX), i.e., the coupling of oxygen reduction to proton translocation without the production of ROS (reactive oxygen species) remained unchanged demonstrating its robustness. A new regulation of respiration by the ATP/ADP ratio was introduced in eukaryotes based on nucleotide interaction with the added COX subunit IV. This allosteric ATP-inhibition was proposed to keep the mitochondrial membrane potential (ΔΨ(m)) at low healthy values and thus prevents the formation of ROS at complexes I and III. ROS have been implicated in various degenerative diseases. The allosteric ATP-inhibition of COX is reversibly switched on and off by phosphorylation of COX at a serine or threonine. In more than 100 individual preparations of rat heart and liver mitochondria, prepared under identical conditions, the extent of allosteric ATP-inhibition varied. This variability correlates with the variable inhibition of uncoupled respiration in intact isolated mitochondria by ATP. It is concluded that in higher organisms the allosteric ATP-inhibition is continually switched on and off by neuronal signalling in order to change oxidative phosphorylation from optimal efficiency with lower rate of ATP synthesis under resting conditions (low ΔΨ(m) and ROS production) to maximal rate of ATP synthesis under active (working, stress) conditions (elevated ΔΨ(m) and ROS production).


Subject(s)
Electron Transport Complex IV/physiology , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Allosteric Regulation , Amino Acid Sequence , Animals , Humans , Membrane Potential, Mitochondrial , Molecular Sequence Data , Phosphorylation
13.
Proteomics ; 12(7): 950-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22522801

ABSTRACT

Cytochrome c oxidase (COX), the terminal enzyme of the mitochondrial electron transport chain, is regulated by isozyme expression, allosteric effectors such as the ATP/ADP ratio, and reversible phosphorylation. Of particular interest is the "allosteric ATP-inhibition," which has been hypothesized to keep the mitochondrial membrane potential at low healthy values (<140 mV), thus preventing the formation of superoxide radical anions, which have been implicated in multiple degenerative diseases. It has been proposed that the "allosteric ATP-inhibition" is switched on by the protein kinase A-dependent phosphorylation of COX. The goal of this study was to identify the phosphorylation site(s) involved in the "allosteric ATP-inhibition" of COX. We report the mass spectrometric identification of four new phosphorylation sites in bovine heart COX. The identified phosphorylation sites include Tyr-218 in subunit II, Ser-1 in subunit Va, Ser-2 in subunit Vb, and Ser-1 in subunit VIIc. With the exception of Ser-2 in subunit Vb, the identified phosphorylation sites were found in enzyme samples with and without "allosteric ATP inhibition," making Ser-2 of subunit Vb a candidate site enabling allosteric regulation. We therefore hypothesize that additional phosphorylation(s) may be required for the "allosteric ATP-inhibition," and that these sites may be easily dephosphorylated or difficult to identify by mass spectrometry.


Subject(s)
Electron Transport Complex IV/metabolism , Phosphopeptides/metabolism , Allosteric Regulation , Amino Acid Sequence , Animals , Cattle , Chromatography, High Pressure Liquid , Electron Transport Complex IV/chemistry , Molecular Sequence Data , Myocardium/enzymology , Phosphopeptides/chemistry , Phosphorylation , Protein Subunits/chemistry , Protein Subunits/metabolism , Tandem Mass Spectrometry
14.
Mitochondrion ; 11(5): 700-6, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21703366

ABSTRACT

The molecular events occurring during myocardial infarction and cardioprotection are described with an emphasis on the changes of the mitochondrial membrane potential (ΔΨ(m)). The low ΔΨ(m) values of the normal beating heart (100-140 mV) are explained by the allosteric ATP-inhibition of cytochrome c oxidase (CcO) through feedback inhibition by ATP at high [ATP]/[ADP] ratios. During ischemia the mechanism is reversibly switched off by signaling through reactive oxygen species (ROS). At reperfusion high ΔΨ(m) values cause a burst of ROS production leading to apoptosis and/or necrosis. Ischemic preconditioning is suggested to cause additional phosphorylation of CcO, protecting the enzyme from immediate dephosphorylation via ROS signaling.


Subject(s)
Heart Failure/metabolism , Membrane Potential, Mitochondrial , Myocardial Ischemia/metabolism , Adenosine Triphosphate/metabolism , Allosteric Regulation , Animals , Cytoprotection , Electron Transport Complex IV/antagonists & inhibitors , Electron Transport Complex IV/metabolism , Heart Failure/physiopathology , Humans , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/physiopathology , Oxygen Consumption , Permeability , Reactive Oxygen Species/metabolism , Signal Transduction
15.
Biochim Biophys Acta ; 1797(9): 1672-80, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20599681

ABSTRACT

This paper describes the problems of measuring the allosteric ATP-inhibition of cytochrome c oxidase (CcO) in isolated mitochondria. Only by using the ATP-regenerating system phosphoenolpyruvate and pyruvate kinase full ATP-inhibition of CcO could be demonstrated by kinetic measurements. The mechanism was proposed to keep the mitochondrial membrane potential (DeltaPsi(m)) in living cells and tissues at low values (100-140 mV), when the matrix ATP/ADP ratios are high. In contrast, high DeltaPsi(m) values (180-220 mV) are generally measured in isolated mitochondria. By using a tetraphenyl phosphonium electrode we observed in isolated rat liver mitochondria with glutamate plus malate as substrates a reversible decrease of DeltaPsi(m) from 233 to 123 mV after addition of phosphoenolpyruvate and pyruvate kinase. The decrease of DeltaPsi(m) is explained by reversal of the gluconeogenetic enzymes pyruvate carboxylase and phosphoenolpyruvate carboxykinase yielding ATP and GTP, thus increasing the matrix ATP/ADP ratio. With rat heart mitochondria, which lack these enzymes, no decrease of DeltaPsi(m) was found. From the data we conclude that high matrix ATP/ADP ratios keep DeltaPsi(m) at low values by the allosteric ATP-inhibition of CcO, thus preventing the generation of reactive oxygen species which could generate degenerative diseases. It is proposed that respiration in living eukaryotic organisms is normally controlled by the DeltaPsi(m)-independent "allosteric ATP-inhibition of CcO." Only when the allosteric ATP-inhibition is switched off under stress, respiration is regulated by "respiratory control," based on DeltaPsi(m) according to the Mitchell Theory.


Subject(s)
Adenosine Triphosphate/pharmacology , Cell Respiration , Electron Transport Complex IV/antagonists & inhibitors , Membrane Potential, Mitochondrial , Mitochondria, Heart/physiology , Mitochondria, Liver/physiology , Adenosine Diphosphate/pharmacology , Adenosine Triphosphate/metabolism , Animals , Rats
16.
Biochim Biophys Acta ; 1800(3): 205-12, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19409964

ABSTRACT

The Mitchell Theory implies the proton motive force Deltap across the inner mitochondrial membrane as the energy-rich intermediate of oxidative phosphorylation. Deltap is composed mainly of an electrical (DeltaPsi(m)) and a chemical part (DeltapH) and generated by the respiratory chain complexes I, III and IV. It is consumed mostly by the ATP synthase (complex V) to produce ATP. The free energy of electron transport within the proton pumps is sufficient to generate Deltap of about 240 mV. The proton permeability of biological membranes, however, increases exponentially above 130 mV leading to a waste of energy at high values (DeltaPsi(m)>140 mV). In addition, at DeltaPsi(m)>140 mV, the production of the superoxide radical anion O(2)(-) at complexes I, II and III increases exponentially with increasing DeltaPsi(m). O(2)(-) and its neutral product H(2)O(2) (=ROS, reactive oxygen species) induce oxidative stress which participates in aging and in the generation of degenerative diseases. Here we describe a new mechanism which acts independently of the Mitchell Theory and keeps DeltaPsi(m) at low values through feedback inhibition of complex IV (cytochrome c oxidase) at high ATP/ADP ratios, thus preventing the formation of ROS and maintaining high efficiency of oxidative phosphorylation.


Subject(s)
Mitochondria/metabolism , Oxidative Phosphorylation , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Disease , Humans , Mitochondrial Membranes/metabolism , Models, Theoretical , Oxidative Stress , Oxygen Consumption , Protein Kinases/metabolism , Protein Subunits/metabolism , Proton Pumps , Reactive Oxygen Species/metabolism
17.
Trends Mol Med ; 15(4): 139-47, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19303362

ABSTRACT

Aging and degenerative diseases are associated with increased levels of reactive oxygen species (ROS). ROS are mostly produced in mitochondria, and their levels increase with higher mitochondrial membrane potential. Cellular respiratory control is based on inhibition of respiration by high membrane potentials. However, we have described a second mechanism of respiratory control based on allosteric inhibition of cytochrome c oxidase (CcO), the terminal enzyme of the respiratory chain, at high ATP:ADP ratios. The mechanism is independent of membrane potential. We have proposed that feedback inhibition of CcO by ATP keeps the membrane potential and ROS production at low levels. Various forms of stress switch off allosteric ATP-inhibition via reversible dephosphorylation of CcO, resulting in increased membrane potential and cellular ROS levels. This mechanism is proposed to represent a missing molecular link between stress and degenerative diseases.


Subject(s)
Electron Transport Complex IV/metabolism , Oxidative Stress/physiology , Adenosine Triphosphate/metabolism , Animals , Electron Transport Complex IV/chemistry , Electron Transport Complex IV/genetics , Humans , Membrane Potential, Mitochondrial/physiology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Models, Biological , Reactive Oxygen Species/metabolism
18.
Mol Cell Proteomics ; 7(9): 1714-24, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18541608

ABSTRACT

The influence of protein phosphorylation on the kinetics of cytochrome c oxidase was investigated by applying Western blotting, mass spectrometry, and kinetic measurements with an oxygen electrode. The isolated enzyme from bovine heart exhibited serine, threonine, and/or tyrosine phosphorylation in various subunits, except subunit I, by using phosphoamino acid-specific antibodies. The kinetics revealed slight inhibition of oxygen uptake in the presence of ATP, as compared with the presence of ADP. Mass spectrometry identified the phosphorylation of Ser-34 at subunit IV and Ser-4 and Thr-35 at subunit Va. Incubation of the isolated enzyme with protein kinase A, cAMP, and ATP resulted in serine and threonine phosphorylation of subunit I, which was correlated with sigmoidal inhibition kinetics in the presence of ATP. This allosteric ATP-inhibition of cytochrome c oxidase was also found in rat heart mitochondria, which had been rapidly prepared in the presence of protein phosphatase inhibitors. The isolated rat heart enzyme, prepared from the mitochondria by blue native gel electrophoresis, showed serine, threonine, and tyrosine phosphorylation of subunit I. It is concluded that the allosteric ATP-inhibition of cytochrome c oxidase, previously suggested to keep the mitochondrial membrane potential and thus the reactive oxygen species production in cells at low levels, occurs in living cells and is based on phosphorylation of cytochrome c oxidase subunit I.


Subject(s)
Adenosine Triphosphate/metabolism , Electron Transport Complex IV/antagonists & inhibitors , Electron Transport Complex IV/metabolism , Adenosine Triphosphate/pharmacology , Allosteric Regulation , Animals , Cattle , Kinetics , Mitochondria, Heart/enzymology , Myocardium/enzymology , Myocardium/ultrastructure , Phosphorylation , Phosphoserine/analysis , Phosphothreonine/analysis , Phosphotyrosine/analysis , Rats , Serine/metabolism , Spectrometry, Mass, Electrospray Ionization , Threonine/metabolism
19.
Biochim Biophys Acta ; 1655(1-3): 400-8, 2004 Apr 12.
Article in English | MEDLINE | ID: mdl-15100056

ABSTRACT

Apoptotic cell death can occur by two different pathways. Type 1 is initiated by the activation of death receptors (Fas, TNF-receptor-family) on the plasma membrane followed by activation of caspase 8. Type 2 involves changes in mitochondrial integrity initiated by various effectors like Ca(2+), reactive oxygen species (ROS), Bax, or ceramide, leading to the release of cytochrome c and activation of caspase 9. The release of cytochrome c is followed by a decrease of the mitochondrial membrane potential DeltaPsi(m). Recent publications have demonstrated, however, that induction of apoptosis by various effectors involves primarily a transient increase of DeltaPsi(m) for unknown reason. Here we propose a new mechanism for the increased DeltaPsi(m) based on experiments on the allosteric ATP-inhibition of cytochrome c oxidase at high matrix ATP/ADP ratios, which was concluded to maintain low levels of DeltaPsi(m) in vivo under relaxed conditions. This regulatory mechanism is based on the potential-dependency of the ATP synthase, which has maximal activity at DeltaPsi(m)=100-120 mV. The mechanism is turned off either through calcium-activated dephosphorylation of cytochrome c oxidase or by 3,5-diiodo-L-thyronine, palmitate, and probably other so far unknown effectors. Consequently, energy metabolism changes to an excited state. We propose that this change causes an increase in DeltaPsi(m), a condition for the formation of ROS and induction of apoptosis.


Subject(s)
Apoptosis/physiology , Electron Transport Complex IV/physiology , Animals , Apoptosis/drug effects , Calcium/metabolism , Electron Transport , Humans , Membrane Potentials , Mitochondria/metabolism , Neoplasms/enzymology , Neoplasms/pathology , Oxidative Stress , Palmitic Acid/metabolism , Palmitic Acid/pharmacology , Thyroid Hormones/physiology , Tumor Suppressor Protein p53/physiology , fas Receptor/physiology
20.
Biochim Biophys Acta ; 1604(2): 77-94, 2003 Jun 05.
Article in English | MEDLINE | ID: mdl-12765765

ABSTRACT

This article reviews parameters of extrinsic uncoupling of oxidative phosphorylation (OxPhos) in mitochondria, based on induction of a proton leak across the inner membrane. The effects of classical uncouplers, fatty acids, uncoupling proteins (UCP1-UCP5) and thyroid hormones on the efficiency of OxPhos are described. Furthermore, the present knowledge on intrinsic uncoupling of cytochrome c oxidase (decrease of H(+)/e(-) stoichiometry=slip) is reviewed. Among the three proton pumps of the respiratory chain of mitochondria and bacteria, only cytochrome c oxidase is known to exhibit a slip of proton pumping. Intrinsic uncoupling was shown after chemical modification, by site-directed mutagenesis of the bacterial enzyme, at high membrane potential DeltaPsi, and in a tissue-specific manner to increase thermogenesis in heart and skeletal muscle by high ATP/ADP ratios, and in non-skeletal muscle tissues by palmitate. In addition, two mechanisms of respiratory control are described. The first occurs through the membrane potential DeltaPsi and maintains high DeltaPsi values (150-200 mV). The second occurs only in mitochondria, is suggested to keep DeltaPsi at low levels (100-150 mV) through the potential dependence of the ATP synthase and the allosteric ATP inhibition of cytochrome c oxidase at high ATP/ADP ratios, and is reversibly switched on by cAMP-dependent phosphorylation. Finally, the regulation of DeltaPsi and the production of reactive oxygen species (ROS) in mitochondria at high DeltaPsi values (150-200 mV) are discussed.


Subject(s)
Oxidative Phosphorylation , Uncoupling Agents/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Animals , Biological Evolution , Electron Transport Complex IV/metabolism , Energy Metabolism , Fatty Acids/metabolism , Intracellular Membranes/metabolism , Membrane Potentials , Membrane Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Oxidative Phosphorylation/drug effects , Proton Pumps/drug effects , Proton Pumps/metabolism , Proton-Translocating ATPases/metabolism , Protons , Reactive Oxygen Species/metabolism , Thyroid Hormones/metabolism , Uncoupling Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...