ArticlePDF Available

Cyanide resistant respiration and the alternative oxidase pathway: A journey from plants to mammals

Authors:

Abstract

In a large number of organisms covering all phyla, the mitochondrial respiratory chain harbors, in addition to the conventional elements, auxiliary proteins that confer adaptive metabolic plasticity. The alternative oxidase (AOX) represents one of the most studied auxiliary proteins, initially identified in plants. In contrast to the standard respiratory chain, the AOX mediates a thermogenic cyanide-resistant respiration; a phenomenon that has been of great interest for over 2 centuries in that energy is not conserved when electrons flow through it. Here we summarize centuries of studies starting from the early observations of thermogenicity in plants and the identification of cyanide resistant respiration, to the fascinating discovery of the AOX and its current applications in animals under normal and pathological conditions.
BBA - Bioenergetics 1863 (2022) 148567
Available online 29 April 2022
0005-2728/© 2022 The Author(s). Published by Elsevier B.V. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
Cyanide resistant respiration and the alternative oxidase pathway: A
journey from plants to mammals
Riyad El-Khoury
a
, Malgorzata Rak
b
, Paule B´
enit
b
, Howard T. Jacobs
c
,
d
,
*
, Pierre Rustin
b
,
**
a
American University of Beirut Medical Center, Pathology and Laboratory Medicine Department, Cairo Street, Hamra, Beirut, Lebanon
b
Universit´
e Paris Cit´
e, Inserm, Maladies neurod´
eveloppementales et neurovasculaires, F-75019 Paris, France
c
Faculty of Medicine and Health Technology, FI-33014, Tampere University, Finland
d
Institute of Biotechnology, University of Helsinki, Helsinki, Finland
ARTICLE INFO
Keywords:
Plants
Mitochondria
Thermogenicity
AOX
Cyanide resistant respiration
Mammals
ABSTRACT
In a large number of organisms covering all phyla, the mitochondrial respiratory chain harbors, in addition to the
conventional elements, auxiliary proteins that confer adaptive metabolic plasticity. The alternative oxidase
(AOX) represents one of the most studied auxiliary proteins, initially identied in plants. In contrast to the
standard respiratory chain, the AOX mediates a thermogenic cyanide-resistant respiration; a phenomenon that
has been of great interest for over 2 centuries in that energy is not conserved when electrons ow through it.
Here we summarize centuries of studies starting from the early observations of thermogenicity in plants and the
identication of cyanide resistant respiration, to the fascinating discovery of the AOX and its current applications
in animals under normal and pathological conditions.
1. Introduction
Mitochondria are multifunctional organelles that play a key role in
many essential cellular processes, as well as being major sites of heat
production. They are a main source of the high-energy phosphate
molecule, adenosine triphosphate (ATP) that is synthesized by the pro-
cess of oxidative phosphorylation (OXPHOS), wherein substrate oxida-
tion via mitochondrial respiratory chain (RC) is coupled to ADP
phosphorylation [1]. In addition to the canonical elements of the RC,
mitochondria of many different organisms from all kingdoms contain
auxiliaryproteins that participate in electron transport under specic
conditions (Fig. 1). Alternative oxidases (AOXs) and alternative NAD(P)
H dehydrogenases (NDX) represent two classes of these RC auxiliary
proteins that branch electron ow at various quinone reduction or
oxidation sites. AOXs complete electron transfer from quinones to oxy-
gen, bypassing RC complexes III and IV, while NDXs oxidize NADH/
NADPH, bypassing RC complex I or even providing a substitute for
complex I, whenever it is absent. [2,3]. Thus, the activity of auxiliary
proteins has a signicant impact on the tightness of coupling between
carbon catabolism and ATP synthesis. In various circumstances, they
confer signicant exibility to the RC, enabling organisms endowed
with such proteins to survive uctuating environmental conditions.
In this review, we summarize over a century of research conducted
on the cyanide-insensitive respiration mediated by the terminal oxidase
AOX. We will start from the initial observations of heat production in
plants. We will then describe the discovery of cyanide resistant respi-
ration and the formulation of the concept of parallel respiratory path-
ways, leading to the discovery of AOX itself. Finally, we will explore the
widespread occurrence of the enzyme beyond plants and review its
recent applications and future potential, notably in metazoans.
2. The road to the discovery of the alternative oxidase
2.1. Cyanide-resistant respiration
Thermogenicity in plants was rst described by Lamarck in 1778 in
the sterile part (spadix) of the inorescence of an arum lily, Arum itali-
cum [4] (Fig. 2). It was not until 1851 that a close association between
heat production and oxygen consumption was made by Garreau in the
same genus [5,6]. Although, it was long known that the oral spadix of
arum lilies has a remarkably high rate of respiration, the earliest pub-
lished measurements of respiration rates and of temperature therein
were provided in 1906 by Pfeffer [7]. Two years later, in 1908, Church
suggested that the excessive rise in temperature, amounting almost to
* Correspondence to: H. T Jacobs, Faculty of Medicine and Health Technology, FI-33014, Tampere University, Finland.
** Correspondence to: P. Rustin, Inserm UMR 1141, Bat Bingen, Hˆ
opital Robert Debr´
e, 48 Boulevard S´
erurier, 75019 Paris, France.
E-mail addresses: howard.jacobs@tuni. (H.T. Jacobs), pierre.rustin@inserm.fr (P. Rustin).
Contents lists available at ScienceDirect
BBA - Bioenergetics
journal homepage: www.elsevier.com/locate/bbabio
https://doi.org/10.1016/j.bbabio.2022.148567
Received 14 December 2021; Received in revised form 6 April 2022; Accepted 18 April 2022
BBA - Bioenergetics 1863 (2022) 148567
2
20 C above ambient, might be vital for pollination by attracting polli-
nator insects to the owers [8,9].
Cyanide-resistant respiration was initially discovered not in plants,
but in protists, rst in Paramecium caudatum, by Lund in 1918 [10] then,
a year later, by Warburg in the unicellular green alga Chlorella sp. [11].
In 1929, concomitantly with the discovery of cytochromes, Genevois
described the rst respiration that was resistant to cyanide in plants, in
studies of sweet pea (Lathyrus odoratus) embryos (Fig. 2). The chemical
mechanism behind this respiration was, however, not addressed
[1214]. Shortly thereafter, Okunuki described cyanide- and carbon
monoxide(CO)-resistant respiration in the pollen of Lilium auratum and
went on to formulate explicitly the concept of a dual respiratory
pathway, with a true alternative path for electron ow parallel to the
cytochrome pathway [1517]. Okunuki did not, however, address the
issue of whether the proposed alternative pathway was coupled to ATP
production. The coupling of ATP formation to electron transfer in the
respiratory chain had been discovered around the same time by Engel-
hardt in avian erythrocytes [18]. In 1937, Van Herck reported the
marked resistance to cyanide of respiration by slices of the spadix of the
voodoo lily (Sauromatum), noting the high respiratory rates, but
ascribing the phenomenon to the existence of auto-oxidizable avo-
proteins [19,20].
In 1950, James and Beevers extended these discoveries to another
species of lily, Arum maculatum [9], showing that the respiratory system
in the spadix did not saturate even at 100% oxygen whereas that in the
peduncle saturated at 21% oxygen. They ascribed the observed differ-
ences to the existence of different oxidation mechanisms. They found no
evidence for the operation of a cytochrome system in the spadix, but
suggested that their ndings instead pointed to the existence of a low
oxygen-afnity, avin-dependent terminal oxidase.
2.2. Discovery of the alternative oxidase
James and Beevers demonstrated that cell-free extracts consumed
oxygen at high rates and reacted to different inhibitors similarly to the
intact tissues that they had studied [9]. Subsequently, Hackett and
Simon showed that particles prepared from spadix homogenates were
able to consume oxygen and to oxidize succinate as well as various Krebs
cycle intermediates [21]. In 1955, James and Elliott showed that the
particles isolated from Arum maculatum spadix were metabolically very
similar to mitochondria obtained from other plant and animal tissues
(Fig. 2) except that their respiration was resistant to cyanide [22]. In the
same year, Bendall and Hill showed that mitochondria from the Arum
spadix did contain the classical RC cytochromes, a, b and c. But in
addition, they identied a novel cytochrome component that did not
react with carbon monoxide, that they called b
7
, and proposed that it
could represent the alternate path of electrons to oxygen during cyanide-
resistant respiration [23]. Similar suggestions were put forward by in-
vestigators studying other plant species [17,24] but were challenged by
Chance and Hackett [25] who reported evidence for the partial reduc-
tion of cytochromes c and a in skunk cabbage mitochondria even in the
presence of high amounts of cyanide and azide, supporting the excess
oxidasehypothesis in place of a genuine alternate pathway [17].
However, Ikuma and colleagues soon obtained direct evidence for the
existence of two independent oxidases in mung bean hypocotyl mito-
chondria, distinguishable by their oxygen afnities [26]. Flux through
the alternative oxidase that they identied was characterized by a very
low apparent K
M
for O
2
(0.5
μ
M) [26]. They suggested that a ferro-
sulfoprotein with high oxygen afnity, rather than a terminal avo-
protein, was involved in the alternative pathway. Bendall and colleagues
then postulated that the alternate pathway in skunk cabbage mito-
chondria is a non-heme iron-containing protein of yet unknown struc-
ture [27].
Bendall and Bonner showed in 1971 that residual cytochrome oxi-
dase activity contributed only minimally to the large succinate-
dependent oxygen consumption rate observed after cyanide inhibition,
thus laying to rest the excess oxidase hypothesis. Furthermore, since
all three b-type cytochromes were almost completely reduced in
antimycin-inhibited mitochondria, the concept of a ‘b-shuntto oxygen
must also be abandoned. The addition of metal complexing agents
inhibited respiration in the presence of cyanide and antimycin, implying
that the alternate pathway involved an autooxidizable non-heme metal
complex [28]. These results were supported by kinetic experiments
conducted by Storey and Bahr in 1969 [29]. It was subsequently shown
that the cyanide-insensitive alternate pathway could be specically
inhibited by salicylhydroxamic acid (SHAM), and that it is signicantly
more active in state 4 (ADP-exhausted) substrate oxidation than in state
3 (in the presence of ADP) [30]. Although not a avoprotein as such, the
cyanide-resistant alternative oxidase did appear to be a metalloprotein.
Based on Mitchells 1975 hypothesis of the protonmotive ubiquinone
cycle [31], Rich and Moore proposed a similar scheme whereby the
alternate oxidase is in close association with the ubiquinone cycle in
Fig. 1. A schematic view of the proton-
motive and non-proton-motive respiratory
chain segments. The gure integrates the
alternative ux of electrons and the build of
proton-motive force, highlighting the rele-
vance or signicance of having respiration
coupled or uncoupled to proton-motive
force-generation. Note that the composi-
tion of each segment could be different ac-
cording to biological material, with a
number of components being present or
absent in specic cases (e.g. complex I being
absent in the yeast Saccharomyces cer-
evisiae). Action sites of inhibitors are indi-
cated in red. AOX, alternative oxidase; I-V,
the various complexes of the respiratory
chain; CN-, cyanide; ME, NAD+-dependent
malic enzyme; Nd
ex
, external NAD(P)H de-
hydrogenases; Nd
i
, internal rotenone-
resistant NADH dehydrogenase; SHAM,
salicyl hydroxamic acid; TCA, tricarboxylic
acid; UQ, ubiquinone. (For interpretation of
the references to colour in this gure
legend, the reader is referred to the web
version of this article).
R. El-Khoury et al.
BBA - Bioenergetics 1863 (2022) 148567
3
higher plants and that the alternate pathway oxidizes the QH/QH2
couple whilst succinate dehydrogenase reduces it [32]. In 1978, Rich
succeeded in solubilizing and partially purifying the alternative oxidase
from A. maculatum. Moreover, it was stable in the solubilized state,
consistent with it being a protein, with its nal product being water
[33], not H
2
O
2
as initially thought [34]. Several other studies suggested,
however, that the involvement of lipoxygenase [35] or even fatty acid
peroxidation reactions could account for the observed cyanide-resistant
oxygen consumption in plants [3638]. Such debates came to an end
when the enzyme was nally puried [3942], and antibodies raised
against it [43,44] showed that homologues were widely distributed in
the plant kingdom and beyond [4548].
Rhoads and McIntosh made a further major contribution by isolating
and characterizing the cDNA encoding the S. guttatum alternative oxi-
dase, which they designated aox1 [49] (Fig. 2). A homologous diiron
carboxylate protein, termed the plastoquinol terminal oxidase, was later
identied in Arabidopsis thaliana chloroplasts, indicating that the alter-
native oxidase is not conned to mitochondria [50,51].
2.3. Cyanide-insensitive respiration beyond the plant kingdom
In parallel to studies performed in plants, cyanide-insensitive respi-
ration was described in a number of other species prior to the actual
characterization of AOX. In 1950, Sanborn and Williams demonstrated
that the cytochrome pathway undergoes signicant quantitative and
qualitative adjustments during the lifespan of the giant silkworm, Hya-
plophora cecropia [52]. Specically, they showed that cytochromes b and
c are replaced by an unknown cytochrome component, which they
termed cytochrome x, in the larval stages, which disappears abruptly in
the prepupa [52]. In 1954, Pappenheimer and Williams showed that the
cytochrome x of the silkworm Platysamia cecropia was a b-type cyto-
chrome, subsequently referred to as cytochrome b
5
. A considerable
fraction of total electron ux linked to NADH oxidation was shown to
pass through cytochrome b
5
in this [53] and in a second silkworm spe-
cies, Bombyx mori even in the presence of cyanide or antimycin A [54].
In the body muscle of the parasitic nematode Ascaris suum, malate- and
succinate-driven respiration was also found to be insensitive to cyanide
and antimycin A [5557]. However, cyanide-resistant oxygen con-
sumption in an organism does not necessarily imply the existence of an
alternative oxidase, as illustrated by the unique metabolic features of
A. suum [58]. As is now clear, AOX and the gene that encodes it is absent
from insects as well as from A. suum, though present in some other
nematodes [59].
Cyanide-insensitive respiration has also been noted in many protists.
In 1960, Grant and Sargent described a cyanide-resistant alternate
pathway represented by an L-
α
-glycerophosphate oxidase (GPO) system
that consumes oxygen independently from the classical cytochrome
pathway in the euglenozoan Trypanosoma rhodesiense [60,61]. The GPO
system was later shown to be linked to a plant-like alternative oxidase
[62]. In 1995, Chaudhuri et al. identied and partially puried the
trypanosome enzyme using antibodies raised against AOX from
S. guttatum [48]. Enzymatic activity was subsequently demonstrated in
E. coli for the putative AOX both from a trypanosome [63] and from an
apicomplexan [64]. AOX is now recognized as the exclusive terminal
oxidase responsible for oxygen-dependent respiration in the blood-
stream form of T. brucei [65,66] making it attractive as a potential drug
target [6769].
AOX genes have been identied in many other eukaryotic protists,
such as in the green alga Chlamydomonas reinhardtii [70] and in jakobids
[2]. Whole genome analyses, taking advantage of high-throughput
sequencing combined with the development of powerful tools in bio-
informatics, have greatly extended the number of species that could
potentially harbor an AOX. Putative AOX genes may today be found
easily by BLAST searching in diverse stramenopiles, alveolates, haptista,
rhizaria, amoebozoa, other excavates and many other groups.
A major advance came with the discovery of the alternative oxidase
in bacteria (Fig. 2), initially in the Gram-negative bacterium Novos-
phingobium aromaticivorans in 2003 [71]. When expressed in E. coli the
recombinant enzyme was shown to confer cyanide-resistant terminal
oxidase activity [71]. Subsequent in silico exploration of metagenomic
data from ocean sampling expeditions indicated that the AOX gene is
widely distributed among bacteria [7274], though has not been re-
ported in archaea. A functional, NO-resistant, terminal oxidase has
subsequently been identied in the marine bacterium Vibrio scheri
ES114 [75].
The discovery of the alternative oxidase in metazoans in 2004 [76]
represented another major breakthrough (Fig. 2). Today, AOX protein or
AOX-encoding genes have been found in over 150 metazoan species
distributed across many phyla, although are missing in vertebrates [3].
This widespread occurrence of AOX, including in the simplest multi-
cellular animals such as sponges, strongly favors its ancestral character
and implies that a gene loss event has taken place in vertebrates [3,59].
These observations highlight the issue of why AOX has been retained so
widely, yet not in vertebrates. Some clues are suggested from studies in
plants, whilst the impact of expressing invertebrate or fungal AOX in
model vertebrates has also been informative, as discussed below (Sec-
tion 4).
3. Structure, function and regulation of the alternative oxidase
The structural and functional biochemistry of AOX has been exten-
sively reviewed elsewhere, and the reader is referred to the relevant
Fig. 2. Timeline chart depicting some of the key discoveries from early observations of heat production and cyanide-resistant respiration in plants, to the discovery of
the AOX pathway and its recent application in mammals. (For interpretation of the references to colour in this gure legend, the reader is referred to the web version
of this article).
R. El-Khoury et al.
BBA - Bioenergetics 1863 (2022) 148567
4
literature for details. Here we trace the history of studies of the enzyme
sourced from different taxa, and how it has impacted the most recent
work using AOX.
Early structure predictions, conrmed later by X-ray crystallog-
raphy, postulated that AOX is a non-heme diiron protein that belongs to
the diiron carboxylate superfamily [7782]. The elucidation of the
crystal structures of the trypanosomal and plant (A. thaliana) enzymes
have provided structural insights into the nature of the active site, cat-
alytic mechanism and supramolecular organization of the enzyme
[8385]. Both in plants and protists, the active site, buried in a hydro-
phobic cavity, is composed of the diiron centre with four glutamate and
two histidine residues. The GluXXHis motifs stabilize the diiron
center and participate in catalysis [78,8284,86,87], while other
conserved residues are essential for ubiquinol/ubiquinone and oxygen
binding [8386,88]. Because of the recent interest in AOX as a potential
drug target, structural studies of the enzyme from different taxa will
undoubtedly intensify.
The supramolecular organization of AOX is more variable. Initial
Western blotting experiments using an AOX monoclonal antibody
detected multiple species [43,47,89], which were subsequently shown
to represent monomeric and dimeric forms of the enzyme, with dimers
linked by disulde bridges [79,90]. This organization appears to be
universal in plants, whereas fungal AOX exists as a monomer [91].
Following the discovery that AOX is widespread phylogenetically,
interest has turned to its biochemical and physiological functions. The
extensive literature on plant AOX provides a paradigm for understand-
ing its role(s) in other taxa, including metazoans [92] and for its use as a
tool to elucidate aspects of respiratory biology in species from which it
has been lost.
Like cytochrome oxidase, AOX catalyses a four-electron reduction of
oxygen to water, but does not compete with the canonical cytochrome
pathway, due to its very low afnity for its substrate, ubiquinol [9395].
The partitioning of electron ux between the two pathways, in the
absence of inhibitors, is mainly governed by the reduction level of the
ubiquinone pools, which is inuenced by the available substrates and
the metabolic state of the cell. Oxygen concentration and the levels of
tricarboxylic acid (TCA) cycle intermediates are important regulators
(Fig. 3) [9499]. This regulation enables signicant respiratory activity
to be maintained even in the presence of potent inhibitors of the cyto-
chrome pathway, such as cyanide, nitric oxide and sulphide [100,101].
The function of AOX in plant thermogenesis was the rst indication
of its existence, though this aspect has not been widely studied in other
taxa. However, it is an unavoidable consequence of its non-proton-
motive nature, and may have many other manifestations in biology.
Note that, even in organisms not endowed with AOX, up to 60% of the
energy released during respiration is converted to heat under normal
physiological conditions [102,103]. The thermogenic role of AOX is
likely to be a lively topic of future research, especially given the nding
that the operating temperature of mitochondria, at least in mammals, is
of the order of 50 C [104].
In plants, AOX is essential for maintaining cellular homeostasis by
preserving mitochondrial activity in daylight, when ATP is efciently
produced by photosynthesis. AOX activation under such conditions
prevents mitochondrial RC blockage, which would otherwise inhibit all
metabolic reactions contingent on RC function, notably the TCA cycle
[105107]. Similar considerations undoubtedly apply to photosynthetic
protists, but an abundance or over-abundance of ATP may occur also in
heterotrophic organisms where AOX could play a similar function. In
plants, this action of AOX limits the overproduction of reactive oxygen
species (ROS), which has been postulated as a more general role of the
enzyme in other taxa as well [108111].
In plants, AOX also confers metabolic plasticity, enabling organisms
to adapt to various biotic and abiotic stress factors such as drought,
salinity, osmotic stress, nutrient deprivation, infection by pathogens or
extreme conditions of light and temperature: many of which have been
linked to ROS [112126]. AOX also inuences fungal biology in ways
linked to ROS, in many cases with effects on pathogenicity [126132].
How broadly AOX protects animals endowed with it against similar
stress conditions remains an open question.
Studies in plants and subsequently in other organisms have shown
that AOX expression and activity are usually under tight control [133],
which is easily understandable due to the potentially drastic effects of
the enzyme on cellular bioenergetics and heat generation. Regulatory
mechanisms so far elucidated can be divided into those that modulate
AOX gene expression and those that modify AOX enzyme activity post-
translationally. The number of nuclear genes encoding AOX varies, but
typically, as in Arabidopsis, the various AOX isoforms are functionally
distinct, differentially expressed in the tissues and life cycle, and respond
differently to environmental cues [92,134138]. Similarly in fungi,
except where AOX is absent altogether [2,133], its expression can be
constitutive [133,139], variable according to life cycle stages [127,140],
or regulated by environmental factors and substrates [14,139,141143].
Plant AOX is regulated chiey by organic acids [144], whereas both
fungal [141,144147] and protistan [148,149] AOXs are modulated by
purine nucleotides, with a fungal-specic N-terminal tract of 2025
amino acids suggested to confer this sensitivity [91]. Excellent reviews
dealing extensively with AOX regulation are available [92,126,133].
Fig. 3. Cyanide-resistance substrate oxidation by isolated mitochondria from
various plant species. The percentage of cyanide-resistant respiration was
measured under state 3 conditions (presence of ADP) using NADH (exogenous
source), succinate or malate (endogenous source of NADH). All substrates were
oxidized at roughly similar rates in the absence of cyanide.
R. El-Khoury et al.
BBA - Bioenergetics 1863 (2022) 148567
5
4. Heterologous expression of the AOX in animals: what have we
learned?
With the discovery of the AOX in metazoans in 2004, it became
possible to take up the challenge of expressing the enzyme in verte-
brates. Such an initiative sought to characterize the biochemical prop-
erties and physiological functions of metazoan AOX in different
contexts, using model organisms. The effects of AOX expression therein
might also shed light onto the conditions that could have led to the loss
of AOX in the course of evolution.
Shortly after the identication of the rst animal species harbouring
the AOX gene [76], Hakkaart and colleagues succeeded in expressing the
AOX of the tunicate Ciona intestinalis, a member of a sister group to the
vertebrates, in cultured human embryonic kidney (HEK-293T)-derived
cells [150]. The AOX protein was successfully targeted to mitochondria
and was engaged in respiratory electron ow only in the presence of
inhibitors that impose a high degree of reduction of ubiquinone. The
data suggested that metazoan AOX has similar biochemical properties to
the known plant enzymes, namely, low afnity for reduced ubiquinone
and possible post-translational regulation by organic acids [150]. AOXs
from a lamentous fungus and from plants were soon expressed suc-
cessfully in mammalian cells, as well [151154].
C. intestinalis AOX was next expressed in pathological models. In
human cells harbouring specic mutations known to be responsible for a
number of cytochrome oxidase (COX)-related diseases, AOX expression
was able to alleviate various cellular consequences of COX deciency,
including impaired growth, compromised respiration and sensitivity to
oxidative stress [111,155,156]. Ubiquitous AOX expression in the fruit-
y Drosophila melanogaster produced no deleterious effects in wild-type
animals [157], but was able to counteract an array of deleterious con-
sequences at the whole-organism level. This included lethality triggered
by genetic manipulation of the cytochrome pathway [158] or by RC
inhibitors, such as cyanide and antimycin A [157], neurodegenerative
effects whether caused by genetic lesions affecting various COX subunits
[158] or induced by other mechanisms suggested to mirror aspects of
Alzheimers [159] or Parkinsons diseases [157,160]. Decreased ROS
production was suggested as a mechanism, but AOX-expressing ies did
not show increased lifespan [161], contradicting at least the simplest
formulation of the ‘mitochondrial free-radical theory of ageing. Unex-
pectedly, AOX expression also rescued cell migration defects during
development [162], but also had both negative and positive effects on
reproductive success under specic conditions [163165], that remain
poorly understood.
AOX-expressing cells have also been used to explore important issues
in mitochondrial biochemistry. For example, expression of a fungal AOX
in a mouse cell-line bearing a mutation in the mt-CYTb gene, and thus
decient in complex III, established a link between ROS-induced com-
plex I degradation and the redox status of ubiquinone [166], enabling
RC organization to be remodelled according to the availability of
different substrates [166]. In contrast, ROS signaling was inferred not to
play a role in the stabilization of the transcription factor HIF1-
α
under
hypoxia in cultured HEK293-derived cells expressing the Ciona enzyme
[167]. AOX also promoted the migration of mammalian cultured cells
[162].
A further advance was achieved in 2013 by the rst successful
expression of AOX in a mammal [168]. As previously observed in
cultured human cells, the C. intestinalis AOX protein was successfully
targeted to mitochondria in different mouse tissues without competing
biochemically, under standard conditions, with the canonical RC, but
becoming functional upon blockade of the cytochrome segment. Mice
harbouring widely expressed AOX showed normal growth and devel-
opment, produced healthy offspring in normal numbers, and exhibited
increased resistance to otherwise lethal doses of gaseous cyanide [168].
However, since the mice were generated using a lentivector transduction
strategy, which resulted in the insertion of the AOX transgene at mul-
tiple genomic sites, long-term maintenance of the model was not
feasible. To overcome this problem, Szibor and colleagues generated a
genetically tractable mouse expressing a single copy of the C. instestinalis
AOX inserted into the non-essential Rosa26 locus [169], as well as a
conditionally activatable version thereof [170]. The AOX
Rosa26
mouse
showed stable, ubiquitous, and substantial level of expression of the
AOX. Moreover, the enzyme was functional and conferred resistance to
inhibitors of the cytochrome pathway.
Widespread expression of AOX in the AOX
Rosa26
mouse again did not
affect physiological processes under unstressed conditions [169].
However, when combined with various pathological models, AOX
expression produced a variety of outcomes, some of them unexpected,
providing insights into pathophysiological mechanisms. For example,
the use of the AOX
Rosa26
mouse demonstrated mitochondrial involve-
ment in lung damage caused by chronic exposure to toxic smoke [171],
as well as in a sepsis model [173]. Conversely, AOX was unable to rescue
a model of inammatory cardiomyopathy, and even exacerbated the
pathology [172].
In mouse models of RC disease a similarly mixed picture has
emerged. AOX prevented lethal mitochondrial cardiomyopathy in a
mouse model of GRACILE syndrome, caused by a mutation in an as-
sembly factor of RC complex III, Bcs1l [174], but worsened the pheno-
type of mouse models of COX-decient mitochondrial myopathy [175]
and of cardiac ischemia/reperfusion injury [176]. In the latter cases,
AOX abrogated ROS signaling, blocking repair and regeneration pro-
cesses, and providing a possible clue as to why AOX was lost from ver-
tebrates. ROS production from complex III was similarly implicated as
an essential trigger in hypoxia signaling in the lung, using AOX
Rosa26
mice in an ex vivo model of hypoxic pulmonary vasoconstriction [177].
AOX-expressing mice have also begun to be exploited to address
longstanding issues in mammalian mitochondrial bioenergetics. For
example, mice expressing a fungal AOX have been used to conrm that
the complex I +III supercomplex sequesters a portion of the mitochon-
drial ubiquinone, which modulates the oxidative capacity of complex I
[178]. Similar ndings have been made in mice expressing the Ciona
enzyme [179].
5. Conclusions and perspectives
The discovery of the AOX brought to an end a century of vigorous
debate on the exact nature of the machinery responsible for mitochon-
drial cyanide-resistant respiration, initially described in plants. The
development of powerful sequencing technologies combined with sig-
nicant progress in bioinformatics, ushered in a new era that led to the
realization that AOX is extremely widespread phylogenetically. X-ray
crystallography and functional enzymology then allowed its structure
and reaction mechanism to be elucidated. The discovery of AOX in
metazoans, but excluding vertebrates, has enabled heterologous
expression of the enzyme in mammals. Leveraging the unique metabolic
properties of the AOX has opened a new window on mitochondrial
biochemistry and enabled pathophysiological mechanisms associated
with RC dysfunction to be explored in a novel way. AOX may yet have
therapeutic potential [180], if the ways in which it interferes with cell
signaling and repair can be fully understood and mitigated.
In the light of the available and accumulating data in mammals, the
issue of the evolutionary loss of AOX can be divided into two rather
independent questions: rst, what are the potential detrimental effects
of AOX that are inextricably linked with vertebrate biology, and second,
if AOX was lost simply because the benets that it provides to organisms
such as plants and lower eukaryotes were no longer operative, what was
it about the vertebrate lifestyle that removed these constraints? Only a
better understanding of the biochemical and thermodynamic properties
of the AOX of lower metazoans and its physiological repercussions in
mammalian models will provide us with clear answers to these
questions.
Lastly, the history of research on AOX is a vivid illustration of the
importance of serendipity in science. Whilst genome analysis would
R. El-Khoury et al.
BBA - Bioenergetics 1863 (2022) 148567
6
have revealed the existence of the AOX gene in any case, without the
century of prior research on plant respiration and thermogenesis it
would have remained one of the thousands of orphan genes awaiting
proper characterization. Its use as a tool to understand mammalian
biochemistry and cellular functions, as well as human pathophysiology,
would likely be far in the future. Instead, research on the respiration of
plant tissue slices and on how organisms generate heat has led to
unpredicted insights into human biology with potential applications in
medicine.
Declaration of competing interest
The authors declare that they have no known competing nancial
interests or personal relationships that could have appeared to inuence
the work reported in this paper.
References
[1] D.C. Wallace, W. Fan, V. Procaccio, Mitochondrial energetics and therapeutics,
Annu. Rev. Pathol. 5 (2010) 297348, https://doi.org/10.1146/annurev.
pathol.4.110807.092314. PMID: 20078222; PMCID: PMC3245719.
[2] A.E. McDonald, G.C. Vanlerberghe, Origins, evolutionary history, and taxonomic
distribution of alternative oxidase and plastoquinol terminal oxidase, Comp.
Biochem. Physiol. D: Genomics Proteomics 1 (3) (2006 Sep) 357364, https://
doi.org/10.1016/j.cbd.2006.08.001. Epub 2006 Aug 9 PMID: 20483267.
[3] A.E. McDonald, D.V. Gospodaryov, Alternative NAD(P)H dehydrogenase and
alternative oxidase: proposed physiological roles in animals, Mitochondrion 45
(2019 Mar) 717, https://doi.org/10.1016/j.mito.2018.01.009. Epub 2018 Feb 6
PMID: 29421444.
[4] J.B. Lamarck (Ed.), Flore française,Flore française, ou, Descriptions succinctes de
toutes les plantes qui croissent naturellement en France: dispos´
ees selon une
nouvelle m´
ethode danalyse, et pr´
ec´
ed´
ees par un expos´
e des principes
´
el´
ementaires de la botanique, Limprimerie Royale, Paris, 1778, pp. 536539,
https://doi.org/10.5962/bhl.title.112968.
[5] L. Garreau, De la respiration chez les plantes, M´
emoire sur les relations qui
existent entre loxyg`
ene, consomm´
e par le spadice de l"Arum italicum" en ´
etat de
paroxysme, et la chaleur qui se produit, Ann. Sci. Nat. Ser. III 15 (1851) 536.
[6] L. Garreau, Nouvelles recherche sur la respiration des plantes, Ann. Sci. Nat. Ser.
III 16 (1852) 271292. https://catalogue.bnf.fr/ark:/12148/cb304833844.
[7] W. Pfeffer, The Physiology of Plants: A Treatise Upon the Metabolism and Sources
of Energy in Plants, Clarendon Press, Oxford, 1906, https://doi.org/10.1038/
075049b0.
[8] A.H. Church, Types of oral mechanism: a selection of diagrams and descriptions
of common owers arranged as an introduction to the systematic study of
angiosperms, Clarendon Press, Oxford, 1908. Epub 2009 Jan 31. PMID:
19366609, http://www.archive.org/details/cu31924000658413.
[9] W.O. James, H. Beevers, The respiration of Arum spadix. A rapid respiration,
resistant to cyanide, New Phytol. 49 (1950) 353374, https://doi.org/10.1111/
j.1469-8137.1950.tb05160.x.
[10] E.J. Lund, Quantitative studies on intracellular respiration: ii. The rate of
oxidations in Paramecium caudatum and its independence of the toxic action of
KNC, Am. J. Phys. 45 (1918) 365373, https://doi.org/10.1152/
ajplegacy.1918.45.4.365.
[11] O. Warburg, Uber die Geschwindigkeit der photochemischen
Kohlens¨
aurezersetzung in lebenden Zellen, S, Biochem. Z. 100 (1919) 230270,
https://doi.org/10.1007/978-3-642-47774-4_2. Accessible as doi:.
[12] M. Genevois, Sur la fermentation et Sur la respiration chez les v´
eg´
etaux
chlorophyliens, Rev. Gen. Bot. 41 (1929) 252271.
[13] C Lance M Chauveau P Dizengremel . The cyanide-resistant pathway of plant
mitochondria. In. R Douce DA Day (Eds.), Encyclopedia of Plant Physiology, vol
18, Springer, Berlin, Heidelberg. pp. 202247. doi: 10.1007/978-3-642-70101-6_
9.
[14] A. Veiga, J.D. Arrabaça, M.C. Loureiro-Dias, Cyanide-resistant respiration, a very
frequent metabolic pathway in yeasts, FEMS Yeast Res. 3 (3) (2003 May)
239245, https://doi.org/10.1016/S1567-1356(03)00036-9. PMID: 12689632.
[15] K. Okunuki, Über den gaswechsel der pollen, Bot. Mag. Tokyo 46 (551) (1932)
701721, https://doi.org/10.15281/jplantres1887.46.701.
[16] K. Okunuki, Über den gaswechsel der pollen II, Acta Phytochim. Japan 11 (1939)
2764.
[17] C.S. Yocum, D.P. Hackett, Participation of cytochromes in the respiration of the
aroid spadix, Plant Physiol. 32 (3) (1957 May) 186191, https://doi.org/
10.1104/pp.32.3.186. PMID: 16654973; PMCID: PMC540895.
[18] W.A. Engelhardt, Ortho- und pyrophosphat im aeroben und anaeroben
stoffwechsel der blutzellen, Biochem. Z. 227 (1930) 1638.
[19] A.W.H. Van Herk, Die chemischen vorgange im sauromatum kolben III.
Mitteilung, Proc. K. Ned. Akad. Wet. (1937) 709719.
[20] B.J.D. Meeuse, Thermogenic respiration in aroids, Annu. Rev. Plant Physiol. 26
(1975) 117126, https://doi.org/10.1146/annurev, pp. 26.060175.001001.
[21] D.P. Hackett, E.W. Simon, Oxidative activity of particles prepared from the spadix
of Arum maculatum, Nature 173 (4395) (1954 Jan 23) 162163, https://doi.org/
10.1038/173162a0. PMID: 13132893.
[22] W.O. James, D.C. Elliott, Cyanide-resistant mitochondria from the spadix of an
arum, Nature 175 (1955) 89, https://doi.org/10.1038/175089a0.
[23] D.S. Bendall, R. Hill, Cytochrome components in the spadix of Arum maculatum,
New Phytol. 55 (2) (1956) 206212, https://doi.org/10.1111/j.1469-8137.1956.
tb05281.x.
[24] H. Lundegårdh, The cytochrome-cytochrome oxidase system, in: W. Ruhland
(Ed.), Handbuch der Panzenphysiologie Bd 12(1), 1960, pp. 311364.
[25] B. Chance, D.P. Hackett, The electron transfer system of skunk cabbage
mitochondria, Plant Physiol. 34 (1) (1959 Jan) 3349, https://doi.org/10.1104/
pp.34.1.33. PMID: 16655173; PMCID: PMC541140.
[26] H. Ikuma, F.J. Schindler, W.D. Bonner, Kinetic analysis of oxidases in tightly
coupled plant mitochondria, Plant Physiol. 39 (1964). S-1x.
[27] D.S. Bendall, W.D. Bonner, M. Plesnicar, Cyanide insensitive respiration, Fed.
Proc. 26 (1967) 731.
[28] D.S. Bendall, W.D. Bonner, Cyanide-insensitive respiration in plant mitochondria,
Plant Physiol. 47 (2) (1971 Feb) 236245, https://doi.org/10.1104/pp.47.2.236.
PMID: 16657603; PMCID: PMC365849.
[29] B.T. Storey, J.T. Bahr, The respiratory chain of plant mitochondria. I. Electron
transport between succinate and oxygen in skunk cabbage mitochondria, Plant
Physiol. 44 (1) (1969 Jan) 115125, https://doi.org/10.1104/pp.44.1.115.
[30] G.R. Schonbaum, W.D. Bonner Jr., B.T. Storey, J.T. Bahr, Specic inhibition of
the cyanide-insensitive respiratory pathway in plant mitochondria by hydroxamic
acids, Plant Physiol. 47 (1) (1971 Jan) 124128, https://doi.org/10.1104/
pp.47.1.124. PMID: 5543780; PMCID: PMC365824.
[31] P. Mitchell, Protonmotive redox mechanism of the cytochrome b-c1 complex in
the respiratory chain: protonmotive ubiquinone cycle, FEBS Lett. 56 (1) (1975
Aug 1) 16, https://doi.org/10.1016/0014-5793(75)80098-6. PMID: 239860.
[32] P.R. Rich, A.L. Moore, The involvement of the protonmotive ubiquinone cycle in
the respiratory chain of higher plants and its relation to the branchpoint of the
alternate pathway, FEBS Lett. 65 (3) (1976 Jun 15) 339344, https://doi.org/
10.1016/0014-5793(76)80142-1. PMID: 182535.
[33] P.R. Rich, W.D. Bonner Jr., EPR studies of higher plant mitochondria. II. Center S-
3 of succinate dehydrogenase and its relation to alternative respiratory
oxidations, Biochim. Biophys. Acta 501 (3) (1978 Mar 13) 381395, https://doi.
org/10.1016/0005-2728(78)90106-8. PMID: 204341.
[34] P.R. Rich, A. Boveris, W.D. Bonner Jr., A.L. Moore, Hydrogen peroxide generation
by the alternate oxidase of higher plants, Biochem. Biophys. Res. Commun. 71 (3)
(1976 Aug 9) 695703, https://doi.org/10.1016/0006-291x(76)90887-1. PMID:
962948.
[35] D.J. Parrish, A.C. Leopold, Confounding of alternate respiration by lipoxygenase
activity, Plant Physiol. 62 (3) (1978 Sep) 470472, https://doi.org/10.1104/
pp.62.3.470. PMID: 16660540; PMCID: PMC1092149.
[36] P. Rustin, J. Dupont, C. Lance, Oxidative interactions between fatty acid peroxy
radicals and quinones: possible involvement in cyanide-resistant electron
transport in plant mitochondria, Arch. Biochem. Biophys. 225 (2) (1983 Sep)
630639, https://doi.org/10.1016/0003-9861(83)90074-7. PMID: 6414377.
[37] P. Rustin, J. Dupont, C. Lance, A role for fatty acid peroxy radicals in the cyanide-
insensitive pathway of plant mitochondria? Trends Biochem. Sci. 8 (1983)
155157, https://doi.org/10.1016/0968-0004(83)90160-3.
[38] J.N. Siedow, M.E. Girvin, Alternative respiratory pathway: its role in seed
respiration and its inhibition by propyl gallate, Plant Physiol. 65 (4) (1980 Apr)
669674, https://doi.org/10.1104/pp.65.4.669. PMID: 16661259; PMCID:
PMC440403.
[39] P.R. Rich, Quinol oxidation in Arum maculatum mitochondria and its application
to the assay, solubilisation and partial purication of the alternative oxidase,
FEBS Lett. 96 (1978) 252256, https://doi.org/10.1016/0014-5793(78)80412-8.
[40] S. Huq, J.M. Palmer, Isolation of a cyanide-resistant duroquinol oxidase from
Arum maculatum mitochondria, FEBS Lett. 95 (2) (1978 Nov 15) 217220,
https://doi.org/10.1016/0014-5793(78)80997-1. PMID: 720613.
[41] W.D. Bonner, S.D. Clarke, P.R. Rich, Partial purication and characterization of
the quinol oxidase activity of Arum maculatum mitochondria, Plant Physiol. 80
(4) (1986 Apr) 838842, https://doi.org/10.1104/pp.80.4.838. PMID:
16664728; PMCID: PMC1075216.
[42] T.E. Elthon, L. McIntosh, Characterization and solubilization of the alternative
oxidase of sauromatum guttatum mitochondria, Plant Physiol. 82 (1) (1986 Sep)
16, https://doi.org/10.1104/pp.82.1.1. PMID: 16664973; PMCID:
PMC1056056.
[43] T.E. Elthon, L. McIntosh, Identication of the alternative terminal oxidase of
higher plant mitochondria, Proc. Natl. Acad. Sci. U. S. A. 84 (23) (1987 Dec)
83998403, https://doi.org/10.1073/pnas.84.23.8399. PMID: 16593898;
PMCID: PMC299550.
[44] T.E. Elthon, R.L. Nickels, L. McIntosh, Monoclonal antibodies to the alternative
oxidase of higher plant mitochondria, Plant Physiol. 89 (4) (1989 Apr)
13111317, https://doi.org/10.1104/pp.89.4.1311. PMID: 16666702; PMCID:
PMC1056014.
[45] A.M. Lambowitz, J.R. Sabourin, H. Bertrand, R. Nickels, L. McIntosh,
Immunological identication of the alternative oxidase of neurospora crassa
mitochondria, Mol. Cell. Biol. 9 (3) (1989 Mar) 13621364, https://doi.org/
10.1128/mcb.9.3.1362-1364.1989. PMID: 2524649; PMCID: PMC362733.
[46] C. Hiser, L. McIntosh, Alternative oxidase of potato is an integral membrane
protein synthesized de novo during aging of tuber slices, Plant Physiol. 93 (1)
(1990 May) 312318, https://doi.org/10.1104/pp.93.1.312. PMID: 16667452;
PMCID: PMC1062504.
[47] A. Kearns, J. Whelan, S. Young, T.E. Elthon, D.A. Day, Tissue-specic expression
of the alternative oxidase in soybean and siratro, Plant Physiol. 99 (2) (1992 Jun)
R. El-Khoury et al.
BBA - Bioenergetics 1863 (2022) 148567
7
712717, https://doi.org/10.1104/pp.99.2.712. PMID: 16668944; PMCID:
PMC1080523.
[48] M. Chaudhuri, W. Ajayi, S. Temple, G.C. Hill, Identication and partial
purication of a stage-specic 33 kDa mitochondrial protein as the alternative
oxidase of the Trypanosoma brucei brucei bloodstream trypomastigotes,
J. Eukaryot. Microbiol. 42 (5) (1995 Sep-Oct) 467472, https://doi.org/10.1111/
j.1550-7408.1995.tb05892.x. PMID: 7581322.
[49] D.M. Rhoads, L. McIntosh, Isolation and characterization of a cDNA clone
encoding an alternative oxidase protein of sauromatum guttatum (Schott), Proc.
Natl. Acad. Sci. U. S. A. 88 (6) (1991 Mar 15) 21222126, https://doi.org/
10.1073/pnas.88.6.2122. PMID: 1706518; PMCID: PMC51181.
[50] D. Wu, D.A. Wright, C. Wetzel, D.F. Voytas, S. Rodermel, The IMMUTANS
variegation locus of arabidopsis denes a mitochondrial alternative oxidase
homolog that functions during early chloroplast biogenesis, Plant Cell 11 (1)
(1999 Jan) 4355, https://doi.org/10.1105/tpc.11.1.43. PMID: 9878631;
PMCID: PMC144093.
[51] P. Carol, M. Kuntz, A plastid terminal oxidase comes to light: implications for
carotenoid biosynthesis and chlororespiration, Trends Plant Sci. 6 (1) (2001 Jan)
3136, https://doi.org/10.1016/s1360-1385(00)01811-2. PMID: 11164375.
[52] R.C. Sanborn, C.M. Williams, The cytochrome system in the cecropia silkworm,
with special reference to the properties of a new component, J. Gen. Physiol. 33
(5) (1950 May 20) 579588, https://doi.org/10.1085/jgp.33.5.579. PMID:
15422111; PMCID: PMC2147201.
[53] A.M. Pappenheimer Jr., C.M. Williams, Cytochrome b5 and the dihydro-
coenzyme I-oxidase system in the cecropia silkworm, J. Biol. Chem. 209 (2) (1954
Aug) 915929, https://doi.org/10.1016/S0021-9258(18)65520-6. PMID:
13192146.
[54] T. Ito, Y. Horie, S. Ishikawa, Oxidative enzymes of the midgut of the silkworm
Bombyx mori, J. Insect Physiol. 2 (4) (1958) 313.323, https://doi.org/10.1016/
0022-1910(58)90016-7.
[55] E. Kmetec, E. Bueding, Succinic and reduced diphosphopyridine nucleotide
oxidase systems of ascaris muscle, J. Biol. Chem. 236 (1961 Feb) 584591,
https://doi.org/10.1016/s0021-9258(18)64408-4. PMID: 13757025.
[56] P. K¨
ohler, R. Bachmann, Mechanisms of respiration and phosphorylation in
ascaris muscle mitochondria, Mol. Biochem. Parasitol. 1 (2) (1980 Apr) 7590,
https://doi.org/10.1016/0166-6851(80)90002-x. PMID: 7442710.
[57] G.E. Rodrick, S.D. Long, W.A. Sodeman Jr., D.L. Smith, Ascaris suum: oxidative
phosphorylation in mitochondria from developing eggs and adult muscle, Exp.
Parasitol. 54 (2) (1982 Oct) 235242, https://doi.org/10.1016/0014-4894(82)
90132-1. PMID: 6889977.
[58] C. Sakai, E. Tomitsuka, H. Esumi, S. Harada, K. Kita, Mitochondrial fumarate
reductase as a target of chemotherapy: from parasites to cancer cells, Biochim.
Biophys. Acta 1820 (5) (2012 May) 643651, https://doi.org/10.1016/j.
bbagen.2011.12.013. Epub 2011 Dec 29 PMID: 22226661.
[59] A.E. McDonald, G.C. Vanlerberghe, J.F. Staples, Alternative oxidase in animals:
unique characteristics and taxonomic distribution, J. Exp. Biol. 212 (Pt 16) (2009
Aug) 26272634, https://doi.org/10.1242/jeb.032151. PMID: 19648408.
[60] P.T. Grant, J.R. Sargent, Properties of L-alpha-glycerophosphate oxidase and its
role in the respiration of trypanosoma rhodesiense, Biochem. J. 76 (2) (1960 Aug)
229237, https://doi.org/10.1042/bj0760229. PMID: 13828972; PMCID:
PMC1204698.
[61] P.T. Grant, J.R. Sargent, L-alpha-glycerophosphate dehydrogenase, a component
of an oxidase system in trypanosoma rhodesiense, Biochem. J. 81 (1) (1961 Oct)
206214, https://doi.org/10.1042/bj0810206. PMID: 13901161; PMCID:
PMC1243319.
[62] A.B. Clarkson Jr., E.J. Bienen, G. Pollakis, R.W. Grady, Respiration of
bloodstream forms of the parasite trypanosoma brucei brucei is dependent on a
plant-like alternative oxidase, J. Biol. Chem. 264 (30) (1989 Oct 25)
1777017776. PMID: 2808350.
[63] C. Nihei, Y. Fukai, K. Kawai, A. Osanai, Y. Yabu, T. Suzuki, N. Ohta, N. Minagawa,
K. Nagai, K. Kita, Purication of active recombinant trypanosome alternative
oxidase, FEBS Lett. 538 (13) (2003 Mar 13) 3540, https://doi.org/10.1016/
s0014-5793(03)00120-0. PMID: 12633849.
[64] T. Suzuki, T. Hashimoto, Y. Yabu, Y. Kido, K. Sakamoto, C. Nihei, M. Hato,
S. Suzuki, Y. Amano, K. Nagai, T. Hosokawa, N. Minagawa, N. Ohta, K. Kita,
Direct evidence for cyanide-insensitive quinol oxidase (alternative oxidase) in
apicomplexan parasite Cryptosporidium parvum: phylogenetic and therapeutic
implications, Biochem. Biophys. Res. Commun. 313 (4) (2004 Jan 23)
10441052, https://doi.org/10.1016/j.bbrc.2003.12.038. PMID: 14706648.
[65] J.W. Priest, S.L. Hajduk, Developmental regulation of mitochondrial biogenesis in
trypanosoma brucei, J. Bioenerg. Biomembr. 26 (2) (1994 Apr) 179191, https://
doi.org/10.1007/BF00763067. PMID: 8056785.
[66] T.K. Smith, F. Bringaud, D.P. Nolan, L.M. Figueiredo, Metabolic reprogramming
during the Trypanosoma brucei life cycle, F1000 Faculty Rev-683, F1000Res 6
(2017 May 16), https://doi.org/10.12688/f1000research.10342.2. PMID:
28620452; PMCID: PMC5461901.
[67] S.K. Menzies, L.B. Tulloch, G.J. Florence, T.K. Smith, The trypanosome
alternative oxidase: a potential drug target? Parasitology 145 (2) (2018 Feb)
175183, https://doi.org/10.1017/S0031182016002109. Epub 2016 Nov 29
PMID: 27894362.
[68] R.A. West, T. Cunningham, L.E. Pennicott, S.P.S. Rao, S.E. Ward, Toward more
drug like inhibitors of trypanosome alternative oxidase, ACS Infect Dis. 4 (4)
(2018 Apr 13) 592604, https://doi.org/10.1021/acsinfecdis.7b00218. Epub
2018 Jan 30 PMID: 29353481.
[69] G.U. Ebiloma, E.O. Balogun, E.J. Cueto-Díaz, H.P. de Koning, C. Dardonville,
Alternative oxidase inhibitors: mitochondrion-targeting as a strategy for new
drugs against pathogenic parasites and fungi, Med. Res. Rev. 39 (5) (2019 Sep)
15531602, https://doi.org/10.1002/med.21560. Epub 2019 Jan 29 PMID:
30693533.
[70] M. Dinant, D. Baurain, N. Coosemans, B. Joris, R.F. Matagne, Characterization of
two genes encoding the mitochondrial alternative oxidase in Chlamydomonas
reinhardtii, Curr. Genet. 39 (2) (2001 Apr) 101108, https://doi.org/10.1007/
s002940000183. PMID: 11405094.
[71] P. Stenmark, P. Nordlund, A prokaryotic alternative oxidase present in the
bacterium novosphingobium aromaticivorans, FEBS Lett. 552 (23) (2003 Sep
25) 189192, https://doi.org/10.1016/s0014-5793(03)00920-7. PMID:
14527685.
[72] J.C. Venter, K. Remington, J.F. Heidelberg, A.L. Halpern, D. Rusch, J.A. Eisen,
D. Wu, I. Paulsen, K.E. Nelson, W. Nelson, D.E. Fouts, S. Levy, A.H. Knap, M.
W. Lomas, K. Nealson, O. White, J. Peterson, J. Hoffman, R. Parsons, H. Baden-
Tillson, C. Pfannkoch, Y.H. Rogers, H.O. Smith, Environmental genome shotgun
sequencing of the Sargasso Sea, Science 304 (5667) (2004 Apr 2) 6674, https://
doi.org/10.1126/science.1093857. Epub 2004 Mar 4 PMID: 15001713.
[73] A.E. McDonald, G.C. Vanlerberghe, Alternative oxidase and plastoquinol terminal
oxidase in marine prokaryotes of the Sargasso Sea, Gene 11 (349) (2005 Apr)
1524, https://doi.org/10.1016/j.gene.2004.12.049. PMID: 15777727.
[74] D.B. Rusch, A.L. Halpern, G. Sutton, K.B. Heidelberg, S. Williamson, S. Yooseph,
D. Wu, J.A. Eisen, J.M. Hoffman, K. Remington, K. Beeson, B. Tran, H. Smith,
H. Baden-Tillson, C. Stewart, J. Thorpe, J. Freeman, C. Andrews-Pfannkoch, J.
E. Venter, K. Li, S. Kravitz, J.F. Heidelberg, T. Utterback, Y.H. Rogers, L.I. Falc´
on,
V. Souza, G. Bonilla-Rosso, L.E. Eguiarte, D.M. Karl, S. Sathyendranath, T. Platt,
E. Bermingham, V. Gallardo, G. Tamayo-Castillo, M.R. Ferrari, R.L. Strausberg,
K. Nealson, R. Friedman, M. Frazier, J.C. Venter, The sorcerer II Global Ocean
sampling expedition: Northwest Atlantic through eastern tropical Pacic, PLoS
Biol. 5 (3) (2007 Mar), e77, https://doi.org/10.1371/journal.pbio.0050077.
PMID: 17355176; PMCID: PMC1821060.
[75] A.K. Dunn, E.A. Karr, Y. Wang, A.R. Batton, E.G. Ruby, E.V. Stabb, The alternative
oxidase (AOX) gene in Vibrio scheri is controlled by NsrR and upregulated in
response to nitric oxide, Mol. Microbiol. 77 (1) (2010 Jul 1) 4455, https://doi.
org/10.1111/j.1365-2958.2010.07194.x. Epub 2010 May 4. PMID: 20487270;
PMCID: PMC3682826.
[76] A. McDonald, G. Vanlerberghe, Branched mitochondrial electron transport in the
animalia: presence of alternative oxidase in several animal phyla, IUBMB Life 56
(6) (2004 Jun) 333341, https://doi.org/10.1080/1521-6540400000876. PMID:
15370881.
[77] A.L. Moore, A.L. Umbach, J.N. Siedow, Structure-function relationships of the
alternative oxidase of plant mitochondria: a model of the active site, J. Bioenerg.
Biomembr. 27 (4) (1995 Aug) 367377, https://doi.org/10.1007/BF02109999.
PMID: 8595972.
[78] J.N. Siedow, A.L. Umbach, A.L. Moore, The active site of the cyanide-resistant
oxidase from plant mitochondria contains a binuclear iron center, FEBS Lett. 362
(1) (1995 Mar 27) 1014, https://doi.org/10.1016/0014-5793(95)00196-g.
PMID: 7698344.
[79] M.E. Andersson, P. Nordlund, A revised model of the active site of alternative
oxidase, FEBS Lett. 449 (1) (1999 Apr 16) 1722, https://doi.org/10.1016/
s0014-5793(99)00376-2. PMID: 10225419.
[80] D.A. Berthold, P. Stenmark, Membrane-bound diiron carboxylate proteins, Annu.
Rev. Plant Biol. 54 (2003) 497517, https://doi.org/10.1146/annurev.
arplant.54.031902.134915. PMID: 14503001.
[81] A.L. Moore, J.E. Carr´
e, C. Affourtit, M.S. Albury, P.G. Crichton, K. Kita,
P. Heathcote, Compelling EPR evidence that the alternative oxidase is a diiron
carboxylate protein, Biochim. Biophys. Acta 1777 (4) (2008 Apr) 327330,
https://doi.org/10.1016/j.bbabio.2008.01.004. Epub 2008 Jan 17 PMID:
18243125.
[82] M.S. Albury, C. Elliott, A.L. Moore, Towards a structural elucidation of the
alternative oxidase in plants, Physiol. Plant. 137 (4) (2009 Dec) 316327, https://
doi.org/10.1111/j.1399-3054.2009.01270.x. Epub 2009 Jul 14 PMID: 19719482.
[83] T. Shiba, Y. Kido, K. Sakamoto, D.K. Inaoka, C. Tsuge, R. Tatsumi, G. Takahashi,
E.O. Balogun, T. Nara, T. Aoki, T. Honma, A. Tanaka, M. Inoue, S. Matsuoka,
H. Saimoto, A.L. Moore, S. Harada, K. Kita, Structure of the trypanosome cyanide-
insensitive alternative oxidase, Proc. Natl. Acad. Sci. U. S. A. 110 (12) (2013 Mar
19) 45804585, https://doi.org/10.1073/pnas.1218386110.
[84] R. Pennisi, D. Salvi, V. Brandi, R. Angelini, P. Ascenzi, F. Polticelli, Molecular
evolution of alternative oxidase proteins: a phylogenetic and structure modeling
approach, J. Mol. Evol. 82 (45) (2016 May) 207218, https://doi.org/10.1007/
s00239-016-9738-8. Epub 2016 Apr 18 PMID: 27090422.
[85] T. Shiba, D.K. Inaoka, G. Takahashi, C. Tsuge, Y. Kido, L. Young, S. Ueda, E.
O. Balogun, T. Nara, T. Honma, A. Tanaka, M. Inoue, H. Saimoto, S. Harada, A.
L. Moore, K. Kita, Insights into the ubiquinol/dioxygen binding and proton relay
pathways of the alternative oxidase, Biochim. Biophys. Acta Bioenerg. 1860 (5)
(2019 May 1) 375382, https://doi.org/10.1016/j.bbabio.2019.03.008. Epub
2019 Mar 22 PMID: 30910528.
[86] D.A. Berthold, Isolation of mutants of the Arabidopsis thaliana alternative oxidase
(ubiquinol:oxygen oxidoreductase) resistant to salicylhydroxamic acid, Biochim.
Biophys. Acta 1364 (1) (1998 Apr 14) 7383, https://doi.org/10.1016/s0005-
2728(98)00015-2. PMID: 9554960.
[87] K. Nakamura, K. Sakamoto, Y. Kido, Y. Fujimoto, T. Suzuki, M. Suzuki, Y. Yabu,
N. Ohta, A. Tsuda, M. Onuma, K. Kita, Mutational analysis of the trypanosoma
vivax alternative oxidase: the E(X)6Y motif is conserved in both mitochondrial
alternative oxidase and plastid terminal oxidase and is indispensable for enzyme
activity, Biochem. Biophys. Res. Commun. 334 (2) (2005 Aug 26) 593600,
https://doi.org/10.1016/j.bbrc.2005.06.131. PMID: 16009344.
R. El-Khoury et al.
BBA - Bioenergetics 1863 (2022) 148567
8
[88] L. Young, A. Rosell-Hidalgo, D.K. Inaoka, F. Xu, M. Albury, B. May, K. Kita, A.
L. Moore, Kinetic and structural characterisation of the ubiquinol-binding site and
oxygen reduction by the trypanosomal alternative oxidase, Biochim. Biophys.
Acta Bioenerg. 1861 (10) (2020 Oct 1), 148247, https://doi.org/10.1016/j.
bbabio.2020.148247. Epub 2020 Jun 18. PMID: 32565080.
[89] D. Obenland, R. Diethelm, R. Shibles, C. Stewart, Relationship of alternative
respiratory capacity and alternative oxidase amount during soybean seedling
growth, Plant Cell Physiol. 31 (6) (1990) 897901, https://doi.org/10.1093/
oxfordjournals.pcp.a077995.
[90] A.L. Umbach, J.N. Siedow, Covalent and noncovalent dimers of the cyanide-
resistant alternative oxidase protein in higher plant mitochondria and their
relationship to enzyme activity, Plant Physiol. 103 (3) (1993 Nov) 845854,
https://doi.org/10.1104/pp.103.3.845. PMID: 12231983; PMCID: PMC159055.
[91] A.L. Umbach, J.N. Siedow, The cyanide-resistant alternative oxidases from the
fungi pichia stipitis and neurospora crassa are monomeric and lack regulatory
features of the plant enzyme, Arch. Biochem. Biophys. 378 (2) (2000 Jun 15)
234245, https://doi.org/10.1006/abbi.2000.1834. PMID: 10860541.
[92] A.E. McDonald, Alternative oxidase: an inter-kingdom perspective on the function
and regulation of this broadly distributed cyanide-resistantterminal oxidase,
Funct. Plant Biol. 35 (2008) 535, https://doi.org/10.1071/FP08025.
[93] J.T. Bahr, W.D. Bonner Jr., Cyanide-insensitive respiration. II. Control of the
alternate pathway, J. Biol. Chem. 248 (10) (1973 May 25) 34463450. PMID:
4702873.
[94] M.H. Hoefnagel, J.T. Wiskich, Activation of the plant alternative oxidase by high
reduction levels of the Q-pool and pyruvate, Arch. Biochem. Biophys. 355 (2)
(1998 Jul 15) 262270, https://doi.org/10.1006/abbi.1998.0737. PMID:
9675036.
[95] N.A. Castro-Guerrero, K. Krab, R. Moreno-S´
anchez, The alternative respiratory
pathway of euglena mitochondria, J. Bioenerg. Biomembr. 36 (5) (2004 Oct)
459469, https://doi.org/10.1023/B:JOBB.0000047328.82733.ef. PMID:
15534393.
[96] I.B. Dry, A.L. Moore, D.A. Day, J.T. Wiskich, Regulation of alternative pathway
activity in plant mitochondria: nonlinear relationship between electron ux and
the redox poise of the quinone pool, Arch. Biochem. Biophys. 273 (1) (1989 Aug
15) 148157, https://doi.org/10.1016/0003-9861(89)90173-2. PMID: 2757390.
[97] M. Ribas-Carbo, J.A. Berry, D. Yakir, L. Giles, S.A. Robinson, A.M. Lennon, J.
N. Siedow, Electron partitioning between the cytochrome and alternative
pathways in plant mitochondria, Plant Physiol. 109 (3) (1995 Nov) 829837,
https://doi.org/10.1104/pp.109.3.829. PMID: 12228636; PMCID: PMC161383.
[98] J. Selinski, A. Hartmann, A. Kordes, G. Deckers-Hebestreit, J. Whelan, R. Scheibe,
Analysis of posttranslational activation of alternative oxidase isoforms, Plant
Physiol. 174 (4) (2017 Aug) 21132127, https://doi.org/10.1104/pp.17.00681.
Epub 2017 Jun 8. PMID: 28596420; PMCID: PMC5543971.
[99] J. Selinski, A. Hartmann, G. Deckers-Hebestreit, D.A. Day, J. Whelan, R. Scheibe,
Alternative oxidase isoforms are differentially activated by tricarboxylic acid
cycle intermediates, Plant Physiol. 176 (2) (2018 Feb) 14231432, https://doi.
org/10.1104/pp.17.01331. Epub 2017 Dec 5. PMID: 29208641; PMCID:
PMC5813554.
[100] J. Azc´
on-Bieto, M. Ribas-Carb´
o, M.A. Gonz´
alez-Meler, J. Pe˜
nuelas, Sulde-
resistant respiration in leaves of Elodea canadensis michx: comparison with
cyanide-resistant respiration, Plant Physiol. 90 (4) (1989 Aug) 12491251,
https://doi.org/10.1104/pp.90.4.1249. PMID: 16666916; PMCID: PMC1061876.
[101] X. Huang, U. von Rad, J. Durner, Nitric oxide induces transcriptional activation of
the nitric oxide-tolerant alternative oxidase in arabidopsis suspension cells, Planta
215 (6) (2002 Oct) 914923, https://doi.org/10.1007/s00425-002-0828-z. Epub
2002 Jul 25 PMID: 12355151.
[102] M.D. Brand, The efciency and plasticity of mitochondrial energy transduction,
Biochem. Soc. Trans. 33 (Pt 5) (2005 Nov) 897904, https://doi.org/10.1042/
BST0330897.
[103] The thermodynamic efciency of ATP synthesis in oxidative phosphorylation,
Nath S. Biophys. Chem. 219 (2016 Dec) 6974, https://doi.org/10.1016/j.
bpc.2016.10.002. Epub 2020 Feb 10. PMID: 32040259; PMCID: PMC7131948.
[104] D. Chr´
etien, P. B´
enit, H.H. Ha, S. Keipert, R. El-Khoury, Y.T. Chang, M. Jastroch,
H.T. Jacobs, P. Rustin, M. Rak, Mitochondria are physiologically maintained at
close to 50 C, PLoS Biol. 16 (1) (2018 Jan 25), e2003992, https://doi.org/
10.1371/journal.pbio.2003992. PMID: 29370167; PMCID: PMC5784887.
[105] R. Hampp, M. Goller, H. Ziegler, Adenylate levels, energy charge, and
phosphorylation potential during dark-light and light-dark transition in
chloroplasts, mitochondria, and cytosol of mesophyll protoplasts from Avena
sativa L, Plant Physiol. 69 (2) (1982 Feb) 448455, https://doi.org/10.1104/
pp.69.2.448. PMID: 16662227; PMCID: PMC426228.
[106] A.S. Raghavendra, K. Padmasree, Benecial interactions of mitochondrial
metabolism with photosynthetic carbon assimilation, Trends Plant Sci. 8 (11)
(2003 Nov) 546553, https://doi.org/10.1016/j.tplants.2003.09.015. PMID:
14607100.
[107] P. Rustin, C. Queiroz-Claret, Changes in oxidative properties of Kalanchoe
blossfeldiana leaf mitochondria during development of crassulacean acid
metabolism, Planta 164 (3) (1985 Jun) 415422, https://doi.org/10.1007/
BF00402955. PMID: 24249613.
[108] A.C. Purvis, Role of the alternative oxidase in limiting superoxide production by
plant mitochondria, Physiol. Plant. 100 (1997) 165170, https://doi.org/
10.1111/j.1399-3054.1997.tb03468.x.
[109] D.P. Maxwell, Y. Wang, L. McIntosh, The alternative oxidase lowers
mitochondrial reactive oxygen production in plant cells, Proc. Natl. Acad. Sci. U.
S. A. 96 (14) (1999 Jul 6) 82718276, https://doi.org/10.1073/pnas.96.14.8271.
PMID: 10393984; PMCID: PMC22224.
[110] A.K. Dunn, Alternative oxidase activity reduces stress in Vibrio scheri cells
exposed to nitric oxide, J. Bacteriol. 200 (15) (2018 Jul 10), https://doi.org/
10.1128/JB.00797-17. PMID: 29760206; PMCID: PMC6040193.
[111] R. El-Khoury, K.K. Kemppainen, E. Dufour, M. Szibor, H.T. Jacobs, P. Rustin,
Engineering the alternative oxidase gene to better understand and counteract
mitochondrial defects: state of the art and perspectives, Br. J. Pharmacol. 171 (8)
(2014 Apr) 22432249, https://doi.org/10.1111/bph.12570. PMID: 24383965;
PMCID: PMC3976633.
[112] M. Hilal, A.M. Zenoff, G. Ponessa, H. Moreno, E.M. Massa, Saline stress alters the
temporal patterns of xylem differentiation and alternative oxidase expression in
developing soybean roots, Plant Physiol. 117 (2) (1998 Jun) 695701, https://
doi.org/10.1104/pp.117.2.695. PMID: 9625723; PMCID: PMC34990.
[113] D.P. Singh, C.A. Moore, A. Gilliland, J.P. Carr, Activation of multiple antiviral
defence mechanisms by salicylic acid, Mol. Plant Pathol. 5 (1) (2004 Jan 1)
5763, https://doi.org/10.1111/j.1364-3703.2004.00203.x. PMID: 20565582.
[114] S. Rachmilevitch, Y. Xu, M.A. Gonzalez-Meler, B. Huang, H. Lambers, Cytochrome
and alternative pathway activity in roots of thermal and non-thermal agrostis
species in response to high soil temperature, Physiol. Plant. 129 (2007) 163174,
https://doi.org/10.1111/j.1399-3054.2006.00784.x.
[115] Y. Murakami, K. Toriyama, Enhanced high temperature tolerance in transgenic
rice seedlings with elevated levels of alternative oxidase, OsAOX1a, Plant
Biotechnol. 25 (2008) 361364, https://doi.org/10.5511/
plantbiotechnology.25.361.
[116] A.G. Rasmusson, A.R. Fernie, J.T. van Dongen, Alternative oxidase: a defence
against metabolic uctuations? Physiol. Plant. 137 (4) (2009 Dec) 371382,
https://doi.org/10.1111/j.1399-3054.2009.01252.x. Epub 2009 May 21 PMID:
19558416.
[117] F. Hanqing, S. Kun, L. Mingquan, L. Hongyu, L. Xin, L. Yan, W. Yifeng, The
expression, function and regulation of mitochondrial alternative oxidase under
biotic stresses, Mol. Plant Pathol. 11 (3) (2010 May) 429440, https://doi.org/
10.1111/j.1364-3703.2010.00615.x. PMID: 20447290; PMCID: PMC6640418.
[118] J. Wang, N. Rajakulendran, S. Amirsadeghi, G.C. Vanlerberghe, Impact of
mitochondrial alternative oxidase expression on the response of Nicotiana
tabacum to cold temperature, Physiol. Plant. 142 (4) (2011 Aug) 339351,
https://doi.org/10.1111/j.1399-3054.2011.01471.x. Epub 2011 Apr 13 PMID:
21401618.
[119] L. Zhang, Y. Oh, H. Li, I.T. Baldwin, I. Galis, Alternative oxidase in resistance to
biotic stresses: Nicotiana attenuata AOX contributes to resistance to a pathogen
and a piercing-sucking insect but not Manduca sexta larvae, Plant Physiol. 160 (3)
(2012 Nov) 14531467, https://doi.org/10.1104/pp.112.200865. Epub 2009 Jan
31. PMID: 19366609.
[120] C. Dinakar, A. Vishwakarma, A.S. Raghavendra, K. Padmasree, Alternative
oxidase pathway optimizes photosynthesis during osmotic and temperature stress
by regulating cellular ROS, malate valve and antioxidative systems, Front. Plant
Sci. 9 (7) (2016 Feb) 68, https://doi.org/10.3389/fpls.2016.00068. PMID:
26904045; PMCID: PMC4747084.
[121] K. Dahal, G.C. Vanlerberghe, Alternative oxidase respiration maintains both
mitochondrial and chloroplast function during drought, New Phytol. 213 (2)
(2017 Jan) 560571, https://doi.org/10.1111/nph.14169. Epub 2016 Aug 31
PMID: 27579773.
[122] Z. Zalutskaya, V. Filina, M. Ostroukhova, E. Ermilova, Regulation of alternative
oxidase 1 in Chlamydomonas reinhardtii during sulfur starvation, Eur. J.
Protistol. 63 (2018 Apr) 2633, https://doi.org/10.1016/j.ejop.2018.01.001.
Epub 2018 Jan 12 PMID: 29407609.
[123] Y. Kaye, W. Huang, S. Clowez, S. Saroussi, A. Idoine, E. Sanz-Luque, A.
R. Grossman, The mitochondrial alternative oxidase from Chlamydomonas
reinhardtii enables survival in high light, J. Biol. Chem. 294 (4) (2019 Jan 25)
13801395, https://doi.org/10.1074/jbc.RA118.004667. Epub 2018 Dec 3.
PMID: 30510139; PMCID: PMC6349123.
[124] A.C. Purvis, R.L. Shewfelt, Does the alternative pathway ameliorate chilling injury
in sensitive plant tissues? Physiol. Plant. 88 (4) (1993 Aug) 712718, https://doi.
org/10.1111/j.1399-3054.1993.tb01393.x. PMID: 28741777.
[125] S. Aubert, R. Bligny, D.A. Day, J. Whelan, R. Douce, Induction of alternative
oxidase synthesis by herbicides inhibiting branched-chain amino acid synthesis,
Plant J. 11 (1997) 649657, https://doi.org/10.1046/j.1365-
313X.1997.11040649.x.
[126] D.M. Rhoads, C.C. Subbaiah, Mitochondrial retrograde regulation in plants,
Mitochondrion 7 (3) (2007 May) 177194, https://doi.org/10.1016/j.
mito.2007.01.002. Epub 2007 Jan 18 PMID: 17320492.
[127] O.H. Ruiz, A. Gonzalez, A.J. Almeida, D. Tamayo, A.M. Garcia, A. Restrepo, J.
G. McEwen, Alternative oxidase mediates pathogen resistance in Paracoccidioides
brasiliensis infection, PLoS Negl. Trop. Dis. 5 (10) (2011 Oct), e1353, https://doi.
org/10.1371/journal.pntd.0001353. Epub 2011 Oct 25. PMID: 22039556;
PMCID: PMC3201906.
[128] S. Lorin, E. Dufour, J. Boulay, O. Begel, S. Marsy, A. Sainsard-Chanet,
Overexpression of the alternative oxidase restores senescence and fertility in a
long-lived respiration-decient mutant of podospora anserina, Mol. Microbiol. 42
(5) (2001 Dec) 12591267, https://doi.org/10.1046/j.1365-2958.2001.02690.x.
PMID: 11886557.
[129] S. Akhter, H.C. McDade, J.M. Gorlach, G. Heinrich, G.M. Cox, J.R. Perfect, Role of
alternative oxidase gene in pathogenesis of Cryptococcus neoformans, Infect.
Immun. 71 (10) (2003 Oct) 57945802, https://doi.org/10.1128/
IAI.71.10.5794-5802.2003. PMID: 14500501; PMCID: PMC201089.
[130] T. Magnani, F.M. Soriani, P. Martins Vde, A.C. Policarpo, C.A. Sorgi, L.H. Faccioli,
C. Curti, S.A. Uyemura, Silencing of mitochondrial alternative oxidase gene of
aspergillus fumigatus enhances reactive oxygen species production and killing of
R. El-Khoury et al.
BBA - Bioenergetics 1863 (2022) 148567
9
the fungus by macrophages, J. Bioenerg. Biomembr. 40 (6) (2008 Dec) 631636,
https://doi.org/10.1007/s10863-008-9191-5. Epub 2009 Jan 16 PMID:
19148712.
[131] L. Yan, M. Li, Y. Cao, P. Gao, Y. Cao, Y. Wang, Y. Jiang, The alternative oxidase of
Candida albicans causes reduced uconazole susceptibility, J. Antimicrob.
Chemother. 64 (4) (2009 Oct) 764773, https://doi.org/10.1093/jac/dkp273.
Epub 2009 Aug 5 PMID: 19656781.
[132] V.P. Martins, T.M. Dinamarco, F.M. Soriani, V.G. Tudella, S.C. Oliveira, G.
H. Goldman, C. Curti, S.A. Uyemura, Involvement of an alternative oxidase in
oxidative stress and mycelium-to-yeast differentiation in Paracoccidioides
brasiliensis, Eukaryot. Cell 10 (2) (2011 Feb) 237248, https://doi.org/10.1128/
EC.00194-10. Epub 2010 Dec 23. PMID: 21183691; PMCID: PMC3067407.
[133] A.G. Rogov, E.I. Sukhanova, L.A. Uralskaya, D.A. Aliverdieva, R.A. Zvyagilskaya,
Alternative oxidase: distribution, induction, properties, structure, regulation, and
functions, Biochemistry (Mosc) 79 (13) (2014 Dec) 16151634, https://doi.org/
10.1134/S0006297914130112. PMID: 25749168.
[134] M.J. Considine, R.C. Holtzapffel, D.A. Day, J. Whelan, A.H. Millar, Molecular
distinction between alternative oxidase from monocots and dicots, Plant Physiol.
129 (3) (2002 Jul) 949953, https://doi.org/10.1104/pp.004150. PMID:
12114550; PMCID: PMC1540239.
[135] R. Clifton, A.H. Millar, J. Whelan, Alternative oxidases in arabidopsis: a
comparative analysis of differential expression in the gene family provides new
insights into function of non-phosphorylating bypasses, Biochim. Biophys. Acta
1757 (7) (2006 Jul) 730741, https://doi.org/10.1016/j.bbabio.2006.03.009.
Epub 2006 May 19 PMID: 16859634.
[136] I. Strodtk¨
otter, K. Padmasree, C. Dinakar, B. Speth, P.S. Niazi, J. Wojtera, I. Voss,
P.T. Do, A. Nunes-Nesi, A.R. Fernie, V. Linke, A.S. Raghavendra, R. Scheibe,
Induction of the AOX1D isoform of alternative oxidase in a. Thaliana T-DNA
insertion lines lacking isoform AOX1A is insufcient to optimize photosynthesis
when treated with antimycin a, Mol. Plant 2 (2) (2009 Mar) 284297, https://doi.
org/10.1093/mp/ssn089. Epub 2009 Jan 21. PMID: 19825614.
[137] K. Kühn, G. Yin, O. Duncan, S.R. Law, S. Kubiszewski-Jakubiak, P. Kaur, E. Meyer,
Y. Wang, C.C. Small, E. Giraud, R. Narsai, J. Whelan, Decreasing electron ux
through the cytochrome and/or alternative respiratory pathways triggers
common and distinct cellular responses dependent on growth conditions, Plant
Physiol. 167 (1) (2015 Jan) 228250, https://doi.org/10.1104/pp.114.249946.
Epub 2014 Nov 6. Erratum in: Plant Physiol. 2019 Jun;180(2):1242-1243. PMID:
25378695; PMCID: PMC4281006.
[138] J. Selinski, R. Scheibe, D.A. Day, J. Whelan, Alternative oxidase is positive for
plant performance, Trends Plant Sci. 23 (7) (2018 Jul) 588597, https://doi.org/
10.1016/j.tplants.2018.03.012. Epub 2018 Apr 14 PMID: 29665989.
[139] W.K. Huh, S.O. Kang, Characterization of the gene family encoding alternative
oxidase from Candida albicans, Biochem. J. 356 (Pt 2) (2001 Jun 1) 595604,
https://doi.org/10.1042/0264-6021:3560595. PMID: 11368790; PMCID:
PMC1221874.
[140] D. Uribe, G.G. Khachatourians, Identication and characterization of an
alternative oxidase in the entomopathogenic fungus metarhizium anisopliae, Can.
J. Microbiol. 54 (2) (2008 Feb) 119127, https://doi.org/10.1139/w07-127.
PMID: 18388981.
[141] O. Ju´
arez, G. Guerra, I. Vel´
azquez, O. Flores-Herrera, R.E. Rivera-P´
erez, J.
P. Pardo, The physiologic role of alternative oxidase in Ustilago maydis, FEBS J.
273 (20) (2006 Oct) 46034615, https://doi.org/10.1111/j.1742-
4658.2006.05463.x. Epub 2006 Sep 11 PMID: 16965537.
[142] T. Magnani, F.M. Soriani, V.P. Martins, A.M. Nascimento, V.G. Tudella, C. Curti,
S.A. Uyemura, Cloning and functional expression of the mitochondrial alternative
oxidase of aspergillus fumigatus and its induction by oxidative stress, FEMS
Microbiol. Lett. 271 (2) (2007 Jun) 230238, https://doi.org/10.1111/j.1574-
6968.2007.00716.x. Epub 2007 Apr 10 PMID: 17425662.
[143] K.J. Gupta, J.K. Shah, Y. Brotman, K. Jahnke, L. Willmitzer, W.M. Kaiser,
H. Bauwe, A.U. Igamberdiev, Inhibition of aconitase by nitric oxide leads to
induction of the alternative oxidase and to a shift of metabolism towards
biosynthesis of amino acids, J. Exp. Bot. 63 (4) (2012 Feb) 17731784, https://
doi.org/10.1093/jxb/ers053. PMID: 22371326.
[144] D.A. Day, J. Whelan, A.H. Millar, J.N. Siedow, J.T. Wiskich, Regulation of the
alternative oxidase in plants and fungi, Aus. J. Plant Biol. 22 (3) (1995) 497509,
https://doi.org/10.1071/PP9950497.
[145] J. Vanderleyden, C. Peeters, H. Verachtert, H. Bertrand, Stimulation of the
alternative oxidase of neurospora crassa by nucleoside phosphates, Biochem. J.
188 (1) (1980 Apr 15) 141144, https://doi.org/10.1042/bj1880141. PMID:
6447503; PMCID: PMC1162548.
[146] M. Mich´
ea-Hamzehpour, G. Turian, GMP-stimulation of the cyanide-insensitive
mitochondrial respiration in heat-shocked conidia of neurospora crassa,
Experientia 43 (4) (1987 Apr 15) 439440, https://doi.org/10.1007/
BF01940445. PMID: 3032673.
[147] S. Sakajo, N. Minagawa, A. Yoshimoto, Effects of nucleotides on cyanide-resistant
respiratory activity in mitochondria isolated from antimycin A-treated yeast
Hansenula anomala, Biosci. Biotechnol. Biochem. 61 (2) (1997 Feb) 396399,
https://doi.org/10.1271/bbb.61.396. PMID: 9058986.
[148] J. Doussi`
ere, P.V. Vignais, AMP-dependence of the cyanide-insensitive pathway in
the respiratory chain of paramecium tetraurelia, Biochem. J. 220 (3) (1984 Jun
15) 787794, https://doi.org/10.1042/bj2200787. PMID: 6547837; PMCID:
PMC1153697.
[149] A.M. Woyda-Ploszczyca, F.E. Sluse, W. Jarmuszkiewicz, Regulation of
Acanthamoeba castellanii alternative oxidase activity by mutual exclusion of
purine nucleotides; ATPs inhibitory effect, Biochim. Biophys. Acta 1787 (4)
(2009 Apr) 264271, https://doi.org/10.1016/j.bbabio.2009.01.017. Epub 2009
Jan 31. PMID: 19366609.
[150] G.A. Hakkaart, E.P. Dassa, H.T. Jacobs, P. Rustin, Allotopic expression of a
mitochondrial alternative oxidase confers cyanide resistance to human cell
respiration, EMBO Rep. 7 (3) (2006 Mar) 341345, https://doi.org/10.1038/sj.
embor.7400601. Epub 2005 Dec 2. PMID: 16322757; PMCID: PMC1456879.
[151] E. Perales-Clemente, M.P. Bayona-Bafaluy, A. P´
erez-Martos, A. Barrientos,
P. Fern´
andez-Silva, J.A. Enriquez, Restoration of electron transport without
proton pumping in mammalian mitochondria, Proc. Natl. Acad. Sci. U. S. A. 105
(48) (2008 Dec 2) 1873518739, https://doi.org/10.1073/pnas.0810518105.
Epub 2008 Nov 19. PMID: 19020091; PMCID: PMC2585044.
[152] K. Matsukawa, T. Kamata, K. Ito, Functional expression of plant alternative
oxidase decreases antimycin A-induced reactive oxygen species production in
human cells, FEBS Lett. 583 (1) (2009 Jan 5) 148152, https://doi.org/10.1016/
j.febslet.2008.11.040. Epub 2008 Dec 6 PMID: 19059403.
[153] Y. Kakizaki, R.S. Seymour, K. Ito, A novel functional element in the N-terminal
region of Arum concinnatum alternative oxidase is indispensable for catalytic
activity of the enzyme in HeLa cells, Biochim. Biophys. Acta 1797 (1) (2010 Jan)
2028, https://doi.org/10.1016/j.bbabio.2009.07.006. Epub 2009 Jul 28.
Erratum in: Biochim Biophys Acta. 2011 May;1807(5):530-1. PMID: 19643077.
[154] Y. Kakizaki, K. Ito, Engineering plant alternative oxidase function in mammalian
cells: substitution of the motif-like sequence ENV for QDT diminishes catalytic
activity of Arum concinnatum AOX1a expressed in HeLa cells, Appl. Biochem.
Biotechnol. 170 (5) (2013 Jul) 12291240, https://doi.org/10.1007/s12010-013-
0235-x. Epub 2013 May 8 PMID: 23653140.
[155] E.P. Dassa, E. Dufour, S. Gonçalves, V. Paupe, G.A. Hakkaart, H.T. Jacobs,
P. Rustin, Expression of the alternative oxidase complements cytochrome c
oxidase deciency in human cells, EMBO Mol. Med. 1 (1) (2009 Apr) 3036,
https://doi.org/10.1002/emmm.200900001. PMID: 20049701; PMCID:
PMC3378104.
[156] E.P. Dassa, E. Dufour, S. Goncalves, H.T. Jacobs, P. Rustin, The alternative
oxidase, a tool for compensating cytochrome c oxidase deciency in human cells,
Physiol. Plant. 137 (4) (2009 Dec) 427434, https://doi.org/10.1111/j.1399-
3054.2009.01248.x. Epub 2009 Apr 25 PMID: 19493305.
[157] D.J. Fernandez-Ayala, A. Sanz, S. Vartiainen, K.K. Kemppainen, M. Babusiak,
E. Mustalahti, R. Costa, T. Tuomela, M. Zeviani, J. Chung, K.M. ODell, P. Rustin,
H.T. Jacobs, Expression of the Ciona intestinalis alternative oxidase (AOX) in
drosophila complements defects in mitochondrial oxidative phosphorylation, Cell
Metab. 9 (5) (2009 May) 449460, https://doi.org/10.1016/j.cmet.2009.03.004.
PMID: 19416715.
[158] K.K. Kemppainen, J. Rinne, A. Sriram, M. Lakanmaa, A. Zeb, T. Tuomela,
A. Popplestone, S. Singh, A. Sanz, P. Rustin, H.T. Jacobs, Expression of alternative
oxidase in Drosophila ameliorates diverse phenotypes due to cytochrome oxidase
deciency, Hum. Mol. Genet. 23 (8) (2014 Apr 15) 20782093, https://doi.org/
10.1093/hmg/ddt601. Epub 2013 Nov 29. PMID: 24293544; PMCID:
PMC3959817.
[159] R. El-Khoury, E. Kaulio, K.A. Lassila, D.C. Crowther, H.T. Jacobs, P. Rustin,
Expression of the alternative oxidase mitigates beta-amyloid production and
toxicity in model systems, Free Radic. Biol. Med. 96 (2016 Jul) 5766, https://
doi.org/10.1016/j.freeradbiomed.2016.04.006. Epub 2016 Apr 14 PMID:
27094492.
[160] D.M. Humphrey, R.B. Parsons, Z.N. Ludlow, T. Riemensperger, G. Esposito,
P. Verstreken, H.T. Jacobs, S. Birman, F. Hirth, Alternative oxidase rescues
mitochondria-mediated dopaminergic cell loss in drosophila, Hum. Mol. Genet.
21 (12) (2012 Jun 15) 26982712, https://doi.org/10.1093/hmg/dds096. Epub
2012 Mar 7 PMID: 22398207.
[161] A. Sanz, D.J. Fern´
andez-Ayala, R.K. Stefanatos, H.T. Jacobs, Mitochondrial ROS
production correlates with, but does not directly regulate lifespan in drosophila,
Aging (Albany NY) 2 (4) (2010 Apr) 200223, https://doi.org/10.18632/
aging.100137. PMID: 20453260; PMCID: PMC2880708.
[162] A. Andjelkovi´
c, A. Mordas, L. Bruinsma, A. Ketola, G. Cannino, L. Giordano, P.
K. Dhandapani, M. Szibor, E. Dufour, H.T. Jacobs, Expression of the alternative
oxidase inuences Jun N-terminal kinase signaling and cell migration, Mol. Cell.
Biol. 38 (24) (2018 Nov 28), https://doi.org/10.1128/MCB.00110-18. PMID:
30224521; PMCID: PMC6275184.
[163] S. Saari, G.S. Garcia, K. Bremer, M.M. Chioda, A. Andjelkovi´
c, P.V. Debes,
M. Nikinmaa, M. Szibor, E. Dufour, P. Rustin, M.T. Oliveira, H.T. Jacobs,
Alternative respiratory chain enzymes: therapeutic potential and possible pitfalls,
Biochim. Biophys. Acta Mol. basis Dis. 1865 (4) (2019 Apr 1) 854866, https://
doi.org/10.1016/j.bbadis.2018.10.012. Epub 2018 Oct 17 PMID: 30342157.
[164] S. Saari, A. Andjelkovi´
c, G.S. Garcia, H.T. Jacobs, M.T. Oliveira, Expression of
Ciona intestinalis AOX causes male reproductive defects in Drosophila
melanogaster, BMC Dev. Biol. 17 (1) (2017 Jul 3) 9, https://doi.org/10.1186/
s12861-017-0151-3. PMID: 28673232; PMCID: PMC5496232.
[165] B.R. Turnell, L. Kumpitsch, K. Reinhardt, Production and scavenging of reactive
oxygen species both affect reproductive success in male and female Drosophila
melanogaster, Biogerontology 22 (4) (2021 Aug) 379396, https://doi.org/
10.1007/s10522-021-09922-1. Epub 2021 Apr 26. PMID: 33903991; PMCID:
PMC8266701.
[166] A. Guar´
as, E. Perales-Clemente, E. Calvo, R. Acín-P´
erez, M. Loureiro-Lopez,
C. Pujol, I. Martínez-Carrascoso, E. Nu˜
nez, F. García-Marqu´
es, M.A. Rodríguez-
Hern´
andez, A. Cort´
es, F. Diaz, A. P´
erez-Martos, C.T. Moraes, P. Fern´
andez-Silva,
A. Trifunovic, P. Navas, J. Vazquez, J.A. Enríquez, The CoQH2/CoQ ratio serves
as a sensor of respiratory chain efciency, Cell Rep. 15 (1) (2016 Apr 5) 197209,
https://doi.org/10.1016/j.celrep.2016.03.009. Epub 2016 Mar 24 PMID:
27052170.
R. El-Khoury et al.
BBA - Bioenergetics 1863 (2022) 148567
10
[167] Y.L. Chua, E. Dufour, E.P. Dassa, P. Rustin, H.T. Jacobs, C.T. Taylor, T. Hagen,
Stabilization of hypoxia-inducible factor-1alpha protein in hypoxia occurs
independently of mitochondrial reactive oxygen species production, J. Biol.
Chem. 285 (41) (2010 Oct 8) 3127731284, https://doi.org/10.1074/jbc.
M110.158485. Epub 2010 Jul 30. PMID: 20675386; PMCID: PMC2951202.
[168] R. El-Khoury, E. Dufour, M. Rak, N. Ramanantsoa, N. Grandchamp, Z. Csaba,
B. Duvilli´
e, P. B´
enit, J. Gallego, P. Gressens, C. Sarkis, H.T. Jacobs, P. Rustin,
Alternative oxidase expression in the mouse enables bypassing cytochrome c
oxidase blockade and limits mitochondrial ROS overproduction, PLoS Genet. 9 (1)
(2013), e1003182, https://doi.org/10.1371/journal.pgen.1003182. Epub 2013
Jan 3. PMID: 23300486; PMCID: PMC3536694.
[169] M. Szibor, P.K. Dhandapani, E. Dufour, K.M. Holmstr¨
om, Y. Zhuang, I. Salwig,
I. Wittig, J. Heidler, Z. Gizatullina, T. Gainutdinov, German Mouse Clinic
Consortium, H. Fuchs, V. Gailus-Durner, M.H. de Angelis, J. Nandania,
V. Velagapudi, A. Wietelmann, P. Rustin, F.N. Gellerich, H.T. Jacobs, T. Braun,
Broad AOX expression in a genetically tractable mouse model does not disturb
normal physiology, Dis. Model. Mech. 10 (2) (2017 Feb 1) 163171, https://doi.
org/10.1242/dmm.027839. Epub 2016 Dec 14. PMID: 28067626; PMCID:
PMC5312010.
[170] P.K. Dhandapani, I.M. Begines-Moreno, G. Brea-Calvo, U. G¨
artner, T.G. Graeber,
G. Javier Sanchez, R.E. Morty, K. Sch¨
onig, J.T. Hoeve, A. Wietelmann, T. Braun,
H.T. Jacobs, M. Szibor, Hyperoxia but not AOX expression mitigates pathological
cardiac remodeling in a mouse model of inammatory cardiomyopathy, Sci. Rep.
9 (1) (2019 Sep 4) 12741, https://doi.org/10.1038/s41598-019-49231-9. PMID:
31484989; PMCID: PMC6726756.
[171] L. Giordano, A. Farnham, P.K. Dhandapani, L. Salminen, J. Bhaskaran,
R. Voswinckel, P. Rauschkolb, S. Scheibe, N. Sommer, C. Beisswenger,
N. Weissmann, T. Braun, H.T. Jacobs, R. Bals, C. Herr, M. Szibor, Alternative
oxidase attenuates cigarette smoke-induced lung dysfunction and tissue damage,
Am. J. Respir. Cell Mol. Biol. 60 (5) (2019 May) 515522, https://doi.org/
10.1165/rcmb.2018-0261OC. PMID: 30339461; PMCID: PMC6503618.
[172] P.K. Dhandapani, I.M. Begines-Moreno, G. Brea-Calvo, U. G¨
artner, T.G. Graeber,
G. Javier Sanchez, R.E. Morty, K. Sch¨
onig, J.T. Hoeve, A. Wietelmann, T. Braun,
H.T. Jacobs, M. Szibor, Hyperoxia but not AOX expression mitigates pathological
cardiac remodeling in a mouse model of inammatory cardiomyopathy, Sci. Rep.
9 (1) (2019 Sep 4) 12741, https://doi.org/10.1038/s41598-019-49231-9. PMID:
31484989; PMCID: PMC6726756.
[173] E.L. Mills, B. Kelly, A. Logan, Costa ASH, M. Varma, C.E. Bryant, P. Tourlomousis,
J.H.M. D¨
abritz, E. Gottlieb, I. Latorre, S.C. Corr, D. Ryan, H.T. Jacobs, M. Szibor,
R.J. Xavier, T. Braun, C. Frezza, M.P. Murphy, L.A. ONeill, G. McManus,
Succinate dehydrogenase supports metabolic repurposing of mitochondria to
drive inammatory macrophages, Cell 167 (2) (2016 Oct 6) 457470, https://doi.
org/10.1016/j.cell.2016.08.064. Epub 2014 Nov 6. Erratum in: Plant Physiol.
2019 Jun;180(2):1242-1243. PMID: 25378695; PMCID: PMC4281006.
[174] J. Rajendran, J. Purhonen, S. Tegelberg, O.P. Smolander, M. M¨
orgelin, J. Rozman,
V. Gailus-Durner, H. Fuchs, M. Hrabe de Angelis, P. Auvinen, E. Mervaala, H.
T. Jacobs, M. Szibor, V. Fellman, J. Kallij¨
arvi, Alternative oxidase-mediated
respiration prevents lethal mitochondrial cardiomyopathy, EMBO Mol. Med. 11
(1) (2019 Jan), e9456, https://doi.org/10.15252/emmm.201809456. PMID:
30530468; PMCID: PMC6328925.
[175] S.A. Dogan, R. Cerutti, C. Beninc´
a, G. Brea-Calvo, H.T. Jacobs, M. Zeviani,
M. Szibor, C. Viscomi, Perturbed redox signaling exacerbates a mitochondrial
myopathy, e5, Cell Metab. 28 (5) (2018 Nov 6) 764775, https://doi.org/
10.1016/j.cmet.2018.07.012. Epub 2018 Aug 16. PMID: 30122554; PMCID:
PMC6224544.
[176] M. Szibor, R. Schreckenberg, Z. Gizatullina, E. Dufour, M. Wiesnet, P.
K. Dhandapani, G. Debska-Vielhaber, J. Heidler, I. Wittig, T.A. Nyman,
U. G¨
artner, A.R. Hall, V. Pell, C. Viscomi, T. Krieg, M.P. Murphy, T. Braun, F.
N. Gellerich, K.D. Schlüter, H.T. Jacobs, Respiratory chain signalling is essential
for adaptive remodelling following cardiac ischaemia, J. Cell. Mol. Med. 24 (6)
(2020 Mar) 35343548, https://doi.org/10.1111/jcmm.15043. Epub 2020 Feb
10. PMID: 32040259; PMCID: PMC7131948.
[177] N. Sommer, N. Alebrahimdehkordi, O. Pak, F. Knoepp, I. Strielkov, S. Scheibe,
E. Dufour, A. Andjelkovi´
c, A. Sydykov, A. Saraji, A. Petrovic, K. Quanz,
M. Hecker, M. Kumar, J. Wahl, S. Kraut, W. Seeger, R.T. Schermuly, H.
A. Ghofrani, K. Ramser, T. Braun, H.T. Jacobs, N. Weissmann, M. Szibor,
Bypassing mitochondrial complex III using alternative oxidase inhibits acute
pulmonary oxygen sensing, Sci. Adv. 6 (16) (2020 Apr 15), eaba0694, https://
doi.org/10.1126/sciadv.aba0694. PMID: 32426457; PMCID: PMC7159913.
[178] E. Calvo, S. Cogliati, P. Hernansanz-Agustín, M. Loureiro-L´
opez, A. Guar´
as, R.
A. Casuso, F. García-Marqu´
es, R. Acín-P´
erez, Y. Martí-Mateos, J.C. Silla-Castro,
M. Carro-Alvarellos, J.R. Huertas, J. V´
azquez, J.A. Enríquez, Functional role of
respiratory supercomplexes in mice: SCAF1 relevance and segmentation of the
Qpool, Sci. Adv. 6 (26) (2020 Jun 24), eaba7509, https://doi.org/10.1126/
sciadv.aba7509. PMID: 32637615; PMCID: PMC7314541.
[179] M. Szibor, T. Gainutdinov, E. Fernandez-Vizarra, E. Dufour, Z. Gizatullina,
G. Debska-Vielhaber, J. Heidler, I. Wittig, C. Viscomi, F. Gellerich, A.L. Moore,
Bioenergetic consequences from xenotopic expression of a tunicate AOX in mouse
mitochondria: switch from RET and ROS to FET, Biochim. Biophys. Acta
Bioenerg. 1861 (2) (2020 Feb 1), 148137, https://doi.org/10.1016/j.
bbabio.2019.148137. Epub 2019 Dec 9. PMID: 31825809.
[180] L. Giordano, M.K. Aneja, N. Sommer, N. Alebrahimdehkordi, A. Seraji,
N. Weissmann, C. Rudolph, C. Plank, H.T. Jacobs, M. Szibor, Alternative oxidase
encoded by sequence-optimized and chemically-modied RNA transfected into
mammalian cells is catalytically active, Gene Ther. (2021 Mar 4), https://doi.org/
10.1038/s41434-021-00235-z. Epub 2016 Dec 14. PMID: 28067626; PMCID:
PMC5312010.
R. El-Khoury et al.
... AOX is the terminal oxidase of the cyanide resistant respiratory pathway and its expression at the protein level reflects the activity of cyanide resistant respiratory pathways [36]. As shown in Figure 7, AOX was highly increased in response to 0.1 and 1 µM 2 ,4 -dichloro-chalcone in both F. tricinctum and T. roseum. ...
... AOX is the terminal oxidase of the cyanide resistant respiratory pathway and its expression at the protein level reflects the activity of cyanide resistant respiratory pathways [36]. As shown in Figure 7, AOX was highly increased in response to 0.1 and 1 µM 2′,4′dichloro-chalcone in both F. tricinctum and T. roseum. ...
Article
Full-text available
Chalcones are a class of flavonoids possessing antimicrobial properties and have potential for use as coatings of plant products for the control of postharvest diseases. The effects of 2′,4′-dichloro-chalcone on the in vitro growth and in vivo pathogenicity of Fusarium tricinctum and Trichothecium roseum were investigated. First, 1 µM of 2′,4′-dichloro-chalcone strongly inhibited the mycelial growth and conidial production of F. tricinctum (32.3%) and T. roseum (65.2%) in vitro. Meanwhile, the cell membrane permeability was increased by 25% and 22.5% and the accumulation of reactive oxygen species was increased by 41.7 and 65.4%, respectively, of F. tricinctum and T. roseum. This treatment also significantly inhibited the total respiration rate and activated the cyanide-resistant respiratory pathway in both pathogens. The expression level of AOX was enhanced in F. tricinctum and T. roseum by 52.76 and 39.13%, respectively. This treatment also significantly inhibited the expansion of potato dry rot from F. tricinctum (48.6%) and apple rot spot from T. roseum (36.2%). Therefore, 2′,4′-dichloro-chalcone has potential use as an alternative safety method in the control of postharvest diseases by F. tricinctum and T. roseum in agricultural practices.
... AOX is the terminal oxidase of the cyanide resistant respiratory pathway and its expression at the protein level reflects the activity of cyanide resistant respiratory pathway [34]. As shown in Figure 7, AOX was highly increased in response to 10 and 100 µM 2',4'-dichloro-chalcone in both F. tricinctum and T. roseum. ...
Preprint
Full-text available
Chalcones are a class of flavonoids possessing a variety of biological activities, including antimicrobial and antioxidant effects. Therefore, chalcones and their derivatives have potential use in the control of postharvest diseases. In this work, the effects of 2ʹ,4ʹ-dichloro-chalcone on the in vitro growth and in vivo pathogenicity of F. tricinctum and T. roseum were investigated. The results showed that 100 µM of 2ʹ,4ʹ-dichloro-chalcone strongly inhibited mycelial growth and conidial production of F. tricinctum (32.3%) and T. roseum (65.2%) in vitro. This treatment also significantly inhibited the expansion of potato dry rot from F. tricinctum (48.6%) and apple rot spot from T. roseum (36.2%). The incubation of F. tricinctum and T. roseum conidia with this agent for 2 h increased their cell membrane permeability by 25% and 22.5%, respectively and conidial membrane permeability by 41.7 and 65.4%, respectively. This treatment also significantly inhibited the total respiration rate and activated the cyanide-resistant respiratory pathway in both pathogens. Protein immunoblotting showed that the treatment increased alternative oxidase (AOX) levels after 4 h in F. tricinctum and T. roseum by 52.76% and 39.13%, respectively. Conclusion: 100 µM 2ʹ,4ʹ-dichloro-chalcone significantly inhibited mycelial growth and spore production in F. tricinctum and T. roseum in vitro and reduced their pathogenicity in postharvest potato and apple crops. Further analysis indicated that 2ʹ,4ʹ-dichloro-chalcone disrupted the integrity of cell membranes and the mitochondrial respiratory electron transport chain which leaded to ROS burst and oxidative stress, AOXs were induced directly or indirectly, and the cyanide resistant respiratory pathway was activated. The in vitro growth and pathogenicity of F. tricinctum and T. roseum were inhibited.
Article
Full-text available
The inflorescence (spadix) of skunk cabbage (Symplocarpus renifolius) is strongly thermogenic and can regulate its temperature at around 23 °C even when the ambient temperature drops below freezing. To elucidate the mechanisms underlying developmentally controlled thermogenesis and thermoregulation in skunk cabbage, we conducted a comprehensive transcriptome and metabolome analysis across 3 developmental stages of spadix development. Our RNA-seq analysis revealed distinct groups of expressed genes, with selenium-binding protein 1/methanethiol oxidase (SBP1/MTO) exhibiting the highest levels in thermo-genic florets. Notably, the expression of alternative oxidase (AOX) was consistently high from the prethermogenic stage through the thermogenic stage in the florets. Metabolome analysis showed that alterations in nucleotide levels correspond with the developmentally controlled and tissue-specific thermogenesis of skunk cabbage, evident by a substantial increase in AMP levels in thermogenic florets. Our study also reveals that hydrogen sulfide, a product of SBP1/MTO, inhibits cytochrome c oxidase (COX)-mediated mitochondrial respiration, while AOX-mediated respiration remains relatively unaffected. Specifically, at lower temperatures, the inhibitory effect of hydrogen sulfide on COX-mediated respiration increases, promoting a shift toward the dominance of AOX-mediated respiration. Finally, despite the differential regulation of genes and metabolites throughout spa-dix development, we observed a convergence of gene expression and metabolite accumulation patterns during thermogenesis. This synchrony may play a key role in developmentally regulated thermogenesis. Moreover, such convergence during the thermogenic stage in the spadix may provide a solid molecular basis for thermoregulation in skunk cabbage.
Article
The increasing global population projected to reach 9.7 billion by 2050 from the current 7.7 billion, which is going to significantly impact food availability. Therefore, current global agricultural production needs to be increased to feed the unconstrained growing population. The changing climatic condition due to anthropogenic activities also makes the conditions more challenging to meet the required crop productivity in the future. Potato is third most consumed staple food. The current cultivation and demand of potato is particularly more in developing countries with high levels of poverty, hunger, and malnutrition because it is cheap source of nutrition and easily available. Potato is severely infected by a number of diseases, insect pests, and abiotic environmental conditions. Amidst the backdrop of climate change, the situation is deteriorating. Projections indicate that the average potato productivity in India's primary potato-growing states, responsible for approximately 90% of the nation's total potato output, is expected to decline by 2.0% in the 2050s and a more substantial 6.4% in the 2080s. Therefore to develop tolerance in plants to cope with changing environmental condition is need of hour to feed the overgrowing population. In this review, we discussed the application of CRISPR to enhance the crop productivity and develop biotic and abiotic stress-tolerant in potato to face the current changing climatic conditions is also discussed. The review also sheds light on the future prospects of CRISPR/Cas technology for potato breeding, including potential challenges and opportunities.
Article
Full-text available
The inflorescence (spadix) of skunk cabbage (Symplocarpus renifolius) is strongly thermogenic and can regulate its temperature at around 23°C even when the ambient temperature drops below freezing. To elucidate the mechanisms underlying developmentally controlled thermogenesis and thermoregulation in skunk cabbage, we conducted a comprehensive transcriptome and metabolome analysis across three developmental stages of spadix development. Our RNA-seq analysis revealed distinct groups of expressed genes, with selenium-binding protein 1/methanethiol oxidase (SBP1/MTO) exhibiting the highest levels in thermogenic florets. Notably, the expression of alternative oxidase (AOX) was consistently high from the pre-thermogenic stage through the thermogenic stage in the florets. Metabolome analysis showed that alterations in nucleotide levels correspond with the developmentally controlled and tissue-specific thermogenesis of skunk cabbage, evident by a substantial increase in AMP levels in thermogenic florets. Our study also reveals that hydrogen sulfide, a product of SBP1/MTO, inhibits cytochrome c (COX)-mediated mitochondrial respiration while AOX-mediated respiration remains relatively unaffected. Specifically, at lower temperatures, the inhibitory effect of hydrogen sulfide on COX-mediated respiration increases, promoting a shift towards the dominance of AOX-mediated respiration. Finally, despite the differential regulation of genes and metabolites throughout spadix development, we observed a convergence of gene expression and metabolite accumulation patterns during thermogenesis. This synchrony may play a key role in developmentally regulated thermogenesis. Moreover, such convergence during the thermogenic stage in the spadix may provide a solid molecular basis for thermoregulation in skunk cabbage.
Article
Full-text available
Hydrogen cyanide (HCN) occurs in living organisms and in the environment. This is a widely known poison but is also considered as a gasotransmitter. For most higher plants, microorganisms and animals HCN is toxic, especially at elevated concentrations. However, plants’ sensitivity to this compound is lower than animals’ due to the activity of an alternative oxidase in the mitochondrial respiration chain. All higher plants synthesize HCN as a co-product during the final step of ethylene biosynthesis, whilst some plant species release it from cyanogenic compounds, accumulated for diverse physiological purposes. This molecule is used as a toxic bomb against herbivores, as a source of nitrogen in N-deficient plants, or as a regulator of seed dormancy state. The toxicity of HCN is mainly due to the inhibition of the activity of several metalloenzymes: iron-containing enzymes, molybdoenzymes and enzymes that contain zinc or copper. HCN impacts cellular metabolism by modulation of the reactive oxygen species and reactive nitrogen species levels, and via modifications of proteins (S-cyanylation, oxidation). The aim of this work is to describe the dual (toxic and signalling) mode of cyanide action in plants at a cellular level.
Article
The alternative oxidase (AOX) is a terminal oxidase in the electron transport system that plays a role in mitochondrial bioenergetics. The past 20 years of research shows AOX has a wide yet patchy distribution across the tree of life. AOX has been suggested to have a role in stress tolerance, growth, and development in plants, but less is known about its function in other groups, including animals. In this study, we analyzed the taxonomic distribution of AOX across >2800 species representatives from prokaryotes and eukaryotes and developed a standardized workflow for finding and verifying the authenticity of AOX sequences. We found that AOX is limited to proteobacteria among prokaryotes, but is widely distributed in eukaryotes, with the highest prevalence in plants, fungi, and protists. AOX is present in many invertebrates, but is absent in others including most arthropods, and is absent from vertebrates. We found aberrant AOX sequences associated with some animal groups. Some of these aberrant AOXs were contaminants, but we also found putative cases of lateral gene transfer of AOX from fungi and protists to nematodes, springtails, fungus gnats, and rotifers. Our findings provide a robust and detailed analysis of the distribution of AOX and a method for identifying and verifying putative AOX sequences, which will be useful as more sequence data becomes available on public repositories.
Article
The mitochondrial respiratory chain or electron transport chain (ETC) facilitates redox reactions which ultimately lead to the reduction of oxygen to water (respiration). Energy released by this process is used to establish a proton electrochemical gradient which drives ATP formation (oxidative phosphorylation, OXPHOS). It also plays an important role in vital processes beyond ATP formation and cellular metabolism, such as heat production, redox and ion homeostasis. Dysfunction of the ETC can thus impair cellular and organismal viability and is thought to be the underlying cause of a heterogeneous group of so-called mitochondrial diseases. Plants, yeasts, and many lower organisms, but not insects and vertebrates, possess an enzymatic mechanism that confers resistance to respiratory stress conditions, i.e. the alternative oxidase (AOX). Even in cells that naturally lack AOX, it is autonomously imported into the mitochondrial compartment upon xenotopic expression, where it refolds and becomes catalytically engaged when the cytochrome segment of the ETC is blocked. AOX was therefore proposed as a tool to study disease etiologies. To this end, AOX has been xenotopically expressed in mammalian cells and disease models of the fruit fly and mouse. Surprisingly, AOX showed remarkable rescue effects in some cases, whilst in others it had no effect or even exacerbated a condition. Here we summarize what has been learnt from the use of AOX in various disease models and discuss issues which still need to be addressed in order to understand the role of the ETC in health and disease.
Article
Hypochlorite (ClO⁻) and cyanide (CN⁻) are closely associated with various pathophysiological processes, it's necessary to develop an efficient method for their detection. In view of the advantages of fluorescent probes and the convenience of organic synthesis, we rationally constructed a novel near-infrared “turn-off” fluorescent probe BPQO based on phenothiazine and 8-hydroxyquinaldine. Probe BPQO can specifically and sensitively detect ClO⁻ (LOD = 2.0 μM) and CN⁻ (LOD = 3.3 μM), and the response processes can be completed in 42 s and 50 s, respectively. In addition, it was proved to be so low toxicity that it has been successfully used for imaging detection of exogenous CN⁻ in HepG2 cells.
Article
Full-text available
Sperm aging is accelerated by the buildup of reactive oxygen species (ROS), which cause oxidative damage to various cellular components. Aging can be slowed by limiting the production of mitochondrial ROS and by increasing the production of antioxidants, both of which can be generated in the sperm cell itself or in the surrounding somatic tissues of the male and female reproductive tracts. However, few studies have compared the separate contributions of ROS production and ROS scavenging to sperm aging, or to cellular aging in general. We measured reproductive fitness in two lines of Drosophila melanogaster genetically engineered to (1) produce fewer ROS via expression of alternative oxidase (AOX), an alternative respiratory pathway; or (2) scavenge fewer ROS due to a loss-of-function mutation in the antioxidant gene dj-1β. Wild-type females mated to AOX males had increased fecundity and longer fertility durations, consistent with slower aging in AOX sperm. Contrary to expectations, fitness was not reduced in wild-type females mated to dj-1β males. Fecundity and fertility duration were increased in AOX and decreased in dj-1β females, indicating that female ROS levels may affect aging rates in stored sperm and/or eggs. Finally, we found evidence that accelerated aging in dj-1β sperm may have selected for more frequent mating. Our results help to clarify the relative roles of ROS production and ROS scavenging in the male and female reproductive systems.
Article
Full-text available
Plants and other organisms, but not insects or vertebrates, express the auxiliary respiratory enzyme alternative oxidase (AOX) that bypasses mitochondrial respiratory complexes III and/or IV when impaired. Persistent expression of AOX from Ciona intestinalis in mammalian models has previously been shown to be effective in alleviating some metabolic stresses produced by respiratory chain inhibition while exacerbating others. This implies that chronic AOX expression may modify or disrupt metabolic signaling processes necessary to orchestrate adaptive remodeling, suggesting that its potential therapeutic use may be confined to acute pathologies, where a single course of treatment would suffice. One possible route for administering AOX transiently is AOX-encoding nucleic acid constructs. Here we demonstrate that AOX-encoding chemically-modified RNA (cmRNA), sequence-optimized for expression in mammalian cells, was able to support AOX expression in immortalized mouse embryonic fibroblasts (iMEFs), human lung carcinoma cells (A549) and primary mouse pulmonary arterial smooth muscle cells (PASMCs). AOX protein was detectable as early as 3 h after transfection, had a half-life of ~4 days and was catalytically active, thus supporting respiration and protecting against respiratory inhibition. Our data demonstrate that AOX-encoding cmRNA optimized for use in mammalian cells represents a viable route to investigate and possibly treat mitochondrial respiratory disorders.
Article
Full-text available
Mitochondrial respiratory complexes assemble into supercomplexes (SC). Q-respirasome (III 2 + IV) requires the supercomplex assembly factor (SCAF1) protein. The role of this factor in the N-respirasome (I + III 2 + IV) and the physiological role of SCs are controversial. Here, we study C57BL/6J mice harboring nonfunctional SCAF1, the full knockout for SCAF1, or the wild-type version of the protein and found that exercise performance is SCAF1 dependent. By combining quantitative data–independent proteomics, 2D Blue native gel electrophoresis, and functional analysis of enriched respirasome fractions, we show that SCAF1 confers structural attachment between III 2 and IV within the N-respirasome, increases NADH-dependent respiration, and reduces reactive oxygen species (ROS). Furthermore, the expression of AOX in cells and mice confirms that CI-CIII superassembly segments the CoQ in two pools and modulates CI-NADH oxidative capacity.
Article
Full-text available
Mitochondria play an important role in sensing both acute and chronic hypoxia in the pulmonary vasculature, but their primary oxygen-sensing mechanism and contribution to stabilization of the hypoxia-inducible factor (HIF) remains elusive. Alteration of the mitochondrial electron flux and increased superoxide release from complex III has been proposed as an essential trigger for hypoxic pulmonary vasoconstriction (HPV). We used mice expressing a tunicate alternative oxidase, AOX, which maintains electron flux when respiratory complexes III and/or IV are inhibited. Respiratory restoration by AOX prevented acute HPV and hypoxic responses of pulmonary arterial smooth muscle cells (PASMC), acute hypoxia-induced redox changes of NADH and cytochrome c, and superoxide production. In contrast, AOX did not affect the development of chronic hypoxia-induced pulmonary hypertension and HIF-1α stabilization. These results indicate that distal inhibition of the mitochondrial electron transport chain in PASMC is an essential initial step for acute but not chronic oxygen sensing.
Article
Full-text available
Cardiac ischaemia‐reperfusion (I/R) injury has been attributed to stress signals arising from an impaired mitochondrial electron transport chain (ETC), which include redox imbalance, metabolic stalling and excessive production of reactive oxygen species (ROS). The alternative oxidase (AOX) is a respiratory enzyme, absent in mammals, that accepts electrons from a reduced quinone pool to reduce oxygen to water, thereby restoring electron flux when impaired and, in the process, blunting ROS production. Hence, AOX represents a natural rescue mechanism from respiratory stress. This study aimed to determine how respiratory restoration through xenotopically expressed AOX affects the re‐perfused post‐ischaemic mouse heart. As expected, AOX supports ETC function and attenuates the ROS load in post‐anoxic heart mitochondria. However, post‐ischaemic cardiac remodelling over 3 and 9 weeks was not improved. AOX blunted transcript levels of factors known to be up‐regulated upon I/R such as the atrial natriuretic peptide (Anp) whilst expression of pro‐fibrotic and pro‐apoptotic transcripts were increased. Ex vivo analysis revealed contractile failure at nine but not 3 weeks after ischaemia whilst label‐free quantitative proteomics identified an increase in proteins promoting adverse extracellular matrix remodelling. Together, this indicates an essential role for ETC‐derived signals during cardiac adaptive remodelling and identified ROS as a possible effector.
Article
Full-text available
Constitutive expression of the chemokine Mcp1 in mouse cardiomyocytes creates a model of inflammatory cardiomyopathy, with death from heart failure at age 7–8 months. A critical pathogenic role has previously been proposed for induced oxidative stress, involving NADPH oxidase activation. To test this idea, we exposed the mice to elevated oxygen levels. Against expectation, this prevented, rather than accelerated, the ultrastructural and functional signs of heart failure. This result suggests that the immune signaling initiated by Mcp1 leads instead to the inhibition of cellular oxygen usage, for which mitochondrial respiration is an obvious target. To address this hypothesis, we combined the Mcp1 model with xenotopic expression of the alternative oxidase (AOX), which provides a sink for electrons blocked from passage to oxygen via respiratory complexes III and IV. Ubiquitous AOX expression provided only a minor delay to cardiac functional deterioration and did not prevent the induction of markers of cardiac and metabolic remodeling considered a hallmark of the model. Moreover, cardiomyocyte-specific AOX expression resulted in exacerbation of Mcp1-induced heart failure, and failed to rescue a second cardiomyopathy model directly involving loss of cIV. Our findings imply that mitochondrial involvement in the pathology of inflammatory cardiomyopathy is multifaceted and complex.
Article
Neurospora crassa mitochondria use a branched electron transport system in which one branch is a conventional cytochrome system and the other is an alternative cyanide-resistant, hydroxamic acid-sensitive oxidase that is induced when the cytochrome system is impaired. We used a monoclonal antibody to the alternative oxidase of the higher plant Sauromatum guttatum to identify a similar set of related polypeptides (Mr, 36,500 and 37,000) that was associated with the alternative oxidase activity of N. crassa mitochondria. These polypeptides were not present constitutively in the mitochondria of a wild-type N. crassa strain, but were produced in high amounts under conditions that induced alternative oxidase activity. Under the same conditions, mutants in the aod-1 gene, with one exception, produced apparently inactive alternative oxidase polypeptides, whereas mutants in the aod-2 gene failed to produce these polypeptides. The latter findings support the hypothesis that aod-1 is a structural gene for the alternative oxidase and that the aod-2 gene encodes a component that is required for induction of alternative oxidase activity. Finally, our results indicate that the alternative oxidase is highly conserved, even between plant and fungal species.
Article
The alternative oxidase (AOX) is a monotopic di‑iron carboxylate protein which acts as a terminal respiratory chain oxidase in a variety of plants, fungi and protists. Of particular importance is the finding that both emerging infectious diseases caused by human and plant fungal pathogens, the majority of which are multi-drug resistant, appear to be dependent upon AOX activity for survival. Since AOX is absent in mammalian cells, AOX is considered a viable therapeutic target for the design of specific fungicidal and anti-parasitic drugs. In this work, we have mutated conserved residues within the hydrophobic channel (R96, D100, R118, L122, L212, E215 and T219), which crystallography has indicated leads to the active site. Our data shows that all mutations result in a drastic reduction in Vmax and catalytic efficiency whilst some also affected the Km for quinol and oxygen. The extent to which mutation effects inhibitor sensitivity was also investigated, with mutation of R118 and T219 leading to a complete loss of inhibitor potency. However, only a slight reduction in IC50 values was observed when R96 was mutated, implying that this residue is less important in inhibitor binding. In silico modelling has been used to provide insight into the reason for such changes, which we suggest is due to disruptions in the proton transfer network, resulting in a reduction in overall reaction kinetics. We discuss our results in terms of the structural features of the ubiquinol binding site and consider the implications of such findings on the nature of the catalytic cycle. Significance The alternative oxidase is a ubiquinol oxidoreductase enzyme that catalyses the oxidation of ubiquinol and the reduction of oxygen to water. It is widely distributed amongst the plant, fungal and parasitic kingdoms and plays a central role in metabolism through facilitating the turnover of the TCA cycle whilst reducing ROS production.
Article
Electron transfer from all respiratory chain dehydrogenases of the electron transport chain (ETC) converges at the level of the quinone (Q) pool. The Q redox state is thus a function of electron input (reduction) and output (oxidation) and closely reflects the mitochondrial respiratory state. Disruption of electron flux at the level of the cytochrome bc1 complex (cIII) or cytochrome c oxidase (cIV) shifts the Q redox poise to a more reduced state which is generally sensed as respiratory stress. To cope with respiratory stress, many species, but not insects and vertebrates, express alternative oxidase (AOX) which acts as an electron sink for reduced Q and by-passes cIII and cIV. Here, we used Ciona intestinalis AOX xenotopically expressed in mouse mitochondria to study how respiratory states impact the Q poise and how AOX may be used to restore respiration. Particularly interesting is our finding that electron input through succinate dehydrogenase (cII), but not NADH:ubiquinone oxidoreductase (cI), reduces the Q pool almost entirely (>90%) irrespective of the respiratory state. AOX enhances the forward electron transport (FET) from cII thereby decreasing reverse electron transport (RET) and ROS specifically when non-phosphorylating. AOX is not engaged with cI substrates, however, unless a respiratory inhibitor is added. This sheds new light on Q poise signaling, the biological role of cII which enigmatically is the only ETC complex absent from respiratory supercomplexes but yet participates in the tricarboxylic acid (TCA) cycle. Finally, we delineate potential risks and benefits arising from therapeutic AOX transfer.
Article
The alternative oxidase (AOX) is a monotopic diiron carboxylate protein which catalyzes the four-electron reduction of dioxygen to water by ubiquinol. Although we have recently determined the crystal structure of Trypanosoma brucei AOX (TAO) in the presence and absence of ascofuranone (AF) derivatives (which are potent mixed type inhibitors) the mechanism by which ubiquinol and dioxygen binds to TAO remain inconclusive. In this article, ferulenol was identified as the first competitive inhibitor of AOX which has been used to probe the binding of ubiquinol. Surface plasmon resonance reveals that AF is a quasi-irreversible inhibitor of TAO whilst ferulenol binding is completely reversible. The structure of the TAO-ferulenol complex, determined at 2.7 Å, provided insights into ubiquinol binding and has also identified a potential dioxygen molecule bound in a side-on conformation to the diiron center for the first time.