Ecological Significance of Mitochondrial Toxicants - 2017 - Toxicology

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Toxicology 391 (2017) 64–74

Contents lists available at ScienceDirect

Toxicology
journal homepage: www.elsevier.com/locate/toxicol

Ecological significance of mitochondrial toxicants MARK


Nishad Jayasundara
University of Maine, Orono, ME, 04469, United States

A R T I C L E I N F O A B S T R A C T

Keywords: Mitochondrial dysfunction with exposure to anthropogenic contaminants is a critical concern in biomedical and
Mitochondria ecological health. Importantly, understanding the role of mitotoxicants in modulating organismal fitness and
Pollutants survival, particularly in the context of species’ resilience to shifts in abiotic factors with climate change, is
Climate change emerging as a key consideration. To date, only a few studies have focused on effects of mitotoxicants on or-
Stress responses
ganismal responses to additional abiotic stressors (e.g., shifts in temperature, levels of oxygen, and salinity);
Ecotoxicology
however, these serve to demonstrate that chemical exposure could alter stress responses. Here a theoretical
framework is presented to integrate mitochondrial dysfunction into organismal response to additional stressors,
highlighting the importance of sustaining the capacity to conserve or regenerate a healthy population of mi-
tochondria, i.e., maintaining mitochondrial plasticity, in a multi-stressor environment. The complexity of mi-
tochondria-associated processes and impact of mitotoxicants in the context of shifts in fate and transport of
anthropogenic chemicals with climate change are also considered. Considering the crucial role of mitochondria
in multi-stressor environments and their vulnerability to physical and chemical stressors, this review proposes
the use of mitochondrial function as a physiological marker of species resilience to environmental change.
Finally, impacts of contaminants on mitochondrial health of wildlife are important in their own right, but the
potential use of these studies, especially mitochondrial DNA analyses, to inform long-term impacts of chemicals
on mitochondria in a human health context is also discussed.

1. Introduction mitochondrial function and integrity with exposure to multiple abiotic


stressors, is critical when exposed to environmental perturbations.
Mitochondria are key cellular organelles involved in a number of The increasing recognition of mitochondrial etiology of a range of
biological processes and mitochondrial health is critical to organismal human health disorders has led to an emergence of studies focusing on
survival and fitness. In addition to serving as the energy powerhouse, mitochondrial toxicants (Meyer et al., 2013) and remain a critical area
mitochondria are associated with apoptotic signaling including initia- of research as discussed in detail in this issue. In addition to research on
tion and amplification of apoptosis (Raimundo et al., 2012; Tann et al., human health implications of mitotoxicants, a number of studies de-
2011), calcium signaling and calcium homeostasis (Clapham, 2007), monstrate that environmental contaminants have significant effects on
copper and iron homeostasis (Askwith and Kaplan, 1998), non-shi- mitochondrial structure and function in organisms across taxa. For
vering thermogenesis (Nedergaard et al., 2001), protein activating iron- example, studies in amphibians (Stabenau et al., 2008; Swann et al.,
sulfur cluster assembly (Gakh et al., 2016), and synthesis of macro- 1996), aquatic and terrestrial invertebrates (de Quadros et al., 2016;
molecular structures (Ahn and Metallo, 2015), ion exchange (Shoshan- Xie et al., 2006; Mommaerts et al., 2011; Cutkomp et al., 1971), and
Barmatz et al., 2015), and metabolite transport (Palmieri and Pierri, fish from the tropics to the polar regions (Cutkomp et al., 1971;
2010). Mitochondria also communicate with the nucleus and cytoplasm Dussauze et al., 2014; Tissier et al., 2015; Padmini et al., 2009; Knecht
via reactive oxygen and nitric oxide species signaling (ROS and NOS, et al., 2013; Cambier et al., 2009; Smolders et al., 2003; Pandey et al.,
respectively) (Schulz et al., 2014; Storz, 2006) and cell cycle arrest 2008), show that exposure to heavy metals, pharmaceuticals, pesti-
signaling (Koczor et al., 2009). Considering the complex and important cides, complex hydrocarbon mixtures, nanomaterials, UV radiation and
roles of these organelles, maintaining a healthy population of mi- various other organic and inorganic contaminants can disrupt mi-
tochondria is crucial for organismal survival and fitness. In particular, tochondrial structure and function. Examples of pollution effects on
mitochondrial plasticity, the capacity to maintain optimum plant mitochondria also exist (Schulte Hostede et al., 1987). However,

Abbreviations: mtDNA, mitochondrial DNA; NOS, nitric oxide species; OXPHOS, oxidative phosphorylation; PAH, polycyclic aromatic hydrocarbons; PCB, polychlorinated biphenyls;
POP, persistent organic pollutant; ROS, reactive oxygen species; TCDD, 2,3,7,8-tetrachlorodibenzo-p-dioxin
E-mail address: nishad.jayasundra@maine.edu.

http://dx.doi.org/10.1016/j.tox.2017.07.015
Received 21 April 2017; Received in revised form 19 July 2017; Accepted 24 July 2017
Available online 01 August 2017
0300-483X/ © 2017 Elsevier B.V. All rights reserved.
N. Jayasundara Toxicology 391 (2017) 64–74

effects of anthropogenic chemicals on mitochondrial health of wildlife chemical environment of the cell. In contrast to nuclear DNA, mtDNA is
in the context of organismal responses to multiple abiotic stressors have not protected by histone proteins and is thought to be damaged at a
received less attention. This remains a key area of research considering higher rate by ROS that are routinely generated in the mitochondria
rapid environmental change due to anthropogenic activities. In parti- (Yakes et al., 1997) (e.g., loss of electrons during electron transport
cular, sub-lethal concentrations of mitotoxicants could affect orga- chain can lead to transformation of some oxygen (∼0.1–0.4%) into
nismal aerobic performance and perturb food-web dynamics, re- superoxide anion radicals (Hermes-Lima, 2004)). Furthermore, com-
productive outcomes, and response to other abiotic stressors, leading to pared to nuclear DNA, mtDNA is susceptible to higher mutation rates
significant ecosystem level impacts (Seiler, 2002; Di Giulio and Clark, (∼10–20 times) and the rates of mutation accumulation depends on the
2015). Given the importance of mitochondrial plasticity in organismal region of mtDNA that is being tested (Neiman and Taylor, 2009) and
fitness and survival, especially when exposure to multiple stressors, may also depend on the type of mutagen (Valente et al., 2016). Ad-
understanding the ecological impact of mitochondrial toxicants remain ditionally, protein products of altered mtDNA and mitochondrial
a crucial consideration and warrants further analysis on impacts of structural damage can lead to mitochondrial dysfunction resulting in
mitochondrial dysfunction at the organismal, population, and eco- increased production of ROS (e.g., H2O2) (∼10× more than normal
system level. As discussed later, these studies may also provide critical mitochondria), and in some conditions may potentially lead to systemic
insights into effects of mitotoxicants on human health. Accordingly, this mitochondrial oxidative damage and dysfunction (Yakes et al., 1997;
review considers the implications of mitochondrial toxicants with a Grivennikova et al., 2010). It is noteworthy that the extent of mi-
specific focus on their effects on organismal response to shifts in their tochondrial perturbation is influenced by the tissue type, since tissues
abiotic environment (Løkke et al., 2013) and briefly discusses the po- differ in their bioenergetic capacity and ability to remove ROS and
tential implications of these studies on human mitochondrial health repair damaged mtDNA (Szczesny et al., 2010).
research. Despite decades of research, large gaps in knowledge exist in our
understanding of the complex nature of mitochondria, including their
2. Complexity of mitochondria origin, structure, function, and repair and removal processes (e.g., mi-
tophagy—selective removal of damaged organelles (Kim and Lemasters,
Mitochondria contain their own genome and each organelle may 2011)). Nonetheless, it is clear that these vital organelles are vulnerable
consist of several copies of mitochondrial DNA (mtDNA). mtDNA is to changes in the surrounding physical and chemical environment,
transcribed at a high rate, especially in highly energetically active tis- implying that mitotoxicants are an important consideration in assessing
sues such as hearts (Mercer et al., 2011), and encodes for ∼13 proteins organismal fitness and survival in their natural environment.
that contribute to the enzyme complexes of the oxidative phosphor-
ylation (OXPHOS) system (Friedman and Nunnari, 2014). Since some 3. Mitochondrial toxicants in the environment
proteins of the electron transport chain complexes are encoded in the
nuclear genome, a tight regulation of mitochondrial function by the Mitochondria are vulnerable to certain anthropogenic chemicals
nucleus and cytoplasm is required to maintain optimal mitochondrial due in part to the membranous high lipid content that facilitates ac-
function (Rand et al., 2004). cumulation of lipophilic compounds, as well as the negative charge of
Additionally, many gene products (∼1000–1500 proteins) (Calvo the matrix that facilitates accumulation of cationic metals (Meyer et al.,
and Mootha, 2010) that are encoded in the nuclear genome are im- 2013). Various classes of anthropogenic contaminants have emerged as
ported into the mitochondria to perform various functions. Mitochon- potent mitochondrial toxicants and Meyer et al. (2013) provide an
drial interactions and biogenesis (growth and division of pre-existing excellent review of these chemicals and their mechanisms of toxicity.
mitochondria) are highly tissue dependent (Jornayvaz and Shulman, These compounds include persistent organic pollutants (POP) (e.g.,
2010) and this specificity is reflected in mitochondrial morphology polycyclic aromatic hydrocarbons (PAH), 2,3,7,8-tetrachlorodibenzo-p-
(spherical vs. elongated) and organization (discrete organelles vs. dioxin (TCDD), and bisphenol A (BPA)) and various metals (e.g., Cd, Cu,
highly interconnected dynamic networks), as well as the total mi- Pb, Hg, As, Methyl-Hg, Mn, Se, and Zn) as well as chemicals designed to
tochondrial amount, content, and transcriptome of these organelles target mitochondrial complexes (e.g., rotenone). Furthermore, a com-
(Okamoto and Shaw, 2005; Vafai and Mootha, 2012; Benard et al., prehensive analysis of Comparative Toxicogenomics Database that
2007). Ontogenetic shifts and environmental factors can also affect contains manually curated information about chemical–gene/protein
mitochondrial morphology, content, and copy number (Jendrach et al., interactions, chemical–disease and gene–disease relationships
2008). Mitochondrial biogenesis is a highly coordinated process with (Mattingly et al., 2006), indicated that over 6000 chemicals from many
mtDNA replication, fusion and fission, and nuclear-cytoplasmic sig- classes of compounds affect mitochondrial oxidative phosphorylation
naling and is heavily influenced by environmental stressors. Precise (Supplemental Fig. S1).
regulation of biogenesis, degradation, fission and fusion, as well as Certain mitotoxicants could present a higher risk for wildlife com-
maintenance of nuclear-cytoplasmic signaling are critical for optimum pared to human health due to elevated bioavailability in the environ-
mitochondrial function (Meyer et al., 2013; Green and Van Houten, ment, especially in aquatic ecosystems. Notably, selenium toxicity has
2011; Piantadosi and Suliman, 2012; Youle and Van Der Bliek, 2012; gained recent attention as a coal combustion waste product released
Blajszczak and Bonini, 2017; Roubicek and de Souza Pinto, 2017; Chan, into the aquatic environment and is shown to affect fish mitochondrial
2017; Meyer et al., 2017). membrane potential leading to apoptosis (Selvaraj et al., 2013). BPA
The aforementioned mitochondria-associated cellular processes, and phthalates, commonly known for their endocrine disrupting prop-
including OXPHOS, are highly sensitive to changes in the chemical and erties, are ubiquitous environmental contaminants, especially in the
physical properties of the cellular milieu. In addition, anthropogenic aquatic environment (Kang et al., 2007). Mounting evidence suggests
chemicals and other stressors could impact macromolecular mi- that these chemicals affect mitochondrial membrane potential, mtDNA
tochondrial structures via molecular interference or mechanical da- copy number, mitochondrial mass, mtDNA mutations, and ATP content
mage. For example, the ion impermeability of the inner mitochondrial (Lin et al., 2013; Kaur et al., 2014). Furthermore, interactive effects of
membrane is critical for the generation of the electrochemical gradient chemical mixtures and other environmental factors are a significant
for oxidative phosphorylation. However, it is a likely target of xeno- concern in an ecological context. For instance, exposure to UV radiation
biotic elements (Meyer et al., 2013; Smith et al., 2003) and physical (de Quadros et al., 2016) induces mitochondrial toxicity and is likely to
stressors (as reviewed by Somero et al., 2017) as they can damage be further exacerbated by synergistic effects with other mitoxicants
membrane structure and/or alter membrane potential. such as PAHs (de Quadros et al., 2016).
mtDNA is also highly susceptible to changes in physical and In addition to chemicals released from various industrial and

65
N. Jayasundara Toxicology 391 (2017) 64–74

agricultural activities, pharmaceuticals are an environmentally im- mitochondrial dysfunction with anthropogenic chemicals can be at-
portant category of mitotoxicants. Evidence of drug-induced mi- tributed to five processes: (i) direct inhibition of mitochondrial bio-
tochondrial dysfunction (e.g., effects on electron transport chain, mi- chemical processes; (ii) structural perturbation (e.g., membrane per-
tochondrial transport pathways, and mtDNA replication etc.) is rapidly turbation and mitochondrial DNA damage and mutation) from
accumulating (Begriche et al., 2011) and specific examples are re- exposure to parent compounds and metabolites (e.g., free radicals); (iii)
viewed by Meyer et al. (2013). Certain bactericidal antibiotics are of direct effects on nuclear-cytoplasmic processes involved in regulation of
particular concern, as they can cause mitochondrial dysfunction and mitochondrial function (e.g., hormonal activity, oxygen availability,
ROS overproduction, potentially via disruption of mitochondrial pro- and ion regulation etc.) and signaling (e.g., ROS, apoptotic, and Ca2+
tein synthesis—given the common evolutionary origins of mitochondria signaling); (iv) compensatory mitochondrial changes to meet the in-
and bacteria (Schägger, 2002), antibiotics that are designed to target creased energy demand following a chemical insult to induce an ade-
bacterial ribosomes could also target mitochondrial ribosomes (Meyer quate stress response; and (v) compensatory mitochondrial changes
et al., 2013). Antibiotics, other pharmaceuticals and their metabolites with altered organismal energy metabolic phenotype due to shifts in
are often detected in the environment, especially in aquatic habitats, substrate (e.g., fatty acids) availability as a result of pollutant effects on
sometimes at concentrations that are orders of magnitude higher than predator-prey interactions and food availability. A combination of these
prescribed for human consumption (Bernhardt et al., 2017). Pharma- direct and indirect effects of exposure to an individual chemical or a
ceuticals, especially the lipophilic and persistent ones, have the po- mixture can affect mitochondrial function in a life-history dependent
tential to accumulate in the mitochondria and biomagnify in non-target manner. Of particular importance are the potential for effects of ex-
aquatic species. In addition, as reviewed previously (Seiler, 2002) cer- posure to mitotoxicants during development that may have long-term
tain pharmaceuticals, potentially including compounds that target mi- consequences across generations (Kozal et al., 2017; Stapleton et al.,
tochondria, that have negligible biological effects at prescribed con- 2015).
centrations in human health contexts may have unintended chemical Regardless of the mechanism, mitochondrial impairment via the
and physical interactions in the environment. They may potentially aforementioned processes is likely to have a significant impact on or-
lead to organismal level consequences. ganismal fitness and survival. Mitochondrial function and integrity are
Risk of mitoxicity can also increase with shifts in fate and transport common targets of both chemical (Meyer et al., 2013) and physical
of chemicals in ecosystems due to natural and anthropogenic activities. (Somero et al., 2017) stressors. For example, as reviewed by Somero
For example, toxic and trace metal exposures can vary rapidly as a et al. (2017), temperature stress alters mitochondrial reaction rates,
result of predicted shifts in fate and transport of chemicals in the abiotic affects mitochondrial membrane fluidity, increases ROS production,
and biotic environment with climate change (Gambrell, 1994). Several induces mtDNA mutations, and alters other macromolecular structures
reviews have described mechanisms and predicted consequences of and functions that are important for peak mitochondrial performance.
altered fate and transport of chemicals due to changes in temperature, Consequently, optimum mitochondrial capacity is critical in inducing
salinity, and pH as well changes in frequency of weather events such as acute, acclimatory, and adaptive responses to enable organisms mini-
precipitation and floods, cloud cover, and forest fires (Noyes et al., mize the impact of climate change. Here these effects are discussed with
2009; Hooper et al., 2013; Noyes and Lema, 2015). For instance, metal a focus on the impact on aquatic organisms, since water bodies often
distribution, speciation, and biotransformation will be affected with serve as ‘sinks’ for anthropogenic waste products and are exceedingly
shifts in geophysical and chemical properties (e.g., temperature), and vulnerable to impacts of climate change. Aquatic organisms are not
bioavailability and accumulation of these chemicals is likely to change only exposed to pollutants due to agricultural runoff, industrial ef-
as a result (Noyes and Lema, 2015; Anawar, 2013). An indirect con- fluents, and other processes, they can experience daily and seasonal
sequence of warming is elevated release and bioavailability of seques- fluctuations in abiotic factors such as temperature, salinity, oxygen, and
tered mitochondrial toxicants such as polychlorinated biphenyls (PCBs) pH (Harley et al., 2006; Schulte, 2007).
and methyl mercury with melting sea ice and snow, especially in the Several recent studies have considered the role of mitochondrial
polar regions (Noyes et al., 2009). Furthermore, climate-induced shifts toxicants in multi-stressor experiments. Ivanina et al. (2012) demon-
in behavior and physiology in polar organisms results in altered bio- strated that exposure to Cd and acute change in temperature affects
logical toxicokinetics within an organism as well as across trophic mitochondrial capacity to maintain function with changes in environ-
cascades with rapid environmental change (Noyes et al., 2009) (ex- mental oxygen concentrations in oysters (Crassostrea virginica). Studies
amples are discussed later). Overall, it is evident that changes in abiotic in oysters exposed to Cd also indicate that mitochondrial vulnerability
factors have significant effects on fate and transport of chemicals in- to chemical insults with thermal stress may depend on the latitudinal
cluding mitotoxicants in the abiotic and biotic environment remains an positioning (i.e., prior thermal history and thermal adaption) of a po-
important, yet highly complex and challenging consideration when pulation (Cherkasov et al., 2010). Furthermore, Cd and temperature co-
determining ecological relevance of these chemicals. exposure studies showed higher sensitivity of mitochondrial enzymes to
the inhibitory effects of Cd, a decrease in activation energy of mi-
4. Consequences of mitotoxicant exposure on organismal tochondrial enzymes (Ivanina et al., 2008), and increased degradation
response to effects of climate change of oxidatively damaged protein as a protective mechanism against Cd
mitotoxicity (Sanni et al., 2008).
As highlighted previously, a number of studies demonstrate sig- Similar studies in rainbow trout (Oncorhynchus mykiss) indicated
nificant effects of anthropogenic chemicals on mitochondrial function that electron transport chain, tricarboxylic acid cycle, and proton leak
and structure, especially in aquatic organisms. A key area of emerging are common targets of metals such as Cd and Cu and exposure to these
research is beginning to focus on effects of mitochondrial toxicants on chemicals affect responses to temperature changes and hypoxia-reox-
organismal response to additional abiotic stressors. Here, the existing ygenation. In these fish, warm acclimation induced electron transport
research examining effects of mitotoxicants on organismal response to chain shifts to compensate for acute thermal stress, but resulted in
multiple stressors are presented with a discussion on five broad me- mitochondria more sensitized to Cu and Cd toxicity. However, mi-
chanisms potentially underlying ecologically relevant effects of mito- tochondrial hormesis—mild mitochondrial stress leaving the mi-
toxicants. tochondria less susceptible to subsequent perturbations—was detected
Previous studies show that, while certain compounds such as rote- with low dose Cd exposure, such that the effects of temperature and
none directly (and on some cases are designed to) target mitochondria, hypoxia-reoxygenation on proton leak and complex 1 were rescued
other chemicals affect mitochondrial function via various indirect me- (Sappal et al., 2015a, 2016, 2015b, 2014; Onukwufor et al., 2016a,
chanisms. Therefore, based on previous research, ecologically relevant 2016b). In addition to metals, on going studies suggest that

66
N. Jayasundara Toxicology 391 (2017) 64–74

Fig. 1. Conceptual figure demonstrating mitochon-


dria as a target of chemical and physical stressors,
consequences of exposure to these stressors and the
use of mitochondrial health index (MHI) as a marker
of optimum mitochondrial function.

developmental and adult exposure to PAHs can affect mitochondrial and Schulte, 2004). Furthermore, as discussed by Noyes and Lema
response to temperature response later in life and across several gen- (2015) exposure to metals and organochlorine pesticides can alter
erations in zebrafish (Danio rerio) (Kozal et al., 2017) and killifish metabolic rates in fish, crustaceans, and mollusks. It is possible that
(Fundulus heteroclitus) (Jayasundara et al., 2017). Collectively, these these observed changes are independent of mitochondrial toxicity and
studies provide evidence for a direct link between mitochondrial toxi- are due to altered metabolic demand and signaling (Casals-Casas and
city and thermal, hypoxia, and other stress tolerance and limits. Im- Desvergne, 2011; Grun and Blumberg, 2006) or substrate (e.g., fatty
portantly, it is evident that mitochondrial effects of chemical and acids, glucose) composition shifts as a result of behavioral and phy-
physical stressors were dependent on magnitude and duration of the siological changes with chemical exposure (De Smet and Blust, 2001;
chemical exposure and the history of exposure and adaptation to other Fabbri et al., 2003). For example, as discussed by Sokolova et al.
stressors. (2012), elevated basal metabolic function observed in aquatic in-
vertebrates with metal exposure is thought to result from increased ATP
demand to maintain cellular integrity. In contrast, higher levels of ex-
5. Potential mechanisms that mediate ecological impact of
posure to metals are thought to reduce aerobic scope via direct effects
mitotoxicants
on mitochondrial function. Overall, it is evident that chemical exposure
can alter organismal energy balance that may or may not depend on
Based on aforementioned studies and previous studies on orga-
direct mitochondrial toxicity of pollutants. Nonetheless, the consequent
nismal response to abiotic stressors (Somero et al., 2017; Schulte, 2007;
shifts in ATP demand and synthesis may affect mitochondrial function
Ivanina and Sokolova, 2016; Haider et al., 2016), three key con-
and the capacity to meet this demand is considered critical in ecological
siderations emerge as likely mediators of mitochondrial toxicity in a
success across eukaryotic taxa (Sokolova et al., 2012).
multi-stressor environment. These, addressed in more detail below, are
Shifts in ATP demand and synthesis with chemical exposure are
(i) bioenergetic imbalance; (ii) oxidative stress; and (iii) common mo-
likely to reduce energy allocation for somatic maintenance, cellular
lecular pathways (e.g. apoptotic response) that are targets of mitotox-
stress responses, growth and reproduction, and aerobic performance,
icants and other abiotic stressors (Fig. 1).
and the magnitude of this effect depends on a number of factors
(Sokolova et al., 2012). Perturbed ATP allocation is further exacerbated
5.1. Bioenergetic imbalance by direct and indirect effects on bioenergetics and mitochondrial
function and integrity via shifts in physical abiotic factors such as
A direct consequence of mitochondrial dysfunction is altered energy temperature, pH, oxygen, and salinity that are anticipated with climate
homeostasis (Sokolova et al., 2012, 2011) and this is a critical para- change (Somero et al., 2017). This energy imbalance with exposure to
meter in determining ecological significance of mitotoxicants. Previous multiple abiotic stressors may have significant population level fitness
studies have demonstrated whole organismal bioenergetic effects of consequences and ecosystem impacts (Calow and Forbes, 1998).
chemical exposures across taxa. For instance, developmental and acute To this end, several useful models have incorporated altered orga-
exposure to individual PAH compounds and complex PAH mixtures nismal bioenergetics to evaluate ecosystem level impacts of anthro-
affects routine (or standard) and maximum metabolic rates, leading to pogenic stressors, particularly focusing on thermal stress. As summar-
reduced aerobic scope in adult and juvenile teleosts (Davoodi and ized by Noyes and Lema (2015), a number of studies indicate that
Claireaux, 2007; Gerger et al., 2015; Gerger and Weber, 2015; Mager chemical exposure can alter thermal limits in ectotherms, though the
et al., 2014). Ongoing studies also show that dietary exposure to PAHs underlying mechanisms are not always clear. As described by the
has transgenerational effects on fish metabolic rates (Kozal et al., oxygen- and capacity-limited thermal tolerance framework, the capa-
2017). PCB exposure also resulted in reduced standard metabolic rates city to maintain an adequate oxygen supply to meet the increased
in European eel (Anguilla anguilla) (van Ginneken et al., 2009) and al- oxygen demand at different temperatures is thought to define thermal
tered energy metabolic enzymes in killifish (F. heteroclitus) (Kraemer

67
N. Jayasundara Toxicology 391 (2017) 64–74

limits in aquatic ectotherms. Accordingly, further studies suggest that are ROS inducers, eliciting oxidative stress responses. In Antarctic
aerobic metabolism at a given temperature and the thermal acclimatory scallop (Adamussium colbecki), exposure to temperature, pH, and Cd
capacity of aerobic metabolic processes are directly associated with stress showed tissue specific oxidative stress response and damage de-
thermal limits and tolerance in ectotherms (Pörtner and Farrell, 2008; pending on the magnitude of the stressors (Benedetti et al., 2016).
Jayasundara and Somero, 2013). Studies in mollusks indicate that Studies in oysters (Crassostrea angulate) showed that As effects on an-
thermal tolerance limits are indeed reduced by decreased aerobic scope tioxidant capacity are increased at higher salinities (Moreira et al.,
with metal exposure in aquatic invertebrates (Lannig et al., 2008; 2016). Differential effects on oxidative stress response markers were
Bagwe et al., 2015; Sokolova and Lannig, 2008). Notably, mitochondria detected in coral (Porites astreoides) larvae with exposure to pesticides
play an important role in defining these limits, where, in teleost fish, and thermal stress (Ross et al., 2015). Falfushynska et al. (2015) de-
cardiac bioenergetics and mitochondria are linked with thermal monstrated that antioxidant response pathways in freshwater mussels
boundaries and optimum performance (Jayasundara and Somero, 2013; (Unio tumidus) are overwhelmed by combined exposure to temperature
Iftikar and Hickey, 2013). stress, organics pollutants, and ZnO nanoparticles and suggested that
Noyes and Lema (2015) present a synthesis of incorporating pollu- toxicity of ZnO particles was modulated by the presence of other
tant effects on organismal energetics and temperature tolerance, by stressors. In European bullhead (Cottus gobio), a teleost fish, oxidative
focusing on thermal performance curves (TPC) (Pörtner and Farrell, stress responses were tissue and stressor specific and were influenced by
2008; Huey et al., 2012)—TPC presents a mechanistic framework to elevated temperature and Cd (Dorts et al., 2014).
examine the temperature vs fitness (e.g., aerobic scope, reproductive As exemplified in these studies, evaluating ROS levels, oxidative
capacity, swimming performance) relationship. TPC of an ectothermic stress responses, and oxidative damage remains important, but is likely
organism characterizes the upper and lower thermal limits and the to be challenging. For example, mitochondria are both a source and
temperature at which an organism maintains optimal performance. target of ROS. Thus, changes in oxidative stress response and oxidative
Extending this analysis beyond thermal stress, Sokolova et al. (2012) damage could be a result of direct effects of pollutants on mitochondria
present an excellent analysis integrating organismal level bioenergetic leading to ROS generation, but also due to ROS generated via bio-
effects of multiple stressors with long-term, population-level con- transformation of xenobiotic elements, which may in turn lead to mi-
sequences accounting for magnitude and duration of stress exposure. In tochondrial damage (Banerjee et al., 2016). Another major challenge is
the context of mitotoxicants, this approach provides a framework to distinguishing whether ROS production is an effect of stress exposure or
incorporate mitochondrial dysfunction in assessment of population a secondary stress response including a cell death response110. In ad-
level impact of anthropogenic stressors (Fig. 2) and is discussed later. dition, ROS signaling is crucial in physiological responses to regulate
cellular homeostasis. Thus, changes in ROS levels and antioxidant re-
sponses may reflect adaptive changes following exposure to con-
5.2. Oxidative stress
taminants (Shadel and Horvath, 2015). These aspects underscore the
complexity in uncovering specific effects of chemical and physical
In addition to bioenergetic constraints, ROS production, oxidative
stressors on ROS production and oxidative stress responses. None-
stress response and oxidative damage derived from mitochondrial
theless, ROS-induced damage, especially mtDNA and membrane da-
dysfunction may play an important role organismal fitness in multi-
mage, can be important in assessing the impact of mitochondrial tox-
stressor environments (Abele and Puntarulo, 2004; Lesser, 2006). Di-
icants on organismal response to multi-stressor environments (Lesser,
rect inhibition of mitochondrial function (e.g., rotenone inhibition of
2006).
complex I, which is a major source of intercellular ROS) and indirect
effects on mitochondria as well as pollutant-specific biotransformation
and detoxification processes can lead to increased ROS production. A 5.3. Common targets of cellular stress response and cell death
number of studies show that both chemical (e.g., metals and POPs) and
physical (e.g., temperature and UV radiation) environmental stressors There are number of common mitochondria-associated pathways

Fig. 2. Impacts of mitochondrial toxicants on aerobic activity of organisms and its relationship with organismal fitness. (a) A theoretical framework for integrating bioenergetic analysis
with thermal tolerance based on thermal performance curves [adapted from Huey (1982) and Pörtner and Farrell (2008)]. Solid blue line depicts the natural thermal range of an
ectotherm defined based on organismal aerobic fitness. Solid green lines illustrate effects of thermal acclimation shifting thermal range towards cold or warm tolerance. Dotted blue line
represents narrowed range for thermal tolerance with exposure to mitotoxicants due to direct and indirect effects on energy metabolism as discussed in the text. This suppression can
affect cold or warm tolerance (solid arrow 1 and 3 respectively), thermal optimum (solid arrow 2) or the overall thermal tolerance (dotted blue line) (b) Mitochondrial function may serve
as a marker of organismal fitness in multi-stressor environment. As discussed in ovo and ex vivo functional studies (e.g., XFe24 Extracellular Flux Analyzer assays) will be useful in these
analyses and utilized for developing a mitochondrial health index (MHI). MHI may vary depending on the duration and the magnitude of the stressor (Δ stressor) e.g., temperature, pH,
salinity (solid blue line). Exposure to mitotoxicants is expected to shift MHI affecting the response to Δ stressor (dotted blue line). The direction of this suppression is toxicant dependent
and is depicted by solid arrows. (c) Mitochondrial plasticity can be measured as the capacity to maintain MHI in a multi-stressor environment. The duration and the magnitude of a given
stressor (Δ stressor) are likely to determine mitochondrial plasticity. Identification of the flexion point of this relationship, where mitochondria fails to maintain plasticity can reveal
vulnerability of an organism to environmental stressors. The flexion point is likely to be right shifted with exposure to mitotoxicants, except for potentially in mitohormesis scenarios
(dotted orange line). (For interpretation of the references to colour in this figure legend, the reader is referred to the web version of this article.)

68
N. Jayasundara Toxicology 391 (2017) 64–74

(e.g., induction of apoptosis) that are involved in xenobiotic response While mitochondrial toxicity is likely an important concern for
and cellular response to physical stressors. For example, an important health and fitness of mammals and birds, they may pose a greater risk
consequence of mitochondrial ROS is apoptotic and necrotic cell death. for organisms that have evolved specialized mitochondrial functions to
Mitochondria-generated ROS lead to release of cytochrome C and other inhabit challenging habitats. For example, organisms have evolved
pro-apoptotic proteins that elicit cellular apoptotic response (e.g., Ca2+ various energetic adaptations to inhabit higher altitudes, polar-regions,
dependent mitochondrial permeability transition (MPT) and caspase and cold ocean depths. With changes in abiotic environmental factors
activation) (Ott et al., 2007). Induction of apoptotic pathways is also a (e.g., temperature), mitochondrial dysfunction could manifest via (i)
key element of abiotic stress responses including temperature and hy- increased mitotoxicant potency with altered biological and environ-
poxia/reoxygenation response (Kültz, 2005). Acute and chronic ex- mental toxicokinetics and (ii) increased mitochondrial demand to
posure to warm temperature leads to apoptotic gene and protein ex- maintain homeostasis during dramatic seasonal or thermocline fluc-
pression level changes in fish hearts and gills (Logan and Buckley, 2015; tuations. For example, higher levels of organohalogen and PCBs, known
Jayasundara et al., 2015a). These responses include protein expression mitotoxicants, are detected in polar bears (Ursus maritimus) and were
changes in voltage-dependent anion channel (VDAC) proteins that are particularly prominent in females during their reproductive and cub
involved in apoptotic signaling, as well as global regulation and sup- rearing period. Elevated contaminant levels appear to be associated
pression of mitochondria (Jayasundara et al., 2015a). Hypoxic and with shifts in diet, mating, feeding, and birth and survival rates that are
anoxic conditions also activate similar cascades of apoptotic pathways, linked to direct impacts of climate change (e.g., levels of sea ice) (Noyes
including regulators of VDAC via induction of ROS and NOS, cytokines, et al., 2009). The capacity to maintain bioenergetic homeostasis for
kinases, and increased levels of NADH (Lemasters and Holmuhamedov, periods of fasting is critical to the survival of adult and juvenile polar
2006). Importantly, VDAC forms a multimeric complex with adenine bears and may lead females to abandon reproductive efforts (Derocher
nucleotide translocase and cyclophilin D that forms the MPT pore, that and Stirling, 1996; Robbins et al., 2012).
is a target of various pharmaceuticals (e.g., doxorubicin and arsenic Studies show that extreme cold-tolerant organisms, such as polar
trioxide), as well as ROS that are generated from pollutant exposure and bears, have evolved specific mitochondrial adaptations to arctic habi-
metabolism (Oliveira and Wallace, 2006; Andreau et al., 2012). The tats (Welch et al., 2014). Thus, even subtle changes in mitochondrial
precise composition of the MPT pore complex and the role of its com- function may have significant energetic consequences, especially
ponents are yet to be fully resolved. However, the ROS generated from during hibernation. In addition to bioenergetics, mitochondria play an
chemical and physical factors are likely to affect the MPT pore complex important role in thermal regulation of polar and high-altitude organ-
leading to cell death and presents an example of a common mi- isms. Notably, heat production in hibernators as well as in mammalian
tochondrial target in a multi-stressor environment. neonates is dependent on non-shivering thermogenesis that occurs in
Besides ROS, interactions among environmental sensor molecules large deposits of brown adipose tissue (BAT) (Stephens et al., 2011).
may play an important role in mediating mitochondrial dysfunction in BAT has high mitochondrial content and activity, and they produce
multi-stressor environments. Notably, the crosstalk between the aryl heat via uncoupling of oxidative phosphorylation mediated by un-
hydrocarbon receptor (AHR) and hypoxia inducible factor-1α (HIF-1α), coupling protein 1 (UCP1) (Stephens et al., 2011). Perturbed mi-
and their common binding partner heat shock protein 90 are targets of tochondrial function may significantly alter BAT activity, thus ther-
exposure to xenobiotics, hypoxia stress and temperature changes mogenesis. In addition, organisms such as hummingbirds that maintain
(Gabriely et al., 2017; Ramsay and oreen Cantrell, 2015). Importantly, remarkably high metabolic rates (Suarez et al., 1991), flying bats (Shen
the recent discovery of the presence of the (AHR) in the inner mi- et al., 2010), and Antarctic killer whales (Foote et al., 2011), have
tochondrial membrane space suggests a unique role for this protein in evolved specialized mitochondria to maintain unique life-history stra-
mitochondrial regulation. AHR is well known for xenobiotic metabo- tegies or inhabit challenging environments are likely to be directly
lism of its agonists (e.g., TCDD and certain PAHs). Studies show that impacted by mitochondrial toxicants. Besides mammals, partially en-
exposure to AHR agonists have profound effects on mitochondrial dothermic (mesothermic), apex oceanic predators (e.g., bluefin tunas
membrane potential and retrograde signaling (Hwang et al., 2016a, and lamnid sharks) also maintain high metabolic rates and specialized
2016b; Tappenden et al., 2011) and that fish resistant to PAHs, an AHR cardiac Ca2+ homeostatic processes to maintain a highly aerobic life
agonist, have evolved specialized mitochondria (Du et al., 2016). style. Thus, optimal mitochondrial function is critical for their success
Considering the detection of AHR in the mitochondria and its interac- as apex predators (Jayasundara et al., 2013). Due to their positions on
tion with HSP90 and HIF-1α, it is possible that AHR is an important the trophic chain, these organisms contain high levels of mitotoxicants
mitochondria-associated common target in a multi stressor environ- such as methy-Hg and POPs (Hisamichi et al., 2010) and are likely to be
ment. susceptible to mitochondrial damage from exposure. These examples
Collectively, it is evident that anthropogenic chemicals have direct indicate the impact of mitotoxicants across taxa and food webs, po-
and indirect effects on mitochondrial function that may impair orga- tentially leading to significant ecological consequences.
nismal capacity to respond to additional abiotic stressors.
7. Mitochondria as markers of species’ resilience to global change
6. Mitotoxicity relevance to mammalian and avian species
Though characterizing the effects of mitotoxicants on organismal
Compared to lower vertebrates, especially in the aquatic environ- health is complex and warrants further studies, mitochondrial function
ment, impacts of mitotoxicants on mammalian and bird species are less may serve as a marker of impact of mitotoxicant exposure on orga-
well studied in ecological contexts. Nevertheless, rodent studies have nismal response to multiple stressors.
significantly contributed to the current understanding of mitochondrial Most studies have focused on in vitro model systems to quantify
toxicants. Considering the taxonomic similarity, it is likely that the function in isolated mitochondria or primary cell-culture. While these
response to mitotoxicants is shared among rodents, humans, and other approaches provide important insights under varying substrate and
mammals, potentially extending to bird species, and exposure may lead ADP levels, particularly into altered mitochondrial membrane poten-
to numerous health disorders (e.g., neurodegenerative diseases and tial, they are often cumbersome and low throughput, and the results are
metabolic syndromes). Furthermore, mitochondrial toxicity during ge- likely to be confounded by perturbed cellular conditions (Kuznetsov
station is an important consideration in mammals and several studies et al., 2008). Several recent studies have utilized XFe Extracellular Flux
indicate that mitochondrial function in the placenta is disrupted with Analyzer as a semi- and high-throughput method for mitochondrial
exposure to air pollutants, lead and endocrine disruptors, and may have function analysis in ovo (Stackley et al., 2011), ex vivo (Jayasundara
multi generational consequences (Brunst et al., 2015). et al., 2015b) and in vivo (whole larvae) (Raftery et al., 2017). In these

69
N. Jayasundara Toxicology 391 (2017) 64–74

the population level warrants careful control studies to define these


parameters. Another limitation is that altered substrate availability due
to an environmental perturbation may affect ATP linked respiration. In
addition, certain organisms are facultative anaerobes (e.g., blue mussel,
Mytilus edulis when under hypoxia) (Müller et al., 2012), limiting the
potential use of MHI as an appropriate indicator of organismal level
bioenergetic effects of exposure to an environmental perturbation.
Despite these challenges, this approach provides a comparative analysis
in multi-stressor contexts to assess mitochondrial health. Accordingly, a
theoretical framework based on TPCs (Fig. 3a) for utilizing MHI as an
indicator for mitochondrial function and plasticity, thus organismal
fitness, in the context of other stressors is presented in Fig. 2b and c. A
similar framework can be utilized for other physical stressors (e.g.,
salinity, pH, and hypoxia changes) that are likely to alter organismal
metabolic rates via direct effects on mitochondrial function and indirect
effects on metabolic signaling. Integrating mitochondrial plasticity
under multiple abiotic stressors will be crucial for assessing population
Fig. 3. Parameters of mitochondrial health index. Ex vivo and in ovo mitochondrial studies vulnerability and may further contribute to various quantitative bioe-
utilizing pharmacological inhibitors can indicate oxygen consumption associated with nergetic modeling approaches (e.g., dynamic energy budget modeling).
mitochondrial and non-mitochondrial respiration, ATP-linked respiration, reserve capa- One disadvantage of functional analyses, especially the methods
city and proton leak. Oligomycin inhibits complex V; FCCP (carbonyl cyanide 4-(tri-
that are based on oxygen consumption rate measurements, is that
fluoromethoxy) phenylhydrazone) is an uncoupler; Antimycin A and rotenone mixture
inhibits oxidative phosphorylation via inhibiting complex III and complex I respectively.
chronic toxicant exposure effects may be masked by compensatory re-
Sodium azide can be used as a substitute for mitochondrial inhibition. Figure adapted sponses by the organism. Integrating functional analyses with changes
from Jayasundara et al. (2015a,b). in protein, lipid and nucleic acid molecules associated with mitochon-
dria may better indicate altered cellular metabolic status and mi-
studies, different pharmacological agents were used to quantify total tochondrial dysfunction, though they may reflect secondary effects.
mitochondrial and non-mitochondrial respiration, maximal respiration, Furthermore, there may be masking effects in multi-stressor contexts.
and reserve mitochondrial capacity (Fig. 3). The ex vivo analysis on For example, studies in oysters indicated that hypoxia induced com-
whole heart, brain and liver (Jayasundara et al., 2015b) tissues of tel- pensatory responses in mitochondrial enzymes were suppressed by Cd
eosts is particularly useful in an ecological context, considering the exposure (Ivanina et al., 2011). Similarly, while markers of ROS pro-
tissue-specificity of mitochondrial function, structure and susceptibility duction and oxidative stress responses are typical indicators of mi-
to chemical exposure. The capability to examine extra cellular acid- tochondrial stress, they may not be useful in the context of assessing
ification rates (an index for glycolysis) in developing embryos and mitochondrial health in multi-stressor contexts, since both chemical
whole tissues is an added advantage of this method and can be a useful and physical stressors are known to disrupt the balance in ROS gen-
metric in assessing environmental conditions that trigger anaerobic eration and removal. For example, Cd exposure masked the tempera-
metabolism due to or independent of mitochondrial impairment. ture effect on mitochondrial aconitase, a marker of oxidative stress in
To this end, focusing on mitochondrial plasticity may provide im- oysters (Sanni et al., 2008). However, a battery of oxidative stress re-
portant insights into overall organismal fitness in changing environ- sponse enzymes (e.g., manganese superoxide dismutase, catalase, glu-
ments. The capacity for mitochondria to repair damage and alter mi- tathione peroxidase etc.) and markers of oxidative damage (e.g., lipid
tochondrial structure, content, and copy number (i.e., conserving a peroxidation) are often used and may serve as indicators of mi-
health population of mitochondria by regenerative biogenesis) is cri- tochondrial vulnerability to ROS production and ROS-induced damage
tical in maintaining mitochondrial plasticity. This can be assessed based (Massarsky et al., 2017).
on the following parameters (Szendroedi et al., 2012; Jelenik and Although molecular changes, including mtDNA sequence changes as
Roden, 2013): (i) In vivo resting oxidative phosphorylation at low ADP discussed next, are excellent indicators of mitochondrial health, cou-
concentrations; (ii) In vivo submaximal ADP-stimulated oxidative pling such markers with functional assays will be crucial in assessing
phosphorylation; (iii) Ex vivo maximal ADP-stimulated oxidative mitochondrial function at a population level. The in ovo and ex vivo
phosphorylation in isolated mitochondria or permeabilized fibers, or functional assays provide a rapid and relatively simple approach to
whole tissues assessed based on oxygen consumption rate measure- estimate mitochondrial bioenergetics. An important consideration in
ments; (iv) Ex vivo mitochondrial reserve capacity—the difference be- functional studies is life history stages, since periods of gametogenesis
tween ATP coupled oxygen consumption rate and uncoupled oxygen and embryonic development are critical windows for exposure to mi-
consumption rates. Specifically, ex vivo and in ovo measurements using totoxicants (Stackley et al., 2011). Species’ vulnerability to mitotox-
the XFe Flux Analyzer will be highly useful in evaluating the mi- icants is likely to be highest during the developmental window of surge
tochondrial plasticity of wild-caught fish species and potentially others in mitochondrial biogenesis, where a profound increase in dynamics
organisms, thereby enabling a population level multi-stress response (mitophagy, fusion, and fission), and consequent shifts in distribution
assessment. and density of mitochondria occurs (Dorn et al., 2015). In fact in heart
Mitochondrial health index (MHI)—a concept introduced by Chacko tissues, mitochondrial function and architecture are defined during
et al. (2014) to describe a bioenergetic health index (BHI) in human developmental mitochondrial biogenesis (Dorn et al., 2015). Thus,
populations—derived from bioenergetic reserve capacity, ATP-linked considering the role of hearts in setting stress tolerance limits as dis-
respiration, proton leak (Fig. 3) and non-mitochondrial respiration can cussed previously, cardiac mitochondria and its plasticity (the capacity
potentially serve as an indicator of mitochondrial health and plasticity to maintain function and integrity with exposure to stressors) may serve
of an organism. Similar to BHI (Chacko et al., 2014), MHI is calculated as a useful ecologically important marker of mitochondrial toxicity, at
(reserve capacity )a × (ATP linked)b least in teleost fish. Collectively, in ovo and ex vivo bioenergetic studies
as following: MHI = log , where a, b, c, and d
(non-mitochindrial)c × (proto leak )d are a powerful tool in assessment of population level mitochondrial
are predefined parameters based on control groups. In fact a major fitness across temporal and spatial scales, i.e., landscape bioenergetic
limitation in this approach is that MHI calculation is highly dependent analyses.
on parameters a,b,c, and d. Thus, the application of MHI approach at

70
N. Jayasundara Toxicology 391 (2017) 64–74

8. Canneries in the coalmine: human health implications of populations or subpopulations of species inhabiting contaminated sites
ecotoxicology of mitotoxicants with clean conditions present unique natural experimental setups for
examining mutagenesis with pollution exposure. For example, a mtDNA
As discussed in the previous section, certain mitochondrial bio- analysis showed an increase in haplotypes and polymorphic loci, ge-
markers can help to evaluate population level impacts of mitochondrial netic diversity, and base pair substitutions in rodents exposed to low
toxicants in wildlife, but can also be informative in human health levels of radiation over 50 generations (Baker et al., 2017). Similar
contexts. Especially, mtDNA sequence analyses can provide important studies showed altered mitochondrial genome diversity in marsh frogs
insights into transgenerational effects of exposure and later life con- (Rana ridibunda) in an industrial waste site (Matson et al., 2006) and in
sequences of developmental exposure to mitotoxicants in wildlife and kangaroo rats (Dipodomys merriami) inhabiting an atomic blast site
human health studies, though may present some caveats. For example, (Theodorakis and Bickham, 2004). With the advent of next generation
in evolutionary biology mtDNA is often used as a genetic marker due to sequencing capabilities, these analyses are likely to become more ac-
their maternal inheritance, absence or very low levels (Neiman and curate and informative, and will enable studies in non-model organisms
Taylor, 2009) of recombination, the presence of highly conserved as to better inform effects of contaminants on mtDNA. Thus, in accordance
well as less conserved regions within the same circular DNA strand, and with the “One Health” and “One Toxicology” concepts that are cur-
the relatively higher mutation rates compared to the nuclear DNA rently being discussed to integrate human and wildlife health impacts
(Ladoukakis and Zouros, 2017). However, these fundamental premises of chemical exposures(Beasley, 2009; Buttke, 2011), assessment of
are complicated by emerging evidence on paternal transfer (or leakage) mtDNA as well as other mitochondrial markers in animal populations
of mtDNA (Schwartz and Vissing, 2002) and occurrence of re- can potentially serve as key indicators of long-term human health ef-
combination (Neiman and Taylor, 2009; Kraytsberg et al., 2004) across fects of exposure to anthropogenic chemicals.
various taxa including in humans. Despite these challenges, evolu-
tionary analyses based on mtDNA in organisms exposed to anthro- 9. Future directions
pogenic chemicals over multiple generations may contribute to ex-
amining effects of contaminants on mtDNA. Additionally, with greater Two emerging aspects of mitochondrial biology that warrant sig-
understanding of a population baseline, these studies can serve as in- nificant attention in the context of mitotoxicants are mitochondrial
dicators of historical exposure to mitotoxicants and other stressors. epigenetics and cross talk between the commensal microbiome and
Furthermore, mtDNA analyses provide important insights into popula- mitochondria. The latter may have profound ecological implications
tion connectivity and migration. Collectively, in addition to informing considering the rapid shifts in microbial communities that may occur
population dynamics within a species, including in the contexts of or- with physical and chemical stressors including increased release of anti-
ganisms inhabiting contaminated sites (Bickham, 2011), these analyses microbial compounds (e.g. antibiotics and nano materials) to the en-
may have important implications in mitochondrial mutagenesis re- vironment. Mitochondrial epigenetics, includes transcriptional regula-
search. tion of mtDNA via differential methylation, and the mitochondrial-
More specifically, these studies may help understand sources of epigenome cross talk. Mitochondrial epigenetics is likely to be a central
mitochondrial mutations and mutation accumulation rates with con- consideration in determining the role of mitochondria and their dys-
taminant exposure—areas of research that are particularly con- function in modulation of acute and acclimatization responses to
troversial in human mitochondrial health studies as highlighted in this abiotic stressors.
issue (Meyer et al., 2017; Chan, 2017) and elsewhere (Valente et al., Methylation of mitochondrial DNA remains contentious with studies
2016). However, several challenges are associated with these ap- showing contradictory evidence (Liu et al., 2016; van der Wijst and
proaches. Naturally occurring mutations, especially at a higher rate in Rots, 2015) and is discussed in detail in this issue (Weinhouse, 2017).
mitochondria, migration or mixing of populations, and diversifying Nonetheless, the discovery of the DNA methyltransferase 1 in mi-
selection confound these analyses adding a layer of complexity in de- tochondria (Shock et al., 2011) and that ROS can alter DNA methyla-
termination of new mutations resulting from pollution exposures tion, suggest that epigenetic changes of mtDNA are likely and could be
(Bickham, 2011). For example, mitochondrial polymorphisms, a key altered with chemical exposures. Furthermore, role of mitotoxicants on
evolutionary marker indicating genetic diversity (haplotype diversity) the mitochondrial modulation of nuclear DNA methylation is of further
of a population (Moore, 1995) can reveal increased mutation rates due significance (Guha and Avadhani, 2013). As discussed in detail by
to mitotoxicant exposure and may show greater genetic diversity, but Weinhouse (2017), mitochondria-epigenome crosstalk is an important
also may mask loss of genetic diversity (Bickham, 2011). Thus, asses- target of mitotoxicants, and the ecological implications of such changes
sing both nucleotide and haplotype diversity in these contexts is im- are yet to be studied. Via mitochondria-epigenome crosstalk, it is pos-
portant in providing further insights into shifts in overall genetic di- sible that a diverse mitochondrial population may lead to differential
versity. In addition to mtDNA polymorphisms, mtDNA damage levels, organismal gene expression patterns (Hill, 2015; Hoekstra et al., 2013),
changes in copy number, and shifts in mtDNA heterogeneity, also although a recent study in killifish Fundulus heteroclitus indicated only
known as mitochondrial heteroplasmy, could be used in these studies as modest changes in gene expression patterns with cold temperature
markers of altered mitochondria. However, it is thought that as a result exposure between populations with different mitochondrial genotypes
of increased mutation rates and limited protection against oxidative (Healy et al., 2017). Nonetheless, a greater understanding of environ-
stress, a baseline level of mitochondrial mutations and heteroplasmy mental regulation of mitochondrial epigenetics will be critical in de-
are present within a cell or a tissue (Chinnery et al., 2012). Thus, to termining organismal response to multiple anthropogenic stressors.
evaluate effects of mitotoxicants on mtDNA, well-controlled studies are
necessary to determine baseline mtDNA mutation rates, damage, copy 10. Conclusions
number, and heteroplasmy. Furthermore, a potential increase in mu-
tation rates with chemical exposure may generate a mixed population Mitochondria are key organelles involved in maintaining cellular
of mtDNAs, resulting in increased heteroplasmy. As discussed pre- homeostasis and also are targets of physical and chemical abiotic
viously by Brunst et al. (2015), despite the advantages mitochondrial stressors. Emergence of mitochondrial toxicants as important category
heterogeneity within an organism, beyond a certain threshold, in- of anthropogenic chemicals indicate the need to better understand the
creased damage and heteroplasmy affect optimal function; a trait that effects of these compounds on organismal response to global climate
might be selected against in a population which may lead to under- change. To that end, further research is necessary to gain mechanistic
estimations of mtDNA mutations in a population. insights into differential effects of mitotoxicants based on the magni-
Despite these challenges, mtDNA sequence comparisons between tude and duration of the exposure and especially history of exposure

71
N. Jayasundara Toxicology 391 (2017) 64–74

(e.g., the protective role of mitohormesis) and methods to incorporate Int. J. Biochem. Cell Biol. 41 (4), 791–799.
Casals-Casas, C., Desvergne, B., 2011. Endocrine disruptors: from endocrine to metabolic
such findings into population level analyses. Importantly, a greater disruption. Annu. Rev. Physiol. 73, 135–162.
understanding of mitochondrial epigenetics and impact of mitotox- Chacko, B.K., Kramer, P.A., Ravi, S., Benavides, G.A., Mitchell, T., Dranka, B.P., Ferrick,
icants on this process may provide crucial insights into mitochondrial, D., Singal, A.K., Ballinger, S.W., Bailey, S.M., 2014. The Bioenergetic Health Index: a
new concept in mitochondrial translational research. Clin. Sci. 127 (6), 367–373.
cellular, and organismal responses to biotic and abiotic stressors. Chan, S.S.L., 2017. Inherited mitochondrial genomic instability and chemical exposures.
Accordingly, the discussion presented here indicates that mitochondria Toxicology 391, 75–83.
may in fact serve as a potential marker of physiological resilience of Cherkasov, A., Taylor, C., Sokolova, I., 2010. Seasonal variation in mitochondrial re-
sponses to cadmium and temperature in eastern oysters Crassostrea virginica (Gmelin)
organisms to environmental shifts and may help to portend species’ from different latitudes. Aquat. Toxicol. 97 (1), 68–78.
fitness and survival with climate change. Additionally, these analyses Chinnery, P.F., Elliott, H.R., Hudson, G., Samuels, D.C., Relton, C.L., 2012. Epigenetics,
can be important in predicting human health impacts of mitotoxicants epidemiology and mitochondrial DNA diseases. Int. J. Epidemiol dyr232.
Clapham, D.E., 2007. Calcium signaling. Cell 131 (6), 1047–1058.
and lends to the “One Health” concept that is being widely discussed as
Cutkomp, L., Yap, H., Vea, E., Koch, R., 1971. Inhibition of oligomycin-sensitive (mi-
an important approach in toxicology and human health (Beasley, 2009; tochondrial) Mg2+ ATPase by DDT and selected analogs in fish and insect tissue. Life
Buttke, 2011). Sci. 10 (21), 1201–1209.
Davoodi, F., Claireaux, G., 2007. Effects of exposure to petroleum hydrocarbons upon the
metabolism of the common sole Solea solea. Mar. Pollut. Bull. 54 (7), 928–934.
Acknowledgments de Quadros, T., Schramm, H., Zeni, E.C., Simioni, C., Allodi, S., Müller, Y.M., Ammar, D.,
Nazari, E.M., 2016. Developmental effects of exposure to ultraviolet B radiation on
The author would like to thank the editors, the reviewers and Dr. the freshwater prawn Macrobrachium olfersi: Mitochondria as a target of environ-
mental UVB radiation. Ecotoxicol. Environ. Saf. 132, 279–287.
Andrey Massarsky, Duke University, NC for their critical insights and De Smet, H., Blust, R., 2001. Stress responses and changes in protein metabolism in carp
feedback. Cyprinus carpio during cadmium exposure. Ecotoxicol. Environ. Saf. 48 (3), 255–262.
Derocher, A.E., Stirling, I., 1996. Aspects of survival in juvenile polar bears. Can. J. Zool.
74 (7), 1246–1252.
Appendix A. Supplementary data Di Giulio, R.T., Clark, B.W., 2015. The Elizabeth River story: a case study in evolutionary
toxicology. J. Toxicol. Environ. Health B: Crit. Rev. 18 (6), 259–298.
Supplementary data associated with this article can be found, in the Dorn, G.W., Vega, R.B., Kelly, D.P., 2015. Mitochondrial biogenesis and dynamics in the
developing and diseased heart. Genes. Dev. 29 (19), 1981–1991.
online version, at http://dx.doi.org/10.1016/j.tox.2017.07.015.
Dorts, J., Kestemont, P., Thézenas, M.-L., Raes, M., Silvestre, F., 2014. Effects of cadmium
exposure on the gill proteome of Cottus gobio: modulatory effects of prior thermal
References acclimation. Aquat. Toxicol. 154, 87–96.
Du, X., Crawford, D.L., Nacci, D.E., Oleksiak, M.F., 2016. Heritable oxidative phosphor-
ylation differences in a pollutant resistant Fundulus heteroclitus population. Aquat.
Abele, D., Puntarulo, S., 2004. Formation of reactive species and induction of antioxidant Toxicol. 177, 44–50.
defence systems in polar and temperate marine invertebrates and fish. Comp. Dussauze, M., Camus, L., Le Floch, S., Pichavant-Rafini, K., Geraudie, P., Coquillé, N.,
Biochem. Physiol. A: Mol. Integr. Physiol. 138 (4), 405–415. Amérand, A., Lemaire, P., Theron, M., 2014. Impact of dispersed fuel oil on cardiac
Ahn, C.S., Metallo, C.M., 2015. Mitochondria as biosynthetic factories for cancer pro- mitochondrial function in polar cod Boreogadus saida. Environ. Sci. Pollut. Res. 21
liferation. Cancer Metab. 3 (1), 1. (24), 13779–13788.
Anawar, H.M., 2013. Impact of climate change on acid mine drainage generation and Fabbri, E., Caselli, F., Piano, A., Sartor, G., Capuzzo, A., 2003. Cd 2+ and Hg 2+ affect
contaminant transport in water ecosystems of semi-arid and arid mining areas. Phys. glucose release and cAMP-dependent transduction pathway in isolated eel hepato-
Chem. Earth A/B/C 58, 13–21. cytes. Aquat. Toxicol. 62 (1), 55–65.
Andreau, K., Leroux, M., Bouharrour, A., 2012. Health and cellular impacts of air pol- Falfushynska, H., Gnatyshyna, L., Yurchak, I., Sokolova, I., Stoliar, O., 2015. The effects of
lutants: from cytoprotection to cytotoxicity. Biochem. Res. Int. 2012. zinc nanooxide on cellular stress responses of the freshwater mussels Unio tumidus
Askwith, C., Kaplan, J., 1998. Iron and copper transport in yeast and its relevance to are modulated by elevated temperature and organic pollutants. Aquat. Toxicol. 162,
human disease. Trends Biochem. Sci. 23 (4), 135–138. 82–93.
Bagwe, R., Beniash, E., Sokolova, I.M., 2015. Effects of cadmium exposure on critical Foote, A.D., Morin, P.A., Durban, J.W., Pitman, R.L., Wade, P., Willerslev, E., Gilbert,
temperatures of aerobic metabolism in eastern oysters Crassostrea virginica (Gmelin, M.T.P., da Fonseca, R.R., 2011. Positive selection on the killer whale mitogenome.
1791). Aquat. Toxicol. 167, 77–89. Biol. Lett. 7 (1), 116–118.
Baker, R.J., Dickins, B., Wickliffe, J.K., et al., 2017. Elevated mitochondrial genome Friedman, J.R., Nunnari, J., 2014. Mitochondrial form and function. Nature 505 (7483),
variation after 50 generations of radiation exposure in a wild rodent. Evol Appl. 335–343.
00, 1–8. Gabriely, G., Wheeler, M.A., Takenaka, M.C., Quintana, F.J., 2017. Role of AHR and HIF-
Banerjee, S., Ghosh, J., Sil, P., 2016. Drug metabolism and oxidative stress: cellular 1α in Glioblastoma Metabolism. Trends in Endocrinology & Metabolism 28 (6),
mechanism and new therapeutic insights. Biochem. Anal. Biochem. 5, 255. 428–436.
Beasley, V., 2009. ‘One toxicology’, ‘ecosystem health’ and ‘one health’. Vet. Ital. 45 (1), Gakh, O., Ranatunga, W., Smith, D.Y., Ahlgren, E.-C., Al-Karadaghi, S., Thompson, J.R.,
97–110. Isaya, G., 2016. Architecture of the human mitochondrial iron-sulfur cluster assembly
Begriche, K., Massart, J., Robin, M.-A., Borgne-Sanchez, A., Fromenty, B., 2011. Drug- machinery. J. Biol. Chem. 291 (40), 21296–21321.
induced toxicity on mitochondria and lipid metabolism: mechanistic diversity and Gambrell, R., 1994. Trace and toxic metals in wetlands—a review. J. Environ. Qual. 23
deleterious consequences for the liver. J. Hepatol. 54 (4), 773–794. (5), 883–891.
Benard, G., Bellance, N., James, D., Parrone, P., Fernandez, H., Letellier, T., Rossignol, R., Gerger, C.J., Weber, L.P., 2015. Comparison of the acute effects of benzo-a-pyrene on
2007. Mitochondrial bioenergetics and structural network organization. J. Cell Sci. adult zebrafish (Danio rerio) cardiorespiratory function following intraperitoneal in-
120 (5), 838–848. jection versus aqueous exposure. Aquat. Toxicol. 165, 19–30.
Benedetti, M., Lanzoni, I., Nardi, A., d'Errico, G., Di Carlo, M., Fattorini, D., Nigro, M., Gerger, C.J., Thomas, J.K., Janz, D.M., Weber, L.P., 2015. Acute effects of β-naphtho-
Regoli, F., 2016. Oxidative responsiveness to multiple stressors in the key Antarctic flavone on cardiorespiratory function and metabolism in adult zebrafish (Danio rerio).
species, Adamussium colbecki: interactions between temperature, acidification and Fish. Physiol. Biochem. 41 (1), 289–298.
cadmium exposure. Mar. Environ. Res. 121, 20–30. Green, D.R., Van Houten, B., 2011. Mitochondrial quality control. Cell 147 (4), 950.
Bernhardt, E.S., Rosi, E.J., Gessner, M.O., 2017. Synthetic chemicals as agents of global Grivennikova, V.G., Kareyeva, A.V., Vinogradov, A.D., 2010. What are the sources of
change. Front. Ecol. Environ. 15 (2), 84–90. hydrogen peroxide production by heart mitochondria? Biochimica et Biophysica Acta
Bickham, J.W., 2011. The four cornerstones of evolutionary toxicology. Ecotoxicology 20 (BBA)-Bioenergetics 1797 (6), 939–944.
(3), 497–502. Grun, F., Blumberg, B., 2006. Environmental obesogens: organotins and endocrine dis-
Blajszczak, C., Bonini, M.G., 2017. Mitochondria targeting by environmental stressors: ruption via nuclear receptor signaling. Endocrinology 147 (6), s50–s55.
implications for redox cellular signaling. Toxicology 391, 84–89. Guha, M., Avadhani, N.G., 2013. Mitochondrial retrograde signaling at the crossroads of
Brunst, K.J., Baccarelli, A.A., Wright, R.J., 2015. Integrating mitochondriomics in chil- tumor bioenergetics, genetics and epigenetics. Mitochondrion 13 (6), 577–591.
dren's environmental health. J. Appl. Toxicol. 35 (9), 976–991. Haider, F., Falfushynska, H., Ivanina, A.V., Sokolova, I.M., 2016. Effects of pH and bi-
Buttke, D.E., 2011. Toxicology, environmental health, and the One Health concept. J. carbonate on mitochondrial functions of marine bivalves. Comp. Biochem. Physiol. A:
Med. Toxicol. 7 (4), 329–332. Mol. Integr. Physiol. 198, 41–50.
Calow, P., Forbes, V.E., 1998. How do physiological responses to stress translate into Harley, C.D., Randall Hughes, A., Hultgren, K.M., Miner, B.G., Sorte, C.J., Thornber, C.S.,
ecological and evolutionary processes? Comp. Biochem. Physiol. A: Mol. Integr. Rodriguez, L.F., Tomanek, L., Williams, S.L., 2006. The impacts of climate change in
Physiol. 120 (1), 11–16. coastal marine systems. Ecol. Lett. 9 (2), 228–241.
Calvo, S.E., Mootha, V.K., 2010. The mitochondrial proteome and human disease. Annu. Healy, T.M., Bryant, H.J., Schulte, P.M., 2017. Mitochondrial genotype and phenotypic
Rev. Genomics Hum. Genet. 11, 25–44. plasticity of gene expression in response to cold acclimation in killifish. Mol. Ecol. 26
Cambier, S., Benard, G., Mesmer-Dudons, N., Gonzalez, P., Rossignol, R., Brethes, D., (3), 814–830.
Bourdineaud, J.-P., 2009. At environmental doses, dietary methylmercury inhibits Hermes-Lima, M., 2004. Oxygen in biology and biochemistry: role of free radicals. Funct.
mitochondrial energy metabolism in skeletal muscles of the zebra fish (Danio rerio). Metab. Regul. Adapt. 1, 319–366.

72
N. Jayasundara Toxicology 391 (2017) 64–74

Hill, G.E., 2015. Mitonuclear ecology. Mol. Biol. Evol. 32 (8), 1917–1927. 304 (5673), 981.
Hisamichi, Y., Haraguchi, K., Endo, T., 2010. Levels of mercury and organochlorine Kuznetsov, A.V., Veksler, V., Gellerich, F.N., Saks, V., Margreiter, R., Kunz, W.S., 2008.
compounds and stable isotope ratios in three tuna species taken from different re- Analysis of mitochondrial function in situ in permeabilized muscle fibers, tissues and
gions of Japan. Environ. Sci. Technol. 44 (15), 5971–5978. cells. Nat. Protoc. 3 (6), 965–976.
Hoekstra, L.A., Siddiq, M.A., Montooth, K.L., 2013. Pleiotropic effects of a mitochon- Løkke, H., Ragas, A.M., Holmstrup, M., 2013. Tools and perspectives for assessing che-
drial?nuclear incompatibility depend upon the accelerating effect of temperature in mical mixtures and multiple stressors. Toxicology 313 (2), 73–82.
Drosophila. Genetics 195 (3), 1129–1139. Ladoukakis, E.D., Zouros, E., 2017. Evolution and inheritance of animal mitochondrial
Hooper, M.J., Ankley, G.T., Cristol, D.A., Maryoung, L.A., Noyes, P.D., Pinkerton, K.E., DNA: rules and exceptions. J. Biol. Res. Thessaloniki 24 (1), 2.
2013. Interactions between chemical and climate stressors: a role for mechanistic Lannig, G., Cherkasov, A.S., Pörtner, H.-O., Bock, C., Sokolova, I.M., 2008. Cadmium-
toxicology in assessing climate change risks. Environ. Toxicol. Chem. 32 (1), 32–48. dependent oxygen limitation affects temperature tolerance in eastern oysters
Huey, R., 1982. Temperature, physiology, and the ecology of reptiles. In: In: Gans, C., (Crassostrea virginica Gmelin). Am. J. Physiol. Regul. Integr. Comp. Physiol. 294 (4),
Pough, F.H. (Eds.), Biology of the Reptilia Vol. 12. Physiology C. Physiological R1338–R1346.
ecology. Academic Press, New York, pp. 25–91. Lemasters, J.J., Holmuhamedov, E., 2006. Voltage-dependent anion channel (VDAC) as
Huey, R.B., Kearney, M.R., Krockenberger, A., Holtum, J.A., Jess, M., Williams, S.E., mitochondrial governator—thinking outside the box. Biochimica et Biophysica Acta
2012. Predicting organismal vulnerability to climate warming: roles of behaviour, (BBA)-Molecular Basis of Disease 1762 (2), 181–190.
physiology and adaptation. Phil. Trans. R. Soc. B 367 (1596), 1665–1679. Lesser, M.P., 2006. Oxidative stress in marine environments: biochemistry and physio-
Hwang, H.J., Dornbos, P., LaPres, J.J., 2016a. AHR-dependent changes in the mi- logical ecology. Annu. Rev. Physiol. 68, 253–278.
tochondrial proteome in response to 2, 3, 7, 8-tetrachlorodibenzo-p-dioxin. Data in Lin, Y., Sun, X., Qiu, L., Wei, J., Huang, Q., Fang, C., Ye, T., Kang, M., Shen, H., Dong, S.,
Brief 8, 191–195. 2013. Exposure to bisphenol A induces dysfunction of insulin secretion and apoptosis
Hwang, H.J., Dornbos, P., Steidemann, M., Dunivin, T.K., Rizzo, M., LaPres, J.J., 2016b. through the damage of mitochondria in rat insulinoma (INS-1) cells. Cell. Death. Dis.
Mitochondrial-targeted aryl hydrocarbon receptor and the impact of 2, 3, 7, 8-tet- 4 (1), e460.
rachlorodibenzo-p-dioxin on cellular respiration and the mitochondrial proteome. Liu, B., Du, Q., Chen, L., Fu, G., Li, S., Fu, L., Zhang, X., Ma, C., Bin, C., 2016. CpG
Toxicol. Appl. Pharmacol. 304, 121–132. methylation patterns of human mitochondrial DNA. Sci. Rep. 6.
Iftikar, F.I., Hickey, A.J., 2013. Do mitochondria limit hot fish hearts? Understanding the Logan, C.A., Buckley, B.A., 2015. Transcriptomic responses to environmental temperature
role of mitochondrial function with heat stress in Notolabrus celidotus. PLoS One 8 (5), in eurythermal and stenothermal fishes. J. Exp. Biol. 218 (12), 1915–1924.
e64120. Müller, M., Mentel, M., van Hellemond, J.J., Henze, K., Woehle, C., Gould, S.B., Yu, R.-Y.,
Ivanina, A.V., Sokolova, I.M., 2016. Effects of intermittent hypoxia on oxidative stress and van der Giezen, M., Tielens, A.G., Martin, W.F., 2012. Biochemistry and evolution of
protein degradation in molluscan mitochondria. J. Exp. Biol. 219 (23), 3794–3802. anaerobic energy metabolism in eukaryotes. Microbiol. Mol. Biol. Rev. 76 (2),
Ivanina, A.V., Habinck, E., Sokolova, I.M., 2008. Differential sensitivity to cadmium of 444–495.
key mitochondrial enzymes in the eastern oyster, Crassostrea virginica Gmelin Mager, E.M., Esbaugh, A.J., Stieglitz, J.D., Hoenig, R., Bodinier, C., Incardona, J.P.,
(Bivalvia: ostreidae). Comp. Biochem. Physiol. C: Toxicol. Pharmacol. 148 (1), 72–79. Scholz, N.L., Benetti, D.D., Grosell, M., 2014. Acute embryonic or juvenile exposure
Ivanina, A.V., Froelich, B., Williams, T., Sokolov, E.P., Oliver, J.D., Sokolova, I.M., 2011. to Deepwater Horizon crude oil impairs the swimming performance of mahi-mahi
Interactive effects of cadmium and hypoxia on metabolic responses and bacterial (Coryphaena hippurus). Environ. Sci. Technol. 48 (12), 7053–7061.
loads of eastern oysters Crassostrea virginica Gmelin. Chemosphere 82 (3), 377–389. Massarsky, A., Kozal, J.S., Di Giulio, R.T., 2017. Glutathione and zebrafish: old assays to
Ivanina, A.V., Kurochkin, I.O., Leamy, L., Sokolova, I.M., 2012. Effects of temperature and address a current issue. Chemosphere 168, 707–715.
cadmium exposure on the mitochondria of oysters (Crassostrea virginica) exposed to Matson, C.W., Lambert, M.M., McDonald, T.J., Autenrieth, R.L., Donnelly, K.C.,
hypoxia and subsequent reoxygenation. J. Exp. Biol. 215 (18), 3142–3154. Islamzadeh, A., Politov, D.I., Bickham, J.W., 2006. Evolutionary toxicology: popu-
Jayasundara, N., Somero, G.N., 2013. Physiological plasticity of cardiorespiratory func- lation-level effects of chronic contaminant exposure on the marsh frogs (Rana ridi-
tion in a eurythermal marine teleost, the longjaw mudsucker, Gillichthys mirabilis. J. bunda) of Azerbaijan. Environ. Health Perspect. 114 (4), 547.
Exp. Biol. 216 (11), 2111–2121. Mattingly, C., Rosenstein, M., Colby, G., Forrest Jr, J., Boyer, J., 2006. The Comparative
Jayasundara, N., Gardner, L.D., Block, B.A., 2013. Effects of temperature acclimation on Toxicogenomics Database (CTD): a resource for comparative toxicological studies. J.
Pacific bluefin tuna (Thunnus orientalis) cardiac transcriptome. Am. J. Physiol. Regul. Exp. Zool. A: Ecol. Genet. Physiol. 305 (9), 689–692.
Integr. Comp. Physiol. 305 (9), R1010–R1020. Mercer, T.R., Neph, S., Dinger, M.E., Crawford, J., Smith, M.A., Shearwood, A.-M.J.,
Jayasundara, N., Tomanek, L., Dowd, W.W., Somero, G.N., 2015a. Proteomic analysis of Haugen, E., Bracken, C.P., Rackham, O., Stamatoyannopoulos, J.A., 2011. The human
cardiac response to thermal acclimation in the eurythermal goby fish Gillichthys mitochondrial transcriptome. Cell 146 (4), 645–658.
mirabilis. J. Exp. Biol. 218 (9), 1359–1372. Meyer, J.N., Leung, M.C., Rooney, J.P., Sendoel, A., Hengartner, M.O., Kisby, G.E., Bess,
Jayasundara, N., Kozal, J.S., Arnold, M.C., Chan, S.S., Di Giulio, R.T., 2015b. High- A.S., 2013. Mitochondria as a target of environmental toxicants. Toxicol. Sci. 134 (1),
throughput tissue bioenergetics analysis reveals identical metabolic allometric 1–17.
scaling for teleost hearts and whole organisms. PLoS One 10 (9), e0137710. Meyer, J.N., Leuthner, T.C., Luz, A.L., 2017. Mitochondrial fusion, fission, and mi-
Jayasundara, N., Fernando, P.W., Osterberg, J.S., Cammen, K.M., Schultz, T.F., Di Giulio, tochondrial toxicity. Toxicology 391, 42–53.
R.T., 2017. Cost of tolerance: physiological consequences of evolved resistance to Mommaerts, V., Hagenaars, A., Meyer, J., De Coen, W., Swevers, L., Mosallanejad, H.,
inhabit a polluted environment in teleost fish Fundulus heteroclitus. Environmental Smagghe, G., 2011. Impact of a perfluorinated organic compound PFOS on the ter-
Science & Technology 51 (15), 8763–8772. restrial pollinator Bombus terrestris (Insecta, Hymenoptera). Ecotoxicology 20 (2),
Jelenik, T., Roden, M., 2013. Mitochondrial plasticity in obesity and diabetes mellitus. 447–456.
Antioxid. Redox Signal. 19 (3), 258–268. Moore, W.S., 1995. Inferring phylogenies from mtDNA variation: mitochondrial-gene
Jendrach, M., Mai, S., Pohl, S., Vöth, M., Bereiter-Hahn, J., 2008. Short-and long-term trees versus nuclear-gene trees. Evolution 718–726.
alterations of mitochondrial morphology, dynamics and mtDNA after transient oxi- Moreira, A., Figueira, E., Soares, A.M., Freitas, R., 2016. Salinity influences the bio-
dative stress. Mitochondrion 8 (4), 293–304. chemical response of Crassostrea angulata to Arsenic. Environ. Pollut. 214, 756–766.
Jornayvaz, F.R., Shulman, G.I., 2010. Regulation of mitochondrial biogenesis. Essays Nedergaard, J., Golozoubova, V., Matthias, A., Asadi, A., Jacobsson, A., Cannon, B., 2001.
Biochem. 47, 69–84. UCP1: the only protein able to mediate adaptive non-shivering thermogenesis and
Kültz, D., 2005. Molecular and evolutionary basis of the cellular stress response. Annu. metabolic inefficiency. Biochimica et Biophysica Acta (BBA)-Bioenergetics 1504 (1),
Rev. Physiol. 67, 225–257. 82–106.
Kang, J.-H., Aasi, D., Katayama, Y., Bisphenol, A., 2007. in the aquatic environment and Neiman, M., Taylor, D.R., 2009. The causes of mutation accumulation in mitochondrial
its endocrine-disruptive effects on aquatic organisms. Crit. Rev. Toxicol. 37 (7), genomes. Proc. R. Soc. Lond. B: Biol. Sci rspb. 2008.1758.
607–625. Noyes, P.D., Lema, S.C., 2015. Forecasting the impacts of chemical pollution and climate
Kaur, K., Chauhan, V., Gu, F., Chauhan, A., Bisphenol, A., 2014. induces oxidative stress change interactions on the health of wildlife. Curr. Zool. 61 (4), 669–689.
and mitochondrial dysfunction in lymphoblasts from children with autism and un- Noyes, P.D., McElwee, M.K., Miller, H.D., Clark, B.W., Van Tiem, L.A., Walcott, K.C.,
affected siblings. Free Radic. Biol. Med. 76, 25–33. Erwin, K.N., Levin, E.D., 2009. The toxicology of climate change: environmental
Kim, I., Lemasters, J.J., 2011. Mitophagy selectively degrades individual damaged mi- contaminants in a warming world. Environ. Int. 35 (6), 971–986.
tochondria after photoirradiation. Antioxid. Redox Signal. 14 (10), 1919–1928. Okamoto, K., Shaw, J.M., 2005. Mitochondrial morphology and dynamics in yeast and
Knecht, A.L., Goodale, B.C., Truong, L., Simonich, M.T., Swanson, A.J., Matzke, M.M., multicellular eukaryotes. Annu. Rev. Genet. 39, 503–536.
Anderson, K.A., Waters, K.M., Tanguay, R.L., 2013. Comparative developmental Oliveira, P.J., Wallace, K.B., 2006. Depletion of adenine nucleotide translocator protein in
toxicity of environmentally relevant oxygenated PAHs. Toxicol. Appl. Pharmacol. heart mitochondria from doxorubicin-treated rats—relevance for mitochondrial
271 (2), 266–275. dysfunction. Toxicology 220 (2), 160–168.
Koczor, C.A., Shokolenko, I.N., Boyd, A.K., Balk, S.P., Wilson, G.L., LeDoux, S.P., 2009. Onukwufor, J.O., Kibenge, F., Stevens, D., Kamunde, C., 2016a. Hypoxia-reoxygenation
Mitochondrial DNA damage initiates a cell cycle arrest by a Chk2-associated me- differentially alters the thermal sensitivity of complex I basal and maximal mi-
chanism in mammalian cells. J. Biol. Chem. 284 (52), 36191–36201. tochondrial oxidative capacity. Comp. Biochem. Physiol. A: Mol. Integr. Physiol. 201,
Kozal, J.S., Jayasundara, N.J., Di Giulio, R.T., 2017. Transgenerational exposure to benzo 87–94.
(a)pyrene affects metabolic plasticity and thermal stress response capacity. In: Onukwufor, J.O., Stevens, D., Kamunde, C., 2016b. Bioenergetic and volume regulatory
International Symposium on Pollutant Responses in Marine Organisms. Matsuyama, effects of mitoKATP channel modulators protect against hypoxia–reoxygenation-in-
Japan. duced mitochondrial dysfunction. J. Exp. Biol. 219 (17), 2743–2751.
Kraemer, L., Schulte, P., 2004. Prior PCB. exposure suppresses hypoxia-induced up-reg- Ott, M., Gogvadze, V., Orrenius, S., Zhivotovsky, B., 2007. Mitochondria, oxidative stress
ulation of glycolytic enzymes in Fundulus heteroclitus. Comp. Biochem. Physiol. C: and cell death. Apoptosis 12 (5), 913–922.
Toxicol. Pharmacol. 139 (1), 23–29. Pörtner, H.O., Farrell, A.P., 2008. Physiology and climate change. Science 322 (5902),
Kraytsberg, Y., Schwartz, M., Brown, T.A., Ebralidse, K., Kunz, W.S., Clayton, D.A., 690–692.
Vissing, J., Khrapko, K., 2004. Recombination of human mitochondrial DNA. Science Padmini, E., Geetha, B.V., Rani, M.U., 2009. Pollution induced nitrative stress and heat

73
N. Jayasundara Toxicology 391 (2017) 64–74

shock protein 70 overexpression in fish liver mitochondria. Sci. Total Environ. 407 Study Mitochondrial Radical Production and Its Consequences. Portland Press
(4), 1307–1317. Limited.
Palmieri, F., Pierri, C.L., 2010. Mitochondrial metabolite transport. Essays Biochem. 47, Smolders, R., De Boeck, G., Blust, R., 2003. Changes in cellular energy budget as a
37–52. measure of whole effluent toxicity in zebrafish (Danio rerio). Environ. Toxicol. Chem.
Pandey, S., Parvez, S., Ansari, R.A., Ali, M., Kaur, M., Hayat, F., Ahmad, F., Raisuddin, S., 22 (4), 890–899.
2008. Effects of exposure to multiple trace metals on biochemical, histological and Sokolova, I.M., Lannig, G., 2008. Interactive effects of metal pollution and temperature on
ultrastructural features of gills of a freshwater fish, Channa punctata Bloch. Chem. metabolism in aquatic ectotherms: implications of global climate change. Clim. Res.
Biol. Interact. 174 (3), 183–192. 37 (2–3), 181–201.
Piantadosi, C.A., Suliman, H.B., 2012. Redox regulation of mitochondrial biogenesis. Free Sokolova, I.M., Sukhotin, A.A., Lannig, G., 2011. Stress effects on metabolism and energy
Radic. Biol. Med. 53 (11), 2043–2053. budgets in mollusks. Oxid. Stress Aquatic Ecosyst. 263–280.
Raftery, T.D., Jayasundara, N., Di Giulio, R.T., 2017. A bioenergetics assay for studying Sokolova, I.M., Frederich, M., Bagwe, R., Lannig, G., Sukhotin, A.A., 2012. Energy
the effects of environmental stressors on mitochondrial function in vivo in zebrafish homeostasis as an integrative tool for assessing limits of environmental stress toler-
larvae. Comp. Biochem. Physiol. C: Toxicol. Pharmacol. 192, 23–32. ance in aquatic invertebrates. Mar. Environ. Res. 79, 1–15.
Raimundo, N., Song, L., Shutt, T.E., McKay, S.E., Cotney, J., Guan, M.-X., Gilliland, T.C., Somero, G.N., Lockwood, B.L., Tomanek, L., 2017. Biochemical Adaptation: Response to
Hohuan, D., Santos-Sacchi, J., Shadel, G.S., 2012. Mitochondrial stress engages E2F1 Environmental Challenges from Life’s Origins to the Anthropocene. Sinauer
apoptotic signaling to cause deafness. Cell 148 (4), 716–726. Associates, Inc, Sunderland, MA.
Ramsay, G., oreen Cantrell, D., 2015. Environmental and metabolic sensors that control T Stabenau, E.K., Sasser, A., Schulte, C., 2008. The effects of pyrene exposure on exercise
cell biology. T Cell Regul. Environ. 16. performance, muscle contraction, and mitochondrial O2 consumption in the leopard
Rand, D.M., Haney, R.A., Fry, A.J., 2004. Cytonuclear coevolution: the genomics of co- frog (Rana pipiens). J. Environ. Sci. Health A 43 (6), 576–583.
operation. Trends Ecol. Evol. 19 (12), 645–653. Stackley, K.D., Beeson, C.C., Rahn, J.J., Chan, S.S., 2011. Bioenergetic profiling of zeb-
Robbins, C.T., Lopez-Alfaro, C., Rode, K.D., Tøien, Ø., Nelson, O.L., 2012. Hibernation rafish embryonic development. PLoS One 6 (9), e25652.
and seasonal fasting in bears: the energetic costs and consequences for polar bears. J. Stapleton, P.A., Nichols, C.E., Yi, J., McBride, C.R., Minarchick, V.C., Shepherd, D.L.,
Mammal. 93 (6), 1493–1503. Hollander, J.M., Nurkiewicz, T.R., 2015. Microvascular and mitochondrial dysfunc-
Ross, C., Olsen, K., Henry, M., Pierce, R., 2015. Mosquito control pesticides and sea tion in the female F1 generation after gestational TiO2 nanoparticle exposure.
surface temperatures have differential effects on the survival and oxidative stress Nanotoxicology 9 (8), 941–951.
response of coral larvae. Ecotoxicology 24 (3), 540–552. Stephens, M., Ludgate, M., Rees, D.A., 2011. Brown fat and obesity: the next big thing?
Roubicek, D.A., de Souza Pinto, N.C., 2017. Mitochondria and mitochondrial DNA as Clin. Endocrinol. 74 (6), 661–670.
relevant targets for environmental contaminants. Toxicology 391, 100–108. Storz, P., 2006. Reactive oxygen species–mediated mitochondria-to-nucleus signaling: a
Sanni, B., Williams, K., Sokolov, E.P., Sokolova, I.M., 2008. Effects of acclimation tem- key to aging and radical-caused diseases. Sci. STKE 2006 re3–re3.
perature and cadmium exposure on mitochondrial aconitase and LON protease from a Suarez, R., Lighton, J., Brown, G., Mathieu-Costello, O., 1991. Mitochondrial respiration
model marine ectotherm, Crassostrea virginica. Comp. Biochem. Physiol. C: Toxicol. in hummingbird flight muscles. Proc. Natl. Acad. Sci. 88 (11), 4870–4873.
Pharmacol. 147 (1), 101–112. Swann, J., Schultz, T., Kennedy, J., 1996. The effects of the organophosphorous in-
Sappal, R., MacDonald, N., Fast, M., Stevens, D., Kibenge, F., Siah, A., Kamunde, C., 2014. secticides Dursban™ and Lorsban™ on the ciliated epithelium of the frog palatein vitro.
Interactions of copper and thermal stress on mitochondrial bioenergetics in rainbow Arch. Environ. Contam. Toxicol. 30 (2), 188–194.
trout, Oncorhynchus mykiss. Aquat. Toxicol. 157, 10–20. Szczesny, B., Tann, A.W., Mitra, S., 2010. Age-and tissue-specific changes in mitochon-
Sappal, R., MacDougald, M., Fast, M., Stevens, D., Kibenge, F., Siah, A., Kamunde, C., drial and nuclear DNA base excision repair activity in mice: susceptibility of skeletal
2015a. Alterations in mitochondrial electron transport system activity in response to muscles to oxidative injury. Mech. Ageing Dev. 131 (5), 330–337.
warm acclimation, hypoxia-reoxygenation and copper in rainbow trout, Szendroedi, J., Phielix, E., Roden, M., 2012. The role of mitochondria in insulin resistance
Oncorhynchus mykiss. Aquat. Toxicol. 165, 51–63. and type 2 diabetes mellitus. Nat. Rev. Endocrinol. 8 (2), 92–103.
Sappal, R., Fast, M., Stevens, D., Kibenge, F., Siah, A., Kamunde, C., 2015b. Effects of Tann, A.W., Boldogh, I., Meiss, G., Qian, W., Van Houten, B., Mitra, S., Szczesny, B., 2011.
copper, hypoxia and acute temperature shifts on mitochondrial oxidation in rainbow Apoptosis induced by persistent single-strand breaks in mitochondrial genome cri-
trout (Oncorhynchus mykiss) acclimated to warm temperature. Aquat. Toxicol. 169, tical role of EXOG (5′-EXO/endonuclease) in their repair. J. Biol. Chem. 286 (37),
46–57. 31975–31983.
Sappal, R., Fast, M., Purcell, S., MacDonald, N., Stevens, D., Kibenge, F., Siah, A., Tappenden, D.M., Lynn, S.G., Crawford, R.B., Lee, K., Vengellur, A., Kaminski, N.E.,
Kamunde, C., 2016. Copper and hypoxia modulate transcriptional and mitochondrial Thomas, R.S., LaPres, J.J., 2011. The aryl hydrocarbon receptor interacts with
functional-biochemical responses in warm acclimated rainbow trout (Oncorhynchus ATP5(1, a subunit of the ATP synthase complex, and modulates mitochondrial
mykiss). Environ. Pollut. 211, 291–306. function. Toxicol. Appl. Pharmacol. 254 (3), 299–310.
Schägger, H., 2002. Respiratory chain supercomplexes of mitochondria and bacteria. Theodorakis, C.W., Bickham, J.W., 2004. Molecular characterization of contaminant-in-
Biochimica et Biophysica Acta (BBA)-Bioenergetics 1555 (1), 154–159. dicative RAPD markers. Ecotoxicology 13 (4), 303–309.
Schulte Hostede, S., Darrall, N.M., Blank, L.W., Wellburn, A., 1987. Air pollution and Tissier, F., Dussauze, M., Lefloch, N., Theron, M., Lemaire, P., Le Floch, S., Pichavant-
plant metabolism. In: Proc. 2nd Intn. Symp. Air Pollution & Plant Metabolism. Rafini, K., 2015. Effect of dispersed crude oil on cardiac function in seabass
Munich, April. Dicentrarchus labrax. Chemosphere 134, 192–198.
Schulte, P.M., 2007. Responses to environmental stressors in an estuarine fish: interacting Vafai, S.B., Mootha, V.K., 2012. Mitochondrial disorders as windows into an ancient
stressors and the impacts of local adaptation. J. Therm. Biol 32 (3), 152–161. organelle. Nature 491 (7424), 374–383.
Schulz, E., Wenzel, P., Münzel, T., Daiber, A., 2014. Mitochondrial redox signaling: in- Valente, W.J., Ericson, N.G., Long, A.S., White, P.A., Marchetti, F., Bielas, J.H., 2016.
teraction of mitochondrial reactive oxygen species with other sources of oxidative Mitochondrial DNA exhibits resistance to induced point and deletion mutations.
stress. Antioxid. Redox Signal. 20 (2), 308–324. Nucleic Acids Res. 44 (18), 8513–8524.
Schwartz, M., Vissing, J., 2002. Paternal inheritance of mitochondrial DNA. New Engl. J. van Ginneken, V., Palstra, A., Leonards, P., Nieveen, M., van den Berg, H., Flik, G.,
Med. 347 (8), 576–580. Spanings, T., Niemantsverdriet, P., van den Thillart, G., Murk, A., 2009. PCBs and the
Seiler, J.P., 2002. Pharmacodynamic activity of drugs and ecotoxicology—can the two be energy cost of migration in the European eel (Anguilla anguilla L.). Aquat. Toxicol. 92
connected? Toxicol. Lett. 131 (1), 105–115. (4), 213–220.
Selvaraj, V., Tomblin, J., Armistead, M.Y., Murray, E., 2013. Selenium (sodium selenite) van der Wijst, M.G., Rots, M.G., 2015. Mitochondrial epigenetics: an overlooked layer of
causes cytotoxicity and apoptotic mediated cell death in PLHC-1 fish cell line through regulation? Trends Genet. 31 (7), 353–356.
DNA and mitochondrial membrane potential damage. Ecotoxicol. Environ. Saf. 87, Weinhouse, C., 2017. Mitochondrial-epigenetic crosstalk in environmental toxicology.
80–88. Toxicology 391, 5–17.
Shadel, G.S., Horvath, T.L., 2015. Mitochondrial ROS signaling in organismal home- Welch, A.J., Bedoya-Reina, O.C., Carretero-Paulet, L., Miller, W., Rode, K.D., Lindqvist,
ostasis. Cell 163 (3), 560–569. C., 2014. Polar bears exhibit genome-wide signatures of bioenergetic adaptation to
Shen, Y.-Y., Liang, L., Zhu, Z.-H., Zhou, W.-P., Irwin, D.M., Zhang, Y.-P., 2010. Adaptive life in the arctic environment. Genome Biol. Evol. 6 (2), 433–450.
evolution of energy metabolism genes and the origin of flight in bats. Proc. Natl. Xie, F., Koziar, S.A., Lampi, M.A., Dixon, D.G., Norwood, W.P., Borgmann, U., Huang,
Acad. Sci. 107 (19), 8666–8671. X.D., Greenberg, B.M., 2006. Assessment of the toxicity of mixtures of copper, 9, 10-
Shock, L.S., Thakkar, P.V., Peterson, E.J., Moran, R.G., Taylor, S.M., 2011. DNA me- phenanthrenequinone, and phenanthrene to Daphnia magna: evidence for a reactive
thyltransferase 1, cytosine methylation, and cytosine hydroxymethylation in mam- oxygen mechanism. Environ. Toxicol. Chem. 25 (2), 613–622.
malian mitochondria. Proc. Natl. Acad. Sci. 108 (9), 3630–3635. Yakes, F.M., Van Houten, B., Mitochondrial, 1997. damage is more extensive and persists
Shoshan-Barmatz, V., Ben-Hail, D., Admoni, L., Krelin, Y., Tripathi, S.S., 2015. The mi- longer than nuclear DNA damage in human cells following oxidative stress. Proc.
tochondrial voltage-dependent anion channel 1 in tumor cells. Bioch. Biophys. Acta Natl. Acad. Sci. 94 (2), 514–519.
(BBA)-Biomembr. 1848 (10), 2547–2575. Youle, R.J., Van Der Bliek, A.M., 2012. Mitochondrial fission, fusion, and stress. Science
Smith, R., Kelso, G., Blaikie, F., Porteous, C., Ledgerwood, E., Hughes, G., James, A., Ross, 337 (6098), 1062–1065.
M., Asin-Cayuela, J., Cocheme, H., 2003. Using Mitochondria-Targeted Molecules to

74

You might also like