Hostname: page-component-76fb5796d-x4r87 Total loading time: 0 Render date: 2024-04-27T17:28:17.141Z Has data issue: false hasContentIssue false

Crosstalk between mitochondrial metabolism and oxidoreductive homeostasis: a new perspective for understanding the effects of bioactive dietary compounds

Published online by Cambridge University Press:  16 October 2019

Mariangela Di Giacomo
Affiliation:
Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
Vincenzo Zara
Affiliation:
Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
Paolo Bergamo
Affiliation:
Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, Italy
Alessandra Ferramosca*
Affiliation:
Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
*
*Corresponding author: Alessandra Ferramosca, fax +39 0832 298698, email alessandra.ferramosca@unisalento.it
Rights & Permissions [Opens in a new window]

Abstract

Mitochondria play an important role in a number of fundamental cellular processes, including energy production, biosynthetic pathways and cellular oxidoreductive homeostasis (redox status), and their dysfunction can lead to numerous pathophysiological consequences. As the biochemical mechanisms orchestrating mitochondrial metabolism and redox homeostasis are functionally linked, mitochondria have been identified as a potential therapeutic target. Consequently, considerable effort has been made to evaluate the efficacy of natural compounds that modulate mitochondrial function. Molecules produced by plants (for example, polyphenols and isothiocyanates) have been shown to modulate mitochondrial metabolism/biogenesis and redox status; however, despite the existence of a functional link, few studies have considered the combined efficacy of these mitochondrial functions. The present review provides a complete overview of the molecular pathways involved in modulating mitochondrial metabolism/biogenesis and redox status. Crosstalk between these critical mechanisms is also discussed, whilst major data from the literature regarding their antioxidant abilities are described and critically analysed. We also provide a summary of recent evidence regarding the ability of several plant-derived compounds to target these mitochondrial functions. An in-depth understanding of the functional link between mitochondrial metabolism/biogenesis and redox status could facilitate the analysis of the biological effects of natural compounds as well as the development of new therapeutic approaches.

Type
Review Article
Copyright
© The Authors 2019

Introduction

Mitochondria play a critical role in the generation of metabolic energy in eukaryotic cells. These double membrane-enclosed organelles are characterised by the presence of enzymes that are involved in many distinct metabolic pathways (pyruvate and fatty acid oxidation and the Krebs cycle) and are responsible for producing the reducing equivalents (NADH and FADH2) required to generate ATP via oxidative phosphorylation (OXPHOS)(Reference Duchen1).

NADH and FADH2 are used in the electron transport chain (ETC), which is composed of four distinct multi-subunit complexes (I, II, III and IV) and two electron shuttle molecules (ubiquinone or coenzyme Q (CoQ) and cytochrome c). The ETC is responsible for transporting reducing equivalents from electron donors to oxygen molecules, ultimately forming water. The energy released from these oxidation/reduction reactions is used to drive ATP synthesis. This sophisticated mechanism, which requires strict coupling between electron transfer through respiratory chain complexes and the phosphorylation of ADP to ATP, provides the majority (80–90 %) of all fatty acid-derived energy.

In addition to their basic role in ATP synthesis, mitochondria are a major source of reactive oxygen species (ROS), which are key mediators of cellular physiology and pathology(Reference Galluzzi, Morselli and Kepp2Reference Galluzzi, Kepp and Trojel-Hansen4). Maintaining mitochondrial function is crucial, since perturbations can lead to negative consequences, such as impaired biomolecule synthesis, disrupted cellular osmolarity and cell death(Reference Zhou, Chen and Peng5,Reference Islam6) ; in fact, mitochondrial dysfunction has been associated with numerous human diseases, including metabolic, cardiovascular and neurodegenerative diseases, cancer, psychiatric disorders and ageing(Reference Bergman and Ben-Shachar7Reference Lunetti, Di Giacomo and Vergara13).

Some natural bioactive compounds offer attractive new therapeutic options, since they can target mitochondria. Thanks to their peculiar properties, these molecules are able to improve mitochondrial functionality and thus regulate the processes in which they are involved(Reference Forbes-Hernández, Giampieri and Gasparrini14). The present review summarises the most recent evidence regarding the ability of new bioactive compounds to target mitochondria and improve and/or restore their defective function, with a particular focus on their involvement in oxidative stress status. Since mitochondrial metabolic processes involve a number of proteins and protein complexes, the introductory paragraphs aim to provide the information necessary to understand the pathways via which these molecules elicit their biological effects, rather than a complete overview of the mechanisms that modulate mitochondrial function.

Nutritional regulation of mitochondrial function

Carbohydrates, lipids and proteins are not only substrates for metabolic bioenergetic pathways, but also modulate mitochondrial function. In fact, adequate nutrient levels are essential for mitochondrial function; an excessive supply (due to high-energy, high-carbohydrate or high-fat diets) or deficiency can be detrimental to mitochondria. Similarly, micronutrients such as vitamins and minerals are essential for mitochondrial function and biological activities, often affecting mitochondrial health as antioxidants, cofactors or coenzymes, or components of mitochondrial biogenesis and/or metabolic functions. A detailed description of how macro- or micronutrients modulate mitochondrial health and the molecular mechanisms involved is beyond the scope of this review because it has been recently and exhaustively analysed(Reference Vasam, Reid, Burelle, Morio, Penicaud and Rigoulet15,Reference Wesselink, Koekkoek and Grefte16) . The modulation of mitochondrial function and redox homeostasis by nutritional components is summarised in Table 1. The molecular targets influenced by nutritional factors will be described in the following paragraphs.

Table 1. Nutritional regulators of mitochondrial functions

OXPHOS, oxidative phosphorylation; SIRT3, sirtuin 3; ETC, electron transport chain; UCP2, uncoupling protein 2; AMPK, AMP-activated protein kinase; ACC, acetyl-CoA carboxylase; SOD2, superoxide dismutase 2.

Another interesting aspect of nutritional regulation is energy restriction, usually defined as a moderate (20–40 %) reduction in energy intake compared with ad libitum feeding, which can induce mitochondrial metabolic reprogramming by preserving oxidative capacity and decreasing oxidative damage(Reference Martin-Montalvo and de Cabo17). Therefore, we will discuss mitochondrial mechanisms that benefit from energy restriction.

Key regulators of mitochondrial function

Mitochondria are responsible for converting food-derived chemical energy into energy that cells can use (ATP). Adequate nutrient levels are essential for mitochondrial function, with the fatty acid oxidation pathway playing a major role in this form of energy production. When fatty acids enter cells via specific cell surface transporters, fatty acyl-CoA synthase (FACS) adds CoA groups to give rise to acyl-CoA, which is then converted into a long-chain acyl-carnitine by carnitine palmitoyltransferase 1 (CPT1). This modification allows acyl groups to be transported across the inner mitochondrial membrane by carnitine translocase, where long-chain acyl-carnitine is converted back into long-chain acyl-CoA by carnitine palmitoyltransferase 2 (CPT2). In the mitochondrial matrix, fatty acids are broken down to acetyl-CoA molecules(Reference Stefanovic-Racic, Perdomo and Mantell18) which enter the Krebs cycle, also known as the tricarboxylic acid (TCA) cycle.

Malonyl-CoA is the product of the acetyl coenzyme A carboxylase (ACC) reaction in fatty acid synthesis and is an important regulator of fatty acid oxidation, with its concentration determining the switch between fatty acid synthesis and oxidation(Reference Ferramosca, Savy and Zara19). AMP-activated protein kinase (AMPK) appears to be extremely important for this process since it modulates cellular energy balance.

In response to an increased AMP:ATP ratio, AMPK phosphorylation inhibits fatty acid biosynthesis by inhibiting ACC activity(Reference Park, Gammon and Knippers20), and thus reduces malonyl-CoA levels. As metabolic disorders can dysregulate fatty acid metabolism (elevated fatty acid synthesis or impaired fatty acid oxidation) and as AMPK plays a key role in regulating fatty acid synthesis and oxidation, this enzyme is considered an attractive target for managing metabolic disorders(Reference Srivastava, Pinkosky and Filippov21). Moreover, AMPK is involved in sirtuin 1 (SIRT1) activation. Sirtuins are a family of NAD+-dependent deacetylases involved in the regulation of many biological processes, including stress responses, metabolism, development and longevity. Like AMPK, SIRT1 is thought to regulate the physiological processes underlying energy restriction; Cohen et al.(Reference Cohen, Miller and Bitterman22) demonstrated that energy restriction induces SIRT1 expression, suggesting that AMPK could be an important link between sensing and adapting to energy restriction(Reference Cantó and Auwerx23). In fact, AMPK can regulate NAD+ levels to cause SIRT1 activation, which can alter the transcriptional activity of PPARγ coactivator-1α (PGC-1α).

PGC-1α belongs to a family of transcriptional coactivators that play a central role in regulating cellular metabolism(Reference Sugden, Caton and Holness24), including maintaining glucose, lipid and energy homeostasis(Reference Lin, Handschin and Spiegelman25) and oxidative metabolism (respiration and mitochondrial biogenesis). PGC-1α levels are low under physiological conditions, but increase in response to increased bioenergetic demands or metabolic alterations. PGC-1α directly interacts with, and coactivates mitochondrial regulators, such as nuclear respiratory factors (NRF) NRF1 and NRF2, which are involved in the transcription of several mitochondrial genes encoding ETC subunits and the mitochondrial transcription factor A (TFAM or mtTFA)(Reference Bouchez and Devin26).

Several studies have shown that sirtuin 3 (SIRT3) also modulates energy homeostasis by regulating mitochondrial ETC activity via interactions with mitochondrial respiratory complexes(Reference Lombard, Tishkoff and Bao27).

In conclusion, the AMPK–SIRT1–PGC-1α axis is a major signalling pathway that orchestrates mitochondrial function and dynamics in mammalian cells; therefore, its deregulation is associated with the onset of several neurological diseases, and it has been suggested as a pharmacological target to prevent or treat such diseases(Reference Valero28).

Modulation of reactive oxygen species yield in mitochondria

In the ETC, electrons (transported from reduced to oxidised subunits according to their redox potential and then tetravalently added to oxygen) can spontaneously undergo side reactions with oxygen to give rise to superoxide and a variety of other downstream ROS. The mitochondrial ETC is a major site of cellular ROS production(Reference Lombard, Tishkoff and Bao27), which is considered an inevitable consequence of oxidative ATP production. Since mitochondrial dysfunction is involved in several metabolic disorders(Reference Bhatti, Bhatti and Reddy29), ETC complexes I and III, which are the major sites of ROS production, could be considered therapeutic targets.

The ROS produced during cellular metabolism can be harmful or beneficial depending on the balance between their yield and the efficiency of antioxidant defences; thus, redox status is crucial for living organisms and has been preserved during evolution. The regulatory role of ROS in physiological processes has become apparent in the past two decades, with their production controlled by a complex network of intracellular signalling pathways and a complex defence system.

For example, various physiological stimuli can increase cellular PGC-1α levels alongside ROS yield, as PGC-1α can up-regulate mitochondrial biogenesis, respiratory capacity, OXPHOS and fatty acid β-oxidation. However, increasing evidence suggests that PGC-1α can also act as a powerful regulator of ROS removal by increasing the expression of the antioxidant enzymes, glutathione peroxidase (GPx) and SOD2 (also known as mitochondrial manganese-dependent superoxide dismutase or Mn-SOD)(Reference Austin and St-Pierre30). Conversely, decreased PGC-1α expression may reduce the expression of NRF-dependent metabolic and mitochondrial genes and contribute to ETC dysfunction and altered redox status.

Mitochondrial redox couples

In mitochondria, as in the cytosol, the redox state of the NADH/NAD+ and NADPH/NADP+ redox pairs are maintained independently as the nucleotides have different metabolic roles. Nicotinamide nucleotide transhydrogenase (NTT) is a mitochondrial NADP-reducing enzyme that catalyses the reduction of NADP+ (at the expense of NADH) and is coupled to H+ transport from the intermembrane space to the mitochondrial matrix. The NADH/NAD+ pair supports electron transfer in the ETC (via respiratory complex I) and the antioxidant system. In mitochondria, high NADH concentrations provide electrons for OXPHOS(Reference Karamanlidis, Lee and Garcia-Menendez31), whilst high NAD+ concentrations in the cytosol promote its role as a cofactor for oxidative reactions (i.e. the glyceraldehyde-3-phosphate dehydrogenase reaction in glycolysis). Notably, these molecules along with GSSG (glutathione disulfide)/GSH (reduced glutathione) and TrxSS (oxidised disulfide thioredoxin)/TrxSH2 (reduced thioredoxin) are the principal redox couples in redox signalling(Reference Jones and Sies32) and are interconnected. In particular, all components of the antioxidative defences, as well as GSSG and the GSH-dependent enzymes, glutathione reductase and GPx1 and GPx4 rely on NADPH as a common reductant for their oxidised forms. NNT maintains the NADPH pool by utilising TCA cycle-derived NADH to reduce NADP+ to NADPH(Reference Rydström33). Other mitochondrial NADPH-regenerating enzymes are the NADP+-dependent isocitrate dehydrogenase and malic enzyme(Reference Sazanov and Jackson34,Reference Jo, Son and Koh35) . NNT activity mainly depends on membrane potential and the activity of complexes I, III and IV, which control the level of mitochondrial NADH, the substrate for the transhydrogenase reaction. Moreover, GSH redox status is probably regulated by the availability of catabolites oxidised in the TCA cycle to generate NADH(Reference Garcia, Han and Sancheti36). Mitochondrial matrix antioxidant defences are very effective at scavenging ROS species generated by the respiratory chain, particularly superoxide anions and H2O2 (Reference Treberg, Quinlan and Brand37,Reference Aon, Stanley and Sivakumaran38) . Thus, it has been suggested that mitochondria are a sink rather than a source of cellular ROS under physiological conditions(Reference Starkov39).

Key mitochondrial regulators of redox status

Mitochondrial respiratory chain

The inner mitochondrial membrane respiratory chain transfers electrons from reducing equivalents to oxygen to generate proton-motive forces that are the primary energy source for cellular ATP synthesis. Recent evidence suggests that the reversible redox modification of protein-thiols is an important response to changes in the cellular redox environment; conversely, ROS generation by respiratory chain complexes may affect the mitochondrial redox balance by reversibly or irreversibly thiol-modifying specific target proteins involved in redox signalling and pathophysiological processes. Moreover, the thiol-based modifications (for example, S-glutathionylation and S-nitrosylation) of mitochondrial respiratory chain complex subunits may regulate respiratory activity(Reference Dröse, Brandt and Wittig40).

Uncoupling proteins

As mentioned above, the mitochondrial ETC is a major site of ROS production, with ROS yield depending on the redox state of its respiratory chain complexes. In particular, increased mitochondrial transmembrane potential (ΔΨm), which determines the proton-motive force along with the proton gradient (ΔpH), results in aberrant electron migration in the ETC and elevated ROS production. Under these conditions, the probability of electrons escaping the respiratory chain and forming superoxide anions increases(Reference Brand, Affourtit and Esteves41). The proton-motive force (ΔΨm or ΔpH) and subsequent ROS production can be reduced either by decreasing substrate oxidation (electron influx) or increasing the consumption of proton-motive forces across the inner mitochondrial membrane.

In 1997, it was proposed that uncoupling proteins (UCP) can modulate the mitochondrial generation of superoxide anions via their uncoupling activity. The expression of UCP isoforms 1–3 may be stimulated by the increased generation of mitochondrial superoxide anions(Reference Echtay, Roussel and St-Pierre42), causing protons to leak from the intermembrane space into the matrix (bypassing ATP synthase in OXPHOS) and decrease the ΔΨm. As a high ΔΨm generates ROS(Reference Korshunov, Skulachev and Starkov43), UCP-mediated proton gradient dissipation may greatly reduce ROS production in a feedback manner by lowering ΔΨm (Reference Miwa and Brand44).

In conclusion, the correlation between ΔΨm and ROS yield(Reference Korshunov, Skulachev and Starkov43) may make uncoupling an acute (energetically costly) mechanism for modulating redox homeostasis by reducing ROS production(Reference Mailloux and Harper45).

AMP-activated protein kinase–sirtuin 1–PPARγ coactivator-1α axis

The transcriptional coactivator PGC-1α is known to regulate mitochondrial biogenesis(Reference Puigserver and Spiegelman46), and can induce ROS removal by up-regulating UCP2 and UCP3 expression(Reference St-Pierre, Lin and Krauss47). Moreover, its ability to regulate ROS homeostasis was confirmed by its role in activating antioxidant enzyme expression (namely GPx1 and SOD2)(Reference St-Pierre, Drori and Uldry48). Furthermore, PGC-1α activation may up-regulate antioxidant enzyme expression (catalase (CAT) or SOD) by non-canonically activating AMPK via mitochondrial ROS(Reference Rabinovitch, Samborska and Faubert49).

A substantial body of evidence has suggested that SIRT1, like AMPK, responds to variations in nutrient availability, and its regulation has been attributed to changes in NAD+ abundance (the NAD+:NADH ratio). It has been shown that the SIRT1 protein is involved in mitochondrial adaptation to redox alterations(Reference Cantó, Houtkooper and Pirinen50). The cytoprotective activity of SIRT3 could underlie its increased expression in tissues with high metabolism, which are, consequently, more exposed to the potentially damaging effects of mitochondrial ROS(Reference Jin, Galonek and Israelian51). Both SIRT1 and SIRT3 were found to regulate PGC-1α; SIRT1 deacetylates and increases the transcriptional activity of PGC-1α(Reference Nemoto, Fergusson and Finkel52,Reference Kong, Wang and Xue53) , whilst SIRT3 is an important regulator of mitochondrial metabolism that plays a role in adaptation to metabolic stress (for example, fasting or energy restriction). In fact, SIRT3 activation by energy restriction has been shown to reduce ROS levels by activating mitochondrial SOD2(Reference Sundaresan, Gupta and Kim54,Reference Qiu, Brown and Hirschey55) or deacetylating or activating mitochondrial isocitrate dehydrogenase 2 (IDH2), which catalyses the conversion of NADP+ to NADPH and increases the mitochondrial GSH:GSSG ratio(Reference Someya, Yu and Hallows56). Thus, sirtuins are classified as vitagenes, a group of genes that preserve cellular homeostasis under stressful conditions(Reference Calabrese, Cornelius and Dinkova-Kostova57).

Nuclear factor erythroid-derived 2-like

The activation of nuclear factor erythroid-derived 2-like (Nrf2) signalling plays a pivotal role in cellular protection and adaptation to external stressors, as well as a number of cytoprotective molecules produced by plants to protect themselves from microbial infection and other environmental conditions. The transcription of some vitagenes, such as haeme oxygenase (HO-1) and thioredoxin reductase (TrxR), is activated via the Nrf2 pathway. Under basal conditions, Kelch-like ECH-associated protein 1 (Keap1) prevents Nrf2 from binding to the antioxidant responsive element (ARE), thereby preventing its nuclear translocation and facilitating the degradation of Nrf2-Keap1 by the proteasome. The cysteine residues of Keap1 are susceptible to oxidative modification; thus, it can act as a redox sensor(Reference Taguchi, Motohashi and Yamamoto58). Nrf2 inducers can therefore improve the activity of cellular antioxidant/detoxifying defences via their mild pro-oxidant activity (eustress). In fact, when ROS levels are low (physiological conditions), they can activate ‘oxidative eustress’(Reference Sarsour, Kalen and Goswami59) which underlies their function as second messengers in several biological and physiological processes(Reference Finkel60). This process is distinct from excessive ROS load (‘oxidative stress’), which can damage cells and organs(Reference Sies, Berndt and Jones61). Under mild stress conditions, ROS can oxidise the cysteine residues of Keap1, thereby allowing Nrf2 to translocate into the nucleus where it activates the transcription of antioxidant/detoxifying enzymes (for example, GSR, GPx, CAT, HO-1 and TrxR; phase II enzymes) that protect cells against cytotoxic and oxidative damage(Reference Kensler, Wakabayashi and Biswal62).

The ability of a molecule to induce opposite biological effects depending on its dose (beneficial at low doses and toxic at high doses) is named hormesis(Reference Calabrese, Cornelius and Dinkova-Kostova63). The activation of the Nrf2 pathway plays a key role in protecting cells from external stressors and its physiologically relevant hormetic (dose-dependent) effects may prevent or mitigate chronic diseases by activating adaptive stress response signalling pathways(Reference Trewavas and Stewart64). Moreover, Nrf2 also modulates the expression of enzymes responsible for NADPH synthesis (i.e. malic enzyme 1 (ME1), isocitrate dehydrogenase 1 (IDH1), glucose-6-phosphate dehydrogenase (G6PD) and 6-phosphogluconate dehydrogenase (6PGD))(Reference Holmström, Kostov and Dinkova-Kostova65), which are involved in the biosynthesis and maintenance of the main intracellular antioxidant, GSH.

Functional link between Nrf2 and mitochondrial physiology

In addition to its key role in redox homeostasis, the Nrf2 pathway is linked to the AMPK–SIRT1–PGC-1α axis. Moreover, Nrf2 activation can sustain mitochondrial membrane potential (NADPH, NADH and FADH2 regeneration) and biogenesis (PGC-1α)(Reference Dinkova-Kostova and Abramov66). HO-1, an important enzyme in antioxidant defence and cellular metabolism(Reference Wegiel, Nemeth and Correa-Costa67), is also regulated by Nrf2 activation and can produce carbon monoxide (CO) during the metabolic conversion of haem to biliverdin(Reference Ryter, Alam and Choi68) to stimulate mitochondrial biogenesis(Reference Dinkova-Kostova and Abramov66,Reference Wegiel, Nemeth and Correa-Costa67) . Furthermore, a study demonstrating that Nrf2 acetylation promotes its binding to antioxidant responsive element (ARE)(Reference Suliman, Carraway and Ali69) paved the way for studies supporting the involvement of Nrf2 in the antioxidant activity of SIRT3 and SIRT6(Reference Piantadosi, Carraway and Babiker70Reference Yu, Sun and Song75).

Crosstalk between Nrf2 and mitochondrial homeostasis has been indicated by studies showing that the antidiabetic drug metformin can activate Nrf2 in an AMPK-dependent manner(Reference Ashabi, Khalaj and Khodagholi76) and that SRT1720, a synthetic SIRT1 agonist, can elicit antioxidant/anti-inflammatory effects via the AMPK/Nrf2 pathway in vivo (Reference Wang, Shang and Zhang77). Additionally, under oxidative stress conditions the Nrf2 pathway can up-regulate UCP3 to counteract superoxide production by increasing proton conduction across the inner mitochondrial membrane(Reference Anedda, Lopez-Bernardo and Acosta-Iborra78).

Thus, it can be concluded that Nrf2 activity is linked with many aspects of mitochondrial physiology (mitochondrial biogenesis, fatty acid oxidation, respiration and ATP production; Fig. 1) in addition to its recognised role in redox homeostasis, as detailed in a recent review(Reference Holmström, Kostov and Dinkova-Kostova65).

Fig. 1. Pivotal role of nuclear factor erythroid-derived 2-like (Nrf2) in the regulation of mitochondrial pathways. In addition to its well-known role in redox homeostasis, Nrf2 is involved in the regulation of many aspects of mitochondrial metabolism, such as biogenesis, fatty acid oxidation, oxidative phosphorylation (OXPHOS) and redox homeostasis. The table describes the major players and their cellular location during these processes. AMPK, AMP-activated protein kinase; CAT, catalase; GPx, glutathione peroxidase; mROS, mitochondrial reactive oxygen species; NRF, nuclear respiratory factor; PGC-1α, PPARγ coactivator-1α; ROS, reactive oxygen species; SIRT1, sirtuin 1; SIRT3, sirtuin 3; SOD, superoxide dismutase; SOD2, superoxide dismutase 2; TFAM, mitochondrial transcription factor A; UCP, uncoupling proteins. For a colour figure, see the online version of the paper.

Mechanisms underlying the ability of polyphenols to modulate redox status

ROS function as ‘redox messengers’ under physiological conditions; however, excess ROS can trigger cell injury and oxidative stress, which can damage cellular structures (protein, lipid and DNA) and has been associated with the pathogenesis of human diseases(Reference Duchen1) and ageing(Reference Lesnefsky and Hoppel79). Due to the noxious effects of oxidative stress, cells possess a complex mechanism that fine tunes oxido-reductive homeostasis (redox status) and mitochondrial metabolism to prevent oxidative injury, which could disrupt mitochondrial integrity, impair the ETC and cause mitochondrial DNA damage.

As mentioned above, mitochondria have been identified as the main site of cellular superoxide anion radical production and are the major target of oxidative stress. Therefore, it has been suggested that the dietary consumption of some secondary metabolites could modulate redox status. Among the plethora of bioactive compounds, polyphenols are known to exert intracellular antioxidant effects via both direct and indirect mechanisms (Fig. 2).

Fig. 2. Ability of bioactive compounds to modulate redox status. Several polyphenolic molecules, such as resveratrol, curcumin, quercetin and genistein, can affect redox homeostasis by directly and indirectly exerting antioxidant effects. The direct mechanisms consist of reactive oxygen species scavenging and metal chelation, whilst the indirect mechanisms include activating nuclear factor erythroid-derived 2-like (Nrf2) signalling, regulating inner mitochondrial membrane potential via uncoupling effects (up-regulating uncoupling protein 2 (UCP2) expression), and modulating radical species production via electron transport chain (ETC) complexes. The table describes the different bioactive compounds, where they originate, and their effects on mitochondrial metabolism. AMPK, AMP-activated protein kinase; EGCG, epigallocatechin gallate; MMP, matrix metalloproteinase; OXPHOS, oxidative phosphorylation; PGC-1α, PPARγ coactivator-1α; SIRT1, sirtuin 1. For a colour figure, see the online version of the paper.

Direct antioxidant activity of polyphenols

Reactive oxygen species scavenging

The direct antioxidant activity of polyphenols involves their ability to scavenge mitochondrial ROS or chelate transition metals (for example, Cu and Fe) involved in the formation of free radicals (i.e. superoxide and hydroxyl) via the Fenton reaction.

Research into the bioactivity of quercetin has shown that the phytochemical can affect mitochondrial redox parameters by acting upon mitochondria directly or indirectly. These effects have been attributed to its chemical structure; however, its ability to control mitochondrial respiratory chain function is still under investigation. Some have reported that the enolic 3-OH group in quercetin (C-ring) is responsible for enhancing its antioxidant activity. Additionally, this 3-OH group seems to exhibit the ability to chelate Fe2+, reducing its availability to react with H2O2 via the Fenton reaction. Moreover, it has been reported that quercetin can prevent methylmercury (MeHg) or mercuric chloride (HgCl2) from decreasing reduced GSH levels in mitochondria. Interestingly, quercetin exhibits a more pronounced effect than CAT on decreased hydroperoxide formation levels in mitochondria exposed to MeHg or HgCl2 (Reference Franco, Braga and Stringari80). Thus, quercetin can protect mitochondria against mercurials via a mechanism that involves antioxidant effects against H2O2, since CAT, a H2O2-consuming enzyme, is similarly effective at protecting mitochondria from such agents.

Others have investigated an alternative mechanism via which quercetin directly exerts antioxidant effects on mitochondria(Reference Lagoa, Graziani and Lopez-Sanchez81), demonstrating that quercetin dose-dependently (1–10 μM) inhibits H2O2 generation in intact mitochondria isolated from rat brains and hearts without interfering with O2 consumption. H2O2 is produced in mitochondria by SOD2, which is located in the mitochondrial matrix and converts superoxide anions into H2O2. SOD2 is a crucial determinant of energy homeostasis in mitochondria that acts not only as an antioxidant enzyme but more importantly as a central hub of redox signalling via H2O2 production(Reference Zou, Ratti and O’Brien82). Interestingly, SOD2 overexpression adversely affects mitochondrial oxidative metabolism and greatly stimulates glycolysis(Reference Lunetti, Di Giacomo and Vergara13).

Metal chelation

As mentioned previously, hydroxyl radicals can also be generated via reactions catalysed by redox-active transition metals, such as Cu and Fe. In mitochondria, free Fe can occur under conditions that increase superoxide production and affect the [4Fe–4S] cluster of aconitase and NADH-ubiquinone dehydrogenase. Some polyphenols (flavonoids such as baicalein, quercetin and myricetin, and non-flavonoids such as gallic, 2,3-dihydroxybenzoic and protocatechuic acids) have been shown to chelate Fe and Cu ions particularly well, rendering them unable to participate in reactions that generate free radicals(Reference Sandoval-Acuña, Ferreira and Speisky83). Metal chelation may reduce the pro-oxidant capacity of metal ions; however, polyphenols can also act as pro-oxidants by chelating metals in a way that maintains or increases their catalytic activity(Reference Decker84). These radicals are stabilised by the delocalisation of unpaired electrons around aromatic rings, enabling them to display pro-oxidant activities(Reference Galati and O’Brien85).

Nevertheless, the ROS-scavenging and metal-chelating abilities of polyphenols have only been demonstrated in silico or in vitro (Reference Bors and Michel86); therefore, these results may not necessarily translate to in vivo systems, particularly since these biological effects are highly dependent on the dose reaching the target cell/tissue. Due to the low intrinsic activity of polyphenols (poor absorption, high metabolism or rapid elimination)(Reference Crozier, Jaganath and Clifford87,Reference Chow and Hakim88) , their concentration in vivo is low compared with that of endogenous antioxidants (for example, GSH). In addition, data have indicated that gut microflora may play a major role in their biotransformation from native phytochemicals into more bioavailable/active metabolites(Reference Czank, Cassidy and Zhang89,Reference Espín, González-Sarrías and Tomás-Barberán90) , further reducing the reliability of extrapolating their biological activity from in vitro to in vivo models.

Indirect antioxidant activity of polyphenols implies crosstalk with mitochondrial metabolism

The second mechanism of polyphenol antioxidant activity involves inhibiting ROS yield and stimulating ROS-removing enzymes. The link between mitochondrial metabolism and redox homeostasis is based on the activity of redox pairs (for example, NAD+/NADH, NADP+/NADPH), mitochondrial respiratory chain enzymes/proteins (for example, complex I and III, UCP and sirtuins), and transcriptional regulators such as Nrf2 and PGC-1α, which form a complex network that modulates crucial physiological functions.

The link between redox homeostasis and mitochondrial metabolism is further supported by the metabolic profiling data of healthy human subjects, showing that receiving dietary intervention with broccoli (source of glucoraphanin, a sulforaphane precursor) improved the integration of fatty acid oxidation and TCA cycle activity(Reference Armah, Traka and Dainty91). Since many diseases have an oxidative stress component, the Nrf2-dependent up-regulation of cytoprotective genes is considered a therapeutic target(Reference Cuadrado, Manda and Hassan92).

Complex I inhibition

Complexes I and III are mainly responsible for superoxide anion yield in mitochondria; therefore, their inhibition affects redox status homeostasis. Recent in vitro and ex vivo studies have indicated that polyphenols can modulate mitochondrial superoxide production via ETC complexes(Reference Lagoa, Graziani and Lopez-Sanchez81,Reference Carrasco-Pozo, Gotteland and Speisky93,Reference Sandoval-Acuña, Lopez-Alarcón and Aliaga94) . Thus, quercetin may suppress H2O2 production by modulating complex I activity(Reference Murphy95).

CoQ pre-treatment reduced the inhibitory effect of quercetin on complex I, suggesting that quercetin competitively binds the quinone-binding site of complex I due to structural similarity with the quinone moiety of CoQ. It has been demonstrated that quercetin (10 μM) stimulates complex I activity in a very similar manner to CoQ in an experimental model using mitochondria isolated from rat duodenum epithelium(Reference Crozier, Jaganath and Clifford87). Importantly, complex I inhibition could lead to electron leakage from the ETC and the increased generation of superoxide anions, which are H2O2 precursors(Reference Halliwell96).

Activation of Nrf2-mediated antioxidant/detoxifying enzymes

An increasingly recognised mechanism by which some polyphenols can exert antioxidant effects in vivo is by up-regulating antioxidant enzyme systems. Moreover, some polyphenols can induce certain phase I and II enzymes that detoxify potentially pro-oxidant xenobiotics(Reference Tsuji, Stephenson and Wade97).

Numerous natural products originating from plants, including isothiocyanates (for example, phenethyl isothiocyanate and sulforaphane), alkaloids (for example, berberine and betanin), flavonoids (for example, epigallocatechin gallate and quercetin), stilbenes (for example, resveratrol and piceatannol), diferuloylmethanes (for example, curcumin and caffeic acid phenethyl ester) and organosulfur compounds (for example, allicin and diallyl trisulfide), have been reported to activate antioxidant/detoxifying defences via an indirect mechanism (Nrf2)(Reference Iranshahy, Iranshahi and Abtahi98). Despite their structural diversity, these molecules share the ability to activate this molecular mechanism due to their pro-oxidant and electrophilic properties(Reference Surh99). At doses ingested by humans, these phytochemicals can induce adaptive responses by inducing Nrf2-driven antioxidant gene expression(Reference Son, Camandola and Mattson100), which improves defensive mechanisms to better protect cells and organs against further toxic insults. Like the hormetic mechanism activated by Nrf2, the pro-oxidant effects exhibited by these chemopreventative phytochemicals are not unexpected.

The ability of polyphenols (for example, stilbenes: resveratrol; and flavonoids: epigallocatechin gallate) and curcumin to activate the Nrf2 pathway has been investigated thoroughly(Reference Scapagnini, Vasto and Abraham101Reference Trujillo, Granados-Castro and Zazueta104). In particular, the ability of quercetin to improve antioxidant defences and bioenergetic parameters in mitochondria was demonstrated in vivo using experimental models consisting of the brain, heart, gastric and liver tissues of experimental animals. Quercetin was reported to exert antioxidant effects by decreasing lipid peroxidation and protein carbonylation (important consequences of increased oxidative stress) and preventing GSH oxidation(Reference Waseem and Parvez105). This flavonoid also protects mitochondria by activating Nrf2 pathways in cultured cells and animal tissues(Reference Ji, Sheng and Zheng106), and attenuates hepatic lipid accumulation in mice fed a high-fat diet(Reference Jung, Cho and Ahn107). Moreover, dietary quercetin supplementation (100 mg/kg administered orally for 90 d) up-regulated SOD2 and GPx activity and restored GSH levels in the liver mitochondria of ethanol-treated rats, with the authors observing that quercetin alleviated the effects of ethanol on mitochondrial ROS production and lipid peroxidation(Reference Tang, Gao and Xing108).

Similar effects have been suggested for resveratrol, which exerts antioxidant activity by increasing the expression of mitochondrial proteins or ROS-scavenging enzymes. In fact, dietary supplementation with resveratrol appeared to improve mitochondrial function in mice, with those treated with resveratrol also tolerating oxidative stress induced by exposure to various chemical agents better than untreated mice(Reference Kovacic and Somanathan109). This stilbene decreases mitochondrial ROS levels and inhibits lipid peroxidation by scavenging ROS (superoxide anions, H2O2 and hydroxyl radicals) and replenishing GSH(Reference Guha, Dey and Dhyani110). Moreover, another study revealed that resveratrol can dose-dependently activate antioxidant defences by activating the Nrf2 pathway(Reference Ungvari, Bagi and Feher111).

Several studies have shown that curcumin exerts significant antioxidant activities by ameliorating lipid peroxidation and oxidative stress in different tissues(Reference Ak and Gülçin112). Moreover, curcumin exerts its antioxidant properties via both direct and indirect mechanisms. This polyphenol effectively scavenges free radicals, such as hydroxyl radicals, superoxide anions, NO, H2O2 and peroxynitrite(Reference Barzegar and Moosavi-Movahedi113,Reference Derochette, Franck and Mouithys-Mickalad114) , and is able to up-regulate cytoprotective cell responses by modulating the expression of genes encoding antioxidant proteins, such as SOD, CAT and HO-1, or proteins that replenish the glutathione pool, such as GR, GPx and GST(Reference Reyes-Fermín, González-Reyes and Tarco-Álvarez115). Improved mitochondrial function has also been associated with preventing reduced aconitase activity, a marker of oxidative stress(Reference Granados-Castro, Rodríguez-Rangel and Fernández-Rojas116). Curcumin exerts cytoprotective effects against toxic compounds that can generate ROS and cause lipid peroxidation and DNA damage, including potassium dichromate (K2Cr2O7). Curcumin prevents K2Cr2O7-induced decreases in body weight, increases liver weight and the liver:body ratio, and exerts protective effects against oxidative damage in liver tissue by preventing K2Cr2O7-induced decreases in hepatic antioxidant enzyme levels. These effects appear to be mediated by its protective effects in mitochondria. Studies on isolated organelles have shown that curcumin reduces mitochondrial dysfunction by preventing K2Cr2O7 from reducing complex I activity and opening the mitochondrial permeability transition pore (mPTP), thus inhibiting mitochondria-induced apoptosis(Reference García-Niño, Tapia and Zazueta117).

By investigating antioxidant efficacy, lycopene, a lipid–soluble carotenoid compound, was found to exert a strong protective effect against brain damage. Lycopene pre-treatment was shown to protect SH–SY5Y neuroblastoma cells against H2O2-induced death by inhibiting apoptosis and improving the activity of Nrf2-activated antioxidant enzymes (SOD and CAT). Additionally, lycopene prevented H2O2-induced mitochondrial dysfunction by mitochondrial permeability transition pore (mPTP) opening and attenuating the decline in mitochondrial membrane potential(Reference Feng, Luo and Zhang118).

Sulforaphane activates Nrf2, which induces the expression of cytoprotective genes that play key roles in cellular defence mechanisms, including redox status and detoxification. Both its high bioavailability (higher than polyphenols) and significant ability to induce Nrf2 contribute towards the therapeutic potential of sulforaphane-yielding supplements(Reference Houghton, Fassett and Coombes119).

Nrf2–sirtuins

The association between the health benefits of several polyphenolic compounds, such as resveratrol, fisetin and quercetin, and their ability to activate SIRT1, was recently reviewed(Reference Bai, Yao and Ma120) and their effects on cancer cells have been associated with AMPK activation(Reference Giovannini and Bianchi121).

Resveratrol reduces mitochondrial ROS generation by increasing SIRT3 levels in the mitochondria of endothelial cells, in turn increasing complex I activity and ATP synthesis by up-regulating the mitochondrial proteins ATP6, CO1, Cytb, ND2 and ND5(Reference Zhou, Chen and Zeng122). Moreover, resveratrol can up-regulate the expression of the scavenging enzymes GPx, CAT(Reference Ungvari, Orosz and Rivera123) and SOD2 in endothelial cells in a SIRT1-dependent manner(Reference Ungvari, Labinskyy and Mukhopadhyay124). Resveratrol treatment has also been shown to stimulate SIRT1 and AMPK activity in vivo, both of which influence redox homeostasis in multiple tissues(Reference Moskaug, Carlsen and Myhrstad125,Reference Sarubbo, Esteban and Miralles126) .

Nrf2–PPARγ coactivator-1α

The effects of a phenolic acid on Nrf2–PGC-1α have been studied; in particular, high dietary hydroxytyrosol intake appears to increase PGC-1α expression, indirectly improving mitochondrial function by interacting with and enhancing enzymes that protect cells against oxidative damage due to excessive ROS levels. In fact, mice lacking PGC-1α suffer greater drug-induced oxidative damage in the brain and neural tissues, suggesting that hydroxytyrosol exerts cytoprotective effects by increasing PGC-1α levels and improving mitochondrial and cellular ROS-related functions(Reference St-Pierre, Drori and Uldry48). Similarly, quercetin was recently reported to enhance hepatic mitochondrial oxidative metabolism and biogenesis (PGC-1α) by activating Nrf-2/HO-1(Reference Kim, Kwon and Choe127).

Nrf2–uncoupling proteins

As mentioned previously, Nrf2 activation may be triggered by mild stress conditions via the oxidation of Keap1 cysteine residues; however, Nrf2 may also be activated in an AMPK-dependent manner by the flavonoid-related compound xanthohumol in vitro or the alkaloid berberine in vivo (Reference Mo, Wang and Zhang128,Reference Zimmermann, Baldinger and Mayerhofer129) . UCP can modulate mitochondrial superoxide anion generation by decreasing mitochondrial inner membrane potential via their uncoupling activity. The uncoupling effect exerted by flavonoids has been attributed to their weakly acidic and highly lipophilic nature. Flavonoids can be protonated on the low-pH external side of the inner mitochondrial membrane, but when they pass through the lipid layer they are deprotonated in the high-pH mitochondrial matrix milieu; thus, the proton gradient across the inner mitochondrial membrane is dissipated. However, a more recent study proposed that the uncoupling effect exerted by the isoflavone genistein (1 µM) might be mediated by up-regulated UCP2 expression(Reference Nadal-Serrano, Pons and Sastre-Serra130). The mechanisms that induce OXPHOS uncoupling remain unknown; however, several studies have suggested that the effect could be associated with decreased ROS formation via the ETC(Reference Modrianský and Gabrielová131). Thus, the uncoupling effect of some polyphenols could be viewed as an ROS scavenging-independent mechanism via which they exert their antioxidant activity.

Summary

Mitochondria play pivotal roles in numerous cellular processes and are considered the main source of ROS whilst simultaneously being a major target for these potentially noxious molecules. In fact, excess oxidants can cause oxidative stress which is known to play a causal role in many diseases, such as cancer, metabolic, degenerative and hyper-proliferative diseases, as well as ageing(Reference Brand and Nicholls132,Reference Demine, Reddy and Renard133) .

Several mechanisms are involved in modulating mitochondrial function, with numerous proteins and protein complexes in specific pathways allowing crosstalk between mitochondrial metabolism and oxidoreductive homeostasis. Recent studies have demonstrated that many bioactive dietary compounds, such as polyphenols and carotenoids, could be used alone or in combination to prevent and control disease development(Reference Pandey and Rizvi134). Most of these molecules can target mitochondria to improve and/or restore their function by indirectly modulating redox status. Therefore, we believe that further investigation and an improved understanding of the molecular and biochemical mechanisms underlying the action of these natural compounds are necessary to develop new therapeutic approaches that improve mitochondrial function and restore redox homeostasis.

Acknowledgements

The present review received no specific grant from any funding agency, commercial or not-for-profit sectors.

P. B., M. D. G. and A. F. drafted the manuscript; A. F., P. B. and V. Z. made critical revisions.

There are no conflicts of interest.

Footnotes

These authors contributed equally to the present review.

References

Duchen, MR (2004) Mitochondria in health and disease: perspectives on a new mitochondrial biology. Mol Aspects Med 25, 365451.CrossRefGoogle ScholarPubMed
Galluzzi, L, Morselli, E, Kepp, O, et al. (2011) Mitochondrial liaisons of p53. Antioxid Redox Signal 15, 16911714.CrossRefGoogle ScholarPubMed
Galluzzi, L, Kepp, O & Kroemer, G (2012) Mitochondria: master regulators of danger signaling. Nat Rev Mol Cell Biol 13, 780788.CrossRefGoogle Scholar
Galluzzi, L, Kepp, O, Trojel-Hansen, C, et al. (2012) Mitochondrial control of cellular life, stress, and death. Circ Res 111, 11981207.CrossRefGoogle Scholar
Zhou, L, Chen, P, Peng, Y, et al. (2016) Role of oxidative stress in the neurocognitive dysfunction of obstructive sleep apnea syndrome. Oxid Med Cell Longev 2016, 9626831.CrossRefGoogle ScholarPubMed
Islam, MT (2017) Oxidative stress and mitochondrial dysfunction linked neurodegenerative disorders. Neurol Res 39, 7382.CrossRefGoogle ScholarPubMed
Bergman, O & Ben-Shachar, D (2016) Mitochondrial oxidative phosphorylation system (OXPHOS) deficits in schizophrenia: possible interactions with cellular processes. Can J Psychiatry 61, 457469.CrossRefGoogle ScholarPubMed
Cadonic, C, Sabbir, MG & Albensi, BC (2016) Mechanisms of mitochondrial dysfunction in Alzheimer’s disease. Mol Neurobiol 53, 60786090.CrossRefGoogle ScholarPubMed
Zulian, A, Schiavone, M, Giorgio, V, et al. (2016) Forty years later: mitochondria as therapeutic targets in muscle diseases. Pharmacol Res 113, 563573.CrossRefGoogle ScholarPubMed
Ferramosca, A, Conte, A & Zara, V (2015) Krill oil ameliorates mitochondrial dysfunctions in rats treated with high-fat diet. Biomed Res Int 2015, 645984.CrossRefGoogle ScholarPubMed
Ferramosca, A, Conte, A, Guerra, F, et al. (2016) Metabolites from invasive pests inhibit mitochondrial complex II: a potential strategy for the treatment of human ovarian carcinoma? Biochem Biophys Res Commun 473, 11331138.CrossRefGoogle ScholarPubMed
Hu, H, Tan, CC, Tan, L, et al. (2017) A mitocentric view of Alzheimer’s disease. Mol Neurobiol 54, 60466060.CrossRefGoogle ScholarPubMed
Lunetti, P, Di Giacomo, M, Vergara, D, et al. (2019) Metabolic reprogramming in breast cancer results in distinct mitochondrial bioenergetics between luminal and basal subtypes. FEBS J 286, 688709.CrossRefGoogle ScholarPubMed
Forbes-Hernández, TY, Giampieri, F, Gasparrini, M, et al. (2014) The effects of bioactive compounds from plant foods on mitochondrial function: a focus on apoptotic mechanisms. Food Chem Toxicol 68, 154182.CrossRefGoogle ScholarPubMed
Vasam, G, Reid, K, Burelle, Y, et al. (2019) Nutritional regulation of mitochondrial function. In Mitochondria in Obesity and Type 2 Diabetes Comprehensive Review on Mitochondrial Functioning and Involvement in Metabolic Diseases, chapter 4, pp. 93126 [Morio, B, Penicaud, L and Rigoulet, M, editors]. Cambridge, MA: Academic Press.CrossRefGoogle Scholar
Wesselink, E, Koekkoek, WAC, Grefte, S, et al. (2019) Feeding mitochondria: potential role of nutritional components to improve critical illness convalescence. Clin Nutr 38, 982995.CrossRefGoogle ScholarPubMed
Martin-Montalvo, A & de Cabo, R (2014) Mitochondrial metabolic reprogramming induced by calorie restriction. Antioxid Redox Signal 19, 310320.CrossRefGoogle Scholar
Stefanovic-Racic, M, Perdomo, G, Mantell, BS, et al. (2008) A moderate increase in carnitine palmitoyltransferase 1a activity is sufficient to substantially reduce hepatic triglyceride levels. Am J Physiol Endocrinol Metab 294, E969E977.CrossRefGoogle ScholarPubMed
Ferramosca, A, Savy, V & Zara, V (2008) Olive oil increases the hepatic triacylglycerol content in mice by a distinct influence on the synthesis and oxidation of fatty acids. Biosci Biotechnol Biochem 72, 6269.CrossRefGoogle ScholarPubMed
Park, SH, Gammon, SR, Knippers, JD, et al. (2002) Phosphorylation-activity relationships of AMPK and acetyl-CoA carboxylase in muscle. J Appl Physiol (1985) 92, 24752482.CrossRefGoogle ScholarPubMed
Srivastava, RA, Pinkosky, SL, Filippov, S et al. (2012) AMP-activated protein kinase: an emerging drug target to regulate imbalances in lipid and carbohydrate metabolism to treat cardio-metabolic diseases. J Lipid Res 53, 24902514.CrossRefGoogle ScholarPubMed
Cohen, HY, Miller, C, Bitterman, KJ, et al. (2004) Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science 305, 390392.CrossRefGoogle ScholarPubMed
Cantó, C & Auwerx, J (2011) Calorie restriction: is AMPK as a key sensor and effector? Physiology (Bethesda) 26, 214224.Google ScholarPubMed
Sugden, MC, Caton, PW & Holness, MJ (2010) PPAR control: it’s SIRTainly as easy as PGC. J Endocrinol 204, 93104.CrossRefGoogle ScholarPubMed
Lin, J, Handschin, C & Spiegelman, M (2005) Metabolic control through the PGC-1 family of transcription coactivators. Cell Metab 1, 361370.CrossRefGoogle ScholarPubMed
Bouchez, C & Devin, A (2019) Mitochondrial biogenesis and mitochondrial reactive oxygen species (ROS): a complex relationship regulated by the cAMP/PKA signaling pathway. Cells 8, E287.CrossRefGoogle ScholarPubMed
Lombard, DB, Tishkoff, DX & Bao, J (2011) Mitochondrial sirtuins in the regulation of mitochondrial activity and metabolic adaptation. Handb Exp Pharmacol 206, 163188.CrossRefGoogle ScholarPubMed
Valero, T (2014) Mitochondrial biogenesis: pharmacological approaches. Curr Pharm Des 20, 55075509.CrossRefGoogle ScholarPubMed
Bhatti, JS, Bhatti, GK & Reddy, PH (2017) Mitochondrial dysfunction and oxidative stress in metabolic disorders – a step towards mitochondria based therapeutic strategies. Biochim Biophys Acta Mol Basis Dis 1863, 10661077.CrossRefGoogle ScholarPubMed
Austin, S & St-Pierre, J (2012) PGC1α and mitochondrial metabolism – emerging concepts and relevance in ageing and neurodegenerative disorders. J Cell Sci 125, 49634971.CrossRefGoogle ScholarPubMed
Karamanlidis, G, Lee, CF, Garcia-Menendez, L, et al. (2013) Mitochondrial complex I deficiency increases protein acetylation and accelerates heart failure. Cell Metab 18, 239250.CrossRefGoogle ScholarPubMed
Jones, DP & Sies, H (2015) The Redox Code. Antioxid Redox Signal 23, 734746.CrossRefGoogle ScholarPubMed
Rydström, J (2006) Mitochondrial NADPH, transhydrogenase and disease. Biochim Biophys Acta 1757, 721726.CrossRefGoogle ScholarPubMed
Sazanov, LA & Jackson, JB (1994) Proton-translocating transhydrogenase and NAD-linked and NADP-linked isocitrate dehydrogenases operate in a substrate cycle which contributes to fine regulation of the tricarboxylic-acid cycle activity in mitochondria. FEBS Lett 344, 109116.CrossRefGoogle Scholar
Jo, SH, Son, MK, Koh, HJ, et al. (2001) Control of mitochondrial redox balance and cellular defense against oxidative damage by mitochondrial NADP+-dependent isocitratedehydrogenase. J Biol Chem 276, 1616816176.CrossRefGoogle Scholar
Garcia, J, Han, D, Sancheti, H, et al. (2010) Regulation of mitochondrial glutathione redox status and protein glutathionylation by respiratory substrates. J Biol Chem 285, 3964639654.CrossRefGoogle ScholarPubMed
Treberg, JR, Quinlan, CL & Brand, MD (2010) Hydrogen peroxide efflux from muscle mitochondria underestimatesmatrix superoxide production – a correction using glutathione depletion FEBS J 277, 27662778.CrossRefGoogle ScholarPubMed
Aon, MA, Stanley, BA, Sivakumaran, V, et al. (2012) Glutathione/thioredoxin systems modulate mitochondrial H2O2 emission: an experimental–computational study. J Gen Physiol 139, 479491.CrossRefGoogle Scholar
Starkov, AA (2008) The role of mitochondria in reactive oxygen species metabolism and signalling. Ann N Y Acad Sci 1147, 3752.CrossRefGoogle Scholar
Dröse, S, Brandt, U & Wittig, I (2014) Mitochondrial respiratory chain complexes as sources and targets of thiol-based redox-regulation. Biochim Biophys Acta 1844, 13441345.CrossRefGoogle ScholarPubMed
Brand, MD, Affourtit, C, Esteves, TC, et al. (2004) Mitochondrial superoxide: production, biological effects, and activation of uncoupling proteins. Free Radic Biol Med 37, 755767.CrossRefGoogle ScholarPubMed
Echtay, KS, Roussel, D, St-Pierre, J, et al. (2002) Superoxide activates mitochondrial uncoupling proteins. Nature 415, 9699.CrossRefGoogle ScholarPubMed
Korshunov, SS, Skulachev, VP & Starkov, AA (1997) High protonic potential actuates a mechanism of production of reactive oxygen species in mitochondria. FEBS Lett 416, 1518.CrossRefGoogle Scholar
Miwa, S & Brand, MD (2003) Mitochondrial matrix reactive oxygen species production is very sensitive to mild uncoupling. Biochem Soc Trans 31, 13001301.CrossRefGoogle ScholarPubMed
Mailloux, RJ & Harper, ME (2011) Uncoupling proteins and the control of mitochondrial reactive oxygen species production. Free Radic Biol Med 51, 11061115.CrossRefGoogle ScholarPubMed
Puigserver, P & Spiegelman, BM (2003) Peroxisome proliferator-activated receptor-γ coactivator 1 α (PGC-1 α): transcriptional coactivator and metabolic regulator. Endocr Rev 24, 7890.CrossRefGoogle ScholarPubMed
St-Pierre, J, Lin, J, Krauss, S, et al. (2003) Bioenergetic analysis of peroxisome proliferator-activated receptor γ coactivators 1 α and 1 β (PGC-1α and PGC-1β) in muscle cells. J Biol Chem 278, 2659726603.CrossRefGoogle Scholar
St-Pierre, J, Drori, S, Uldry, M, et al. (2006) Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators. Cell 127, 397408.CrossRefGoogle ScholarPubMed
Rabinovitch, RC, Samborska, B, Faubert, B, et al. (2017) AMPK maintains cellular metabolic homeostasis through regulation of mitochondrial reactive oxygen species. Cell Rep 21, 19.CrossRefGoogle ScholarPubMed
Cantó, C, Houtkooper, RH, Pirinen, E, et al. (2012) The NAD+ precursor nicotinamide riboside enhances oxidative metabolism and protects against high-fat diet-induced obesity. Cell Metab 15, 838847.CrossRefGoogle ScholarPubMed
Jin, L, Galonek, H, Israelian, K, et al. (2009) Biochemical characterization, localization, and tissue distribution of the longer form of mouse SIRT3. Protein Sci 18, 514525.CrossRefGoogle ScholarPubMed
Nemoto, S, Fergusson, MM & Finkel, T (2005) SIRT1 functionally interacts with the metabolic regulator and transcriptional coactivator PGC-1α. J Biol Chem 280, 1645616460.CrossRefGoogle Scholar
Kong, X, Wang, R, Xue, Y, et al. (2010) Sirtuin 3, a new target of PGC-1α, plays an important role in the suppression of ROS and mitochondrial biogenesis. PLoS ONE 5, e11707.CrossRefGoogle ScholarPubMed
Sundaresan, NR, Gupta, M, Kim, G, et al. (2009) Sirt3 blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent antioxidant defense mechanisms in mice. J Clin Invest 119, 27582771.Google ScholarPubMed
Qiu, X, Brown, K, Hirschey, MD, et al. (2010) Calorie restriction reduces oxidative stress by SIRT3-mediated SOD2 activation. Cell Metab 12, 662667.CrossRefGoogle ScholarPubMed
Someya, S, Yu, W, Hallows, WC, et al. (2010) Sirt3 mediates reduction of oxidative damage and prevention of age-related hearing loss under caloric restriction. Cell 143, 802812.CrossRefGoogle ScholarPubMed
Calabrese, V, Cornelius, C, Dinkova-Kostova, AT, et al. (2010) Cellular stress responses, the hormesis paradigm, and vitagenes: novel targets for therapeutic intervention in neurodegenerative disorders. Antioxid Redox Signal 13, 17631811.CrossRefGoogle ScholarPubMed
Taguchi, K, Motohashi, H & Yamamoto, M (2011) Molecular mechanisms of the Keap1–Nrf2 pathway in stress response and cancer evolution. Genes Cells 16, 123140.CrossRefGoogle ScholarPubMed
Sarsour, EH, Kalen, AL & Goswami, PC (2014) Manganese superoxide dismutase regulates a redox cycle within the cell cycle. Antioxid Redox Signal 20, 16181627.CrossRefGoogle ScholarPubMed
Finkel, T (2011) Signal transduction by reactive oxygen species. J Cell Biol 194, 715.CrossRefGoogle ScholarPubMed
Sies, H, Berndt, C & Jones, DP (2017) Oxidative stress. Annu Rev Biochem 86, 715748.CrossRefGoogle ScholarPubMed
Kensler, TW, Wakabayashi, N & Biswal, S (2007) Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol 47, 89116.CrossRefGoogle ScholarPubMed
Calabrese, V, Cornelius, C, Dinkova-Kostova, AT, et al. (2012) Cellular stress responses, hormetic phytochemicals and vitagenes in aging and longevity. Biochim Biophys Acta 1822, 753783.CrossRefGoogle ScholarPubMed
Trewavas, A & Stewart, D (2003) Paradoxical effects of chemicals in the diet on health. Curr Opin Plant Biol 6, 185190.CrossRefGoogle ScholarPubMed
Holmström, KM, Kostov, RV & Dinkova-Kostova, AT (2016) The multifaceted role of Nrf2 in mitochondrial function. Curr Opin Toxicol 1, 8091.CrossRefGoogle ScholarPubMed
Dinkova-Kostova, AT & Abramov, AY (2015) The emerging role of Nrf2 in mitochondrial function. Free Radic Biol Med 88, 179188.CrossRefGoogle ScholarPubMed
Wegiel, B, Nemeth, Z, Correa-Costa, M, et al. (2014) Hemeoxygenase-1: a metabolic nike. Antioxid Redox Signal 20, 17091722.CrossRefGoogle ScholarPubMed
Ryter, SW, Alam, J & Choi, AM (2006) Heme oxygenase-1/carbon monoxide: from basic science to therapeutic applications. Physiol Rev 86, 583650.CrossRefGoogle ScholarPubMed
Suliman, HB, Carraway, MS, Ali, AS, et al. (2007) The CO/HO system reverses inhibition of mitochondrial biogenesis and prevents murine doxorubicin cardiomyopathy. J Clin Invest 117, 37303741.Google ScholarPubMed
Piantadosi, CA, Carraway, MS, Babiker, A, et al. (2008) Heme oxygenase-1 regulates cardiac mitochondrial biogenesis via Nrf2-mediated transcriptional control of nuclear respiratory factor-1. Circ Res 103, 12321240.CrossRefGoogle ScholarPubMed
Kawai, Y, Garduño, L, Theodore, M, et al. (2011) Acetylation–deacetylation of the transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2) regulates its transcriptional activity and nucleocytoplasmic localization. J Biol Chem 286, 76297640.CrossRefGoogle ScholarPubMed
Zhang, W, Wei, R, Zhang, L, et al. (2017) Sirtuin 6 protects the brain from cerebral ischemia/reperfusion injury through NRF2 activation. Neuroscience 366, 95104.CrossRefGoogle ScholarPubMed
Ka, SO, Bang, IH, Bae, EJ, et al. (2017) Hepatocyte-specific sirtuin 6 deletion predisposes to non-alcoholic steatohepatitis by up-regulation of Bach1, a Nrf2 repressor. FASEB J 3, 39994010.CrossRefGoogle Scholar
Oh, JY, Choi, GE, Lee, HJ, et al. (2019) 17β-Estradiol protects mesenchymal stem cells against high glucose-induced mitochondrial oxidants production via Nrf2/Sirt3/MnSOD signaling. Free Radic Biol Med 130, 328342.CrossRefGoogle ScholarPubMed
Yu, J, Sun, W, Song, Y, et al. (2019) SIRT6 protects retinal ganglion cells against hydrogen peroxide-induced apoptosis and oxidative stress by promoting Nrf2/ARE signaling via inhibition of Bach1. Chem Biol Interact 300, 151158.CrossRefGoogle ScholarPubMed
Ashabi, G, Khalaj, L, Khodagholi, F, et al. (2015) Pretreatment with metformin activates Nrf2 antioxidant pathways and inhibits inflammatory responses through induction of AMPK after transient global cerebral ischemia. Metab Brain Dis 30, 747754.CrossRefGoogle ScholarPubMed
Wang, F, Shang, Y, Zhang, R, et al. (2018) A SIRT1 agonist reduces cognitive decline in type 2 diabetic rats through antioxidative and anti-inflammatory mechanisms. Mol Med Rep 19, 10401048.Google Scholar
Anedda, A, Lopez-Bernardo, E, Acosta-Iborra, B, et al. (2013) The transcription factor Nrf2 promotes survival by enhancing the expression of uncoupling protein 3 under conditions of oxidative stress. Free Radic Biol Med 61, 395407.CrossRefGoogle ScholarPubMed
Lesnefsky, EJ & Hoppel, CL (2006) Oxidative phosphorylation and aging. Ageing Res Rev 5, 402433.CrossRefGoogle ScholarPubMed
Franco, JL, Braga, HC, Stringari, J, et al. (2007) Mercurial-induced hydrogen peroxide generation in mouse brain mitochondria: protective effects of quercetin. Chem Res Toxicol 20, 19191926.CrossRefGoogle ScholarPubMed
Lagoa, R, Graziani, I, Lopez-Sanchez, C, et al. (2011) Complex I and cytochrome c are molecular targets of flavonoids that inhibit hydrogen peroxide production by mitochondria. Biochim Biophys Acta Bioenerg 1807, 15621572.CrossRefGoogle ScholarPubMed
Zou, X, Ratti, BA, O’Brien, JG, et al. (2017) Manganese superoxide dismutase (SOD2): is there a center in the universe of mitochondrial redox signaling? J Bioenerg Biomembr 49, 325333.CrossRefGoogle Scholar
Sandoval-Acuña, C, Ferreira, J & Speisky, H (2014) Polyphenols and mitochondria: an update on their increasingly emerging ROS-scavenging independent actions. Arch Biochem Biophys 559, 7590.CrossRefGoogle ScholarPubMed
Decker, EA (1997) Phenolics: prooxidants or antioxidants? Nutr Rev 55, 396398.CrossRefGoogle ScholarPubMed
Galati, G & O’Brien, PJ (2004) Potential toxicity of flavonoids and other dietary phenolics: significance for their chemopreventive and anticancer properties. Free Radic Biol Med 37, 287303.CrossRefGoogle ScholarPubMed
Bors, W & Michel, C (2002) Chemistry of the antioxidant effect of polyphenols. Ann N Y Acad Sci 957, 5769.CrossRefGoogle ScholarPubMed
Crozier, A, Jaganath, IB & Clifford, MN (2009) Dietary phenolics: chemistry, bioavailability and effects on health. Nat Prod Rep 26, 10011043.CrossRefGoogle ScholarPubMed
Chow, HH & Hakim, IA (2011) Pharmacokinetic and chemoprevention studies on tea in humans. Pharmacol Res A64, 105112.Google Scholar
Czank, C, Cassidy, A, Zhang, Q, et al. (2013) Human metabolism and elimination of the anthocyanin, cyanidin-3-glucoside: a 13C-tracer study. Am J Clin Nutr 97, 995–100.CrossRefGoogle Scholar
Espín, JC, González-Sarrías, A & Tomás-Barberán, FA (2017) The gut microbiota: a key factor in the therapeutic effects of (poly)phenols. Biochem Pharmacol 139, 8293.CrossRefGoogle ScholarPubMed
Armah, CN, Traka, MH, Dainty, JR, et al. (2013) A diet rich in high-glucoraphanin broccoli interacts with genotype to reduce discordance in plasma metabolite profiles by modulating mitochondrial function. Am J Clin Nutr 98, 712722.CrossRefGoogle Scholar
Cuadrado, A, Manda, G, Hassan, A, et al. (2018) Transcription factor NRF2 as a therapeutic target for chronic diseases: a systems medicine approach. Pharmacol Rev 70, 348383.CrossRefGoogle ScholarPubMed
Carrasco-Pozo, C, Gotteland, M & Speisky, H (2011) Apple peel polyphenol extract protects against indomethacin-induced damage in Caco-2 cells by preventing mitochondrial complex I inhibition. J Agric Food Chem 59, 1150111508.CrossRefGoogle ScholarPubMed
Sandoval-Acuña, C, Lopez-Alarcón, C, Aliaga, ME, et al. (2012) Inhibition of mitochondrial complex I by various non-steroidal anti-inflammatory drugs and its protection by quercetin via coenzyme Q-like action. Chem Biol Interact 199, 1828.CrossRefGoogle ScholarPubMed
Murphy, M (2009) How mitochondria produce reactive oxygen species. Biochem J 417, 113.CrossRefGoogle ScholarPubMed
Halliwell, B (2006) Oxidative stress and neurodegeneration: where are we now? J Neurochem 97, 16341658.CrossRefGoogle ScholarPubMed
Tsuji, PA, Stephenson, KK, Wade, KL, et al. (2013) Structure–activity analysis of flavonoids: direct and indirect antioxidant, and antiinflammatory potencies and toxicities. Nutr Cancer 65, 10141025.CrossRefGoogle ScholarPubMed
Iranshahy, M, Iranshahi, M, Abtahi, SR, et al. (2018) The role of nuclear factor erythroid 2-related factor 2 in hepatoprotective activity of natural products: a review. Food Chem Toxicol 120, 261276.CrossRefGoogle ScholarPubMed
Surh, YJ (2011) Xenohormesis mechanisms underlying chemopreventive effects of some dietary phytochemicals. Ann N Y Acad Sci 1229, 16.CrossRefGoogle ScholarPubMed
Son, TG, Camandola, S & Mattson, MP (2008) Hormetic dietary phytochemicals. Neuromolecular Med 10, 236246.CrossRefGoogle ScholarPubMed
Scapagnini, G, Vasto, S, Abraham, NG et al. (2011) Modulation of Nrf2/ARE pathway by food polyphenols: a nutritional neuroprotective strategy for cognitive and neurodegenerative disorders. Mol Neurobiol 44, 192201.CrossRefGoogle ScholarPubMed
Eggler, AL & Savinov, SN (2013) Chemical and biological mechanisms of phytochemical activation of Nrf2 and importance in disease prevention. Recent Adv Phytochem 43, 121155.Google ScholarPubMed
Stefanson, AL & Bakovic, M (2014) Dietary regulation of Keap1/Nrf2/ARE pathway: focus on plant-derived compounds and trace minerals. Nutrients 6, 37773801.CrossRefGoogle ScholarPubMed
Trujillo, J, Granados-Castro, LF, Zazueta, C, et al. (2014) Mitochondria as a target in the therapeutic properties of curcumin. Arch Pharm (Weinheim) 347, 873884.CrossRefGoogle ScholarPubMed
Waseem, M & Parvez, S (2015) Neuroprotective activities of curcumin and quercetin with potential relevance to mitochondrial dysfunction induced by oxaliplatin. Protoplasma 253, 417430.CrossRefGoogle ScholarPubMed
Ji, LL, Sheng, YC, Zheng, ZY, et al. (2015) The involvement of p62–Keap1–Nrf2 antioxidative signaling pathway and JNK in the protection of natural flavonoid quercetin against hepatotoxicity. Free Radic Biol Med 85, 1223.CrossRefGoogle ScholarPubMed
Jung, CH, Cho, I, Ahn, J, et al. (2013) Quercetin reduces high-fat diet induced fat accumulation in the liver by regulating lipid metabolism genes. Phytother Res 27, 139143.CrossRefGoogle ScholarPubMed
Tang, Y, Gao, C, Xing, M, et al. (2012) Quercetin prevents ethanol induced dyslipidemia and mitochondrial oxidative damage. Food Chem Toxicol 50, 11941200.CrossRefGoogle ScholarPubMed
Kovacic, P & Somanathan, R (2010) Multifaceted approach to resveratrol bioactivity: focus on antioxidant action, cell signaling and safety. Oxid Med Cell Longev 3, 86100.CrossRefGoogle ScholarPubMed
Guha, P, Dey, A, Dhyani, MV, et al. (2010) Calpain and caspase orchestrated death signal to accomplish apoptosis induced by resveratrol and its novel analog hydroxstilbene-1 in cancer cells. J Pharmacol Exp Ther 334, 381394.CrossRefGoogle ScholarPubMed
Ungvari, Z, Bagi, Z, Feher, A, et al. (2010) Resveratrol confers endothelial protection via activation of the antioxidant transcription factor Nrf2. Am J Physiol Heart Circ Physiol 299, H18H24.CrossRefGoogle ScholarPubMed
Ak, T & Gülçin, I (2008) Antioxidant and radical scavenging properties of curcumin. Chem Biol Interact 174, 2737.CrossRefGoogle ScholarPubMed
Barzegar, A & Moosavi-Movahedi, AA (2011) Intracellular ROS protection efficiency and free radical-scavenging activity of curcumin. PLoS ONE 6, e26012.CrossRefGoogle ScholarPubMed
Derochette, S, Franck, T, Mouithys-Mickalad, A, et al. (2013) Curcumin and resveratrol act by different ways on NADPH oxidase activity and reactive oxygen species produced by equine neutrophils. Chem Biol Interact 206, 186193.CrossRefGoogle ScholarPubMed
Reyes-Fermín, LM, González-Reyes, S, Tarco-Álvarez, NG, et al. (2012) Neuroprotective effect of α-mangostin and curcumin against iodoacetate-induced cell death. Nutr Neurosci 15, 3441.CrossRefGoogle ScholarPubMed
Granados-Castro, LF, Rodríguez-Rangel, DS, Fernández-Rojas, B, et al. (2016) Curcumin prevents paracetamol-induced liver mitochondrial alterations. J Pharm Pharmacol 68, 245256.CrossRefGoogle ScholarPubMed
García-Niño, WR, Tapia, E, Zazueta, C, et al. (2013) Curcumin pretreatment prevents potassium dichromate-induced hepatotoxicity, oxidative stress, decreased respiratory complex I activity, and membrane permeability transition pore opening. Evid Based Complementary Altern Med 2013, 424692.CrossRefGoogle ScholarPubMed
Feng, C, Luo, T, Zhang, S, et al. (2016) Lycopene protects human SH–SY5Y neuroblastoma cells against hydrogen peroxide-induced death via inhibition of oxidative stress and mitochondria–associated apoptotic pathways. Mol Med Rep 13, 42054214.CrossRefGoogle Scholar
Houghton, CA, Fassett, RG & Coombes, JS (2016) Sulforaphane and other nutrigenomic Nrf2 activators: can the clinician’s expectation be matched by the reality? Oxid Med Cell Longev 2016, 7857186.CrossRefGoogle ScholarPubMed
Bai, X, Yao, L, Ma, X, et al. (2018) Small molecules as SIRT modulators. Mini-Rev Med Chem 18, 11511157.CrossRefGoogle ScholarPubMed
Giovannini, L & Bianchi, S (2017) Role of nutraceutical SIRT1 modulators in AMPK and mTOR pathway: evidence of a synergistic effect. Nutrition 34, 8296.CrossRefGoogle ScholarPubMed
Zhou, X, Chen, M, Zeng, X, et al. (2014) Resveratrol regulates mitochondrial reactive oxygen species homeostasis through Sirt3 signaling pathway in human vascular endothelial cells. Cell Death Dis 5, e1576.CrossRefGoogle ScholarPubMed
Ungvari, Z, Orosz, Z, Rivera, A, et al. (2007) Resveratrol increases vascular oxidative stress resistance. Am J Physiol Heart Circ Physiol 292, H2417H2424.CrossRefGoogle ScholarPubMed
Ungvari, Z, Labinskyy, N, Mukhopadhyay, P, et al. (2009) Resveratrol attenuates mitochondrial oxidative stress in coronary arterial endothelial cells. Am J Physiol Heart Circ Physiol 297, H1876H1881.CrossRefGoogle ScholarPubMed
Moskaug, , Carlsen, H, Myhrstad, MC, et al. (2005) Polyphenols and glutathione synthesis regulation. Am J Clin Nutr 81, 277S283S.CrossRefGoogle ScholarPubMed
Sarubbo, F, Esteban, S, Miralles, A, et al. (2018) Effects of resveratrol and other polyphenols on Sirt1: relevance to brain function during aging. Curr Neuropharmacol 16, 126136.CrossRefGoogle ScholarPubMed
Kim, CS, Kwon, Y, Choe, SY, et al. (2015) Quercetin reduces obesity-induced hepatosteatosis by enhancing mitochondrial oxidative metabolism via heme oxygenase-1. Nutr Metab (Lond) 12, 33.CrossRefGoogle ScholarPubMed
Mo, C, Wang, L, Zhang, J, et al. (2014) The crosstalk between Nrf2 and AMPK signal pathways is important for the anti-inflammatory effect of berberine in LPS-stimulated macrophages and endotoxin-shocked mice. Antioxid Redox Signal 20, 574588.CrossRefGoogle ScholarPubMed
Zimmermann, K, Baldinger, J, Mayerhofer, B, et al. (2015) Activated AMPK boosts the Nrf2/HO-1 signaling axis – a role for the unfolded protein response. Free Radic Biol Med 88, 417426.CrossRefGoogle Scholar
Nadal-Serrano, M, Pons, DG, Sastre-Serra, J, et al. (2013) Genistein modulates oxidative stress in breast cancer cell lines according to ERα/ERβ ratio: effects on mitochondrial functionality, sirtuins, uncoupling protein 2 and antioxidant enzymes. Int J Biochem Cell Biol 45, 20452051.CrossRefGoogle ScholarPubMed
Modrianský, M & Gabrielová, E (2009) Uncouple my heart: the benefits of inefficiency. J Bioenerg Biomembr 41, 133136.CrossRefGoogle ScholarPubMed
Brand, MD & Nicholls, DG (2011) Assessing mitochondrial dysfunction in cells. Biochem J 435, 297312.CrossRefGoogle ScholarPubMed
Demine, S, Reddy, N, Renard, P, et al. (2014) Unraveling biochemical pathways affected by mitochondrial dysfunctions using metabolomic approaches. Metabolites 4, 831878.CrossRefGoogle ScholarPubMed
Pandey, KB & Rizvi, SI (2009) Plant polyphenols as dietary antioxidants in human health and disease. Oxid Med Cell Longev 2, 270278.CrossRefGoogle ScholarPubMed
Figure 0

Table 1. Nutritional regulators of mitochondrial functions

Figure 1

Fig. 1. Pivotal role of nuclear factor erythroid-derived 2-like (Nrf2) in the regulation of mitochondrial pathways. In addition to its well-known role in redox homeostasis, Nrf2 is involved in the regulation of many aspects of mitochondrial metabolism, such as biogenesis, fatty acid oxidation, oxidative phosphorylation (OXPHOS) and redox homeostasis. The table describes the major players and their cellular location during these processes. AMPK, AMP-activated protein kinase; CAT, catalase; GPx, glutathione peroxidase; mROS, mitochondrial reactive oxygen species; NRF, nuclear respiratory factor; PGC-1α, PPARγ coactivator-1α; ROS, reactive oxygen species; SIRT1, sirtuin 1; SIRT3, sirtuin 3; SOD, superoxide dismutase; SOD2, superoxide dismutase 2; TFAM, mitochondrial transcription factor A; UCP, uncoupling proteins. For a colour figure, see the online version of the paper.

Figure 2

Fig. 2. Ability of bioactive compounds to modulate redox status. Several polyphenolic molecules, such as resveratrol, curcumin, quercetin and genistein, can affect redox homeostasis by directly and indirectly exerting antioxidant effects. The direct mechanisms consist of reactive oxygen species scavenging and metal chelation, whilst the indirect mechanisms include activating nuclear factor erythroid-derived 2-like (Nrf2) signalling, regulating inner mitochondrial membrane potential via uncoupling effects (up-regulating uncoupling protein 2 (UCP2) expression), and modulating radical species production via electron transport chain (ETC) complexes. The table describes the different bioactive compounds, where they originate, and their effects on mitochondrial metabolism. AMPK, AMP-activated protein kinase; EGCG, epigallocatechin gallate; MMP, matrix metalloproteinase; OXPHOS, oxidative phosphorylation; PGC-1α, PPARγ coactivator-1α; SIRT1, sirtuin 1. For a colour figure, see the online version of the paper.