Next Article in Journal
Downregulation of PPARα during Experimental Left Ventricular Hypertrophy is Critically Dependent on Nox2 NADPH Oxidase Signalling
Next Article in Special Issue
Involvement of Mitochondrial Mechanisms in the Cytostatic Effect of Desethylamiodarone in B16F10 Melanoma Cells
Previous Article in Journal
Investigating the Role of PPARβ/δ in Retinal Vascular Remodeling Using Pparβ/δ-Deficient Mice
Previous Article in Special Issue
Bioinformatics Analyses of the Transcriptome Reveal Ube3a-Dependent Effects on Mitochondrial-Related Pathways
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Multifaceted Roles of Mitochondrial Components and Metabolites in Metabolic Diseases and Cancer

by
Jean Nakhle
1,2,
Anne-Marie Rodriguez
3,4,5,6,* and
Marie-Luce Vignais
1,*
1
Institute for Regenerative Medicine & Biotherapy (IRMB), INSERM, Univ Montpellier, F-34090 Montpellier, France
2
Institute of Molecular Genetics of Montpellier (IGMM), CNRS, Univ Montpellier, F-34090 Montpellier, France
3
Univ Paris Est Creteil, INSERM, IMRB, F-94010 Creteil, France
4
EnvA, IMRB, F-94700 Maisons-Alfort, France
5
EFS, Mondor Institute for Biomedical Research (IMRB), F-94010 Creteil, France
6
AP-HP, Hopital Mondor, Service d’histologie, F-94010 Creteil, France
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2020, 21(12), 4405; https://doi.org/10.3390/ijms21124405
Submission received: 30 May 2020 / Revised: 12 June 2020 / Accepted: 17 June 2020 / Published: 20 June 2020
(This article belongs to the Special Issue Mitochondria-Targeted Approaches in Health and Disease)

Abstract

:
Mitochondria are essential cellular components that ensure physiological metabolic functions. They provide energy in the form of adenosine triphosphate (ATP) through the electron transport chain (ETC). They also constitute a metabolic hub in which metabolites are used and processed, notably through the tricarboxylic acid (TCA) cycle. These newly generated metabolites have the capacity to feed other cellular metabolic pathways; modify cellular functions; and, ultimately, generate specific phenotypes. Mitochondria also provide intracellular signaling cues through reactive oxygen species (ROS) production. As expected with such a central cellular role, mitochondrial dysfunctions have been linked to many different diseases. The origins of some of these diseases could be pinpointed to specific mutations in both mitochondrial- and nuclear-encoded genes. In addition to their impressive intracellular tasks, mitochondria also provide intercellular signaling as they can be exchanged between cells, with resulting effects ranging from repair of damaged cells to strengthened progression and chemo-resistance of cancer cells. Several therapeutic options can now be envisioned to rescue mitochondria-defective cells. They include gene therapy for both mitochondrial and nuclear defective genes. Transferring exogenous mitochondria to target cells is also a whole new area of investigation. Finally, supplementing targeted metabolites, possibly through microbiota transplantation, appears as another therapeutic approach full of promises.

1. Introduction

Mitochondria are descendants from alpha-proteobacteria that survived after their endocytosis by eukaryotic progenitors, more than 1.5 billion years ago. These bacterial ancestors became symbiotic with their host cells and gradually gave rise to the permanent organelles found today in almost all eukaryotic cells [1]. As a vestige of their bacterial endosymbiotic origin, mitochondria have retained their double membranes and circular genome [2]. However, this mitochondrial DNA (mtDNA) has been dramatically reduced over evolution, rendering mitochondria dependent on the nucleus for the expression of the vast majority of their proteins [2]. As a result, the biogenesis and functionality of mitochondria are tightly regulated by the continuous crosstalk with the nucleus [3]. The function usually assigned to mitochondria is their capacity to produce high amounts of ATP, from glucose breakdown, through oxidative phosphorylation (OXPHOS). OXPHOS-dependent ATP synthesis is supported by the mitochondrial electron transport chain (ETC), composed of protein complexes and organic molecules, which conveys electrons to molecular oxygen and creates the electrochemical gradient needed for the functioning of ATP synthase [4]. In tight connection with the ETC, metabolites are produced in the mitochondria through the tricarboxylic acid (TCA) cycle. The TCA cycle is composed of eight enzymes catalyzing the chemical breakdown of carbohydrates, fats, and proteins to produce either ATP or building blocks for the synthesis of nucleic acids, amino-acids, and lipids.
Beyond their role as energy and building-block producers, mitochondria also act as signaling organelles that govern cell fate by regulating essential biological processes, such as cell growth, differentiation, and apoptosis, as well as Ca2+ and redox homeostasis. Mitochondria are thus implicated in essential physiological and pathophysiological processes including tissue healing, inflammation, and cancer [5,6,7,8]. The complexity and diversity of mitochondrial functions are reflected by their wide-ranging proteome of more than 1000 proteins [9,10]. The already-extensive panel of mitochondrial functions has been further increased following the recent discovery of their capacity to translocate to other cells and to alter their behavior [11,12]. This signaling role of mitochondria is also supported by their recently-documented capacity to release metabolites as well as mitochondrial fragments, also called damage-associated molecular patterns (DAMPs) [13,14,15].
The severity of the diseases caused by mitochondrial dysfunctions confirms the critical importance of mitochondria for living cells. These diseases can originate from genetic mutations directly affecting the ETC activity or other mitochondrial functions. However, they can also be owing to unbalanced production of metabolites and mitochondrial components. The challenging identification of mitochondrial dysfunctions will need to be overcome in order to develop therapeutic strategies for the related metabolic, inflammatory, and malignant pathologies [14,16,17,18].
Here, we present an overview of recent findings on the critical involvement of mitochondrial metabolites/compounds in the signaling functions of mitochondria and on the emergence of novel therapies for diseases caused by mitochondrial dysfunctions.

2. Role of the Mitochondrial ETC and OXPHOS in Physiology and Disease

The respiratory machinery responsible for OXPHOS is located in the inner mitochondrial membrane. It includes the four complexes (I–IV) of the electron transport chain (ETC), the free-electron carriers ubiquinone and cytochrome c, and the ATP synthase (complex V) that produces OXPHOS-dependent ATP (Figure 1). The ETC machinery is encoded by both the nuclear DNA (nDNA) (80 proteins) and mitochondrial DNA (mtDNA) (13 proteins). Nuclear-encoded proteins (roughly 70% of total mitochondrial proteins) also have functions in ETC supercomplex (or respirasome) assembly [19] and in mtDNA replication and repair [20]. Therefore, nuclear and mitochondrial gene expressions need to be tightly coordinated to ensure effective mitochondrial functioning. Alterations in either result in mitochondrial defects, which makes their treatment particularly challenging. Mitochondrial DNA exhibits high mutation rates owing to (1) direct exposure to ETC-derived reactive oxygen species (ROS), (2) asymmetric replication leading to higher exposure of the single H-strand, (3) intramitochondrial dNTP pool imbalance favoring dGTP incorporation, and (4) low-efficiency DNA repair mechanisms [21]. Mitochondrial DNA mutations range from point mutations to large-scale DNA rearrangements. They can be found in cells together with their wild-type counterpart, a feature called heteroplasmy. Exceeding the critical heteroplasmy thresholds causes mutant mtDNA to trigger mitochondria-related pathologies [22].
Mitochondrial diseases are characterized by genetic alterations that primarily compromise oxidative phosphorylation (OXPHOS) and OXPHOS-dependent ATP generation. Mitochondrial diseases thus affect organs that mainly rely on OXPHOS for energy supply, including the eye, ear, liver, kidney, heart, and skeletal muscles, as well as the brain. These diseases are associated with syndromes such as CPEO (chronic progressive external ophthalmoplegia), KSS (Kearns–Sayre syndrome), LHON (Leber hereditary optic neuropathy), MIDD (maternally inherited diabetes and deafness), MELAS (mitochondrial encephalomyopathy with lactic acidosis and stroke-like episodes), mitochondrial encephalomyopathy (Leigh syndrome), MERRF (myoclonic epilepsy with ragged-red fibers), and NARP (syndrome neuropathy, ataxia, and retinitis pigmentosa). Mitochondrial disorders have also been associated with cancers in different organs [17,22,23], as detailed below.

3. Effects of Mutations in the Subunits of Complexes I and II of the ETC in Cancer

Mitochondrial disorders play a pivotal role in tumorigenesis by orchestrating metabolic reprogramming, apoptosis, and ROS signaling at virtually all stages, from tumor initiation to metastasis, and mtDNA mutations have been proposed as drivers/modifiers of tumorigenesis in many cancers [24,25,26]. Sequencing of mtDNAs from over 2000 tumor samples across over 30 tumor types revealed important features of the mitochondrial mutational signature [27,28]. These large-scale studies identified intrinsic replicative errors, characterized by an asymmetric bias towards C > T and T > C transitions on the mtDNA heavy and light strands respectively, as the most prominent source of mtDNA mutations, overpowering the 4% caused by ROS exposure. While neutral missense mutations were found to gradually drift towards homoplasmy, deleterious frameshift or nonsense mutations, often resulting in truncated proteins, were almost exclusively heteroplasmic, thus highlighting a selective pressure in cancer cells to retain residual mitochondrial functionality [27,28].

3.1. Complex I Mutation-Induced ROS Production Promotes Tumorigenesis through Phosphatidylinositol 3-Kinase (PI3K)/Protein Kinase B (AKT) Signaling, Hypoxia-Inducible Factor 1 Alpha (HIF1α) Stabilization, and NADPH-Oxidase 1 (NOX1) Signaling

Complex I of the ETC transfers electrons from NADH produced in the TCA cycle and from the β-oxidation of fatty acids to ubiquinone. This reaction is accompanied by the translocation of four protons from the mitochondrial matrix into the intermembrane space, which generates a transmembrane electrochemical gradient and drives final ATP production by complex V (Figure 1). Complex I is the largest (1 MDa) and most elaborate complex of the ETC. X-ray crystallography revealed a structure with two L-shaped arms: a peripheral arm that catalyzes the redox reaction and an inner membrane-embedded arm containing the proton-translocating machinery. Complex I is composed of 14 conserved core subunits, encoded by both the mitochondrial and nuclear DNAs, and of at least 30 additional nuclear-encoded accessory subunits. Half the core subunits (ND1, ND2, ND3, ND4, ND4L, ND5, ND6) are mitochondria-encoded and constitute the membrane-embedded arm, while the other half (NDUFS1, NDUFV1, NDUFV2, NDUFS2, NDUFS3, NDUFS8, NDUFS7) are nuclear-encoded and constitute the peripheral arm [19,29,30]. Complex I is the main target of the currently-identified mtDNA mutations. Taking into account both mtDNA and nDNA origins, mutations are found in all 14 catalytic core subunits, in 13 accessory subunits, and in at least 11 assembly factors [31]. They have been described in cancers [32] as diverse as head and neck [33,34], breast [27,35,36], thyroid [37,38,39], prostate [40], renal [39,41], and hepatocellular cancers [42]. The technique of transmitochondrial cybrids, which consists of repopulating ρ0 cells (depleted of their mtDNA) with exogenous mitochondria, has been widely used to establish the role of mtDNA variants in pathologies, independent of nDNA backgrounds [43]. Transmitochondrial cybrid studies thus showed that complex I mutants can exhibit both pro- and anti-tumorigenic effects in cancer, in an OXPHOS- and ROS-dependent fashion, as detailed below.

3.1.1. PI3K/AKT Signaling

The ND5 m.12418insA frameshift mutation, found in colorectal cancers, leads to a truncated ND5 protein, which destabilizes the assembly of the membrane-embedded arm of complex I [44,45]. As shown in 143B osteosarcoma transmitochondrial cybrids, the effects of this mutant highly depended on its heteroplasmy level. While a 72% ND5 mutant heteroplasmy still retained 46% residual complex I activity, ND5 mutant homoplasmy (96%) led to loss of complex I function [44]. These effects were associated, as expected, with a gradual decrease of OXPHOS-dependent oxygen consumption and ATP production [46]. Heteroplasmic cybrids exhibited increased levels of mitochondria-specific ROS (but not of intracellular ROS), which correlated with the overexpression of the catalase, the glutathione peroxidase 4 (Gpx4), and the Cu-Zn superoxide dismutase (SOD1) anti-oxidative enzymes. This enhanced ROS production also activated the PI3K/AKT signaling pathway and the expression of downstream genes encoding the hypoxia-inducible factor 1 alpha (HIF1α) and the anti-apoptotic proteins B-cell lymphoma-extra large (BCL-XL) and induced myeloid leukemia cell differentiation protein (MCL1) (Figure 2) [47]. Osteosarcoma cybrids harboring a heteroplasmic ND5 mutation were thus endowed with resistance to oxidative stress and, subsequently, enhanced tumorigenicity upon subcutaneous injection in nude mice [46,47]. ND5-mutant homoplasmic cybrids, however, showed increased levels in both mitochondrial and intracellular ROS, which led to apoptosis and prevented tumor formation in vivo [46].

3.1.2. HIF1α Stabilization

The ND1m.3571insC frameshift mutation, observed in thyroid oncocytic carcinoma, also generates a truncated ND1 protein, which leads to complex I disassembly [48]. Transmitochondrial osteosarcoma cybrids expressing this ND1 mutant totally lost their complex I activity and their basal oxygen consumption, revealing a dramatic OXPHOS deficit [49]. Complex I dysfunction also induced the accumulation of NADH, an allosteric inhibitor of α-ketoglutarate dehydrogenase, which led to increased α-ketoglutarate levels and to HIF1α destabilization (Figure 2). These osteosarcoma cybrids did not form tumors in vivo [49,50]. Another ND1mutation (missense m.3460G>A, A52T) showed a weaker phenotype as homoplasmic osteosarcoma cybrids retained 50% of complex I activity, with only a partial reduction in basal oxygen consumption [49]. These mutants were still able to form tumors upon xenograft in mice, a tumorigenic potential linked to cytoplasmic ROS accumulation and HIF1α stabilization [49]. A third type of mutation (m.4776G>A; A103T), found in the ND2 subunit of complex I in head and neck squamous cell carcinoma, also illustrates the role of ROS accumulation and HIF1α stabilization in tumorigenesis [33,34]. This mutation, which impaired cell respiration and increased ROS production, led to a feedback metabolic signaling involving pyruvate dehydrogenase (PDH) and pyruvate dehydrogenase kinase 2 (PDK2) [34]. ROS-induced PDK2 activity resulted in increased pyruvate, HIF1α activation and tumor growth (Figure 2) [34]. Finally, the highly-metastatic capacities of murine Lewis lung carcinoma and fibrosarcoma were attributed to the missense m.13997G>A (P25L) and frameshift m.13885insC mutations, respectively, in the mtDNA ND6-encoding gene, as further confirmed in cybrids [51]. The increased metastatic capacity of these complex I mutants was also associated with a higher ROS production, which led to high levels of HIF1α, of the anti-apoptotic MCL-1 protein, and of the vascular endothelial growth factor (VEGF), thus nurturing neoangiogenesis. Consistent with this role of ROS, pretreatment of highly-metastatic cybrids with the ROS scavenger N-acetylcysteine (NAC) decreased MCL-1 expression in vitro and reduced metastasis in vivo [51].

3.1.3. NOX1 Signaling

NADPH-oxidase 1 (NOX1) activity was also shown to contribute to tumorigenesis, together with increased mitochondrial ROS, as a result of complex I dysfunction [52]. The various osteosarcoma cybrids analyzed, carrying the m.3460G>A/MT-ND1 (A52T), m.11778G>A/MT-ND4 (R340H), and m.14484T>C/MT-ND6 (M64V) point mutations, respectively, exhibited decreased OXPHOS and ATP production. Their enhanced tumorigenicity, upon xenograft in nude mice, was associated with increased levels of both mitochondrial and cytoplasmic ROS as well as increased activity of NOX1, resulting in the production of superoxide and hydrogen peroxide, major sources of cytoplasmic ROS (Figure 2) [52]. Overall, mutations in complex I appear to exert anti-tumorigenic effects only upon complete abolishment of complex I function, which prevents ROS production. Milder mutations, which preserve some complex I activity, enhance ROS production and stimulate tumor growth [46,49,52].

3.2. OXPHOS-Harmful Complex I Mutations Elicit Metabolic Compensation

3.2.1. Shift towards Glycolysis

Several studies reported that OXPHOS dysfunction, as a consequence of complex I impairment, leads to a metabolic shift towards glycolysis, which enables cells to maintain cell growth and ATP production [33,34,46,49,52,53]. This metabolic shift was reported in transmitochondrial osteosarcoma cybrids harboring the m.12418insA/MT-ND5 mutation. Increasing the mutational load from 72% of mutant ND5 in heteroplasmic cybrids to 96% in homoplasmic cybrids gradually decreased respiration (from 53% to 17% of wild type (WT)) and OXPHOS-linked ATP production (from 21% to 2% of WT), while it concomitantly increased glucose uptake and lactate production (from 28% to 56% of WT) [46]. Likewise, as shown for another subunit of complex I, osteosarcoma cells harboring the severe m.3571insC/MT-ND1 mutation showed increased glucose uptake and lactate production, while the milder m.3460G>A/MT-ND1 mutant had a glucose metabolism similar to that of the wild-type cells [49]. This glycolytic shift observed in transmitochondrial osteosarcoma cybrids was ascribed to HIF1α activation [50]. HIF1α stimulated the expression of the glucose transporters 1 (SLC2A1) and 3 (SLC2A3) as well as a series of glycolytic enzymes, including phosphofructokinase (PFKP), glyceraldehyde 3-phosphate dehydrogenase (GAPDH), phosphoglycerate kinase (PGK1), and lactate dehydrogenase (LDHA) [50]. Taken together, these data indicate that complex I mutations could induce a Warburg-like effect in cancer cells, fostering tumor growth.

3.2.2. Shift towards Complex II-Dependent Succinate Oxidation

Beyond the Warburg effect, a metabolic shift towards complex II-dependent succinate oxidation was recently described as an adaptive mechanism to compensate for OXPHOS dysfunction in high-grade prostate cancers, mutant for the ND1 subunit of complex I [54]. As a response to the inhibition of glutamate and malate oxidation and to the subsequent electron transport loss across complex I, ND1-mutant cancer cells were found to produce ATP by aerobic respiration through complex II-mediated succinate oxidation [54]. This metabolic reprogramming was associated with shorter patient survival, highlighting the critical role of complex II and succinate in malignancies associated with complex I dysfunction [54].

3.2.3. Shift towards Serine Catabolism

The catabolism of serine, a precursor for nucleic acids, proteins, and fatty acids [55,56], was also shown to maintain OXPHOS activity in colon cancer cells with mutant complex I, by producing NADH and feeding it to the ETC [57]. Higher NADH levels than expected could be reached through serine catabolism because of the insensitivity to NADH concentrations of MTHFD2 (methylenetetrahydrofolate dehydrogenase 2), the enzyme involved in this conversion [57]. Overall, these findings demonstrate the various ways tumor cells manage to get around complex I dysfunction to maintain efficient OXPHOS.

3.2.4. Complex I Loss Alleviates Complex V Dysfunction

By applying a genome-wide CRISPR screening on human K562 chronic myeloid leukemia cells concomitantly treated with oligomycin, an inhibitor of complex V, Mootha and colleagues identified a series of both synthetic lethal mutants and suppressors showing that complex I mutations can alleviate complex V dysfunction [58]. Mechanistically, concomitant inhibition of complexes I and V was accompanied by an increased reductive carboxylation of α-ketoglutarate [58]. Experimental depletion of cytosolic NADPH, known to drive reductive pathways, suppressed the protective effect of complex I loss against complex V inactivation, thus supporting the role of reductive metabolism in these effects [58].

3.3. Complex II Dysfunction Induces Tumorigenicity via Succinate Accumulation

Complex II, also known as succinate dehydrogenase (SDH) and succinate ubiquinone oxidoreductase, has the unique property of linking both the TCA cycle and the ETC by coupling succinate oxidation to fumarate within the TCA cycle with ubiquinone (coenzyme Q10) reduction to ubiquinol in the ETC. Complex II is composed of four subunits solely encoded by nuclear genes (SDHA, SDHB, SDHC, and SDHD). SDHA oxidizes succinate to fumarate, while reducing FAD to FADH2. Electrons from FADH2 are then transferred sequentially to SDHB and to ubiquinone at the inner membrane-embedded site formed by SDHC and SDHD. Contrary to complexes I, III, and IV, complex II-mediated electron transport is not accompanied by proton translocation into the mitochondrial intermembrane space [19,59].
Deleterious mutations in any of the four subunits of complex II decrease SDH activity and result in abnormal accumulation of succinate, as observed in complex II-mutated cells in vitro and in the extracellular fluids (plasma, urine, saliva, and feces) of complex II-deficient patients. Complex II mutations, principally affecting SDHB and SDHD subunits, have been associated with various cancers including hereditary paraganglioma and pheochromocytoma (i.e., neuroendocrine tumors of the paraganglionic tissue), gastrointestinal stromal tumors [60], and renal cell carcinoma [61]. Therefore, SDH has now been defined as a tumor suppressor and succinate as an oncometabolite [62,63].

3.3.1. Complex II Mutations Inhibit 2-Oxoglutarate-Dependent Dioxygenases

The R22X nonsense SDHD mutation found in hereditary paraganglioma and pheochromocytoma generates a truncated SDHD protein of 21 amino acids (instead of 159), resulting in the loss of complex II electron transfer and enzymatic activities and in the activation of the HIF1α signaling pathway [64,65]. The R22X nonsense SDHD mutation leads to succinate accumulation, which inhibits the activity of prolyl hydroxylase (PHD) and, consequently, induces HIF1α stabilization [65]. PHD is a 2-oxoglutarate (2-OG, also known as α-ketoglutarate)-dependent dioxygenase that, in normal settings, hydroxylates HIF1α and leads to its poly-ubiquitylation by the Von Hippel–Lindau protein (pVHL) complex and, ultimately, to its degradation by proteasomes [66] (Figure 3). Succinate is structurally similar to 2-OG. It thus acts as a competitive inhibitor for PHD and promotes HIF1α stabilization [67,68].
Succinate accumulation resulting from complex II mutations was shown to inhibit other 2-OG-dependent dioxygenases, including the Jumonji C (JmjC)-domain containing histone lysine demethylases (KDMs) [69] and the ten-eleven translocation (TET) family of DNA hydroxylases [70] (Figure 3). TET enzymes mediate DNA oxidative demethylation through 5-methylcytosine (5mC) hydroxylation into 5-hydroxy-methylcytosine (5hmC) [70,71]. Succinate accumulation was shown to reduce TET-induced levels of 5hmC in human embryonic kidney cells (HEK293T) with SDHA/B knockdown as well as in mice livers with transient SDHA knockdown [72]. In line with these observations, DNA hypermethylation was found in SDHx-mutated paraganglioma and pheochromocytoma samples and in SDH-deficient gastrointestinal stromal samples, as a result of succinate inhibition of KDM and TET enzymes [73,74,75].

3.3.2. The Hypermethylator Phenotype: A Double-Edged Sword for Complex II-Mutated Cancers

As mentioned above, SDH mutations can promote tumorigenesis by a succinate-mediated genome-wide epigenetic remodeling [74,76]. As shown in mouse SDHB-knockout chromaffin cells, increased succinate levels were associated with CpG island hypermethylation in the promoters of genes encoding proteins such as the matrix-metalloprotease inhibitor SPOCK2, the metastasis suppressor DNAJA4, and the cell adhesion marker KRT19 [74], thus resulting in a derepression of epithelial-to-mesenchymal transition (EMT). Succinate accumulation following SDHB-knockdown also led to an EMT-like phenotype in a murine serous ovarian carcinoma cell line, which was attributed to H3K27 hypermethylation [77]. The now-established succinate-mediated “hypermethylator” phenotype was also recently shown to synergize with HIF2α in establishing a mesenchymal-like phenotype and enhancing the metastatic potential of SDHB-knockout mouse chromaffin cells in vivo [75].
Yet, the hypermethylator phenotype of succinate accumulation was also found to sensitize complex II-deficient cancer cells to chemotherapy, as shown in paragangliomas and pheochromocytomas following treatment with the alkylating agent temozolomide [78]. This was attributed to the decreased expression of the DNA repair enzyme O6-methylguanine-DNA methyltransferase (MGMT) consequent to its promoter hypermethylation, as detected in patient SDHB-mutated paraganglioma and pheochromocytoma metastasis [78,79]. Similar inhibition of DNA repair processes was reported for SDHB-deficient YUNK1 kidney cell lines as a result of histone lysine demethylase (KDM) inhibition [80]. Deficiency in homologous recombination rendered SDHB-knockdown cells sensitive to synthetic lethality with poly(ADP)-ribose polymerase (PARP) inhibitor drugs olaparib and BMN-673, as shown both in vitro and in vivo, thus suggesting potential therapies for renal cell cancer (HLRCC) [80]. Taken together, these studies indicate that the succinate-induced hypermethylator phenotype acts as a double-edged sword in the physiopathology of cancers harboring SDH mutations, as it can either promote or inhibit cancer progression, depending on the genes targeted for methylation.

3.4. Metabolic Compensation Following Complex II Mutations Affecting OXPHOS

3.4.1. Preferential Usage of Glucose and Glutamine

As complex II links the ETC to the TCA cycle, alterations of its functions expectedly result in important metabolic rewiring, found to support the bioenergetic needs of tumors. In particular, murine SDHB-deficient serous ovarian carcinoma cells were shown to exhibit an unbalanced TCA cycle, with low production of fumarate and malate, owing to high succinate levels. In addition, these complex II-deficient cells preferentially used glucose for anaerobic ATP production (through glycolysis) and glutamate for fueling the TCA cycle [77]. Similar preferential incorporation of glutamine carbons in the TCA cycle has been reported in murine SDHB-knockout chromaffin cells [81]. These data suggest that SDH-deficient cells rewire their central carbon metabolism through differential metabolite usage, with glucose as a source of ATP production and glutamine as a major fuel for the TCA cycle.

3.4.2. Dependence on Pyruvate Carboxylation

In addition to its effects on glucose and glutamine metabolism, complex II deficiency leads to alterations in the metabolism of aspartate, a major precursor for non-essential amino acid, protein, and nucleotide biosynthesis. Seminal studies elegantly demonstrated that SDH-deficient cells use oxaloacetate, generated by pyruvate carboxylation, to produce aspartate, while wild-type cells rely on acetyl-CoA originating from pyruvate oxidation. These results suggest that SDH-deficient cells switch from pyruvate oxidation to carboxylation to sustain their aspartate anabolic needs. These data were further confirmed by the growth arrest of SDH-deficient cells following pyruvate carboxylation inhibition [81,82]. Taken together, these results confirm previous studies on the role of the ETC for cell proliferation, owing to its support of the synthesis of aspartate [83,84].

3.4.3. CIIlow, an Energy Consumption Regulator

An interesting study by the Neuzil team recently identified an alternative assembly of complex II, termed CIIlow, and composed only of the SDHA subunit. This CIIlow complex, detected in patients with SDHB-mutated paragangliomas, was linked to poor survival [62]. Expression of CIIlow complex in SDHB-deficient cancer cells was associated with a decrease in de novo pyrimidine synthesis and cell proliferation along with an upregulation of catabolic and salvage pathways. The expression of such a CIIlow complex may thus result from the metabolic adaptation of SDHB-deficient cancer cells, which are less proliferative, but endowed with high invasive and metastatic capacities [62].

4. Role of Complex III of the ETC for Physiological Functions

4.1. Links between Complex III Activity in Treg Cells and Immune Function

As developed above, the diverse complexes of the electron transport chain (ETC) have an essential role for the effective coupling of mitochondrial respiration and energy production. Nonetheless, the defective activity of ETC complexes can have further functional consequences that depend on mitochondria-produced metabolites and reach beyond mere mitochondrial activity. T regulatory (Treg) cells are a subset of CD4+ T cells characterized by a mitochondrial metabolism. Chandel and collaborators recently showed that the activity of complex III (ubiquinol-cytochrome c reductase) of the ETC is needed for the immune suppressive function of Treg cells and that its ablation leads to fatal inflammatory disease in treated mice, through modified metabolite production [85]. Mice whose Treg cells are deficient in complex III were generated, by deleting the gene encoding the Rieske iron-sulfur protein (RISP), one of its essential subunits. Although Treg cell proliferation and survival, as well as Foxp3 expression, were maintained in these mice, their immune suppressive capacity was lost. Increased concentrations of the 2-hydroxyglutarate (2-HG) and succinate metabolites, which are known inhibitors of the TET-family of DNA demethylases (Figure 3), were found responsible for the DNA hypermethylation phenotype, for the altered gene expression in Treg cells and, eventually, for the disruption of their immune suppressive function [85].

4.2. Links between Complex III Activity in Haematopoietic Stem Cells and Haematopoiesis

Haematopoietic stem cells (HSCs) display a mainly glycolytic phenotype. However, the importance of HSC mitochondrial activity for hematopoiesis was demonstrated by the same team, by depleting complex III in murine fetal hematopoietic stem cells. As for RISP-null Treg cells, RISP-null fetal HSCs maintained their proliferation. However, HSC impaired respiration was accompanied by a decreased NAD+/NADH ratio and, as observed for RISP-null Treg cells, levels of the metabolites 2-hydroxyglutarate and succinate were increased, as that of fumarate. The functional consequences of RISP-dependent complex III depletion and the resulting unbalanced metabolite production were the inability of HSCs to generate multipotent progenitors, leading to mice anemia and fetal death [86].

4.3. Links between Complex III Activity in Endothelial Cells and Angiogenesis

Endothelial cells (ECs) also primarily use glycolysis for ATP production, vessel sprouting, and angiogenesis. However, in addition to glucose that fuels glycolysis, endothelial cells can also take up fatty acids and glutamine that fuel the TCA cycle and, further, the ETC. By antimycin inhibition of complex III in human umbilical vein endothelial cells (HUVECs) in vitro, Chandel and collaborators showed that complex III is necessary for angiogenesis through the maintenance of NAD+/NADH ratios, aspartate concentrations, and proliferation of endothelial cells. These effects of a dysfunctional complex III in reducing NAD+/NADH ratios, proliferation, and angiogenesis were confirmed in vivo in mice harboring a deletion of the Uqcrq gene, encoding the ubiquinol-binding protein QPC, a critical subunit of complex III. They were accompanied by a loss of postnatal retinal and lung angiogenesis, as well as melanoma angiogenesis in a B16-F10 melanoma model [87].
These studies highlighted the physiological consequences of a dysfunctional complex III of the mitochondrial ETC, for immunity, hematopoiesis, or angiogenesis. Some of these effects were linked to the overproduction of metabolites like 2-hydroxyglutarate and succinate, or fumarate, which interestingly appeared to be cell-type dependent, suggesting other levels of regulation.

5. Versatile Roles of Mitochondrial Components in Physiology and Disease

5.1. The Role of Ubiquinone (Coenzyme Q10), Activated by the Mevalonate Pathway, in Cancer

Ubiquinone, also known as coenzyme Q10 (CoQ10), is an important electron carrier located in the inner mitochondrial membrane, where it transfers electrons from complexes I and II to complex III of the electron transport chain (ETC) (Figure 1). Ubiquinone is thus involved in the regulation of oxidative stress and ROS production. Ubiquinone is also a downstream metabolite of the mevalonate pathway. The mevalonate pathway uses acetyl-CoA, derived from glucose, glutamine, and/or acetate metabolism, to produce mevalonate; farnesyl-pyrophosphate (FPP); and, thereafter, different metabolites including cholesterol and ubiquinone [88] (Figure 4). The mevalonate pathway is often upregulated in cancers, which leads to increased mitochondrial concentrations of CoQ10. Statin inhibition of the mevalonate pathway is beneficial and statin treatment has been correlated with tumor cell apoptosis and reduced mortality in diverse cancers, notably breast cancer, pancreatic adenocarcinoma, and hepatocellular carcinoma [88]. As shown for pancreatic ductal adenocarcinoma (PDAC) tumor cells, ubiquinone levels are lowered by statin treatment, resulting in increased oxidative stress and ROS production. However, this oxidative stress triggers redox-active metabolic pathways aimed at lowering ROS levels, including the increased import of cystine for downstream glutathione production [89]. Therefore, the dysfunctional role of ubiquinone in the mitochondria of PDAC cells can be addressed by the concomitant targeting of the upstream mevalonate pathway (with statins) and of the metabolic glutathione-based compensation of excessive ROS production (with cystine/glutamate xCT antiporter inhibitors). As demonstrated in PDAC murine models, this effective approach triggers cancer cell death while sparing the mitochondrial functions [89].

5.2. Changing Dogmas about the Mitochondrial Role of CPT1, in both Synthesis and Oxidation of Fatty Acids

Lipids are important metabolites for membrane building and, therefore, for cell proliferation. They also provide cellular energy, act as signaling entities, and are involved in intercellular communication. All these functions allow lipid metabolism to contribute to cancer progression [90]. Both activation of fatty acid (FA) synthesis and FA oxidation have been linked to cancer progression. As recently reviewed [90], enzymes involved in fatty acid β-oxidation were overexpressed in diverse cancers and their inhibition was shown to curb cancer progression. Such is the case for CPT1 (carnitine palmitoyltransferase 1), a protein associated with the outer mitochondrial membrane, allowing the transport of long-chain fatty acids into the mitochondrial matrix. CPT1-dependent transfer of long-chain acyl groups from coenzyme A to carnitine constitutes the rate-limiting enzymatic process for the oxidative degradation of fatty acids [91].
However, CPT1 is now demonstrated to affect cancer cell proliferation by mechanisms relying on anabolic FA synthesis rather than FA β-oxidation (FAO) [92]. This novel role for CPT1 was uncovered by changing the tools to inhibit CPT1 activity. Instead of the etomoxir-dependent pharmacological inhibition of CPT1, CPT1 knock downs were performed [92]. Studying the effects of CPT1 using high concentrations of etomoxir was marred by its off-target inhibition of complex I of the electron transport chain, a feature that was reported for the BT549 breast cancer cell line [92] as well as for T cells [93,94,95]. CPT1 knock down in the BT549 breast cancer cell line demonstrated the role of CPT1 for cell proliferation, independently of FAO. CPT1 was actually found necessary for mitochondrial morphology maintenance, regulated mitochondrial lipid levels, polarized mitochondrial membrane, and efficient respiratory chain coupling [92]. As suggested by the authors, an important CPT1 function may thus be to provide long-chain fatty acids for anabolic processes in the mitochondria, needed for healthy cells and exploited in enhanced cancer cell proliferation [92].

5.3. Different Types of Mitochondria Linked to Lipid Metabolism

Attributing both antagonistic functions of FA synthesis and FA oxidation to mitochondria might be counter-intuitive. However, the recently-reported existence of metabolically-distinct mitochondrial subpopulations—one endowed with FA oxidation activity, the other involved in lipid droplet formation—could reconcile these two different mitochondrial activities [96,97]. A mitochondrial population associated with lipid droplets was characterized in brown adipose tissue (BAT). These peridroplet mitochondria (PDM) showed a high bioenergetic capacity that supported triacylglyceride (TAG) synthesis and lipid droplet formation. The protein composition of these mitochondria and the structure of their cristae appeared different from that of cytosolic mitochondria. They remained as an isolated pool different from cytosolic mitochondria owing to their low fusion-fission activity. In BAT cells isolated from cold-exposed mice, PDM abundance was reduced two fold in association with enhanced β-oxidation activity [97]. Mitochondria recruitment to lipid droplets is not limited to BAT, as also reported in mouse embryonic fibroblasts (MEFs) and in striated muscle upon nutrient deprivation, as reviewed by Benador et al. [96]. Lipid droplet accumulation has been associated with therapy resistance for various cancers, including glioma, colon, kidney, lung, and prostate cancers [98]. Whether PDM represent a population of mitochondria that can be targeted therapeutically in these pathologies will be worth investigating.

6. Mitochondria as Signaling Organelles: Functional Effects of Mitochondrial Exchange between Cells

Mitochondria are known to act as signaling organelles owing to their production of reactive oxygen species, structural compounds, and metabolites. As described above, metabolites produced by the TCA cycle can be used for the biosynthesis of macromolecules such as lipids, proteins, and nucleotides, as well as for epigenome modifications and post-translational protein changes [99]. As a result, mitochondria regulate a wide range of biological functions, including survival, growth, and differentiation.
However, the effects of mitochondria are not restricted to the cells that originally produced them, as we now know that, quite unexpectedly, mitochondria can translocate between cells. By their capacity to get transferred between cells, mitochondria can thus provide signaling cues to other cells. Numerous studies have now documented the diverse effects generated by intercellular mitochondria transfers and shown that they depend on the physiological/pathophysiological conditions and, more specifically, on the state of both mitochondria-provider and -recipient cells. The parts below provide an overview of the functional effects of intercellular mitochondria transfers in tissue repair, inflammation, and cancer progression, with a focus on the role of metabolites and structural compounds generated in the mitochondria-recipient cells.

6.1. Biological Effects of Intercellular Mitochondria Transfers

The past two decades revealed that mitochondria constantly communicate with both the cytosol and the nucleus under normal and stress conditions, thus eliciting adaptive cellular biological responses. More recently, the concept that mitochondria could exert biological effects outside from their original cells emerged from the observation that whole mitochondria, or parts of mitochondria, can translocate from one cell to the other. One example of intercellular mitochondrial communication is provided by the crosstalk of dying cells, following tissue injury, with the innate immune system of recipient cells, through the release of mitochondrial molecules including mitochondrial DNA (mtDNA), N-formyl peptides, ATP, or cardiolipin [100,101]. These mitochondrial products, recognized as damage-associated molecular patterns (DAMPs) by specific receptors on innate immune cells, are capable of directly activating the innate immune system and triggering adaptive inflammatory responses [102,103]. In addition to the crosstalk mediated by mitochondrial fragments, cells have been shown to communicate with each other by exchanging whole mitochondria.
Intercellular transfers of whole mitochondria have been reported to occur both in vitro and in vivo, and in both physiological and pathological conditions. This phenomenon is involved in several biological processes such as tissue repair, inflammation, and cancer progression (see reviews [11,12,104,105]). Mitochondria can translocate from one cell to the other by different modalities. These include thin membrane channels called tunneling nanotubes (TNTs) that ensure the connection and the mitochondria transfer between mitochondria-donor and -acceptor cells [11,12,104,105]. Mitochondria can also be transferred through their release in the extracellular space, either encapsulated inside microvesicles or as free organelles [104,106,107]. Importantly, the biological effects promoted by intercellular mitochondria transfers are highly dependent on the state of both mitochondria-donor and mitochondria-recipient cells. The outcome of these transfers can be beneficial, for instance, in tissue healing; on the other hand, they can also enhance tumor progression [11,12,104,105].
The vast majority of the studies addressing the process of intercellular mitochondria transfers have been conducted on mesenchymal stem cells (MSCs) as donor or acceptor cells for the conveyed mitochondria. These studies revealed that, compared with other cells, MSCs demonstrate a high capacity to donate their mitochondria to neighboring cells. This capacity is attributed to high expression of Miro-1, an outer mitochondrial membrane Rho-GTPase that mediates mitochondria trafficking through TNTs [108,109,110]. Mitochondria donation from MSCs to damaged or cancer cells has invariably been shown to improve recipient cell survival (see reviews [11,12,104,105]). Therefore, this process has important consequences in tissue regeneration and in cancer progression and aggressiveness (resistance to chemotherapy). MSCs have also been reported to transfer mitochondria to immune cells including T cells and macrophages. In these settings, transferred mitochondria decreased inflammation by promoting recipient immune cell differentiation towards an anti-inflammatory phenotype [111,112,113,114,115,116]. On the contrary, the transfer to T cells of mitochondria originating from myeloid cells was shown to exacerbate inflammation and to lead to asthma aggravation [117]. In addition, mitochondria transfer from pro-inflammatory monocytes to endothelial cells was associated with vessel inflammation observed in cardiovascular diseases [118]. It is worth mentioning that intercellular mitochondria transfers can be bidirectional, eliciting distinct biological responses in the two communicating cells [119]. For instance, and as mentioned above, MSCs can transfer mitochondria to damaged cells, which results in improved survival. However, MSCs can also acquire mitochondria released from damaged cells. These mitochondria function as signaling organelles that alert MSCs of a danger situation and trigger an adaptive wound-healing response [119].
The presence in the bloodstream of free or membrane-encapsulated respiratory-competent mitochondria has also been recently reported. Although the biological impact of these circulating mitochondria has yet to be determined, a role in regulating systemic inflammation through their interactions with immune cells could be speculated [107,120,121]. This pro-inflammatory role of circulating mitochondria is supported by observations made in organs from deceased patients and used for allotransplantation. In particular, circulating whole mitochondria and mtDNA levels were strongly correlated with the inflammation initiated by neutrophil activation and early allograft dysfunction, as shown for liver transplantation [120]. Besides, the concentration of free mitochondria in the blood of cancer patients correlated with metastasis, suggesting that circulating mitochondria are involved in cancer cell-to-cell communication processes [107].

6.2. OXPHOS Induced by Mitochondria Transfer in Tissue Repair and Cancer

The first discovered biological effect assigned to intercellular mitochondria transfers was their capacity to improve the survival of recipient cells, either damaged following a stress injury [122,123] or, for cancer cells, treated by chemotherapy [124]. Increased recipient cell survival was invariably correlated with enhanced OXPHOS activity and ATP production. The most convincing proof of the importance of functional OXPHOS restoration in the mitochondria-recipient cells was provided by studies using, as either mitochondria-donor or -recipient cells, ρ0 cells, which are devoid of mtDNA and have an impaired respiratory chain. The pioneering work of Prockop and collaborators thus demonstrated the OXPHOS activity restoration in ρ0 A549 lung adenocarcinoma cells owing to mitochondria transfer from cocultivated MSCs [125]. Likewise, Neuzil and collaborators found that mitochondria transfer occurred in vivo, from host stroma cells to engrafted ρ0 melanoma or ρ0 breast cancer cells, and that this process fully restored the respiration in the acceptor ρ0 cancer cells [126,127].
Beside the studies conducted with ρ0 cells, many works reported that mitochondria are transferred during cocultures from MSCs to damaged cells, including cardiomyocytes [122,128], endothelial cells [129], bronchial epithelial cells [130], corneal epithelial cells [131], and neuronal cells [132], or with leukemic, bladder, and breast cancer cells [133,134,135]. In addition, the physiological relevance of this process was strengthened by the in vivo demonstration of an increase in OXPHOS activity and ATP production following mitochondria transfer from mouse lung alveolar epithelial cells injured through exposure to LPS [123], to rotenone [110], or to cigarette smoke [130].
Finally, the prerequisite for transferred mitochondria to harbor functional respiration in order to exert their cytoprotective effects was reinforced by using ρ0 MSCs, which were unable to protect damaged cells against apoptosis [122].

6.3. Metabolic Reprogramming of MSCs and Cancer Cells by Metabolites Supplied by Mitochondra Transfer

As mentioned above, the acquisition of functional mitochondria leads to the enhancement of OXPHOS activity and ATP production in mitochondria-recipient cells. This process is of critical importance in damaged and dying cells owing to the role of ATP in restoring cellular bioenergetics. Beyond its impact on ATP levels, OXPHOS activation may also reflect an activation of the TCA cycle with an increased generation of some metabolites, with the OXPHOS and the TCA cycle being tightly coordinated [14,99]. It is still poorly documented to what extent intercellular mitochondria transfers contribute to the metabolic reprogramming of recipient cells through enhanced production of TCA cycle metabolites. However, two recent works demonstrated that TCA cycle metabolites are involved in the signaling mediated by the transferred mitochondria in recipient cancer cells and MSCs. As shown by Neuzil’s group, ρ0 melanoma and breast cancer cells lacking functional OXPHOS were unable to form tumors in mice. However, acquisition of respiratory-competent mitochondria from stromal cells allowed these ρ0 malignant cells to become highly proliferative, leading to tumor formation and progression [127]. Mechanistically, the authors demonstrated that OXPHOS-mediated ATP production was not required for tumor progression, but that, instead, OXPHOS function was critical for the generation of orotate, which is an essential intermediate for pyrimidine de novo synthesis [136,137]. In particular, OXPHOS restoration in ρ0 cells was found to re-activate the enzymatic activity of dihydroorotate dehydrogenase (DHODH), which is the ubiquinone-oxidoreductase responsible for orotate formation through dihydroorotate oxidation (Figure 1) [136,138]. As pyrimidine serves as building blocks for DNA replication and transcription, this explains why restoration of OXPHOS in ρ0 cells is essential to sustain cell proliferation [137]. In the absence of a functional respiratory chain, as is the case in ρ0 cells prior to intercellular mitochondria transfers, DHODH is unable to oxidize dihydroorotate, thus blocking pyrimidine synthesis and, as a result, arresting DNA replication and cell division [136].
Another example of the role of TCA cycle metabolites in mediating the effects of exogenous mitochondria is provided by recent observations from our laboratory indicating that the respiratory-competent mitochondria released by activated platelets can be engulfed by MSCs and can stimulate the MSC pro-angiogenic activity [139]. Citrate levels were increased in MSCs following platelet mitochondria transfer. Citrate was identified as a key metabolite initiating metabolic remodeling and functional alterations in recipient MSCs. Citrate was shown to stimulate recipient MSCs through its export to the cytosol where it fueled the ATP citrate lyase enzyme (ACLY), leading to the activation of fatty acid synthesis (Figure 4) and, subsequently, to the stimulation of the angiogenic activity of MSCs [139].

6.4. Role of DAMPs, Generated by the Transfer of Damaged Mitochondria, in Regulating Inflammation and MSC Activation

Following tissue injury, damaged cells have been reported to release their mitochondria to the extracellular environment [119,140,141]. As discussed below, the translocation of these damaged mitochondria to target cells, such as immune cells or reparative progenitors cells, functions as signaling cues, able to alert the rest of the body of a danger situation [140,142,143]. In contrast to the above-mentioned studies where transferred mitochondria needed to be fully functional to behave as pro-survival factors or as metabolite suppliers, mitochondria transferred from suffering cells mediate their effects by the means of mitochondrial fragments or DAMPs generated by recipient cells. Several DAMPs produced by recipient cells following the intercellular transfer of damaged mitochondria have been involved in inflammation [106,141]. One example is provided by the role of mitochondria released by activated platelets to neutrophils in the stimulation of the innate immune system [106]. Following their engulfment by neutrophils, platelet-derived mitochondria have been shown to have their membranes hydrolyzed by the phospholipase A2 enzyme, this mitochondria degradation leading to the production of various DAMPs, including lysophospholipids, fatty acids, and mtDNA, which are known to trigger leukocyte activation to have their membranes hydrolyzed by the phospholipase A2 enzyme [106].
More recently, Zhu and colleagues reported that both apoptotic and necrotic cells released whole mitochondria in the extracellular compartment [141]. Strikingly, the authors observed that the mitochondria released by the two types of dying cells did not support the same systemic effects following their internalization in cultivated macrophages, with mitochondria from apoptotic cells generating more inflammation than those derived from necrotic cells. Mechanistically, cardiolipin was identified, in the recipient macrophages, as the DAMP responsible for the activation of the inflammasome, a multi-protein complex able to detect danger signals and to trigger the secretion of the pro-inflammatory IL-1α and IL-18 cytokines [141]. How mitochondrial cardiolipin exerts its pro-inflammatory effects in these settings remains to be formally demonstrated. One possible explanation suggested by the authors is that the mitochondrial cardiolipin was externalized from the inner to the outer mitochondrial membrane in the apoptotic mitochondria. This conformational change would allow the binding of cardiolipin to the inflammasome sensor NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3), as previously reported [144,145].
Besides their role as pro-inflammatory mediators, mitochondria released by damaged cells can also be involved in tissue repair processes, following an injury. In particular, we recently reported that the mitochondria transfer from apoptotic endothelial or cardiac cells to MSCs constitutes a signaling messenger that triggers a cytoprotective response in the recipient MSCs, consisting of the enhanced donation of mitochondria by MSCs towards damaged cells to rescue them [119]. Interestingly, this study underlined that two DAMPs, namely reactive oxygen species (ROS) and heme, mediate the effects of damaged mitochondria in the recipient MSCs. These two DAMPS are involved at different levels of the cascade of events leading to the activation of the MSCs. First, our findings indicated that the ROS produced by damaged cells are critical regulators of the transfer of the mitochondria from the damaged cells towards MSCs, as the use of ROS scavenger abrogated both the mitochondrial transfer from the injured cells to the MSCs and the resulting MSC rescuing function [119]. The role of ROS signaling as modulator of mitochondria transfer and its associated effects have also been reported in the context of cancer, between leukemic cells and bone marrow-derived MSCs [146]. The molecular mechanisms whereby mitochondrial ROS produced by injured cells activate the cytoprotective functions of MSCs remain to be fully investigated. However, one possible mechanism is that ROS produced in excess by damaged mitochondria are sensed by MSCs as the signal triggering the degradation of dysfunctional organelles, as previously reported in other experimental settings [147,148]. The second DAMP involved in the activation of the recipient MSCs was the mitochondrial heme, released in the cytosol following the degradation of the transferred damaged mitochondria. In response to the increased level of cytosolic free heme, which has powerful pro-oxidant and toxic capacities, MSCs were found to enhance their expression of the heme oxygenase I (HO-1) enzyme to catalyze the degradation of heme, thus showing the role of HO-1 signaling in the stimulation of the pro-healing properties of MSCs [119].

7. Mitochondria and Microbiota: Two Sources of Metabolites for Cell Metabolism and Functions

7.1. Endosymbiotic Origin of the Mitochondria

Mitochondria and their host eukaryotic cells have a now well-documented endosymbiotic history showing the bacterial origin of the mitochondria. Over time, this endosymbiosis resulted in gene transfers between the mitochondrial and nuclear genomes [9,149]. As described above, this concerns the genes encoding the various subunits of the complexes of the electron transport chain, which are found in both the nucleus and the mitochondria. Many other mitochondrial proteins are encoded in the nucleus, translated in the cytoplasm, and transported to mitochondria where they exert their biological functions. Reciprocally, through the metabolites they produce, mitochondria have the capacity to exert retrograde controls on nuclear gene expression. Such is the case for alpha-ketoglutarate, 2-hydroxyglutarate, succinate, and fumarate, which regulate nuclear gene expression via the activity of DNA demethylases (Figure 3) [14].

7.2. Role of the Gut Microbiota in Physiology and Disease

Bacteria are actually present in mammals, in particular in their gut. The complex set of bacteria constituting the gut microbiota provides essential functions for the host mammalian body, as shown by numerous publications in this rapidly evolving area of research [150,151,152,153,154]. Its effects are not restricted to organs located near the gut, but extended to all organs as shown by liquid chromatography–tandem mass spectrometry (LC–MS/MS) of samples collected from 29 organs from either germ-free (GF) or specific-pathogens-free (SPF) mice [155]. The gut microbiota contributes to metabolically balanced physiological conditions, including the regulation of the immune system. The gut microbiota was found to alter the efficacy of immunotherapies based on the blockade of programmed cell death 1 (PD-1) or its ligand (PD-L1). These immune effects were observed for cancers whose primary sites were distant from the gut, like melanoma, non-small cell lung carcinoma (NSCLC), and renal cell carcinoma (RCC) [154]. In NSCLC and RCC patients, the presence of Akkermansia muciniphila in their gut microbiota was associated to a better response to PD-1 blockade [156]. Santoni and collaborators proposed that the role of A. muciniphila in promoting an immune response to anti PD-1 treatment could be attributed to the short-chain fatty acids (SCFAs), mainly acetate and propionate, produced by these bacteria. These SCFAs can activate the G-protein-coupled receptors (GPRCs), GPR41 and GPR43, with expected downstream effects on both cancer cell apoptosis and immune response [157].
The composition of the gut microbiota has also been recently tightly linked to malignancies. While the interaction of fecal bacteria with human colorectal cancers was expected, much less expected was the persistent association of bacteria like Fusobacterium nucleatum and its associated gram-negative microbiota with the colon cancer metastatic cells, at sites distant from the primary tumor. This was the case, for instance, for metastases in liver, which was associated with tumor progression [154,158]. As pointed out by the authors, the findings that the bacteria Fusobacterium travel with the primary tumor cells to their metastatic sites suggest that tumor microbiota might constitute an essential component of the tumor microenvironment [158].

7.3. Role of the Short-Chain Fatty Acids (SCFAs) Secreted by the Microbiota

Outside from physiological conditions, the gut microbiota can also either support or mitigate the metabolic functions of mammalian cells, in pathological conditions or when under metabolic stress. A lot of the effects of the microbiota, through its secreted metabolites, actually go through the functioning of their “distant cousins, the mitochondria” as recently reviewed by Agrawal and collaborators [150].
An important part of the metabolites circulating in mammals are not produced by the mitochondria-containing mammalian cells, but instead originate from the commensal bacteria found in the gut. These microbiota metabolites are mainly produced through the bacterial metabolism of dietary products and host molecules. These microbiota metabolites include lactate and the short-chain fatty acids (SCFA) acetate, propionate, and butyrate, in roughly a 3:1:1 ratio [150,152]. They provide the means for the metabolic interactions between the gut microbiota and the human host and play a key role for the overall metabolism in physiological and pathological conditions, as well as for the response to therapy treatments [150,151,152,159].
Acetate can be processed to acetyl-CoA through the enzymatic activity of the nucleocytosolic acetyl-CoA synthetase 2 (ACSS2). The overexpression of ACSS2 was observed in diverse cancers including glioblastoma and brain metastases, hepatocellular carcinoma, and breast and colorectal cancers and correlated with tumor progression both in humans and in murine models [160,161,162]. ACSS2-dependant production of Acetyl-CoA is used by the cells for lipid synthesis and acetylation of histones [161]. Therefore, as expected, high levels of acetyl-CoA lead to the increased acetylation of histones and, consequently, to the expression of genes, including those involved in cell growth. When nutrients are fully available, the acetyl-CoA comes primarily from the citrate produced from the TCA cycle in the mitochondria. However, in glioblastoma and brain metastases, the amount of the acetyl-CoA originating from acetate and feeding in the TCA cycle was substantially increased [160]. As shown by Gottlieb and collaborators [162], acetate can be processed to acetyl-CoA in conditions of low oxygen and low lipid concentrations, and thus can provide an alternative carbon source for the synthesis of fatty acids and cholesterol. In these metabolically stressed conditions, the upregulated expression of ACSS2 was responsible for cancer cell growth and survival by increasing lipid biomass. Therefore, there too, the acetate produced by the gut microbiota is expected to play a determinant role in supporting lipid biosynthesis and cancer progression.

7.4. Role of Other Microbiota-Secreted Metabolites

Outside from the SCFA major classes of microbiota-secreted metabolites, other metabolites produced by the gut bacteria from dietary products can influence the host cell metabolism. Such is the case for isovanillic acid 3-O-sulfate (IVAS), a metabolite produced by the microbiota upon consumption of the cyanidin 3-O-glucoside found in berries and that is detected in the blood. Houghton and coworkers showed that this compound increased the uptake of glucose and the metabolism of the differentiated human skeletal muscle myoblast line LHCN-M2 through an increased concentration and activity of the glucose transporter GLUT4, along with an activated PI3K/AKT signaling [153].

8. Current Therapeutic Approaches and Clinical Trials for the Treatment of Mitochondria Dysfunctions

As outlined above, diverse biological processes can account for mitochondrial dysfunctions with their load of adverse biological consequences. From the knowledge of the biological mechanisms at play, at least four types of strategies emerge for keeping control of diseases linked to mitochondrial dysfunctions:
  • directly target and repair the genes, encoded either by the mitochondrial or nuclear genomes, which are responsible for the defects;
  • use whole mitochondria to restore metabolic activities;
  • supplement the unbalanced production of metabolites from the deficient mitochondria by adding purified metabolites;
  • exploit the diversity of metabolites produced by the gut microbiota and supplement with a subset of this microbiota to provide the missing metabolites.
A number of therapeutic strategies, with their related clinical trials, have been considered in the past few years to counteract mitochondrial dysfunctions and their associated pathologies [163,164,165]. These therapies have as goals to restore the functioning of the electron transport chain, to reduce the overall oxidative stress, to supply the defective metabolites, and to enhance mitochondrial biogenesis. For most of them, however, the sought-after objective is to alleviate the symptoms rather than permanently cure the disease. The more advanced (phase III) clinical trials target genetic diseases such as mitochondrial myopathies, pyruvate dehydrogenase complex deficiency (PDCD), and Leber’s hereditary optic neuropathy (LHON) [165].

8.1. Gene Therapy for Both Mitochondrial and Nuclear-Encoded Proteins

One example of transient gene therapy is the expression of the wild-type and functional subunit 4 of NADH dehydrogenase (ND4), via adeno-associated virus (AAV) vectors and by intravitreal injection, in patients suffering from LHON disease in connection with the point mutation G11778A in their mitochondrial ND4 encoding gene [166]. The allotropic expression of the mitochondria-targeted ND4 in the retinal ganglion cell nuclei was followed by the import of the mitochondria-tagged ND4 protein in the mitochondria-imbedded complex I. This process restored complex I function and ATP production and preserved the visual function, as shown in both murine and rat models [167,168]. Parkinson’s disease has been linked to mitochondria dysfunction and to defective mitophagy-dependent mitochondria clearance in the dopaminergic neurons of the substantia nigra pars compacta. This is notably owing to the non-functional ubiquitin ligase Parkin and PTEN-induced putative kinase 1 (PINK1) in connection with the numerous mutations found in the corresponding PARK2 and PINK1 genes in patients with Parkinson’s disease [169,170,171]. The conditions for the viral delivery of the wild-type genes PARK2 and PINK1 in the brain have been set up in preclinical murine and rat models. However, conclusive preclinical data showing a therapeutic effect for Parkinson disease phenotypes are still lacking, thus presently precluding gene therapy clinical trials [169].

8.2. Therapy by Mitochondria Replacement

As detailed above, the intercellular transfers of mitochondria from MSCs to damaged recipient cells have been shown to promote beneficial wound healing effects in a wide range of pathophysiological conditions. As a result, novel mitochondria-targeted therapeutic approaches have emerged, consisting of the replacement of dysfunctional mitochondria in the injured/diseased organs through the transplantation of exogenous functional mitochondria. Mitochondrial dysfunction, leading to decreased ATP production, increased oxidative stress, apoptosis, and loss of tissue function, is the hallmark of aging and of numerous pathologies, including those caused by ischemia injury, mtDNA mutations, and metabolic disorders [172]. As a matter of fact, mitochondria transplantation has been successfully tested in several animal models for mitochondrial diseases. In particular, this therapeutic approach has been invariably shown to significantly reduce hypoxic/ischemic insult and restore tissue function following myocardial infarction [173,174,175,176], acute kidney injury [177], stroke [178,179], spinal cord injury [180,181], or optic nerve crush leading to glaucoma [182] by improving the bioenergetics and cell survival and by decreasing oxidative stress and mitochondrial DNA damages. Similarly, mitochondria transplantation has been reported to exert beneficial effects in animal models for either metabolic syndromes, including diabetic ischemic heart [183] and non-alcoholic fatty liver [184], or for neurological disorders, such as Parkinson’s disease [185,186] and schizophrenia [187]. Beyond mitochondrial disease treatment, the transplantation of exogenous mitochondrial has been evaluated in the attempt to mitigate mitochondrial dysfunction of cancer cells. This approach has been shown to attenuate the Warburg effect and to enhance the sensitivity to anti-tumoral treatments (i.e., chemotherapy or radiotherapy) in breast cancer [188] or glioma cells [189].
Overall, and similar to what is observed in the rescue of damaged cells following intercellular mitochondria transfer from MSCs, studies testing the therapeutic efficacy of mitochondria transplantation indicate that mitochondria need to be functional to exert their beneficial effects [178,182,188]. Strikingly, mitochondria used for therapeutic purposes may be of autologous, allogeneic, or xenogeneic origin, because, in all the cases, transplanted mitochondria have been reported to be non-immunogenic [178,185,186,187,188,189,190]. Although the mitochondrial transplantation procedure remains largely experimental, this technique has been successfully applied in the damaged hearts of five pediatric patients sharing ischemia-reperfusion-associated myocardial dysfunction [191].

8.3. Therapy by Metabolites Supplementation

A number of antioxidant molecules, such as quinone derivatives, are currently used for patients with mitochondrial diseases [165]. A water-soluble analog of coenzyme Q10, idebenone, was reported to improve the ATP production in the affected retinal ganglion cells from LHON patients and to improve the disease symptoms. Current efforts are devoted to modify these compounds so as to reduce their lipophilicity, with the goal of increasing their bioavailability. However, safety issues also need to be monitored as anti-oxidants such as N-acetylcysteine (NAC) and vitamin E are also likely to promote tumor progression [165].
Supplementation with coenzyme Q10 may also be a therapeutic option for patients suffering from heart failure, whose severity is associated with reduced levels of CoQ10. There have been numerous clinical trials testing CoQ10 supplementation for heart failure patients, notably based on the anti-oxidant capacity of CoQ10 and on the assumption that it could support the production of ATP in the defective cardiomyocytes, through the activation of the electron transport chain [192]. The Q-SYMBIO latest trial and longer (two-year end-point) to date suggested that the supplementation with CoQ10 reduced cardiovascular death, but will definitely need to be confirmed owing to the small number of patients tested [192].
Still, despite all current clinical trials, there are no FDA-approved treatments for curing mitochondrial diseases or even mitigating their effects [165].

8.4. Therapy by Metabolite-Producing Microbiota

Depending on the properties of the metabolites produced by specific microbiota, which can be either beneficial or harmful for humans, the corresponding microbiota can be therapeutically supplemented to the patients, or instead specifically removed with adapted antibodies. Current clinical trials actually include the supplementation of the microbiota of cancer patients undergoing strong antibiotic treatments prior to bone marrow transplantation [159]. Microbiota supplementation can be performed either by heterologous or homologous fecal microbiota transplant (FMT). The latter possibility can be used, for instance, with the microbiota of patients that can be stored during the patient microbiota-damaging treatment, such as radio- or chemo-therapy, in order to prevent further microbiota-related therapeutic side effects [159].

9. Concluding Remarks

While merely considered as the powerhouse of the cells for a long time, mitochondria have provoked great enthusiasm in the scientific community in this past decade. A wide number of recent investigations have recognized mitochondria as essential hubs governing cell fate, through the regulation of their bioenergetics and the production of various metabolites. On top of that, mitochondria were shown to have the capacity to translocate between cells. As a result, these fascinating organelles control multifaceted biological processes including wound healing and inflammation and their dysfunctions are associated with a broad range of diseases including inherited mitochondrial disorders and metabolic diseases such as diabetes and cancer. The full understanding of the fine metabolic mechanisms that allow mitochondria to exert their biological/signaling effects will undoubtedly need further investigations. However, the rapid scientific progress in this field highlight the key role played by the metabolites produced through the tightly interconnected ETC and TCA cycles and by mitochondrial components like DAMPs. Although additional research is needed to identify the specific metabolites/mitochondrial components responsible for the given effects of mitochondria, several therapeutic options can now be envisioned to treat mitochondrial diseases and cancer and to promote wound healing in injured/degenerative tissues, by focusing attention on whole mitochondria as well as on the metabolites/compounds they produce.

Author Contributions

Writing, J.N., A.-M.R. and M.-L.V. Figure design with BioRender.com, J.N. All authors have read and agreed to the published version of the manuscript.

Funding

J.N. is supported by a PhD fellowship from the French Ministry of Research. This work was supported by a grant from the Ligue Nationale contre le Cancer-Comité du Gard and from the Fondation ARC pour la recherche sur le cancer (M.-L.V.).

Acknowledgments

A.-M.R. and M.-L.V. are staff scientists from the National Institute for Health and Medical Research (INSERM) and the National Center for scientific research (CNRS), respectively.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AAVAdeno-associated virus
ACSS2Acetyl-CoA synthetase 2
AGCAspartate-glutamate carrier
aKGAlpha-ketoglutarate
AKTProtein kinase B
ALALipoic acid (1,2-dithiolane-3-pentanoic acid or thioctic acid)
ATPAdenosine triphosphate
BATBrown adipose tissue
BCL-XLB-cell lymphoma-extra large
CoQ10Coenzyme Q10
CPEOChronic progressive external ophthalmoplegia
CPT1Carnitine palmitoyltransferase 1
DAMPMitochondrial damage-associated molecular patterns
dGTPDeoxyguanosine triphosphate
DHODHDihydroorotate dehydrogenase
DNAJA4Dnaj heat shock protein family (Hsp40) member A4
dNTPDeoxynucleotide triphosphate
EMTEpithelial-to-mesenchymal transition
ERKExtracellular signal-regulated kinase
ETCElectron transport chain
FADFlavin adenine dinucleotide
FAOFatty acids beta-oxidation
FMTFecal microbiota transplant
FOXP3Forkhead box P3
FPPFarnesyl pyrophosphate (or diphosphate)
GAPDHGlyceraldehyde 3-phosphate dehydrogenase
GFGerm-free
GOT2Glutamate-oxaloacetate transaminase
Gpx4Glutathione peroxidase 4
HCCHepatocellular carcinoma
2-HG2-hydroxyglutarate
HIFHypoxia-inducible factor
HLRCCHereditary leiomyomatosis and renal cell cancer
5hmc5-hydroxymethylcytosine
HMGCRHMG-CoA reductase
HO-1Heme oxygenase- 1
JmjcJumonji C
KDMHistone lysine demethylase
KRT19Keratin 19
KSSKearns–Sayre syndrome
LC–MS/MSLiquid chromatography–tandem mass spectrometry
LDHALactate dehydrogenase A
LHONLeber’s hereditary optic neuropathy
5mc5-methylcytosine
MCL1Induced myeloid leukemia cell differentiation protein
MEFsMouse embryonic fibroblasts
MELASMitochondrial encephalomyopathy with lactic acidosis and stroke-like episodes
MERRFMyoclonic epilepsy with ragged-red fibers
MGMTO-6-methylguanine-DNA methyltransferase
MIDDMaternally inherited diabetes and deafness
MSCMesenchymal stem cell
mtDNAMitochondrial DNA
MTHFDMethylenetetrahydrofolate dehydrogenase
NACN-acetylcysteine
NADHNicotinamide adenine dinucleotide
NARPSyndrome neuropathy, ataxia, and retinitis pigmentosa
ND4NADH dehydrogenase subunit 4
nDNANuclear DNA
NOXNicotinamide adenine dinucleotide phosphate oxidase
NSCLCNon-small-cell lung carcinoma
2-OG2-oxoglutarate
OXPHOSOxidative phosphorylation
PARPPoly(ADP)-ribose polymerase
PCPyruvate carboxylase
PD1Programmed cell death 1
PDACPancreatic ductal adenocarcinoma
PDCDPyruvate dehydrogenase complex deficiency
PDHPyruvate dehydrogenase
PDK2Pyruvate dehydrogenase kinase 2
PDMPeridroplet mitochondria
PFKPPhosphofructokinase
PGK1Phosphoglycerate kinase
PHDProlyl hydroxylase
PI3KPhosphatidylinositol 3-kinase
PINK1PTEN-induced putative kinase 1
PPPPentose phosphate pathway
pVHLVon Hippel–Lindau protein
RCCRenal cell carcinoma
RISPRieske iron-sulfur protein
ROSReactive oxygen species
SCFAShort-chain fatty acid
SDHSuccinate dehydrogenase
SLC2ASolute carrier family 2
SMADMothers against decapentaplegic homolog 1
SNAILSnail family transcriptional repressor
SODSuperoxide dismutase
SPFSpecific pathogens-free
SPOCKSPARC (Osteonectin), Cwcv, and Kazal like domains proteoglycan
STATSignal transducer and activator of transcription
TAGTriacylglyceride
TCATricarboxylic acid
TETTen-eleven-translocation
TGFβTransforming growth factor beta
TNTTunneling nanotube
TregRegulatory T cell
VEGFVascular endothelial growth factor

References

  1. Gray, M.W. The endosymbiont hypothesis revisited. Int. Rev. Cytol. 1992, 141, 233–357. [Google Scholar] [CrossRef] [PubMed]
  2. Taanman, J.W. The mitochondrial genome: Structure, transcription, translation and replication. Biochim. Biophys. Acta 1999, 1410, 103–123. [Google Scholar] [CrossRef] [Green Version]
  3. Eisenberg-Bord, M.; Schuldiner, M. Ground control to major TOM: Mitochondria-nucleus communication. FEBS J. 2017, 284, 196–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Tzameli, I. The evolving role of mitochondria in metabolism. Trends Endocrinol. Metab. Tem. 2012, 23, 417–419. [Google Scholar] [CrossRef]
  5. Spinelli, J.B.; Haigis, M.C. The multifaceted contributions of mitochondria to cellular metabolism. Nat. Cell Biol. 2018, 20, 745–754. [Google Scholar] [CrossRef]
  6. Bock, F.J.; Tait, S.W.G. Mitochondria as multifaceted regulators of cell death. Nat. Rev. Mol. Cell Biol. 2020, 21, 85–100. [Google Scholar] [CrossRef]
  7. de Souza Breda, C.N.; Davanzo, G.G.; Basso, P.J.; Saraiva Câmara, N.O.; Moraes-Vieira, P.M.M. Mitochondria as central hub of the immune system. Redox Biol. 2019, 26, 101255. [Google Scholar] [CrossRef]
  8. Papa, L.; Djedaini, M.; Hoffman, R. Mitochondrial Role in Stemness and Differentiation of Hematopoietic Stem Cells. Stem Cells Int. 2019, 2019, 4067162. [Google Scholar] [CrossRef] [Green Version]
  9. Roger, A.J.; Muñoz-Gómez, S.A.; Kamikawa, R. The Origin and Diversification of Mitochondria. Curr. Biol. 2017, 27, R1177–R1192. [Google Scholar] [CrossRef] [Green Version]
  10. Calvo, S.E.; Mootha, V.K. The Mitochondrial Proteome and Human Disease. Annu. Rev. Genom. Hum. Genet. 2010, 11, 25–44. [Google Scholar] [CrossRef] [Green Version]
  11. Rodriguez, A.-M.; Nakhle, J.; Griessinger, E.; Vignais, M.-L. Intercellular mitochondria trafficking highlighting the dual role of mesenchymal stem cells as both sensors and rescuers of tissue injury. Cell Cycle 2018, 17, 1–25. [Google Scholar] [CrossRef] [PubMed]
  12. Hekmatshoar, Y.; Nakhle, J.; Galloni, M.; Vignais, M.-L. The role of metabolism and tunneling nanotube-mediated intercellular mitochondria exchange in cancer drug resistance. Biochem. J. 2018, 475, 2305–2328. [Google Scholar] [CrossRef] [PubMed]
  13. Frezza, C. Mitochondrial metabolites: Undercover signalling molecules. Interface Focus 2017, 7, 20160100. [Google Scholar] [CrossRef] [PubMed]
  14. Martínez-Reyes, I.; Chandel, N.S. Mitochondrial TCA cycle metabolites control physiology and disease. Nat. Commun. 2020, 11, 102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Grazioli, S.; Pugin, J. Mitochondrial Damage-Associated Molecular Patterns: From Inflammatory Signaling to Human Diseases. Front. Immunol. 2018, 9, 832. [Google Scholar] [CrossRef]
  16. Bhatti, J.S.; Bhatti, G.K.; Reddy, P.H. Mitochondrial dysfunction and oxidative stress in metabolic disorders—A step towards mitochondria based therapeutic strategies. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1066–1077. [Google Scholar] [CrossRef]
  17. Gorman, G.S.; Chinnery, P.F.; DiMauro, S.; Hirano, M.; Koga, Y.; McFarland, R.; Suomalainen, A.; Thorburn, D.R.; Zeviani, M.; Turnbull, D.M. Mitochondrial diseases. Nat. Rev. Dis. Primer 2016, 2, 16080. [Google Scholar] [CrossRef]
  18. Zhu, Y.; Dean, A.E.; Horikoshi, N.; Heer, C.; Spitz, D.R.; Gius, D. Emerging evidence for targeting mitochondrial metabolic dysfunction in cancer therapy. J. Clin. Investig. 2018, 128, 3682–3691. [Google Scholar] [CrossRef]
  19. Mukherjee, S.; Ghosh, A. Molecular mechanism of mitochondrial respiratory chain assembly and its relation to mitochondrial diseases. Mitochondrion 2020, S1567724919303484. [Google Scholar] [CrossRef]
  20. Zinovkina, L.A. Mechanisms of Mitochondrial DNA Repair in Mammals. Biochem. Mosc. 2018, 83, 233–249. [Google Scholar] [CrossRef]
  21. Sharma, P.; Sampath, H. Mitochondrial DNA Integrity: Role in Health and Disease. Cells 2019, 8, 100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Russell, O.M.; Gorman, G.S.; Lightowlers, R.N.; Turnbull, D.M. Mitochondrial Diseases: Hope for the Future. Cell 2020, 181, 168–188. [Google Scholar] [CrossRef] [PubMed]
  23. Pfeffer, G.; Majamaa, K.; Turnbull, D.M.; Thorburn, D.; Chinnery, P.F. Treatment for mitochondrial disorders. Cochrane Database Syst. Rev. 2012, CD004426. [Google Scholar] [CrossRef] [PubMed]
  24. Gammage, P.A.; Frezza, C. Mitochondrial DNA: The overlooked oncogenome? BMC Biol. 2019, 17, 53. [Google Scholar] [CrossRef] [Green Version]
  25. Roth, K.G.; Mambetsariev, I.; Kulkarni, P.; Salgia, R. The Mitochondrion as an Emerging Therapeutic Target in Cancer. Trends Mol. Med. 2020, 26, 119–134. [Google Scholar] [CrossRef] [PubMed]
  26. Zong, W.-X.; Rabinowitz, J.D.; White, E. Mitochondria and Cancer. Mol. Cell 2016, 61, 667–676. [Google Scholar] [CrossRef] [Green Version]
  27. Ju, Y.S.; Alexandrov, L.B.; Gerstung, M.; Martincorena, I.; Nik-Zainal, S.; Ramakrishna, M.; Davies, H.R.; Papaemmanuil, E.; Gundem, G.; Shlien, A.; et al. Origins and functional consequences of somatic mitochondrial DNA mutations in human cancer. Elife 2014, 3, e02935. [Google Scholar] [CrossRef] [Green Version]
  28. Stewart, J.B.; Alaei-Mahabadi, B.; Sabarinathan, R.; Samuelsson, T.; Gorodkin, J.; Gustafsson, C.M.; Larsson, E. Simultaneous DNA and RNA Mapping of Somatic Mitochondrial Mutations across Diverse Human Cancers. PLoS Genet. 2015, 11, e1005333. [Google Scholar] [CrossRef] [Green Version]
  29. Zickermann, V.; Wirth, C.; Nasiri, H.; Siegmund, K.; Schwalbe, H.; Hunte, C.; Brandt, U. Mechanistic insight from the crystal structure of mitochondrial complex I. Science 2015, 347, 44–49. [Google Scholar] [CrossRef] [Green Version]
  30. Zhu, J.; Vinothkumar, K.R.; Hirst, J. Structure of mammalian respiratory complex I. Nature 2016, 536, 354–358. [Google Scholar] [CrossRef] [Green Version]
  31. Frazier, A.E.; Thorburn, D.R.; Compton, A.G. Mitochondrial energy generation disorders: Genes, mechanisms, and clues to pathology. J. Biol. Chem. 2019, 294, 5386–5395. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kim, H.K.; Noh, Y.H.; Nilius, B.; Ko, K.S.; Rhee, B.D.; Kim, N.; Han, J. Current and upcoming mitochondrial targets for cancer therapy. Semin. Cancer Biol. 2017, 47, 154–167. [Google Scholar] [CrossRef] [PubMed]
  33. Zhou, S.; Kachhap, S.; Sun, W.; Wu, G.; Chuang, A.; Poeta, L.; Grumbine, L.; Mithani, S.K.; Chatterjee, A.; Koch, W.; et al. Frequency and phenotypic implications of mitochondrial DNA mutations in human squamous cell cancers of the head and neck. Proc. Natl. Acad. Sci. USA 2007, 104, 7540–7545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sun, W.; Zhou, S.; Chang, S.S.; McFate, T.; Verma, A.; Califano, J.A. Mitochondrial Mutations Contribute to HIF1 Accumulation via Increased Reactive Oxygen Species and Up-regulated Pyruvate Dehydrogenease Kinase 2 in Head and Neck Squamous Cell Carcinoma. Clin. Cancer Res. 2009, 15, 476–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Imanishi, H.; Hattori, K.; Wada, R.; Ishikawa, K.; Fukuda, S.; Takenaga, K.; Nakada, K.; Hayashi, J.-I. Mitochondrial DNA Mutations Regulate Metastasis of Human Breast Cancer Cells. PLoS ONE 2011, 6, e23401. [Google Scholar] [CrossRef] [Green Version]
  36. Yu, Y.; Lv, F.; Lin, H.; Qian, G.; Jiang, Y.S.; Pang, L.X.; Wang, Y.P.; Wang, X.F.; Kang, Y.M.; Li, C.B.; et al. Mitochondrial ND3 G10398A mutation: A biomarker for breast cancer. Genet. Mol. Res. 2015, 14, 17426–17431. [Google Scholar] [CrossRef]
  37. Gasparre, G.; Porcelli, A.M.; Bonora, E.; Pennisi, L.F.; Toller, M.; Iommarini, L.; Ghelli, A.; Moretti, M.; Betts, C.M.; Martinelli, G.N.; et al. Disruptive mitochondrial DNA mutations in complex I subunits are markers of oncocytic phenotype in thyroid tumors. Proc. Natl. Acad. Sci. USA 2007, 104, 9001–9006. [Google Scholar] [CrossRef] [Green Version]
  38. Evangelisti, C.; de Biase, D.; Kurelac, I.; Ceccarelli, C.; Prokisch, H.; Meitinger, T.; Caria, P.; Vanni, R.; Romeo, G.; Tallini, G.; et al. A mutation screening of oncogenes, tumor suppressor gene TP53 and nuclear encoded mitochondrial complex I genes in oncocytic thyroid tumors. BMC Cancer 2015, 15, 157. [Google Scholar] [CrossRef] [Green Version]
  39. Gopal, R.K.; Kübler, K.; Calvo, S.E.; Polak, P.; Livitz, D.; Rosebrock, D.; Sadow, P.M.; Campbell, B.; Donovan, S.E.; Amin, S.; et al. Widespread Chromosomal Losses and Mitochondrial DNA Alterations as Genetic Drivers in Hürthle Cell Carcinoma. Cancer Cell 2018, 34, 242–255.e5. [Google Scholar] [CrossRef] [Green Version]
  40. Philley, J.V.; Kannan, A.; Qin, W.; Sauter, E.R.; Ikebe, M.; Hertweck, K.L.; Troyer, D.A.; Semmes, O.J.; Dasgupta, S. Complex-I Alteration and Enhanced Mitochondrial Fusion Are Associated With Prostate Cancer Progression: Mitochondrial DNA Alterations in Prostate Cancer. J. Cell. Physiol. 2016, 231, 1364–1374. [Google Scholar] [CrossRef] [Green Version]
  41. Kim, H.; Komiyama, T.; Inomoto, C.; Kamiguchi, H.; Kajiwara, H.; Kobayashi, H.; Nakamura, N.; Terachi, T. Mutations in the Mitochondrial ND1 Gene Are Associated with Postoperative Prognosis of Localized Renal Cell Carcinoma. Int. J. Mol. Sci. 2016, 17, 2049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Gao, W.; Xu, K.; Li, P.; Tang, B. Functional roles of superoxide and hydrogen peroxide generated by mitochondrial DNA mutation in regulating tumorigenicity of HepG2 cells: Roles of Superoxide and Hydrogen Peroxide. Cell Biochem. Funct. 2011, 29, 400–407. [Google Scholar] [CrossRef] [PubMed]
  43. King, M.; Attardi, G. Human cells lacking mtDNA: Repopulation with exogenous mitochondria by complementation. Science 1989, 246, 500–503. [Google Scholar] [CrossRef] [PubMed]
  44. Hofhaus, G.; Attardi, G. Efficient selection and characterization of mutants of a human cell line which are defective in mitochondrial DNA-encoded subunits of respiratory NADH dehydrogenase. Mol. Cell. Biol. 1995, 15, 964–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Polyak, K.; Li, Y.; Zhu, H.; Lengauer, C.; Willson, J.K.; Markowitz, S.D.; Trush, M.A.; Kinzler, K.W.; Vogelstein, B. Somatic mutations of the mitochondrial genome in human colorectal tumours. Nat. Genet. 1998, 20, 291–293. [Google Scholar] [CrossRef]
  46. Park, J.S.; Sharma, L.K.; Li, H.; Xiang, R.; Holstein, D.; Wu, J.; Lechleiter, J.; Naylor, S.L.; Deng, J.J.; Lu, J.; et al. A heteroplasmic, not homoplasmic, mitochondrial DNA mutation promotes tumorigenesis via alteration in reactive oxygen species generation and apoptosis. Hum. Mol. Genet. 2009, 18, 1578–1589. [Google Scholar] [CrossRef]
  47. Sharma, L.K.; Fang, H.; Liu, J.; Vartak, R.; Deng, J.; Bai, Y. Mitochondrial respiratory complex I dysfunction promotes tumorigenesis through ROS alteration and AKT activation. Hum. Mol. Genet. 2011, 20, 4605–4616. [Google Scholar] [CrossRef] [Green Version]
  48. Bonora, E.; Porcelli, A.M.; Gasparre, G.; Biondi, A.; Ghelli, A.; Carelli, V.; Baracca, A.; Tallini, G.; Martinuzzi, A.; Lenaz, G.; et al. Defective Oxidative Phosphorylation in Thyroid Oncocytic Carcinoma Is Associated with Pathogenic Mitochondrial DNA Mutations Affecting Complexes I and III. Cancer Res. 2006, 66, 6087–6096. [Google Scholar] [CrossRef] [Green Version]
  49. Iommarini, L.; Kurelac, I.; Capristo, M.; Calvaruso, M.A.; Giorgio, V.; Bergamini, C.; Ghelli, A.; Nanni, P.; De Giovanni, C.; Carelli, V.; et al. Different mtDNA mutations modify tumor progression in dependence of the degree of respiratory complex I impairment. Hum. Mol. Genet. 2014, 23, 1453–1466. [Google Scholar] [CrossRef] [Green Version]
  50. Calabrese, C.; Iommarini, L.; Kurelac, I.; Calvaruso, M.A.; Capristo, M.; Lollini, P.-L.; Nanni, P.; Bergamini, C.; Nicoletti, G.; Giovanni, C.D.; et al. Respiratory complex I is essential to induce a Warburg profile in mitochondria-defective tumor cells. Cancer Metab. 2013, 1, 11. [Google Scholar] [CrossRef] [Green Version]
  51. Ishikawa, K.; Takenaga, K.; Akimoto, M.; Koshikawa, N.; Yamaguchi, A.; Imanishi, H.; Nakada, K.; Honma, Y.; Hayashi, J.-I. ROS-Generating Mitochondrial DNA Mutations Can Regulate Tumor Cell Metastasis. Science 2008, 320, 661–664. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Cruz-Bermúdez, A.; Vallejo, C.G.; Vicente-Blanco, R.J.; Gallardo, M.E.; Fernández-Moreno, M.Á.; Quintanilla, M.; Garesse, R. Enhanced tumorigenicity by mitochondrial DNA mild mutations. Oncotarget 2015, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Singh, R.K.; Saini, S.; Verma, D.; Kalaiarasan, P.; Bamezai, R.N.K. Mitochondrial ND5 mutation mediated elevated ROS regulates apoptotic pathway epigenetically in a P53 dependent manner for generating pro-cancerous phenotypes. Mitochondrion 2017, 35, 35–43. [Google Scholar] [CrossRef] [PubMed]
  54. Schöpf, B.; Weissensteiner, H.; Schäfer, G.; Fazzini, F.; Charoentong, P.; Naschberger, A.; Rupp, B.; Fendt, L.; Bukur, V.; Giese, I.; et al. OXPHOS remodeling in high-grade prostate cancer involves mtDNA mutations and increased succinate oxidation. Nat. Commun. 2020, 11, 1487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Amelio, I.; Cutruzzolá, F.; Antonov, A.; Agostini, M.; Melino, G. Serine and glycine metabolism in cancer. Trends Biochem. Sci. 2014, 39, 191–198. [Google Scholar] [CrossRef] [PubMed]
  56. Ducker, G.S.; Rabinowitz, J.D. One-Carbon Metabolism in Health and Disease. Cell Metab. 2017, 25, 27–42. [Google Scholar] [CrossRef] [Green Version]
  57. Yang, L.; Garcia Canaveras, J.C.; Chen, Z.; Wang, L.; Liang, L.; Jang, C.; Mayr, J.A.; Zhang, Z.; Ghergurovich, J.M.; Zhan, L.; et al. Serine Catabolism Feeds NADH when Respiration Is Impaired. Cell Metab. 2020, 31, 809–821.e6. [Google Scholar] [CrossRef]
  58. To, T.-L.; Cuadros, A.M.; Shah, H.; Hung, W.H.W.; Li, Y.; Kim, S.H.; Rubin, D.H.F.; Boe, R.H.; Rath, S.; Eaton, J.K.; et al. A Compendium of Genetic Modifiers of Mitochondrial Dysfunction Reveals Intra-organelle Buffering. Cell 2019, 179, 1222–1238.e17. [Google Scholar] [CrossRef]
  59. Rutter, J.; Winge, D.R.; Schiffman, J.D. Succinate dehydrogenase—Assembly, regulation and role in human disease. Mitochondrion 2010, 10, 393–401. [Google Scholar] [CrossRef] [Green Version]
  60. Flavahan, W.A.; Drier, Y.; Johnstone, S.E.; Hemming, M.L.; Tarjan, D.R.; Hegazi, E.; Shareef, S.J.; Javed, N.M.; Raut, C.P.; Eschle, B.K.; et al. Altered chromosomal topology drives oncogenic programs in SDH-deficient GISTs. Nature 2019, 575, 229–233. [Google Scholar] [CrossRef]
  61. Aghamir, S.M.K.; Heshmat, R.; Ebrahimi, M.; Ketabchi, S.E.; Parichehreh Dizaji, S.; Khatami, F. The Impact of Succinate Dehydrogenase Gene (SDH) Mutations In Renal Cell Carcinoma (RCC): A Systematic Review. Oncotargets Ther. 2019, 12, 7929–7940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Bezawork-Geleta, A.; Wen, H.; Dong, L.; Yan, B.; Vider, J.; Boukalova, S.; Krobova, L.; Vanova, K.; Zobalova, R.; Sobol, M.; et al. Alternative assembly of respiratory complex II connects energy stress to metabolic checkpoints. Nat. Commun. 2018, 9, 2221. [Google Scholar] [CrossRef] [PubMed]
  63. Dalla Pozza, E.; Dando, I.; Pacchiana, R.; Liboi, E.; Scupoli, M.T.; Donadelli, M.; Palmieri, M. Regulation of succinate dehydrogenase and role of succinate in cancer. Semin. Cell Dev. Biol. 2020, 98, 4–14. [Google Scholar] [CrossRef] [PubMed]
  64. Gimenez-Roqueplo, A.P.; Favier, J.; Rustin, P.; Mourad, J.J.; Plouin, P.F.; Corvol, P.; Rötig, A.; Jeunemaitre, X. The R22X mutation of the SDHD gene in hereditary paraganglioma abolishes the enzymatic activity of complex II in the mitochondrial respiratory chain and activates the hypoxia pathway. Am. J. Hum. Genet. 2001, 69, 1186–1197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Selak, M.A.; Armour, S.M.; MacKenzie, E.D.; Boulahbel, H.; Watson, D.G.; Mansfield, K.D.; Pan, Y.; Simon, M.C.; Thompson, C.B.; Gottlieb, E. Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-alpha prolyl hydroxylase. Cancer Cell 2005, 7, 77–85. [Google Scholar] [CrossRef] [Green Version]
  66. Movafagh, S.; Crook, S.; Vo, K. Regulation of Hypoxia-Inducible Factor-1α by Reactive Oxygen Species: New Developments in an Old Debate: Regulation of Hypoxia-Inducible Factor-1α. J. Cell. Biochem. 2015, 116, 696–703. [Google Scholar] [CrossRef]
  67. Favier, J.; Amar, L.; Gimenez-Roqueplo, A.-P. Paraganglioma and phaeochromocytoma: From genetics to personalized medicine. Nat. Rev. Endocrinol. 2015, 11, 101–111. [Google Scholar] [CrossRef]
  68. Kluckova, K.; Tennant, D.A. Metabolic implications of hypoxia and pseudohypoxia in pheochromocytoma and paraganglioma. Cell Tissue Res. 2018, 372, 367–378. [Google Scholar] [CrossRef] [Green Version]
  69. Tsukada, Y.; Fang, J.; Erdjument-Bromage, H.; Warren, M.E.; Borchers, C.H.; Tempst, P.; Zhang, Y. Histone demethylation by a family of JmjC domain-containing proteins. Nature 2006, 439, 811–816. [Google Scholar] [CrossRef]
  70. Ito, S.; D’Alessio, A.C.; Taranova, O.V.; Hong, K.; Sowers, L.C.; Zhang, Y. Role of Tet proteins in 5mC to 5hmC conversion, ES-cell self-renewal and inner cell mass specification. Nature 2010, 466, 1129–1133. [Google Scholar] [CrossRef] [Green Version]
  71. Melamed, P.; Yosefzon, Y.; David, C.; Tsukerman, A.; Pnueli, L. Tet Enzymes, Variants, and Differential Effects on Function. Front. Cell Dev. Biol. 2018, 6, 22. [Google Scholar] [CrossRef] [Green Version]
  72. Xiao, M.; Yang, H.; Xu, W.; Ma, S.; Lin, H.; Zhu, H.; Liu, L.; Liu, Y.; Yang, C.; Xu, Y.; et al. Inhibition of -KG-dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors. Genes Dev. 2012, 26, 1326–1338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Killian, J.K.; Kim, S.Y.; Miettinen, M.; Smith, C.; Merino, M.; Tsokos, M.; Quezado, M.; Smith, W.I.; Jahromi, M.S.; Xekouki, P.; et al. Succinate dehydrogenase mutation underlies global epigenomic divergence in gastrointestinal stromal tumor. Cancer Discov. 2013, 3, 648–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Letouzé, E.; Martinelli, C.; Loriot, C.; Burnichon, N.; Abermil, N.; Ottolenghi, C.; Janin, M.; Menara, M.; Nguyen, A.T.; Benit, P.; et al. SDH mutations establish a hypermethylator phenotype in paraganglioma. Cancer Cell 2013, 23, 739–752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Morin, A.; Goncalves, J.; Moog, S.; Castro-Vega, L.-J.; Job, S.; Buffet, A.; Fontenille, M.-J.; Woszczyk, J.; Gimenez-Roqueplo, A.-P.; Letouzé, E.; et al. TET-Mediated Hypermethylation Primes SDH-Deficient Cells for HIF2α-Driven Mesenchymal Transition. Cell Rep. 2020, 30, 4551–4566. [Google Scholar] [CrossRef] [PubMed]
  76. Loriot, C.; Domingues, M.; Berger, A.; Menara, M.; Ruel, M.; Morin, A.; Castro-Vega, L.-J.; Letouzé, É.; Martinelli, C.; Bemelmans, A.-P.; et al. Deciphering the molecular basis of invasiveness in Sdhb-deficient cells. Oncotarget 2015, 6, 32955–32965. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Aspuria, P.-J.P.; Lunt, S.Y.; Väremo, L.; Vergnes, L.; Gozo, M.; Beach, J.A.; Salumbides, B.; Reue, K.; Wiedemeyer, W.R.; Nielsen, J.; et al. Succinate dehydrogenase inhibition leads to epithelial-mesenchymal transition and reprogrammed carbon metabolism. Cancer Metab. 2014, 2, 21. [Google Scholar] [CrossRef]
  78. Hadoux, J.; Favier, J.; Scoazec, J.-Y.; Leboulleux, S.; Al Ghuzlan, A.; Caramella, C.; Déandreis, D.; Borget, I.; Loriot, C.; Chougnet, C.; et al. SDHB mutations are associated with response to temozolomide in patients with metastatic pheochromocytoma or paraganglioma. Int. J. Cancer 2014, 135, 2711–2720. [Google Scholar] [CrossRef]
  79. Fan, C.-H.; Liu, W.-L.; Cao, H.; Wen, C.; Chen, L.; Jiang, G. O6-methylguanine DNA methyltransferase as a promising target for the treatment of temozolomide-resistant gliomas. Cell Death Dis. 2013, 4, e876. [Google Scholar] [CrossRef] [Green Version]
  80. Sulkowski, P.L.; Sundaram, R.K.; Oeck, S.; Corso, C.D.; Liu, Y.; Noorbakhsh, S.; Niger, M.; Boeke, M.; Ueno, D.; Kalathil, A.N.; et al. Krebs-cycle-deficient hereditary cancer syndromes are defined by defects in homologous-recombination DNA repair. Nat. Genet. 2018, 50, 1086–1092. [Google Scholar] [CrossRef]
  81. Lussey-Lepoutre, C.; Hollinshead, K.E.R.; Ludwig, C.; Menara, M.; Morin, A.; Castro-Vega, L.-J.; Parker, S.J.; Janin, M.; Martinelli, C.; Ottolenghi, C.; et al. Loss of succinate dehydrogenase activity results in dependency on pyruvate carboxylation for cellular anabolism. Nat. Commun. 2015, 6, 8784. [Google Scholar] [CrossRef] [PubMed]
  82. Cardaci, S.; Zheng, L.; MacKay, G.; van den Broek, N.J.F.; MacKenzie, E.D.; Nixon, C.; Stevenson, D.; Tumanov, S.; Bulusu, V.; Kamphorst, J.J.; et al. Pyruvate carboxylation enables growth of SDH-deficient cells by supporting aspartate biosynthesis. Nat. Cell Biol. 2015, 17, 1317–1326. [Google Scholar] [CrossRef] [PubMed]
  83. Birsoy, K.; Wang, T.; Chen, W.W.; Freinkman, E.; Abu-Remaileh, M.; Sabatini, D.M. An Essential Role of the Mitochondrial Electron Transport Chain in Cell Proliferation Is to Enable Aspartate Synthesis. Cell 2015, 162, 540–551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Sullivan, L.B.; Gui, D.Y.; Hosios, A.M.; Bush, L.N.; Freinkman, E.; Vander Heiden, M.G. Supporting Aspartate Biosynthesis Is an Essential Function of Respiration in Proliferating Cells. Cell 2015, 162, 552–563. [Google Scholar] [CrossRef] [Green Version]
  85. Weinberg, S.E.; Singer, B.D.; Steinert, E.M.; Martinez, C.A.; Mehta, M.M.; Martínez-Reyes, I.; Gao, P.; Helmin, K.A.; Abdala-Valencia, H.; Sena, L.A.; et al. Mitochondrial complex III is essential for suppressive function of regulatory T cells. Nature 2019, 565, 495–499. [Google Scholar] [CrossRef]
  86. Ansó, E.; Weinberg, S.E.; Diebold, L.P.; Thompson, B.J.; Malinge, S.; Schumacker, P.T.; Liu, X.; Zhang, Y.; Shao, Z.; Steadman, M.; et al. The mitochondrial respiratory chain is essential for haematopoietic stem cell function. Nat. Cell Biol. 2017, 19, 614–625. [Google Scholar] [CrossRef]
  87. Diebold, L.P.; Gil, H.J.; Gao, P.; Martinez, C.A.; Weinberg, S.E.; Chandel, N.S. Mitochondrial complex III is necessary for endothelial cell proliferation during angiogenesis. Nat. Metab. 2019, 1, 158–171. [Google Scholar] [CrossRef]
  88. Mullen, P.J.; Yu, R.; Longo, J.; Archer, M.C.; Penn, L.Z. The interplay between cell signalling and the mevalonate pathway in cancer. Nat. Rev. Cancer 2016, 16, 718–731. [Google Scholar] [CrossRef]
  89. McGregor, G.H.; Campbell, A.D.; Fey, S.K.; Tumanov, S.; Sumpton, D.; Blanco, G.R.; Mackay, G.; Nixon, C.; Vazquez, A.; Sansom, O.J.; et al. Targeting the Metabolic Response to Statin-Mediated Oxidative Stress Produces a Synergistic Antitumor Response. Cancer Res. 2020, 80, 175–188. [Google Scholar] [CrossRef] [Green Version]
  90. Snaebjornsson, M.T.; Janaki-Raman, S.; Schulze, A. Greasing the Wheels of the Cancer Machine: The Role of Lipid Metabolism in Cancer. Cell Metab. 2020, 31, 62–76. [Google Scholar] [CrossRef]
  91. Carracedo, A.; Cantley, L.C.; Pandolfi, P.P. Cancer metabolism: Fatty acid oxidation in the limelight. Nat. Rev. Cancer 2013, 13, 227–232. [Google Scholar] [CrossRef]
  92. Yao, C.-H.; Liu, G.-Y.; Wang, R.; Moon, S.H.; Gross, R.W.; Patti, G.J. Identifying off-target effects of etomoxir reveals that carnitine palmitoyltransferase I is essential for cancer cell proliferation independent of β-oxidation. PLoS Biol. 2018, 16, e2003782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. O’Connor, R.S.; Guo, L.; Ghassemi, S.; Snyder, N.W.; Worth, A.J.; Weng, L.; Kam, Y.; Philipson, B.; Trefely, S.; Nunez-Cruz, S.; et al. The CPT1a inhibitor, etomoxir induces severe oxidative stress at commonly used concentrations. Sci. Rep. 2018, 8, 6289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Raud, B.; Roy, D.G.; Divakaruni, A.S.; Tarasenko, T.N.; Franke, R.; Ma, E.H.; Samborska, B.; Hsieh, W.Y.; Wong, A.H.; Stüve, P.; et al. Etomoxir Actions on Regulatory and Memory T Cells Are Independent of Cpt1a-Mediated Fatty Acid Oxidation. Cell Metab. 2018, 28, 504–515.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Van den Bossche, J.; van der Windt, G.J.W. Fatty Acid Oxidation in Macrophages and T Cells: Time for Reassessment? Cell Metab. 2018, 28, 538–540. [Google Scholar] [CrossRef] [Green Version]
  96. Benador, I.Y.; Veliova, M.; Liesa, M.; Shirihai, O.S. Mitochondria Bound to Lipid Droplets: Where Mitochondrial Dynamics Regulate Lipid Storage and Utilization. Cell Metab. 2019, 29, 827–835. [Google Scholar] [CrossRef] [Green Version]
  97. Benador, I.Y.; Veliova, M.; Mahdaviani, K.; Petcherski, A.; Wikstrom, J.D.; Assali, E.A.; Acín-Pérez, R.; Shum, M.; Oliveira, M.F.; Cinti, S.; et al. Mitochondria Bound to Lipid Droplets Have Unique Bioenergetics, Composition, and Dynamics that Support Lipid Droplet Expansion. Cell Metab. 2018, 27, 869–885.e6. [Google Scholar] [CrossRef] [Green Version]
  98. Cruz, A.L.S.; de Barreto, E.A.; Fazolini, N.P.B.; Viola, J.P.B.; Bozza, P.T. Lipid droplets: Platforms with multiple functions in cancer hallmarks. Cell Death Dis. 2020, 11, 105. [Google Scholar] [CrossRef] [Green Version]
  99. Patananan, A.N.; Wu, T.-H.; Chiou, P.-Y.; Teitell, M.A. Modifying the Mitochondrial Genome. Cell Metab. 2016, 23, 785–796. [Google Scholar] [CrossRef] [Green Version]
  100. Wenceslau, C.F.; McCarthy, C.G.; Szasz, T.; Spitler, K.; Goulopoulou, S.; Webb, R.C. Working Group on DAMPs in Cardiovascular Disease Mitochondrial damage-associated molecular patterns and vascular function. Eur. Heart J. 2014, 35, 1172–1177. [Google Scholar] [CrossRef] [Green Version]
  101. Zhang, Q.; Raoof, M.; Chen, Y.; Sumi, Y.; Sursal, T.; Junger, W.; Brohi, K.; Itagaki, K.; Hauser, C.J. Circulating mitochondrial DAMPs cause inflammatory responses to injury. Nature 2010, 464, 104–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Galluzzi, L.; Bravo-San Pedro, J.M.; Kepp, O.; Kroemer, G. Regulated cell death and adaptive stress responses. Cell. Mol. Life Sci. 2016, 73, 2405–2410. [Google Scholar] [CrossRef] [PubMed]
  103. Nakahira, K.; Hisata, S.; Choi, A.M.K. The Roles of Mitochondrial Damage-Associated Molecular Patterns in Diseases. Antioxid. Redox Signal. 2015, 23, 1329–1350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Murray, L.M.A.; Krasnodembskaya, A.D. Concise Review: Intercellular Communication via Organelle Transfer in the Biology and Therapeutic Applications of Stem Cells. Stem Cells Dayt. Ohio 2019, 37, 14–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Berridge, M.V.; Neuzil, J. The mobility of mitochondria: Intercellular trafficking in health and disease. Clin. Exp. Pharm. Physiol. 2017, 44 (Suppl. 1), 15–20. [Google Scholar] [CrossRef] [Green Version]
  106. Boudreau, L.H.; Duchez, A.-C.; Cloutier, N.; Soulet, D.; Martin, N.; Bollinger, J.; Paré, A.; Rousseau, M.; Naika, G.S.; Lévesque, T.; et al. Platelets release mitochondria serving as substrate for bactericidal group IIA-secreted phospholipase A2 to promote inflammation. Blood 2014, 124, 2173–2183. [Google Scholar] [CrossRef] [Green Version]
  107. Al Amir Dache, Z.; Otandault, A.; Tanos, R.; Pastor, B.; Meddeb, R.; Sanchez, C.; Arena, G.; Lasorsa, L.; Bennett, A.; Grange, T.; et al. Blood contains circulating cell-free respiratory competent mitochondria. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2020, 34, 3616–3630. [Google Scholar] [CrossRef] [Green Version]
  108. Fransson, S.; Ruusala, A.; Aspenström, P. The atypical Rho GTPases Miro-1 and Miro-2 have essential roles in mitochondrial trafficking. Biochem. Biophys. Res. Commun. 2006, 344, 500–510. [Google Scholar] [CrossRef]
  109. Saotome, M.; Safiulina, D.; Szabadkai, G.; Das, S.; Fransson, A.; Aspenstrom, P.; Rizzuto, R.; Hajnóczky, G. Bidirectional Ca2+-dependent control of mitochondrial dynamics by the Miro GTPase. Proc. Natl. Acad. Sci. USA 2008, 105, 20728–20733. [Google Scholar] [CrossRef] [Green Version]
  110. Ahmad, T.; Mukherjee, S.; Pattnaik, B.; Kumar, M.; Singh, S.; Kumar, M.; Rehman, R.; Tiwari, B.K.; Jha, K.A.; Barhanpurkar, A.P.; et al. Miro1 regulates intercellular mitochondrial transport & enhances mesenchymal stem cell rescue efficacy. EMBO J. 2014, 33, 994–1010. [Google Scholar] [CrossRef]
  111. Wang, Y.; Chen, X.; Cao, W.; Shi, Y. Plasticity of mesenchymal stem cells in immunomodulation: Pathological and therapeutic implications. Nat. Immunol. 2014, 15, 1009–1016. [Google Scholar] [CrossRef] [PubMed]
  112. Castro-Manrreza, M.E.; Montesinos, J.J. Immunoregulation by mesenchymal stem cells: Biological aspects and clinical applications. J. Immunol. Res. 2015, 2015, 394917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Court, A.C.; Le-Gatt, A.; Luz-Crawford, P.; Parra, E.; Aliaga-Tobar, V.; Bátiz, L.F.; Contreras, R.A.; Ortúzar, M.I.; Kurte, M.; Elizondo-Vega, R.; et al. Mitochondrial transfer from MSCs to T cells induces Treg differentiation and restricts inflammatory response. EMBO Rep. 2020, 21, e48052. [Google Scholar] [CrossRef] [PubMed]
  114. Luz-Crawford, P.; Hernandez, J.; Djouad, F.; Luque-Campos, N.; Caicedo, A.; Carrère-Kremer, S.; Brondello, J.-M.; Vignais, M.-L.; Pène, J.; Jorgensen, C. Mesenchymal stem cell repression of Th17 cells is triggered by mitochondrial transfer. Stem Cell Res. 2019, 10, 232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Jackson, M.V.; Morrison, T.J.; Doherty, D.F.; McAuley, D.F.; Matthay, M.A.; Kissenpfennig, A.; O’Kane, C.M.; Krasnodembskaya, A.D. Mitochondrial Transfer via Tunneling Nanotubes is an Important Mechanism by Which Mesenchymal Stem Cells Enhance Macrophage Phagocytosis in the In Vitro and In Vivo Models of ARDS. Stem Cells Dayt. Ohio 2016, 34, 2210–2223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Morrison, T.J.; Jackson, M.V.; Cunningham, E.K.; Kissenpfennig, A.; McAuley, D.F.; O’Kane, C.M.; Krasnodembskaya, A.D. Mesenchymal Stromal Cells Modulate Macrophages in Clinically Relevant Lung Injury Models by Extracellular Vesicle Mitochondrial Transfer. Am. J. Respir. Crit. Care Med. 2017, 196, 1275–1286. [Google Scholar] [CrossRef]
  117. Hough, K.P.; Trevor, J.L.; Strenkowski, J.G.; Wang, Y.; Chacko, B.K.; Tousif, S.; Chanda, D.; Steele, C.; Antony, V.B.; Dokland, T.; et al. Exosomal transfer of mitochondria from airway myeloid-derived regulatory cells to T cells. Redox Biol. 2018, 18, 54–64. [Google Scholar] [CrossRef]
  118. Puhm, F.; Afonyushkin, T.; Resch, U.; Obermayer, G.; Rohde, M.; Penz, T.; Schuster, M.; Wagner, G.; Rendeiro, A.F.; Melki, I.; et al. Mitochondria Are a Subset of Extracellular Vesicles Released by Activated Monocytes and Induce Type I IFN and TNF Responses in Endothelial Cells. Circ. Res. 2019, 125, 43–52. [Google Scholar] [CrossRef]
  119. Mahrouf-Yorgov, M.; Augeul, L.; Da Silva, C.C.; Jourdan, M.; Rigolet, M.; Manin, S.; Ferrera, R.; Ovize, M.; Henry, A.; Guguin, A.; et al. Mesenchymal stem cells sense mitochondria released from damaged cells as danger signals to activate their rescue properties. Cell Death Differ. 2017, 24, 1224–1238. [Google Scholar] [CrossRef] [Green Version]
  120. Pollara, J.; Edwards, R.W.; Lin, L.; Bendersky, V.A.; Brennan, T.V. Circulating mitochondria in deceased organ donors are associated with immune activation and early allograft dysfunction. JCI Insight 2018, 3. [Google Scholar] [CrossRef]
  121. Song, X.; Hu, W.; Yu, H.; Wang, H.; Zhao, Y.; Korngold, R.; Zhao, Y. Existence of Circulating Mitochondria in Human and Animal Peripheral Blood. Int. J. Mol. Sci. 2020, 21, 2122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Acquistapace, A.; Bru, T.; Lesault, P.-F.; Figeac, F.; Coudert, A.E.; le Coz, O.; Christov, C.; Baudin, X.; Auber, F.; Yiou, R.; et al. Human mesenchymal stem cells reprogram adult cardiomyocytes toward a progenitor-like state through partial cell fusion and mitochondria transfer. Stem Cells Dayt. Ohio 2011, 29, 812–824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Islam, M.N.; Das, S.R.; Emin, M.T.; Wei, M.; Sun, L.; Westphalen, K.; Rowlands, D.J.; Quadri, S.K.; Bhattacharya, S.; Bhattacharya, J. Mitochondrial transfer from bone-marrow-derived stromal cells to pulmonary alveoli protects against acute lung injury. Nat. Med. 2012, 18, 759–765. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Pasquier, J.; Guerrouahen, B.S.; Al Thawadi, H.; Ghiabi, P.; Maleki, M.; Abu-Kaoud, N.; Jacob, A.; Mirshahi, M.; Galas, L.; Rafii, S.; et al. Preferential transfer of mitochondria from endothelial to cancer cells through tunneling nanotubes modulates chemoresistance. J. Transl. Med. 2013, 11, 94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Spees, J.L.; Olson, S.D.; Whitney, M.J.; Prockop, D.J. Mitochondrial transfer between cells can rescue aerobic respiration. Proc. Natl. Acad. Sci. USA 2006, 103, 1283–1288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Dong, L.-F.; Kovarova, J.; Bajzikova, M.; Bezawork-Geleta, A.; Svec, D.; Endaya, B.; Sachaphibulkij, K.; Coelho, A.R.; Sebkova, N.; Ruzickova, A.; et al. Horizontal transfer of whole mitochondria restores tumorigenic potential in mitochondrial DNA-deficient cancer cells. Elife 2017, 6. [Google Scholar] [CrossRef] [Green Version]
  127. Tan, A.S.; Baty, J.W.; Dong, L.-F.; Bezawork-Geleta, A.; Endaya, B.; Goodwin, J.; Bajzikova, M.; Kovarova, J.; Peterka, M.; Yan, B.; et al. Mitochondrial Genome Acquisition Restores Respiratory Function and Tumorigenic Potential of Cancer Cells without Mitochondrial DNA. Cell Metab. 2015, 21, 81–94. [Google Scholar] [CrossRef] [Green Version]
  128. Han, H.; Hu, J.; Yan, Q.; Zhu, J.; Zhu, Z.; Chen, Y.; Sun, J.; Zhang, R. Bone marrow-derived mesenchymal stem cells rescue injured H9c2 cells via transferring intact mitochondria through tunneling nanotubes in an in vitro simulated ischemia/reperfusion model. Mol. Med. Rep. 2016, 13, 1517–1524. [Google Scholar] [CrossRef] [Green Version]
  129. Liu, K.; Ji, K.; Guo, L.; Wu, W.; Lu, H.; Shan, P.; Yan, C. Mesenchymal stem cells rescue injured endothelial cells in an in vitro ischemia-reperfusion model via tunneling nanotube like structure-mediated mitochondrial transfer. Microvasc. Res. 2014, 92, 10–18. [Google Scholar] [CrossRef]
  130. Li, X.; Zhang, Y.; Yeung, S.C.; Liang, Y.; Liang, X.; Ding, Y.; Ip, M.S.M.; Tse, H.-F.; Mak, J.C.W.; Lian, Q. Mitochondrial Transfer of Induced Pluripotent Stem Cell–Derived Mesenchymal Stem Cells to Airway Epithelial Cells Attenuates Cigarette Smoke–Induced Damage. Am. J. Respir. Cell Mol. Biol. 2014, 51, 455–465. [Google Scholar] [CrossRef]
  131. Jiang, D.; Gao, F.; Zhang, Y.; Wong, D.S.H.; Li, Q.; Tse, H.; Xu, G.; Yu, Z.; Lian, Q. Mitochondrial transfer of mesenchymal stem cells effectively protects corneal epithelial cells from mitochondrial damage. Cell Death Dis. 2016, 7, e2467. [Google Scholar] [CrossRef] [Green Version]
  132. Babenko, V.A.; Silachev, D.N.; Zorova, L.D.; Pevzner, I.B.; Khutornenko, A.A.; Plotnikov, E.Y.; Sukhikh, G.T.; Zorov, D.B. Improving the Post-Stroke Therapeutic Potency of Mesenchymal Multipotent Stromal Cells by Cocultivation With Cortical Neurons: The Role of Crosstalk Between Cells. Stem Cells Transl. Med. 2015, 4, 1011–1020. [Google Scholar] [CrossRef] [PubMed]
  133. Moschoi, R.; Imbert, V.; Nebout, M.; Chiche, J.; Mary, D.; Prebet, T.; Saland, E.; Castellano, R.; Pouyet, L.; Collette, Y.; et al. Protective mitochondrial transfer from bone marrow stromal cells to acute myeloid leukemic cells during chemotherapy. Blood 2016, 128, 253–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Lu, J.; Zheng, X.; Li, F.; Yu, Y.; Chen, Z.; Liu, Z.; Wang, Z.; Xu, H.; Yang, W. Tunneling nanotubes promote intercellular mitochondria transfer followed by increased invasiveness in bladder cancer cells. Oncotarget 2017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Caicedo, A.; Fritz, V.; Brondello, J.-M.; Ayala, M.; Dennemont, I.; Abdellaoui, N.; de Fraipont, F.; Moisan, A.; Prouteau, C.A.; Boukhaddaoui, H.; et al. MitoCeption as a new tool to assess the effects of mesenchymal stem/stromal cell mitochondria on cancer cell metabolism and function. Sci. Rep. 2015, 5, 9073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Bajzikova, M.; Kovarova, J.; Coelho, A.R.; Boukalova, S.; Oh, S.; Rohlenova, K.; Svec, D.; Hubackova, S.; Endaya, B.; Judasova, K.; et al. Reactivation of Dihydroorotate Dehydrogenase-Driven Pyrimidine Biosynthesis Restores Tumor Growth of Respiration-Deficient Cancer Cells. Cell Metab. 2019, 29, 399–416.e10. [Google Scholar] [CrossRef] [Green Version]
  137. Löffler, M.; Carrey, E.A.; Zameitat, E. Orotic Acid, More Than Just an Intermediate of Pyrimidine de novo Synthesis. J. Genet. Genom. Yi Chuan Xue Bao 2015, 42, 207–219. [Google Scholar] [CrossRef]
  138. Zameitat, E.; Freymark, G.; Dietz, C.D.; Löffler, M.; Bölker, M. Functional expression of human dihydroorotate dehydrogenase (DHODH) in pyr4 mutants of ustilago maydis allows target validation of DHODH inhibitors in vivo. Appl. Env. Microbiol. 2007, 73, 3371–3379. [Google Scholar] [CrossRef] [Green Version]
  139. Levoux, J.; Prola, A.; Lafuste, P.; Gervais, M.; Braud, L.; Schirmann, A.; Hersant, B.; Sid-Ahmed, M.; Ben-Larbi, S.; Komrskova, K.; et al. Platelets Promote Pro-Angiogenic Activity of Mesenchymal Stem Cells via Mitochondrial Transfer and Metabolic Reprogramming; Social Science Research Network: Rochester, NY, USA, 2020. [Google Scholar]
  140. Maeda, A.; Fadeel, B. Mitochondria released by cells undergoing TNF-α-induced necroptosis act as danger signals. Cell Death Dis. 2014, 5, e1312. [Google Scholar] [CrossRef] [Green Version]
  141. Zhu, M.; Barbas, A.S.; Lin, L.; Scheuermann, U.; Bishawi, M.; Brennan, T.V. Mitochondria Released by Apoptotic Cell Death Initiate Innate Immune Responses. ImmunoHorizons 2018, 2, 384–397. [Google Scholar] [CrossRef]
  142. Spencer, D.M.; Dye, J.R.; Piantadosi, C.A.; Pisetsky, D.S. The release of microparticles and mitochondria from RAW 264.7 murine macrophage cells undergoing necroptotic cell death in vitro. Exp. Cell Res. 2018, 363, 151–159. [Google Scholar] [CrossRef] [PubMed]
  143. Singh, A.; Periasamy, S.; Malik, M.; Bakshi, C.S.; Stephen, L.; Ault, J.G.; Mannella, C.A.; Sellati, T.J. Necroptotic debris including damaged mitochondria elicits sepsis-like syndrome during late-phase tularemia. Cell Death Discov. 2017, 3, 17056. [Google Scholar] [CrossRef] [PubMed]
  144. Chu, C.T.; Ji, J.; Dagda, R.K.; Jiang, J.F.; Tyurina, Y.Y.; Kapralov, A.A.; Tyurin, V.A.; Yanamala, N.; Shrivastava, I.H.; Mohammadyani, D.; et al. Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nat. Cell Biol. 2013, 15, 1197–1205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Iyer, S.S.; He, Q.; Janczy, J.R.; Elliott, E.I.; Zhong, Z.; Olivier, A.K.; Sadler, J.J.; Knepper-Adrian, V.; Han, R.; Qiao, L.; et al. Mitochondrial cardiolipin is required for NLRP3 inflammasome activation. Immunity 2013, 39, 311–323. [Google Scholar] [CrossRef] [Green Version]
  146. Marlein, C.R.; Zaitseva, L.; Piddock, R.E.; Robinson, S.; Edwards, D.; Shafat, M.S.; Zhou, Z.; Lawes, M.; Bowles, K.M.; Rushworth, S.A. NADPH oxidase-2 derived superoxide drives mitochondrial transfer from bone marrow stromal cells to leukemic blasts. Blood 2017, blood-2017-03-772939. [Google Scholar] [CrossRef]
  147. Ryter, S.W.; Choi, A.M.K. Regulation of autophagy in oxygen-dependent cellular stress. Curr. Pharm. Des. 2013, 19, 2747–2756. [Google Scholar] [CrossRef]
  148. Shadel, G.S.; Horvath, T.L. Mitochondrial ROS signaling in organismal homeostasis. Cell 2015, 163, 560–569. [Google Scholar] [CrossRef] [Green Version]
  149. Archibald, J.M. Endosymbiosis and Eukaryotic Cell Evolution. Curr. Biol. 2015, 25, R911–R921. [Google Scholar] [CrossRef] [Green Version]
  150. Bajpai, P.; Darra, A.; Agrawal, A. Microbe-mitochondrion crosstalk and health: An emerging paradigm. Mitochondrion 2018, 39, 20–25. [Google Scholar] [CrossRef]
  151. Zitvogel, L.; Daillère, R.; Roberti, M.P.; Routy, B.; Kroemer, G. Anticancer effects of the microbiome and its products. Nat. Rev. Microbiol. 2017, 15, 465–478. [Google Scholar] [CrossRef]
  152. Cani, P.D.; Van Hul, M.; Lefort, C.; Depommier, C.; Rastelli, M.; Everard, A. Microbial regulation of organismal energy homeostasis. Nat. Metab. 2019, 1, 34–46. [Google Scholar] [CrossRef]
  153. Houghton, M.J.; Kerimi, A.; Mouly, V.; Tumova, S.; Williamson, G. Gut microbiome catabolites as novel modulators of muscle cell glucose metabolism. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2019, 33, 1887–1898. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Kroemer, G.; Zitvogel, L. Cancer immunotherapy in 2017: The breakthrough of the microbiota. Nat. Rev. Immunol. 2018, 18, 87–88. [Google Scholar] [CrossRef] [PubMed]
  155. Quinn, R.A.; Melnik, A.V.; Vrbanac, A.; Fu, T.; Patras, K.A.; Christy, M.P.; Bodai, Z.; Belda-Ferre, P.; Tripathi, A.; Chung, L.K.; et al. Global chemical effects of the microbiome include new bile-acid conjugations. Nature 2020, 579, 123–129. [Google Scholar] [CrossRef] [PubMed]
  156. Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef] [Green Version]
  157. Santoni, M.; Piva, F.; Conti, A.; Santoni, A.; Cimadamore, A.; Scarpelli, M.; Battelli, N.; Montironi, R. Re: Gut Microbiome Influences Efficacy of PD-1-based Immunotherapy Against Epithelial Tumors. Eur. Urol. 2018, 74, 521–522. [Google Scholar] [CrossRef]
  158. Bullman, S.; Pedamallu, C.S.; Sicinska, E.; Clancy, T.E.; Zhang, X.; Cai, D.; Neuberg, D.; Huang, K.; Guevara, F.; Nelson, T.; et al. Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer. Science 2017, 358, 1443–1448. [Google Scholar] [CrossRef] [Green Version]
  159. Xavier, J.B.; Young, V.B.; Skufca, J.; Ginty, F.; Testerman, T.; Pearson, A.T.; Macklin, P.; Mitchell, A.; Shmulevich, I.; Xie, L.; et al. The Cancer Microbiome: Distinguishing Direct and Indirect Effects Requires a Systemic View. Trends Cancer 2020, 6, 192–204. [Google Scholar] [CrossRef] [Green Version]
  160. Mashimo, T.; Pichumani, K.; Vemireddy, V.; Hatanpaa, K.J.; Singh, D.K.; Sirasanagandla, S.; Nannepaga, S.; Piccirillo, S.G.; Kovacs, Z.; Foong, C.; et al. Acetate is a bioenergetic substrate for human glioblastoma and brain metastases. Cell 2014, 159, 1603–1614. [Google Scholar] [CrossRef] [Green Version]
  161. Comerford, S.A.; Huang, Z.; Du, X.; Wang, Y.; Cai, L.; Witkiewicz, A.K.; Walters, H.; Tantawy, M.N.; Fu, A.; Manning, H.C.; et al. Acetate dependence of tumors. Cell 2014, 159, 1591–1602. [Google Scholar] [CrossRef] [Green Version]
  162. Schug, Z.T.; Peck, B.; Jones, D.T.; Zhang, Q.; Grosskurth, S.; Alam, I.S.; Goodwin, L.M.; Smethurst, E.; Mason, S.; Blyth, K.; et al. Acetyl-CoA synthetase 2 promotes acetate utilization and maintains cancer cell growth under metabolic stress. Cancer Cell 2015, 27, 57–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. El-Hattab, A.W.; Zarante, A.M.; Almannai, M.; Scaglia, F. Therapies for mitochondrial diseases and current clinical trials. Mol. Genet. Metab. 2017, 122, 1–9. [Google Scholar] [CrossRef] [PubMed]
  164. Khayat, D.; Kurtz, T.L.; Stacpoole, P.W. The changing landscape of clinical trials for mitochondrial diseases: 2011 to present. Mitochondrion 2020, 50, 51–57. [Google Scholar] [CrossRef]
  165. Weissig, V. Drug Development for the Therapy of Mitochondrial Diseases. Trends Mol. Med. 2020, 26, 40–57. [Google Scholar] [CrossRef]
  166. Vignal, C.; Uretsky, S.; Fitoussi, S.; Galy, A.; Blouin, L.; Girmens, J.-F.; Bidot, S.; Thomasson, N.; Bouquet, C.; Valero, S.; et al. Safety of rAAV2/2-ND4 Gene Therapy for Leber Hereditary Optic Neuropathy. Ophthalmology 2018, 125, 945–947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Koilkonda, R.D.; Yu, H.; Chou, T.-H.; Feuer, W.J.; Ruggeri, M.; Porciatti, V.; Tse, D.; Hauswirth, W.W.; Chiodo, V.; Boye, S.L.; et al. Safety and effects of the vector for the Leber hereditary optic neuropathy gene therapy clinical trial. JAMA Ophthalmol. 2014, 132, 409–420. [Google Scholar] [CrossRef]
  168. Cwerman-Thibault, H.; Augustin, S.; Lechauve, C.; Ayache, J.; Ellouze, S.; Sahel, J.-A.; Corral-Debrinski, M. Nuclear expression of mitochondrial ND4 leads to the protein assembling in complex I and prevents optic atrophy and visual loss. Mol. Methods Clin. Dev. 2015, 2, 15003. [Google Scholar] [CrossRef]
  169. Choong, C.-J.; Mochizuki, H. Gene therapy targeting mitochondrial pathway in Parkinson’s disease. J. Neural Transm. Vienna Austria 1996 2017, 124, 193–207. [Google Scholar] [CrossRef]
  170. Miller, S.; Muqit, M.M.K. Therapeutic approaches to enhance PINK1/Parkin mediated mitophagy for the treatment of Parkinson’s disease. Neurosci. Lett. 2019, 705, 7–13. [Google Scholar] [CrossRef]
  171. Ge, P.; Dawson, V.L.; Dawson, T.M. PINK1 and Parkin mitochondrial quality control: A source of regional vulnerability in Parkinson’s disease. Mol. Neurodegener. 2020, 15, 1–18. [Google Scholar] [CrossRef] [Green Version]
  172. Roushandeh, A.M.; Kuwahara, Y.; Roudkenar, M.H. Mitochondrial transplantation as a potential and novel master key for treatment of various incurable diseases. Cytotechnology 2019, 71, 647–663. [Google Scholar] [CrossRef] [PubMed]
  173. Masuzawa, A.; Black, K.M.; Pacak, C.A.; Ericsson, M.; Barnett, R.J.; Drumm, C.; Seth, P.; Bloch, D.B.; Levitsky, S.; Cowan, D.B.; et al. Transplantation of autologously derived mitochondria protects the heart from ischemia-reperfusion injury. Am. J. Physiol. Heart Circ. Physiol. 2013, 304, H966–H982. [Google Scholar] [CrossRef] [PubMed]
  174. Kaza, A.K.; Wamala, I.; Friehs, I.; Kuebler, J.D.; Rathod, R.H.; Berra, I.; Ericsson, M.; Yao, R.; Thedsanamoorthy, J.K.; Zurakowski, D.; et al. Myocardial rescue with autologous mitochondrial transplantation in a porcine model of ischemia/reperfusion. J. Thorac. Cardiovasc. Surg. 2017, 153, 934–943. [Google Scholar] [CrossRef] [Green Version]
  175. Cowan, D.B.; Yao, R.; Akurathi, V.; Snay, E.R.; Thedsanamoorthy, J.K.; Zurakowski, D.; Ericsson, M.; Friehs, I.; Wu, Y.; Levitsky, S.; et al. Intracoronary Delivery of Mitochondria to the Ischemic Heart for Cardioprotection. PLoS ONE 2016, 11, e0160889. [Google Scholar] [CrossRef] [Green Version]
  176. Shin, B.; Saeed, M.Y.; Esch, J.J.; Guariento, A.; Blitzer, D.; Moskowitzova, K.; Ramirez-Barbieri, G.; Orfany, A.; Thedsanamoorthy, J.K.; Cowan, D.B.; et al. A Novel Biological Strategy for Myocardial Protection by Intracoronary Delivery of Mitochondria: Safety and Efficacy. JACC Basic Transl. Sci. 2019, 4, 871–888. [Google Scholar] [CrossRef]
  177. Jabbari, H.; Roushandeh, A.M.; Rostami, M.K.; Razavi-Toosi, M.T.; Shokrgozar, M.A.; Jahanian-Najafabadi, A.; Kuwahara, Y.; Roudkenar, M.H. Mitochondrial transplantation ameliorates ischemia/reperfusion-induced kidney injury in rat. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165809. [Google Scholar] [CrossRef] [PubMed]
  178. Huang, P.-J.; Kuo, C.-C.; Lee, H.-C.; Shen, C.-I.; Cheng, F.-C.; Wu, S.-F.; Chang, J.-C.; Pan, H.-C.; Lin, S.-Z.; Liu, C.-S.; et al. Transferring Xenogenic Mitochondria Provides Neural Protection Against Ischemic Stress in Ischemic Rat Brains. Cell Transpl. 2016, 25, 913–927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Zhang, Z.; Ma, Z.; Yan, C.; Pu, K.; Wu, M.; Bai, J.; Li, Y.; Wang, Q. Muscle-derived autologous mitochondrial transplantation: A novel strategy for treating cerebral ischemic injury. Behav. Brain Res. 2019, 356, 322–331. [Google Scholar] [CrossRef]
  180. Gollihue, J.L.; Patel, S.P.; Eldahan, K.C.; Cox, D.H.; Donahue, R.R.; Taylor, B.K.; Sullivan, P.G.; Rabchevsky, A.G. Effects of Mitochondrial Transplantation on Bioenergetics, Cellular Incorporation, and Functional Recovery after Spinal Cord Injury. J. Neurotrauma 2018, 35, 1800–1818. [Google Scholar] [CrossRef]
  181. Fang, S.-Y.; Roan, J.-N.; Lee, J.-S.; Chiu, M.-H.; Lin, M.-W.; Liu, C.-C.; Lam, C.-F. Transplantation of viable mitochondria attenuates neurologic injury after spinal cord ischemia. J. Thorac. Cardiovasc. Surg. 2019. [Google Scholar] [CrossRef]
  182. Nascimento-Dos-Santos, G.; de-Souza-Ferreira, E.; Lani, R.; Faria, C.C.; Araújo, V.G.; Teixeira-Pinheiro, L.C.; Vasconcelos, T.; Gonçalo, T.; Santiago, M.F.; Linden, R.; et al. Neuroprotection from optic nerve injury and modulation of oxidative metabolism by transplantation of active mitochondria to the retina. Biochim. Biophys. Acta Mol. Basis Dis. 2020, 1866, 165686. [Google Scholar] [CrossRef] [PubMed]
  183. Doulamis, I.P.; Guariento, A.; Duignan, T.; Orfany, A.; Kido, T.; Zurakowski, D.; Del Nido, P.J.; McCully, J.D. Mitochondrial transplantation for myocardial protection in diabetic hearts. Eur. J. Cardio-Thorac. Surg. Off. J. Eur. Assoc. Cardio-Thorac. Surg. 2020, 57, 836–845. [Google Scholar] [CrossRef] [PubMed]
  184. Fu, A.; Shi, X.; Zhang, H.; Fu, B. Mitotherapy for Fatty Liver by Intravenous Administration of Exogenous Mitochondria in Male Mice. Front. Pharm. 2017, 8, 241. [Google Scholar] [CrossRef] [Green Version]
  185. Chang, J.-C.; Wu, S.-L.; Liu, K.-H.; Chen, Y.-H.; Chuang, C.-S.; Cheng, F.-C.; Su, H.-L.; Wei, Y.-H.; Kuo, S.-J.; Liu, C.-S. Allogeneic/xenogeneic transplantation of peptide-labeled mitochondria in Parkinson’s disease: Restoration of mitochondria functions and attenuation of 6-hydroxydopamine-induced neurotoxicity. Transl. Res. J. Lab. Clin. Med. 2016, 170, 40–56.e3. [Google Scholar] [CrossRef] [PubMed]
  186. Shi, X.; Zhao, M.; Fu, C.; Fu, A. Intravenous administration of mitochondria for treating experimental Parkinson’s disease. Mitochondrion 2017, 34, 91–100. [Google Scholar] [CrossRef]
  187. Robicsek, O.; Ene, H.M.; Karry, R.; Ytzhaki, O.; Asor, E.; McPhie, D.; Cohen, B.M.; Ben-Yehuda, R.; Weiner, I.; Ben-Shachar, D. Isolated Mitochondria Transfer Improves Neuronal Differentiation of Schizophrenia-Derived Induced Pluripotent Stem Cells and Rescues Deficits in a Rat Model of the Disorder. Schizophr. Bull. 2018, 44, 432–442. [Google Scholar] [CrossRef] [Green Version]
  188. Chang, J.-C.; Chang, H.-S.; Wu, Y.-C.; Cheng, W.-L.; Lin, T.-T.; Chang, H.-J.; Kuo, S.-J.; Chen, S.-T.; Liu, C.-S. Mitochondrial transplantation regulates antitumour activity, chemoresistance and mitochondrial dynamics in breast cancer. J. Exp. Clin. Cancer Res. Cr 2019, 38, 30. [Google Scholar] [CrossRef] [Green Version]
  189. Sun, C.; Liu, X.; Wang, B.; Wang, Z.; Liu, Y.; Di, C.; Si, J.; Li, H.; Wu, Q.; Xu, D.; et al. Endocytosis-mediated mitochondrial transplantation: Transferring normal human astrocytic mitochondria into glioma cells rescues aerobic respiration and enhances radiosensitivity. Theranostics 2019, 9, 3595–3607. [Google Scholar] [CrossRef]
  190. Ali Pour, P.; Kenney, M.C.; Kheradvar, A. Bioenergetics Consequences of Mitochondrial Transplantation in Cardiomyocytes. J. Am. Heart Assoc. 2020, 9, e014501. [Google Scholar] [CrossRef]
  191. Emani, S.M.; Piekarski, B.L.; Harrild, D.; Del Nido, P.J.; McCully, J.D. Autologous mitochondrial transplantation for dysfunction after ischemia-reperfusion injury. J. Thorac. Cardiovasc. Surg. 2017, 154, 286–289. [Google Scholar] [CrossRef] [Green Version]
  192. Sharma, A.; Fonarow, G.C.; Butler, J.; Ezekowitz, J.A.; Felker, G.M. Coenzyme Q10 and Heart Failure: A State-of-the-Art Review. Circ. Heart Fail. 2016, 9, e002639. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Oxidative phosphorylation (OXPHOS) through the electron transport chain (ETC) and adenosine triphosphate (ATP) production. OXPHOS is the process leading to ATP synthesis through the transport of electrons released by the oxidation of nicotinamide adenine dinucleotide (NADH) and succinate generated by the tricarboxylic acid (TCA) cycle. The transport of electrons is mediated by the ETC located in the mitochondrial inner membrane. The ETC is composed of the four complexes NADH–ubiquinone oxidoreductase (complex I), succinate–CoQ oxidoreductase (complex II), ubiquinol–cytochrome c oxidoreductase (complex III), and cytochrome c oxidase (complex IV), and of the free-electron carriers ubiquinone (CoQ10) and cytochrome c (CytC). Electrons are transferred through the ETC and finally to oxygen (O2) (dashed lines). This electron transfer is accompanied by a flow of protons (H+) from the mitochondrial matrix into the intermembrane space, across complexes I, III, and IV. The generated transmembrane electrochemical proton gradient allows the ATP synthase (complex V) to produce ATP. Also shown is the dihydroorotate dehydrogenase (DHODH), which participates in the electron transfer process through the oxidation of dihydroorotate to orotate, leading to de novo pyrimidine synthesis.
Figure 1. Oxidative phosphorylation (OXPHOS) through the electron transport chain (ETC) and adenosine triphosphate (ATP) production. OXPHOS is the process leading to ATP synthesis through the transport of electrons released by the oxidation of nicotinamide adenine dinucleotide (NADH) and succinate generated by the tricarboxylic acid (TCA) cycle. The transport of electrons is mediated by the ETC located in the mitochondrial inner membrane. The ETC is composed of the four complexes NADH–ubiquinone oxidoreductase (complex I), succinate–CoQ oxidoreductase (complex II), ubiquinol–cytochrome c oxidoreductase (complex III), and cytochrome c oxidase (complex IV), and of the free-electron carriers ubiquinone (CoQ10) and cytochrome c (CytC). Electrons are transferred through the ETC and finally to oxygen (O2) (dashed lines). This electron transfer is accompanied by a flow of protons (H+) from the mitochondrial matrix into the intermembrane space, across complexes I, III, and IV. The generated transmembrane electrochemical proton gradient allows the ATP synthase (complex V) to produce ATP. Also shown is the dihydroorotate dehydrogenase (DHODH), which participates in the electron transfer process through the oxidation of dihydroorotate to orotate, leading to de novo pyrimidine synthesis.
Ijms 21 04405 g001
Figure 2. Effects of complex I mutations. (1) As a result of complete loss of complex I, NADH inhibits the TCA cycle enzyme α-ketoglutarate dehydrogenase (αKGDH). This increases α-ketoglutarate levels and results in HIF1α inhibition. (24) Complex I mutations increase reactive oxygen species (ROS) production. (2) ROS activate pyruvate dehydrogenase kinase (PDK2), which inhibits pyruvate dehydrogenase (PDH). Pyruvate accumulation activates HIF1α. (3) ROS activate the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/hypoxia-inducible factor 1 alpha (HIF1α) signaling pathway. (4) Mitochondrial ROS activate the NADPH oxidase, which produces cytoplasmic superoxide. ROS lead to genomic instability via double-strand breaks (DSBs) and activate the cellular anti-oxidant responses.
Figure 2. Effects of complex I mutations. (1) As a result of complete loss of complex I, NADH inhibits the TCA cycle enzyme α-ketoglutarate dehydrogenase (αKGDH). This increases α-ketoglutarate levels and results in HIF1α inhibition. (24) Complex I mutations increase reactive oxygen species (ROS) production. (2) ROS activate pyruvate dehydrogenase kinase (PDK2), which inhibits pyruvate dehydrogenase (PDH). Pyruvate accumulation activates HIF1α. (3) ROS activate the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/hypoxia-inducible factor 1 alpha (HIF1α) signaling pathway. (4) Mitochondrial ROS activate the NADPH oxidase, which produces cytoplasmic superoxide. ROS lead to genomic instability via double-strand breaks (DSBs) and activate the cellular anti-oxidant responses.
Ijms 21 04405 g002
Figure 3. Role of TCA cycle intermediates in HIF1α stabilization and nuclear epigenetic modifications. Following its hydroxylation by prolyl-hydroxylases (PHDs), HIF1α is recognized by the von Hippel–Lindau (VHL) complex that targets it for proteasomal degradation. The inactivation of complex II of the ETC leads to ROS accumulation and inhibition of PHD activity by succinate, preventing HIF1α hydroxylation and degradation. Upon translocation to the nucleus, HIF1α binds to the hypoxic response element (HRE) and activates gene transcription, contributing to tumorigenesis. Complex II-deficient cells primarily rely on glutamine to fuel the TCA cycle and utilize oxaloacetate to generate aspartate for their biosynthetic pathways. The TCA cycle also produces metabolites that act as epigenetic modifiers. The α-ketoglutarate is a cofactor of 2-oxoglutarate-dependent dioxygenases, including the ten-eleven translocation (TET) family of DNA demethylases and the histone lysine demethylase (KDM) family. The metabolites 2-hydroxyglutarate, succinate, and fumarate, which are structurally similar to α-ketoglutarate, act as antagonists of TET- and KDM-catalyzed reactions.
Figure 3. Role of TCA cycle intermediates in HIF1α stabilization and nuclear epigenetic modifications. Following its hydroxylation by prolyl-hydroxylases (PHDs), HIF1α is recognized by the von Hippel–Lindau (VHL) complex that targets it for proteasomal degradation. The inactivation of complex II of the ETC leads to ROS accumulation and inhibition of PHD activity by succinate, preventing HIF1α hydroxylation and degradation. Upon translocation to the nucleus, HIF1α binds to the hypoxic response element (HRE) and activates gene transcription, contributing to tumorigenesis. Complex II-deficient cells primarily rely on glutamine to fuel the TCA cycle and utilize oxaloacetate to generate aspartate for their biosynthetic pathways. The TCA cycle also produces metabolites that act as epigenetic modifiers. The α-ketoglutarate is a cofactor of 2-oxoglutarate-dependent dioxygenases, including the ten-eleven translocation (TET) family of DNA demethylases and the histone lysine demethylase (KDM) family. The metabolites 2-hydroxyglutarate, succinate, and fumarate, which are structurally similar to α-ketoglutarate, act as antagonists of TET- and KDM-catalyzed reactions.
Ijms 21 04405 g003
Figure 4. The mevalonate and the fatty acid synthesis pathways. Acetyl-CoA, derived from glucose, glutamine, or citrate following its export to the cytosol, is converted through the mevalonate pathway into several metabolites including cholesterol and coenzyme Q10. Coenzyme Q10 transfers electrons from complexes I and II of the electron transport chain, as well as from dihydroorotate dehydrogenase (DHODH), to complex III. Acetyl-coA also acts a precursor for fatty acid synthesis, through its conversion to malonyl-CoA, and then to palmitate. The mevalonate pathway is represented in yellow boxes. The fatty acid synthesis pathway is represented in blue boxes. Dashed arrows represent multiple steps. HMG-CoA, 3-hydroxy-3-methylglutaryl CoA; IPP, isopentenyl-diphosphate; FPP, farnesyl diphosphate; GGPP, geranylgeranyl-diphosphate; TCA cycle, tricarboxylic acid cycle.
Figure 4. The mevalonate and the fatty acid synthesis pathways. Acetyl-CoA, derived from glucose, glutamine, or citrate following its export to the cytosol, is converted through the mevalonate pathway into several metabolites including cholesterol and coenzyme Q10. Coenzyme Q10 transfers electrons from complexes I and II of the electron transport chain, as well as from dihydroorotate dehydrogenase (DHODH), to complex III. Acetyl-coA also acts a precursor for fatty acid synthesis, through its conversion to malonyl-CoA, and then to palmitate. The mevalonate pathway is represented in yellow boxes. The fatty acid synthesis pathway is represented in blue boxes. Dashed arrows represent multiple steps. HMG-CoA, 3-hydroxy-3-methylglutaryl CoA; IPP, isopentenyl-diphosphate; FPP, farnesyl diphosphate; GGPP, geranylgeranyl-diphosphate; TCA cycle, tricarboxylic acid cycle.
Ijms 21 04405 g004

Share and Cite

MDPI and ACS Style

Nakhle, J.; Rodriguez, A.-M.; Vignais, M.-L. Multifaceted Roles of Mitochondrial Components and Metabolites in Metabolic Diseases and Cancer. Int. J. Mol. Sci. 2020, 21, 4405. https://doi.org/10.3390/ijms21124405

AMA Style

Nakhle J, Rodriguez A-M, Vignais M-L. Multifaceted Roles of Mitochondrial Components and Metabolites in Metabolic Diseases and Cancer. International Journal of Molecular Sciences. 2020; 21(12):4405. https://doi.org/10.3390/ijms21124405

Chicago/Turabian Style

Nakhle, Jean, Anne-Marie Rodriguez, and Marie-Luce Vignais. 2020. "Multifaceted Roles of Mitochondrial Components and Metabolites in Metabolic Diseases and Cancer" International Journal of Molecular Sciences 21, no. 12: 4405. https://doi.org/10.3390/ijms21124405

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop