The Mitochondrial ATP Synthase/IF1 Axis in Cancer Progression: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Mitochondria are central hubs in cellular physiology integrating cellular metabolism, bioenergetics, the execution of cell death and signaling through different effectors like Ca2+, reactive oxygen species (mtROS), mtDNA and different metabolites. Studies have summarized the different functions that the mitochondrial ATP synthase and its inhibitor protein, Inhibitory Factor 1 (IF1), play in cellular biology and in cancer progression. It has overviewed the mechanism by which the ATP synthase/IF1 axis contributes to metabolic reprogramming to an enhanced glycolytic phenotype, both in cancer cells and in the maintenance of stemness, and its potential both as biomarkers of prognosis and as targets for therapy. Moreover, it have highlighted how the ATP synthase/IF1 axis contributes to the signaling of cell-type specific programs that allow the adaptation of the cell/organisms to different changing cues, and finally, how the ATP synthase/IF1 axis also participates in preventing the execution of cell death and hence, in therapeutic resistance of the carcinomas. It has emphasized that the relative low activity of mitochondrial metabolic pathways, such as oxidation of pyruvate coupled to oxidative phosphorylation (OXPHOS) and β-oxidation in lung adenocarcinomas, contribute to cancer progression.

  • cancer
  • OXPHOS
  • Warburg effect
  • mitochondrial ATP synthase
  • ATPase inhibitory factor 1
  • metabolic reprogramming
  • metastasis
  • RNA binding proteins
  • mitohormesis
  • cell death

1. Metabolic Rewiring and Cancer Progression

Carcinomas undergo a coordinated reprogramming of their metabolic pathways to satisfy the demand for high energy and for the building blocks that are required to sustain cellular proliferation [1][2][3][4][5][6]. The metabolic reprogramming experienced by cancer is generally known as the Warburg effect, and it is characterized by the sharp increase in the rates of glycolysis and of the fermentation of pyruvate into lactate, which allows the regeneration of NAD+ for glycolysis to proceed at high rates (Figure 1). Glycolysis and fermentation do not require oxygen to function. Concurrent with the hyperactivation of glycolysis and fermentation, the oxidation of pyruvate coupled to oxidative phosphorylation (OXPHOS) in mitochondria, which is strictly dependent on the availability of oxygen, is marginally increased and/or partially inhibited in cancer cells, to spare the backbone of carbon skeletons for the synthesis of precursors (Figure 1) [3][6][7][8]. In fact, the citrate formed in the tricarboxylic acid (TCA) cycle is exported to the cytoplasm for providing the acetyl-CoA that is used for the synthesis of fatty acids and cholesterol (Figure 1) [9]. However, cancer cells replenish carbon skeletons in the TCA cycle in the form of glutamate by increasing the metabolism of glutamine (Figure 1) [10]. Simultaneously, the metabolism of glucose through the pentose phosphate pathway (PPP) is also significantly augmented, to produce the ribose that is used for building nucleotides and producing the NADPH that is used in biosynthetic processes and in the maintenance of the cellular redox state (Figure 1) [11]. Consistent with the reprogramming of metabolism in carcinomas, the sharp increase in different enzymes of glycolysis provide biomarkers that predict a bad patient prognosis [12], whereas, for example, the silencing of glycolytic enzymes in glioblastoma (GBM) xenografts dramatically increase the survival of mice [13]. In contrast, the loss of mitochondrial proteins involved in pyruvate oxidation, β-oxidation and OXPHOS predicts bad patient prognosis [12][14][15][16], supporting the theory that a diminished metabolic activity of the organelle compromises survival. Likewise, the overexpression of enzymes of metabolic pathways that are induced in carcinomas, such as glutamine metabolism [12][17][18] and the biosynthesis of fatty acids [12][19][20][21][22], predicts bad patient prognosis.
Figure 1. Metabolic pathways in cancer. The color-coded scheme illustrates the major cancer-promoted changes in relevant metabolic pathways observed in human carcinomas. Glycolysis, fermentation, pentose phosphate pathway (PPP), lipogenesis and glutaminolysis are enhanced (red circles) in carcinomas. However, the mitochondrial oxidation of pyruvate, β-oxidation (β-Ox.), tricarboxylic acid (TCA) cycle and oxidative phosphorylation (OXPHOS) are marginally increased (pink circles) or diminished when compared to the activation of metabolic pathway in the cytoplasm. Glucose enters the cell by specific transporters and is metabolized at high rate through glycolysis to provide precursors such as glycerol-3-P and amino acids for lipid and protein synthesis (green box), respectively. Pyruvate is reduced to lactate in fermentation to regenerate NAD+ to allow glycolysis to proceed and is exported from the cell. In addition, glucose is catabolized through the PPP to obtain NADPH (orange box), for biosynthetic processes and the maintenance of the cellular redox state, and ribose-5-P, for nucleic acid biosynthesis (green box). In the mitochondria, pyruvate is oxidized to generate acetyl-CoA that feeds the TCA cycle. Fatty acids (FA) oxidation also feeds acetyl-CoA to the TCA cycle. Citrate in the TCA cycle is drained to the cytoplasm for the biosynthesis of FA and cholesterol. Glutaminolysis feeds the TCA cycle with glutamate. The electrons obtained in the oxidation of pyruvate and FA are transferred to the electron transport chain system for the synthesis of the ATP by OXPHOS (purple).

2. The Mitochondrial ATP Synthase/IF1 Axis

The OXPHOS system integrates the complexes of the electron transport chain (ETC) (complex I–IV), a variety of different dehydrogenases, the ATP synthase, two diffusible electron carriers such as CoQ and cytochrome c, and substrate transporters in the inner mitochondrial membrane (IMM) (Figure 2a) [7][23][24]. Respiratory complexes of the ETC, and a variety of FAD-dependent dehydrogenases that feed electrons directly to CoQ, transfer electrons obtained in biological oxidations in the form of NADH and FADH2 down to molecular oxygen, to generate the proton electrochemical gradient responsible for the proton-motive force (∆p) across the IMM (Figure 2a). The ATP synthase utilizes ∆p by coupling the backflow of protons into the matrix of the organelle for the generation of ATP from ADP and inorganic phosphate (Pi) (Figure 2a).
Figure 2. The mitochondrial OXPHOS system: (a) The OXPHOS system consists of five protein complexes: complexes I–IV (CI–IV) of the electron transport chain (ETC) and complex V (CV) or ATP synthase. In addition, there are mobile electron carriers such as coenzyme Q (CoQ) and cytochrome c (cyt c), as well as membrane transporters that include among others the pyruvate carrier (not shown), adenine nucleotide translocase (ANT) and the phosphate carrier (PiC). Other FAD-dependent dehydrogenases transfer electrons to CoQ, such as glycerol-3-phosphate dehydrogenase (G3PDH), dihydroorotate dehydrogenase (DHOH), acyl-CoA dehydrogenase complex (ACDH), electron transfer flavoprotein (ETF) and electron transfer flavoprotein dehydrogenase (ETFDH). The electron transfer from NADH and FADH2 to molecular oxygen (O2), the final electron acceptor, builds a proton-motive force (∆p) by the pumping of protons through complexes I, III and IV, which is used by ATP synthase to produce ATP; (b) Structure of monomeric bovine active ATP synthase (left) and bound to the N-terminal inhibitory fragment of IF1 in the inhibited ATP synthase state (right). The soluble F1 domain is composed of α3β3 subunits (purple/blue) and the central stalk (γ subunit, light blue, and δ, ε subunits, light yellow), while the Fo domain is formed by a ring of 8 c subunits (pink) and a subunit (light blue). These two domains are linked by a peripheral stalk, composed of b, d, F6 subunits (light green), and the oligomycin sensitivity-conferring protein (OSCP; grey). The supernumerary subunits include e (orange), f, g, A6L, the protein associated with diabetes in insulin-sensitive tissues (DAPIT), and the 6.8 kDa proteolipid (6.8PL) (light blue). Right, the interaction between the N-terminal inhibitory fragment of IF1 (red) and the α3β3 and γ subunits is shown. The Ala14 residue of IF1 (Ser14 in human and mouse IF1) is highlighted in yellow. The active ATP synthase is able to synthetize ATP using ADP and Pi. When IF1 is bound it is inhibited and no longer synthetizes nor hydrolyzes ATP. Regulation of the ATP synthase is exerted by IF1. IF1 can be inactivated by forming oligomers that mask the inhibitory regions that bind to the F1 domain when the matrix pH is above neutrality. However, in mitochondrial de-energization conditions the pH drops below neutrality and IF1 becomes activated. On the other hand, IF1 can be phosphorylated in S39 by PKA preventing its interaction with the ATP synthase. IF1 has a short-half life and there is no evidence yet for the existence of an IF1 protein phosphatase (PPase). Molecular reconstruction from PDB: 6ZPO, 1OHH and 1GMJ. Images created using the PyMOL molecular graphics system.
The ATP synthase, a protein complex composed of 18 different proteins in humans, drives the production of ATP using the free energy stored in the proton-motive force (Figure 2a,b) [25]. It consists of two main domains, the Fo embedded in the IMM and the F1 domain projected into the matrix, containing the α and β subunits of the catalytic core (Figure 2b) [26]. The Fo and F1 domains are linked together by a central (subunit γ, ε and δ) and a peripheral (subunit b, d, OSCP and F6) stalk (Figure 2b). The protons are imported into the matrix through subunit a causing the rotation of the c-ring in the Fo domain. The central stalk transfers this rotational energy to the α3β3 subunits in the F1 domain, leading to the synthesis of ATP (Figure 2b).
The ATP synthase has a small inhibitory protein, the ATPase Inhibitory Factor 1 (IF1), that for long time has been considered an inhibitor only (unidirectional) of the hydrolytic activity of the enzyme by binding the αβ interface in the F1 domain (Figure 2b) [27][28]. IF1 is a highly conserved protein among mammalian species and is encoded in the nuclear ATP5IF1 gene [29].
The IF1-mediated inhibition of ATP synthase results in partial blockade of the import of protons into mitochondria, the limitation of OXPHOS activity and the metabolic reprogramming of the cells to an enhanced glycolysis (Figure 3) [30][31][32][33]. Moreover, the blockage in proton import also promotes mitochondrial hyperpolarization and the subsequent production of ROS (mtROS) at respiratory complexes of the ETC (Figure 3) [31][34]. The generated mtROS modify the activity of protein kinases and of transcription factors that signal to the nucleus different programs that allow adaptation of the cell to changing cues (Figure 3) [31][34][35][36][37]. In some mouse tissues, the IF1-mediated activation of mtROS signaling promotes long-lasting metabolic and molecular cytoprotective mechanisms that allow cells to withstand subsequent insults (Figure 3) [35][36][37], a process known as mitohormesis [38][39][40][41]. However, in other mouse tissues which are naturally devoid of IF1 [42], the overexpression of IF1 is detrimental [36][43][44]. In sharp contrast, the overexpression of IF1 in neurons protects from excitotoxic insults [35] and increases the exploratory activity, motor coordination and cognition of mice (Figure 3) [34]. Similarly, the overexpression of IF1 in intestinal epithelial cells provides protection against inflammation (Figure 3) [37], whereas IF1 ablation results in a pro-inflammatory phenotype of fatal consequences [45]. Overall, these results emphasized the relevance of the ATP synthase/IF1 axis in signaling and controlling tissue-specific and non-cell autonomous cell fate decisions.
/media/item_content/202308/64caf28aef85ecancers-15-03775-g003.png
Figure 3. The role of ATP synthase/IF1 axis in metabolic reprogramming and signaling. Binding of IF1 to the ATP synthase limits the production of ATP and promotes metabolic reprogramming towards an enhanced glycolytic phenotype by allosteric regulation of the enzymes of glycolysis and the activation of AMPK. Inhibition of ATP synthase prevents the backflow of protons into the matrix increasing the mitochondrial membrane potential (∆Ψm). The increase in ∆Ψm leads to an increase in the generation of mitochondrial reactive oxygen species (mtROS) at the electron transport chain that signal to the nucleus through kinases and transcription factors. Nuclear reprogramming promotes the activation of cellular responses that include proliferation, prevention of cell death allowing adaptation of the cell to changing cues in mitohormetic responses. The cellular signaling response mediated by IF1 overexpression is cell type specific in mouse tissues and promotes antioxidant responses, anti-inflammatory phenotypes and motor coordination and cognition. In contrast, IF1 overexpression in skeletal muscle and heart is deleterious.
Besides its function in energy provision and in signaling, the ATP synthase also plays a structural role in the IMM by forming dimers and oligomers to shape cristae at its rims (Figure 4a,b) [26]. Recent cryo-EM structures of mammalian tetrameric ATP synthases (Figure 4b) [46][47] and isobaric quantitative Protein Interaction Reporter (iqPIR) cross-linking technologies [32], have uncovered the structural role that IF1 plays in oligomerization and inhibition of ATP synthase, and in reprogramming of the tissue to an enhanced glycolysis [32].
Figure 4. Oligomeric assemblies of the ATP synthase and the permeability transition pore (PTP): (a) Structure of bovine dimeric ATP synthase promotes the curvature of the inner mitochondrial membrane at its tips. Molecular reconstruction from PDB: 7AJD; (b) Cryo-electron microscopy structure of porcine IF1-inhibited ATP synthase tetramers viewed from the matrix side. The IF1 dimers (red) bind to two adjacent antiparallel ATP synthase dimers. Molecular reconstruction from PDB: 6J5K. Images created using the PyMOL molecular graphics system. Both in a, and b, the color coding of the subunits is the same as in Figure 2; (c) Two major models explain the participation of the ATP synthase in the PTP. The “death finger” model proposes that Ca2+ binding to β subunit causes a rearrangement in the F1 domain increasing its rigidity that is transmitted through OSCP (grey) to the peripheral stalk (green) and to the e subunit (orange). Subunit e exerts a pulling force on the outer lipids that plug the center of the c-ring, leading to the formation of a channel that allows the entrance of ions. Mild Ca2+ levels lead to closed and open states in a low-conductance mode of opening (“flickering”) in the PTP. However, when Ca2+ increases the channel is more prone to opening, promoting complete displacement of inner lipids in the c-rotor and of the central stalk of the ATP synthase, committing cells to death by the high-conductance PTP. In the “monomer/monomer interfaces” model, the Ca2+ induced conformational changes in dimers of the ATP synthase destabilize the dimeric assembly opening a high conductance pore in the interface of the dimer (subunits e and g).
Moreover, the ATP synthase is also a critical component in the regulation and execution of cell death since its components [48] and activity [49][50][51] are required for the efficient execution of cell death. In fact, the ATP synthase is structurally and functionally implicated in permeability transition (mPT) [52][53][54]. mPT is exerted through a high-conductance channel in the IMM, known as the Permeability Transition Pore (PTP) [55][56][57]. PTP opening leads to mitochondrial swelling and cell death and is regulated by Ca2+ and/or cyclophilin D-dependent structural changes on ATP synthase [53][58], whereas it is inhibited by cyclosporine A (CsA) [59][60]. The ATP synthase is postulated as a key component of the PTP [52][53][61][62][63][64], although its participation in mPT has been questioned [65][66]. Nowadays, the participation of the ATP synthase in PTP is undisputed after the reunification of the different alternatives that implicated the enzyme in the “death finger” and “monomer/monomer interfaces” models of the PTP (Figure 4c) [52][53][54][58].

3. ATP Synthase and the Bioenergetic Signature of Cancer

As previously mentioned, the high rates of pyruvate fermentation observed in cancer cells under aerobic conditions lead Warburg to suggest that carcinomas had an impaired bioenergetic activity of mitochondria [67]. However, while the upregulation of glycolysis in carcinomas is nowadays out of the question, the alteration of mitochondrial bioenergetics is still a matter of debate, despite the obvious evidence linking the down-regulation of mitochondrial metabolism and bioenergetics in cancer progression [12][68][69][70][71][72][73][74][75]. Perhaps the debate emerges due to the metabolic behavior of some cancer cells when growing in culture [76], differences in the mitochondrial proteome and activity in different tissues [77][78], the heterogeneity of the microenvironment of carcinomas [79][80][81][82][83] and/or the finding that metastatic disease requires an enhanced expression of OXPHOS proteins [84][85][86][87][88][89][90].
Indeed, the down-regulation of the catalytic subunit of mitochondrial ATP synthase (β-F1-ATPase) is observed in a large number of human carcinomas [12][68][70][91], either when expressed in absolute levels or normalized relative to other mitochondrial proteins, such as Hsp60 and/or to the glycolytic GAPDH. In fact, the quotient of the expression level of these three biomarkers β-F1-ATPase/Hsp60/GAPDH offers a bioenergetic cellular (BEC) index that informs about the overall capacity of mitochondria in the tissue or carcinoma [3][70].
Remarkably, a high-throughput quantitative analysis of 27 biomarkers of metabolism—using Reverse Phase Protein Array (RPPA) technology, in a cohort of 128 tumors and the corresponding paired NAT of patients bearing lung adenocarcinomas (LUAD)— confirmed that the BEC index is significantly diminished in the carcinomas, suggesting that OXPHOS activity is limited in LUAD [68], in agreement with previous reports [92][93][94].
The downregulation of the mitochondrial ATP synthase in cancer can be mediated at the expression and activity levels [7]. The expression of the catalytic subunit of ATP synthase (β-F1-ATPase) in mammalian liver is complex, including the subcellular localization of its mRNA in a structure that is frequently found attached to the outer mitochondrial membrane [95][96]. β-F1-ATPase mRNA localization and translation requires at least two cis-acting elements and a large set of RNABPs [95][97] and the 3′-UTR of the mRNA that acts as a translational enhancer sequence [98][99][100][101]
An affinity purification approach of RNABPs using as bait the full-length β-F1-ATPase mRNA allowed the identification of nine bona fide mRNA binding proteins [102]. Among them, G3BP1 (Ras-GTPase-Activating Protein SH3-Domain-Binding Protein) was found to interact both in vivo and in vitro with the 3′UTR of β-F1-ATPase mRNA, to promote inhibition of the synthesis of the protein by preventing mRNA translation [7][102][103]

5. ATPase Inhibitory Factor 1, the Physiological Inhibitor of the ATP Synthase

As mentioned above, the control of the overall ATP synthase activity is not only exerted at the protein level, but also by regulation of the content and activity of IF1, its inhibitor protein. For many years, it has been considered that under normal physiological situations, i.e., when the mitochondrial matrix pH is above neutrality, IF1 forms inactive oligomers by masking the inhibitory regions that bind to the F1 domain of the enzyme (Figure 2b) [104][105]. Only when mitochondria become de-energized, and matrix pH drops below neutrality, IF1 is considered to become activated by depolymerization and binds to the ATP synthase to inhibit its hydrolase activity (Figure 2b) [104][105].
Studies in metabolic reprograming in human carcinomas revealed that normal lung, colon and breast tissues express negligible quantities of IF1 [33] but showed a sharp increase in the expression of the protein in the carcinomas and in cultured cancer cells [30][31][33]. These findings raised the reasonable hypothesis that IF1 might represent a sort of mitochondrial “oncogene” [106], not only to control the hydrolytic activity of the ATP synthase, as it was postulated many years ago, but also its ATP synthetic activity. In other words, the overexpression of IF1 could be mediating the partial inhibition of OXPHOS contributing to the metabolic reprogramming experienced in oncogenesis. Indeed, the overexpression of IF1 in several cancer cells [30][31][33][107][108], mouse models of gain of function of IF1 [32][34][35][36][37][44] and pharmacologic in vivo approaches [109], amply demonstrated that IF1 overexpression inhibits the ATP synthetic activity of ATP synthase in isolated mitochondria, diminished tissue ATP levels, activated AMPK and promoted the metabolic rewiring of cells and tissues to an enhanced glycolytic phenotype, i.e., induced the Warburg phenotype. Conversely, silencing or knocking out IF1 in cancer cells [68][110] and mouse models of loss of function of IF1 [34][45] resulted in and enhanced activity of the ATP synthase.
Stem cells, iPSC and cancer cells have a predominant glycolytic phenotype whereas OXPHOS prevails in differentiated cells [111][112]. Interestingly, IF1 also plays a relevant role in metabolic reprogramming during differentiation of human mesenchymal stem cells (hMSC) and in the maintenance/acquisition of stemness [113]. In fact, hMSC also present high levels of IF1, altered mitochondrial structure and molecular composition, low rates of OXPHOS and an enhanced glycolytic metabolism [113].
Remarkably, the results in tissues of mouse models with regulated expression of IF1 further suggest that mitochondria in non-stressed physiological conditions of some cellular types such as cardiomyocytes [109], neurons [34] and intestinal epithelial cells [45] contain a fraction of IF1-bound and -inhibited ATP synthase, in agreement with recent cryo-EM studies of mammalian heart ATP synthases [46][47].
The regulation of IF1 activity as an inhibitor of ATP synthase is also exerted by covalent modification of the protein by phosphorylation (Figure 2b). Phosphoproteomic studies indicated that serine residues of IF1 are phosphorylated in cancer cells [114][115][116]
Indeed, the phosphorylation of IF1 has been observed when there is an increase in energy demand in the heart in vivo or in cells in cultures when they are forced to increase OXPHOS [109]. Likewise, IF1 is phosphorylated when cells progress through the OXPHOS-dependent G1-phase of the cell cycle [109]. In contrast, hypoxia and progression through the reductive phases of the cell cycle (S/G2/M) promote dephosphorylation of IF1, the reduction of OXPHOS and an enhanced glycolytic flux [109]. Consistently, dephosphorylated IF1 is the prevalent form of the inhibitor protein in lung, colon, and breast carcinomas, contributing to the inhibition of ATP synthase and the reprogramming of metabolism to an enhanced glycolysis and fermentation [109]
Most available evidence suggests that regulation of IF1 expression is exerted at post-transcriptional levels by controlling the rates of its synthesis and degradation [113]. In fact, the increase in protein content of IF1 in carcinomas [33] or its sharp reduction during cellular differentiation [113], are exerted in the absence of relevant changes in the abundance of IF1-mRNA. In fact, the IF1 protein has a very high turnover rate (2–3 h), both in cancer cells [33] and in differentiated osteocytes [113], which is much faster than the turnover of many other subunits of ATP synthase in cancer cells (18 h) [117]. IF1 degradation is mediated by several mitochondrial serine proteases [33][113] and metalloproteases [118], but the specific genes involved have not been identified yet [113].

6. Tissue-Specific Activity of IF1: Tumor Promotor and Tumor Suppressor

A fundamental characteristic of IF1 is that it is a tissue-specific and species-specific expressed protein in mouse and human tissues [42]. Human normal tissues such as the heart, brain, kidney, stomach, endometrium and liver have a high IF1 content, whereas breast, colon, and lung epithelia contain negligible levels of IF1 [33][42]. Likewise, in human carcinomas the expression of IF1 is also variable [33]. Whereas oncogenesis in epithelial cells of the endometrium, stomach and kidney does not promote an increase in IF1 expression, carcinomas in the colon, lung and breast show a very sharp increase in the content of IF1 [33].
In agreement with the pro-oncogenic role of IF1, studies in human hepatocellular carcinomas (HCC) showed that high levels of IF1 predict worse overall and progression free survival for the patients (Figure 5) [43]. Mechanistically, IF1 activates the non-canonical NFκB pathway, driving angiogenesis and metastasis through the activation of SNAI1 and VEGF [43]. Consistent with these findings, transgenic mice overexpressing in the liver the constitutively active mutant IF1-H49K promoted the inhibition of OXPHOS and a state of metabolic preconditioning guided by the activation of the stress kinases AMPK and p38 MAPK [36]. Diethylnitrosamine-induced liver carcinogenesis in IF1-H49K transgenic mice contributed to an enhanced liver carcinogenesis by augmenting proliferation and apoptotic resistance of carcinoma cells [36]. Likewise, the overexpression of IF1 in bladder carcinomas [119] and in gliomas [120] also predict a worse prognosis for the patients and a shorter time to disease recurrence (Figure 5). Mechanistically, the silencing of IF1 diminished the rates of proliferation, migration and invasive capacities of the cells, preventing epithelial mesenchymal transition (EMT) (Figure 5) [43][119][120]. In contrast, the overexpression of IF1 in these cancer cells promoted angiogenesis and activation of EMT, increasing the expression of E-cadherin and diminishing that of vimentin, to promote their metastatic capacity (Figure 5) [43][119][120].
Figure 5. The role of IF1 as tumor promotor or tumor suppressor. In bladder carcinomas, hepatocarcinomas and gliomas, high expression levels of IF1 (red) predict a poor prognosis for the patients when compared to carcinomas with low levels of IF1 (blue). In these carcinomas, IF1 acts as a tissue-specific tumor promotor facilitating proliferation, angiogenesis, epithelial mesenchymal transition (EMT) and metastasis. In contrast, in breast, colon and lung carcinomas, high expression levels of IF1 (red) predict a good prognosis for the patients when compared to carcinomas with low levels of IF1 (blue). In these carcinomas, IF1 acts as a tissue-specific tumor suppressor preventing epithelial mesenchymal transition (EMT) and metastasis by favoring cell death upon cellular detachment and immune surveillance.

7. Mitochondria as s Promising Target for Cancer Treatment

Novel therapeutic approaches based on personalized medicine are required to minimize the social and economic burden caused by cancer. The enzymes that control metabolism are promising targets to combat progression of the disease [3][121][122][123][124]. In this regard, several glycolytic inhibitors have been proposed and some of them are actually in clinical trials [3][69][124][125][126][127][128]. Mitochondrial metabolism also offers other effective targets to restrain tumor growth [124][127][129][130][131][132]. In this regard, antibiotics such as doxycycline that target the mitochondrial ribosome and inhibit translation prevent mitochondrial biogenesis which is required for the clonal expansion and survival of cancer stem cells (CSCs) [133]. Doxycycline has also been used in combination with other anticancer therapies [134][135][136][137] and their effectiveness lies in their ability to prevent metastasis by inhibiting the growth of CSCs. 

In this line, it recently found that nebivolol, a third-generation β1-blocker, halts colon and breast tumor growth in vivo by a severe inhibition of OXPHOS [138]. Mechanistically, nebivolol binds to β1-adrenergic receptors in cancer cells, causing the increase in IF1 content bound to the ATP synthase and promoting its inhibition. Concurrently, nebivolol prevents the phosphorylation of NDUFS7, a Complex I subunit, which promotes partial inhibition of the activity of the enzyme [138]. In addition, nebivolol also arrests proliferation of endothelial cells by blocking β1-adrenergic receptors impeding tumor angiogenesis [138]

This entry is adapted from the peer-reviewed paper 10.3390/cancers15153775

References

  1. Wang, R.; Green, D.R. Metabolic reprogramming and metabolic dependency in T cells. Immunol. Rev. 2012, 249, 14–26.
  2. Vasan, K.; Werner, M.; Chandel, N.S. Mitochondrial Metabolism as a Target for Cancer Therapy. Cell Metab. 2020, 32, 341–352.
  3. Cuezva, J.M.; Ortega, A.D.; Willers, I.; Sanchez-Cenizo, L.; Aldea, M.; Sanchez-Arago, M. The tumor suppressor function of mitochondria: Translation into the clinics. Biochim. Biophys. Acta 2009, 1792, 1145–1158.
  4. Vander Heiden, M.G.; Lunt, S.Y.; Dayton, T.L.; Fiske, B.P.; Israelsen, W.J.; Mattaini, K.R.; Vokes, N.I.; Stephanopoulos, G.; Cantley, L.C.; Metallo, C.M.; et al. Metabolic pathway alterations that support cell proliferation. Cold Spring Harb. Symp. Quant. Biol. 2013, 76, 325–334.
  5. Lunt, S.Y.; Vander Heiden, M.G. Aerobic glycolysis: Meeting the metabolic requirements of cell proliferation. Annu. Rev. Cell Dev. Biol. 2011, 27, 441–464.
  6. DeBerardinis, R.J.; Chandel, N.S. We need to talk about the Warburg effect. Nat Metab. 2020, 2, 127–129.
  7. Esparza-Molto, P.B.; Cuezva, J.M. Reprogramming oxidative phosphorylation in cancer: A role for RNA binding proteins. Antioxid. Redox Signal. 2020, 33, 927–945.
  8. Paul, S.; Ghosh, S.; Kumar, S. Tumor glycolysis, an essential sweet tooth of tumor cells. Semin. Cancer Biol. 2022, 86, 1216–1230.
  9. Zaidi, N.; Swinnen, J.V.; Smans, K. ATP-citrate lyase: A key player in cancer metabolism. Cancer Res. 2012, 72, 3709–3714.
  10. Jin, J.; Byun, J.K.; Choi, Y.K.; Park, K.G. Targeting glutamine metabolism as a therapeutic strategy for cancer. Exp. Mol. Med. 2023, 55, 706–715.
  11. Patra, K.C.; Hay, N. The pentose phosphate pathway and cancer. Trends Biochem. Sci. 2014, 39, 347–354.
  12. Torresano, L.; Nuevo-Tapioles, C.; Santacatterina, F.; Cuezva, J.M. Metabolic reprogramming and disease progression in cancer patients. Biochim. Et Biophys. Acta. Mol. Basis Dis. 2020, 1866, 165721.
  13. Sanzey, M.; Abdul Rahim, S.A.; Oudin, A.; Dirkse, A.; Kaoma, T.; Vallar, L.; Herold-Mende, C.; Bjerkvig, R.; Golebiewska, A.; Niclou, S.P. Comprehensive analysis of glycolytic enzymes as therapeutic targets in the treatment of glioblastoma. PLoS ONE 2015, 10, e0123544.
  14. Zhao, Z.; Lu, J.; Han, L.; Wang, X.; Man, Q.; Liu, S. Prognostic significance of two lipid metabolism enzymes, HADHA and ACAT2, in clear cell renal cell carcinoma. Tumour Biol. 2016, 37, 8121–8130.
  15. Wang, J.; Liu, F.; Ao, P.; Li, X.; Zheng, H.; Wu, D.; Zhang, N.; She, J.; Yuan, J.; Wu, X. Correlation of PDK1 expression with clinicopathologic features and prognosis of hepatocellular carcinoma. Onco Targets Ther. 2016, 9, 5597–5602.
  16. Lin, P.C.; Lin, J.K.; Yang, S.H.; Wang, H.S.; Li, A.F.; Chang, S.C. Expression of beta-F1-ATPase and mitochondrial transcription factor A and the change in mitochondrial DNA content in colorectal cancer: Clinical data analysis and evidence from an in vitro study. Int. J. Color. Dis. 2008, 23, 1223–1232.
  17. Liu, G.; Zhu, J.; Yu, M.; Cai, C.; Zhou, Y.; Yu, M.; Fu, Z.; Gong, Y.; Yang, B.; Li, Y.; et al. Glutamate dehydrogenase is a novel prognostic marker and predicts metastases in colorectal cancer patients. J. Transl. Med. 2015, 13, 144.
  18. Jin, L.; Chun, J.; Pan, C.; Kumar, A.; Zhang, G.; Ha, Y.; Li, D.; Alesi, G.N.; Kang, Y.; Zhou, L.; et al. The PLAG1-GDH1 Axis Promotes Anoikis Resistance and Tumor Metastasis through CamKK2-AMPK Signaling in LKB1-Deficient Lung Cancer. Mol. Cell 2018, 69, 87–99.e7.
  19. Li, K.; Zhang, C.; Chen, L.; Wang, P.; Fang, Y.; Zhu, J.; Chen, S.; Du, J.; Shen, B.; Wu, K.; et al. The role of acetyl-coA carboxylase2 in head and neck squamous cell carcinoma. PeerJ 2019, 7, e7037.
  20. Osugi, J.; Yamaura, T.; Muto, S.; Okabe, N.; Matsumura, Y.; Hoshino, M.; Higuchi, M.; Suzuki, H.; Gotoh, M. Prognostic impact of the combination of glucose transporter 1 and ATP citrate lyase in node-negative patients with non-small lung cancer. Lung Cancer 2015, 88, 310–318.
  21. Wang, D.; Yin, L.; Wei, J.; Yang, Z.; Jiang, G. ATP citrate lyase is increased in human breast cancer, depletion of which promotes apoptosis. Tumour Biol. 2017, 39, 1010428317698338.
  22. Cai, Y.; Wang, J.; Zhang, L.; Wu, D.; Yu, D.; Tian, X.; Liu, J.; Jiang, X.; Shen, Y.; Zhang, L.; et al. Expressions of fatty acid synthase and HER2 are correlated with poor prognosis of ovarian cancer. Med. Oncol. 2015, 32, 391.
  23. Cogliati, S.; Enriquez, J.A.; Scorrano, L. Mitochondrial Cristae: Where Beauty Meets Functionality. Trends Biochem. Sci. 2016, 41, 261–273.
  24. Wittig, I.; Schagger, H. Supramolecular organization of ATP synthase and respiratory chain in mitochondrial membranes. Biochim. Biophys. Acta 2009, 1787, 672–680.
  25. Boyer, P.D. The ATP synthase. A splendid molecular machine. Annu. Rev. Biochem. 1997, 66, 717–749.
  26. Kuhlbrandt, W. Structure and Mechanisms of F-Type ATP Synthases. Annu. Rev. Biochem. 2019, 88, 515–549.
  27. Pullman, M.E.; Monroy, G.C. A Naturally Occurring Inhibitor of Mitochondrial Adenosine Triphosphatase. J. Biol. Chem. 1963, 238, 3762–3769.
  28. Walker, J.E. The ATP synthase: The understood, the uncertain and the unknown. Biochem. Soc. Trans. 2013, 41, 1–16.
  29. Garcia-Bermudez, J.; Cuezva, J.M. The ATPase Inhibitory Factor 1 (IF1): A master regulator of energy metabolism and of cell survival. Biochim. Biophys. Acta 2016, 1857, 1167–1182.
  30. Sanchez-Cenizo, L.; Formentini, L.; Aldea, M.; Ortega, A.D.; Garcia-Huerta, P.; Sanchez-Arago, M.; Cuezva, J.M. Up-regulation of the ATPase inhibitory factor 1 (IF1) of the mitochondrial H+-ATP synthase in human tumors mediates the metabolic shift of cancer cells to a Warburg phenotype. J. Biol. Chem. 2010, 285, 25308–25313.
  31. Formentini, L.; Sánchez-Aragó, M.; Sánchez-Cenizo, L.; Cuezva, J.M. The mitochondrial ATPase Inhibitory Factor 1 (IF1) triggers a ROS-mediated retrograde pro-survival and proliferative response. Mol. Cell 2012, 45, 731–742.
  32. Zhou, B.; Caudal, A.; Tang, X.; Chavez, J.D.; McMillen, T.S.; Keller, A.; Villet, O.; Zhao, M.; Liu, Y.; Ritterhoff, J.; et al. Upregulation of mitochondrial ATPase inhibitory factor 1 (ATPIF1) mediates increased glycolysis in mouse hearts. J. Clin. Investig. 2022, 132, e155333.
  33. Sanchez-Arago, M.; Formentini, L.; Martinez-Reyes, I.; Garcia-Bermudez, J.; Santacatterina, F.; Sanchez-Cenizo, L.; Willers, I.M.; Aldea, M.; Najera, L.; Juarranz, A.; et al. Expression, regulation and clinical relevance of the ATPase inhibitory factor 1 in human cancers. Oncogenesis 2013, 2, e46.
  34. Esparza-Molto, P.B.; Romero-Carraminana, I.; Nunez de Arenas, C.; Pereira, M.P.; Blanco, N.; Pardo, B.; Bates, G.R.; Sanchez-Castillo, C.; Artuch, R.; Murphy, M.P.; et al. Generation of mitochondrial reactive oxygen species is controlled by ATPase inhibitory factor 1 and regulates cognition. PLoS Biol. 2021, 19, e3001252.
  35. Formentini, L.; Pereira, M.P.; Sanchez-Cenizo, L.; Santacatterina, F.; Lucas, J.J.; Navarro, C.; Martinez-Serrano, A.; Cuezva, J.M. In vivo inhibition of the mitochondrial H+-ATP synthase in neurons promotes metabolic preconditioning. EMBO J. 2014, 33, 762–778.
  36. Santacatterina, F.; Sanchez-Cenizo, L.; Formentini, L.; Mobasher, M.A.; Casas, E.; Rueda, C.B.; Martinez-Reyes, I.; Nunez de Arenas, C.; Garcia-Bermudez, J.; Zapata, J.M.; et al. Down-regulation of oxidative phosphorylation in the liver by expression of the ATPase inhibitory factor 1 induces a tumor-promoter metabolic state. Oncotarget 2016, 7, 490–508.
  37. Formentini, L.; Santacatterina, F.; Nunez de Arenas, C.; Stamatakis, K.; Lopez-Martinez, D.; Logan, A.; Fresno, M.; Smits, R.; Murphy, M.P.; Cuezva, J.M. Mitochondrial ROS Production Protects the Intestine from Inflammation through Functional M2 Macrophage Polarization. Cell Rep. 2017, 19, 1202–1213.
  38. Esparza-Molto, P.B.; Nuevo-Tapioles, C.; Cuezva, J.M. Regulation of the H(+)-ATP synthase by IF1: A role in mitohormesis. Cell. Mol. Life Sci. 2017, 74, 2151–2166.
  39. Chandel, N.S. Evolution of Mitochondria as Signaling Organelles. Cell Metab. 2015, 22, 204–206.
  40. Shadel, G.S.; Horvath, T.L. Mitochondrial ROS Signaling in Organismal Homeostasis. Cell 2015, 163, 560–569.
  41. Yun, J.; Finkel, T. Mitohormesis. Cell Metab. 2014, 19, 757–766.
  42. Esparza-Molto, P.B.; Nuevo-Tapioles, C.; Chamorro, M.; Najera, L.; Torresano, L.; Santacatterina, F.; Cuezva, J.M. Tissue-specific expression and post-transcriptional regulation of the ATPase inhibitory factor 1 (IF1) in human and mouse tissues. FASEB J. 2019, 33, 1836–1851.
  43. Song, R.; Song, H.; Liang, Y.; Yin, D.; Zhang, H.; Zheng, T.; Wang, J.; Lu, Z.; Song, X.; Pei, T.; et al. Reciprocal activation between ATPase inhibitory factor 1 and NF-kappaB drives hepatocellular carcinoma angiogenesis and metastasis. Hepatology 2014, 60, 1659–1673.
  44. Sanchez-Gonzalez, C.; Nuevo-Tapioles, C.; Herrero Martin, J.C.; Pereira, M.P.; Serrano Sanz, S.; Ramirez de Molina, A.; Cuezva, J.M.; Formentini, L. Dysfunctional oxidative phosphorylation shunts branched-chain amino acid catabolism onto lipogenesis in skeletal muscle. EMBO J. 2020, 39, e103812.
  45. Dominguez-Zorita, S.; Romero-Carraminana, I.; Santacatterina, F.; Esparza-Molto, P.B.; Simo, C.; Del-Arco, A.; Nunez de Arenas, C.; Saiz, J.; Barbas, C.; Cuezva, J.M. IF1 ablation prevents ATP synthase oligomerization, enhances mitochondrial ATP turnover and promotes an adenosine-mediated pro-inflammatory phenotype. Cell Death Dis. 2023, 14, 413.
  46. Gu, J.; Zhang, L.; Zong, S.; Guo, R.; Liu, T.; Yi, J.; Wang, P.; Zhuo, W.; Yang, M. Cryo-EM structure of the mammalian ATP synthase tetramer bound with inhibitory protein IF1. Science 2019, 364, 1068–1075.
  47. Pinke, G.; Zhou, L.; Sazanov, L.A. Cryo-EM structure of the entire mammalian F-type ATP synthase. Nat. Struct. Mol. Biol. 2020, 27, 1077–1085.
  48. Gross, A.; Pilcher, K.; Blachly-Dyson, E.; Basso, E.; Jockel, J.; Bassik, M.C.; Korsmeyer, S.J.; Forte, M. Biochemical and genetic analysis of the mitochondrial response of yeast to BAX and BCL-X(L). Mol. Cell. Biol. 2000, 20, 3125–3136.
  49. Santamaria, G.; Martinez-Diez, M.; Fabregat, I.; Cuezva, J.M. Efficient execution of cell death in non-glycolytic cells requires the generation of ROS controlled by the activity of mitochondrial H+-ATP synthase. Carcinogenesis 2006, 27, 925–935.
  50. Dey, R.; Moraes, C.T. Lack of oxidative phosphorylation and low mitochondrial membrane potential decrease susceptibility to apoptosis and do not modulate the protective effect of Bcl-x(L) in osteosarcoma cells. J. Biol. Chem. 2000, 275, 7087–7094.
  51. Matsuyama, S.; Xu, Q.; Velours, J.; Reed, J.C. The Mitochondrial F0F1-ATPase proton pump is required for function of the proapoptotic protein Bax in yeast and mammalian cells. Mol. Cell 1998, 1, 327–336.
  52. Mnatsakanyan, N.; Park, H.A.; Wu, J.; He, X.; Llaguno, M.C.; Latta, M.; Miranda, P.; Murtishi, B.; Graham, M.; Weber, J.; et al. Mitochondrial ATP synthase c-subunit leak channel triggers cell death upon loss of its F1 subcomplex. Cell Death Differ. 2022, 29, 1874–1887.
  53. Bernardi, P.; Carraro, M.; Lippe, G. The mitochondrial permeability transition: Recent progress and open questions. FEBS J. 2021, 289, 7051–7074.
  54. Bonora, M.; Giorgi, C.; Pinton, P. Molecular mechanisms and consequences of mitochondrial permeability transition. Nat. Rev. Mol. Cell Biol. 2022, 23, 266–285.
  55. Hunter, D.R.; Haworth, R.A. The Ca2+-induced membrane transition in mitochondria. I. The protective mechanisms. Arch. Biochem. Biophys. 1979, 195, 453–459.
  56. Kinnally, K.W.; Campo, M.L.; Tedeschi, H. Mitochondrial channel activity studied by patch-clamping mitoplasts. J. Bioenerg. Biomembr. 1989, 21, 497–506.
  57. Petronilli, V.; Szabo, I.; Zoratti, M. The inner mitochondrial membrane contains ion-conducting channels similar to those found in bacteria. FEBS Lett. 1989, 259, 137–143.
  58. Gerle, C. Mitochondrial F-ATP synthase as the permeability transition pore. Pharmacol. Res. 2020, 160, 105081.
  59. Baines, C.P.; Kaiser, R.A.; Purcell, N.H.; Blair, N.S.; Osinska, H.; Hambleton, M.A.; Brunskill, E.W.; Sayen, M.R.; Gottlieb, R.A.; Dorn, G.W.; et al. Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature 2005, 434, 658–662.
  60. Crompton, M.; Ellinger, H.; Costi, A. Inhibition by cyclosporin A of a Ca2+-dependent pore in heart mitochondria activated by inorganic phosphate and oxidative stress. Biochem. J. 1988, 255, 357–360.
  61. Giorgio, V.; von Stockum, S.; Antoniel, M.; Fabbro, A.; Fogolari, F.; Forte, M.; Glick, G.D.; Petronilli, V.; Zoratti, M.; Szabo, I.; et al. Dimers of mitochondrial ATP synthase form the permeability transition pore. Proc. Natl. Acad. Sci. USA 2013, 110, 5887–5892.
  62. Alavian, K.N.; Beutner, G.; Lazrove, E.; Sacchetti, S.; Park, H.A.; Licznerski, P.; Li, H.; Nabili, P.; Hockensmith, K.; Graham, M.; et al. An uncoupling channel within the c-subunit ring of the F1FO ATP synthase is the mitochondrial permeability transition pore. Proc. Natl. Acad. Sci. USA 2014, 111, 10580–10585.
  63. Bonora, M.; Bononi, A.; De Marchi, E.; Giorgi, C.; Lebiedzinska, M.; Marchi, S.; Patergnani, S.; Rimessi, A.; Suski, J.M.; Wojtala, A.; et al. Role of the c subunit of the FO ATP synthase in mitochondrial permeability transition. Cell Cycle 2013, 12, 674–683.
  64. Mnatsakanyan, N.; Llaguno, M.C.; Yang, Y.; Yan, Y.; Weber, J.; Sigworth, F.J.; Jonas, E.A. A mitochondrial megachannel resides in monomeric F1FO ATP synthase. Nat Commun. 2019, 10, 5823.
  65. He, J.; Ford, H.C.; Carroll, J.; Ding, S.; Fearnley, I.M.; Walker, J.E. Persistence of the mitochondrial permeability transition in the absence of subunit c of human ATP synthase. Proc. Natl. Acad. Sci. USA 2017, 114, 3409–3414.
  66. Carroll, J.; He, J.; Ding, S.; Fearnley, I.M.; Walker, J.E. Persistence of the permeability transition pore in human mitochondria devoid of an assembled ATP synthase. Proc. Natl. Acad. Sci. USA 2019, 116, 12816–12821.
  67. Warburg, O. On respiratory impairment in cancer cells. Science 1956, 124, 269–270.
  68. Torresano, L.; Santacatterina, F.; Dominguez-Zorita, S.; Nuevo-Tapioles, C.; Nunez-Salgado, A.; Esparza-Molto, P.B.; Gonzalez-Llorente, L.; Romero-Carraminana, I.; Nunez de Arenas, C.; Sanchez-Garrido, B.; et al. Analysis of the metabolic proteome of lung adenocarcinomas by reverse-phase protein arrays (RPPA) emphasizes mitochondria as targets for therapy. Oncogenesis 2022, 11, 24.
  69. Sanchez-Arago, M.; Chamorro, M.; Cuezva, J.M. Selection of cancer cells with repressed mitochondria triggers colon cancer progression. Carcinogenesis 2010, 31, 567–576.
  70. Cuezva, J.M.; Krajewska, M.; de Heredia, M.L.; Krajewski, S.; Santamaria, G.; Kim, H.; Zapata, J.M.; Marusawa, H.; Chamorro, M.; Reed, J.C. The bioenergetic signature of cancer: A marker of tumor progression. Cancer Res. 2002, 62, 6674–6681.
  71. Wang, X.; Moraes, C.T. Increases in mitochondrial biogenesis impair carcinogenesis at multiple levels. Mol. Oncol. 2011, 5, 399–409.
  72. D’Errico, I.; Salvatore, L.; Murzilli, S.; Lo Sasso, G.; Latorre, D.; Martelli, N.; Egorova, A.V.; Polishuck, R.; Madeyski-Bengtson, K.; Lelliott, C.; et al. Peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC1alpha) is a metabolic regulator of intestinal epithelial cell fate. Proc. Natl. Acad. Sci. USA 2011, 108, 6603–6608.
  73. Xing, F.; Luan, Y.; Cai, J.; Wu, S.; Mai, J.; Gu, J.; Zhang, H.; Li, K.; Lin, Y.; Xiao, X.; et al. The Anti-Warburg Effect Elicited by the cAMP-PGC1alpha Pathway Drives Differentiation of Glioblastoma Cells into Astrocytes. Cell Rep. 2017, 18, 468–481.
  74. LaGory, E.L.; Wu, C.; Taniguchi, C.M.; Ding, C.C.; Chi, J.T.; von Eyben, R.; Scott, D.A.; Richardson, A.D.; Giaccia, A.J. Suppression of PGC-1alpha Is Critical for Reprogramming Oxidative Metabolism in Renal Cell Carcinoma. Cell Rep. 2015, 12, 116–127.
  75. Liu, W.; Beck, B.H.; Vaidya, K.S.; Nash, K.T.; Feeley, K.P.; Ballinger, S.W.; Pounds, K.M.; Denning, W.L.; Diers, A.R.; Landar, A.; et al. Metastasis suppressor KISS1 seems to reverse the Warburg effect by enhancing mitochondrial biogenesis. Cancer Res. 2014, 74, 954–963.
  76. Ikari, R.; Mukaisho, K.I.; Kageyama, S.; Nagasawa, M.; Kubota, S.; Nakayama, T.; Murakami, S.; Taniura, N.; Tanaka, H.; Kushima, R.P.; et al. Differences in the Central Energy Metabolism of Cancer Cells between Conventional 2D and Novel 3D Culture Systems. Int. J. Mol. Sci. 2021, 22, 1805.
  77. Mootha, V.K.; Bunkenborg, J.; Olsen, J.V.; Hjerrild, M.; Wisniewski, J.R.; Stahl, E.; Bolouri, M.S.; Ray, H.N.; Sihag, S.; Kamal, M.; et al. Integrated analysis of protein composition, tissue diversity, and gene regulation in mouse mitochondria. Cell 2003, 115, 629–640.
  78. Fernandez-Vizarra, E.; Enriquez, J.A.; Perez-Martos, A.; Montoya, J.; Fernandez-Silva, P. Tissue-specific differences in mitochondrial activity and biogenesis. Mitochondrion 2011, 11, 207–213.
  79. Pagliarini, D.J.; Calvo, S.E.; Chang, B.; Sheth, S.A.; Vafai, S.B.; Ong, S.E.; Walford, G.A.; Sugiana, C.; Boneh, A.; Chen, W.K.; et al. A mitochondrial protein compendium elucidates complex I disease biology. Cell 2008, 134, 112–123.
  80. Kim, J.; DeBerardinis, R.J. Mechanisms and Implications of Metabolic Heterogeneity in Cancer. Cell Metab. 2019, 30, 434–446.
  81. Labuschagne, C.F.; Cheung, E.C.; Blagih, J.; Domart, M.C.; Vousden, K.H. Cell Clustering Promotes a Metabolic Switch that Supports Metastatic Colonization. Cell Metab. 2019, 30, 720–734.
  82. Hensley, C.T.; Faubert, B.; Yuan, Q.; Lev-Cohain, N.; Jin, E.; Kim, J.; Jiang, L.; Ko, B.; Skelton, R.; Loudat, L.; et al. Metabolic Heterogeneity in Human Lung Tumors. Cell 2016, 164, 681–694.
  83. Momcilovic, M.; Jones, A.; Bailey, S.T.; Waldmann, C.M.; Li, R.; Lee, J.T.; Abdelhady, G.; Gomez, A.; Holloway, T.; Schmid, E.; et al. In vivo imaging of mitochondrial membrane potential in non-small-cell lung cancer. Nature 2019, 575, 380–384.
  84. Weinberg, F.; Hamanaka, R.; Wheaton, W.W.; Weinberg, S.; Joseph, J.; Lopez, M.; Kalyanaraman, B.; Mutlu, G.M.; Budinger, G.R.; Chandel, N.S. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc. Natl. Acad. Sci. USA 2010, 107, 8788–8793.
  85. Vazquez, F.; Lim, J.H.; Chim, H.; Bhalla, K.; Girnun, G.; Pierce, K.; Clish, C.B.; Granter, S.R.; Widlund, H.R.; Spiegelman, B.M.; et al. PGC1alpha expression defines a subset of human melanoma tumors with increased mitochondrial capacity and resistance to oxidative stress. Cancer Cell 2013, 23, 287–301.
  86. LeBleu, V.S.; O’Connell, J.T.; Gonzalez Herrera, K.N.; Wikman, H.; Pantel, K.; Haigis, M.C.; de Carvalho, F.M.; Damascena, A.; Domingos Chinen, L.T.; Rocha, R.M.; et al. PGC-1alpha mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to promote metastasis. Nat. Cell Biol. 2014, 16, 992–1003, 1001-1015.
  87. Viale, A.; Pettazzoni, P.; Lyssiotis, C.A.; Ying, H.; Sanchez, N.; Marchesini, M.; Carugo, A.; Green, T.; Seth, S.; Giuliani, V.; et al. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature 2014, 514, 628–632.
  88. Sancho, P.; Burgos-Ramos, E.; Tavera, A.; Bou Kheir, T.; Jagust, P.; Schoenhals, M.; Barneda, D.; Sellers, K.; Campos-Olivas, R.; Grana, O.; et al. MYC/PGC-1alpha Balance Determines the Metabolic Phenotype and Plasticity of Pancreatic Cancer Stem Cells. Cell Metab. 2015, 22, 590–605.
  89. Martinez-Reyes, I.; Diebold, L.P.; Kong, H.; Schieber, M.; Huang, H.; Hensley, C.T.; Mehta, M.M.; Wang, T.; Santos, J.H.; Woychik, R.; et al. TCA Cycle and Mitochondrial Membrane Potential Are Necessary for Diverse Biological Functions. Mol. Cell 2016, 61, 199–209.
  90. Basu, S.; Gnanapradeepan, K.; Barnoud, T.; Kung, C.P.; Tavecchio, M.; Scott, J.; Watters, A.; Chen, Q.; Kossenkov, A.V.; Murphy, M.E. Mutant p53 controls tumor metabolism and metastasis by regulating PGC-1alpha. Genes Dev. 2018, 32, 230–243.
  91. Isidoro, A.; Martinez, M.; Fernandez, P.L.; Ortega, A.D.; Santamaria, G.; Chamorro, M.; Reed, J.C.; Cuezva, J.M. Alteration of the bioenergetic phenotype of mitochondria is a hallmark of breast, gastric, lung and oesophageal cancer. Biochem. J. 2004, 378, 17–20.
  92. Lopez-Rios, F.; Sanchez-Arago, M.; Garcia-Garcia, E.; Ortega, A.D.; Berrendero, J.R.; Pozo-Rodriguez, F.; Lopez-Encuentra, A.; Ballestin, C.; Cuezva, J.M. Loss of the mitochondrial bioenergetic capacity underlies the glucose avidity of carcinomas. Cancer Res. 2007, 67, 9013–9017.
  93. Xu, J.Y.; Zhang, C.; Wang, X.; Zhai, L.; Ma, Y.; Mao, Y.; Qian, K.; Sun, C.; Liu, Z.; Jiang, S.; et al. Integrative Proteomic Characterization of Human Lung Adenocarcinoma. Cell 2020, 182, 245–261.e17.
  94. Cuezva, J.M.; Chen, G.; Alonso, A.M.; Isidoro, A.; Misek, D.E.; Hanash, S.M.; Beer, D.G. The bioenergetic signature of lung adenocarcinomas is a molecular marker of cancer diagnosis and prognosis. Carcinogenesis 2004, 25, 1157–1163.
  95. Ricart, J.; Egea, G.; Izquierdo, J.M.; San Martin, C.; Cuezva, J.M. Subcellular structure containing mRNA for beta subunit of mitochondrial H+-ATP synthase in rat hepatocytes is translationally active. Biochem. J. 1997, 324 Pt 2, 635–643.
  96. Lithgow, T.; Cuezva, J.M.; Silver, P.A. Highways for protein delivery to the mitochondria. Trends Biochem. Sci. 1997, 22, 110–113.
  97. Egea, G.; Izquierdo, J.M.; Ricart, J.; San Martín, C.; Cuezva, J.M. mRNA encoding the beta-subunit of the mitochondrial F1-ATPase complex is a localized mRNA in rat hepatocytes. Biochem. J. 1997, 322, 557–565.
  98. Izquierdo, J.M.; Cuezva, J.M. Control of the translational efficiency of beta-F1-ATPase mRNA depends on the regulation of a protein that binds the 3’ untranslated region of the mRNA. Mol. Cell. Biol. 1997, 17, 5255–5568.
  99. Willers, I.M.; Isidoro, A.; Ortega, A.D.; Fernandez, P.L.; Cuezva, J.M. Selective inhibition of beta-F1-ATPase mRNA translation in human tumours. Biochem. J. 2010, 426, 319–326.
  100. Izquierdo, J.M.; Cuezva, J.M. Internal-ribosome-entry-site functional activity of the 3’-untranslated region of the mRNA for the beta subunit of mitochondrial H+-ATP synthase. Biochem. J. 2000, 346, 849–855.
  101. Di Liegro, C.M.; Bellafiore, M.; Izquierdo, J.M.; Rantanen, A.; Cuezva, J.M. 3’-untranslated regions of oxidative phosphorylation mRNAs function in vivo as enhancers of translation. Biochem. J. 2000, 352 Pt 1, 109–115.
  102. Ortega, A.D.; Willers, I.M.; Sala, S.; Cuezva, J.M. Human G3BP1 interacts with beta-F1-ATPase mRNA and inhibits its translation. J. Cell Sci. 2010, 123, 2685–2696.
  103. Willers, I.M.; Cuezva, J.M. Post-transcriptional regulation of the mitochondrial H(+)-ATP synthase: A key regulator of the metabolic phenotype in cancer. Biochim. Biophys. Acta 2011, 1807, 543–551.
  104. Cabezon, E.; Butler, P.J.; Runswick, M.J.; Walker, J.E. Modulation of the oligomerization state of the bovine F1-ATPase inhibitor protein, IF1, by pH. J. Biol. Chem. 2000, 275, 25460–25464.
  105. Cabezon, E.; Butler, P.J.; Runswick, M.J.; Carbajo, R.J.; Walker, J.E. Homologous and heterologous inhibitory effects of ATPase inhibitor proteins on F-ATPases. J. Biol. Chem. 2002, 277, 41334–41341.
  106. Sanchez-Arago, M.; Formentini, L.; Garcia-Bermudez, J.; Cuezva, J.M. IF1 reprograms energy metabolism and signals the oncogenic phenotype in cancer. Cell Cycle 2012, 11, 2963–2964.
  107. Kahancova, A.; Sklenar, F.; Jezek, P.; Dlaskova, A. Regulation of glucose-stimulated insulin secretion by ATPase Inhibitory Factor 1 (IF1). FEBS Lett. 2018, 592, 999–1009.
  108. Kahancova, A.; Sklenar, F.; Jezek, P.; Dlaskova, A. Overexpression of native IF1 downregulates glucose-stimulated insulin secretion by pancreatic INS-1E cells. Sci. Rep. 2020, 10, 1551.
  109. Garcia-Bermudez, J.; Sanchez-Arago, M.; Soldevilla, B.; Del Arco, A.; Nuevo-Tapioles, C.; Cuezva, J.M. PKA Phosphorylates the ATPase Inhibitory Factor 1 and Inactivates Its Capacity to Bind and Inhibit the Mitochondrial H(+)-ATP Synthase. Cell Rep. 2015, 12, 2143–2155.
  110. Gonzalez-Llorente, L.; Santacatterina, F.; Garcia-Aguilar, A.; Nuevo-Tapioles, C.; Gonzalez-Garcia, S.; Tirpakova, Z.; Toribio, M.L.; Cuezva, J.M. Overexpression of Mitochondrial IF1 Prevents Metastatic Disease of Colorectal Cancer by Enhancing Anoikis and Tumor Infiltration of NK Cells. Cancers 2019, 12, 22.
  111. Tormos, K.V.; Anso, E.; Hamanaka, R.B.; Eisenbart, J.; Joseph, J.; Kalyanaraman, B.; Chandel, N.S. Mitochondrial complex III ROS regulate adipocyte differentiation. Cell Metab. 2011, 14, 537–544.
  112. Chen, C.T.; Shih, Y.R.; Kuo, T.K.; Lee, O.K.; Wei, Y.H. Coordinated changes of mitochondrial biogenesis and antioxidant enzymes during osteogenic differentiation of human mesenchymal stem cells. Stem Cells 2008, 26, 960–968.
  113. Sanchez-Arago, M.; Garcia-Bermudez, J.; Martinez-Reyes, I.; Santacatterina, F.; Cuezva, J.M. Degradation of IF1 controls energy metabolism during osteogenic differentiation of stem cells. EMBO Rep. 2013, 14, 638–644.
  114. Sharma, K.; D’Souza, R.C.; Tyanova, S.; Schaab, C.; Wisniewski, J.R.; Cox, J.; Mann, M. Ultradeep human phosphoproteome reveals a distinct regulatory nature of Tyr and Ser/Thr-based signaling. Cell Rep. 2014, 8, 1583–1594.
  115. Zhou, H.; Di Palma, S.; Preisinger, C.; Peng, M.; Polat, A.N.; Heck, A.J.; Mohammed, S. Toward a comprehensive characterization of a human cancer cell phosphoproteome. J. Proteome Res. 2013, 12, 260–271.
  116. Christensen, G.L.; Kelstrup, C.D.; Lyngso, C.; Sarwar, U.; Bogebo, R.; Sheikh, S.P.; Gammeltoft, S.; Olsen, J.V.; Hansen, J.L. Quantitative phosphoproteomics dissection of seven-transmembrane receptor signaling using full and biased agonists. Mol. Cell. Proteom. 2010, 9, 1540–1553.
  117. Garcia-Aguilar, A.; Martinez-Reyes, I.; Cuezva, J.M. Changes in the turnover of the cellular proteome during metabolic reprogramming: A role for mtROS in proteostasis. J. Proteome Res. 2019, 18, 3142–3155.
  118. Shen, L.; Zhi, L.; Hu, W.; Wu, M.X. IEX-1 targets mitochondrial F1Fo-ATPase inhibitor for degradation. Cell Death Differ. 2009, 16, 603–612.
  119. Wei, S.; Fukuhara, H.; Kawada, C.; Kurabayashi, A.; Furihata, M.; Ogura, S.; Inoue, K.; Shuin, T. Silencing of ATPase Inhibitory Factor 1 Inhibits Cell Growth via Cell Cycle Arrest in Bladder Cancer. Pathobiology 2015, 82, 224–232.
  120. Wu, J.; Shan, Q.; Li, P.; Wu, Y.; Xie, J.; Wang, X. ATPase inhibitory factor 1 is a potential prognostic marker for the migration and invasion of glioma. Oncol. Lett. 2015, 10, 2075–2080.
  121. Vyas, S.; Zaganjor, E.; Haigis, M.C. Mitochondria and Cancer. Cell 2016, 166, 555–566.
  122. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674.
  123. Weinberg, S.E.; Chandel, N.S. Targeting mitochondria metabolism for cancer therapy. Nat. Chem. Biol. 2015, 11, 9–15.
  124. Martinez-Outschoorn, U.E.; Peiris-Pages, M.; Pestell, R.G.; Sotgia, F.; Lisanti, M.P. Cancer metabolism: A therapeutic perspective. Nat. Rev. Clin. Oncol. 2017, 14, 11–31.
  125. Sanchez-Arago, M.; Cuezva, J.M. The bioenergetic signature of isogenic colon cancer cells predicts the cell death response to treatment with 3-bromopyruvate, iodoacetate or 5-fluorouracil. J. Transl. Med. 2011, 9, 19.
  126. Hay, N. Reprogramming glucose metabolism in cancer: Can it be exploited for cancer therapy? Nat. Rev. Cancer 2016, 16, 635–649.
  127. Luengo, A.; Gui, D.Y.; Vander Heiden, M.G. Targeting Metabolism for Cancer Therapy. Cell Chem. Biol. 2017, 24, 1161–1180.
  128. Li, L.; Liang, Y.; Kang, L.; Liu, Y.; Gao, S.; Chen, S.; Li, Y.; You, W.; Dong, Q.; Hong, T.; et al. Transcriptional Regulation of the Warburg Effect in Cancer by SIX1. Cancer Cell 2018, 33, 368–385.
  129. 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.
  130. Wheaton, W.W.; Weinberg, S.E.; Hamanaka, R.B.; Soberanes, S.; Sullivan, L.B.; Anso, E.; Glasauer, A.; Dufour, E.; Mutlu, G.M.; Budigner, G.S.; et al. Metformin inhibits mitochondrial complex I of cancer cells to reduce tumorigenesis. Elife 2014, 3, e02242.
  131. Shi, Y.; Lim, S.K.; Liang, Q.; Iyer, S.V.; Wang, H.Y.; Wang, Z.; Xie, X.; Sun, D.; Chen, Y.J.; Tabar, V.; et al. Gboxin is an oxidative phosphorylation inhibitor that targets glioblastoma. Nature 2019, 567, 341–346.
  132. Leanza, L.; Romio, M.; Becker, K.A.; Azzolini, M.; Trentin, L.; Manago, A.; Venturini, E.; Zaccagnino, A.; Mattarei, A.; Carraretto, L.; et al. Direct Pharmacological Targeting of a Mitochondrial Ion Channel Selectively Kills Tumor Cells In Vivo. Cancer Cell 2017, 31, 516–531.
  133. Lamb, R.; Ozsvari, B.; Lisanti, C.L.; Tanowitz, H.B.; Howell, A.; Martinez-Outschoorn, U.E.; Sotgia, F.; Lisanti, M.P. Antibiotics that target mitochondria effectively eradicate cancer stem cells, across multiple tumor types: Treating cancer like an infectious disease. Oncotarget 2015, 6, 4569–4584.
  134. Lamb, R.; Fiorillo, M.; Chadwick, A.; Ozsvari, B.; Reeves, K.J.; Smith, D.L.; Clarke, R.B.; Howell, S.J.; Cappello, A.R.; Martinez-Outschoorn, U.E.; et al. Doxycycline down-regulates DNA-PK and radiosensitizes tumor initiating cells: Implications for more effective radiation therapy. Oncotarget 2015, 6, 14005–14025.
  135. De Francesco, E.M.; Maggiolini, M.; Tanowitz, H.B.; Sotgia, F.; Lisanti, M.P. Targeting hypoxic cancer stem cells (CSCs) with Doxycycline: Implications for optimizing anti-angiogenic therapy. Oncotarget 2017, 8, 56126–56142.
  136. De Francesco, E.M.; Bonuccelli, G.; Maggiolini, M.; Sotgia, F.; Lisanti, M.P. Vitamin C and Doxycycline: A synthetic lethal combination therapy targeting metabolic flexibility in cancer stem cells (CSCs). Oncotarget 2017, 8, 67269–67286.
  137. Ozsvari, B.; Magalhaes, L.G.; Latimer, J.; Kangasmetsa, J.; Sotgia, F.; Lisanti, M.P. A Myristoyl Amide Derivative of Doxycycline Potently Targets Cancer Stem Cells (CSCs) and Prevents Spontaneous Metastasis, without Retaining Antibiotic Activity. Front. Oncol. 2020, 10, 1528.
  138. Nuevo-Tapioles, C.; Santacatterina, F.; Stamatakis, K.; Nunez de Arenas, C.; Gomez de Cedron, M.; Formentini, L.; Cuezva, J.M. Coordinate beta-adrenergic inhibition of mitochondrial activity and angiogenesis arrest tumor growth. Nat Commun. 2020, 11, 3606.
More
This entry is offline, you can click here to edit this entry!
ScholarVision Creations