Skip to main content
Current Urology logoLink to Current Urology
. 2022 Aug 31;16(4):207–212. doi: 10.1097/CU9.0000000000000120

Androgen receptor signaling–mitochondrial DNA–oxidative phosphorylation: A critical triangle in early prostate cancer

Minas Sakellakis a,, Laura Jacqueline Flores b
PMCID: PMC9875216  PMID: 36714229

Abstract

Mitochondria are more than just the cellular powerhouse. They also play key roles in vital functions such as apoptosis, metabolism regulation, and other intracellular interactions. The mitochondrial DNA (mtDNA) encodes for 12 subunits of the oxidative phosphorylation (OXPHOS) system. Depletion of mtDNA in androgen-dependent prostate cancer (PCa) cell lines renders them androgen-independent and more aggressive. Paradoxically, pharmaceutical inhibition of OXPHOS is lethal for subsets of PCa cells, whereas others become dependent on androgen receptor (AR) signaling for survival. Given that the AR-mitochondria interaction is critical for early PCa, it is crucial to understand the details of this interaction. Technical hurdles have made mitochondria traditionally difficult to study, with many techniques used for isolation masking the properties of given individual mitochondria. Although the isolation of mitochondria enables us to study OXPHOS, we miss the context in which mitochondria interact with the rest of the cell. Both AR signaling and mtDNA affect apoptosis, metabolism regulation, cellular calcium storage and homeostasis, intracellular calcium signaling, and redox homeostasis. In this review, we will attempt to understand how the crosstalk between AR-mtDNA-OXPHOS is responsible for “life or death” decisions inside the cells. Our aim is to point toward potential vulnerabilities that can lead to the discovery of novel therapeutic targets.

Keywords: Androgen receptor, Mitochondria, Oxidative phosphorylation, Prostate cancer

1. Introduction

Mitochondria are mostly known for their role as the cellular powerhouses [1]. The mitochondrial oxidative phosphorylation (OXPHOS) system consists of 5 multisubunit complexes.[2] These complexes play a critical role in electron transfer and cellular energy production in the form of ATP. Mitochondria also regulate cellular metabolism, cellular calcium homeostasis, cell proliferation, and apoptosis.[1] They also play a role in steroid synthesis, hormonal signaling, immune signaling, heme synthesis, and so on.[1] Moreover, mitochondrial defects have been implicated in tumorigenesis and cancer aggressiveness.[35] The resulting oxygen accumulation has been linked to the expression and activation of protumorigenic signaling such as Ras, ERK, Akt, and nuclear factor κB.[69] Mitochondrial dysfunction also has a direct inhibitory effect on apoptosis and the function of tumor suppressors (eg, p53).[1012] In addition, OXPHOS dysfunction favors a metabolic shift toward glycolysis, which is known as the Warburg effect, a well-established hallmark of cancer.[13]

2. Literature review

2.1. Mitochondrial DNA

The vast majority of mammalian mitochondrial proteins (approximately 1200) are encoded and expressed by the nuclear genome, but a small subset of these proteins is encoded by mitochondrial DNA (mtDNA).[2,14] The protein components of complexes I, III, IV, and V are encoded both from nuclear DNA and mtDNA.[15,16] The subunits of complex II are encoded only by nuclear DNA.[16] The mtDNA encodes for 12 subunits of the OXPHOS system, 2 ribosomal RNAs, and 14 tRNAs.[17,18] It encodes for subunits 1, 2, 3, 4, 5, and 6 of complex I, cytochrome b of complex III; Subunits 1, 2, and 3 for complex IV; and F0 subunits 6 and 8 of complex V (ATP synthase).[1922] Reduced levels or defective mtDNA can cause imbalances in the structure of the OXPHOS complexes and result in defective mitochondrial respiration. Although the main activity of these proteins is related to OXPHOS and energy production, they are not limited solely to their role as parts of the respiratory chain. For example, protein 1 of complex I is a substrate of caspase-3 and plays a critical role in the induction of apoptosis; complex IV has been observed in extra-mitochondrial locations, and so on.[23,24] It has also been proposed that mutations or deletions in mtDNA might play a causative role in tumorigenesis and cancer aggressiveness, but robust experimental evidence is still lacking.[25,26]

2.2. Prostate cancer and mitochondrial DNA

Prostate cancer (PCa) is among the most frequently diagnosed cancers in the world. One of the most notable characteristics of the disease is the tumor cell dependence on the androgen receptor (AR) for activation of luminal differentiation, proliferation, and survival.[27] The role of mitochondria in PCa is also very important.[28] Depletion of mtDNA prevents apoptosis and induces PCa progression.[29] It also induces epithelial-to-mesenchymal transition and promotes tumor cell motility by upregulating phosphatidylinositol-3-kinase/Akt2 signaling.[30] Reduction of mtDNA can also result in a more invasive phenotype via increased migration onto the basement membrane protein laminin-1 and decreased expression of poly(ADP-ribose) polymerase 1.[31] In vitro studies have shown that the depletion of mtDNA from androgen-dependent lymph node carcinoma of the prostate (LNCaP) cells results in androgen-dependence loss.[32,33] Strikingly, the depleted cells grow significantly faster than the parental cell line. Restoration of mtDNA results in restoration of androgen dependence. The androgen-independent cell line C4-2 established by inoculation of LNCaP cells in castrated mice has 8 times less amount of normal mtDNA than the parental LNCaP. Androgen-independent cell lines PC3 and DU125 exhibit less mtDNA content than LNCaP cells as well. These results indicate that mtDNA is connected to androgen dependence in PCa cells. Interestingly, mtDNA-depleted LNCaP cells in the absence of androgens grow faster than both LNCaP cells and C4-2 cells.[32,33] This is particularly surprising because most mtDNA-depleted cells grow slower than their parental cells, because of loss of energy generation via normal cellular respiration. In vivo studies in athymic nude mice were also in line with these findings, where mtDNA depletion was sufficient to render the cells androgen-independent and resistant to the effects of androgen deprivation.[32]

2.3. Oxidative phosphorylation inhibition in prostate cancer

It is well known that PCa metabolism relies more and more on glycolysis as it evolves toward the aggressive phenotype.[2833] Based on the previous results, one would expect that the pharmaceutical OXPHOS inhibition in androgen-dependent cancer cells will result in a cellular reprogramming that renders them androgen-independent, more aggressive, and more dependent on aerobic glycolysis. What is interesting is that when we inhibit OXPHOS in LNCaP cells, we observe the exact opposite effect in vitro and in vivo. In the context of androgen deprivation and OXPHOS inhibition, almost all LNCaP cells eventually die, mostly through apoptosis.[3437] The cells not only do not become androgen-independent when exposed to oligomycin (complex V inhibitor) or IACS-010729 (complex I inhibitor), but also the addition of androgens has a prosurvival effect. Oxidative phosphorylation inhibition in C4-2 and C4-2B cells (androgen-independent derivatives of LNCaP cells) poses a similar antitumor effect, albeit smaller. The addition of androgens also has a prosurvival effect in C4-2 and C4-2B cells under OXPHOS inhibition. Moreover, in vertebral cancer of the prostate (VCaP) cells, the combination of OXPHOS inhibition with oligomycin and androgen deprivation is lethal for all cells within 72 hours in vitro. When VCaP cells under oligomycin are exposed to androgens, not only do they escape death, but they also continue growing. VCaP cells harbor increased AR expression via AR gene amplification.

These results are interesting and suggest that OXPHOS inhibition might be used to synergize the antitumor effects of androgen deprivation in subsets of androgen-dependent PCa cells. One must be careful, though, because complex V inhibition does not equal complex I or II or III or IV inhibition. For example, in PCa, succinate anaplerosis has a tumor-promoting effect and restores cellular respiration through complex II.[38] This can overcome complex I inhibition. We also mentioned previously that subunit 1 of complex I is a substrate of caspase-3.[23] Although inhibition of complexes III, IV, or V can trigger proapoptotic signals, complex I inhibition can potentially sabotage apoptosis.[39] It would be preferable to target complex III, IV, or V, but so far, toxicity has been limiting our efforts. At this time, mostly complex I inhibitors are being studied in clinical trials.[40] Only time will tell if there is a therapeutic window to use various OXPHOS inhibitors in clinical practice for the treatment of PCa patients. Interestingly, reformulated niclosamide, a splice-variant AR-V7 inhibitor, has shown promising synergistic antitumor effects with androgen deprivation in PCa patients.[41] Niclosamide is also a potent mitochondrial respiratory chain uncoupler, suggesting that at least part of its activity might be due to OXPHOS inhibition.[42]

2.4. Androgen receptor effects in mitochondria

We previously mentioned that mtDNA mutation or depletion contributes to PCa tumorigenesis, induces androgen independence, and enhances proliferation. However, pharmaceutical OXPHOS inhibition is detrimental for androgen-dependent cells, whereas androgens have a positive survival effect. What is the cause of this paradox?

Complete depletion of mtDNA makes LNCaP cells proliferate faster, but pharmaceutical OXPHOS inhibition makes them undergo apoptosis or necrosis. Thus, the presence of some mitochondrial activity causes LNCaP cells to undergo apoptosis under pharmaceutical OXPHOS inhibition and/or prevents a metabolic shift toward glycolysis to generate ATP to avoid necrosis. And this activity is (at least partly) inhibited by AR. It is well known that AR activity and mitochondria are connected.[43,44] Upon activation, AR is imported into the cells and localized to the mitochondria. There it plays multiple roles in regulating multiple mitochondrial processes. The import of AR in the mitochondria is dependent on a 36-amino-acid-long mitochondrial localization sequence. Increased expression of AR decreases OXPHOS. Mitochondrial impairment increases AR expression and in turn increases its localization inside the mitochondria.[44] This creates a loop that enables cancer cells to survive AR ablation or OXPHOS inhibition by upregulating OXPHOS or AR signaling, respectively.

2.5. Apoptosis

It is well known that AR inactivation or inhibition has proapoptotic effects in PCa cell lines.[45] The 2 most understood apoptotic mechanisms include the intrinsic pathway and the extrinsic pathway. The intrinsic pathway is also known as the mitochondrial pathway. Intracellular signals generated from cellular stress, such as energy depletion (eg, from OXPHOS inhibition), hypoxia, heat, radiation, nutrient deprivation, infection, or increased intracellular calcium concentration activate the intrinsic pathway of apoptosis.[4548] A multitude of Bax/Bak homodimers and heterodimers are then inserted into the outer mitochondrial membrane, which results in the release of cytochrome c from the mitochondria. Cytochrome c binds with apoptotic protease activating factor-1 and ATP to create the apoptosome. The apoptosome cleaves procaspases to create caspase-3 and caspase-9. Mitochondria also release proteins known as second mitochondria-derived activator of caspases, which bind to cytosolic factors, which inhibit apoptosis, thereby inactivating them and further promoting apoptosis.[49] Hence, the presence of functional mitochondria is critical for the execution of apoptosis after the energy depletion that OXPHOS inhibition causes. Several studies demonstrated that mtDNA depletion prevents apoptosis and contributes to tumor progression and metastasis.[5054] Subunit 1 of complex I is a substrate of caspase-3. Caspase-3, apart from its essential role in the apoptotic body formation, also functions before or at the stage when commitment to loss of cell viability is made.[23,55] Androgen withdrawal triggers the programmed cell death in androgen-dependent PCa cells.[56] Hence, both AR signaling and mtDNA depletion have prosurvival effects. However, the interplay between AR and apoptosis is complex.[57] For example, the apoptosis-inducing activity of tumor suppressor protein retinoblastoma is AR-dependent.[58] Androgen-independent PCa cells do not initiate apoptosis during androgen deprivation, but they retain the ability to do so when sufficiently damaged by exogenous factors.[59]

2.6. Calcium cell storage and homeostasis

Mitochondria regulate calcium cell homeostasis and calcium storage.[60,61] Calcium ions regulate every aspect of cell function, including gene expression, movement, secretion, proliferation, metabolism, and so on. Calcium signals have to be tightly regulated to prevent cell injury.[62] Prostate cancer cells are characterized by calcium signals that are different from those in normal cells with regard to subcellular localization, amplitude or kinetics.[63,64] Calcium-dependent modifications in PCa cells rely on altered calcium homeostasis, which is caused by alterations in plasma membrane and endoplasmic reticulum channels, as well as gap junctions.[65] These changes result in influx/efflux ratio changes and altered calcium storage and sequestration.[64,66] It is known that mtDNA mutations/polymorphisms result in defective mitochondrial calcium regulation.[67,68] Increased levels of cytosolic calcium might overactivate calcium-binding proteins involved in tumor cell progression. One example is calcium/calmodulin-dependent kinase II (CAMKKII), which seems to play an important role in the ability of PCa cells to evade apoptosis (inhibits caspase-7 and caspase-8) and to progress into an androgen-independent state.[69] Androgen receptor signaling also promotes CAMKKII signaling. Calcium signaling is involved in the activity of AR on PCa proliferation.[69,70] Moreover, in LNCaP cells, androgen-dependent increases in intracellular calcium concentrations have been observed.[71] Upregulation of T-type calcium channels increases several proliferative signals such as Akt kinase, mTOR, CDK4, and others.[72,73] Androgen-dependent activation of CAMKKII signaling also promotes the glucose transporter GLUT12 trafficking to the plasma membrane and shifts cellular metabolism toward glycolysis.[74] In addition, CAMKKII overexpression promotes PCa growth via de novo lipogenesis.[75]

2.7. Metabolism regulation

Normal prostate epithelial cells halt tricarboxylic acid cycle to increase the secretion of citrate, which enhances sperm viability. In order to cover their energy needs, they use a relatively glycolytic metabolism.[76,77] On the other hand, early-stage prostate adenocarcinoma reprograms metabolism to enable tumor progression, by consuming citrate to power OXPHOS and to fuel lipogenesis. Despite the strict definition of the Warburg effect as aerobic glycolysis, early PCa displays both increased glycolytic and mitochondrial activity.[78] Androgen receptor signaling directly or indirectly regulates the expression and activity of several metabolism-related enzymes, such as fatty acid synthase, α-methylacyl-CoA-racemase or hexokinase 1/2. It also regulates the axis CAMKK2-AMPK-phosphofructokinase, Myc, SLC2A1 (facilitated glucose transporter), and so on. Hence, AR activity is a major regulator of metabolism in PCa and promotes both glycolysis and OXPHOS.[79] However, metabolism regulation in PCa cells is very complex and does not rely solely on AR. For example, c-Myc contributes to metabolic adaptations favoring glycolysis and glutaminolysis, often with the cooperation of hypoxia-inducible factor α and/or a mutated p53.[78,80,81] It is known that androgen-dependent PCa cells use a metabolic switch to survive androgen ablation.[36] Respiration-deficient LNCaP cells due to mtDNA depletion begin to grow in an androgen-independent manner.[28,29] It is evident that androgen-independent mechanisms rewire cellular metabolism to promote tumor cell survival and proliferation. However, potent OXPHOS inhibition with oligomycin makes subsets of PCa cells dependent on AR for survival.[34] This suggests that, in these cells, the mechanism that enables cells to switch their metabolism and become aggressive is likely still present, but these cells appear (for reasons not yet fully elucidated) to “choose” to die, unless AR signaling prevents them.

2.8. Reactive oxygen species formation

Mitochondrial respiration is a leading source of reactive oxygen species (ROS) that can cause considerable cell damage and even trigger cell death if levels become too high. Cancer cells have higher ROS than their noncancerous cells of origin.[82] The presence of basal levels of ROS is essential for protumorigenic signaling, important for cell survival, differentiation, and proliferation.[83] In PCa, chronically increased intracellular ROS (mainly via 5-lipoxygenase activation) results in ligand-independent Src-mediated activation of prosurvival epidermal growth factor receptor signaling.[84] The proliferative activity of LNCaP cells increases when exposed to low H2O2.[85] On the other hand, excessive amounts of ROS can trigger oxidative damage and cellular death.[83] Cancer cells counterbalance the detrimental effects of ROS by upregulating the production of antioxidant molecules, such as reduced glutathione and thioredoxin, which rely on the reducing power of nicotinamide adenine dinucleotide phosphate to maintain their antioxidative activities.[86] Mutations or deletions in mtDNA may result in defective respiratory chains and increased ROS production.[87] On the other hand, mtDNA depletion results in decreased ROS levels. This can affect cell proliferation and survival in variable ways, depending on the cell type.[82,83] Pharmacological inhibition of OXPHOS can also have a variable effect in ROS production. This depends on the specific site of the respiratory chain that the drug acts on. However, the most widely used OXPHOS inhibitors, such as rotenone, antimycin-C, or oligomycin, impede proton flow and promote ROS production.[88] Increased AR signaling has been shown to increase basal ROS levels in PCa cells.[89] Moreover, AR signaling enhances tumor cell proliferation, which in turn increases ROS levels.[90] The presence of ROS is required for androgen stimulation in androgen-dependent cells.[91] Increased ROS levels upregulate AR, whereas decreased ROS levels downregulate AR.[92,93] Moreover, AR signaling contributes to the redox balance by upregulating 6-phosphogluconate dehydrogenase, thus activating the pentose phosphate pathway and the production of nicotinamide adenine dinucleotide phosphate.[94] Androgen receptor also regulates glutathione S-transferases and attenuates oxidative stress in a redox environment.[95]

2.9.Role of tumor suppressors

Various PCa cell lines have different metabolism regulation mechanisms and different metabolic dependencies. The tumor suppressor status is a well-known determinant of how cells respond to mitochondrial damage and metabolic stress.[11,12]PTEN, retinoblastoma, and TP53 are the most recurrently altered tumor suppressor genes in treatment-resistant PCa.[96] Cooperative loss of 2 or more of them is frequently a sign that the disease has progressed into an aggressive variant.[97] PTEN loss is found in more than 40% of metastatic castrate-resistant PCs.[98] PTEN protein is a well-known metabolism regulator.[98] PTEN loss can promote tumor growth via increased metabolic flux of glycolysis, glutaminolysis, and fatty acid metabolism.[99101] Interestingly, mitochondrial complex I inhibition with deguelin was found to be selectively toxic for PTEN-deficient PCa cells.[102] This is in line with experiments showing that LNCaP cells do not survive pharmacologic complex I or complex V inhibition. LNCaP cells harbor one mutated PTEN allele and one deleted allele and do not express PTEN protein.[103]TP53 is not only the guardian of the genome that protects against ROS formation, but also an important metabolic regulator.[104,105] Aberrations in TP53 can result in increased aerobic glycolysis. TP53 protects against mtDNA mutations or deletions, and loss of TP53 results in mtDNA depletion.[106] This further promotes an energy flux toward aerobic glycolysis. VCaP cells harbor mutated TP53.[107] In the presence of AR signaling, VCaP cells not only survive ATP synthase inhibition with oligomycin, but they also continue growing. Retinoblastoma deletion in PCa also increases tumorigenic growth by reprogramming lipid and amino acid metabolism.[108] It also protects against ROS formation through increased glutathione synthesis.[108] Other tumor suppressors such as ATM, BRCA1/2, and so on, can also play a role in the metabolism regulation of PCa cells.[109111] Sometimes, the activity of tumor suppressor genes can be affected by the status of other suppressor genes or other regulatory genes.[112,113] In this case, the combination of genetic aberrations becomes more important than the status of individual genes. Given that cancer cells usually harbor numerous mutations, it is evident that every cancer carries a unique genetic signature. This implies that the tumor models that we usually use in research laboratories have limitations as they are only approximations of subsets of real-life cancers.

3. Conclusions

Technical hurdles have made mitochondria traditionally difficult to study, with many techniques used for isolation masking the properties of given individual mitochondria. Given that AR-mitochondria interaction in early PCa cells is responsible for life or death decisions, it is crucial to understand the details of this interaction. This will enable us to identify mechanisms that can be targeted alone or in combination with antiandrogen therapy. The selective toxicity of antiandrogen therapy to PCa cells can be exploited either via the discovery of a synergistic mechanism or by reducing the dose of other drugs to levels nontoxic to normal cells.

Conflict of interest statement

No conflict of interest has been declared by the authors.

Acknowledgments

None.

Statement of ethics

Not applicable.

Funding source

All authors received no financial support for this work.

Author contributions

MS: Concept of the review, review of the literature, drafting of and critical review of the final version of the manuscript;

LJF: Review of the literature, drafting of and critical review of the final version of the manuscript.

Footnotes

How to cite this article: Sakellakis M, Flores LJ. Androgen receptor signaling–mitochondrial DNA–oxidative phosphorylation: A critical triangle in early prostate cancer. Curr Urol 2022;16(4):207–212. doi: 10.1097/CU9.0000000000000120.

References

  • 1.McBride HM, Neuspiel M, Wasiak S. Mitochondria: More than just a powerhouse. Curr Biol 2006;16(14):R551–R560. [DOI] [PubMed] [Google Scholar]
  • 2.Signes A, Fernandez-Vizarra E. Assembly of mammalian oxidative phosphorylation complexes I–V and supercomplexes. Essays Biochem 2018;62(3):255–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Carew JS, Huang P. Mitochondrial defects in cancer. Mol Cancer 2002;1:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Zong WX, Rabinowitz JD, White E. Mitochondria and cancer. Mol Cell 2016;61(5):667–676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hsu CC, Tseng LM, Lee HC. Role of mitochondrial dysfunction in cancer progression. Exp Biol Med (Maywood) 2016;241(12):1281–1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Koundouros N, Poulogiannis G. Phosphoinositide 3-kinase/Akt signaling and redox metabolism in cancer. Front Oncol 2018;8:160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Rezatabar S Karimian A Rameshknia V, et al. RAS/MAPK signaling functions in oxidative stress, DNA damage response and cancer progression. J Cell Physiol 2019;234(9):14951–14965. [DOI] [PubMed] [Google Scholar]
  • 8.Morgan MJ, Liu ZG. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res 2011;21(1):103–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Cook CC, Kim A, Terao S, Gotoh A, Higuchi M. Consumption of oxygen: A mitochondrial-generated progression signal of advanced cancer. Cell Death Dis 2012;3(1):e258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wang C, Youle RJ. The role of mitochondria in apoptosis. Annu Rev Genet 2009;43:95–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Vaseva AV, Moll UM. The mitochondrial p53 pathway. Biochim Biophys Acta 2009;1787(5):414–420. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Mihara M Erster S Zaika A, et al. p53 has a direct apoptogenic role at the mitochondria. Mol Cell 2003;11(3):577–590. [DOI] [PubMed] [Google Scholar]
  • 13.Chandra D, Singh KK. Genetic insights into OXPHOS defect and its role in cancer. Biochim Biophys Acta 2011;1807(6):620–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Gammage PA, Frezza C. Mitochondrial DNA: The overlooked oncogenome? BMC Biol 2019;17(1):53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Taanman JW. The mitochondrial genome: Structure, transcription, translation and replication. Biochim Biophys Acta 1999;1410(2):103–123. [DOI] [PubMed] [Google Scholar]
  • 16.Procaccio V Mousson B Beugnot R, et al. Nuclear DNA origin of mitochondrial complex I deficiency in fatal infantile lactic acidosis evidenced by transnuclear complementation of cultured fibroblasts. J Clin Invest 1999;104(1):83–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Barshad G, Marom S, Cohen T, Mishmar D. Mitochondrial DNA transcription and its regulation: An evolutionary perspective. Trends Genet 2018;34(9):682–692. [DOI] [PubMed] [Google Scholar]
  • 18.Barchiesi A, Vascotto C. Transcription, processing, and decay of mitochondrial RNA in health and disease. Int J Mol Sci 2019;20(9):2221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Mimaki M, Wang X, McKenzie M, Thorburn DR, Ryan MT. Understanding mitochondrial complex I assembly in health and disease. Biochim Biophys Acta 2012;1817(6):851–862. [DOI] [PubMed] [Google Scholar]
  • 20.Gil Borlado MC Moreno Lastres D Gonzalez Hoyuela M, et al. Impact of the mitochondrial genetic background in complex III deficiency. PLoS One 2010; 5(9):e12801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lazarou M, Smith SM, Thorburn DR, Ryan MT, McKenzie M. Assembly of nuclear DNA-encoded subunits into mitochondrial complex IV, and their preferential integration into supercomplex forms in patient mitochondria. FEBS J 2009;276(22):6701–6713. [DOI] [PubMed] [Google Scholar]
  • 22.Song J, Pfanner N, Becker T. Assembling the mitochondrial ATP synthase. Proc Natl Acad Sci U S A 2018;115(12):2850–2852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ricci JE Muñoz-Pinedo C Fitzgerald P, et al. Disruption of mitochondrial function during apoptosis is mediated by caspase cleavage of the p75 subunit of complex I of the electron transport chain. Cell 2004;117(6):773–786. [DOI] [PubMed] [Google Scholar]
  • 24.Sadacharan SK, Singh B, Bowes T, Gupta RS. Localization of mitochondrial DNA encoded cytochrome c oxidase subunits I and II in rat pancreatic zymogen granules and pituitary growth hormone granules. Histochem Cell Biol 2005;124(5):409–421. [DOI] [PubMed] [Google Scholar]
  • 25.Schon EA, DiMauro S, Hirano M. Human mitochondrial DNA: Roles of inherited and somatic mutations. Nat Rev Genet 2012;13(12):878–890. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Larman TC DePalma SR Hadjipanayis AG, et al. Spectrum of somatic mitochondrial mutations in five cancers. Proc Natl Acad Sci U S A 2012;109(35):14087–14091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Heinlein CA, Chang C. Androgen receptor in prostate cancer. Endocr Rev 2004;25(2):276–308. [DOI] [PubMed] [Google Scholar]
  • 28.Petros JA Baumann AK Ruiz-Pesini E, et al. mtDNA mutations increase tumorigenicity in prostate cancer. Proc Natl Acad Sci U S A 2005;102(3):719–724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chaudhary AK O'Malley J Kumar S, et al. Mitochondrial dysfunction and prostate cancer racial disparities among American men. Front Biosci (Schol Ed) 2017;9(1):154–164. [DOI] [PubMed] [Google Scholar]
  • 30.Moro L, Arbini AA, Yao JL, di Sant'Agnese PA, Marra E, Greco M. Mitochondrial DNA depletion in prostate epithelial cells promotes anoikis resistance and invasion through activation of PI3K/Akt2. Cell Death Differ 2009;16(4):571–583. [DOI] [PubMed] [Google Scholar]
  • 31.Moro L, Arbini AA, Marra E, Greco M. Mitochondrial DNA depletion reduces PARP-1 levels and promotes progression of the neoplastic phenotype in prostate carcinoma. Cell Oncol 2008;30(4):307–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Higuchi M Kudo T Suzuki S, et al. Mitochondrial DNA determines androgen dependence in prostate cancer cell lines. Oncogene 2006;25(10):1437–1445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Naito A Cook CC Mizumachi T, et al. Progressive tumor features accompany epithelial-mesenchymal transition induced in mitochondrial DNA-depleted cells. Cancer Sci 2008;99(8):1584–1588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Sakellakis M, Ramachandran S, Zhang XQ, Logothetis C, Titus M. PO-017 androgen deprivation and oxidative phosphorylation inhibition pose synergistic antitumor effects in subsets of prostate tumours in vitro. ESMO Open 2018;3(suppl 2):A234. [Google Scholar]
  • 35.Basu H Wilganowski N Robertson S, et al. Metabolic switch from glycolysis to oxidative phosphorylation (ox-phos) provides survival advantage to anti-androgen-treated prostate cancer cells and make them vulnerable to mitochondrial metabolism inhibitors IACS-010759 and CB-839. Cancer Res 2020;80(suppl 16):4791.32855208 [Google Scholar]
  • 36.Basu HS Wilganowski N Robertson S, et al. Prostate cancer cells survive anti-androgen and mitochondrial metabolic inhibitors by modulating glycolysis and mitochondrial metabolic activities. Prostate 2021;81(12):799–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Yadav N Kumar S Marlowe T, et al. Oxidative phosphorylation-dependent regulation of cancer cell apoptosis in response to anticancer agents. Cell Death Dis 2015;6(11):e1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Sant'Anna-Silva ACB Perez-Valencia JA Sciacovelli M, et al. Succinate anaplerosis has an onco-driving potential in prostate cancer cells. Cancers (Basel) 2021;13(7):1727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kwong JQ, Henning MS, Starkov AA, Manfredi G. The mitochondrial respiratory chain is a modulator of apoptosis. J Cell Biol 2007;179(6):1163–1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Yap T Rodon Ahnert J Piha-Paul SA, et al. Phase I trial of IACS-010759 (IACS), a potent, selective inhibitor of complex I of the mitochondrial electron transport chain, in patients (pts) with advanced solid tumors. J Clin Oncol 2019;37(suppl 15):3014–3014. [Google Scholar]
  • 41.Parikh M Liu C Wu CY, et al. Phase Ib trial of reformulated niclosamide with abiraterone/prednisone in men with castration-resistant prostate cancer. Sci Rep 2021;11(1):6377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Figarola JL, Singhal J, Singhal S, Kusari J, Riggs A. Bioenergetic modulation with the mitochondria uncouplers SR4 and niclosamide prevents proliferation and growth of treatment-naïve and vemurafenib-resistant melanomas. Oncotarget 2018;9(97):36945–36965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kobayashi A, Azuma K, Ikeda K, Inoue S. Mechanisms underlying the regulation of mitochondrial respiratory chain complexes by nuclear steroid receptors. Int J Mol Sci 2020;21(18):6683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bajpai P, Koc E, Sonpavde G, Singh R, Singh KK. Mitochondrial localization, import, and mitochondrial function of the androgen receptor. J Biol Chem 2019;294(16):6621–6634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Elmore S. Apoptosis: A review of programmed cell death. Toxicol Pathol 2007;35(4):495–516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Loreto C La Rocca G Anzalone R, et al. The role of intrinsic pathway in apoptosis activation and progression in Peyronie's disease. Biomed Res Int 2014;2014:616149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Eguchi Y, Shimizu S, Tsujimoto Y. Intracellular ATP levels determine cell death fate by apoptosis or necrosis. Cancer Res 1997;57(10):1835–1840. [PubMed] [Google Scholar]
  • 48.Nirmala JG, Lopus M. Cell death mechanisms in eukaryotes. Cell Biol Toxicol 2020;36(2):145–164. [DOI] [PubMed] [Google Scholar]
  • 49.Fesik SW, Shi Y. Structural biology. Controlling the caspases. Science 2001;294(5546):1477–1478. [DOI] [PubMed] [Google Scholar]
  • 50.Chen H Wang J Liu Z, et al. Mitochondrial DNA depletion causes decreased ROS production and resistance to apoptosis. Int J Mol Med 2016;38(4):1039–1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.El-Hattab AW, Scaglia F. Mitochondrial DNA depletion syndromes: Review and updates of genetic basis, manifestations, and therapeutic options. Neurotherapeutics 2013;10(2):186–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Rizzo F Ronchi D Salani S, et al. Selective mitochondrial depletion, apoptosis resistance, and increased mitophagy in human Charcot-Marie-Tooth 2A motor neurons. Hum Mol Genet 2016;25(19):4266–4281. [DOI] [PubMed] [Google Scholar]
  • 53.Kenny TC, Germain D. mtDNA, metastasis, and the mitochondrial unfolded protein response (UPRmt). Front Cell Dev Biol 2017;5:37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Ferraresi R Troiano L Pinti M, et al. Resistance of mtDNA-depleted cells to apoptosis. Cytometry A 2008;73(6):528–537. [DOI] [PubMed] [Google Scholar]
  • 55.Porter AG, Jänicke RU. Emerging roles of caspase-3 in apoptosis. Cell Death Differ 1999;6(2):99–104. [DOI] [PubMed] [Google Scholar]
  • 56.Amirghofran Z, Monabati A, Gholijani N. Androgen receptor expression in relation to apoptosis and the expression of cell cycle related proteins in prostate cancer. Pathol Oncol Res 2004;10(1):37–41. [DOI] [PubMed] [Google Scholar]
  • 57.Lin Y Kokontis J Tang F, et al. Androgen and its receptor promote Bax-mediated apoptosis. Mol Cell Biol 2006;26(5):1908–1916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Wang X, Deng H, Basu I, Zhu L. Induction of androgen receptor-dependent apoptosis in prostate cancer cells by the retinoblastoma protein. Cancer Res 2004;64(4):1377–1385. [DOI] [PubMed] [Google Scholar]
  • 59.McKenzie S, Kyprianou N. Apoptosis evasion: The role of survival pathways in prostate cancer progression and therapeutic resistance. J Cell Biochem 2006;97(1):18–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Brini M, Calì T, Ottolini D, Carafoli E. Intracellular calcium homeostasis and signaling. Met Ions Life Sci 2013;12:119–168. [DOI] [PubMed] [Google Scholar]
  • 61.Romero-Garcia S, Prado-Garcia H. Mitochondrial calcium: Transport and modulation of cellular processes in homeostasis and cancer [review]. Int J Oncol 2019;54(4):1155–1167. [DOI] [PubMed] [Google Scholar]
  • 62.Santulli G, Xie W, Reiken SR, Marks AR. Mitochondrial calcium overload is a key determinant in heart failure. Proc Natl Acad Sci U S A 2015;112(36):11389–11394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Ardura JA, Álvarez-Carrión L, Gutiérrez-Rojas I, Alonso V. Role of calcium signaling in prostate cancer progression: Effects on cancer hallmarks and bone metastatic mechanisms. Cancers (Basel) 2020;12(5):1071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Flourakis M, Prevarskaya N. Insights into Ca2+ homeostasis of advanced prostate cancer cells. Biochim Biophys Acta 2009;1793(6):1105–1109. [DOI] [PubMed] [Google Scholar]
  • 65.Dubois C Vanden Abeele F Lehen'kyi V, et al. Remodeling of channel-forming ORAI proteins determines an oncogenic switch in prostate cancer. Cancer Cell 2014;26(1):19–32. [DOI] [PubMed] [Google Scholar]
  • 66.De Stefani D, Rizzuto R, Pozzan T. Enjoy the trip: Calcium in mitochondria back and forth. Annu Rev Biochem 2016;85:161–192. [DOI] [PubMed] [Google Scholar]
  • 67.McKenzie M, Duchen MR. Impaired cellular bioenergetics causes mitochondrial calcium handling defects in MT-ND5 mutant cybrids. PLoS One 2016;11(4):e0154371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Kato T. Role of mitochondrial DNA in calcium signaling abnormality in bipolar disorder. Cell Calcium 2008;44(1):92–102. [DOI] [PubMed] [Google Scholar]
  • 69.Cohen MB, Rokhlin OW. Mechanisms of prostate cancer cell survival after inhibition of AR expression. J Cell Biochem 2009;106(3):363–371. [DOI] [PubMed] [Google Scholar]
  • 70.Dadwal UC, Chang ES, Sankar U. Androgen receptor-CaMKK2 axis in prostate cancer and bone microenvironment. Front Endocrinol (Lausanne) 2018;9:335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Sun YH, Gao X, Tang YJ, Xu CL, Wang LH. Androgens induce increases in intracellular calcium via a G protein–coupled receptor in LNCaP prostate cancer cells. J Androl 2006;27(5):671–678. [DOI] [PubMed] [Google Scholar]
  • 72.Silvestri R Pucci P Venalainen E, et al. T-type calcium channels drive the proliferation of androgen-receptor negative prostate cancer cells. Prostate 2019;79(13):1580–1586. [DOI] [PubMed] [Google Scholar]
  • 73.Sallán MC, Visa A, Shaikh S, Nàger M, Herreros J, Cantí C. T-type Ca2+ channels: T for targetable. Cancer Res 2018;78(3):603–609. [DOI] [PubMed] [Google Scholar]
  • 74.White MA Tsouko E Lin C, et al. GLUT12 promotes prostate cancer cell growth and is regulated by androgens and CaMKK2 signaling. Endocr Relat Cancer 2018;25(4):453–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Penfold L Woods A Muckett P, et al. CAMKK2 promotes prostate cancer independently of AMPK via increased lipogenesis. Cancer Res 2018;78(24):6747–6761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Bader DA, McGuire SE. Tumour metabolism and its unique properties in prostate adenocarcinoma. Nat Rev Urol 2020;17(4):214–231. [DOI] [PubMed] [Google Scholar]
  • 77.Eidelman E, Twum-Ampofo J, Ansari J, Siddiqui MM. The metabolic phenotype of prostate cancer. Front Oncol 2017;7:131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Barfeld SJ, Itkonen HM, Urbanucci A, Mills IG. Androgen-regulated metabolism and biosynthesis in prostate cancer. Endocr Relat Cancer 2014;21(4):T57–T66. [DOI] [PubMed] [Google Scholar]
  • 79.Zacharias N Lee J Ramachandran S, et al. Androgen receptor signaling in castration-resistant prostate cancer alters hyperpolarized pyruvate to lactate conversion and lactate levels in vivo. Mol Imaging Biol 2019;21(1):86–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Zawacka-Pankau J Grinkevich VV Hünten S, et al. Inhibition of glycolytic enzymes mediated by pharmacologically activated p53: Targeting Warburg effect to fight cancer. J Biol Chem 2011;286(48):41600–41615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Jiang P Du W Wang X, et al. p53 Regulates biosynthesis through direct inactivation of glucose-6-phosphate dehydrogenase. Nat Cell Biol 2011;13(3):310–316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Liou GY, Storz P. Reactive oxygen species in cancer. Free Radic Res 2010;44(5):479–496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Boese AC, Kang S. Mitochondrial metabolism-mediated redox regulation in cancer progression. Redox Biol 2021;42:101870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Giannoni E, Fiaschi T, Ramponi G, Chiarugi P. Redox regulation of anoikis resistance of metastatic prostate cancer cells: Key role for Src and EGFR-mediated pro-survival signals. Oncogene 2009; 28(20):2074–2086. [DOI] [PubMed] [Google Scholar]
  • 85.Polytarchou C, Hatziapostolou M, Papadimitriou E. Hydrogen peroxide stimulates proliferation and migration of human prostate cancer cells through activation of activator protein-1 and up-regulation of the heparin affin regulatory peptide gene. J Biol Chem 2005;280(49):40428–40435. [DOI] [PubMed] [Google Scholar]
  • 86.Jaganjac M, Milkovic L, Sunjic SB, Zarkovic N. The NRF2, thioredoxin, and glutathione system in tumorigenesis and anticancer therapies. Antioxidants (Basel) 2020;9(11):1151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hahn A, Zuryn S. Mitochondrial genome (mtDNA) mutations that generate reactive oxygen species. Antioxidants (Basel) 2019;8(9):392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zhao RZ, Jiang S, Zhang L, Yu ZB. Mitochondrial electron transport chain, ROS generation and uncoupling [review]. Int J Mol Med 2019;44(1):3–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Lu JP Monardo L Bryskin I, et al. Androgens induce oxidative stress and radiation resistance in prostate cancer cells though NADPH oxidase. Prostate Cancer Prostatic Dis 2010;13(1):39–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Aggarwal V Tuli HS Varol A, et al. Role of reactive oxygen species in cancer progression: Molecular mechanisms and recent advancements. Biomolecules 2019;9(11):735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Han C Wang Z Xu Y, et al. Roles of reactive oxygen species in biological behaviors of prostate cancer. Biomed Res Int 2020;2020:1269624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Shigemura K Sung SY Kubo H, et al. Reactive oxygen species mediate androgen receptor- and serum starvation-elicited downstream signaling of ADAM9 expression in human prostate cancer cells. Prostate 2007;67(7):722–731. [DOI] [PubMed] [Google Scholar]
  • 93.Shiota M, Yokomizo A, Naito S. Pro-survival and anti-apoptotic properties of androgen receptor signaling by oxidative stress promote treatment resistance in prostate cancer. Endocr Relat Cancer 2012;19(6):R243–253. [DOI] [PubMed] [Google Scholar]
  • 94.Gillis JL Hinneh JA Ryan NK, et al. A feedback loop between the androgen receptor and 6-phosphogluoconate dehydrogenase (6PGD) drives prostate cancer growth. Elife 2021;10:e62592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Tinti F, Soory M. Mechanisms for redox actions of nicotine and glutathione in cell culture, relevant to periodontitis. Sci Rep 2012;2:566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Hamid AA Gray KP Shaw G, et al. Compound genomic alterations of TP53, PTEN, and RB1 tumor suppressors in localized and metastatic prostate cancer. Eur Urol 2019;76(1):89–97. [DOI] [PubMed] [Google Scholar]
  • 97.Aparicio AM Shen L Tapia EL, et al. Combined tumor suppressor defects characterize clinically defined aggressive variant prostate cancers. Clin Cancer Res 2016;22(6):1520–1530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Ferraldeschi R Nava Rodrigues D Riisnaes R, et al. PTEN protein loss and clinical outcome from castration-resistant prostate cancer treated with abiraterone acetate. Eur Urol 2015;67(4):795–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Chen CY, Chen J, He L, Stiles BL. PTEN: Tumor suppressor and metabolic regulator. Front Endocrinol (Lausanne) 2018;9:338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Qian X Li X Shi Z, et al. PTEN suppresses glycolysis by dephosphorylating and inhibiting autophosphorylated PGK1. Mol Cell 2019;76(3):516–527.e7. [DOI] [PubMed] [Google Scholar]
  • 101.Zhou X Yang X Sun X, et al. Effect of PTEN loss on metabolic reprogramming in prostate cancer cells. Oncol Lett 2019;17(3):2856–2866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Naguib A Mathew G Reczek CR, et al. Mitochondrial complex I inhibitors expose a vulnerability for selective killing of PTEN-null cells. Cell Rep 2018;23(1):58–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Lotan TL Gurel B Sutcliffe S, et al. PTEN protein loss by immunostaining: Analytic validation and prognostic indicator for a high risk surgical cohort of prostate cancer patients. Clin Cancer Res 2011;17(20):6563–6573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Simabuco FM, Morale MG, Pavan ICB, Morelli AP, Silva FR, Tamura RE. p53 And metabolism: From mechanism to therapeutics. Oncotarget 2018;9(34):23780–23823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Liu J, Zhang C, Hu W, Feng Z. Tumor suppressor p53 and metabolism. J Mol Cell Biol 2019;11(4):284–292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Lebedeva MA, Eaton JS, Shadel GS. Loss of p53 causes mitochondrial DNA depletion and altered mitochondrial reactive oxygen species homeostasis. Biochim Biophys Acta 2009;1787(5):328–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Guedes LB Almutairi F Haffner MC, et al. Analytic, preanalytic, and clinical validation of p53 IHC for detection of TP53 missense mutation in prostate cancer. Clin Cancer Res 2017;23(16):4693–4703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Mandigo AC Yuan W Xu K, et al. RB/E2F1 as a master regulator of cancer cell metabolism in advanced disease. Cancer Discov 2021;11(9):2334–2353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Dahl ES, Aird KM. Ataxia-telangiectasia mutated modulation of carbon metabolism in cancer. Front Oncol 2017;7:291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Schayek H, Haugk K, Sun S, True LD, Plymate SR, Werner H. Tumor suppressor BRCA1 is expressed in prostate cancer and controls insulin-like growth factor I receptor (IGF-IR) gene transcription in an androgen receptor–dependent manner. Clin Cancer Res 2009;15(5):1558–1565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Castro E, Eeles R. The role of BRCA1 and BRCA2 in prostate cancer. Asian J Androl 2012;14(3):409–414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Nyquist MD Corella A Coleman I, et al. Combined TP53 and RB1 loss promotes prostate cancer resistance to a spectrum of therapeutics and confers vulnerability to replication stress. Cell Rep 2020;31(8):107669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Stodden GR Lindberg ME King ML, et al. Loss of Cdh1 and Trp53 in the uterus induces chronic inflammation with modification of tumor microenvironment. Oncogene 2015;34(19):2471–2482. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Current Urology are provided here courtesy of Wolters Kluwer Health

RESOURCES