Next Article in Journal
The Superiority of Fibroblast Activation Protein Inhibitor (FAPI) PET/CT Versus FDG PET/CT in the Diagnosis of Various Malignancies
Next Article in Special Issue
Contribution of Mitochondrial Activity to Doxorubicin-Resistance in Osteosarcoma Cells
Previous Article in Journal
Modulating T Cell Responses by Targeting CD3
Previous Article in Special Issue
Metabolic Health, Mitochondrial Fitness, Physical Activity, and Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unraveling the Peculiar Features of Mitochondrial Metabolism and Dynamics in Prostate Cancer

Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(4), 1192; https://doi.org/10.3390/cancers15041192
Submission received: 13 January 2023 / Revised: 8 February 2023 / Accepted: 10 February 2023 / Published: 13 February 2023
(This article belongs to the Special Issue Mitochondrial Functions in Cancer)

Abstract

:

Simple Summary

Mitochondria are organelles involved in different biological processes, including tumorigenesis. Accumulating evidence strongly supports that, despite the presence of an active glycolytic pathway (the Warburg effect), cancer cells undergo a mitochondrial metabolic rewiring towards the OXPHOS pathway, leading to the production of high levels of ATP to sustain their uncontrolled proliferation and aggressive behavior. Alterations of the mitochondrial structural dynamics (biogenesis, fusion, fission, mitophagy) are also involved in cancer growth and progression. The mechanisms underlying this mitochondrial plasticity, now considered as a hallmark of cancer, were shown to occur with specific features in different cancer cell types and contexts. In this review, we provide an incisive description of the peculiar metabolic rewiring and structural dynamics occurring in prostate cancer cells during the different phases of their transformation from healthy cells to early- and late-stage cancer cells. We also address the role of these mitochondrial dynamics as effective targets of therapeutic approaches.

Abstract

Prostate cancer (PCa) is the second leading cause of cancer deaths among men in Western countries. Mitochondria, the “powerhouse” of cells, undergo distinctive metabolic and structural dynamics in different types of cancer. PCa cells experience peculiar metabolic changes during their progression from normal epithelial cells to early-stage and, progressively, to late-stage cancer cells. Specifically, healthy cells display a truncated tricarboxylic acid (TCA) cycle and inefficient oxidative phosphorylation (OXPHOS) due to the high accumulation of zinc that impairs the activity of m-aconitase, the enzyme of the TCA cycle responsible for the oxidation of citrate. During the early phase of cancer development, intracellular zinc levels decrease leading to the reactivation of m-aconitase, TCA cycle and OXPHOS. PCa cells change their metabolic features again when progressing to the late stage of cancer. In particular, the Warburg effect was consistently shown to be the main metabolic feature of late-stage PCa cells. However, accumulating evidence sustains that both the TCA cycle and the OXPHOS pathway are still present and active in these cells. The androgen receptor axis as well as mutations in mitochondrial genes involved in metabolic rewiring were shown to play a key role in PCa cell metabolic reprogramming. Mitochondrial structural dynamics, such as biogenesis, fusion/fission and mitophagy, were also observed in PCa cells. In this review, we focus on the mitochondrial metabolic and structural dynamics occurring in PCa during tumor development and progression; their role as effective molecular targets for novel therapeutic strategies in PCa patients is also discussed.

1. Introduction

Mitochondria are organelles involved in different cellular processes, including cell proliferation and intrinsic apoptosis, redox and Ca2+ homeostasis as well as cell stemness. They are also known as the master producers of ATP, being deeply involved in cellular energy metabolism; in addition, being dynamic organelles, they often undergo structural changes, including biogenesis, fusion/fission and mitophagy. Specifically, it is now well recognized that mitochondria undergo complex functional and structural dynamics in cancer cells during the different phases of tumor growth and progression.

1.1. Mitochondrial Metabolism in Cancer Cells

It is now recognized that cancer cells, growing in a hypoxic and hyponutrient microenvironment, are forced to adapt their metabolism (“metabolic reprogramming”) to obtain the required amount of biomass and energy to sustain their uncontrolled proliferation and aggressive behavior.
According to the theory proposed by Otto Warburg in the 1920s (named the Warburg effect), cancer cells are characterized by high rates of glucose uptake and preferentially metabolize it through the glycolytic pathway, even in the presence of adequate amounts of oxygen (aerobic glycolysis) and functional mitochondria [1,2]. Although a small amount of ATP per mole of glucose is produced through glycolysis, it is believed that this metabolic process rapidly produces high levels of metabolites to sustain the biosynthesis of the molecules (i.e., amino acids, fatty acids and nucleotides) required for cancer cell growth and division [3,4,5,6,7,8]. Moreover, the high levels of lactate produced at the end of glycolysis by lactate dehydrogenase (LDH) are secreted by cancer cells to generate an acidic tumor microenvironment promoting their transition to the most aggressive (i.e., migratory, invasive) phenotype and affecting the immune microenvironment to induce an immune tolerant condition [9,10].
Despite the presence of an active glycolytic pathway, several recent data strongly support the coexistence of functional mitochondria in cancer cells, even in the metastatic phase [6,11,12,13,14,15,16,17,18,19,20]. Mitochondria, known as the “powerhouse of the cell”, are deeply involved in the cellular metabolic dynamics, being the major intracellular producers of ATP through the oxidative phosphorylation (OXPHOS) pathway; these organelles are also the “venue” where the tricarboxylic acid (TCA) cycle takes place to provide the most building blocks for the synthesis of biomolecules. A high level of fatty acid β-oxidation also occurs in mitochondria to sustain the production of citrate. Moreover, glutaminolysis is activated to convert glutamine into intermediates for the synthesis of amino acids and nucleotides, as well as into glutamate to fuel the TCA cycle. Together, these metabolic pathways are necessary for cell anabolism to trigger cell proliferation and metastasis in cancers [8,20,21,22,23,24,25,26,27,28,29,30].
A metabolic remodeling based on a shift towards the OXPHOS machinery has also been shown to be involved in the development of drug resistance in tumor cells [31,32,33,34] and to occur in the subpopulation of cancer stem cells (CSCs), known to play a key role in tumor relapse, to provide sufficient amounts of energy and metabolites for their self-renewal and evasion from cell death induced by anticancer drugs [35,36,37,38,39,40,41,42,43].
Cancer cell metabolic plasticity has also been reported to be regulated by neighboring cells in the tumor microenvironment through both mechanical and chemical factors [18,44]. For instance, it has been shown that cancer-associated fibroblasts (CAFs) secrete lactate that is taken up by tumor cells to trigger their metabolic switch towards the OXPHOS energy-producing pathway and reactive oxygen species (ROS) generation [45,46,47,48].
The key role played by mitochondrial metabolism in cancer is strongly supported by the observation that different compounds were shown to exert their anticancer activity by targeting the oxidative phosphorylation pathways [42,49,50,51,52,53,54,55,56].
In addition to their central role in energy production, these organelles are also involved in different biological processes, such as ROS production and signaling, Ca2+ homeostasis and apoptosis [57,58,59,60,61]. ROS are generated as by-products of an impaired activity of the mitochondrial electron transport chain (ETC), specifically of respiratory complexes I, II and III, leading to an excessive production of electrons that are directly transferred to oxygen-producing superoxides [62]. It is now clear that the effects of ROS on oxidative cell signaling depend on the specific type of generated ROS, their localization and, in particular, their concentration [63,64,65,66,67]. In cancer cells, low or moderate levels of ROS were shown to be associated with activated mitogenic pathways, supporting cell growth and metastatic behavior, and with a survival advantage to stress-inducing stimuli from the microenvironment [57,64,68,69]. On the other hand, high levels of ROS were widely reported to induce DNA damage and to trigger pro-death mechanisms (i.e., apoptosis, paraptosis, necroptosis) [57,70,71] as well as to mediate the activity of several anticancer drugs [72]. ROS overproduction is frequently observed as a consequence of increased endoplasmic reticulum (ER)-derived Ca2+ levels in mitochondria. Ca2+ overload and ROS-associated mitochondrial dysfunctions have been reported to be linked to the induction of apoptosis and paraptosis [54,73,74,75,76].

1.2. Mitochondrial Dynamics in Cancer Cells

Mitochondria are also highly dynamic organelles undergoing changes in number and structure through different processes such as biogenesis, fusion, fission (fragmentation) and mitophagy (removal of impaired mitochondria). A balance between these processes is required for the maintenance of homeostasis in healthy cells; on the other hand, adaptations of mitochondrial dynamics have been widely reported in cells undergoing metabolic and stressful conditions (i.e., glucose starvation, hypoxia), such as cancer cells [8,77,78,79,80,81,82].
Mitochondrial biogenesis is the generation of new organelles from pre-existing ones. In cancer cells, an increase in mitochondrial mass is induced by a variety of stressful signals and has been found to correlate with cell growth, invasiveness, metastasis and drug resistance [83,84]. The master regulator of mitochondrial biogenesis is peroxisome proliferator-activated receptor gamma coactivator 1α (PGC1α) which cooperates with different transcription factors to increase the expression of the mitochondrial transcription factor (TFAM), the final effector of the increase of mitochondrial mass [85]. PGC1α, activated by phosphorylation by the energy sensor adenosine monophosphate-activated protein kinase (AMPK) and by deacetylation by silent information regulator 1 (SIRT1), also triggers the transcription of both nuclear and mitochondrial genes, leading to increased mitochondrial mass, OXPHOS activity and ATP production [86,87,88]. Mitochondrial biogenesis has been shown to mediate the ability of CSCs to overcome antitumor therapies [89,90,91].
An imbalance in the mitochondrial fusion/fission leads to peculiar changes of the morphological features of these organelles (interconnected vs. fragmented). GTPases belonging to the dynamin family play a pivotal role in mediating both of these processes [16,60,79,82]. Mitochondrial fusion is the physical merging of the outer membranes (OMM) and the inner membranes (IMM) of distinct mitochondria and depends on GTP hydrolysis. This process foresees the activity of three GTPases, mitofusin (MFN) 1 and 2 on the OMM and optic atrophy protein 1 (OPA1) on the IMM. MFN 1 and 2 interact to induce the formation of strict connections between adjacent mitochondria, leading to the fusion of the OMM. Then, OPA1 interacts with MFNs forming intermembrane protein complexes, thus coupling the fusion of OMMs with IMMs [92,93,94,95]. Mitochondrial fission is a multi-step process allowing the division of one mitochondrion, leading to the formation of new organelles. The key protein involved in this process is dynamin-related protein 1 (DRP1), a cytosolic GTPase. Endoplasmic reticulum (ER) membranes get in touch with mitochondria allowing a Ca2+ flux from the ER into the mitochondria, thus favoring actin polymerization and inner mitochondrial membrane constriction at this site. At the level of ER-mitochondria contact sites, different proteins such as mitochondrial dynamics 49 and 51 (MID49 and MID51), MFF and mitochondrial fission 1 (FIS1) proteins, identified as DRP1 receptors, are also located. DRP1 binds to these proteins to encircle and shrink the mitochondria, finally leading to their fission. Accumulating evidence demonstrates that an imbalance in these mitochondrial dynamics occurs in different types of cancer [79,96,97,98,99].
Mitophagy is the process by which damaged mitochondria are selectively removed, thus representing a key mechanism of mitochondrial quantity and quality control. This process foresees the engulfment of mitochondria in autophagosomes by which the organelles are transferred to lysosomes where they are degraded [100]. The PTEN-induced serine/threonine kinase 1 (PINK1)/Parkin (an E3 ubiquitin ligase) pathway plays a pivotal role in detecting dysfunctional mitochondria and in activating mechanisms leading to their removal and replacement. In the presence of damaged mitochondria, PINK1 localizes at the OMM level where it phosphorylates (i.e., activates) Parkin, which triggers the ubiquitination of different target proteins (mitophagy receptors), such as FIS1, MFN1/2, Miro and voltage-dependent anion channel-1 (VDAC1). These proteins, in turn, recruit the autophagic adaptors p62 and optineurin (OPTN) and selectively interact with the autophagosomal marker LC3 that mediates mitochondria engulfment into autophagosomes, thus, ultimately, leading to their degradation at the lysosomal level [101,102,103]. Dysregulations of the mitochondrial mitophagic process have been reported to occur in different diseases, including cancer [102,104,105,106]. Last but not least, it has been reported that changes in the mitochondrial structural dynamics together with mtDNA damage (i.e., mutations, altered copy number and transcription) correlate with changes in mitochondrial respiration and ATP production in cancer cells.

2. Mitochondrial Metabolism and Dynamics in PCa

Prostate cancer (PCa) represents the most common malignancy among men and the second leading cause of cancer deaths in developed countries [107]. Androgen-deprivation therapy (ADT), based on gonadotropin-releasing hormone (GnRH) agonists and antagonists, either alone or in combination with androgen receptor antagonists (enzalutamide, apalutamide, darolutamide), still remains the most common treatment for androgen-dependent PCa patients [108,109,110,111,112,113,114]. However, PCa often progresses towards the castration-resistant phase (CRPC), a condition characterized by the acquisition of a more aggressive and metastatic behavior even in the absence of circulating androgens [115,116]. The standard treatment of CRPC patients is presently based on chemotherapy (docetaxel) either alone or in combination with GnRH analogs, antiandrogens or inhibitors of androgen synthesis (abiraterone) [117,118,119,120,121,122,123,124,125,126,127]. Novel therapeutic strategies, such as immune check-point inhibitors or CAR-T approaches, are under investigation [128,129,130,131]. The elucidation of novel molecular hallmarks of tumor progression (i.e., mitochondrial metabolism and dynamics) will likely pave the way for the development of novel therapeutic approaches for PCa patients. Based on the above considerations, as well as on the progressively accumulating data in the literature, in this review we aim to highlight and discuss the recent findings on the involvement of mitochondrial functional reprogramming and structural dynamics in PCa, specifically in its CRPC stage; we also address the impact of these mechanisms as molecular targets of novel and effective antitumor strategies for this aggressive disease.

2.1. Mitochondrial Metabolism

Metabolic reprogramming is a well-recognized hallmark of cancer, enabling cancer cells to acquire properties that support cell survival, proliferation and acquisition of aggressive (invasive, metastatic) features. However, peculiar molecular mechanisms of this metabolic rewiring have been reported to occur in different types of tumor cells, and this metabolic heterogeneity confers differences in their proliferative/metastatic potential. In prostate epithelial cells, distinctive changes of cell metabolism have been highlighted during the different phases of their conversion from healthy cells to early-stage and, progressively, to late-stage cancer cells [31,132,133,134,135].

2.1.1. Metabolic Rewiring

Healthy epithelial cells, located in the peripheral zone of the prostate, exhibit a peculiar metabolic programming aimed at producing and secreting citrate into the prostatic fluid, one of the most relevant functions of this gland [136]. In most mammalian cells, pyruvate, the end product of the glycolytic pathway, is transported into the mitochondria where it is decarboxylated to acetyl-CoA. Acetyl-CoA subsequently binds to oxaloacetic acid to form citrate that enters the TCA cycle. Citrate is then converted into its isomer isocitrate that is further oxidized into the TCA cycle for the progression to OXPHOS and ATP production; mitochondrial aconitase (m-aconitase) is the ROS-sensitive key enzyme responsible for the citrate-isocitrate conversion. In normal prostate epithelial cells, m-aconitase activity is inhibited, resulting in the impairment of citrate oxidation followed by its accumulation and secretion [137]. The inhibition of m-aconitase strictly correlates with the ability of these cells to accumulate zinc, due to their elevated expression of its transporter ZIP1; high intramitochondrial zinc levels increase ROS generation, leading to the inhibition of m-aconitase activity and resulting in a truncated TCA cycle [138]. As a consequence, the healthy “zinc-accumulating, citrate-producing” epithelial cells are characterized by an inefficient OXPHOS which is compensated by an increased glycolytic pathway to support citrate production [139]. High zinc levels were also found to be associated with a mitochondrial apoptotic phenotype mediated by the release of cytochrome c and caspase activation [140].
On other hand, it is now well established that prostate epithelial cells undergo a peculiar metabolic rewiring during the early phases of cancer development. Specifically, elevated levels of the TCA cycle enzymes and intermediate metabolites could be detected in prostate cancer tissues in comparison to adjacent normal tissues [141,142,143]. Intracellular zinc levels were found to be significantly reduced in PCa cells, thus leading to the reactivation of m-aconitase, citrate oxidation, TCA cycle pathways and oxidative phosphorylation [144]. This reduction was shown to be related to a decreased expression of zinc transporters, such as ZIP1 and 3 [145], mediated by the hypermethylation of their gene promoters [146]. The low levels of zinc also allow cancer cells to avoid apoptosis; actually, it has been reported that zinc treatments trigger cell death and promote chemosensitivity in PCa cells [147]. In line with these observations, very low zinc levels were observed in PCa tissues [148]. Taken together, these data support that the transformation of prostate epithelial cells into their tumoral phenotype is associated with an efficient reactivation of the TCA cycle/OXPHOS metabolic pathway to meet their increased energy and metabolite demand [149].
Interestingly, prostate epithelial cells seem to possess a markedly metabolic plasticity by changing their mitochondrial metabolic features again when progressing from the early-stage towards the late-stage (i.e., metastasis) of cancer, even in the presence of low intracellular zinc levels. Specifically, the Warburg effect (i.e., increased glycolytic activity) has been proposed as the prominent metabolic feature of metastatic prostate tumors [149,150]. Mechanistically, the PI3K-AKT-mammalian target of the rapamycin (mTOR) pathway, a key driver of tumor progression, was shown to play a causal role in prostate tumorigenesis through the up-regulation of pyruvate kinase isoenzyme type M2 (PKM2), the rate-limiting enzyme catalyzing the final reaction of the glycolytic pathway [151,152]. Mutations of the tumor suppressor p53, frequently occurring in advanced prostate cancers, were reported to trigger the Warburg effect. Moreover, deletions of the tumor suppressor PTEN, often observed in aggressive prostate tumors, were demonstrated to correlate with an increased expression of hexokinase 2 (HK2), the initial enzyme of glycolysis, catalyzing the phosphorylation of glucose by ATP to glucose-6-P through the AKT-mTOR pathway [149,153]. Taken together, these observations support that PTEN and p53 tumor suppressors, together with the PI3K-AKT-mTOR pathway, are essential drivers of the Warburg effect to maintain a sufficient energy and metabolites supply for PCa growth and progression [154]. In addition, it has been demonstrated that, in prostatic carcinoma cell lines, the hypoxic conditions of the tumor microenvironment trigger the expression of SUMO1/sentrin-specific peptidase 1 (SENP1) that in turn interacts with HIF1α to promote the Warburg effect and sustain cell proliferation [155]. In line with these data, Sun and coworkers recently reported that HK2 and HIF1α are highly expressed in PCa tissues and their expression correlates with tumor growth and metastasis [156].
There is also consistent evidence that an association exists between obesity and the risk of PCa growth [157]. A deleterious bidirectional cross-talk between PCa cells and adipocytes in their microenvironment has been demonstrated [158]. Specifically, it has been reported that PCa cells educate neighboring adipocytes towards a lipolytic phenotype, resulting in free glycerol production and secretion; adipocyte-derived glycerol is then uptaken by PCa cells to enter and fuel the glycolytic pathway [159]. Moreover, adipocyte conditioning of PCa cells leads to an increased expression of glycolytic genes, resulting in lactate production and OXPHOS inhibition [159,160]. In line with these observations, we recently reported that adipocyte-released extracellular vesicles significantly decrease the sensitivity of PCa cells to the chemotherapeutic drug docetaxel and this effect is associated with an AKT/HIF-1α axis-related Warburg effect, which is characterized by enhanced glucose consumption, lactate release and ATP production [161].
To confirm that PCa cells undergo dynamic metabolic changes at each stage of tumor development, Vayalil and Landar introduced the “mitochondrial oncobioenergetic index (MOBI)” (i.e., the mathematical representation of the oncobioenergetic features of a tumor cell). In PCa cells with progressive malignant behaviors, they demonstrated that MOBI values (representative of OXPHOS activity) are high in premalignant prostate cells and significantly decrease with increasing malignancy [133].
Taken together, these observations support that PCa cells reprogram their metabolism towards the aerobic glycolysis (the Warburg effect) in the context of tumor progression. However, contrasting results, demonstrating that the high levels of OXPHOS activity observed in primary tumors still persists during the progression of the pathology towards its metastatic stage, have also been reported in the literature [162,163,164,165].
Galbraith and coworkers recently demonstrated an association between peroxisome proliferator-activated receptor gamma (PPARG) expression and metastatic features in PCa. By means of in vitro and in vivo studies, these authors could show that, in PCa cells, PPARG overexpression induces AKT3 expression leading to increased mitochondrial biogenesis and ATP production, finally fueling tumor cell epithelial-to-mesenchymal transition (EMT) and metastatic behavior [166]. Pyruvate dehydrogenase complex (PDC) is the multi-protein complex that catalyzes the conversion of pyruvate to acetyl-CoA, thus fostering mitochondrial activity. It was reported that, in PCa cells, knockout of the major subunit of PDC (PDHA1) is accompanied by lower levels of the TCA cycle activity, resulting in impaired OXPHOS activity and growth of these cells when xenografted in nude mice [163]. Pyruvate kinase isozyme 2 (PKM2) has been shown to be highly expressed in many types of cancer cells, including PCa cells. Interestingly, in these cells, PKM2 has been reported to be also deeply involved in glucose metabolism (OXPHOS activity) and to mediate proliferation, metastatic behavior and acquisition of stem cell properties [167,168,169]. Mitochondrial pyruvate carrier (MPC) is the hetero-dimeric complex (formed by MPC1 and MPC2) responsible for the import of pyruvate into the mitochondria where it is converted to acetyl-CoA and then further enters the TCA cycle to fuel the OXPHOS machinery. MPC2 expression was found to correlate with tumor aggressiveness in PCa specimens [170,171]. Transcriptional enhanced associate domain 4 (TEAD4) is a transcription factor previously shown to be involved in the regulation of the expression of mitochondrial genes involved in the OXPHOS pathways [172]. TEAD4 is expressed in PCa cells, and its expression has been reported to be critical in increasing OXPHOS activity. In a recent paper, Chen and coworkers reported that TEAD4 expression is epigenetically regulated by the semi-essential amino acid arginine to modulate OXPHOS functions in hormone-refractory PCa cells [173].
Interestingly, elevated OXPHOS and mitochondrial mass have been observed in the aggressive stem cell subpopulation of different tumors, including PCa [174]. Sotgia and coworkers proposed the development of a “mitochondrial based oncology platform” for specifically targeting CSC metabolism [175,176]. In line with this observation, metformin has been proposed as an effective anticancer agent based on its ability to specifically target OXPHOS and ATP production in prostate CSCs [177]. On the other hand, impaired mitochondrial OXPHOS and upregulated glycolysis were observed in these cells [178]. Thus, the presence of exacerbated OXPHOS in PCa stem cells still remains a controversial issue.
Given the crucial role of the tumor-stroma cross-talk in shaping cancer cell metabolism, Ippolito et al. investigated how CAFs might regulate mitochondrial dynamics in PCa cells. They found that tumor-associated CAFs significantly enhance mitochondrial respiration, mediated by a lactate shuttle, and favor mitochondria transfer in PCa cells, thus promoting their malignant behavior [46]. In line with these results, Grupp et al. demonstrated that a high mitochondria content in PCa specimens correlates positively with PCa progression and represents an effective predictor of a poor clinical prognosis and outcome [132]. Last but not least, a switch from glycolysis to OXPHOS activity has been observed in PCa cells undergoing the development of resistance to standard therapies (i.e., enzalutamide, docetaxel) [179].
Based on these observations, it is now accepted that targeting both glycolysis and mitochondrial OXPHOS pathway might represent an effective therapeutic strategy for advanced, metastatic and drug-resistant PCa [180].
A schematic overview of the metabolic rewiring occurring in prostate epithelial cells during the different stages of cancer progression is given in Figure 1.

2.1.2. The AR-Mitochondria Axis

ADT still represents the therapy of choice for early-stage, androgen-dependent PCa. However, most patients progress towards the aggressive CRPC stage characterized by a high rate of cell proliferation, invasiveness and metastatic behavior; interestingly, in most CRPC patients (about 80%) a reactivation of the androgen receptor (AR) has been observed. The persistent activity of AR in this stage has been shown to involve different mechanisms, including AR gene amplification, AR mutations, AR gene alternative splicing, generating different receptor splice variants and intratumoral synthesis of androgens. Since PCa progression is also associated with a peculiar metabolic reprogramming, as discussed above, it has been postulated that AR might be a master regulator of PCa cell metabolism. In line with this hypothesis, genomic, transcriptomic and metabolomic functional studies pointed out that AR regulates different metabolic pathways, including glucose uptake (through the induction of different glucose transporters), glycolysis, TCA cycle, mitochondrial biogenesis and respiration, de novo lipid synthesis and fatty acid β-oxidation [143,170,181,182,183,184].
Different intracellular signaling pathways were reported to be involved in this AR-driven metabolic reprogramming. Importantly, AR activation was demonstrated to induce mTOR translocation into the nucleus where it binds to the promoter regions of metabolic genes (such as HK2), thereby regulating their expression; accordingly, inhibition of the mTOR pathway resulted in impaired glycolytic activities and reduced proliferation in PCa cells [183]. In AR-expressing PCa cells, it was shown that androgens promote the activity of the AMPK/PGC1α signaling cascade, leading to increased glycolytic rates, mitochondrial biogenesis, OXPHOS, intracellular ATP levels and cell growth [182].
An additional interesting mediator of the AR metabolic activity is MPC (specifically the MPC2 isoform), reported to be highly expressed in AR-positive PCa cells (both hormone-dependent and CRPC cells) but almost absent in AR-negative PCa cells and to play a pivotal role in supporting a functional TCA cycle. Bader and coworkers demonstrated that MPC is transcriptionally upregulated by AR in PCa cells, and its inhibition impairs O2 consumption, TCA cycle metabolite levels and oxidative phosphorylation, thus halting cell proliferation. Moreover, these authors could show that targeting MPC with the MPC inhibitors UK5099 and MSDC0160 (this inhibitor is orally administered and clinically viable) results in the suppression of the growth of AR-expressing, but not AR-negative, PCa cells in in vitro and in vivo studies [143,170,185]. In line with these results, it has been reported that, in androgen-sensitive and CRPC cells, activation of the AR signaling upregulates the expression of DRP1 (the mediator of the mitochondrial fission) to induce the formation of the VDAC/MPC2 complex and thereby the pyruvate transport into the mitochondria and sustains mitochondrial metabolic pathways, such as OXPHOS.
Ultimately, DRP1 overexpression was observed to positively correlate with cancer cell proliferation and survival in different conditions of metabolic stress, such as oxidative stress and hypoxia; moreover, high DRP1 expression levels in patients with CRPC were found to be suggestive of poor prognosis [171]. Interestingly, by means of in vitro and in vivo models, Xu et al. recently reported a loss of MPC expression during the progression of PCa cells towards the most aggressive and AR-negative neuroendocrine phenotype (NEPC); in these models, MPC loss induces the expression of PKM2 that, in turn, translocates into the nucleus to regulate the expression of EMT markers [186].
On the other hand, it has been shown that knockout of KDM4B, a transcriptional activator of AR, in CRPC cells reduces proliferation and triggers a metabolic switch towards OXPHOS [187]. Bajpai and coworkers found that, in experimental models of AR overexpression in AR-negative PCa cells, AR localizes to mitochondria where it decreases mitochondrial DNA (mtDNA) content and impairs OXPHOS activity [188].
Taken together, these data suggest that, even if most of the published data support that in the PCa models in which it is expressed and active (i.e., AR-driven hormone-sensitive and castration-resistant PCa) AR plays a pivotal role as a master regulator of the cancer cell metabolic reprogramming towards mitochondrial respiration, further studies are needed to definitely assess this issue.

2.1.3. mtDNA Mutations

So far, most studies addressing the relevance of genetic mutations in PCa development have been focused on the nuclear genome. Mitochondria, maternally inherited organelles, are deeply involved in the process of tumorigenesis by orchestrating metabolic and energy production pathways, ROS signaling and apoptosis [8,57,189]. Thus, dissecting the mitochondrial genome is currently considered an essential step to obtain a complete view of the genetic alteration profile in PCas.
The majority of the proteins of the four ETC complexes (COI-IV) involved in OXPHOS are encoded by nuclear DNA; however, 13 proteins in these complexes are encoded by mtDNA, the small circular DNA molecule found inside mitochondria. The mtDNA is characterized by a high mutation rate, mainly linked to high levels of ETC-derived ROS and a low efficient DNA repair system in these organelles [190]. Mutations of mtDNA have been found in different types of human cancers, including PCa [162,191,192,193,194,195], although their functional role still needs to be fully elucidated. Gomez-Zaera and coworkers analyzed the presence of mtDNA sequence variants in human PCa tissues; they reported that the most frequent variants were present in the following genes: mt-RNR2, encoding the large 16S mitochondrial ribosomal RNA (rRNA) subunit; mt-D-loop (displacement loop, control sites for the expression of the mitochondrial genome); and mt-ND4, encoding the protein NADH dehydrogenase 4, part of the COI of the ETC pathway [196].
The analysis of somatic mutations in tumor tissues from PCa patients pointed out their presence in genes coding for rRNA (mt-RNR1 and mt-RNR2), transfer RNA (tRNA) and the protein-coding gene mitochondrially encoded ATP synthase membrane subunit 6 (mt-ATP6), that encodes the ATP synthase Fo subunit 6 (or subunit/chain A). Moreover, somatic mutations in the entire mitochondrial genome were found to be associated with high PSA levels in PCa patients [197]. By using a yeast model organism, it was shown that the mutation mt-ATP6-P136S specifically found in PCa tissues positively correlates with tumor progression and may be involved in cancer cell escape from apoptosis [198]. Hopkins et al. reported that mutations in mitochondrial rRNA, tRNA as well as in the protein-coding genes mt-ATP6, mt-ND1, mt-ND2 and mitochondrially encoded cytochrome c oxidase I (mt-CO1), component of the complex IV, cytochrome c oxidase, the last enzyme in the mitochondrial electron transport chain which drives OXPHOS, are frequent in PCa tissues and are drivers of PCa aggressive behavior [195]. In line with these observations, mutations in genes encoding for proteins of the mitochondrial complex I (mt-NDs) were reported to be frequent in high grade PCa tissues and to be associated with a reduced activity of the NADH dehydrogenase pathway and an increased, compensatory, activity of the succinate-using FADH2 pathway [199]. Interestingly, Sun and coworkers demonstrated that the presence of a mutant mt-CO1 gene results in the resistance of PCa cells to the pro-death activity of simvastatin [200].
Taken together, these data support that in PCa cells undergoing stressful conditions, activation of the ROS signaling might have a deleterious effect on mtDNA driving mutations at the level of genes coding for proteins deeply involved in the metabolic rewiring.

2.2. Mitochondrial Dynamics

It is now well established that alterations of the mitochondrial structural dynamics (biogenesis, fusion, fission and mitophagy) are deeply involved in the different steps of cancer growth, progression and development of drug resistance [16,80,82,201,202]. However, current data on the role of the mitochondrial structural alterations in PCa are still scanty.
PGC1α is the well-recognized master regulator of mitochondrial biogenesis [203]; it is also involved in the control of the mitochondrial fusion/fission balance by promoting fusion, through the activation of MFN1 and 2, and impairing DRP1 expression, through the binding to its promoter region [204]. The expression of this gene, together with the mitochondrial number, was found to be upregulated in tumors, including PCa, of African American cancer patients known to be exposed to a higher risk of cancer and mortality compared to European American patients [205]. PGC1α has been observed to be highly expressed in PCa cells harboring either deletion or mutation of the classic tumor suppressor protein p53, and its expression positively correlates with cancer cell metastatic behavior [206]. It has been demonstrated that, in CRPC PC3 cells, overexpression of p53 decreases the expression and activity (i.e., nuclear localization) of PGC-1α, thereby leading to a reduced mitochondrial mass and a significant change in the expression levels of genes and proteins involved in the fusion/fission balance [207]. More recently, Galbraith and coworkers reported that, in PCa cells, the activity of PGC1α is also regulated by the PPARG/AKT3 axis. Specifically, these authors found that, in CRPC cells, overexpression of the transcription factor PPARG induces the expression of the AKT3 kinase that, in turn, triggers the nuclear localization of PGC1α, thereby driving mitochondrial biogenesis and ATP production which may fuel the metastatic behavior of tumor cells [166].
Burch et al. investigated the expression of mitochondrial biogenesis and bioenergetics genes in PCa cells and tissues. They reported that 47 genes involved in mitochondrial biogenesis, fusion/fission and energy metabolism are differentially expressed in malignant vs. non-malignant PCa cells. Specifically, the mitochondrial carrier uncoupling protein 2 (UCP2) gene, involved in energy homeostasis linking mitochondrial metabolism and redox (ROS detoxification), was observed to be overexpressed in malignant PCa cells as well as in clinical PCa specimens [208].
Alterations of the fusion/fission balance have also shown to be deeply involved in tumorigenesis, although the data so far available on this issue in PCa cells are still limited. Generally, it is accepted that mitochondria fission, the division of mitochondria in smaller organelles, is a typical feature of cells undergoing apoptosis; moreover, this process foresees the translocation of the cytoplasmic DRP1 protein to the mitochondria where it interacts with its receptor FIS1. In PCa cells, it has been reported that the overload of Ca2+ in mitochondria triggers the interaction of DRP1 with FIS1, thereby leading to mitochondrial fragmentation and enhanced cell response to pro-apoptotic agents [209]. In line with these observations, we observed that, in CRPC cells, mitochondrial Ca2+ and ROS overload triggers mitochondrial fission and mitophagy to mediate the pro-death (apoptotic, paraptotic) activities of natural anticancer compounds [78]. Moreover, enhanced mitochondrial fusion, together with mutations of the complex I mtDNA, were found to be associated with PCa progression, as evaluated in cancer cell lines as well as in mice and human tissue samples [210].
On the other hand, mitochondrial fission has been recently reported to play a key role in the maintenance of stemness features in prostate CSCs. Specifically, Civenni and coworkers focused their attention on bromodomain and extra-terminal domain (BET) proteins, such as bromodomain containing 4 (BRD4), well known as epigenetic modifiers of gene transcription. These authors showed that the DRP1 receptor, and fission factor, MFF is upregulated in hormone-refractory human prostate tumors as well as in prostate CSCs. Moreover, they could show that BRD4 acts as a key driver of MFF transcription and, therefore, of mitochondrial fission, which is an essential biological event for the survival and self-renewal of CSCs; accordingly, they observed that the inhibition of the BRD4 activity and of the subsequent MFF transcription results in the accumulation of dysfunctional mitochondria and, consequently, in the acquisition of the senescent phenotype in these cells. Thus, mitochondrial fission is a crucial process for the maintenance of the self-renewal and tumorigenic potential of the CSC subpopulation in prostate tumors. The authors conclude that pharmacological inhibition of BRD4 may represent a promising effective therapeutic strategy for PCa patients [211,212].
Mitophagy is the intracellular removal of mitochondria by autophagy. It has been shown both to promote cancer cell survival and drug resistance through the degradation of damaged mitochondria and to prevent tumor progression mediating the pro-death activity of different anticancer drugs. Thus, its specific role in regulating the balance between cell survival vs. cell death is still a matter of debate in most tumor types, including PCa [105,106,213,214,215,216]. Han and coworkers reported that pharmacological targeting of the androgen/AR axis promotes apoptosis of PCa cells by triggering the mitophagic pathway [217]. It is well known that the oncogenic epidermal growth factor receptor (EGFR) is overexpressed in different types of tumors to exert its peculiar pro-survival effects. It has been shown that, in PCa cells, downregulation of EGFR expression, but not its tyrosine kinase activity, results in cell death mediated by mitophagy through the activation of the AKT/mTOR [218]. In line with these observations, it has been reported that elevated levels of the mitophagy inhibitor leucine-rich pentatricopeptide repeat motif-containing protein (LRPPRC) positively correlate with poor prognosis and shorter survival in PCa patients, supporting a protective effect of mitophagy against PCa growth [219]. On the other hand, Balvan et al. demonstrated that, in CRPC PC3 cells, activation of mitophagy (degradation of mitochondria upon oxidative stress) represents a crucial mechanism of protection and resistance against pro-oxidant conditions [220].
In summary, mounting evidence supports that mitochondrial dynamics are involved in prostate carcinogenesis; however, further research is needed to definitely assess their role in the growth and development of PCa.

3. Targeting Mitochondrial Reprogramming and Dynamics in PCa

The data discussed so far support that mitochondrial metabolic reprogramming and structural dynamics represent promising molecular targets for novel and effective therapeutic strategies for PCa patients. Actually, several “mitochondrial metabolism-interfering agents” are currently being investigated for their anticancer activity in different tumors, including PCa (Table 1).
Docetaxel in combination with ADT therapy as a neoadjuvant intervention before radical prostatectomy in patients with localized, high-risk PCa is now well recognized as an effective strategy to increase overall and progression-free survival. To dissect the molecular mechanisms underlying the beneficial effects of this therapeutic intervention, Qu and coworkers recently investigated the effects of neoadjuvant docetaxel in combination with a GnRH analog on different metabolic pathways in cancer tissues. They reported that the neoadjuvant therapy induces a significant downregulation of metabolic pathways, such as the TCA cycle and lipid synthesis; a decrease of the GSH/GSSG ratio, supporting a reduced oxidative stress condition, was also observed. Glutamine was downregulated in the treatment group, supporting that the energy supply was significantly reduced in these patients as the consequence of a reduced glutamate production and impairment of the TCA cycle [221].
Metformin, an FDA-approved biguanide hypoglycemic compound, is the first-line drug for the treatment of type 2 diabetes [222,223]. It is administered orally and is well tolerated, being associated with a low incidence of side effects [224]. Recently, several studies have pointed out that metformin exerts a significant antitumor activity in different types of cancer cells, including PCa cells, both in vitro and in preclinical xenograft models [225,226,227,228,229]. In line with these data, epidemiologic studies evidenced a decrease in the incidence of cancer in metformin-treated diabetic patients [230,231,232]. In PCa cells, this compound has been shown to decrease cell viability, induce cell cycle arrest and promote apoptosis through modulation of different intracellular signaling pathways, including glucose metabolism [233,234,235,236]. Specifically, metformin was reported to interfere with the IGF-1 (insulin-like growth factor-1), the VEGF (vascular-endothelial growth factor) and the androgen receptor signaling pathways [236,237,238,239,240,241] as well as increase ROS production, causing a rapid imbalance in cellular redox homeostasis and causing cell death [242]. Focusing on cell glucose metabolism, it was found to induce the activation (i.e., phosphorylation) of the energy sensor AMPK thus inhibiting downstream mTOR activity, increasing glucose uptake, lactate production, glycolytic rate and, more importantly, inhibiting O2 consumption and the complex I of the mitochondrial ETC, thereby impairing the OXPHOS pathway [229,236,241,243,244,245]. Thus, in PCa cells, this drug decreases ATP levels, triggering energy deficiency and reduced lipogenesis [246]. Metformin was also found to target the viability of CSCs deeply involved in drug resistance and cancer relapse through the inhibition of mitochondrial bioenergetics and the subsequent compensative increase of the glycolytic pathway, and by triggering mitochondrial ROS production ultimately leading to apoptosis [177,243,247,248]. In line with these data, Ippolito and coworkers demonstrated that CRPC cells made resistant to docetaxel acquire an invasive phenotype associated both with a decreased glycolytic rate and with an overactivation of oxidative phosphorylation; by targeting mitochondrial complex I, metformin suppresses the proliferative and invasive behavior of drug-resistant cells [31]. Moreover, by addressing the issue of the stroma/tumor interplay in PCa, Giannoni et al. observed that CAFs induce an aggressive phenotype in PCa cells by triggering PKM2 nuclear translocation and consequent metabolic reprogramming towards oxidative phosphorylation; pharmacological targeting of PKM2 (by DASA-58) and OXPHOS activity (by metformin) significantly counteracts CAF-induced PCa cell growth in vitro and in vivo [167]. Similar observations were reported with other antidiabetic drugs, such as the metformin analog phenformin and the SGTL2 (sodium-glucose transporter 2) inhibitor canagliflozin [52,249]. The antitumor activity of metformin and phenformin in PCa cells was also reported to be potentiated by co-treating the cells with drugs shown to impair different aspects of cell metabolism, such as 2-deoxyglucose, an inhibitor of HK2 activity and inducer of intracellular ATP depletion [250]; gossypol, an inhibitor of ALDH (cytosolic aldehyde dehydrogenase) and, therefore, of cytosolic NADH production, which selectively reduces electron supply for ATP production [52]; and salicylate, the aspirin metabolite known to activate the AMPK pathway [251]. Recent clinical studies investigated the activity of metformin, either alone or in combination with other anticancer compounds, in PCa patients. Metformin was reported to be associated with improved disease-specific survival in PCa patients, with treatment starting at tumor diagnosis or after prostatectomy [252,253,254]; moreover, it was shown to improve the outcome in PCa patients co-treated with ADT [255,256]. However, contrasting results were also reported. Cui and coworkers, by performing a systematic literature search and meta-analysis, reported the lack of association between metformin and the risk of PCa in diabetic patients [257]. Moreover, this drug also failed to improve the efficacy of standard treatment regimens, such as bicalutamide and docetaxel, in overweight and in metastatic castration-resistant PCa patients, respectively [258,259]. Several clinical trials aimed to definitely dissect the antitumor activity of metformin and its analog phenformin, either alone or in combination with common treatments, are currently being conducted in PCa patients (https://www.clinicaltrials.gov/) (accessed on 21 December 2022).
Many other compounds were reported to counteract PCa growth by targeting the mitochondrial OXPHOS pathway through the inhibition of the ETC complexes. In CRPC DU145 cells, the class III antiarrhythmic drug amiodarone, currently available in the clinical setting, was found to impair the activity of complex I and II, thereby suppressing ATP production and potentiating the antitumor activity of standard therapies (docetaxel and cisplatin) [260]. PCa cells made resistant to the AR antagonist enzalutamide (ENZA) switch from glycolysis to OXPHOS. Basu et al. demonstrated that the complex I inhibitor IACS-010759 exerts a significant antiproliferative activity in ENZA-resistant PCa cells, when compared to non-resistant cells; similar results were observed with the mitochondrial glutaminase inhibitor CB-839, which acts by decreasing the glutamate supply to the TCA cycle [179].
Mitochondrial ATP synthase, or complex V, is the fifth OXPHOS complex, responsible for the synthesis of ATP. High levels of ATP synthase were observed in different tumors, including PCa. In cancer cells, ATP synthase has also been shown to translocate from the mitochondria to the cell membrane where it contributes to many processes, such as tumorigenesis, angiogenesis and metastasis. Several ATP synthase synthetic inhibitors were reported to suppress cancer growth and are currently undergoing clinical investigation [261]. Specifically, Elbehairi et al. reported that, in PCa cells, the Pd(II) complex of Gboxin analog-chitooligosaccharides conjugate (Pd(II)COS@GbA) effectively impairs ATP synthase expression and activity, leading to the suppression of ATP production, and triggers mitochondrial fragmentation (fission) by increasing the expression levels of the fission protein DRP1 while decreasing those of the fusion protein OPA1 [262].
It is now accepted that some antibiotics can induce death in cancer cells by impairing mitochondrial functions and, based on these properties, are being used as anticancer drugs [263]. According to the endosymbiotic theory, mitochondria are organelles that derive from prokaryotic aerobic cells that were engulfed by eukaryotic cells during their evolution process. In line with this theory, mitochondria contain a DNA different from that of the nucleus and their own protein biosynthesis machinery. Lisanti and coworkers demonstrated that in different types of cancer cells, including PCa cells, and specifically in their stem cell counterpart, some antibiotics (i.e., azithromycin, doxycycline, tigecycline, pyrvinium pamoate, chloramphenicol) impair mitochondrial respiration by binding to mitoribosomes thus inhibiting protein synthesis and by impairing mitochondrial biogenesis, OXPHOS and ATP production [264,265,266,267]. These authors proposed the efficacy of the mitochondrial-based oncology platform (MITO-ONC-RX), a panel of FDA-approved antibiotics, to reduce mitochondrial mass and OXPHOS for the eradication of CSCs in different tumors, including PCa [176]. In line with these observations, the antimicrobial peptide Buforin IIb, in combination with 2-deoxyglucose, displays a synergistic toxic effect on CRPC cells, in vitro and in mouse xenograft tumors. Mechanistically, the anticancer effects of the combination treatment were mediated by a decrease of lactate production (i.e., inhibition of glycolysis) and intracellular ATP levels [268]. Further studies, focusing on the metabolic reprogramming-mediated anticancer activity of antibiotics, either alone or in combination with standard therapies, in PCa patients are warranted.
Different natural compounds were also reported to be endowed with significant anticancer activity in PCa, based on their antioxidant effects but also mediated by a rewiring of mitochondrial functional and structural dynamics.
Tocotrienols (TTs; α-, β-, δ- and γ-TT) are hydrophobic compounds derived from vitamin E and present in some plant sources, such as annatto (Bixa orellana) seeds, rice bran and palm oil [269,270]. These compounds have been widely reported to be endowed with different beneficial health activities in the prevention or treatment of chronic diseases, including osteoporosis, cardiovascular and neurodegenerative diseases [271,272,273,274,275,276,277]. Specifically, over the last couple of decades, TTs have also emerged as potential anticancer agents, based on their antiproliferative, proapoptotic, antimetastatic and antiangiogenic activities in several types of cancer cells, including PCa cells [278,279,280,281,282,283,284,285,286,287]. We recently reported that, in CRPC cells (DU145 and PC3), δ-TT triggers apoptosis, involving ER stress and autophagy, as well as paraptosis, a non-canonical cell death mechanism. [288]. Mechanistically, we could demonstrate that this compound inhibits glucose uptake and lactate production in PTEN-deficient LNCaP and PC3 PCa cells by specifically decreasing HK2 expression, and that it synergizes with metformin in inducing cell death. δ-TT also impairs mitochondrial respiration through the downregulation of the expression of OXPOHS complexes (complex I, II and IV), O2 consumption and ATP production. These energy-depleting effects were associated with the induction of mitochondrial fission, which is associated with the Ca2+- and ROS-mediated mitophagic pathway. Thus, in CRPC cells, δ-TT exerts a significant anticancer activity by impairing the mitochondrial functional and structural dynamics and dysregulating mitochondrial Ca2+-ROS homeostasis [78,289].
Table 1. Anticancer drugs targeting mitochondrial functional rewiring and structural dynamics in PCa cells.
Table 1. Anticancer drugs targeting mitochondrial functional rewiring and structural dynamics in PCa cells.
DrugTherapy ClassEffectsReferences
Docetaxel + ADTTaxane drugs + GnRH analogsInhibition of TCA cycle;
decrease of the GSH/GSSG ratio;
downregulation of glutamine
[221]
Metformin, phenformin
canagliflozin
Antidiabetic (type 2 diabetes)
drugs
Inhibition of ETC complex I;
decreased O2 consumption;
AMPK activation;
suppression of the OXPHOS pathway
and ATP production;
upregulation of ROS levels
[31,52,167,177,229,236,242,243,244,245,246,247,248,249,250,251]
AmiodaroneClass III antiarrythmic drugsInhibition of ETC complex I and II;
suppression of ATP production
[260]
IACS-010759, CB-839Small molecule inhibitorsInhibition of ETC complex I;
inhibition of mitochondrial glutaminase
and glutamate supply to the TCA cycle;
decreased OXPHOS pathway
[179]
Pd(II)COS@GbAPd(II) anticancer complexesInhibition of ETC complex V (ATP synthase);
suppression of ATP production;
mitochondrial fission
[262]
Doxycycline, azithromycin,
tigecycline, pyrvinium
pamoate, chloramphenicol
AntibioticsInhibition of the OXPHOS pathway
and mitochondrial respiration;
dysregulations of ETC complexes;
suppression of ATP production;
impairment of mitochondrial biogenesis
[176,264,265,266,267]
Buforin IIb Antimicrobial peptidesInhibition of glycolysis and ATP production[268]
Vitamin E-derived
tocotrienols, curcumin, resveratrol, triterpenoids,
sulforaphane, phenethyl isothiocyanate (PEITC), silibinin, atpenin A5
analogs, jasmonates,
alternol
Natural antioxidantsReduced O2 consumption, ETC protein (complexes COI-V) levels and activity;
suppression of the OXPHOS pathway
and ATP production;
decreased glycolytic pathway;
AMPK activation;
altered Ca2+/ROS homeostasis;
mitochondrial fission, mitophagy
[78,180,261,289,290,291,292,293,294,295,296,297,298,299,300,301,302,303,304,305,306]
Curcumin, also called diferuloylmethane, is the main natural curcuminoid present in the Indian spice turmeric. It has been shown to exert a growth-suppressive activity, which is mainly based on its ability to induce oxidative stress in different types of cancers, including PCa [307,308,309]. Recent articles highlighted the role of mitochondrial reprogramming as a novel molecular mechanism underlying the antitumor activity of this polyphenol. Specifically, Ossikbayeva and coworkers recently reported that curcumin, either alone or in association with carnosic acid, inhibits cell proliferation in CRPC cells by inducing cellcycle arrest. These anticancer effects were associated with a dysfunction of mitochondrial respiration, as evidenced by a decrease of O2 consumption and of the activities of all the complexes (COI-IV) of the ETC, and were counteracted by cotreating the cells with the mTOR inhibitor rapamycin [290]. In line with these data, curcumin was recently shown to impair mitochondrial phosphorylation in PCa cells by specifically targeting mitochondrial ATP synthase, thus suppressing ATP production [261,291].
Resveratrol, a polyphenolic compound found in different vegetables such as grape, berry and peanut fruits, has been shown to have anti-inflammatory, antioxidant, immunomodulatory and antitumor activities [310,311,312,313]. In cancer cells, this polyphenol was widely shown to induce apoptosis by promoting ROS overproduction [292]. By dissecting the biochemical mechanisms underlying the resveratrol anticancer activity, Rodriguez-Enriquez et al. reported that, in cervix cancer HeLa cells, resveratrol inhibits cell proliferation by significantly decreasing both glycolysis and OXPHOS while triggering ROS production and mitophagy [293]. However, contrasting results were reported in PCa cells; specifically, it was observed in CRPC PC3 cells that resveratrol significantly impairs cell growth and that this is coincident with increased mitochondrial biogenesis, fusion and respiration [314]. Thus, further studies are needed to definitely highlight the involvement of mitochondrial metabolic reprogramming and dynamics in the anticancer activity of resveratrol in PCa.
Triterpenoids are plant-derived isopentenyl pyrophosphate oligomers; so far, more than 20,000 triterpenoids have been identified [315]. These compounds have been widely reported to have pharmacological properties, including anti-inflammatory, antioxidant, antipyretic and cardioprotective activities; moreover, several studies revealed their ability to induce cell cycle arrest, cell cytotoxicity/apoptosis, anti-invasion and autophagy [316,317]. In line with these observations, in vitro and in vivo studies pointed out that a great number of triterpenoids exhibit a potent antitumor activity against various types of cancer cells, including PCa cells, through the regulation of different molecular pathways [318,319,320,321,322,323,324]. Among the various molecular mechanisms, they were reported to exert their anticancer activity by impacting tumor metabolism [325,326,327]. Specifically, in PCa cells, triterpenoids were shown to reduce glucose uptake, aerobic glycolysis, O2 consumption and intracellular ATP levels and to induce a sustained activation of AMPK and consequent suppression of mTOR signaling [180,294,295,296,297,298].
1,4-Naphthoquinones represent the most important class of quinones found to be endowed with anti-inflammatory, antiallergic, antibacterial, radical scavenging and, importantly, anticancer activity [328]. Dyshlovoy and coworkers recently reported that glucose-conjugated 1,4-naphthoquinones significantly impair CRPC PC3 cell proliferation through the inhibition of pro-survival and drug resistance mechanisms, such as AR signaling and autophagy. These compounds also inhibit glucose uptake, disrupt mitochondrial OXPHOS by suppressing the expression of components of the five complexes of the ETC (COI-COV) and trigger a mitochondrial ROS overload [299].
Sulforaphane is a natural plant isothiocyanate found in many cruciferous vegetables like broccoli, cabbage, cauliflower and kale [329]. In CRPC cells, sulforaphane has been reported to induce mitochondrial apoptosis mediated by an increased expression of the Bax protein. This compound also impairs mitochondrial structural dynamics by inducing mitochondrial biogenesis and fragmentation through the stabilization of the transcription factor Nrf2 (nuclear factor E2-related factor) and the increase of PGC1α expression levels [300]. In line with these data, it was shown that the natural compound PEITC (phenethyl isothiocyanate), another isothiocyanate found in cruciferous vegetables, is a specific inhibitor of complex III that acts by impairing oxidative phosphorylation and ATP synthesis, leading to ROS overproduction and cell death, in PCa cells [301].
Silibinin, also known as silybin (both from Silybum, the name of the plant from which it is extracted), is the major active constituent of silymarin, a standardized extract of the milk thistle seeds. It has been shown to induce apoptosis in different cancer cells, both in vitro and in vivo [330,331]. In PC3 cells, silibinin was observed to induce apoptosis by triggering mitochondrial ROS generation and disruption of Ca2+ homeostasis, leading to ER stress response [302].
Other natural compounds reported to induce PCa cell death by impairing mitochondrial functional plasticity and dynamics include atpenin A5 analogs (specific inhibitors of complex II activity) [303], jasmonates (inducers of mitochondrial ATP depletion) [304], alternol (inhibitor of TCA cycle, mitochondrial respiration and ATP production) [305] and deguelin (COI inhibitor) [306]. Some of these compounds are presently under investigation in clinical trials aimed to assess their bioavailability, lack of side effects and chemopreventive activity as well as their ability to prevent the progression to metastatic disease in PCa patients (https://www.clinicaltrials.gov/; accessed on 21 December 2022).

4. Conclusions

Mitochondria are known to play a pivotal role in crucial biological processes, such as cell metabolism, cell proliferation/death and cell signaling pathways as well as Ca2+ and redox homeostasis. In addition, it is now well accepted that these organelles also undergo a peculiar functional and structural rewiring during the process of tumorigenesis to support cancer growth and progression; however, this plasticity seems to occur specifically in the different types of tumor cells and cell contexts.
Prostate epithelial cells are characterized by a distinctive metabolic reprogramming during the different phases of their transformation from healthy cells to early-stage and, sequentially, to late-stage tumor cells. Specifically, healthy cells accumulate high levels of zinc due to elevated levels of its ZIP1 transporter at the plasma membrane. High intramitochondrial zinc levels inhibit the activity of m-aconitase, the enzyme responsible for the conversion of citrate into isocitrate in the TCA cycle, thus leading to an impairment of its oxidation and to its accumulation and secretion out of the cells. Consequently, healthy epithelial cells are characterized by a truncated TCA cycle and an impaired OXPHOS pathway, which is compensated by an increased glycolytic rate. Moreover, in these cells high intracellular zinc levels were also found to correlate with the mitochondria-mediated intrinsic apoptotic pathway. On the other hand, in early-stage prostate cancer cells the expression of ZIP1 decreases significantly, leading to low intramitochondrial zinc levels. As a consequence, early-stage PCa cells display a reactivation of m-aconitase and citrate oxidation as well as the TCA cycle and OXPHOS pathways, to meet their high demand for energy and building blocks for the synthesis of biomolecules. The low intracellular levels of zinc also allow these cells to avoid apoptosis, thus sustaining cancer cell proliferation. Interestingly, PCa cells undergo an additional mitochondrial metabolic rewiring during their progression toward the late stage of tumorigenesis. The Warburg effect was initially proposed as the main metabolic feature of aggressive/metastatic PCa cells. However, accumulating evidence strongly supports that high levels of the TCA cycle as well as of OXPHOS activity and ATP production are still present in late-stage as well as in drug-resistant PCa cells. Interestingly, it has been shown that both the presence of an active AR axis and mutations in mitochondrial genes are deeply involved in these mechanisms.
As reported for different types of tumors, distinctive mitochondrial structural dynamics (biogenesis, fusion/fission, mitophagy) have also been observed in PCa cells and specifically in late-stage/CRPC cells. Based on the data discussed here, it has become widely accepted during the last years that mitochondrial metabolic rewiring and dynamics might represent novel molecular markers of PCa growth and progression, as well as interesting targets for novel anticancer therapeutic strategies. Accordingly, it was demonstrated that, in PCa cells, a wide range of drugs, including both synthetic and natural compounds, exert a significant anticancer activity by specifically targeting both the rewired mitochondrial metabolic pathways (O2 consumption, TCA cycle, OXPHOS and ATP production) and the altered mitochondrial dynamics. However, most of these results are still derived from in vitro and preclinical studies; thus, clinical trials are urgently required to definitely assess the effectiveness of these compounds in improving treatment options for PCa patients.

Author Contributions

Conceptualization, P.L.; investigation, P.L.; writing—original draft preparation, P.L.; writing—review and editing, P.L. and F.F.; visualization, F.F., M.A. and P.L.; supervision, P.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by MIUR Progetto di Eccellenza (Department of Pharmacological and Biomolecular Sciences, University of Milan). F.F. was supported by an AIRC fellowship for Italy.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Warburg, O.; Wind, F.; Negelein, E. The metabolism of tumors in the body. J. Gen. Physiol. 1927, 8, 519–530. [Google Scholar] [CrossRef]
  2. Warburg, O. On the Origin of Cancer Cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef]
  3. Koppenol, W.H.; Bounds, P.L.; Dang, C.V. Otto Warburg’s contributions to current concepts of cancer metabolism. Nat. Rev. Cancer 2011, 11, 325–337. [Google Scholar] [CrossRef]
  4. Locasale, J.W.; Grassian, A.R.; Melman, T.; Lyssiotis, C.A.; Mattaini, K.R.; Bass, A.J.; Heffron, G.; Metallo, C.M.; Muranen, T.; Sharfi, H.; et al. Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to oncogenesis. Nat. Genet. 2011, 43, 869–874. [Google Scholar] [CrossRef]
  5. Granchi, C.; Fancelli, D.; Minutolo, F. An update on therapeutic opportunities offered by cancer glycolytic metabolism. Bioorg. Med. Chem. Lett. 2014, 24, 4915–4925. [Google Scholar] [CrossRef] [PubMed]
  6. Lu, J. The Warburg metabolism fuels tumor metastasis. Cancer Metastasis Rev. 2019, 38, 157–164. [Google Scholar] [CrossRef]
  7. Vaupel, P.; Multhoff, G. Revisiting the Warburg effect: Historical dogma versus current understanding. J. Physiol. 2020, 599, 1745–1757. [Google Scholar] [CrossRef]
  8. Fontana, F.; Limonta, P. The multifaceted roles of mitochondria at the crossroads of cell life and death in cancer. Free. Radic. Biol. Med. 2021, 176, 203–221. [Google Scholar] [CrossRef]
  9. Niu, D.; Wu, Y.; Lei, Z.; Zhang, M.; Xie, Z.; Tang, S. Lactic acid, a driver of tumor-stroma interactions. Int. Immunopharmacol. 2022, 106, 108597. [Google Scholar] [CrossRef]
  10. Wang, X.; Liu, H.; Ni, Y.; Shen, P.; Han, X. Lactate shuttle: From substance exchange to regulatory mechanism. Hum. Cell 2022, 35, 1–14. [Google Scholar] [CrossRef] [PubMed]
  11. Morandi, A.; Taddei, M.L.; Chiarugi, P.; Giannoni, E. Targeting the Metabolic Reprogramming That Controls Epithelial-to-Mesenchymal Transition in Aggressive Tumors. Front. Oncol. 2017, 7, 40. [Google Scholar] [CrossRef]
  12. Porporato, P.E.; Filigheddu, N.; Pedro, J.M.B.-S.; Kroemer, G.; Galluzzi, L. Mitochondrial metabolism and cancer. Cell Res. 2018, 28, 265–280. [Google Scholar] [CrossRef] [PubMed]
  13. Jia, D.; Lu, M.; Jung, K.H.; Park, J.H.; Yu, L.; Onuchic, J.N.; Kaipparettu, B.A.; Levine, H. Elucidating cancer metabolic plasticity by coupling gene regulation with metabolic pathways. Proc. Natl. Acad. Sci. USA 2019, 116, 3909–3918. [Google Scholar] [CrossRef] [PubMed]
  14. Cassim, S.; Vučetić, M.; Ždralević, M.; Pouyssegur, J. Warburg and Beyond: The Power of Mitochondrial Metabolism to Collaborate or Replace Fermentative Glycolysis in Cancer. Cancers 2020, 12, 1119. [Google Scholar] [CrossRef]
  15. DeBerardinis, R.J.; Chandel, N.S. We need to talk about the Warburg effect. Nat. Metab. 2020, 2, 127–129. [Google Scholar] [CrossRef] [PubMed]
  16. Grasso, D.; Zampieri, L.X.; Capelôa, T.; Van De Velde, J.A.; Sonveaux, P. Mitochondria in cancer. Cell Stress 2020, 4, 114–146. [Google Scholar] [CrossRef] [PubMed]
  17. Roth, K.G.; Mambetsariev, I.; Kulkarni, P.; Salgia, R. The Mitochondrion as an Emerging Therapeutic Target in Cancer. Trends Mol. Med. 2020, 26, 119–134. [Google Scholar] [CrossRef]
  18. Mosier, J.A.; Schwager, S.C.; Boyajian, D.A.; Reinhart-King, C.A. Cancer cell metabolic plasticity in migration and metastasis. Clin. Exp. Metastasis 2021, 38, 343–359. [Google Scholar] [CrossRef] [PubMed]
  19. Tan, Y.Q.; Zhang, X.; Zhang, S.; Zhu, T.; Garg, M.; Lobie, P.E.; Pandey, V. Mitochondria: The metabolic switch of cellular oncogenic transformation. Biochim. Biophys. Acta (BBA)—Rev. Cancer 2021, 1876, 188534. [Google Scholar] [CrossRef]
  20. Zhang, X.; Su, Q.; Zhou, J.; Yang, Z.; Liu, Z.; Ji, L.; Gao, H.; Jiang, G. To betray or to fight? The dual identity of the mitochondria in cancer. Futur. Oncol. 2021, 17, 723–743. [Google Scholar] [CrossRef]
  21. Ward, P.S.; Thompson, C.B. Metabolic Reprogramming: A Cancer Hallmark Even Warburg Did Not Anticipate. Cancer Cell 2012, 21, 297–308. [Google Scholar] [CrossRef] [PubMed]
  22. Yoshida, G.J. Metabolic reprogramming: The emerging concept and associated therapeutic strategies. J. Exp. Clin. Cancer Res. 2015, 34, 111. [Google Scholar] [CrossRef]
  23. Scalise, M.; Pochini, L.; Galluccio, M.; Console, L.; Indiveri, C. Glutamine Transport and Mitochondrial Metabolism in Cancer Cell Growth. Front. Oncol. 2017, 7, 306. [Google Scholar] [CrossRef] [PubMed]
  24. Herst, P.M.; Grasso, C.; Berridge, M.V. Metabolic reprogramming of mitochondrial respiration in metastatic cancer. Cancer Metastasis Rev. 2018, 37, 643–653. [Google Scholar] [CrossRef] [PubMed]
  25. Bacci, M.; Lorito, N.; Ippolito, L.; Ramazzotti, M.; Luti, S.; Romagnoli, S.; Parri, M.; Bianchini, F.; Cappellesso, F.; Virga, F.; et al. Reprogramming of Amino Acid Transporters to Support Aspartate and Glutamate Dependency Sustains Endocrine Resistance in Breast Cancer. Cell Rep. 2019, 28, 104–118 e108. [Google Scholar] [CrossRef] [PubMed]
  26. Lee, A.; Lau, P.; Kwan, Y.; Kong, S. Mitochondrial Fuel Dependence on Glutamine Drives Chemo-Resistance in the Cancer Stem Cells of Hepatocellular Carcinoma. Int. J. Mol. Sci. 2021, 22, 3315. [Google Scholar] [CrossRef] [PubMed]
  27. Faubert, B.; Solmonson, A.; DeBerardinis, R.J. Metabolic reprogramming and cancer progression. Science 2020, 368, eaaw5473. [Google Scholar] [CrossRef]
  28. Icard, P.; Coquerel, A.; Wu, Z.; Gligorov, J.; Fuks, D.; Fournel, L.; Lincet, H.; Simula, L. Understanding the Central Role of Citrate in the Metabolism of Cancer Cells and Tumors: An Update. Int. J. Mol. Sci. 2021, 22, 6587. [Google Scholar] [CrossRef]
  29. Mortezaee, K. Redox tolerance and metabolic reprogramming in solid tumors. Cell Biol. Int. 2021, 45, 273–286. [Google Scholar] [CrossRef]
  30. Oliveira, G.L.; Coelho, A.R.; Marques, R.; Oliveira, P.J. Cancer cell metabolism: Rewiring the mitochondrial hub. Biochim. et Biophys. Acta (BBA)—Mol. Basis Dis. 2020, 1867, 166016. [Google Scholar] [CrossRef]
  31. Ippolito, L.; Marini, A.; Cavallini, L.; Morandi, A.; Pietrovito, L.; Pintus, G.; Giannoni, E.; Schrader, T.; Puhr, M.; Chiarugi, P.; et al. Metabolic shift toward oxidative phosphorylation in docetaxel resistant prostate cancer cells. Oncotarget 2016, 7, 61890–61904. [Google Scholar] [CrossRef] [PubMed]
  32. Pan, Y.; Cao, M.; Liu, J.; Yang, Q.; Miao, X.; Go, V.L.W.; Lee, P.W.N.; Xiao, G.G. Metabolic Regulation in Mitochondria and Drug Resistance. Adv. Exp. Med. Biol. 2017, 1038, 149–171. [Google Scholar] [CrossRef] [PubMed]
  33. Cocetta, V.; Ragazzi, E.; Montopoli, M. Mitochondrial Involvement in Cisplatin Resistance. Int. J. Mol. Sci. 2019, 20, 3384. [Google Scholar] [CrossRef] [PubMed]
  34. Salunkhe, S.; Mishra, S.V.; Ghorai, A.; Hole, A.; Chandrani, P.; Dutt, A.; Chilakapati, M.; Dutt, S. Metabolic rewiring in drug resistant cells exhibit higher OXPHOS and fatty acids as preferred major source to cellular energetics. Biochim. Biophys. Acta (BBA)—Bioenerg. 2020, 1861, 148300. [Google Scholar] [CrossRef]
  35. De Francesco, E.M.; Sotgia, F.; Lisanti, M.P. Cancer stem cells (CSCs): Metabolic strategies for their identification and eradication. Biochem. J. 2018, 475, 1611–1634. [Google Scholar] [CrossRef]
  36. Fiorillo, M.; Sotgia, F.; Lisanti, M.P. “Energetic” Cancer Stem Cells (e-CSCs): A New Hyper-Metabolic and Proliferative Tumor Cell Phenotype, Driven by Mitochondrial Energy. Front. Oncol. 2018, 8, 677. [Google Scholar] [CrossRef] [PubMed]
  37. Bonekamp, N.A.; Peter, B.; Hillen, H.S.; Felser, A.; Bergbrede, T.; Choidas, A.; Horn, M.; Unger, A.; Di Lucrezia, R.; Atanassov, I.; et al. Small-molecule inhibitors of human mitochondrial DNA transcription. Nature 2020, 588, 712–716. [Google Scholar] [CrossRef]
  38. Marzagalli, M.; Raimondi, M.; Fontana, F.; Marelli, M.M.; Moretti, R.M.; Limonta, P. Cellular and molecular biology of cancer stem cells in melanoma: Possible therapeutic implications. Semin. Cancer Biol. 2019, 59, 221–235. [Google Scholar] [CrossRef]
  39. Sotgia, F.; Fiorillo, M.; Lisanti, M.P. Hallmarks of the cancer cell of origin: Comparisons with “energetic” cancer stem cells (e-CSCs). Aging 2019, 11, 1065–1068. [Google Scholar] [CrossRef]
  40. García-Heredia, J.M.; Carnero, A. Role of Mitochondria in Cancer Stem Cell Resistance. Cells 2020, 9, 1693. [Google Scholar] [CrossRef] [PubMed]
  41. Liu, G.; Luo, Q.; Li, H.; Liu, Q.; Ju, Y.; Song, G. Increased Oxidative Phosphorylation is Required for Stemness Maintenance in Liver Cancer Stem Cells from Hepatocellular Carcinoma Cell Line HCCLM3 Cells. Int. J. Mol. Sci. 2020, 21, 5276. [Google Scholar] [CrossRef] [PubMed]
  42. Kaur, J.; Bhattacharyya, S. Cancer Stem Cells: Metabolic Characterization for Targeted Cancer Therapy. Front. Oncol. 2021, 11, 756888. [Google Scholar] [CrossRef] [PubMed]
  43. Marzagalli, M.; Fontana, F.; Raimondi, M.; Limonta, P. Cancer Stem Cells—Key Players in Tumor Relapse. Cancers 2021, 13, 376. [Google Scholar] [CrossRef] [PubMed]
  44. Zanotelli, M.R.; Goldblatt, Z.E.; Miller, J.P.; Bordeleau, F.; Li, J.; VanderBurgh, J.A.; Lampi, M.C.; King, M.R.; Reinhart-King, C.A. Regulation of ATP utilization during metastatic cell migration by collagen architecture. Mol. Biol. Cell 2018, 29, 1–9. [Google Scholar] [CrossRef] [PubMed]
  45. Chiarugi, P.; Cirri, P. Metabolic exchanges within tumor microenvironment. Cancer Lett. 2016, 380, 272–280. [Google Scholar] [CrossRef] [PubMed]
  46. Ippolito, L.; Morandi, A.; Taddei, M.L.; Parri, M.; Comito, G.; Iscaro, A.; Raspollini, M.R.; Magherini, F.; Rapizzi, E.; Masquelier, J.; et al. Cancer-associated fibroblasts promote prostate cancer malignancy via metabolic rewiring and mitochondrial transfer. Oncogene 2019, 38, 5339–5355. [Google Scholar] [CrossRef] [PubMed]
  47. Bacci, M.; Ippolito, L.; Magnelli, L.; Giannoni, E.; Chiarugi, P. Stromal-induced mitochondrial re-education: Impact on epithelial-to-mesenchymal transition and cancer aggressiveness. Semin. Cell Dev. Biol. 2020, 98, 71–79. [Google Scholar] [CrossRef]
  48. Li, Z.; Sun, C.; Qin, Z. Metabolic reprogramming of cancer-associated fibroblasts and its effect on cancer cell reprogramming. Theranostics 2021, 11, 8322–8336. [Google Scholar] [CrossRef]
  49. Modica-Napolitano, J.S.; Weissig, V. Treatment Strategies that Enhance the Efficacy and Selectivity of Mitochondria-Targeted Anticancer Agents. Int. J. Mol. Sci. 2015, 16, 17394–17421. [Google Scholar] [CrossRef]
  50. Ashton, T.M.; McKenna, W.G.; Kunz-Schughart, L.A.; Higgins, G.S. Oxidative Phosphorylation as an Emerging Target in Cancer Therapy. Clin. Cancer Res. 2018, 24, 2482–2490. [Google Scholar] [CrossRef] [Green Version]
  51. Fontana, F.; Raimondi, M.; Marzagalli, M.; Di Domizio, A.; Limonta, P. Natural Compounds in Prostate Cancer Prevention and Treatment: Mechanisms of Action and Molecular Targets. Cells 2020, 9, 460. [Google Scholar] [CrossRef] [PubMed]
  52. Lee, J.-S.; Lee, H.; Jang, H.; Woo, S.M.; Park, J.B.; Lee, S.-H.; Kang, J.H.; Kim, H.Y.; Song, J.; Kim, S.-Y. Targeting Oxidative Phosphorylation Reverses Drug Resistance in Cancer Cells by Blocking Autophagy Recycling. Cells 2020, 9, 2013. [Google Scholar] [CrossRef]
  53. Iessi, E.; Vona, R.; Cittadini, C.; Matarrese, P. Targeting the Interplay between Cancer Metabolic Reprogramming and Cell Death Pathways as a Viable Therapeutic Path. Biomedicines 2021, 9, 1942. [Google Scholar] [CrossRef]
  54. Raimondi, M.; Fontana, F.; Marzagalli, M.; Audano, M.; Beretta, G.; Procacci, P.; Sartori, P.; Mitro, N.; Limonta, P. Ca(2+) overload- and ROS-associated mitochondrial dysfunction contributes to delta-tocotrienol-mediated paraptosis in melanoma cells. Apoptosis 2021, 26, 277–292. [Google Scholar] [CrossRef]
  55. Wu, Z.; Ho, W.S.; Lu, R. Targeting Mitochondrial Oxidative Phosphorylation in Glioblastoma Therapy. NeuroMol. Med. 2022, 24, 18–22. [Google Scholar] [CrossRef]
  56. Yu, H.-J.; Xiao, G.-L.; Zhao, Y.-Y.; Wang, X.-X.; Lan, R. Targeting Mitochondrial Metabolism and RNA Polymerase POLRMT to Overcome Multidrug Resistance in Cancer. Front. Chem. 2021, 9, 775226. [Google Scholar] [CrossRef]
  57. Wallace, D.C. Mitochondria and cancer. Nat. Rev. Cancer 2012, 12, 685–698. [Google Scholar] [CrossRef] [PubMed]
  58. Halestrap, A.P.; Richardson, A.P. The mitochondrial permeability transition: A current perspective on its identity and role in ischaemia/reperfusion injury. J. Mol. Cell. Cardiol. 2015, 78, 129–141. [Google Scholar] [CrossRef]
  59. DeBerardinis, R.J.; Chandel, N.S. Fundamentals of cancer metabolism. Sci. Adv. 2016, 2, e1600200. [Google Scholar] [CrossRef] [PubMed]
  60. Brillo, V.; Chieregato, L.; Leanza, L.; Muccioli, S.; Costa, R. Mitochondrial Dynamics, ROS, and Cell Signaling: A Blended Overview. Life 2021, 11, 332. [Google Scholar] [CrossRef]
  61. Choudhury, F.K. Mitochondrial Redox Metabolism: The Epicenter of Metabolism during Cancer Progression. Antioxidants 2021, 10, 1838. [Google Scholar] [CrossRef]
  62. Quinlan, C.L.; Perevoshchikova, I.V.; Hey-Mogensen, M.; Orr, A.L.; Brand, M.D. Sites of reactive oxygen species generation by mitochondria oxidizing different substrates. Redox Biol. 2013, 1, 304–312. [Google Scholar] [CrossRef]
  63. Aggarwal, V.; Tuli, H.S.; Varol, A.; Thakral, F.; Yerer, M.B.; Sak, K.; Varol, M.; Jain, A.; Khan, M.A.; Sethi, G. Role of Reactive Oxygen Species in Cancer Progression: Molecular Mechanisms and Recent Advancements. Biomolecules 2019, 9, 735. [Google Scholar] [CrossRef]
  64. Parascandolo, A.; Laukkanen, M.O. Carcinogenesis and Reactive Oxygen Species Signaling: Interaction of the NADPH Oxidase NOX1-5 and Superoxide Dismutase 1-3 Signal Transduction Pathways. Antioxid. Redox Signal. 2019, 30, 443–486. [Google Scholar] [CrossRef] [PubMed]
  65. Hayes, J.D.; Dinkova-Kostova, A.T.; Tew, K.D. Oxidative Stress in Cancer. Cancer Cell 2020, 38, 167–197. [Google Scholar] [CrossRef]
  66. Kohan, R.; Collin, A.; Guizzardi, S.; Tolosa De Talamoni, N.; Picotto, G. Reactive oxygen species in cancer: A paradox between pro- and anti-tumour activities. Cancer Chemother. Pharmacol. 2020, 86, 1–13. [Google Scholar] [CrossRef] [PubMed]
  67. Arfin, S.; Jha, N.K.; Jha, S.K.; Kesari, K.K.; Ruokolainen, J.; Roychoudhury, S.; Rathi, B.; Kumar, D. Oxidative Stress in Cancer Cell Metabolism. Antioxidants 2021, 10, 642. [Google Scholar] [CrossRef]
  68. Wallace, D.C.; Fan, W.; Procaccio, V. Mitochondrial Energetics and Therapeutics. Annu. Rev. Pathol. 2010, 5, 297–348. [Google Scholar] [CrossRef] [PubMed]
  69. Gupta, S.C.; Hevia, D.; Patchva, S.; Park, B.; Koh, W.; Aggarwal, B.B. Upsides and Downsides of Reactive Oxygen Species for Cancer: The Roles of Reactive Oxygen Species in Tumorigenesis, Prevention, and Therapy. Antioxid. Redox Signal. 2012, 16, 1295–1322. [Google Scholar] [CrossRef]
  70. Nakamura, H.; Takada, K. Reactive oxygen species in cancer: Current findings and future directions. Cancer Sci. 2021, 112, 3945–3952. [Google Scholar] [CrossRef]
  71. Zahra, K.F.; Lefter, R.; Ali, A.; Abdellah, E.-C.; Trus, C.; Ciobica, A.; Timofte, D. The Involvement of the Oxidative Stress Status in Cancer Pathology: A Double View on the Role of the Antioxidants. Oxid. Med. Cell. Longev. 2021, 2021, 9965916. [Google Scholar] [CrossRef]
  72. Bekhet, O.H.; Eid, M.E. The interplay between reactive oxygen species and antioxidants in cancer progression and therapy: A narrative review. Transl. Cancer Res. 2021, 10, 4196–4206. [Google Scholar] [CrossRef]
  73. Delierneux, C.; Kouba, S.; Shanmughapriya, S.; Potier-Cartereau, M.; Trebak, M.; Hempel, N. Mitochondrial Calcium Regulation of Redox Signaling in Cancer. Cells 2020, 9, 432. [Google Scholar] [CrossRef] [PubMed]
  74. Macasoi, I.; Mioc, A.; Mioc, M.; Racoviceanu, R.; Soica, I.; Chevereșan, A.; Dehelean, C.; Dumitrașcu, V. Targeting Mitochondria through the Use of Mitocans as Emerging Anticancer Agents. Curr. Med. Chem. 2019, 27, 5730–5757. [Google Scholar] [CrossRef] [PubMed]
  75. Sterea, A.M.; El Hiani, Y. The Role of Mitochondrial Calcium Signaling in the Pathophysiology of Cancer Cells. Adv. Exp. Med. Biol. 2019, 1131, 747–770. [Google Scholar] [CrossRef]
  76. Genovese, I.; Carinci, M.; Modesti, L.; Aguiari, G.; Pinton, P.; Giorgi, C. Mitochondria: Insights into Crucial Features to Overcome Cancer Chemoresistance. Int. J. Mol. Sci. 2021, 22, 4770. [Google Scholar] [CrossRef]
  77. Ploumi, C.; Daskalaki, I.; Tavernarakis, N. Mitochondrial biogenesis and clearance: A balancing act. FEBS J. 2017, 284, 183–195. [Google Scholar] [CrossRef]
  78. Fontana, F.; Raimondi, M.; Marzagalli, M.; Audano, M.; Beretta, G.; Procacci, P.; Sartori, P.; Mitro, N.; Limonta, P. Mitochondrial functional and structural impairment is involved in the antitumor activity of delta-tocotrienol in prostate cancer cells. Free. Radic. Biol. Med. 2020, 160, 376–390. [Google Scholar] [CrossRef]
  79. Ma, Y.; Wang, L.; Jia, R. The role of mitochondrial dynamics in human cancers. Am. J. Cancer Res. 2020, 10, 1278–1293. [Google Scholar]
  80. Lee, Y.G.; Park, D.H.; Chae, Y.C. Role of Mitochondrial Stress Response in Cancer Progression. Cells 2022, 11, 771. [Google Scholar] [CrossRef] [PubMed]
  81. Liu, H.; Zang, C.; Yuan, F.; Ju, C.; Shang, M.; Ning, J.; Yang, Y.; Ma, J.; Li, G.; Bao, X.; et al. The role of FUNDC1 in mitophagy, mitochondrial dynamics and human diseases. Biochem. Pharmacol. 2022, 197, 114891. [Google Scholar] [CrossRef]
  82. Wang, S.; Tan, J.; Miao, Y.; Zhang, Q. Mitochondrial Dynamics, Mitophagy, and Mitochondria–Endoplasmic Reticulum Contact Sites Crosstalk Under Hypoxia. Front. Cell Dev. Biol. 2022, 10, 848214. [Google Scholar] [CrossRef]
  83. Grasso, D.; Medeiros, H.; Zampieri, L.X.; Bol, V.; Danhier, P.; Van Gisbergen, M.W.; Bouzin, C.; Brusa, D.; Grégoire, V.; Smeets, H.; et al. Fitter Mitochondria Are Associated with Radioresistance in Human Head and Neck SQD9 Cancer Cells. Front. Pharmacol. 2020, 11, 263. [Google Scholar] [CrossRef]
  84. Popov, L. Mitochondrial biogenesis: An update. J. Cell. Mol. Med. 2020, 24, 4892–4899. [Google Scholar] [CrossRef]
  85. Ekstrand, M.I.; Falkenberg, M.; Rantanen, A.; Park, C.B.; Gaspari, M.; Hultenby, K.; Rustin, P.; Gustafsson, C.M.; Larsson, N.-G. Mitochondrial transcription factor A regulates mtDNA copy number in mammals. Hum. Mol. Genet. 2004, 13, 935–944. [Google Scholar] [CrossRef]
  86. Canto, C.; Auwerx, J. PGC-1α, SIRT1 and AMPK, an energy sensing network that controls energy expenditure. Curr. Opin. Lipidol. 2009, 20, 98–105. [Google Scholar] [CrossRef]
  87. Bost, F.; Kaminski, L. The metabolic modulator PGC-1α in cancer. Am. J. Cancer Res. 2019, 9, 198–211. [Google Scholar]
  88. Zhang, Q.; Chen, W.; Xie, C.; Dai, X.; Ma, J.; Lu, J. The Role of PGC-1α in Digestive System Malignant Tumours. Anti-Cancer Agents Med. Chem. 2020, 20, 276–285. [Google Scholar] [CrossRef]
  89. Farnie, G.; Sotgia, F.; Lisanti, M.P. High mitochondrial mass identifies a sub-population of stem-like cancer cells that are chemo-resistant. Oncotarget 2015, 6, 30472–30486. [Google Scholar] [CrossRef]
  90. Raggi, C.; Taddei, M.L.; Sacco, E.; Navari, N.; Correnti, M.; Piombanti, B.; Pastore, M.; Campani, C.; Pranzini, E.; Iorio, J.; et al. Mitochondrial oxidative metabolism contributes to a cancer stem cell phenotype in cholangiocarcinoma. J. Hepatol. 2021, 74, 1373–1385. [Google Scholar] [CrossRef]
  91. Praharaj, P.P.; Panigrahi, D.P.; Bhol, C.S.; Patra, S.; Mishra, S.R.; Mahapatra, K.K.; Behera, B.P.; Singh, A.; Patil, S.; Bhutia, S.K. Mitochondrial rewiring through mitophagy and mitochondrial biogenesis in cancer stem cells: A potential target for anti-CSC cancer therapy. Cancer Lett. 2021, 498, 217–228. [Google Scholar] [CrossRef] [PubMed]
  92. Chen, H.C.; Detmer, S.A.; Ewald, A.J.; Griffin, E.E.; Fraser, S.E.; Chan, D.C. Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J. Cell Biol. 2003, 160, 189–200. [Google Scholar] [CrossRef]
  93. Koshiba, T.; Detmer, S.A.; Kaiser, J.T.; Chen, H.; McCaffery, J.M.; Chan, D.C. Structural Basis of Mitochondrial Tethering by Mitofusin Complexes. Science 2004, 305, 858–862. [Google Scholar] [CrossRef]
  94. Martens, S.; McMahon, H.T. Mechanisms of membrane fusion: Disparate players and common principles. Nat. Rev. Mol. Cell Biol. 2008, 9, 543–556. [Google Scholar] [CrossRef] [PubMed]
  95. Ban, T.; Ishihara, T.; Kohno, H.; Saita, S.; Ichimura, A.; Maenaka, K.; Oka, T.; Mihara, K.; Ishihara, N. Molecular basis of selective mitochondrial fusion by heterotypic action between OPA1 and cardiolipin. Nat. Cell Biol. 2017, 19, 856–863. [Google Scholar] [CrossRef] [PubMed]
  96. Friedman, J.R.; Lackner, L.L.; West, M.; DiBenedetto, J.R.; Nunnari, J.; Voeltz, G.K. ER Tubules Mark Sites of Mitochondrial Division. Science 2011, 334, 358–362. [Google Scholar] [CrossRef]
  97. Lee, H.; Yoon, Y. Mitochondrial fission and fusion. Biochem. Soc. Trans. 2016, 44, 1725–1735. [Google Scholar] [CrossRef]
  98. Steffen, J.; Koehler, C.M. ER–mitochondria contacts: Actin dynamics at the ER control mitochondrial fission via calcium release. J. Cell Biol. 2017, 217, 15–17. [Google Scholar] [CrossRef]
  99. Banerjee, R.; Mukherjee, A.; Nagotu, S. Mitochondrial dynamics and its impact on human health and diseases: Inside the DRP1 blackbox. J. Mol. Med. 2021, 100, 1–21. [Google Scholar] [CrossRef]
  100. Ding, W.-X.; Yin, X.-M. Mitophagy: Mechanisms, pathophysiological roles, and analysis. Biol. Chem. 2012, 393, 547–564. [Google Scholar] [CrossRef]
  101. Kulikov, A.V.; Luchkina, E.A.; Gogvadze, V.; Zhivotovsky, B. Mitophagy: Link to cancer development and therapy. Biochem. Biophys. Res. Commun. 2017, 482, 432–439. [Google Scholar] [CrossRef]
  102. Denisenko, T.V.; Gogvadze, V.; Zhivotovsky, B. Mitophagy in carcinogenesis and cancer treatment. Discov. Oncol. 2021, 12, 58. [Google Scholar] [CrossRef]
  103. Iorio, R.; Celenza, G.; Petricca, S. Mitophagy: Molecular Mechanisms, New Concepts on Parkin Activation and the Emerging Role of AMPK/ULK1 Axis. Cells 2021, 11, 30. [Google Scholar] [CrossRef]
  104. Poole, L.P.; Macleod, K.F. Mitophagy in tumorigenesis and metastasis. Cell. Mol. Life Sci. 2021, 78, 3817–3851. [Google Scholar] [CrossRef]
  105. Qiu, Y.-H.; Zhang, T.-S.; Wang, X.-W.; Wang, M.-Y.; Zhao, W.-X.; Zhou, H.-M.; Zhang, C.-H.; Cai, M.-L.; Chen, X.-F.; Zhao, W.-L.; et al. Mitochondria autophagy: A potential target for cancer therapy. J. Drug Target. 2021, 29, 576–591. [Google Scholar] [CrossRef]
  106. Ray, S.K.; Mukherjee, S. Mitophagy in Carcinogenesis and Tumour Progression- A New Paradigm with Emerging Importance. Anti-Cancer Agents Med. Chem. 2021, 21, 2130–2141. [Google Scholar] [CrossRef]
  107. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef]
  108. Labrie, F.; Candas, B.; Gomez, J.-L.; Cusan, L. Can combined androgen blockade provide long-term control or possible cure of localized prostate cancer? Urology 2002, 60, 115–119. [Google Scholar] [CrossRef]
  109. James, N.D.; De Bono, J.S.; Spears, M.R.; Clarke, N.W.; Mason, M.D.; Dearnaley, D.P.; Ritchie, A.W.S.; Amos, C.L.; Gilson, C.; Jones, R.J.; et al. Abiraterone for Prostate Cancer Not Previously Treated with Hormone Therapy. N. Engl. J. Med. 2017, 377, 338–351. [Google Scholar] [CrossRef]
  110. Litwin, M.S.; Tan, H.-J. The Diagnosis and Treatment of Prostate Cancer: A Review. JAMA 2017, 317, 2532–2542. [Google Scholar] [CrossRef]
  111. Onozawa, M.; Akaza, H.; Hinotsu, S.; Oya, M.; Ogawa, O.; Kitamura, T.; Suzuki, K.; Naito, S.; Namiki, M.; Nishimura, K.; et al. Combined androgen blockade achieved better oncological outcome in androgen deprivation therapy for prostate cancer: Analysis of community-based multi-institutional database across Japan using propensity score matching. Cancer Med. 2018, 7, 4893–4902. [Google Scholar] [CrossRef]
  112. Armstrong, A.J.; Szmulewitz, R.Z.; Petrylak, D.P.; Holzbeierlein, J.; Villers, A.; Azad, A.; Alcaraz, A.; Alekseev, B.; Iguchi, T.; Shore, N.D.; et al. ARCHES: A Randomized, Phase III Study of Androgen Deprivation Therapy With Enzalutamide or Placebo in Men With Metastatic Hormone-Sensitive Prostate Cancer. J. Clin. Oncol. 2019, 37, 2974–2986. [Google Scholar] [CrossRef]
  113. Chi, K.N.; Agarwal, N.; Bjartell, A.; Chung, B.H.; Pereira de Santana Gomes, A.J.; Given, R.; Juarez Soto, A.; Merseburger, A.S.; Ozguroglu, M.; Uemura, H.; et al. Apalutamide for Metastatic, Castration-Sensitive Prostate Cancer. N. Engl. J. Med. 2019, 381, 13–24. [Google Scholar] [CrossRef]
  114. Kim, T.; Lee, Y.; Koo, K. Current Status and Future Perspectives of Androgen Receptor Inhibition Therapy for Prostate Cancer: A Comprehensive Review. Biomolecules 2021, 11, 492. [Google Scholar] [CrossRef]
  115. Perner, S.; Cronauer, M.V.; Schrader, A.J.; Klocker, H.; Culig, Z.; Baniahmad, A. Adaptive responses of androgen receptor signaling in castration-resistant prostate cancer. Oncotarget 2015, 6, 35542–35555. [Google Scholar] [CrossRef]
  116. Galletti, G.; Leach, B.I.; Lam, L.; Tagawa, S.T. Mechanisms of resistance to systemic therapy in metastatic castration-resistant prostate cancer. Cancer Treat. Rev. 2017, 57, 16–27. [Google Scholar] [CrossRef]
  117. Marelli, M.M.; Moretti, R.M.; Januszkiewicz-Caulier, J.; Motta, M.; Limonta, P. Gonadotropin-Releasing Hormone (GnRH) Receptors in Tumors: A New Rationale for the Therapeutical Application of GnRH Analogs in Cancer Patients? Curr. Cancer Drug Targets 2006, 6, 257–269. [Google Scholar] [CrossRef]
  118. Limonta, P.; Marelli, M.M.; Mai, S.; Motta, M.; Martini, L.; Moretti, R.M. GnRH Receptors in Cancer: From Cell Biology to Novel Targeted Therapeutic Strategies. Endocr. Rev. 2012, 33, 784–811. [Google Scholar] [CrossRef]
  119. Limonta, P.; Manea, M. Gonadotropin-releasing hormone receptors as molecular therapeutic targets in prostate cancer: Current options and emerging strategies. Cancer Treat. Rev. 2013, 39, 647–663. [Google Scholar] [CrossRef]
  120. Manea, M.; Marelli, M.M.; Moretti, R.; Maggi, R.; Marzagalli, M.; Limonta, P. Targeting Hormonal Signaling Pathways in Castration Resistant Prostate Cancer. Recent Patents Anti-Cancer Drug Discov. 2014, 9, 267–285. [Google Scholar] [CrossRef]
  121. Jang, H.S.; Koo, K.C.; Cho, K.S.; Chung, B.H. Survival Outcomes of Concurrent Treatment with Docetaxel and Androgen Deprivation Therapy in Metastatic Castration-Resistant Prostate Cancer. Yonsei Med. J. 2016, 57, 1070–1078. [Google Scholar] [CrossRef]
  122. Komura, K.; Sweeney, C.J.; Inamoto, T.; Ibuki, N.; Azuma, H.; Kantoff, P.W. Current treatment strategies for advanced prostate cancer. Int. J. Urol. 2018, 25, 220–231. [Google Scholar] [CrossRef] [Green Version]
  123. Saad, F.; Shore, N.; Zhang, T.; Sharma, S.; Cho, H.K.; Jacobs, I.A. Emerging therapeutic targets for patients with advanced prostate cancer. Cancer Treat. Rev. 2019, 76, 1–9. [Google Scholar] [CrossRef]
  124. Fontana, F.; Marzagalli, M.; Marelli, M.M.; Raimondi, M.; Moretti, R.; Limonta, P. Gonadotropin-Releasing Hormone Receptors in Prostate Cancer: Molecular Aspects and Biological Functions. Int. J. Mol. Sci. 2020, 21, 9511. [Google Scholar] [CrossRef]
  125. Fontana, F.; Limonta, P. Dissecting the Hormonal Signaling Landscape in Castration-Resistant Prostate Cancer. Cells 2021, 10, 1133. [Google Scholar] [CrossRef]
  126. Hou, Z.; Huang, S.; Li, Z. Androgens in prostate cancer: A tale that never ends. Cancer Lett. 2021, 516, 1–12. [Google Scholar] [CrossRef] [PubMed]
  127. Fontana, F.; Anselmi, M.; Limonta, P. Molecular mechanisms and genetic alterations in prostate cancer: From diagnosis to targeted therapy. Cancer Lett. 2022, 534, 215619. [Google Scholar] [CrossRef] [PubMed]
  128. Grigor, E.J.; Fergusson, D.; Kekre, N.; Montroy, J.; Atkins, H.; Seftel, M.D.; Daugaard, M.; Presseau, J.; Thavorn, K.; Hutton, B.; et al. Risks and Benefits of Chimeric Antigen Receptor T-Cell (CAR-T) Therapy in Cancer: A Systematic Review and Meta-Analysis. Transfus. Med. Rev. 2019, 33, 98–110. [Google Scholar] [CrossRef] [PubMed]
  129. Markowski, M.C.; Shenderov, E.; Eisenberger, M.A.; Kachhap, S.; Pardoll, D.M.; Denmeade, S.R.; Antonarakis, E.S. Extreme responses to immune checkpoint blockade following bipolar androgen therapy and enzalutamide in patients with metastatic castration resistant prostate cancer. Prostate 2020, 80, 407–411. [Google Scholar] [CrossRef] [PubMed]
  130. Sun, B.L. Immunotherapy in treatment of metastatic prostate cancer: An approach to circumvent immunosuppressive tumor microenvironment. Prostate 2021, 81, 1125–1134. [Google Scholar] [CrossRef]
  131. Siewe, N.; Friedman, A. Combination therapy for mCRPC with immune checkpoint inhibitors, ADT and vaccine: A mathematical model. PLoS ONE 2022, 17, e0262453. [Google Scholar] [CrossRef]
  132. Grupp, K.; Jedrzejewska, K.; Tsourlakis, M.C.; Koop, C.; Wilczak, W.; Adam, M.; Quaas, A.; Sauter, G.; Simon, R.; Izbicki, J.R.; et al. High mitochondria content is associated with prostate cancer disease progression. Mol. Cancer 2013, 12, 145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Vayalil, P.K.; Landar, A. Mitochondrial oncobioenergetic index: A potential biomarker to predict progression from indolent to aggressive prostate cancer. Oncotarget 2015, 6, 43065–43080. [Google Scholar] [CrossRef]
  134. Kelly, R.S.; Sinnott, J.A.; Rider, J.R.; Ebot, E.M.; Gerke, T.; Bowden, M.; Pettersson, A.; Loda, M.; Sesso, H.D.; Kantoff, P.W.; et al. The role of tumor metabolism as a driver of prostate cancer progression and lethal disease: Results from a nested case-control study. Cancer Metab. 2016, 4, 22. [Google Scholar] [CrossRef] [PubMed]
  135. Eidelman, E.; Twum-Ampofo, J.; Ansari, J.; Siddiqui, M.M. The Metabolic Phenotype of Prostate Cancer. Front. Oncol. 2017, 7, 131. [Google Scholar] [CrossRef]
  136. Costello, L.C.; Feng, P.; Milon, B.; Tan, M.; Franklin, R.B. Role of zinc in the pathogenesis and treatment of prostate cancer: Critical issues to resolve. Prostate Cancer Prostatic Dis. 2004, 7, 111–117. [Google Scholar] [CrossRef] [PubMed]
  137. Costello, L.C.; Franklin, R.B. The clinical relevance of the metabolism of prostate cancer; zinc and tumor suppression: Connecting the dots. Mol. Cancer 2006, 5, 17. [Google Scholar] [CrossRef]
  138. Costello, L.C.; Franklin, R.B.; Zou, J.; Feng, P.; Bok, R.; Swanson, M.G.; Kurhanewicz, J. Human prostate cancer ZIP1/zinc/citrate genetic/metabolic relationship in the TRAMP prostate cancer animal model. Cancer Biol. Ther. 2011, 12, 1078–1084. [Google Scholar] [CrossRef]
  139. Costello, L.C.; Franklin, R.B. A comprehensive review of the role of zinc in normal prostate function and metabolism; and its implications in prostate cancer. Arch. Biochem. Biophys. 2016, 611, 100–112. [Google Scholar] [CrossRef]
  140. Hacioglu, C.; Kacar, S.; Kar, F.; Kanbak, G.; Sahinturk, V. Concentration-Dependent Effects of Zinc Sulfate on DU-145 Human Prostate Cancer Cell Line: Oxidative, Apoptotic, Inflammatory, and Morphological Analyzes. Biol. Trace Element Res. 2020, 195, 436–444. [Google Scholar] [CrossRef] [PubMed]
  141. Latonen, L.; Afyounian, E.; Jylhä, A.; Nättinen, J.; Aapola, U.; Annala, M.; Kivinummi, K.K.; Tammela, T.T.L.; Beuerman, R.W.; Uusitalo, H.; et al. Integrative proteomics in prostate cancer uncovers robustness against genomic and transcriptomic aberrations during disease progression. Nat. Commun. 2018, 9, 1176. [Google Scholar] [CrossRef] [PubMed]
  142. Shao, Y.; Ye, G.; Ren, S.; Piao, H.-L.; Zhao, X.; Lu, X.; Wang, F.; Ma, W.; Li, J.; Yin, P.; et al. Metabolomics and transcriptomics profiles reveal the dysregulation of the tricarboxylic acid cycle and related mechanisms in prostate cancer. Int. J. Cancer 2018, 143, 396–407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Bader, D.A.; McGuire, S.E. Tumour metabolism and its unique properties in prostate adenocarcinoma. Nat. Rev. Urol. 2020, 17, 214–231. [Google Scholar] [CrossRef]
  144. Kolenko, V.; Teper, E.; Kutikov, A.; Uzzo, R. Zinc and zinc transporters in prostate carcinogenesis. Nat. Rev. Urol. 2013, 10, 219–226. [Google Scholar] [CrossRef] [PubMed]
  145. Franz, M.-C.; Anderle, P.; Bürzle, M.; Suzuki, Y.; Freeman, M.; Hediger, M.; Kovacs, G. Zinc transporters in prostate cancer. Mol. Asp. Med. 2013, 34, 735–741. [Google Scholar] [CrossRef] [PubMed]
  146. Makhov, P.B.; Golovine, K.V.; Kutikov, A.; Canter, D.J.; Rybko, V.A.; Roshchin, D.A.; Matveev, V.B.; Uzzo, R.G.; Kolenko, V.M. Reversal of epigenetic silencing of AP-2alpha results in increased zinc uptake in DU-145 and LNCaP prostate cancer cells. Carcinogenesis 2011, 32, 1773–1781. [Google Scholar] [CrossRef]
  147. Xue, Y.N.; Yu, B.B.; Liu, Y.N.; Guo, R.; Li, L.J.; Zhang, L.C.; Su, J.; Sun, L.K.; Li, Y. Zinc promotes prostate cancer cell chemosensitivity to paclitaxel by inhibiting epithelial-mesenchymal transition and inducing apoptosis. Prostate 2019, 79, 647–656. [Google Scholar] [CrossRef] [PubMed]
  148. Sauer, A.K.; Vela, H.; Vela, G.; Stark, P.; Barrera-Juarez, E.; Grabrucker, A.M. Zinc Deficiency in Men Over 50 and Its Implications in Prostate Disorders. Front. Oncol. 2020, 10, 1293. [Google Scholar] [CrossRef] [PubMed]
  149. Ahmad, F.; Cherukuri, M.K.; Choyke, P.L. Metabolic reprogramming in prostate cancer. Br. J. Cancer 2021, 125, 1185–1196. [Google Scholar] [CrossRef]
  150. Bazylianska, V.; Kalpage, H.A.; Wan, J.; Vaishnav, A.; Mahapatra, G.; Turner, A.A.; Chowdhury, D.D.; Kim, K.; Morse, P.T.; Lee, I.; et al. Lysine 53 Acetylation of Cytochrome c in Prostate Cancer: Warburg Metabolism and Evasion of Apoptosis. Cells 2021, 10, 802. [Google Scholar] [CrossRef] [PubMed]
  151. Fang, M.; Shen, Z.; Huang, S.; Zhao, L.; Chen, S.; Mak, T.W.; Wang, X. The ER UDPase ENTPD5 Promotes Protein N-Glycosylation, the Warburg Effect, and Proliferation in the PTEN Pathway. Cell 2010, 143, 711–724. [Google Scholar] [CrossRef]
  152. Sun, Q.; Chen, X.; Ma, J.; Peng, H.; Wang, F.; Zha, X.; Wang, Y.; Jing, Y.; Yang, H.; Chen, R.; et al. Mammalian target of rapamycin up-regulation of pyruvate kinase isoenzyme type M2 is critical for aerobic glycolysis and tumor growth. Proc. Natl. Acad. Sci. USA 2011, 108, 4129–4134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Wang, L.; Xiong, H.; Wu, F.; Zhang, Y.; Wang, J.; Zhao, L.; Guo, X.; Chang, L.-J.; Zhang, Y.; You, M.J.; et al. Hexokinase 2-Mediated Warburg Effect Is Required for PTEN- and p53-Deficiency-Driven Prostate Cancer Growth. Cell Rep. 2014, 8, 1461–1474. [Google Scholar] [CrossRef] [PubMed]
  154. Singh, R.; Mills, I.G. The Interplay Between Prostate Cancer Genomics, Metabolism, and the Epigenome: Perspectives and Future Prospects. Front. Oncol. 2021, 11, 704353. [Google Scholar] [CrossRef]
  155. Wang, C.; Tao, W.; Ni, S.; Chen, Q. SENP1 Interacts with HIF1α to Regulate Glycolysis of Prostatic Carcinoma Cells. Int. J. Biol. Sci. 2019, 15, 395–403. [Google Scholar] [CrossRef] [PubMed]
  156. Sun, X.; Huang, Q.; Peng, F.; Wang, J.; Zhao, W.; Guo, G. Expression and Clinical Significance of HKII and HIF-1α in Grade Groups of Prostate Cancer. Front. Genet. 2021, 12, 680928. [Google Scholar] [CrossRef] [PubMed]
  157. Nassar, Z.D.; Aref, A.T.; Miladinovic, D.; Mah, C.Y.; Raj, G.V.; Hoy, A.J.; Butler, L.M. Peri-prostatic adipose tissue: The metabolic microenvironment of prostate cancer. BJU Int. 2018, 121 (Suppl. S3), 9–21. [Google Scholar] [CrossRef] [PubMed]
  158. Himbert, C.; Delphan, M.; Scherer, D.; Bowers, L.W.; Hursting, S.; Ulrich, C.M. Signals from the Adipose Microenvironment and the Obesity–Cancer Link—A Systematic Review. Cancer Prev. Res. 2017, 10, 494–506. [Google Scholar] [CrossRef]
  159. Diedrich, J.D.; Rajagurubandara, E.; Herroon, M.K.; Mahapatra, G.; Hüttemann, M.; Podgorski, I. Bone marrow adipocytes promote the Warburg phenotype in metastatic prostate tumorsviaHIF-1α activation. Oncotarget 2016, 7, 64854–64877. [Google Scholar] [CrossRef] [PubMed]
  160. Cutruzzolà, F.; Giardina, G.; Marani, M.; Macone, A.; Paiardini, A.; Rinaldo, S.; Paone, A. Glucose Metabolism in the Progression of Prostate Cancer. Front. Physiol. 2017, 8, 97. [Google Scholar] [CrossRef] [PubMed]
  161. Fontana, F.; Anselmi, M.; Carollo, E.; Sartori, P.; Procacci, P.; Carter, D.; Limonta, P. Adipocyte-Derived Extracellular Vesicles Promote Prostate Cancer Cell Aggressiveness by Enabling Multiple Phenotypic and Metabolic Changes. Cells 2022, 11, 2388. [Google Scholar] [CrossRef] [PubMed]
  162. Dakubo, G.D.; Parr, R.L.; Costello, L.C.; Franklin, R.B.; Thayer, R.E. Altered metabolism and mitochondrial genome in prostate cancer. J. Clin. Pathol. 2006, 59, 10–16. [Google Scholar] [CrossRef] [Green Version]
  163. Chen, J.; Guccini, I.; Di Mitri, D.; Brina, D.; Revandkar, A.; Sarti, M.; Pasquini, E.; Alajati, A.; Pinton, S.; Losa, M.; et al. Compartmentalized activities of the pyruvate dehydrogenase complex sustain lipogenesis in prostate cancer. Nat. Genet. 2018, 50, 219–228. [Google Scholar] [CrossRef]
  164. Oberhuber, M.; Pecoraro, M.; Rusz, M.; Oberhuber, G.; Wieselberg, M.; Haslinger, P.; Gurnhofer, E.; Schlederer, M.; Limberger, T.; Lagger, S.; et al. STAT 3 -dependent analysis reveals PDK 4 as independent predictor of recurrence in prostate cancer. Mol. Syst. Biol. 2020, 16, e9247. [Google Scholar] [CrossRef] [PubMed]
  165. Chen, C.-L.; Lin, C.-Y.; Kung, H.-J. Targeting Mitochondrial OXPHOS and Their Regulatory Signals in Prostate Cancers. Int. J. Mol. Sci. 2021, 22, 13435. [Google Scholar] [CrossRef] [PubMed]
  166. Galbraith, L.C.A.; Mui, E.; Nixon, C.; Hedley, A.; Strachan, D.; MacKay, G.; Sumpton, D.; Sansom, O.J.; Leung, H.Y.; Ahmad, I. PPAR-gamma induced AKT3 expression increases levels of mitochondrial biogenesis driving prostate cancer. Oncogene 2021, 40, 2355–2366. [Google Scholar] [CrossRef]
  167. Giannoni, E.; Taddei, M.L.; Morandi, A.; Comito, G.; Calvani, M.; Bianchini, F.; Richichi, B.; Raugei, G.; Wong, N.; Tang, D.; et al. Targeting stromal-induced pyruvate kinase M2 nuclear translocation impairs OXPHOS and prostate cancer metastatic spread. Oncotarget 2015, 6, 24061–24074. [Google Scholar] [CrossRef]
  168. Dayton, T.L.; Jacks, T.; Vander Heiden, M.G. PKM 2, cancer metabolism, and the road ahead. EMBO Rep. 2016, 17, 1721–1730. [Google Scholar] [CrossRef]
  169. Hsu, M.-C.; Hung, W.-C. Pyruvate kinase M2 fuels multiple aspects of cancer cells: From cellular metabolism, transcriptional regulation to extracellular signaling. Mol. Cancer 2018, 17, 35. [Google Scholar] [CrossRef]
  170. Bader, D.A.; Hartig, S.M.; Putluri, V.; Foley, C.; Hamilton, M.P.; Smith, E.A.; Saha, P.K.; Panigrahi, A.; Walker, C.; Zong, L.; et al. Mitochondrial pyruvate import is a metabolic vulnerability in androgen receptor-driven prostate cancer. Nat. Metab. 2018, 1, 70–85. [Google Scholar] [CrossRef]
  171. Lee, Y.G.; Nam, Y.; Shin, K.J.; Yoon, S.; Park, W.S.; Joung, J.Y.; Seo, J.K.; Jang, J.; Lee, S.; Nam, D.; et al. Androgen-induced expression of DRP1 regulates mitochondrial metabolic reprogramming in prostate cancer. Cancer Lett. 2020, 471, 72–87. [Google Scholar] [CrossRef]
  172. Kumar, R.P.; Ray, S.; Home, P.; Saha, B.; Bhattacharya, B.; Wilkins, H.M.; Chavan, H.; Ganguly, A.; Milano-Foster, J.; Paul, A.; et al. Regulation of energy metabolism during early mammalian development: TEAD4 controls mitochondrial transcription. Development 2018, 145, dev162644. [Google Scholar] [CrossRef] [Green Version]
  173. Chen, C.-L.; Hsu, S.-C.; Chung, T.-Y.; Chu, C.-Y.; Wang, H.-J.; Hsiao, P.-W.; Yeh, S.-D.; Ann, D.K.; Yen, Y.; Kung, H.-J. Arginine is an epigenetic regulator targeting TEAD4 to modulate OXPHOS in prostate cancer cells. Nat. Commun. 2021, 12, 2398. [Google Scholar] [CrossRef]
  174. Skvortsov, S.; Skvortsova, I.-I.; Tang, D.G.; Dubrovska, A. Concise Review: Prostate Cancer Stem Cells: Current Understanding. Stem Cells 2018, 36, 1457–1474. [Google Scholar] [CrossRef] [PubMed]
  175. Sotgia, F.; Martinez-Outschoorn, U.E.; Lisanti, M.P. Cancer Metabolism: New Validated Targets for Drug Discovery. Oncotarget 2013, 4, 1309–1316. [Google Scholar] [CrossRef] [PubMed]
  176. Sotgia, F.; Ozsvari, B.; Fiorillo, M.; De Francesco, E.M.; Bonuccelli, G.; Lisanti, M.P. A mitochondrial based oncology platform for targeting cancer stem cells (CSCs): MITO-ONC-RX. Cell Cycle 2018, 17, 2091–2100. [Google Scholar] [CrossRef] [PubMed]
  177. Mayer, M.J.; Klotz, L.H.; Venkateswaran, V. Metformin and prostate cancer stem cells: A novel therapeutic target. Prostate Cancer Prostatic Dis. 2015, 18, 303–309. [Google Scholar] [CrossRef]
  178. Zhong, Y.; Li, X.; Yu, D.; Li, X.; Li, Y.; Long, Y.; Yuan, Y.; Ji, Z.; Zhang, M.; Wen, J.-G.; et al. Application of mitochondrial pyruvate carrier blocker UK5099 creates metabolic reprogram and greater stem-like properties in LnCap prostate cancer cells in vitro. Oncotarget 2015, 6, 37758–37769. [Google Scholar] [CrossRef]
  179. Basu, H.S.; Wilganowski, N.; Robertson, S.; Reuben, J.M.; Cohen, E.N.; Zurita, A.; Ramachandran, S.; Xiao, L.; Titus, M.; Wilding, G. Prostate cancer cells survive anti-androgen and mitochondrial metabolic inhibitors by modulating glycolysis and mitochondrial metabolic activities. Prostate 2021, 81, 799–811. [Google Scholar] [CrossRef]
  180. Mamouni, K.; Kallifatidis, G.; Lokeshwar, B. Targeting Mitochondrial Metabolism in Prostate Cancer with Triterpenoids. Int. J. Mol. Sci. 2021, 22, 2466. [Google Scholar] [CrossRef]
  181. Massie, C.E.; Lynch, A.; Ramos-Montoya, A.; Boren, J.; Stark, R.; Fazli, L.; Warren, A.; Scott, H.; Madhu, B.; Sharma, N.; et al. The androgen receptor fuels prostate cancer by regulating central metabolism and biosynthesis. EMBO J. 2011, 30, 2719–2733. [Google Scholar] [CrossRef] [PubMed]
  182. Tennakoon, J.B.; Shi, Y.; Han, J.J.; Tsouko, E.; White, M.A.; Burns, A.R.; Zhang, A.; Xia, X.; Ilkayeva, O.R.; Xin, L.; et al. Androgens regulate prostate cancer cell growth via an AMPK-PGC-1α-mediated metabolic switch. Oncogene 2013, 33, 5251–5261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Audet-Walsh, É.; Vernier, M.; Yee, T.; E Laflamme, C.; Li, S.; Chen, Y.; Giguère, V. SREBF1 Activity Is Regulated by an AR/mTOR Nuclear Axis in Prostate Cancer. Mol. Cancer Res. 2018, 16, 1396–1405. [Google Scholar] [CrossRef] [PubMed]
  184. Gonthier, K.; Poluri, R.T.K.; Audet-Walsh, E. Functional genomic studies reveal the androgen receptor as a master regulator of cellular energy metabolism in prostate cancer. J. Steroid Biochem. Mol. Biol. 2019, 191, 105367. [Google Scholar] [CrossRef] [PubMed]
  185. Stone, L. Mitochondrial metabolism: A target in AR-driven disease. Nat. Rev. Urol. 2019, 16, 1. [Google Scholar] [CrossRef]
  186. Xu, H.; Liu, Z.; Gao, D.; Li, P.; Shen, Y.; Sun, Y.; Xu, L.; Song, N.; Wang, Y.; Zhan, M.; et al. Reprogramming hormone-sensitive prostate cancer to a lethal neuroendocrine cancer lineage by mitochondrial pyruvate carrier (MPC). Mol. Metab. 2022, 59, 101466. [Google Scholar] [CrossRef]
  187. Wu, M.-J.; Chen, C.-J.; Lin, T.-Y.; Liu, Y.-Y.; Tseng, L.-L.; Cheng, M.-L.; Chuu, C.-P.; Tsai, H.-K.; Kuo, W.-L.; Kung, H.-J.; et al. Targeting KDM4B that coactivates c-Myc-regulated metabolism to suppress tumor growth in castration-resistant prostate cancer. Theranostics 2021, 11, 7779–7796. [Google Scholar] [CrossRef]
  188. Bajpai, P.; Koc, E.; Sonpavde, G.; Singh, R.; Singh, K.K. Mitochondrial localization, import, and mitochondrial function of the androgen receptor. J. Biol. Chem. 2019, 294, 6621–6634. [Google Scholar] [CrossRef]
  189. Valcarcel-Jimenez, L.; Gaude, E.; Torrano, V.; Frezza, C.; Carracedo, A. Mitochondrial Metabolism: Yin and Yang for Tumor Progression. Trends Endocrinol. Metab. 2017, 28, 748–757. [Google Scholar] [CrossRef] [PubMed]
  190. Quinlan, C.L.; Treberg, J.R.; Perevoshchikova, I.V.; Orr, A.L.; Brand, M.D. Native rates of superoxide production from multiple sites in isolated mitochondria measured using endogenous reporters. Free. Radic. Biol. Med. 2012, 53, 1807–1817. [Google Scholar] [CrossRef]
  191. Chen, J.Z.; Gokden, N.; Greene, G.F.; Mukunyadzi, P.; Kadlubar, F.F. Extensive somatic mitochondrial mutations in primary prostate cancer using laser capture microdissection. Cancer Res. 2002, 62, 6470–6474. [Google Scholar] [PubMed]
  192. Chen, J.Z.; Kadlubar, F.F.; Chen, J.Z. Mitochondrial Mutagenesis and Oxidative Stress in Human Prostate Cancer. J. Environ. Sci. Health C Environ. Carcinog. Ecotoxicol. Rev. 2004, 22, 1–12. [Google Scholar] [CrossRef] [PubMed]
  193. Lindberg, J.; Mills, I.G.; Klevebring, D.; Liu, W.; Neiman, M.; Xu, J.; Wikström, P.; Wiklund, P.; Wiklund, F.; Egevad, L.; et al. The Mitochondrial and Autosomal Mutation Landscapes of Prostate Cancer. Eur. Urol. 2013, 63, 702–708. [Google Scholar] [CrossRef]
  194. McCrow, J.P.; Petersen, D.C.; Louw, M.; Chan, E.K.F.; Harmeyer, K.; Vecchiarelli, S.; Lyons, R.J.; Bornman, M.S.R.; Hayes, V.M. Spectrum of mitochondrial genomic variation and associated clinical presentation of prostate cancer in South African men. Prostate 2016, 76, 349–358. [Google Scholar] [CrossRef]
  195. Hopkins, J.F.; Sabelnykova, V.Y.; Weischenfeldt, J.; Simon, R.; Aguiar, J.A.; Alkallas, R.; Heisler, L.E.; Zhang, J.; Watson, J.D.; Chua, M.L.K.; et al. Mitochondrial mutations drive prostate cancer aggression. Nat. Commun. 2017, 8, 656. [Google Scholar] [CrossRef]
  196. Gómez-Zaera, M.; Abril, J.; González, L.; Aguiló, F.; Condom, E.; Nadal, M.; Nunes, V. Identification of somatic and germline mitochondrial DNA sequence variants in prostate cancer patients. Mutat. Res. 2006, 595, 42–51. [Google Scholar] [CrossRef] [PubMed]
  197. Kloss-Brandstätter, A.; Schäfer, G.; Erhart, G.; Hüttenhofer, A.; Coassin, S.; Seifarth, C.; Summerer, M.; Bektic, J.; Klocker, H.; Kronenberg, F. Somatic Mutations throughout the Entire Mitochondrial Genome Are Associated with Elevated PSA Levels in Prostate Cancer Patients. Am. J. Hum. Genet. 2010, 87, 802–812. [Google Scholar] [CrossRef]
  198. Niedzwiecka, K.; Tisi, R.; Penna, S.; Lichocka, M.; Plochocka, D.; Kucharczyk, R. Two mutations in mitochondrial ATP6 gene of ATP synthase, related to human cancer, affect ROS, calcium homeostasis and mitochondrial permeability transition in yeast. Biochim. Biophys. Acta (BBA)—Mol. Cell Res. 2018, 1865, 117–131. [Google Scholar] [CrossRef]
  199. Schöpf, B.; Weissensteiner, H.; Schäfer, G.; Fazzini, F.; Charoentong, P.; Naschberger, A.; Rupp, B.; Fendt, L.; Bukur, V.; Giese, I.; et al. OXPHOS remodeling in high-grade prostate cancer involves mtDNA mutations and increased succinate oxidation. Nat. Commun. 2020, 11, 1487. [Google Scholar] [CrossRef]
  200. Sun, Q.; Arnold, R.S.; Sun, C.Q.; Petros, J.A. A mitochondrial DNA mutation influences the apoptotic effect of statins on prostate cancer. Prostate 2015, 75, 1916–1925. [Google Scholar] [CrossRef]
  201. Zhao, J.; Zhang, J.; Yu, M.; Xie, Y.; Huang, Y.; Wolff, D.W.; Abel, P.W.; Tu, Y. Mitochondrial dynamics regulates migration and invasion of breast cancer cells. Oncogene 2013, 32, 4814–4824. [Google Scholar] [CrossRef] [PubMed]
  202. Audano, M.; Pedretti, S.; Ligorio, S.; Crestani, M.; Caruso, D.; De Fabiani, E.; Mitro, N. “The Loss of Golden Touch”: Mitochondria-Organelle Interactions, Metabolism, and Cancer. Cells 2020, 9, 2519. [Google Scholar] [CrossRef]
  203. Wu, Z.; Puigserver, P.; Andersson, U.; Zhang, C.; Adelmant, G.; Mootha, V.; Troy, A.; Cinti, S.; Lowell, B.; Scarpulla, R.C.; et al. Mechanisms Controlling Mitochondrial Biogenesis and Respiration through the Thermogenic Coactivator PGC-1. Cell 1999, 98, 115–124. [Google Scholar] [CrossRef] [PubMed]
  204. Dabrowska, A.; Venero, J.L.; Iwasawa, R.; Hankir, M.-K.; Rahman, S.; Boobis, A.; Hajji, N. PGC-1α controls mitochondrial biogenesis and dynamics in lead-induced neurotoxicity. Aging 2015, 7, 629–647. [Google Scholar] [CrossRef]
  205. Piyarathna, D.W.B.; Balasubramanian, A.; Arnold, J.M.; Lloyd, S.M.; Karanam, B.; Castro, P.; Ittmann, M.M.; Putluri, N.; Navone, N.; Jones, J.A.; et al. ERR1- and PGC1α-associated mitochondrial alterations correlate with pan-cancer disparity in African Americans. J. Clin. Investig. 2019, 129, 2351–2356. [Google Scholar] [CrossRef]
  206. Shiota, M.; Yokomizo, A.; Tada, Y.; Inokuchi, J.; Tatsugami, K.; Kuroiwa, K.; Uchiumi, T.; Fujimoto, N.; Seki, N.; Naito, S. Peroxisome Proliferator-Activated Receptor γ Coactivator-1α Interacts with the Androgen Receptor (AR) and Promotes Prostate Cancer Cell Growth by Activating the AR. Mol. Endocrinol. 2010, 24, 114–127. [Google Scholar] [CrossRef]
  207. Li, J.; Li, Y.; Chen, L.; Yu, B.; Xue, Y.; Guo, R.; Su, J.; Liu, Y.; Sun, L. p53/PGC-1α-mediated mitochondrial dysfunction promotes PC3 prostate cancer cell apoptosis. Mol. Med. Rep. 2020, 22, 155–164. [Google Scholar] [CrossRef]
  208. Burch, T.C.; Rhim, J.S.; Nyalwidhe, J.O. Mitochondria Biogenesis and Bioenergetics Gene Profiles in Isogenic Prostate Cells with Different Malignant Phenotypes. BioMed Res. Int. 2016, 2016, 1785201. [Google Scholar] [CrossRef]
  209. Kaddour-Djebbar, I.; Choudhary, V.; Brooks, C.; Ghazaly, T.; Lakshmikanthan, V.; Dong, Z.; Kumar, M.V. Specific mitochondrial calcium overload induces mitochondrial fission in prostate cancer cells. Int. J. Oncol. 2010, 36, 1437–1444. [Google Scholar] [CrossRef]
  210. Philley, J.V.; Kannan, A.; Qin, W.; Sauter, E.R.; Ikebe, M.; Hertweck, K.L.; Troyer, D.A.; Semmes, O.J.; Dasgupta, S. Complex-I Alteration and Enhanced Mitochondrial Fusion Are Associated With Prostate Cancer Progression. J. Cell. Physiol. 2016, 231, 1364–1374. [Google Scholar] [CrossRef]
  211. Civenni, G.; Bosotti, R.; Timpanaro, A.; Vàzquez, R.; Merulla, J.; Pandit, S.; Rossi, S.; Albino, D.; Allegrini, S.; Mitra, A.; et al. Epigenetic Control of Mitochondrial Fission Enables Self-Renewal of Stem-like Tumor Cells in Human Prostate Cancer. Cell Metab. 2019, 30, 303–318 e306. [Google Scholar] [CrossRef] [PubMed]
  212. Civenni, G.; Carbone, G.M.; Catapano, C.V. Mitochondrial fission promotes self-renewal and tumorigenic potential in prostate cancer. Mol. Cell. Oncol. 2019, 6, e1644598. [Google Scholar] [CrossRef]
  213. Yan, C.; Li, T.-S. Dual Role of Mitophagy in Cancer Drug Resistance. Anticancer. Res. 2018, 38, 617–621. [Google Scholar] [CrossRef] [PubMed]
  214. Yang, X.; Pan, W.; Xu, G.; Chen, L. Mitophagy: A crucial modulator in the pathogenesis of chronic diseases. Clin. Chim. Acta 2020, 502, 245–254. [Google Scholar] [CrossRef] [PubMed]
  215. Panigrahi, D.P.; Praharaj, P.P.; Bhol, C.S.; Mahapatra, K.K.; Patra, S.; Behera, B.P.; Mishra, S.R.; Bhutia, S.K. The emerging, multifaceted role of mitophagy in cancer and cancer therapeutics. Semin. Cancer Biol. 2020, 66, 45–58. [Google Scholar] [CrossRef]
  216. Dai, K.; Radin, D.P.; Leonardi, D. Deciphering the dual role and prognostic potential of PINK1 across cancer types. Neural Regen. Res. 2021, 16, 659–665. [Google Scholar] [CrossRef]
  217. Han, J.; Zhang, J.; Zhang, W.; Zhang, D.; Li, Y.; Zhang, J.; Zhang, Y.; Diao, T.; Cui, L.; Li, W.; et al. Abiraterone and MDV3100 inhibits the proliferation and promotes the apoptosis of prostate cancer cells through mitophagy. Cancer Cell Int. 2019, 19, 332. [Google Scholar] [CrossRef]
  218. Katreddy, R.R.; Bollu, L.R.; Su, F.; Xian, N.; Srivastava, S.; Thomas, R.; Dai, Y.; Wu, B.; Xu, Y.; Rea, M.A.; et al. Targeted reduction of the EGFR protein, but not inhibition of its kinase activity, induces mitophagy and death of cancer cells through activation of mTORC2 and Akt. Oncogenesis 2018, 7, 5. [Google Scholar] [CrossRef]
  219. Zou, J.; Yue, F.; Li, W.; Song, K.; Jiang, X.; Yi, J.; Liu, L. Autophagy Inhibitor LRPPRC Suppresses Mitophagy through Interaction with Mitophagy Initiator Parkin. PLoS ONE 2014, 9, e94903. [Google Scholar] [CrossRef]
  220. Balvan, J.; Gumulec, J.; Raudenska, M.; Krizova, A.; Štěpka, P.; Babula, P.; Kizek, R.; Adam, V.; Masarik, M. Oxidative Stress Resistance in Metastatic Prostate Cancer: Renewal by Self-Eating. PLoS ONE 2015, 10, e0145016. [Google Scholar] [CrossRef]
  221. Qu, F.; Gu, Y.; Xue, M.; He, M.; Zhou, F.; Wang, G.; Peng, Y. Impact of therapy on cancer metabolism in high-risk localized prostate cancer treated with neoadjuvant docetaxel and androgen deprivation therapy. Prostate 2021, 81, 560–571. [Google Scholar] [CrossRef]
  222. Hatoum, D.; McGowan, E.M. Recent Advances in the Use of Metformin: Can Treating Diabetes Prevent Breast Cancer? BioMed Res. Int. 2015, 2015, 548436. [Google Scholar] [CrossRef] [Green Version]
  223. Maruthur, N.M.; Tseng, E.; Hutfless, S.; Wilson, L.M.; Suarez-Cuervo, C.; Berger, Z.; Chu, Y.; Iyoha, E.; Segal, J.B.; Bolen, S. Diabetes Medications as Monotherapy or Metformin-Based Combination Therapy for Type 2 Diabetes: A Systematic Review and Meta-analysis. Ann. Intern. Med. 2016, 164, 740–751. [Google Scholar] [CrossRef] [PubMed]
  224. Triggle, C.R.; Ding, H. Metformin is not just an antihyperglycaemic drug but also has protective effects on the vascular endothelium. Acta Physiol. 2017, 219, 138–151. [Google Scholar] [CrossRef] [PubMed]
  225. Ben Sahra, I.; Laurent, K.; Loubat, A.; Giorgetti-Peraldi, S.; Colosetti, P.; Auberger, P.; Tanti, J.F.; Le Marchand-Brustel, Y.; Bost, F. The antidiabetic drug metformin exerts an antitumoral effect in vitro and in vivo through a decrease of cyclin D1 level. Oncogene 2008, 27, 3576–3586. [Google Scholar] [CrossRef]
  226. Del Barco, S.; Vazquez-Martin, A.; Cufí, S.; Oliveras-Ferraros, C.; Bosch-Barrera, J.; Joven, J.; Martín-Castillo, B.; Menendez, J.A. Metformin: Multi-faceted protection against cancer. Oncotarget 2011, 2, 896–917. [Google Scholar] [CrossRef] [PubMed]
  227. Pollak, M.N. Investigating Metformin for Cancer Prevention and Treatment: The End of the Beginning. Cancer Discov. 2012, 2, 778–790. [Google Scholar] [CrossRef]
  228. 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. [Google Scholar] [CrossRef]
  229. Zaidi, S.; Gandhi, J.; Joshi, G.; Smith, N.L.; Khan, S.A. The anticancer potential of metformin on prostate cancer. Prostate Cancer Prostatic Dis. 2019, 22, 351–361. [Google Scholar] [CrossRef]
  230. DeCensi, A.; Puntoni, M.; Goodwin, P.; Cazzaniga, M.; Gennari, A.; Bonanni, B.; Gandini, S. Metformin and Cancer Risk in Diabetic Patients: A Systematic Review and Meta-analysis. Cancer Prev. Res. 2010, 3, 1451–1461. [Google Scholar] [CrossRef]
  231. Noto, H.; Goto, A.; Tsujimoto, T.; Noda, M. Cancer Risk in Diabetic Patients Treated with Metformin: A Systematic Review and Meta-analysis. PLoS ONE 2012, 7, e33411. [Google Scholar] [CrossRef]
  232. León-González, A.J.; Jiménez-Vacas, J.M.; Fuentes-Fayos, A.C.; Sarmento-Cabral, A.; Herrera-Martínez, A.D.; Gahete, M.D.; Luque, R.M. Role of metformin and other metabolic drugs in the prevention and therapy of endocrine-related cancers. Curr. Opin. Pharmacol. 2021, 60, 17–26. [Google Scholar] [CrossRef]
  233. Li, H.-X.; Gao, J.-M.; Liang, J.-Q.; Xi, J.-M.; Fu, M.; Wu, Y.-J. Vitamin D3potentiates the growth inhibitory effects of metformin in DU145 human prostate cancer cells mediated by AMPK/mTOR signalling pathway. Clin. Exp. Pharmacol. Physiol. 2015, 42, 711–717. [Google Scholar] [CrossRef] [PubMed]
  234. Chen, X.; Li, C.; He, T.; Mao, J.; Li, C.; Lyu, J.; Meng, Q.H. Metformin inhibits prostate cancer cell proliferation, migration, and tumor growth through upregulation of PEDF expression. Cancer Biol. Ther. 2016, 17, 507–514. [Google Scholar] [CrossRef] [PubMed]
  235. Kang, J.; Li, C.; Gao, X.; Liu, Z.; Chen, C.; Luo, D. Metformin inhibits tumor growth and affects intestinal flora in diabetic tumor-bearing mice. Eur. J. Pharmacol. 2021, 912, 174605. [Google Scholar] [CrossRef]
  236. Morale, M.G.; Tamura, R.E.; Rubio, I.G.S. Metformin and Cancer Hallmarks: Molecular Mechanisms in Thyroid, Prostate and Head and Neck Cancer Models. Biomolecules 2022, 12, 357. [Google Scholar] [CrossRef] [PubMed]
  237. Demir, Ü.; Köhler, A.; Schneider, R.; Schweiger, S.; Klocker, H. Metformin anti-tumor effect via disruption of the MID1 translational regulator complex and AR downregulation in prostate cancer cells. BMC Cancer 2014, 14, 52. [Google Scholar] [CrossRef] [PubMed]
  238. Kato, H.; Sekine, Y.; Furuya, Y.; Miyazawa, Y.; Koike, H.; Suzuki, K. Metformin inhibits the proliferation of human prostate cancer PC-3 cells via the downregulation of insulin-like growth factor 1 receptor. Biochem. Biophys. Res. Commun. 2015, 461, 115–121. [Google Scholar] [CrossRef] [PubMed]
  239. Wang, Y.; Liu, G.; Tong, D.; Parmar, H.; Hasenmayer, D.; Yuan, W.; Zhang, D.; Jiang, J. Metformin represses androgen-dependent and androgen-independent prostate cancers by targeting androgen receptor. Prostate 2015, 75, 1187–1196. [Google Scholar] [CrossRef]
  240. Sun, S.; Gong, F.; Liu, P.; Miao, Q. Metformin combined with quercetin synergistically repressed prostate cancer cells via inhibition of VEGF/PI3K/Akt signaling pathway. Gene 2018, 664, 50–57. [Google Scholar] [CrossRef]
  241. Yang, B.; Damodaran, S.; Khemees, T.A.; Filon, M.J.; Schultz, A.; Gawdzik, J.; Etheridge, T.; Malin, D.; Richards, K.A.; Cryns, V.L.; et al. Synthetic Lethal Metabolic Targeting of Androgen-Deprived Prostate Cancer Cells with Metformin. Mol. Cancer Ther. 2020, 19, 2278–2287. [Google Scholar] [CrossRef] [PubMed]
  242. Kim, J.H.; Ofori, S.; Mertens, R.T.; Parkin, S.; Awuah, S.G. Water-Soluble Gold(III)–Metformin Complex Alters Mitochondrial Bioenergetics in Breast Cancer Cells. Chemmedchem 2021, 16, 3222–3230. [Google Scholar] [CrossRef]
  243. Zannella, V.E.; Dal Pra, A.; Muaddi, H.; McKee, T.D.; Stapleton, S.; Sykes, J.; Glicksman, R.; Chaib, S.; Zamiara, P.; Milosevic, M.; et al. Reprogramming Metabolism with Metformin Improves Tumor Oxygenation and Radiotherapy Response. Clin. Cancer Res. 2013, 19, 6741–6750. [Google Scholar] [CrossRef] [PubMed]
  244. Ahn, H.; Lee, Y.; Koo, K. Current Status and Application of Metformin for Prostate Cancer: A Comprehensive Review. Int. J. Mol. Sci. 2020, 21, 8540. [Google Scholar] [CrossRef] [PubMed]
  245. Jiménez-Vacas, J.M.; Herrero-Aguayo, V.; Montero-Hidalgo, A.J.; Sáez-Martínez, P.; Gómez-Gómez, E.; León-González, A.J.; Fuentes-Fayos, A.C.; Yubero-Serrano, E.M.; Requena-Tapia, M.J.; López, M.; et al. Clinical, Cellular, and Molecular Evidence of the Additive Antitumor Effects of Biguanides and Statins in Prostate Cancer. J. Clin. Endocrinol. Metab. 2021, 106, e696–e710. [Google Scholar] [CrossRef] [PubMed]
  246. Loubière, C.; Goiran, T.; Laurent, K.; Djabari, Z.; Tanti, J.-F.; Bost, F. Metformin-induced energy deficiency leads to the inhibition of lipogenesis in prostate cancer cells. Oncotarget 2015, 6, 15652–15661. [Google Scholar] [CrossRef]
  247. Zhang, J.; Shen, C.; Wang, L.; Ma, Q.; Xia, P.; Qi, M.; Yang, M.; Han, B. Metformin inhibits epithelial–mesenchymal transition in prostate cancer cells: Involvement of the tumor suppressor miR30a and its target gene SOX4. Biochem. Biophys. Res. Commun. 2014, 452, 746–752. [Google Scholar] [CrossRef] [PubMed]
  248. Tsao, T.; Beretov, J.; Ni, J.; Bai, X.; Bucci, J.; Graham, P.; Li, Y. Cancer stem cells in prostate cancer radioresistance. Cancer Lett. 2019, 465, 94–104. [Google Scholar] [CrossRef]
  249. Villani, L.A.; Smith, B.K.; Marcinko, K.; Ford, R.J.; Broadfield, L.A.; Green, A.E.; Houde, V.P.; Muti, P.; Tsakiridis, T.; Steinberg, G.R. The diabetes medication Canagliflozin reduces cancer cell proliferation by inhibiting mitochondrial complex-I supported respiration. Mol. Metab. 2016, 5, 1048–1056. [Google Scholar] [CrossRef]
  250. Ben Sahra, I.; Laurent, K.; Giuliano, S.; Larbret, F.; Ponzio, G.; Gounon, P.; Le Marchand-Brustel, Y.; Giorgetti-Peraldi, S.; Cormont, M.; Bertolotto, C.; et al. Targeting Cancer Cell Metabolism: The Combination of Metformin and 2-Deoxyglucose Induces p53-Dependent Apoptosis in Prostate Cancer Cells. Cancer Res 2010, 70, 2465–2475. [Google Scholar] [CrossRef]
  251. Tsakiridis, E.E.; Broadfield, L.; Marcinko, K.; Biziotis, O.-D.; Ali, A.; Mekhaeil, B.; Ahmadi, E.; Singh, K.; Mesci, A.; Zacharidis, P.G.; et al. Combined metformin-salicylate treatment provides improved anti-tumor activity and enhanced radiotherapy response in prostate cancer; drug synergy at clinically relevant doses. Transl. Oncol. 2021, 14, 101209. [Google Scholar] [CrossRef] [PubMed]
  252. Joentausta, R.M.; Rannikko, A.; Murtola, T.J. Prostate Cancer–specific Survival After Radical Prostatectomy Is Improved Among Metformin Users but Not Among Other Antidiabetic Drug Users. Eur. Urol. Open Sci. 2021, 34, 86–93. [Google Scholar] [CrossRef] [PubMed]
  253. Khan, S.; Chang, S.-H.; Hicks, V.; Wang, M.; Grubb, R.L.; Drake, B.F., 3rd. Improved survival with post-diagnostic metformin and statin use in a racially diverse cohort of US Veterans with advanced prostate cancer. Prostate Cancer Prostatic Dis. 2021, 25, 707–712. [Google Scholar] [CrossRef]
  254. Yao, X.; Liu, H.; Xu, H. The Impact of Metformin Use with Survival Outcomes in Urologic Cancers: A Systematic Review and Meta-Analysis. BioMed Res. Int. 2021, 2021, 5311828. [Google Scholar] [CrossRef]
  255. Aboelnaga, E.M.; Aboelnaga, M.M.; Elkalla, H.M. Metformin addition to androgen deprivation therapy effect on cancer prostate patients with type 2 diabetes. Diabetes Metab. Syndr. 2021, 15, 102251. [Google Scholar] [CrossRef]
  256. Alghandour, R.; Ebrahim, M.A.; Elshal, A.M.; Ghobrial, F.; Elzaafarany, M.; Elbaiomy, M.A. Repurposing metformin as anticancer drug: Randomized controlled trial in advanced prostate cancer (MANSMED). Urol. Oncol. 2021, 39, 831.e1–831.e10. [Google Scholar] [CrossRef]
  257. Cui, H.; Wang, Y.; Yang, S.; He, G.; Jiang, Z.; Gang, X.; Wang, G. Antidiabetic medications and the risk of prostate cancer in patients with diabetes mellitus: A systematic review and meta-analysis. Pharmacol. Res. 2022, 177, 106094. [Google Scholar] [CrossRef]
  258. Bilusic, M.; Toney, N.J.; Donahue, R.N.; Wroblewski, S.; Zibelman, M.; Ghatalia, P.; Ross, E.A.; Karzai, F.; Madan, R.A.; Dahut, W.L.; et al. A randomized phase 2 study of bicalutamide with or without metformin for biochemical recurrence in overweight or obese prostate cancer patients (BIMET-1). Prostate Cancer Prostatic Dis. 2022, 25, 735–740. [Google Scholar] [CrossRef]
  259. Martin, M.P.; Borchiellini, D.; Thamphya, B.; Guillot, A.; Paoli, J.-B.; Besson, D.; Hilgers, W.; Priou, F.; El Kouri, C.; Hoch, B.; et al. TAXOMET: A French Prospective Multicentric Randomized Phase II Study of Docetaxel Plus Metformin Versus Docetaxel Plus Placebo in Metastatic Castration-Resistant Prostate Cancer. Clin. Genitourin. Cancer 2021, 19, 501–509. [Google Scholar] [CrossRef]
  260. Mitrakas, A.G.; Kalamida, D.; Koukourakis, M.I. Effect of mitochondrial metabolism-interfering agents on cancer cell mitochondrial function and radio/chemosensitivity. Anti-Cancer Drugs 2014, 25, 1182–1191. [Google Scholar] [CrossRef]
  261. Wang, T.; Ma, F.; Qian, H.-L. Defueling the cancer: ATP synthase as an emerging target in cancer therapy. Mol. Ther.—Oncolytics 2021, 23, 82–95. [Google Scholar] [CrossRef] [PubMed]
  262. Elbehairi, S.E.I.; Alfaifi, M.Y.; Shati, A.A.; AlShehri, M.A.; Elshaarawy, R.F.; Hafez, H.S. Role of Pd(II)–chitooligosaccharides–Gboxin analog in oxidative phosphorylation inhibition and energy depletion: Targeting mitochondrial dynamics. Chem. Biol. Drug Des. 2020, 96, 1148–1161. [Google Scholar] [CrossRef]
  263. Gao, Y.; Shang, Q.; Li, W.; Guo, W.; Stojadinovic, A.; Mannion, C.; Man, Y.-G.; Chen, T. Antibiotics for cancer treatment: A double-edged sword. J. Cancer 2020, 11, 5135–5149. [Google Scholar] [CrossRef] [PubMed]
  264. 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. [Google Scholar] [CrossRef] [PubMed]
  265. Ozsvari, B.; Fiorillo, M.; Bonuccelli, G.; Cappello, A.R.; Frattaruolo, L.; Sotgia, F.; Trowbridge, R.; Foster, R.; Lisanti, M.P. Mitoriboscins: Mitochondrial-based therapeutics targeting cancer stem cells (CSCs), bacteria and pathogenic yeast. Oncotarget 2017, 8, 67457–67472. [Google Scholar] [CrossRef]
  266. Ozsvari, B.; Bonuccelli, G.; Sanchez-Alvarez, R.; Foster, R.; Sotgia, F.; Lisanti, M.P. Targeting flavin-containing enzymes eliminates cancer stem cells (CSCs), by inhibiting mitochondrial respiration: Vitamin B2 (Riboflavin) in cancer therapy. Aging 2017, 9, 2610–2628. [Google Scholar] [CrossRef]
  267. Fiorillo, M.; Tóth, F.; Sotgia, F.; Lisanti, M.P. Doxycycline, Azithromycin and Vitamin C (DAV): A potent combination therapy for targeting mitochondria and eradicating cancer stem cells (CSCs). Aging 2019, 11, 2202–2216. [Google Scholar] [CrossRef]
  268. Wanyan, Y.; Xu, X.; Liu, K.; Zhang, H.; Zhen, J.; Zhang, R.; Wen, J.; Liu, P.; Chen, Y. 2-Deoxy-d-glucose Promotes Buforin IIb-Induced Cytotoxicity in Prostate Cancer DU145 Cells and Xenograft Tumors. Molecules 2020, 25, 5778. [Google Scholar] [CrossRef]
  269. Shahidi, F.; De Camargo, A.C. Tocopherols and Tocotrienols in Common and Emerging Dietary Sources: Occurrence, Applications, and Health Benefits. Int. J. Mol. Sci. 2016, 17, 1745. [Google Scholar] [CrossRef]
  270. Beretta, G.; Gelmini, F.; Fontana, F.; Moretti, R.M.; Marelli, M.M.; Limonta, P. Semi-preparative HPLC purification of δ-tocotrienol (δ-T3) from Elaeis guineensis Jacq. and Bixa orellana L. and evaluation of its in vitro anticancer activity in human A375 melanoma cells. Nat. Prod. Res. 2018, 32, 1130–1135. [Google Scholar] [CrossRef]
  271. Chin, K.-Y.; Pang, K.-L.; Soelaiman, I.-N. Tocotrienol and Its Role in Chronic Diseases. Adv. Exp. Med. Biol. 2016, 928, 97–130. [Google Scholar] [CrossRef]
  272. Comitato, R.; Ambra, R.; Virgili, F. Tocotrienols: A Family of Molecules with Specific Biological Activities. Antioxidants 2017, 6, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  273. Chin, K.-Y.; Tay, S.S. A Review on the Relationship between Tocotrienol and Alzheimer Disease. Nutrients 2018, 10, 881. [Google Scholar] [CrossRef] [PubMed]
  274. Ramanathan, N.; Tan, E.; Loh, L.J.; Soh, B.S.; Yap, W.N. Tocotrienol is a cardioprotective agent against ageing-associated cardiovascular disease and its associated morbidities. Nutr. Metab. 2018, 15, 6. [Google Scholar] [CrossRef]
  275. Casati, L.; Pagani, F.; Limonta, P.; Vanetti, C.; Stancari, G.; Sibilia, V. Beneficial effects of δ-tocotrienol against oxidative stress in osteoblastic cells: Studies on the mechanisms of action. Eur. J. Nutr. 2019, 59, 1975–1987. [Google Scholar] [CrossRef]
  276. Wong, S.K.; Kamisah, Y.; Mohamed, N.; Muhammad, N.; Masbah, N.; Fahami, N.A.M.; Mohamed, I.N.; Shuid, A.N.; Saad, Q.M.; Abdullah, A.; et al. Potential Role of Tocotrienols on Non-Communicable Diseases: A Review of Current Evidence. Nutrients 2020, 12, 259. [Google Scholar] [CrossRef] [PubMed]
  277. Naomi, R.; Shafie, N.H.; Kaniappan, P.; Bahari, H. An Interactive Review on the Role of Tocotrienols in the Neurodegenerative Disorders. Front. Nutr. 2021, 8, 754086. [Google Scholar] [CrossRef]
  278. Montagnani Marelli, M.; Marzagalli, M.; Moretti, R.M.; Beretta, G.; Casati, L.; Comitato, R.; Gravina, G.L.; Festuccia, C.; Limonta, P. Vitamin E delta-tocotrienol triggers endoplasmic reticulum stress-mediated apoptosis in human melanoma cells. Sci. Rep. 2016, 6, 30502. [Google Scholar] [CrossRef]
  279. Marzagalli, M.; Moretti, R.M.; Messi, E.; Marelli, M.M.; Fontana, F.; Anastasia, A.; Bani, M.R.; Beretta, G.; Limonta, P. Targeting melanoma stem cells with the Vitamin E derivative δ-tocotrienol. Sci. Rep. 2018, 8, 587. [Google Scholar] [CrossRef] [PubMed]
  280. Sailo, B.L.; Banik, K.; Padmavathi, G.; Javadi, M.; Bordoloi, D.; Kunnumakkara, A.B. Tocotrienols: The promising analogues of vitamin E for cancer therapeutics. Pharmacol. Res. 2018, 130, 259–272. [Google Scholar] [CrossRef]
  281. Aggarwal, V.; Kashyap, D.; Sak, K.; Tuli, H.S.; Jain, A.; Chaudhary, A.; Garg, V.K.; Sethi, G.; Yerer, M.B. Molecular Mechanisms of Action of Tocotrienols in Cancer: Recent Trends and Advancements. Int. J. Mol. Sci. 2019, 20, 656. [Google Scholar] [CrossRef]
  282. Fontana, F.; Raimondi, M.; Marzagalli, M.; Moretti, R.M.; Marelli, M.M.; Limonta, P. Tocotrienols and Cancer: From the State of the Art to Promising Novel Patents. Recent Patents Anti-Cancer Drug Discov. 2019, 14, 5–18. [Google Scholar] [CrossRef]
  283. Limonta, P.; Moretti, R.M.; Marzagalli, M.; Fontana, F.; Raimondi, M.; Marelli, M.M. Role of Endoplasmic Reticulum Stress in the Anticancer Activity of Natural Compounds. Int. J. Mol. Sci. 2019, 20, 961. [Google Scholar] [CrossRef] [PubMed]
  284. Montagnani Marelli, M.; Marzagalli, M.; Fontana, F.; Raimondi, M.; Moretti, R.M.; Limonta, P. Anticancer properties of tocotrienols: A review of cellular mechanisms and molecular targets. J. Cell. Physiol. 2019, 234, 1147–1164. [Google Scholar] [CrossRef] [PubMed]
  285. Fontana, F.; Marzagalli, M.; Raimondi, M.; Zuco, V.; Zaffaroni, N.; Limonta, P. δ-Tocotrienol sensitizes and re-sensitizes ovarian cancer cells to cisplatin via induction of G1 phase cell cycle arrest and ROS/MAPK-mediated apoptosis. Cell Prolif. 2021, 54, e13111. [Google Scholar] [CrossRef]
  286. Theodosis-Nobelos, P.; Papagiouvannis, G.; Rekka, E.A. A Review on Vitamin E Natural Analogues and on the Design of Synthetic Vitamin E Derivatives as Cytoprotective Agents. Mini-Reviews Med. Chem. 2021, 21, 10–22. [Google Scholar] [CrossRef] [PubMed]
  287. Trujillo, M.; Kharbanda, A.; Corley, C.; Simmons, P.; Allen, A.R. Tocotrienols as an Anti-Breast Cancer Agent. Antioxidants 2021, 10, 1383. [Google Scholar] [CrossRef] [PubMed]
  288. Fontana, F.; Moretti, R.M.; Raimondi, M.; Marzagalli, M.; Beretta, G.; Procacci, P.; Sartori, P.; Marelli, M.M.; Limonta, P. δ-Tocotrienol induces apoptosis, involving endoplasmic reticulum stress and autophagy, and paraptosis in prostate cancer cells. Cell Prolif. 2019, 52, e12576. [Google Scholar] [CrossRef]
  289. Fontana, F.; Anselmi, M.; Limonta, P. Exploiting the Metabolic Consequences of PTEN Loss and Akt/Hexokinase 2 Hyperactivation in Prostate Cancer: A New Role for δ-Tocotrienol. Int. J. Mol. Sci. 2022, 23, 5269. [Google Scholar] [CrossRef]
  290. Ossikbayeva, S.; Khanin, M.; Sharoni, Y.; Trachtenberg, A.; Tuleukhanov, S.; Sensenig, R.; Rom, S.; Danilenko, M.; Orynbayeva, Z. Curcumin and Carnosic Acid Cooperate to Inhibit Proliferation and Alter Mitochondrial Function of Metastatic Prostate Cancer Cells. Antioxidants 2021, 10, 1591. [Google Scholar] [CrossRef]
  291. Chen, Q.-H. Curcumin-based anti-prostate cancer agents. Anti-Cancer Agents Med. Chem. 2015, 15, 138–156. [Google Scholar] [CrossRef] [PubMed]
  292. García-Zepeda, S.P.; García-Villa, E.; Díaz-Chávez, J.; Hernández-Pando, R.; Gariglio, P. Resveratrol induces cell death in cervical cancer cells through apoptosis and autophagy. Eur. J. Cancer Prev. 2013, 22, 577–584. [Google Scholar] [CrossRef] [PubMed]
  293. Rodriguez-Enriquez, S.; Pacheco-Velazquez, S.C.; Marin-Hernandez, A.; Gallardo-Perez, J.C.; Robledo-Cadena, D.X.; Hernandez-Resendiz, I.; Garcia-Garcia, J.D.; Belmont-Diaz, J.; Lopez-Marure, R.; Hernandez-Esquivel, L.; et al. Resveratrol inhibits cancer cell proliferation by impairing oxidative phosphorylation and inducing oxidative stress. Toxicol. Appl. Pharmacol. 2019, 370, 65–77. [Google Scholar] [CrossRef] [PubMed]
  294. Liu, J.; Wu, N.; Ma, L.; Liu, M.; Liu, G.; Zhang, Y.; Lin, X. Oleanolic Acid Suppresses Aerobic Glycolysis in Cancer Cells by Switching Pyruvate Kinase Type M Isoforms. PLoS ONE 2014, 9, e91606. [Google Scholar] [CrossRef]
  295. Lodi, A.; Saha, A.; Lu, X.; Wang, B.; Sentandreu, E.; Collins, M.; Kolonin, M.G.; DiGiovanni, J.; Tiziani, S. Erratum: Combinatorial treatment with natural compounds in prostate cancer inhibits prostate tumor growth and leads to key modulations of cancer cell metabolism. NPJ Precis. Oncol. 2017, 1, 30. [Google Scholar] [CrossRef] [PubMed]
  296. Liu, J.; Zheng, L.; Wu, N.; Ma, L.; Zhong, J.; Liu, G.; Lin, X. Oleanolic Acid Induces Metabolic Adaptation in Cancer Cells by Activating the AMP-Activated Protein Kinase Pathway. J. Agric. Food Chem. 2014, 62, 5528–5537. [Google Scholar] [CrossRef]
  297. Akhtar, N.; Syed, D.N.; Khan, M.I.; Adhami, V.M.; Mirza, B.; Mukhtar, H. The pentacyclic triterpenoid, plectranthoic acid, a novel activator of AMPK induces apoptotic death in prostate cancer cells. Oncotarget 2015, 7, 3819–3831. [Google Scholar] [CrossRef]
  298. Younis, T.; Khan, M.I.; Khan, M.R.; Rasul, A.; Majid, M.; Adhami, V.M.; Mukhtar, H. Nummularic acid, a triterpenoid, from the medicinal plant Fraxinus xanthoxyloides, induces energy crisis to suppress growth of prostate cancer cells. Mol. Carcinog. 2018, 57, 1267–1277. [Google Scholar] [CrossRef]
  299. Dyshlovoy, S.A.; Pelageev, D.N.; Hauschild, J.; Borisova, K.L.; Kaune, M.; Krisp, C.; Venz, S.; Sabutskii, Y.E.; Khmelevskaya, E.A.; Busenbender, T.; et al. Successful Targeting of the Warburg Effect in Prostate Cancer by Glucose-Conjugated 1,4-Naphthoquinones. Cancers 2019, 11, 1690. [Google Scholar] [CrossRef]
  300. Negrette-Guzmán, M.; Huerta-Yepez, S.; Vega, M.I.; León-Contreras, J.C.; Hernández-Pando, R.; Medina-Campos, O.N.; Rodríguez, E.; Tapia, E.; Pedraza-Chaverri, J. Sulforaphane induces differential modulation of mitochondrial biogenesis and dynamics in normal cells and tumor cells. Food Chem. Toxicol. 2017, 100, 90–102. [Google Scholar] [CrossRef]
  301. Xiao, D.; Powolny, A.A.; Moura, M.B.; Kelley, E.E.; Bommareddy, A.; Kim, S.-H.; Hahm, E.-R.; Normolle, D.; Van Houten, B.; Singh, S.V. Phenethyl Isothiocyanate Inhibits Oxidative Phosphorylation to Trigger Reactive Oxygen Species-mediated Death of Human Prostate Cancer Cells. J. Biol. Chem. 2010, 285, 26558–26569. [Google Scholar] [CrossRef] [PubMed]
  302. Kim, S.-H.; Kim, K.-Y.; Yu, S.-N.; Seo, Y.-K.; Chun, S.-S.; Yu, H.-S.; Ahn, S.-C. Silibinin induces mitochondrial NOX4-mediated endoplasmic reticulum stress response and its subsequent apoptosis. BMC Cancer 2016, 16, 452. [Google Scholar] [CrossRef] [Green Version]
  303. Wang, H.; Huwaimel, B.; Verma, K.; Miller, J.; Germain, T.M.; Kinarivala, N.; Pappas, D.; Brookes, P.S.; Trippier, P.C. Synthesis and Antineoplastic Evaluation of Mitochondrial Complex II (Succinate Dehydrogenase) Inhibitors Derived from Atpenin A5. Chemmedchem 2017, 12, 1033–1044. [Google Scholar] [CrossRef] [PubMed]
  304. Goldin, N.; Heyfets, A.; Reischer, D.; Flescher, E. Mitochondria-mediated ATP depletion by anti-cancer agents of the jasmonate family. J. Bioenerg. Biomembr. 2007, 39, 51–57. [Google Scholar] [CrossRef] [PubMed]
  305. Li, C.; He, C.; Xu, Y.; Xu, H.; Tang, Y.; Chavan, H.; Duan, S.; Artigues, A.; Forrest, M.L.; Krishnamurthy, P.; et al. Alternol eliminates excessive ATP production by disturbing Krebs cycle in prostate cancer. Prostate 2019, 79, 628–639. [Google Scholar] [CrossRef]
  306. Naguib, A.; Mathew, G.; Reczek, C.R.; Watrud, K.; Ambrico, A.; Herzka, T.; Salas, I.C.; Lee, M.F.; El-Amine, N.; Zheng, W.; et al. Mitochondrial Complex I Inhibitors Expose a Vulnerability for Selective Killing of Pten-Null Cells. Cell Rep. 2018, 23, 58–67. [Google Scholar] [CrossRef]
  307. Khan, M.A.; Gahlot, S.; Majumdar, S. Oxidative Stress Induced by Curcumin Promotes the Death of Cutaneous T-cell Lymphoma (HuT-78) by Disrupting the Function of Several Molecular Targets. Mol. Cancer Ther. 2012, 11, 1873–1883. [Google Scholar] [CrossRef]
  308. Larasati, Y.A.; Yoneda-Kato, N.; Nakamae, I.; Yokoyama, T.; Meiyanto, E.; Kato, J.-Y. Curcumin targets multiple enzymes involved in the ROS metabolic pathway to suppress tumor cell growth. Sci. Rep. 2018, 8, 2039. [Google Scholar] [CrossRef]
  309. Mortezaee, K.; Salehi, E.; Mirtavoos-Mahyari, H.; Motevaseli, E.; Najafi, M.; Farhood, B.; Rosengren, R.J.; Sahebkar, A. Mechanisms of apoptosis modulation by curcumin: Implications for cancer therapy. J. Cell. Physiol. 2019, 234, 12537–12550. [Google Scholar] [CrossRef]
  310. Jiang, Z.; Chen, K.; Cheng, L.; Yan, B.; Qian, W.; Cao, J.; Li, J.; Wu, E.; Ma, Q.; Yang, W. Resveratrol and cancer treatment: Updates. Ann. N. Y. Acad. Sci. 2017, 1403, 59–69. [Google Scholar] [CrossRef]
  311. Gal, R.; Deres, L.; Toth, K.; Halmosi, R.; Habon, T. The Effect of Resveratrol on the Cardiovascular System from Molecular Mechanisms to Clinical Results. Int. J. Mol. Sci. 2021, 22, 10152. [Google Scholar] [CrossRef]
  312. Zhang, L.-X.; Li, C.-X.; Kakar, M.U.; Khan, M.S.; Wu, P.-F.; Amir, R.M.; Dai, D.-F.; Naveed, M.; Li, Q.-Y.; Saeed, M.; et al. Resveratrol (RV): A pharmacological review and call for further research. Biomed. Pharmacother. 2021, 143, 112164. [Google Scholar] [CrossRef]
  313. Rudrapal, M.; Khairnar, S.J.; Khan, J.; Bin Dukhyil, A.; Ansari, M.A.; Alomary, M.N.; Alshabrmi, F.M.; Palai, S.; Deb, P.K.; Devi, R. Dietary Polyphenols and Their Role in Oxidative Stress-Induced Human Diseases: Insights Into Protective Effects, Antioxidant Potentials and Mechanism(s) of Action. Front. Pharmacol. 2022, 13, 806470. [Google Scholar] [CrossRef] [PubMed]
  314. Fonseca, J.; Moradi, F.; Maddalena, L.A.; Ferreira-Tollstadius, B.; Selim, S.; Stuart, J.A. Resveratrol integrates metabolic and growth effects in PC3 prostate cancer cells-involvement of prolyl hydroxylase and hypoxia inducible factor-1. Oncol. Lett. 2019, 17, 697–705. [Google Scholar] [CrossRef] [PubMed]
  315. Liby, K.; Yore, M.M.; Sporn, M.B. Triterpenoids and rexinoids as multifunctional agents for the prevention and treatment of cancer. Nat. Rev. Cancer 2007, 7, 357–369. [Google Scholar] [CrossRef]
  316. Banno, N.; Akihisa, T.; Yasukawa, K.; Tokuda, H.; Tabata, K.; Nakamura, Y.; Nishimura, R.; Kimura, Y.; Suzuki, T. Anti-inflammatory activities of the triterpene acids from the resin of Boswellia carteri. J. Ethnopharmacol. 2006, 107, 249–253. [Google Scholar] [CrossRef] [PubMed]
  317. Dembitsky, V.M. In Silico Prediction of Steroids and Triterpenoids as Potential Regulators of Lipid Metabolism. Mar. Drugs 2021, 19, 650. [Google Scholar] [CrossRef] [PubMed]
  318. Bishayee, A.; Ahmed, S.; Brankov, N.; Perloff, M. Triterpenoids as potential agents for the chemoprevention and therapy of breast cancer. Front. Biosci. 2011, 16, 980–996. [Google Scholar] [CrossRef]
  319. Shanmugam, M.K.; Nguyen, A.H.; Kumar, A.P.; Tan, B.K.; Sethi, G. Targeted inhibition of tumor proliferation, survival, and metastasis by pentacyclic triterpenoids: Potential role in prevention and therapy of cancer. Cancer Lett. 2012, 320, 158–170. [Google Scholar] [CrossRef]
  320. Ghante, M.H.; Jamkhande, P.G. Role of Pentacyclic Triterpenoids in Chemoprevention and Anticancer Treatment: An Overview on Targets and Underling Mechanisms. J. Pharmacopunct. 2019, 22, 55–67. [Google Scholar] [CrossRef]
  321. Li, S.; Kuo, H.-C.D.; Yin, R.; Wu, R.; Liu, X.; Wang, L.; Hudlikar, R.; Peter, R.M.; Kong, A.-N. Epigenetics/epigenomics of triterpenoids in cancer prevention and in health. Biochem. Pharmacol. 2020, 175, 113890. [Google Scholar] [CrossRef]
  322. Şoica, C.; Voicu, M.; Ghiulai, R.; Dehelean, C.; Racoviceanu, R.; Trandafirescu, C.; Roșca, O.-J.; Nistor, G.; Mioc, M.; Mioc, A. Natural Compounds in Sex Hormone-Dependent Cancers: The Role of Triterpenes as Therapeutic Agents. Front. Endocrinol. 2020, 11, 612396. [Google Scholar] [CrossRef] [PubMed]
  323. El-Baba, C.; Baassiri, A.; Kiriako, G.; Dia, B.; Fadlallah, S.; Moodad, S.; Darwiche, N. Terpenoids’ anti-cancer effects: Focus on autophagy. Apoptosis 2021, 26, 491–511. [Google Scholar] [CrossRef] [PubMed]
  324. Singla, R.K.; Sharma, P.; Dubey, A.K.; Gundamaraju, R.; Kumar, D.; Kumar, S.; Madaan, R.; Shri, R.; Tsagkaris, C.; Parisi, S.; et al. Natural Product-Based Studies for the Management of Castration-Resistant Prostate Cancer: Computational to Clinical Studies. Front. Pharmacol. 2021, 12, 732266. [Google Scholar] [CrossRef] [PubMed]
  325. Pandita, A.; Kumar, B.; Manvati, S.; Vaishnavi, S.; Singh, S.K.; Bamezai, R.N.K. Synergistic Combination of Gemcitabine and Dietary Molecule Induces Apoptosis in Pancreatic Cancer Cells and Down Regulates PKM2 Expression. PLoS ONE 2014, 9, e107154. [Google Scholar] [CrossRef] [PubMed]
  326. Amara, S.; Zheng, M.; Tiriveedhi, V. Oleanolic Acid Inhibits High Salt-Induced Exaggeration of Warburg-like Metabolism in Breast Cancer Cells. Cell Biochem. Biophys. 2016, 74, 427–434. [Google Scholar] [CrossRef]
  327. Guerra, A.R.; Duarte, M.F.; Duarte, I.F. Targeting Tumor Metabolism with Plant-Derived Natural Products: Emerging Trends in Cancer Therapy. J. Agric. Food Chem. 2018, 66, 10663–10685. [Google Scholar] [CrossRef]
  328. Lu, J.J.; Bao, J.L.; Wu, G.S.; Xu, W.S.; Huang, M.Q.; Chen, X.P.; Wang, Y.T. Quinones derived from plant secondary metabolites as anti-cancer agents. Anticancer. Agents Med. Chem. 2013, 13, 456–463. [Google Scholar]
  329. Negrette-Guzmán, M.; Huerta-Yepez, S.; Tapia, E.; Pedraza-Chaverri, J. Modulation of mitochondrial functions by the indirect antioxidant sulforaphane: A seemingly contradictory dual role and an integrative hypothesis. Free. Radic. Biol. Med. 2013, 65, 1078–1089. [Google Scholar] [CrossRef]
  330. Delmas, D.; Xiao, J.; Vejux, A.; Aires, V. Silymarin and Cancer: A Dual Strategy in Both in Chemoprevention and Chemosensitivity. Molecules 2020, 25, 2009. [Google Scholar] [CrossRef]
  331. Koltai, T.; Fliegel, L. Role of Silymarin in Cancer Treatment: Facts, Hypotheses, and Questions. J. Evid.-Based Integr. Med. 2022, 27, 2515690X211068826. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic overview of the mitochondrial metabolic rewiring occurring in prostate epithelial cells during the different stages of cancer progression. (a) Healthy prostate epithelial cells accumulate high levels of zinc (due to the overexpression of its transporter ZIP1), resulting in the inhibition of mitochondrial m-aconitase, the key enzyme responsible for the citrate-isocitrate conversion in the TCA cycle (1). This inhibition ultimately leads to the truncation of the TCA cycle and to citrate accumulation and secretion. As a result, normal prostate epithelial cells are characterized by an inefficient OXPHOS machinery (complexes I-V, COI-V) (2). (b) In the early stage of tumor progression, intracellular zinc levels are significantly reduced (due to a decreased expression of its transporter) (1); this leads to the reactivation of m-aconitase, restoring the citrate-isocitrate conversion, and consequently of the TCA cycle and OXPHOS metabolic pathways (2). (c) In the advanced stage of tumor progression, PCa cells exhibit the Warburg effect, an active aerobic glycolysis accompanied by high levels of lactate production (1). However, it is now well established that a high activity of the TCA cycle/OXPHOS pathways still persists in these cells, and it is even exacerbated in PCa stem cells as well as in drug-resistant PCa cells (2).
Figure 1. Schematic overview of the mitochondrial metabolic rewiring occurring in prostate epithelial cells during the different stages of cancer progression. (a) Healthy prostate epithelial cells accumulate high levels of zinc (due to the overexpression of its transporter ZIP1), resulting in the inhibition of mitochondrial m-aconitase, the key enzyme responsible for the citrate-isocitrate conversion in the TCA cycle (1). This inhibition ultimately leads to the truncation of the TCA cycle and to citrate accumulation and secretion. As a result, normal prostate epithelial cells are characterized by an inefficient OXPHOS machinery (complexes I-V, COI-V) (2). (b) In the early stage of tumor progression, intracellular zinc levels are significantly reduced (due to a decreased expression of its transporter) (1); this leads to the reactivation of m-aconitase, restoring the citrate-isocitrate conversion, and consequently of the TCA cycle and OXPHOS metabolic pathways (2). (c) In the advanced stage of tumor progression, PCa cells exhibit the Warburg effect, an active aerobic glycolysis accompanied by high levels of lactate production (1). However, it is now well established that a high activity of the TCA cycle/OXPHOS pathways still persists in these cells, and it is even exacerbated in PCa stem cells as well as in drug-resistant PCa cells (2).
Cancers 15 01192 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fontana, F.; Anselmi, M.; Limonta, P. Unraveling the Peculiar Features of Mitochondrial Metabolism and Dynamics in Prostate Cancer. Cancers 2023, 15, 1192. https://doi.org/10.3390/cancers15041192

AMA Style

Fontana F, Anselmi M, Limonta P. Unraveling the Peculiar Features of Mitochondrial Metabolism and Dynamics in Prostate Cancer. Cancers. 2023; 15(4):1192. https://doi.org/10.3390/cancers15041192

Chicago/Turabian Style

Fontana, Fabrizio, Martina Anselmi, and Patrizia Limonta. 2023. "Unraveling the Peculiar Features of Mitochondrial Metabolism and Dynamics in Prostate Cancer" Cancers 15, no. 4: 1192. https://doi.org/10.3390/cancers15041192

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

Article Metrics

Back to TopTop