Next Article in Journal
Towards a Molecular Classification of Sinonasal Carcinomas: Clinical Implications and Opportunities
Next Article in Special Issue
Autophagy, Oxidative Stress and Cancer Development
Previous Article in Journal
Taking Advantage of the Senescence-Promoting Effect of Olaparib after X-ray and Proton Irradiation Using the Senolytic Drug, ABT-263
Previous Article in Special Issue
The Role of Decorin Proteoglycan in Mitophagy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Chemotherapy Resistance: Role of Mitochondrial and Autophagic Components

1
College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Korea
2
Department of Convergence Medical Science and Biochemistry, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Korea
3
Department of Pathology, Ilsan Paik Hospital, College of Medicine, Inje University, Goyang 10380, Korea
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(6), 1462; https://doi.org/10.3390/cancers14061462
Submission received: 3 February 2022 / Revised: 10 March 2022 / Accepted: 10 March 2022 / Published: 12 March 2022
(This article belongs to the Special Issue Targeting Autophagy for Cancer Treatment)

Abstract

:

Simple Summary

Chemotherapy resistance is a common occurrence during cancer treatment that cancer researchers are attempting to understand and overcome. Mitochondria are a crucial intracellular signaling core that are becoming important determinants of numerous aspects of cancer genesis and progression, such as metabolic reprogramming, metastatic capability, and chemotherapeutic resistance. Mitophagy, or selective autophagy of mitochondria, can influence both the efficacy of tumor chemotherapy and the degree of drug resistance. Regardless of the fact that mitochondria are well-known for coordinating ATP synthesis from cellular respiration in cellular bioenergetics, little is known its mitophagy regulation in chemoresistance. Recent advancements in mitochondrial research, mitophagy regulatory mechanisms, and their implications for our understanding of chemotherapy resistance are discussed in this review.

Abstract

Cancer chemotherapy resistance is one of the most critical obstacles in cancer therapy. One of the well-known mechanisms of chemotherapy resistance is the change in the mitochondrial death pathways which occur when cells are under stressful situations, such as chemotherapy. Mitophagy, or mitochondrial selective autophagy, is critical for cell quality control because it can efficiently break down, remove, and recycle defective or damaged mitochondria. As cancer cells use mitophagy to rapidly sweep away damaged mitochondria in order to mediate their own drug resistance, it influences the efficacy of tumor chemotherapy as well as the degree of drug resistance. Yet despite the importance of mitochondria and mitophagy in chemotherapy resistance, little is known about the precise mechanisms involved. As a consequence, identifying potential therapeutic targets by analyzing the signal pathways that govern mitophagy has become a vital research goal. In this paper, we review recent advances in mitochondrial research, mitophagy control mechanisms, and their implications for our understanding of chemotherapy resistance.

1. Introduction

Chemotherapy resistance is a prevalent phenomenon during cancer treatment that cancer researchers are working to understand and conquer [1]. It alters cancer cell signals that are needed for cellular growth or immune response stimulation through a wide range of molecular pathways [2,3]. In our previous literature review and experimental study, we revealed that endoplasmic reticulum stress-dependent signaling contributes to chemotherapy resistance [4,5,6]. Mitochondria are a key intracellular signaling hub that are emerging as important determinants of several aspects of cancer’s development and progression, including metabolic reprogramming, acquisition of metastatic capability, and response to chemotherapeutic drugs. Mitochondrial dysfunction has been linked to the onset and progression of cancer. However, recent evidence suggests that pathogenic mutations or mitochondrial genome depletion may play a role in the development of chemoresistance in cancerous tumors. Despite the fact that mitochondria are well-known for their role in cellular bioenergetics, coordinating adenosine triphosphate (ATP) production from cellular respiration, little is known about their involvement and activity in chemoresistance [7].
Mitophagy, or selective autophagy of mitochondria, is important for quality control because it can efficiently degrade, remove, and recycle defective or damaged mitochondria [8]. In many cancers, severe mitophagy abnormalities are linked to total impairment of mitochondrial functioning, oncogenesis, and tumor development. In addition, mitophagy serves a dual role in cancer therapy, inducing cell death while also promoting cell survival [9]. According to a recent study, mitophagy influences both the efficacy of tumor chemotherapy and the degree of drug resistance, because cancer cells use mitophagy to rapidly sweep away damaged mitochondria in order to mediate their own drug resistance [10]. Although substantial progress has been made in defining mitophagy regulation in mitochondria and the role of mitophagy in chemoresistance, understanding how mitophagy acts as a chemo-resistant in cancer therapy remains an unsolved topic. Therefore, in this review, the role of autophagy and mitochondrial functionality in determining chemotherapeutic resistance will be discussed in light of recent research.

2. Potential Conventional Mitochondrial Targets for Chemotherapy

Mitochondria play a critical role in the generation of metabolic energy in eukaryotic cells, and they are responsible for the energy derived from the breakdown of carbohydrates and fatty acids, which is converted to ATP by the process of oxidative phosphorylation (OXPHOS) [11,12]. Most mitochondrial proteins are translated on free cytosolic ribosomes and imported into the organelle by specific targeting signals [13]. In addition, mitochondria are unique among the cytoplasmic organelles already discussed in that they contain their own deoxyribonucleic acid (DNA) called mtDNA, which encodes transfer ribonucleic acids (tRNAs), ribosomal ribonucleic acids (rRNAs), and some mitochondrial proteins [14]. The assembly of mitochondria thus involves proteins encoded by their own genomes and translated within the organelle, as well as proteins encoded by the nuclear genome and imported from the cytosol [15]. Since mitochondria have distinct structural and functional properties, therapies designed to influence the activity of this organelle can be targeted specifically for therapeutic benefit. The mitochondrion is engaged in a number of vital functions in eukaryotic cells, including energy metabolism and cell cycle regulation, which is critical in cancer physiology. The argument for developing anticancer therapies based on mitochondrial targeting is backed up by substantial evidence. Molecules found on or inside mitochondria are regarded as excellent pharmacological targets, and many attempts are being made to exploit these targets in order to develop tailored therapeutics for a variety of disorders, including cancer [16]. For example, mitochondria are recognized to play a significant part in the complicated apoptotic pathway, causing cell death by a variety of ways such as interrupting electron transport and energy metabolism, releasing or activating apoptotic proteins, and changing the cellular redox potential [17,18,19,20]. Therefore, mitochondrial drug-targeting approaches could create more opportunities for manipulating metabolic activity and allowing for the selective protection or destruction of cells for therapeutic benefit in a range of disorders (Figure 1).

2.1. Mitochondrial Calcium Ion (Ca2+)

Calcium ion (Ca2+) signaling is necessary for a range of cell processes; hence, it must be maintained in check to ensure proper cellular homeostasis [21,22]. Mitochondria are important organelles in the control of Ca2+ homeostasis in cells. Although Ca2+ regulates mitochondrial respiration, which is dependent on ATP synthesis, excess Ca2+ can cause mitochondrion-mediated death [23]. Mitochondrial Ca2+ signaling pathways are receiving more attention due to their diverse role in regulating cell survival and death, and the proteins implicated in those processes may represent alternate targets for cancer therapy [24,25]. The outer mitochondrial membrane (OMM), which is engaged in the permeabilization process, and the inner mitochondrial membrane (IMM) channel are in charge of maintaining and adapting membrane potential (ΔΨ); hence, controlling mitochondrial respiration and reactive oxygen species (ROS) production efficiency [26,27]. As a result, the ion channels residing both in the mitochondrial membranes may have an impact on death resistance.

2.1.1. The Voltage-Dependent Anion Channel (VDAC)

The voltage-dependent anion channel 1 (VDAC1), the most abundant protein on the outer membrane of mitochondria, is one of the major proteins that govern mitochondrial function [28,29,30,31]. VDAC1 is the gatekeeper for the passage of metabolites, nucleotides, and ions; it regulates apoptosis through interactions with apoptotic and anti-apoptotic proteins, including the BCL-2 family of proteins and hexokinase [32,33,34,35]. To coordinate cellular metabolism and maintain OMM integrity, Ca2+ from the sarcoplasmic reticulum (SR) or endoplasmic reticulum (ER) enters mitochondria via the VDAC, which is positioned inside the mitochondrial-associated membrane (MAM) domain near the SR [36,37]. VDAC1 is thought to selectively convey apoptotic Ca2+ signals from the SR to the mitochondria [29,38,39]. In addition, VDACs were assumed to be a component of the mitochondrial permeability transition pore (mPTP), a giant channel complex implicated in cell protection and cell death, together with other mitochondrial proteins (ANT, PiC, and TSPO) [40,41,42,43]. Moreover, VDACs are thought to be important in the release of ROS, providing an anchoring site for HK binding, cytochrome c release caused by apoptosis, and (4) interacting with the BCL-2 family of proteins [44,45,46,47,48]. However, Parkin can be recruited to defective mitochondria by VDACs, which promotes mitochondrial autophagy [49].

2.1.2. The Mitochondrial Calcium Uniporter (MCU) Complex

The activities of the mitochondrial calcium uniporter (MCU) complex, which consists of the pore-forming subunit of the MCU channel as well as many regulatory proteins (MICU1, MICU2, MICU3, MCUR1, MCUb, and EMRE), are commonly regarded as a mechanism for Ca2+ entrance into mitochondria [50,51,52]. MCU plays a crucial function in the regulation of aerobic metabolism, ATP production and cell viability by modulating mitochondrial Ca2+ concentration [53,54,55]. In HEK293T and HeLa cells, MCUR1 knockdown (identical to MCU knockdown) inhibits mitochondrial Ca2+ absorption, impairs OXPHOS, reduces cellular ATP, and triggers AMPK-dependent pro-survival autophagy [56]. MICU1 promotes cell survival by acting as a gatekeeper for MCU-mediated mitochondrial Ca2+ uptake [57,58]. MCU or MCUR1 deficiency impairs the MCU heterooligomeric complex, altering mitochondrial bioenergetics, cell proliferation, and migration, as well as initiating autophagy via AMPK [59,60,61].

2.2. Mitochondrial Permeability Transition Pore Complex (mPTPC)

The mitochondrial permeability transition pore complex (mPTPC), a multiprotein complex produced at the contact region between the mitochondrial inner and outer membranes, regulates mitochondrial membrane permeabilization, which occupies a central role as a selective target for anti-cancer therapy [62,63]. Mitochondrial permeability transition (mPT) is a key process in the control of cell death and survival, and is a potential target for cytotoxic drug development [64,65,66]. Alteration in mPT by chemotherapy might influence cell destiny by altering the physiological activity of mPTPC. As a result, it is suggested that forcing the mitochondrial permeability transition pore (mPTP) to open or close for an extended period of time is toxic to the cell [26,67]. For example, pretreatment of mPT with a pore inhibitor, Cyclosporin A (CsA), or a pore opener sensitizer, PK11195, has been discovered to increase mitochondrial apoptosis triggered by resveratrol, a natural polyphenol [68]. It has been demonstrated that modulation of mPT affects multidrug resistance in human hepatocellular carcinoma cells [69].

2.3. ATP Synthase and Mitochondrial Reactive Oxygen Species (mtROS)

ATP synthase is a mitochondrial energy conversion enzyme that is found in the inner mitochondrial membrane and is required for OXPHOS under normal conditions [70,71]. In mammals, ATP synthase is responsible for the majority of cellular ATP generation, and its biogenesis pathway can be affected by either structural changes or insufficient quantities [72,73]. The ATP synthase inhibitor causes cytotoxicity in breast and lung cancer cells but not in healthy cells, suggesting that it could be employed as a cancer treatment candidate [74,75]. It has been demonstrated that combination therapy targeting ectopic ATP synthase and 26S proteasome induces ER stress in breast cancer cells [76]. Various anticancer drugs that target mitochondria diminish ATP levels and/or increase ROS generation [77,78,79]. Oligomycin, an inhibitor of the reversible mitochondrial ATP synthase, induces cell death which is thought to be due to ATP depletion caused by inhibition of the FOF1-ATP synthase (FO and F1 are the main subunit of ATP synthase) enzyme complex [77,80]. Citreoviridin inhibits the growth and proliferation by targeting the F1-ATP synthase in breast and lung cancer cells [75,76]. Polyphenolic phytochemicals including quercetin, genistein, and resveratrol inhibit mitochondrial proton FOF1-ATPase/ATP synthase, which has health benefits as well as possible cytotoxicity [77].
Mitochondria are primarily responsible for energy production, but they also regulate the levels of mitochondria-derived reactive oxygen species (mtROS) [26,81]. Superoxide anion (O2•−), hydroxyl radical (OH), hydrogen peroxide (H2O2), and singlet oxygen (1O2) are all examples of mtROS [26,82,83,84,85]. In mitochondria, mtROS are largely produced by electron transport chain (ETC) complex I and III; however, increased ROS is also caused by inhibition of complex IV or V (ATP synthase) [86,87]. Inhibition of ATP synthase causes a rise in mitochondrial membrane potential (ΔΨm), resulting in increased electron leak to superoxide [88]. The mtROS are required for cell proliferation and cancer growth, but they can also cause DNA damage, protein oxidation, and lipid peroxidation, which can lead to cell death [88,89,90,91,92]. Drug options to combat mtROS activity as a cancer treatment are still being developed, as it can both aid and inhibit tumor cell proliferation. The antioxidant system is engaged to ensure that ROS levels do not reach a deadly threshold, while tumor cell lines increase their ROS production [93,94,95,96,97,98,99]. As a result, while some studies have found dietary antioxidants to be useful in treating cancer, others have found them to be ineffective or even harmful. For instance, carotenoids may increase mortality in breast cancer patients, while vitamin C and E intake have been linked to a lower recurrence rate in the same patients [100,101]. Vitamin C enhances the anti-proliferative activity of doxorubicin in breast cancer patients, while high vitamin D levels are linked to improved survival in colorectal cancer patients [102,103].

2.4. Mitochondrial DNA (mtDNA)

Mammalian mitochondria have their own mtDNA, which encodes 13 oxidative phosphorylation complex polypeptides, 12S and 16S rRNAs, and 22 tRNAs necessary for mitochondrial activity [104]. The mtDNA, a tiny circular chromosome found inside mitochondria, functions as a biomarker of mitochondrial dysfunction and has been linked to the progression of several diseases, including cancer, suggesting that it may be used as a chemotherapeutic target [105]. It has been reported that mtDNA acts as the critical target of cisplatin in its anticancer activity [106,107]. For example, cisplatin binds to mtDNA and VDAC protein in the mitochondrial membrane to induce apoptosis in head and neck squamous cell carcinoma (HNSCC) [108]. Cisplatin induced mitochondrial DNA damage, impaired respiratory activity, increased oxidative stress, and activated caspase-9 in cultured hippocampal neurons and hippocampal neural stem cells (NSCs) [109]. In cisplatin-treated cells, mtDNA damage is visible, and cells with depleted mtDNA derived from a cisplatin-sensitive ovarian cancer cell line developed resistance to cisplatin-induced cell death [110,111].

2.5. DNA Polymerase Subunit Gamma (PolG)

The only DNA polymerase known in human mitochondria, PolG, contains a large catalytic component, PolG1 (140 kDa), as well as two smaller identical accessory subunits, PolG2 (55 kDa), both of which are essential for embryonic development [112,113,114]. The PolG1 mutations have been linked to mtDNA depletion, mitochondrial dysfunction, aging, cancer, and a variety of other illnesses [115,116]. The PolG gene mutations have been found in human malignancies, suggesting that PolG may have a role in decreasing OXPHOS and boosting tumorigenicity in tumor cells [115]. The POLG1 has been shown to have altered genetic and epigenetic regulation in human malignancies, implying that POLG1 germline variations may play a role in tumorigenic characteristics [117]. For example, the breast tumorigenesis is aided by mutations in mitochondrial DNA POLG1 [115]. It is also a crucial component in mtDNA replication and repair [118,119]. It has been reported that targeting mitochondrial DNA PolG could be a unique therapy method for acute myeloid leukemia [120,121]. For examples, in acute myeloid leukemia, the thymidine dideoxynucleoside analog alovudine inhibits mitochondrial DNA polymerase, hinders OXPHOS, and promotes monocytic differentiation [120].

2.6. Mitochondrial Ribosome and Ribosomal Proteins (MRP)

The mitochondrial ribosome is the organelle in eukaryotic cells that synthesize 13 inner membrane proteins of the oligomeric complex required for OXPHOS [122,123]. Mitochondrial ribosome is essential for ribosomal RNA processing, modification, and binding to ribosomal proteins and cellular respiration, a process linked to cell growth and proliferation [124]. Several mitochondrial ribosomal proteins, notably the death-associated proteins 3 (DAP3) and programmed cell death protein 9 (PDCD9), have been identified as apoptosis-inducing agents in several investigations [125,126]. The mitochondrial ribosomal protein L41 (MRPL41) suppresses tumor cell growth through inducing p53-induced mitochondrion-dependent apoptosis, whereas the activation of p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) signaling by the S29 ribosomal protein (RPS29) causes apoptosis in a human laryngeal carcinoma cell line [127,128]. The X-linked ribosomal protein S4 (RPS4X), which is involved in cellular translation and proliferation in breast cancer cell lines, has been identified as a partner of the overexpressed multifunctional protein YB-1, and RPS4X depletion leads to consistent resistance to cisplatin [129]. It has been revealed that RPS4X deletion in ovarian cancer cell lines decreased their proliferative rate but more critically, increased their cisplatin resistance [130]. Higher expression of mitochondrial ribosomal protein L65 (MRPL65), also known as PDCD9 or mitochondrial ribosomal protein S30 (MRPS30) in murine fibroblasts, can cause apoptosis by activating the JNK1 and upregulating other transcription factors [131,132].

2.7. Mitochondrial Proteins

Apoptosis of cancer cells is a desirable result of chemotherapeutic treatment. Inducing apoptosis by targeting mitochondrial proteins is one of the strategies through which chemotherapeutics kill cancer cells. Mitochondrial apoptosis is regulated by the B-cell lymphoma 2 (BCL-2) family proteins, which are classified into three groups based on their primary function, including (i) anti-apoptotic proteins (BCL-2, BCL-xL, BCL-W, MCL-1, etc.), (ii) pro-apoptotic pore-formers (Bax, Bak, BOK, etc.), and (ii) pro-apoptotic BH3-only proteins (Bad, Bid, Bik, Bim, Bmf, HRK, PUMA, etc.) [133,134,135]. The BCL-2 protein family regulates cell death mainly through direct binding interactions that govern mitochondrial outer membrane permeabilization (MOMP), which result in the irreversible release of intermembrane space proteins, caspase activation, and apoptosis [133,136]. One of the aims of chemotherapy is to induce apoptosis by targeting BCL-2 family proteins and creating MOMP. The MOMP causes the release of pro-apoptogenic substances (such as cytochrome c and Smac/DIABLO) from the mitochondrial intermembrane space (IMS) into the cytosol, which activates a caspase cascade that demolishes and kills the cell [136,137,138]. Hundreds of proteins essential for appropriate cellular function and homeostasis are cleaved by caspases, resulting in the biochemical and morphological characteristics of apoptosis [139,140]. Practically all of the developed targeted medicines that have the potential to be lethal to cancer cells only, rather than normal healthy cells, work by activating the same mitochondrial apoptotic pathways as standard chemotherapeutic agents [141,142,143,144].

3. Novel Metabolic Switch Targeted Anti-Cancer Therapeutic Approaches

3.1. Oxidative Phosphorylation (OXPHOS)

Evidence shows that mitochondrial metabolism is critical in cancer cell survival and growth [145]. Glycolysis and OXPHOS are two of the most important metabolic processes for cells to obtain energy [146]. Even when sufficient oxygen is available, cancer cells prefer to employ glycolysis, an inefficient metabolic pathway for energy metabolism that promotes carcinogenesis and cancer growth [147,148]. Glycolysis is significantly simpler than OXPHOS, often known as aerobic mitochondrial respiration. In normal cells, metabolic activities depend mostly on mitochondrial OXPHOS to create ATP, while cancer cells have increased glycolysis and decreased OXPHOS capability (Figure 2). Aerobic glycolysis is used by cancer cells as a metabolic adaptation relative to normal cells, leading some to believe that OXPHOS is downregulated in most of the cancers [149]. Cancer cells typically create pyruvate through the glycolysis route, which produces lactic acid instead of entering mitochondria and converting to acetyl-CoA to produce ATP in hypoxic conditions [149]. Even in the presence of abundant oxygen, cancer cells preferentially use glycolysis to create ATP, known as aerobic glycolysis or the very well-known Warburg effect [150]. In this process, cancer cells would utilize amino acids, fatty acids, and lipid substances to increase their own proliferation in addition to decomposing glucose for ATP [151]. The unique mitochondrial physiology potentially offers anticancer properties because cancer cells reduce their function by utilizing glycolysis [152]. Therefore, restricting cancer growth by targeting several enzymes involved in aerobic glycolysis has emerged as a potential therapy [153,154]. For example, fasentin is an anticancer agent that works by targeting the glucose transporter (GLUT) and preventing glucose uptake [155]. Treatment of pancreatic CD133+ cancer cells with a selective GLUT1 inhibitor, STF-31, increased the sensitivity to cytotoxic drugs such as gemcitabine, paclitaxel, and 5-fluorouracil, resulting in extensive cell death [156]. In many hexokinase 2 (a key enzyme of aerobic glycolysis) overexpressed malignant tumors, lonidamine, 3-bromopyruvate (3-BrPA) and 2-deoxy-D-glucose (2-DG) have been used as effective hexokinase inhibitors [157,158,159,160,161]. Clinical trials have demonstrated that combining lonidamine with a chemotherapy, such as doxorubicin, improves therapeutic efficacy for breast, prostate, melanoma, brain, and ovarian malignancies [162,163].
Conventionally, it has been suggested that tumor cell proliferation is linked to a wide range of OXPHOS deficits and decreased ATP availability. However, some recent studies have shown that OXPHOS can be also upregulated in certain cancers, even in the face of active glycolysis [164,165]. While OXPHOS produces the most ATP, limiting metabolism to glycolysis provides the biomolecule precursors needed by tumors to maintain a high level of growth [166,167,168,169,170,171,172,173]. OXPHOS is thought to be a process implicated in a variety of malignancies, and it is being investigated as a possible target for cancer treatment. Blocking mitochondrial biogenesis, utilizing drugs that impact mitochondrial function, or using direct high-affinity inhibitors of respiratory chain complexes are all options for inhibiting OXPHOS. The OXPHOS inhibitors are used in various tumors, such as metformin for prostate cancer, phenformin for melanoma, carboxyamidotriazole orotate for glioblastoma, and atovaquone for acute promyelocytic leukemia [174,175]. However, due to inherent or acquired resistance of cancer cells to OXPHOS suppression, there are only a few cases in which OXPHOS inhibition would be sufficient to permanently suppress tumorigenesis. Therefore, the combination therapies could be devised to enhance the efficacy of OXPHOS inhibition in cancer treatment. BRAF inhibition, for example, can be coupled with phenformin to slow the progression of melanoma, and the cKIT inhibitor imatinib can be used with VLX600 to decrease the growth of gastrointestinal stromal tumors in mice [176]. The glucose oxidation checkpoint pyruvate dehydrogenase (PDH) and its inhibitor pyruvate dehydrogenase kinase (PDK) can be targeted to modify glucose oxidation and OXPHOS [177,178,179]. The glioblastoma growth and proliferation are inhibited when PDK1 and epidermal growth factor receptor (EGFR) are targeted together, reversing the Warburg effect through decreased HIF-1 expression, shifting the Warburg phenotype to OXPHOS, and inhibiting glioblastoma multiforme growth and proliferation [180]. In addition, several dietary and medicinal plants, such as oleanolic acid and curcumin, inhibit the Warburg effect by altering pyruvate kinase (PK), which reduces aerobic glycolysis in cancer cells [181,182]. Overall, inhibiting the Warburg signature at various checkpoints of the glycolysis could be a highly effective strategy for chemotherapy.

3.2. The Mitochondrial Respiratory Chain (mRC)

The proportional contribution of glycolysis and OXPHOS to ATP production differs substantially between cell types; for example, the typical contribution of OXPHOS to ATP synthesis in normal cells is 80%, but it is less than that in cancer cells [175,183]. Contrary to popular perception, it has been demonstrated that the availability of oxygen (O2) in solid tumors is the most important driver of the oxygen consumption rate (OCR), where respiratory ability is not significantly diminished as compared to normal cells [184]. The heterogeneity among the cancer types in OXPHOS contribution could be due to mtDNA content. For example, acute lymphoblastic leukemia, non-Hodgkin lymphoma, endometrial cancer, colorectal cancer, ovarian cancer, prostate cancer, head and neck cancer, lung adenocarcinoma, esophageal squamous cell carcinoma, and thyroid cancer all have higher mtDNA content than normal tissue [185,186].
The OXPHOS deficiency has been suspected in mitochondrial-based myopathies due to genetic abnormalities in the RCC (either complex I, complex II, or complex IV) [187,188,189]. Metformin, the first-line medication for type 2 diabetes, has shown to have exceptional therapeutic potential in preventing a range of human cancers, as well as anti-cancer activity in preclinical animal models [190]. In addition, metformin has been linked to mitochondrial toxicity due to its complex I inhibitory action [191,192].

3.3. Mitochondrial Metabolic Regulation and Drug Resistance

The anticancer drugs that target thymidylate synthase and folate-dependent enzymes disturb metabolic pathways, potentially leading to new cancer treatments [193]. A growing body of research implies that defective or abnormal glycolysis is frequently linked to drug resistance in cancer treatment [194,195]. Glycolysis dysregulation is caused by abnormal expression of glycolysis-related enzymes, including hexokinase 2 (HK2), phosphoglycerate mutase 1 (PGAM1), pyruvate kinase M2 (PKM2), pyruvate dehydrogenase (PDH) complex, and lactate dehydrogenase (LDH), which contributes to tumorigenesis, tumor growth, and tumor therapeutic resistance [196,197,198,199,200]. Therefore, targeting such enzymes could be novel strategies to overcome cancer chemoresistance (Figure 3). For example, hexokinase 2 is a crucial enzyme that aids the progression and resistance of cancer cells by promoting tumor glycolysis [201]. Hexokinase 2 has two functions: (1) up-regulation of HK2 causes enhanced glycolysis rates, and (2) the interaction between VDAC and HKII inhibits apoptosis by inhibiting the creation of mitochondrial permeability transition pores [202]. Table 1 shows a list of drugs that target glycolysis-related and OXPHOS proteins to aid the effects of conventional chemotherapy.

4. Autophagy: A Potential Target in Cancer Treatment

4.1. Autophagy

Autophagy is a self-digestion mechanism that allows cells to sequester internal and extracellular substrates and merge with lysosomes for degradation to maintain cellular homeostasis, biosynthesis, and energy requirements amid nutritional scarcity or metabolic stress [226,238,239]. This is a complex biochemical process that involves multiple proteins, notably the autophagy-related (ATG) proteins, in sequential phases of initiation, nucleation–elongation–maturation, fusion, and degradation (Figure 4) [240].
Autophagy can be initiated by a variety of intracellular and extracellular factors, including nutrient deprivation, which depletes total amino acids and serum fasting; oxidative stress, which stimulates autophagy to recycle damaged organelles (e.g., mitochondria) and remove protein aggregates; as well as the target of rapamycin (TOR) inhibitors like rapamycin and commodity channel index-779 (CCI-779) [239,241,242]. The mammalian target of rapamycin complex 1 (mTORC1) suppresses autophagy by inactivating the unc-51-like kinase 1 (ULK1) complex, which includes ULK kinase, ATG13, focal adhesion kinase family interacting protein of 200 kDa (FIP200), and ATG101, in nutrient-rich circumstances [243,244]. The ULK complex is activated by the dissociation of mTORC1, which then promotes autophagy via the class III phosphatidylinositol 3-kinase (PI3KC3) complex as a result of a nutritional deficiency [244,245,246]. The PI3KC3 complex regulates the development of autophagic membranes by recruiting many proteins, including vacuolar protein sorting 34 (VPS34), beclin-1, VPS15, autophagy and beclin-1 regulator 1 (AMBRA1), and ATG14-like (ATG14L), which have been identified as new phagophore regulators [247,248,249,250]. Phagophores work by engulfing senescent cytosolic components and elongating into spherical autophagosomes, which are controlled by two ubiquitin-like conjugation pathways: the ATG12 conjugation system and the ATG8 conjugation system, which are two different types of conjugation systems [251,252,253,254]. The covalent conjugation of ATG12 to the ATG5 protein is catalyzed in one system by the E1- and E2-like activities of ATG7 and ATG10, forming the ATG12–ATG5 complex [255,256,257]. The ATG12–ATG5 complex directly associates with ATG16-like (ATG16L), forming the ATG12–ATG5–ATG16L complex, which has an E3-like function in the second ubiquitin-like conjugation pathway [258,259,260,261]. The oligomerization state of ATG16 was shown to affect the development of the approximately 350-kDa complex and autophagic activity [262]. These findings imply that the ATG12–ATG5 conjugate and ATG16 form a multimeric complex regulated by the ATG16 homo-oligomer, and that the 350-kDa complex is necessary for yeast autophagy. The microtubule-associated proteins 1A/1B light chain 3 (LC3) (or ATG8 in yeast) protein is conjugated to a lipid molecule called phosphatidylethanolamine (PE) in the second system [263,264]. The LC3 is first synthesized as a precursor protein (proLC3), and then ATG4 converts proLC3 to LC3-I, which is then conjugated with phosphatidylethanolamine (PE) phospholipid by ATG7 and ATG3, as well as the ATG12–ATG5–ATG16 complex, to create LC3-II [264,265,266,267]. Most outer membrane proteins, including ATG proteins, are dissociated once the autophagosome is formed and matured. The autophagosome joins the lysosome in the perinuclear area to produce the autolysosome [268]. Finally, breakdown products can be recycled and used to support cell growth.
The primary modulator of autophagy, mTOR, is an evolutionarily conserved serine/threonine kinase that transmits a range of autophagy-stimulating signals [269,270]. There are two types of mTOR complexes—mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2)—and these are important nodes in signaling pathways that maintain cellular energy homeostasis by coordinating anabolic and catabolic activities [271,272]. The Ras homolog enriched in the brain (RHEB) is a lysosomal GTPase that interacts directly with mTOR and activates it, which is essential for mTORC1 growth factor activation [273,274]. Activated mTORC1 inhibits the autophagic cascade response by phosphorylating ATG13 and ULK1, and thus suppressing ATG14-containing PI3KC3 activity to inhibit the formation of the pre-initiation ULK complex (ULK1, ATG101-FIP200-ATG13) [275,276].
As part of the autophagosome formation process, the cytosolic microtubule-associated protein light chain 3 (LC3-I) is conjugated with phosphatidylethanolamine to create LC3-phosphatidylethanolamine (LC3-II). As a result, the ratio of LC3-II to LC3-I levels can be utilized as a marker for autophagy activation [277,278]. It has been demonstrated that when HepG2 cells were given a combination of lentiviral shLC3 and epirubicin, their survival rate was much lower than when they were given either agent alone [279]. The findings of this investigation reveal that LC3 plays a critical role in hepatocellular carcinoma chemoresistance, and is a novel therapeutic target for hepatocellular carcinoma. ATG3 is required for autophagy by the E3-like activity of the ATG12-ATG5 conjugate, which is produced by ATG12 activation by ATG7 and conjugation to ATG5 via E2-like ATG10. In colorectal cancer, the c-Myc/miR-27b-3p/ATG10 regulatory axis modulates chemoresistance [280].

4.2. The Role of Autophagy in Cancer Treatment

Autophagy modulation serves as both a tumor suppressant and a tumor promoter in cancer [257,265,281,282]. There has been a lot of interest in inhibiting autophagy for cancer therapy since it was discovered that autophagy is a survival route for tumor cells [283,284]. For example, the lysosomotropic and anti-malarial drug hydroxychloroquine (HCQ), which inhibits lysosome function and thus limits the breakdown of autophagy products, is now being investigated in the clinic trial [283,285]. In addition, human cancers frequently activate different pathways, including mTOR, PI3K, and AKT, and numerous pathway component-specific activation or inhibition are currently under research or in clinical trials [286]. For example, the use of PI3K pathway inhibitors in combination with HCQ or autophagy genetic ablation has resulted in increased tumor regression [285,287,288,289]. In combination with autophagy inhibition, dietary adjustment may help improve cancer therapy. For example, in a mouse melanoma model, leucine deprivation fails to activate autophagy, resulting in tumorigenesis impairment [290,291]. The survival function of autophagy aids tumor cells in the face of increased metabolic demands, a hypoxic microenvironment, or cancer therapy [292]. On the other hand, persistent or excessive autophagy has been demonstrated to increase cell death after treatment with particular chemotherapeutic drugs, either by boosting apoptosis induction or facilitating “autophagic cell death” [293,294,295]. Although the molecular mechanisms by which autophagy mediates its effects on both normal and malignant cells are not fully understood, different signaling pathways have been linked to autophagy upregulation or downregulation [239,296,297,298]. In normal biological conditions, autophagy operates at a basal rate to preserve cellular viability and homeostasis [299]. When autophagy is disrupted by chemotherapy or other environmental stresses, several pathways influence autophagy for cell death (cytotoxic autophagy) or adaptation and survival (cytoprotective autophagy), including the mTOR, phosphoinositide-dependent protein kinase B/AKT (PKB/AKT) pathway, AMP-activated protein kinase (AMPK), tumor suppressor 53 pathway, mitogen-activated protein kinase (MAPK) signaling, FOXO3A-PUMA signaling, and the ER stress pathway (Figure 5) [300,301,302,303,304,305,306,307,308,309,310,311,312].
Upon chemotherapy, the growth factors like epidermal growth factor (EGF) and insulin-like growth factor (IGF) activate the growth factor receptors (EGFR, IGFR, etc.), which in turn activate the PI3K-PDK1-Akt signaling cascade [247,248]. The upstream signaling of PI3K/AKT, AMPK, p53 and MAPKs is integrated by mTORC1 to regulate the autophagic pathway [313,314,315,316,317,318,319,320]. The activation of pro-oncogenic class-I PI3K (PI3KC1) by insulin receptor substrate (IRS) proteins, IRS1 and IRS2 catalyzes the conversion of phosphatidylinositol-4,5-bisphosphate (PIP2) to phosphatidylinositol-3,4,5-trisphosphate (PIP3), which binds both AKT and phosphoinositide-dependent protein kinase 1 (PDK1) and triggers PDK1 to phosphorylate AKT at Thr308 [313,314,315,316,317,318,319,320]. Tuberous sclerosis complex 2 (TSC2) is phosphorylated by active Akt, which suppresses the TSC complex, a GTPase-activating protein (GAP) complex composed of TSC1/2. The TSC complex inhibits mTOR by inactivating RHEB [321,322].
The energy-sensing kinase AMPK is a crucial pathway in eukaryotic cells for maintaining energy balance, coordination metabolism, and the regulation of autophagy, apoptosis, epithelial–mesenchymal transition (EMT), etc. [323,324,325]. The AMPK cascade induces autophagy by recognizing the amount of AMP and ATP [326,327,328,329]. The AMPK is also activated by calcium/calmodulin-dependent protein kinase kinase (CaMKK) and liver kinase B1 (LKB1) when they phosphorylate the Thr-172 residue in response to chemotherapy [330,331,332,333,334,335,336,337,338]. Activated AMPK promotes the formation of TSC1/TSC2 complex to inhibit mTOR activity, resulting in autophagy induction [317,339,340,341,342]. The AMPK can directly phosphorylate ULK1 by separating mTORC1 from ULK1 and promote autophagy in the starving condition, whereas mTORC1 can limit AMPK’s activity by phosphorylating ULK1 in a glucose abundant situation [317,339,340,341,342].
The p53 tumor suppressor gene regulates autophagy either by nuclear p53 to stimulate autophagy via transcriptional activities or cytoplasmic p53 to restrain autophagy via cytoplasmic functions [343,344,345,346,347,348]. The activated AMPK causes autophagy through the TSC1/TSC2 complex and the mTOR pathway when nuclear p53 production is boosted by external stress [349,350]. It was discovered that p53 increases the expression of its downstream substrates Sestrin, which then activates AMPK, resulting in the deactivation of mTORC1 and the induction of autophagy [351]. Other key downstream substrates of nuclear p53 include the damage regulated autophagy modulator (DRAM) and death-associated protein kinase 1 (DAPK1), both of which have been shown to activate autophagy [352,353,354,355,356]. The DAPK-1 induces autophagy and membrane blebbing via binding to a linear peptide motif in microtubule-associated protein 1B (MAP1B) or directly phosphorylates Beclin1, a necessary component of the Vps34 complex [357,358,359,360]. On the other hand, cytoplasmic p53 suppresses autophagy through blocking AMPK, TIGAR (TP53-induced glycolysis and apoptosis regulator), and Beclin-1 [361,362,363,364,365,366].
MAPKs are involved in a variety of biological activities, including cell development, proliferation, and autophagy [367]. MAPK signaling can be triggered by a variety of cell surface growth factor receptors (i.e., HER2/EGFR, and HER2) in response to external stimuli including chemotherapy [368,369,370,371,372]. The activation of extracellular signal regulated kinases (ERK) through the Ras-Raf-MEK-ERK signal pathway can induce autophagy by the blocking of the TSC1/TSC2 complex, leading to the increased expression of Rheb-GTPase to induce mTORC1, which can result in the inhibition of autophagy [373,374,375,376].
Autophagy’s ability to recycle macromolecules gives cancer cells a competitive edge in harsh situations including metabolic stress, glucose deprivation, oxidative damage, hypoxia, expression of aggregate-prone proteins, ER stress, and so on. Among various stressors, ER stress can successfully trigger autophagy because tumor cells must reuse their organelles to maintain growth [377,378]. Autophagy is induced by ER stress via the protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK)/eukaryotic initiation factor 2 (eIF2) and IRE1/JNK1 pathways [379,380,381]. The unfolded-protein response (UPR) is dominated by the phosphorylation of eIF2, translation suppression, and selective translation of the transcription factor (TF) ATF4 [379]. Phosphorylation of PERK/eIF2 has been found to be required for the transcription of critical autophagy-associated genes (ATG) during ER stress, and may also play a role in the formation of LC3-II [308,380]. Melatonin-induced suppression of gastric cancer (GC) cell proliferation is mediated by activation of the IRE/JNK/Beclin1 signaling pathway, according to recent research [382]. JNK pathway activation has been shown to cause selective phosphorylation of c-Jun and up-regulation of c-Fos [383,384]. The JNK also controls autophagy by regulating ATG transcription via FOXO [385,386,387].
Through its established roles in genomic stability, elimination of endogenous sources of ROS, maintenance of bioenergetic functions, degradation of oncogenic proteins, and induction of immune response mechanisms against malignant transformations, autophagy has been involved as a desirable pathway for repressing cancer initiation at distinct steps [388,389,390]. For example, genetic studies of autophagic machinery, such as Atg7, Atg5, and Beclin1, have found that when autophagy is disrupted, tumor initiation accelerates [391,392,393,394]. Autophagic cell death (ACD) is a type II cell death process that does not cause real apoptosis, and involves an increase in autophagic flux before or during cell death rather than only an increase in autophagy markers [395,396,397]. Chloroquine has been shown to destroy breast and pancreatic cancer stem cells (CSCs) by inhibiting autophagy via the Janus kinase 2 (JAK2) and DNA methyltransferase 1 (DNMT1) pathways [398,399,400]. However, in another study, autophagy was observed to promote the mortality of CSCs, which is caused by an increase in autophagy-related proteins and autophagosomes in CSCs from malignant gliomas [401,402]. Autophagy induces cell death and regulates cell proliferation by accelerating genetic stability through removing oncogenic proteins [391]. For example, autophagy has been found to destroy a number of proteins implicated in oncogenesis, including the mutant form of p53, DRAM, p62, NF-κB, promyelocytic leukemia/retinoic acid receptor α (PML/RARα), BCR-ABL1, and sequestosome 1 (SQSTM1) [352,403,404,405,406,407,408,409]. In addition, autophagy’s inducement stress-related response is one possible method by which autophagy serves as a tumor suppressor, including oncogenic stress, ER-stress and DNA-damage-stress, etc. [410,411]. Cellular senescence is a program of permanent cell division arrest that can be produced in response to oncogenic stress to avoid malignant transformation [412,413]. Increased levels of ER-stress and DNA-damage-stress resulted from autophagy suppression, which in turn contributed to oncogenesis through increased levels of p62 accumulation [405,414,415,416].
In addition to its tumor suppressive role, autophagy supports the tumor microenvironment of cancer cells in stress conditions, including anti-cancer treatment, metabolic stress, oxygen and glucose deprivation, genotoxic, and oxidative stress. This tumor-promoting role of autophagy is achieved through supplying nutrients and energy to cancer cells, mediating adaptation to hypoxia, oxidative stress and DNA damage, supporting stromal cells to maintain homeostasis and tumor growth, and regulating UPR [417]. In the stroma, autophagy’s activation protects nearby cancer cells from cellular damage and death by recycling energy-rich metabolites such as the ketones and L-lactate, leading to improved mitochondrial biogenesis [418]. Autophagy is an important method for cancer cells to cope with UPR and to promote their survival [419]. In the majority of cancer cells, the PERK-arm of the UPR is critical for autophagy’s activation; for example, both phospho-mutant eIF2 and dominant-negative PERK have been found to impede LC3-I to LC3-II conversion [308,379].

5. Cancer Chemoresistance: Mitochondrial Fusion, Fission, Biogenesis, and Mitophagy

5.1. Mitochondrial Fusion and Fission

Mitochondrial biogenesis, fusion, fission and mitophagy, collectively referred to as mitochondrial dynamics, are important in maintaining mitochondrial homeostasis in response to variations in energy and stress [420]. The beginning and advancement of various cancer types, as well as cancer metastasis, CSC survival, and drug resistance, have all been linked to defective mitochondrial dynamics (Figure 6) [421,422,423]. Fusion happens when two adjacent mitochondria come together, whereas fission occurs when one mitochondrion splits into two [424,425,426]. These two processes are constantly counterbalanced to maintain mitochondrial homeostasis without interfering with each other’s functions [427]. Mitochondrial fusion and fission have a role in a number of cellular activities, including cell cycle development and apoptosis [428,429]. Fusion is frequently linked to high energy demand, resulting in a hyperfused mitochondrial network that produces more ATP and protects against autophagy [424,430,431,432,433,434]. Fission is mostly associated with apoptosis, facilitating mitophagy, promoting mitotic mitochondrial fragmentation and segregating damaged mitochondrial subdomains for elimination [430,435,436,437]. Notably, fusion and fission can be proapoptotic or antiapoptotic depending on the condition of the cell, confounding their involvement in cancer [438,439,440]. The formation of homotypic and heterotypic oligomers of MFN1 and MFN2, which leads to membrane clustering in a GTP-dependent manner, is primarily responsible for fusion of the OMM, whereas OPA1 requires MFN1 to mediate mitochondrial fusion at the IMM [441,442,443,444,445,446]. Fusion requires the co-existence of long and short OPA1 isoforms (L-OPA1 and S-OPA1), although only the L-OPA1 has been shown to be fusion competent [447,448]. Mitochondrial fission is predominantly regulated by dynamin-related protein 1 (DRP1), a cytosolic guanosine triphosphatase (GTPase), recruited by OMM receptors, such as fission protein homolog 1 (FIS1), mitochondrial fission factor (MFF), and mitochondrial dynamics proteins (MIDs) 49/51 [449,450]. DRP1 oligomerizes and coils around the constriction sites of dividing mitochondria when it reaches the OMM, facilitating mitochondrial fission [451].
It has been revealed that chemoresistant ovarian cancer cells had more interconnected mitochondrial networks than their chemosensitive counterparts, implying that mitochondrial fusion plays a role in chemoresistance [452,453,454,455]. The mitochondrial membrane potential collapses early in apoptosis, allowing total cytochrome c release from mitochondria to the cytosol [456]. The fusion GTPase OPA1 has the ability to prevent cytochrome c mobilization and release [457]. It has been demonstrated that venetoclax-resistant AML cells are also resistance to cytochrome c release whenever stimulated due to upregulation of OPA1 [458,459]. Since MFN2 deletion greatly enhances Jurkat doxorubicin sensitivity, MFN2 and OXPHOS have been discovered to be considerably elevated in surviving leukemia cells [460,461]. It has been discovered that suppressing MFN1 improves cisplatin sensitivity in human neuroblastoma cells by inhibiting mitochondrial fusion [462]. Even though mitochondrial fission is the polar opposite of mitochondrial fusion, mounting data suggests that it is similarly important in chemoresistance [463]. For example, the chemosensitivity of nasopharyngeal cancer (NPC) cells to cisplatin was boosted by metformin or cucurbitacin E, which inhibited the DRP1 upstream kinase AMPK or cyclin B1/Cdk1 [464]. Similarly, mitochondrial fission driven by DRP1 phosphorylation enhances cell survival and cisplatin resistance in EBV-LMP1-positive nasopharyngeal cancer [465]. As cancer cells adapt to the hypoxic tumor microenvironment, chemoresistant cells may be able to target mitochondrial fission. For example, inhibition of DRP 1 by either a putative DRP inhibitor (e.g., Mdivi-1) or DRP1 silencing increased the cisplatin sensitivity of hypoxic ovarian cancer cells [455,466]. Mdivi, a Drp1 inhibitor, abolished invasion and chemoresistance induced by LASS2 depletion. The mesenchymal stem cells (MSCs)-induced drug resistance is linked to ERK/Drp1-dependent mitochondrial fission [467]. PD98059, an ERK inhibitor, inhibits LASS2-deficient bladder cancer invasion and chemoresistance by suppression of Drp1 phosphorylation [468,469]. Anticancer drugs that increase the release of the high-mobility group box 1 protein (HMGB1) trigger ERK1/2-mediated DRP1 phosphorylation in colorectal cancers, or increased ROS production and hypoxia in ovarian tumors, and are usually associated with fission-driven chemoresistance [470]. Conversely, induction of DRP1 through inhibiting anti-apoptotic proteins BCL-2 or activating Sirt3 (histone deacetylase) by ABT737 (an inhibitor of antiapoptotic BCL-2/BCL-X) promotes fission, leading to apoptosis and mitophagy in ovarian cancer cells resistant to cisplatin [453,455,471].

5.2. Mitochondrial Biogenesis and Mitochondrial Selective Autophagy (Mitophagy)

Mitochondrial biogenesis and mitochondrial selective autophagy (mitophagy), two opposing cellular processes, coordinately control mitochondrial content to support energy metabolism, in response to the cellular metabolic conditions, stress, and other intracellular or external signals [472]. Mitochondrial biogenesis is a multistep biological process that includes mtDNA transcription and translation, as well as the translation of nucleus-derived transcripts [473]. The recruitment of newly synthesized proteins and lipids, import and assembly of mitochondrial and nuclear products in the expanding mitochondrial network are the principal roles of mitochondrial biogenesis. The peroxisome proliferator-activated receptor-gamma co-activator 1-alpha (PGC-1α), the well-studied member of the PGC family, coordinates the activity of many transcription factors involved in mitochondrial biogenesis and function [474]. For example, PGC-1 overexpression improves mitochondrial function and promotes mitochondrial proliferation in mouse models [475]. Furthermore, the expression of PGC-1, and/or its homolog PGC-1, promotes mitochondrial respiration in complex III or IV impairment. As a result, PGC-1 has been dubbed the “master regulator” of mitochondrial biosynthesis and function. In addition, PGC1-expressing cells have a better chance of surviving against metabolic stressors such as oxidative damage, energy deprivation, and even cancer therapy [476]. Based on the evidence, PGC1 is thought to play a function in chemotherapy resistance. The most common techniques for disrupting PGC1 signaling have been to block critical enzymes in PGC1-dependent metabolic pathways or to target PGC1-related transcription factors as PGC1 inhibitors are not yet available. For instance, inhibitors of OXPHOS, FAO, or ERR all inhibited cell proliferation in PGC1-positive cancer cells [477]. Suppressing PGC1 or OXPHOS expression in melanoma cells re-sensitizes therapeutic-resistant cells to oxidative damage [478]. The PGC1 suppression increases ROS generation, which stabilizes HIF1 protein and causes a metabolic transition from OXPHOS to glycolysis, which enhances cell survival. It has been reported that triple suppression of PGC1α, HIF1α, and glutamine utilization results in complete blockage of tumor growth [479]. The PGC1 was found to be a tumor suppressor in several cancer types in a few investigations [480,481,482]. Low PGC1 expression, for example, has been linked to poor prognosis in VHL-deficient clear cell renal cell carcinoma (ccRCC) and breast cancer patients [480]. PGC1 increases Bax-mediated apoptosis in colorectal and ovarian epithelial carcinoma cells and works as a stabilizer of the tumor suppressor mitostatin, which triggers mitophagy in breast cancer [482,483,484].
Mitophagy is the process of selective removal of damaged mitochondria by the autophagic machinery. In many mammalian cell types, mitochondrial fission is shown to be synchronized with mitophagy, implying that it is a precondition for the mitochondrial breakdown process [485]. The mitophagy induction process begins with the engulfment of damaged mitochondrial components by an isolating membrane (phagophore). Mitophagy involves vesicle nucleation, vesicle elongation, and retrieval, which is followed by the creation of a mitophagosome, a double-membrane structure that sequesters the organelle and allows mitophagosomal destruction after fusion with a lysosome [486,487]. During the degradation process, amino acids and fatty acids are produced, which can be used for protein synthesis or oxidized by the mitochondrial ETC to produce ATP that can be used for cell survival [488]. Essential components for cell viability (e.g., mitochondria) can be degraded with a lethal process, leading to autophagic cell death [489,490,491,492]. Many studies have found that the mitophagy-related molecules phosphatase and tensin (PTEN)-induced kinase 1 (PINK1), parkin, Bcl-2/adenovirus E1B 19 kDa interacting protein 3 (BNIP3), FK506-binding protein 8 (FKBP8) and FUN14 domain-containing protein 1 (FUNDC1) promote tumor formation and can be used as therapeutic targets in a variety of malignancies [493,494,495,496]. For example, PINK1 overexpression is linked to a poor response to chemotherapy and a poor prognosis in patients with esophageal squamous cell carcinoma treated with neoadjuvant chemotherapy, while silencing PINK1 inhibits lung cancer cell growth and migration while also inducing cell death [495,497]. In addition, polyphyllin I, a natural compound, decreased tumor growth and promoted apoptosis in MDA-MB-231 xenografts, and PINK1 knockdown boosted these effects [498]. Furthermore, FUNDC1 overexpression predicts a poor prognosis and is a possible target for improving chemoradiotherapy effects in cervical cancer patients. In contrast, a study found that the migraine medicine dihydroergotamine tartrate (DHE) could cause lung cancer cell death via mitophagy and mitochondria-dependent cell apoptosis through the activation of the PINK1/Parkin pathway [499]. In addition, translocation of BNIP3 to the mitochondria causes cytochrome c to be released, resulting in caspase activation and apoptosis in cardiomyocytes [500]. Arsenic trioxide, ceramide, and concanavalin A can stimulate BNIP3, and BNIP3 is involved in the induction of autophagy and cell death in response to these stimuli [501,502,503].
Drug resistance frequently results in treatment failure due to autophagy or mitophagy. Mitophagy has a dual role in cancer therapy: on the one hand, it can cause cancer cell death, and on the other, it can help cancer cells survive [504]. Importantly, drug resistance to major chemotherapy medications, including cisplatin, doxorubicin, 5-fluorouracil, and paclitaxel, typically results in treatment failure due to autophagy or mitophagy [505]. The PINK1/Parkin pathway is the well-studied ubiquitin-mediated system involved in cancer chemoresistance. PINK1 aggregation at the OMM is triggered by mitochondrial malfunction or depolarization, which recruits and phosphorylates Parkin, an E3 ligase, boosting its E3 ligase activity [506,507,508,509,510]. B5G1, a novel betulinic acid analog, sensitizes multidrug-resistant cancer cells via inhibiting PINK1/ Parkin -dependent mitophagy [511]. Several mitophagy receptors, including BNIP3, BNIP3-like (BNIP3L, also known as NIX)), and FUNDC1, have been linked to chemoresistance in PINK/ Parkin -independent mitophagy [512,513,514,515,516]. Mitophagy produced by doxorubicin leads to drug resistance, and BNIP3L silencing improves doxorubicin sensitivity in human colorectal CSCs isolated from HCT8 cells, implying that mitophagy plays a role in drug resistance [517]. FUNDC1 overexpression stimulates the development of cervical cancer cells, while inhibition of its expression improves susceptibility to cisplatin and ionizing radiation [518].

5.3. Autophagy Acts as a Chemotherapy Resistance Machinery in the Mitochondrial Pathway

The main principles of cancer therapy are the induction of cell death and the inhibition of cell survival. According to an increasing body of research, autophagy contributes to the anticancer efficiency of chemotherapy by inducing the apoptotic pathway, but it also confers drug resistance via its paradoxical protective role in many circumstances (Figure 7) [519]. Recent research shows that increased autophagy not only improves tumor survival, but also improves tumor drug resistance in a variety of tumor types. Autophagy influences both the efficacy of tumor chemotherapy and the degree of drug resistance, and this is likely because cancer cells use mitophagy to rapidly sweep away damaged mitochondria in order to mediate their own drug resistance [497,520]. According to several studies, the ATG has a crucial role in drug resistance mediated by mitochondria. For example, ATG12 conjugation regulates mitochondrial homeostasis and cell death, whereas altering the ATG12–ATG3 complex leads to increased mitochondrial mass, fragmentation of the mitochondrial network, and resistance to cell death mediated by mitochondrial pathways [521]. It has been demonstrated that miR-1 overexpression increased the cisplatin sensitivity by suppressing the ATG3-related autophagy of NSCLC cells [522]. Similarly, by inhibiting ATG5 expression and autophagy, miRNA-153-3p increases gefitinib sensitivity in NSCLC. Recently, ATG5 has been found to be implicated in doxorubicin resistance in gall bladder cancer [523]. It has been shown that knocking down ATG5 in paclitaxel-resistant cells causes a G2/M arrest and makes cells susceptible to paclitaxel-induced necrosis [524]. Another study showed that direct knockdown of ATG5 increased paclitaxel resistance in Ras-NIH 3T3 (v-Ha-ras-transformed NIH 3T3) cells by reducing the frequency of early apoptotic cells [525]. Direct control of beclin-1 (ATG6) is also critical in the development of drug resistance. For example, in ER (estrogen receptor)-positive breast cancer cells, suppressing beclin-1 boosted tamoxifen sensitivity in vitro, and lower beclin-1 expression predicts a better prognosis in patients with ER-positive breast cancer [526]. MicroRNA-30a downregulation enhanced osteosarcoma cell chemoresistance by activating beclin-1-mediated autophagy, which decreased cell proliferation and invasion [527]. In addition, miR-30a has been shown to sensitize tumor cells to cisplatin by lowering beclin-1-mediated autophagy, suggesting that boosting miR-30a levels in tumor cells could be a novel way to improve chemotherapy’s efficacy during cancer treatment [528]. In MCF-7 cells, co-treatment with docetaxel and siATG7 resulted in a higher cytotoxicity and apoptosis rate than docetaxel treatment alone, demonstrating that silencing of ATG7 increased the effectiveness and apoptotic effect of docetaxel [529]. ATG12 is important for autophagy, and studies have shown that miR-23b-3p can suppress autophagy in gastric cancer cells, restoring the proliferation of chemoresistant cells in vitro and in vivo [530]. Through Nrf2-dependent metabolic reprogramming, p62/SQSTM1 increases the malignancy of HCV-positive hepatocellular carcinoma [531]. The abnormal amplification and phosphorylation of p62/SQSTM1 has been linked to tumor growth and cisplatin resistance in patient-derived high-grade serous ovarian cancer cells [532].
Several upstream target genes and signaling pathways have been shown to influence autophagy-mediated chemoresistance, in addition to directly influencing ATG as indicated above, including DNA-dependent protein kinase catalytic subunit (DNA-PKcs), high mobility group box 1 (HMGB1), high-mobility group nucleosome-binding domain 5 (HMGN5), heat shock protein 90AA1 (HSP90AA1), insulin growth factor 2 (IGF2), and N-myc downstream regulated gene 1 (NDRG1) [533,534,535,536,537]. The induction of autophagy by oxidative stress is thought to contribute to the long-term survival of breast cancer cells by regulating DNA repair via ataxia telangiectasia mutated (ATM), DNA-PKcs, and poly (ADP-ribose) polymerase (PARP)-1 [538,539]. HMGB1 has become a new target for chemotherapy because of its autophagy regulation in response to oxidative stress [540,541]. HMGB1 is a prototypical damage-associated molecular pattern (DAMP) molecule that is released by induced autophagy and enhances treatment resistance in ovarian cancer, colorectal cancer, and lung cancer [470,542]. The release of the HMGB1 and the receptor for the advanced glycation end-product (RAGE) triggers autophagy, which leads to chemoresistance and regeneration of cancer cells [543,544]. The use of an HMGB1 inhibitor or a RAGE blocker to improve chemotherapeutic sensitivity in colorectal cancer is well-accepted [470]. HMGN5 is a novel target for enhancing osteosarcoma therapy because it regulates autophagy, a key component in the development of chemoresistance [535]. HSP90AA1 increases chemotherapy (doxorubicin, cisplatin, and methotrexate) resistance in osteosarcoma through upregulation autophagy [534]. The pro-survival action of HSP90AA1 could be reversed by 3-methyladenine (3-MA) through inhibiting HSP90AA1 and increased chemosensitivity via blocking autophagy. In human osteosarcoma cell lines, IGF1R-targeted treatment improved doxorubicin chemosensitivity [536]. Activation of IGF or insulin signaling has been shown to prolong the lifespan of osteosarcoma cells under chemotherapeutic stress, resulting in a drug-resistance [545]. The anti-metastatic activity of cells expressing NDRG1 makes this protein an important therapeutic target for cancer chemotherapy [537]. NDRG1 inhibits both basal and hypoxia-induced autophagy at the initiation and degradation stages, as well as making pancreatic cancer cells more susceptible to lysosomal membrane permeabilization [546].
The Ca2+ is an important intracellular second messenger that regulates significant cell fate decisions like metabolism, growth, and death [547]. Mitochondria are considered as a crucial cell death checkpoint, and mitochondrial Ca2+ excess is thought to be a powerful inducer of apoptosis through the intrinsic pathway. One of the ways to trigger mitochondrial-mediated intrinsic apoptotic pathway is mitochondrial Ca2+ overload, which is caused by a persistent rise of calcium ion [24,548,549]. According to a growing number of studies, Ca2+ is also involved in the latest hallmarks of cancer like autophagy, which has been linked to tumor growth and cancer therapy [25,292,550]. As a result, targeting mitochondria, specifically mitochondrial Ca2+ homeostasis and autophagy, is a main focus of anti-cancer therapy in order to sensitize cancer cells to death and overcome drug resistance [551].
Uncontrolled Ca2+ dynamics affect practically every aspect of cell function, including proliferation, gene expression, cell death, and protein phosphorylation and dephosphorylation due to the complicated network of Ca2+ transporters or pumps is disrupted [552,553,554]. The downregulation of calcium release-activated calcium modulator 1 (CRACM1, also called Orai1) has been demonstrated to delay cytoplasmic Ca2+ clearance, boosting the activation of many Ca2+-dependent kinases and enzymes, leading to activation of the cyclin-dependent kinase inhibitor p21, which results in autophagy activation, cell growth arrest, and enhanced cell survival [555]. Inhibition of ORAI1-mediated Ca2+ entry, on the other hand, increases the chemosensitivity of HepG2 hepatocarcinoma cells to 5-fluorouracil [556]. Furthermore, several Ca2+-mobilizing substances (thapsigargin, ATP, ionomycin, and chemotherapeutic drugs) and nutritional deprivation cause increases in Ca2+ levels, as well as the activation of pro-survival autophagy in cancer cells [557,558,559].
The inner mitochondrial membrane of diverse tissues has been found to contain the following types of potassium channels: ATP-sensitive, Ca2+-activated, voltage-gated, and two-pore domain potassium channels [557,560,561,562,563,564,565,566]. The regulation of mitochondrial respiration, membrane potential, and ROS production are all direct functions of these channels [567]. The mitochondrial potassium channel act as important factors in various pro-life and pro-death reactions that are triggered by changes in these channel activities in distinct cell types. In response to a change in mitochondrial Ca2+ concentration, Ca2+ regulates three types of mitochondrial potassium channels: mitoBKCa, mitoIKCa, and mitoSKCa [567,568]. Because of their putative role in cytoprotective events, potassium channels found in heart or brain mitochondria have received a lot of interest [567,568,569,570,571,572,573]. For example, in cardiac and neural tissue, activation of mitochondrial potassium channels, such as ATP-regulated or calcium-activated large conductance potassium channels may have cytoprotective effects [574]. It is critical to design a therapy that encourages cancer cells to undergo apoptosis in spite of such limitations, like Bax or Bak, which causes apoptosis resistance and limits the activity of chemotherapeutics. The Bax protein blocks the mitochondrial Kv1.3 potassium channel, making it a potential therapeutic target [575]. For example, three inhibitors (Psora-4, PAP-1, and clofazimine) of mitochondrial Kv1.3 channels cause cancer cells to die in a Bax/Bak-independent manner [576].

6. Conclusions

Overall, this work provides strong evidence for the roles of autophagy and mitochondria in cancer chemoresistance. In many malignancies, autophagy modulation serves as both a tumor suppressor and a tumor promoter. Autophagy helps tumor cells survive and confers drug resistance in numerous situations when they are subjected to increased metabolic demands during cancer therapy. Survivability of cancer cells in the presence of abnormal glucose metabolism, fatty acid synthesis, and glutamine metabolic rates is linked to glycolysis and OXPHOS, which is important not only for carcinogenesis, but also for chemoresistance and recurrence, and especially for the emergence of treatment failure. Chemotherapy-induced mitophagy aids mitochondrial biogenesis by allowing damaged proteins or organelles to be recycled, preventing DNA damage and enhancing drug resistance. As a result, discovering autophagy inhibitors or activators, as well as targeting glycolysis and OXPHOS, could be critical in reversing drug resistance and enhancing chemosensitivity. However, combining autophagy and glycolysis, as well as OXPHOS modulators and chemotherapeutic medicines, will offer new hope for cancer treatment.

Author Contributions

E.B., conceptualization, methodology, data curation, software and formal analysis, original draft preparation; J.-Y.K., conceptualization, review and editing of the manuscript; S.-Y.H., validation, investigation, resources, supervision, project administration, writing—review and editing of the manuscript; H.Y., validation, investigation, resources, supervision, project administration, funding acquisition and writing—review and editing of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF), funded by the Ministry of Science and ICT (2019R1F1A1059148, 2022R1A2C1009939, 2022R1C1C1008833).

Institutional Review Board Statement

Not applicable for this study.

Informed Consent Statement

Not applicable for this study.

Data Availability Statement

The data presented in this study are available in this article Cancers.

Acknowledgments

Not applicable for this study.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviation

ADPadenosine biphosphate
ATPadenosine triphosphate
ATGsautophagy genes
AMPKAMP-activated protein kinase
BCL-2B-cell lymphoma 2
Ca2+calcium ion
CaMKKcalcium/calmodulin-dependent protein kinase kinase
CsAcyclosporin A
DNAdeoxyribonucleic acid
mtDNAmitochondrial DNA
DNA-PKcsDNA-dependent protein kinase catalytic subunit
Drp1dynamin-related protein 1
DRAMdamage regulated autophagy modulator
DAPK1death-associated protein kinase 1
DNA-PKcsDNA-dependent protein kinase catalytic subunit
mtDNAmitochondrial deoxyribonucleic acid (DNA)
ERKextracellular signal regulated kinases
ETCelectron transport chain
eIF2eukaryotic initiation factor 2
FADH2adenine dinucleotide
FIP200focal adhesion kinase family interacting protein of 200 kDa
GDPguanosine diphosphate
GTguanosine triphosphate
GLUTglucose transporter
HKhexokinase
HMGB1high mobility group box 1
HMGN5high-mobility group nucleosome-binding domain 5
HSP90AA1heat shock protein 90AA1
IGF2insulin growth factor 2
IMMinner mitochondrial membrane
IRS1/2insulin receptor substrate 1 and 2
JNKc-Jun N-terminal kinase
LC3 I/IIlight chain I and II
LDHlactate dehydrogenase
LKB1liver kinase B1
MCTmonocarboxylate transporters
MCUmitochondrial calcium uniporter
MEKmitogen-activated protein kinase/ERK kinase
MAMmitochondrial-associated membrane
mtROSmitochondria-derived reactive oxygen species
MOMPmitochondrial outer membrane permeabilization
Mnf1/2mitofusin 1 and 2
NADHnicotinamide adenine dinucleotide (NAD) hydrogen
NDRG1N-myc down-stream regulated gene 1
OPA1optic atrophy 1
OXPHOSoxidative phosphorylation
OMMouter mitochondrial membrane
PI3Kphosphatidylinositol 3-kinase
PDK1phosphoinositide-dependent protein kinase 1
PERKprotein kinase R (PKR)-like endoplasmic reticulum kinase
PEphosphatidylethanolamine
PTENphosphatase and tensin
PIP2phosphatidylinositol-4,5-bisphosphate
PIP3phosphatidylinositol-3,4,5-trisphosphate
PKBphosphoinositide-dependent protein kinase B
PGC1αproliferator-activated receptor-gamma co-activator 1-alpha
PFKphosphofructokinase
PGAMphosphoglycerate mutase
PI3Kphosphatidylinositol 3-kinase
PolGDNA polymerase gamma subunit
mPPTmitochondrial permeability transition pore complex
RASrat sarcoma
RAFrapidly accelerated fibrosarcoma
RCCrespiratory chain complex
Rhebras homolog enriched in the brain
ROSreactive oxygen species
mRNAmessenger ribonucleic acid
tRNAtransfer ribonucleic acid
TCAtricarboxylic acid
TIGARTP53-induced glycolysis and apoptosis regulator
TSC1/2tuberous sclerosis complex 1 and 2
mTORmammalian target of rapamycin complex
ULK1unc-51-like kinase 1
VDAC1voltage-dependent anion channel 1
VPS34vacuolar protein sorting 34

References

  1. Yeldag, G.; Rice, A.; Del Rio Hernandez, A. Chemoresistance and the Self-Maintaining Tumor Microenvironment. Cancers 2018, 10, 471. [Google Scholar] [CrossRef] [Green Version]
  2. Assaraf, Y.G.; Brozovic, A.; Goncalves, A.C.; Jurkovicova, D.; Line, A.; Machuqueiro, M.; Saponara, S.; Sarmento-Ribeiro, A.B.; Xavier, C.P.R.; Vasconcelos, M.H. The multi-factorial nature of clinical multidrug resistance in cancer. Drug Resist. Updates 2019, 46, 100645. [Google Scholar] [CrossRef] [PubMed]
  3. Vaidya, F.U.; Sufiyan Chhipa, A.; Mishra, V.; Gupta, V.K.; Rawat, S.G.; Kumar, A.; Pathak, C. Molecular and Cellular Paradigms of Multidrug Resistance in Cancer. In Cancer Reports; Wiley Online Library: Hoboken, NJ, USA, 2020; p. e1291. [Google Scholar] [CrossRef]
  4. Bahar, E.; Kim, J.Y.; Yoon, H. Chemotherapy Resistance Explained through Endoplasmic Reticulum Stress-Dependent Signaling. Cancers 2019, 11, 338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Bahar, E.; Kim, J.Y.; Kim, H.S.; Yoon, H. Establishment of Acquired Cisplatin Resistance in Ovarian Cancer Cell Lines Characterized by Enriched Metastatic Properties with Increased Twist Expression. Int. J. Mol. Sci. 2020, 21, 7613. [Google Scholar] [CrossRef]
  6. Bahar, E.; Kim, J.Y.; Kim, D.C.; Kim, H.S.; Yoon, H. Combination of Niraparib, Cisplatin and Twist Knockdown in Cisplatin-Resistant Ovarian Cancer Cells Potentially Enhances Synthetic Lethality through ER-Stress Mediated Mitochondrial Apoptosis Pathway. Int. J. Mol. Sci. 2021, 22, 3916. [Google Scholar] [CrossRef] [PubMed]
  7. Bohovych, I.; Chan, S.S.; Khalimonchuk, O. Mitochondrial protein quality control: The mechanisms guarding mitochondrial health. Antioxid. Redox Signal. 2015, 22, 977–994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Ding, W.X.; Yin, X.M. Mitophagy: Mechanisms, pathophysiological roles, and analysis. Biol. Chem. 2012, 393, 547–564. [Google Scholar] [CrossRef] [Green Version]
  9. Wang, Y.; Liu, H.H.; Cao, Y.T.; Zhang, L.L.; Huang, F.; Yi, C. The Role of Mitochondrial Dynamics and Mitophagy in Carcinogenesis, Metastasis and Therapy. Front. Cell Dev. Biol. 2020, 8, 413. [Google Scholar] [CrossRef] [PubMed]
  10. Yang, Z.; Feng, Z.; Gu, J.; Li, X.; Dong, Q.; Liu, K.; Li, Y.; OuYang, L. microRNA-488 inhibits chemoresistance of ovarian cancer cells by targeting Six1 and mitochondrial function. Oncotarget 2017, 8, 80981–80993. [Google Scholar] [CrossRef] [Green Version]
  11. Osellame, L.D.; Blacker, T.S.; Duchen, M.R. Cellular and molecular mechanisms of mitochondrial function. Best Pract. Res. Clin. Endocrinol. Metab. 2012, 26, 711–723. [Google Scholar] [CrossRef] [Green Version]
  12. Alberts, B.; Johnson, A.; Lewis, J.; Raff, M.; Roberts, K.; Walter, P. The Mitochondrion. In Molecular Biology of the Cell, 4th ed.; Garland Science: New York, NY, USA, 2002. Available online: https://www.ncbi.nlm.nih.gov/books/NBK26894/ (accessed on 2 February 2022).
  13. Avendano-Monsalve, M.C.; Ponce-Rojas, J.C.; Funes, S. From cytosol to mitochondria: The beginning of a protein journey. Biol. Chem. 2020, 401, 645–661. [Google Scholar] [CrossRef]
  14. Scaglia, F.; Wong, L.J. Human mitochondrial transfer RNAs: Role of pathogenic mutation in disease. Muscle Nerve 2008, 37, 150–171. [Google Scholar] [CrossRef] [PubMed]
  15. Fox, T.D. Mitochondrial protein synthesis, import, and assembly. Genetics 2012, 192, 1203–1234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Seyfried, T.N.; Flores, R.E.; Poff, A.M.; D’Agostino, D.P. Cancer as a metabolic disease: Implications for novel therapeutics. Carcinogenesis 2014, 35, 515–527. [Google Scholar] [CrossRef]
  17. D’Souza, G.G.; Wagle, M.A.; Saxena, V.; Shah, A. Approaches for targeting mitochondria in cancer therapy. Biochim. Biophys. Acta 2011, 1807, 689–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Hiendleder, S.; Schmutz, S.M.; Erhardt, G.; Green, R.D.; Plante, Y. Transmitochondrial differences and varying levels of heteroplasmy in nuclear transfer cloned cattle. Mol. Reprod. Dev. 1999, 54, 24–31. [Google Scholar] [CrossRef]
  19. Gulbins, E.; Dreschers, S.; Bock, J. Role of mitochondria in apoptosis. Exp. Physiol. 2003, 88, 85–90. [Google Scholar] [CrossRef]
  20. Waterhouse, N.J.; Goldstein, J.C.; Kluck, R.M.; Newmeyer, D.D.; Green, D.R. The (Holey) study of mitochondria in apoptosis. Methods Cell Biol. 2001, 66, 365–391. [Google Scholar] [CrossRef]
  21. Bagur, R.; Hajnoczky, G. Intracellular Ca2+ Sensing: Its Role in Calcium Homeostasis and Signaling. Mol. Cell 2017, 66, 780–788. [Google Scholar] [CrossRef] [Green Version]
  22. Duchen, M.R. Mitochondria and calcium: From cell signalling to cell death. J. Physiol. 2000, 529 Pt 1, 57–68. [Google Scholar] [CrossRef]
  23. Sterea, A.M.; El Hiani, Y. The Role of Mitochondrial Calcium Signaling in the Pathophysiology of Cancer Cells. Adv. Exp. Med. Biol. 2020, 1131, 747–770. [Google Scholar] [CrossRef] [PubMed]
  24. Pinton, P.; Giorgi, C.; Siviero, R.; Zecchini, E.; Rizzuto, R. Calcium and apoptosis: ER-mitochondria Ca2+ transfer in the control of apoptosis. Oncogene 2008, 27, 6407–6418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Patergnani, S.; Danese, A.; Bouhamida, E.; Aguiari, G.; Previati, M.; Pinton, P.; Giorgi, C. Various Aspects of Calcium Signaling in the Regulation of Apoptosis, Autophagy, Cell Proliferation, and Cancer. Int. J. Mol. Sci. 2020, 21, 8323. [Google Scholar] [CrossRef]
  26. Zorov, D.B.; Juhaszova, M.; Sollott, S.J. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol. Rev. 2014, 94, 909–950. [Google Scholar] [CrossRef] [Green Version]
  27. Stowe, D.F.; Camara, A.K. Mitochondrial reactive oxygen species production in excitable cells: Modulators of mitochondrial and cell function. Antioxid. Redox Signal. 2009, 11, 1373–1414. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Shoshan-Barmatz, V.; Krelin, Y.; Shteinfer-Kuzmine, A. VDAC1 functions in Ca2+ homeostasis and cell life and death in health and disease. Cell Calcium 2018, 69, 81–100. [Google Scholar] [CrossRef]
  29. Shoshan-Barmatz, V.; Ben-Hail, D.; Admoni, L.; Krelin, Y.; Tripathi, S.S. The mitochondrial voltage-dependent anion channel 1 in tumor cells. Biochim. Biophys. Acta 2015, 1848, 2547–2575. [Google Scholar] [CrossRef] [Green Version]
  30. Shoshan-Barmatz, V.; Krelin, Y.; Shteinfer-Kuzmine, A.; Arif, T. Voltage-Dependent Anion Channel 1 As an Emerging Drug Target for Novel Anti-Cancer Therapeutics. Front. Oncol. 2017, 7, 154. [Google Scholar] [CrossRef]
  31. Camara, A.K.S.; Zhou, Y.; Wen, P.C.; Tajkhorshid, E.; Kwok, W.M. Mitochondrial VDAC1: A Key Gatekeeper as Potential Therapeutic Target. Front. Physiol. 2017, 8, 460. [Google Scholar] [CrossRef] [Green Version]
  32. Guzun, R.; Gonzalez-Granillo, M.; Karu-Varikmaa, M.; Grichine, A.; Usson, Y.; Kaambre, T.; Guerrero-Roesch, K.; Kuznetsov, A.; Schlattner, U.; Saks, V. Regulation of respiration in muscle cells in vivo by VDAC through interaction with the cytoskeleton and MtCK within Mitochondrial Interactosome. Biochim. Biophys. Acta 2012, 1818, 1545–1554. [Google Scholar] [CrossRef]
  33. Colombini, M. Voltage gating in the mitochondrial channel, VDAC. J. Membr. Biol. 1989, 111, 103–111. [Google Scholar] [CrossRef] [PubMed]
  34. Shoshan-Barmatz, V.; Zakar, M.; Rosenthal, K.; Abu-Hamad, S. Key regions of VDAC1 functioning in apoptosis induction and regulation by hexokinase. Biochim. Biophys. Acta 2009, 1787, 421–430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Hodge, T.; Colombini, M. Regulation of metabolite flux through voltage-gating of VDAC channels. J. Membr. Biol. 1997, 157, 271–279. [Google Scholar] [CrossRef]
  36. Vance, J.E. MAM (mitochondria-associated membranes) in mammalian cells: Lipids and beyond. Biochim. Biophys. Acta 2014, 1841, 595–609. [Google Scholar] [CrossRef] [PubMed]
  37. Gomez, L.; Thiebaut, P.A.; Paillard, M.; Ducreux, S.; Abrial, M.; Crola Da Silva, C.; Durand, A.; Alam, M.R.; Van Coppenolle, F.; Sheu, S.S.; et al. The SR/ER-mitochondria calcium crosstalk is regulated by GSK3beta during reperfusion injury. Cell Death Differ. 2016, 23, 313–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. De Stefani, D.; Bononi, A.; Romagnoli, A.; Messina, A.; De Pinto, V.; Pinton, P.; Rizzuto, R. VDAC1 selectively transfers apoptotic Ca2+ signals to mitochondria. Cell Death Differ. 2012, 19, 267–273. [Google Scholar] [CrossRef] [Green Version]
  39. Keinan, N.; Pahima, H.; Ben-Hail, D.; Shoshan-Barmatz, V. The role of calcium in VDAC1 oligomerization and mitochondria-mediated apoptosis. Biochim. Biophys. Acta 2013, 1833, 1745–1754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Camara, A.K.; Aldakkak, M.; Heisner, J.S.; Rhodes, S.S.; Riess, M.L.; An, J.; Heinen, A.; Stowe, D.F. ROS scavenging before 27 degrees C ischemia protects hearts and reduces mitochondrial ROS, Ca2+ overload, and changes in redox state. Am. J. Physiol. Cell Physiol. 2007, 292, C2021–C2031. [Google Scholar] [CrossRef]
  41. Camara, A.K.; Bienengraeber, M.; Stowe, D.F. Mitochondrial approaches to protect against cardiac ischemia and reperfusion injury. Front. Physiol. 2011, 2, 13. [Google Scholar] [CrossRef] [Green Version]
  42. Javadov, S.; Kuznetsov, A. Mitochondrial permeability transition and cell death: The role of cyclophilin d. Front. Physiol. 2013, 4, 76. [Google Scholar] [CrossRef] [Green Version]
  43. Javadov, S.; Karmazyn, M.; Escobales, N. Mitochondrial permeability transition pore opening as a promising therapeutic target in cardiac diseases. J. Pharmacol. Exp. Ther. 2009, 330, 670–678. [Google Scholar] [CrossRef] [Green Version]
  44. Azoulay-Zohar, H.; Israelson, A.; Abu-Hamad, S.; Shoshan-Barmatz, V. In self-defence: Hexokinase promotes voltage-dependent anion channel closure and prevents mitochondria-mediated apoptotic cell death. Biochem. J. 2004, 377, 347–355. [Google Scholar] [CrossRef]
  45. Zaid, H.; Abu-Hamad, S.; Israelson, A.; Nathan, I.; Shoshan-Barmatz, V. The voltage-dependent anion channel-1 modulates apoptotic cell death. Cell Death Differ. 2005, 12, 751–760. [Google Scholar] [CrossRef] [PubMed]
  46. Shimizu, S.; Konishi, A.; Kodama, T.; Tsujimoto, Y. BH4 domain of antiapoptotic Bcl-2 family members closes voltage-dependent anion channel and inhibits apoptotic mitoch.hondrial changes and cell death. Proc. Natl. Acad. Sci. USA 2000, 97, 3100–3105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Shimizu, S.; Matsuoka, Y.; Shinohara, Y.; Yoneda, Y.; Tsujimoto, Y. Essential role of voltage-dependent anion channel in various forms of apoptosis in mammalian cells. J. Cell Biol. 2001, 152, 237–250. [Google Scholar] [CrossRef]
  48. Tsujimoto, Y.; Shimizu, S. VDAC regulation by the Bcl-2 family of proteins. Cell Death Differ. 2000, 7, 1174–1181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Sun, Y.; Vashisht, A.A.; Tchieu, J.; Wohlschlegel, J.A.; Dreier, L. Voltage-dependent anion channels (VDACs) recruit Parkin to defective mitochondria to promote mitochondrial autophagy. J. Biol. Chem. 2012, 287, 40652–40660. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Kamer, K.J.; Mootha, V.K. The molecular era of the mitochondrial calcium uniporter. Nat. Rev. Mol. Cell Biol. 2015, 16, 545–553. [Google Scholar] [CrossRef]
  51. Chaudhuri, D.; Clapham, D.E. Outstanding questions regarding the permeation, selectivity, and regulation of the mitochondrial calcium uniporter. Biochem. Biophys. Res. Commun. 2014, 449, 367–369. [Google Scholar] [CrossRef]
  52. Pendin, D.; Greotti, E.; Pozzan, T. The elusive importance of being a mitochondrial Ca2+ uniporter. Cell Calcium 2014, 55, 139–145. [Google Scholar] [CrossRef]
  53. Giorgi, C.; Baldassari, F.; Bononi, A.; Bonora, M.; De Marchi, E.; Marchi, S.; Missiroli, S.; Patergnani, S.; Rimessi, A.; Suski, J.M.; et al. Mitochondrial Ca2+ and apoptosis. Cell Calcium 2012, 52, 36–43. [Google Scholar] [CrossRef] [Green Version]
  54. Hajnoczky, G.; Csordas, G.; Das, S.; Garcia-Perez, C.; Saotome, M.; Sinha Roy, S.; Yi, M. Mitochondrial calcium signalling and cell death: Approaches for assessing the role of mitochondrial Ca2+ uptake in apoptosis. Cell Calcium 2006, 40, 553–560. [Google Scholar] [CrossRef] [Green Version]
  55. Bonora, M.; Patergnani, S.; Rimessi, A.; De Marchi, E.; Suski, J.M.; Bononi, A.; Giorgi, C.; Marchi, S.; Missiroli, S.; Poletti, F.; et al. ATP synthesis and storage. Purinergic Signal. 2012, 8, 343–357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Mallilankaraman, K.; Cardenas, C.; Doonan, P.J.; Chandramoorthy, H.C.; Irrinki, K.M.; Golenar, T.; Csordas, G.; Madireddi, P.; Yang, J.; Muller, M.; et al. MCUR1 is an essential component of mitochondrial Ca2+ uptake that regulates cellular metabolism. Nat. Cell Biol. 2012, 14, 1336–1343. [Google Scholar] [CrossRef] [Green Version]
  57. Mallilankaraman, K.; Doonan, P.; Cardenas, C.; Chandramoorthy, H.C.; Muller, M.; Miller, R.; Hoffman, N.E.; Gandhirajan, R.K.; Molgo, J.; Birnbaum, M.J.; et al. MICU1 is an essential gatekeeper for MCU-mediated mitochondrial Ca2+ uptake that regulates cell survival. Cell 2012, 151, 630–644. [Google Scholar] [CrossRef] [Green Version]
  58. Alam, M.R.; Groschner, L.N.; Parichatikanond, W.; Kuo, L.; Bondarenko, A.I.; Rost, R.; Waldeck-Weiermair, M.; Malli, R.; Graier, W.F. Mitochondrial Ca2+ uptake 1 (MICU1) and mitochondrial Ca2+ uniporter (MCU) contribute to metabolism-secretion coupling in clonal pancreatic beta-cells. J. Biol. Chem. 2012, 287, 34445–34454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Tomar, D.; Dong, Z.; Shanmughapriya, S.; Koch, D.A.; Thomas, T.; Hoffman, N.E.; Timbalia, S.A.; Goldman, S.J.; Breves, S.L.; Corbally, D.P.; et al. MCUR1 Is a Scaffold Factor for the MCU Complex Function and Promotes Mitochondrial Bioenergetics. Cell Rep. 2016, 15, 1673–1685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Yamamoto, T.; Ozono, M.; Watanabe, A.; Maeda, K.; Nara, A.; Hashida, M.; Ido, Y.; Hiroshima, Y.; Yamada, A.; Terada, H.; et al. Functional analysis of coiled-coil domains of MCU in mitochondrial calcium uptake. Biochim. Biophys. Acta Bioenerg. 2019, 1860, 148061. [Google Scholar] [CrossRef] [PubMed]
  61. Garg, V.; Suzuki, J.; Paranjpe, I.; Unsulangi, T.; Boyman, L.; Milescu, L.S.; Lederer, W.J.; Kirichok, Y. The mechanism of MICU-dependent gating of the mitochondrial Ca2+uniporter. eLife 2021, 10, e69312. [Google Scholar] [CrossRef]
  62. Suh, D.H.; Kim, M.K.; Kim, H.S.; Chung, H.H.; Song, Y.S. Mitochondrial permeability transition pore as a selective target for anti-cancer therapy. Front. Oncol. 2013, 3, 41. [Google Scholar] [CrossRef] [Green Version]
  63. Brenner, C.; Grimm, S. The permeability transition pore complex in cancer cell death. Oncogene 2006, 25, 4744–4756. [Google Scholar] [CrossRef] [Green Version]
  64. Bernardi, P.; Krauskopf, A.; Basso, E.; Petronilli, V.; Blachly-Dyson, E.; Di Lisa, F.; Forte, M.A. The mitochondrial permeability transition from in vitro artifact to disease target. FEBS J. 2006, 273, 2077–2099. [Google Scholar] [CrossRef] [PubMed]
  65. Grimm, S.; Brdiczka, D. The permeability transition pore in cell death. Apoptosis 2007, 12, 841–855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Rasola, A.; Bernardi, P. The mitochondrial permeability transition pore and its adaptive responses in tumor cells. Cell Calcium 2014, 56, 437–445. [Google Scholar] [CrossRef] [PubMed]
  67. Kent, A.C.; El Baradie, K.B.Y.; Hamrick, M.W. Targeting the Mitochondrial Permeability Transition Pore to Prevent Age-Associated Cell Damage and Neurodegeneration. Oxid. Med. Cell. Longev. 2021, 2021, 6626484. [Google Scholar] [CrossRef]
  68. Zunino, S.J.; Storms, D.H. Resveratrol-induced apoptosis is enhanced in acute lymphoblastic leukemia cells by modulation of the mitochondrial permeability transition pore. Cancer Lett. 2006, 240, 123–134. [Google Scholar] [CrossRef]
  69. Ling, X.; Zhou, Y.; Li, S.W.; Yan, B.; Wen, L. Modulation of mitochondrial permeability transition pore affects multidrug resistance in human hepatocellular carcinoma cells. Int. J. Biol. Sci. 2010, 6, 773–783. [Google Scholar] [CrossRef] [Green Version]
  70. Huttemann, M.; Lee, I.; Samavati, L.; Yu, H.; Doan, J.W. Regulation of mitochondrial oxidative phosphorylation through cell signaling. Biochim. Biophys. Acta 2007, 1773, 1701–1720. [Google Scholar] [CrossRef] [Green Version]
  71. Garcia-Bermudez, J.; Cuezva, J.M. The ATPase Inhibitory Factor 1 (IF1): A master regulator of energy metabolism and of cell survival. Biochim. Biophys. Acta 2016, 1857, 1167–1182. [Google Scholar] [CrossRef]
  72. Seelert, H.; Dencher, N.A. ATP synthase superassemblies in animals and plants: Two or more are better. Biochim. Biophys. Acta 2011, 1807, 1185–1197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Kucharczyk, R.; Zick, M.; Bietenhader, M.; Rak, M.; Couplan, E.; Blondel, M.; Caubet, S.D.; di Rago, J.P. Mitochondrial ATP synthase disorders: Molecular mechanisms and the quest for curative therapeutic approaches. Biochim. Biophys. Acta 2009, 1793, 186–199. [Google Scholar] [CrossRef] [Green Version]
  74. Huang, T.C.; Chang, H.Y.; Hsu, C.H.; Kuo, W.H.; Chang, K.J.; Juan, H.F. Targeting therapy for breast carcinoma by ATP synthase inhibitor aurovertin B. J. Proteome Res. 2008, 7, 1433–1444. [Google Scholar] [CrossRef] [PubMed]
  75. Chang, H.Y.; Huang, H.C.; Huang, T.C.; Yang, P.C.; Wang, Y.C.; Juan, H.F. Ectopic ATP synthase blockade suppresses lung adenocarcinoma growth by activating the unfolded protein response. Cancer Res. 2012, 72, 4696–4706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Chang, H.Y.; Huang, T.C.; Chen, N.N.; Huang, H.C.; Juan, H.F. Combination therapy targeting ectopic ATP synthase and 26S proteasome induces ER stress in breast cancer cells. Cell Death Dis. 2014, 5, e1540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Zheng, J.; Ramirez, V.D. Inhibition of mitochondrial proton F0F1-ATPase/ATP synthase by polyphenolic phytochemicals. Br. J. Pharmacol. 2000, 130, 1115–1123. [Google Scholar] [CrossRef] [PubMed]
  78. Tuquet, C.; Dupont, J.; Mesneau, A.; Roussaux, J. Effects of tamoxifen on the electron transport chain of isolated rat liver mitochondria. Cell Biol. Toxicol 2000, 16, 207–219. [Google Scholar] [CrossRef]
  79. Kallio, A.; Zheng, A.; Dahllund, J.; Heiskanen, K.M.; Harkonen, P. Role of mitochondria in tamoxifen-induced rapid death of MCF-7 breast cancer cells. Apoptosis 2005, 10, 1395–1410. [Google Scholar] [CrossRef]
  80. Shchepina, L.A.; Pletjushkina, O.Y.; Avetisyan, A.V.; Bakeeva, L.E.; Fetisova, E.K.; Izyumov, D.S.; Saprunova, V.B.; Vyssokikh, M.Y.; Chernyak, B.V.; Skulachev, V.P. Oligomycin, inhibitor of the F0 part of H+-ATP-synthase, suppresses the TNF-induced apoptosis. Oncogene 2002, 21, 8149–8157. [Google Scholar] [CrossRef] [Green Version]
  81. Sena, L.A.; Chandel, N.S. Physiological roles of mitochondrial reactive oxygen species. Mol. Cell 2012, 48, 158–167. [Google Scholar] [CrossRef] [Green Version]
  82. Shadel, G.S.; Horvath, T.L. Mitochondrial ROS signaling in organismal homeostasis. Cell 2015, 163, 560–569. [Google Scholar] [CrossRef] [Green Version]
  83. Gammella, E.; Recalcati, S.; Cairo, G. Dual Role of ROS as Signal and Stress Agents: Iron Tips the Balance in favor of Toxic Effects. Oxid. Med. Cell. Longev. 2016, 2016, 8629024. [Google Scholar] [CrossRef] [Green Version]
  84. Diebold, L.; Chandel, N.S. Mitochondrial ROS regulation of proliferating cells. Free Radic. Biol. Med. 2016, 100, 86–93. [Google Scholar] [CrossRef]
  85. Mailloux, R.J. Teaching the fundamentals of electron transfer reactions in mitochondria and the production and detection of reactive oxygen species. Redox Biol. 2015, 4, 381–398. [Google Scholar] [CrossRef] [Green Version]
  86. Sipos, I.; Tretter, L.; Adam-Vizi, V. Quantitative relationship between inhibition of respiratory complexes and formation of reactive oxygen species in isolated nerve terminals. J. Neurochem. 2003, 84, 112–118. [Google Scholar] [CrossRef]
  87. Zhen, D.; Chen, Y.; Tang, X. Metformin reverses the deleterious effects of high glucose on osteoblast function. J. Diabetes Complicat. 2010, 24, 334–344. [Google Scholar] [CrossRef] [PubMed]
  88. Murphy, M.P. How mitochondria produce reactive oxygen species. Biochem. J. 2009, 417, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Weinberg, F.; Hamanaka, R.; Wheaton, W.W.; Weinberg, S.; Joseph, J.; Lopez, M.; Kalyanaraman, B.; Mutlu, G.M.; Budinger, G.R.; Chandel, N.S. Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc. Natl. Acad. Sci. USA 2010, 107, 8788–8793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Kohen, R.; Nyska, A. Oxidation of biological systems: Oxidative stress phenomena, antioxidants, redox reactions, and methods for their quantification. Toxicol. Pathol. 2002, 30, 620–650. [Google Scholar] [CrossRef] [Green Version]
  91. Kruspig, B.; Valter, K.; Skender, B.; Zhivotovsky, B.; Gogvadze, V. Targeting succinate:ubiquinone reductase potentiates the efficacy of anticancer therapy. Biochim. Biophys. Acta 2016, 1863, 2065–2071. [Google Scholar] [CrossRef] [PubMed]
  92. Di Meo, S.; Reed, T.T.; Venditti, P.; Victor, V.M. Role of ROS and RNS Sources in Physiological and Pathological Conditions. Oxid. Med. Cell. Longev. 2016, 2016, 1245049. [Google Scholar] [CrossRef] [PubMed]
  93. Sullivan, L.B.; Chandel, N.S. Mitochondrial reactive oxygen species and cancer. Cancer Metab. 2014, 2, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Schenk, B.; Fulda, S. Reactive oxygen species regulate Smac mimetic/TNFalpha-induced necroptotic signaling and cell death. Oncogene 2015, 34, 5796–5806. [Google Scholar] [CrossRef] [PubMed]
  95. Chandel, N.S.; Tuveson, D.A. The promise and perils of antioxidants for cancer patients. N. Engl. J. Med. 2014, 371, 177–178. [Google Scholar] [CrossRef] [PubMed]
  96. Dall’Acqua, S.; Linardi, M.A.; Bortolozzi, R.; Clauser, M.; Marzocchini, S.; Maggi, F.; Nicoletti, M.; Innocenti, G.; Basso, G.; Viola, G. Natural daucane esters induces apoptosis in leukaemic cells through ROS production. Phytochemistry 2014, 108, 147–156. [Google Scholar] [CrossRef]
  97. Castaldo, S.A.; Freitas, J.R.; Conchinha, N.V.; Madureira, P.A. The Tumorigenic Roles of the Cellular REDOX Regulatory Systems. Oxid. Med. Cell. Longev. 2016, 2016, 8413032. [Google Scholar] [CrossRef] [Green Version]
  98. Isaia, G.C.; Limone, P.; Bodrato, L.; Avataneo, T.; Potenzoni, F.; Gremo, L.; Molinatti, G.M. Is magnetic resonance imaging more sensitive than computed tomography in the location of corticotropin secreting pituitary adenomas? Ann. Ital. Med. Int. 1989, 4, 122–124. [Google Scholar]
  99. Hayes, J.D.; McMahon, M. NRF2 and KEAP1 mutations: Permanent activation of an adaptive response in cancer. Trends Biochem. Sci. 2009, 34, 176–188. [Google Scholar] [CrossRef]
  100. Greenlee, H.; Kwan, M.L.; Kushi, L.H.; Song, J.; Castillo, A.; Weltzien, E.; Quesenberry, C.P., Jr.; Caan, B.J. Antioxidant supplement use after breast cancer diagnosis and mortality in the Life After Cancer Epidemiology (LACE) cohort. Cancer 2012, 118, 2048–2058. [Google Scholar] [CrossRef] [Green Version]
  101. Harris, H.R.; Orsini, N.; Wolk, A. Vitamin C and survival among women with breast cancer: A meta-analysis. Eur. J. Cancer 2014, 50, 1223–1231. [Google Scholar] [CrossRef]
  102. Bober, P.; Alexovic, M.; Talian, I.; Tomkova, Z.; Viscorova, Z.; Benckova, M.; Andrasina, I.; Ciccocioppo, R.; Petrovic, D.; Adamek, M.; et al. Proteomic analysis of the vitamin C effect on the doxorubicin cytotoxicity in the MCF-7 breast cancer cell line. J. Cancer Res. Clin. Oncol. 2017, 143, 35–42. [Google Scholar] [CrossRef]
  103. Printz, C. High vitamin D levels increase survival rates in patients with metastatic colorectal cancer. Cancer 2015, 121, 2105. [Google Scholar] [CrossRef]
  104. DiMauro, S. Mitochondrial diseases. Biochim. Biophys. Acta 2004, 1658, 80–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Wisnovsky, S.P.; Wilson, J.J.; Radford, R.J.; Pereira, M.P.; Chan, M.R.; Laposa, R.R.; Lippard, S.J.; Kelley, S.O. Targeting mitochondrial DNA with a platinum-based anticancer agent. Chem. Biol. 2013, 20, 1323–1328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Cullen, K.J.; Yang, Z.; Schumaker, L.; Guo, Z. Mitochondria as a critical target of the chemotheraputic agent cisplatin in head and neck cancer. J. Bioenerg. Biomembr. 2007, 39, 43–50. [Google Scholar] [CrossRef] [PubMed]
  107. Murata, T.; Hibasami, H.; Maekawa, S.; Tagawa, T.; Nakashima, K. Preferential binding of cisplatin to mitochondrial DNA and suppression of ATP generation in human malignant melanoma cells. Biochem. Int. 1990, 20, 949–955. [Google Scholar]
  108. Yang, Z.; Schumaker, L.M.; Egorin, M.J.; Zuhowski, E.G.; Guo, Z.; Cullen, K.J. Cisplatin preferentially binds mitochondrial DNA and voltage-dependent anion channel protein in the mitochondrial membrane of head and neck squamous cell carcinoma: Possible role in apoptosis. Clin. Cancer Res. 2006, 12, 5817–5825. [Google Scholar] [CrossRef] [Green Version]
  109. Lomeli, N.; Di, K.; Czerniawski, J.; Guzowski, J.F.; Bota, D.A. Cisplatin-induced mitochondrial dysfunction is associated with impaired cognitive function in rats. Free Radic. Biol. Med. 2017, 102, 274–286. [Google Scholar] [CrossRef] [Green Version]
  110. Podratz, J.L.; Knight, A.M.; Ta, L.E.; Staff, N.P.; Gass, J.M.; Genelin, K.; Schlattau, A.; Lathroum, L.; Windebank, A.J. Cisplatin induced mitochondrial DNA damage in dorsal root ganglion neurons. Neurobiol. Dis. 2011, 41, 661–668. [Google Scholar] [CrossRef] [Green Version]
  111. Montopoli, M.; Bellanda, M.; Lonardoni, F.; Ragazzi, E.; Dorigo, P.; Froldi, G.; Mammi, S.; Caparrotta, L. “Metabolic reprogramming” in ovarian cancer cells resistant to cisplatin. Curr. Cancer Drug Targets 2011, 11, 226–235. [Google Scholar] [CrossRef]
  112. Copeland, W.C. Inherited mitochondrial diseases of DNA replication. Annu. Rev. Med. 2008, 59, 131–146. [Google Scholar] [CrossRef] [Green Version]
  113. Hance, N.; Ekstrand, M.I.; Trifunovic, A. Mitochondrial DNA polymerase gamma is essential for mammalian embryogenesis. Hum. Mol. Genet. 2005, 14, 1775–1783. [Google Scholar] [CrossRef] [PubMed]
  114. Cohen, B.H.; Naviaux, R.K. The clinical diagnosis of POLG disease and other mitochondrial DNA depletion disorders. Methods 2010, 51, 364–373. [Google Scholar] [CrossRef] [PubMed]
  115. Singh, K.K.; Ayyasamy, V.; Owens, K.M.; Koul, M.S.; Vujcic, M. Mutations in mitochondrial DNA polymerase-gamma promote breast tumorigenesis. J. Hum. Genet. 2009, 54, 516–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Armstrong, J.S. Mitochondrial medicine: Pharmacological targeting of mitochondria in disease. Br. J. Pharmacol. 2007, 151, 1154–1165. [Google Scholar] [CrossRef] [PubMed]
  117. Singh, B.; Owens, K.M.; Bajpai, P.; Desouki, M.M.; Srinivasasainagendra, V.; Tiwari, H.K.; Singh, K.K. Mitochondrial DNA Polymerase POLG1 Disease Mutations and Germline Variants Promote Tumorigenic Properties. PLoS ONE 2015, 10, e0139846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Copeland, W.C.; Longley, M.J. DNA polymerase gamma in mitochondrial DNA replication and repair. Sci. World J. 2003, 3, 34–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Pinz, K.G.; Bogenhagen, D.F. The influence of the DNA polymerase gamma accessory subunit on base excision repair by the catalytic subunit. DNA Repair. 2006, 5, 121–128. [Google Scholar] [CrossRef]
  120. Yehudai, D.; Liyanage, S.U.; Hurren, R.; Rizoska, B.; Albertella, M.; Gronda, M.; Jeyaraju, D.V.; Wang, X.; Barghout, S.H.; MacLean, N.; et al. The thymidine dideoxynucleoside analog, alovudine, inhibits the mitochondrial DNA polymerase gamma, impairs oxidative phosphorylation and promotes monocytic differentiation in acute myeloid leukemia. Haematologica 2019, 104, 963–972. [Google Scholar] [CrossRef] [Green Version]
  121. Chan, S.S.; Copeland, W.C. DNA polymerase gamma and mitochondrial disease: Understanding the consequence of POLG mutations. Biochim. Biophys. Acta 2009, 1787, 312–319. [Google Scholar] [CrossRef] [Green Version]
  122. Attardi, G. Animal mitochondrial DNA: An extreme example of genetic economy. Int. Rev. Cytol. 1985, 93, 93–145. [Google Scholar] [CrossRef]
  123. Chomyn, A.; Cleeter, M.W.; Ragan, C.I.; Riley, M.; Doolittle, R.F.; Attardi, G. URF6, last unidentified reading frame of human mtDNA, codes for an NADH dehydrogenase subunit. Science 1986, 234, 614–618. [Google Scholar] [CrossRef] [PubMed]
  124. Pecoraro, A.; Pagano, M.; Russo, G.; Russo, A. Ribosome Biogenesis and Cancer: Overview on Ribosomal Proteins. Int. J. Mol. Sci. 2021, 22, 5496. [Google Scholar] [CrossRef] [PubMed]
  125. Cavdar Koc, E.; Ranasinghe, A.; Burkhart, W.; Blackburn, K.; Koc, H.; Moseley, A.; Spremulli, L.L. A new face on apoptosis: Death-associated protein 3 and PDCD9 are mitochondrial ribosomal proteins. FEBS Lett. 2001, 492, 166–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Miller, J.L.; Koc, H.; Koc, E.C. Identification of phosphorylation sites in mammalian mitochondrial ribosomal protein DAP3. Protein Sci. 2008, 17, 251–260. [Google Scholar] [CrossRef] [Green Version]
  127. Yoo, Y.A.; Kim, M.J.; Park, J.K.; Chung, Y.M.; Lee, J.H.; Chi, S.G.; Kim, J.S.; Yoo, Y.D. Mitochondrial ribosomal protein L41 suppresses cell growth in association with p53 and p27Kip1. Mol. Cell. Biol. 2005, 25, 6603–6616. [Google Scholar] [CrossRef] [Green Version]
  128. Saini, N.; Balhara, J.; Adlakha, Y.K. Singh N: S29 Ribosomal protein induces mitochondria mediated apoptosis of Hep2 cells via the activation of p38 MAPK and JNK signaling. Int. J. Integr. Biol. 2009, 5, 49–57. [Google Scholar]
  129. Garand, C.; Guay, D.; Sereduk, C.; Chow, D.; Tsofack, S.P.; Langlois, M.; Perreault, E.; Yin, H.H.; Lebel, M. An integrative approach to identify YB-1-interacting proteins required for cisplatin resistance in MCF7 and MDA-MB-231 breast cancer cells. Cancer Sci. 2011, 102, 1410–1417. [Google Scholar] [CrossRef]
  130. Tsofack, S.P.; Meunier, L.; Sanchez, L.; Madore, J.; Provencher, D.; Mes-Masson, A.M.; Lebel, M. Low expression of the X-linked ribosomal protein S4 in human serous epithelial ovarian cancer is associated with a poor prognosis. BMC Cancer 2013, 13, 303. [Google Scholar] [CrossRef] [Green Version]
  131. Sun, L.; Liu, Y.; Fremont, M.; Schwarz, S.; Siegmann, M.; Matthies, R.; Jost, J.P. A novel 52 kDa protein induces apoptosis and concurrently activates c-Jun N-terminal kinase 1 (JNK1) in mouse C3H10T1/2 fibroblasts. Gene 1998, 208, 157–166. [Google Scholar] [CrossRef]
  132. Gilkerson, R.W.; Margineantu, D.H.; Capaldi, R.A.; Selker, J.M. Mitochondrial DNA depletion causes morphological changes in the mitochondrial reticulum of cultured human cells. FEBS Lett. 2000, 474, 1–4. [Google Scholar] [CrossRef] [Green Version]
  133. Kale, J.; Osterlund, E.J.; Andrews, D.W. BCL-2 family proteins: Changing partners in the dance towards death. Cell Death Differ. 2018, 25, 65–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Lomonosova, E.; Chinnadurai, G. BH3-only proteins in apoptosis and beyond: An overview. Oncogene 2008, 27 (Suppl. S1), S2–S19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Chipuk, J.E.; Moldoveanu, T.; Llambi, F.; Parsons, M.J.; Green, D.R. The BCL-2 family reunion. Mol. Cell 2010, 37, 299–310. [Google Scholar] [CrossRef] [PubMed]
  136. Tait, S.W.; Green, D.R. Mitochondria and cell death: Outer membrane permeabilization and beyond. Nat. Rev. Mol. Cell Biol. 2010, 11, 621–632. [Google Scholar] [CrossRef] [PubMed]
  137. Deng, Y.; Lin, Y.; Wu, X. TRAIL-induced apoptosis requires Bax-dependent mitochondrial release of Smac/DIABLO. Genes Dev. 2002, 16, 33–45. [Google Scholar] [CrossRef] [Green Version]
  138. Renault, T.T.; Floros, K.V.; Chipuk, J.E. BAK/BAX activation and cytochrome c release assays using isolated mitochondria. Methods 2013, 61, 146–155. [Google Scholar] [CrossRef] [Green Version]
  139. McIlwain, D.R.; Berger, T.; Mak, T.W. Caspase functions in cell death and disease. Cold Spring Harb. Perspect. Biol. 2015, 7, a026716. [Google Scholar] [CrossRef] [Green Version]
  140. Kesavardhana, S.; Malireddi, R.K.S.; Kanneganti, T.D. Caspases in cell death, inflammation, and pyroptosis. Annu. Rev. Immunol. 2020, 38, 567–595. [Google Scholar] [CrossRef] [Green Version]
  141. Imai, K.; Takaoka, A. Comparing antibody and small-molecule therapies for cancer. Nat. Rev. Cancer 2006, 6, 714–727. [Google Scholar] [CrossRef]
  142. Bender, A.; Opel, D.; Naumann, I.; Kappler, R.; Friedman, L.; von Schweinitz, D.; Debatin, K.M.; Fulda, S. PI3K inhibitors prime neuroblastoma cells for chemotherapy by shifting the balance towards pro-apoptotic Bcl-2 proteins and enhanced mitochondrial apoptosis. Oncogene 2011, 30, 494–503. [Google Scholar] [CrossRef] [Green Version]
  143. Deng, J.; Shimamura, T.; Perera, S.; Carlson, N.E.; Cai, D.; Shapiro, G.I.; Wong, K.K.; Letai, A. Proapoptotic BH3-only BCL-2 family protein BIM connects death signaling from epidermal growth factor receptor inhibition to the mitochondrion. Cancer Res. 2007, 67, 11867–11875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Gojo, I.; Zhang, B.; Fenton, R.G. The cyclin-dependent kinase inhibitor flavopiridol induces apoptosis in multiple myeloma cells through transcriptional repression and down-regulation of Mcl-1. Clin. Cancer Res. 2002, 8, 3527–3538. [Google Scholar] [PubMed]
  145. Sotgia, F.; Whitaker-Menezes, D.; Martinez-Outschoorn, U.E.; Flomenberg, N.; Birbe, R.C.; Witkiewicz, A.K.; Howell, A.; Philp, N.J.; Pestell, R.G.; Lisanti, M.P. Mitochondrial metabolism in cancer metastasis: Visualizing tumor cell mitochondria and the “reverse Warburg effect” in positive lymph node tissue. Cell Cycle 2012, 11, 1445–1454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Pearce, E.L.; Pearce, E.J. Metabolic pathways in immune cell activation and quiescence. Immunity 2013, 38, 633–643. [Google Scholar] [CrossRef] [Green Version]
  147. Zheng, J. Energy metabolism of cancer: Glycolysis versus oxidative phosphorylation (Review). Oncol. Lett. 2012, 4, 1151–1157. [Google Scholar] [CrossRef] [Green Version]
  148. Martinez-Outschoorn, U.E.; Pavlides, S.; Sotgia, F.; Lisanti, M.P. Mitochondrial biogenesis drives tumor cell proliferation. Am. J. Pathol. 2011, 178, 1949–1952. [Google Scholar] [CrossRef] [Green Version]
  149. Rodriguez, C.; Puente-Moncada, N.; Reiter, R.J.; Sanchez-Sanchez, A.M.; Herrera, F.; Rodriguez-Blanco, J.; Duarte-Olivenza, C.; Turos-Cabal, M.; Antolin, I.; Martin, V. Regulation of cancer cell glucose metabolism is determinant for cancer cell fate after melatonin administration. J. Cell. Physiol. 2021, 236, 27–40. [Google Scholar] [CrossRef]
  150. Callao, V.; Montoya, E. Toxohormone-like factor from microorganisms with impaired respiration. Science 1961, 134, 2041–2042. [Google Scholar] [CrossRef]
  151. Altman, B.J.; Stine, Z.E.; Dang, C.V. From Krebs to clinic: Glutamine metabolism to cancer therapy. Nat. Rev. Cancer 2016, 16, 749. [Google Scholar] [CrossRef]
  152. Akram, M. Mini-review on glycolysis and cancer. J. Cancer Educ. 2013, 28, 454–457. [Google Scholar] [CrossRef]
  153. Pelicano, H.; Martin, D.S.; Xu, R.H.; Huang, P. Glycolysis inhibition for anticancer treatment. Oncogene 2006, 25, 4633–4646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Xu, R.H.; Pelicano, H.; Zhou, Y.; Carew, J.S.; Feng, L.; Bhalla, K.N.; Keating, M.J.; Huang, P. Inhibition of glycolysis in cancer cells: A novel strategy to overcome drug resistance associated with mitochondrial respiratory defect and hypoxia. Cancer Res. 2005, 65, 613–621. [Google Scholar] [PubMed]
  155. Wood, T.E.; Dalili, S.; Simpson, C.D.; Hurren, R.; Mao, X.; Saiz, F.S.; Gronda, M.; Eberhard, Y.; Minden, M.D.; Bilan, P.J.; et al. A novel inhibitor of glucose uptake sensitizes cells to FAS-induced cell death. Mol. Cancer Ther. 2008, 7, 3546–3555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Kraus, D.; Reckenbeil, J.; Veit, N.; Kuerpig, S.; Meisenheimer, M.; Beier, I.; Stark, H.; Winter, J.; Probstmeier, R. Targeting glucose transport and the NAD pathway in tumor cells with STF-31: A re-evaluation. Cell. Oncol. 2018, 41, 485–494. [Google Scholar] [CrossRef]
  157. Chen, X.S.; Li, L.Y.; Guan, Y.D.; Yang, J.M.; Cheng, Y. Anticancer strategies based on the metabolic profile of tumor cells: Therapeutic targeting of the Warburg effect. Acta Pharmacol. Sin. 2016, 37, 1013–1019. [Google Scholar] [CrossRef] [PubMed]
  158. Walcher, W.; Petru, E. The obstetric risk in adolescent primiparous females. Geburtshilfe Frauenheilkd. 1989, 49, 491–493. [Google Scholar] [CrossRef]
  159. Hadjiolov, D. Glucose-6-phosphatase activity in primary rat liver tumors induced by high doses of 4-dimethylaminoazo-benzene. Z. Krebsforsch. 1969, 72, 43–46. [Google Scholar] [CrossRef]
  160. Weber, G.; Cantero, A. Glucose-6-phosphatase activity in normal, pre-cancerous, and neoplastic tissues. Cancer Res. 1955, 15, 105–108. [Google Scholar]
  161. Sancho, P.; Barneda, D.; Heeschen, C. Hallmarks of cancer stem cell metabolism. Br. J. Cancer 2016, 114, 1305–1312. [Google Scholar] [CrossRef] [Green Version]
  162. Amadori, D.; Frassineti, G.L.; De Matteis, A.; Mustacchi, G.; Santoro, A.; Cariello, S.; Ferrari, M.; Nascimben, O.; Nanni, O.; Lombardi, A.; et al. Modulating effect of lonidamine on response to doxorubicin in metastatic breast cancer patients: Results from a multicenter prospective randomized trial. Breast Cancer Res. Treat. 1998, 49, 209–217. [Google Scholar] [CrossRef]
  163. Nath, K.; Nelson, D.S.; Heitjan, D.F.; Leeper, D.B.; Zhou, R.; Glickson, J.D. Lonidamine induces intracellular tumor acidification and ATP depletion in breast, prostate and ovarian cancer xenografts and potentiates response to doxorubicin. NMR Biomed. 2015, 28, 281–290. [Google Scholar] [CrossRef] [Green Version]
  164. Kroemer, G.; Pouyssegur, J. Tumor cell metabolism: Cancer’s Achilles’ heel. Cancer Cell 2008, 13, 472–482. [Google Scholar] [CrossRef] [PubMed]
  165. Weinberg, S.E.; Chandel, N.S. Targeting mitochondria metabolism for cancer therapy. Nat. Chem. Biol. 2015, 11, 9–15. [Google Scholar] [CrossRef] [Green Version]
  166. Hatzivassiliou, G.; Zhao, F.; Bauer, D.E.; Andreadis, C.; Shaw, A.N.; Dhanak, D.; Hingorani, S.R.; Tuveson, D.A.; Thompson, C.B. ATP citrate lyase inhibition can suppress tumor cell growth. Cancer Cell 2005, 8, 311–321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. 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] [PubMed]
  168. Griguer, C.E.; Oliva, C.R.; Gillespie, G.Y. Glucose metabolism heterogeneity in human and mouse malignant glioma cell lines. J. Neurooncol. 2005, 74, 123–133. [Google Scholar] [CrossRef] [PubMed]
  169. Scott, D.A.; Richardson, A.D.; Filipp, F.V.; Knutzen, C.A.; Chiang, G.G.; Ronai, Z.A.; Osterman, A.L.; Smith, J.W. Comparative metabolic flux profiling of melanoma cell lines: Beyond the Warburg effect. J. Biol. Chem. 2011, 286, 42626–42634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Hsu, P.P.; Sabatini, D.M. Cancer cell metabolism: Warburg and beyond. Cell 2008, 134, 703–707. [Google Scholar] [CrossRef] [Green Version]
  171. Menendez, J.A.; Lupu, R. Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat. Rev. Cancer 2007, 7, 763–777. [Google Scholar] [CrossRef] [PubMed]
  172. Christofk, H.R.; Vander Heiden, M.G.; Harris, M.H.; Ramanathan, A.; Gerszten, R.E.; Wei, R.; Fleming, M.D.; Schreiber, S.L.; Cantley, L.C. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature 2008, 452, 230–233. [Google Scholar] [CrossRef]
  173. Christofk, H.R.; Vander Heiden, M.G.; Wu, N.; Asara, J.M.; Cantley, L.C. Pyruvate kinase M2 is a phosphotyrosine-binding protein. Nature 2008, 452, 181–186. [Google Scholar] [CrossRef]
  174. Koritzinsky, M. Metformin: A Novel Biological Modifier of Tumor Response to Radiation Therapy. Int. J. Radiat. Oncol. Biol. Phys. 2015, 93, 454–464. [Google Scholar] [CrossRef] [PubMed]
  175. 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] [PubMed] [Green Version]
  176. Vitiello, G.A.; Medina, B.D.; Zeng, S.; Bowler, T.G.; Zhang, J.Q.; Loo, J.K.; Param, N.J.; Liu, M.; Moral, A.J.; Zhao, J.N.; et al. Mitochondrial Inhibition Augments the Efficacy of Imatinib by Resetting the Metabolic Phenotype of Gastrointestinal Stromal Tumor. Clin. Cancer Res. 2018, 24, 972–984. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Kinnaird, A.; Dromparis, P.; Saleme, B.; Gurtu, V.; Watson, K.; Paulin, R.; Zervopoulos, S.; Stenson, T.; Sutendra, G.; Pink, D.B.; et al. Metabolic Modulation of Clear-cell Renal Cell Carcinoma with Dichloroacetate, an Inhibitor of Pyruvate Dehydrogenase Kinase. Eur. Urol. 2016, 69, 734–744. [Google Scholar] [CrossRef]
  178. Stacpoole, P.W. Therapeutic Targeting of the Pyruvate Dehydrogenase Complex/Pyruvate Dehydrogenase Kinase (PDC/PDK) Axis in Cancer. J. Natl. Cancer Inst. 2017, 109, djx071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Sutendra, G.; Michelakis, E.D. Pyruvate dehydrogenase kinase as a novel therapeutic target in oncology. Front. Oncol. 2013, 3, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Velpula, K.K.; Bhasin, A.; Asuthkar, S.; Tsung, A.J. Combined targeting of PDK1 and EGFR triggers regression of glioblastoma by reversing the Warburg effect. Cancer Res. 2013, 73, 7277–7289. [Google Scholar] [CrossRef] [Green Version]
  181. 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] [Green Version]
  182. Yadav, S.; Bhagat, S.D.; Gupta, A.; Samaiya, A.; Srivastava, A.; Shukla, S. Dietary-phytochemical mediated reversion of cancer-specific splicing inhibits Warburg effect in head and neck cancer. BMC Cancer 2019, 19, 1031. [Google Scholar] [CrossRef]
  183. Zu, X.L.; Guppy, M. Cancer metabolism: Facts, fantasy, and fiction. Biochem. Biophys. Res. Commun. 2004, 313, 459–465. [Google Scholar] [CrossRef] [PubMed]
  184. Vaupel, P.; Mayer, A. Availability, not respiratory capacity governs oxygen consumption of solid tumors. Int. J. Biochem. Cell Biol. 2012, 44, 1477–1481. [Google Scholar] [CrossRef] [PubMed]
  185. Yu, M. Generation, function and diagnostic value of mitochondrial DNA copy number alterations in human cancers. Life Sci. 2011, 89, 65–71. [Google Scholar] [CrossRef] [PubMed]
  186. Reznik, E.; Miller, M.L.; Senbabaoglu, Y.; Riaz, N.; Sarungbam, J.; Tickoo, S.K.; Al-Ahmadie, H.A.; Lee, W.; Seshan, V.E.; Hakimi, A.A.; et al. Mitochondrial DNA copy number variation across human cancers. eLife 2016, 5, e10769. [Google Scholar] [CrossRef] [PubMed]
  187. DiMauro, S.; Garone, C.; Naini, A. Metabolic myopathies. Curr. Rheumatol. Rep. 2010, 12, 386–393. [Google Scholar] [CrossRef] [PubMed]
  188. DiMauro, S. Pathogenesis and treatment of mitochondrial myopathies: Recent advances. Acta Myol. 2010, 29, 333–338. [Google Scholar]
  189. Dimauro, S.; Rustin, P. A critical approach to the therapy of mitochondrial respiratory chain and oxidative phosphorylation diseases. Biochim. Biophys. Acta 2009, 1792, 1159–1167. [Google Scholar] [CrossRef] [Green Version]
  190. Bao, B.; Azmi, A.S.; Ali, S.; Zaiem, F.; Sarkar, F.H. Metformin may function as anti-cancer agent via targeting cancer stem cells: The potential biological significance of tumor-associated miRNAs in breast and pancreatic cancers. Ann. Transl. Med. 2014, 2, 59. [Google Scholar] [CrossRef]
  191. Brunmair, B.; Staniek, K.; Gras, F.; Scharf, N.; Althaym, A.; Clara, R.; Roden, M.; Gnaiger, E.; Nohl, H.; Waldhausl, W.; et al. Thiazolidinediones, like metformin, inhibit respiratory complex I: A common mechanism contributing to their antidiabetic actions? Diabetes 2004, 53, 1052–1059. [Google Scholar] [CrossRef] [Green Version]
  192. Owen, M.R.; Doran, E.; Halestrap, A.P. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem. J. 2000, 348 Pt 3, 607–614. [Google Scholar] [CrossRef]
  193. Taddia, L.; D’Arca, D.; Ferrari, S.; Marraccini, C.; Severi, L.; Ponterini, G.; Assaraf, Y.G.; Marverti, G.; Costi, M.P. Inside the biochemical pathways of thymidylate synthase perturbed by anticancer drugs: Novel strategies to overcome cancer chemoresistance. Drug Resist. Updates 2015, 23, 20–54. [Google Scholar] [CrossRef]
  194. Apicella, M.; Giannoni, E.; Fiore, S.; Ferrari, K.J.; Fernandez-Perez, D.; Isella, C.; Granchi, C.; Minutolo, F.; Sottile, A.; Comoglio, P.M.; et al. Increased Lactate Secretion by Cancer Cells Sustains Non-cell-autonomous Adaptive Resistance to MET and EGFR Targeted Therapies. Cell Metab. 2018, 28, 848–865.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Icard, P.; Shulman, S.; Farhat, D.; Steyaert, J.M.; Alifano, M.; Lincet, H. How the Warburg effect supports aggressiveness and drug resistance of cancer cells? Drug Resist. Updates 2018, 38, 1–11. [Google Scholar] [CrossRef] [PubMed]
  196. Xu, M.; Chen, S.; Yang, W.; Cheng, X.; Ye, Y.; Mao, J.; Wu, X.; Huang, L.; Ji, J. FGFR4 Links Glucose Metabolism and Chemotherapy Resistance in Breast Cancer. Cell. Physiol. Biochem. 2018, 47, 151–160. [Google Scholar] [CrossRef] [Green Version]
  197. Pelicano, H.; Zhang, W.; Liu, J.; Hammoudi, N.; Dai, J.; Xu, R.H.; Pusztai, L.; Huang, P. Mitochondrial dysfunction in some triple-negative breast cancer cell lines: Role of mTOR pathway and therapeutic potential. Breast Cancer Res. 2014, 16, 434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  198. Varghese, E.; Samuel, S.M.; Liskova, A.; Samec, M.; Kubatka, P.; Busselberg, D. Targeting Glucose Metabolism to Overcome Resistance to Anticancer Chemotherapy in Breast Cancer. Cancers 2020, 12, 2252. [Google Scholar] [CrossRef]
  199. Samuel, S.M.; Varghese, E.; Koklesova, L.; Liskova, A.; Kubatka, P.; Busselberg, D. Counteracting Chemoresistance with Metformin in Breast Cancers: Targeting Cancer Stem Cells. Cancers 2020, 12, 2482. [Google Scholar] [CrossRef]
  200. Peng, J.; Cui, Y.; Xu, S.; Wu, X.; Huang, Y.; Zhou, W.; Wang, S.; Fu, Z.; Xie, H. Altered glycolysis results in drug-resistant in clinical tumor therapy. Oncol. Lett. 2021, 21, 369. [Google Scholar] [CrossRef] [PubMed]
  201. Liu, X.; Miao, W.; Huang, M.; Li, L.; Dai, X.; Wang, Y. Elevated Hexokinase II Expression Confers Acquired Resistance to 4-Hydroxytamoxifen in Breast Cancer Cells. Mol. Cell. Proteom. 2019, 18, 2273–2284. [Google Scholar] [CrossRef] [PubMed]
  202. Krasnov, G.S.; Dmitriev, A.A.; Lakunina, V.A.; Kirpiy, A.A.; Kudryavtseva, A.V. Targeting VDAC-bound hexokinase II: A promising approach for concomitant anti-cancer therapy. Expert Opin. Ther. Targets 2013, 17, 1221–1233. [Google Scholar] [CrossRef]
  203. Catanzaro, D.; Gabbia, D.; Cocetta, V.; Biagi, M.; Ragazzi, E.; Montopoli, M.; Carrara, M. Silybin counteracts doxorubicin resistance by inhibiting GLUT1 expression. Fitoterapia 2018, 124, 42–48. [Google Scholar] [CrossRef]
  204. Scambia, G.; De Vincenzo, R.; Ranelletti, F.O.; Panici, P.B.; Ferrandina, G.; D’Agostino, G.; Fattorossi, A.; Bombardelli, E.; Mancuso, S. Antiproliferative effect of silybin on gynaecological malignancies: Synergism with cisplatin and doxorubicin. Eur. J. Cancer 1996, 32, 877–882. [Google Scholar] [CrossRef]
  205. Colombo, V.; Lupi, M.; Falcetta, F.; Forestieri, D.; D’Incalci, M.; Ubezio, P. Chemotherapeutic activity of silymarin combined with doxorubicin or paclitaxel in sensitive and multidrug-resistant colon cancer cells. Cancer Chemother. Pharmacol. 2011, 67, 369–379. [Google Scholar] [CrossRef] [PubMed]
  206. Chuang, J.H.; Chou, M.H.; Tai, M.H.; Lin, T.K.; Liou, C.W.; Chen, T.; Hsu, W.M.; Wang, P.W. 2-Deoxyglucose treatment complements the cisplatin- or BH3-only mimetic-induced suppression of neuroblastoma cell growth. Int. J. Biochem. Cell Biol. 2013, 45, 944–951. [Google Scholar] [CrossRef] [PubMed]
  207. Maschek, G.; Savaraj, N.; Priebe, W.; Braunschweiger, P.; Hamilton, K.; Tidmarsh, G.F.; De Young, L.R.; Lampidis, T.J. 2-deoxy-D-glucose increases the efficacy of adriamycin and paclitaxel in human osteosarcoma and non-small cell lung cancers in vivo. Cancer Res. 2004, 64, 31–34. [Google Scholar] [CrossRef] [Green Version]
  208. Gupta, S.; Mathur, R.; Dwarakanath, B.S. The glycolytic inhibitor 2-deoxy-D-glucose enhances the efficacy of etoposide in ehrlich ascites tumor-bearing mice. Cancer Biol. Ther. 2005, 4, 87–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. De Lena, M.; Lorusso, V.; Latorre, A.; Fanizza, G.; Gargano, G.; Caporusso, L.; Guida, M.; Catino, A.; Crucitta, E.; Sambiasi, D.; et al. Paclitaxel, cisplatin and lonidamine in advanced ovarian cancer. A phase II study. Eur. J. Cancer 2001, 37, 364–368. [Google Scholar] [CrossRef]
  210. Angioli, R.; Janicek, M.; Sevin, B.; Estape, R.; Averette, H.; Koechli, O.; Untch, M.; Penalver, M. Use of lonidamine to potentiate the effect of cisplatin and carboplatin on platinum resistant human ovarian cancer cells. Int. J. Oncol. 1997, 11, 777–780. [Google Scholar] [CrossRef] [PubMed]
  211. Biroccio, A.; Del Bufalo, D.; Fanciulli, M.; Bruno, T.; Zupi, G.; Floridi, A. bcl-2 inhibits mitochondrial metabolism and lonidamine-induced apoptosis in adriamycin-resistant MCF7 cells. Int. J. Cancer 1999, 82, 125–130. [Google Scholar] [CrossRef]
  212. Li, Y.C.; Fung, K.P.; Kwok, T.T.; Lee, C.Y.; Suen, Y.K.; Kong, S.K. Mitochondrial targeting drug lonidamine triggered apoptosis in doxorubicin-resistant HepG2 cells. Life Sci. 2002, 71, 2729–2740. [Google Scholar] [CrossRef]
  213. Calvino, E.; Estan, M.C.; Simon, G.P.; Sancho, P.; Boyano-Adanez Mdel, C.; de Blas, E.; Breard, J.; Aller, P. Increased apoptotic efficacy of lonidamine plus arsenic trioxide combination in human leukemia cells. Reactive oxygen species generation and defensive protein kinase (MEK/ERK, Akt/mTOR) modulation. Biochem. Pharmacol. 2011, 82, 1619–1629. [Google Scholar] [CrossRef] [PubMed]
  214. Chong, D.; Ma, L.; Liu, F.; Zhang, Z.; Zhao, S.; Huo, Q.; Zhang, P.; Zheng, H.; Liu, H. Synergistic antitumor effect of 3-bromopyruvate and 5-fluorouracil against human colorectal cancer through cell cycle arrest and induction of apoptosis. Anticancer Drugs 2017, 28, 831–840. [Google Scholar] [CrossRef]
  215. Attia, Y.M.; El-Abhar, H.S.; Al Marzabani, M.M.; Shouman, S.A. Targeting glycolysis by 3-bromopyruvate improves tamoxifen cytotoxicity of breast cancer cell lines. BMC Cancer 2015, 15, 838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Rai, Y.; Yadav, P.; Kumari, N.; Kalra, N.; Bhatt, A.N. Hexokinase II inhibition by 3-bromopyruvate sensitizes myeloid leukemic cells K-562 to anti-leukemic drug, daunorubicin. Biosci. Rep. 2019, 39, BSR20190880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Bhattacharya, S.K. Inhibition of liver hexokinase by dehydroascorbic acid and alloxan. Nature 1959, 184 (Suppl. S21), 1638–1640. [Google Scholar] [CrossRef] [PubMed]
  218. Cohen, S.; Flescher, E. Methyl jasmonate: A plant stress hormone as an anti-cancer drug. Phytochemistry 2009, 70, 1600–1609. [Google Scholar] [CrossRef] [PubMed]
  219. Huang, K.; Liang, Q.; Zhou, Y.; Jiang, L.L.; Gu, W.M.; Luo, M.Y.; Tang, Y.B.; Wang, Y.; Lu, W.; Huang, M.; et al. A Novel Allosteric Inhibitor of Phosphoglycerate Mutase 1 Suppresses Growth and Metastasis of Non-Small-Cell Lung Cancer. Cell Metab. 2019, 30, 1107–1119.e1108. [Google Scholar] [CrossRef]
  220. Qian, Y.; Bi, L.; Yang, Y.; Wang, D. Effect of pyruvate kinase M2-regulating aerobic glycolysis on chemotherapy resistance of estrogen receptor-positive breast cancer. Anticancer. Drugs 2018, 29, 616–627. [Google Scholar] [CrossRef]
  221. Wang, X.; Zhang, F.; Wu, X.R. Inhibition of Pyruvate Kinase M2 Markedly Reduces Chemoresistance of Advanced Bladder Cancer to Cisplatin. Sci. Rep. 2017, 7, 45983. [Google Scholar] [CrossRef] [Green Version]
  222. Shang, D.; Wu, J.; Guo, L.; Xu, Y.; Liu, L.; Lu, J. Metformin increases sensitivity of osteosarcoma stem cells to cisplatin by inhibiting expression of PKM2. Int. J. Oncol. 2017, 50, 1848–1856. [Google Scholar] [CrossRef]
  223. Gatti, M.; Solari, A.; Pattarozzi, A.; Campanella, C.; Thellung, S.; Maniscalco, L.; De Maria, R.; Wurth, R.; Corsaro, A.; Bajetto, A.; et al. In vitro and in vivo characterization of stem-like cells from canine osteosarcoma and assessment of drug sensitivity. Exp. Cell Res. 2018, 363, 48–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Michelakis, E.D.; Webster, L.; Mackey, J.R. Dichloroacetate (DCA) as a potential metabolic-targeting therapy for cancer. Br. J. Cancer 2008, 99, 989–994. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Woo, S.H.; Seo, S.K.; Park, Y.; Kim, E.K.; Seong, M.K.; Kim, H.A.; Song, J.Y.; Hwang, S.G.; Lee, J.K.; Noh, W.C.; et al. Dichloroacetate potentiates tamoxifen-induced cell death in breast cancer cells via downregulation of the epidermal growth factor receptor. Oncotarget 2016, 7, 59809–59819. [Google Scholar] [CrossRef] [Green Version]
  226. Lu, H.; Lu, Y.; Xie, Y.; Qiu, S.; Li, X.; Fan, Z. Rational combination with PDK1 inhibition overcomes cetuximab resistance in head and neck squamous cell carcinoma. JCI Insight 2019, 4, e131106. [Google Scholar] [CrossRef] [PubMed]
  227. Zhou, M.; Zhao, Y.; Ding, Y.; Liu, H.; Liu, Z.; Fodstad, O.; Riker, A.I.; Kamarajugadda, S.; Lu, J.; Owen, L.B.; et al. Warburg effect in chemosensitivity: Targeting lactate dehydrogenase-A re-sensitizes taxol-resistant cancer cells to taxol. Mol. Cancer 2010, 9, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  228. Farabegoli, F.; Vettraino, M.; Manerba, M.; Fiume, L.; Roberti, M.; Di Stefano, G. Galloflavin, a new lactate dehydrogenase inhibitor, induces the death of human breast cancer cells with different glycolytic attitude by affecting distinct signaling pathways. Eur. J. Pharm. Sci. 2012, 47, 729–738. [Google Scholar] [CrossRef]
  229. 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]
  230. Lee, S.; Lee, J.S.; Seo, J.; Lee, S.H.; Kang, J.H.; Song, J.; Kim, S.Y. Targeting Mitochondrial Oxidative Phosphorylation Abrogated Irinotecan Resistance in NSCLC. Sci. Rep. 2018, 8, 15707. [Google Scholar] [CrossRef]
  231. Masoud, R.; Reyes-Castellanos, G.; Lac, S.; Garcia, J.; Dou, S.; Shintu, L.; Abdel Hadi, N.; Gicquel, T.; El Kaoutari, A.; Dieme, B.; et al. Targeting Mitochondrial Complex I Overcomes Chemoresistance in High OXPHOS Pancreatic Cancer. Cell Rep. Med. 2020, 1, 100143. [Google Scholar] [CrossRef]
  232. Odier, F.; Vago, P.; Quiot, J.M.; Devauchelle, G.; Bureau, J.P. Cytometric study of the effects of destruxin E on leukemic cells in mice. Comptes Rendus Acad. Sci. III 1987, 305, 575–578. [Google Scholar]
  233. Giddings, E.L.; Champagne, D.P.; Wu, M.H.; Laffin, J.M.; Thornton, T.M.; Valenca-Pereira, F.; Culp-Hill, R.; Fortner, K.A.; Romero, N.; East, J.; et al. Mitochondrial ATP fuels ABC transporter-mediated drug efflux in cancer chemoresistance. Nat. Commun. 2021, 12, 2804. [Google Scholar] [CrossRef]
  234. Oliva, C.R.; Zhang, W.; Langford, C.; Suto, M.J.; Griguer, C.E. Repositioning chlorpromazine for treating chemoresistant glioma through the inhibition of cytochrome c oxidase bearing the COX4-1 regulatory subunit. Oncotarget 2017, 8, 37568–37583. [Google Scholar] [CrossRef]
  235. Li, Y.C.; Fung, K.P.; Kwok, T.T.; Lee, C.Y.; Suen, Y.K.; Kong, S.K. Mitochondria-targeting drug oligomycin blocked P-glycoprotein activity and triggered apoptosis in doxorubicin-resistant HepG2 cells. Chemotherapy 2004, 50, 55–62. [Google Scholar] [CrossRef]
  236. Shi, Y.; Lim, S.K.; Liang, Q.; Iyer, S.V.; Wang, H.Y.; Wang, Z.; Xie, X.; Sun, D.; Chen, Y.J.; Tabar, V.; et al. Gboxin is an oxidative phosphorylation inhibitor that targets glioblastoma. Nature 2019, 567, 341–346. [Google Scholar] [CrossRef] [PubMed]
  237. Dartier, J.; Lemaitre, E.; Chourpa, I.; Goupille, C.; Servais, S.; Chevalier, S.; Maheo, K.; Dumas, J.F. ATP-dependent activity and mitochondrial localization of drug efflux pumps in doxorubicin-resistant breast cancer cells. Biochim. Biophys. Acta Gen. Subj. 2017, 1861, 1075–1084. [Google Scholar] [CrossRef]
  238. Xie, Z.; Klionsky, D.J. Autophagosome formation: Core machinery and adaptations. Nat. Cell Biol. 2007, 9, 1102–1109. [Google Scholar] [CrossRef] [PubMed]
  239. Glick, D.; Barth, S.; Macleod, K.F. Autophagy: Cellular and molecular mechanisms. J. Pathol. 2010, 221, 3–12. [Google Scholar] [CrossRef] [Green Version]
  240. Li, X.; He, S.; Ma, B. Autophagy and autophagy-related proteins in cancer. Mol. Cancer 2020, 19, 12. [Google Scholar] [CrossRef]
  241. Mazure, N.M.; Pouyssegur, J. Hypoxia-induced autophagy: Cell death or cell survival? Curr. Opin. Cell Biol. 2010, 22, 177–180. [Google Scholar] [CrossRef]
  242. Ureshino, R.P.; Rocha, K.K.; Lopes, G.S.; Bincoletto, C.; Smaili, S.S. Calcium signaling alterations, oxidative stress, and autophagy in aging. Antioxid. Redox Signal. 2014, 21, 123–137. [Google Scholar] [CrossRef] [PubMed]
  243. Hosokawa, N.; Hara, T.; Kaizuka, T.; Kishi, C.; Takamura, A.; Miura, Y.; Iemura, S.; Natsume, T.; Takehana, K.; Yamada, N.; et al. Nutrient-dependent mTORC1 association with the ULK1-Atg13-FIP200 complex required for autophagy. Mol. Biol. Cell 2009, 20, 1981–1991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Chang, Y.Y.; Juhasz, G.; Goraksha-Hicks, P.; Arsham, A.M.; Mallin, D.R.; Muller, L.K.; Neufeld, T.P. Nutrient-dependent regulation of autophagy through the target of rapamycin pathway. Biochem. Soc. Trans. 2009, 37, 232–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Chen, Y.; Klionsky, D.J. The regulation of autophagy-unanswered questions. J. Cell Sci. 2011, 124, 161–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Pyo, J.O.; Nah, J.; Jung, Y.K. Molecules and their functions in autophagy. Exp. Mol. Med. 2012, 44, 73–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  247. Zachari, M.; Ganley, I.G. The mammalian ULK1 complex and autophagy initiation. Essays Biochem. 2017, 61, 585–596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Kraft, C.; Martens, S. Mechanisms and regulation of autophagosome formation. Curr. Opin. Cell Biol. 2012, 24, 496–501. [Google Scholar] [CrossRef]
  249. Shibutani, S.T.; Yoshimori, T. A current perspective of autophagosome biogenesis. Cell Res. 2014, 24, 58–68. [Google Scholar] [CrossRef]
  250. Mizushima, N.; Yoshimori, T.; Ohsumi, Y. The role of Atg proteins in autophagosome formation. Annu. Rev. Cell Dev. Biol. 2011, 27, 107–132. [Google Scholar] [CrossRef]
  251. Hanada, T.; Noda, N.N.; Satomi, Y.; Ichimura, Y.; Fujioka, Y.; Takao, T.; Inagaki, F.; Ohsumi, Y. The Atg12-Atg5 conjugate has a novel E3-like activity for protein lipidation in autophagy. J. Biol. Chem. 2007, 282, 37298–37302. [Google Scholar] [CrossRef] [Green Version]
  252. Noda, N.N.; Fujioka, Y.; Hanada, T.; Ohsumi, Y.; Inagaki, F. Structure of the Atg12-Atg5 conjugate reveals a platform for stimulating Atg8-PE conjugation. EMBO Rep. 2013, 14, 206–211. [Google Scholar] [CrossRef] [Green Version]
  253. Sakoh-Nakatogawa, M.; Matoba, K.; Asai, E.; Kirisako, H.; Ishii, J.; Noda, N.N.; Inagaki, F.; Nakatogawa, H.; Ohsumi, Y. Atg12-Atg5 conjugate enhances E2 activity of Atg3 by rearranging its catalytic site. Nat. Struct. Mol. Biol. 2013, 20, 433–439. [Google Scholar] [CrossRef] [PubMed]
  254. Geng, J.; Klionsky, D.J. The Atg8 and Atg12 ubiquitin-like conjugation systems in macroautophagy. ‘Protein modifications: Beyond the usual suspects’ review series. EMBO Rep. 2008, 9, 859–864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  255. Mizushima, N.; Yoshimori, T.; Ohsumi, Y. Role of the Apg12 conjugation system in mammalian autophagy. Int. J. Biochem. Cell Biol. 2003, 35, 553–561. [Google Scholar] [CrossRef]
  256. Mizushima, N.; Sugita, H.; Yoshimori, T.; Ohsumi, Y. A new protein conjugation system in human. The counterpart of the yeast Apg12p conjugation system essential for autophagy. J. Biol. Chem. 1998, 273, 33889–33892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Mizushima, N.; Noda, T.; Yoshimori, T.; Tanaka, Y.; Ishii, T.; George, M.D.; Klionsky, D.J.; Ohsumi, M.; Ohsumi, Y. A protein conjugation system essential for autophagy. Nature 1998, 395, 395–398. [Google Scholar] [CrossRef]
  258. Matsushita, M.; Suzuki, N.N.; Obara, K.; Fujioka, Y.; Ohsumi, Y.; Inagaki, F. Structure of Atg5.Atg16, a complex essential for autophagy. J. Biol. Chem. 2007, 282, 6763–6772. [Google Scholar] [CrossRef] [Green Version]
  259. Xiong, Q.; Li, W.; Li, P.; Yang, M.; Wu, C.; Eichinger, L. The Role of ATG16 in Autophagy and The Ubiquitin Proteasome System. Cells 2018, 8, 2. [Google Scholar] [CrossRef] [Green Version]
  260. Romanov, J.; Walczak, M.; Ibiricu, I.; Schuchner, S.; Ogris, E.; Kraft, C.; Martens, S. Mechanism and functions of membrane binding by the Atg5-Atg12/Atg16 complex during autophagosome formation. EMBO J 2012, 31, 4304–4317. [Google Scholar] [CrossRef]
  261. Fujioka, Y.; Noda, N.N.; Nakatogawa, H.; Ohsumi, Y.; Inagaki, F. Dimeric coiled-coil structure of Saccharomyces cerevisiae Atg16 and its functional significance in autophagy. J. Biol. Chem. 2010, 285, 1508–1515. [Google Scholar] [CrossRef] [Green Version]
  262. Kuma, A.; Mizushima, N.; Ishihara, N.; Ohsumi, Y. Formation of the approximately 350-kDa Apg12-Apg5.Apg16 multimeric complex, mediated by Apg16 oligomerization, is essential for autophagy in yeast. J. Biol. Chem. 2002, 277, 18619–18625. [Google Scholar] [CrossRef] [Green Version]
  263. Tanida, I.; Ueno, T.; Kominami, E. Human light chain 3/MAP1LC3B is cleaved at its carboxyl-terminal Met121 to expose Gly120 for lipidation and targeting to autophagosomal membranes. J. Biol. Chem. 2004, 279, 47704–47710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  264. Kirisako, T.; Ichimura, Y.; Okada, H.; Kabeya, Y.; Mizushima, N.; Yoshimori, T.; Ohsumi, M.; Takao, T.; Noda, T.; Ohsumi, Y. The reversible modification regulates the membrane-binding state of Apg8/Aut7 essential for autophagy and the cytoplasm to vacuole targeting pathway. J. Cell Biol. 2000, 151, 263–276. [Google Scholar] [CrossRef] [PubMed]
  265. Ichimura, Y.; Kirisako, T.; Takao, T.; Satomi, Y.; Shimonishi, Y.; Ishihara, N.; Mizushima, N.; Tanida, I.; Kominami, E.; Ohsumi, M.; et al. A ubiquitin-like system mediates protein lipidation. Nature 2000, 408, 488–492. [Google Scholar] [CrossRef]
  266. Hanada, T.; Satomi, Y.; Takao, T.; Ohsumi, Y. The amino-terminal region of Atg3 is essential for association with phosphatidylethanolamine in Atg8 lipidation. FEBS Lett. 2009, 583, 1078–1083. [Google Scholar] [CrossRef] [Green Version]
  267. Kabeya, Y.; Mizushima, N.; Ueno, T.; Yamamoto, A.; Kirisako, T.; Noda, T.; Kominami, E.; Ohsumi, Y.; Yoshimori, T. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J. 2000, 19, 5720–5728. [Google Scholar] [CrossRef] [PubMed]
  268. Lorincz, P.; Juhasz, G. Autophagosome-Lysosome Fusion. J. Mol. Biol. 2020, 432, 2462–2482. [Google Scholar] [CrossRef]
  269. Wang, X.; Proud, C.G. mTORC2 is a tyrosine kinase. Cell Res. 2016, 26, 1–2. [Google Scholar] [CrossRef] [PubMed]
  270. Yin, Y.; Hua, H.; Li, M.; Liu, S.; Kong, Q.; Shao, T.; Wang, J.; Luo, Y.; Wang, Q.; Luo, T.; et al. mTORC2 promotes type I insulin-like growth factor receptor and insulin receptor activation through the tyrosine kinase activity of mTOR. Cell Res. 2016, 26, 46–65. [Google Scholar] [CrossRef] [Green Version]
  271. Yoon, M.S. The Role of Mammalian Target of Rapamycin (mTOR) in Insulin Signaling. Nutrients 2017, 9, 1176. [Google Scholar] [CrossRef]
  272. Kroemer, G.; Marino, G.; Levine, B. Autophagy and the integrated stress response. Mol. Cell 2010, 40, 280–293. [Google Scholar] [CrossRef] [Green Version]
  273. Deng, L.; Chen, L.; Zhao, L.; Xu, Y.; Peng, X.; Wang, X.; Ding, L.; Jin, J.; Teng, H.; Wang, Y.; et al. Ubiquitination of Rheb governs growth factor-induced mTORC1 activation. Cell Res. 2019, 29, 136–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  274. Angarola, B.; Ferguson, S.M. Coordination of Rheb lysosomal membrane interactions with mTORC1 activation. F1000Research 2020, 9, 450. [Google Scholar] [CrossRef] [PubMed]
  275. Ganley, I.G.; Lam, D.H.; Wang, J.; Ding, X.; Chen, S.; Jiang, X. ULK1.ATG13.FIP200 complex mediates mTOR signaling and is essential for autophagy. J. Biol. Chem. 2009, 284, 12297–12305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  276. Jung, C.H.; Jun, C.B.; Ro, S.H.; Kim, Y.M.; Otto, N.M.; Cao, J.; Kundu, M.; Kim, D.H. ULK-Atg13-FIP200 complexes mediate mTOR signaling to the autophagy machinery. Mol. Biol. Cell 2009, 20, 1992–2003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  277. Tanida, I.; Ueno, T.; Kominami, E. LC3 conjugation system in mammalian autophagy. Int. J. Biochem. Cell Biol. 2004, 36, 2503–2518. [Google Scholar] [CrossRef]
  278. Klionsky, D.J.; Emr, S.D. Autophagy as a regulated pathway of cellular degradation. Science 2000, 290, 1717–1721. [Google Scholar] [CrossRef]
  279. Peng, W.; Du, T.; Zhang, Z.; Du, F.; Jin, J.; Gong, A. Knockdown of autophagy-related gene LC3 enhances the sensitivity of HepG2 cells to epirubicin. Exp. Ther. Med. 2015, 9, 1271–1276. [Google Scholar] [CrossRef]
  280. Sun, W.; Li, J.; Zhou, L.; Han, J.; Liu, R.; Zhang, H.; Ning, T.; Gao, Z.; Liu, B.; Chen, X.; et al. The c-Myc/miR-27b-3p/ATG10 regulatory axis regulates chemoresistance in colorectal cancer. Theranostics 2020, 10, 1981–1996. [Google Scholar] [CrossRef]
  281. Zaffagnini, G.; Martens, S. Mechanisms of Selective Autophagy. J. Mol. Biol. 2016, 428, 1714–1724. [Google Scholar] [CrossRef] [Green Version]
  282. Mancias, J.D.; Kimmelman, A.C. Mechanisms of Selective Autophagy in Normal Physiology and Cancer. J. Mol. Biol. 2016, 428, 1659–1680. [Google Scholar] [CrossRef] [Green Version]
  283. Amaravadi, R.K.; Lippincott-Schwartz, J.; Yin, X.M.; Weiss, W.A.; Takebe, N.; Timmer, W.; DiPaola, R.S.; Lotze, M.T.; White, E. Principles and current strategies for targeting autophagy for cancer treatment. Clin. Cancer Res. 2011, 17, 654–666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  284. Garber, K. Inducing indigestion: Companies embrace autophagy inhibitors. J. Natl. Cancer Inst. 2011, 103, 708–710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  285. White, E.; DiPaola, R.S. The double-edged sword of autophagy modulation in cancer. Clin. Cancer Res. 2009, 15, 5308–5316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  286. White, E. Deconvoluting the context-dependent role for autophagy in cancer. Nat. Rev. Cancer 2012, 12, 401–410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  287. Amaravadi, R.K.; Yu, D.; Lum, J.J.; Bui, T.; Christophorou, M.A.; Evan, G.I.; Thomas-Tikhonenko, A.; Thompson, C.B. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J. Clin. Investig. 2007, 117, 326–336. [Google Scholar] [CrossRef] [Green Version]
  288. Altman, B.J.; Jacobs, S.R.; Mason, E.F.; Michalek, R.D.; MacIntyre, A.N.; Coloff, J.L.; Ilkayeva, O.; Jia, W.; He, Y.W.; Rathmell, J.C. Autophagy is essential to suppress cell stress and to allow BCR-Abl-mediated leukemogenesis. Oncogene 2011, 30, 1855–1867. [Google Scholar] [CrossRef] [Green Version]
  289. Feldman, M.E.; Shokat, K.M. New inhibitors of the PI3K-Akt-mTOR pathway: Insights into mTOR signaling from a new generation of Tor Kinase Domain Inhibitors (TORKinibs). Curr. Top. Microbiol. Immunol. 2010, 347, 241–262. [Google Scholar] [CrossRef]
  290. Sheen, J.H.; Zoncu, R.; Kim, D.; Sabatini, D.M. Defective regulation of autophagy upon leucine deprivation reveals a targetable liability of human melanoma cells in vitro and in vivo. Cancer Cell 2011, 19, 613–628. [Google Scholar] [CrossRef] [Green Version]
  291. Diaz-Meco, M.T. Targeting leucine addiction and autophagy in melanoma. Pigment Cell Melanoma Res. 2011, 24, 588–589. [Google Scholar] [CrossRef] [Green Version]
  292. Yang, Z.J.; Chee, C.E.; Huang, S.; Sinicrope, F.A. The role of autophagy in cancer: Therapeutic implications. Mol. Cancer Ther. 2011, 10, 1533–1541. [Google Scholar] [CrossRef] [Green Version]
  293. Bursch, W.; Karwan, A.; Mayer, M.; Dornetshuber, J.; Frohwein, U.; Schulte-Hermann, R.; Fazi, B.; Di Sano, F.; Piredda, L.; Piacentini, M.; et al. Cell death and autophagy: Cytokines, drugs, and nutritional factors. Toxicology 2008, 254, 147–157. [Google Scholar] [CrossRef] [Green Version]
  294. Dasari, S.K.; Bialik, S.; Levin-Zaidman, S.; Levin-Salomon, V.; Merrill, A.H., Jr.; Futerman, A.H.; Kimchi, A. Signalome-wide RNAi screen identifies GBA1 as a positive mediator of autophagic cell death. Cell Death Differ. 2017, 24, 1288–1302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  295. Dasari, S.K.; Schejter, E.; Bialik, S.; Shkedy, A.; Levin-Salomon, V.; Levin-Zaidman, S.; Kimchi, A. Death by over-eating: The Gaucher disease associated gene GBA1, identified in a screen for mediators of autophagic cell death, is necessary for developmental cell death in Drosophila midgut. Cell Cycle 2017, 16, 2003–2010. [Google Scholar] [CrossRef] [Green Version]
  296. Cecconi, F.; Levine, B. The role of autophagy in mammalian development: Cell makeover rather than cell death. Dev. Cell 2008, 15, 344–357. [Google Scholar] [CrossRef] [Green Version]
  297. Le Bot, N. Autophagy: A new regulator of development. Nat. Cell Biol. 2007, 9, 741. [Google Scholar] [CrossRef] [PubMed]
  298. Kelekar, A. Autophagy. Ann. N. Y. Acad. Sci. 2005, 1066, 259–271. [Google Scholar] [CrossRef] [PubMed]
  299. Musiwaro, P.; Smith, M.; Manifava, M.; Walker, S.A.; Ktistakis, N.T. Characteristics and requirements of basal autophagy in HEK 293 cells. Autophagy 2013, 9, 1407–1417. [Google Scholar] [CrossRef]
  300. Roberts, P.J.; Der, C.J. Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene 2007, 26, 3291–3310. [Google Scholar] [CrossRef] [Green Version]
  301. Nagaria, T.S.; Williams, J.L.; Leduc, C.; Squire, J.A.; Greer, P.A.; Sangrar, W. Flavopiridol synergizes with sorafenib to induce cytotoxicity and potentiate antitumorigenic activity in EGFR/HER-2 and mutant RAS/RAF breast cancer model systems. Neoplasia 2013, 15, 939–951. [Google Scholar] [CrossRef] [Green Version]
  302. Nagaria, T.S.; Shi, C.; Leduc, C.; Hoskin, V.; Sikdar, S.; Sangrar, W.; Greer, P.A. Combined targeting of Raf and Mek synergistically inhibits tumorigenesis in triple negative breast cancer model systems. Oncotarget 2017, 8, 80804–80819. [Google Scholar] [CrossRef] [Green Version]
  303. Zhang, L.; Wang, H.; Li, W.; Zhong, J.; Yu, R.; Huang, X.; Wang, H.; Tan, Z.; Wang, J.; Zhang, Y. Pazopanib, a novel multi-kinase inhibitor, shows potent antitumor activity in colon cancer through PUMA-mediated apoptosis. Oncotarget 2017, 8, 3289–3303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  304. Codogno, P.; Morel, E. FOXO3a Provides a Quickstep from Autophagy Inhibition to Apoptosis in Cancer Therapy. Dev. Cell 2018, 44, 537–539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  305. Fitzwalter, B.E.; Thorburn, A. FOXO3 links autophagy to apoptosis. Autophagy 2018, 14, 1467–1468. [Google Scholar] [CrossRef] [PubMed]
  306. Fitzwalter, B.E.; Towers, C.G.; Sullivan, K.D.; Andrysik, Z.; Hoh, M.; Ludwig, M.; O’Prey, J.; Ryan, K.M.; Espinosa, J.M.; Morgan, M.J.; et al. Autophagy Inhibition Mediates Apoptosis Sensitization in Cancer Therapy by Relieving FOXO3a Turnover. Dev. Cell 2018, 44, 555–565.e553. [Google Scholar] [CrossRef] [Green Version]
  307. Sun, L.; Huang, Y.; Liu, Y.; Zhao, Y.; He, X.; Zhang, L.; Wang, F.; Zhang, Y. Ipatasertib, a novel Akt inhibitor, induces transcription factor FoxO3a and NF-kappaB directly regulates PUMA-dependent apoptosis. Cell Death Dis. 2018, 9, 911. [Google Scholar] [CrossRef] [Green Version]
  308. Kouroku, Y.; Fujita, E.; Tanida, I.; Ueno, T.; Isoai, A.; Kumagai, H.; Ogawa, S.; Kaufman, R.J.; Kominami, E.; Momoi, T. ER stress (PERK/eIF2alpha phosphorylation) mediates the polyglutamine-induced LC3 conversion, an essential step for autophagy formation. Cell Death Differ. 2007, 14, 230–239. [Google Scholar] [CrossRef] [Green Version]
  309. Din, F.V.; Valanciute, A.; Houde, V.P.; Zibrova, D.; Green, K.A.; Sakamoto, K.; Alessi, D.R.; Dunlop, M.G. Aspirin inhibits mTOR signaling, activates AMP-activated protein kinase, and induces autophagy in colorectal cancer cells. Gastroenterology 2012, 142, 1504–1515.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  310. Kwon, B.; Querfurth, H.W. Palmitate activates mTOR/p70S6K through AMPK inhibition and hypophosphorylation of raptor in skeletal muscle cells: Reversal by oleate is similar to metformin. Biochimie 2015, 118, 141–150. [Google Scholar] [CrossRef]
  311. Iksen; Pothongsrisit, S.; Pongrakhananon, V. Targeting the PI3K/AKT/mTOR Signaling Pathway in Lung Cancer: An Update Regarding Potential Drugs and Natural Products. Molecules 2021, 26, 4100. [Google Scholar] [CrossRef]
  312. Zhang, H.; Yang, S.; Wang, J.; Jiang, Y. Blockade of AMPK-Mediated cAMP-PKA-CREB/ATF1 Signaling Synergizes with Aspirin to Inhibit Hepatocellular Carcinoma. Cancers 2021, 13, 1738. [Google Scholar] [CrossRef]
  313. Fattahi, S.; Amjadi-Moheb, F.; Tabaripour, R.; Ashrafi, G.H.; Akhavan-Niaki, H. PI3K/AKT/mTOR signaling in gastric cancer: Epigenetics and beyond. Life Sci. 2020, 262, 118513. [Google Scholar] [CrossRef] [PubMed]
  314. Alzahrani, A.S. PI3K/Akt/mTOR inhibitors in cancer: At the bench and bedside. Semin. Cancer Biol. 2019, 59, 125–132. [Google Scholar] [CrossRef] [PubMed]
  315. Costa, R.L.B.; Han, H.S.; Gradishar, W.J. Targeting the PI3K/AKT/mTOR pathway in triple-negative breast cancer: A review. Breast Cancer Res. Treat. 2018, 169, 397–406. [Google Scholar] [CrossRef] [PubMed]
  316. Jia, J.; Abudu, Y.P.; Claude-Taupin, A.; Gu, Y.; Kumar, S.; Choi, S.W.; Peters, R.; Mudd, M.H.; Allers, L.; Salemi, M.; et al. Galectins control MTOR and AMPK in response to lysosomal damage to induce autophagy. Autophagy 2019, 15, 169–171. [Google Scholar] [CrossRef] [Green Version]
  317. Shaw, R.J. LKB1 and AMP-activated protein kinase control of mTOR signalling and growth. Acta Physiol. 2009, 196, 65–80. [Google Scholar] [CrossRef]
  318. Alers, S.; Loffler, A.S.; Wesselborg, S.; Stork, B. Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: Cross talk, shortcuts, and feedbacks. Mol. Cell. Biol. 2012, 32, 2–11. [Google Scholar] [CrossRef] [Green Version]
  319. Anerillas, C.; Abdelmohsen, K.; Gorospe, M. Regulation of senescence traits by MAPKs. Geroscience 2020, 42, 397–408. [Google Scholar] [CrossRef]
  320. Maruyama, J.; Naguro, I.; Takeda, K.; Ichijo, H. Stress-activated MAP kinase cascades in cellular senescence. Curr. Med. Chem. 2009, 16, 1229–1235. [Google Scholar] [CrossRef]
  321. Tee, A.R.; Fingar, D.C.; Manning, B.D.; Kwiatkowski, D.J.; Cantley, L.C.; Blenis, J. Tuberous sclerosis complex-1 and -2 gene products function together to inhibit mammalian target of rapamycin (mTOR)-mediated downstream signaling. Proc. Natl. Acad. Sci. USA 2002, 99, 13571–13576. [Google Scholar] [CrossRef] [Green Version]
  322. Tee, A.R.; Manning, B.D.; Roux, P.P.; Cantley, L.C.; Blenis, J. Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase-activating protein complex toward Rheb. Curr. Biol. 2003, 13, 1259–1268. [Google Scholar] [CrossRef] [Green Version]
  323. Hardie, D.G. AMP-activated protein kinase: A cellular energy sensor with a key role in metabolic disorders and in cancer. Biochem. Soc. Trans. 2011, 39, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  324. Carling, D. AMPK signalling in health and disease. Curr. Opin. Cell Biol. 2017, 45, 31–37. [Google Scholar] [CrossRef] [PubMed]
  325. Carling, D.; Mayer, F.V.; Sanders, M.J.; Gamblin, S.J. AMP-activated protein kinase: Nature’s energy sensor. Nat. Chem. Biol. 2011, 7, 512–518. [Google Scholar] [CrossRef] [PubMed]
  326. Hardie, D.G.; Ross, F.A.; Hawley, S.A. AMPK: A nutrient and energy sensor that maintains energy homeostasis. Nat. Rev. Mol. Cell Biol. 2012, 13, 251–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  327. Ke, R.; Xu, Q.; Li, C.; Luo, L.; Huang, D. Mechanisms of AMPK in the maintenance of ATP balance during energy metabolism. Cell Biol. Int. 2018, 42, 384–392. [Google Scholar] [CrossRef] [Green Version]
  328. Xiao, B.; Sanders, M.J.; Underwood, E.; Heath, R.; Mayer, F.V.; Carmena, D.; Jing, C.; Walker, P.A.; Eccleston, J.F.; Haire, L.F.; et al. Structure of mammalian AMPK and its regulation by ADP. Nature 2011, 472, 230–233. [Google Scholar] [CrossRef] [Green Version]
  329. Chen, L.; Xin, F.J.; Wang, J.; Hu, J.; Zhang, Y.Y.; Wan, S.; Cao, L.S.; Lu, C.; Li, P.; Yan, S.F.; et al. Conserved regulatory elements in AMPK. Nature 2013, 498, E8–E10. [Google Scholar] [CrossRef]
  330. Viollet, B.; Mounier, R.; Leclerc, J.; Yazigi, A.; Foretz, M.; Andreelli, F. Targeting AMP-activated protein kinase as a novel therapeutic approach for the treatment of metabolic disorders. Diabetes Metab. 2007, 33, 395–402. [Google Scholar] [CrossRef]
  331. Stahmann, N.; Woods, A.; Carling, D.; Heller, R. Thrombin activates AMP-activated protein kinase in endothelial cells via a pathway involving Ca2+/calmodulin-dependent protein kinase kinase beta. Mol. Cell. Biol. 2006, 26, 5933–5945. [Google Scholar] [CrossRef] [Green Version]
  332. Woods, A.; Dickerson, K.; Heath, R.; Hong, S.P.; Momcilovic, M.; Johnstone, S.R.; Carlson, M.; Carling, D. Ca2+/calmodulin-dependent protein kinase kinase-beta acts upstream of AMP-activated protein kinase in mammalian cells. Cell Metab. 2005, 2, 21–33. [Google Scholar] [CrossRef] [Green Version]
  333. Hawley, S.A.; Pan, D.A.; Mustard, K.J.; Ross, L.; Bain, J.; Edelman, A.M.; Frenguelli, B.G.; Hardie, D.G. Calmodulin-dependent protein kinase kinase-beta is an alternative upstream kinase for AMP-activated protein kinase. Cell Metab. 2005, 2, 9–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  334. Willows, R.; Sanders, M.J.; Xiao, B.; Patel, B.R.; Martin, S.R.; Read, J.; Wilson, J.R.; Hubbard, J.; Gamblin, S.J.; Carling, D. Phosphorylation of AMPK by upstream kinases is required for activity in mammalian cells. Biochem. J. 2017, 474, 3059–3073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  335. Hawley, S.A.; Boudeau, J.; Reid, J.L.; Mustard, K.J.; Udd, L.; Makela, T.P.; Alessi, D.R.; Hardie, D.G. Complexes between the LKB1 tumor suppressor, STRAD alpha/beta and MO25 alpha/beta are upstream kinases in the AMP-activated protein kinase cascade. J. Biol. 2003, 2, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  336. Ellingson, W.J.; Chesser, D.G.; Winder, W.W. Effects of 3-phosphoglycerate and other metabolites on the activation of AMP-activated protein kinase by LKB1-STRAD-MO25. Am. J. Physiol. Endocrinol. Metab. 2007, 292, E400–E407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  337. Alessi, D.R.; Sakamoto, K.; Bayascas, J.R. LKB1-dependent signaling pathways. Annu. Rev. Biochem. 2006, 75, 137–163. [Google Scholar] [CrossRef] [PubMed]
  338. Forcet, C.; Billaud, M. Dialogue between LKB1 and AMPK: A hot topic at the cellular pole. Sci. STKE 2007, 2007, pe51. [Google Scholar] [CrossRef]
  339. Inoki, K.; Zhu, T.; Guan, K.L. TSC2 mediates cellular energy response to control cell growth and survival. Cell 2003, 115, 577–590. [Google Scholar] [CrossRef] [Green Version]
  340. Corradetti, M.N.; Inoki, K.; Bardeesy, N.; DePinho, R.A.; Guan, K.L. Regulation of the TSC pathway by LKB1: Evidence of a molecular link between tuberous sclerosis complex and Peutz-Jeghers syndrome. Genes Dev. 2004, 18, 1533–1538. [Google Scholar] [CrossRef] [Green Version]
  341. Nobukini, T.; Thomas, G. The mTOR/S6K signalling pathway: The role of the TSC1/2 tumour suppressor complex and the proto-oncogene Rheb. Novartis Found. Symp. 2004, 262, discussion 154–149, 265–148. [Google Scholar]
  342. Kim, J.; Kundu, M.; Viollet, B.; Guan, K.L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 2011, 13, 132–141. [Google Scholar] [CrossRef] [Green Version]
  343. Tasdemir, E.; Chiara Maiuri, M.; Morselli, E.; Criollo, A.; D’Amelio, M.; Djavaheri-Mergny, M.; Cecconi, F.; Tavernarakis, N.; Kroemer, G. A dual role of p53 in the control of autophagy. Autophagy 2008, 4, 810–814. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  344. Denisenko, T.V.; Pivnyuk, A.D.; Zhivotovsky, B. p53-Autophagy-Metastasis Link. Cancers 2018, 10, 148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  345. Hafner, A.; Bulyk, M.L.; Jambhekar, A.; Lahav, G. The multiple mechanisms that regulate p53 activity and cell fate. Nat. Rev. Mol. Cell Biol. 2019, 20, 199–210. [Google Scholar] [CrossRef]
  346. Kenzelmann Broz, D.; Spano Mello, S.; Bieging, K.T.; Jiang, D.; Dusek, R.L.; Brady, C.A.; Sidow, A.; Attardi, L.D. Global genomic profiling reveals an extensive p53-regulated autophagy program contributing to key p53 responses. Genes Dev. 2013, 27, 1016–1031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  347. Napoli, M.; Flores, E.R. The family that eats together stays together: New p53 family transcriptional targets in autophagy. Genes Dev. 2013, 27, 971–974. [Google Scholar] [CrossRef] [Green Version]
  348. Liu, Y.; Tavana, O.; Gu, W. p53 modifications: Exquisite decorations of the powerful guardian. J. Mol. Cell Biol. 2019, 11, 564–577. [Google Scholar] [CrossRef] [Green Version]
  349. Hu, W.; Chen, S.; Thorne, R.F.; Wu, M. TP53, TP53 Target Genes (DRAM, TIGAR), and Autophagy. Adv. Exp. Med. Biol. 2019, 1206, 127–149. [Google Scholar] [CrossRef]
  350. Chollat-Namy, M.; Ben Safta-Saadoun, T.; Haferssas, D.; Meurice, G.; Chouaib, S.; Thiery, J. The pharmalogical reactivation of p53 function improves breast tumor cell lysis by granzyme B and NK cells through induction of autophagy. Cell Death Dis. 2019, 10, 695. [Google Scholar] [CrossRef]
  351. Budanov, A.V.; Karin, M. p53 target genes sestrin1 and sestrin2 connect genotoxic stress and mTOR signaling. Cell 2008, 134, 451–460. [Google Scholar] [CrossRef] [Green Version]
  352. Cui, L.; Song, Z.; Liang, B.; Jia, L.; Ma, S.; Liu, X. Radiation induces autophagic cell death via the p53/DRAM signaling pathway in breast cancer cells. Oncol. Rep. 2016, 35, 3639–3647. [Google Scholar] [CrossRef] [Green Version]
  353. Garufi, A.; Pistritto, G.; Baldari, S.; Toietta, G.; Cirone, M.; D’Orazi, G. p53-Dependent PUMA to DRAM antagonistic interplay as a key molecular switch in cell-fate decision in normal/high glucose conditions. J. Exp. Clin. Cancer Res. 2017, 36, 126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  354. Crighton, D.; Wilkinson, S.; O’Prey, J.; Syed, N.; Smith, P.; Harrison, P.R.; Gasco, M.; Garrone, O.; Crook, T.; Ryan, K.M. DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell 2006, 126, 121–134. [Google Scholar] [CrossRef] [Green Version]
  355. Pei, L.; Shang, Y.; Jin, H.; Wang, S.; Wei, N.; Yan, H.; Wu, Y.; Yao, C.; Wang, X.; Zhu, L.Q.; et al. DAPK1-p53 interaction converges necrotic and apoptotic pathways of ischemic neuronal death. J. Neurosci. 2014, 34, 6546–6556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  356. Kim, N.; Chen, D.; Zhou, X.Z.; Lee, T.H. Death-Associated Protein Kinase 1 Phosphorylation in Neuronal Cell Death and Neurodegenerative Disease. Int. J. Mol. Sci. 2019, 20, 3131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  357. Harrison, B.; Kraus, M.; Burch, L.; Stevens, C.; Craig, A.; Gordon-Weeks, P.; Hupp, T.R. DAPK-1 binding to a linear peptide motif in MAP1B stimulates autophagy and membrane blebbing. J. Biol. Chem. 2008, 283, 9999–10014. [Google Scholar] [CrossRef] [Green Version]
  358. Steinmann, S.; Scheibe, K.; Erlenbach-Wuensch, K.; Neufert, C.; Schneider-Stock, R. Death-associated protein kinase: A molecule with functional antagonistic duality and a potential role in inflammatory bowel disease (Review). Int. J. Oncol. 2015, 47, 5–15. [Google Scholar] [CrossRef] [Green Version]
  359. Singh, P.; Ravanan, P.; Talwar, P. Death Associated Protein Kinase 1 (DAPK1): A Regulator of Apoptosis and Autophagy. Front. Mol. Neurosci. 2016, 9, 46. [Google Scholar] [CrossRef] [Green Version]
  360. Zalckvar, E.; Berissi, H.; Mizrachy, L.; Idelchuk, Y.; Koren, I.; Eisenstein, M.; Sabanay, H.; Pinkas-Kramarski, R.; Kimchi, A. DAP-kinase-mediated phosphorylation on the BH3 domain of beclin 1 promotes dissociation of beclin 1 from Bcl-XL and induction of autophagy. EMBO Rep. 2009, 10, 285–292. [Google Scholar] [CrossRef] [Green Version]
  361. Mrakovcic, M.; Frohlich, L.F. p53-Mediated Molecular Control of Autophagy in Tumor Cells. Biomolecules 2018, 8, 14. [Google Scholar] [CrossRef] [Green Version]
  362. Tasdemir, E.; Maiuri, M.C.; Galluzzi, L.; Vitale, I.; Djavaheri-Mergny, M.; D’Amelio, M.; Criollo, A.; Morselli, E.; Zhu, C.; Harper, F.; et al. Regulation of autophagy by cytoplasmic p53. Nat. Cell Biol. 2008, 10, 676–687. [Google Scholar] [CrossRef] [Green Version]
  363. Sui, X.; Jin, L.; Huang, X.; Geng, S.; He, C.; Hu, X. p53 signaling and autophagy in cancer: A revolutionary strategy could be developed for cancer treatment. Autophagy 2011, 7, 565–571. [Google Scholar] [CrossRef] [PubMed]
  364. Li, B.; Wang, Z.; Xie, J.M.; Wang, G.; Qian, L.Q.; Guan, X.M.; Shen, X.P.; Qin, Z.H.; Shen, G.H.; Li, X.Q.; et al. TIGAR knockdown enhanced the anticancer effect of aescin via regulating autophagy and apoptosis in colorectal cancer cells. Acta Pharmacol. Sin. 2019, 40, 111–121. [Google Scholar] [CrossRef] [Green Version]
  365. Ye, L.; Zhao, X.; Lu, J.; Qian, G.; Zheng, J.C.; Ge, S. Knockdown of TIGAR by RNA interference induces apoptosis and autophagy in HepG2 hepatocellular carcinoma cells. Biochem. Biophys. Res. Commun. 2013, 437, 300–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  366. Spoerri, P.E.; Srivastava, N.; Vernadakis, A. Role of GABAA receptors in the GABA attenuation of ethanol neurotoxicity. J. Neurosci. Res. 1996, 44, 499–506. [Google Scholar] [CrossRef]
  367. Cargnello, M.; Roux, P.P. Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases. Microbiol. Mol. Biol. Rev. 2011, 75, 50–83. [Google Scholar] [CrossRef] [Green Version]
  368. Katz, M.; Amit, I.; Yarden, Y. Regulation of MAPKs by growth factors and receptor tyrosine kinases. Biochim. Biophys. Acta 2007, 1773, 1161–1176. [Google Scholar] [CrossRef] [Green Version]
  369. Sun, Y.; Liu, W.Z.; Liu, T.; Feng, X.; Yang, N.; Zhou, H.F. Signaling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis. J. Recept. Signal Transduct. Res. 2015, 35, 600–604. [Google Scholar] [CrossRef]
  370. Hatzivassiliou, G.; Song, K.; Yen, I.; Brandhuber, B.J.; Anderson, D.J.; Alvarado, R.; Ludlam, M.J.; Stokoe, D.; Gloor, S.L.; Vigers, G.; et al. RAF inhibitors prime wild-type RAF to activate the MAPK pathway and enhance growth. Nature 2010, 464, 431–435. [Google Scholar] [CrossRef] [Green Version]
  371. Burotto, M.; Chiou, V.L.; Lee, J.M.; Kohn, E.C. The MAPK pathway across different malignancies: A new perspective. Cancer 2014, 120, 3446–3456. [Google Scholar] [CrossRef] [Green Version]
  372. Lee, D.; Kim, Y.M.; Chin, Y.W.; Kang, K.S. Schisandrol A Exhibits Estrogenic Activity via Estrogen Receptor alpha-Dependent Signaling Pathway in Estrogen Receptor-Positive Breast Cancer Cells. Pharmaceutics 2021, 13, 1082. [Google Scholar] [CrossRef]
  373. Komatsu, N.; Fujita, Y.; Matsuda, M.; Aoki, K. mTORC1 upregulation via ERK-dependent gene expression change confers intrinsic resistance to MEK inhibitors in oncogenic KRas-mutant cancer cells. Oncogene 2015, 34, 5607–5616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  374. Ogier-Denis, E.; Pattingre, S.; El Benna, J.; Codogno, P. Erk1/2-dependent phosphorylation of Galpha-interacting protein stimulates its GTPase accelerating activity and autophagy in human colon cancer cells. J. Biol. Chem. 2000, 275, 39090–39095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  375. Pattingre, S.; Bauvy, C.; Codogno, P. Amino acids interfere with the ERK1/2-dependent control of macroautophagy by controlling the activation of Raf-1 in human colon cancer HT-29 cells. J. Biol. Chem. 2003, 278, 16667–16674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  376. Ranek, M.J.; Kokkonen-Simon, K.M.; Chen, A.; Dunkerly-Eyring, B.L.; Vera, M.P.; Oeing, C.U.; Patel, C.H.; Nakamura, T.; Zhu, G.; Bedja, D.; et al. PKG1-modified TSC2 regulates mTORC1 activity to counter adverse cardiac stress. Nature 2019, 566, 264–269. [Google Scholar] [CrossRef] [PubMed]
  377. Mahoney, E.; Lucas, D.M.; Gupta, S.V.; Wagner, A.J.; Herman, S.E.; Smith, L.L.; Yeh, Y.Y.; Andritsos, L.; Jones, J.A.; Flynn, J.M.; et al. ER stress and autophagy: New discoveries in the mechanism of action and drug resistance of the cyclin-dependent kinase inhibitor flavopiridol. Blood 2012, 120, 1262–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  378. Lin, Y.; Jiang, M.; Chen, W.; Zhao, T.; Wei, Y. Cancer and ER stress: Mutual crosstalk between autophagy, oxidative stress and inflammatory response. Biomed. Pharmacother. 2019, 118, 109249. [Google Scholar] [CrossRef]
  379. Lv, S.; Sun, E.C.; Xu, Q.Y.; Zhang, J.K.; Wu, D.L. Endoplasmic reticulum stress-mediated autophagy contributes to bluetongue virus infection via the PERK-eIF2alpha pathway. Biochem. Biophys. Res. Commun. 2015, 466, 406–412. [Google Scholar] [CrossRef]
  380. Avivar-Valderas, A.; Salas, E.; Bobrovnikova-Marjon, E.; Diehl, J.A.; Nagi, C.; Debnath, J.; Aguirre-Ghiso, J.A. PERK integrates autophagy and oxidative stress responses to promote survival during extracellular matrix detachment. Mol. Cell. Biol. 2011, 31, 3616–3629. [Google Scholar] [CrossRef] [Green Version]
  381. Liu, C.; Yan, D.Y.; Wang, C.; Ma, Z.; Deng, Y.; Liu, W.; Xu, B. IRE1 signaling pathway mediates protective autophagic response against manganese-induced neuronal apoptosis in vivo and in vitro. Sci. Total Environ. 2020, 712, 136480. [Google Scholar] [CrossRef]
  382. Zheng, Y.; Tu, J.; Wang, X.; Yu, Y.; Li, J.; Jin, Y.; Wu, J. The Therapeutic Effect of Melatonin on GC by Inducing Cell Apoptosis and Autophagy Induced by Endoplasmic Reticulum Stress. OncoTargets Ther. 2019, 12, 10187–10198. [Google Scholar] [CrossRef] [Green Version]
  383. Borsello, T.; Croquelois, K.; Hornung, J.P.; Clarke, P.G. N-methyl-d-aspartate-triggered neuronal death in organotypic hippocampal cultures is endocytic, autophagic and mediated by the c-Jun N-terminal kinase pathway. Eur. J. Neurosci. 2003, 18, 473–485. [Google Scholar] [CrossRef] [PubMed]
  384. Walton, M.; MacGibbon, G.; Young, D.; Sirimanne, E.; Williams, C.; Gluckman, P.; Dragunow, M. Do c-Jun, c-Fos, and amyloid precursor protein play a role in neuronal death or survival? J. Neurosci. Res. 1998, 53, 330–342. [Google Scholar] [CrossRef]
  385. Calnan, D.R.; Brunet, A. The FoxO code. Oncogene 2008, 27, 2276–2288. [Google Scholar] [CrossRef] [Green Version]
  386. Daitoku, H.; Fukamizu, A. FOXO transcription factors in the regulatory networks of longevity. J. Biochem. 2007, 141, 769–774. [Google Scholar] [CrossRef]
  387. Bogoyevitch, M.A.; Kobe, B. Uses for JNK: The many and varied substrates of the c-Jun N-terminal kinases. Microbiol. Mol. Biol. Rev. 2006, 70, 1061–1095. [Google Scholar] [CrossRef] [Green Version]
  388. Galluzzi, L.; Pietrocola, F.; Bravo-San Pedro, J.M.; Amaravadi, R.K.; Baehrecke, E.H.; Cecconi, F.; Codogno, P.; Debnath, J.; Gewirtz, D.A.; Karantza, V.; et al. Autophagy in malignant transformation and cancer progression. EMBO J. 2015, 34, 856–880. [Google Scholar] [CrossRef]
  389. Kimmelman, A.C. The dynamic nature of autophagy in cancer. Genes Dev. 2011, 25, 1999–2010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  390. Maiuri, M.C.; Tasdemir, E.; Criollo, A.; Morselli, E.; Vicencio, J.M.; Carnuccio, R.; Kroemer, G. Control of autophagy by oncogenes and tumor suppressor genes. Cell Death Differ. 2009, 16, 87–93. [Google Scholar] [CrossRef] [Green Version]
  391. Qu, X.; Yu, J.; Bhagat, G.; Furuya, N.; Hibshoosh, H.; Troxel, A.; Rosen, J.; Eskelinen, E.L.; Mizushima, N.; Ohsumi, Y.; et al. Promotion of tumorigenesis by heterozygous disruption of the beclin 1 autophagy gene. J. Clin. Investig. 2003, 112, 1809–1820. [Google Scholar] [CrossRef] [Green Version]
  392. Takamura, A.; Komatsu, M.; Hara, T.; Sakamoto, A.; Kishi, C.; Waguri, S.; Eishi, Y.; Hino, O.; Tanaka, K.; Mizushima, N. Autophagy-deficient mice develop multiple liver tumors. Genes Dev. 2011, 25, 795–800. [Google Scholar] [CrossRef] [Green Version]
  393. Yue, Z.; Jin, S.; Yang, C.; Levine, A.J.; Heintz, N. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc. Natl. Acad. Sci. USA 2003, 100, 15077–15082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  394. Liang, X.H.; Yu, J.; Brown, K.; Levine, B. Beclin 1 contains a leucine-rich nuclear export signal that is required for its autophagy and tumor suppressor function. Cancer Res. 2001, 61, 3443–3449. [Google Scholar]
  395. Lenardo, M.J.; McPhee, C.K.; Yu, L. Autophagic cell death. Methods Enzymol. 2009, 453, 17–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  396. Denton, D.; Nicolson, S.; Kumar, S. Cell death by autophagy: Facts and apparent artefacts. Cell Death Differ. 2012, 19, 87–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  397. Shimizu, S.; Yoshida, T.; Tsujioka, M.; Arakawa, S. Autophagic cell death and cancer. Int. J. Mol. Sci. 2014, 15, 3145–3153. [Google Scholar] [CrossRef] [Green Version]
  398. Choi, D.S.; Blanco, E.; Kim, Y.S.; Rodriguez, A.A.; Zhao, H.; Huang, T.H.; Chen, C.L.; Jin, G.; Landis, M.D.; Burey, L.A.; et al. Chloroquine eliminates cancer stem cells through deregulation of Jak2 and DNMT1. Stem Cells 2014, 32, 2309–2323. [Google Scholar] [CrossRef] [Green Version]
  399. Balic, A.; Sorensen, M.D.; Trabulo, S.M.; Sainz, B., Jr.; Cioffi, M.; Vieira, C.R.; Miranda-Lorenzo, I.; Hidalgo, M.; Kleeff, J.; Erkan, M.; et al. Chloroquine targets pancreatic cancer stem cells via inhibition of CXCR4 and hedgehog signaling. Mol. Cancer Ther. 2014, 13, 1758–1771. [Google Scholar] [CrossRef] [Green Version]
  400. Matsumoto, S.; Nakata, K.; Sagara, A.; Guan, W.; Ikenaga, N.; Ohuchida, K.; Nakamura, M. Efficient pre-treatment for pancreatic cancer using chloroquine-loaded nanoparticles targeting pancreatic stellate cells. Oncol. Lett. 2021, 22, 633. [Google Scholar] [CrossRef]
  401. Jiang, H.; Gomez-Manzano, C.; Aoki, H.; Alonso, M.M.; Kondo, S.; McCormick, F.; Xu, J.; Kondo, Y.; Bekele, B.N.; Colman, H.; et al. Examination of the therapeutic potential of Delta-24-RGD in brain tumor stem cells: Role of autophagic cell death. J. Natl. Cancer Inst. 2007, 99, 1410–1414. [Google Scholar] [CrossRef] [Green Version]
  402. Camuzard, O.; Trojani, M.C.; Santucci-Darmanin, S.; Pagnotta, S.; Breuil, V.; Carle, G.F.; Pierrefite-Carle, V. Autophagy in Osteosarcoma Cancer Stem Cells Is Critical Process which Can Be Targeted by the Antipsychotic Drug Thioridazine. Cancers 2020, 12, 3675. [Google Scholar] [CrossRef]
  403. Choudhury, S.; Kolukula, V.K.; Preet, A.; Albanese, C.; Avantaggiati, M.L. Dissecting the pathways that destabilize mutant p53: The proteasome or autophagy? Cell Cycle 2013, 12, 1022–1029. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  404. Moscat, J.; Diaz-Meco, M.T. p62: A versatile multitasker takes on cancer. Trends Biochem. Sci. 2012, 37, 230–236. [Google Scholar] [CrossRef] [Green Version]
  405. Duran, A.; Linares, J.F.; Galvez, A.S.; Wikenheiser, K.; Flores, J.M.; Diaz-Meco, M.T.; Moscat, J. The signaling adaptor p62 is an important NF-kappaB mediator in tumorigenesis. Cancer Cell 2008, 13, 343–354. [Google Scholar] [CrossRef] [Green Version]
  406. Isakson, P.; Bjoras, M.; Boe, S.O.; Simonsen, A. Autophagy contributes to therapy-induced degradation of the PML/RARA oncoprotein. Blood 2010, 116, 2324–2331. [Google Scholar] [CrossRef] [PubMed]
  407. Goussetis, D.J.; Gounaris, E.; Wu, E.J.; Vakana, E.; Sharma, B.; Bogyo, M.; Altman, J.K.; Platanias, L.C. Autophagic degradation of the BCR-ABL oncoprotein and generation of antileukemic responses by arsenic trioxide. Blood 2012, 120, 3555–3562. [Google Scholar] [CrossRef] [Green Version]
  408. Goussetis, D.J.; Gounaris, E.; Platanias, L.C. BCR-ABL1-induced leukemogenesis and autophagic targeting by arsenic trioxide. Autophagy 2013, 9, 93–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  409. Auberger, P. BCR-ABL/p62/SQSTM1: A cannibal embrace. Blood 2012, 120, 3389–3390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  410. Kung, C.P.; Budina, A.; Balaburski, G.; Bergenstock, M.K.; Murphy, M. Autophagy in tumor suppression and cancer therapy. Crit. Rev. Eukaryot. Gene Expr. 2011, 21, 71–100. [Google Scholar] [CrossRef]
  411. Avalos, Y.; Canales, J.; Bravo-Sagua, R.; Criollo, A.; Lavandero, S.; Quest, A.F. Tumor suppression and promotion by autophagy. Biomed. Res. Int. 2014, 2014, 603980. [Google Scholar] [CrossRef]
  412. Perez-Mancera, P.A.; Young, A.R.; Narita, M. Inside and out: The activities of senescence in cancer. Nat. Rev. Cancer 2014, 14, 547–558. [Google Scholar] [CrossRef] [Green Version]
  413. Kang, C.; Xu, Q.; Martin, T.D.; Li, M.Z.; Demaria, M.; Aron, L.; Lu, T.; Yankner, B.A.; Campisi, J.; Elledge, S.J. The DNA damage response induces inflammation and senescence by inhibiting autophagy of GATA4. Science 2015, 349, aaa5612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  414. Moscat, J.; Diaz-Meco, M.T. p62 at the crossroads of autophagy, apoptosis, and cancer. Cell 2009, 137, 1001–1004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  415. Moscat, J.; Diaz-Meco, M.T.; Wooten, M.W. Signal integration and diversification through the p62 scaffold protein. Trends Biochem. Sci. 2007, 32, 95–100. [Google Scholar] [CrossRef]
  416. Liu, E.Y.; Ryan, K.M. Autophagy and cancer--issues we need to digest. J. Cell Sci. 2012, 125, 2349–2358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  417. Kocaturk, N.M.; Akkoc, Y.; Kig, C.; Bayraktar, O.; Gozuacik, D.; Kutlu, O. Autophagy as a molecular target for cancer treatment. Eur. J. Pharm. Sci. 2019, 134, 116–137. [Google Scholar] [CrossRef] [PubMed]
  418. Zhou, Z.; Chen, Y.; Zhang, H.; Min, S.; Yu, B.; He, B.; Jin, A. Comparison of mesenchymal stromal cells from human bone marrow and adipose tissue for the treatment of spinal cord injury. Cytotherapy 2013, 15, 434–448. [Google Scholar] [CrossRef]
  419. Nagelkerke, A.; Bussink, J.; Sweep, F.C.; Span, P.N. The unfolded protein response as a target for cancer therapy. Biochim. Biophys. Acta 2014, 1846, 277–284. [Google Scholar] [CrossRef]
  420. Chan, D.C. Mitochondrial fusion and fission in mammals. Annu. Rev. Cell Dev. Biol. 2006, 22, 79–99. [Google Scholar] [CrossRef] [Green Version]
  421. Chen, H.; Chan, D.C. Mitochondrial Dynamics in Regulating the Unique Phenotypes of Cancer and Stem Cells. Cell Metab. 2017, 26, 39–48. [Google Scholar] [CrossRef] [Green Version]
  422. 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]
  423. Mishra, P.; Chan, D.C. Metabolic regulation of mitochondrial dynamics. J. Cell Biol. 2016, 212, 379–387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  424. Tilokani, L.; Nagashima, S.; Paupe, V.; Prudent, J. Mitochondrial dynamics: Overview of molecular mechanisms. Essays Biochem. 2018, 62, 341–360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  425. Wai, T.; Langer, T. Mitochondrial Dynamics and Metabolic Regulation. Trends Endocrinol. Metab. 2016, 27, 105–117. [Google Scholar] [CrossRef] [PubMed]
  426. Srinivasan, S.; Guha, M.; Kashina, A.; Avadhani, N.G. Mitochondrial dysfunction and mitochondrial dynamics-The cancer connection. Biochim. Biophys. Acta Bioenerg. 2017, 1858, 602–614. [Google Scholar] [CrossRef]
  427. Scorrano, L. Keeping mitochondria in shape: A matter of life and death. Eur. J. Clin. Investig. 2013, 43, 886–893. [Google Scholar] [CrossRef] [PubMed]
  428. Ishihara, N.; Nomura, M.; Jofuku, A.; Kato, H.; Suzuki, S.O.; Masuda, K.; Otera, H.; Nakanishi, Y.; Nonaka, I.; Goto, Y.; et al. Mitochondrial fission factor Drp1 is essential for embryonic development and synapse formation in mice. Nat. Cell Biol. 2009, 11, 958–966. [Google Scholar] [CrossRef]
  429. Bertholet, A.M.; Delerue, T.; Millet, A.M.; Moulis, M.F.; David, C.; Daloyau, M.; Arnaune-Pelloquin, L.; Davezac, N.; Mils, V.; Miquel, M.C.; et al. Mitochondrial fusion/fission dynamics in neurodegeneration and neuronal plasticity. Neurobiol. Dis. 2016, 90, 3–19. [Google Scholar] [CrossRef]
  430. van der Bliek, A.M.; Shen, Q.; Kawajiri, S. Mechanisms of mitochondrial fission and fusion. Cold Spring Harb. Perspect. Biol. 2013, 5, a011072. [Google Scholar] [CrossRef]
  431. Quintana-Cabrera, R.; Quirin, C.; Glytsou, C.; Corrado, M.; Urbani, A.; Pellattiero, A.; Calvo, E.; Vazquez, J.; Enriquez, J.A.; Gerle, C.; et al. The cristae modulator Optic atrophy 1 requires mitochondrial ATP synthase oligomers to safeguard mitochondrial function. Nat. Commun. 2018, 9, 3399. [Google Scholar] [CrossRef]
  432. Skulachev, V.P. Mitochondrial filaments and clusters as intracellular power-transmitting cables. Trends Biochem. Sci. 2001, 26, 23–29. [Google Scholar] [CrossRef]
  433. Rambold, A.S.; Kostelecky, B.; Elia, N.; Lippincott-Schwartz, J. Tubular network formation protects mitochondria from autophagosomal degradation during nutrient starvation. Proc. Natl. Acad. Sci. USA 2011, 108, 10190–10195. [Google Scholar] [CrossRef] [Green Version]
  434. Youle, R.J.; van der Bliek, A.M. Mitochondrial fission, fusion, and stress. Science 2012, 337, 1062–1065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  435. Lee, Y.J.; Jeong, S.Y.; Karbowski, M.; Smith, C.L.; Youle, R.J. Roles of the mammalian mitochondrial fission and fusion mediators Fis1, Drp1, and Opa1 in apoptosis. Mol. Biol. Cell 2004, 15, 5001–5011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  436. Taguchi, N.; Ishihara, N.; Jofuku, A.; Oka, T.; Mihara, K. Mitotic phosphorylation of dynamin-related GTPase Drp1 participates in mitochondrial fission. J. Biol. Chem. 2007, 282, 11521–11529. [Google Scholar] [CrossRef] [Green Version]
  437. Burman, J.L.; Pickles, S.; Wang, C.; Sekine, S.; Vargas, J.N.S.; Zhang, Z.; Youle, A.M.; Nezich, C.L.; Wu, X.; Hammer, J.A.; et al. Mitochondrial fission facilitates the selective mitophagy of protein aggregates. J. Cell Biol. 2017, 216, 3231–3247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  438. Ferreira-da-Silva, A.; Valacca, C.; Rios, E.; Populo, H.; Soares, P.; Sobrinho-Simoes, M.; Scorrano, L.; Maximo, V.; Campello, S. Mitochondrial dynamics protein Drp1 is overexpressed in oncocytic thyroid tumors and regulates cancer cell migration. PLoS ONE 2015, 10, e0122308. [Google Scholar] [CrossRef] [Green Version]
  439. Westrate, L.M.; Sayfie, A.D.; Burgenske, D.M.; MacKeigan, J.P. Persistent mitochondrial hyperfusion promotes G2/M accumulation and caspase-dependent cell death. PLoS ONE 2014, 9, e91911. [Google Scholar] [CrossRef]
  440. Qian, W.; Choi, S.; Gibson, G.A.; Watkins, S.C.; Bakkenist, C.J.; Van Houten, B. Mitochondrial hyperfusion induced by loss of the fission protein Drp1 causes ATM-dependent G2/M arrest and aneuploidy through DNA replication stress. J. Cell Sci. 2012, 125, 5745–5757. [Google Scholar] [CrossRef] [Green Version]
  441. Santel, A.; Frank, S.; Gaume, B.; Herrler, M.; Youle, R.J.; Fuller, M.T. Mitofusin-1 protein is a generally expressed mediator of mitochondrial fusion in mammalian cells. J. Cell Sci. 2003, 116, 2763–2774. [Google Scholar] [CrossRef] [Green Version]
  442. Cao, Y.L.; Meng, S.; Chen, Y.; Feng, J.X.; Gu, D.D.; Yu, B.; Li, Y.J.; Yang, J.Y.; Liao, S.; Chan, D.C.; et al. MFN1 structures reveal nucleotide-triggered dimerization critical for mitochondrial fusion. Nature 2017, 542, 372–376. [Google Scholar] [CrossRef]
  443. Hoppins, S.; Nunnari, J. The molecular mechanism of mitochondrial fusion. Biochim. Biophys. Acta 2009, 1793, 20–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  444. Ishihara, N.; Eura, Y.; Mihara, K. Mitofusin 1 and 2 play distinct roles in mitochondrial fusion reactions via GTPase activity. J. Cell Sci. 2004, 117, 6535–6546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  445. Cipolat, S.; Martins de Brito, O.; Dal Zilio, B.; Scorrano, L. OPA1 requires mitofusin 1 to promote mitochondrial fusion. Proc. Natl. Acad. Sci. USA 2004, 101, 15927–15932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  446. Santel, A.; Fuller, M.T. Control of mitochondrial morphology by a human mitofusin. J. Cell Sci. 2001, 114, 867–874. [Google Scholar] [CrossRef] [PubMed]
  447. Ishihara, N.; Fujita, Y.; Oka, T.; Mihara, K. Regulation of mitochondrial morphology through proteolytic cleavage of OPA1. EMBO J. 2006, 25, 2966–2977. [Google Scholar] [CrossRef] [PubMed]
  448. Duvezin-Caubet, S.; Jagasia, R.; Wagener, J.; Hofmann, S.; Trifunovic, A.; Hansson, A.; Chomyn, A.; Bauer, M.F.; Attardi, G.; Larsson, N.G.; et al. Proteolytic processing of OPA1 links mitochondrial dysfunction to alterations in mitochondrial morphology. J. Biol. Chem. 2006, 281, 37972–37979. [Google Scholar] [CrossRef] [Green Version]
  449. Chan, D.C. Fusion and fission: Interlinked processes critical for mitochondrial health. Annu. Rev. Genet. 2012, 46, 265–287. [Google Scholar] [CrossRef] [Green Version]
  450. Wang, S.; Xiao, W.; Shan, S.; Jiang, C.; Chen, M.; Zhang, Y.; Lu, S.; Chen, J.; Zhang, C.; Chen, Q.; et al. Multi-patterned dynamics of mitochondrial fission and fusion in a living cell. PLoS ONE 2012, 7, e19879. [Google Scholar] [CrossRef] [Green Version]
  451. Smirnova, E.; Griparic, L.; Shurland, D.L.; van der Bliek, A.M. Dynamin-related protein Drp1 is required for mitochondrial division in mammalian cells. Mol. Biol. Cell 2001, 12, 2245–2256. [Google Scholar] [CrossRef] [Green Version]
  452. Kong, B.; Tsuyoshi, H.; Orisaka, M.; Shieh, D.B.; Yoshida, Y.; Tsang, B.K. Mitochondrial dynamics regulating chemoresistance in gynecological cancers. Ann. N. Y. Acad. Sci. 2015, 1350, 1–16. [Google Scholar] [CrossRef]
  453. Hou, L.; Wang, R.; Wei, H.; Li, S.; Liu, L.; Lu, X.; Yu, H.; Liu, Z. ABT737 enhances ovarian cancer cells sensitivity to cisplatin through regulation of mitochondrial fission via Sirt3 activation. Life Sci. 2019, 232, 116561. [Google Scholar] [CrossRef] [PubMed]
  454. Kong, B.; Wang, Q.; Fung, E.; Xue, K.; Tsang, B.K. p53 is required for cisplatin-induced processing of the mitochondrial fusion protein L-Opa1 that is mediated by the mitochondrial metallopeptidase Oma1 in gynecologic cancers. J. Biol. Chem. 2014, 289, 27134–27145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  455. Kingnate, C.; Charoenkwan, K.; Kumfu, S.; Chattipakorn, N.; Chattipakorn, S.C. Possible Roles of Mitochondrial Dynamics and the Effects of Pharmacological Interventions in Chemoresistant Ovarian Cancer. EBioMedicine 2018, 34, 256–266. [Google Scholar] [CrossRef] [PubMed]
  456. Burke, P.J. Mitochondria, Bioenergetics and Apoptosis in Cancer. Trends Cancer 2017, 3, 857–870. [Google Scholar] [CrossRef] [PubMed]
  457. Frezza, C.; Cipolat, S.; Martins de Brito, O.; Micaroni, M.; Beznoussenko, G.V.; Rudka, T.; Bartoli, D.; Polishuck, R.S.; Danial, N.N.; De Strooper, B.; et al. OPA1 controls apoptotic cristae remodeling independently from mitochondrial fusion. Cell 2006, 126, 177–189. [Google Scholar] [CrossRef] [Green Version]
  458. Chen, X.; Glytsou, C.; Zhou, H.; Narang, S.; Reyna, D.E.; Lopez, A.; Sakellaropoulos, T.; Gong, Y.; Kloetgen, A.; Yap, Y.S.; et al. Targeting Mitochondrial Structure Sensitizes Acute Myeloid Leukemia to Venetoclax Treatment. Cancer Discov. 2019, 9, 890–909. [Google Scholar] [CrossRef]
  459. Ramsey, H.E.; Fischer, M.A.; Lee, T.; Gorska, A.E.; Arrate, M.P.; Fuller, L.; Boyd, K.L.; Strickland, S.A.; Sensintaffar, J.; Hogdal, L.J.; et al. A Novel MCL1 Inhibitor Combined with Venetoclax Rescues Venetoclax-Resistant Acute Myelogenous Leukemia. Cancer Discov. 2018, 8, 1566–1581. [Google Scholar] [CrossRef] [Green Version]
  460. Decker, C.W.; Garcia, J.; Gatchalian, K.; Arceneaux, D.; Choi, C.; Han, D.; Hernandez, J.B. Mitofusin-2 mediates doxorubicin sensitivity and acute resistance in Jurkat leukemia cells. Biochem. Biophys. Rep. 2020, 24, 100824. [Google Scholar] [CrossRef]
  461. Otasevic, V.; Surlan, L.; Vucetic, M.; Tulic, I.; Buzadzic, B.; Stancic, A.; Jankovic, A.; Velickovic, K.; Golic, I.; Markelic, M.; et al. Expression patterns of mitochondrial OXPHOS components, mitofusin 1 and dynamin-related protein 1 are associated with human embryo fragmentation. Reprod. Fertil. Dev. 2016, 28, 319–327. [Google Scholar] [CrossRef]
  462. Casinelli, G.; LaRosa, J.; Sharma, M.; Cherok, E.; Banerjee, S.; Branca, M.; Edmunds, L.; Wang, Y.; Sims-Lucas, S.; Churley, L.; et al. N-Myc overexpression increases cisplatin resistance in neuroblastoma via deregulation of mitochondrial dynamics. Cell Death Discov. 2016, 2, 16082. [Google Scholar] [CrossRef] [Green Version]
  463. Rodrigues, T.; Ferraz, L.S. Therapeutic potential of targeting mitochondrial dynamics in cancer. Biochem. Pharmacol. 2020, 182, 114282. [Google Scholar] [CrossRef] [PubMed]
  464. Xie, L.; Shi, F.; Li, Y.; Li, W.; Yu, X.; Zhao, L.; Zhou, M.; Hu, J.; Luo, X.; Tang, M.; et al. Drp1-dependent remodeling of mitochondrial morphology triggered by EBV-LMP1 increases cisplatin resistance. Signal Transduct. Target. Ther. 2020, 5, 56. [Google Scholar] [CrossRef] [PubMed]
  465. Yang, G.D.; Huang, T.J.; Peng, L.X.; Yang, C.F.; Liu, R.Y.; Huang, H.B.; Chu, Q.Q.; Yang, H.J.; Huang, J.L.; Zhu, Z.Y.; et al. Epstein-Barr Virus_Encoded LMP1 upregulates microRNA-21 to promote the resistance of nasopharyngeal carcinoma cells to cisplatin-induced Apoptosis by suppressing PDCD4 and Fas-L. PLoS ONE 2013, 8, e78355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  466. Han, Y.; Kim, B.; Cho, U.; Park, I.S.; Kim, S.I.; Dhanasekaran, D.N.; Tsang, B.K.; Song, Y.S. Mitochondrial fission causes cisplatin resistance under hypoxic conditions via ROS in ovarian cancer cells. Oncogene 2019, 38, 7089–7105. [Google Scholar] [CrossRef] [PubMed]
  467. Cai, J.; Wang, J.; Huang, Y.; Wu, H.; Xia, T.; Xiao, J.; Chen, X.; Li, H.; Qiu, Y.; Wang, Y.; et al. ERK/Drp1-dependent mitochondrial fission is involved in the MSC-induced drug resistance of T-cell acute lymphoblastic leukemia cells. Cell Death Dis. 2016, 7, e2459. [Google Scholar] [CrossRef] [PubMed]
  468. Huang, L.; Luan, T.; Chen, Y.; Bao, X.; Huang, Y.; Fu, S.; Wang, H.; Wang, J. LASS2 regulates invasion and chemoresistance via ERK/Drp1 modulated mitochondrial dynamics in bladder cancer cells. J. Cancer 2018, 9, 1017–1024. [Google Scholar] [CrossRef]
  469. Luan, T.; Fu, S.; Huang, L.; Zuo, Y.; Ding, M.; Li, N.; Chen, J.; Wang, H.; Wang, J. MicroRNA-98 promotes drug resistance and regulates mitochondrial dynamics by targeting LASS2 in bladder cancer cells. Exp. Cell Res. 2018, 373, 188–197. [Google Scholar] [CrossRef]
  470. Huang, C.Y.; Chiang, S.F.; Chen, W.T.; Ke, T.W.; Chen, T.W.; You, Y.S.; Lin, C.Y.; Chao, K.S.C.; Huang, C.Y. HMGB1 promotes ERK-mediated mitochondrial Drp1 phosphorylation for chemoresistance through RAGE in colorectal cancer. Cell Death Dis. 2018, 9, 1004. [Google Scholar] [CrossRef] [Green Version]
  471. Yu, Y.; Xu, L.; Qi, L.; Wang, C.; Xu, N.; Liu, S.; Li, S.; Tian, H.; Liu, W.; Xu, Y.; et al. ABT737 induces mitochondrial pathway apoptosis and mitophagy by regulating DRP1-dependent mitochondrial fission in human ovarian cancer cells. Biomed. Pharmacother. 2017, 96, 22–29. [Google Scholar] [CrossRef]
  472. Palikaras, K.; Lionaki, E.; Tavernarakis, N. Balancing mitochondrial biogenesis and mitophagy to maintain energy metabolism homeostasis. Cell Death Differ. 2015, 22, 1399–1401. [Google Scholar] [CrossRef] [Green Version]
  473. Zhu, J.; Wang, K.Z.; Chu, C.T. After the banquet: Mitochondrial biogenesis, mitophagy, and cell survival. Autophagy 2013, 9, 1663–1676. [Google Scholar] [CrossRef] [Green Version]
  474. Dominy, J.E.; Puigserver, P. Mitochondrial biogenesis through activation of nuclear signaling proteins. Cold Spring Harb. Perspect. Biol. 2013, 5, a015008. [Google Scholar] [CrossRef] [PubMed]
  475. Viscomi, C.; Bottani, E.; Civiletto, G.; Cerutti, R.; Moggio, M.; Fagiolari, G.; Schon, E.A.; Lamperti, C.; Zeviani, M. In vivo correction of COX deficiency by activation of the AMPK/PGC-1alpha axis. Cell Metab. 2011, 14, 80–90. [Google Scholar] [CrossRef] [Green Version]
  476. Haq, R.; Shoag, J.; Andreu-Perez, P.; Yokoyama, S.; Edelman, H.; Rowe, G.C.; Frederick, D.T.; Hurley, A.D.; Nellore, A.; Kung, A.L.; et al. Oncogenic BRAF regulates oxidative metabolism via PGC1alpha and MITF. Cancer Cell 2013, 23, 302–315. [Google Scholar] [CrossRef] [Green Version]
  477. McGuirk, S.; Gravel, S.P.; Deblois, G.; Papadopoli, D.J.; Faubert, B.; Wegner, A.; Hiller, K.; Avizonis, D.; Akavia, U.D.; Jones, R.G.; et al. PGC-1alpha supports glutamine metabolism in breast cancer. Cancer Metab. 2013, 1, 22. [Google Scholar] [CrossRef] [Green Version]
  478. Vazquez, F.; Lim, J.H.; Chim, H.; Bhalla, K.; Girnun, G.; Pierce, K.; Clish, C.B.; Granter, S.R.; Widlund, H.R.; Spiegelman, B.M.; et al. PGC1alpha expression defines a subset of human melanoma tumors with increased mitochondrial capacity and resistance to oxidative stress. Cancer Cell 2013, 23, 287–301. [Google Scholar] [CrossRef] [Green Version]
  479. Lim, J.H.; Luo, C.; Vazquez, F.; Puigserver, P. Targeting mitochondrial oxidative metabolism in melanoma causes metabolic compensation through glucose and glutamine utilization. Cancer Res. 2014, 74, 3535–3545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  480. LaGory, E.L.; Wu, C.; Taniguchi, C.M.; Ding, C.C.; Chi, J.T.; von Eyben, R.; Scott, D.A.; Richardson, A.D.; Giaccia, A.J. Suppression of PGC-1alpha Is Critical for Reprogramming Oxidative Metabolism in Renal Cell Carcinoma. Cell Rep. 2015, 12, 116–127. [Google Scholar] [CrossRef] [Green Version]
  481. Feilchenfeldt, J.; Brundler, M.A.; Soravia, C.; Totsch, M.; Meier, C.A. Peroxisome proliferator-activated receptors (PPARs) and associated transcription factors in colon cancer: Reduced expression of PPARgamma-coactivator 1 (PGC-1). Cancer Lett. 2004, 203, 25–33. [Google Scholar] [CrossRef] [PubMed]
  482. Zhang, Y.; Ba, Y.; Liu, C.; Sun, G.; Ding, L.; Gao, S.; Hao, J.; Yu, Z.; Zhang, J.; Zen, K.; et al. PGC-1alpha induces apoptosis in human epithelial ovarian cancer cells through a PPARgamma-dependent pathway. Cell Res. 2007, 17, 363–373. [Google Scholar] [CrossRef]
  483. Neill, T.; Torres, A.; Buraschi, S.; Owens, R.T.; Hoek, J.B.; Baffa, R.; Iozzo, R.V. Decorin induces mitophagy in breast carcinoma cells via peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha) and mitostatin. J. Biol. Chem. 2014, 289, 4952–4968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  484. Ba, Y.; Zhang, Y.; Zhang, C.Y. Peroxisome proliferator activated receptor gamma coactivator-1 induces apoptosis in human epithelial ovarian cancer cells. Zhonghua Yi Xue Za Zhi 2007, 87, 1430–1433. [Google Scholar] [PubMed]
  485. Twig, G.; Shirihai, O.S. The interplay between mitochondrial dynamics and mitophagy. Antioxid. Redox Signal. 2011, 14, 1939–1951. [Google Scholar] [CrossRef] [Green Version]
  486. Lemasters, J.J. Selective mitochondrial autophagy, or mitophagy, as a targeted defense against oxidative stress, mitochondrial dysfunction, and aging. Rejuvenation Res. 2005, 8, 3–5. [Google Scholar] [CrossRef] [PubMed]
  487. Okamoto, K.; Kondo-Okamoto, N.; Ohsumi, Y. Mitochondria-anchored receptor Atg32 mediates degradation of mitochondria via selective autophagy. Dev. Cell 2009, 17, 87–97. [Google Scholar] [CrossRef] [Green Version]
  488. Chen, Y.; Azad, M.B.; Gibson, S.B. Methods for detecting autophagy and determining autophagy-induced cell death. Can. J. Physiol. Pharmacol. 2010, 88, 285–295. [Google Scholar] [CrossRef]
  489. Levine, B.; Klionsky, D.J. Development by self-digestion: Molecular mechanisms and biological functions of autophagy. Dev. Cell 2004, 6, 463–477. [Google Scholar] [CrossRef]
  490. Mizushima, N.; Levine, B.; Cuervo, A.M.; Klionsky, D.J. Autophagy fights disease through cellular self-digestion. Nature 2008, 451, 1069–1075. [Google Scholar] [CrossRef] [Green Version]
  491. Levine, B.; Yuan, J. Autophagy in cell death: An innocent convict? J. Clin. Investig. 2005, 115, 2679–2688. [Google Scholar] [CrossRef]
  492. Maiuri, M.C.; Criollo, A.; Kroemer, G. Crosstalk between apoptosis and autophagy within the Beclin 1 interactome. EMBO J. 2010, 29, 515–516. [Google Scholar] [CrossRef] [Green Version]
  493. Tan, E.Y.; Campo, L.; Han, C.; Turley, H.; Pezzella, F.; Gatter, K.C.; Harris, A.L.; Fox, S.B. BNIP3 as a progression marker in primary human breast cancer; opposing functions in in situ versus invasive cancer. Clin. Cancer Res. 2007, 13, 467–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  494. D’Amico, A.G.; Maugeri, G.; Magro, G.; Salvatorelli, L.; Drago, F.; D’Agata, V. Expression pattern of parkin isoforms in lung adenocarcinomas. Tumour Biol. 2015, 36, 5133–5141. [Google Scholar] [CrossRef] [PubMed]
  495. Liu, L.; Zuo, Z.; Lu, S.; Wang, L.; Liu, A.; Liu, X. Silencing of PINK1 represses cell growth, migration and induces apoptosis of lung cancer cells. Biomed. Pharmacother. 2018, 106, 333–341. [Google Scholar] [CrossRef] [PubMed]
  496. Hui, L.; Wu, H.; Wang, T.W.; Yang, N.; Guo, X.; Jang, X.J. Hydrogen peroxide-induced mitophagy contributes to laryngeal cancer cells survival via the upregulation of FUNDC1. Clin. Transl. Oncol. 2019, 21, 596–606. [Google Scholar] [CrossRef] [PubMed]
  497. Yamashita, K.; Miyata, H.; Makino, T.; Masuike, Y.; Furukawa, H.; Tanaka, K.; Miyazaki, Y.; Takahashi, T.; Kurokawa, Y.; Yamasaki, M.; et al. High Expression of the Mitophagy-Related Protein Pink1 is Associated with a Poor Response to Chemotherapy and a Poor Prognosis for Patients Treated with Neoadjuvant Chemotherapy for Esophageal Squamous Cell Carcinoma. Ann. Surg. Oncol. 2017, 24, 4025–4032. [Google Scholar] [CrossRef]
  498. Li, G.B.; Fu, R.Q.; Shen, H.M.; Zhou, J.; Hu, X.Y.; Liu, Y.X.; Li, Y.N.; Zhang, H.W.; Liu, X.; Zhang, Y.H.; et al. Polyphyllin I induces mitophagic and apoptotic cell death in human breast cancer cells by increasing mitochondrial PINK1 levels. Oncotarget 2017, 8, 10359–10374. [Google Scholar] [CrossRef] [Green Version]
  499. Chang, S.H.; Lee, A.Y.; Yu, K.N.; Park, J.; Kim, K.P.; Cho, M.H. Dihydroergotamine Tartrate Induces Lung Cancer Cell Death through Apoptosis and Mitophagy. Chemotherapy 2016, 61, 304–312. [Google Scholar] [CrossRef]
  500. Regula, K.M.; Ens, K.; Kirshenbaum, L.A. Inducible expression of BNIP3 provokes mitochondrial defects and hypoxia-mediated cell death of ventricular myocytes. Circ. Res. 2002, 91, 226–231. [Google Scholar] [CrossRef] [Green Version]
  501. Kanzawa, T.; Zhang, L.; Xiao, L.; Germano, I.M.; Kondo, Y.; Kondo, S. Arsenic trioxide induces autophagic cell death in malignant glioma cells by upregulation of mitochondrial cell death protein BNIP3. Oncogene 2005, 24, 980–991. [Google Scholar] [CrossRef] [Green Version]
  502. Daido, S.; Kanzawa, T.; Yamamoto, A.; Takeuchi, H.; Kondo, Y.; Kondo, S. Pivotal role of the cell death factor BNIP3 in ceramide-induced autophagic cell death in malignant glioma cells. Cancer Res. 2004, 64, 4286–4293. [Google Scholar] [CrossRef] [Green Version]
  503. Chang, C.P.; Yang, M.C.; Liu, H.S.; Lin, Y.S.; Lei, H.Y. Concanavalin A induces autophagy in hepatoma cells and has a therapeutic effect in a murine in situ hepatoma model. Hepatology 2007, 45, 286–296. [Google Scholar] [CrossRef]
  504. 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] [PubMed]
  505. Oun, R.; Moussa, Y.E.; Wheate, N.J. The side effects of platinum-based chemotherapy drugs: A review for chemists. Dalton Trans. 2018, 47, 6645–6653. [Google Scholar] [CrossRef] [PubMed]
  506. Okatsu, K.; Oka, T.; Iguchi, M.; Imamura, K.; Kosako, H.; Tani, N.; Kimura, M.; Go, E.; Koyano, F.; Funayama, M.; et al. PINK1 autophosphorylation upon membrane potential dissipation is essential for Parkin recruitment to damaged mitochondria. Nat. Commun. 2012, 3, 1016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  507. Kondapalli, C.; Kazlauskaite, A.; Zhang, N.; Woodroof, H.I.; Campbell, D.G.; Gourlay, R.; Burchell, L.; Walden, H.; Macartney, T.J.; Deak, M.; et al. PINK1 is activated by mitochondrial membrane potential depolarization and stimulates Parkin E3 ligase activity by phosphorylating Serine 65. Open Biol. 2012, 2, 120080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  508. Kazlauskaite, A.; Kondapalli, C.; Gourlay, R.; Campbell, D.G.; Ritorto, M.S.; Hofmann, K.; Alessi, D.R.; Knebel, A.; Trost, M.; Muqit, M.M. Parkin is activated by PINK1-dependent phosphorylation of ubiquitin at Ser65. Biochem. J. 2014, 460, 127–139. [Google Scholar] [CrossRef] [Green Version]
  509. Kazlauskaite, A.; Martinez-Torres, R.J.; Wilkie, S.; Kumar, A.; Peltier, J.; Gonzalez, A.; Johnson, C.; Zhang, J.; Hope, A.G.; Peggie, M.; et al. Binding to serine 65-phosphorylated ubiquitin primes Parkin for optimal PINK1-dependent phosphorylation and activation. EMBO Rep. 2015, 16, 939–954. [Google Scholar] [CrossRef]
  510. Matsuda, N.; Sato, S.; Shiba, K.; Okatsu, K.; Saisho, K.; Gautier, C.A.; Sou, Y.S.; Saiki, S.; Kawajiri, S.; Sato, F.; et al. PINK1 stabilized by mitochondrial depolarization recruits Parkin to damaged mitochondria and activates latent Parkin for mitophagy. J. Cell Biol. 2010, 189, 211–221. [Google Scholar] [CrossRef]
  511. Yao, N.; Wang, C.; Hu, N.; Li, Y.; Liu, M.; Lei, Y.; Chen, M.; Chen, L.; Chen, C.; Lan, P.; et al. Inhibition of PINK1/Parkin-dependent mitophagy sensitizes multidrug-resistant cancer cells to B5G1, a new betulinic acid analog. Cell Death Dis. 2019, 10, 232. [Google Scholar] [CrossRef]
  512. Zhu, Y.; Massen, S.; Terenzio, M.; Lang, V.; Chen-Lindner, S.; Eils, R.; Novak, I.; Dikic, I.; Hamacher-Brady, A.; Brady, N.R. Modulation of serines 17 and 24 in the LC3-interacting region of Bnip3 determines pro-survival mitophagy versus apoptosis. J. Biol. Chem. 2013, 288, 1099–1113. [Google Scholar] [CrossRef] [Green Version]
  513. Novak, I.; Kirkin, V.; McEwan, D.G.; Zhang, J.; Wild, P.; Rozenknop, A.; Rogov, V.; Lohr, F.; Popovic, D.; Occhipinti, A.; et al. Nix is a selective autophagy receptor for mitochondrial clearance. EMBO Rep. 2010, 11, 45–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  514. Bhujabal, Z.; Birgisdottir, A.B.; Sjottem, E.; Brenne, H.B.; Overvatn, A.; Habisov, S.; Kirkin, V.; Lamark, T.; Johansen, T. FKBP8 recruits LC3A to mediate Parkin-independent mitophagy. EMBO Rep. 2017, 18, 947–961. [Google Scholar] [CrossRef] [PubMed]
  515. Liu, L.; Feng, D.; Chen, G.; Chen, M.; Zheng, Q.; Song, P.; Ma, Q.; Zhu, C.; Wang, R.; Qi, W.; et al. Mitochondrial outer-membrane protein FUNDC1 mediates hypoxia-induced mitophagy in mammalian cells. Nat. Cell Biol. 2012, 14, 177–185. [Google Scholar] [CrossRef] [PubMed]
  516. Novak, I.; Dikic, I. Autophagy receptors in developmental clearance of mitochondria. Autophagy 2011, 7, 301–303. [Google Scholar] [CrossRef]
  517. Yan, C.; Luo, L.; Guo, C.Y.; Goto, S.; Urata, Y.; Shao, J.H.; Li, T.S. Doxorubicin-induced mitophagy contributes to drug resistance in cancer stem cells from HCT8 human colorectal cancer cells. Cancer Lett. 2017, 388, 34–42. [Google Scholar] [CrossRef]
  518. Hou, H.; Er, P.; Cheng, J.; Chen, X.; Ding, X.; Wang, Y.; Chen, X.; Yuan, Z.; Pang, Q.; Wang, P.; et al. High expression of FUNDC1 predicts poor prognostic outcomes and is a promising target to improve chemoradiotherapy effects in patients with cervical cancer. Cancer Med. 2017, 6, 1871–1881. [Google Scholar] [CrossRef]
  519. Sui, X.; Chen, R.; Wang, Z.; Huang, Z.; Kong, N.; Zhang, M.; Han, W.; Lou, F.; Yang, J.; Zhang, Q.; et al. Autophagy and chemotherapy resistance: A promising therapeutic target for cancer treatment. Cell Death Dis. 2013, 4, e838. [Google Scholar] [CrossRef]
  520. Yun, C.W.; Lee, S.H. The Roles of Autophagy in Cancer. Int. J. Mol. Sci. 2018, 19, 3466. [Google Scholar] [CrossRef] [Green Version]
  521. Radoshevich, L.; Murrow, L.; Chen, N.; Fernandez, E.; Roy, S.; Fung, C.; Debnath, J. ATG12 conjugation to ATG3 regulates mitochondrial homeostasis and cell death. Cell 2010, 142, 590–600. [Google Scholar] [CrossRef] [Green Version]
  522. Hua, L.; Zhu, G.; Wei, J. MicroRNA-1 overexpression increases chemosensitivity of non-small cell lung cancer cells by inhibiting autophagy related 3-mediated autophagy. Cell Biol. Int. 2018, 42, 1240–1249. [Google Scholar] [CrossRef]
  523. Zhang, W.; Dong, Y.Z.; Du, X.; Peng, X.N.; Shen, Q.M. MiRNA-153-3p promotes gefitinib-sensitivity in non-small cell lung cancer by inhibiting ATG5 expression and autophagy. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 2444–2452. [Google Scholar] [CrossRef] [PubMed]
  524. Hwang, S.H.; Yeom, H.; Lee, M. ATG5 knockout promotes paclitaxel sensitivity in drug-resistant cells via induction of necrotic cell death. Korean J. Physiol. Pharmacol. 2020, 24, 233–240. [Google Scholar] [CrossRef] [PubMed]
  525. Eom, S.Y.; Hwang, S.H.; Yeom, H.; Lee, M. An ATG5 knockout promotes paclitaxel resistance in v-Ha-ras-transformed NIH 3T3 cells. Biochem. Biophys. Res. Commun. 2019, 513, 234–241. [Google Scholar] [CrossRef] [PubMed]
  526. Gu, Y.; Chen, T.; Li, G.; Xu, C.; Xu, Z.; Zhang, J.; He, K.; Zheng, L.; Guan, Z.; Su, X.; et al. Lower Beclin 1 downregulates HER2 expression to enhance tamoxifen sensitivity and predicts a favorable outcome for ER positive breast cancer. Oncotarget 2017, 8, 52156–52177. [Google Scholar] [CrossRef] [Green Version]
  527. Xu, R.; Liu, S.; Chen, H.; Lao, L. MicroRNA-30a downregulation contributes to chemoresistance of osteosarcoma cells through activating Beclin-1-mediated autophagy. Oncol. Rep. 2016, 35, 1757–1763. [Google Scholar] [CrossRef] [Green Version]
  528. Zou, Z.; Wu, L.; Ding, H.; Wang, Y.; Zhang, Y.; Chen, X.; Chen, X.; Zhang, C.Y.; Zhang, Q.; Zen, K. MicroRNA-30a sensitizes tumor cells to cis-platinum via suppressing beclin 1-mediated autophagy. J. Biol. Chem. 2012, 287, 4148–4156. [Google Scholar] [CrossRef] [Green Version]
  529. Gong, C.; Hu, C.; Gu, F.; Xia, Q.; Yao, C.; Zhang, L.; Qiang, L.; Gao, S.; Gao, Y. Co-delivery of autophagy inhibitor ATG7 siRNA and docetaxel for breast cancer treatment. J. Control. Release 2017, 266, 272–286. [Google Scholar] [CrossRef]
  530. An, Y.; Zhang, Z.; Shang, Y.; Jiang, X.; Dong, J.; Yu, P.; Nie, Y.; Zhao, Q. miR-23b-3p regulates the chemoresistance of gastric cancer cells by targeting ATG12 and HMGB2. Cell Death Dis. 2015, 6, e1766. [Google Scholar] [CrossRef] [Green Version]
  531. Saito, T.; Ichimura, Y.; Taguchi, K.; Suzuki, T.; Mizushima, T.; Takagi, K.; Hirose, Y.; Nagahashi, M.; Iso, T.; Fukutomi, T.; et al. p62/Sqstm1 promotes malignancy of HCV-positive hepatocellular carcinoma through Nrf2-dependent metabolic reprogramming. Nat. Commun. 2016, 7, 12030. [Google Scholar] [CrossRef]
  532. Nguyen, E.V.; Huhtinen, K.; Goo, Y.A.; Kaipio, K.; Andersson, N.; Rantanen, V.; Hynninen, J.; Lahesmaa, R.; Carpen, O.; Goodlett, D.R. Hyper-phosphorylation of Sequestosome-1 Distinguishes Resistance to Cisplatin in Patient Derived High Grade Serous Ovarian Cancer Cells. Mol. Cell. Proteom. 2017, 16, 1377–1392. [Google Scholar] [CrossRef] [Green Version]
  533. Daido, S.; Yamamoto, A.; Fujiwara, K.; Sawaya, R.; Kondo, S.; Kondo, Y. Inhibition of the DNA-dependent protein kinase catalytic subunit radiosensitizes malignant glioma cells by inducing autophagy. Cancer Res. 2005, 65, 4368–4375. [Google Scholar] [CrossRef] [Green Version]
  534. Xiao, X.; Wang, W.; Li, Y.; Yang, D.; Li, X.; Shen, C.; Liu, Y.; Ke, X.; Guo, S.; Guo, Z. HSP90AA1-mediated autophagy promotes drug resistance in osteosarcoma. J. Exp. Clin. Cancer Res. 2018, 37, 201. [Google Scholar] [CrossRef] [PubMed]
  535. Yang, C.; Gao, R.; Wang, J.; Yuan, W.; Wang, C.; Zhou, X. High-mobility group nucleosome-binding domain 5 increases drug resistance in osteosarcoma through upregulating autophagy. Tumour Biol. 2014, 35, 6357–6363. [Google Scholar] [CrossRef] [PubMed]
  536. Luk, F.; Yu, Y.; Walsh, W.R.; Yang, J.L. IGF1R-targeted therapy and its enhancement of doxorubicin chemosensitivity in human osteosarcoma cell lines. Cancer Investig. 2011, 29, 521–532. [Google Scholar] [CrossRef] [PubMed]
  537. Bae, D.H.; Jansson, P.J.; Huang, M.L.; Kovacevic, Z.; Kalinowski, D.; Lee, C.S.; Sahni, S.; Richardson, D.R. The role of NDRG1 in the pathology and potential treatment of human cancers. J. Clin. Pathol. 2013, 66, 911–917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  538. Bryant, H.E.; Helleday, T. Inhibition of poly (ADP-ribose) polymerase activates ATM which is required for subsequent homologous recombination repair. Nucleic Acids Res. 2006, 34, 1685–1691. [Google Scholar] [CrossRef] [PubMed]
  539. Golla, R.M.; Li, M.; Shen, Y.; Ji, M.; Yan, Y.; Fu, K.; Greiner, T.C.; McKeithan, T.W.; Chan, W.C. Inhibition of poly(ADP-ribose) polymerase (PARP) and ataxia telangiectasia mutated (ATM) on the chemosensitivity of mantle cell lymphoma to agents that induce DNA strand breaks. Hematol. Oncol. 2012, 30, 175–179. [Google Scholar] [CrossRef] [PubMed]
  540. Tang, D.; Kang, R.; Livesey, K.M.; Cheh, C.W.; Farkas, A.; Loughran, P.; Hoppe, G.; Bianchi, M.E.; Tracey, K.J.; Zeh, H.J., III; et al. Endogenous HMGB1 regulates autophagy. J. Cell Biol. 2010, 190, 881–892. [Google Scholar] [CrossRef] [PubMed]
  541. Tang, D.; Kang, R.; Livesey, K.M.; Zeh, H.J., III; Lotze, M.T. High mobility group box 1 (HMGB1) activates an autophagic response to oxidative stress. Antioxid. Redox Signal. 2011, 15, 2185–2195. [Google Scholar] [CrossRef]
  542. Cicco, S.; Cicco, G.; Racanelli, V.; Vacca, A. Neutrophil Extracellular Traps (NETs) and Damage-Associated Molecular Patterns (DAMPs): Two Potential Targets for COVID-19 Treatment. Mediat. Inflamm. 2020, 2020, 7527953. [Google Scholar] [CrossRef]
  543. Zhou, J.; Chen, X.; Gilvary, D.L.; Tejera, M.M.; Eksioglu, E.A.; Wei, S.; Djeu, J.Y. HMGB1 induction of clusterin creates a chemoresistant niche in human prostate tumor cells. Sci. Rep. 2015, 5, 15085. [Google Scholar] [CrossRef] [PubMed]
  544. Azizian-Farsani, F.; Abedpoor, N.; Hasan Sheikhha, M.; Gure, A.O.; Nasr-Esfahani, M.H.; Ghaedi, K. Receptor for Advanced Glycation End Products Acts as a Fuel to Colorectal Cancer Development. Front. Oncol. 2020, 10, 552283. [Google Scholar] [CrossRef] [PubMed]
  545. Shimizu, T.; Sugihara, E.; Yamaguchi-Iwai, S.; Tamaki, S.; Koyama, Y.; Kamel, W.; Ueki, A.; Ishikawa, T.; Chiyoda, T.; Osuka, S.; et al. IGF2 preserves osteosarcoma cell survival by creating an autophagic state of dormancy that protects cells against chemotherapeutic stress. Cancer Res. 2014, 74, 6531–6541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  546. Sahni, S.; Gillson, J.; Park, K.C.; Chiang, S.; Leck, L.Y.W.; Jansson, P.J.; Richardson, D.R. NDRG1 suppresses basal and hypoxia-induced autophagy at both the initiation and degradation stages and sensitizes pancreatic cancer cells to lysosomal membrane permeabilization. Biochim. Biophys. Acta Gen. Subj. 2020, 1864, 129625. [Google Scholar] [CrossRef]
  547. Rossi, A.; Pizzo, P.; Filadi, R. Calcium, mitochondria and cell metabolism: A functional triangle in bioenergetics. Biochim. Biophys. Acta Mol. Cell Res. 2019, 1866, 1068–1078. [Google Scholar] [CrossRef]
  548. Zhivotovsky, B.; Orrenius, S. Calcium and cell death mechanisms: A perspective from the cell death community. Cell Calcium 2011, 50, 211–221. [Google Scholar] [CrossRef] [PubMed]
  549. Orrenius, S.; Zhivotovsky, B.; Nicotera, P. Regulation of cell death: The calcium-apoptosis link. Nat. Rev. Mol. Cell Biol. 2003, 4, 552–565. [Google Scholar] [CrossRef]
  550. Bong, A.H.L.; Monteith, G.R. Calcium signaling and the therapeutic targeting of cancer cells. Biochim. Biophys. Acta Mol. Cell Res. 2018, 1865, 1786–1794. [Google Scholar] [CrossRef]
  551. Dubois, C.; Kondratskyi, A.; Bidaux, G.; Noyer, L.; Vancauwenberghe, E.; Farfariello, V.; Toillon, R.A.; Roudbaraki, M.; Tierny, D.; Bonnal, J.L.; et al. Co-targeting Mitochondrial Ca2+ Homeostasis and Autophagy Enhances Cancer Cells’ Chemosensitivity. iScience 2020, 23, 101263. [Google Scholar] [CrossRef]
  552. Modesti, L.; Danese, A.; Angela Maria Vitto, V.; Ramaccini, D.; Aguiari, G.; Gafa, R.; Lanza, G.; Giorgi, C.; Pinton, P. Mitochondrial Ca2+ Signaling in Health, Disease and Therapy. Cells 2021, 10, 1317. [Google Scholar] [CrossRef]
  553. Giorgi, C.; Danese, A.; Missiroli, S.; Patergnani, S.; Pinton, P. Calcium Dynamics as a Machine for Decoding Signals. Trends Cell Biol. 2018, 28, 258–273. [Google Scholar] [CrossRef] [PubMed]
  554. Kondratskyi, A.; Kondratska, K.; Skryma, R.; Klionsky, D.J.; Prevarskaya, N. Ion channels in the regulation of autophagy. Autophagy 2018, 14, 3–21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  555. Abdelmohsen, K.; Srikantan, S.; Tominaga, K.; Kang, M.J.; Yaniv, Y.; Martindale, J.L.; Yang, X.; Park, S.S.; Becker, K.G.; Subramanian, M.; et al. Growth inhibition by miR-519 via multiple p21-inducing pathways. Mol. Cell. Biol. 2012, 32, 2530–2548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  556. Tang, B.D.; Xia, X.; Lv, X.F.; Yu, B.X.; Yuan, J.N.; Mai, X.Y.; Shang, J.Y.; Zhou, J.G.; Liang, S.J.; Pang, R.P. Inhibition of Orai1-mediated Ca2+ entry enhances chemosensitivity of HepG2 hepatocarcinoma cells to 5-fluorouracil. J. Cell Mol. Med. 2017, 21, 904–915. [Google Scholar] [CrossRef]
  557. Hoyer-Hansen, M.; Bastholm, L.; Szyniarowski, P.; Campanella, M.; Szabadkai, G.; Farkas, T.; Bianchi, K.; Fehrenbacher, N.; Elling, F.; Rizzuto, R.; et al. Control of macroautophagy by calcium, calmodulin-dependent kinase kinase-beta, and Bcl-2. Mol. Cell 2007, 25, 193–205. [Google Scholar] [CrossRef]
  558. Mathiasen, I.S.; Sergeev, I.N.; Bastholm, L.; Elling, F.; Norman, A.W.; Jaattela, M. Calcium and calpain as key mediators of apoptosis-like death induced by vitamin D compounds in breast cancer cells. J. Biol. Chem. 2002, 277, 30738–30745. [Google Scholar] [CrossRef] [Green Version]
  559. Decuypere, J.P.; Welkenhuyzen, K.; Luyten, T.; Ponsaerts, R.; Dewaele, M.; Molgo, J.; Agostinis, P.; Missiaen, L.; De Smedt, H.; Parys, J.B.; et al. Ins(1,4,5)P3 receptor-mediated Ca2+ signaling and autophagy induction are interrelated. Autophagy 2011, 7, 1472–1489. [Google Scholar] [CrossRef] [Green Version]
  560. Bednarczyk, P.; Kicinska, A.; Laskowski, M.; Kulawiak, B.; Kampa, R.; Walewska, A.; Krajewska, M.; Jarmuszkiewicz, W.; Szewczyk, A. Evidence for a mitochondrial ATP-regulated potassium channel in human dermal fibroblasts. Biochim. Biophys. Acta Bioenerg. 2018, 1859, 309–318. [Google Scholar] [CrossRef]
  561. Laskowski, M.; Kulawiak, B. Topology of the mitochondrial potassium ion channels. Postepy Biochem. 2016, 62, 199–205. [Google Scholar]
  562. Nakaya, H. Role of ATP-sensitive K+ channels in cardiac arrhythmias. J. Cardiovasc. Pharmacol. Ther. 2014, 19, 237–243. [Google Scholar] [CrossRef]
  563. Galecka, S.; Kulawiak, B.; Bednarczyk, P.; Singh, H.; Szewczyk, A. Single channel properties of mitochondrial large conductance potassium channel formed by BK-VEDEC splice variant. Sci. Rep. 2021, 11, 10925. [Google Scholar] [CrossRef] [PubMed]
  564. Bednarczyk, P.; Kowalczyk, J.E.; Beresewicz, M.; Dolowy, K.; Szewczyk, A.; Zablocka, B. Identification of a voltage-gated potassium channel in gerbil hippocampal mitochondria. Biochem. Biophys. Res. Commun. 2010, 397, 614–620. [Google Scholar] [CrossRef] [PubMed]
  565. Toczylowska-Maminska, R.; Olszewska, A.; Laskowski, M.; Bednarczyk, P.; Skowronek, K.; Szewczyk, A. Potassium channel in the mitochondria of human keratinocytes. J. Investig. Dermatol. 2014, 134, 764–772. [Google Scholar] [CrossRef] [Green Version]
  566. Kang, D.; Kim, S.H.; Hwang, E.M.; Kwon, O.S.; Yang, H.Y.; Kim, E.S.; Choi, T.H.; Park, J.Y.; Hong, S.G.; Han, J. Expression of thermosensitive two-pore domain K+ channels in human keratinocytes cell line HaCaT cells. Exp. Dermatol. 2007, 16, 1016–1022. [Google Scholar] [CrossRef]
  567. Rotko, D.; Kunz, W.S.; Szewczyk, A.; Kulawiak, B. Signaling pathways targeting mitochondrial potassium channels. Int. J. Biochem. Cell Biol. 2020, 125, 105792. [Google Scholar] [CrossRef] [PubMed]
  568. Krabbendam, I.E.; Honrath, B.; Culmsee, C.; Dolga, A.M. Mitochondrial Ca2+-activated K(+) channels and their role in cell life and death pathways. Cell Calcium 2018, 69, 101–111. [Google Scholar] [CrossRef]
  569. Testai, L.; Rapposelli, S.; Martelli, A.; Breschi, M.C.; Calderone, V. Mitochondrial potassium channels as pharmacological target for cardioprotective drugs. Med. Res. Rev. 2015, 35, 520–553. [Google Scholar] [CrossRef] [PubMed]
  570. Foster, D.B.; Ho, A.S.; Rucker, J.; Garlid, A.O.; Chen, L.; Sidor, A.; Garlid, K.D.; O’Rourke, B. Mitochondrial ROMK channel is a molecular component of mitoK(ATP). Circ. Res. 2012, 111, 446–454. [Google Scholar] [CrossRef] [Green Version]
  571. Laskowski, M.; Augustynek, B.; Bednarczyk, P.; Zochowska, M.; Kalisz, J.; O’Rourke, B.; Szewczyk, A.; Kulawiak, B. Single-Channel Properties of the ROMK-Pore-Forming Subunit of the Mitochondrial ATP-Sensitive Potassium Channel. Int. J. Mol. Sci. 2019, 20, 5323. [Google Scholar] [CrossRef] [Green Version]
  572. Papanicolaou, K.N.; Ashok, D.; Liu, T.; Bauer, T.M.; Sun, J.; Li, Z.; da Costa, E.; D’Orleans, C.C.; Nathan, S.; Lefer, D.J.; et al. Global knockout of ROMK potassium channel worsens cardiac ischemia-reperfusion injury but cardiomyocyte-specific knockout does not: Implications for the identity of mitoKATP. J. Mol. Cell. Cardiol. 2020, 139, 176–189. [Google Scholar] [CrossRef]
  573. Kulawiak, B.; Bednarczyk, P.; Szewczyk, A. Multidimensional Regulation of Cardiac Mitochondrial Potassium Channels. Cells 2021, 10, 1554. [Google Scholar] [CrossRef]
  574. Wrzosek, A.; Augustynek, B.; Zochowska, M.; Szewczyk, A. Mitochondrial Potassium Channels as Druggable Targets. Biomolecules 2020, 10, 1200. [Google Scholar] [CrossRef] [PubMed]
  575. Checchetto, V.; Prosdocimi, E.; Leanza, L. Mitochondrial Kv1.3: A New Target in Cancer Biology? Cell. Physiol. Biochem. 2019, 53, 52–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  576. Leanza, L.; Henry, B.; Sassi, N.; Zoratti, M.; Chandy, K.G.; Gulbins, E.; Szabo, I. Inhibitors of mitochondrial Kv1.3 channels induce Bax/Bak-independent death of cancer cells. EMBO Mol. Med. 2012, 4, 577–593. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemotherapy in development targeting mitochondrial components. The mitochondrial components are being targeted by chemotherapeutics that regulate metabolic activity, enabling for the selective killing of cancer cells for therapeutic benefit. To achieve therapeutic benefit, the calcium ion (Ca2+) channels or other ion complexes are predominantly targeted by chemotherapy, such as the voltage-dependent anion channel 1 (VDAC1), the mitochondrial calcium uniporter (MCU) complex, and the mitochondrial permeability transition pore complex (mPTPC), etc. Chemotherapy also targeted various mitochondrial enzymes or proteins, including mitochondrial ribosome and ribosomal proteins (MRP), DNA polymerase subunit gamma (PolG) and ATP synthase, etc. The mitochondrial deoxyribonucleic acid (mtDNA) and mitochondria-derived reactive oxygen species (mtROS) are considered to be potential mitochondrial components for chemotherapy. mPPT: mitochondrial permeability transition pore complex; ADP: adenosine diphosphate; ATP: adenosine triphosphate; mtDNA: mitochondrial deoxyribonucleic acid (DNA); mRNA: messenger ribonucleic acid; tRNA: transfer ribonucleic acid; VDAC1: voltage-dependent anion channel1; MCU: mitochondrial calcium uniporter; PolG: DNA polymerase gamma subunit.
Figure 1. Chemotherapy in development targeting mitochondrial components. The mitochondrial components are being targeted by chemotherapeutics that regulate metabolic activity, enabling for the selective killing of cancer cells for therapeutic benefit. To achieve therapeutic benefit, the calcium ion (Ca2+) channels or other ion complexes are predominantly targeted by chemotherapy, such as the voltage-dependent anion channel 1 (VDAC1), the mitochondrial calcium uniporter (MCU) complex, and the mitochondrial permeability transition pore complex (mPTPC), etc. Chemotherapy also targeted various mitochondrial enzymes or proteins, including mitochondrial ribosome and ribosomal proteins (MRP), DNA polymerase subunit gamma (PolG) and ATP synthase, etc. The mitochondrial deoxyribonucleic acid (mtDNA) and mitochondria-derived reactive oxygen species (mtROS) are considered to be potential mitochondrial components for chemotherapy. mPPT: mitochondrial permeability transition pore complex; ADP: adenosine diphosphate; ATP: adenosine triphosphate; mtDNA: mitochondrial deoxyribonucleic acid (DNA); mRNA: messenger ribonucleic acid; tRNA: transfer ribonucleic acid; VDAC1: voltage-dependent anion channel1; MCU: mitochondrial calcium uniporter; PolG: DNA polymerase gamma subunit.
Cancers 14 01462 g001
Figure 2. Metabolic activity of normal and cancer cells. The cells use glucose as their primary metabolic substrate, which is broken down into pyruvate by glycolytic enzymes, which ultimately produces adenosine triphosphate (ATP) either by glycolysis or oxidative phosphorylation (OXPHOS). In normal cells, most pyruvate is transported into mitochondria for further oxidation to produce ATP. In cancer cells, most pyruvate is converted to lactate and transported out through the MCT transporter. GLUT: glucose transporter; MCT: monocarboxylate transporters.
Figure 2. Metabolic activity of normal and cancer cells. The cells use glucose as their primary metabolic substrate, which is broken down into pyruvate by glycolytic enzymes, which ultimately produces adenosine triphosphate (ATP) either by glycolysis or oxidative phosphorylation (OXPHOS). In normal cells, most pyruvate is transported into mitochondria for further oxidation to produce ATP. In cancer cells, most pyruvate is converted to lactate and transported out through the MCT transporter. GLUT: glucose transporter; MCT: monocarboxylate transporters.
Cancers 14 01462 g002
Figure 3. Drugs targeting glycolysis and oxidative phosphorylation (OXPHOS) to sensitize chemotherapy resistance cancer cells. The cells use glucose as their primary metabolic substrate, which is broken down into pyruvate by glycolysis, which produces adenosine triphosphate (ATP), and nicotinamide adenine dinucleotide (NAD) plus hydrogen (H) (NADH) in the presence of various glycolysis enzymes, including hexokinase (HK), phosphofructokinase (PFK), and phosphoglycerate mutase (PGAM), etc. In normal cells, most pyruvate is transported into mitochondria for further oxidation, whereas in cancer cells most pyruvate is converted to lactate in the presence of lactate dehydrogenase (LDH). The tricarboxylic acid (TCA) cycle is fueled by acetyl-CoA, which produces NADH, flavin adenine dinucleotide (FADH2), ATP, and metabolic intermediates required for proliferation. The mitochondrial respiratory chain complex (RCC) or electron transport chains (ETC) are composed of complex I, complex II, complex III, complex IV and complex V (ATP synthase), that catalyzes the oxidation of reducing equivalents, primarily NADH and FADH2, in the IMM using the terminal electron acceptor oxygen (O2). The ETC receives NADH and FADH2 from the TCA cycle in the mitochondrial matrix, each of which provides a pair of electrons to the ETC via complexes I and II, respectively. The ETC complexes (complexes I–IV), the ATP synthase, the IMM, two diffusible electron carriers, and substrate transporters in IMM form the OXPHOS system. Ubiquinone (Q), which can be reduced to ubiquinol (QH2), is the product of electron transfer from complex I or II. Complex III oxidizes ubiquinol, allowing one electron to continue its journey via cytochrome c at a time. Cytochrome c transfers electrons to complex IV, where they are reduced to water by acting as a terminal electron acceptor. The reduction of O2 to H2O results in the pumping of protons (H+) to the IMS at complex IV. The re-entry of H+ into the matrix via complex V is related to the generation of ATP from ADP. OXPHOS: oxidative phosphorylation; GLUT1: glucose transporter 1; NADH: nicotinamide adenine dinucleotide (NAD) hydrogen; HK: hexokinase; PFK: phosphofructokinase; PGAM: phosphoglycerate mutase; LDH: lactate dehydrogenase; TCA: tricarboxylic acid; FADH2: adenine dinucleotide; RCC: respiratory chain complex; 2-DG: 2-deoxyglucose; 3-BP: 3-bromopyruvate; DHA: dehydroascorbic acid; MJ: methyl jasmonate; PKM: Pyruvate kinase M; PDH: pyruvate dehydrogenase; DCA: dichloroacetate; FCCP: carbonyl cyanide-p-trifluoromethoxyphenylhydrazone.
Figure 3. Drugs targeting glycolysis and oxidative phosphorylation (OXPHOS) to sensitize chemotherapy resistance cancer cells. The cells use glucose as their primary metabolic substrate, which is broken down into pyruvate by glycolysis, which produces adenosine triphosphate (ATP), and nicotinamide adenine dinucleotide (NAD) plus hydrogen (H) (NADH) in the presence of various glycolysis enzymes, including hexokinase (HK), phosphofructokinase (PFK), and phosphoglycerate mutase (PGAM), etc. In normal cells, most pyruvate is transported into mitochondria for further oxidation, whereas in cancer cells most pyruvate is converted to lactate in the presence of lactate dehydrogenase (LDH). The tricarboxylic acid (TCA) cycle is fueled by acetyl-CoA, which produces NADH, flavin adenine dinucleotide (FADH2), ATP, and metabolic intermediates required for proliferation. The mitochondrial respiratory chain complex (RCC) or electron transport chains (ETC) are composed of complex I, complex II, complex III, complex IV and complex V (ATP synthase), that catalyzes the oxidation of reducing equivalents, primarily NADH and FADH2, in the IMM using the terminal electron acceptor oxygen (O2). The ETC receives NADH and FADH2 from the TCA cycle in the mitochondrial matrix, each of which provides a pair of electrons to the ETC via complexes I and II, respectively. The ETC complexes (complexes I–IV), the ATP synthase, the IMM, two diffusible electron carriers, and substrate transporters in IMM form the OXPHOS system. Ubiquinone (Q), which can be reduced to ubiquinol (QH2), is the product of electron transfer from complex I or II. Complex III oxidizes ubiquinol, allowing one electron to continue its journey via cytochrome c at a time. Cytochrome c transfers electrons to complex IV, where they are reduced to water by acting as a terminal electron acceptor. The reduction of O2 to H2O results in the pumping of protons (H+) to the IMS at complex IV. The re-entry of H+ into the matrix via complex V is related to the generation of ATP from ADP. OXPHOS: oxidative phosphorylation; GLUT1: glucose transporter 1; NADH: nicotinamide adenine dinucleotide (NAD) hydrogen; HK: hexokinase; PFK: phosphofructokinase; PGAM: phosphoglycerate mutase; LDH: lactate dehydrogenase; TCA: tricarboxylic acid; FADH2: adenine dinucleotide; RCC: respiratory chain complex; 2-DG: 2-deoxyglucose; 3-BP: 3-bromopyruvate; DHA: dehydroascorbic acid; MJ: methyl jasmonate; PKM: Pyruvate kinase M; PDH: pyruvate dehydrogenase; DCA: dichloroacetate; FCCP: carbonyl cyanide-p-trifluoromethoxyphenylhydrazone.
Cancers 14 01462 g003
Figure 4. The process of autophagy. The process of autophagy is divided into three major stages: (i) initiation (formation of phagophore assembly site (PAS); (ii) nucleation, elongation and maturation (phagophore regulation and elongating into spherical autophagosomes); and (iii) fusion and degradation (autophagolysosome and autolysosome). mTOR: mammalian target of rapamycin; ULK1: unc-51-like kinase 1; ATGs: autophagy genes; FIP 200: focal adhesion kinase family interacting protein of 200 kDa; PI3K: phosphatidylinositol 3-kinase; PKB: phosphoinositide-dependent protein kinase B; VPS34: vacuolar protein sorting 34; AMBRA1: autophagy and beclin-1 regulator 1; LC3 I/II: light chain I and II; PE: phosphatidylethanolamine.
Figure 4. The process of autophagy. The process of autophagy is divided into three major stages: (i) initiation (formation of phagophore assembly site (PAS); (ii) nucleation, elongation and maturation (phagophore regulation and elongating into spherical autophagosomes); and (iii) fusion and degradation (autophagolysosome and autolysosome). mTOR: mammalian target of rapamycin; ULK1: unc-51-like kinase 1; ATGs: autophagy genes; FIP 200: focal adhesion kinase family interacting protein of 200 kDa; PI3K: phosphatidylinositol 3-kinase; PKB: phosphoinositide-dependent protein kinase B; VPS34: vacuolar protein sorting 34; AMBRA1: autophagy and beclin-1 regulator 1; LC3 I/II: light chain I and II; PE: phosphatidylethanolamine.
Cancers 14 01462 g004
Figure 5. Possible pathways involved in autophagy-mediated cell survival or cell death. Chemotherapy can be activated cytotoxic or cytoprotective autophagy by a variety of signaling pathways; including the mTOR, phosphoinositide-dependent protein kinase B/AKT (PKB/AKT) pathway, AMP-activated protein kinase (AMPK), tumor suppressor p53 pathway, mitogen-activated protein kinase (MAPK) signaling, FOXO3A-PUMA signaling, and ER stress, etc. IRS1/2: insulin receptor substrate 1 and 2; PI3K; phosphatidylinositol 3-kinase; PDK1: phosphoinositide-dependent protein kinase 1; PTEN: phosphatase and tensin; PIP2: phosphatidylinositol-4,5-bisphosphate; PIP3: phosphatidylinositol-3,4,5-trisphosphate; PKB: phosphoinositide-dependent protein kinase B; RAS: rat sarcoma; RAF: rapidly accelerated fibrosarcoma; ERK: extracellular signal regulated kinases; MEK: mitogen-activated protein kinase/ERK kinase; TSC1/2: tuberous sclerosis complex 1 and 2; GDP: guanosine diphosphate; GT: guanosine triphosphate; mTOR: mammalian target of rapamycin; LC3-I/II: light chain 3 I and II; PERK: protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK); eIF2: eukaryotic initiation factor 2; DAPK1: death-associated protein kinase 1; DRAM: damage regulated autophagy modulator; TIGAR: TP53-induced glycolysis and apoptosis regulator; LKB1: liver kinase B1; CaMKK: calcium/calmodulin-dependent protein kinase kinase; Rheb: ras homolog enriched in the brain; AMPK: AMP-activated protein kinase; ATG: autophagy gene.
Figure 5. Possible pathways involved in autophagy-mediated cell survival or cell death. Chemotherapy can be activated cytotoxic or cytoprotective autophagy by a variety of signaling pathways; including the mTOR, phosphoinositide-dependent protein kinase B/AKT (PKB/AKT) pathway, AMP-activated protein kinase (AMPK), tumor suppressor p53 pathway, mitogen-activated protein kinase (MAPK) signaling, FOXO3A-PUMA signaling, and ER stress, etc. IRS1/2: insulin receptor substrate 1 and 2; PI3K; phosphatidylinositol 3-kinase; PDK1: phosphoinositide-dependent protein kinase 1; PTEN: phosphatase and tensin; PIP2: phosphatidylinositol-4,5-bisphosphate; PIP3: phosphatidylinositol-3,4,5-trisphosphate; PKB: phosphoinositide-dependent protein kinase B; RAS: rat sarcoma; RAF: rapidly accelerated fibrosarcoma; ERK: extracellular signal regulated kinases; MEK: mitogen-activated protein kinase/ERK kinase; TSC1/2: tuberous sclerosis complex 1 and 2; GDP: guanosine diphosphate; GT: guanosine triphosphate; mTOR: mammalian target of rapamycin; LC3-I/II: light chain 3 I and II; PERK: protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK); eIF2: eukaryotic initiation factor 2; DAPK1: death-associated protein kinase 1; DRAM: damage regulated autophagy modulator; TIGAR: TP53-induced glycolysis and apoptosis regulator; LKB1: liver kinase B1; CaMKK: calcium/calmodulin-dependent protein kinase kinase; Rheb: ras homolog enriched in the brain; AMPK: AMP-activated protein kinase; ATG: autophagy gene.
Cancers 14 01462 g005
Figure 6. The role of mitochondrial biogenesis and mitophagy in chemotherapy resistance. Mitochondrial biogenesis, fusion, fission, and mitophagy are cellular mechanisms that coordinately control mitochondrial content to maintain energy metabolism in response to cellular metabolic circumstances, stress, and other intracellular or external signals. The mitochondria increase their population by biogenesis process through utilizing fusion to fuse two neighboring mitochondria and fission to split two mitochondria from one, whereas the mitophagy (selective autophagic machinery) selectively removes damaged mitochondria to maintain cellular homeostasis. The pro-survival process of mitophagy utilized recycled mitochondrial components produced during the degradation process to use amino acids and fatty acids for protein synthesis and ATP generation, followed by cell survival (chemotherapy resistance). PGC1α: proliferator-activated receptor-gamma co-activator 1-alpha; OPA1: optic atrophy 1; Mnf1/2: mitofusin 1 and 2; Drp1: dynamin-related protein 1; FIS1: fission protein homolog 1; MFF: mitochondrial fission factor; MIDs: mitochondrial dynamics proteins; PINK1: PTEN-induced kinase 1; BNIP3: Bcl-2/adenovirus E1B 19 kDa interacting protein 3; FKBP8: FK506-binding protein 8; FUNDC1: FUN14 domain-containing protein 1; TCA: tricarboxylic acid.
Figure 6. The role of mitochondrial biogenesis and mitophagy in chemotherapy resistance. Mitochondrial biogenesis, fusion, fission, and mitophagy are cellular mechanisms that coordinately control mitochondrial content to maintain energy metabolism in response to cellular metabolic circumstances, stress, and other intracellular or external signals. The mitochondria increase their population by biogenesis process through utilizing fusion to fuse two neighboring mitochondria and fission to split two mitochondria from one, whereas the mitophagy (selective autophagic machinery) selectively removes damaged mitochondria to maintain cellular homeostasis. The pro-survival process of mitophagy utilized recycled mitochondrial components produced during the degradation process to use amino acids and fatty acids for protein synthesis and ATP generation, followed by cell survival (chemotherapy resistance). PGC1α: proliferator-activated receptor-gamma co-activator 1-alpha; OPA1: optic atrophy 1; Mnf1/2: mitofusin 1 and 2; Drp1: dynamin-related protein 1; FIS1: fission protein homolog 1; MFF: mitochondrial fission factor; MIDs: mitochondrial dynamics proteins; PINK1: PTEN-induced kinase 1; BNIP3: Bcl-2/adenovirus E1B 19 kDa interacting protein 3; FKBP8: FK506-binding protein 8; FUNDC1: FUN14 domain-containing protein 1; TCA: tricarboxylic acid.
Cancers 14 01462 g006
Figure 7. Chemotherapy resistance, mitochondrial dysfunction and autophagic machinery. Mitochondria act as a powerful inducer of apoptosis via the intrinsic pathway, while mitochondrial calcium ion (Ca2+) regulates significant cell fate decisions like metabolism, growth, and death. The cytoprotective autophagy activated by chemotherapy through several autophagy regulating proteins that can effectively blocked Ca2+ channels on the outer mitochondrial membrane (OMM) and/or intramitochondrial membrane (IMM) to enter Ca2+. Inhibition of mitochondrial Ca2+ signaling can block the release of mitochondrial cytochrome c (Cyto C), followed by suppression of interaction between Cyto C and apoptosome, subsequently inhibit the apoptosis pathway. In addition, Ca2+ and Ca2+-activated potassium channel (mKca) has the ability to block the apoptosis pathway through the regulation of reactive oxygen species (ROS). Ca2+: calcium ion; HMGB1: high mobility group box 1; HMGN5: high-mobility group nucleosome-binding domain 5; HSP90AA1: heat shock protein 90AA1; IGF2: insulin growth factor 2; NDRG1: N-myc down-stream regulated gene 1; DNA-PKcs: DNA-dependent protein kinase catalytic subunit; mKca: Ca2+-activated potassium channel; Cyto C: cytochrome c; Bcl-2: B-cell lymphoma 2; Bax: BCL2-associated X; Apaf-1: apoptotic protease activating factor 1; Cas-9: caspase-9; Cas-3: caspase-3.
Figure 7. Chemotherapy resistance, mitochondrial dysfunction and autophagic machinery. Mitochondria act as a powerful inducer of apoptosis via the intrinsic pathway, while mitochondrial calcium ion (Ca2+) regulates significant cell fate decisions like metabolism, growth, and death. The cytoprotective autophagy activated by chemotherapy through several autophagy regulating proteins that can effectively blocked Ca2+ channels on the outer mitochondrial membrane (OMM) and/or intramitochondrial membrane (IMM) to enter Ca2+. Inhibition of mitochondrial Ca2+ signaling can block the release of mitochondrial cytochrome c (Cyto C), followed by suppression of interaction between Cyto C and apoptosome, subsequently inhibit the apoptosis pathway. In addition, Ca2+ and Ca2+-activated potassium channel (mKca) has the ability to block the apoptosis pathway through the regulation of reactive oxygen species (ROS). Ca2+: calcium ion; HMGB1: high mobility group box 1; HMGN5: high-mobility group nucleosome-binding domain 5; HSP90AA1: heat shock protein 90AA1; IGF2: insulin growth factor 2; NDRG1: N-myc down-stream regulated gene 1; DNA-PKcs: DNA-dependent protein kinase catalytic subunit; mKca: Ca2+-activated potassium channel; Cyto C: cytochrome c; Bcl-2: B-cell lymphoma 2; Bax: BCL2-associated X; Apaf-1: apoptotic protease activating factor 1; Cas-9: caspase-9; Cas-3: caspase-3.
Cancers 14 01462 g007
Table 1. The drugs targeting glycolysis and oxidative phosphorylation (OXPHOS) to overcome different chemotherapy resistance.
Table 1. The drugs targeting glycolysis and oxidative phosphorylation (OXPHOS) to overcome different chemotherapy resistance.
TargetsDrugs Augment the Effects of ChemotherapyRefs.
GLUT1SilybinDoxorubicin, cisplatin and paclitaxel[203,204,205]
STF-31Gemcitabine, paclitaxel and 5-fluorouracil[156]
Hexokinase (HK)2-deoxyglucose (2-DG)Cisplatin[206]
Doxorubicin[207]
Etoposide[208]
LonidamineCisplatin and paclitaxel[209]
Carboplatin[210]
Doxorubicin[150,151,211,212,213]
3-bromopyruvate (3-BP)5-fluorouracil[214]
Tamoxifen[215]
Daunorubicin[216]
Dehydroascorbic acid (DHA)Cisplatin or sorafenib[202,217]
HK2—VDAC complexesMethyl jasmonate (MJ)Cisplatin and doxorubicin[218]
Phosphoglycerate mutase 1 (PGAM1)HKB99Erlotinib[219]
Pyruvate kinase M2 (PKM2)2-DGDoxorubicin[220]
ShikoninTamoxifen[221]
MetforminCisplatin, doxorubicin and 5-fluorouracil[222,223]
Pyruvate dehydrogenase (PDH) complexDichloroacetate (DCA)Tamoxifen[224,225]
Cetuximab[226]
Lactate dehydrogenase (LDH)OxalateTaxol[227]
Galloflavin4-hydroxy-tamoxifen[167,228]
Aldehyde dehydrogenase (ALDH)GossypolIrinotecan[229,230]
Complex IPhenforminGemcitabine and irinotecan[229,230,231]
DeguelinVemurafenib[232]
RotenoneDoxorubicin[233]
Complex IVChlorpromazineTemozolomide[234]
ATP synthaseOligomycinDoxorubicin[233,235]
C-GboxinGboxin [236]
Mitochondrial uncouplerFCCPDoxorubicin[237]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Bahar, E.; Han, S.-Y.; Kim, J.-Y.; Yoon, H. Chemotherapy Resistance: Role of Mitochondrial and Autophagic Components. Cancers 2022, 14, 1462. https://doi.org/10.3390/cancers14061462

AMA Style

Bahar E, Han S-Y, Kim J-Y, Yoon H. Chemotherapy Resistance: Role of Mitochondrial and Autophagic Components. Cancers. 2022; 14(6):1462. https://doi.org/10.3390/cancers14061462

Chicago/Turabian Style

Bahar, Entaz, Sun-Young Han, Ji-Ye Kim, and Hyonok Yoon. 2022. "Chemotherapy Resistance: Role of Mitochondrial and Autophagic Components" Cancers 14, no. 6: 1462. https://doi.org/10.3390/cancers14061462

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