Next Article in Journal
Impedance Analysis to Evaluate Nutritional Status in Physiological and Pathological Conditions
Previous Article in Journal
Eight-Hour Time-Restricted Feeding: A Strong Candidate Diet Protocol for First-Line Therapy in Polycystic Ovary Syndrome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

p53 at the Crossroads between Doxorubicin-Induced Cardiotoxicity and Resistance: A Nutritional Balancing Act

1
School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
2
Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan 250014, China
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(10), 2259; https://doi.org/10.3390/nu15102259
Submission received: 13 March 2023 / Revised: 19 April 2023 / Accepted: 3 May 2023 / Published: 10 May 2023
(This article belongs to the Section Phytochemicals and Human Health)

Abstract

:
Doxorubicin (DOX) is a highly effective chemotherapeutic drug, but its long-term use can cause cardiotoxicity and drug resistance. Accumulating evidence demonstrates that p53 is directly involved in DOX toxicity and resistance. One of the primary causes for DOX resistance is the mutation or inactivation of p53. Moreover, because the non-specific activation of p53 caused by DOX can kill non-cancerous cells, p53 is a popular target for reducing toxicity. However, the reduction in DOX-induced cardiotoxicity (DIC) via p53 suppression is often at odds with the antitumor advantages of p53 reactivation. Therefore, in order to increase the effectiveness of DOX, there is an urgent need to explore p53-targeted anticancer strategies owing to the complex regulatory network and polymorphisms of the p53 gene. In this review, we summarize the role and potential mechanisms of p53 in DIC and resistance. Furthermore, we focus on the advances and challenges in applying dietary nutrients, natural products, and other pharmacological strategies to overcome DOX-induced chemoresistance and cardiotoxicity. Lastly, we present potential therapeutic strategies to address key issues in order to provide new ideas for increasing the clinical use of DOX and improving its anticancer benefits.

1. Introduction

The clinical application of doxorubicin (DOX) can often be a double-edged sword. DOX mainly exerts its antitumor effects by inserting itself into the DNA helix structure, thereby preventing DNA replication and RNA synthesis [1]. DOX can also interfere with DNA repair and synthesis by inhibiting topoisomerase 2 [2,3]. Furthermore, inside cells, DOX is converted into an unstable intermediate called semiquinone. Semiquinone generates reactive oxygen species (ROS), which cause cell membrane and DNA damage, ultimately inducing apoptosis [4].While DOX is considered one of the most effective drugs for lung, breast, ovarian, uterine, and hematological malignancies, its cumulative dose-limiting toxicity and drug resistance limit its success in anticancer therapy [5]. Owing to its non-specific nature, DOX exerts cytotoxic effects against not only tumors but also healthy tissues. Compared with other organs, cardiac tissue has a low antioxidant capacity and limited regenerative potential. Therefore, it is highly sensitive to DOX-induced oxidative damage, and this cardiotoxicity is usually progressive and irreversible [6]. In the last 50 years, several studies have extensively investigated the mechanisms of DOX-induced cardiotoxicity (DIC), focusing on oxidative stress, endoplasmic reticulum stress, inflammation, calcium dyshomeostasis, and the dysregulation of apoptosis and autophagy [5,7]. Apart from its dose-limiting toxicities, the efficacy of DOX is also limited by inherent or acquired multidrug resistance (MDR), which may lead to tumor recurrence and metastasis [8]. The mechanisms of chemoresistance can be roughly divided into primary and acquired drug resistance. Primary chemoresistance occurs due to the natural resistance of some tumors to DOX and is not a result of treatment pressure [9]. Acquired drug resistance can be of two types: cancer stem cell (CSC)-mediated and non-CSC-mediated. CSC-mediated drug resistance can occur due to the proliferation and differentiation of CSCs, leading to tumor recurrence or metastasis. Non-CSC-mediated drug resistance mechanisms include increased DOX efflux due to multidrug resistance proteins (MRPs) 1–9 [10], increased DNA damage repair [11], apoptosis escape [12], and autophagy imbalance [13].
Over the years, the underlying causes of DIC and chemoresistance have been the subjects of extensive research. In particular, the tumor suppressor gene p53 has garnered much attention due to its crucial role in coordinating the intricate regulatory network in the tumor environment. By activating p53 and its target genes (p21, BAX, and PUMA), DOX promotes DNA repair, cell cycle arrest, and tumor cell death [14]. However, p53 activation can also cause non-specific chemotoxicity in normal cells. In addition, the mutation or inactivation of p53 in most human cancers causes DOX resistance and accelerates tumor development [15]. Therefore, restoring the tumor-inhibitory function of wild-type p53 (wtp53) has become an appealing anticancer strategy. Paradoxically, p53 inhibition also appears to be beneficial in a variety of DIC models, although it is obviously not effective in preventing chemotherapeutic resistance.
Several new dietary nutrients, natural products, and drugs that can simultaneously boost the sensitivity of DOX and reduce its adverse effects have attracted attention. In this review, we provide a new perspective on enhancing the efficacy of DOX chemotherapy by summarizing the intricate role of p53 in this process. Further, we highlight the development of anticancer strategies that target p53 to limit DIC and overcome chemoresistance and the challenges associated with these strategies.

2. Mechanisms of p53 in DIC

p53 has been identified as a key transcriptomic regulator of DIC in a transcriptomic profiling study [16]. Moreover, p53-induced cell death receptor upregulation has been proposed to play a role in regulating cardiomyocyte apoptosis and autophagy and in inducing myocardial atrophy (Figure 1a).

2.1. p53 Induces Cardiomyocyte Apoptosis

Under physiological conditions, p53 is maintained at low levels via proteasomal degradation [17]. The expression of p53 increases during stress injury in the heart through transcription-dependent and -independent pathways. The transcription-dependent pathways involve the regulation of mitochondrial apoptotic signals, including the upregulation of pro-apoptotic genes such as PUMA, NOXA, and BAX and the downregulation of anti-apoptotic genes such as Bcl-2 [18]. The transcription-independent effects involve direct interactions with factors such as BAX [19] and superoxide dismutase 2 (SOD2) [20]. BAX induces cardiomyocyte apoptosis through a caspase cascade mediated by the mitochondrial release of cytochrome C (Cyt C). SOD2 interacts with p53, leading to a decrease in superoxide anion scavenging and mitochondrial membrane potential, which results in the activation of apoptosis.
Several studies have demonstrated that DOX mediates cardiomyocyte apoptosis by activating p53 transcriptional activity in the mitochondria. p53 overexpression can also induce apoptosis through its transactivation in the autologous nucleus [21]. This pro-apoptotic activity has been implicated in the relationship between the dose- and treatment duration-dependent effects of DOX and the status of the p53 gene. Higher DOX concentrations mainly induce p53-dependent cell apoptosis, while lower DOX concentrations cause cardiotoxicity via bioenergetic dysfunction [22]. p53 suppression prevents acute cardiotoxicity following DOX treatment. In contrast, inhibiting p53 activity can increase cardiomyocyte apoptosis during the late stage of DOX treatment. Notably, p53 regulates the normal transcription of genes involved in cardiac mitochondrial biogenesis and bioenergetics, and it plays a complex role in DIC [23,24]. In DOX-induced mouse models, p53 mutants (R172H) lack the typical tumor-suppressor effects of p53, such as apoptosis. However, mitochondrial biogenesis is preserved, and cardiac dysfunction is prevented [25]. Further, p53 has similar protective effects for mitochondrial DNA in cardiomyocytes derived from DOX-treated human induced pluripotent stem cells (iPSCs) [25].

2.2. p53 Regulates Cardiac Autophagy and Mitophagy

Autophagy is generally considered a mechanism for the recycling of intracellular components, and excessive autophagy is believed to be maladaptive and detrimental. At present, the mechanism of autophagy induction in the heart is not completely clear. Some studies have found that p53 exerts pro-autophagy and anti-autophagy effects under different conditions [26,27]. Under physiological conditions, the basal level of p53 (p53b) inhibits adenosine 5‘-monophosphate (AMP)-activated protein kinase (AMPK) to alleviate rapamycin complex 1 (mTORC1)-suppressed autophagy, which mTORC1 affects the formation of the ULK1/2 complex by interacting with FIP200. When stress occurs, activated p53 (p53a) translocates to the nucleus, where it promotes the transcription of autophagy genes (such as AMPK, PTEN, DRAM1, and Sestrins), and this p53-induced autophagy has no cardioprotective effect. Under high-stress conditions, when the intensity of pro-apoptotic stimuli exceeds the cell’s protective ability, the translocation of p53 to the mitochondria triggers mitochondrial outer membrane permeabilization, which is followed by the activation of the mitochondrial apoptotic pathway [26,27]. Interestingly, previous studies have shown that p53 does not have a direct effect on the expression levels of autophagy genes (ATG5, ATG12, and BECN-1). However, it indirectly contributes to autophagy and cell death in ventricular myocytes by directly altering mitochondrial membrane permeability [28] or causing mitochondrial interference via its downstream effector, the hypoxia-inducible death protein BNIP3 (Bcl-2/adenovirus E1B 19-kDa interacting protein 3) [21]. Moreover, Pan et al. demonstrated that miR-146a targeted TATA-binding protein (TBP) associated factor 9b (TAF9b) partially reverses DIC by indirectly destabilizing p53, inhibiting apoptosis, and regulating autophagy [29].
Mitophagy plays a role in the development of DIC by selectively removing structurally damaged and dysfunctional mitochondria [30]. Damaged mitochondria are cleared through the regulation of key mitochondrial fission and fusion proteins (Mfn1/2, OPA1 and DRP1) and mitophagy receptors (PINK1/Parkin, BNIP3/NIX, and FUNDC1) that directly or indirectly interact with the positive autophagosomal LC3, thereby contributing to mitochondrial encapsulation in autophagic vesicles to induce mitochondrial clearance [31,32]. The binding of cytoplasmic p53 to Parkin in DIC impairs the autophagic degradation of damaged mitochondria by inhibiting its mitochondrial transport [33]. Therefore, activating mitochondrial phagocytosis by inhibiting cytosolic p53 may be a potential therapeutic strategy for reducing DIC.

2.3. p53 Regulates Myocardial Atrophy

DOX exposure has been reported to reduce cardiac weight in mice [34]. Acute DOX-induced systolic dysfunction and reductions in heart weight and cardiomyocyte size have been found to be mediated by the p53-dependent inhibition of mammalian target of rapamycin (mTOR) signaling [35]. Similar DOX-induced adverse effects, including weakness, fatigue, dysfunction, and atrophy, have been observed in skeletal muscle tissue. The strong induction of p53/p21/REDD1 signaling is common to both effects. The downstream target genes in this axis induce cell cycle arrest and increase the levels of ROS, causing cardiac and skeletal muscle atrophy [36].

2.4. Upstream Targets Regulating p53 Protein Modification and Activity

A complex regulatory network upstream of p53 promotes its rapid activation during DOX stimulation. Multiple post-translational modifications (PTMs) have been shown to directly influence p53-induced target activation, and ultimately, p53-induced cell damage [37]. p53 phosphorylation is a key modification that exerts regulatory effects on apoptosis. In DIC, ROS and DNA damage activate the downstream checkpoint kinases Chk2 and Chk1 via ataxia telangiectasia mutated (ATM) and ataxia telangiectasia and Rad3-related (ATR) proteins, which together activate p53 phosphorylation. This promotes the split between p53 and MDM2 and allows transcription factors to bind to DNA [38]. The inhibition of Rac1, the major subunit of NADPH oxidase, enhances DIC via the ATM/p53/apoptotic pathway [39]. In addition, DOX activates mitogen-activated protein kinase (MAPK) signaling pathway components, including extracellular signal-regulated kinase 1/2 (ERK1/2), JNK, and p38, which then activate p53 phosphorylation and induce the mitochondrial apoptotic pathway [40]. The acetylation of p53, another important PTM, is essential for its activation, and it occurs through a reversible enzymatic process. Sirtuins (SIRT, class III histone deacetylases) such as SIRT1 [41,42], SIRT3 [43], and SIRT6 [44] have been shown to function as control points for several mechanisms in DIC [45]. Furthermore, Rac1 attenuates the DOX-mediated inhibition of histone deacetylase (HDAC), thereby ameliorating DIC [46]. Meanwhile, the ubiquitination of p53 is mutually exclusive with acetylation, and the ubiquitin E3 ligase ITCH degrades the ROS scavenger thioredoxin negative regulator, thereby inhibiting p38 MAPK/p53 pathway-mediated apoptosis in cardiomyocytes [47]. Apart from PTMs, Joiner et al. found that CaMKII induces apoptosis by promoting mitochondrial Ca2+ flow and p53, and the chaperone glucose-regulated protein 78 (GRP78) can restore DOX-induced cardiomyocyte apoptosis by inhibiting Ca2+-dependent CaMKII activation and p53 accumulation [48,49]. Interestingly, Huang et al. identified a novel stress-inducible protein, insulin-like growth factor receptor type IIα (IGF-IIRα), which activates the p53 apoptotic pathway [50].

3. Role of p53 in DOX Resistance

It has been established that p53 inhibits tumor formation and provides protection against DNA damage by inducing apoptosis, cell cycle arrest, and DNA repair. The overexpression of wtp53 promotes sensitivity to chemotherapeutic agents, while the expression of mutated p53 genes leads to DOX resistance. The involvement of p53 in the mechanism of chemoresistance has become a hot topic in recent reviews [51].

3.1. Mutant p53 (mutp53) Mediates DOX Resistance

p53 is the most commonly mutated gene in human cancers. The mutp53 gene has been identified in 42% of the 12 tumor types studied using cancer genome sequencing [52]. The majority of mutations occur in the core domain of p53, leading to a loss of DNA binding ability [50]. Mutp53 can also exhibit functional changes, such as loss-of-function (LOF), gain-of-function (GOF), or dominant negative effects [53]. Particularly, GOF mutations turn p53 into a proto-oncogene, which can further promote malignancy and chemotherapeutic resistance [53]. Furthermore, mutp53 is more stable than wtp53 because the binding of heat shock protein (HSP) 90 to mutp53 prevents its MDM2- and carboxy-terminus of Hsp70-interacting protein (CHIP)-mediated degradation following ubiquitination [54]. In addition, HSP70 and HSP40 have also been reported to stabilize mutp53 [55,56]. The various mechanisms of chemoresistance associated with p53 mutations have been extensively researched. These are summarized briefly in Figure 1b.

3.1.1. Interaction between p53 and P-gp

The MDR transporter P-glycoprotein (P-gp, more commonly referred to as MDR1) can eliminate almost all drugs from within cells. It is the main cause of drug resistance in cancer cells. P-gp expression is most closely related to that of p53 [57]. As a transcriptional activator, wtp53 directly inhibits P-gp gene expression by binding to its head-to-tail p53 binding sequence and downregulating P-gp [58]. Moreover, ROR1, an upstream regulator of P-gp, is overexpressed in DOX-resistant breast cancers and regulates chemoresistance by modulating P-gp expression via ERK and p53 [59]. Adenovirus-mediated p53 overexpression can significantly reduce P-gp levels and increase DOX sensitivity [60]. Notably, mutp53 positively regulates P-gp expression in a variety of cancers, including osteosarcoma and breast, colorectal, and ovarian cancers [61,62]. For example, mutp53 (R248Q) can induce combined resistance against DOX and paclitaxel by upregulating P-gp [63]. Additionally, mutp53 accumulation in DOX-resistant MCF-7 cell lines increases P-gp expression [64]. These findings suggest that P-gp inhibitors are an effective tool for reducing or preventing MDR.

3.1.2. Mutp53 Disrupts Cell Cycle Arrest and Apoptosis

In the presence of mutp53 expression or p53 deletions, tumor cells cannot be blocked in the G1 phase, abnormal cell proliferation increases, and apoptosis is inhibited, leading to drug resistance. DOX significantly activates caspase 3/7 in wtp53 cells, but not in migratory carcinoma cells expressing mutp53 [65]. In addition, unlike wtp53, mutp53 fails to activate BAX and downregulate Bcl-2 [51,66], and the overexpression of Bcl-2 can inhibit BAX and BAK activity, leading to DOX resistance [67]. Mutp53 (R273H) has been shown to induce cross-resistance to DOX and methotrexate by downregulating procaspase-3 [68]. Dominant-negative proteins or mutp53 also cause anti-apoptosis effects in tumor cells via the inhibition of p73 function [69]. Lin et al. revealed a novel mechanism in which mutp53 promotes intrinsic DOX resistance by downregulating miR-30c and subsequently upregulating the DNA damage repair proteins FANCY and REV1 [70].

3.1.3. Mutp53 Increases CSC Differentiation

CSCs are a rare subpopulation of tumor cells with the ability to self-renew and multiply differentiate, and are thought to be the cause of drug-resistant relapse after chemotherapy [71]. Although DOX kills most rapidly proliferating tumor cells, numerous preclinical and clinical studies have shown that DOX resistance is often associated with CSC enrichment and that DOX is largely ineffective at eliminating CSCs [72]. Wtp53, a potent stem cell inhibitor, prevents suboptimal cells that carry DNA damage from becoming iPSCs by inducing their apoptotic elimination [73]. Functional p53 also inhibits the epithelial–mesenchymal transition (EMT), and escape from chemotherapeutic toxicity through EMT acquisition is an essential feature of CSCs [74,75]. Recent studies have revealed a close association between p53 and EMT, in which the mutational status of p53 decides the outcome of the EMT process [76]. In contrast to wtp53, which negatively regulates EMT, mutp53 can drive part of the EMT process via the EMT transcription factors Twist and Slug [77]. Indeed, in addition to regulating the EMT program, p53 can also modulate the extracellular matrix (ECM). Accumulating evidence has demonstrated that ECM remodeling promotes the development and maintenance of stemness and chemoresistance [78,79,80,81]. ECM is composed of collagen, non-collagenous glycoproteins (mainly fibronectin and laminin), and proteoglycans. ECM components broadly regulate cellular processes and functions, including cell proliferation and survival, migration, differentiation, autophagy, angiogenesis, and immune regulation [82]. There is growing evidence that mutp53 promotes tumor neovascularization and regulates the secretion of secreted proteins and chemokines [83]. Mechanistically, p53 LOF is associated with the upregulation of ECM components (e.g., fibronectin, extracellular matrix metalloproteinase inducer, and matrix metalloproteinases), and these changes are accompanied by invasion-metastasis and the inhibition of apoptosis in tumor cells [79,84]. Notably, mutp53 expression is often associated with a high incidence of CSCs in tumors, and mutp53 GOF contributes to their development [85]. The expression of iPSCs generated by p53 mutant precursors is considered a marker gene for CSCs, and confers DOX resistance to these cells.

3.1.4. m6A Methylation of mutp53

m6A RNA methylation is the most common internal modification in mammalian mRNAs, and lncRNAs that regulate oncogenic signaling pathways contribute to carcinogenesis [86]. The Gb3-cSrc complex within sphingolipid-enriched microdomains in cancer cell membranes can upregulate m6A modifications in the p53 pre-mRNA point mutation codon 273 by increasing the expression of methyltransferase-like 3, producing DOX resistance with increasing ceramide glycosylation [87]. The inhibition of ceramide glycosylation suppresses m6A methylation in p53 pre-mRNA, activates the function of the wtp53 gene, and restores the sensitivity of drug-resistant cancer cells to DOX [88].

3.2. Degradation and Inactivation of wtp53

The inactivation or inhibition of certain p53 functions is believed to be a prerequisite for chemoresistance in most human cancers. Lin et al. reported that RNA-binding protein 28 inhibits p53 transcription by interacting with the DNA-binding structural domain of the p53 gene via Chk1/Chk2 translocation from the nucleolus to the nucleoplasm, further explaining the stronger DOX resistance seen in wtp53 cancer cells [89]. Below, we summarize and describe the mechanisms associated with wtp53 in DOX-induced tumor drug resistance.

3.2.1. Mechanism of Regulating wtp53 Degradation

p53 ubiquitination and its subsequent degradation can affect the response to DOX therapy and promote tumor formation, progression, and chemoresistance. In 10–20% of human cancers, this occurs due to the inactivation of p53 ubiquitination-mediated degradation via the amplification of MDM2 or MDM4 [90]. MDM2-mediated p53 degradation is a key factor in DOX-acquired resistance in HepG2 cells. By developing small-molecule inhibitors or degraders that block the interaction between p53 and MDM2, the oncogenic activity of p53 can be restored in patients carrying wtp53 [91]. Other deubiquitinases have also been implicated in the regulation of p53 during DOX resistance. For example, OTU dismutase 5 (OTUD5) and C-Jun activation domain-binding protein 1 (JAB1) enhance DOX and cisplatin resistance via p53 ubiquitination [92,93]. Ribosomal L1 structural domain protein 1 (RSL1D1) has been identified as a novel negative regulator of p53 in human colorectal cancer cells. Through direct interaction, it inhibits the E3 ligase activity of HDM2, enhancing the ubiquitination-mediated degradation of p53 and decreasing p53 protein levels [94]. The oncogene Gankyrin (Gank), a key molecule in hepatoblastoma, induces tumor growth by triggering the degradation of p53 [95]. Mechanistically, Gank interacts with MDM2 to amplify MDM2-mediated p53 poly-ubiquitylation, leading to p53 degradation. Additionally, the hyper-phosphorylated retinoblastoma protein (pRb) stabilizes p53 by inhibiting the interaction of the Gank-MDM2-p53 complex via binding to the central domain of MDM2. Gank and MDM2 overexpression leads to pRb inactivation, which, in turn, enhances the formation of the Gank-MDM2-p53 complex and subsequent p53 degradation [96].

3.2.2. Wtp53 and Non-Coding RNAs (ncRNAs)

NcRNAs, including microRNAs, lncRNAs, piRNAs, and circular RNAs, can regulate the therapeutic sensitivity of cancer cells [97]. Hence, they can play a role in the acquisition of drug resistance. In recent years, accumulating evidence has shown that various miRNAs, including miR-125b, miR-504, miR-25, miR-30d, miR-1285, miR-27a, and miR-380-5p, can downregulate p53 protein levels [98,99]. The overexpression of the lncRNA LINP1 is positively associated with proliferation, drug resistance, and metastasis in breast cancer cells. LINP1 knockdown can promote BC cell metastasis and improve resistance to DOX by blocking the effects of p53 [100]. Compared with microRNAs and lncRNAs, piRNAs have been studied to a lower extent in this field. By screening for piRNA-36712, a novel tumor suppressor, using cancer genome atlas data analysis, researchers found that piRNA-36712 expression is significantly lower in breast cancer cells than in normal breast tissues. Moreover, this piRNA suppresses p53 through high selenoprotein W pseudogene expression, thus promoting the proliferation, invasion, and migration of cancer cells. Moreover, piRNA-36712 also produces synergistic anticancer effects with paclitaxel and DOX [101].

3.2.3. Tumor Microenvironment Regulates wtp53

The tumor microenvironment refers to the internal environment wherein tumor cells arise and survive. It contains cancer-associated fibroblasts (CAFs), ECM, microvasculature, and various cytokines and chemokines that can modulate chemosensitivity [102,103]. The ECM and CAFs form a protective barrier, preventing drug diffusion and activating resistance pathways through anti-apoptotic effects and microenvironmental remodeling (e.g., hypoxia and metabolic stress) [81]. When cancer cells are co-cultured with CAFs or in CAF-conditioned medium, DNA damage and the p53-mediated response to chemotherapy drugs are reduced, and drug resistance is enhanced via the inhibition of drug accumulation and the antagonization of drug-induced oxidative stress [104]. Moreover, the interleukin-6 (IL-6) produced by CAFs can reduce the response of p53 to DOX [105]. In addition to increasing the levels of MRPs and anti-apoptotic proteins, chronic exposure to DOX induces an MDR phenotype in drug-resistant MCF-7 cells through the upregulation of the ECM glycoprotein pp-GalNAc-T6 (which mainly constitutes carcinoembryonic fibronectin) [80]. The integrin-dependent downregulation of p53 levels in melanoma, sarcoma, and fibroblasts results in increased cell survival and resistance to apoptosis [106]. Furthermore, in breast cancer cells, ECM stiffness affects the p53 activation induced by DOX. A soft ECM downregulates RHO kinase 2 (ROCK2), which drives chemoresistance by inhibiting p53 activation [107]. EMT plays an important role in cancer cell plasticity, leading to tumor heterogeneity. Studies have shown that activation of the EMT program plays a key role in drug resistance by converting non-CSCs to CSCs. Leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5), an EMT inducer, promotes DOX resistance by regulating the programmed cell death 5 (PDCD5)/p53 pathway to inhibit apoptosis [108]. Tight junctions (TJs) maintain cell polarity, permeability, and adhesion and participate in the regulation of cell proliferation and differentiation. The Claudin (CLDN) family is an integral part of TJs. CLDN6 is mainly expressed in tumor tissues. CLDN6 interacts with p53 and promotes its translocation from the nucleus to the cytoplasm, where p53 positively regulates glutathione S-transferase-p1 (GSTP1) to promote DOX resistance in MCF-7 cells [109]. Hypoxia, a common feature of all solid tumors, can downregulate p53 and promote vascularization, EMT, mobility, and metastasis in cancer cells. Further, it can alter metabolism and promote resistance to anticancer therapies [110].

4. Nutrients Can Attenuate DOX Toxicity and Drug Resistance by Targeting p53

There are two major obstacles that limit the clinical application of DOX: tumor drug resistance and cardiotoxicity (Figure 1). Hence, researchers have attempted to use the dual model of cardiac cytotoxicity/chemotherapeutic efficacy to screen effective chemotherapeutic adjuvants that can be administered in combination with DOX. Figure 2 summarizes recent findings on the use of various dietary nutrients and natural products for preventing DOX toxicity and resistance via p53 modulation. Their differential effects on p53 in different settings indicate different cellular outcomes, which may explain the possible mechanisms underlying these effects.

4.1. Dietary Nutrients

In recent years, antitumor dietary therapy has become an important approach in cancer treatment. Some dietary nutrients and their derivatives have been experimentally shown to increase DOX antitumor activity and are considered safe and convenient as supplements to established therapies. Dietary changes do not alter nutrient levels in tumors and healthy tissues in a balanced manner. Further, similar to new drug combinations, the combination of dietary nutrients and DOX must be adapted based on the specific tumor and its metabolic activity, and clinical trials are required before an optimal anticancer regimen may be identified [111]. Here, we summarize the specific mechanisms by which vitamins, fatty acids, glucose analogs, and the micronutrient selenium regulate DIC and DOX resistance via p53.

4.1.1. Vitamins

Vitamin C (L-ascorbic acid) is a well-known antioxidant, and several studies have investigated the beneficial effects of vitamin C in combination with DOX [112]. One of these studies noted that that low concentrations of L-ascorbic acid (below 7 mM) can downregulate some apoptotic, cell survival-, and cell cycle-related proteins, without affecting the RNA levels of p53, ultimately increasing the sensitivity of HeLa cells to cisplatin and DOX via the induction of a DNA damage response [113]. Significant attenuation of DIC via the inhibition of apoptotic proteins and antioxidants was also observed in isolated rat cardiomyocytes pretreated with vitamin C [114].
Vitamin D is a fat-soluble vitamin. Vitamin D2 coupled with DOX (VitD-DOX) has emerged as a novel DOX administration modality. The treatment of human osteosarcoma cells with 10 µM VitD-DOX for 24 h was found to significantly activate the expression of p53 [115]. Likewise, vitamin D3 analogue EB 1089 could interfere with MAPK activity and promote apoptosis in wtp53 MCF-7 cells when administered in combination with DOX, but it was ineffective in mutp53 MCF-7 cells [116]. Furthermore, in female mice with triple-negative mammary tumors, vitamin D supplementation (10,000 IU/kg) was found to reduce DIC in vivo by reducing ROS and mitochondrial damage without decreasing the anticancer effects of DOX [117].
Nicotinamide (NAM) belongs to the vitamin B group. Studies have shown that exogenous NAM selectively inhibits the proliferation of DOX-resistant K562 cells and promotes apoptosis by activating p53 [118,119]. A recent comparative study revealed that the administration of NAM (600 mg/kg) and the active vitamin D analogue 1α(OH)D3 (0.5 μg/kg) for 4 weeks effectively reverses DIC in rats. Mechanistically, the combination of NAM and vitamin D prevented the myocardial mitochondria-mediated apoptotic cascade by maintaining Ca2+ endostasis. In addition, the NAM-induced blockade of inflammatory signaling provides a better cardioprotective effect than 1α(OH)D3 [120].
α-lipoic acid (LA) is called a “super antioxidant” and is a vitamin-like water- and fat-soluble metabolic antioxidant. Mutp53 degradation may attenuate its pro-tumor and metastatic activity. A recent clinical study showed that LA treatment (600 mg daily) can significantly decrease oxidative stress and inflammatory markers [121]. In this context, LA may enable supportive treatment for cancer and may represent a promising adjuvant therapy for mitigating DIC.

4.1.2. Glucose Analogs

Cancer cells use glycolysis as their major mode of energy metabolism, and this metabolic phenotype is an important factor in the progression of MDR to DOX [122]. The glucose analogue 2-deoxy-D-glucose (2-DG) antagonizes DIC by mimicking the effects of caloric restriction at the cellular level. It achieves this by blocking ATP depletion, inhibiting myocardial apoptosis and AMPK activation, and inhibiting DOX-induced deleterious autophagy, without affecting caloric intake [123]. 2-DG significantly inhibits drug resistance in MCF-7 cells via the upregulation of p53 and downregulation of P-gp [122]. In addition, the combination of metformin and 2-DG can reverse drug resistance in MCF-7 cells by restoring p53 function and increasing DOX accumulation through the inhibition of murine MDM2 and MDM4 [124]. The rational design of energy inhibitors or caloric restriction mimetics for DOX and tumor cells’ energy metabolism characteristics warrants further investigation.

4.1.3. Fatty Acids

The dysregulation of lipid metabolism is one of the most prominent metabolic alterations in cancer, and many experts believe that saturated fatty acids are an important cause of human cancers. Treatment with the saturated fatty acids palmitic acid, stearic acid, and myristic acid reduces the accumulation of p53 and promotes tumorigenesis [125]. In contrast, physiological concentrations of exogenous palmitic acid (10–200 μM) can act as adjuvants in endometrial cancer treatment, potentially by significantly inhibiting the cell cycle blockade proteins FAS, p53, and Cyclin D1 and inducing DNA damage, autophagy, and apoptosis [126]. Different polyunsaturated fatty acids (PUFAs) have different effects on the treatment of malignancies. Epidemiological evidence suggests that Ω-6 PUFAs promote cancer, while Ω-3 PUFAs prevent it [127]. Jing et al. found that Ω-3 PUFA treatment results in autophagy via p53-mediated AMPK/mTOR signaling and sensitizes tumor cells to apoptosis [128]. In addition, a population study showed that long-term supplementation with Ω-3 PUFA attenuates DIC in pediatric patients with acute lymphoblastic leukemia [129]. In contrast, Bose et al. proposed that Ω-6 PUFAs have a diametrically opposite effect in cancer prevention and treatment. In their study, the intake of Ω-6 PUFAs increased the formation of the metabolite 4-hydroxynonenal, increased lipid peroxidation, and improved DOX chemosensitivity in mutp53 breast cancer cells [127].

4.1.4. Micronutrients

The micronutrient selenium inhibits thioredoxin reductase activity, thus interfering with oxidative stress. As a result, it boosts the efficacy of anticancer drugs and is considered as an adjuvant to chemotherapy [130]. However, large doses (cumulative 1.3 g) of selenium can produce serious toxicity [131]. Therefore, selenium polymer nanocarriers were recently developed to induce cell cycle arrest and p53-mediated caspase-independent apoptosis in order to arrest cancer cell proliferation, which could reduce the cytotoxicity of many chemotherapeutic drugs, including DOX [130]. Notably, mice fed selenium-supplemented diets showed significantly lower DOX-induced oxidative damage and inflammation in cardiac tissues. In other words, controlled selenium supplementation and the development of nanoformulations could help in the application of selenium in adjuvant strategies for chemotherapy [132].

4.2. Natural Products

The wide-ranging pharmacological properties of natural products have encouraged scientists to seek effective natural substances, or their more effective derivatives or prodrugs, as adjuvants to DOX treatment [133]. Natural products are typically considered to be relatively inexpensive, less toxic, and easy to use. Flavonoids, sesquiterpenoids, alkaloids, diterpenoids, and saponins have all demonstrated excellent potential in combating the toxicity and adverse effects of DOX.
Flavonoids are a class of compounds with a 2-phenyl chromogenic ketone structure and are widely found in nature [134]. Quercetin (QUE), the most abundant dietary flavonol, shows excellent efficacy in combination with DOX due to its potent biological activity. On the one hand, QUE significantly increases the sensitivity of pancreatic cancer and hepatocellular carcinoma cells to gemcitabine and DOX by decreasing hypoxia-inducible factor-1α (HIF-1α) expression and inhibiting P-gp [135]. On the other hand, the anti-inflammatory and antioxidant activity of QUE provides protective effects to normal cells. QUE selectively enhances the toxic effects of DOX against hepatocellular carcinoma cells without damaging normal hepatocytes [136]. In a mouse oral QUE model and H9c2 cells, QUE was found to reduce oxidative stress and myocardial apoptosis by downregulating p53 and NADPH oxidase 1 [137]. The combination of QUE and DOX also provided p53-related hepatoprotective effects to the liver and immune system in a rat model [138,139].
Epigallocatechin-3-gallate (EGCG) is the most abundant and biologically active flavanole in tea and is widely used as a health-promoting food ingredient. EGCG in combination with DOX can inhibit bladder cancer cell proliferation by increasing p53 expression [140]. In a DIC model, EGCG (40 mg/kg) pretreatment for 4 weeks exerted anti-inflammatory and apoptotic effects on heart cells by inhibiting the expression of Nuclear factor-κB (NF-κB), p53, caspase 3, and caspase 12 [141].
Dihydromyricetin (DHM) is a special flavonoid compound obtained from vine tea (ampelopsis grossedentata). DHM (25 μM) can increase the sensitivity of drug-resistant human ovarian cancer cells to paclitaxel and DOX via the p53-mediated downregulation of survivin [142]. DHM (50 μM) was also found to attenuate cardiomyocyte proliferation arrest and apoptosis by regulating MDM2 and promoting the accumulation of ARC, an important anti-apoptotic factor, in H9c2 cells [143].
Apigenin is a naturally occurring flavone and is also known as “phytoestrogen”. The synergistic anticancer activity of apigenin and DOX in K562 cells was linked to the increased expression of p53 and downregulation of BAX and Bcl-2 [144]. Apigenin (100 mg/kg) not only reduced DIC in Wistar rats by modulating the p38/JNK/p53 signaling pathway, but also significantly upregulated antioxidant responses [145].
Isorhamnetin is a promising and novel antitumor flavonoid extracted from the sea-buckthorn (Hippophae rhamnoides L.). When co-administered with DOX, it was found to decrease viability and increase apoptosis in MCF-7, HepG2, and Hep2 cells, and these effects were more prominent than those in the DOX alone group [146]. Isorhamnetin has also been demonstrated to provide antioxidant effects against DOX-induced oxidative stress and to inhibit the mitochondrial apoptosis (decreasing p53) pathway [146].
Dihydroartemisinin contains peroxisomal sesquiterpene lactones with potent antimalarial and anticancer activities. The combination of dihydroartemisinin and DOX at an optimal concentration of 10 µg/mL increases chemosensitivity to DOX [147]. In hepatocellular carcinoma cells expressing mutp53 (R248Q), dihydroartemisinin can reduce P-gp expression and restore DOX sensitivity by inhibiting the p53/ERK1/2/NF-κB signaling pathway [63]. In an animal study, no toxic side effects were observed in the livers, kidneys, spleens, or hearts of tumor-bearing mice treated with dihydroartemisinin (15 mg/kg) [147]. In contrast, dihydroartemisinin was reported to be a pharmacological translational control tumor protein inhibitor (30 mg/kg), inducing heart failure and cardiomyocyte death in mice following continuous intraperitoneal injection for 4 weeks. Hence, its dose-dependent toxicity still needs further investigation [148].
Curcumin (CUR) and resveratrol (RES) are natural polyphenol compounds with multiple activities, including anti-inflammatory, antioxidant, anti-proliferative, and anti-angiogenic effects. CUR is classified by the National Cancer Institute as a third-generation oncotherapeutic agent [149], with a unique role as a p300 histone acetyltransferase inhibitor in DOX-resistant cancer cells. Thus, it can enhance the transcriptional activity of p53 and endogenous caspase cascade activation by relieving the inhibition of the NF-κB pathway, and thus, triggering p53-p300 crosstalk [150]. Furthermore, CUR (below 50 μM) treatment induces p53/p21-dependent apoptosis and significantly inhibits SH-SY5Y cell growth and migration [151]. An in vitro study showed that CUR treatment (200 mg/kg/day, gavage) can significantly downregulate the expression of genes such as Rac1 and p53 in myocardial tissue, highlighting a potential application of CUR in the prevention of DIC [152]. Meanwhile, RES, an anthraquinone terpenoid that is highly abundant in grapes, was first reported to have an anticancer effect in 1997 [153]. In recent years, the mechanism underlying the inhibitory effects of RES and DOX on the production, proliferation, and metastasis of various tumor cells has been discovered. For example, RES (10 μg/mL) triggered growth inhibition in lymphocytic leukemia cell lines through the hyperactivation of phosphorylation at the phosphatase and tensin homolog (PTEN) and p53 ser15 [154]. RES (50–100 µM) treatment increased the expression of both BAX and Bcl-2 and inhibited P-gp expression in HepG2 and MCF-7 cell lines, while upregulating p53 only marginally [155]. Further, the effects of RES treatment (10–100 μM) on DOX B16 melanoma cell subline growth were shown to be induced by increased p53 expression levels [156]. In normal cells, RES acted as a SIRT1 agonist and improved cardiac function and survival by restoring SIRT1 activity, thereby reducing p53 acetylation. RES attenuated USP7, a p53 deubiquitinating protein, and inhibited the pro-apoptotic markers p53, BAX, and caspase 3 by activating SIRT1 [157].
Berberine (BER), mainly obtained from the Chinese herbal medicine Huanglian, is a quaternary alkaloid with yellow needle-like crystals. Daily administration of BER (5 mg/kg) via oral gavage can enhance sensitivity to DOX by inhibiting P-gp [158]. Moreover, high-dose BER (200 mg/kg) directly induces apoptosis in cancer cells by activating the AMPK/p53 pathway [158]. In addition, BER induces autophagy and apoptosis in p53-negative leukemia cells in a non-p53-dependent manner by down-regulating the expression of MDM2 at the transcriptional and post-transcriptional levels [159]. Notably, BER pretreatment promotes p53 deacetylation via SIRT1/3 levels, thereby preventing cardiomyocyte apoptosis, providing a promising approach against DIC [160].
Thymoquinone (TQ), a quinone isolated from black seed (Nigella sativa), has been demonstrated to have anticancer properties. TQ stimulates ROS to a greater extent in mutp53 cells than in wtp53 cells, and this could be because the activation of p53 induces cell cycle arrest to repair TQ-induced damage [161]. TQ upregulates the transcriptional levels of PTEN, a tumor suppressor that exerts anti-proliferative effects, leading to the phosphorylation of p53/p21. Thus, it induces a G2/M phase block and apoptosis in MCF-7 cells [162]. Moreover, El-Far et al. showed that combined treatment with TQ (50 µM), CUR (15 µM), and caffeine (10 mM) can result in significant upregulation of the senescence marker β-galactosidase, as well as p53/p21, increasing DOX efficacy [163]. Correspondingly, the administration of 50 mg/kg TQ for 8 weeks in rats inhibits DOX-induced apoptosis and fibrosis, suggesting that TQ may be a potential therapeutic agent for reducing DIC [164].
Even though some molecules have shown promise, due to the potential of unknown toxicity, low bioavailability, and DOX interactions, clinical studies are required to validate the results seen in preclinical models. Subsequent studies should concentrate on the synergistic effects of natural products in combination with DOX.

5. Dual-Drug Strategy to Overcome both DOX Resistance and Cardiotoxic Side Effects by Targeting p53

Given that p53 is a key nexus for various tumor suppression pathways, extensive research has been devoted to determining how p53 function can be restored in human cancers in order to reduce cellular resistance to DOX therapy. Many anti-DIC therapeutic strategies have also been investigated to gain cardioprotective effects by inhibiting p53 activity. To date, in addition to natural products and nutrients, several DOX dual-drug strategies have been developed to improve chemosensitivity while reducing DOX dose and improving its efficacy, as summarized and discussed in Figure 3.
In recent years, nanotechnology-based carrier co-delivery systems have attracted great interest for the delivery of chemotherapeutic drugs [165]. DOX nano-formulations (e.g., liposomes [166], polymeric micelles [167], and DOX nanogels [168]) not only promote drug accumulation in cancer cells through enhanced permeation and retention, but also minimize non-specific DOX toxicity due to their unique molecular structure. This novel and simple model of direct intervention during DOX release control and delivery can directly increase p53 levels and reverse MDR, and has significant translational potential. In addition, DOX can be directly co-delivered with p53, other chemotherapeutic agents (e.g., methotrexate and rapamycin) [169], or MDR reversal agents (e.g., efflux pump inhibitors or redox cell state modulators) via nanocarriers to directly or indirectly synergistically induce cancer cell apoptosis by increasing p53 [167,170,171]. However, most drugs are still in the research stage. This is because there are various limitations to the clinical application of nanoparticle-mediated therapy, including high production costs, batch inconsistency in nanoparticles, and unformulated test protocols and models.
Similar to nanosystems, cell-penetrating peptides (CPPs) can bind to conventional chemotherapeutic drugs such as DOX, enabling preferential DOX uptake and growth inhibition. They can also be directly coupled to p53, leading to p53 overexpression in tumor cells [172]. The use of CPPs is limited by issues such as dose toxicity, poor water solubility, and immunogenic resistance, and more studies and clinical trials are required to address these drawbacks and test their effects on different tumor types [173].
Cyclotherapy is an emerging therapeutic strategy. In p53 gene-based cyclotherapies, the apoptosis of mutp53 cancer cells can be specifically achieved using low doses of p53 gene activators, such as nutlin-3 and actinomycin D, without affecting the cell cycle in normal cells carrying wtp53 [174].
In recent years, epigenetic mechanisms have become a hot topic in the development of antitumor drugs, and HDAC inhibitors (HDACi) are the first class of epigenetic drugs approved for tumor treatment [175]. In cancer cells, HDAC overexpression leads to enhanced histone deacetylation in the p53 gene, making the nucleosomes compact and decreasing p53 gene expression. HDAC inhibition may promote cell death via p53 deacetylation. Clinical trials have reported the synergistic effects of HDACi and DOX combination therapy [176], and studies using animal models have found that butyroyloxymethyl not only increases the anticancer activity of DOX, but also exerts cardioprotective effects [177]. However, Sonnemann et al. demonstrated that the HDACi vorinostat and entinostat exhibit p53-independent cytotoxic activity [178]. Therefore, there is a need to delineate the mechanisms of HDACi and optimize anticancer regimens to make them faster and more effective for clinical use.
In addition to exogenous agents, endogenous small molecules play multiple interesting roles in antitumor effects and protection against DOX toxicity. The redox-metabolizing enzyme pyruvate kinase 2 (PKM2) has been a hot topic of research in recent years [179]. Saleme et al. used PKM2 to treat mice with lung tumors and found that PKM2 combined with DOX could activate p53 expression in tumor tissues and induce tumor cell death. By taking advantage of the differences in oxygen tension between the myocardium and tumors, PKM2 could inhibit the activity of the apoptotic protein p53 in myocardial tissues, thus preventing the apoptosis of myocardial cells while enhancing tumor regression in a lung cancer model [180]. Thus, PKM2 could be a new target for enhancing the safety and efficacy of DOX chemotherapy, but further experimental and clinical studies are needed to determine its safety and suitability.

6. Conclusions and Future Perspectives

As a tumor inhibitor, p53 greatly reduces the efficacy of chemotherapy following its extensive mutation and degradation inactivation. This is one of the main reasons for DOX resistance. Therefore, p53 reactivation is a promising strategy to overcome DOX resistance. It is widely accepted that the inhibition of p53 attenuates DOX-induced damage in non-cancer cells [181,182], but we cannot ignore that inhibiting p53 itself can promote tumor growth and reduce the antitumor activity of DOX. Owing to the potent antitumor efficacy of p53, which inevitably leads to the death of non-cancer cells, and polymorphisms of the p53 gene in the tumor environment, antitumor strategies targeting p53 are an attractive option for improving the clinical utility of DOX. However, this appears to be challenging due to the complexity of p53 gene polymorphisms and regulatory networks [85]. In general, the dynamic and interdisciplinary exchange of knowledge is required among biologists, chemists, materials scientists, and oncologists. Closer collaboration between academia, the pharmaceutical industry, clinical institutions, and regulatory agencies would also support this goal. Combining diverse and complementary expertise will be crucial for successfully addressing the toxicity and resistance challenges in the application of DOX and for better leveraging its antitumor activity.

Author Contributions

Systematic search, Y.G.; writing—original draft preparation, Y.G.; writing—review and editing, Y.G., Y.T. and G.L.; supervision, J.G.; project administration, J.G. and Y.T.; funding acquisition J.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China (82272601), the Natural Science Foundation of Shandong Province (ZR2021MH330), and the Qilu Young Scholar’s Program of Shandong University (21330089963007).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Frederick, C.A.; Williams, L.D.; Ughetto, G.; Van Der Marel, G.A.; Van Boom, J.H.; Rich, A.; Wang, A.H. Structural comparison of anticancer drug-DNA complexes: Adriamycin and daunomycin. Biochemistry 1990, 29, 2538–2549. [Google Scholar] [CrossRef] [PubMed]
  2. Chen, S.H.; Chan, N.-L.; Hsieh, T.-S. New Mechanistic and Functional Insights into DNA Topoisomerases. Annu. Rev. Biochem. 2013, 82, 139–170. [Google Scholar] [CrossRef] [PubMed]
  3. Delgado, J.L.; Hsieh, C.-M.; Chan, N.-L.; Hiasa, H. Topoisomerases as anticancer targets. Biochem. J. 2018, 475, 373–398. [Google Scholar] [CrossRef] [PubMed]
  4. Begleiter, A.; Leith, M.K. Activity of quinone alkylating agents in quinone-resistant cells. Cancer Res. 1990, 50, 2872–2876. [Google Scholar] [PubMed]
  5. Liu, C.; Ma, X.; Zhuang, J.; Liu, L.; Sun, C. Cardiotoxicity of doxorubicin-based cancer treatment: What is the protective cognition that phytochemicals provide us? Pharmacol. Res. 2020, 160, 105062. [Google Scholar] [CrossRef]
  6. Goormaghtigh, E.; Chatelain, P.; Caspers, J.; Ruysschaert, J. Evidence of a complex between adriamycin derivatives and cardiolipin: Possible role in cardiotoxicity. Biochem. Pharmacol. 1980, 29, 3003–3010. [Google Scholar] [CrossRef]
  7. Wang, J.; Zhang, J.; Xiao, M.; Wang, S.; Wang, J.; Guo, Y.; Tang, Y.; Gu, J. Molecular mechanisms of doxorubicin-induced cardiotoxicity: Novel roles of sirtuin 1-mediated signaling pathways. Cell. Mol. Life Sci. 2021, 78, 3105–3125. [Google Scholar] [CrossRef]
  8. Duan, J.; Mansour, H.M.; Zhang, Y.; Deng, X.; Chen, Y.; Wang, J.; Pan, Y.; Zhao, J. Reversion of multidrug resistance by co-encapsulation of doxorubicin and curcumin in chitosan/poly(butyl cyanoacrylate) nanoparticles. Int. J. Pharm. 2012, 426, 193–201. [Google Scholar] [CrossRef]
  9. Szakacs, G.; Paterson, J.K.; Ludwig, J.A.; Booth-Genthe, C.; Gottesman, M.M. Targeting multidrug resistance in cancer. Nat. Rev. Drug Discov. 2006, 5, 219–234. [Google Scholar] [CrossRef]
  10. Gottesman, M.M.; Fojo, T.; Bates, S.E. Multidrug resistance in cancer: Role of ATP–dependent transporters. Nat. Rev. Cancer 2002, 2, 48–58. [Google Scholar] [CrossRef]
  11. Nitiss, J.L. Targeting DNA topoisomerase II in cancer chemotherapy. Nat. Rev. Cancer 2009, 9, 338–350. [Google Scholar] [CrossRef] [PubMed]
  12. Mohammad, R.M.; Muqbil, I.; Lowe, L.; Yedjou, C.; Hsu, H.-Y.; Lin, L.-T.; Siegelin, M.D.; Fimognari, C.; Kumar, N.B.; Dou, Q.P.; et al. Broad targeting of resistance to apoptosis in cancer. Semin. Cancer Biol. 2015, 35, S78–S103. [Google Scholar] [CrossRef] [PubMed]
  13. Kumar, A.; Singh, U.K.; Chaudhary, A. Targeting autophagy to overcome drug resistance in cancer therapy. Future Med. Chem. 2015, 7, 1535–1542. [Google Scholar] [CrossRef] [PubMed]
  14. Lee, D.-H.; Kim, C.; Zhang, L.; Lee, Y.J. Role of p53, PUMA, and Bax in wogonin-induced apoptosis in human cancer cells. Biochem. Pharmacol. 2008, 75, 2020–2033. [Google Scholar] [CrossRef] [PubMed]
  15. Gomes, A.S.; Ramos, H.; Inga, A.; Sousa, E.; Saraiva, L. Structural and Drug Targeting Insights on Mutant p53. Cancers 2021, 13, 3344. [Google Scholar] [CrossRef]
  16. McSweeney, K.M.; Bozza, W.P.; Alterovitz, W.-L.; Zhang, B. Transcriptomic profiling reveals p53 as a key regulator of doxorubicin-induced cardiotoxicity. Cell Death Discov. 2019, 5, 1–11. [Google Scholar] [CrossRef]
  17. Sun, A.; Zou, Y.; Wang, P.; Xu, D.; Gong, H.; Wang, S.; Qin, Y.; Zhang, P.; Chen, Y.; Harada, M.; et al. Mitochondrial Aldehyde Dehydrogenase 2 Plays Protective Roles in Heart Failure After Myocardial Infarction via Suppression of the Cytosolic JNK/p53 Pathway in Mice. J. Am. Heart Assoc. 2014, 3, e000779. [Google Scholar] [CrossRef]
  18. Feng, X.; Liu, X.; Zhang, W.; Xiao, W. p53 directly suppresses BNIP3 expression to protect against hypoxia-induced cell death. EMBO J. 2011, 30, 3397–3415. [Google Scholar] [CrossRef]
  19. Chipuk, J.E.; Kuwana, T.; Bouchier-Hayes, L.; Droin, N.M.; Newmeyer, D.D.; Schuler, M.; Green, D.R. Direct Activation of Bax by p53 Mediates Mitochondrial Membrane Permeabilization and Apoptosis. Science 2004, 303, 1010–1014. [Google Scholar] [CrossRef]
  20. Candas, D.; Li, J.J. MnSOD in oxidative stress response-potential regulation via mitochondrial protein influx. Antioxid. Redox Signal. 2014, 20, 1599–1617. [Google Scholar] [CrossRef]
  21. Wang, E.Y.; Gang, H.; Aviv, Y.; Dhingra, R.; Margulets, V.; Kirshenbaum, L.A. p53 Mediates Autophagy and Cell Death by a Mechanism Contingent On Bnip3. Hypertension 2013, 62, 70–77. [Google Scholar] [CrossRef] [PubMed]
  22. Cunha-Oliveira, T.; Ferreira, L.L.; Coelho, A.R.; Deus, C.M.; Oliveira, P.J. Doxorubicin triggers bioenergetic failure and p53 activation in mouse stem cell-derived cardiomyocytes. Toxicol. Appl. Pharmacol. 2018, 348, 1–13. [Google Scholar] [CrossRef] [PubMed]
  23. Zhu, W.; Zhang, W.; Shou, W.; Field, L.J. P53 inhibition exacerbates late-stage anthracycline cardiotoxicity. Cardiovasc. Res. 2014, 103, 81–89. [Google Scholar] [CrossRef] [PubMed]
  24. Mak, T.W.; Hauck, L.; Grothe, D.; Billia, F. p53 regulates the cardiac transcriptome. Proc. Natl. Acad. Sci. USA 2017, 114, 2331–2336. [Google Scholar] [CrossRef]
  25. Li, J.; Wang, P.-Y.; Long, N.A.; Zhuang, J.; Springer, D.A.; Zou, J.; Lin, Y.; Bleck, C.K.E.; Park, J.-H.; Kang, J.-G.; et al. p53 prevents doxorubicin cardiotoxicity independently of its prototypical tumor suppressor activities. Proc. Natl. Acad. Sci. USA 2019, 116, 19626–19634. [Google Scholar] [CrossRef]
  26. Moe, G.W.; Marín-García, J. Role of cell death in the progression of heart failure. Heart Fail. Rev. 2016, 21, 157–167. [Google Scholar] [CrossRef]
  27. Mariño, G.; Niso-Santano, M.; Baehrecke, E.H.; Kroemer, G. Self-consumption: The interplay of autophagy and apoptosis. Nat. Rev. Mol. Cell Biol. 2014, 15, 81–94. [Google Scholar] [CrossRef]
  28. Zheng, K.; Zhang, Q.; Sheng, Z.; Li, Y.; Lu, H.H. Ciliary Neurotrophic Factor (CNTF) Protects Myocardial Cells from Oxygen Glucose Deprivation (OGD)/Re-Oxygenation via Activation of Akt-Nrf2 Signaling. Cell. Physiol. Biochem. 2018, 51, 1852–1862. [Google Scholar] [CrossRef]
  29. Pan, J.-A.; Tang, Y.; Yu, J.-Y.; Zhang, H.; Zhang, J.-F.; Wang, C.-Q.; Gu, J. miR-146a attenuates apoptosis and modulates autophagy by targeting TAF9b/P53 pathway in doxorubicin-induced cardiotoxicity. Cell Death Dis. 2019, 10, 1–15. [Google Scholar] [CrossRef]
  30. Chen, Q.; Thompson, J.; Hu, Y.; Das, A.; Lesnefsky, E.J. Cardiac Specific Knockout of p53 Decreases ER Stress-Induced Mitochondrial Damage. Front. Cardiovasc. Med. 2019, 6, 10. [Google Scholar] [CrossRef] [PubMed]
  31. Liang, Z.; He, Y.; Hu, X. Cardio-Oncology: Mechanisms, Drug Combinations, and Reverse Cardio-Oncology. Int. J. Mol. Sci. 2022, 23, 10617. [Google Scholar] [CrossRef] [PubMed]
  32. Koleini, N.; Kardami, E. Autophagy and mitophagy in the context of doxorubicin-induced cardiotoxicity. Oncotarget 2017, 8, 46663–46680. [Google Scholar] [CrossRef] [PubMed]
  33. Hoshino, A.; Mita, Y.; Okawa, Y.; Ariyoshi, M.; Iwai-Kanai, E.; Ueyama, T.; Ikeda, K.; Ogata, T.; Matoba, S. Cytosolic p53 inhibits Parkin-mediated mitophagy and promotes mitochondrial dysfunction in the mouse heart. Nat. Commun. 2013, 4, 2308. [Google Scholar] [CrossRef]
  34. Antoniak, S.; Phungphong, S.; Cheng, Z.; Jensen, B.C. Novel Mechanisms of Anthracycline-Induced Cardiovascular Toxicity: A Focus on Thrombosis, Cardiac Atrophy, and Programmed Cell Death. Front. Cardiovasc. Med. 2022, 8, 2196. [Google Scholar] [CrossRef]
  35. Zhu, W.; Soonpaa, M.H.; Chen, H.; Shen, W.; Payne, R.M.; Liechty, E.A.; Caldwell, R.L.; Shou, W.; Field, L.J. Acute Doxorubicin Cardiotoxicity Is Associated With p53-Induced Inhibition of the Mammalian Target of Rapamycin Pathway. Circulation 2009, 119, 99–106. [Google Scholar] [CrossRef] [PubMed]
  36. Hulmi, J.J.; Nissinen, T.A.; Räsänen, M.; Degerman, J.; Lautaoja, J.H.; Hemanthakumar, K.A.; Backman, J.T.; Ritvos, O.; Silvennoinen, M.; Kivelä, R. Prevention of chemotherapy-induced cachexia by ACVR2B ligand blocking has different effects on heart and skeletal muscle. J. Cachex Sarcopenia Muscle 2018, 9, 417–432. [Google Scholar] [CrossRef]
  37. Chen, L.; Liu, S.; Tao, Y. Regulating tumor suppressor genes: Post-translational modifications. Signal Transduct. Target. Ther. 2020, 5, 90. [Google Scholar] [CrossRef]
  38. Roos, W.P.; Thomas, A.D.; Kaina, B. DNA damage and the balance between survival and death in cancer biology. Nat. Rev. Cancer 2016, 16, 20–33. [Google Scholar] [CrossRef]
  39. Yoshida, M.; Shiojima, I.; Ikeda, H.; Komuro, I. Chronic doxorubicin cardiotoxicity is mediated by oxidative DNA damage-ATM-p53-apoptosis pathway and attenuated by pitavastatin through the inhibition of Rac1 activity. J. Mol. Cell. Cardiol. 2009, 47, 698–705. [Google Scholar] [CrossRef]
  40. Zerikiotis, S.; Angelidis, C.; Dhima, I.; Naka, K.K.; Kasioumi, P.; Kalfakakou, V.; Peschos, D.; Vezyraki, P. The increased expression of the inducible Hsp70 (HSP70A1A) in serum of patients with heart failure and its protective effect against the cardiotoxic agent doxorubicin. Mol. Cell. Biochem. 2019, 455, 41–59. [Google Scholar] [CrossRef]
  41. Zhang, C.; Qu, S.; Wei, X.; Feng, Y.; Zhu, H.; Deng, J.; Wang, K.; Liu, K.; Liu, M.; Zhang, H.; et al. HSP25 down-regulation enhanced p53 acetylation by dissociation of SIRT1 from p53 in doxorubicin-induced H9c2 cell apoptosis. Cell Stress Chaperones 2015, 21, 251–260. [Google Scholar] [CrossRef] [PubMed]
  42. Piegari, E.; Russo, R.; Cappetta, D.; Esposito, G.; Urbanek, K.; Dell’aversana, C.; Altucci, L.; Berrino, L.; Rossi, F.; De Angelis, A. MicroRNA-34a regulates doxorubicin-induced cardiotoxicity in rat. Oncotarget 2015, 7, 62312–62326. [Google Scholar] [CrossRef]
  43. Tabrizi, F.B.; Yarmohammadi, F.; Hayes, A.W.; Karimi, G. The modulation of SIRT1 and SIRT3 by natural compounds as a therapeutic target in doxorubicin-induced cardiotoxicity: A review. J. Biochem. Mol. Toxicol. 2022, 36, e22946. [Google Scholar] [CrossRef] [PubMed]
  44. Wu, S.; Lan, J.; Li, L.; Wang, X.; Tong, M.; Fu, L.; Zhang, Y.; Xu, J.; Chen, X.; Chen, H.; et al. Sirt6 protects cardiomyocytes against doxorubicin-induced cardiotoxicity by inhibiting P53/Fas-dependent cell death and augmenting endogenous antioxidant defense mechanisms. Cell Biol. Toxicol. 2021, 28, 1–22. [Google Scholar] [CrossRef] [PubMed]
  45. Matsushima, S.; Sadoshima, J. The role of sirtuins in cardiac disease. Am. J. Physiol. Circ. Physiol. 2015, 309, H1375–H1389. [Google Scholar] [CrossRef]
  46. Ma, J.; Wang, Y.; Zheng, D.; Wei, M.; Xu, H.; Peng, T. Rac1 signalling mediates doxorubicin-induced cardiotoxicity through both reactive oxygen species-dependent and -independent pathways. Cardiovasc. Res. 2013, 97, 77–87. [Google Scholar] [CrossRef]
  47. Otaki, Y.; Takahashi, H.; Watanabe, T.; Funayama, A.; Netsu, S.; Honda, Y.; Narumi, T.; Kadowaki, S.; Hasegawa, H.; Honda, S.; et al. HECT-Type Ubiquitin E3 Ligase ITCH Interacts With Thioredoxin-Interacting Protein and Ameliorates Reactive Oxygen Species–Induced Cardiotoxicity. J. Am. Heart Assoc. 2016, 5, e002485. [Google Scholar] [CrossRef]
  48. Joiner, M.-L.A.; Koval, O.M.; Li, J.; He, B.J.; Allamargot, C.; Gao, Z.; Luczak, E.D.; Hall, D.D.; Fink, B.D.; Chen, B.; et al. CaMKII determines mitochondrial stress responses in heart. Nature 2012, 491, 269–273. [Google Scholar] [CrossRef]
  49. Toko, H.; Takahashi, H.; Kayama, Y.; Oka, T.; Minamino, T.; Okada, S.; Morimoto, S.; Zhan, D.Y.; Terasaki, F.; Anderson, M.E.; et al. Ca2+/calmodulin-dependent kinase IIdelta causes heart failure by accumulation of p53 in dilated cardiomyopathy. Circulation 2010, 122, 891–899. [Google Scholar] [CrossRef]
  50. Pandey, S.; Kuo, W.W.; Ho, T.J.; Yeh, Y.L.; Shen, C.Y.; Chen, R.J.; Chang, R.L.; Pai, P.Y.; Padma, V.V.; Huang, C.Y.; et al. Upregulation of IGF-IIRα intensifies doxorubicin-induced cardiac damage. J. Cell. Biochem. 2019, 120, 16956–16966. [Google Scholar] [CrossRef]
  51. Xu, J.; Patel, N.H.; Gewirtz, D.A. Triangular Relationship between p53, Autophagy, and Chemotherapy Resistance. Int. J. Mol. Sci. 2020, 21, 8991. [Google Scholar] [CrossRef] [PubMed]
  52. Kandoth, C.; McLellan, M.D.; Vandin, F.; Ye, K.; Niu, B.; Lu, C.; Xie, M.; Zhang, Q.; McMichael, J.F.; Wyczalkowski, M.A.; et al. Mutational landscape and significance across 12 major cancer types. Nature 2013, 502, 333–339. [Google Scholar] [CrossRef] [PubMed]
  53. Sabapathy, K.; Lane, D. Therapeutic targeting of p53: All mutants are equal, but some mutants are more equal than others. Nat. Rev. Clin. Oncol. 2018, 15, 13–30. [Google Scholar] [CrossRef]
  54. Wang, F.; Zhang, H.; Wang, H.; Qiu, T.; He, B.; Yang, Q. Combination of AURKA inhibitor and HSP90 inhibitor to treat breast cancer with AURKA overexpression and TP53 mutations. Med Oncol. 2022, 39, 180. [Google Scholar] [CrossRef] [PubMed]
  55. Parrales, A.; Ranjan, A.; Iyer, S.; Padhye, S.; Weir, S.J.; Roy, A.; Iwakuma, T. DNAJA1 controls the fate of misfolded mutant p53 through the mevalonate pathway. Nat. Cell Biol. 2016, 18, 1233–1243. [Google Scholar] [CrossRef] [PubMed]
  56. Wiech, M.; Olszewski, M.B.; Tracz-Gaszewska, Z.; Wawrzynow, B.; Zylicz, M.; Zylicz, A. Molecular Mechanism of Mutant p53 Stabilization: The Role of HSP70 and MDM2. PLoS ONE 2012, 7, e51426. [Google Scholar] [CrossRef]
  57. Nanayakkara, A.K.; Follit, C.A.; Chen, G.; Williams, N.S.; Vogel, P.D.; Wise, J.G. Targeted inhibitors of P-glycoprotein increase chemotherapeutic-induced mortality of multidrug resistant tumor cells. Sci. Rep. 2018, 8, 967. [Google Scholar] [CrossRef]
  58. Johnson, R.A.; Ince, T.A.; Scotto, K.W. Transcriptional Repression by p53 through Direct Binding to a Novel DNA Element. J. Biol. Chem. 2001, 276, 27716–27720. [Google Scholar] [CrossRef]
  59. Fultang, N.; Illendula, A.; Lin, J.; Pandey, M.K.; Klase, Z.; Peethambaran, B. ROR1 regulates chemoresistance in Breast Cancer via modulation of drug efflux pump ABCB1. Sci. Rep. 2020, 10, 1821. [Google Scholar] [CrossRef]
  60. Qi, X.; Chang, Z.; Song, J.; Gao, G.; Shen, Z. Adenovirus-mediated p53 gene therapy reverses resistance of breast cancer cells to adriamycin. Anti-Cancer Drugs 2011, 22, 556–562. [Google Scholar] [CrossRef]
  61. Sampath, J.; Sun, D.; Kidd, V.J.; Grenet, J.; Gandhi, A.; Shapiro, L.H.; Wang, Q.; Zambetti, G.P.; Schuetz, J.D. Mutant p53 Cooperates with ETS and Selectively Up-regulates Human MDR1 Not MRP1. J. Biol. Chem. 2001, 276, 39359–39367. [Google Scholar] [CrossRef] [PubMed]
  62. Stiewe, T.; Haran, T.E. How mutations shape p53 interactions with the genome to promote tumorigenesis and drug resistance. Drug Resist. Updat. 2018, 38, 27–43. [Google Scholar] [CrossRef] [PubMed]
  63. Yang, Y.; He, J.; Chen, J.; Lin, L.; Liu, Y.; Zhou, C.; Su, Y.; Wei, H. Dihydroartemisinin Sensitizes Mutant p53 (R248Q)-Expressing Hepatocellular Carcinoma Cells to Doxorubicin by Inhibiting P-gp Expression. BioMed Res. Int. 2019, 2019, 8207056. [Google Scholar] [CrossRef] [PubMed]
  64. Kanagasabai, R.; Krishnamurthy, K.; Druhan, L.J.; Ilangovan, G. Forced Expression of Heat Shock Protein 27 (Hsp27) Reverses P-Glycoprotein (ABCB1)-mediated Drug Efflux and MDR1 Gene Expression in Adriamycin-resistant Human Breast Cancer Cells. J. Biol. Chem. 2011, 286, 33289–33300. [Google Scholar] [CrossRef]
  65. Pandey, S.; Bourn, J.; Cekanova, M. Mutations of p53 decrease sensitivity to the anthracycline treatments in bladder cancer cells. Oncotarget 2018, 9, 28514–28531. [Google Scholar] [CrossRef] [PubMed]
  66. Kojima, K.; Duvvuri, S.; Ruvolo, V.; Samaniego, F.; Younes, A.; Andreeff, M. Decreased sensitivity of 17p-deleted chronic lymphocytic leukemia cells to a small molecule BCL-2 antagonist ABT-737. Cancer 2012, 118, 1023–1031. [Google Scholar] [CrossRef]
  67. Panaretakis, T.; Pokrovskaja, K.; Shoshan, M.C.; Grandér, D. Activation of Bak, Bax, and BH3-only Proteins in the Apoptotic Response to Doxorubicin. J. Biol. Chem. 2002, 277, 44317–44326. [Google Scholar] [CrossRef]
  68. Wong, R.P.C.; Tsang, W.P.; Chau, P.Y.; Na Co, N.; Tsang, T.Y.; Kwok, T.T. p53-R273H gains new function in induction of drug resistance through down-regulation of procaspase-3. Mol. Cancer Ther. 2007, 6, 1054–1061. [Google Scholar] [CrossRef]
  69. Bergamaschi, D.; Gasco, M.; Hiller, L.; Sullivan, A.; Syed, N.; Trigiante, G.; Yulug, I.; Merlano, M.; Numico, G.; Comino, A.; et al. p53 polymorphism influences response in cancer chemotherapy via modulation of p73-dependent apoptosis. Cancer Cell 2003, 3, 387–402. [Google Scholar] [CrossRef]
  70. Lin, S.; Yu, L.; Song, X.; Bi, J.; Jiang, L.; Wang, Y.; He, M.; Xiao, Q.; Sun, M.; Olopade, O.I.; et al. Intrinsic adriamycin resistance in p53-mutated breast cancer is related to the miR-30c/FANCF/REV1-mediated DNA damage response. Cell Death Dis. 2019, 10, 666. [Google Scholar] [CrossRef]
  71. Xiang, D.; Shigdar, S.; Bean, A.G.; Bruce, M.; Yang, W.; Mathesh, M.; Wang, T.; Yin, W.; Tran, P.; Al Shamaileh, H.; et al. Transforming doxorubicin into a cancer stem cell killer via EpCAM aptamer-mediated delivery. Theranostics 2017, 7, 4071–4086. [Google Scholar] [CrossRef] [PubMed]
  72. Chan, K.S. Molecular Pathways: Targeting Cancer Stem Cells Awakened by Chemotherapy to Abrogate Tumor Repopulation. Clin. Cancer Res. 2016, 22, 802–806. [Google Scholar] [CrossRef] [PubMed]
  73. Marión, R.M.; Strati, K.; Li, H.; Murga, M.; Blanco, R.; Ortega, S.; Fernandez-Capetillo, O.; Serrano, M.; Blasco, M.A. A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity. Nature 2009, 460, 1149–1153. [Google Scholar] [CrossRef] [PubMed]
  74. Koren, E.; Fuchs, Y. The bad seed: Cancer stem cells in tumor development and resistance. Drug Resist. Updates 2016, 28, 1–12. [Google Scholar] [CrossRef]
  75. Carnero, A.; Garcia-Mayea, Y.; Mir, C.; Lorente, J.; Rubio, I.; Lleonart, M. The cancer stem-cell signaling network and resistance to therapy. Cancer Treat. Rev. 2016, 49, 25–36. [Google Scholar] [CrossRef]
  76. Powell, E.; Piwnica-Worms, D.; Piwnica-Worms, H. Contribution of p53 to Metastasis. Cancer Discov. 2014, 4, 405–414. [Google Scholar] [CrossRef]
  77. Wang, S.-P.; Wang, W.-L.; Chang, Y.-L.; Wu, C.-T.; Chao, Y.-C.; Kao, S.-H.; Yuan, A.; Lin, C.-W.; Yang, S.-C.; Chan, W.-K.; et al. p53 controls cancer cell invasion by inducing the MDM2-mediated degradation of Slug. Nat. Cell Biol. 2009, 11, 694–704. [Google Scholar] [CrossRef]
  78. Safa, A.R. Resistance to drugs and cell death in cancer stem cells (CSCs). J. Transl. Sci. 2020, 6, 341. [Google Scholar] [CrossRef]
  79. Yokoi, A.; Matsumoto, T.; Oguri, Y.; Hasegawa, Y.; Tochimoto, M.; Nakagawa, M.; Saegusa, M. Upregulation of fibronectin following loss of p53 function is a poor prognostic factor in ovarian carcinoma with a unique immunophenotype. Cell Commun. Signal. 2020, 18, 1–15. [Google Scholar] [CrossRef]
  80. Dos Reis, J.S.; da Costa Santos, M.A.R.; da Costa, K.M.; Freire-de-Lima, C.G.; Morrot, A.; Previato, J.O.; Previato, L.M.; da Fonseca, L.M.; Freire-de-Lima, L. Increased expression of the pathological O-glycosylated form of oncofetal fibronectin in the multidrug resistance phenotype of cancer cells. Matrix Biol. 2023, 118, 47–68. [Google Scholar] [CrossRef]
  81. Henke, E.; Nandigama, R.; Ergün, S. Extracellular Matrix in the Tumor Microenvironment and Its Impact on Cancer Therapy. Front. Mol. Biosci. 2020, 6, 160. [Google Scholar] [CrossRef] [PubMed]
  82. Karamanos, N.K.; Theocharis, A.D.; Piperigkou, Z.; Manou, D.; Passi, A.; Skandalis, S.S.; Vynios, D.H.; Orian-Rousseau, V.; Ricard-Blum, S.; Schmelzer, C.E.; et al. A guide to the composition and functions of the extracellular matrix. FEBS J. 2021, 288, 6850–6912. [Google Scholar] [CrossRef] [PubMed]
  83. Mantovani, F.; Collavin, L.; Del Sal, G. Mutant p53 as a guardian of the cancer cell. Cell Death Differ. 2019, 26, 199–212. [Google Scholar] [CrossRef] [PubMed]
  84. Zhu, H.; Evans, B.; O’neill, P.; Ren, X.; Xu, Z.; Hait, W.N.; Yang, J.-M. A role for p53 in the regulation of extracellular matrix metalloproteinase inducer in human cancer cells. Cancer Biol. Ther. 2009, 8, 1722–1728. [Google Scholar] [CrossRef] [PubMed]
  85. Cao, X.; Hou, J.; An, Q.; Assaraf, Y.G.; Wang, X. Towards the overcoming of anticancer drug resistance mediated by p53 mutations. Drug Resist. Updates 2020, 49, 100671. [Google Scholar] [CrossRef]
  86. Uddin, M.B.; Wang, Z.; Yang, C. The m6A RNA methylation regulates oncogenic signaling pathways driving cell malignant transformation and carcinogenesis. Mol. Cancer 2021, 20, 1–18. [Google Scholar] [CrossRef]
  87. Roy, K.R.; Uddin, M.B.; Roy, S.C.; Hill, R.A.; Marshall, J.; Li, Y.T.; Chamcheu, J.C.; Lu, H.; Liu, Y.Y. Gb3-cSrc complex in glycosphingolipid-enriched microdomains contributes to the expression of p53 mutant protein and cancer drug resistance via β-catenin-activated RNA methylation. FASEB Bioadv. 2020, 2, 653–667. [Google Scholar] [CrossRef]
  88. Uddin, M.B.; Roy, K.R.; Hosain, S.B.; Khiste, S.K.; Hill, R.A.; Jois, S.D.; Zhao, Y.; Tackett, A.J.; Liu, Y.Y. An N(6)-methyladenosine at the transited codon 273 of p53 pre-mRNA promotes the expression of R273H mutant protein and drug resistance of cancer cells. Biochem. Pharmacol. 2019, 160, 134–145. [Google Scholar] [CrossRef]
  89. Lin, X.; Zhou, L.; Zhong, J.; Zhong, L.; Zhang, R.; Kang, T.; Wu, Y. RNA-binding protein RBM28 can translocate from the nucleolus to the nucleoplasm to inhibit the transcriptional activity of p53. J. Biol. Chem. 2022, 298, 101524. [Google Scholar] [CrossRef]
  90. Guo, L.; Xu, Y.; Zhou, A.; Zhang, L.; Sun, L.; Gao, Y.; Chen, J.; Shan, X.; Zhang, J.; Ge, J.; et al. A stimuli-responsive combination therapy for recovering p53-inactivation associated drug resistance. Mater. Sci. Eng. C 2020, 108, 110403. [Google Scholar] [CrossRef]
  91. Hientz, K.; Mohr, A.; Bhakta-Guha, D.; Efferth, T. The role of p53 in cancer drug resistance and targeted chemotherapy. Oncotarget 2016, 8, 8921–8946. [Google Scholar] [CrossRef] [PubMed]
  92. Kang, X.; Zhang, J.; Tang, L.; Huang, L.; Tong, J.; Fu, Q. OTU deubiquitinase 5 inhibits the progression of non-small cell lung cancer via regulating p53 and PDCD5. Chem. Biol. Drug Des. 2020, 96, 790–800. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, G.; Yu, M.; Wu, B.; Guo, S.; Huang, X.; Zhou, F.; Claret, F.X.; Pan, Y. Jab1/Cops5 contributes to chemoresistance in breast cancer by regulating Rad51. Cell Signal. 2019, 53, 39–48. [Google Scholar] [CrossRef] [PubMed]
  94. Ding, L.; Zhang, Z.; Zhao, C.; Chen, L.; Chen, Z.; Zhang, J.; Liu, Y.; Nie, Y.; He, Y.; Liao, K.; et al. Ribosomal L1 domain-containing protein 1 coordinates with HDM2 to negatively regulate p53 in human colorectal Cancer cells. J. Exp. Clin. Cancer Res. 2021, 40, 245. [Google Scholar] [CrossRef]
  95. Valanejad, L.; Lewis, K.; Wright, M.; Jiang, Y.; D’souza, A.; Karns, R.; Sheridan, R.; Gupta, A.; Bove, K.; Witte, D.; et al. FXR-Gankyrin axis is involved in development of pediatric liver cancer. Carcinogenesis 2017, 38, 738–747. [Google Scholar] [CrossRef]
  96. Dawson, S.P. Hepatocellular carcinoma and the ubiquitin–proteasome system. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2008, 1782, 775–784. [Google Scholar] [CrossRef]
  97. Toledo, F.; Wahl, G.M. Regulating the p53 pathway: In vitro hypotheses, in vivo veritas. Nat. Rev. Cancer 2006, 6, 909–923. [Google Scholar] [CrossRef]
  98. Tian, J.-H.; Liu, S.-H.; Yu, C.-Y.; Wu, L.-G.; Wang, L.-B. The Role of Non-Coding RNAs in Breast Cancer Drug Resistance. Front. Oncol. 2021, 11, 702082. [Google Scholar] [CrossRef]
  99. Cao, Z.; Xu, L.; Zhao, S. Exosome-derived miR-27a produced by PSC-27 cells contributes to prostate cancer chemoresistance through p53. Biochem. Biophys. Res. Commun. 2019, 515, 345–351. [Google Scholar] [CrossRef]
  100. Jiang, B.; Zhao, W.; Shi, M.; Zhang, J.; Chen, A.; Ma, H.; Suleman, M.; Lin, F.; Zhou, L.; Wang, J.; et al. IDH1 Arg-132 mutant promotes tumor formation through down-regulating p53. J. Biol. Chem. 2018, 293, 9747–9758. [Google Scholar] [CrossRef]
  101. Tan, L.; Mai, D.; Zhang, B.; Jiang, X.; Zhang, J.; Bai, R.; Ye, Y.; Li, M.; Pan, L.; Su, J.; et al. PIWI-interacting RNA-36712 restrains breast cancer progression and chemoresistance by interaction with SEPW1 pseudogene SEPW1P RNA. Mol. Cancer 2019, 18, 9. [Google Scholar] [CrossRef] [PubMed]
  102. Parker, T.M.; Gupta, K.; Palma, A.M.; Yekelchyk, M.; Fisher, P.B.; Grossman, S.R.; Won, K.J.; Madan, E.; Moreno, E.; Gogna, R. Cell competition in intratumoral and tumor microenvironment interactions. EMBO J. 2021, 40, e107271. [Google Scholar] [CrossRef]
  103. Chen, S.-H.; Chang, J.-Y. New Insights into Mechanisms of Cisplatin Resistance: From Tumor Cell to Microenvironment. Int. J. Mol. Sci. 2019, 20, 4136. [Google Scholar] [CrossRef] [PubMed]
  104. Cheteh, E.H.; Augsten, M.; Rundqvist, H.; Bianchi, J.; Sarne, V.; Egevad, L.; Bykov, V.J.; Östman, A.; Wiman, K.G. Human cancer-associated fibroblasts enhance glutathione levels and antagonize drug-induced prostate cancer cell death. Cell Death Dis. 2017, 8, e2848. [Google Scholar] [CrossRef]
  105. Cheteh, E.H.; Sarne, V.; Ceder, S.; Bianchi, J.; Augsten, M.; Rundqvist, H.; Egevad, L.; Östman, A.; Wiman, K.G. Interleukin-6 derived from cancer-associated fibroblasts attenuates the p53 response to doxorubicin in prostate cancer cells. Cell Death Discov. 2020, 6, 42. [Google Scholar] [CrossRef]
  106. Lewis, J.M.; Truong, T.N.; Schwartz, M.A. Integrins regulate the apoptotic response to DNA damage through modulation of p53. Proc. Natl. Acad. Sci. USA 2002, 99, 3627–3632. [Google Scholar] [CrossRef]
  107. Ebata, T.; Mitsui, Y.; Sugimoto, W.; Maeda, M.; Araki, K.; Machiyama, H.; Harada, I.; Sawada, Y.; Fujita, H.; Hirata, H.; et al. Substrate Stiffness Influences Doxorubicin-Induced p53 Activation via ROCK2 Expression. BioMed Res. Int. 2017, 2017, 5158961. [Google Scholar] [CrossRef]
  108. Ma, Z.; Guo, D.; Wang, Q.; Liu, P.; Xiao, Y.; Wu, P.; Wang, Y.; Chen, B.; Liu, Z.; Liu, Q. Lgr5-mediated p53 Repression through PDCD5 leads to doxorubicin resistance in Hepatocellular Carcinoma. Theranostics 2019, 9, 2967–2983. [Google Scholar] [CrossRef]
  109. Yang, M.; Li, Y.; Shen, X.; Ruan, Y.; Lu, Y.; Jin, X.; Song, P.; Guo, Y.; Zhang, X.; Qu, H.; et al. CLDN6 promotes chemoresistance through GSTP1 in human breast cancer. J. Exp. Clin. Cancer Res. 2017, 36, 157. [Google Scholar] [CrossRef]
  110. Zhang, Y.; Yapryntseva, M.A.; Vdovin, A.; Maximchik, P.; Zhivotovsky, B.; Gogvadze, V. Modeling hypoxia facilitates cancer cell survival through downregulation of p53 expression. Chem. Interact. 2021, 345, 109553. [Google Scholar] [CrossRef]
  111. Werida, R.H.; Elshafiey, R.A.; Ghoneim, A.; Elzawawy, S.; Mostafa, T.M. Role of alpha-lipoic acid in counteracting paclitaxel- and doxorubicin-induced toxicities: A randomized controlled trial in breast cancer patients. Support. Care Cancer 2022, 30, 7281–7292. [Google Scholar] [CrossRef]
  112. 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]
  113. Wu, T.-M.; Liu, S.-T.; Chen, S.-Y.; Chen, G.-S.; Wu, C.-C.; Huang, S.-M. Mechanisms and Applications of the Anti-cancer Effect of Pharmacological Ascorbic Acid in Cervical Cancer Cells. Front. Oncol. 2020, 10, 1483. [Google Scholar] [CrossRef]
  114. Ludke, A.; Akolkar, G.; Ayyappan, P.; Sharma, A.K.; Singal, P.K. Time course of changes in oxidative stress and stress-induced proteins in cardiomyocytes exposed to doxorubicin and prevention by vitamin C. PLoS ONE 2017, 12, e0179452. [Google Scholar] [CrossRef]
  115. Maayah, Z.H.; Zhang, T.; Forrest, M.L.; Alrushaid, S.; Doschak, M.R.; Davies, N.M.; El-Kadi, A.O.S. DOX-Vit D, a Novel Doxorubicin Delivery Approach, Inhibits Human Osteosarcoma Cell Proliferation by Inducing Apoptosis While Inhibiting Akt and mTOR Signaling Pathways. Pharmaceutics 2018, 10, 144. [Google Scholar] [CrossRef]
  116. Sundaram, S.; Chaudhry, M.; Reardon, D.; Gupta, M.; Gewirtz, D. The vitamin D3 analog EB 1089 enhances the antiproliferative and apoptotic effects of adriamycin in MCF-7 breast tumor cells. Breast Cancer Res. Treat. 2000, 63, 1–10. [Google Scholar] [CrossRef]
  117. Lee, K.J.; Wright, G.; Bryant, H.; Wiggins, L.A.; Zotto, V.L.D.; Schuler, M.; Malozzi, C.; Cohen, M.V.; Gassman, N.R. Cytoprotective Effect of Vitamin D on Doxorubicin-Induced Cardiac Toxicity in Triple Negative Breast Cancer. Int. J. Mol. Sci. 2021, 22, 7439. [Google Scholar] [CrossRef]
  118. Pan, S.; Leng, J.; Deng, X.; Ruan, H.; Zhou, L.; Jamal, M.; Xiao, R.; Xiong, J.; Yin, Q.; Wu, Y.; et al. Nicotinamide increases the sensitivity of chronic myeloid leukemia cells to doxorubicin via the inhibition of SIRT1. J. Cell. Biochem. 2020, 121, 574–586. [Google Scholar] [CrossRef]
  119. Audrito, V.; Vaisitti, T.; Rossi, D.; Gottardi, D.; D’Arena, G.; Laurenti, L.; Gaidano, G.; Malavasi, F.; Deaglio, S. Nicotinamide blocks proliferation and induces apoptosis of chronic lymphocytic leukemia cells through activation of the p53/miR-34a/SIRT1 tumor suppressor network. Cancer Res. 2011, 71, 4473–4483. [Google Scholar] [CrossRef]
  120. Awad, H.H.; El-Derany, M.O.; Mantawy, E.M.; Michel, H.E.; El-Naa, M.M.; El-Din, R.A.S.; El-Brairy, A.I.; El-Demerdash, E. Comparative study on beneficial effects of vitamins B and D in attenuating doxorubicin induced cardiotoxicity in rats: Emphasis on calcium homeostasis. Biomed. Pharmacother. 2021, 140, 111679. [Google Scholar] [CrossRef]
  121. Neitzel, C.; Seiwert, N.; Göder, A.; Diehl, E.; Weber, C.; Nagel, G.; Stroh, S.; Rasenberger, B.; Christmann, M.; Fahrer, J. Lipoic Acid Synergizes with Antineoplastic Drugs in Colorectal Cancer by Targeting p53 for Proteasomal Degradation. Cells 2019, 8, 794. [Google Scholar] [CrossRef] [PubMed]
  122. Ma, S.; Jia, R.; Li, D.; Shen, B. Targeting Cellular Metabolism Chemosensitizes the Doxorubicin-Resistant Human Breast Adenocarcinoma Cells. BioMed Res. Int. 2015, 2015, 453986. [Google Scholar] [CrossRef] [PubMed]
  123. Chen, K.; Xu, X.; Kobayashi, S.; Timm, D.; Jepperson, T.; Liang, Q. Caloric Restriction Mimetic 2-Deoxyglucose Antagonizes Doxorubicin-induced Cardiomyocyte Death by Multiple Mechanisms. J. Biol. Chem. 2011, 286, 21993–22006. [Google Scholar] [CrossRef] [PubMed]
  124. Xue, C.; Wang, C.; Sun, Y.; Meng, Q.; Liu, Z.; Huo, X.; Sun, P.; Sun, H.; Ma, X.; Ma, X.; et al. Targeting P-glycoprotein function, p53 and energy metabolism: Combination of metformin and 2-deoxyglucose reverses the multidrug resistance of MCF-7/Dox cells to doxorubicin. Oncotarget 2017, 8, 8622–8632. [Google Scholar] [CrossRef]
  125. Zeng, L.; Wu, G.-Z.; Goh, K.J.; Lee, Y.M.; Ng, C.C.; You, A.B.; Wang, J.; Jia, D.; Hao, A.; Yu, Q.; et al. Saturated fatty acids modulate cell response to DNA damage: Implication for their role in tumorigenesis. PLoS ONE 2008, 3, e2329. [Google Scholar] [CrossRef]
  126. Wu, Z.-S.; Huang, S.-M.; Wang, Y.-C. Palmitate Enhances the Efficacy of Cisplatin and Doxorubicin against Human Endometrial Carcinoma Cells. Int. J. Mol. Sci. 2021, 23, 80. [Google Scholar] [CrossRef]
  127. Bose, C.; Hindle, A.; Lee, J.; Kopel, J.; Tonk, S.; Palade, P.T.; Singhal, S.S.; Awasthi, S.; Singh, S.P. Anticancer Activity of Ω-6 Fatty Acids through Increased 4-HNE in Breast Cancer Cells. Cancers 2021, 13, 6377. [Google Scholar] [CrossRef]
  128. Jing, K.; Song, K.S.; Shin, S.; Kim, N.; Jeong, S.; Oh, H.R.; Park, J.H.; Seo, K.S.; Heo, J.Y.; Han, J.; et al. Docosahexaenoic acid induces autophagy through p53/AMPK/mTOR signaling and promotes apoptosis in human cancer cells harboring wild-type p53. Autophagy 2011, 7, 1348–1358. [Google Scholar] [CrossRef]
  129. El Amrousy, D.; El-Afify, D.; Khedr, R.; Ibrahim, A.M. Omega 3 fatty acids can reduce early doxorubicin-induced cardiotoxicity in children with acute lymphoblastic leukemia. Pediatr. Blood Cancer 2022, 69, e29496. [Google Scholar] [CrossRef]
  130. Purohit, M.P.; Verma, N.K.; Kar, A.K.; Singh, A.; Ghosh, D.; Patnaik, S. Inhibition of Thioredoxin Reductase by Targeted Selenopolymeric Nanocarriers Synergizes the Therapeutic Efficacy of Doxorubicin in MCF7 Human Breast Cancer Cells. ACS Appl. Mater. Interfaces 2017, 9, 36493–36512. [Google Scholar] [CrossRef]
  131. Aldosary, B.M.; Sutter, M.E.; Schwartz, M.; Morgan, B.W. Case Series Of Selenium Toxicity From A Nutritional Supplement. Clin. Toxicol. 2012, 50, 57–64. [Google Scholar] [CrossRef] [PubMed]
  132. Yang, H.-B.; Lu, Z.-Y.; Yuan, W.; Li, W.-D.; Mao, S. Selenium Attenuates Doxorubicin-Induced Cardiotoxicity Through Nrf2-NLRP3 Pathway. Biol. Trace Element Res. 2021, 200, 2848–2856. [Google Scholar] [CrossRef] [PubMed]
  133. Talib, W.H.; Alsayed, A.R.; Barakat, M.; Abu-Taha, M.I.; Mahmod, A.I. Targeting Drug Chemo-Resistance in Cancer Using Natural Products. Biomedicines 2022, 9, 1353. [Google Scholar] [CrossRef]
  134. Navarro-Hortal, M.D.; Varela-López, A.; Romero-Márquez, J.M.; Rivas-García, L.; Speranza, L.; Battino, M.; Quiles, J.L. Role of flavonoids against adriamycin toxicity. Food Chem. Toxicol. 2020, 146, 111820. [Google Scholar] [CrossRef]
  135. Hassan, S.; Peluso, J.; Chalhoub, S.; Gillet, Y.I.; Benkirane-Jessel, N.; Rochel, N.; Fuhrmann, G.; Ubeaud-Sequier, G. Quercetin potentializes the respective cytotoxic activity of gemcitabine or doxorubicin on 3D culture of AsPC-1 or HepG2 cells, through the inhibition of HIF-1α and MDR1. PLoS ONE 2020, 15, e0240676. [Google Scholar] [CrossRef] [PubMed]
  136. Wang, G.; Zhang, J.; Liu, L.; Sharma, S.; Dong, Q. Quercetin potentiates doxorubicin mediated antitumor effects against liver cancer through p53/Bcl-xl. PLoS ONE 2012, 7, e51764. [Google Scholar] [CrossRef]
  137. Dong, Q.; Chen, L.; Lu, Q.; Sharma, S.; Li, L.; Morimoto, S.; Wang, G. Quercetin attenuates doxorubicin cardiotoxicity by modulating Bmi-1 expression. Br. J. Pharmacol. 2014, 171, 4440–4454. [Google Scholar] [CrossRef] [PubMed]
  138. Farag, M.R.; Moselhy, A.A.A.; El-Mleeh, A.; Aljuaydi, S.H.; Ismail, T.A.; Di Cerbo, A.; Crescenzo, G.; Abou-Zeid, S.M. Quercetin Alleviates the Immunotoxic Impact Mediated by Oxidative Stress and Inflammation Induced by Doxorubicin Exposure in Rats. Antioxidants 2021, 10, 1906. [Google Scholar] [CrossRef]
  139. Ahmed, O.M.; Elkomy, M.H.; Fahim, H.I.; Ashour, M.B.; Naguib, I.A.; Alghamdi, B.S.; Mahmoud, H.U.R.; Ahmed, N.A. Rutin and Quercetin Counter Doxorubicin-Induced Liver Toxicity in Wistar Rats via Their Modulatory Effects on Inflammation, Oxidative Stress, Apoptosis, and Nrf2. Oxidative Med. Cell. Longev. 2022, 2022, 2710607. [Google Scholar] [CrossRef]
  140. Luo, K.W.; Zhu, X.H.; Zhao, T.; Zhong, J.; Gao, H.C.; Luo, X.L.; Huang, W.R. EGCG Enhanced the Anti-tumor Effect of Doxorubicine in Bladder Cancer via NF-κB/MDM2/p53 Pathway. Front. Cell Dev. Biol. 2020, 8, 606123. [Google Scholar] [CrossRef]
  141. Saeed, N.M.; El-Naga, R.N.; El-Bakly, W.M.; Abdel-Rahman, H.M.; ElDin, R.A.S.; El-Demerdash, E. Epigallocatechin-3-gallate pretreatment attenuates doxorubicin-induced cardiotoxicity in rats: A mechanistic study. Biochem. Pharmacol. 2015, 95, 145–155. [Google Scholar] [CrossRef] [PubMed]
  142. Xu, Y.; Wang, S.; Chan, H.F.; Lu, H.; Lin, Z.; He, C.; Chen, M. Dihydromyricetin Induces Apoptosis and Reverses Drug Resistance in Ovarian Cancer Cells by p53-mediated Downregulation of Survivin. Sci. Rep. 2017, 7, srep46060. [Google Scholar] [CrossRef] [PubMed]
  143. Zhu, H.; Luo, P.; Fu, Y.; Wang, J.; Dai, J.; Shao, J.; Yang, X.; Chang, L.; Weng, Q.; Yang, B.; et al. Dihydromyricetin prevents cardiotoxicity and enhances anticancer activity induced by adriamycin. Oncotarget 2015, 6, 3254–3267. [Google Scholar] [CrossRef] [PubMed]
  144. Noorian, M.; Chamani, E.; Salmani, F.; Rezaei, Z.; Khorsandi, K. Effects of doxorubicin and apigenin on chronic myeloid leukemia cells (K562) in vitro: Anti-proliferative and apoptosis induction assessments. Nat. Prod. Res. 2022, 1–9. [Google Scholar] [CrossRef] [PubMed]
  145. Sahu, R.; Dua, T.K.; Das, S.; De Feo, V.; Dewanjee, S. Wheat phenolics suppress doxorubicin-induced cardiotoxicity via inhibition of oxidative stress, MAP kinase activation, NF-κB pathway, PI3K/Akt/mTOR impairment, and cardiac apoptosis. Food Chem. Toxicol. 2019, 125, 503–519. [Google Scholar] [CrossRef] [PubMed]
  146. Sun, J.; Sun, G.; Meng, X.; Wang, H.; Luo, Y.; Qin, M.; Ma, B.; Wang, M.; Cai, D.; Guo, P.; et al. Isorhamnetin Protects against Doxorubicin-Induced Cardiotoxicity In Vivo and In Vitro. PLoS ONE 2013, 8, e64526. [Google Scholar] [CrossRef]
  147. Tai, X.; Cai, X.-B.; Zhang, Z.; Wei, R. In vitro and in vivo inhibition of tumor cell viability by combined dihydroartemisinin and doxorubicin treatment, and the underlying mechanism. Oncol. Lett. 2016, 12, 3701–3706. [Google Scholar] [CrossRef]
  148. Cai, W.; Fujita, T.; Hidaka, Y.; Jin, H.; Suita, K.; Shigeta, M.; Kiyonari, H.; Umemura, M.; Yokoyama, U.; Sadoshima, J.; et al. Translationally controlled tumor protein (TCTP) plays a pivotal role in cardiomyocyte survival through a Bnip3-dependent mechanism. Cell Death Dis. 2019, 10, 549. [Google Scholar] [CrossRef]
  149. Odot, J.; Albert, P.; Carlier, A.; Tarpin, M.; Madoulet, C. In vitro andin vivo anti-tumoral effect of curcumin against melanoma cells. Int. J. Cancer 2004, 111, 381–387. [Google Scholar] [CrossRef]
  150. Sen, G.S.; Mohanty, S.; Hossain, D.M.S.; Bhattacharyya, S.; Banerjee, S.; Chakraborty, J.; Saha, S.; Ray, P.; Bhattacharjee, P.; Mandal, D.; et al. Curcumin enhances the efficacy of chemotherapy by tailoring p65NFκB-p300 cross-talk in favor of p53-p300 in breast cancer. J. Biol. Chem. 2011, 286, 42232–42247. [Google Scholar] [CrossRef]
  151. Namkaew, J.; Jaroonwitchawan, T.; Rujanapun, N.; Saelee, J.; Noisa, P. Combined effects of curcumin and doxorubicin on cell death and cell migration of SH-SY5Y human neuroblastoma cells. Vitr. Cell. Dev. Biol. Anim. 2018, 54, 629–639. [Google Scholar] [CrossRef] [PubMed]
  152. Soliman, N.A.; Abo El Gheit, R.E.; Abdel Ghafar, M.T.; AbuoHashish, N.A.; Ibrahim, M.A.; Abo Safia, H.S.; El-Saka, M.H.; Elshamy, A.M. Unraveling the biomechanistic role of Rac1/TWEAK/Fn14/NF-κB intricate network in experimentally doxorubicin-induced cardiotoxicity in rats: The role of curcumin. J. Biochem. Mol. Toxicol. 2021, 35, e22829. [Google Scholar] [CrossRef] [PubMed]
  153. Jang, M.; Cai, L.; Udeani, G.O.; Slowing, K.V.; Thomas, C.F.; Beecher, C.W.W.; Fong, H.H.S.; Farnsworth, N.R.; Kinghorn, A.D.; Mehta, R.G.; et al. Cancer Chemopreventive Activity of Resveratrol, a Natural Product Derived from Grapes. Science 1997, 275, 218–220. [Google Scholar] [CrossRef] [PubMed]
  154. Ghorbani, A.; Zand, H.; Jeddi-Tehrani, M.; Koohdani, F.; Shidfar, F.; Keshavarz, S.A. PTEN over-expression by resveratrol in acute lymphoblastic leukemia cells along with suppression of AKT/PKB and ERK1/2 in genotoxic stress. J. Nat. Med. 2015, 69, 507–512. [Google Scholar] [CrossRef]
  155. De Angelis, A.; Piegari, E.; Cappetta, D.; Russo, R.; Esposito, G.; Ciuffreda, L.P.; Ferraiolo, F.A.V.; Frati, C.; Fagnoni, F.; Berrino, L.; et al. SIRT1 activation rescues doxorubicin-induced loss of functional competence of human cardiac progenitor cells. Int. J. Cardiol. 2015, 189, 30–44. [Google Scholar] [CrossRef]
  156. Gatouillat, G.; Balasse, E.; Joseph-Pietras, D.; Morjani, H.; Madoulet, C. Resveratrol induces cell-cycle disruption and apoptosis in chemoresistant B16 melanoma. J. Cell. Biochem. 2010, 110, 893–902. [Google Scholar] [CrossRef]
  157. Sin, T.K.; Tam, B.T.; Yung, B.Y.; Yip, S.P.; Chan, L.W.; Wong, C.S.; Ying, M.; Rudd, J.A.; Siu, P.M. Resveratrol protects against doxorubicin-induced cardiotoxicity in aged hearts through the SIRT1-USP7 axis. J. Physiol. 2015, 593, 1887–1899. [Google Scholar] [CrossRef]
  158. Pan, Y.; Zhang, F.; Zhao, Y.; Shao, D.; Zheng, X.; Chen, Y.; He, K.; Li, J.; Chen, L. Berberine Enhances Chemosensitivity and Induces Apoptosis Through Dose-orchestrated AMPK Signaling in Breast Cancer. J. Cancer 2017, 8, 1679–1689. [Google Scholar] [CrossRef]
  159. Liu, J.; Zhu, Z.; Liu, Y.; Wei, L.; Li, B.; Mao, F.; Zhang, J.; Wang, Y.; Liu, Y. MDM2 inhibition-mediated autophagy contributes to the pro-apoptotic effect of berberine in p53-null leukemic cells. Life Sci. 2020, 242, 117228. [Google Scholar] [CrossRef]
  160. Coelho, A.R.; Martins, T.R.; Couto, R.; Deus, C.; Pereira, C.V.; Simões, R.F.; Rizvanov, A.A.; Silva, F.; Cunha-Oliveira, T.; Oliveira, P.J.; et al. Berberine-induced cardioprotection and Sirt3 modulation in doxorubicin-treated H9c2 cardiomyoblasts. Biochim. Biophys. Acta (BBA) Mol. Basis Dis. 2017, 1863, 2904–2923. [Google Scholar] [CrossRef]
  161. Jehan, S.; Zhong, C.; Li, G.; Bakhtiar, S.Z.; Li, D.; Sui, G. Thymoquinone Selectively Induces Hepatocellular Carcinoma Cell Apoptosis in Synergism with Clinical Therapeutics and Dependence of p53 Status. Front. Pharmacol. 2020, 11, 555283. [Google Scholar] [CrossRef] [PubMed]
  162. Arafa, E.S.A.; Zhu, Q.; Shah, Z.I.; Wani, G.; Barakat, B.M.; Racoma, I.; El-Mahdy, M.A.; Wani, A.A. Thymoquinone up-regulates PTEN expression and induces apoptosis in doxorubicin-resistant human breast cancer cells. Mutat. Res. 2011, 706, 28–35. [Google Scholar] [CrossRef] [PubMed]
  163. El-Far, A.H.; Darwish, N.H.E.; Mousa, S.A. Senescent Colon and Breast Cancer Cells Induced by Doxorubicin Exhibit Enhanced Sensitivity to Curcumin, Caffeine, and Thymoquinone. Integr. Cancer Ther. 2020, 19, 1534735419901160. [Google Scholar] [CrossRef] [PubMed]
  164. Pei, Z.; Hu, J.; Bai, Q.; Liu, B.; Cheng, D.; Liu, H.; Na, R.; Yu, Q. Thymoquinone protects against cardiac damage from doxorubicin-induced heart failure in Sprague-Dawley rats. RSC Adv. 2018, 8, 14633–14639. [Google Scholar] [CrossRef] [PubMed]
  165. Curcio, M.; Farfalla, A.; Saletta, F.; Valli, E.; Pantuso, E.; Nicoletta, F.P.; Iemma, F.; Vittorio, O.; Cirillo, G. Functionalized Carbon Nanostructures Versus Drug Resistance: Promising Scenarios in Cancer Treatment. Molecules 2020, 25, 2102. [Google Scholar] [CrossRef]
  166. Richter, M.; Yumul, R.; Wang, H.; Saydaminova, K.; Ho, M.; May, D.; Baldessari, A.; Gough, M.; Drescher, C.; Urban, N.; et al. Preclinical safety and efficacy studies with an affinity-enhanced epithelial junction opener and PEGylated liposomal doxorubicin. Mol. Ther. Methods Clin. Dev. 2015, 2, 15005. [Google Scholar] [CrossRef]
  167. Chen, L.; Ji, F.; Bao, Y.; Xia, J.; Guo, L.; Wang, J.; Li, Y. Biocompatible cationic pullulan-g-desoxycholic acid-g-PEI micelles used to co-deliver drug and gene for cancer therapy. Mater. Sci. Eng. C 2017, 70, 418–429. [Google Scholar] [CrossRef]
  168. Huang, D.; Qian, H.; Qiao, H.; Chen, W.; Feijen, J.; Zhong, Z. Bioresponsive functional nanogels as an emerging platform for cancer therapy. Expert Opin. Drug Deliv. 2018, 15, 703–716. [Google Scholar] [CrossRef]
  169. Bhattarai, J.K.; Neupane, D.; Nepal, B.; Demchenko, A.V.; Stine, K.J. Nanoporous Gold Monolith for High Loading of Unmodified Doxorubicin and Sustained Co-Release of Doxorubicin-Rapamycin. Nanomaterials 2021, 11, 208. [Google Scholar] [CrossRef]
  170. Li, L.; Tian, H.; He, J.; Zhang, M.; Li, Z.; Ni, P. Fabrication of aminated poly(glycidyl methacrylate)-based polymers for co-delivery of anticancer drugs and the p53 gene. J. Mater. Chem. B 2020, 8, 9555–9565. [Google Scholar] [CrossRef]
  171. Wang, G.-H.; Huang, G.-L.; Zhao, Y.; Pu, X.-X.; Li, T.; Deng, J.-J.; Lin, J.-T. ATP triggered drug release and DNA co-delivery systems based on ATP responsive aptamers and polyethylenimine complexes. J. Mater. Chem. B 2016, 4, 3832–3841. [Google Scholar] [CrossRef] [PubMed]
  172. Zhang, T.; Wu, K.; Ding, C.; Sun, K.; Guan, Z.; Wang, X.; Hsieh, J.-T.; He, D.; Fan, J. Inhibiting bladder tumor growth with a cell penetrating R11 peptide derived from the p53 C-terminus. Oncotarget 2015, 6, 37782–37791. [Google Scholar] [CrossRef] [PubMed]
  173. Vale, N.; Duarte, D.; Silva, S.; Correia, A.S.; Costa, B.; Gouveia, M.J.; Ferreira, A. Cell-penetrating peptides in oncologic pharmacotherapy: A review. Pharmacol. Res. 2020, 162, 105231. [Google Scholar] [CrossRef] [PubMed]
  174. Rao, B.; Lain, S.; Thompson, A.M. p53-Based cyclotherapy: Exploiting the ‘guardian of the genome’ to protect normal cells from cytotoxic therapy. Br. J. Cancer 2013, 109, 2954–2958. [Google Scholar] [CrossRef] [PubMed]
  175. Karagiannis, D.; Rampias, T. HDAC Inhibitors: Dissecting Mechanisms of Action to Counter Tumor Heterogeneity. Cancers 2021, 13, 3575. [Google Scholar] [CrossRef]
  176. Munster, P.N.; Marchion, D.; Thomas, S.; Egorin, M.; Minton, S.; Springett, G.; Lee, J.-H.; Simon, G.; Chiappori, A.; Sullivan, D.; et al. Phase I trial of vorinostat and doxorubicin in solid tumours: Histone deacetylase 2 expression as a predictive marker. Br. J. Cancer 2009, 101, 1044–1050. [Google Scholar] [CrossRef]
  177. Tarasenko, N.; Kessler-Icekson, G.; Boer, P.; Inbal, A.; Schlesinger, H.; Phillips, D.R.; Cutts, S.M.; Nudelman, A.; Rephaeli, A. The histone deacetylase inhibitor butyroyloxymethyl diethylphosphate (AN-7) protects normal cells against toxicity of anticancer agents while augmenting their anticancer activity. Investig. New Drugs 2012, 30, 130–143. [Google Scholar] [CrossRef]
  178. Sonnemann, J.; Marx, C.; Becker, S.; Wittig, S.; Palani, C.D.; Krämer, O.H.; Beck, J.F. p53-dependent and p53-independent anticancer effects of different histone deacetylase inhibitors. Br. J. Cancer 2014, 110, 656–667. [Google Scholar] [CrossRef]
  179. Wang, Y.; Zhao, H.; Zhao, P.; Wang, X. Targeting PKM2 promotes chemosensitivity of breast cancer cells in vitro and in vivo. Cancer Biomark. 2021, 32, 221–230. [Google Scholar] [CrossRef]
  180. Saleme, B.; Gurtu, V.; Zhang, Y.; Kinnaird, A.; Boukouris, A.E.; Gopal, K.; Ussher, J.R.; Sutendra, G. Tissue-specific regulation of p53 by PKM2 is redox dependent and provides a therapeutic target for anthracycline-induced cardiotoxicity. Sci. Transl. Med. 2019, 11, eaau8866. [Google Scholar] [CrossRef]
  181. Shafiei-Roudbari, S.-K.; Malekinejad, H.; Janbaz-Aciabar, H.; Razi, M. Crosstalk between E2F1 and P53 transcription factors in doxorubicin-induced DNA damage: Evidence for preventive/protective effects of silymarin. J. Pharm. Pharmacol. 2017, 69, 1116–1124. [Google Scholar] [CrossRef] [PubMed]
  182. Chen, C.-C.; Hong, H.-J.; Hao, W.-R.; Cheng, T.-H.; Liu, J.-C.; Sung, L.-C. Nicorandil prevents doxorubicin-induced human umbilical vein endothelial cell apoptosis. Eur. J. Pharmacol. 2019, 859, 172542. [Google Scholar] [CrossRef] [PubMed]
Figure 1. (a) Mechanism of action of p53 in DOX-induced cardiotoxicity. p53 is activated by DOX-induced DNA damage and reactive oxygen species accumulation, which leads to the development of cardiotoxicity by inducing cardiomyocyte apoptosis, regulating autophagy, inhibiting mitochondrial autophagy, and causing cardiac atrophy. (b) Mechanism of p53 in DOX resistance. p53 is frequently mutated, inhibited, or degradation inactivated in tumor tissues, leading to DOX resistance.
Figure 1. (a) Mechanism of action of p53 in DOX-induced cardiotoxicity. p53 is activated by DOX-induced DNA damage and reactive oxygen species accumulation, which leads to the development of cardiotoxicity by inducing cardiomyocyte apoptosis, regulating autophagy, inhibiting mitochondrial autophagy, and causing cardiac atrophy. (b) Mechanism of p53 in DOX resistance. p53 is frequently mutated, inhibited, or degradation inactivated in tumor tissues, leading to DOX resistance.
Nutrients 15 02259 g001
Figure 2. Mechanism by which nutrients overcome DOX cardiotoxicity and resistance by targeting p53. ↑Arrows indicate upregulation or facilitation; ↓Arrows indicate downregulation or suppression.
Figure 2. Mechanism by which nutrients overcome DOX cardiotoxicity and resistance by targeting p53. ↑Arrows indicate upregulation or facilitation; ↓Arrows indicate downregulation or suppression.
Nutrients 15 02259 g002
Figure 3. p53-targeted approaches to combating DOX cardiotoxicity and medication resistance. It has been demonstrated that a variety of nanostructure-based carrier co-delivery systems, cell-penetrating peptides (CPPs), circulating therapies, histone deacetylase (HDAC) inhibitors, endogenous small molecules, dietary nutrients, and natural products can increase the sensitivity of tumor cells to DOX by modulating p53 activity, and these tactics also have cardioprotective effects.
Figure 3. p53-targeted approaches to combating DOX cardiotoxicity and medication resistance. It has been demonstrated that a variety of nanostructure-based carrier co-delivery systems, cell-penetrating peptides (CPPs), circulating therapies, histone deacetylase (HDAC) inhibitors, endogenous small molecules, dietary nutrients, and natural products can increase the sensitivity of tumor cells to DOX by modulating p53 activity, and these tactics also have cardioprotective effects.
Nutrients 15 02259 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Guo, Y.; Tang, Y.; Lu, G.; Gu, J. p53 at the Crossroads between Doxorubicin-Induced Cardiotoxicity and Resistance: A Nutritional Balancing Act. Nutrients 2023, 15, 2259. https://doi.org/10.3390/nu15102259

AMA Style

Guo Y, Tang Y, Lu G, Gu J. p53 at the Crossroads between Doxorubicin-Induced Cardiotoxicity and Resistance: A Nutritional Balancing Act. Nutrients. 2023; 15(10):2259. https://doi.org/10.3390/nu15102259

Chicago/Turabian Style

Guo, Yuanfang, Yufeng Tang, Guangping Lu, and Junlian Gu. 2023. "p53 at the Crossroads between Doxorubicin-Induced Cardiotoxicity and Resistance: A Nutritional Balancing Act" Nutrients 15, no. 10: 2259. https://doi.org/10.3390/nu15102259

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