Review article

Modulating the p53-MDM2 pathway: the therapeutic potential of natural compounds in cancer treatment

Iman Ramli1, Thamere Cheriet2,3, Anna Maria Posadino4, Roberta Giordo4, Grazia Fenu4, Kingsley Chukwuemeka Nwachukwu5,6, Oluwafemi Adebayo Oyewole7, Charles Oluwaseun Adetunji8, Daniela Calina9[*], Javad Sharifi-Rad10,11,12, Gianfranco Pintus4,13

1Laboratory of Applied Biochemistry, Faculté des Sciences de la Nature et de la Vie, Université Constantine 1 Frères Mentouri, 25000 Constantine, Algeria

2Unité de Valorisation des Ressources Naturelles, Molécules Bioactives et Analyses Physicochimiques et Biologiques, Université Constantine 1 Frères Mentouri, 25000 Constantine, Algeria

3Département Science de la Nature et de la Vie, Faculté des Sciences Exactes et Sciencede la Nature et de la Vie, Université Mohammed Boudiaf-Oum El-Bouaghi, 04000 Oum El-Bouaghi, Algeria

4Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, 07100 Sassari, Italy

5Department of Microbiology, Faculty of Biological Sciences, Abia State University, Uturu, Nigeria

6Covenant Applied Informatics Communication African Centre for Excellence (CApIC-ACE), Covenant University, Ota, Ogun State, Nigeria

7Department of Microbiology, Federal University of Technology, Minna, Nigeria

8Laboratory of Applied Microbiology, Biotechnology and Nanotechnology, Department of Microbiology, Edo State University, Uzairue, Edo State, Nigeria

9Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania

10Universidad Espíritu Santo, Samborondón 092301, Ecuador

11Centro de Estudios Tecnológicos y Universitarios del Golfo, Veracruz, Mexico

12Department of Medicine, College of Medicine, Korea University, Seoul 02841, Republic of Korea

13Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, 27272 Sharjah, United Arab Emirates

EXCLI J 2024;23:Doc1397

 

Abstract

The p53-MDM2 pathway plays a crucial role regulating tumor suppression and is a focal point of cancer research. This literature review delves into the complex interplay between the tumor suppressor protein p53 and its main regulator MDM2, highlighting their interaction and implications in cancer development and progression. The review compiles and summarizes the existing understanding of the biology and regulation of p53 and MDM2, emphasizing their roles in various cellular processes, including cell cycle regulation, DNA repair, apoptosis, and metabolism. It also discusses the disruption of the p53-MDM2 interaction in different cancer types and its impact on tumor progression and therapeutic resistance. Recent studies have highlighted natural products as promising avenues for targeting the p53-MDM2 pathway. This review categorizes these natural compounds into three groups based on their mechanisms: those that inhibit MDM2 expression or stability, those that prevent p53-MDM2 binding, and those that stabilize p53 by inhibiting MDM2's E3 ligase activity. Detailed mechanisms of action, structural aspects, and potential therapeutic applications of various natural products, including polyphenols, terpenoids, and alkaloids, are presented. Despite significant advances in understanding the p53-MDM2 interaction and the identification of natural products capable of modulating this pathway, further research is necessary to evaluate the clinical efficacy, toxicity, and bioavailability of these compounds. The promising role of natural products in cancer therapy underscores the importance of ongoing investigation and development of these agents as potential treatments for cancer.

Keywords: p53, MDM2, cancer therapy, natural products, tumor suppression

Abbreviation list

Ac: Acetylation

AKT: Ak mouse strain thymoma protein

ARF: ADP ribosylation factor

Bax: Bcl-2-associated X protein

Bcl-xL: B-cell lymphoma-extra large

Bcl-2: B-cell lymphoma 2 protein

c-FLIP: Cellular FLICE-like inhibitory protein

CDK: Cyclin-dependent kinase

CK2: Casein kinase 2

CSCs: Cancer stem cells

CTD: C-terminal domain

DNA-PK: DNA-dependent protein kinase

EGFR: Epidermal growth factor receptor

EGF: Epidermal growth factor

ER: Endoplasmic reticulum

E2F: E2F transcription factor

ETS2: E26 transformation-specific transcription factor 2

FOXO3a: Forkhead box O3

GADD45: Growth arrest and DNA-damage-inducible protein 45

HCC: Hepatocellular carcinoma

HDM2: Human double minute 2 homolog (MDM2)

HSP: Heat shock protein

HSP90: Heat shock protein 90

JAK1/2: Janus kinase 1/2

MAPK: Mitogen-activated protein kinase

MCL: Myeloid cell leukemia

MDM2: Murine double minute 2

mTOR: Mechanistic target of rapamycin

NADH: Nicotinamide adenine dinucleotide (reduced)

NFAT1: Nuclear factor of activated T cells 1

NF-κB: Nuclear factor kappa-light-chain-enhancer of activated B cells

NLS: Nuclear localization signal

NoLS: Nucleolar localization signal

NQO1: NAD(P)H quinone oxidoreductase 1

PARP: Poly (ADP-ribose) polymerase

PI3K: Phosphoinositide 3-kinase

p21: Cyclin-dependent kinase inhibitor 1

p27: Cyclin-dependent kinase inhibitor 1B

p53: Tumor suppressor 53

PML: Promyelocytic leukemia protein

PTEN: Phosphatase and tensin homolog

pRb1: Retinoblastoma protein 1

RING: Really interesting new gene

ROS: Reactive oxygen species

SIRT1: Sirtuin 1

SNP: Single nucleotide polymorphism

TGF-β: Transforming growth factor beta

TP53: Tumor protein p53

TRAIL: TNF-related apoptosis-inducing ligand

VEGF: Vascular endothelial growth factor

VGFA: Vascular endothelial growth factor A

XIAP: X-linked inhibitor of apoptosis protein

Introduction

During a malignant transformation, the accumulation of genomic lesions leads to alterations in gene expression, cell signaling, and cell cycle progression. These phenomena generally define the malignant nature of a cell, indicating sustained proliferative capacity, evasion of growth suppressors, resistance to cell death and replicative senescence, enhanced angiogenesis, and the initiation of invasion and metastasis (Iqbal et al., 2024[91]). A major focus in cancer research is the activation of oncogenes and the inactivation of tumor suppressor genes, both of which play important roles in cancer initiation and progression (Morla-Barcelo et al., 2024[144]). These processes are also recognized as potential targets for cancer therapy. Among tumor suppressors, p53 has been the most extensively studied over recent decades. Known as the “guardian of the genome”, p53 is a potent transcription factor that responds to oncogenic stresses and environmental insults by inducing a series of mechanisms, including senescence, apoptosis activation, and cell cycle arrest. These mechanisms ensure that damaged cells are either repaired or eliminated before they undergo irreversible transformation into cancer cells (K et al., 2024[96]; Liu et al., 2024[125]; Rusin, 2024[190]). The murine double minute 2 (MDM2) is a ubiquitin ligase that keeps low levels of p53 in normal cells (García-Cano et al., 2020[59]). MDM2 serves as the principal p53 regulator in a negative-feedback loop, where p53 initiates MDM2 expression, which in turn induces the monoubiquitinated degradation of p53, leading to the quenching of p53 biological activity (García-Cano et al., 2020[59]; Rusin, 2024[190]). In humans, the TP53 gene encodes p53, while Trp53 encodes it in mice. Mutations in p53 are found in approximately 50 % of human cancers, while amplification of the MDM2 gene occurs in around 17 % of tumors, either in the presence or absence of p53 mutations. Both conditions are associated with poor prognosis and resistance to chemotherapy (Momand et al., 1998[143]; Zhang and Wang, 2000[265]; Levine and Oren, 2009[115]). Consequently, targeting the MDM2-p53 linkage represents an encouraging strategy for cancer therapy. This review offers a novel and detailed exploration of natural compounds as modulators of the p53-MDM2 pathway, an important target in cancer therapy. This comprehensive review categorizes these compounds based on their mechanisms - such as inhibiting MDM2 expression, preventing p53-MDM2 binding, and stabilizing p53. By focusing on the therapeutic potential of natural agents, it presents a unique perspective on developing more effective and less toxic cancer treatments.

Review Methodology

An extensive literature investigation was conducted on different databases, including Scopus, PubMed/MedLine, TRIP databases. The search terms included a combination of keywords related to cancer and natural compounds, focusing on the p53-MDM2 interaction. The specific search strings used were: (tumor OR tumors OR cancer OR cancers OR neoplasms OR neoplasm OR proliferation OR antiproliferative OR metastasis OR metastatic OR angiogenesis OR carcinoma OR growth OR malignancy OR tumor suppressor OR oncogene OR division OR oncoprotein OR prevention OR in vitro OR in vivo OR treatment); (natural compounds OR natural antioxidants OR polyphenols OR terpenoids OR alkaloids); (p53 OR MDM2 OR p53-MDM2 interaction). To ensure the relevance and quality of the studies included in this review, we implemented the following inclusion and exclusion criteria:

Inclusion criteria:

Exclusion criteria:

To ensure the accuracy and reliability of the data, the taxonomy of plant species mentioned in the included studies was validated using the World Flora Online (WFO) database. Additionally, the chemical structures of the natural compounds were verified using PubChem. The most representative data from the included studies have been synthesized and presented in tables and figures to facilitate a clear understanding of the findings. These visual aids summarize key information on mechanistic insight, structural aspects, and potential therapeutic approaches of various natural products targeting the p53-MDM2 pathway.

p53-MDM2 Biology, Regulation and Interaction: Connecting the Dots

Extensive research on p53 and its regulatory network has revealed a high level of complexity. Beyond its well-established role in cancer, p53 is also implicated in the pathogenesis of several diseases, including cardiovascular and infectious diseases, neurodegenerative and metabolic disorders, and autoimmune conditions (Takatori et al., 2014[215]; Siegl and Rudel, 2015[206]; Kung and Murphy, 2016[106]; Szybińska and Leśniak, 2017[214]; Aloni-Grinstein et al., 2018[2]; Maor-Nof et al., 2021[132]; Men et al., 2021[135]). Additionally, P53 was also found to be essential in driving the pathologic effects of COVID-19 (Cardozo and Hainaut, 2021[25]). The expanding understanding of p53's involvement in these diverse pathologies has provided deeper insights into the molecular mechanisms influenced by this protein, including its roles in metabolism, autophagy, translational regulation, and epigenetic control (Levine, 2019[114]; Boutelle and Attardi, 2021[18]).

p53 biology: the guardian of the genome

It is well-known that the regulatory network governing p53 functions is complex and influenced by a multitude of molecular factors. These include the mutation status and post-translational modifications of p53, the response elements (REs) of p53-target genes, the interactions between p53 and its cofactors, as well as the dynamic heterogeneity of p53 activity (Hafner et al., 2017[75]; Farkas et al., 2021[50]). The process of controlling cell fate is remarkably orchestrated by a vast array of p53-target genes and mechanisms, numbering over 3,500 (Figure 1(Fig. 1)) (Fischer, 2017[52]; Sammons et al., 2020[192]). Primarily, p53 acts as a tumor suppressor by preventing malignant transformation in cells. It achieves this by activating the transcription of targeted genes, which then produce proteins that trigger apoptosis, cell growth arrest, or senescence in response to stress signals (Vousden and Prives, 2009[234]). Additionally, p53 plays a key role in regulating DNA repair systems and is responsible for upregulating genes involved in cell cycle progression, including those related to cell cycle checkpoints and genomic integrity. This regulation facilitates the initiation of cell cycle arrest and/or apoptosis in the presence of DNA damage (Menendez et al., 2009[136]).

In case of genotoxicity and/or genomic instability due to ionizing radiations or chemotherapy, induced p53, elevated levels of p53 work to repair cellular damage by increasing the expression of pro-apoptotic (BAX and PUMA) and cell cycle proteins (GADD45, p21) (Steffens Reinhardt et al., 2023[209]). The induction of p21 and/or GADD45 inhibits the activity of CDC2/cyclin E, halting mitosis and leading to cycle arrest in the G2/M phase (Shangary and Wang, 2009[198]). Another way p53 suppresses tumorigenesis is through the p21-Rb-E2F pathway, which triggers cell senescence in response to oxidative stress, DNA damage, or telomere erosion (Steffens Reinhardt et al., 2023[209]). p53 also promotes base excision repair by upregulating components such as Ape/ref1, OGG1, and Polβ. Additionally, p53 enhances the expression of Ku70, which interacts with BAX, promoting its translocation to the mitochondria, oligomerization, and subsequent cell survival. Other DNA repair systems upregulated by p53 are the mismatch repair and nucleotide excision repair components (Menendez et al., 2009[136]). Beyond these roles, p53 acts as a transcriptional repressor for several genes, including c-fos, myc, VEGF-A, and genes associated with cell survival, all of which are involved in promoting pathways related to survival, proliferation, and angiogenesis (Ginsberg et al., 1991[63]; Zhang et al., 2000[264]; Menendez et al., 2009[136]). Furthermore, p53 regulates the transcription of microRNAs, particularly members of the miR-34 family. Increased levels of miR-34a induced by p53 enhance apoptosis and influence the expression of genes related to DNA repair, apoptosis, cell cycle regulation, and angiogenesis (Fu et al., 2023[55]). These findings highlight p53 as a key regulator that links various cellular and molecular signaling pathways, playing a crucial role in processes such as apoptosis, senescence, cell cycle control, angiogenesis, metabolism, immune response, cell motility, differentiation, migration, and cell-cell communication (Amendolare et al., 2022[5]; Wang et al., 2023[235]).

MDM2 biology: the loyal companion of p53

Elevated levels of MDM2 have been correlated with poor prognosis in several cancer types, including solid tumors of the lung, esophagus, breast, stomach, as well as liposarcomas, glioblastomas, and leukemias (Yao et al., 2024[256]). Various molecular mechanisms contribute to MDM2 overexpression in these cancers, most notably mdm2 gene amplification (Momand et al., 1998[143]). Additionally, a single nucleotide polymorphism at position 309 (SNP309) in the MDM2 gene promoter has been shown to enhance transcription and translation (Yao et al., 2024[256]). MDM2 overexpression is also associated with metastasis and advanced stages of cancers such as osteosarcoma, colon, breast, and prostate cancers, and has been linked to chemotherapy resistance (Lin et al., 2024[123]). During tumorigenesis, MDM2 plays a multifaceted role, regulating key cellular processes such as the cell cycle, apoptosis, angiogenesis, metastasis, metabolism, and DNA synthesis and repair (Zafar et al., 2023[258]) (Figure 2(Fig. 2)).

The MDM2 oncoprotein is regulated through several well-characterized mechanisms (Rayburn et al., 2009[184]). A key regulatory pathway involves p53-induced transcription of mdm2 via the P2 promoter, while the P1 promoter drives basal transcription independently of p53 (Barak et al., 1994[12]). Other transcription factors, including NFAT1 (Zhang et al., 2012[266]), NF-κB (Thomasova et al., 2012[221]), IRF-8, SP1 (Rayburn et al., 2009[184]), Fli-ETS (Truong et al., 2005[223]), and Ras/Raf/MEK/MAPK (Ries et al., 2000[186]), positively modulate MDM2 expression through both the P1 and P2 promoters. Conversely, the tumor suppressor PTEN acts as a negative regulator of MDM2, independent of p53 (Ries et al., 2000[186]). Additionally, microRNAs (miRNAs) such as miR-29, miR-18b, miR-145, and miR-143 initiate epigenetic mechanisms that downregulate MDM2 by inhibiting its mRNA translation (Dar et al., 2013[37]; Zhang et al., 2013[262]). Post-translational modifications, such as phosphorylation, also play an important role in MDM2 regulation. For instance, modifications by the ATM protein reduce MDM2 stability (de Toledo et al., 2000[38]; Maya et al., 2001[133]; Meulmeester et al., 2005[139]), while the Akt pathway facilitates MDM2 translocation from the cytoplasm to the nucleus, promoting p53 degradation (Mayo and Donner, 2001[134]; Zhou et al., 2001[274]; Ogawara et al., 2002[155]; Gama et al., 2009[57]). Other enzymes, such as CK2, DNA-PK, and components of the Ras/Raf/MEK/ MAPK pathway, further regulate MDM2 function (Rayburn et al., 2009[184]). Extensive evidence underscores that MDM2 is a key regulator of multiple cellular processes, independent of its interaction with p53. MDM2 influences DNA synthesis and repair through interactions with DNA polymerase ε (Vlatkovic et al., 2000[231]; Asahara et al., 2003[7]), DHFR (Maguire et al., 2008[129]), centrosome amplification (Carroll et al., 1999[26]), and the MRN DNA repair complex, including Nbs1 (Alt et al., 2005[4]; Bouska et al., 2008[17]). Additionally, MDM2 interacts with cellular proteins such as DNMT3A (Tang et al., 2012[218]), the Rb/E2F-1 complex (Hsieh et al., 1999[82]; Katsube et al., 2003[100]; Uchida et al., 2005[225]), MTBP (Boyd et al., 2000[19]; Brady et al., 2005[20]), p107 (Dubs-Poterszman et al., 1995[43]), and the cyclin-dependent kinase inhibitor p21, promoting cell cycle progression, particularly through the S-phase (Zhang et al., 2004[269]; Xu et al., 2010[249]). MDM2 also plays a role in apoptosis inhibition by modulating both pro-apoptotic and anti-apoptotic proteins. It interacts with the E2F1/Rb pathway (Bouska et al., 2008[17]) and apoptosis mediators such as p73 (through p73 NEDDylation, which prevents p53 transactivation) (Bouska et al., 2008[17]; Malaguarnera et al., 2008[130]), and FOXO3a (by reducing its stability) (Fu et al., 2009[56]). MDM2 further enhances anti-apoptotic signaling by upregulating XIAP, which inactivates caspase-mediated apoptosis (Gu et al., 2009[73]). Beyond its function as a negative regulator of p53, MDM2 is involved in the regulation of proteins important for DNA repair, apoptosis, cell dynamics, and invasion pathways (Bouska et al., 2008[17]; Rayburn et al., 2009[184]; Manfredi, 2010[131]; Li and Lozano, 2013[120]). While the intricate web of MDM2 interactions, both dependent and independent of p53, highlights its multifaceted role in cellular regulation, the MDM2-p53 axis remains central to maintaining normal cellular homeostasis. Over the past decades, research has primarily focused on understanding the MDM2 interactions that influence the cellular and molecular levels of p53, both directly and indirectly (Subhasree et al., 2013[212]).

p53-MDM2 interaction: a tremendous complexity and complementarity

The relationship between MDM2 and p53 is primarily built upon the regulation of the latter and serves as a crucial checkpoint for most stress-mediated signaling pathways that lead to p53 activation and regulation (Levine, 2020[113]). It is well-established that p53 plays a central role in preventing the proliferation of abnormal cells with genetic instabilities. Under normal conditions, p53 levels are kept low by MDM2 to maintain cellular homeostasis (Moll and Petrenko, 2003[142]). In this section, we explore the unique MDM2-p53 interaction, which has been extensively studied as a pivotal process in cancer therapeutics. MDM2 tightly controls a rapid regulatory mechanism that toggles the activation of wild-type (WT) p53 on and off. One of the primary mechanisms by which transformed cells drive tumorigenesis is through the overexpression of MDM2, which inhibits p53's transcriptional activity and reduces its cellular levels (Cahilly-Snyder et al., 1987[22]; Fakharzadeh et al., 1991[46]; Oliner et al., 1993[157]). Reduced MDM2 activity results in p53 mono-ubiquitination and its export from the nucleus, while increased MDM2 activity leads to poly-ubiquitination and subsequent degradation of p53 in the nucleus (Fang et al., 2000[49]; Rodriguez et al., 2000[188]; Lai et al., 200[109]; Lee and Gu, 2010[111]). A widely supported hypothesis regarding the negative autoregulatory loop between p53 and MDM2 is the amplification of the MDM2 gene observed in several human sarcomas with wild-type p53 (Momand et al., 1998[143]). At the molecular level, MDM2 binds to p53 via a primary N-terminal p53-binding domain and contains a C-terminal RING domain, which functions as an E3 ubiquitin ligase. This domain, along with various sequence motifs such as the nucleolar localization signal (NoLS), nuclear localization signal (NLS), and nuclear export signal (NES), facilitates MDM2's localization within the nucleus and its export (Chi et al., 2005[34]; Yu et al., 2006[257]; Poyurovsky et al., 2011[170]). The interaction between MDM2 and p53 is further strengthened by the ability of p53 to bind MDM2 at multiple sites. Through its C-terminal RING domain, MDM2 ubiquitinates p53, leading to its degradation via the proteasome pathway (Haupt et al., 1997[76]; Midgley et al., 2000[140]). However, stress-mediated signaling pathways can disrupt the MDM2-p53 interaction through various mechanisms. These pathways often involve post-translational modifications (PTMs) of p53, such as phosphorylation at serine residues 15, 20, 37, and 106, and threonine 18, which weaken the MDM2-p53 interaction. Acetylation at lysine residues in the C-terminal domain (CTD) of p53 further prevents MDM2-mediated ubiquitination (Shieh et al., 1997[203]; Unger et al., 1999[227]; Nakamura et al., 2000[150]; Rodriguez et al., 2000[188]; Sakaguchi et al., 2000[191]; Li et al., 2002[117]; Hsueh et al., 2013[85]). Additionally, MDM2 can export p53 out of the nucleus (Haupt et al., 1997[76]; Honda et al., 1997[78]), preventing its interaction with transcriptional co-activators (Oliner et al., 1993[157]) and enhancing the transcription of p53 co-repressors (Wu et al., 1993[247]; Thut et al., 1997[222]; Chi et al., 2005[34]). In turn, wild-type (WT) p53 promotes the transcription of MDM2, establishing a regulatory feedback loop (Barak et al., 1994[12]). This loop finely tunes p53 functions through p53-mediated regulation of MDM2 at its promoter (Barak et al., 1994[12]; Shangary and Wang, 2009[198]; Vousden and Prives, 2009[234]). The complexity of this autoregulatory loop ensures the maintenance of physiological p53 levels in normal cells, as the precise homeostatic concentration of p53 is crucial for proper cell growth and development (Vousden and Prives, 2009[234]) (Figure 3(Fig. 3)).

Considering the diverse activities of the MDM2 E3 ligase, which enable it to target various proteins, the p53-MDM2 axis can either suppress or promote tumor development, depending on the cellular context and the factors involved. For example, MDM2 can degrade the tumor-promoting factor HIF-1α (hypoxia-inducible factor 1-alpha) in a p53-dependent manner (Ravi et al., 2000[183]). In contrast, it has been demonstrated that mutant p53 (mutp53) drives tumorigenesis by dissociating HIF-1α from MDM2, leading to HIF-1α upregulation (Kamat et al., 2007[98]). Another tumor-suppressive function of MDM2 is its ability to degrade mutp53, thereby stabilizing its levels in cancer cells (García-Cano et al., 2020[59]). Since mutp53 cannot upregulate MDM2 expression, it interacts with other factors, such as heat shock proteins (HSP), valosin-containing protein (VCP), and chaperones like HSP90, to disrupt the mutp53-MDM2 complex, resulting in pro-tumorigenic activities (García-Cano et al., 2020[59]). This dual role of MDM2-as both a tumor suppressor and a tumor promoter-provides new insights into the regulation of mutp53 by the p53-MDM2 interaction (Kadosh et al., 2020[97]).

Moreover, several MDM2 activities can synergize with or counteract p53 while functioning independently of it. One key role of mitochondrial p53 is regulating mitochondria-mediated apoptosis and mitochondrial respiration during cancer development (Rusin, 2024[190]). Under hypoxic conditions, MDM2 partially relocates to the mitochondria, where it inhibits mitochondrial respiration by reducing the expression of complex I subunit NADH-dehydrogenase 6 (MT-ND6) (García-Cano et al., 2020[59]). This inhibition increases the production of reactive oxygen species (ROS), which subsequently promotes cancer cell migration and invasion (Arena et al., 2018[6]). Conversely, in the cytoplasm, MDM2 binds to the mitochondrial stabilizer NADH ubiquinone oxidoreductase 75 kDa Fe-S protein 1 (NDUFS1), leading to increased ROS generation and the promotion of apoptosis (Elkholi et al., 2019[45]). Despite the growing understanding of MDM2's role in tumorigenesis, particularly through its mitochondrial functions, the precise mechanisms regulating its pro-tumorigenic and anti-tumorigenic activities remain largely unknown. Further research is needed to elucidate how MDM2 mediates these opposing functions within different cellular compartments.

Role of p53 and MDM2 in cancers

Mutations in p53 have been detected in up to 30 % of all breast cancer cases, and individuals with inherited p53 mutations face an elevated risk of developing ovarian, breast, pancreatic, and colorectal cancers (Lacroix et al., 2006[108]; Muller and Vousden, 2013[148]). While p53 retains its wild-type form in nearly 50 % of tumors, TP53 remains the most frequently mutated gene in human cancers, where p53 function is often impaired or compromised (Herrero et al., 2016[77]; Wasylishen and Lozano, 2016[241]). In parallel, MDM2 overexpression, driven by gene amplification or single nucleotide polymorphisms, is well-established in the tumorigenesis of several human cancers (Bond et al., 2004[16]; Oliner et al., 2016[158]). Importantly, MDM2 overexpression and p53 mutations typically occur as mutually exclusive events, reinforcing the idea that cancer phenotypes are often driven by disruptions in the p53-MDM2 interaction (Oliner et al., 1992[156], 2016[158]; Bond et al., 2004[16]). Additionally, MDM2 contributes to tumorigenesis through other oncogenic activities, including its pro-angiogenic effects, induction of chromosomal instability, degradation of cell cycle regulators, and promotion of epithelial-mesenchymal transition (EMT) via the degradation of E-cadherin (Alt et al., 2005[4]; Miwa et al., 2006[141]; Yang et al., 2006[252], 2008[253]; Bouska et al., 2008[17]; Muthumani et al., 2014[149]; Wolf et al., 2020[244]). Recent research has also linked MDM2 overexpression to resistance against conventional chemotherapy (Hou et al., 2019[81]). For example, p53 gene mutations are a key risk factor for breast cancer (Gasco et al., 2002[60]), where mutant p53 enhances migration, invasion, angiogenesis, scattering, stem cell proliferation, survival, and tissue remodeling. The role of p53 in breast cancer progression is complex and multifaceted, with p53 interacting with other signaling pathways, including the Wnt/β-catenin and PI3K/Akt/mTOR pathways, to modulate breast cancer cell behavior (Reddy et al., 2020[185]; Shahcheraghi et al., 2020[194]). Furthermore, p53 mutations are also an important risk factor in hepatocellular carcinoma (HCC). Epidemiological studies have shown that chronic viral infections (such as HBV, HCV, and HIV) and metabolic disorders (such as metabolic syndrome) are associated with disruption of the MDM2-p53 axis in HCC (Cao et al., 2020[24]). Hepatitis viruses employ multiple mechanisms to persist in hepatocytes, including inducing p53 mutations, silencing or overexpressing MDM2, stabilizing MDM2 levels, and accelerating p53 degradation. These processes trigger various stress responses, including oxidative stress, energy metabolism shifts, chronic ER stress, genetic instability, and abnormal antitumor gene expression, ultimately driving the transformation of hepatocytes into hepatoma cells (Cao et al., 2020[24]). The MDM2-p53 interaction has also been implicated in the regulation of glucolipid metabolism in the liver, where its dysregulation contributes to metabolic diseases such as metabolic syndrome and non-alcoholic fatty liver disease (NAFLD), conditions that can progress to HCC under certain circumstances (Guillen-Sacoto et al., 2017[74]). Studies suggest that lipid accumulation in hepatocytes leads to the overexpression of microRNA-21, which enhances the expression of carcinogenesis-related proteins (CCNB1, CCND1, and SREBP1C) by inhibiting p53. MicroRNA-21 targets HBP1, a transcriptional activator of p53, and its overexpression drives G1/S and G2/S transitions in hepatocytes, promoting de novo lipogenesis by modulating the HBP1-p53 axis. Conversely, knocking down microRNA-21 inhibits the G1/S transition and suppresses hepatoma proliferation (Wu et al., 2016[245]).

The disruption of p53-MDM2 interaction in cancers

The dysregulation of the p53-MDM2 feedback loop is one of the most well-documented disruptions in cancer, often manifesting as overexpression of MDM2 and/or p53. This phenomenon is linked to the activation of stress-induced p53 pathways, which fail to adequately induce growth arrest and/or apoptosis (Klein and Vassilev, 2004[103]). Additionally, the impaired activation of various oncogenes, such as Myc, Ras, E2F-1, and β-catenin, has been shown to enhance ARF activation, a known MDM2 suppressor, which negatively modulates MDM2 function (Eischen et al., 1999[44]; Manfredi, 2010[131]; Hu et al., 2012[86]). As a result, activated p53 exerts tumor-suppressive effects in this context. ARF promotes the sequestration of MDM2 in the nucleolus, reducing its E3 ubiquitin ligase activity and ultimately segregating MDM2, thereby diminishing its negative regulatory effect on p53 (Weber et al., 1999[242]; Zhang and Xiong, 1999[268]). Disruptions in the ARF/MDM2/p53 signaling pathway are frequently observed in various cancer types (Sherr, 2006[201]). Other oncogenes, such as AKT and Wip1, directly regulate MDM2. The IGF-1/AKT oncogenic pathway is closely linked to cell fate and proliferation, where AKT kinase phosphorylates MDM2 at Ser-166 and Ser-186, leading to downregulation of p53 activity (Grossman et al., 1998[71]; Zhou et al., 2001[273]). Wip1, an oncogenic serine/threonine phosphatase induced and regulated by p53 following genotoxic stress, dephosphorylates MDM2 at Ser-395, restoring MDM2 activity and maintaining p53 at steady-state levels, thus ensuring proper regulation of the p53-MDM2 feedback loop. Notably, Wip1 is frequently overexpressed in various human cancers, indicating a disruption of the p53-MDM2 interaction (Peuget et al., 2024[162]). To date, no candidate drugs targeting the MDM2-p53 interaction have been successfully validated in pharmacological stages (preclinical/clinical trials) as cancer therapies. This can be attributed to the low efficacy and high toxicity of the tested prodrugs in experimental models. Understanding these challenges is crucial for making informed decisions and advancing the field of drug development (Zanjirband and Rahgozar, 2019[259]; Mullard, 2020[147]). The accumulated data suggest that a deeper understanding of the complexity and intricacy of the p53-MDM2 interaction is important for gaining valuable insights into cancer therapy. The complex biology and regulation of this interaction, along with its implications in cancer, are summarized in Table 1(Tab. 1) (References in Table 1: Asahara et al., 2003[7]; Dar et al., 2013[37]; de Toledo et al., 2000[38]; Farkas et al., 2021[50]; Fischer, 2017[52]; Fu et al., 2023[55]; Hafner et al., 2017[75]; Levine, 2020[113]; Lin et al., 2024[123]; Maya et al., 2001[133]; Menendez et al., 2009[136]; Meulmeester et al., 2005[139]; Mullard, 2020[147]; Peuget et al., 2024[162]; Rayburn et al., 2009[184]; Sammons et al., 2020[192]; Thomasova et al., 2012[221]; Vlatkovic et al., 2000[231]; Vousden and Prives, 2009[234]; Yao et al., 2024[256]; Zafar et al., 2023[258]; Zanjirband and Rahgozar, 2019[259]; Zhang et al., 2013[262]).

Mechanistic Insights into Natural Products Targeting the p53-MDM2 Pathway

Bioactive natural compounds are well-known for producing a wide array of secondary metabolites with diverse structures, which have played a pivotal role in the development of approximately 50 % of anti-cancer drugs over the past several decades (Kamath et al., 2023[99]; Chaachouay and Zidane, 2024[27]; Chaudhry et al., 2024[29]; Chunarkar-Patil et al., 2024[36]; Ijaz et al., 2024[89]; Nandi et al., 2024[151]). Among these natural molecules, several have been reported to target the p53-MDM2 interaction and can be classified into three categories:

a) Direct inhibitors of MDM2 expression and/or protein stability: These compounds reduce MDM2 levels by inhibiting its expression or destabilizing the protein, thereby enhancing p53 activity. Examples include: i) Curcumin: Downregulates MDM2 expression via the PI3K/mTOR/ETS2 pathway, leading to reduced MDM2 levels and increased p53 activity (Sultana et al., 2021[213]); ii) Resveratrol: Acts as a direct inhibitor of MDM2 expression and prevents MDM2-mediated p53 degradation, thereby promoting p53 stability (Merlin et al., 2021[137]); iii) Gambogic Acid: Inhibits MDM2 by downregulating its expression, stabilizing p53, and inducing apoptosis in cancer cells (Foggetti et al., 2017[53]).

b) Inhibitors of the p53-MDM2 binding and activators of wild-type p53: These compounds disrupt the p53-MDM2 interaction, thereby reactivating wild-type p53. Examples include: i) Nutlin-3: A potent MDM2 antagonist that inhibits the p53-MDM2 interaction, leading to the activation of p53 (Lerma Clavero et al., 2023[112]); ii) Epigallocatechin Gallate (EGCG): Disrupts p53-MDM2 binding, preventing p53 degradation and enhancing its accumulation in cells (Bahena Culhuac and Bello, 2024[11]); iii) Leucomalachite Green (LMG): Inhibits the binding of p53 to MDM2, reactivating p53 in cancer cells (Koo et al., 2022[105]).

c) Inhibitors of MDM2's E3 ligase activity, stabilizing p53: These compounds inhibit MDM2's E3 ligase activity, preventing the ubiquitination and degradation of p53, thus stabilizing and activating p53. Examples include: i) MI-219: Blocks MDM2's E3 ligase activity, stabilizing p53 and enhancing its tumor-suppressive functions (Yang et al., 2021[255]); ii) Lithocholic Acid: Inhibits MDM2's E3 ligase function, leading to increased stability and activity of p53 (Yao et al., 2024[256]); iii) Oridonin: A diterpenoid that inhibits MDM2's E3 ligase activity, resulting in the stabilization and accumulation of p53 (Zhu et al., 2019[276]) (Figure 4(Fig. 4)).

This section will explore the data on natural compounds that modulate the p53-MDM2 interaction, focusing on their structural aspects, binding modes, and mechanisms of action (Table 1(Tab. 1)).

Polyphenols

Flavonoids, a significant group of over 10,000 secondary metabolites (Ullah et al., 2020[226]), ave garnered considerable attention for their ability to counteract free radicals, modulate cellular metabolism, and mitigate oxidative stress associated with several severe diseases, including Parkinson's, Alzheimer's, cardiovascular conditions, and cancer (Vazhappilly et al., 2019[228]; Giordo et al., 2021[69], 2022[68]; Sharifi-Rad et al., 2022[199]; Shaito et al., 2023[195]). Numerous studies suggest that natural flavonoids exhibit potent anticancer activities through various mechanisms, including the inhibition of MDM2 expression (Merlin et al., 2021[137]). Genistein (4′,5,7-trihydroxyisoflavone), a natural isoflavone abundantly found in soybeans, has been shown to downregulate MDM2 at both transcriptional and post-translational levels (Bhat et al., 2021[14]). In vitro chemopreventive studies in several human cancer cell lines revealed that genistein decreased MDM2 expression levels independently of p53. It also inhibited the tyrosine kinase pathway regulating MDM2, while simultaneously increasing p21 levels. In vivo studies further confirmed genistein's antitumor activity, which is related to its inhibitory effects on MDM2 expression (Tuli et al., 2019[224]; Gao et al., 2020[58]). Gao et al. (2020[58]) demonstrated that prolonged genistein treatment significantly reduces epidermal growth factor receptor (EGFR) expression and moderates downstream signaling molecules (JAK1/2, MDM2, STAT3, and Akt phosphorylation), leading to the inhibition of the JAK1/2-STAT3 and AKT/MDM2/p53 pathways. This ultimately results in apoptosis, cell cycle arrest, and reduced proliferation of esophageal carcinoma cells (Gao et al., 2020[58]).

Apigenin (4′,5,7-trihydroxyflavone) is a well-known flavone widely distributed in nuts, fruits, vegetables, and herbs. Its low intrinsic toxicity (Tang et al., 2017[217]), combined with its potent effects on cancer cell growth (Yan et al., 2017[251]), survival (Rahmani et al., 2022[178]), or apoptosis (Shukla and Gupta, 2008[205]) has drawn significant interest. A study by Fang et al. (2005[48]) demonstrated that apigenin attenuates angiogenesis and tumor growth by enhancing p53 activity through AKT-mediated phosphorylation of its negative regulator HDM2 in ovarian cancer cells (Fang et al., 2005[48]). The study also indicated that apigenin's effect on HDM2 downregulation is mediated by the PI3K/Akt pathway (Fang et al., 2005[48]). Furthermore, apigenin has been shown to stabilize p53 activation and inhibit metastasis (Sherr, 1998[202]; Zheng et al., 2005[273]).

Quercetin (3,5,7,3′,4′-pentahydroxyflavone), one of the most extensively studied flavonols, is found in fruits, tea, wine, vegetables, and other plants (Hossain et al., 2022[80], Aghababaei and Hadidi, 2023[1]). Research indicates that reactive oxygen species (ROS) activate p53 via upstream signal transduction, promoting programmed cell death in abnormal cells (Asgharian et al., 2022[8]). Quercetin has been shown to inhibit tumor cell proliferation by stimulating p53 and NF-κB (Vidya Priyadarsini et al., 2010[230]). An in vitro study on human leukemia cells demonstrated that quercetin enhances p53 phosphorylation and induces apoptosis in a dose-dependent manner (Mertens-Talcott et al., 2005[138]). Similarly, Tanigawa et al. (2008[219]) found that quercetin increases p53 phosphorylation without upregulating its transcription (Tanigawa et al., 2008[219]). In another study, quercetin accelerated apoptosis and growth arrest in wild-type p53-containing A549 human lung cancer cells (Chan et al., 2013[28]). Quercetin also induced apoptosis in glioblastoma cells by upregulating MDM2 mRNA expression, activating caspase-3, and decreasing p53 levels, affecting the regulation of the MDM2-p53 axis (Wang et al., 2014[236]) Molecular dynamics studies revealed that quercetin binds to the MDM2-p53 hydrophobic groove, altering its conformation and disrupting the MDM2-p53 interaction through π-π stacking between MDM2's Tyr 51 and quercetin (Verma et al., 2013[229]). Furthermore, Yang et al. (2016[254]) demonstrated that quercetin reduces cell viability, triggers apoptosis, and induces cell cycle arrest in HT-29 cells by inhibiting the Akt-CSN6-Myc axis, another pathway regulating the MDM2-p53 interaction (Zhou et al., 2001[274]; Zhao et al., 2011[272]; Yang et al., 2016[254]).

Epigallocatechin gallate (3',4',5,5',7-pentahydroxy 3-gallic acid flavane), (EGCG), the main catechin in green tea, has potent antioxidant and anticancer properties (Johnson et al., 2012[95]). EGCG treatment inhibits anchorage-independent growth in human lung cancer cells by stabilizing p53, promoting its nuclear localization, and reducing MDM2 nuclear accumulation. EGCG also enhances p53 phosphorylation at Ser15 and Ser20, thereby increasing its transcriptional activity. This compound likely promotes MDM2 expression in a p53-dependent manner, preventing the ubiquitination of p53 by MDM2 (Jin et al., 2013[94]). A study using NMR, atomistic simulation, AUC, and SAXS analyses identified p53's N-terminal domain (NTD) as the primary binding site for EGCG, which interrupts the p53-MDM2 interaction and stabilizes p53 by inhibiting its ubiquitination and degradation (Zhao et al., 2021[271]).

Oroxylin A (5,7-dihydroxy-6-methoxyflavone), a natural flavone from Oroxylum and Scutellaria species, has been shown to induce apoptosis in HepG2 hepatocellular carcinoma cells by stabilizing p53 at the post-translational level through the downregulation of MDM2 and inhibition of its E3 ligase activity (Mu et al., 2009[145]).

Involucrasin A, a recently discovered natural flavanone from Shuteria involucrata, has demonstrated significant anticancer effects in colon cancer cells (HCT-116) by inhibiting the phosphorylation of Akt and MDM2, which leads to elevated p53 levels (Wei et al., 2023[243]).

Chrysin (5,7-dihydroxyflavone) and wogonin (5,7-dihydroxy 8-methoxyflavone) are similar flavones. TRAIL is a promising antitumor agent that inhibits various tumor cell growth without causing any damage to the peripheral normal tissues (Ding et al., 2012[42]). However, several cancers remain resistant to TRAIL, including TRAIL-resistant human T-cell leukemia virus type 1 (HTLV-1) and Adult T-cell leukemia/lymphoma (ATL) cells. Ding et al. (2012[42]) demonstrated that both chrysin and wogonin inhibit the p53 antagonist MDM2 by increasing p53 levels and upregulating TRAIL-R2. TRAIL-R2 encodes the receptor responsible for the expression of TRAIL protein, a key target gene of p53. This mechanism successfully overcame TRAIL resistance in HTLV-1-associated ATL cells by downregulating the anti-apoptotic FLICE-inhibitory protein (c-FLIP), which is a key inhibitor of the death receptor signaling pathway and blocks caspase 8 activation (Ding et al., 2012[42]).

Tricetin (3′,4′,5′,5,7-pentahydroxyflavone) is a multi-hydroxylated flavone found in certain medicinal plants (Wu et al., 2022[246]). In studies on MCF-7 breast cancer cells, tricetin inhibited cell growth by arresting the cell cycle in the G2/M phase and inducing apoptosis. This was associated with the activation of ATM, which phosphorylates p53 at Ser15, leading to increased p53 stability and reduced MDM2-p53 interaction (Hsu et al., 2009[84]).

Hinokiflavone, a natural bioflavonoid with potent anticancer properties (Patel, 2024[160]), was investigated by Zhang et al. (2022[267]), who concluded that it suppresses MDM2 mRNA synthesis at the transcriptional level. This inhibition results in increased p53 expression, activation of the p53 pathway, and reduced survival of HCT116 colon cancer cells via apoptosis induction and G2/M phase arrest (Zhang et al., 2022[267]).

Curcumin, a dietary polyphenol derived from Curcuma species, is renowned for its biological properties, particularly its anti-inflammatory and anti-angiogenic effects (Quispe et al., 2022[176]; Azzini et al., 2024[10]). To explore how curcumin influences gene expression and carcinogenesis, Li et al. (2007[119]) investigated its impact on various cancer cell lines, including prostate cancer LNCaP (p53 wild type), breast cancer MCF-7 (p53 wild type and p53 knockout), and PC3 (p53 null). Their findings revealed that curcumin inhibits MDM2 expression in a dose-dependent manner, with inhibition occurring at the transcriptional level and affecting MDM2 promoter activity (Li et al., 2007[119]). Further in vitro and in vivo studies showed that curcumin downregulates MDM2 expression in both p53-wild-type and p53-null prostate cancer cells by inhibiting the PI3K/mTOR/ETS2 pathway (Li et al., 2007[119]). Additionally, curcumin induces apoptosis through cell cycle arrest by upregulating the expression of p27, p21, and p16, increasing ER stress, and reducing MDM2 levels (Srivastava et al., 2007[208]; Rivera et al., 2017[187]). A similar study on multiple myeloma RPMI 8226 cells demonstrated that curcumin downregulates MDM2 expression while upregulating p53 and Bax expression (Li et al., 2015[122]). According to Patiño-Morales et al., curcumin stabilizes and extends the active period of p53 by enhancing its interaction with NAD(P)H quinone oxidoreductase 1 (NQO1), ultimately leading to cervical cancer cell death in vitro (Patiño-Morales et al., 2020[161]). Interestingly, curcumin was found to be less effective against breast cancer cell lines compared to cervical cancer cells, likely due to the presence of wild-type p53 in cervical cancer cells, whereas breast cancer cells often contain mutated p53 (Patiño-Morales et al., 2020[161]).

Resveratrol, a natural stilbene monomer, possesses potent antioxidant, anti-inflammatory, neuroprotective, vasculoprotective, and anticancer properties (Giordo et al., 2020[65], 2021[66], 2022[68]; Ramli et al., 2023[179]). The report by She et al. was the first to demonstrate the effect of resveratrol on p53 increase in epidermal JB6 cells, especially in the phosphorylated state (She et al., 2001[200]). In p53-positive Hep G2 cells, resveratrol inhibited cell growth by inducing p53-activated apoptosis. Additionally, resveratrol caused cell cycle arrest in the G1 phase and concurrently upregulated p21 protein expression ((Kuo et al., 2002[107]). Resveratrol was also found to activate the ERK and/or p38 kinase pathways, which promote p53 activation, induce cell cycle arrest, and facilitate DNA repair (Hsieh et al., 2011[83]). Furthermore, resveratrol influenced p53-mediated mitochondrial functions (Delmas et al., 2011[39]).

A study by Ferraz da Costa et al. (2012[51]) reported that resveratrol increased p53 levels in MCF-7 cells without affecting its transcriptional activity. Moreover, transient transfection of wild-type p53 into p53-negative H1299 cells dramatically enhanced susceptibility to apoptosis in resveratrol-treated cells (Ferraz da Costa et al., 2012[51]). Resveratrol also inhibited the viability of CO115, HCT116, and SW480 cells while upregulating p53 and its target genes, including PUMA and Bax (Liu et al., 2019[126]). Bioinformatics analysis further revealed that resveratrol elevated p53 expression in a dose-dependent manner by inhibiting p-Akt and p-MDM2 signaling (Fan et al., 2020[47]). Another study demonstrated that resveratrol preserved AEC₂ cell integrity by activating Sirt1 expression, promoting p53 instability, and stimulating the phosphorylation of both Akt and MDM2 (Navarro et al., 2017[153]). However, resveratrol at concentrations greater than 10 µM was shown to downregulate Sirt1 expression, inhibit cellular plasticity, and induce apoptosis. This effect was accompanied by simultaneous acetylation of p53 in CRC cells, prompting the activation of p53, p21, Bax, and cytochrome C, as well as cleavage of caspase-3 (Brockmueller et al., 2023[21]).

Gossypol, a naturally occurring phytochemical derived from cotton plants (Gossypium species), appears to be a promising anticancer agent (Stein et al., 1992[210]). One study showed that the viability of LAPC4, PC3, and DU145 cancer cells was reduced through the induction of DNA damage and activation of p53 (Volate et al., 2010[233]). Xiong et al. (2017[248]) also reported gossypol's ability to inhibit both VEGF and MDM2 expression in human breast cancer cells, irrespective of whether p53 was mutant or wild-type (Xiong et al., 2017[248]).

Gambogic acid, a naturally prenylated xanthone, was found to suppress tumor growth by inhibiting MDM2 expression while promoting p53 activation (Gu et al., 2008[72]). A related study demonstrated that gambogic acid inhibits Bcl-2 expression in MCF-7 cells by increasing p53 levels, ultimately inducing cell death (Zhai et al., 2008[260]). Further investigation identified a negative correlation between p53 activation and the promotion of p21Waf1/CIP1 expression, which enhances apoptosis in gambogic acid-treated MCF-7 cells via suppression of MDM2 (Rong et al., 2009[189]). A parallel molecular docking study supported these findings, indicating that gambogic acid binds directly to MDM2, functioning as a direct MDM2 inhibitor (Leão et al., 2013[110]).

Terpenoids

With more than 80,000 structures discovered, terpenoids, also known as terpenes or isoprenoids, represent the most prominent family of natural products in all living organisms (Christianson, 2017[35]). They are essential for supporting human health and have been employed as antioxidant, anti-inflammatory, anti-aggregator, anticoagulant, anticancer, antimicrobial, neuroprotective, sedative, anti-allergic, and analgesic agents (Zhao et al., 2016[270]). Numerous studies have highlighted the potent anticancer properties of natural terpenoids, particularly their ability to inhibit MDM2 expression through regulation of p53 levels.

Using a structure-based computational screening method designed to identify molecules that specifically target MDM2, Qin and collaborators identified three natural dimeric sesquiterpene lactones from Inula japonica-namely japonicone A, inulanolide A, and lineariifolianoid A-as potent inhibitors of MDM2 expression in breast cancer cells. Japonicone A was shown to inhibit cell growth, reduce cell proliferation, and induce apoptosis and G₂/M phase cell cycle arrest via an MDM2-dependent mechanism, independent of p53 status. Moreover, no toxicity was observed in breast cancer xenograft models treated with japonicone A, which effectively inhibited tumor growth and lung metastasis (Qin et al., 2015[174]). Inulanolide A underwent both in vitro and in vivo anticancer experiments, demonstrating its dual inhibitory effects on MDM2 and NFAT1 in breast cancer cells. This anticancer activity was selective in both p53-dependent and p53-independent manners, leading to apoptosis induction, reduced cell proliferation, and G₂/M phase arrest. Furthermore, a reduction in MDM2, NFAT1, and cell proliferation-related proteins was observed, alongside an increase in apoptosis-related proteins (Qin et al., 2016[173]). Another study examined the anti-tumorigenic effects of inulanolide A in prostate cancer, showing its ability to inhibit migration, invasion, and proliferation of prostate cancer cells, regardless of androgen receptor (AR) responsiveness and p53 status. This study demonstrated a high affinity for binding to the RING domains of both MDM2 and MDMX proteins (Qin et al., 2017[172]). A similar study on breast cancer revealed that lineariifolianoid A significantly influenced apoptosis, cell cycle progression, proliferation, and colony formation in MCF7 and MDA-MB-231 cells in a dose-dependent and p53-independent manner (Jiang-Jiang et al., 2016[93]). Another sesquiterpene lactone, parthenolide, isolated from Tanacetum parthenium, was reported to induce ATM-dependent MDM2 ubiquitination and proteasomal degradation, leading to p53 activation and the activation of other tumor suppressors that regulate MDM2 (Nasim and Crooks, 2008[152]; Gopal et al., 2009[70]).

Several reports have demonstrated the potent anticancer activity of natural diterpenoids, particularly triptolide, which is extracted from the Chinese plant Tripterygium wilfordii. This diterpene has shown potent antitumor activity against various cancer cells through different mechanisms (Huang et al., 2012[88]; Tamgue and Lei, 2017[216]). One study revealed that ionizing radiation-resistant (IR-resistant) acute lymphoblastic leukemia (ALL) cells are sensitive to triptolide, which reversed IR resistance in ALL cells by inducing an MDM2-overexpressing phenotype. The accumulation and activation of p53-induced by many chemotherapeutic drugs that kill cancer cells through DNA damage and cellular stress-lead to increased p53 activation, which subsequently induces MDM2 expression. Inhibition of p53 and induction of XIAP are key mechanisms involved in the development of IR- or chemo-resistance in wild-type p53/MDM2-overexpressing ALL cells (Huang et al., 2013[87]). To determine whether the effect of triptolide on MDM2 expression is p53-dependent, Xiong and colleagues investigated its impact on paired MDA-MB-468 (p53 mutant) and MCF-7 (wild-type p53) cell lines. Their results showed that triptolide inhibited MDM2 protein expression in a time- and dose-dependent manner, while increasing p53 accumulation without activating its function. Thus, the inhibitory effect of triptolide on MDM2 mRNA and protein expression was independent of p53 status (Xiong et al., 2017[248]). Epoxy clerodane diterpene, isolated from the stems of Tinospora cordifolia, has been found to exhibit remarkable anticancer effects (Dhanasekaran et al., 2009[40]). A study led by Subash-Babu and collaborators on the antitumor activity of epoxy clerodane diterpene against MCF-7 cells demonstrated its ability to upregulate Cdkn2A, pRb1, and p53 proteins, while simultaneously downregulating MDM2. The increase in p53 expression activated the Bax apoptotic pathway, contributing to the suppression of MDM2 expression (Subash-Babu et al., 2017[211]). Another bioactive diterpene, oridonin, derived from the traditional Chinese herb Rabdosia rubescens, exhibits a wide range of biological activities, particularly anticancer, antibacterial, and anti-inflammatory effects (Xu et al., 2018[250]). A study by Zhu et al. (2019[276]) reported that oridonin stimulates p53-mediated cell cycle arrest and apoptosis in neuroblastoma cells by promoting the cleavage of MDM2-p60 (Zhu et al., 2019[276]).

Studies on the ortho-diphenolic diterpene carnosol, found in sage (Salvia officinalis) and rosemary (Rosmarinus officinalis), have demonstrated its potent antioxidant and anticancer effects (O'Neill et al., 2020[154]). In vitro anticancer studies using the U87MG human glioblastoma cell line model revealed that carnosol modulates cellular proliferation by elevating intracellular p53 levels. This was achieved by promoting the transcriptional reactivation of p53, disrupting the p53-MDM2 interaction, and inducing cell cycle arrest and apoptosis (Giacomelli et al., 2016[62]). Continuing their research, Giacomelli and collaborators reported that carnosol decreases CD44 gene expression. This effect is associated with inhibition of the MDM2-p53 complex and the subsequent increase in intracellular p53 levels (Giacomelli et al., 2017[61]).

Ginsenosides, a group of steroid glycosides and triterpene saponins found exclusively in the roots of Panax ginseng, a highly renowned herb in traditional Asian medicine, have demonstrated strong anticancer activity in both in vitro and in vivo studies. Compounds such as 20(R)-dammarane-3β,12β,20,25-tetrol and 20(S)-25-methoxyldammarane-3β,12β,20-triol (also known as 25-OH-PPD and 25-OCH₃-PPD, respectively) have shown potent anti-prostate cancer effects by regulating cell proliferation, apoptosis, cell cycle progression, and tumor growth. 25-OH-PPD also decreased MDM2 levels without affecting p53 expression, decreased cell survival, suppressed proliferation, and triggered apoptosis, leading to G1 cell cycle arrest in both LNCaP and PC3 cells (Wang et al., 2008[239]). Furthermore, 25-OCH₃-PPD was found to decrease the levels of cyclin D1, CDK2, E2F1, and MDM2 while increasing or activating cleaved caspase-3, -8, -9, and cleaved PARP (Wang et al., 2008[238]). Similar results were observed in breast cancer cells, where 25-OCH₃-PPD downregulated MDM2 expression at both transcriptional and posttranslational levels in a time- and dose-dependent manner, irrespective of p53 status (Wang et al., 2012[240]). A study on the antiproliferative and pro-apoptotic effects of 20(S)-ginsenoside Rg3 in MDA-MB-231 cells demonstrated that it reduced mutant p53 levels in both a concentration- and time-dependent fashion. Concurrently, 20(S)-ginsenoside Rg3 increased the association of MDM2 with p53 in these cells (Kim et al., 2014[101]). Another study reported that the survival of NOZ and GBC-SD gallbladder cancer cells was inhibited in a dose-dependent manner by 20(S)-ginsenoside Rg3. This inhibition was achieved through G₁ phase arrest, promoting senescence and apoptosis by inhibiting MDM2 levels, leading to the accumulation of p53 and p21 1 (Zhang et al., 2015[261]).

Ganoderic acids, a group of triterpenes isolated from Ganoderma mushrooms, have been shown to possess various biological activities, including antitumor properties (Kimura et al., 2002[102]). Bin et al. demonstrated that ganoderic acid A has an inhibitory effect on LNCaP prostate cancer cells in a concentration-dependent manner by promoting p53-mediated apoptosis (Bin et al., 2019[15]). A previous virtual screening study of Ganoderma lucidum triterpenoids predicted a strong binding affinity of ganoderic acid A for MDM2 (Froufe et al., 2013[54]). Recent research confirmed these findings, showing that ganoderic acid and its amide derivatives regulate the MDM2-p53 pathway in MCF-7 cells (Jia et al., 2023[92]). Chen et al. demonstrated that ganoderic acid T promotes cell aggregation, suppresses cell migration, and inhibits cell adhesion in HCT-116 human colon cancer cells in a concentration-dependent manner, highlighting the important role of p53 in its anti-invasion effects (Chen and Zhong, 2011[31]). A study on the cytotoxicity and cell cycle arrest capabilities of ganoderic acid against highly metastatic human colon tumor HCT-116 cells, p53-null lung cancer H1299 cells, and lung cancer 95-D cells demonstrated remarkable effects in both a concentration- and time-dependent manner. In 95-D and HCT-116 p53+/+ cells, the cell cycle was arrested at the G₁ phase, while in H1299 and HCT-116 p53−/− cells, ganoderic acid was able to arrest the cell cycle in the S phase or at the G₁/S transition. Based on these findings, Chen and Zhong suggested that ganoderic acid may target p53 (Chen and Zhong, 2009[30]). Aqueous and methanol extracts from Ganoderma lucidum have been shown to inhibit interleukin-3-dependent lymphoma cell (DA-1) proliferation (Calviño et al., 2011[23]). Western blot analysis revealed that the aqueous extracts elevated Bax levels after 13 hours, as well as p53 and Mdm2 levels after 19 hours, with a subsequent reduction in all these proteins at 24 hours. Similarly, the methanol extract increased p53 and Mdm2 levels at 19 hours, followed by a decrease at 24 hours (Calviño et al., 2011[23]).

Cucurbitacins are another class of terpenoids with notable anticancer activity (Attar et al., 2022[9]). Among the more than 10 groups of cucurbitacins, the most commonly isolated and studied are A, B, E, and I (Attar et al., 2022[9]). Zhou et al. demonstrated that cucurbitacin B inhibits the proliferation of benign prostatic hyperplasia epithelial cell line (BPH-1). Molecular analysis showed that cucurbitacin B increased the mRNA levels of MDM2 and thrombospondin 1 (THBS1). Immunocytochemistry results further indicated that cucurbitacin B treatment elevated the protein expressions of p53 and MDM2 (Zhou et al., 2023[275]).

An in vitro and in vivo study on the triterpenoid saponin platycodin D demonstrated its ability to inhibit cell growth in human breast cancer MDA-MB-231 cells by suppressing MDM2 and MDMX, and by reducing mutant p53 expression levels (Kong et al., 2016[104]). Another study found that platycodin D treatment induced apoptosis in MDA-MB-231 cells by upregulating PUMA, a modulator of p53-mediated apoptosis (Chen et al., 2022[33]).

An in silico study reported the binding affinity of fucoxanthin to the p53 gene, CDK2, and tubulin (Indra Januar et al., 2012[90]). Wang et al. (2014[237]) evaluated the anticancer activity of fucoxanthin against the human bladder cancer T24 cell line and revealed its inhibitory effects on both cell growth and colony formation. Additionally, fucoxanthin was able to induce apoptosis and G₀/G₁ phase cell cycle arrest by suppressing the mortalin-p53 complex and reactivating p53 (Wang et al., 2014[237]).

Several in silico screening studies have identified new MDM2-p53 inhibitors, including lithocholic acid, which demonstrated dual inhibitory activity against both MDMX-p53 and MDM2-p53 interactions. Another study showed that lithocholic acid induced apoptosis in wild-type p53 HCT116 cells in vitro (Vogel et al., 2012[232]). Similarly, Muhseen and Li reported the strong binding affinity of 3-trans-p-coumaroyl maslinic acid, betulonic acid, and silvestrol to the active site of MDM2, comparable to the binding affinity exhibited by Nutlin-3a, a known inhibitor of the p53-MDM2 interaction. Results suggest that these compounds occupied the p53 binding regions of MDM2, thereby inhibiting the p53-MDM2 interaction (Muhseen and Li, 2019[146]). Comparable results were found in a recent study, which demonstrated the strong binding affinity of three compounds-justin A, 6-hydroxy justicidin A, and 6′-hydroxy justicidin B-at the active site of MDM2, surpassing the binding affinity of Nutlin-3a (Shoaib et al., 2023[204]).

Alkaloids

Despite the relatively low number of alkaloids identified from plants (approximately 3,000 molecules), many of them are considered potent anticancer agents. Numerous reports have demonstrated the ability of alkaloids to induce self-ubiquitination and degradation of MDM2 by disrupting the MDM2-DAXX-HAUSP interactions (Dhyani et al., 2022[41]). The natural isoquinoline alkaloid berberine has been shown to downregulate the MDM2 oncoprotein in wild-type p53 acute lymphoblastic leukemia (ALL) cell lines, leading to the induction of apoptosis (Zhang et al., 2010[263]). A similar study confirmed that the berberine-induced downregulation of MDM2 expression also reduced XIAP levels, promoting apoptosis in ALL cells independent of p53 status (Liu et al., 2013[124]).

Another alkaloid, matrine, has been reported to inhibit MDM2 expression by reducing MDM2 mRNA synthesis in liver cancer cells. Additionally, matrine sensitizes MDM2-overexpressing liver cancers to etoposide-induced apoptosis, independent of p53 levels. The monoamine alkaloid melatonin has been found to inhibit MDM2 transcription and post-transcriptional expression, reduce MDM2 phosphorylation, and promote p53 acetylation, resulting in p53 activation in MCF-7 cells (Proietti et al., 2014[171]). In human gastric cancer cells, melatonin induced cell cycle arrest and downregulated CDC25A, phospho-CDC25A, and p21. Moreover, melatonin upregulated Bax, downregulated Bcl-xL, activated caspase-3, and increased levels of cleaved caspase-9. Melatonin also increased p53 levels by inhibiting MDM2 phosphorylation at Ser166 and Akt phosphorylation at Thr308 (Song et al., 2018[207]). Another study reported that melatonin upregulated the Nrf2 signaling pathway by activating the MDM2-p53-p21 signaling cascade (Tao et al., 2022[220]).

Overall, significant progress has been made in studying natural products that target the p53-MDM2 interaction for cancer prevention and therapy. However, there remains an urgent need to address key challenges related to in vivo efficacy, bioavailability, potential toxicity, and mechanisms of action in clinical models to develop efficient and safe preventive therapies. A comprehensive summary of the aforementioned natural products and their anticancer activities mediated through the p53-MDM2 interaction is presented in Table 2(Tab. 2) (References in Table 2: Bin et al., 2019[15]; Brockmueller et al., 2023[21]; Chan et al., 2013[28]; Chen and Zhong, 2011[31]; Chen et al., 2022[32][33]; Ding et al., 2012[42]; Fang et al., 2005[48]; Ferraz da Costa et al., 2012[51]; Gao et al., 2020[58]; Giacomelli et al., 2016[62]; Gopal et al., 2009[70]; Gu et al., 2008[72]; Hsieh et al., 2011[83]; Hsu et al., 2009[84]; Huang et al., 2013[87]; Jiang-Jiang et al., 2016[93]; Jin et al., 2013[94]; Kong et al., 2016[104]; Kuo et al., 2002[107]; Li et al., 2005[118], 2007[119], 2015[122]; Liu et al., 2013[124], 2019[126]; Mertens-Talcott et al., 2005[138]; Mu et al., 2009[145]; Nasim and Crooks, 2008[152]; Navarro et al., 2017[153]; Proietti et al., 2014[171]; Qin et al., 2014[174], 2016[173], 2017[172]; Rivera et al., 2017[187]; Rong et al., 2009[189]; She et al., 2001[200]; Sherr, 1998[202]; Song et al., 2018[207]; Srivastava et al., 2007[208]; Subash-Babu et al., 2017[211]; Tanigawa et al., 2008[219]; Tao et al., 2022[220]; Vidya Priyadarsini et al., 2010[230]; Volate et al., 2010[233]; Wang et al., 2008[238], 2008[239], 2012[240], 2014[236]; Wei et al., 2023[243]; Xiong et al., 2017[248]; Yang et al., 2016[254]; Zhai et al., 2008[260]; Zhang et al., 2010[263], 2022[267]; Zheng et al., 2005[273]; Zhu et al., 2019[276]).

Synergistic and Combinatorial Approaches Using Natural Products

Rationale for combining different natural products or natural products with conventional cancer therapies

Natural compounds are an inexhaustible source of potential pharmaceuticals (Bhagani et al., 2020[13]; Hossain et al., 2022[79]; Popović-Djordjević et al., 2022[163]; Posadino et al., 2023[167][168], 2024[169]; Ramli et al., 2023[181][182], 2024[180]). In this context, it is becoming increasingly evident that combining different natural products may produce a synergistic effect, which is greater than the sum of their individual effects. This synergy can enhance their health-related properties, such as antioxidant potential and the ability to inhibit cancer cell growth. The combination of various natural compounds also increases the likelihood of simultaneously targeting multiple signaling pathways, thereby improving the chances of inhibiting cancer progression by affecting several stages, including apoptosis, cell proliferation, angiogenesis, and metastasis.

Synergistic combinations can achieve the desired therapeutic effect at lower doses, thus reducing the risk of side effects and toxicity compared to high doses of a single natural compound, which generally exhibit lower toxicity than synthetic drugs. Furthermore, the interaction between different natural compounds may improve the bioavailability and absorption of each compound, enhancing their overall effectiveness. Combining natural compounds may also reduce the potential for cancer cells to develop resistance, a common issue with single-agent therapies. In addition, natural compounds can be used in conjunction with conventional cancer treatments, such as chemotherapy and radiotherapy, to augment their efficacy.

Evidence supporting the synergistic effects of natural compounds on the p53-MDM2 pathway and cancer cell proliferation

A study by Li et al. (2021[116]) demonstrated that the administration of green tea polyphenols combined with broccoli sprouts inhibited cancer cell growth by inducing apoptosis and cell cycle arrest in HER2/neu transgenic mice. At the molecular level, the authors highlighted that this combination upregulated the expression of phosphatase and tensin homolog (PTEN), p53, and p16, while downregulating the myelocytomatosis oncogene (MYC), polycomb ring finger oncogene Bmi1, and the reverse transcriptase of telomerase, compared to the control group (Li et al., 2021[121]). A similar study used a transgenic mouse model to investigate the effect of combining withaferin A-rich Ashwagandha and sulforaphane-rich broccoli sprouts on breast cancer prevention (Rahman et al., 2024[178]).

The authors' results revealed that this combination contributes to reducing tumor growth by upregulating the apoptosis-associated proteins (BAX and PUMA), the tumor suppressors (P53, P57), and the BAX:BCL-2 ratio (Rahman et al., 2024[178]). Luo et al. (2020[127]) demonstrated that a mixture of epigallocatechin gallate and doxorubicin upregulated p53 and downregulated MDM2 expression, leading to inhibition of proliferation, induction of doxorubicin-mediated apoptosis, and decreased migration of bladder cancer cells (T24 and SW780) (Luo et al., 2020[127]). Another study revealed the synergistic effect of triptolide and Nutlin-3a (an MDM2 inhibitor) in inhibiting cell proliferation and triggering mitochondrial-mediated apoptosis in vitro and ex vivo in wild-type p53 AML xenograft leukemia cells. This combination delayed tumor growth and reduced the leukemia burden by decreasing mRNA levels of XIAP and Mcl-1 in wild-type p53 cells (Chen et al., 2022[32]).

Icaritin is a naturally occurring flavonoid derived from the Epimedium plant, commonly known as Horny Goat Weed. This plant is rich in several classes of flavonoids, each with specific biological functions (Zhuang et al., 2023[277]). Li and colleagues evaluated the role of icaritin in promoting controlled cell death and inhibiting the proliferation of hepatic cells via the P53/MDM2 and AFP pathways. They discovered that the introduction of icaritin enhanced p53 activity by extending the duration of its response, thereby repressing the expression of AFP genes. Additionally, icaritin stabilized p53, preventing the expression of MDM2 (Li et al., 2021[116]).

An in vitro anticancer analysis using the human glioblastoma cell line model (U87MG) demonstrated that carnosol, a phenolic diterpene found in rosemary, regulated cellular proliferation by increasing intracellular p53 levels, promoting its transcriptional reactivation, degrading the p53-MDM2 interaction, and inducing apoptosis and cell cycle arrest. In the same study, combining carnosol with temozolomide resulted in a synergistic effect, reducing the recurrence of tumor cell proliferation even after the drug was withdrawn (Giacomelli et al., 2016[62]). Table 3(Tab. 3) (References in Table 3: Chen et al., 2022[32]; Giacomelli et al., 2016[62]; Li et al., 2021[121]; Luo et al., 2020[127]; Rahman et al., 2024[177]) summarizes studies demonstrating the synergistic effects of natural products on the p53-MDM2 interaction in various experimental cancer models.

Strategies for optimizing combination therapies regarding dosage, timing and delivery

While generally considered safe in terms of toxicity, natural compounds can become harmful to cells and the body depending on their concentration and environmental conditions (Pasciu et al., 2010[159]; Giordo et al., 2013[64]; Posadino et al., 2013[164], 2015[165], 2019[166]; Shaito et al., 2020[196]). In this regard, determining the optimal concentrations of these compounds is an important step. This can be achieved by conducting dose-response experiments to identify the concentrations at which each compound is most effective in vitro, as well as preclinical studies to establish the effective dose range for each compound. Identifying combinations that exhibit synergistic effects, which allow for lower doses of each compound while maintaining or enhancing efficacy, is essential. The potential toxicity of mixtures should also be checked by conducting in vitro and in vivo toxicity studies to ensure that the combined compounds do not exceed toxicity thresholds. Dosages should be adjusted to minimize adverse effects while maintaining therapeutic benefits. Further optimization could involve exploring the timing of compound administration, which may include: i) Sequential Administration: Analyzing the effects of compounds administered in a specific sequence. ii) Simultaneous Administration: Analyzing the effects of compounds when administered simultaneously to target multiple pathways at once. iii) Chronotherapy: Analyzing the effects of compounds when administered in alignment with the body's biological rhythms (circadian rhythms) to enhance efficacy and reduce side effects. Additional optimization may include improving compound delivery. This can involve using nanoparticles, liposomes, or other delivery systems to target compounds specifically to cancer cells, thereby enhancing efficacy and reducing systemic toxicity (Sanna et al., 2011[193]; Quispe et al., 2021[175]; Giordo et al., 2022[67]). Biodegradable controlled-release formulations can also be developed to provide controlled release of compounds over time, maintaining therapeutic levels and reducing the frequency of administration. In this context, the use of combined delivery vehicles may be explored to carry multiple compounds, ensuring they.

Challenges and Future Directions

Targeting the P53-MDM2 pathway in cancer using natural compounds poses several challenges. The intricate nature of the P53-MDM2 interaction, which encompasses several complex regulatory mechanisms and feedback loops, necessitates a thorough understanding of these interactions to develop effective therapies. Detailed mechanistic studies are necessary to uncover how natural compounds affect the P53-MDM2 pathway and related cellular processes. Variability in the biological activity and therapeutic efficacy of natural products due to differences in plant sources, cultivation conditions, and extraction methods poses a significant challenge, resulting in reproducibility issues (Maaliki et al., 2019[128]; Shaito et al., 2020[196][197]; Alsamri et al., 2021[3]). Moreover, their poor bioavailability and stability hinder many natural compounds' clinical efficacy. Addressing these issues necessitates the creation of advanced delivery systems, including nanoparticles and liposomes, and other nanocarriers, to enhance the delivery and absorption of these compounds (Shaito et al., 2020[196][197]; Posadino et al., 2024[169]).

Although natural products are perceived as safer alternatives to synthetic drugs, they can still cause toxicity and adverse effects at therapeutic doses (Pasciu et al., 2010[159]; Giordo et al., 2013[64]; Posadino et al., 2013[164], 2015[165], 2019[166]; Shaito et al., 2020[197]). Consequently, ensuring safety requires comprehensive toxicity assessments. In addition, cancer cells can develop resistance to therapies targeting the P53-MDM2 pathway, necessitating an in-depth understanding of resistance mechanisms to develop effective counterstrategies. The absence of standardized protocols for the extraction, preparation, and clinical evaluation of natural products further complicates their development and approval as anticancer therapies. Advanced screening methods are essential to overcome many of these challenges. High-throughput screening and computational modeling can aid in identifying and optimizing natural compounds that effectively target the P53-MDM2 pathway. These methods can also facilitate the exploration of synergistic effects when natural compounds are combined with each other or with conventional cancer therapies, potentially enhancing efficacy and reducing side effects. Preclinical and clinical trials are vital to assess the safety, efficacy, and optimal dosage of natural compounds targeting the P53-MDM2 pathway, facilitating the translation of promising compounds from bench to bedside. Personalized medicine approaches, which tailor natural compound-based therapies to individual patients' genetic and molecular profiles, hold promise for improving treatment outcomes. Finally, efforts towards international regulatory harmonization are necessary to streamline the approval process for natural compound-based therapies, thereby facilitating their development and accessibility to patients.

Conclusion

The intricate interplay between the p53 tumor suppressor and MDM2 ubiquitin ligase is vital for regulating cell cycle, apoptosis, and genomic stability. Disruption of this interaction is a hallmark of various cancers, contributing to poor prognosis and resistance to conventional therapies. An in-depth understanding of this interaction's molecular mechanisms may yield valuable insights into potential therapeutic targets. Natural products have emerged as promising candidates for targeting the p53-MDM2 pathway, offering various compounds capable of modulating this important interaction. Polyphenols, terpenoids, and alkaloids have shown significant potential in inhibiting MDM2 expression, preventing the p53-MDM2 binding, and stabilizing p53, thereby restoring its tumor suppressor functions. These compounds not only exhibit anticancer properties but also lay the groundwork for the development of novel chemotherapeutic agents with reduced toxicity. Despite the promising results, translating these findings into clinical applications remains challenging. These natural compounds' bioavailability, efficacy, and safety need to be addressed through rigorous preclinical and clinical studies. Furthermore, a deeper understanding of the complex regulatory networks involving p53 and MDM2 will aid in the creation of more effective therapeutic strategies. In conclusion, the p53-MDM2 axis represents an important target in cancer therapy, and natural products offer a valuable reservoir of bioactive compounds for therapeutic intervention. Continued research in this area holds great promise for developing innovative treatments that can improve cancer prognosis and patient outcomes.

Notes

Daniela Calina, Javad Sharifi-Rad (Universidad Espíritu Santo, Samborondón 092301, Ecuador; E-mail: javad.sharifirad@gmail.com) and Gianfranco Pintus (Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43B, 07100 Sassari, Italy; E-mail: gpintus@uniss.it) contributed equally as corresponding author.

Declaration

Competing interests

The authors wish to confirm that there are no known conflicts of interest associated with this publication and that there has been no significant financial support for this work that could have influenced its outcome.

Funding

This work has been developed within the framework of the project eINS-Ecosystem of Innovation for Next Generation Sardinia (cod. ECS 00000038), funded by the Italian Ministry for Research and Education (MUR) under the National Recovery and Resilience Plan (PNRR). This work was also made possible thanks to “Progetto Fondazione di Sardegna -Bando 2022-2023” and “DM 737/2021 resources 2021-2022, funded by the European Union-NextGenerationEU”.

Acknowledgments

The authors would like to express their gratitude to: Dr. Irina Zamfir, MD, MRCP London, Basildon University Hospital UK for providing professional English editing of this manuscript and for editorial support.

 

References

1. Aghababaei F, Hadidi M. Recent advances in potential health benefits of quercetin. Pharmaceuticals. 2023;16 (7):1020
2. Aloni-Grinstein R, Charni-Natan M, Solomon H, Rotter V. p53 and the viral connection: back into the future. Cancers (Basel). 2018;10:178
3. Alsamri H, Athamneh K, Pintus G, Eid AH, Iratni R. Pharmacological and antioxidant activities of Rhus coriaria L.(Sumac). Antioxidants. 2021;10(1):73
4. Alt JR, Bouska A, Fernandez MR, Cerny RL, Xiao H, Eischen CM. Mdm2 binds to Nbs1 at sites of DNA damage and regulates double strand break repair. J Biol Chem. 2005;280:18771-81. doi: 10.1074/jbc.M413387200
5. Amendolare A, Marzano F, Petruzzella V, Vacca RA, Guerrini L, Pesole G, et al. The underestimated role of the p53 pathway in renal cancer. Cancers (Basel). 2022;14(23):5733. doi: 10.3390/cancers14235733
6. Arena G, Cissé MY, Pyrdziak S, Chatre L, Riscal R, Fuentes M, et al. Mitochondrial MDM2 regulates respiratory complex I activity independently of p53. Mol Cell. 2018;69:594-609
7. Asahara H, Li Y, Fuss J, Haines DS, Vlatkovic N, Boyd MT, et al. Stimulation of human DNA polymerase ϵ by MDM2. Nucleic Acids Res. 2003;31:2451-9. doi: 10.1093/nar/gkg342
8. Asgharian P, Tazekand AP, Hosseini K, Forouhandeh H, Ghasemnejad T, Ranjbar M, et al. Potential mechanisms of quercetin in cancer prevention: focus on cellular and molecular targets. Cancer Cell Int. 2022;22(1):257. doi: 10.1186/s12935-022-02677-w
9. Attar UA, Ghane SG, Chavan NS, Shiragave PD. Simultaneous detection of anticancer compounds (Cucurbitacin I, B and E) and some pharmacological properties of Indian Blastania species. S Afr J Bot. 2022;147:871-81
10. Azzini E, Peña-Corona SI, Hernández-Parra H, Chandran D, Saleena LAK, Sawikr Y, et al. Neuroprotective and anti-inflammatory effects of curcumin in Alzheimer's disease: Targeting neuroinflammation strategies. Phytother Res. 2024;38:3169-89. doi: 10.1002/ptr.8200
11. Bahena Culhuac E, Bello M. Unveiling the mechanisms of EGCG–p53 interactions through molecular dynamics simulations. ACS Omega. 2024;9:20066-85. doi: 10.1021/acsomega.3c10523
12. Barak Y, Gottlieb E, Juven-Gershon T, Oren M. Regulation of mdm2 expression by p53: alternative promoters produce transcripts with nonidentical translation potential. Genes Dev. 1994;8:1739-49
13. Bhagani H, Nasser SA, Dakroub A, El-Yazbi AF, Eid AA, Kobeissy F, et al. The mitochondria: A target of polyphenols in the treatment of diabetic cardiomyopathy. Int J Mol Sci. 2020;21(14):4962
14. Bhat SS, Prasad SK, Shivamallu C, Prasad KS, Syed A, Reddy P, et al. Genistein: a potent anti-breast cancer agent. Curr Issues Mol Biol. 2021;43:1502-17. doi: 10.3390/cimb43030106
15. Bin XU, Wei JIA, Zhong W, He-Nan Z, Di WU, Chuan-Hong T, et al. The effect and mechanism of ganoderic acid A (GA-A) inhibiting the growth of prostate cancer LNCaP cells. Mycosystema. 2019;38:717-27. doi: 10.13346/j.mycosystema.190004
16. Bond GL, Hu W, Bond EE, Robins H, Lutzker SG, Arva NC, et al. A single nucleotide polymorphism in the MDM2 promoter attenuates the p53 tumor suppressor pathway and accelerates tumor formation in humans. Cell. 2004;119:591-602
17. Bouska A, Lushnikova T, Plaza S, Eischen CM. Mdm2 promotes genetic instability and transformation independent of p53. Mol Cell Biol. 2008;28:4862-74. doi: 10.1128/MCB.01584-07
18. Boutelle AM, Attardi LD. p53 and tumor suppression: it takes a network. Trends Cell Biol. 2021;31:298-310
19. Boyd MT, Vlatkovic N, Haines DS. A novel cellular protein (MTBP) binds to MDM2 and induces a G1 arrest that is suppressed by MDM2. J Biol Chem. 2000;275:31883-90. doi: 10.1074/jbc.M004252200
20. Brady M, Vlatković N, Boyd MT. Regulation of p53 and MDM2 activity by MTBP. Mol Cell Biol. 2005;25:545-53. doi: 10.1128/MCB.25.2.545-553.2005
21. Brockmueller A, Buhrmann C, Shayan P, Shakibaei M. Resveratrol induces apoptosis by modulating the reciprocal crosstalk between p53 and Sirt-1 in the CRC tumor microenvironment. Front Immunol. 2023;14:1225530. doi: 10.3389/fimmu.2023.1225530
22. Cahilly-Snyder L, Yang-Feng T, Francke U, George DL. Molecular analysis and chromosomal mapping of amplified genes isolated from a transformed mouse 3T3 cell line. Somat Cell Mol Genet. 1987;13:235-44. doi: 10.1007/BF01535205
23. Calviño E, Pajuelo L, de Eribe Casas JAO, Manjón JL, Tejedor MC, Herráez A, et al. Cytotoxic action of Ganoderma lucidum on interleukin‐3 dependent lymphoma DA‐1 cells: involvement of apoptosis proteins. Phytother Res. 2011;25(1):25-32
24. Cao H, Chen X, Wang Z, Wang L, Xia Q, Zhang W. The role of MDM2–p53 axis dysfunction in the hepatocellular carcinoma transformation. Cell Death Discov. 2020;6(1):53. doi: 10.1038/s41420-020-0287-y
25. Cardozo CM, Hainaut P. Viral strategies for circumventing p53: The case of severe acute respiratory syndrome coronavirus. Curr Opin Oncol. 2021;33:149-58
26. Carroll PE, Okuda M, Horn HF, Biddinger P, Stambrook PJ, Gleich LL, et al. Centrosome hyperamplification in human cancer: chromosome instability induced by p53 mutation and/or Mdm2 overexpression. Oncogene. 1999;18:1935-44. doi: 10.1038/sj.onc.1202515
27. Chaachouay N, Zidane L. Plant-derived natural products: a source for drug discovery and development. Drugs Drug Candidates. 2024;3(1):184-207. doi: 10.3390/ddc3010011
28. Chan S-T, Yang N-C, Huang C-S, Liao J-W, Yeh S-L. Quercetin enhances the antitumor activity of trichostatin a through upregulation of p53 protein expression in vitro and in vivo. PLoS One. 2013;8(1):e54255. doi: 10.1371/journal.pone.0054255
29. Chaudhry GE, Zeenia, Sharifi-Rad J, Calina D. Hispidulin: a promising anticancer agent and mechanistic breakthrough for targeted cancer therapy. Naunyn Schmiedebergs Arch Pharmacol. 2024;397:1919-34. doi: 10.1007/s00210-023-02645-9
30. Chen N-H, Zhong J-J. Ganoderic acid Me induces G1 arrest in wild-type p53 human tumor cells while G1/S transition arrest in p53-null cells. Process Biochem. 2009;44:928-33. doi: 10.1016/j.procbio.2009.03.018
31. Chen N-H, Zhong J-J. p53 is important for the anti-invasion of ganoderic acid T in human carcinoma cells. Phytomedicine. 2011;18:719-25. doi: 10.1016/j.phymed.2011.01.011
32. Chen Q, Deng S, Deng M, Shi Y, Zhong M, Ding L, et al. Therapeutic synergy of triptolide and MDM2 inhibitor against acute myeloid leukemia through modulation of p53-dependent and -independent pathways. Exp Hematol Oncol. 2022;11(1):23. doi: 10.1186/s40164-022-00276-z
33. Chen S, Wang Q, Ming S, Zheng H, Hua B, Yang H-S. Platycodin D induces apoptosis through JNK1/AP-1/PUMA pathway in non-small cell lung cancer cells: A new mechanism for an old compound. Front Pharmacol. 2022;13:1045375. doi: 10.3389/fphar.2022.1045375
34. Chi S-W, Lee S-H, Kim D-H, Ahn M-J, Kim J-S, Woo J-Y, et al. Structural details on mdm2-p53 interaction. J Biol Chem. 2005;280:38795-802
35. Christianson DW. Structural and chemical biology of terpenoid cyclases. Chem Rev. 2017;117:11570-648. doi: 10.1021/acs.chemrev.7b00287
36. Chunarkar-Patil P, Kaleem M, Mishra R, Ray S, Ahmad A, Verma D, et al. Anticancer drug discovery based on natural products: from computational approaches to clinical studies. Biomedicines. 2024;12(1):201. doi: 10.3390/biomedicines12010201
37. Dar AA, Majid S, Rittsteuer C, de Semir D, Bezrookove V, Tong S, et al. The role of miR-18b in MDM2-p53 pathway signaling and melanoma progression. J Natl Cancer Inst. 2013;105:433-42. doi: 10.1093/jnci/djt003
38. de Toledo SM, Azzam EI, Dahlberg WK, Gooding TB, Little JB. ATM complexes with HDM2 and promotes its rapid phosphorylation in a p53-independent manner in normal and tumor human cells exposed to ionizing radiation. Oncogene. 2000;19:6185-93. doi: 10.1038/sj.onc.1204020
39. Delmas D, Solary E, Latruffe N. Resveratrol, a phytochemical inducer of multiple cell death pathways: apoptosis, autophagy and mitotic catastrophe. Curr Med Chem. 2011;18:1100-21. doi: 10.2174/092986711795029708
40. Dhanasekaran M, Baskar A-A, Ignacimuthu S, Agastian P, Duraipandiyan V. Chemopreventive potential of Epoxy clerodane diterpene from Tinospora cordifolia against diethylnitrosamine-induced hepatocellular carcinoma. Invest New Drugs. 2009;27:347-55. doi: 10.1007/s10637-008-9181-9
41. Dhyani P, Quispe C, Sharma E, Bahukhandi A, Sati P, Attri DC, et al. Anticancer potential of alkaloids: a key emphasis to colchicine, vinblastine, vincristine, vindesine, vinorelbine and vincamine. Cancer Cell Int. 2022;22(1):206. doi: 10.1186/s12935-022-02624-9
42. Ding J, Polier G, Köhler R, Giaisi M, Krammer PH, Li-Weber M. Wogonin and related natural flavones overcome tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) protein resistance of tumors by down-regulation of c-FLIP protein and up-regulation of TRAIL receptor 2 expression. J Biol Chem. 2012;287 (1):641-9. doi: 10.1074/jbc.M111.286526
43. Dubs-Poterszman MC, Tocque B, Wasylyk B. MDM2 transformation in the absence of p53 and abrogation of the p107 G1 cell-cycle arrest. Oncogene. 1995;11(11):2445-9
44. Eischen CM, Weber JD, Roussel MF, Sherr CJ, Cleveland JL. Disruption of the ARF–Mdm2–p53 tumor suppressor pathway in Myc-induced lymphomagenesis. Genes Dev. 1999;13:2658-69
45. Elkholi R, Abraham-Enachescu I, Trotta AP, Rubio-Patiño C, Mohammed JN, Luna-Vargas MPA, et al. MDM2 integrates cellular respiration and apoptotic signaling through NDUFS1 and the mitochondrial network. Mol Cell. 2019;74:452-65
46. Fakharzadeh SS, Trusko SP, George DL. Tumorigenic potential associated with enhanced expression of a gene that is amplified in a mouse tumor cell line. EMBO J. 1991;10:1565-9
47. Fan Y, Li J, Yang Y, Zhao X, Liu Y, Jiang Y, et al. Resveratrol modulates the apoptosis and autophagic death of human lung adenocarcinoma A549 cells via a p53‑dependent pathway: Integrated bioinformatics analysis and experimental validation. Int J Oncol. 2020;57:925-38. doi: 10.3892/ijo.2020.5107
48. Fang J, Xia C, Cao Z, Zheng JZ, Reed E, Jiang B-H. Apigenin inhibits VEGF and HIF-1 expression via PI3K/AKT/p70S6K1 and HDM2/p53 pathways. FASEB J. 2005;19:342-53. doi: 10.1096/fj.04-2175com
49. Fang S, Jensen JP, Ludwig RL, Vousden KH, Weissman AM. Mdm2 is a RING finger-dependent ubiquitin protein ligase for itself and p53. J Biol Chem. 2000;275:8945-51. doi: 10.1074/jbc.275.12.8945
50. Farkas M, Hashimoto H, Bi Y, Davuluri RV, Resnick-Silverman L, Manfredi JJ, et al. Distinct mechanisms control genome recognition by p53 at its target genes linked to different cell fates. Nat Commun. 2021;12(1):484
51. Ferraz da Costa DC, Casanova FA, Quarti J, Malheiros MS, Sanches D, dos Santos PS, et al. Transient transfection of a wild-type p53 gene triggers resveratrol-induced apoptosis in cancer cells. PLoS One. 2012;7 (11):e48746. doi: 10.1371/journal.pone.0048746
52. Fischer M. Census and evaluation of p53 target genes. Oncogene. 2017;36:3943-56
53. Foggetti G, Ottaggio L, Russo D, Monti P, Degan P, Fronza G, et al. Gambogic acid counteracts mutant p53 stability by inducing autophagy. Biochim Biophys Acta Mol Cell Res. 2017;1864:382-92. doi: 10.1016/j.bbamcr.2016.11.023
54. Froufe HJC, Abreu RMV, Ferreira ICFR. Virtual screening of low molecular weight mushrooms compounds as potential Mdm2 inhibitors. J Enzyme Inhib Med Chem. 2013;28:569-75. doi: 10.3109/14756366.2012.658787
55. Fu J, Imani S, Wu MY, Wu RC. MicroRNA-34 family in cancers: role, mechanism, and therapeutic potential. Cancers (Basel). 2023;15(19):4723. doi: 10.3390/cancers15194723
56. Fu W, Ma Q, Chen L, Li P, Zhang M, Ramamoorthy S, et al. MDM2 acts downstream of p53 as an E3 ligase to promote FOXO ubiquitination and degradation. J Biol Chem. 2009;284:13987-4000. doi: 10.1074/jbc.M901758200
57. Gama V, Gomez JA, Mayo LD, Jackson MW, Danielpour D, Song K, et al. Hdm2 is a ubiquitin ligase of Ku70-Akt promotes cell survival by inhibiting Hdm2-dependent Ku70 destabilization. Cell Death Differ. 2009;16:758-69. doi: 10.1038/cdd.2009.6
58. Gao J, Xia R, Chen J, Gao J, Luo X, Ke C, et al. Inhibition of esophageal-carcinoma cell proliferation by genistein via suppression of JAK1/2-STAT3 and AKT/MDM2/p53 signaling pathways. Aging (Albany NY). 2020;12:6240-59. doi: 10.18632/aging.103019
59. García-Cano J, Sánchez-Tena S, Sala-Gaston J, Figueras A, Viñals F, Bartrons R, et al. Regulation of the MDM2-p53 pathway by the ubiquitin ligase HERC2. Mol Oncol. 2020;14(1):69-86. doi: 10.1002/1878-0261.12592
60. Gasco M, Shami S, Crook T. The p53 pathway in breast cancer. Breast Cancer Res. 2002;4(2):70-6. doi: 10.1186/bcr426
61. Giacomelli C, Daniele S, Natali L, Iofrida C, Flamini G, Braca A, et al. Carnosol controls the human glioblastoma stemness features through the epithelial-mesenchymal transition modulation and the induction of cancer stem cell apoptosis. Sci Rep. 2017;7(1):15174. doi: 10.1038/s41598-017-15360-2
62. Giacomelli C, Natali L, Trincavelli ML, Daniele S, Bertoli A, Flamini G, et al. New insights into the anticancer activity of carnosol: p53 reactivation in the U87MG human glioblastoma cell line. Int J Biochem Cell Biol. 2016;74:95-108. doi: 10.1016/j.biocel.2016.02.019
63. Ginsberg D, Mechta F, Yaniv M, Oren M. Wild-type p53 can down-modulate the activity of various promoters. Proc Natl Acad Sci U S A. 1991;88:9979-83. doi: 10.1073/pnas.88.22.9979
64. Giordo R, Cossu A, Pasciu V, Hoa PT, Posadino AM, Pintus G. Different redox response elicited by naturally occurring antioxidants in human endothelial cells. Open Biochem J. 2013;7:44
65. Giordo R, Nasrallah GK, Al-Jamal O, Paliogiannis P, Pintus G. Resveratrol inhibits oxidative stress and prevents mitochondrial damage induced by zinc oxide nanoparticles in zebrafish (Danio rerio). Int J Mol Sci. 2020;21(11):3838
66. Giordo R, Nasrallah GK, Posadino AM, Galimi F, Capobianco G, Eid AH, et al. Resveratrol-elicited pkc inhibition counteracts nox-mediated endothelial to mesenchymal transition in human retinal endothelial cells exposed to high glucose. Antioxidants. 2021;10(2):224
67. Giordo R, Wehbe Z, Paliogiannis P, Eid AH, Mangoni AA, Pintus G. Nano-targeting vascular remodeling in cancer: Recent developments and future directions. Semin Cancer Biol. 2022;86:784-804. doi: 10.1016/j.semcancer.2022.03.001
68. Giordo R, Wehbe Z, Posadino AM, Erre GL, Eid AH, Mangoni AA, et al. Disease-associated regulation of non-coding RNAs by resveratrol: molecular insights and therapeutic applications. Front Cell Dev Biol. 2022;10:894305
69. Giordo R, Zinellu A, Eid AH, Pintus G. Therapeutic potential of resveratrol in COVID-19-associated hemostatic disorders. Molecules. 2021;26(4):856
70. Gopal YNV, Chanchorn E, Van Dyke MW. Parthenolide promotes the ubiquitination of MDM2 and activates p53 cellular functions. Mol Cancer Ther. 2009;8:552-62. doi: 10.1158/1535-7163.MCT-08-0661
71. Grossman SR, Perez M, Kung AL, Joseph M, Mansur C, Xiao Z-X, et al. p300/MDM2 complexes participate in MDM2-mediated p53 degradation. Mol Cell. 1998;2:405-15
72. Gu H, Wang X, Rao S, Wang J, Zhao J, Ren FL, et al. Gambogic acid mediates apoptosis as a p53 inducer through down-regulation of mdm2 in wild-type p53-expressing cancer cells. Mol Cancer Ther. 2008;7:3298-305. doi: 10.1158/1535-7163.MCT-08-0212
73. Gu L, Zhu N, Zhang H, Durden DL, Feng Y, Zhou M. Regulation of XIAP translation and induction by MDM2 following irradiation. Cancer Cell. 2009;15:363-75. doi: 10.1016/j.ccr.2009.03.002
74. Guillen-Sacoto MJ, Martinez AF, Abe Y, Kruszka P, Weiss K, Everson JL, et al. Human germline hedgehog pathway mutations predispose to fatty liver. J Hepatol. 2017;67:809-17
75. Hafner A, Stewart-Ornstein J, Purvis JE, Forrester WC, Bulyk ML, Lahav G. p53 pulses lead to distinct patterns of gene expression albeit similar DNA-binding dynamics. Nat Struct Mol Biol. 2017;24:840-7
76. Haupt Y, Maya R, Kazaz A, Oren M. Mdm2 promotes the rapid degradation of p53. Nature. 1997;387(6630):296-9. doi: 10.1038/387296a0
77. Herrero AB, Rojas EA, Misiewicz-Krzeminska I, Krzeminski P, Gutiérrez NC. Molecular mechanisms of p53 deregulation in cancer: an overview in multiple myeloma. Int J Mol Sci. 2016;17(12):2003. doi: 10.3390/ijms17122003
78. Honda R, Tanaka H, Yasuda H. Oncoprotein MDM2 is a ubiquitin ligase E3 for tumor suppressor p53. FEBS Lett. 1997;420(1):25-7. doi: 10.1016/S0014-5793(97)01480-4
79. Hossain R, Quispe C, Khan RA, Saikat ASM, Ray P, Ongalbek D, et al. Propolis: An update on its chemistry and pharmacological applications. Chin Med. 2022;17(1):100
80. Hossain R, Sarkar C, Hassan SMH, Khan RA, Arman M, Ray P, et al. In silico screening of natural products as potential inhibitors of SARS-CoV-2 using molecular docking simulation. Chin J Integr Med. 2022;28:249-56. doi: 10.1007/s11655-021-3504-5
81. Hou H, Sun D, Zhang X. The role of MDM2 amplification and overexpression in therapeutic resistance of malignant tumors. Cancer Cell Int. 2019;19(1):216
82. Hsieh J-K, Chan FSG, O’Connor DJ, Mittnacht S, Zhong S, Lu X. RB regulates the stability and the apoptotic function of p53 via MDM2. Mol Cell. 1999;3:181-93. doi: 10.1016/S1097-2765(00)80309-3
83. Hsieh T-C, Wong C, John Bennett D, Wu JM. Regulation of p53 and cell proliferation by resveratrol and its derivatives in breast cancer cells: An in silico and biochemical approach targeting integrin αvβ3. Int J Cancer. 2011;129:2732-43. doi: 10.1002/ijc.25930
84. Hsu Y-L, Uen Y-H, Chen Y, Liang H-L, Kuo P-L. Tricetin, a dietary flavonoid, inhibits proliferation of human breast adenocarcinoma MCF-7 cells by blocking cell cycle progression and inducing apoptosis. J Agric Food Chem. 2009;57:8688-95. doi: 10.1021/jf901053x
85. Hsueh K-W, Fu S-L, Chang C-B, Chang Y-L, Lin C-H. A novel Aurora-A-mediated phosphorylation of p53 inhibits its interaction with MDM2. Biochim Biophys Acta Prot Proteom. 2013;1834:508-15
86. Hu W, Feng Z, Levine AJ. The regulation of multiple p53 stress responses is mediated through MDM2. Genes Cancer. 2012;3:199-208
87. Huang M, Zhang H, Liu T, Tian D, Gu L, Zhou M. Triptolide inhibits MDM2 and induces apoptosis in acute lymphoblastic leukemia cells through a p53-independent pathway. Mol Cancer Ther. 2013;12:184-94. doi: 10.1158/1535-7163.MCT-12-0425
88. Huang W, He T, Chai C, Yang Y, Zheng Y, Zhou P, et al. Triptolide inhibits the proliferation of prostate cancer cells and down-regulates SUMO-specific protease 1 expression. PLoS One. 2012;7(5):e37693. doi: 10.1371/journal.pone.0037693
89. Ijaz S, Iqbal J, Abbasi BA, Tufail A, Yaseen T, Uddin S, et al. Current stage of preclinical and clinical development of guggulsterone in cancers: Challenges and promises. Cell Biol Int. 2024;48:128-42. doi: 10.1002/cbin.12112
90. Indra Januar H, Dewi A, Marraskuranto E, Wikanta T. In silico study of fucoxanthin as a tumor cytotoxic agent. J Pharm Bioallied Sci. 2012;4:56-9. doi: 10.4103/0975-7406.92733
91. Iqbal MJ, Kabeer A, Abbas Z, Siddiqui HA, Calina D, Sharifi-Rad J, et al. Interplay of oxidative stress, cellular communication and signaling pathways in cancer. Cell Commun Signal. 2024;22(1):7. doi: 10.1186/s12964-023-01398-5
92. Jia Y, Li Y, Shang H, Luo Y, Tian Y. Ganoderic acid A and its amide derivatives as potential anti-cancer agents by regulating the p53-MDM2 pathway: synthesis and biological evaluation. Molecules. 2023;28(5):2374. doi: 10.3390/molecules28052374
93. Jiang-Jiang Q, Sushanta S, Sukesh V, Rajesh A, Wei W, Ruiwen Z. Identification of lineariifolianoid A as a novel dual NFAT1 and MDM2 inhibitor for human cancer therapy. J Biomed Res. 2016;30:322-33. doi: 10.7555/JBR.30.20160018
94. Jin L, Li C, Xu Y, Wang L, Liu J, Wang D, et al. Epigallocatechin gallate promotes p53 accumulation and activity via the inhibition of MDM2-mediated p53 ubiquitination in human lung cancer cells. Oncol Rep. 2013;29:1983-90. doi: 10.3892/or.2013.2343
95. Johnson R, Bryant S, Huntley AL. Green tea and green tea catechin extracts: An overview of the clinical evidence. Maturitas. 2012;73:280-7. doi: 10.1016/j.maturitas.2012.08.008
96. K AR, Arumugam S, Muninathan N, Baskar K, S D, D DR. P53 gene as a promising biomarker and potential target for the early diagnosis of reproductive cancers. Cureus. 2024;16(5):e60125. doi: 10.7759/cureus.60125
97. Kadosh E, Snir-Alkalay I, Venkatachalam A, May S, Lasry A, Elyada E, et al. The gut microbiome switches mutant p53 from tumour-suppressive to oncogenic. Nature. 2020;586(7827):133-8
98. Kamat CD, Green DE, Warnke L, Thorpe JE, Ceriello A, Ihnat MA. Mutant p53 facilitates pro-angiogenic, hyperproliferative phenotype in response to chronic relative hypoxia. Cancer Lett. 2007;249:209-19
99. Kamath AJ, Chandy AS, Joseph AA, Gorantla JN, Donadkar AD, Nath LR, et al. Embelin: A multifaceted anticancer agent with translational potential in targeting tumor progression and metastasis. EXCLI J. 2023;22:1311-29. doi: 10.17179/excli2023-6590
100. Katsube N, Iwashita K, Tsushida T, Yamaki K, Kobori M. Induction of apoptosis in cancer cells by Bilberry (Vaccinium myrtillus) and the anthocyanins. J Agric Food Chem. 2003;51(1):68-75. doi: 10.1021/jf025781x
101. Kim BM, Kim DH, Park JH, Surh YJ, Na HK. Ginsenoside Rg3 inhibits constitutive activation of NF-κB signaling in human breast cancer (MDA-MB-231) cells: ERK and Akt as potential upstream targets. J Cancer Prev. 2014;19(1):23-30. doi: 10.15430/jcp.2014.19.1.23
102. Kimura Y, Taniguchi M, Baba K. Antitumor and antimetastatic effects on liver of triterpenoid fractions of Ganoderma lucidum: mechanism of action and isolation of an active substance. Anticancer Res. 2002;22:3309-18
103. Klein C, Vassilev LT. Targeting the p53–MDM2 interaction to treat cancer. Br J Cancer. 2004;91:1415-9. doi: 10.1038/sj.bjc.6602164
104. Kong Y, Lu Z-L, Wang J-J, Zhou R, Guo J, Liu J, et al. Platycodin D, a metabolite of Platycodin grandiflorum, inhibits highly metastatic MDA-MB-231 breast cancer growth in vitro and in vivo by targeting the MDM2 oncogene. Oncol Rep. 2016;36:1447-56. doi: 10.3892/or.2016.4935
105. Koo N, Sharma AK, Narayan S. Therapeutics targeting p53-MDM2 interaction to induce cancer cell death. Int J Mol Sci. 2022;23(9):5005. doi: 10.3390/ijms23095005
106. Kung C-P, Murphy ME. The role of the p53 tumor suppressor in metabolism and diabetes. J Endocrinol. 2016;231(2):R61-75
107. Kuo P-L, Chiang L-C, Lin C-C. Resveratrol-induced apoptosis is mediated by p53-dependent pathway in Hep G2 cells. Life Sci. 2002;72(1):23-34. doi: 10.1016/S0024-3205(02)02177-X
108. Lacroix M, Toillon R-A, Leclercq G. p53 and breast cancer, an update. Endocr Relat Cancer. 2006;13:293-325. doi: 10.1677/erc.1.01172
109. Lai Z, Ferry KV, Diamond MA, Wee KE, Kim YB, Ma J, et al. Human mdm2 mediates multiple mono-ubiquitination of p53 by a mechanism requiring enzyme isomerization. J Biol Chem. 2001;276):31357-67. doi: 10.1074/jbc.M011517200
110. Leão M, Gomes S, Pedraza-Chaverri J, Machado N, Sousa E, Pinto M, et al. α-Mangostin and gambogic acid as potential inhibitors of the p53–mdm2 interaction revealed by a yeast approach. J Nat Prod. 2013;76:774-8. doi: 10.1021/np400049j
111. Lee JT, Gu W. The multiple levels of regulation by p53 ubiquitination. Cell Death Differ. 2010;17:86-92. doi: 10.1038/cdd.2009.77
112. Lerma Clavero A, Boqvist PL, Ingelshed K, Bosdotter C, Sedimbi S, Jiang L, et al. MDM2 inhibitors, nutlin-3a and navtemadelin, retain efficacy in human and mouse cancer cells cultured in hypoxia. Sci Rep. 2023;13(1):4583. doi: 10.1038/s41598-023-31484-0
113. Levine AJ. p53: 800 million years of evolution and 40 years of discovery. Nat Rev Cancer. 2020;20:471-80
114. Levine AJ. The many faces of p53: something for everyone. J Mol Cell Biol. 2019;11:524-30
115. Levine AJ, Oren M. The first 30 years of p53: growing ever more complex. Nat Rev Cancer. 2009;9:749-58. doi: 10.1038/nrc2723
116. Li H, Liu Y, Jiang W, Xue J, Cheng Y, Wang J, et al. Icaritin promotes apoptosis and inhibits proliferation by down-regulating AFP gene expression in hepatocellular carcinoma. BMC Cancer. 2021;21(1):318. doi: 10.1186/s12885-021-08043-9
117. Li M, Luo J, Brooks CL, Gu W. Acetylation of p53 inhibits its ubiquitination by Mdm2. J Biol Chem. 2002;277:50607-11
118. Li M, Zhang Z, Hill DL, Chen X, Wang H, Zhang R. Genistein, a dietary isoflavone, down-regulates the mdm2 oncogene at both transcriptional and posttranslational levels. Cancer Res. 2005;65:8200-8. doi: 10.1158/0008-5472.CAN-05-1302
119. Li M, Zhang Z, Hill DL, Wang H, Zhang R. Curcumin, a dietary component, has anticancer, chemosensitization, and radiosensitization effects by down-regulating the MDM2 oncogene through the PI3K/mTOR/ETS2 pathway. Cancer Res. 2007;67:1988-96. doi: 10.1158/0008-5472.CAN-06-3066
120. Li Q, Lozano G. Molecular pathways: targeting Mdm2 and Mdm4 in cancer therapy. Clin Cancer Res. 2013;19(1):34-41. doi: 10.1158/1078-0432.CCR-12-0053
121. Li S, Wu H, Tollefsbol TO. Combined broccoli sprouts and green tea polyphenols contribute to the prevention of estrogen receptor–negative mammary cancer via cell cycle arrest and inducing apoptosis in HER2/neu mice. J Nutr. 2021;151(1):73-84
122. Li W, Wang Y, Song Y, Xu L, Zhao J, Fang B. A preliminary study of the effect of curcumin on the expression of p53 protein in a human multiple myeloma cell line. Oncol Lett. 2015;9:1719-24. doi: 10.3892/ol.2015.2946
123. Lin W, Yan Y, Huang Q, Zheng D. MDMX in cancer: a partner of p53 and a p53-independent effector. Biologics. 2024;18:61-78. doi: 10.2147/btt.S436629
124. Liu J, Zhang X, Liu A, Liu S, Zhang L, Wu B, et al. Berberine induces apoptosis in p53-null leukemia cells by down-regulating XIAP at the post-transcriptional level. Cell Physiol Biochem. 2013;32:1213-24. doi: 10.1159/000354520
125. Liu Y, Su Z, Tavana O, Gu W. Understanding the complexity of p53 in a new era of tumor suppression. Cancer Cell. 2024;42:946-67. doi: 10.1016/j.ccell.2024.04.009
126. Liu Z, Wu X, Lv J, Sun H, Zhou F. Resveratrol induces p53 in colorectal cancer through SET7/9. Oncol Lett. 2019;17:3783-9. doi: 10.3892/ol.2019.10034
127. Luo K-W, Zhu X-h, Zhao T, Zhong J, Gao H-c, Luo X-L, et al. EGCG enhanced the anti-tumor effect of doxorubicine in bladder cancer via NF-κB/MDM2/p53 pathway. Front Cell Dev Biol. 2020;8:606123. doi: 10.3389/fcell.2020.606123
128. Maaliki D, Shaito AA, Pintus G, El-Yazbi A, Eid AH. Flavonoids in hypertension: a brief review of the underlying mechanisms. Curr Opin Pharmacol. 2019;45:57-65
129. Maguire M, Nield PC, Devling T, Jenkins RE, Park BK, Polański R, et al. MDM2 regulates dihydrofolate reductase activity through monoubiquitination. Cancer Res. 2008;68:3232-42. doi: 10.1158/0008-5472.CAN-07-5271
130. Malaguarnera R, Vella V, Pandini G, Sanfilippo M, Pezzino V, Vigneri R, et al. TAp73α increases p53 tumor suppressor activity in thyroid cancer cells via the inhibition of mdm2-mediated degradation. Mol Cancer Res. 2008;6(1):64-77. doi: 10.1158/1541-7786.MCR-07-0005
131. Manfredi JJ. The Mdm2–p53 relationship evolves: Mdm2 swings both ways as an oncogene and a tumor suppressor. Genes Dev. 2010;24:1580-9
132. Maor-Nof M, Shipony Z, Lopez-Gonzalez R, Nakayama L, Zhang Y-J, Couthouis J, et al. p53 is a central regulator driving neurodegeneration caused by C9orf72 poly (PR). Cell. 2021;184:689-708
133. Maya R, Balass M, Kim S-T, Shkedy D, Leal J-FM, Shifman O, et al. ATM-dependent phosphorylation of Mdm2 on serine 395: role in p53 activation by DNA damage. Genes Dev. 2001;15:1067-77
134. Mayo LD, Donner DB. A phosphatidylinositol 3-kinase/Akt pathway promotes translocation of Mdm2 from the cytoplasm to the nucleus. Proc Natl Acad Sci U S A. 2001;98:11598-603. doi: 10.1073/pnas.181181198
135. Men H, Cai H, Cheng Q, Zhou W, Wang X, Huang S, et al. The regulatory roles of p53 in cardiovascular health and disease. Cell Mol Life Sci. 2021;78:2001-18
136. Menendez D, Inga A, Resnick MA. The expanding universe of p53 targets. Nat Rev Cancer. 2009;9:724-37. doi: 10.1038/nrc2730
137. Merlin JPJ, Rupasinghe HPV, Dellaire G, Murphy K. Role of dietary antioxidants in p53-mediated cancer chemoprevention and tumor suppression. Oxid Med Cell Longev. 2021;2021:9924328. doi: 10.1155/2021/9924328
138. Mertens-Talcott SU, Bomser JA, Romero C, Talcott ST, Percival SS. Ellagic acid potentiates the effect of quercetin on p21waf1/cip1, p53, and MAP-kinases without affecting intracellular generation of reactive oxygen species in vitro. J Nutr. 2005;135:609-14. doi: 10.1093/jn/135.3.609
139. Meulmeester E, Pereg Y, Shiloh Y, Jochemsen AG. ATM-mediated phosphorylations inhibit Mdmx/ Mdm2 stabilization by HAUSP in favor of p53 activation. Cell Cycle. 2005;4:1166-70. doi: 10.4161/cc.4.9.1981
140. Midgley CA, Desterro JMP, Saville MK, Howard S, Sparks A, Hay RT, et al. An N-terminal p14ARF peptide blocks Mdm2-dependent ubiquitination in vitro and can activate p53 in vivo. Oncogene. 2000;19:2312-23
141. Miwa S, Uchida C, Kitagawa K, Hattori T, Oda T, Sugimura H, et al. Mdm2-mediated pRB downregulation is involved in carcinogenesis in a p53-independent manner. Biochem Biophys Res Commun. 2006;340(1):54-61
142. Moll UM, Petrenko O. The MDM2-p53 interaction. Mol Cancer Res. 2003;1:1001-8
143. Momand J, Jung D, Wilczynski S, Niland J. The MDM2 gene amplification database. Nucleic Acids Res. 1998;26:3453-9. doi: 10.1093/nar/26.15.3453
144. Morla-Barcelo PM, Laguna-Macarrilla D, Cordoba O, Matheu G, Oliver J, Roca P, et al. Unraveling malignant phenotype of peritumoral tissue: transcriptomic insights into early-stage breast cancer. Breast Cancer Res. 2024;26(1):89. doi: 10.1186/s13058-024-01837-2
145. Mu R, Qi Q, Gu H, Wang J, Yang Y, Rong J, et al. Involvement of p53 in oroxylin A-induced apoptosis in cancer cells. Molecular Carcinogenesis. 2009;48:1159-69. doi: 10.1002/mc.20570
146. Muhseen ZT, Li G. Promising terpenes as natural antagonists of cancer: an in-silico approach. Molecules. 2019;25(1):155. doi: 10.3390/molecules25010155
147. Mullard A. p53 programmes plough on. Nat Rev Drug Discov. 2020;19:497-500
148. Muller PAJ, Vousden KH. p53 mutations in cancer. Nat Cell Biol. 2013;15(1):2-8. doi: 10.1038/ncb2641
149. Muthumani P, Alagarsamy K, Dhandayuthapani S, Venkatesan T, Rathinavelu A. Pro-angiogenic effects of MDM2 through HIF-1α and NF-κB mediated mechanisms in LNCaP prostate cancer cells. Mol Biol Rep. 2014;41:5533-41
150. Nakamura S, Roth JA, Mukhopadhyay T. Multiple lysine mutations in the C-terminal domain of p53 interfere with MDM2-dependent protein degradation and ubiquitination. Mol Cell Biol. 2000;20:9391-8
151. Nandi S, Nag A, Khatua S, Sen S, Chakraborty N, Naskar A, et al. Anticancer activity and other biomedical properties of β-sitosterol: Bridging phytochemistry and current pharmacological evidence for future translational approaches. Phytother Res. 2024;38:592-619. doi: 10.1002/ptr.8061
152. Nasim S, Crooks PA. Antileukemic activity of aminoparthenolide analogs. Bioorg Med Chem Lett. 2008;18:3870-3. doi: 10.1016/j.bmcl.2008.06.050
153. Navarro S, Reddy R, Lee J, Warburton D, Driscoll B. Inhaled resveratrol treatments slow ageing-related degenerative changes in mouse lung. Thorax. 2017;72(5):451. doi: 10.1136/thoraxjnl-2016-208964
154. O’Neill EJ, Den Hartogh DJ, Azizi K, Tsiani E. Anticancer properties of carnosol: a summary of in vitro and in vivo evidence. Antioxidants (Basel). 2020;9 (10):961. doi: 10.3390/antiox9100961
155. Ogawara Y, Kishishita S, Obata T, Isazawa Y, Suzuki T, Tanaka K, et al. Akt enhances Mdm2-mediated ubiquitination and degradation of p53. J Biol Chem. 2002;277:21843-50. doi: 10.1074/jbc.M109745200
156. Oliner JD, Kinzler KW, Meltzer PS, George DL, Vogelstein B. Amplification of a gene encoding a p53-associated protein in human sarcomas. Nature. 1992;358 (6381):80-3
157. Oliner JD, Pietenpol JA, Thiagalingam S, Gyuris J, Kinzler KW, Vogelstein B. Oncoprotein MDM2 conceals the activation domain of tumour suppressor p53. Nature. 1993;362(6423):857-60
158. Oliner JD, Saiki AY, Caenepeel S. The role of MDM2 amplification and overexpression in tumorigenesis. Cold Spring Harb Perspect Med. 2016;6(6):a026336
159. Pasciu V, Posadino AM, Cossu A, Sanna B, Tadolini B, Gaspa L, et al. Akt downregulation by flavin oxidase–induced ROS generation mediates dose-dependent endothelial cell damage elicited by natural antioxidants. Toxicol Sci. 2010;114(1):101-12
160. Patel KD. Biological potential and therapeutic effectiveness of hinokiflavone in medicine: the effective components of herbal medicines for treatment of cancers and associated complications. Curr Nutr Food Sci. 2024;20:439-49. doi: 10.2174/1573401319666230602121227
161. Patiño-Morales CC, Soto-Reyes E, Arechaga-Ocampo E, Ortiz-Sánchez E, Antonio-Véjar V, Pedraza-Chaverri J, et al. Curcumin stabilizes p53 by interaction with NAD (P) H: quinone oxidoreductase 1 in tumor-derived cell lines. Redox Biol. 2020;28:101320
162. Peuget S, Zhou X, Selivanova G. Translating p53-based therapies for cancer into the clinic. Nat Rev Cancer. 2024;24:192-215. doi: 10.1038/s41568-023-00658-3
163. Popović-Djordjević J, Quispe C, Giordo R, Kostić A, Stanković JSK, Fokou PVT, et al. Natural products and synthetic analogues against HIV: A perspective to develop new potential anti-HIV drugs. Eur J Med Chem. 2022;233:114217
164. Posadino AM, Cossu A, Giordo R, Zinellu A, Sotgia S, Vardeu A, et al. Coumaric acid induces mitochondrial damage and oxidative-mediated cell death of human endothelial cells. Cardiovasc Toxicol. 2013;13:301-6
165. Posadino AM, Cossu A, Giordo R, Zinellu A, Sotgia S, Vardeu A, et al. Resveratrol alters human endothelial cells redox state and causes mitochondrial-dependent cell death. Food Chem Toxicol. 2015;78:10-6
166. Posadino AM, Giordo R, Cossu A, Nasrallah GK, Shaito A, Abou-Saleh H, et al. Flavin oxidase-induced ROS generation modulates PKC biphasic effect of resveratrol on endothelial cell survival. Biomolecules. 2019;9(6):209
167. Posadino AM, Giordo R, Pintus G, Mohammed SA, Orhan IE, Fokou PVT, et al. Medicinal and mechanistic overview of artemisinin in the treatment of human diseases. Biomed Pharmacother. 2023;163:114866
168. Posadino AM, Giordo R, Ramli I, Zayed H, Nasrallah GK, Wehbe Z, et al. An updated overview of cyanidins for chemoprevention and cancer therapy. Biomed Pharmacother. 2023;163:114783
169. Posadino AM, Maccioccu P, Eid AH, Giordo R, Pintus G, Fenu G. Citrus limon var. pompia Camarda var. nova: a comprehensive review of its botanical characteristics, traditional uses, phytochemical profile, and potential health benefits. Nutrients. 2024;16(16):2619
170. Poyurovsky MV, Katz C, Laptenko O, Beckerman R, Lokshin M, Ahn J, et al. The C terminus of p53 binds the N-terminal domain of MDM2. Nat Struct Mol Biol. 2011;18(4):516
171. Proietti S, Cucina A, Dobrowolny G, D'Anselmi F, Dinicola S, Masiello MG, et al. Melatonin down-regulates MDM2 gene expression and enhances p53 acetylation in MCF-7 cells. J Pineal Res. 2014;57(1):120-9. doi: 10.1111/jpi.12150
172. Qin J-J, Li X, Wang W, Zi X, Zhang R. Targeting the NFAT1-MDM2-MDMX network inhibits the proliferation and invasion of prostate cancer cells, independent of p53 and androgen. Front Pharmacol. 2017;8:917. doi: 10.3389/fphar.2017.00917
173. Qin J-J, Wang W, Sarkar S, Voruganti S, Agarwal R, Zhang R. Inulanolide A as a new dual inhibitor of NFAT1-MDM2 pathway for breast cancer therapy. Oncotarget. 2016;7:32566-78. doi: 10.18632/oncotarget.8873
174. Qin J-J, Wang W, Voruganti S, Wang H, Zhang W-D, Zhang R. Identification of a new class of natural product MDM2 inhibitor: In vitro and in vivo anti-breast cancer activities and target validation. Oncotarget. 2015;6:2623-40. doi: 10.18632/oncotarget.3098
175. Quispe C, Cruz-Martins N, Manca ML, Manconi M, Sytar O, Hudz N, et al. Nano‐derived therapeutic formulations with curcumin in inflammation‐related diseases. Oxid Med Cell Longev. 2021;2021(1):3149223
176. Quispe C, Herrera-Bravo J, Khan K, Javed Z, Semwal P, Painuli S, et al. Therapeutic applications of curcumin nanomedicine formulations in cystic fibrosis. Prog Biomater. 2022;11:321-9. doi: 10.1007/s40204-022-00198-3
177. Rahman MM, Wu H, Tollefsbol TO. A novel combinatorial approach using sulforaphane-and withaferin A-rich extracts for prevention of estrogen receptor-negative breast cancer through epigenetic and gut microbial mechanisms. Sci Rep. 2024;14(1):12091
178. Rahmani AH, Alsahli MA, Almatroudi A, Almogbel MA, Khan AA, Anwar S, et al. The potential role of apigenin in cancer prevention and treatment. Molecules. 2022;27(18):6051. doi: 10.3390/molecules27186051
179. Ramli I, Cheriet T, Posadino AM, Giordo R, Zayed H, Eid AH, et al. Potential therapeutic targets of resveratrol in the prevention and treatment of pulmonary fibrosis. Front Biosci (Landmark Ed). 2023;28(9):198. doi: 10.31083/j.fbl2809198
180. Ramli I, Cheriet T, Thuan DTB, Khoi DN, Thu DNK, Posadino AM, et al. Potential applications of antofine and its synthetic derivatives in cancer therapy: structural and molecular insights. Naunyn-Schmiedeberg's Arch Pharmacol. 2024;397:8231-58. doi: 10.1007/s00210-024-03180-x
181. Ramli I, Posadino AM, Giordo R, Fenu G, Fardoun M, Iratni R, et al. Effect of resveratrol on pregnancy, prenatal complications and pregnancy-associated structure alterations. Antioxidants. 2023;12(2):341
182. Ramli I, Posadino AM, Zerizer S, Spissu Y, Barberis A, Djeghim H, et al. Low concentrations of Ambrosia maritima L. phenolic extract protect endothelial cells from oxidative cell death induced by H2O2 and sera from Crohn's disease patients. J Ethnopharmacol. 2023;300:115722
183. Ravi R, Mookerjee B, Bhujwalla ZM, Sutter CH, Artemov D, Zeng Q, et al. Regulation of tumor angiogenesis by p53-induced degradation of hypoxia-inducible factor 1α. Genes Dev. 2000;14(1):34-44
184. Rayburn RE, Ezell JS, Zhang R. Recent advances in validating mdm2 as a cancer target. Anticancer Agents Med Chem. 2009;9:882-903. doi: 10.2174/187152009789124628
185. Reddy D, Kumavath R, Tan TZ, Ampasala DR, Kumar AP. Peruvoside targets apoptosis and autophagy through MAPK Wnt/β-catenin and PI3K/AKT/mTOR signaling pathways in human cancers. Life Sci. 2020;241:117147. doi: 10.1016/j.lfs.2019.117147
186. Ries S, Biederer C, Woods D, Shifman O, Shirasawa S, Sasazuki T, et al. Opposing effects of ras on p53: transcriptional activation of mdm2 and induction of p19ARF. Cell. 2000;103:321-30. doi: 10.1016/S0092-8674(00)00123-9
187. Rivera M, Ramos Y, Rodríguez-Valentín M, López-Acevedo S, Cubano LA, Zou J, et al. Targeting multiple pro-apoptotic signaling pathways with curcumin in prostate cancer cells. PLoS One. 2017;12(6):e0179587. doi: 10.1371/journal.pone.0179587
188. Rodriguez MS, Desterro JMP, Lain S, Lane DP, Hay RT. Multiple C-terminal lysine residues target p53 for ubiquitin-proteasome-mediated degradation. Mol Cell Biol. 2000;20:8458-67. doi: 10.1128/MCB.20.22.8458-8467.2000
189. Rong J-J, Hu R, Qi Q, Gu H-Y, Zhao Q, Wang J, et al. Gambogic acid down-regulates MDM2 oncogene and induces p21Waf1/CIP1 expression independent of p53. Cancer Lett. 2009;284(1):102-12. doi: 10.1016/j.canlet.2009.04.011
190. Rusin M. The p53 protein - not only the guardian of the genome. Postepy Biochem. 2024;70(1):71-87. doi: 10.18388/pb.2021_518
191. Sakaguchi K, Saito Si, Higashimoto Y, Roy S, Anderson CW, Appella E. Damage-mediated phosphorylation of human p53 threonine 18 through a cascade mediated by a casein 1-like kinase: Effect on Mdm2 binding. J Biol Chem. 2000;275:9278-83
192. Sammons MA, Nguyen T-AT, McDade SS, Fischer M. Tumor suppressor p53: from engaging DNA to target gene regulation. Nucleic Acids Res. 2020;48:8848-69
193. Sanna V, Roggio AM, Posadino AM, Cossu A, Marceddu S, Mariani A, et al. Novel docetaxel-loaded nanoparticles based on poly (lactide-co-caprolactone) and poly (lactide-co-glycolide-co-caprolactone) for prostate cancer treatment: formulation, characterization, and cytotoxicity studies. Nanoscale Res Lett. 2011;6(1):260. doi: 10.1186/1556-276X-6-260
194. Shahcheraghi HS, Tchokonte-Nana V, Lotfi M, Lotfi M, Ghorbani A, Sadeghnia RH. Wnt/beta-catenin and PI3K/Akt/mTOR signaling pathways in glioblastoma: two main targets for drug design: a review. Curr Pharm Design. 2020;26:1729-41. doi: 10.2174/1381612826666200131100630
195. Shaito A, Al-Mansoob M, Ahmad SM, Haider MZ, Eid AH, Posadino AM, et al. Resveratrol-mediated regulation of mitochondria biogenesis-associated pathways in neurodegenerative diseases: Molecular insights and potential therapeutic applications. Curr Neuropharmacol. 2023;21(5):1184
196. Shaito A, Posadino AM, Younes N, Hasan H, Halabi S, Alhababi D, et al. Potential adverse effects of resveratrol: A literature review. Int J Mol Sci. 2020;21(6):2084
197. Shaito A, Thuan DTB, Phu HT, Nguyen THD, Hasan H, Halabi S, et al. Herbal medicine for cardiovascular diseases: efficacy, mechanisms, and safety. Front Pharmacol. 2020;11:422
198. Shangary S, Wang S. Small-molecule inhibitors of the MDM2-p53 protein-protein interaction to reactivate p53 function: a novel approach for cancer therapy. Annu Rev Pharmacol Toxicol. 2009;49:223-41. doi: 10.1146/annurev.pharmtox.48.113006.094723
199. Sharifi-Rad J, Quispe C, Shaheen S, El Haouari M, Azzini E, Butnariu M, et al. Flavonoids as potential anti-platelet aggregation agents: from biochemistry to health promoting abilities. Crit Rev Food Sci Nutr. 2022;62:8045-58. doi: 10.1080/10408398.2021.1924612
200. She Q-B, Bode A, Ma WY, Chen NY, Dong Z. Resveratrol-induced activation of p53 and apoptosis is mediated by extracellular-signal-regulated protein kinases and p38 kinase. Cancer Res. 2001;61:1604-10
201. Sherr CJ. Divorcing ARF and p53: an unsettled case. Nat Rev Cancer. 2006;6:663-73
202. Sherr CJ. Tumor surveillance via the ARF–p53 pathway. Genes Dev. 1998;12:2984-91
203. Shieh S-Y, Ikeda M, Taya Y, Prives C. DNA damage-induced phosphorylation of p53 alleviates inhibition by MDM2. Cell. 1997;91:325-34
204. Shoaib TH, Abdelmoniem N, Mukhtar RM, Alqhtani AT, Alalawi AL, Alawaji R, et al. Molecular docking and molecular dynamics studies reveal the anticancer potential of medicinal-plant-derived lignans as MDM2-P53 interaction inhibitors. Molecules. 2023;28 (18):6665. doi: 10.3390/molecules28186665
205. Shukla S, Gupta S. Apigenin-induced prostate cancer cell death is initiated by reactive oxygen species and p53 activation. Free Radic Biol Med. 2008;44:1833-45. doi: 10.1016/j.freeradbiomed.2008.02.007
206. Siegl C, Rudel T. Modulation of p53 during bacterial infections. Nat Rev Microbiol. 2015;13:741-8
207. Song J, Ma S-J, Luo J-H, Zhang H, Wang R-X, Liu H, et al. Melatonin induces the apoptosis and inhibits the proliferation of human gastric cancer cells via blockade of the AKT/MDM2 pathway. Oncol Rep. 2018;39:1975-83. doi: 10.3892/or.2018.6282
208. Srivastava RK, Chen Q, Siddiqui I, Sarva K, Shankar S. Linkage of curcumin-induced cell cycle arrest and apoptosis by cyclin-dependent kinase inhibitor p21/WAF1/CIP1. Cell Cycle. 2007;6:2953-61. doi: 10.4161/cc.6.23.4951
209. Steffens Reinhardt L, Groen K, Newton C, Avery-Kiejda KA. The role of truncated p53 isoforms in the DNA damage response. Biochim Biophys Acta Rev Cancer. 2023;1878(3):188882. doi: 10.1016/j.bbcan.2023.188882
210. Stein RC, Joseph AEA, Matlin SA, Cunningham DC, Ford HT, Coombes RC. A preliminary clinical study of gossypol in advanced human cancer. Cancer Chemother Pharmacol. 1992;30:480-2. doi: 10.1007/BF00685601
211. Subash-Babu P, Alshammari GM, Ignacimuthu S, Alshatwi AA. Epoxy clerodane diterpene inhibits MCF-7 human breast cancer cell growth by regulating the expression of the functional apoptotic genes Cdkn2A, Rb1, mdm2 and p53. Biomed Pharmacother. 2017;87:388-96. doi: 10.1016/j.biopha.2016.12.091
212. Subhasree N, Jiangjiang Q, Kalkunte S, Minghai W, Ruiwen Z. The MDM2-p53 pathway revisited. J Biomed Res. 2013;27:254-71. doi: 10.7555/JBR.27.20130030
213. Sultana S, Munir N, Mahmood Z, Riaz M, Akram M, Rebezov M, et al. Molecular targets for the management of cancer using Curcuma longa Linn. phytoconstituents: A Review. Biomed Pharmacother. 2021;135:111078. doi: 10.1016/j.biopha.2020.111078
214. Szybińska A, Leśniak W. P53 dysfunction in neurodegenerative diseases-the cause or effect of pathological changes? Aging Dis. 2017;8(4):506
215. Takatori H, Kawashima H, Suzuki K, Nakajima H. Role of p53 in systemic autoimmune diseases. Crit Rev Immunol. 2014;34:509-16. doi: 10.1615/critrevimmunol.2014012193
216. Tamgue O, Lei M. Triptolide promotes senescence of prostate cancer cells through induction of histone methylation and heterochromatin formation. Asian Pacif J Cancer Prev. 2017;18:2519-26. doi: 10.22034/APJCP.2017.18.9.2519
217. Tang D, Chen K, Huang L, Li J. Pharmacokinetic properties and drug interactions of apigenin, a natural flavone. Exp Opin Drug Metab Toxicol. 2017;13:323-30. doi: 10.1080/17425255.2017.1251903
218. Tang Y-A, Lin R-K, Tsai Y-T, Hsu H-S, Yang Y-C, Chen C-Y, et al. MDM2 overexpression deregulates the transcriptional control of RB/E2F leading to dna methyltransferase 3a overexpression in lung cancer. Clin Cancer Res. 2012;18:4325-33. doi: 10.1158/1078-0432.CCR-11-2617
219. Tanigawa S, Fujii M, Hou D-X. Stabilization of p53 is involved in quercetin-induced cell cycle arrest and apoptosis in HepG2 cells. Biosci Biotechnol Biochem. 2008;72:797-804. doi: 10.1271/bbb.70680
220. Tao S, Yang Y, Fan Y, Chu K, Sun J, Wu Q, et al. Melatonin protects against nonylphenol caused pancreatic β-cells damage through MDM2-P53-P21 axis. Toxicol Res. 2022;11:391-401. doi: 10.1093/toxres/tfac016
221. Thomasova D, Mulay SR, Bruns H, Anders H-J. p53-independent roles of MDM2 in NF-κB signaling: implications for cancer therapy, wound healing, and autoimmune diseases. Neoplasia. 2012;14:1097-101. doi: 10.1593/neo.121534
222. Thut CJ, Goodrich JA, Tjian R. Repression of p53-mediated transcription by MDM2: a dual mechanism. Genes Dev. 1997;11:1974-86
223. Truong AHL, Cervi D, Lee J, Ben-David Y. Direct transcriptional regulation of MDM2 by Fli-1. Oncogene. 2005;24:962-9. doi: 10.1038/sj.onc.1208323
224. Tuli HS, Tuorkey MJ, Thakral F, Sak K, Kumar M, Sharma AK, et al. Molecular mechanisms of action of genistein in cancer: recent advances. Front Pharmacol. 2019;10:1336. doi: 10.3389/fphar.2019.01336
225. Uchida C, Miwa S, Kitagawa K, Hattori T, Isobe T, Otani S, et al. Enhanced Mdm2 activity inhibits pRB function via ubiquitin‐dependent degradation. EMBO J. 2005;24(1):160-9. doi: 10.1038/sj.emboj.7600486
226. Ullah A, Munir S, Badshah SL, Khan N, Ghani L, Poulson BG, et al. Important flavonoids and their role as a therapeutic agent. Molecules. 2020;25(22):5243. doi: 10.3390/molecules25225243
227. Unger T, Juven-Gershon T, Moallem E, Berger M, Vogt Sionov R, Lozano G, et al. Critical role for Ser20 of human p53 in the negative regulation of p53 by Mdm2. EMBO J. 1999;18:1805-14. doi: 10.1093/emboj/18.7.1805
228. Vazhappilly CG, Ansari SA, Al-Jaleeli R, Al-Azawi AM, Ramadan WS, Menon V, et al. Role of flavonoids in thrombotic, cardiovascular, and inflammatory diseases. Inflammopharmacology. 2019;27:863-9. doi: 10.1007/s10787-019-00612-6
229. Verma S, Singh A, Mishra A. Molecular dynamics investigation on the inhibition of MDM2-p53 interaction by polyphenols. Mol Inform. 2013;32:203-12. doi: 10.1002/minf.201200113
230. Vidya Priyadarsini R, Senthil Murugan R, Maitreyi S, Ramalingam K, Karunagaran D, Nagini S. The flavonoid quercetin induces cell cycle arrest and mitochondria-mediated apoptosis in human cervical cancer (HeLa) cells through p53 induction and NF-κB inhibition. Eur J Pharmacol. 2010;649(1):84-91. doi: 10.1016/j.ejphar.2010.09.020
231. Vlatkovic N, Guerrera S, Li Y, Linn S, Haines DS, Boyd MT. MDM2 interacts with the C-terminus of the catalytic subunit of DNA polymerase ε. Nucleic Acids Res. 2000;28:3581-6. doi: 10.1093/nar/28.18.3581
232. Vogel SM, Bauer MR, Joerger AC, Wilcken R, Brandt T, Veprintsev DB, et al. Lithocholic acid is an endogenous inhibitor of MDM4 and MDM2. Proc Natl Acad Sci U S A. 2012;109:16906-10. doi: 10.1073/pnas.1215060109
233. Volate SR, Kawasaki BT, Hurt EM, Milner JA, Kim YS, White J, et al. Gossypol induces apoptosis by activating p53 in prostate cancer cells and prostate tumor–initiating cells. Mol Cancer Ther. 2010;9:461-70. doi: 10.1158/1535-7163.MCT-09-0507
234. Vousden KH, Prives C. Blinded by the light: the growing complexity of p53. Cell. 2009;137:413-31. doi: 10.1016/j.cell.2009.04.037
235. Wang H, Guo M, Wei H, Chen Y. Targeting p53 pathways: mechanisms, structures, and advances in therapy. Signal Transduct Target Ther. 2023;8(1):92. doi: 10.1038/s41392-023-01347-1
236. Wang H, Yuan Z, Chen Z, Yao F, Hu Z, Wu B. [Effect of quercetin on glioma cell U87 apoptosis and feedback regulation of MDM2-p53]. Nan fang yi ke da xue xue bao = Journal of Southern Medical University. 2014;34:686-9
237. Wang L, Zeng Y, Liu Y, Hu X, Li S, Wang Y, et al. Fucoxanthin induces growth arrest and apoptosis in human bladder cancer T24 cells by up-regulation of p21 and down-regulation of mortalin. Acta Biochim Biophys Sin (Shanghai). 2014;46:877-84. doi: 10.1093/abbs/gmu080
238. Wang W, Rayburn ER, Hao M, Zhao Y, Hill DL, Zhang R, et al. Experimental therapy of prostate cancer with novel natural product anti-cancer ginsenosides. Prostate. 2008;68:809-19. doi: 10.1002/pros.20742
239. Wang W, Wang H, Rayburn ER, Zhao Y, Hill DL, Zhang R. 20(S)-25-methoxyl-dammarane-3β, 12β, 20-triol, a novel natural product for prostate cancer therapy: activity in vitro and in vivo and mechanisms of action. Br J Cancer. 2008;98:792-802. doi: 10.1038/sj.bjc.6604227
240. Wang W, Zhang X, Qin J-J, Voruganti S, Nag SA, Wang M-H, et al. Natural product ginsenoside 25-OCH3-PPD inhibits breast cancer growth and metastasis through down-regulating MDM2. PLoS One. 2012;7(7):e41586. doi: 10.1371/journal.pone.0041586
241. Wasylishen AR, Lozano G. Attenuating the p53 pathway in human cancers: many means to the same end. Cold Spring Harb Perspect Med. 2016;6(8):a026211
242. Weber JD, Taylor LJ, Roussel MF, Sherr CJ, Bar-Sagi D. Nucleolar Arf sequesters Mdm2 and activates p53. Nat Cell Biol. 1999;1(1):20-6. doi: 10.1038/8991
243. Wei C, Du J, Shen Y, Wang Z, Lin Q, Chen J, et al. Anticancer effect of involucrasin A on colorectal cancer cells by modulating the Akt/MDM2/p53 pathway. Oncol Lett. 2023;25(6):218. doi: 10.3892/ol.2023.13804
244. Wolf ER, Mabry AR, Damania B, Mayo LD. Mdm2-mediated neddylation of pVHL blocks the induction of antiangiogenic factors. Oncogene. 2020;39:5228-39
245. Wu H, Ng R, Chen X, Steer CJ, Song G. MicroRNA-21 is a potential link between non-alcoholic fatty liver disease and hepatocellular carcinoma via modulation of the HBP1-p53-Srebp1c pathway. Gut. 2016;65:1850-60
246. Wu S, Chang L, Tian L. Identification and characterization of two regiospecific tricetin UDP-dependent glycosyltransferases from pomegranate (Punica granatum L.). Plants. 2022;11(6):810
247. Wu X, Bayle JH, Olson D, Levine AJ. The p53-mdm-2 autoregulatory feedback loop. Genes Dev. 1993;7:1126-32. doi: 10.1101/gad.7.7a.1126
248. Xiong J, Li J, Yang Q, Wang J, Su T, Zhou S. Gossypol has anti-cancer effects by dual-targeting MDM2 and VEGF in human breast cancer. Breast Cancer Res. 2017;19(1):27. doi: 10.1186/s13058-017-0818-5
249. Xu H, Zhang Z, Li M, Zhang R. MDM2 promotes proteasomal degradation of p21Waf1 via a conformation change. J Biol Chem. 2010;285:18407-14. doi: 10.1074/jbc.M109.059568
250. Xu J, Wold EA, Ding Y, Shen Q, Zhou J. Therapeutic potential of oridonin and its analogs: from anticancer and antiinflammation to neuroprotection. Molecules. 2018;23(2):474. doi: 10.3390/molecules23020474
251. Yan X, Qi M, Li P, Zhan Y, Shao H. Apigenin in cancer therapy: anti-cancer effects and mechanisms of action. Cell Biosci. 2017;7(1):50. doi: 10.1186/s13578-017-0179-x
252. Yang J-Y, Zong CS, Xia W, Wei Y, Ali-Seyed M, Li Z, et al. MDM2 promotes cell motility and invasiveness by regulating E-cadherin degradation. Mol Cell Biol. 2006;26:7269-82. doi: 10.1128/MCB.00172-06
253. Yang J-Y, Zong CS, Xia W, Yamaguchi H, Ding Q, Xie X, et al. ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2-mediated degradation. Nat Cell Biol. 2008;10(2):138-48
254. Yang L, Liu Y, Wang M, Qian Y, Dong X, Gu H, et al. Quercetin-induced apoptosis of HT-29 colon cancer cells via inhibition of the Akt-CSN6-Myc signaling axis. Mol Med Rep. 2016;14:4559-66. doi: 10.3892/mmr.2016.5818
255. Yang Q, Zhao J, Chen D, Wang Y. E3 ubiquitin ligases: styles, structures and functions. Mol Biomed. 2021;2(1):23. doi: 10.1186/s43556-021-00043-2
256. Yao Y, Zhang Q, Li Z, Zhang H. MDM2: current research status and prospects of tumor treatment. Cancer Cell Int. 2024;24(1):170. doi: 10.1186/s12935-024-03356-8
257. Yu GW, Rudiger S, Veprintsev D, Freund S, Fernandez-Fernandez MR, Fersht AR. The central region of HDM2 provides a second binding site for p53. Proc Natl Acad Sci U S A. 2006;103:1227-32
258. Zafar A, Khan MJ, Naeem A. MDM2- an indispensable player in tumorigenesis. Mol Biol Rep. 2023;50:6871-83. doi: 10.1007/s11033-023-08512-3
259. Zanjirband M, Rahgozar S. Targeting p53-MDM2 interaction using small molecule inhibitors and the challenges needed to be addressed. Curr Drug Targets. 2019;20:1091-111
260. Zhai D, Jin C, Shiau C-w, Kitada S, Satterthwait AC, Reed JC. Gambogic acid is an antagonist of antiapoptotic Bcl-2 family proteins. Mol Cancer Ther. 2008;7:1639-46. doi: 10.1158/1535-7163.MCT-07-2373
261. Zhang F, Li M, Wu X, Hu Y, Cao Y, Wang Xa, et al. 20(S)-ginsenoside Rg3 promotes senescence and apoptosis in gallbladder cancer cells via the p53 pathway. Drug Des Devel Ther. 2015;9:3969-87. doi: 10.2147/DDDT.S84527. Erratum in: Drug Des Devel Ther. 2019;13:3527-8. doi: 10.2147/DDDT.S230835
262. Zhang J, Sun Q, Zhang Z, Ge S, Han ZG, Chen WT. Loss of microRNA-143/145 disturbs cellular growth and apoptosis of human epithelial cancers by impairing the MDM2-p53 feedback loop. Oncogene. 2013;32(1):61-9. doi: 10.1038/onc.2012.28
263. Zhang L, Li J, Hogan S, Chung H, Welbaum GE, Zhou K. Inhibitory effect of raspberries on starch digestive enzyme and their antioxidant properties and phenolic composition. Food Chem. 2010;119:592-9. doi: 10.1016/j.foodchem.2009.06.063
264. Zhang L, Yu D, Hu M, Xiong S, Lang A, Ellis LM, et al. Wild-type p53 suppresses angiogenesis in human leiomyosarcoma and synovial sarcoma by transcriptional suppression of vascular endothelial growth factor expression. Cancer Res. 2000;60:3655-61
265. Zhang R, Wang H. MDM2 oncogene as a novel target for human cancer therapy. Curr Pharm Des. 2000;6:393-416. doi: 10.2174/1381612003400911
266. Zhang X, Zhang Z, Cheng J, Li M, Wang W, Xu W, et al. Transcription factor NFAT1 activates the mdm2 oncogene independent of p53. J Biol Chem. 2012;287:30468-76. doi: 10.1074/jbc.M112.373738
267. Zhang Y, Chen N, Xin N, Li Q, Zhang T, Ye H, et al. Complexation of chlorogenic acid enhances the antiproliferative effect of lactoferrin to colon cancer cells. Food Biosci. 2022;46:101601. doi: 10.1016/j.fbio.2022.101601
268. Zhang Y, Xiong Y. Mutations in human ARF exon 2 disrupt its nucleolar localization and impair its ability to block nuclear export of MDM2 and p53. Mol Cell. 1999;3:579-91
269. Zhang Z, Wang H, Li M, Agrawal S, Chen X, Zhang R. MDM2 is a negative regulator of p21WAF1/CIP1, independent of p53. J Biol Chem. 2004;279:16000-6. doi: 10.1074/jbc.M312264200
270. Zhao D-D, Jiang L-L, Li H-Y, Yan P-F, Zhang Y-L. Chemical components and pharmacological activities of terpene natural products from the genus paeonia. Molecules. 2016;21(10):1362. doi: 10.3390/molecules21101362
271. Zhao J, Blayney A, Liu X, Gandy L, Jin W, Yan L, et al. EGCG binds intrinsically disordered N-terminal domain of p53 and disrupts p53-MDM2 interaction. Nat Commun. 2021;12(1):986. doi: 10.1038/s41467-021-21258-5
272. Zhao R, Yeung S-CJ, Chen J, Iwakuma T, Su C-H, Chen B, et al. Subunit 6 of the COP9 signalosome promotes tumorigenesis in mice through stabilization of MDM2 and is upregulated in human cancers. J Clin Invest. 2011;121:851-65. doi: 10.1172/JCI44111
273. Zheng P-W, Chiang L-C, Lin C-C. Apigenin induced apoptosis through p53-dependent pathway in human cervical carcinoma cells. Life Sci. 2005;76:1367-79. doi: 10.1016/j.lfs.2004.08.023
274. Zhou BP, Liao Y, Xia W, Zou Y, Spohn B, Hung M-C. HER-2/neu induces p53 ubiquitination via Akt-mediated MDM2 phosphorylation. Nat Cell Biol. 2001;3:973-82. doi: 10.1038/ncb1101-973
275. Zhou P, Huang S, Shao C, Huang D, Hu Y, Su X, et al. The antiproliferative and proapoptotic effects of cucurbitacin B on BPH-1 cells via the p53/MDM2 axis. Int J Mol Sci. 2023;25(1):442
276. Zhu HQ, Zhang C, Guo ZY, Yang JM, Guo JH, Chen C, et al. Oridonin induces Mdm2-p60 to promote p53-mediated apoptosis and cell cycle arrest in neuroblastoma. Cancer Med. 2019;8:5313-26. doi: 10.1002/cam4.2393
277. Zhuang W, Sun N, Gu C, Liu S, Zheng Y, Wang H, et al. A literature review on Epimedium, a medicinal plant with promising slow aging properties. Heliyon. 2023;9 (11):e21226. doi: 10.1016/j.heliyon.2023.e21226
 
 

Figure 1: p53 functions against cellular stress signals and target genes. In response to various inducers of cellular stress, the activation of p53 results in its post-translational via phosphorylation (P), acetylation (Ac), or ubiquitination (Ub). This activation contributes to the induction of multiple genes implicated in different cellular processes via regulation of downstream targets and/or signaling pathways, including apoptosis, cell cycle arrest, DNA repair…etc. p21: cyclin-dependent kinase inhibitor 1, 14-3-3a: 14-3-3a protein, Btg2: B-cell translocation gene 2, Reprimo: Reprimo gene, Gadd45a: Growth arrest and DNA-damage-inducible gene, p53r2: p53 inducible ribonucleotide reductase gene, Ddb2: Damage Specific DNA Binding Protein 2 coding gene, Mgmt: Methylguanine methyltransferase coding gene, Puma: P53 Upregulated Modulator of Apoptosis coding gene, Noxa: axotomy-induced motor neuron death, Bax: Bcl-2-associated X coding gene, Pig3: p53 inducible gene 3, Pai1: plasminogen activator inhibitor 1, Pml: Promyelocytic leukemia protein gene, Cddkn1 a: Cyclin Dependent Kinase Inhibitor 1a, Tsp: thrombospondine 1, Bai 1: Brain-specific angiogenesis inhibitor 1, Epha2: ephrin type-A receptor 2 coding gene, Col4a1: Collagen type IV alpha 1 Chain, Atg 10: ATG10 autophagy related 10, Tsc 2: tuberous sclerosis complex 2 gene, Tpp1: Tripeptidyl-peptidase 1 coding gene, Foxo3: transcription factor forkhead box O-3, Tigar: TP53 Induced Glycolysis regulatory phosphatase, Gls2 Glutaminase 2, Gpx1: Glutathione peroxidase 1, Sesn1/2: Sestrin 1 and 2 coding gene

Figure 2: MDM2 functions and targeted cellular processes. The diagram illustrates the diverse roles of MDM2 in cancer progression and cellular processes. MDM2, a fundamental regulator of the p53 tumor suppressor, influences multiple cellular pathways. It inhibits growth via downregulating TGF-β and affects apoptosis through reactive oxygen species (ROS) and p53 modulation. MDM2's role in metabolism includes the regulation of glycolysis and ROS levels. In DNA synthesis and repair, MDM2 interacts with DNA polymerase ε. MDM2 also promotes metastasis by upregulating MMP2/9 and E-cadherin (E-cad) and downregulating N-cadherin (N-cad). Additionally, it contributes to angiogenesis by regulating VGFA and VGEF. The arrows indicate whether MDM2 induces/upregulates (black arrows) or inhibits/downregulates (black lines) specific processes and molecules. The dashed lines represent the interconnected pathways influenced by MDM2, emphasizing its central role in cancer biology.

Figure 3: p53-MDM2 interaction. p53 and MDM2 are part of an autoregulatory feedback loop. p53 increases MDM2 expression, which in turn inhibits p53 activity by promoting its degradation in the nucleus and cytoplasm, inhibiting its transcriptional activity, and facilitating its nuclear export. Various DNA-damaging agents or oncogene inhibitors trigger p53 activity. DNA damage leads to p53 phosphorylation and enhances MDM2, preventing p53 interaction and ubiquitination. In parallel, activated oncogenes induce ARF protein, leading to the sequestration of MDM2 into the nucleus, hence blocking p53 degradation. In contrast, survival signals regulate the nuclear transport of MDM2 via the Akt pathway, leading to the destabilization of p53. p53: Tumor suppressor 53, MDM2: The murine double minute 2, P: phosphorylation, Ub: Ubiquitination, Ac: Acetylation, Arf: ADP ribosylation factor

Figure 4: The impact of natural products on the cellular processes and target genes mediated by p53-MDM2 interaction. Crosstalk of the anticancer activity of natural products mediated by the p53-MDM2 pathway: When the MDM2-p53 interaction is modulated by natural products, p53 accumulates and activates its direct transcriptional targets, leading to protein synthesis. This results in various cellular responses: p21 induces cell cycle arrest; PUMA, NOXA, and BAX activate the intrinsic apoptotic pathway; FAS induces the extrinsic apoptotic pathway; MDM2 and WIP1 regulate p53 feedback and other pathways involved in DNA repair and cell metabolism. Cell cycle progression is primarily regulated by p53 activity via the p21 protein, which binds to and inhibits the CDK/cyclin complexes, blocking cell cycle progression. Consequently, CDK4/6 with cyclin D/E mediates the activity of RB and E2F1. This activation eventually releases E2F1, which then activates its transcriptional program, leading to cell cycle progression. Key Proteins and Pathways: AKT: Ak mouse strain thymoma protein, PTEN: Phosphatase and TENsin homolog, PI3K: phosphoinositide 3-kinase, MAPK: Mitogen-activated protein kinase, HSP27: heat shock protein 27, SRC: Proto-oncogene tyrosine-protein kinase, SCK: Shc-related adaptor protein, MKK3/6: Map kinase kinase isoforms 3 and 6, VRAP:VEGF-receptor-associated protein/T-cell-specific adaptor molecule, PDK1: Phosphoinositide-dependent kinase-1, Csp9: Caspase-9, Csp8: Caspase-8, Csp3: Caspase-3, Arf: ADP-ribosylation factor, MDM2: The murine double minute 2, MDMX: The murine double minute X, p53: Tumor suppressor 53, BCL: B-cell lymphoma 2 protein, MCL: Induced myeloid leukemia cell differentiation protein, PUMA: P53 upregulated modulator of apoptosis, BAX: Bcl-2-associated X protein, Wip1: Wild-type p53-induced phosphatase1, p21: cyclin-dependent kinase inhibitor 1, E2F1: E2F transcription factor 1, FAS: Fas cell surface death receptor, C-Myc: C-MYC proto-oncogene, BHLH transcription factor protein, CDK1: cyclin-dependent kinase 1, CDK2: cyclin-dependent kinase 2, CDK4: cyclin-dependent kinase 4

 

Table 1: Comprehensive overview of p53-MDM2 biology, regulation, and interaction in cancer

Table 2: Mechanistic actions and binding modes of some natural compounds that were found to modulate the p53-MDM2 interaction

Table 3: Synergistic effects of natural compounds on the p53-MDM2 pathway and cancer cell proliferation

[*] Corresponding Author:

Daniela Calina, Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania, eMail: calinadaniela@gmail.com