Next Article in Journal
Effects of CSN1/CSN2 Mutants in Flavonoid Metabolism on Rice (Oryza sativa L.)
Previous Article in Journal
Exploiting Paradoxical Activation of Oncogenic MAPK Signaling by Targeting Mitochondria to Sensitize NRAS Mutant-Melanoma to Vemurafenib
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Prunin: An Emerging Anticancer Flavonoid

by
Juie Nahushkumar Rana
1 and
Sohail Mumtaz
2,*
1
Fels Cancer Institute for Personalized Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
2
Department of Chemical and Biological Engineering, Gachon University, 1342 Seongnamdaero, Sujeong-gu, Seongnam-si 13120, Republic of Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(6), 2678; https://doi.org/10.3390/ijms26062678
Submission received: 13 February 2025 / Revised: 12 March 2025 / Accepted: 14 March 2025 / Published: 16 March 2025
(This article belongs to the Section Bioactives and Nutraceuticals)

Abstract

:
Despite the substantial advances in cancer therapies, developing safe and effective treatment methodologies is critical. Natural (plant-derived compounds), such as flavonoids, might be crucial in developing a safe treatment methodology without toxicity toward healthy tissues. Prunin is a flavonoid with the potential to be used in biomedical applications. Prunin has yet to undergo thorough scientific research, and its precise molecular mechanisms of action remain largely unexplored. This review summarizes the therapeutic potential of prunin for the first time, focusing on its underlying mechanisms as an anticancer compound. Prunin has gained significant attention due to its antioxidant, anti-inflammatory, and anticancer effects. This review aims to unlock how prunin functions at the molecular level to exert its anticancer effects, primarily modulating key cellular pathways. Furthermore, we have discussed the prunin’s potential as an adjunctive therapy with conventional treatments, highlighting its ability to strengthen treatment responses while decreasing drug resistance. Moreover, the discussion probes into innovative delivery methods, particularly nanoformulations, that might address prunin’s bioavailability, solubility, and stability limitations and optimize its therapeutic application. By providing a comprehensive analysis of prunin’s properties, this review aims to stimulate further exploration of using prunin as an anticancer agent, thereby progressing the development of targeted, selective, safe, and effective therapeutic methods.

1. Introduction

1.1. Flavonoids and Their Significance in Biomedical Applications

Flavonoids are polyphenolic compounds synthesized in plants as bioactive with significant biological activities [1,2]. The flavonoids are responsible for various color, flavor, and pharmacological activities [1,3]. These compounds comprise 15 carbon atoms arranged into two aromatic rings coupled by a three-carbon linkage. Based on disparities in the structure of the C-ring, they are classified into distinct subgroups, including flavones, flavanones, isoflavones, flavanols, flavan-3-ols, and anthocyanidins [4,5,6]. Plants contain flavonoids that function as secondary metabolites, fulfilling essential roles like protection against UV radiation, pathogens, and herbivores [7,8]. In the inflammatory process, flavonoids can operate in several ways, including acting as antioxidants, scavenging reactive oxygen species (ROS), or decreasing free radical accumulation [9]. It also acts as an inhibitor of the activity of some regulatory enzymes like protein kinases and phosphodiesterase [10]. They modulate the activity of the immune cells (e.g., inhibition of cell activation, maturation, signaling transduction, and secretory processes). Some flavonols (e.g., quercetin, rutin, and morin), flavanones (e.g., hesperetin and hesperidin), flavonols (e.g., catechin), isoflavones (e.g., genisten), and anthocyanins (e.g., cyanidin) have exhibited anti-inflammatory functions during in vitro and in vivo experiments, as well as clinical studies [11,12]. The anti-inflammatory properties of flavonoids and their relevant compounds also significantly affect cancer development. These compounds show their activity by deactivating carcinogens, inducing apoptosis, triggering cell cycle arrest, and inhibiting angiogenesis through various pathway activation [13]. Identifying natural flavonoids as an effective and safer source of antioxidants opens new perspectives to explore more of these compounds, focusing on new structures using the latest methodologies and technologies and exploiting other new natural sources.
Prunin, a flavonoid glycoside, is a bioactive compound present in many plants and fruits [14]. It has attracted considerable interest due to its potential anticancer effects, including promoting apoptosis, inhibiting cell growth, and preventing tumor development by modulating essential signaling pathways [15]. Like other extensively researched flavonoids, prunin is anticipated to show a positive safety profile owing to its natural origins and structural resemblance to non-toxic flavonoids [16,17]. This selective toxicity probably stems from prunin’s capability to influence certain molecular pathways in cancer cells while sparing normal cells from adverse effects [18,19]. These attributes position prunin as a promising subject for future cancer prevention and treatment research [20,21,22,23].

1.2. Introduction to Prunin: Sources and Structure

Prunin is a flavonoid glycoside primarily found in plants, especially those from the Prunus genus, and is integral to plant growth, development, and defense systems [24,25]. Prunin is mainly found in citrus fruits and in plants belonging to the Prunus genus, including cherries and plums [15,22,26]. These bioactive compounds are produced via the phenylpropanoid pathway, a complex metabolic method including enzymes like chalcone synthase and flavanone 3-hydroxylase [27], which convert phenylalanine into flavonoid precursors [28,29,30,31]. Cherries and plums, for example, are considered some of the richest natural sources of prunin [32,33]. The most common sources of prunin include cherries (Prunus avium and Prunus cerasus) [34], plums (Prunus domestica) [35], peaches (Prunus persica), apricots (Prunus armeniaca), and almonds (Prunus dulcis) [36]. These fruits, especially their seeds and skins, contain significant concentrations of prunin, contributing to their antioxidant, anti-inflammatory, and potential anticancer properties. Besides cherries, other fruits in the Prunus family, such as blackberries and nectarines, may contain prunin, albeit in varying amounts [36]. Fruits are not the only sources of prunin; some medicinal plants within the Prunus genus or related families also contain prunin, which is used in traditional medicine for treating various diseases due to its therapeutic properties [26,36,37].
Prunin represents a broad spectrum of biological activities, such as antioxidant, anti-inflammatory, neuroprotective effects, and anti-diabetic [38]. Recent progressions in extraction and analytical techniques have enabled the isolation and characterization of prunin and other flavonoids, allowing scientists to explore their biomedical effects [39,40,41]. Monoglycoside flavonoid prunin (4,5,7-trihydroxyflavanone-β-D-glucoside) is a hydrolysis product of naringin, and the water solubility of prunin is 7.6-fold that of naringin at 25 °C, which has various biological activities [42,43]. Various methods for prunin production from different sources have been studied, including plant extraction, enzymatic techniques, and microbial and chemical synthesis [44,45].
Prunin, a flavanone glycoside, features a chemical structure characterized by a flavanone backbone accompanied by a benzopyran skeleton [20,46]. This skeleton is typically segmented into three rings: the A-ring (the leftmost phenolic ring that contains the hydroxyl (OH) group), the B-ring (the rightmost phenolic ring), and the C-ring (the central pyran ring comprising an oxygen atom), as illustrated in Figure 1A. The structure also features several OH groups on the A and B rings, which enhances its antioxidant properties [20]. Additionally, a sugar moiety, like rhamnose or a similar glycoside, is connected to the C-ring, aiding its interaction with cellular targets [47,48,49].
Prunin’s selective toxicity toward cancer cells, as demonstrated by in vitro studies, is linked to its capacity to influence particular molecular pathways that are more active in tumor cells [48,50]. The hydroxyl groups on the B ring engage in hydrogen bonding and π-π stacking interactions with proteins and enzymes that are overexpressed in malignant cells, including tyrosine kinases and topoisomerases, eventually disrupting their functionalities [51,52,53]. In contrast, the glycosylated version of prunin appears to reduce its toxicity towards normal cells, likely because of reduced membrane permeability and a lower affinity for non-cancerous cellular targets. This characteristic is similar to that seen in other glycosylated flavonoids, including naringin and hesperidin [54,55]. This selective action is also reinforced by prunin’s structural resemblance to other non-toxic flavonoids, which are naturally occurring compounds that the human body metabolizes efficiently with no or negligible adverse effects [56,57,58].
Prunin’s safety profile is supported by its natural source, usually obtained from citrus fruits [59,60,61], and its structural similarity to well-researched flavonoids like quercetin and kaempferol [62,63,64], which demonstrates low toxicity in normal cells when used at therapeutic levels. The existence of sugar moiety may help lower the risk of oxidative damage and off-target effects, as glycosylation frequently stabilizes flavonoid structures and adjusts their reactivity [65,66]. These structures and mechanistic insights, drawn from a review of the current literature, highlight prunins’ potential as a safe and selective anticancer agent, justifying further research and concentration optimizations at various cancer targets.
Prunin is a flavonoid glycoside that assists as the glucoside derivative of prunetin [67]. The structure of prunin features a flavone backbone with a glucose molecule linked to the hydroxyl group at the C-7 position, as shown in Figure 1. This glycosylation increases its solubility and bioavailability [68], which is essential for its medical use. The biosynthesis of prunin encompasses the enzymatic conjugation of glucose to prunetin [69]. The plant’s enzymatic machinery mainly regulates the synthesis of prunin compounds [70]. The formation of prunin in plants is considered an adaptive mechanism that increases the plant’s ability to decrease or prevent oxidative damage [71].
Bioavailability refers to how rapidly a compound enters systemic circulation and reaches its target site in the body [72]. For prunin, this bioavailability is affected by its chemical structure, particularly its glycosylated form. As a flavonoid glycoside, prunin’s glucose component increases its solubility in water, which is vital for gastrointestinal absorption. Factors like enzymatic hydrolysis play a role, as gut microbiota and intestinal enzymes can convert prunin into its aglycone form (prunetin) or other metabolites [73,74]. This conversion is essential because the bioavailability of the aglycone can differ from that of prunin itself. Once absorbed, prunin and its metabolites undergo further metabolism in the liver, primarily involving phase I reactions (like oxidation and reduction) and phase II reactions (like conjugating with glucuronic acid or sulfate) [75,76]. The metabolites then circulate to various tissues, where they produce pharmacological effects [77]. Grasping prunin’s metabolic pathways and bioavailability is essential for assessing its therapeutic potential, as effective delivery to target tissues is key for its anticancer effects.

1.3. Importance of Prunin in Cancer Research

Cancer is known as a leading cause of death worldwide [78]. Mutations in DNA inside the nucleus of cells, caused by any exogenous or endogenous stimulant anomaly in normal cells, lead to uncontrolled division of cells, leading to tumor development and progression [79,80]. Several medications are available. For example, the most well-known chemotherapeutic agents seem to exert anti-cancer effects by assisting in cell apoptosis. In contrast, therapeutically active plant-derived compounds are regarded as marginally or not toxic while possessing significant pharmacological properties [81]. Prunin is one of the most prominent plant compounds with antioxidant and anti-cancer properties [82,83]. It has drawn considerable attention from researchers due to its structure and ability to modulate various biological pathways involved in cancer development [84]. Prunin possesses significant antioxidant activity, is vital in combating oxidative stress, and is a major carcinogen contributor [85,86,87,88]. ROS activates various apoptotic pathways for cancer treatment [88,89,90,91,92,93,94,95]. Prunin plays a crucial role in inhibiting eukaryotic translation by activating different kinases [96]. Prunin also plays a key role in scavenging ROS and inhibits many pathways like NF-κB, MAPK, STAT1, and the replication of many viruses inside the body [97,98,99,100,101]. These compounds can also inhibit the production of pro-inflammatory cytokines, which contribute to immune homeostasis [102,103]. Moreover, Prunin has substantial properties in enhancing the function of immune cells and supporting their role in both types of immunity—innate and adaptive [104]. Their therapeutic potential and significant effect on the body make them valuable candidates for immune-related interventions [104].
Plant-derived immunotherapies have emerged as a significant area of interest in modern medical research, mainly due to their potential to modulate immune responses with fewer side effects than conventional treatments [105,106]. Key findings highlight the ability of natural compounds, such as prunin, to activate both the innate and adaptive immune systems [17,107]. This compound has demonstrated capabilities in reducing inflammation, enhancing immune cell activity, and promoting antioxidant defenses, making it valuable in treating various conditions, including cancer, infections, and autoimmune disorders [108,109].
One of the most prominent challenges identified is the poor bioavailability of these plant-derived compounds [3]. Though highly active in vitro, many natural compounds are poorly absorbed in the human body, rapidly metabolized, and excreted, limiting their therapeutic potential [110]. This has led researchers to focus on innovative delivery systems like nanoparticles, liposomal encapsulation, and other advanced formulations to improve absorption, prolong circulation time, and enhance overall effectiveness [111,112]. These advancements could transform plant-derived immunotherapies from promising experimental treatments to widely used clinical options [112]. Synergy with existing treatments is another crucial finding. Plant-derived compounds can improve therapeutic outcomes when combined with conventional therapies, particularly immune checkpoint inhibitors (ICIs) [112]. These natural compounds can modulate the tumor microenvironment (TME), reduce inflammation, and enhance immune activation, making ICIs more effective and possibly lowering their side effects [113,114,115,116]. Such combinations could lead to more comprehensive and less toxic cancer treatments. Toxicological concerns are also critical. Although plant-based therapies are generally considered safer, certain compounds can cause adverse effects at high doses or with prolonged use [117,118,119]. For example, saponins may induce hemolysis, and excessive immune stimulation by some plant compounds could trigger autoimmunity or other harmful effects.
The key findings highlight the potential and the challenges associated with plant-derived immunotherapies. While the therapeutic benefits are evident, especially regarding immunomodulation and cancer treatment, additional research is necessary to refine delivery methods, guarantee safety, and comprehensively understand the underlying mechanisms. Addressing these challenges and advanced research will be essential for successfully integrating plant-derived therapies into mainstream medicine [120,121]. By exploring these mechanisms, the review aims to explain how prunin combats oxidative stress and modulates inflammation, two critical factors that contribute to cancer development. In addition to discussing prunin’s biological activities, this review summarizes preclinical studies investigating its anticancer efficacy in various cellular and animal models. Plant-derived compounds have shown immense potential in cancer treatment, offering a complementary approach to traditional therapies such as chemotherapy, radiation, and immune ICIs [122]. With their natural origins and bioactive properties, plant-based compounds can enhance the body’s immune response, modulate the tumor microenvironment, and reduce the toxic side effects of conventional treatments [123]. This has made them an area of growing interest in cancer therapy research. One of the key potential impacts of plant-derived compounds in cancer treatment is their ability to modulate the immune system. A compound such as prunin is known for its immunomodulatory effects, which can boost innate and adaptive immune responses [124]. This compound can activate immune cells, such as T-cells, natural killer (NK) cells, and macrophages, playing crucial roles in recognizing and destroying cancer cells [125,126,127,128]. Plant-based compounds can help the body target and eliminate cancer by enhancing immune surveillance and response [129,130,131].
Another significant impact of plant-derived compounds is their ability to act as adjuvants, enhancing the effectiveness of existing cancer therapies, particularly ICIs [132]. ICIs have revolutionized cancer treatment by removing the inhibitory signals that prevent the immune system from attacking tumors [132,133]. However, not all patients respond to ICIs, and many develop resistance [134,135,136,137]. Research suggests that combining ICIs with plant-derived compounds can improve the efficacy of these treatments by altering the tumor microenvironment, reducing inflammation, and enhancing immune activation [138]. This combination approach may increase the proportion of patients who benefit from ICIs and help overcome resistance. Furthermore, plant-derived compounds such as prunin can reduce the toxicity of conventional cancer therapies. Chemotherapy and radiation often cause severe side effects, including damage to healthy cells, immune suppression, and systemic toxicity [139]. With their antioxidant and anti-inflammatory properties, plant-based compounds may help mitigate these side effects [140,141,142,143]. For example, curcumin has been shown to protect normal cells from oxidative damage during chemotherapy [144], reducing its harmful effects without compromising its efficacy against cancer cells [145,146]. This ability to minimize treatment-related toxicity could improve patients’ quality of life and allow for more aggressive cancer treatment strategies without overwhelming the body.
In summary, integrating plant-derived compound prunin into cancer treatment holds considerable promise. By enhancing immune responses, improving the efficacy of existing therapies, and reducing treatment-related toxicity, these natural compounds could significantly impact how cancer is treated. Future research and clinical trials will be essential in realizing their full potential and optimizing their use in comprehensive cancer care strategies. These studies are necessary to understand prunin’s potential to target and inhibit tumor growth. To the best of our knowledge, there are currently no comprehensive reviews that specifically examine prunin’s role as an anticancer agent. This review serves as the first in-depth examination of the mechanisms through which prunin exerts its therapeutic effects, such as its capacity to modify key signaling pathways, trigger apoptosis, and hinder tumor development. By consolidating and evaluating the existing evidence, this review seeks to address a significant gap in the literature and lay the groundwork for future investigations into prunin’s potential in cancer treatment. Additionally, the review evaluates the current state of clinical research on prunin, assessing its viability as a chemo-preventive or therapeutic agent in cancer treatment. Finally, the review addresses the challenges in translating prunin’s preclinical promise into clinical application, including its pharmacokinetics, bioavailability, and the need for further clinical trials to establish its safety and efficacy. By the end of this review, we aim to provide a comprehensive framework for understanding prunin’s therapeutic potential and outline the future directions for its study in cancer therapy.

2. Therapeutic Potential of Prunin and Mechanism of Action in Cancer

Prunin has garnered significant interest due to its anticancer potential, mainly because it impacts various biological pathways involved in oncogenesis [147,148]. These pathways regulate essential processes such as cell growth, cell cycle regulation, cell death, angiogenesis, and metastasis—crucial factors in cancer [149]. By regulating various pathways, prunin suppresses cancer growth and induces apoptosis. Its multifaceted action underscores its broad-spectrum effects, offering a deeper understanding of its capability to combat cancer at a molecular level [150]. Figure 2 demonstrates that prunin induces apoptosis by influencing multiple molecular targets involved in cellular and receptor-mediated cell death pathways [151]. Thus, targeting these apoptotic pathways offers a hopeful strategy for fighting cancer.

2.1. Effect of Prunin on Cell Cycle

One of the primary ways the prunin exerts its anticancer effects is by influencing the cell cycle [152,153], as shown in Figure 3. The cell cycle is a regulated process that includes several checkpoints (G1/S, G2/M, and M/G1 checkpoints) and regulatory proteins that ensure proper cell division [154,155]. Cell cycle dysregulation can lead to uncontrolled cell growth and eventually induce cancers [154,155]. On the other hand, the arrest of the cell cycle can be used to inhibit cancers [156]. Prunin impacts cell cycle regulation through interactions with various cyclin-dependent kinases (CDKs) and cyclins, as illustrated in Figure 3. Prunin has been demonstrated to influence crucial cell cycle regulators, including cyclins and CDKs [157,158]. Cyclins are essential proteins that regulate CDKs, which phosphorylate target proteins to advance the cell cycle [158]. Excessive cyclin and CDK expression lead to uncontrollable cell division, frequently seen in cancers [159]. Prunin alters the expression levels of several cyclins, especially cyclin D1 and cyclin E, along with their related CDKs, like CDK4 and CDK2 (see Figure 3). By inhibiting these cyclins and CDKs, prunin effectively triggers cell cycle arrests at vital checkpoints, suppressing cancer cells or initiating apoptosis [160]. The G1/S and G2/M phases serve as essential checkpoints in the cell cycle, where the cell primarily determines whether to continue with DNA replication or proceed to mitosis. Prunin has the capability to induce cell cycle arrest at these two points [161,162,163]. During the G1/S phase, prunin can inhibit cyclin D-CDK4 complex activity, activating the retinoblastoma protein (pRb) [164,165]. This activation stops the cell from moving into the S phase. In the G2/M phase, prunin can impact cyclin B-CDK1 complex activity, blocking the cells from beginning mitosis [152]. This dual-phase cell cycle arrest at G1/S and G2/M stages caused by prunin effectively decreases or inhibits tumor growth.

2.2. Intrinsic and Extrinsic Pathways

Cellular stresses like DNA damage or oxidative stress can introduce the intrinsic apoptosis pathway [169], releasing pro-apoptotic proteins from mitochondria. Prunin activates the intrinsic pathway by enhancing mitochondrial membrane permeability, eventually triggering the release of cytochrome c and introducing caspase cascades [170], as illustrated in Figure 4. Prunin can affect intrinsic and extrinsic apoptotic pathways to manage cancer cell death effectively [170]. It upregulates the expression of BAX and BAK genes while downregulating BCL-2, BCL-XL, and MCL1 in the intrinsic pathway [171,172]. Prunin can promote autophagy by blocking the AKT/mTOR/p70S6K pathway, inhibit the development of ovarian cancer cells by activating PARP-1 and caspase-9, and induce apoptosis to treat malignancies [173].
Like other flavonoids, prunin can prevent cell necrosis mainly by lowering mitochondrial ROS production, preserving ATP levels, preventing oxidative harm, and releasing mitochondrial DNA [176]. In contrast, the extrinsic pathway is initiated by activating death receptors on the cell’s surface [177]. Prunin boosts the expression of death receptors like Fas and TRAIL (TNF-related apoptosis-inducing ligand) [176,178,179], thus triggering the extrinsic apoptotic pathway (Figure 4). Caspases are a group of cysteine proteases essential for the process of apoptosis [180]. Prunin stimulates the activation of several caspases, such as caspase-3, caspase-8, and caspase-9, resulting in the cleavage of crucial cellular pathways and the elevation of cell apoptosis [181]. This modulation of the cascade by prunin ultimately causes cell death while inhibiting tumor progression. Conversely, a key mechanism of mitochondria-independent, or extrinsic, apoptosis induction by prunin involves its effects on death receptors [181]. These receptors are essential for transmitting apoptotic signals from the cell membrane to the cytoplasmic signaling pathways [182]. Prunin effectively activates Nrf2, initiating antioxidant genes, and offers protection against oxidative stress in hepatocytes [182]. Prunin may protect against liver oxidative injury by activating Nrf2 and boosting cellular antioxidant responses [66].

2.3. The PI3K/Akt/mTOR Pathway

The PI3K/Akt/mTOR-signaling pathway is crucial and plays a key role in regulating various cellular processes [183,184]. Key proteins involved in this pathway include PI3K and AKT, which are vital to its signaling mechanisms [185,186,187]. Dysregulation of the PI3K/Akt/mTOR-signaling pathway is associated with malignancy initiation, growth, and progression [188,189,190]. Furthermore, PTEN acts as a significant negative regulator by transforming PIP3 back into PIP2, which helps inhibit the activation of the PI3K/Akt/mTOR pathway [189]. Targeting a single pathway like PI3K often leads to toxic side effects in patients. Therefore, the inhibition of the PTR pathway for oncological treatment is greatly improved by using combination techniques. For example, combining natural compounds may reduce toxicity and offer an intriguing approach for more effective targeted cancer treatments [189]. Prunin is a prospective therapeutic agent for cancer treatment since it directly targets essential components of the PI3K/Akt/mTOR pathway [191]. Autophagy is a vital cellular mechanism that breaks down damaged proteins and organelles to preserve stability [20].
In cancers, autophagy can be activated by deregulating the PI3K/Akt/mTOR pathway, permitting tumor cells to survive in environments lacking nutrients [191]. This adaptive mechanism is essential for cancer development and therapy resistance. Anticancer therapies aimed at the PI3K/Akt/mTOR pathway can promote autophagy, which inhibits mTOR, a crucial regulator [191]. mTORC1 suppresses autophagy and lysosomal activity by phosphorylating crucial proteins like ULK1, which is involved in autophagy regulation, and TFEB, which controls lysosomal gene expression. The activity of mTORC1 is influenced by energy status, hypoxia, and other conditions, affecting autophagy through the AMPK/TSC pathway [192]. Prunin can promote autophagy by inhibiting the PI3K/mTOR-signaling molecules [186,193,194,195]. Prunin blocks Akt/mTOR signaling, causing both autophagy and apoptosis [196]. Moreover, prunin can decrease LC3II levels and partially restore p62 degradation in MN9D cells, which controls autophagy.

2.4. Antioxidant and Anti-Inflammatory Mechanisms

Prunin’s antioxidant activity is a key feature that makes it a promising agent against diseases related to oxidative stress, such as cancer [162]. By scavenging ROS and reactive nitrogen species (RNS), prunin helps protect cellular components like DNA, proteins, and lipids from oxidative damage. This protection is essential for cancer prevention, as the accumulation of oxidative damage can lead to genetic mutations and promote the onset of cancer [34]. Prunin’s antioxidant characteristics are mainly attributed to its capability to donate electrons, neutralize free radicals, and prevent cellular damage to preserve cellular homeostasis [197,198]. A recent study by Zhang et al. showed that the prunin isolated from Bauhinia variegate induces a protective effect against diet-induced atherosclerosis by reducing the levels of proinflammatory mediators such as TNF-α and IL-6 [198]. In addition to its antioxidant characteristics, prunin exerts significant anti-inflammatory effects [199]. Chronic inflammation plays a key role in cancer progression, and prunin’s ability to modulate inflammatory pathways is essential in mitigating this risk. One of the significant anti-inflammatory mechanisms of prunin is its inhibition of the NF-κB-signaling pathway, which is generally triggered in several cancers. NF-κB serves as a transcription factor that governs the expression of pro-inflammatory cytokines, chemokines, and adhesion molecules, all of which play a significant role in tumor promotion [200,201,202]. By preventing NF-κB activation, prunin decreases the production of these inflammatory mediators, thereby preventing tumor progression and metastasis processes [203]. Furthermore, pruning modulates various inflammatory pathways, including MAPK (mitogen-activated protein kinase) and COX-2 (cyclooxygenase-2), further contributing to its anti-inflammatory effects [204,205,206].
Prunin decreases ROS and RNS levels by functioning as a direct antioxidant [207]. Prunin efficiently scavenges harmful radicals like superoxide, hydroxyl radicals, and hydrogen peroxide by donating electrons to neutralize them [208,209]. This phenomenon transforms highly reactive species into more stable and less harmful molecules, reducing oxidative stress. By decreasing ROS and RNS, prunin protects cells from oxidative damage. Furthermore, prunin improves the phosphorylation of key signaling molecules, indicating its protective role against oxidative stress [207]. Prunin is a therapeutic agent for improving glucose homeostasis and is involved in glucose uptake [61]. In addition to prunin’s direct scavenging capability, it also triggers the action of several key endogenous antioxidant enzymes, such as superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT) [61,210]. These endogenous antioxidant enzymes are essential in preserving cellular redox balance by converting highly reactive ROS into less reactive or bio-friendly molecules [211,212,213].

2.5. Activation of the P53 Pathway by Prunin

Primarily, many cancers are associated with inactivated P53 expression [214]. P53 is crucial in various ways inside cancer cells (Figure 5). The main biological functions of P53 in cancer cells include triggering apoptosis, regulating cellular senescence, inhibiting angiogenesis, controlling the cell cycle, modulating cellular differentiation, and maintaining DNA metabolism [215]. Within tumor cells, P53 acts as a molecular sensor that inhibits cell proliferation in response to detrimental stimulation. P53 induces apoptosis by activating the Bax gene (a key member of the Bcl-2 family) through the mitochondrial intrinsic pathway [215]. In the intrinsic pathway, Bax binds to Bcl-2, thereby activating the production of apoptotic mediators (e.g., caspase 3/9 and cytochrome C) through mitochondrial activity [215]. Thus, plant flavonoid prunin may be a valuable source to target Bcl-2 through p53, offering an efficient means for combating cancer [216]. Prunin could activate the ATM/ATR pathway by inducing DNA damage inside cells through oxidative stress or cellular homeostasis disruption, generating DNA damage signals and leading to cell death as ATR/ATM is linked with CHK1 and CHK2 cell cycle checkpoints that can be triggered through prunin, which leads these kinases to phosphorylate downstream effectors, including checkpoint kinases CHK1 and CHK2 [217,218,219,220,221]. Subsequently, it will phosphorylate the tumor-suppressor protein P53. Phosphorylation of P53 stabilizes it by avoiding its degradation through MDM2 inhibition, permitting its initiation [222]. P53 coordinates a multifaceted cellular response, promoting cell cycle arrest to allow DNA repair [223]. Purine activity inside cells induces oxidative stress, which leads to apoptosis if the damage is acute and starts cellular senescence to prevent the proliferation of damaged cells. Prunin treatment could potentially increase the expression of death receptors Fas Ligand (FasL) protein in human gastric cancer cells [223]. Additionally, it induced the activation of caspase-8 and caspase-3 and the cleavage of PARP after P53 signaling through the intrinsic pathway [223]. By initiating this pathway, purine increases genomic integrity and exerts its anticancer effects, indicating its therapeutic potential as a modulator of the DNA damage response and tumor-suppressor pathways [224,225].

2.6. Activation of MAPK Pathway by Prunin

Prunin, as a potential flavonoid compound, has gained significant attention due to its potential for biomedical applications for future research [22,32,46,229,230]. Prunin could be a candidate for molecular studies that modulate the mitogen-activated protein kinase (MAPK) pathway, which plays a vital role in the signaling cascade for cellular responses to external stimuli, including oxidative stress, inflammation, and oncogenic signals [231,232,233]. Prunin treatment could increase the phosphorylation and activation of MAPK and act as an anticancer agent by exhibiting pro-apoptotic activities inside cells [234]. There are three key hallmarks in the MAPK pathway, which play different roles according to situations. It comprises (subfamilies) such as ERK (extracellular signal-regulated kinase), JNK (c-Jun N-terminal kinase), and p38, which play a central role in regulating cell functions such as proliferation, apoptosis, differentiation, and stress responses [235,236,237].
In the cells, various transport systems available for prunin could enter inside cells through active or passive transport, which interacts with membrane receptors such as receptor tyrosine kinases (RTKs) or G-protein coupled receptors (GPCRs) upon cellular uptake [238]. This interaction stimulated a cascade of phosphorylation actions, stimulating Ras, a small GTPase protein. This process is conducted through stimulated Ras, which recruits and activates Raf, a serine/threonine kinase. Raf phosphorylates and activates MEK1/2, which in turn phosphorylates and activates ERK1/2 [239]. Further, ERK1/2 translocates to the nucleus, influencing numerous transcription factors and ultimately driving gene expression in cell survival, proliferation, repair processes, and apoptosis according to the signal [239]. Some experimental data show that using different plant flavonoid compounds further demonstrates that they suppress cell growth and migration and induce apoptosis via MAPK/mTOR pathway using combination treatment [240]. Prunin could inhibit the proliferation and migration of cancer cells, possibly by downregulating MAPK14 expression, which is linked to poor prognosis [241]. Therefore, plant flavonoids could be potential therapeutic drugs stemming from their ability to modulate the MAPK pathway, thereby reducing tumor growth and metastasis [242].
In addition to ERK activation, prunin could influence the JNK and p38 MAPK branches, which leads to cell death signaling. Under oxidative stress conditions, prunin mitigates ROS production, stabilizing the intracellular redox balance [243,244]. This antioxidant effect curtails the overactivation of JNK and p38 pathways in cells, avoiding excessive inflammatory responses and apoptosis. Interestingly, prunin could selectively increase JNK and p38 activity in cancer cells, endorsing pro-apoptotic signaling. Prunin accomplishes this by inducing the expression of upstream kinases, which phosphorylate JNK and p38 after treatment [245]. The selective cytotoxicity that prunin could exhibit against malignant cells underscores its therapeutic potential as an anticancer agent. It suggests that prunin could be a potential bioactive compound on the MAPK pathway [245].
Beyond prunin’s antioxidant and apoptotic roles, which play vital roles inside cells that activate several signaling pathways, it exerts potent anti-inflammatory effects by regulating MAPK-driven cytokine production [246,247]. By reducing the activation of NF-κB (a downstream effector of the MAPK pathway), prunin could inhibit the transcription of pro-inflammatory cytokines such as TNF-α, IL-6, and IL-1β [248]. This dual inhibition of MAPK and NF-κB signaling by prunin alleviates inflammation and decreases the tumor-facilitating microenvironment [169,249]. The anticancer properties of prunin are further augmented by its capability to cause cell cycle arrest and apoptosis via MAPK-mediated pathways. For instance, activating JNK by prunin improves the phosphorylation of p53, a tumor-suppressor protein that leads to the transcription of pro-apoptotic genes like BAX and PUMA [248]. Simultaneously, prunin could disrupt the phosphorylation of BCL-2, an anti-apoptotic protein, tilting the cancerous cells’ balance toward programmed cell death [250,251,252,253]. Thus, prunin, a plant bioactive compound, could be a multifaceted regulator of the MAPK pathway, showing its antioxidant, anti-inflammatory, and anticancer potential to modulate various cellular functions [254]. By targeting specific branches of the MAPK cascade context-dependently, prunin displays notable potential as a therapeutic bioactive compound for dealing with oxidative stress, chronic inflammation, and tumor prevention. Further research might be required to unravel its precise molecular interactions and optimize its clinical applications for real applications.

2.7. Modulation of Tumor Microenvironment (TME)

Like other flavonoids, prunin can modulate immune cell influx in the TME [255,256,257]. Research shows it aids in the recruitment of tumor-suppressor cells, such as NK cells and cytotoxic T lymphocytes, while inhibiting the influx of pro-tumorigenic immune cells like tumor-associated macrophages [258,259,260,261]. This immune modulation contributes to the anti-tumor effects of prunin, supporting the body’s natural defense mechanisms against cancer development [262]. The immunomodulatory effects of isorhamnetin on the innate and adaptive immune responses are shown in Figure 6. Prunin boosts the innate immune response by stimulating various immune cells, such as NK cells, macrophages, neutrophils, eosinophils, basophils, and mast cells [262]. This stimulation promotes immune cell infiltration, enhances phagocytosis, and increases NK cell-mediated cytotoxicity, culminating in the apoptosis and death of cancer cells [263,264]. Simultaneously, prunin stimulates adaptive immunity by modulating antigen-presenting cells, B cells, and T cells [265,266]. Enhanced T cell activity promotes cancer cell death through effector mechanisms involving perforin, granzyme B, interferon-gamma, and tumor necrosis factor-alpha [267,268,269,270]. Furthermore, pruning enhances the humoral immune response, boosting antibody production for a strong immune defense [271,272].

2.8. Suppression of Angiogenesis and Metastasis by Prunin

Metastasis is a complex procedure that comprises the transport of tumor cells from the primary site to secondary lesions [273]. Angiogenesis is one of the key mechanisms that support cancer progression processes [273,274]. Flavonoids have shown their potential to disrupt these key cancerous phenomena (angiogenesis and metastasis) and restrict their survival and spread [275,276,277]. Vascular endothelial growth factor (VEGF) is a key pro-angiogenic factor that promotes the development of new blood vessels, which enhances the supply of oxygen and nutrients, subsequently contributing to cancer growth [278]. VEGF enhances angiogenesis by stimulating the development of new blood vessels, which are crucial for delivering oxygen and nutrients essential for tumor growth and aiding metastasis [279,280,281,282]. The inhibition of VEGF expression can restrict the formation of new blood vessels and limit the supply of oxygen and necessary nutrients for cancer cell survival, eventually leading to cell death [283]. The prunin can inhibit VEGF expression, suppressing angiogenesis and tumor vascularization [284]. Prunin inhibits cancer growth and metastases by reducing the expression of VEGF and MMP-2 while increasing endostatin levels, an angiogenesis inhibitor [285,286]. Prunin also elevates immune markers IL-2 and IFN-γ, indicating an improved immune response. Additionally, prunin reduces the expression levels of MMP-9 without cytotoxic effects, suggesting it could be an essential natural antioxidant and MMP inhibitor related to oxidative stress [287,288]. The prunin significantly reduced EMT (epithelial-mesenchymal transition) and lowered STAT3, a transcription factor that promotes tumor invasion, thereby inhibiting blood vessel formation [289,290]. Prunin inhibits several transcription factors crucial for regulating immune cells’ differentiation, proliferation, and activation while promoting T-cell generation [291]. This compound influences various immune system processes and holds potential for therapeutic applications [292].
Angiogenesis is one of the most important factors for cancer progression. Several angiogenic proteins include VEGF, essential fibroblast growth factor, IL8, and TGF-β, whereas anti-angiogenic factors include thrombospondin-1, angiostatin, and endostatin. Flavonoids such as prunin act as effective angiogenesis inhibitors and are a promising treatment for controlling malignancies [293]. Prunin exhibits its angio-inhibitory effect by decreasing VEGF and other related factors. It has also been reported to cause downregulation of the TGF-β pathway, thereby reducing metastasis and invasion [227,234]. Inflammation is a hallmark of cancer progression as the immune system plays a crucial role in combating cancers, and its chronic activation contributes to tumor growth, metastasis, and resistance to chemotherapy. Prunin’s ability to modify inflammatory mediators and signaling molecules makes it a promising candidate for cancer therapy. Additionally, prunin has demonstrated anticancer activity in various in vitro and in vivo models by inducing cell death in cancer cells [294]. It has been found to modify the expression of key proteins involved in apoptosis, such as caspases, Bcl-2 family members, and p53. Through these mechanisms, prunin may help to eliminate transformed cells while sparing normal, healthy cells [295]. Due to these promising properties, prunin is gaining recognition as a potential chemo-preventive agent. However, further clinical studies are necessary to fully establish its efficacy and safety as a therapeutic agent in cancer treatment.

3. Potential for Combining Prunin with Conventional Therapies

Research on flavonoid’s role in improving chemotherapy effectiveness is crucial in cancer studies [296,297]. While chemotherapy is essential for cancer treatment, it often results in side effects and drug-resistance problems [298,299,300]. The flavonoids, especially prunin, exhibit antioxidant, anti-inflammatory, and apoptotic properties that may allow them to enhance chemotherapy outcomes [301]. Prunin can make cancer cells more sensitive to chemotherapy-induced death by regulating several signaling pathways related to chemoresistance (Figure 7) [302]. For example, prunin inhibits the PI3K/Akt/mTOR pathway and promotes apoptosis in cancer cells, which could boost the cytotoxicity of chemotherapeutic drugs [303,304]. Moreover, prunin’s antioxidant characteristics might alleviate the oxidative damage that chemotherapy drugs inflict, protecting healthy cells while enabling cancer cell death. As such, combining prunin with chemotherapy might enhance treatment effectiveness, lower required dosages, and reduce the side effects typically associated with chemotherapy.
Radiotherapy is a common treatment for cancer that uses ionizing radiation to destroy or damage cancer cells [305]. Although it is effective, this therapy can also harm surrounding healthy tissues, resulting in side effects such as fatigue, skin reactions, and immunosuppression [305]. Prunin, known for its antioxidant properties [22,199], might be a beneficial supplement to radiotherapy by enhancing its effectiveness while minimizing harm to normal tissues. By neutralizing free radicals produced during radiation treatment, prunin can help protect healthy cells from oxidative stress caused by radiation, potentially reducing side effects and improving patients’ quality of life [306]. Additionally, prunin can promote apoptosis in cancer cells, increasing their susceptibility to radiation-induced cell death and enhancing treatment efficacy [307,308,309]. Research indicates that flavonoids like prunin can enhance the cytotoxicity of radiation on cancer cells by inducing DNA damage and impairing repair processes, thereby improving the overall success of radiotherapy [234]. In conclusion, when combined with standard cancer treatments like chemotherapy and radiotherapy, prunin’s synergistic effects can boost treatment effectiveness while minimizing side effects and addressing resistance mechanisms. Its diverse actions—antioxidant, apoptotic, and signaling modulation—position prunin as a promising choice for combination therapies to enhance the overall success of cancer treatment.
Figure 7. Overview of prunin for combination treatment and nanoformulation for improved drug delivery. Prunin can be utilized alongside radiation, nanoparticles, various natural compounds, and anticancer chemotherapy drugs to achieve a synergistic effect, helping to reduce drug resistance and enhance cell activity and death. For drug delivery, the nanoformulation increases the bioavailability, solubility, stability, target delivery, and overall efficiency of the prunin [310,311,312]. The figure was prepared using Biorender.
Figure 7. Overview of prunin for combination treatment and nanoformulation for improved drug delivery. Prunin can be utilized alongside radiation, nanoparticles, various natural compounds, and anticancer chemotherapy drugs to achieve a synergistic effect, helping to reduce drug resistance and enhance cell activity and death. For drug delivery, the nanoformulation increases the bioavailability, solubility, stability, target delivery, and overall efficiency of the prunin [310,311,312]. The figure was prepared using Biorender.
Ijms 26 02678 g007

4. Nanotechnology and Drug Delivery Systems for Prunin

One significant research area in harnessing prunin’s therapeutic potential is its increase its bioavailability [313]. As a flavonoid glycoside, prunin’s absorption is restricted due to its low solubility and essential first-pass metabolism [314,315,316,317]. To address these challenges, nanotechnology offers innovative solutions by developing nanoformulations that improve prunin’s solubility, stability, and bioavailability (Figure 7) [313,318,319]. Nanoparticles like liposomes, solid lipid nanoparticles, and mesoporouss can be tailored to encapsulate prunin, protecting from degradation and easing its absorption in the gastrointestinal tract [320,321,322,323]. These nanoformulations can be formulated for controlled release, assisting in maintaining therapeutic concentrations for a long time [324,325,326,327]. Additionally, by improving prunin’s solubility and stability, these formulations can increase tissue penetration, permitting the compound to reach target sites more efficiently [328,329,330,331]. Leveraging nanotechnology to optimize prunin’s pharmacokinetics could notably boost its potential as a potent therapeutic agent for cancer treatment [332,333,334,335]. Furthermore, targeted drug delivery is a cutting-edge approach that aims to deliver therapeutic agents precisely to tumor cells, diminishing damage to normal tissues and improving treatment outcomes [336,337,338,339]. For prunin, targeted delivery systems using nanotechnology could be a breakthrough in increasing its anticancer impacts while decreasing the risk of systemic toxicity [340,341,342,343].
Beyond receptor-mediated targeting, nanoparticles can also be formulated to exploit the enhanced permeability and retention (EPR) effect, where they accumulate in tumor tissue because of the unique vasculature in tumors [344,345,346,347]. This passive targeting approach further elevates prunin concentration at the tumor site, enhancing its anticancer effectiveness [348]. By integrating active and passive targeting methods, prunin can be delivered more efficiently to cancer cells, boosting its therapeutic impact while reducing off-target effects. Creating these targeted delivery systems offers a promising pathway for enhancing the clinical use of prunin in cancer therapeutics [348]. Future research in the delivery of prunin should focus on developing multifunctional nanoparticles that improve bioavailability and targeting and allow for the simultaneous delivery of other therapeutic agents, such as chemotherapeutic drugs or RNA-based therapies [12,349]. Combining prunin with other conventional treatment agents could produce a synergistic effect, improving overall treatment methodology [350]. Advances in nanomedicine, such as using stimuli-responsive nanoparticles that release their payload in response to specific triggers (e.g., pH, temperature, or light), could also offer new ways to control prunin delivery in the TME precisely [351,352,353]. In summary, although nanotechnology shows significant potential for enhancing prunin delivery and boosting its anticancer effects, multiple challenges must be tackled. Ongoing research on designing safe, effective, and scalable nanoformulations and developing targeted delivery systems will be essential for converting prunin’s preclinical potential into successful clinical treatments.

5. Conclusions and Future Perspectives

5.1. Summary of Key Findings

Prunin has significant potential as an anticancer natural compound due to its potential to influence various biological activities, including antioxidant, anti-inflammatory, anti-angiogenesis, anti-metastasis, and pro-apoptotic activities. Prunin effectively modulates key cellular signaling pathways in cancer cell survival, proliferation, and metastasis, making it a capable candidate for cancer treatment and prevention. Remarkably, prunin inhibits the proliferation of cancer cells by regulating cyclins, while CDKs induce apoptosis through caspase activation and mitochondrial dysfunction. Prunin also inhibits metastatic progression by downregulating MMPs, VEGF, and EMT markers. Furthermore, its antioxidant and anti-inflammatory characteristics mitigate endogenous ROS and pro-inflammatory cytokines, inhibiting cancer progression and regulating the tumor microenvironment. Prunin is also a potential candidate as an adjunct to available traditional therapies, increasing their efficacy and overcoming resistance mechanisms. Advances in nanotechnology and targeted drug delivery schemes might further improve its therapeutic potential by improving bioavailability, solubility, and stability.

5.2. Limitations and Future Perspectives

Despite its promise of anticancer potential, numerous limitations hinder Prunin’s clinical translation. Current research lacks comprehensive in vivo studies, and toxicological data remain insufficient, restricting complete understanding. The detailed mechanisms and precise molecular targets have not been thoroughly studied, and there is a limited exploration of their efficacy in combination therapies. Few therapeutic targets were explored; no modern drug delivery systems were tested to improve bioavailability and targeted therapy. These research gaps highlight the need for more preclinical investigations.
Future studies should address these limitations through well-designed in vivo studies, detailed toxicological assessments, and underlying mechanistic studies to identify prunin’s molecular targets. Integrating artificial intelligence (AI) and machine-learning technology into prunin research could accelerate drug discovery by predicting optimized derivatives, classifying novel targets, and enhancing combination therapies. Emerging nanoformulations and personalized medicine methods will also increase prunin’s clinical applicability. Collaborative efforts between scientists, clinicians, and pharmaceutical developers are essential to translate prunin from a promising natural compound into a clinically approved therapeutic. Addressing bioavailability, scalability, and regulatory challenges is necessary for unlocking Prunin’s full anticancer potential.

Author Contributions

Conceptualization, J.N.R. and S.M.; methodology, J.N.R. and S.M.; software, J.N.R. and S.M.; validation, J.N.R. and S.M.; formal analysis, J.N.R. and S.M.; investigation, J.N.R. and S.M.; data curation, J.N.R. and S.M.; writing—original draft preparation, J.N.R. and S.M.; writing—review and editing, J.N.R. and S.M.; visualization, J.N.R. and S.M.; supervision, S.M.; project administration, S.M.; funding acquisition, S.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
ROSReactive oxygen species
ICIsImmune checkpoint inhibitors
NKNatural killer
VEGFVascular endothelial growth factor
CDKCyclin-dependent kinase
RNSReactive nitrogen species
SODSuperoxide dismutase
CATCatalase
GPxGlutathione peroxidase
MAPKMitogen-activated protein kinase
ERKExtracellular signal-regulated kinase
JNKc-Jun N-terminal kinase
RTKsReceptor tyrosine kinases
GPCRsG-protein coupled receptors
EPREnhanced permeability and retention
AIArtificial intelligence
TMETumor microenvironment

References

  1. Nabavi, S.M.; Šamec, D.; Tomczyk, M.; Milella, L.; Russo, D.; Habtemariam, S.; Suntar, I.; Rastrelli, L.; Daglia, M.; Xiao, J.; et al. Flavonoid Biosynthetic Pathways in Plants: Versatile Targets for Metabolic Engineering. Biotechnol. Adv. 2020, 38, 107316. [Google Scholar] [CrossRef] [PubMed]
  2. Jiang, C.-H.; Sun, T.-L.; Xiang, D.-X.; Wei, S.-S.; Li, W.-Q. Anticancer Activity and Mechanism of Xanthohumol: A Prenylated Flavonoid From Hops (Humulus lupulus L.). Front. Pharmacol. 2018, 9, 530. [Google Scholar] [CrossRef]
  3. Kopustinskiene, D.M.; Jakstas, V.; Savickas, A.; Bernatoniene, J. Flavonoids as Anticancer Agents. Nutrients 2020, 12, 457. [Google Scholar] [CrossRef] [PubMed]
  4. Redondo-Blanco, S.; Fernández, J.; López-Ibáñez, S.; Miguélez, E.M.; Villar, C.J.; Lombó, F. Plant Phytochemicals in Food Preservation: Antifungal Bioactivity: A Review. J. Food Prot. 2020, 83, 163–171. [Google Scholar] [CrossRef]
  5. Tzanova, M.; Atanasov, V.; Yaneva, Z.; Ivanova, D.; Dinev, T. Selectivity of Current Extraction Techniques for Flavonoids from Plant Materials. Processes 2020, 8, 1222. [Google Scholar] [CrossRef]
  6. Sun, D.; Li, X.; Nie, S.; Liu, J.; Wang, S. Disorders of Cancer Metabolism: The Therapeutic Potential of Cannabinoids. Biomed. Pharmacother. 2023, 157, 113993. [Google Scholar] [CrossRef] [PubMed]
  7. Julkunen-Tiitto, R.; Nenadis, N.; Neugart, S.; Robson, M.; Agati, G.; Vepsäläinen, J.; Zipoli, G.; Nybakken, L.; Winkler, B.; Jansen, M.A.K. Assessing the Response of Plant Flavonoids to UV Radiation: An Overview of Appropriate Techniques. Phytochem. Rev. 2015, 14, 273–297. [Google Scholar] [CrossRef]
  8. Agati, G.; Azzarello, E.; Pollastri, S.; Tattini, M. Flavonoids as Antioxidants in Plants: Location and Functional Significance. Plant Sci. 2012, 196, 67–76. [Google Scholar] [CrossRef]
  9. Dias, M.C.; Pinto, D.C.G.A.; Silva, A.M.S. Plant Flavonoids: Chemical Characteristics and Biological Activity. Molecules 2021, 26, 5377. [Google Scholar] [CrossRef]
  10. Rahimi, R.; Ghiasi, S.; Azimi, H.; Fakhari, S.; Abdollahi, M. A Review of the Herbal Phosphodiesterase Inhibitors; Future Perspective of New Drugs. Cytokine 2010, 49, 123–129. [Google Scholar] [CrossRef]
  11. Ginwala, R.; Bhavsar, R.; Chigbu, D.G.I.; Jain, P.; Khan, Z.K. Potential Role of Flavonoids in Treating Chronic Inflammatory Diseases with a Special Focus on the Anti-Inflammatory Activity of Apigenin. Antioxidants 2019, 8, 35. [Google Scholar] [CrossRef] [PubMed]
  12. Frenț, O.-D.; Stefan, L.; Morgovan, C.M.; Duteanu, N.; Dejeu, I.L.; Marian, E.; Vicaș, L.; Manole, F. A Systematic Review: Quercetin—Secondary Metabolite of the Flavonol Class, with Multiple Health Benefits and Low Bioavailability. Int. J. Mol. Sci. 2024, 25, 12091. [Google Scholar] [CrossRef] [PubMed]
  13. Terahara, N. Flavonoids in Foods: A Review. Nat. Prod. Commun. 2015, 10, 1934578X1501000334. [Google Scholar] [CrossRef]
  14. Treutter, D.; Feucht, W.; Schmid, P.P.S. Ageing-Dependent Responses of Phloem Flavonoids of Prunus Avium Graftings: Flavanone-, Flavone- and Isoflavone-Glucosides. Sci. Hortic. 1987, 32, 183–193. [Google Scholar] [CrossRef]
  15. Castillo, J.; Benavente, O.; del Rio, J.A. Hesperetin 7-O-Glucoside and Prunin in Citrus Species (C. aurantium and C. paradisi). A Study of Their Quantitative Distribution in Immature Fruits and as Immediate Precursors of Neohesperidin and Naringin in Citrus Aurantium. J. Agric. Food Chem. 1993, 41, 1920–1924. [Google Scholar] [CrossRef]
  16. Dabeek, W.M.; Marra, M.V. Dietary Quercetin and Kaempferol: Bioavailability and Potential Cardiovascular-Related Bioactivity in Humans. Nutrients 2019, 11, 2288. [Google Scholar] [CrossRef]
  17. Jin, T.; Albillos, S.M.; Guo, F.; Howard, A.; Fu, T.-J.; Kothary, M.H.; Zhang, Y.-Z. Crystal Structure of Prunin-1, a Major Component of the Almond (Prunus dulcis) Allergen Amandin. J. Agric. Food Chem. 2009, 57, 8643–8651. [Google Scholar] [CrossRef]
  18. Abotaleb, M.; Samuel, S.M.; Varghese, E.; Varghese, S.; Kubatka, P.; Liskova, A.; Büsselberg, D. Flavonoids in Cancer and Apoptosis. Cancers 2019, 11, 28. [Google Scholar] [CrossRef]
  19. Safe, S.; Jayaraman, A.; Chapkin, R.S.; Howard, M.; Mohankumar, K.; Shrestha, R. Flavonoids: Structure–Function and Mechanisms of Action and Opportunities for Drug Development. Toxicol. Res. 2021, 37, 147–162. [Google Scholar] [CrossRef]
  20. Jung, H.A.; Ali, M.Y.; Bhakta, H.K.; Min, B.-S.; Choi, J.S. Prunin Is a Highly Potent Flavonoid from Prunus davidiana Stems That Inhibits Protein Tyrosine Phosphatase 1B and Stimulates Glucose Uptake in Insulin-Resistant HepG2 Cells. Arch. Pharm. Res. 2017, 40, 37–48. [Google Scholar] [CrossRef]
  21. Choi, J.S.; Yokozawa, T.; Oura, H. Antihyperlipidemic Effect of Flavonoids from Prunus davidiana. J. Nat. Prod. 1991, 54, 218–224. [Google Scholar] [CrossRef] [PubMed]
  22. Céliz, G.; Audisio, M.C.; Daz, M. Antimicrobial Properties of Prunin, a Citric Flavanone Glucoside, and Its Prunin 6″-O-lauroyl Ester. J. Appl. Microbiol. 2010, 109, 1450–1457. [Google Scholar] [CrossRef] [PubMed]
  23. Harikrishna, D.; Appa Rao, A.V.N.; Prabhakar, M.C. Pharmacological Investigation of Prunin-6″-O-p-Coumarate: A Flavonoid Glycoside. Indian J. Pharmacol. 2004, 36, 244–245. [Google Scholar]
  24. Ayub, H.; Nadeem, M.; Mohsin, M.; Ambreen, S.; Khan, F.A.; Oranab, S.; Rahim, M.A.; Zubair Khalid, M.; Zongo, E.; Zarlasht, M.; et al. A Comprehensive Review on the Availability of Bioactive Compounds, Phytochemicals, and Antioxidant Potential of Plum (Prunus domestica). Int. J. Food Prop. 2023, 26, 2388–2406. [Google Scholar] [CrossRef]
  25. Abraão, A.S.; Fernandes, N.; Silva, A.M.; Domínguez-Perles, R.; Barros, A. Prunus lusitanica L. Fruits as a Novel Source of Bioactive Compounds with Antioxidant Potential: Exploring the Unknown. Antioxidants 2022, 11, 1738. [Google Scholar] [CrossRef]
  26. Berhow, M.A.; Vandercook, C.E. Biosynthesis of Naringin and Prunin in Detached Grapefruit. Phytochemistry 1989, 28, 1627–1630. [Google Scholar] [CrossRef]
  27. Dai, M.; Kang, X.; Wang, Y.; Huang, S.; Guo, Y.; Wang, R.; Chao, N.; Liu, L. Functional Characterization of Flavanone 3-Hydroxylase (F3H) and Its Role in Anthocyanin and Flavonoid Biosynthesis in Mulberry. Molecules 2022, 27, 3341. [Google Scholar] [CrossRef]
  28. Rehan, M. Biosynthesis of Diverse Class Flavonoids via Shikimate and Phenylpropanoid Pathway. In Bioactive Compounds-Biosynthesis, Characterization and Applications; Queiroz Zepka, L., Casagrande Do Nascimento, T., Jacob-Lopes, E., Eds.; IntechOpen: Rijeka, Croatia, 2021; ISBN 978-1-83969-270-3. [Google Scholar]
  29. Liu, Y.; Qian, J.; Li, J.; Xing, M.; Grierson, D.; Sun, C.; Xu, C.; Li, X.; Chen, K. Hydroxylation Decoration Patterns of Flavonoids in Horticultural Crops: Chemistry, Bioactivity, and Biosynthesis. Hortic. Res. 2022, 9, uhab068. [Google Scholar] [CrossRef]
  30. Wu, R.; Qian, C.; Yang, Y.; Liu, Y.; Xu, L.; Zhang, W.; Ou, J. Integrative Transcriptomic and Metabolomic Analyses Reveal the Phenylpropanoid and Flavonoid Biosynthesis of Prunus mume. J. Plant Res. 2024, 137, 95–109. [Google Scholar] [CrossRef]
  31. Waris, M.H.; Muzaffar, N.; Mumtaz, M.A.; Afzal, A.M.; Iqbal, M.W.; Mumtaz, S.; Ali, M.; Alaraidh, I.A.; Okla, M.K.; Munnaf, S.A. High Performance Lanthanum-Doped Nickel Cobalt Ferrites on Titanium Carbide MXene Electrode Material for Superior Hybrid Device and Precision Creatinine Sensing. Appl. Phys. A 2025, 131, 220. [Google Scholar] [CrossRef]
  32. Munekata, P.E.S.; Yilmaz, B.; Pateiro, M.; Kumar, M.; Domínguez, R.; Shariati, M.A.; Hano, C.; Lorenzo, J.M. Valorization of By-Products from Prunus Genus Fruit Processing: Opportunities and Applications. Crit. Rev. Food Sci. Nutr. 2023, 63, 7795–7810. [Google Scholar] [CrossRef] [PubMed]
  33. Zan, S.; Wang, R.; Zhang, F.; Zhang, D.; Liu, B.; Meng, X. Composition Analysis of Rootstock Cherry (Prunus mahaleb L.), a Potential Source of Human Nutrition and Dietary Supplements. Eur. Food Res. Technol. 2022, 248, 1421–1435. [Google Scholar] [CrossRef]
  34. Nunes, A.R.; Gonçalves, A.C.; Falcão, A.; Alves, G.; Silva, L.R. Prunus avium L. (Sweet Cherry) By-Products: A Source of Phenolic Compounds with Antioxidant and Anti-Hyperglycemic Properties—A Review. Appl. Sci. 2021, 11, 8516. [Google Scholar] [CrossRef]
  35. Ortega-Vidal, J.; Cobo, A.; Ortega-Morente, E.; Gálvez, A.; Martínez-Bailén, M.; Salido, S.; Altarejos, J. Antimicrobial Activity of Phenolics Isolated from the Pruning Wood Residue of European Plum (Prunus domestica L.). Ind. Crops Prod. 2022, 176, 114296. [Google Scholar] [CrossRef]
  36. Kumar, A.; Sharma, M.K.; Wani, T.F.; Sharma, A.; Nyorak, G. Varietal Wealth of Prunus Species. In Prunus-Recent Advances; Küden, A.B., Kuden, A., Eds.; IntechOpen: Rijeka, Croatia, 2021; ISBN 978-1-83969-582-7. [Google Scholar]
  37. Vila-Real, H.; Alfaia, A.J.; Calado, A.R.; Ribeiro, M.H.L. Improvement of Activity and Stability of Soluble and Sol–Gel Immobilized Naringinase in Co-Solvent Systems. J. Mol. Catal. B Enzym. 2010, 65, 91–101. [Google Scholar] [CrossRef]
  38. Yang, X.; Jiang, Y.; Yang, J.; He, J.; Sun, J.; Chen, F.; Zhang, M.; Yang, B. Prenylated Flavonoids, Promising Nutraceuticals with Impressive Biological Activities. Trends Food Sci. Technol. 2015, 44, 93–104. [Google Scholar] [CrossRef]
  39. Das, S.; Bhattacharya, A.; Maulik, S.R. Chapter 3-Isolation and Characterization of Natural Dyes and Pigments. In Renewable Dyes and Pigments; Islam, U.S., Ed.; Elsevier: Amsterdam, The Netherlands, 2024; pp. 37–48. ISBN 978-0-443-15213-9. [Google Scholar]
  40. Ekalu, A.; Habila, J.D. Flavonoids: Isolation, Characterization, and Health Benefits. Beni-Suef Univ. J. Basic Appl. Sci. 2020, 9, 45. [Google Scholar] [CrossRef]
  41. Arora, S.; Itankar, P. Extraction, Isolation and Identification of Flavonoid from Chenopodium Album Aerial Parts. J. Tradit. Complement. Med. 2018, 8, 476–482. [Google Scholar] [CrossRef]
  42. Youn, S.H.; Kim, H.J.; Kim, T.H.; Shin, C.S. Lipase-Catalyzed Acylation of Naringin with Palmitic Acid in Highly Concentrated Homogeneous Solutions. J. Mol. Catal. B Enzym. 2007, 46, 26–31. [Google Scholar] [CrossRef]
  43. Lyu, Y.; Zeng, W.; Du, G.; Chen, J.; Zhou, J. Efficient Bioconversion of Epimedin C to Icariin by a Glycosidase from Aspergillus nidulans. Bioresour. Technol. 2019, 289, 121612. [Google Scholar] [CrossRef]
  44. Zha, J.; Wu, X.; Gong, G.; Koffas, M.A.G. Pathway Enzyme Engineering for Flavonoid Production in Recombinant Microbes. Metab. Eng. Commun. 2019, 9, e00104. [Google Scholar] [CrossRef]
  45. Yadav, V.; Yadav, P.K.; Yadav, S.; Yadav, K.D.S. α-l-Rhamnosidase: A Review. Process Biochem. 2010, 45, 1226–1235. [Google Scholar] [CrossRef]
  46. Hădărugă, D.-I.; Hădărugă, N.-G. Flavanones in Plants and Humans: Properties and Applications. In Handbook of Food Bioactive Ingredients; Jafari, S.M., Rashidinejad, A., Simal-Gandara, J., Eds.; Springer International Publishing: Cham, Switzerland, 2022; pp. 1–53. ISBN 978-3-030-81404-5. [Google Scholar]
  47. Ribeiro, M.H.L. Glycosides. In Biotechnology of Bioactive Compounds; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2015; pp. 301–344. ISBN 9781118733103. [Google Scholar]
  48. Isidore, E.; Willig, G.; Brunissen, F.; Magro, C.; Monteux, C.; Ioannou, I. Selective Recovery of Glycosylated Phenolic Compounds from Nectarine Tree Branches (Prunus persica Var. nucipersica). Food Chem. Adv. 2024, 4, 100585. [Google Scholar] [CrossRef]
  49. Le Roy, J.; Huss, B.; Creach, A.; Hawkins, S.; Neutelings, G. Glycosylation Is a Major Regulator of Phenylpropanoid Availability and Biological Activity in Plants. Front. Plant Sci. 2016, 7, 735. [Google Scholar] [CrossRef] [PubMed]
  50. Bartmańska, A.; Tronina, T.; Popłoński, J.; Milczarek, M.; Filip-Psurska, B.; Wietrzyk, J. Highly Cancer Selective Antiproliferative Activity of Natural Prenylated Flavonoids. Molecules 2018, 23, 2922. [Google Scholar] [CrossRef]
  51. Li, R.; Huang, L.; Zhang, Z.; Chen, J.; Tang, H. Integrated Multispectroscopic Analysis and Molecular Docking Analyses of the Structure-Affinity Relationship and Mechanism of the Interaction of Flavonoids with Zein. Food Chem. 2022, 386, 132839. [Google Scholar] [CrossRef] [PubMed]
  52. Akher, F.B.; Ebrahimi, A.; Mostafavi, N. Characterization of π-Stacking Interactions between Aromatic Amino Acids and Quercetagetin. J. Mol. Struct. 2017, 1128, 13–20. [Google Scholar] [CrossRef]
  53. Deogratias, G.; Shadrack, D.M.; Munissi, J.J.E.; Kinunda, G.A.; Jacob, F.R.; Mtei, R.P.; Masalu, R.J.; Mwakyula, I.; Kiruri, L.W.; Nyandoro, S.S. Hydrophobic π-π Stacking Interactions and Hydrogen Bonds Drive Self-Aggregation of Luteolin in Water. J. Mol. Graph. Model. 2022, 116, 108243. [Google Scholar] [CrossRef]
  54. Jung, H.A.; Paudel, P.; Seong, S.H.; Min, B.-S.; Choi, J.S. Structure-Related Protein Tyrosine Phosphatase 1B Inhibition by Naringenin Derivatives. Bioorg. Med. Chem. Lett. 2017, 27, 2274–2280. [Google Scholar] [CrossRef]
  55. Shilpa, V.S.; Shams, R.; Dash, K.K.; Pandey, V.K.; Dar, A.H.; Ayaz Mukarram, S.; Harsányi, E.; Kovács, B. Phytochemical Properties, Extraction, and Pharmacological Benefits of Naringin: A Review. Molecules 2023, 28, 5623. [Google Scholar] [CrossRef]
  56. Li, Y.; Paxton, J.W. The Effects of Flavonoids on the ABC Transporters: Consequences for the Pharmacokinetics of Substrate Drugs. Expert Opin. Drug Metab. Toxicol. 2013, 9, 267–285. [Google Scholar] [CrossRef] [PubMed]
  57. Rebai, R.; Jasmin, L.; Boudah, A. Identification of Two Flavonoids as New and Safe Inhibitors of Kynurenine Aminotransferase II via Computational and In Vitro Study. Pharmaceuticals 2025, 18, 76. [Google Scholar] [CrossRef] [PubMed]
  58. Watkins, B.A.; Mitchell, A.E.; Shin, A.C.; Dehghani, F.; Shen, C.-L. Dietary Flavonoid Actions on Senescence, Aging, and Applications for Health. J. Nutr. Biochem. 2025, 139, 109862. [Google Scholar] [CrossRef] [PubMed]
  59. Yadav, V.; Yadav, S.; Yadava, S.; Yadav, K.D.S. α-l-Rhamnosidase from Aspergillus Flavus MTCC-9606 Isolated from Lemon Fruit Peel. Int. J. Food Sci. Technol. 2011, 46, 350–357. [Google Scholar] [CrossRef]
  60. Shakour, Z.T.A.; Fayek, N.M.; Farag, M.A. How Do Biocatalysis and Biotransformation Affect Citrus Dietary Flavonoids Chemistry and Bioactivity? A Review. Crit. Rev. Biotechnol. 2020, 40, 689–714. [Google Scholar] [CrossRef]
  61. Ali, M.Y.; Zamponi, G.W.; Abdul, Q.A.; Seong, S.H.; Min, B.-S.; Jung, H.A.; Choi, J.S. Prunin from Poncirus trifoliata (L.) Rafin Inhibits Aldose Reductase and Glucose-Fructose–Mediated Protein Glycation and Oxidation of Human Serum Albumin. J. Agric. Food Chem. 2024, 72, 7203–7218. [Google Scholar] [CrossRef]
  62. Zhang, Y.; Gong, Y.; Hu, J.; Zhang, L.; Benito, M.J.; Usmanov, D.; Nishanbaev, S.Z.; Song, X.; Zou, L.; Wu, Y. Quercetin and Kaempferol from Saffron Petals Alleviated Hydrogen Peroxide-Induced Oxidative Damage in B16 Cells. J. Sci. Food Agric. 2025, 105, 967–973. [Google Scholar] [CrossRef]
  63. Zhang, L.; Mohankumar, K.; Martin, G.; Mariyam, F.; Park, Y.; Han, S.J.; Safe, S. Flavonoids Quercetin and Kaempferol Are NR4A1 Antagonists and Suppress Endometriosis in Female Mice. Endocrinology 2023, 164, bqad133. [Google Scholar] [CrossRef]
  64. Speisky, H.; Arias-Santé, M.F.; Fuentes, J. Oxidation of Quercetin and Kaempferol Markedly Amplifies Their Antioxidant, Cytoprotective, and Anti-Inflammatory Properties. Antioxidants 2023, 12, 155. [Google Scholar] [CrossRef]
  65. Li, B.-C.; Wu, B.; Hou, X.; Ding, G.-B. Substrate Selectivities of GH78 α-L-Rhamnosidases from Human Gut Bacteria on Dietary Flavonoid Glycosides. Molecules 2025, 30, 980. [Google Scholar] [CrossRef]
  66. Chukwuma, C.I. Antioxidative, Metabolic and Vascular Medicinal Potentials of Natural Products in the Non-Edible Wastes of Fruits Belonging to the Citrus and Prunus Genera: A Review. Plants 2024, 13, 191. [Google Scholar] [CrossRef] [PubMed]
  67. Tan, Y.; Wang, K.; Guo, M.; Zhang, G.; Li, X.; Shi, Y.; He, M.; Xu, D.; Chen, F.; Fan, J. Metabolomics Profiling Reveals P-Aminobenzoic Acid Enhances Resistance to Fusarium Head Blight in Wheat. Food Prod. Process. Nutr. 2025, 7, 14. [Google Scholar] [CrossRef]
  68. Yin, T.; Jiang, Y.; Shi, J. Effects of Alcalase Hydrolysis Combined with TGase-Type Glycosylation of Self-Assembled Zein for Curcumin Delivery: Stability, Bioavailability, and Antioxidant Properties. Int. J. Biol. Macromol. 2025, 303, 140735. [Google Scholar] [CrossRef] [PubMed]
  69. Kooptiwut, S.; Samon, K.; Semprasert, N.; Suksri, K.; Yenchitsomanus, P.-T. Prunetin Protects Against Dexamethasone-Induced Pancreatic Β-Cell Apoptosis via Modulation of P53 Signaling Pathway. Nat. Prod. Commun. 2020, 15, 1934578X20916328. [Google Scholar] [CrossRef]
  70. Veerana, M.; Mumtaz, S.; Rana, J.N.; Javed, R.; Panngom, K.; Ahmed, B.; Akter, K.; Choi, E.H. Recent Advances in Non-Thermal Plasma for Seed Germination, Plant Growth, and Secondary Metabolite Synthesis: A Promising Frontier for Sustainable Agriculture. Plasma Chem. Plasma Process. 2024, 44, 2263–2302. [Google Scholar] [CrossRef]
  71. Ullah, H.; De Filippis, A.; Khan, H.; Xiao, J.; Daglia, M. An Overview of the Health Benefits of Prunus Species with Special Reference to Metabolic Syndrome Risk Factors. Food Chem. Toxicol. 2020, 144, 111574. [Google Scholar] [CrossRef] [PubMed]
  72. Rein, M.J.; Renouf, M.; Cruz-Hernandez, C.; Actis-Goretta, L.; Thakkar, S.K.; da Silva Pinto, M. Bioavailability of Bioactive Food Compounds: A Challenging Journey to Bioefficacy. Br. J. Clin. Pharmacol. 2013, 75, 588–602. [Google Scholar] [CrossRef]
  73. Sugai, T.; Hanaya, K.; Higashibayashi, S. Semisynthesis of Prunetin, a Bioactive O-Methylated Isoflavone from Naringenin, by the Sequential Deacetylation of Chalcone Intermediates and Oxidative Rearrangement. Biosci. Biotechnol. Biochem. 2021, 85, 143–147. [Google Scholar] [CrossRef]
  74. Gordon, P.B.; Holen, I.; Seglen, P.O. Protection by Naringin and Some Other Flavonoids of Hepatocytic Autophagy and Endocytosis against Inhibition by Okadaic Acid. J. Biol. Chem. 1995, 270, 5830–5838. [Google Scholar] [CrossRef]
  75. Gonçalves, M.; Vale, N.; Silva, P. Neuroprotective Effects of Olive Oil: A Comprehensive Review of Antioxidant Properties. Antioxidants 2024, 13, 762. [Google Scholar] [CrossRef]
  76. Sorrenti, V.; Ali, S.; Mancin, L.; Davinelli, S.; Paoli, A.; Scapagnini, G. Cocoa Polyphenols and Gut Microbiota Interplay: Bioavailability, Prebiotic Effect, and Impact on Human Health. Nutrients 2020, 12, 1908. [Google Scholar] [CrossRef] [PubMed]
  77. Shahidi, F.; Peng, H. Bioaccessibility and Bioavailability of Phenolic Compounds. J. Food Bioact. 2018, 4, 11–68. [Google Scholar] [CrossRef]
  78. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global Cancer Statistics 2022: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef] [PubMed]
  79. Tanaka, T.; Kawabata, K.; Kakumoto, M.; Makita, H.; Hara, A.; Mori, H.; Satoh, K.; Hara, A.; Murakami, A.; Kuki, W.; et al. Citrus Auraptene Inhibits Chemically Induced Colonic Aberrant Crypt Foci in Male F344 Rats. Carcinogenesis 1997, 18, 2155–2161. [Google Scholar] [CrossRef]
  80. Arafah, A.; Rehman, M.U.; Mir, T.M.; Wali, A.F.; Ali, R.; Qamar, W.; Khan, R.; Ahmad, A.; Aga, S.S.; Alqahtani, S.; et al. Multi-Therapeutic Potential of Naringenin (4′,5,7-Trihydroxyflavonone): Experimental Evidence and Mechanisms. Plants 2020, 9, 1784. [Google Scholar] [CrossRef]
  81. Dehelean, C.A.; Marcovici, I.; Soica, C.; Mioc, M.; Coricovac, D.; Iurciuc, S.; Cretu, O.M.; Pinzaru, I. Plant-Derived Anticancer Compounds as New Perspectives in Drug Discovery and Alternative Therapy. Molecules 2021, 26, 1109. [Google Scholar] [CrossRef]
  82. Choi, J.S.; Yokozawa, T.; Oura, H. Improvement of Hyperglycemia and Hyperlipemia in Streptozotocin-Diabetic Rats by a Methanolic Extract of Prunus davidiana Stems and Its Main Component, Prunin. Planta Med. 1991, 57, 208–211. [Google Scholar] [CrossRef]
  83. Chen, J.; Chen, A.Y.; Huang, H.; Ye, X.; Rollyson, W.D.; Perry, H.E.; Brown, K.C.; Rojanasakul, Y.; Rankin, G.O.; Dasgupta, P.; et al. The Flavonoid Nobiletin Inhibits Tumor Growth and Angiogenesis of Ovarian Cancers via the Akt Pathway. Int. J. Oncol. 2015, 46, 2629–2638. [Google Scholar] [CrossRef]
  84. Ortuno, A.; Benavente-Garcia, O.; Castillo, J.; Alcaraz, M.; Vicente, V.; Del Rio, J.A. Beneficial Action of Citrus Flavonoids on Multiple Cancer-Related Biological Pathways. Curr. Cancer Drug Targets 2007, 7, 795–809. [Google Scholar] [CrossRef]
  85. Wang, I.-K.; Lin-Shiau, S.-Y.; Lin, J.-K. Induction of Apoptosis by Apigenin and Related Flavonoids through Cytochrome c Release and Activation of Caspase-9 and Caspase-3 in Leukaemia HL-60 Cells. Eur. J. Cancer 1999, 35, 1517–1525. [Google Scholar] [CrossRef]
  86. Galluzzo, P.; Ascenzi, P.; Bulzomi, P.; Marino, M. The Nutritional Flavanone Naringenin Triggers Antiestrogenic Effects by Regulating Estrogen Receptor α-Palmitoylation. Endocrinology 2008, 149, 2567–2575. [Google Scholar] [CrossRef]
  87. Sak, K. Site-Specific Anticancer Effects of Dietary Flavonoid Quercetin. Nutr. Cancer 2014, 66, 177–193. [Google Scholar] [CrossRef] [PubMed]
  88. Rana, J.N.; Mumtaz, S.; Choi, E.H.; Han, I. ROS Production in Response to High-Power Microwave Pulses Induces P53 Activation and DNA Damage in Brain Cells: Radiosensitivity and Biological Dosimetry Evaluation. Front. Cell Dev. Biol. 2023, 11, 1067861. [Google Scholar] [CrossRef] [PubMed]
  89. Farooq, A.; Jia, G.; Rizwan, M.; Imran, M.; Wei, D. Can WGX-50 Be a Potential Therapy to Treat Tumor by Inhibiting Mitochondrial Reactive Oxidative Species? Med. Hypotheses 2025, 196, 111583. [Google Scholar] [CrossRef]
  90. Gao, C.; Zhang, C.; Wen, L.; Zhang, G.; Liu, X.; Wang, J.; Cui, L.; Li, R.; Nie, T.; Duan, J.; et al. Regulation of Reactive Oxygen Species and the Role of Mitochondrial Apoptotic-Related Genes in Rheumatoid Arthritis. Sci. Rep. 2025, 15, 2165. [Google Scholar] [CrossRef]
  91. Zhang, X.; Li, H.; Zhang, H.; Liu, Y.; Huo, L.; Jia, Z.; Xue, Y.; Sun, X.; Zhang, W. Inhibition of Transmembrane Member 16A Calcium-Activated Chloride Channels by Natural Flavonoids Contributes to Flavonoid Anticancer Effects. Br. J. Pharmacol. 2017, 174, 2334–2345. [Google Scholar] [CrossRef]
  92. Mumtaz, S.; Rana, J.N.; Choi, E.H.; Han, I. Microwave Radiation and the Brain: Mechanisms, Current Status, and Future Prospects. Int. J. Mol. Sci. 2022, 23, 9288. [Google Scholar] [CrossRef]
  93. Mumtaz, S.; Rana, J.N.; Lim, J.S.; Javed, R.; Choi, E.H.; Han, I. Effect of Plasma On-Time with a Fixed Duty Ratio on Reactive Species in Plasma-Treated Medium and Its Significance in Biological Applications. Int. J. Mol. Sci. 2023, 24, 5289. [Google Scholar] [CrossRef]
  94. Rana, J.N.; Mumtaz, S.; Han, I.; Choi, E.H. Formation of Reactive Species via High Power Microwave Induced DNA Damage and Promoted Intrinsic Pathway-Mediated Apoptosis in Lung Cancer Cells: An in vitro Investigation. Fundam. Res. 2024, 4, 1542–1556. [Google Scholar] [CrossRef]
  95. Mumtaz, S.; Javed, R.; Rana, J.N.; Iqbal, M.; Choi, E.H. Pulsed High Power Microwave Seeds Priming Modulates Germination, Growth, Redox Homeostasis, and Hormonal Shifts in Barley for Improved Seedling Growth: Unleashing the Molecular Dynamics. Free Radic. Biol. Med. 2024, 222, 371–385. [Google Scholar] [CrossRef]
  96. Gunaseelan, S.; Wong, K.Z.; Min, N.; Sun, J.; Ismail, N.K.B.M.; Tan, Y.J.; Lee, R.C.H.; Chu, J.J.H. Prunin Suppresses Viral IRES Activity and Is a Potential Candidate for Treating Enterovirus A71 Infection. Sci. Transl. Med. 2019, 11, eaar5759. [Google Scholar] [CrossRef] [PubMed]
  97. Morikawa, K.; Nonaka, M.; Mochizuki, H.; Handa, K.; Hanada, H.; Hirota, K. Naringenin and Hesperetin Induce Growth Arrest, Apoptosis, and Cytoplasmic Fat Deposit in Human Preadipocytes. J. Agric. Food Chem. 2008, 56, 11030–11037. [Google Scholar] [CrossRef] [PubMed]
  98. Ren, W.; Qiao, Z.; Wang, H.; Zhu, L.; Zhang, L. Flavonoids: Promising Anticancer Agents. Med. Res. Rev. 2003, 23, 519–534. [Google Scholar] [CrossRef]
  99. Siti, H.N.; Jalil, J.; Asmadi, A.Y.; Kamisah, Y. Rutin Modulates MAPK Pathway Differently from Quercetin in Angiotensin II-Induced H9c2 Cardiomyocyte Hypertrophy. Int. J. Mol. Sci. 2021, 22, 5063. [Google Scholar] [CrossRef] [PubMed]
  100. Yuan, C.; Chen, G.; Jing, C.; Liu, M.; Liang, B.; Gong, G.; Yu, M. Eriocitrin, a Dietary Flavonoid Suppressed Cell Proliferation, Induced Apoptosis through Modulation of JAK2/STAT3 and JNK/P38 MAPKs Signaling Pathway in MCF-7 Cells. J. Biochem. Mol. Toxicol. 2022, 36, e22943. [Google Scholar] [CrossRef]
  101. Cheng, M.; Li, T.; Hu, E.; Yan, Q.; Li, H.; Wang, Y.; Luo, J.; Tang, T. A Novel Strategy of Integrating Network Pharmacology and Transcriptome Reveals Antiapoptotic Mechanisms of Buyang Huanwu Decoction in Treating Intracerebral Hemorrhage. J. Ethnopharmacol. 2024, 319, 117123. [Google Scholar] [CrossRef]
  102. Zhao, C.; Song, W.; Wang, J.; Tang, X.; Jiang, Z. Immunoadjuvant-Functionalized Metal–Organic Frameworks: Synthesis and Applications in Tumor Immune Modulation. Chem. Commun. 2025, 61, 1962–1977. [Google Scholar] [CrossRef]
  103. Liu, Y.; Wang, Z.; Tang, Z.; Fu, Y.; Wang, L. Mirna-383-5p Functions as an Anti-Oncogene in Glioma through the Akt/MTOR Signaling Pathway by Targeting VEGFA. Curr. Cancer Drug Targets 2024, 24, 463–475. [Google Scholar] [CrossRef]
  104. Rao, U.S.M.; Dudekula, J.B.; Bhatt, S.; Kumar, M.S.; Shah, K.; Chauhan, N.S.; Shilpi, S. Chapter 18-Role of Phytopharmaceuticals in Inflammatory Disorders. In Phytopharmaceuticals and Herbal Drugs; Singh, M.R., Singh, D., Eds.; Academic Press: Cambridge, MA, USA, 2023; pp. 433–451. ISBN 978-0-323-99125-4. [Google Scholar]
  105. Yang, Y.; Liu, Q.; Shi, X.; Zheng, Q.; Chen, L.; Sun, Y. Advances in Plant-Derived Natural Products for Antitumor Immunotherapy. Arch. Pharm. Res. 2021, 44, 987–1011. [Google Scholar] [CrossRef]
  106. Rattanapisit, K.; Suwanchaikasem, P.; Bulaon, C.J.I.; Guo, S.; Phoolcharoen, W. Plant-Derived Pembrolizumab in Conjugation with IL-15Rα-IL-15 Complex Shows Effective Anti-Tumor Activity. PLoS ONE 2025, 20, e0316790. [Google Scholar] [CrossRef]
  107. Pyo, Y.; Kwon, K.H.; Jung, Y.J. Anticancer Potential of Flavonoids: Their Role in Cancer Prevention and Health Benefits. Foods 2024, 13, 2253. [Google Scholar] [CrossRef] [PubMed]
  108. Jin, C.; Hinterdorfer, P.; Lee, J.H.; Ko, K. Plant Production Systems for Recombinant Immunotherapeutic Proteins. Plant Biotechnol. Rep. 2025, 19, 1–14. [Google Scholar] [CrossRef]
  109. Trivedi, A.; Hasan, A.; Ahmad, R.; Siddiqui, S.; Srivastava, A.; Misra, A.; Mir, S.S. Flavonoid Myricetin as Potent Anticancer Agent: A Possibility towards Development of Potential Anticancer Nutraceuticals. Chin. J. Integr. Med. 2024, 30, 75–84. [Google Scholar] [CrossRef]
  110. Esmeeta, A.; Adhikary, S.; Dharshnaa, V.; Swarnamughi, P.; Ummul Maqsummiya, Z.; Banerjee, A.; Pathak, S.; Duttaroy, A.K. Plant-Derived Bioactive Compounds in Colon Cancer Treatment: An Updated Review. Biomed. Pharmacother. 2022, 153, 113384. [Google Scholar] [CrossRef] [PubMed]
  111. Elawad, M.A.; Ayaz, M.; Mosa, O.F.; Usman, A.; Hamdoon, A.A.E.; Almawash, S.; Salim, L.H.M.; Ahmed, A.; Elkhalifa, M.E.M. Polyphenols and Their Biogenic Nano-Formulations Targeting BACE1 as Anti-Amyloid Therapies; Meeting the Challenges of Bioavailability, Safety, and Specificity for the Treatment of Alzheimer’s Disease. Mol. Nutr. Food Res. 2024, 68, 2400525. [Google Scholar] [CrossRef]
  112. Marcucci, F.; Corti, A. How to Improve Exposure of Tumor Cells to Drugs—Promoter Drugs Increase Tumor Uptake and Penetration of Effector Drugs. Adv. Drug Deliv. Rev. 2012, 64, 53–68. [Google Scholar] [CrossRef]
  113. Pistollato, F.; Giampieri, F.; Battino, M. The Use of Plant-Derived Bioactive Compounds to Target Cancer Stem Cells and Modulate Tumor Microenvironment. Food Chem. Toxicol. 2015, 75, 58–70. [Google Scholar] [CrossRef]
  114. Zhang, H.; Li, Y. Targeting the Breast Tumor Microenvironment by Plant-Derived Products and Their Nanoformulations. J. Drug Deliv. Sci. Technol. 2024, 93, 105432. [Google Scholar] [CrossRef]
  115. Bharadwaj, K.K.; Rabha, B.; Ahmad, I.; Mathew, S.P.; Bhattacharjee, C.K.; Jaganathan, B.G.; Poddar, S.; Patel, H.; Subramaniyan, V.; Chinni, S.V.; et al. Rhamnetin, a Nutraceutical Flavonoid Arrests Cell Cycle Progression of Human Ovarian Cancer (SKOV3) Cells by Inhibiting the Histone Deacetylase 2 Protein. J. Biomol. Struct. Dyn. 2024, 42, 13421–13436. [Google Scholar] [CrossRef]
  116. Cao, Z.; Zhu, J.; Wang, Z.; Peng, Y.; Zeng, L. Comprehensive Pan-Cancer Analysis Reveals ENC1 as a Promising Prognostic Biomarker for Tumor Microenvironment and Therapeutic Responses. Sci. Rep. 2024, 14, 25331. [Google Scholar] [CrossRef]
  117. Saadh, M.J.; Mustafa, M.A.; Malathi, H.; Ahluwalia, G.; Kaur, S.; Al-Dulaimi, M.A.A.H.; Alubiady, M.H.S.; Zain Al-Abdeen, S.H.; Shakier, H.G.; Ali, M.S.; et al. Targeting the Pancreatic Tumor Microenvironment by Plant-Derived Products and Their Nanoformulations. Med. Oncol. 2024, 41, 201. [Google Scholar] [CrossRef] [PubMed]
  118. Tran, H.K.; Nguyen, N.P.N.; Nguyen, T.T.T.; Nguyen, K.N.; Do, B.D.; Nguyen, T.V.A.; Tran, A.K.; Nguyen, T.K.C.; Ho, Q.T.; Truong, D.-H.; et al. Extraction of Flavonoids from Durian (Durio zibethinus) Fruit Rinds and Evaluation of Their Antioxidant, Antidiabetic and Anticancer Properties. Int. J. Food Sci. Technol. 2024, 59, 1409–1420. [Google Scholar] [CrossRef]
  119. Lotfi, M.-S.; Rassouli, F.B. Natural Flavonoid Apigenin, an Effective Agent Against Nervous System Cancers. Mol. Neurobiol. 2024, 61, 5572–5583. [Google Scholar] [CrossRef] [PubMed]
  120. Chaachouay, N.; Zidane, L. Plant-Derived Natural Products: A Source for Drug Discovery and Development. Drugs Drug Candidates 2024, 3, 184–207. [Google Scholar] [CrossRef]
  121. Anand, U.; Jacobo-Herrera, N.; Altemimi, A.; Lakhssassi, N. A Comprehensive Review on Medicinal Plants as Antimicrobial Therapeutics: Potential Avenues of Biocompatible Drug Discovery. Metabolites 2019, 9, 258. [Google Scholar] [CrossRef]
  122. Mumtaz, S.; Lim, J.; Kaushik, N.K.; Choi, E.H. Biological Effects of Pulsed High-Power Microwaves. In Plasma Biosciences and Medicine; Choi, E.H., Ed.; Springer Nature: Singapore, 2023; pp. 281–307. ISBN 978-981-19-7935-4. [Google Scholar]
  123. Wu, T.-N.; Chen, H.-M.; Shyur, L.-F. Current Advancements of Plant-Derived Agents for Triple-Negative Breast Cancer Therapy through Deregulating Cancer Cell Functions and Reprogramming Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 13571. [Google Scholar] [CrossRef]
  124. Jiang, L.; Zhang, G.; Li, Y.; Shi, G.; Li, M. Potential Application of Plant-Based Functional Foods in the Development of Immune Boosters. Front. Pharmacol. 2021, 12, 637782. [Google Scholar] [CrossRef]
  125. Grudzien, M.; Rapak, A. Effect of Natural Compounds on NK Cell Activation. J. Immunol. Res. 2018, 2018, 4868417. [Google Scholar] [CrossRef]
  126. Ganai, S.A.; Sheikh, F.A.; Baba, Z.A.; Mir, M.A.; Mantoo, M.A.; Yatoo, M.A. Anticancer Activity of the Plant Flavonoid Luteolin against Preclinical Models of Various Cancers and Insights on Different Signalling Mechanisms Modulated. Phyther. Res. 2021, 35, 3509–3532. [Google Scholar] [CrossRef]
  127. Ferdous, U.T.; Balia Yusof, Z.N. Insight into Potential Anticancer Activity of Algal Flavonoids: Current Status and Challenges. Molecules 2021, 26, 6844. [Google Scholar] [CrossRef]
  128. Qi, Y.-K.; Zheng, J.-S.; Liu, L. Mirror-Image Protein and Peptide Drug Discovery through Mirror-Image Phage Display. Chem 2024, 10, 2390–2407. [Google Scholar] [CrossRef]
  129. Saklani, A.; Kutty, S.K. Plant-Derived Compounds in Clinical Trials. Drug Discov. Today 2008, 13, 161–171. [Google Scholar] [CrossRef] [PubMed]
  130. Prasher, P.; Sharma, M.; Singh, S.K.; Gulati, M.; Chellappan, D.K.; Zacconi, F.; De Rubis, G.; Gupta, G.; Sharifi-Rad, J.; Cho, W.C.; et al. Luteolin: A Flavonoid with a Multifaceted Anticancer Potential. Cancer Cell Int. 2022, 22, 386. [Google Scholar] [CrossRef]
  131. Pandey, P.; Khan, F. A Mechanistic Review of the Anticancer Potential of Hesperidin, a Natural Flavonoid from Citrus Fruits. Nutr. Res. 2021, 92, 21–31. [Google Scholar] [CrossRef] [PubMed]
  132. Afshari, A.R.; Sanati, M.; Ahmadi, S.S.; Kesharwani, P.; Sahebkar, A. Harnessing the Capacity of Phytochemicals to Enhance Immune Checkpoint Inhibitor Therapy of Cancers: A Focus on Brain Malignancies. Cancer Lett. 2024, 593, 216955. [Google Scholar] [CrossRef]
  133. Lee, J.; Han, Y.; Wang, W.; Jo, H.; Kim, H.; Kim, S.; Yang, K.-M.; Kim, S.-J.; Dhanasekaran, D.N.; Song, Y.S. Phytochemicals in Cancer Immune Checkpoint Inhibitor Therapy. Biomolecules 2021, 11, 1107. [Google Scholar] [CrossRef]
  134. Mariappan, B.; Kaliyamurthi, V.; Binesh, A. Chapter 8-Medicinal Plants or Plant Derived Compounds Used in Aquaculture. In Recent Advances in Aquaculture Microbial Technology; Mathew, J., Jose, M.S., Radhakrishnan, E.K., Kumar, A., Eds.; Academic Press: Cambridge, MA, USA, 2023; pp. 153–207. ISBN 978-0-323-90261-8. [Google Scholar]
  135. Rahmani, A.H.; Almatroudi, A.; Allemailem, K.S.; Khan, A.A.; Almatroodi, S.A. The Potential Role of Fisetin, a Flavonoid in Cancer Prevention and Treatment. Molecules 2022, 27, 9009. [Google Scholar] [CrossRef]
  136. Tavsan, Z.; Kayali, H.A. Flavonoids Showed Anticancer Effects on the Ovarian Cancer Cells: Involvement of Reactive Oxygen Species, Apoptosis, Cell Cycle and Invasion. Biomed. Pharmacother. 2019, 116, 109004. [Google Scholar] [CrossRef]
  137. Batra, P.; Sharma, A.K. Anti-Cancer Potential of Flavonoids: Recent Trends and Future Perspectives. 3 Biotech 2013, 3, 439–459. [Google Scholar] [CrossRef]
  138. Budi, H.S.; Farhood, B. Tumor Microenvironment Remodeling in Oral Cancer: Application of Plant Derived-Natural Products and Nanomaterials. Environ. Res. 2023, 233, 116432. [Google Scholar] [CrossRef]
  139. Raffa, D.; Maggio, B.; Raimondi, M.V.; Plescia, F.; Daidone, G. Recent Discoveries of Anticancer Flavonoids. Eur. J. Med. Chem. 2017, 142, 213–228. [Google Scholar] [CrossRef]
  140. Wan, H.; Zhou, S.; Li, C.; Zhou, H.; Wan, H.; Yang, J.; Yu, L. Ant Colony Algorithm-Enabled Back Propagation Neural Network and Response Surface Methodology Based Ultrasonic Optimization of Safflower Seed Alkaloid Extraction and Antioxidant. Ind. Crops Prod. 2024, 220, 119191. [Google Scholar] [CrossRef]
  141. Gao, T.-H.; Liao, W.; Lin, L.-T.; Zhu, Z.-P.; Lu, M.-G.; Fu, C.-M.; Xie, T. Curcumae rhizoma and Its Major Constituents against Hepatobiliary Disease: Pharmacotherapeutic Properties and Potential Clinical Applications. Phytomedicine 2022, 102, 154090. [Google Scholar] [CrossRef] [PubMed]
  142. Guo, D.; Pan, Y.; Wang, S.; Ming, K.; Chi, Q.; Wang, C.; Xu, K. Rapid Identification of Astragalus Membranaceus Processing with Rice Water Based on Intelligent Color Recognition and Multi-Source Information Fusion Technology. Chin. Herb. Med. 2025. [Google Scholar] [CrossRef]
  143. Guo, Y.; Han, Z.; Zhang, J.; Lu, Y.; Li, C.; Liu, G. Development of a High-Speed and Ultrasensitive UV/Vis-CM for Detecting Total Triterpenes in Traditional Chinese Medicine and Its Application. Heliyon 2024, 10, e32239. [Google Scholar] [CrossRef] [PubMed]
  144. Liu, Z.; Huang, P.; Law, S.; Tian, H.; Leung, W.; Xu, C. Preventive Effect of Curcumin Against Chemotherapy-Induced Side-Effects. Front. Pharmacol. 2018, 9, 1374. [Google Scholar] [CrossRef]
  145. Zhu, J.; Jiang, X.; Luo, X.; Zhao, R.; Li, J.; Cai, H.; Ye, X.-Y.; Bai, R.; Xie, T. Combination of Chemotherapy and Gaseous Signaling Molecular Therapy: Novel β-Elemene Nitric Oxide Donor Derivatives against Leukemia. Drug Dev. Res. 2023, 84, 718–735. [Google Scholar] [CrossRef]
  146. Lodi, R.S.; Dong, X.; Wang, X.; Han, Y.; Liang, X.; Peng, C.; Peng, L. Current Research on the Medical Importance of Trametes Species. Fungal Biol. Rev. 2025, 51, 100413. [Google Scholar] [CrossRef]
  147. Wang, T.; Zhang, F.; Zhuang, W.; Shu, X.; Wang, Z. Metabolic Variations of Flavonoids in Leaves of T. media and T. mairei Obtained by UPLC-ESI-MS/MS. Molecules 2019, 24, 3323. [Google Scholar] [CrossRef]
  148. De Marco, F.; Altieri, F.; Giuliani, S.; Falcone, I.; Falcucci, S.; Tedesco, M.; Becelli, R. A Combination of Flavonoids Suppresses Cell Proliferation and the E6 Oncogenic Pathway in Human Papillomavirus-Transformed Cells. Pathogens 2025, 14, 221. [Google Scholar] [CrossRef]
  149. Rajakumar, T.; Pugalendhi, P. Allyl Isothiocyanate Regulates Oxidative Stress, Inflammation, Cell Proliferation, Cell Cycle Arrest, Apoptosis, Angiogenesis, Invasion and Metastasis via Interaction with Multiple Cell Signaling Pathways. Histochem. Cell Biol. 2024, 161, 211–221. [Google Scholar] [CrossRef] [PubMed]
  150. Wang, Z.; Ren, M.; Liu, W.; Wu, J.; Tang, P. Role of Cell Division Cycle-Associated Proteins in Regulating Cell Cycle and Promoting Tumor Progression. Biochim. Biophys. Acta-Rev. Cancer 2024, 1879, 189147. [Google Scholar] [CrossRef] [PubMed]
  151. Li, S.; Mu, R.; Guo, X. Defensins Regulate Cell Cycle: Insights of Defensins on Cellular Proliferation and Division. Life Sci. 2024, 349, 122740. [Google Scholar] [CrossRef] [PubMed]
  152. Park, C.; Cha, H.-J.; Choi, E.O.; Lee, H.; Hwang-Bo, H.; Ji, S.Y.; Kim, M.Y.; Kim, S.Y.; Hong, S.H.; Cheong, J.; et al. Isorhamnetin Induces Cell Cycle Arrest and Apoptosis Via Reactive Oxygen Species-Mediated AMP-Activated Protein Kinase Signaling Pathway Activation in Human Bladder Cancer Cells. Cancers 2019, 11, 1494. [Google Scholar] [CrossRef]
  153. Wu, J.; Song, Y.; Wang, J.; Wang, T.; Yang, L.; Shi, Y.; Song, B.; Yu, Z. Isorhamnetin inhibits hypertrophic scar formation through TGF-β1/Smad and TGF-β1/CREB3L1 signaling pathways. Heliyon 2024, 10, e33802. [Google Scholar] [CrossRef]
  154. Diehl, F.F.; Sapp, K.M.; Vander Heiden, M.G. The Bidirectional Relationship between Metabolism and Cell Cycle Control. Trends Cell Biol. 2024, 34, 136–149. [Google Scholar] [CrossRef]
  155. Stallaert, W.; Taylor, S.R.; Kedziora, K.M.; Taylor, C.D.; Sobon, H.K.; Young, C.L.; Limas, J.C.; Varblow Holloway, J.; Johnson, M.S.; Cook, J.G.; et al. The Molecular Architecture of Cell Cycle Arrest. Mol. Syst. Biol. 2022, 18, e11087. [Google Scholar] [CrossRef]
  156. Lu, R.; Liu, J.; Thakur, K.; Cao, H.; Mejuto, J.C.; Gandara, J.S.; Zhang, J.-G. Protopanaxadiol Triggers G0/G1 Cell Cycle Arrest and Apoptosis in Human Cervical Cancer HeLa Cells through the PPER Pathway. Food Biosci. 2024, 62, 105388. [Google Scholar] [CrossRef]
  157. Wang, J.-L.; Quan, Q.; Ji, R.; Guo, X.-Y.; Zhang, J.-M.; Li, X.; Liu, Y.-G. Isorhamnetin Suppresses PANC-1 Pancreatic Cancer Cell Proliferation through S Phase Arrest. Biomed. Pharmacother. 2018, 108, 925–933. [Google Scholar] [CrossRef]
  158. Hu, D.; Wang, H.-J.; Yu, L.-H.; Guan, Z.-R.; Jiang, Y.-P.; Hu, J.-H.; Yan, Y.-X.; Zhou, Z.-H.; Lou, J.-S. The Role of Ginkgo Folium on Antitumor: Bioactive Constituents and the Potential Mechanism. J. Ethnopharmacol. 2024, 321, 117202. [Google Scholar] [CrossRef]
  159. Deshpande, A.; Sicinski, P.; Hinds, P.W. Cyclins and Cdks in Development and Cancer: A Perspective. Oncogene 2005, 24, 2909–2915. [Google Scholar] [CrossRef]
  160. Wu, N.; Zhang, X.; Fang, C.; Zhu, M.; Wang, Z.; Jian, L.; Tan, W.; Wang, Y.; Li, H.; Xu, X.; et al. Progesterone Enhances Niraparib Efficacy in Ovarian Cancer by Promoting Palmitoleic-Acid-Mediated Ferroptosis. Research 2025, 7, 371. [Google Scholar] [CrossRef] [PubMed]
  161. Koyu, H.; Kazan, A.; Nalbantsoy, A.; Yalcin, H.T.; Yesil-Celiktas, O. Cytotoxic, Antimicrobial and Nitric Oxide Inhibitory Activities of Supercritical Carbon Dioxide Extracted Prunus Persica Leaves. Mol. Biol. Rep. 2020, 47, 569–581. [Google Scholar] [CrossRef]
  162. Na, E.J.; Ryu, J.Y. Anti-Inflammatory Effects of Prunin on UVB-Irradiated Human Keratinocytes. Biomed. Dermatol. 2018, 2, 14. [Google Scholar] [CrossRef]
  163. Oliveira Lino, L.; Pacheco, I.; Mercier, V.; Faoro, F.; Bassi, D.; Bornard, I.; Quilot-Turion, B. Brown Rot Strikes Prunus Fruit: An Ancient Fight Almost Always Lost. J. Agric. Food Chem. 2016, 64, 4029–4047. [Google Scholar] [CrossRef]
  164. Huang, M.-F.; Wang, Y.-X.; Chou, Y.-T.; Lee, D.-F. Therapeutic Strategies for RB1-Deficient Cancers: Intersecting Gene Regulation and Targeted Therapy. Cancers 2024, 16, 1558. [Google Scholar] [CrossRef] [PubMed]
  165. Chang, C.-H.; Liu, F.; Militi, S.; Hester, S.; Nibhani, R.; Deng, S.; Dunford, J.; Rendek, A.; Soonawalla, Z.; Fischer, R.; et al. The PRb/RBL2-E2F1/4-GCN5 Axis Regulates Cancer Stem Cell Formation and G0 Phase Entry/Exit by Paracrine Mechanisms. Nat. Commun. 2024, 15, 3580. [Google Scholar] [CrossRef] [PubMed]
  166. Huang, W.; Hickson, L.J.; Eirin, A.; Kirkland, J.L.; Lerman, L.O. Cellular Senescence: The Good, the Bad and the Unknown. Nat. Rev. Nephrol. 2022, 18, 611–627. [Google Scholar] [CrossRef]
  167. Casagrande, F.; Darbon, J.-M. Effects of structurally related flavonoids on cell cycle progression of human melanoma cells: Regulation of cyclin-dependent kinases CDK2 and CDK1. Biochem. Pharmacol. 2001, 61, 1205–1215. [Google Scholar] [CrossRef]
  168. Pan, M.-H.; Chen, W.-J.; Lin-Shiau, S.-Y.; Ho, C.-T.; Lin, J.-K. Tangeretin Induces Cell-Cycle G1 Arrest through Inhibiting Cyclin-Dependent Kinases 2 and 4 Activities as Well as Elevating Cdk Inhibitors P21 and P27 in Human Colorectal Carcinoma Cells. Carcinogenesis 2002, 23, 1677–1684. [Google Scholar] [CrossRef]
  169. AL-Ishaq, R.K.; Abotaleb, M.; Kubatka, P.; Kajo, K.; Büsselberg, D. Flavonoids and Their Anti-Diabetic Effects: Cellular Mechanisms and Effects to Improve Blood Sugar Levels. Biomolecules 2019, 9, 430. [Google Scholar] [CrossRef] [PubMed]
  170. Hogan, F.S.; Krishnegowda, N.K.; Mikhailova, M.; Kahlenberg, M.S. Flavonoid, Silibinin, Inhibits Proliferation and Promotes Cell-Cycle Arrest of Human Colon Cancer. J. Surg. Res. 2007, 143, 58–65. [Google Scholar] [CrossRef]
  171. Yang, S.; Chu, G.; Wu, J.; Zhang, G.; Du, L.; Lin, R. Enrichment and Evaluation of Antitumor Properties of Total Flavonoids from Juglans mandshurica Maxim. Molecules 2024, 29, 1976. [Google Scholar] [CrossRef]
  172. Kashyap, D.; Garg, V.K.; Goel, N. Chapter Four-Intrinsic and Extrinsic Pathways of Apoptosis: Role in Cancer Development and Prognosis. In Apoptosis in Health and Disease-Part A; Donev, R., Ed.; Academic Press: Cambridge, MA, USA, 2021; Volume 125, pp. 73–120. ISBN 1876-1623. [Google Scholar]
  173. Liu, L.; Pang, Y.; Zhao, X.; Li, R.; Jin, C.; Xue, J.; Dong, R.; Liu, P. Curcumin Induces Apoptotic Cell Death and Protective Autophagy by Inhibiting AKT/MTOR/P70S6K Pathway in Human Ovarian Cancer Cells. Arch. Gynecol. Obstet. 2019, 299, 1627–1639. [Google Scholar] [CrossRef] [PubMed]
  174. Yang, X.; Bai, Z.-F.; Zhang, Y.; Cui, H.; Zhou, H.-L. Flavonoids-Rich Extract from Bidens bipinnata L. Protects Pancreatic β-Cells against Oxidative Stress-Induced Apoptosis through Intrinsic and Extrinsic Pathways. J. Ethnopharmacol. 2021, 275, 114097. [Google Scholar] [CrossRef]
  175. Hongmei, Z. Extrinsic and Intrinsic Apoptosis Signal Pathway Review. In Apoptosis and Medicine; Ntuli, T.M., Ed.; IntechOpen: Rijeka, Croatia, 2012. [Google Scholar]
  176. Li, X.; Wang, T.; Zhou, Q.; Li, F.; Liu, T.; Zhang, K.; Wen, A.; Feng, L.; Shu, X.; Tian, S.; et al. Isorhamnetin Alleviates Mitochondrial Injury in Severe Acute Pancreatitis via Modulation of KDM5B/HtrA2 Signaling Pathway. Int. J. Mol. Sci. 2024, 25, 3784. [Google Scholar] [CrossRef]
  177. Liu, M.; Lu, J.; Chen, Y.; Zhang, D.; Huang, W.; Shi, M.; Zhang, Y.; Wu, T.; Chen, Z.; Wu, L.; et al. Investigation of the Underlying Mechanism of Huangqi-Dangshen for Myasthenia Gravis Treatment via Molecular Docking and Network Pharmacology. Evid.-Based Complement. Altern. Med. 2023, 2023, 5301024. [Google Scholar] [CrossRef] [PubMed]
  178. An, X.; Yu, W.; Liu, J.; Tang, D.; Yang, L.; Chen, X. Oxidative Cell Death in Cancer: Mechanisms and Therapeutic Opportunities. Cell Death Dis. 2024, 15, 556. [Google Scholar] [CrossRef]
  179. Chen, L.; Wu, L.; Zhang, L.; Sun, B.; Wu, W.; Lei, Y.; Zhu, L.; Sun, T.; Liang, B.; Zhao, H.; et al. Effect of Metformin on Hepatocellular Carcinoma Patients with Type II Diabetes Receiving Transarterial Chemoembolization: A Multicenter Retrospective Cohort Study. Int. J. Surg. 2025, 111, 828–838. [Google Scholar] [CrossRef]
  180. Rodríguez, L.; Badimon, L.; Méndez, D.; Padró, T.; Vilahur, G.; Peña, E.; Carrasco, B.; Vogel, H.; Palomo, I.; Fuentes, E. Antiplatelet Activity of Isorhamnetin via Mitochondrial Regulation. Antioxidants 2021, 10, 666. [Google Scholar] [CrossRef]
  181. Zhu, J.T.T.; Choi, R.C.Y.; Chu, G.K.Y.; Cheung, A.W.H.; Gao, Q.T.; Li, J.; Jiang, Z.Y.; Dong, T.T.X.; Tsim, K.W.K. Flavonoids Possess Neuroprotective Effects on Cultured Pheochromocytoma PC12 Cells:  A Comparison of Different Flavonoids in Activating Estrogenic Effect and in Preventing β-Amyloid-Induced Cell Death. J. Agric. Food Chem. 2007, 55, 2438–2445. [Google Scholar] [CrossRef]
  182. Schulze-Osthoff, K.; Ferrari, D.; Los, M.; Wesselborg, S.; Peter, M.E. Apoptosis Signaling by Death Receptors. Eur. J. Biochem. 1998, 254, 439–459. [Google Scholar] [CrossRef] [PubMed]
  183. Ahmad, I.; Hoque, M.; Alam, S.S.M.; Zughaibi, T.A.; Tabrez, S. Curcumin and Plumbagin Synergistically Target the PI3K/Akt/MTOR Pathway: A Prospective Role in Cancer Treatment. Int. J. Mol. Sci. 2023, 24, 6651. [Google Scholar] [CrossRef] [PubMed]
  184. Tian, L.-Y.; Smit, D.J.; Jücker, M. The Role of PI3K/AKT/MTOR Signaling in Hepatocellular Carcinoma Metabolism. Int. J. Mol. Sci. 2023, 24, 2652. [Google Scholar] [CrossRef] [PubMed]
  185. Lee, J.H.; Chinnathambi, A.; Alharbi, S.A.; Shair, O.H.M.; Sethi, G.; Ahn, K.S. Farnesol Abrogates Epithelial to Mesenchymal Transition Process through Regulating Akt/MTOR Pathway. Pharmacol. Res. 2019, 150, 104504. [Google Scholar] [CrossRef]
  186. Tewari, D.; Patni, P.; Bishayee, A.; Sah, A.N.; Bishayee, A. Natural Products Targeting the PI3K-Akt-MTOR Signaling Pathway in Cancer: A Novel Therapeutic Strategy. Semin. Cancer Biol. 2022, 80, 1–17. [Google Scholar] [CrossRef]
  187. Chen, S.; Long, S.; Liu, Y.; Wang, S.; Hu, Q.; Fu, L. Evaluation of a Three-Gene Methylation Model for Correlating Lymph Node Metastasis in Postoperative Early Gastric Cancer Adjacent Samples. Front. Oncol. 2024, 14, 1432869. [Google Scholar] [CrossRef]
  188. Zhong, J.; Ding, S.; Zhang, X.; Di, W.; Wang, X.; Zhang, H.; Chen, Y.; Zhang, Y.; Hu, Y. To Investigate the Occurrence and Development of Colorectal Cancer Based on the PI3K/AKT/MTOR Signaling Pathway. Front. Biosci. 2023, 28, 37. [Google Scholar] [CrossRef]
  189. Yu, L.; Wei, J.; Liu, P. Attacking the PI3K/Akt/MTOR Signaling Pathway for Targeted Therapeutic Treatment in Human Cancer. Semin. Cancer Biol. 2022, 85, 69–94. [Google Scholar] [CrossRef]
  190. Lee, J.H.; Kim, C.; Um, J.-Y.; Sethi, G.; Ahn, K.S. Casticin-Induced Inhibition of Cell Growth and Survival Are Mediated through the Dual Modulation of Akt/MTOR Signaling Cascade. Cancers 2019, 11, 254. [Google Scholar] [CrossRef]
  191. Zheng, X.; Zhang, X.; Zeng, F. Biological Functions and Health Benefits of Flavonoids in Fruits and Vegetables: A Contemporary Review. Foods 2025, 14, 155. [Google Scholar] [CrossRef] [PubMed]
  192. Al-Bari, M.A.A.; Xu, P. Molecular Regulation of Autophagy Machinery by MTOR-Dependent and-Independent Pathways. Ann. N. Y. Acad. Sci. 2020, 1467, 3–20. [Google Scholar] [CrossRef] [PubMed]
  193. Granato, M.; Rizzello, C.; Gilardini Montani, M.S.; Cuomo, L.; Vitillo, M.; Santarelli, R.; Gonnella, R.; D’Orazi, G.; Faggioni, A.; Cirone, M. Quercetin Induces Apoptosis and Autophagy in Primary Effusion Lymphoma Cells by Inhibiting PI3K/AKT/MTOR and STAT3 Signaling Pathways. J. Nutr. Biochem. 2017, 41, 124–136. [Google Scholar] [CrossRef]
  194. Bai, D.; Zhao, Y.; Zhu, Q.; Zhou, Y.; Zhao, Y.; Zhang, T.; Guo, Q.; Lu, N. LZ205, a Newly Synthesized Flavonoid Compound, Exerts Anti-Inflammatory Effect by Inhibiting M1 Macrophage Polarization through Regulating PI3K/AKT/MTOR Signaling Pathway. Exp. Cell Res. 2018, 364, 84–94. [Google Scholar] [CrossRef]
  195. Saini, S.; Tuli, H.S.; Saini, R.V.; Saini, A.K.; Sak, K.; Kaur, D.; Shahwan, M.; Chauhan, R.; Chauhan, A. Flavonoid-Mediated Suppression of Tumor Angiogenesis: Roles of Ang-Tie/PI3K/AKT. Pathophysiology 2024, 31, 596–607. [Google Scholar] [CrossRef]
  196. Zhang, B.; Zhang, J.; Chen, H.; Qiao, D.; Guo, F.; Hu, X.; Qin, C.; Jin, X.; Zhang, K.; Wang, C.; et al. Role of FMRP in AKT/MTOR Pathway-Mediated Hippocampal Autophagy in Fragile X Syndrome. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2024, 134, 111036. [Google Scholar] [CrossRef]
  197. Araújo, C.B.; Alves Júnior, J.D.; Sato, M.R.; Costa, K.M.; Lima, J.R.; Damasceno, B.P.; Lima Junior, F.J.; Andréo, B.G.; Santos, V.L.; Oshiro-Junior, J.A. The Development and Pre-Clinical Anti-Inflammatory Efficacy of a New Transdermal Ureasil–Polyether Hybrid Matrix Loaded with Flavonoid-Rich Annona Muricata Leaf Extract. Pharmaceutics 2024, 16, 1097. [Google Scholar] [CrossRef] [PubMed]
  198. Zhang, X.; Ye, B. Isolation of Prunin From Bauhinia Variegata and Its Antioxidant Activity in Rats Fed an Atherogenic Diet. Nat. Prod. Commun. 2020, 15, 1934578X20967875. [Google Scholar] [CrossRef]
  199. Ortega-Vidal, J.; Cobo, A.; Ortega-Morente, E.; Gálvez, A.; Alejo-Armijo, A.; Salido, S.; Altarejos, J. Antimicrobial and Antioxidant Activities of Flavonoids Isolated from Wood of Sweet Cherry Tree (Prunus avium L.). J. Wood Chem. Technol. 2021, 41, 104–117. [Google Scholar] [CrossRef]
  200. Al-Khayri, J.M.; Sahana, G.R.; Nagella, P.; Joseph, B.V.; Alessa, F.M.; Al-Mssallem, M.Q. Flavonoids as Potential Anti-Inflammatory Molecules: A Review. Molecules 2022, 27, 2901. [Google Scholar] [CrossRef]
  201. Lu, Y.; Zhou, R.; Zhu, R.; Wu, X.; Liu, J.; Ma, Y.; Zhang, X.; Zhang, Y.; Yang, L.; Li, Y.; et al. Baicalin Ameliorates Neuroinflammation by Targeting TLR4/MD2 Complex on Microglia via PI3K/AKT/NF-ΚB Signaling Pathway. Neuropharmacology 2025, 267, 110296. [Google Scholar] [CrossRef] [PubMed]
  202. Guo, Y.; Zhang, J.; Yuan, T.; Yang, C.; Zhou, Q.; Shaukat, A.; Deng, G.; Wang, X. Luteolin Alleviates Inflammation Induced by Staphylococcus Aureus in Bovine Mammary Epithelial Cells by Attenuating NF-ΚB and MAPK Activation. Vet. Sci. 2025, 12, 96. [Google Scholar] [CrossRef] [PubMed]
  203. Huangfu, L.; Wang, J.; Li, D.; Fei, H.; Chen, X.; Dong, J.; Sun, L. Fraxetin Inhibits IKKβ, Blocks NF-ΚB Pathway and NLRP3 Inflammasome Activation, and Alleviates Spleen Injury in Sepsis. Chem. Biol. Interact. 2025, 408, 111406. [Google Scholar] [CrossRef]
  204. Hou, X.L.; Tong, Q.; Wang, W.Q.; Shi, C.Y.; Xiong, W.; Chen, J.; Liu, X.; Fang, J.G. Suppression of Inflammatory Responses by Dihydromyricetin, a Flavonoid from Ampelopsis grossedentata, via Inhibiting the Activation of NF-ΚB and MAPK Signaling Pathways. J. Nat. Prod. 2015, 78, 1689–1696. [Google Scholar] [CrossRef] [PubMed]
  205. Ren, Q.; Guo, F.; Tao, S.; Huang, R.; Ma, L.; Fu, P. Flavonoid Fisetin Alleviates Kidney Inflammation and Apoptosis via Inhibiting Src-Mediated NF-ΚB P65 and MAPK Signaling Pathways in Septic AKI Mice. Biomed. Pharmacother. 2020, 122, 109772. [Google Scholar] [CrossRef]
  206. Tian, C.; Zhang, P.; Yang, J.; Zhang, Z.; Wang, H.; Guo, Y.; Liu, M. The Protective Effect of the Flavonoid Fraction of Abutilon Theophrasti Medic. Leaves on LPS-Induced Acute Lung Injury in Mice via the NF-ΚB and MAPK Signalling Pathways. Biomed. Pharmacother. 2019, 109, 1024–1031. [Google Scholar] [CrossRef]
  207. Yang, J.H.; Shin, B.Y.; Han, J.Y.; Kim, M.G.; Wi, J.E.; Kim, Y.W.; Cho, I.J.; Kim, S.C.; Shin, S.M.; Ki, S.H. Isorhamnetin Protects against Oxidative Stress by Activating Nrf2 and Inducing the Expression of Its Target Genes. Toxicol. Appl. Pharmacol. 2014, 274, 293–301. [Google Scholar] [CrossRef]
  208. Seo, K.; Yang, J.H.; Kim, S.C.; Ku, S.K.; Ki, S.H.; Shin, S.M. The Antioxidant Effects of Isorhamnetin Contribute to Inhibit COX-2 Expression in Response to Inflammation: A Potential Role of HO-1. Inflammation 2014, 37, 712–722. [Google Scholar] [CrossRef]
  209. Wang, M.; Zhang, X.; Zhang, Z.; Tong, L.; Yu, S.; Liu, Y.; Yang, F. Flavonoid Compounds in Hippophae rhamnoides L. Protect Endothelial Cells from Oxidative Damage Through the PI3K/AKT-ENOS Pathway. Chem. Biodivers. 2024, 21, e202400300. [Google Scholar] [CrossRef]
  210. Pal, C. Small Molecules Targeting Mitochondria: A Mechanistic Approach to Combating Doxorubicin-Induced Cardiotoxicity. Cardiovasc. Toxicol. 2024, 25, 216–247. [Google Scholar] [CrossRef]
  211. Mihaylova, R.; Gevrenova, R.; Petrova, A.; Savov, Y.; Zheleva-Dimitrova, D.; Balabanova, V.; Momekov, G.; Simeonova, R. Mitigating Effects of Tanacetum balsamita L. on Metabolic Dysfunction-Associated Fatty Liver Disease (MAFLD). Plants 2024, 13, 2086. [Google Scholar] [CrossRef]
  212. Sattari, M.; Amri, J.; Shahaboddin, M.E.; Sattari, M.; Tabatabaei-Malazy, O.; Azmon, M.; Meshkani, R.; Panahi, G. The Protective Effects of Fisetin in Metabolic Disorders: A Focus on Oxidative Stress and Associated Events. J. Diabetes Metab. Disord. 2024, 23, 1753–1771. [Google Scholar] [CrossRef] [PubMed]
  213. Mao, Y.; Zha, Y.; Zang, Y.; Gao, Y.; Sun, J.; Liu, Y.; Wang, Z.; Wei, Z.; Wang, M.; Yang, Y. Isorhamnetin Improves Diabetes-Induced Erectile Dysfunction in Rats through Activation of the PI3K/AKT/ENOS Signaling Pathway. Biomed. Pharmacother. 2024, 177, 116987. [Google Scholar] [CrossRef] [PubMed]
  214. Yang, T.; Xiao, Y.; Liu, S.; Luo, F.; Tang, D.; Yu, Y.; Xie, Y. Isorhamnetin Induces Cell Cycle Arrest and Apoptosis by Triggering DNA Damage and Regulating the AMPK/MTOR/P70S6K Signaling Pathway in Doxorubicin-Resistant Breast Cancer. Phytomedicine 2023, 114, 154780. [Google Scholar] [CrossRef]
  215. Song, B.; Yang, P.; Zhang, S. Cell Fate Regulation Governed by P53: Friends or Reversible Foes in Cancer Therapy. Cancer Commun. 2024, 44, 297–360. [Google Scholar] [CrossRef]
  216. Ajiboye, B.O.; Famusiwa, C.D.; Falode, J.A.; Ojelabi, A.O.; Mistura, A.N.; Ogunbiyi, D.O.; Jeje, T.O.; Akinlolu, O.S.; Ogedengbe, O.O.; Ojo, O.A. Ocimum gratissimum L. Leaf Flavonoid-Rich Extracts Reduced the Expression of P53 and VCAM in Streptozotocin-Induced Cardiomyopathy Rats. Phytomedicine Plus 2024, 4, 100548. [Google Scholar] [CrossRef]
  217. Efe, G.; Rustgi, A.K.; Prives, C. P53 at the Crossroads of Tumor Immunity. Nat. Cancer 2024, 5, 983–995. [Google Scholar] [CrossRef]
  218. Peuget, S.; Zhou, X.; Selivanova, G. Translating P53-Based Therapies for Cancer into the Clinic. Nat. Rev. Cancer 2024, 24, 192–215. [Google Scholar] [CrossRef]
  219. Fischer, M.; Sammons, M.A. Determinants of P53 DNA Binding, Gene Regulation, and Cell Fate Decisions. Cell Death Differ. 2024, 31, 836–843. [Google Scholar] [CrossRef]
  220. Rana, J.N.; Mumtaz, S.; Han, I.; Choi, E.H. Harnessing the Synergy of Nanosecond High-Power Microwave Pulses and Cisplatin to Increase the Induction of Apoptosis in Cancer Cells through the Activation of ATR/ATM and Intrinsic Pathways. Free Radic. Biol. Med. 2024, 225, 221–235. [Google Scholar] [CrossRef]
  221. Rana, J.N.; Mumtaz, S.; Han, I.; Choi, E.H. Unveiling the Therapeutic Potential of Soft Plasma Jet and Nitric-Oxide Enriched Plasma-Activated Water (NO-PAW) on Oral Cancer YD-10B Cells: A Comprehensive Investigation of Direct and Indirect Treatments. Plasma Chem. Plasma Process. 2025, 1–28. [Google Scholar] [CrossRef]
  222. Wang, X.; Yang, J.; Yang, W.; Sheng, H.; Jia, B.; Cheng, P.; Xu, S.; Hong, X.; Jiang, C.; Yang, Y.; et al. Multiple Roles of P53 in Cancer Development: Regulation of Tumor Microenvironment, M6A Modification and Diverse Cell Death Mechanisms. J. Adv. Res. 2024. [Google Scholar] [CrossRef] [PubMed]
  223. Zhang, H.; Xu, J.; Long, Y.; Maimaitijiang, A.; Su, Z.; Li, W.; Li, J. Unraveling the Guardian: P53’s Multifaceted Role in the DNA Damage Response and Tumor Treatment Strategies. Int. J. Mol. Sci. 2024, 25, 12928. [Google Scholar] [CrossRef] [PubMed]
  224. Liu, Y.; Su, Z.; Tavana, O.; Gu, W. Understanding the Complexity of P53 in a New Era of Tumor Suppression. Cancer Cell 2024, 42, 946–967. [Google Scholar] [CrossRef]
  225. Indeglia, A.; Murphy, M.E. Elucidating the Chain of Command: Our Current Understanding of Critical Target Genes for P53-Mediated Tumor Suppression. Crit. Rev. Biochem. Mol. Biol. 2024, 59, 128–138. [Google Scholar] [CrossRef]
  226. Prakash, D.; Sudhandiran, G. Dietary Flavonoid Fisetin Regulates Aluminium Chloride-Induced Neuronal Apoptosis in Cortex and Hippocampus of Mice Brain. J. Nutr. Biochem. 2015, 26, 1527–1539. [Google Scholar] [CrossRef]
  227. Benavente-García, O.; Castillo, J. Update on Uses and Properties of Citrus Flavonoids: New Findings in Anticancer, Cardiovascular, and Anti-Inflammatory Activity. J. Agric. Food Chem. 2008, 56, 6185–6205. [Google Scholar] [CrossRef]
  228. Wang, H.; Guo, M.; Wei, H.; Chen, Y. Targeting P53 Pathways: Mechanisms, Structures and Advances in Therapy. Signal Transduct. Target. Ther. 2023, 8, 92. [Google Scholar] [CrossRef]
  229. Carceller, J.M.; Martínez Galán, J.P.; Monti, R.; Bassan, J.C.; Filice, M.; Iborra, S.; Yu, J.; Corma, A. Selective Synthesis of Citrus Flavonoids Prunin and Naringenin Using Heterogeneized Biocatalyst on Graphene Oxide. Green Chem. 2019, 21, 839–849. [Google Scholar] [CrossRef]
  230. Vila-Real, H.; Alfaia, A.J.; Bronze, M.R.; Calado, A.R.T.; Ribeiro, M.H.L. Enzymatic Synthesis of the Flavone Glucosides, Prunin and Isoquercetin, and the Aglycones, Naringenin and Quercetin, with Selective α-L-Rhamnosidase and β-D-Glucosidase Activities of Naringinase. Enzym. Res. 2011, 2011, 692618. [Google Scholar] [CrossRef]
  231. Uchida, Y.; Ferdousi, F.; Zheng, Y.-W.; Oda, T.; Isoda, H. Global Gene Expression Profiling Reveals Isorhamnetin Induces Hepatic-Lineage Specific Differentiation in Human Amniotic Epithelial Cells. Front. Cell Dev. Biol. 2020, 8, 578036. [Google Scholar] [CrossRef]
  232. Mohamed, E.M.; Hetta, M.H.; Rateb, M.E.; Selim, M.A.; AboulMagd, A.M.; Badria, F.A.; Abdelmohsen, U.R.; Alhadrami, H.A.; Hassan, H.M. Bioassay-Guided Isolation, Metabolic Profiling, and Docking Studies of Hyaluronidase Inhibitors from Ravenala madagascariensis. Molecules 2020, 25, 1714. [Google Scholar] [CrossRef]
  233. Chau, T.P.; Devanesan, S.; Ayub, R.; Perumal, K. Identification and Characterization of Major Bioactive Compounds from Andrographis paniculata (Burm. f.) Extracts Showed Multi-Biomedical Applications. Environ. Res. 2024, 242, 117763. [Google Scholar] [CrossRef] [PubMed]
  234. Mojzis, J.; Varinska, L.; Mojzisova, G.; Kostova, I.; Mirossay, L. Antiangiogenic Effects of Flavonoids and Chalcones. Pharmacol. Res. 2008, 57, 259–265. [Google Scholar] [CrossRef] [PubMed]
  235. Kim, E.K.; Choi, E.-J. Pathological Roles of MAPK Signaling Pathways in Human Diseases. Biochim. Biophys. Acta-Mol. Basis Dis. 2010, 1802, 396–405. [Google Scholar] [CrossRef] [PubMed]
  236. Saleem, S. Targeting MAPK Signaling: A Promising Approach for Treating Inflammatory Lung Disease. Pathol.-Res. Pract. 2024, 254, 155122. [Google Scholar] [CrossRef]
  237. Edvinsson, L.; Krause, D.N. Switching Off Vascular MAPK Signaling: A Novel Strategy to Prevent Delayed Cerebral Ischemia Following Subarachnoid Hemorrhage. Transl. Stroke Res. 2024. [Google Scholar] [CrossRef]
  238. Lin, H.-H. An Alternative Mode of GPCR Transactivation: Activation of GPCRs by Adhesion GPCRs. Int. J. Mol. Sci. 2025, 26, 552. [Google Scholar] [CrossRef]
  239. Roskoski, R. ERK1/2 MAP Kinases: Structure, Function, and Regulation. Pharmacol. Res. 2012, 66, 105–143. [Google Scholar] [CrossRef]
  240. Martínez-Rodríguez, O.P.; Thompson-Bonilla, M.d.R.; Jaramillo-Flores, M.E. Association between Obesity and Breast Cancer: Molecular Bases and the Effect of Flavonoids in Signaling Pathways. Crit. Rev. Food Sci. Nutr. 2020, 60, 3770–3792. [Google Scholar] [CrossRef]
  241. Shi, X.; Yu, Q.; Wang, K.; Fu, Y.; Zhang, S.; Liao, Z.; Li, Y.; Cai, T. Active Ingredients Isorhamnetin of Croci srigma Inhibit Stomach Adenocarcinomas Progression by MAPK/MTOR Signaling Pathway. Sci. Rep. 2023, 13, 12607. [Google Scholar] [CrossRef] [PubMed]
  242. Qiu, S.; Sun, G.; Zhang, Y.; Li, X.; Wang, R. Involvement of the NF-ΚB Signaling Pathway in the Renoprotective Effects of Isorhamnetin in a Type 2 Diabetic Rat Model. Biomed. Rep. 2016, 4, 628–634. [Google Scholar] [CrossRef]
  243. Gao, L.; Yao, R.; Liu, Y.; Wang, Z.; Huang, Z.; Du, B.; Zhang, D.; Wu, L.; Xiao, L.; Zhang, Y. Isorhamnetin Protects against Cardiac Hypertrophy through Blocking PI3K–AKT Pathway. Mol. Cell. Biochem. 2017, 429, 167–177. [Google Scholar] [CrossRef] [PubMed]
  244. Lu, X.; Liu, T.; Chen, K.; Xia, Y.; Dai, W.; Xu, S.; Xu, L.; Wang, F.; Wu, L.; Li, J.; et al. Isorhamnetin: A Hepatoprotective Flavonoid Inhibits Apoptosis and Autophagy via P38/PPAR-α Pathway in Mice. Biomed. Pharmacother. 2018, 103, 800–811. [Google Scholar] [CrossRef] [PubMed]
  245. Hwang, S.-L.; Yen, G.-C. Modulation of Akt, JNK, and P38 Activation Is Involved in Citrus Flavonoid-Mediated Cytoprotection of PC12 Cells Challenged by Hydrogen Peroxide. J. Agric. Food Chem. 2009, 57, 2576–2582. [Google Scholar] [CrossRef]
  246. Hwang, S.-L.; Shih, P.-H.; Yen, G.-C. Neuroprotective Effects of Citrus Flavonoids. J. Agric. Food Chem. 2012, 60, 877–885. [Google Scholar] [CrossRef]
  247. Jayashankar, B.; Mishra, K.P.; Kumar, M.S.Y.; Udayasankar, K.; Misra, K.; Ganju, L.; Singh, S.B. A Supercritical CO2 Extract from Seabuckthorn Leaves Inhibits Pro-Inflammatory Mediators via Inhibition of Mitogen Activated Protein Kinase P38 and Transcription Factor Nuclear Factor-ΚB. Int. Immunopharmacol. 2012, 13, 461–467. [Google Scholar] [CrossRef]
  248. Lian, J.-J.; Cheng, B.-F.; Gao, Y.-X.; Xue, H.; Wang, L.; Wang, M.; Yang, H.-J.; Feng, Z.-W. Protective Effect of Kaempferol, a Flavonoid Widely Present in Varieties of Edible Plants, on IL-1β-Induced Inflammatory Response via Inhibiting MAPK, Akt, and NF-ΚB Signalling in SW982 Cells. J. Funct. Foods 2016, 27, 214–222. [Google Scholar] [CrossRef]
  249. Cai, J.; Wen, H.; Zhou, H.; Zhang, D.; Lan, D.; Liu, S.; Li, C.; Dai, X.; Song, T.; Wang, X.; et al. Naringenin: A Flavanone with Anti-Inflammatory and Anti-Infective Properties. Biomed. Pharmacother. 2023, 164, 114990. [Google Scholar] [CrossRef]
  250. Abdel Bar, F.M.; Alonazi, R.; Elekhnawy, E.; Samra, R.M.; Alqarni, M.H.; Badreldin, H.; Magdy, G. HPLC-PDA and in vivo Anti-Inflammatory Potential of Isorhamnetin-3-O-β-D-Glucoside from Zygophyllum simplex L. J. Ethnopharmacol. 2025, 338, 119089. [Google Scholar] [CrossRef]
  251. Jnawali, H.N.; Jeon, D.; Jeong, M.-C.; Lee, E.; Jin, B.; Ryoo, S.; Yoo, J.; Jung, I.D.; Lee, S.J.; Park, Y.-M.; et al. Antituberculosis Activity of a Naturally Occurring Flavonoid, Isorhamnetin. J. Nat. Prod. 2016, 79, 961–969. [Google Scholar] [CrossRef] [PubMed]
  252. Jazvinšćak Jembrek, M.; Oršolić, N.; Mandić, L.; Sadžak, A.; Šegota, S. Anti-Oxidative, Anti-Inflammatory and Anti-Apoptotic Effects of Flavonols: Targeting Nrf2, NF-ΚB and P53 Pathways in Neurodegeneration. Antioxidants 2021, 10, 1628. [Google Scholar] [CrossRef]
  253. Asgharian, P.; Tazekand, A.P.; Hosseini, K.; Forouhandeh, H.; Ghasemnejad, T.; Ranjbar, M.; Hasan, M.; Kumar, M.; Beirami, S.M.; Tarhriz, V.; et al. Potential Mechanisms of Quercetin in Cancer Prevention: Focus on Cellular and Molecular Targets. Cancer Cell Int. 2022, 22, 257. [Google Scholar] [CrossRef]
  254. Kumari, N.; Radha; Kumar, M.; Puri, S.; Zhang, B.; Rais, N.; Pundir, A.; Chandran, D.; Raman, P.; Dhumal, S.; et al. Peach (Prunus persica (L.) Batsch) Seeds and Kernels as Potential Plant-Based Functional Food Ingredients: A Review of Bioactive Compounds and Health-Promoting Activities. Food Biosci. 2023, 54, 102914. [Google Scholar] [CrossRef]
  255. Chi, G.; Zhong, W.; Liu, Y.; Lu, G.; Lü, H.; Wang, D.; Sun, F. Isorhamnetin Protects Mice from Lipopolysaccharide-Induced Acute Lung Injury via the Inhibition of Inflammatory Responses. Inflamm. Res. 2016, 65, 33–41. [Google Scholar] [CrossRef] [PubMed]
  256. Yang, B.; Ma, L.; Wei, Y.; Cui, Y.; Li, X.; Wei, Y.; Zhang, S.; Zhang, L.; Zhou, H.; Wang, G.; et al. Isorhamnetin Alleviates Lipopolysaccharide-Induced Acute Lung Injury by Inhibiting MTOR Signaling Pathway. Immunopharmacol. Immunotoxicol. 2022, 44, 387–399. [Google Scholar] [CrossRef]
  257. Liu, G.; Jiang, C.; Li, D.; Yao, L.; Lin, Y.; Wang, B.; Qiu, J.; Wang, W.; Wang, W. Isorhamnetin Alleviates Esophageal Mucosal Injury in a Chronic Model of Reflux Esophagitis. Eur. J. Pharmacol. 2019, 864, 172720. [Google Scholar] [CrossRef] [PubMed]
  258. Sun, Q.; Liu, Q.; Zhou, X.; Wang, X.; Li, H.; Zhang, W.; Yuan, H.; Sun, C. Flavonoids Regulate Tumor-Associated Macrophages–From Structure-Activity Relationship to Clinical Potential (Review). Pharmacol. Res. 2022, 184, 106419. [Google Scholar] [CrossRef]
  259. Shahrezaei, A.; Sohani, M.; Sohouli, M.; Taherkhani, S.; Nasirinezhad, F. The Involvement and Significance of M2 Macrophages in Neuropathic Pain Following Spinal Cord Injury: A Systematic Review. J. Physiol. Sci. 2024, 74, 45. [Google Scholar] [CrossRef]
  260. Zhang, Y.; Zhu, K.; Wang, X.; Zhao, Y.; Shi, J.; Liu, Z. Roles of IL-4, IL-13, and Their Receptors in Lung Cancer. J. Interferon Cytokine Res. 2024, 44, 399–407. [Google Scholar] [CrossRef]
  261. Guo, J.; Yan, W.; Duan, H.; Wang, D.; Zhou, Y.; Feng, D.; Zheng, Y.; Zhou, S.; Liu, G.; Qin, X. Therapeutic Effects of Natural Products on Liver Cancer and Their Potential Mechanisms. Nutrients 2024, 16, 1642. [Google Scholar] [CrossRef]
  262. Hoskin, D.W.; Coombs, M.R.P. Editorial: Immune Modulation by Flavonoids. Front. Immunol. 2022, 13, 899577. [Google Scholar] [CrossRef] [PubMed]
  263. Gong, G.; Guan, Y.-Y.; Zhang, Z.-L.; Rahman, K.; Wang, S.-J.; Zhou, S.; Luan, X.; Zhang, H. Isorhamnetin: A Review of Pharmacological Effects. Biomed. Pharmacother. 2020, 128, 110301. [Google Scholar] [CrossRef]
  264. Wang, H.; Zhang, Q.; Liang Cheng, M.; Ma, L.; Zhi Meng, Q.; Duan, L.; Chen, Y.; Wu Tan, J.; Chen, M.; Ting Liang, T.; et al. Effect of the Miaoyao Fanggan Sachet-Derived Isorhamnetin on TLR2/4 and NKp46 Expression in Mice. J. Ethnopharmacol. 2012, 144, 138–144. [Google Scholar] [CrossRef] [PubMed]
  265. Upadhaya, P.; Lamenza, F.F.; Shrestha, S.; Roth, P.; Jagadeesha, S.; Pracha, H.; Horn, N.A.; Oghumu, S. Berry Extracts and Their Bioactive Compounds Mitigate LPS and DNFB-Mediated Dendritic Cell Activation and Induction of Antigen Specific T-Cell Effector Responses. Antioxidants 2023, 12, 1667. [Google Scholar] [CrossRef] [PubMed]
  266. Hosseinzade, A.; Sadeghi, O.; Naghdipour Biregani, A.; Soukhtehzari, S.; Brandt, G.S.; Esmaillzadeh, A. Immunomodulatory Effects of Flavonoids: Possible Induction of T CD4+ Regulatory Cells Through Suppression of MTOR Pathway Signaling Activity. Front. Immunol. 2019, 10, 51. [Google Scholar] [CrossRef]
  267. Chen, Q.; Song, S.; Wang, Z.; Shen, Y.; Xie, L.; Li, J.; Jiang, L.; Zhao, H.; Feng, X.; Zhou, Y.; et al. Isorhamnetin Induces the Paraptotic Cell Death through ROS and the ERK/MAPK Pathway in OSCC Cells. Oral Dis. 2021, 27, 240–250. [Google Scholar] [CrossRef]
  268. Jaramillo, S.; Lopez, S.; Varela, L.M.; Rodriguez-Arcos, R.; Jimenez, A.; Abia, R.; Guillen, R.; Muriana, F.J.G. The Flavonol Isorhamnetin Exhibits Cytotoxic Effects on Human Colon Cancer Cells. J. Agric. Food Chem. 2010, 58, 10869–10875. [Google Scholar] [CrossRef]
  269. Yuan, J.; Ofengeim, D. A Guide to Cell Death Pathways. Nat. Rev. Mol. Cell Biol. 2024, 25, 379–395. [Google Scholar] [CrossRef]
  270. Meier, P.; Legrand, A.J.; Adam, D.; Silke, J. Immunogenic Cell Death in Cancer: Targeting Necroptosis to Induce Antitumour Immunity. Nat. Rev. Cancer 2024, 24, 299–315. [Google Scholar] [CrossRef]
  271. Pham, D.-C.; Shibu, M.A.; Mahalakshmi, B.; Velmurugan, B.K. Effects of Phytochemicals on Cellular Signaling: Reviewing Their Recent Usage Approaches. Crit. Rev. Food Sci. Nutr. 2020, 60, 3522–3546. [Google Scholar] [CrossRef] [PubMed]
  272. Jayashankar, B.; Singh, D.; Tanwar, H.; Mishra, K.P.; Murthy, S.; Chanda, S.; Mishra, J.; Tulswani, R.; Misra, K.; Singh, S.B.; et al. Augmentation of Humoral and Cellular Immunity in Response to Tetanus and Diphtheria Toxoids by Supercritical Carbon Dioxide Extracts of Hippophae rhamnoides L. Leaves. Int. Immunopharmacol. 2017, 44, 123–136. [Google Scholar] [CrossRef] [PubMed]
  273. Hosseinzadeh, A.; Poursoleiman, F.; Biregani, A.N.; Esmailzadeh, A. Flavonoids Target Different Molecules of Autophagic and Metastatic Pathways in Cancer Cells. Cancer Cell Int. 2023, 23, 114. [Google Scholar] [CrossRef] [PubMed]
  274. Rajabi, M.; Mousa, S.A. The Role of Angiogenesis in Cancer Treatment. Biomedicines 2017, 5, 34. [Google Scholar] [CrossRef]
  275. Liskova, A.; Koklesova, L.; Samec, M.; Smejkal, K.; Samuel, S.M.; Varghese, E.; Abotaleb, M.; Biringer, K.; Kudela, E.; Danko, J.; et al. Flavonoids in Cancer Metastasis. Cancers 2020, 12, 1498. [Google Scholar] [CrossRef]
  276. Wei, Q.; Zhang, Y. Flavonoids with Anti-Angiogenesis Function in Cancer. Molecules 2024, 29, 1570. [Google Scholar] [CrossRef]
  277. Weng, C.-J.; Yen, G.-C. Flavonoids, a Ubiquitous Dietary Phenolic Subclass, Exert Extensive in vitro Anti-Invasive and in vivo Anti-Metastatic Activities. Cancer Metastasis Rev. 2012, 31, 323–351. [Google Scholar] [CrossRef]
  278. Masarkar, N.; Pal, M.; Roy, M.; Yadav, A.K.; Pandya, B.; Lokhande, S.; Kanwar, J.R.; Ray, S.K.; Mukherjee, S. In-silico Screening of Bioactive Compounds of Moringa oleifera as Potential Inhibitors Targeting HIF-1α/VEGF/GLUT-1 Pathway against Breast Cancer. J. Complement. Integr. Med. 2024, 22, 149–164. [Google Scholar] [CrossRef]
  279. Carmeliet, P. VEGF as a Key Mediator of Angiogenesis in Cancer. Oncology 2005, 69, 4–10. [Google Scholar] [CrossRef]
  280. Shaw, P.; Dwivedi, S.K.D.; Bhattacharya, R.; Mukherjee, P.; Rao, G. VEGF Signaling: Role in Angiogenesis and Beyond. Biochim. Biophys. Acta-Rev. Cancer 2024, 1879, 189079. [Google Scholar] [CrossRef]
  281. Bhattacharya, R.; Brown, J.S.; Gatenby, R.A.; Ibrahim-Hashim, A. A Gene for All Seasons: The Evolutionary Consequences of HIF-1 in Carcinogenesis, Tumor Growth and Metastasis. Semin. Cancer Biol. 2024, 102–103, 17–24. [Google Scholar] [CrossRef] [PubMed]
  282. Razzaque, S.; Abubakar, M.; Farid, M.A.; Zia, R.; Nazir, S.; Razzaque, H.; Ali, A.; Ali, Z.; Mahmood, A.; Al-Masry, W.; et al. Detection of Toxic Cypermethrin Pesticides in Drinking Water by Simple Graphitic Electrode Modified with Kraft Lignin@Ni@g-C3N4 Nano-Composite. J. Mater. Chem. B 2024, 12, 9364–9374. [Google Scholar] [CrossRef] [PubMed]
  283. Laack, E.; Scheffler, A.; Burkholder, I.; Boeters, I.; Andritzky, B.; Schuch, G.; Görn, M.; Vohwinkel, G.; Edler, L.; Fiedler, W.; et al. Pretreatment Vascular Endothelial Growth Factor (VEGF) and Matrix Metalloproteinase-9 (MMP-9) Serum Levels in Patients with Metastatic Non-Small Cell Lung Cancer (NSCLC). Lung Cancer 2005, 50, 51–58. [Google Scholar] [CrossRef]
  284. Kpeli, G.W.; Conrad, K.M.; Bralower, W.; Byrne, C.E.; Boue, S.M.; Burow, M.E.; Mondrinos, M.J. Xenohormetic Phytochemicals Inhibit Neovascularization in Microphysiological Models of Vasculogenesis and Tumor Angiogenesis. Adv. Biol. 2024, 8, 2300480. [Google Scholar] [CrossRef]
  285. Zhu, Y.; Sun, L.; Zhang, H.; Li, Y.; Lai, S. Effects of Isorhamnetin on Protein Expression of VEGF, MMP-2 and Endostatin in Lewis Lung Cancer Mouse. Int. J. Clin. Exp. Med. 2017, 10, 11488–11495. [Google Scholar]
  286. Kang, L.; Gao, X.-H.; Liu, H.-R.; Men, X.; Wu, H.-N.; Cui, P.-W.; Oldfield, E.; Yan, J.-Y. Structure–Activity Relationship Investigation of Coumarin–Chalcone Hybrids with Diverse Side-Chains as Acetylcholinesterase and Butyrylcholinesterase Inhibitors. Mol. Divers. 2018, 22, 893–906. [Google Scholar] [CrossRef]
  287. Biswas, P.; Kaium, M.A.; Islam Tareq, M.M.; Tauhida, S.J.; Hossain, M.R.; Siam, L.S.; Parvez, A.; Bibi, S.; Hasan, M.H.; Rahman, M.M.; et al. The Experimental Significance of Isorhamnetin as an Effective Therapeutic Option for Cancer: A Comprehensive Analysis. Biomed. Pharmacother. 2024, 176, 116860. [Google Scholar] [CrossRef] [PubMed]
  288. Zeng, G.; Wu, Z.; Cao, W.; Wang, Y.; Deng, X.; Zhou, Y. Identification of Anti-Nociceptive Constituents from the Pollen of Typha angustifolia L. Using Effect-Directed Fractionation. Nat. Prod. Res. 2020, 34, 1041–1045. [Google Scholar] [CrossRef]
  289. Hui, Q.; Yang, N.; Xiong, C.; Zhou, S.; Zhou, X.; Jin, Q.; Xu, X. Isorhamnetin Suppresses the Epithelial-Mesenchymal Transition of the Retinal Pigment Epithelium Both in Vivo and in Vitro through Nrf2-Dependent AKT/GSK-3β Pathway. Exp. Eye Res. 2024, 240, 109823. [Google Scholar] [CrossRef]
  290. Zhang, Z.; Zhang, H.; Shi, J.; Wang, Z.; Liang, Y.; Yu, J.; Wang, H.; Song, Z.; Tang, Z.; Zhang, D.; et al. Isorhamnetin Alleviates Renal Fibrosis by Inducing Endogenous Hydrogen Sulfide and Regulating Thiol-Based Redox State in Obstructed Kidneys. Biomolecules 2024, 14, 1233. [Google Scholar] [CrossRef]
  291. Martínez, G.; Mijares, M.R.; De Sanctis, J.B. Effects of Flavonoids and Its Derivatives on Immune Cell Responses. Recent Pat. Inflamm. Allergy Drug Discov. 2019, 13, 84–104. [Google Scholar] [CrossRef]
  292. Shakoor, H.; Feehan, J.; Apostolopoulos, V.; Platat, C.; Al Dhaheri, A.S.; Ali, H.I.; Ismail, L.C.; Bosevski, M.; Stojanovska, L. Immunomodulatory Effects of Dietary Polyphenols. Nutrients 2021, 13, 728. [Google Scholar] [CrossRef]
  293. Kim, M.H. Flavonoids Inhibit VEGF/BFGF-Induced Angiogenesis in Vitro by Inhibiting the Matrix-Degrading Proteases. J. Cell. Biochem. 2003, 89, 529–538. [Google Scholar] [CrossRef] [PubMed]
  294. Khater, M.; Greco, F.; Osborn, H.M.I. Antiangiogenic Activity of Flavonoids: A Systematic Review and Meta-Analysis. Molecules 2020, 25, 4712. [Google Scholar] [CrossRef]
  295. Shanmugavadivu, A.; Balagangadharan, K.; Selvamurugan, N. Angiogenic and Osteogenic Effects of Flavonoids in Bone Regeneration. Biotechnol. Bioeng. 2022, 119, 2313–2330. [Google Scholar] [CrossRef] [PubMed]
  296. Dewanjee, S.; Chakraborty, P.; Bhattacharya, H.; Singh, S.K.; Dua, K.; Dey, A.; Jha, N.K. Recent Advances in Flavonoid-Based Nanocarriers as an Emerging Drug Delivery Approach for Cancer Chemotherapy. Drug Discov. Today 2023, 28, 103409. [Google Scholar] [CrossRef] [PubMed]
  297. Cai, J.; Tan, X.; Hu, Q.; Pan, H.; Zhao, M.; Guo, C.; Zeng, J.; Ma, X.; Zhao, Y. Flavonoids and Gastric Cancer Therapy: From Signaling Pathway to Therapeutic Significance. Drug Des. Dev. Ther. 2024, 18, 3233–3253. [Google Scholar] [CrossRef]
  298. van den Boogaard, W.M.C.; Komninos, D.S.J.; Vermeij, W.P. Chemotherapy Side-Effects: Not All DNA Damage Is Equal. Cancers 2022, 14, 627. [Google Scholar] [CrossRef]
  299. Słonimska, P.; Sachadyn, P.; Zieliński, J.; Skrzypski, M.; Pikuła, M. Chemotherapy-Mediated Complications of Wound Healing: An Understudied Side Effect. Adv. Wound Care 2024, 13, 187–199. [Google Scholar] [CrossRef]
  300. Labe, S.; Jones, G.; Dailey, H.; Bhasker, J.; Kanwar, R.; Crago, M.; Fitzgerald, B.; Mikhail, D.; Hafiz, S.; Kramer, C.; et al. D-CRSE: Diminishing Chemotherapy-Related Side Effects through Patient Education, a Mixed-Methods Pilot Study. J. Psychosoc. Oncol. 2025, 43, 1–15. [Google Scholar] [CrossRef]
  301. Zhai, K.; Mazurakova, A.; Koklesova, L.; Kubatka, P.; Büsselberg, D. Flavonoids Synergistically Enhance the Anti-Glioblastoma Effects of Chemotherapeutic Drugs. Biomolecules 2021, 11, 1841. [Google Scholar] [CrossRef] [PubMed]
  302. Hussain, S.A.; Sulaiman, A.A.; Balch, C.; Chauhan, H.; Alhadidi, Q.M.; Tiwari, A.K. Natural Polyphenols in Cancer Chemoresistance. Nutr. Cancer 2016, 68, 879–891. [Google Scholar] [CrossRef]
  303. Zughaibi, T.A.; Suhail, M.; Tarique, M.; Tabrez, S. Targeting PI3K/Akt/MTOR Pathway by Different Flavonoids: A Cancer Chemopreventive Approach. Int. J. Mol. Sci. 2021, 22, 12455. [Google Scholar] [CrossRef]
  304. Jiang, C.; Xie, N.; Sun, T.; Ma, W.; Zhang, B.; Li, W. Xanthohumol Inhibits TGF-Β1-Induced Cardiac Fibroblasts Activation via Mediating PTEN/Akt/MTOR Signaling Pathway. Drug Des. Devel. Ther. 2020, 14, 5431–5439. [Google Scholar] [CrossRef]
  305. Talapko, J.; Talapko, D.; Katalinić, D.; Kotris, I.; Erić, I.; Belić, D.; Vasilj Mihaljević, M.; Vasilj, A.; Erić, S.; Flam, J.; et al. Health Effects of Ionizing Radiation on the Human Body. Medicina 2024, 60, 653. [Google Scholar] [CrossRef] [PubMed]
  306. Lohani, M.; Ahuja, M.; Buabeid, M.A.; Schwartz, D.; Shannon, D.; Suppiramaniam, V.; Kemppainen, B.; Dhanasekaran, M. Anti-Oxidative and DNA Protecting Effects of Flavonoids-Rich Scutellaria lateriflora. Nat. Prod. Commun. 2013, 8, 1934578X1300801019. [Google Scholar] [CrossRef]
  307. Arcas, M.C.; Botía, J.M.; Ortuño, A.M.; Del Río, J.A. UV Irradiation Alters the Levels of Flavonoids Involved in the Defence Mechanism of Citrus Aurantium Fruits against Penicillium Digitatum. Eur. J. Plant Pathol. 2000, 106, 617–622. [Google Scholar] [CrossRef]
  308. Yahyapour, R.; Shabeeb, D.; Cheki, M.; Musa, A.E.; Farhood, B.; Rezaeyan, A.; Amini, P.; Fallah, H.; Najafi, M. Radiation Protection and Mitigation by Natural Antioxidants and Flavonoids: Implications to Radiotherapy and Radiation Disasters. Curr. Mol. Pharmacol. 2018, 11, 285–304. [Google Scholar] [CrossRef]
  309. Tiwari, P.; Mishra, K.P. Flavonoids Sensitize Tumor Cells to Radiation: Molecular Mechanisms and Relevance to Cancer Radiotherapy. Int. J. Radiat. Biol. 2020, 96, 360–369. [Google Scholar] [CrossRef]
  310. Singh, S.; Ahuja, A.; Sharma, H.; Maheshwari, P. An Overview of Dietary Flavonoids as a Nutraceutical Nanoformulation Approach to Life-Threatening Diseases. Curr. Pharm. Biotechnol. 2023, 24, 1740–1773. [Google Scholar] [CrossRef]
  311. Fonseca, M.; Rehman, M.; Soares, R.; Fonte, P. The Impact of Flavonoid-Loaded Nanoparticles in the UV Protection and Safety Profile of Topical Sunscreens. Biomolecules 2023, 13, 493. [Google Scholar] [CrossRef] [PubMed]
  312. Jasim, A.J.; Sulaiman, G.M.; Ay, H.; Mohammed, S.A.A.; Mohammed, H.A.; Jabir, M.S.; Khan, R.A. Preliminary Trials of the Gold Nanoparticles Conjugated Chrysin: An Assessment of Anti-Oxidant, Anti-Microbial, and in Vitro Cytotoxic Activities of a Nanoformulated Flavonoid. Nanotechnol. Rev. 2022, 11, 2726–2741. [Google Scholar] [CrossRef]
  313. Teng, H.; Zheng, Y.; Cao, H.; Huang, Q.; Xiao, J.; Chen, L. Enhancement of Bioavailability and Bioactivity of Diet-Derived Flavonoids by Application of Nanotechnology: A Review. Crit. Rev. Food Sci. Nutr. 2023, 63, 378–393. [Google Scholar] [CrossRef] [PubMed]
  314. Zhao, J.; Yang, J.; Xie, Y. Improvement Strategies for the Oral Bioavailability of Poorly Water-Soluble Flavonoids: An Overview. Int. J. Pharm. 2019, 570, 118642. [Google Scholar] [CrossRef]
  315. Zverev, Y.F.; Rykunova, A.Y. Modern Nanocarriers as a Factor in Increasing the Bioavailability and Pharmacological Activity of Flavonoids. Appl. Biochem. Microbiol. 2022, 58, 1002–1020. [Google Scholar] [CrossRef] [PubMed]
  316. Mohite, P.; Puri, A.; Bharati, D.; Singh, S. Polyphenol-Encapsulated Nanoparticles for the Treatment of Chronic Metabolic Diseases. In Role of Flavonoids in Chronic Metabolic Diseases; Scrivener Publishing LLC: Austin, TX, USA, 2024; pp. 375–416. ISBN 9781394238071. [Google Scholar]
  317. Chen, H.; Wang, G.; Li, X.; Wang, J.; Wang, X.; Wang, Y.; Liu, Z.; Liu, J.; Ding, Y.; Guo, J.; et al. Multi-Functional D-Alpha-Tocopheryl Polyethylene Glycol Succinate Surface Modified Nanocrystals Improve the Stability and Oral Bioavailability of Pueraria Flavonoids. J. Drug Deliv. Sci. Technol. 2024, 95, 105623. [Google Scholar] [CrossRef]
  318. Puspadewi, R.; Milanda, T.; Muhaimin, M.; Chaerunisaa, A.Y. Nanoparticle-Encapsulated Plant Polyphenols and Flavonoids as an Enhanced Delivery System for Anti-Acne Therapy. Pharmaceuticals 2025, 18, 209. [Google Scholar] [CrossRef]
  319. Mustafa, G.M.; Younas, B.; Waseem, M.; Ateeq-ur-Rehman; Noor, N.A.; Elhindi, K.M.; Mumtaz, S. Investigation of Optoelectronic and Thermoelectric Characteristics of Tl2Os(Cl/Br)6 Double Perovskites for Renewable Energy Applications. Mater. Sci. Semicond. Process. 2025, 192, 109420. [Google Scholar] [CrossRef]
  320. Ciceu, A.; Fenyvesi, F.; Hermenean, A.; Ardelean, S.; Dumitra, S.; Puticiu, M. Advancements in Plant-Based Therapeutics for Hepatic Fibrosis: Molecular Mechanisms and Nanoparticulate Drug Delivery Systems. Int. J. Mol. Sci. 2024, 25, 9346. [Google Scholar] [CrossRef]
  321. Li, N.; Wang, M.; Lyu, Z.; Shan, K.; Chen, Z.; Chen, B.; Chen, Y.; Hu, X.; Dou, B.; Zhang, J.; et al. Medicinal Plant-Based Drug Delivery System for Inflammatory Bowel Disease. Front. Pharmacol. 2023, 14, 1158945. [Google Scholar] [CrossRef]
  322. Sezgin-Bayindir, Z.; Losada-Barreiro, S.; Bravo-Díaz, C.; Sova, M.; Kristl, J.; Saso, L. Nanotechnology-Based Drug Delivery to Improve the Therapeutic Benefits of NRF2 Modulators in Cancer Therapy. Antioxidants 2021, 10, 685. [Google Scholar] [CrossRef] [PubMed]
  323. Zhang, D.; Song, J.; Jing, Z.; Qin, H.; Wu, Y.; Zhou, J.; Zang, X. Stimulus Responsive Nanocarrier for Enhanced Antitumor Responses Against Hepatocellular Carcinoma. Int. J. Nanomed. 2024, 19, 13339–13355. [Google Scholar] [CrossRef]
  324. Sharma, A.; Wairkar, S. Flavonoids for Treating Pulmonary Fibrosis: Present Status and Future Prospects. Phyther. Res. 2024, 38, 4406–4423. [Google Scholar] [CrossRef] [PubMed]
  325. Majid, I.; Majid, D.; Makroo, H.A.; Dar, B.N. Enhancing the Bioavailability and Gut Health Benefits of Quercetin from Sprouted Onions: A Comprehensive Review in the Context of Food-Derived Bioactives. Food Chem. Adv. 2024, 4, 100725. [Google Scholar] [CrossRef]
  326. Attar, E.S.; Chaudhari, V.H.; Deokar, C.G.; Dyawanapelly, S.; Devarajan, P. V Nano Drug Delivery Strategies for an Oral Bioenhanced Quercetin Formulation. Eur. J. Drug Metab. Pharmacokinet. 2023, 48, 495–514. [Google Scholar] [CrossRef]
  327. Arif, M.; Rauf, A.; Akhter, T. A Comprehensive Review on Crosslinked Network Systems of Zinc Oxide-Organic Polymer Composites. Int. J. Biol. Macromol. 2024, 274, 133250. [Google Scholar] [CrossRef]
  328. Shree Harini, K.; Ezhilarasan, D. Flavonoids-Based Nanomedicines for the Treatment of Liver Fibrosis: A Recent Progress. J. Drug Deliv. Sci. Technol. 2024, 93, 105467. [Google Scholar] [CrossRef]
  329. Gervasi, T.; Calderaro, A.; Barreca, D.; Tellone, E.; Trombetta, D.; Ficarra, S.; Smeriglio, A.; Mandalari, G.; Gattuso, G. Biotechnological Applications and Health-Promoting Properties of Flavonols: An Updated View. Int. J. Mol. Sci. 2022, 23, 1710. [Google Scholar] [CrossRef]
  330. Sharma, H.; Anand, A.; Halagali, P.; Inamdar, A.; Pathak, R.; Taghizadeh-Hesary, F.; Ashique, S. Advancement of Nanoengineered Flavonoids for Chronic Metabolic Diseases. In Role of Flavonoids in Chronic Metabolic Diseases; Scrivener Publishing LLC: Austin, TX, USA, 2024; pp. 459–510. ISBN 9781394238071. [Google Scholar]
  331. Arif, M.; Rauf, A.; Raza, H.; Moussa, S.B.; Haroon, S.M.; Alzahrani, A.Y.A.; Akhter, T. Catalytic Reduction of Nitroarenes by Palladium Nanoparticles Decorated Silica@poly(Chitosan-N-Isopropylacrylamide-Methacrylic Acid) Hybrid Microgels. Int. J. Biol. Macromol. 2024, 275, 133633. [Google Scholar] [CrossRef]
  332. Patel, D.; Sethi, N.; Patel, P.; Shah, S.; Patel, K. Exploring the Potential of P-Glycoprotein Inhibitors in the Targeted Delivery of Anti-Cancer Drugs: A Comprehensive Review. Eur. J. Pharm. Biopharm. 2024, 198, 114267. [Google Scholar] [CrossRef]
  333. Alexander, A.; Ajazuddin; Patel, R.J.; Saraf, S.; Saraf, S. Recent Expansion of Pharmaceutical Nanotechnologies and Targeting Strategies in the Field of Phytopharmaceuticals for the Delivery of Herbal Extracts and Bioactives. J. Control. Release 2016, 241, 110–124. [Google Scholar] [CrossRef] [PubMed]
  334. Wang, Y.; Tao, B.; Wan, Y.; Sun, Y.; Wang, L.; Sun, J.; Li, C. Drug Delivery Based Pharmacological Enhancement and Current Insights of Quercetin with Therapeutic Potential against Oral Diseases. Biomed. Pharmacother. 2020, 128, 110372. [Google Scholar] [CrossRef] [PubMed]
  335. Arif, M.; Rauf, A.; Akhter, T. A Review on Ag Nanoparticles Fabricated in Microgels. RSC Adv. 2024, 14, 19381–19399. [Google Scholar] [CrossRef]
  336. Vlachopoulos, A.; Karlioti, G.; Balla, E.; Daniilidis, V.; Kalamas, T.; Stefanidou, M.; Bikiaris, N.D.; Christodoulou, E.; Koumentakou, I.; Karavas, E.; et al. Poly(Lactic Acid)-Based Microparticles for Drug Delivery Applications: An Overview of Recent Advances. Pharmaceutics 2022, 14, 359. [Google Scholar] [CrossRef]
  337. Samborska, K.; Boostani, S.; Geranpour, M.; Hosseini, H.; Dima, C.; Khoshnoudi-Nia, S.; Rostamabadi, H.; Falsafi, S.R.; Shaddel, R.; Akbari-Alavijeh, S.; et al. Green Biopolymers from By-Products as Wall Materials for Spray Drying Microencapsulation of Phytochemicals. Trends Food Sci. Technol. 2021, 108, 297–325. [Google Scholar] [CrossRef]
  338. Khursheed, R.; Singh, S.K.; Wadhwa, S.; Kapoor, B.; Gulati, M.; Kumar, R.; Ramanunny, A.K.; Awasthi, A.; Dua, K. Treatment Strategies against Diabetes: Success so Far and Challenges Ahead. Eur. J. Pharmacol. 2019, 862, 172625. [Google Scholar] [CrossRef]
  339. Zhu, Q.; Sun, J.; An, C.; Li, X.; Xu, S.; He, Y.; Zhang, X.; Liu, L.; Hu, K.; Liang, M. Mechanism of LncRNA Gm2044 in Germ Cell Development. Front. Cell Dev. Biol. 2024, 12, 1410914. [Google Scholar] [CrossRef]
  340. Birnbaum, D.T.; Brannon-Peppas, L. Microparticle Drug Delivery Systems. In Drug Delivery Systems in Cancer Therapy; Brown, D.M., Ed.; Humana Press: Totowa, NJ, USA, 2004; pp. 117–135. ISBN 978-1-59259-427-6. [Google Scholar]
  341. Kohane, D.S. Microparticles and Nanoparticles for Drug Delivery. Biotechnol. Bioeng. 2007, 96, 203–209. [Google Scholar] [CrossRef]
  342. Kállai-Szabó, N.; Farkas, D.; Lengyel, M.; Basa, B.; Fleck, C.; Antal, I. Microparticles and Multi-Unit Systems for Advanced Drug Delivery. Eur. J. Pharm. Sci. 2024, 194, 106704. [Google Scholar] [CrossRef]
  343. Dong, Q.; Jiang, Z. Platinum–Iron Nanoparticles for Oxygen-Enhanced Sonodynamic Tumor Cell Suppression. Inorganics 2024, 12, 331. [Google Scholar] [CrossRef]
  344. Alizadeh-Sani, M.; Mohammadian, E.; Rhim, J.-W.; Jafari, S.M. PH-Sensitive (Halochromic) Smart Packaging Films Based on Natural Food Colorants for the Monitoring of Food Quality and Safety. Trends Food Sci. Technol. 2020, 105, 93–144. [Google Scholar] [CrossRef]
  345. Chanaj-Kaczmarek, J.; Paczkowska, M.; Osmałek, T.; Kaproń, B.; Plech, T.; Szymanowska, D.; Karaźniewicz-Łada, M.; Kobus-Cisowska, J.; Cielecka-Piontek, J. Hydrogel Delivery System Containing Calendulae Flos Lyophilized Extract with Chitosan as a Supporting Strategy for Wound Healing Applications. Pharmaceutics 2020, 12, 634. [Google Scholar] [CrossRef] [PubMed]
  346. Kar, K. Functionalization of Food Polyphenols for Nanodeliveries. In Polyphenols: Food, Nutraceutical, and Nanotherapeutic Applications; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2023; pp. 134–154. ISBN 9781394188864. [Google Scholar]
  347. Arif, M.; Raza, H.; Moussa, S.B.; Alzahrani, A.Y.A.; Akhter, T. Poly(Chitosan-N-Vinylcaprolactam-Methacrylic Acid) Microgels as Microreactor for Ag(I) Ions Extraction and in-Situ Silver Nanoparticles Formation to Reduce the Toxins. Int. J. Biol. Macromol. 2024, 282, 136906. [Google Scholar] [CrossRef] [PubMed]
  348. Wang, C.; Xu, J.; Zhang, Y.; Nie, G. Emerging Nanotechnological Approaches to Regulating Tumor Vasculature for Cancer Therapy. J. Control. Release 2023, 362, 647–666. [Google Scholar] [CrossRef]
  349. Jia, S.; Huang, S.; Jimo, R.; AXi, Y.; Lu, Y.; Kong, Z.; Ma, J.; Li, H.; Luo, X.; Qu, Y.; et al. In-Situ Forming Carboxymethyl Chitosan Hydrogel Containing Paeonia Suffruticosa Andr. Leaf Extract for Mixed Infectious Vaginitis Treatment by Reshaping the Micro-Biota. Carbohydr. Polym. 2024, 339, 122255. [Google Scholar] [CrossRef]
  350. Kaushal, N.; Singh, M.; Singh Sangwan, R. Flavonoids: Food Associations, Therapeutic Mechanisms, Metabolism and Nanoformulations. Food Res. Int. 2022, 157, 111442. [Google Scholar] [CrossRef]
  351. Soares Mateus, A.R.; Pena, A.; Sendón, R.; Almeida, C.; Nieto, G.A.; Khwaldia, K.; Sanches Silva, A. By-Products of Dates, Cherries, Plums and Artichokes: A Source of Valuable Bioactive Compounds. Trends Food Sci. Technol. 2023, 131, 220–243. [Google Scholar] [CrossRef]
  352. Anusha Siddiqui, S.; Redha, A.A.; Esmaeili, Y.; Mehdizadeh, M. Novel Insights on Extraction and Encapsulation Techniques of Elderberry Bioactive Compounds. Crit. Rev. Food Sci. Nutr. 2023, 63, 5937–5952. [Google Scholar] [CrossRef]
  353. Borah, M.S.; Tiwari, A.; Sridhar, K.; Narsaiah, K.; Nayak, P.K.; Stephen Inbaraj, B. Recent Trends in Valorization of Food Industry Waste and By-Products: Encapsulation and In Vitro Release of Bioactive Compounds. Foods 2023, 12, 3823. [Google Scholar] [CrossRef]
Figure 1. The chemical structure of Prunin and its primary biological effects. (A) Chemical structure of prunin. (B) Primary biological effects of prunin. Prunin demonstrates various pharmacological properties, including anticancer activity, anti-inflammatory effects, antimicrobial effects, immune regulation, anti-osteoporosis, anti-hypoxia, and protective effects. These benefits highlight the potential of Prunin as a versatile therapeutic compound. The figure was prepared using Biorender (BioRender.com accessed on 13 March 2025).
Figure 1. The chemical structure of Prunin and its primary biological effects. (A) Chemical structure of prunin. (B) Primary biological effects of prunin. Prunin demonstrates various pharmacological properties, including anticancer activity, anti-inflammatory effects, antimicrobial effects, immune regulation, anti-osteoporosis, anti-hypoxia, and protective effects. These benefits highlight the potential of Prunin as a versatile therapeutic compound. The figure was prepared using Biorender (BioRender.com accessed on 13 March 2025).
Ijms 26 02678 g001
Figure 2. The biological significance of prunin. Prunin influences multiple pathways in tumor cells at the molecular level, ultimately inducing key therapeutic effects such as cell cycle arrest, apoptosis, antiproliferation, and anti-angiogenesis, as well as anti-metastasis and invasion. The figure was prepared using Biorender.
Figure 2. The biological significance of prunin. Prunin influences multiple pathways in tumor cells at the molecular level, ultimately inducing key therapeutic effects such as cell cycle arrest, apoptosis, antiproliferation, and anti-angiogenesis, as well as anti-metastasis and invasion. The figure was prepared using Biorender.
Ijms 26 02678 g002
Figure 3. Prunin influences cell cycle regulation by interacting with various CDKs and cyclins. Prunin is helpful for modifying CDKs and cyclins during different cycle phases. It can inhibit CDK4, CDK6, and cyclin D action by increasing the expressions of the p21 marker in the G1 phase. At the G1/S checkpoint, prunin suppresses CDK2-Cyclin E by increasing p21 expressions. Similarly, CDK2-Cyclin A activity is restricted by the p27 marker [166,167,168]. This highlights the potential of flavonoids such as prunin to influence or induce cell cycle arrest, which contributes to cancer treatment. The figure was prepared using Biorender.
Figure 3. Prunin influences cell cycle regulation by interacting with various CDKs and cyclins. Prunin is helpful for modifying CDKs and cyclins during different cycle phases. It can inhibit CDK4, CDK6, and cyclin D action by increasing the expressions of the p21 marker in the G1 phase. At the G1/S checkpoint, prunin suppresses CDK2-Cyclin E by increasing p21 expressions. Similarly, CDK2-Cyclin A activity is restricted by the p27 marker [166,167,168]. This highlights the potential of flavonoids such as prunin to influence or induce cell cycle arrest, which contributes to cancer treatment. The figure was prepared using Biorender.
Ijms 26 02678 g003
Figure 4. The prunin compound induces apoptosis by triggering intrinsic and extrinsic-mediated pathways. In the extrinsic pathway, the prunin compound activates the FAS receptor, triggering FADD and cleaving pro-caspase-8/10. The activated caspase-8 processes BID into tBID, effectively linking the extrinsic pathway to the intrinsic pathway by enhancing the permeabilization of the outer mitochondrial membrane. In the intrinsic pathway, prunin can cause mitochondrial dysfunction by influencing endogenous ROS levels and altering the balance between pro-apoptotic and anti-apoptotic proteins [172,174,175]. The figure was prepared using Biorender.
Figure 4. The prunin compound induces apoptosis by triggering intrinsic and extrinsic-mediated pathways. In the extrinsic pathway, the prunin compound activates the FAS receptor, triggering FADD and cleaving pro-caspase-8/10. The activated caspase-8 processes BID into tBID, effectively linking the extrinsic pathway to the intrinsic pathway by enhancing the permeabilization of the outer mitochondrial membrane. In the intrinsic pathway, prunin can cause mitochondrial dysfunction by influencing endogenous ROS levels and altering the balance between pro-apoptotic and anti-apoptotic proteins [172,174,175]. The figure was prepared using Biorender.
Ijms 26 02678 g004
Figure 5. The prunin compound induces activation of P53 pathways. The prunin stimulates the P53 pathways after receiving the DNA damage signal, which further leads to multiple tasks, including cell cycle arrest, DNA repair if the damage is moderate, senescence, and apoptosis [84,226,227]. The apoptosis mechanism is highlighted when DNA damage stimulation occurs by prunin. It increases the activation of ATM, ATR, CHK1, and CHK2, which further leads to the activation of its downstream marker P53. The P53 further activates BAX and caspase cascades to induce apoptosis in cancer cells [228]. The figure was prepared using Biorender.
Figure 5. The prunin compound induces activation of P53 pathways. The prunin stimulates the P53 pathways after receiving the DNA damage signal, which further leads to multiple tasks, including cell cycle arrest, DNA repair if the damage is moderate, senescence, and apoptosis [84,226,227]. The apoptosis mechanism is highlighted when DNA damage stimulation occurs by prunin. It increases the activation of ATM, ATR, CHK1, and CHK2, which further leads to the activation of its downstream marker P53. The P53 further activates BAX and caspase cascades to induce apoptosis in cancer cells [228]. The figure was prepared using Biorender.
Ijms 26 02678 g005
Figure 6. Overview of the immunomodulatory effects of prunin on innate and adaptive immune responses. Activation (black arrow), increased response (red arrow). The figure was prepared using Biorender.
Figure 6. Overview of the immunomodulatory effects of prunin on innate and adaptive immune responses. Activation (black arrow), increased response (red arrow). The figure was prepared using Biorender.
Ijms 26 02678 g006
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Rana, J.N.; Mumtaz, S. Prunin: An Emerging Anticancer Flavonoid. Int. J. Mol. Sci. 2025, 26, 2678. https://doi.org/10.3390/ijms26062678

AMA Style

Rana JN, Mumtaz S. Prunin: An Emerging Anticancer Flavonoid. International Journal of Molecular Sciences. 2025; 26(6):2678. https://doi.org/10.3390/ijms26062678

Chicago/Turabian Style

Rana, Juie Nahushkumar, and Sohail Mumtaz. 2025. "Prunin: An Emerging Anticancer Flavonoid" International Journal of Molecular Sciences 26, no. 6: 2678. https://doi.org/10.3390/ijms26062678

APA Style

Rana, J. N., & Mumtaz, S. (2025). Prunin: An Emerging Anticancer Flavonoid. International Journal of Molecular Sciences, 26(6), 2678. https://doi.org/10.3390/ijms26062678

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

Article Metrics

Back to TopTop