Examining the evolution of pharmaceutical development over the preceding fifty years reveals that natural resources constitute a noteworthy sector within the pharmaceutical industry when synthesized without specific design.1,2 Natural extracts and medicinal plants constitute a significant reservoir for advancing pharmaceutical research and developing therapeutic agents. TCM is a prominent exemplar among natural remedies, enjoying extensive global utilization as an adjunctive therapeutic approach across diverse maladies. TCMs have been extensively employed for millennia in China and neighboring Asian regions as integral modalities for preventive measures and therapeutic interventions across various diseases.3–9 Naturally occurring substances and medicinal plants represent a significant reservoir of biologically active compounds with therapeutic potential. Sourcing pharmaceutical agents from natural origins has enjoyed a longstanding tradition within medicine. Despite being employed for millennia in China and having significantly advanced human health, TCM continues to harbor substantial ambiguity regarding its molecular-level therapeutic mechanisms. Therefore, it is imperative to ascertain the primary components and comprehend the molecular mechanisms underlying the observed effectiveness. In the preceding twenty years, an escalating trend has emerged in which an increasing array of bioactive compounds originated from TCM herbs. A predominant proportion of the identified bioactive compounds fall within the classification of plant secondary metabolites. These agents exhibit diverse effects on cell death pathways. In tumors, specific agents induce apoptosis in cancer cells. Conversely, some agents may inhibit apoptosis in non-malignant conditions like cardiovascular and neurological diseases, thereby protecting cells from further deterioration.10–14
MiRNAs, a subset of small ncRNAs that regulate target gene expression via post-transcriptional mechanisms, have emerged as pivotal regulatory molecules in TCM-induced therapeutic responses. In this context, recent in silico and in vitro analysis demonstrated that Danggui Buxue Decoction (DGBXD) ameliorates idiopathic pulmonary fibrosis (IPF) through several miRNA-mRNA regulatory networks, including the miR-493-Olr1 and miR-338-Hif1a axes.15 TCM has demonstrated efficacy in regulating apoptosis via the modulation of particular miRNAs, thereby substantiating its therapeutic efficacy across a spectrum of health disorders.16 Herein, we comprehensively examine contemporary research investigating the correlation between miRNAs and anti-tumor TCMs alongside their bioactive constituents. This endeavor aims to offer fresh insights into the mechanisms underlying TCMs’ anti-tumor properties. In addition, we examine a novel proposition regarding the regulation of genes facilitated by exogenous miRNAs, potentially presenting a novel avenue in cancer therapy through the utilization of Chinese medicinal herbs.
Major Apoptosis Signaling PathwayApoptosis is a fundamental biological phenomenon observed in multicellular organisms. It entails the regulated removal of cells. It is pivotal in embryonic morphogenesis and maintaining tissue homeostasis in adult organisms. Apoptosis is primarily mediated by two distinct signaling pathways: the extrinsic pathway, initiated by death receptor engagement, and the intrinsic pathway, triggered by mitochondrial stress.17,18 The engagement of death receptors through either their endogenous ligands (eg, TNFα, CD95L, and TRAIL) or agonistic antibodies (eg, anti-APO-1) initiates a stepwise activation cascade involving caspase-8 and −3. This cascade results in the proteolytic cleavage of specific substrates and ultimately triggers apoptosis. Intrinsic triggers of cellular demise, such as reactive oxygen species (ROS), agents inducing DNA damage, and stimuli facilitating calcium ion mobilization, initiate the mitochondrial pathway. This leads to the liberation of cytochrome c and the assembly of the apoptosome complex, comprising cytochrome c, Apaf-1, and caspase-9, either through direct activation or indirect mechanisms.19 In the interplay of receptor-mediated and mitochondrial signaling pathways, the pro-apoptotic protein Bid assumes a pivotal role as an intermediary agent after its cleavage by activated caspase-8. This cleavage event facilitates the translocation of the pro-apoptotic proteins Bax and/or Bak to the mitochondrial membrane. Cellular fate, including survival and demise, is critically mediated by the interplay between intrinsic pro-apoptotic and anti-apoptotic protein families. In the context of the receptor-mediated pathway, c-FLIP and XIAP function as negative regulators, exerting control over the enzymatic actions of caspase-8 and caspase-3. In the mitochondria-mediated pathway of apoptosis, regulation predominantly ensues through a network of pro-apoptotic proteins such as Bax, Bid, Bak, and Smac, alongside anti-apoptotic counterparts including Mcl-1, Bcl-xL, B-cell lymphoma 2 (Bcl-2), and XIAP.20,21
NF-κB: In 1986, the identification of the transcription factor nuclear factor of kappa light polypeptide gene enhancer in B cells (NF-κB) marked a pivotal discovery in molecular biology. It elucidated its role as a nuclear entity that selectively interacts with the enhancer region of the immunoglobulin kappa light chain within activated B cells.22 This seminal observation consequently led to the establishment of the abbreviated designation NF-κB. Five constituents of this transcription factor cohort have been delineated, denoted as p65 (RelA), RelB, c-Rel, NF-κB1, and NF-κB2.23 In cellular signaling, NFκB activation is facilitated through two distinct pathways: the canonical pathway, the classical route, and the non-canonical pathway, commonly referred to as the alternative pathway. In the canonical activation cascade, stimulatory signaling pathways may be facilitated via Interleukin-1 receptor (IL-1R), Toll-like receptors (TLRs), tumor necrosis factor receptor (TNFR), and antigen receptors. Commonly encountered signaling molecules that provoke stimulation include tumor necrosis factor α (TNFα), lipopolysaccharides (LPS)—constituents of bacterial cell walls—and interleukin-1 β (IL-1β). Upon stimulation via these receptors, activation of the IκB kinase (IKK) complex ensues, resulting in the phosphorylation of IκBα primarily by IKK2. This pathway is pivotal in initiating innate immune responses and inflammatory processes while suppressing apoptosis.24,25 The Non-canonical pathway plays a pivotal role in p100/Rel-B complexes activation, predominantly manifesting during the ontogenesis of lymphoid organs pivotal for B and T lymphocyte production, with its activation modulated solely by B cell activating factor (BAFF). This signaling route employs an IKK complex composed of two IKKα subunits, excluding NEMO from its composition. In the non-canonical signaling pathway, ligands stimulate NFκB activation, inducing kinase (NIK), initiating a cascade whereby the IKKα complex becomes phosphorylated and activated. This activated complex subsequently phosphorylates the p100 protein, resulting in its processing and subsequent release of the p52/Rel-B active heterodimers. Contrary to p100, which undergoes cleavage to generate p50, the inducible processing potential of p105 remains a subject of contention. The initiation of p100 processing via BAFFR plays a pivotal role in bolstering the viability of transitional and mature B cells, ostensibly by prompting the activation of anti-apoptotic genes such as bcl-x and bcl-2.26 Notably, the observation that NF-κB becomes activated concurrently with or near cellular apoptosis in response to stimulatory stimuli has prompted the proposition that NF-κB activation is closely associated with suppressing apoptosis. In the majority of cellular contexts, the activation of NF-κB serves to safeguard the cell against apoptosis, primarily achieved by upregulating the expression of various survival genes, including but not limited to TRAF1, Bcl-xL, TRAF2, BfI-1, c-IAP1, c-IAP2, and IEX-IL. NF-κB binding motifs have been discerned within the regulatory regions of the interleukin-1b converting enzyme protease, c-myc, and TNFα genes, all of which play integral roles in the induction of apoptotic cell death.27 NF-κB also mediates the transcription of genes encoding various anti-apoptotic proteins belonging to the bcl-2 family, including Nrl3, bfl-1/A1, and bcl-xl. These proteins collectively inhibit the release of cytochrome-c and the consequent activation of caspase-9, thereby impeding the apoptotic process.28,29
PTEN/PI3K/AKT/mTOR Signaling PathwayPI3K, also known as phosphatidylinositol-3-kinase, is a protein kinase located on the plasma membrane, which functions as a pivotal mediator linking extracellular growth factor and cytokine signals with intracellular signal transduction pathways downstream.30 Typically, the PI3K activation occurs via receptor tyrosine kinases (RTKs); however, alternative mechanisms exist involving G-protein-coupled receptors and oncogenes, including small GTPase RAS. The activation of PI3K is contingent upon the specific interaction context and the affinity towards particular PI3K subunits. Upon activation, PI3K catalyzes the phosphorylation of phosphatidylinositol-4,5-biphosphate (PI (4,5)P2) to produce phosphatidylinositol-3,4,5-triphosphate (PIP3).31 IP3 initiates intracellular signaling pathways by enlisting and interacting with various proteins, notably phosphoinositide-dependent kinase 1 (PDK1) and AKT, which possess pleckstrin homology (PH) domains. This recruitment event triggers AKT phosphorylation by PDK1, subsequently inducing activation of AKT.32 The activation of AKT exerts regulatory control over numerous cellular processes through the phosphorylation of a multitude of substrates, including but not limited to IKKα, FOXO, PRAS40, GSK3, MDM2, eNOS, TSC2, WNK1, ASK1, AMPK, RAF1, CHK1, p27, p21, and BAD. The downstream effectors establish a connection between AKT activity and regulating processes such as protein synthesis, transcription, cell survival, apoptosis, and proliferation.33,34 Furthermore, the mammalian target of Rapamycin (mTOR) denotes a serine/threonine protein kinase that functions as a prominent downstream mediator within the AKT signaling pathway. The PI3K-AKT-mTOR pathway assumes a pivotal function in regulating pro-survival cellular signaling mechanisms.35 Phosphatase and tensin homologue (PTEN) functions as a regulator within the PI3K-AKT-mTOR signaling cascade by catalyzing the dephosphorylation of PIP3 to PI(4,5)P2. This enzymatic activity ultimately impedes cellular proliferation. So, PTEN inactivation stimulates the PI3K-AKT-mTOR pathway, consequently promoting cellular survival.36 AKT is evidently the principal determinant governing the pro-survival/anti-apoptotic function of the PI3K-AKT-mTOR pathway. It employs various mechanisms to regulate cell survival and apoptosis, either through direct interaction with proteins involved in the apoptotic pathway or by modulating transcription factors responsible for transcribing genes involved in apoptosis. One of the immediate targets of AKT is BAD, an apoptosis-promoting factor and constituent of the BCL-2 protein family. BAD interacts with and suppresses the activity of anti-apoptotic factors such as BCL-XL or BCL-2. BAD inactivation occurs via the phosphorylation of Ser136 by the enzymatically active AKT, activating BCL-2 or BCL-XL and promoting cellular survival.35,37
MAPK Signaling PathwayMitogen-activated protein kinases (MAPKs) are serine-threonine protein kinases that exert regulatory control over diverse cellular processes, encompassing proliferation, differentiation, apoptosis, survival, inflammation, and innate immunity.38,39 In mammalian organisms, MAPKs encompass c-Jun NH2-terminal kinase (JNK), p38 MAPK, and extracellular signal-regulated kinase (ERK), comprising multiple isoforms.40 JNK1 and JNK2 exhibit widespread tissue expression, whereas JNK3 is primarily expressed in neuronal cells.41 The most comprehensive investigations among the four p38 MAPK isoforms (namely p38-α, p38-β, p38-γ, and p38-δ) have predominantly focused on p38-α and p38-β. MAPKs that have been activated catalyze the phosphorylation of a range of target proteins, encompassing transcription factors such as c-Jun, c-Myc, and ATF2, as well as antiapoptotic protein Bcl-2 and the proapoptotic protein Bad.42 Furthermore, various substrates for the JNK and p38 MAPK signaling pathways, known to facilitate apoptosis, have been discovered and confirmed through validation processes. Many transcription factors have been documented as subject to regulation by the signaling molecules JNK and p38, leading to upregulation of pro-apoptotic proteins and downregulation of anti-apoptotic proteins.43 A prominent target of the JNK pathway is the transcription factor AP-1, known as activator protein 1. AP-1 is a dimeric complex composed of various combinations of proteins from the Fos (including Fra2, Fra1, c-Fos, and FosB), Jun (such as c-Jun, JunB, and JunD), ATF (activating transcription factor), and MAF (V-maf musculoaponeurotic fibrosarcoma) families.44 In addition, one of the most prominent transcription factors modulated by the JNK and p38 MAPK signaling cascades in the context of apoptosis is the p53 tumor suppressor protein. Within cells experiencing stress, the process of phosphorylation mediated by JNK can lead to the stabilization and activation of the p53 protein, thereby facilitating the initiation of programmed cell death. Similar to the c-Jun subunit of the AP-1 complex, the transcription factor p53 collaborates with various other proteins. According to the available literature, the dimerization of p53 and p73 has been identified as a pivotal factor in initiating apoptotic cell death, particularly in cellular stress response mediated by the JNK pathway. JNK activation leads to the phosphorylation of p53 at Thr81 within its proline-rich domain (PRD), facilitating the subsequent dimerization of p73 and p53. The formation of the p53-p73 dimer promotes the transcriptional activation of numerous pro-apoptotic genes, including but not limited to puma and bax45,46(Figure 1).
Figure 1 Major signaling pathways involved in apoptosis.
Apoptosis Dysregulation in Human DiseasesMalfunctioning apoptosis is linked to various illnesses such as autoimmune disorders, neurodegenerative conditions, bacterial and viral infections, cancer, and cardiovascular diseases. In this regard, Kun et al investigated the involvement of apoptosis in the pathogenesis of varicosities. In their study, the apoptotic ratio observed within the walls (intima and media) of the afflicted splenic vein and varicose great saphenous vein groups exhibited a notable reduction compared to the equivalent anatomical sites within the non-afflicted splenic vein and great saphenous vein groups, respectively. Notably, histopathological examination of diseased splenic and varicose great saphenous veins revealed apoptotic features within endothelial and smooth muscle cells of their respective walls. These characteristic morphological changes included fuzzy mitochondrial cristae, alterations in the medullary area, and peripheralization of nuclear chromatin. The results indicate that reduced apoptosis in the vascular wall is implicated in the pathogenesis of splenic vein disorders and varicose great saphenous veins.47 As well, apoptosis appears to be a pivotal factor in the advancement of numerous neurological conditions, including Alzheimer’s disease (AD). In this regard, platelet levels of caspase-3, caspase-9, Bax, and Bad markedly increased among individuals diagnosed with AD and mild cognitive impairment (MCI). Apoptosis proteins were also upregulated in individuals with amnestic MCI in comparison to both AD patients and cognitively normal controls. In individuals with amnestic MCI, the levels of the anti-apoptotic protein Bcl2 were elevated, whereas in AD, a reduction in Bcl2 expression was noted. In this manner, elevated levels of apoptosis are observed in platelets, potentially reflecting similar apoptotic processes occurring in the brain, with aberrant apoptosis occurring early in Alzheimer’s disease, and the balance between pro- and anti-apoptotic protein levels contributing to the platelet’s susceptibility to apoptotic signals.48 Thus, modulating apoptosis towards a homeostatic equilibrium presents a potential therapeutic strategy for various diseases.
MiRNAs: Biosynthesis in Functioning in ApoptosismiRNAs are a class of small, highly conserved, ncRNAs, approximately 22 nucleotides in length, that play crucial roles in modulating gene expression. RNA polymerase II (Pol II) facilitates the production of a hairpin intermediate termed “pri-miRNA” during the transcription of a nuclear miRNA gene. The recognition of pri-miRNA involves a microprocessor complex comprising double-stranded RNA-specific endoribonuclease (DROSHA) and DiGeorge syndrome critical region gene 8 (DGCR8) enzymes, which cleave the molecule to generate precursor miRNA (pre-miRNA).49 Pre-miRNAs are subsequently translocated to the cytoplasm for additional processing facilitated by the enzyme DICER, in conjunction with cofactors, including protein activator of protein kinase R (PACT) and Trans-activation response RNA-binding protein (TRBP). The mature miRNA duplex is ultimately incorporated into the RISC, where one of the miRNA strands (−5p or −3p) associates with the Argonaute (AGO) protein, directing the complex to interact with its target mRNA. A single mRNA molecule can possess numerous binding sites accommodating various miRNAs, thereby establishing a complex network of interactions between miRNAs and mRNA.50,51 MiRNAs exhibit tissue-specific distributions and can modulate the expression of roughly 30% of the human genome.52 MiRNAs, as integral cellular components, are increasingly recognized for their pivotal involvement in a diverse array of fundamental biological processes. Despite lacking complete understanding, multiple strands of evidence suggest that miRNAs modulate apoptosis. It is believed that they regulate the expression of genes involved in both promoting and inhibiting apoptosis. In this context, employing a combination of bioinformatics techniques and experimental methodologies, Druz et al predicted and identified the anti-apoptotic targets of mmu-miR-466h. Their experimental investigation revealed that inhibiting mmu-miR-466h led to heightened expression levels of genes associated with apoptosis, thereby leading to enhanced cellular viability and reduced activation of Caspase-3/7. They additionally revealed that the increase in expression of mmu-miR-466h following nutrient depletion suppresses multiple anti-apoptotic genes. Their results indicate that mmu-miR-466h exhibits a pro-apoptotic function and can regulate the apoptotic pathway within mammalian cells.53 Similarly, through the application of bioinformatics analysis and luciferase reporter assays, Ouyang et al confirmed that miR-181, a miRNA predominantly expressed in the brain, can selectively target the 3′-untranslated regions (3′-UTRs) of Bcl-2 family members, namely Bcl-2-L11/Bim, Mcl-1, and Bcl-2. Their subsequent investigation revealed that reduced expression levels of miR-181a mitigated apoptosis triggered by glucose deprivation, alleviated mitochondrial dysfunction, and preserved mitochondrial membrane potential in astrocytes.54 Thus, the involvement of miRNAs in regulating apoptosis indicates that therapeutic interventions directed towards these molecules hold promise for addressing both malignant and non-malignant conditions.
Targeting miRNAs-Apoptosis Regulatory Network via TCM, and TCM Extracts in Malignant DisordersCancer is characterized by the uncontrolled proliferation of abnormal cells in the body. It is currently regarded as a major global health challenge and one of the most prevalent diseases worldwide, with cancer-related mortality is being increased. Regulating the survival and death of cancer cells is a critical strategy in the effective management and treatment of cancer.55 Traditional Chinese Medicine can either promote or inhibit the expression of cancer-associated miRNAs, and thus modulating tumorigenicity and cancer progression.
Targeting miRNAs-Apoptosis Regulatory Network via BaicalinScutellaria baicalensis Georgi (Lamiaceae or Labiatae) is a widely recognized TCM herb, commonly referred to as Huang-Qin or Chinese skullcap (Radix Scutellariae).56 Baicalin, a phytochemical derived from the roots of Scutellaria baicalensis Georgi, exhibits various pharmacological properties in neurodegenerative diseases and multiple malignancies through several biological processes, such as anti-inflammatory, antioxidant, and anti-apoptotic effects.57,58 Tao et al investigated the molecular pathways implicated in the therapeutic effects of baicalin on colorectal cancer (CRC). They validated that baicalin can efficiently stimulate and increase apoptosis in HT-29 cells in a manner dependent on dosage while also inhibiting tumor proliferation in xenografted nude mice. They conducted a miRNA microarray analysis on HT-29 cells treated with baicalin, revealing significant suppression of numerous oncomiRs, such as miR-205, miR-151a, miR-31, miR-30c, miR-10a, and miR-23a compared to untreated cells. Furthermore, their in vitro and in vivo investigations demonstrated that baicalin inhibited oncomiRs by decreasing c-Myc expression levels. Their research demonstrates a novel pathway through which baicalin exerts its anti-cancer effects by promoting apoptosis in colon cancer cells and inhibiting tumor growth via downregulation of c-Myc and oncomiRs expression.59 In addition, Duan et al conducted a study to examine the precise function and potential underlying mechanism of baicalin in breast cancer (BC). Baicalin administration was observed to suppress cellular viability while stimulating apoptosis in BC cells. In their study, reduced miR-338-3p expression was observed in both BC tissues and cells, with overexpression of miR-338-3p leading to reduced cell viability and increased apoptosis. Baicalin exposure resulted in the reversal of miR-338-3p expression, and the inhibition of miR-338-3p mitigated the effects of baicalin on cell viability and apoptosis. Subsequent functional investigations revealed that the mRNA expression of MORC4, a target of miR-338-3p, was elevated in both BC tissues and cells but reduced upon exposure to baicalin. They finally revealed that the upregulation of MORC4 mitigated the impact of miR-338-3p on cellular viability and apoptosis. Their findings suggest that baicalin inhibits cell viability while inducing apoptosis in BC cells by modulating miR-338-3p and MORC4, underscoring its potential as a pharmacological agent for BC therapy.60
Targeting miRNAs-Apoptosis Regulatory Network via BaicaleinBaicalein, a flavone compound, serves as a bioactive constituent within the traditional medicinal plant known as Scutellaria baicalensis Georgi. A growing body of evidence indicates that baicalein possesses therapeutic potential for treating and preventing diverse forms of cancer.61 Ma et al explored the mechanisms through which baicalein inhibits pancreatic cancer progression. Initially, they confirmed the significant involvement of baicalein in the suppression of pancreatic tumorigenesis through experimentation both in vitro and in vivo. Through high-throughput sequencing analysis, they observed differential expression of 20 up-regulated and 39 down-regulated miRNAs in Panc-1 cells treated with 100 μM baicalein compared to the control group. The qPCR analysis substantiated that miR-139-3p exhibited the highest upregulation following treatment with baicalein, whereas miR-196b-5p demonstrated the most pronounced downregulation. Their subsequent investigations into functionality demonstrated that miR-139-3p promoted apoptosis in Panc-1 cells by targeting NOB1, while miR-196b-5p suppressed apoptosis by targeting ING5. They determined that baicalein’s ability to regulate the expression of miR-139-3p or miR-196b-5p induces apoptosis and inhibits tumor growth in pancreatic cancer, indicating its potential as a potent inhibitor in this context.62
Targeting miRNAs-Apoptosis Regulatory Network via MatrineMatrine, an alkaloid, is extracted from the traditional Chinese medicinal plant Sophora flavescens Aiton. Numerous studies have demonstrated the anticancer properties of matrine, showcasing its ability to impede cancer cell proliferation and trigger apoptosis.63 An investigation conducted by An et al explored the therapeutic impacts and molecular mechanisms of matrine on the A549 non-small cell lung cancer (NSCLC) cell line. In their study, a substantial matrine concentration of 1.0 mg/mL resulted in a noteworthy 52.68±3.32% inhibition of cellular proliferation, accompanied by observable phenomena such as cell shrinkage and disruption. Flow cytometric analysis demonstrated a notable rise in the proportion of G1/G0 cells and a decrease in the proportions of S and G2/M cells after 48-hour matrine treatment. These findings suggest that matrine administration led to the initiation of cellular arrest. Also, via both DNA fragmentation assay and TUNEL assay, they examine the impact of matrine on apoptosis induction in A549 cells, revealing that treatment with matrine resulted in apoptosis of approximately 30% of the A549 cell population. Importantly, treatment with a low concentration of matrine (0.2 mg/mL) resulted in an observed increase in miR-126 expression levels, subsequently leading to a decrease in the expression of its target gene, vascular endothelial growth factor. In conclusion, TCM matrine, possessing antitumor properties, prompted cell cycle arrest and apoptosis while restoring miR-126 expression in the A549 NSCLC cell line.64 Furthermore, Fu et al sought to examine the anticancer properties of matrine against papillary thyroid cancer (PTC) in human subjects and elucidate the associated molecular mechanisms. Initially, they provided evidence indicating a notable increase in the expression levels of miR-182-5p in both PTC tissues and cell lines. Matrine suppressed miR-182-5p expression and triggered apoptosis in TCP-1 and BCPAP cells, varying the extent of this effect according to the dosage administered. Matrine elevated the caspase-3 expression and decreased the expression levels of Bcl-2 in both BCPAP and TCP-1 cells. Subsequent functional investigations revealed that upregulation of miR-182-5p attenuated matrine-induced apoptosis and caspase-3 activation while preventing the decrease in Bcl-2 expression induced by matrine. Their study ultimately demonstrated that miR-182-5p upregulation antagonized the suppressive impact of matrine on PTC tumor proliferation. In this manner, Matrine potentially elicits anti-tumor properties through mechanisms such as the promotion of apoptosis in TCP-1 and BCPAP cells, reduction of Bcl-2 levels, activation of caspase 3, and inhibition of PTC tumor progression via the downregulation of miR-182-5p expression. These results elucidate the anti-cancer properties of matrine in PTC and highlight miR-182-5p as a promising therapeutic target for matrine in PTC management.65
Targeting miRNAs-Apoptosis Regulatory Network via BerberineBerberine (BBR) is a benzylisoquinoline alkaloid isolated from the Chinese herb Coptis chinensis Franch. It is extensively utilized in TCM and exhibits pharmacological effects such as anti-inflammatory and antioxidant properties.66,67 Chen et al sought to elucidate the mechanisms underlying the anti-tumor effects of BBR in NSCLC. In their study, BBR inhibited the proliferation of NSCLC and facilitated apoptosis in NSCLC cells by regulating the expression of TF and miR-19a. Their luciferase assays demonstrated that TF was directly targeted and inhibited by miR-19a in NSCLC cells. Their subsequent experiments revealed that BBR triggered apoptosis via modulation of the miR-19a/TF/MAPK axis. Their findings indicate that BBR prompts apoptosis in NSCLC cells by activating the miR-19a/TF/MAPK signaling cascade.68
Targeting miRNAs-Apoptosis Regulatory Network via Hd-SbThe combination of Hedyotis diffusa (HD) and Scutellaria barbata (SB) is extensively utilized in clinical antitumor prescriptions as a commonly employed herb pair in China. Pan et al sought to examine the antitumor efficacy of the herb-pair (Hedyotis diffusa plus Scutellaria barbata (Hd-Sb)) in bladder cancer cells. Their findings demonstrate that Hd-Sb suppressed the growth and colony formation of bladder cancer cells in a manner dependent on both dose and time. They also observed that the herb-pair induced cell apoptosis by inhibiting Akt activation and decreasing antiapoptotic protein expression Mcl-1 and Bcl-2. Their subsequent experiments revealed that the herb-pair decreased miR-155 levels, leading to induction of cell apoptosis and inhibition of Akt activation by miR-155 inhibitor. Notably, exogenous miR-155 abrogated the pro-apoptotic effects of the herbal combination by stimulating the Akt signaling pathway in bladder cancer cell lines. Taken together, Hd-Sb suppressed miR-155 expression and Akt pathways, induced apoptosis, and exhibited anticancer properties in bladder cancer cells.69
Targeting miRNAs-Apoptosis Regulatory Network via ArtesunateArtesunate (ART) is a semi-synthetic compound derived from artemisinin, a natural substance obtained from the leaves of Chinese herb Artemisia annua L. Artemisinin and its derivatives exhibit activity against cancer cells.70 ART effectively suppressed orthotopic tumor growth in rats with bladder cancer, concomitant with upregulation of miR-16 expression and downregulation of cyclooxygenase-2 (COX-2) expression. In vitro studies revealed that ART exhibited selective cytotoxicity towards bladder cancer cells, inducing apoptosis. However, normal human urothelial cells demonstrated significantly lower susceptibility to ART-mediated toxicity. ART notably elevated miR-16 expression and reduced the expression of COX-2 and the production of prostaglandin E2 (PGE2). More importantly, the downregulation of miR-16 expression could counteract the apoptotic effect of ART and the downregulation of COX-2 expression in bladder cells. Moreover, exogenously administered PGE2 demonstrated the ability to abrogate apoptosis in bladder cancer cells previously exposed to ART-based therapy. In conclusion, ART induces an anti-tumor effect against bladder cancer by increasing miR-16 expression, promoting apoptosis, and decreasing COX-2 expression and PGE2 production. Hence, ART shows promise as a potent therapeutic agent for the management of bladder cancer.71
Targeting miRNAs-Apoptosis Regulatory Network via ShikoninShikonin, the principal bioactive compound derived from the roots of Lithospermum erythrorhizon Siebold & Zucc, also referred to as “Zicao” in TCM. Recent research indicates that shikonin exhibits diverse bioactive properties relevant to cancer therapy.72 Shikonin suppresses proliferation in human endometrioid endometrial cancer (EEC) cell lines in a manner dependent on dosage. Moreover, shikonin induces apoptosis by increasing the expression of pro-apoptotic proteins cleaved-Caspase-3 and Bax while decreasing the expression of the anti-apoptotic protein Bcl-2. Shikonin induces dysregulation of numerous miRNAs, with miR-106b being notably downregulated. Furthermore, the reintroduction of miR-106b expression nullifies the anti-proliferative and pro-apoptotic effects induced by shikonin. Mechanistically, miR-106b targets PTEN, thereby regulating the AKT/mTOR signaling pathway. Therefore, shikonin suppresses proliferation and triggers apoptosis in human EEC cells by modulating the miR-106b/PTEN/AKT/mTOR signaling pathway, underscoring its promise as a therapeutic intervention for EEC.73
Targeting miRNAs-Apoptosis Regulatory Network via CurcuminCurcumin, sourced from the rhizome of Curcuma longa L., has been utilized for centuries in TCM and Ayurvedic medicine throughout Asia for cancer therapy.74 Curcumin suppressed cell proliferation, triggered apoptosis, and enhanced caspase-3 activity in A549 cells. The relative expression of miR-192-5p in NCL-H460 cells decreased compared to that in A549 cells, which exhibited higher expression, while BEAS-2E cells demonstrated the highest expression. Furthermore, miR-192-5p mimics attenuated cell proliferation and augmented cell apoptosis in A549 cells, whereas anti-miR-192-5p mimics promoted cell proliferation and hindered cell apoptosis in A549 cells. Curcumin efficiently upregulated the relative expression of miR-192-5p and attenuated the PI3K/Akt signaling pathway. Notably, miR-192-5p mimics potentiated the impact of curcumin on apoptosis and cell viability while inhibiting the PI3K/Akt signaling pathway in A549 cells. Also, anti-miR-192-5p mimics reversed the impact of curcumin on A549 cells and the expression of PI3K/Akt. Therefore, curcumin triggers apoptosis and suppresses cell proliferation in human non-small cell lung cancer cells by increasing miR-192-5p and inhibiting the PI3K/Akt signaling pathway.75
Targeting miRNAs-Apoptosis Regulatory Network via TriptolideTriptolide (TP), identified by Kupchan et al in 1972 from the root of Tripterygium wilfordii Hook F. (TWHF), is regarded as the most potent epoxide diterpene lactone compound among various CHM. TP has garnered significant research attention due to its potent anti-cancer properties.76 In PC‑9 cells, triptolide treatment decreased cell proliferation, enhanced apoptosis, and increased caspase‑9 and 3 activity. Triptolide decreased miR‑21 expression and increased PTEN protein levels in PC‑9 cells. Significantly, increased expression of miR‑21 mitigated the impact of Triptolide on cell viability and PTEN protein levels in PC‑9 cells. In this manner, Triptolide attenuated the proliferation and promoted the apoptosis of human NSCLC cells via modulation of PTEN through targeting miR-21.77
Targeting miRNAs-Apoptosis Regulatory Network via IsoliquiritigeninIsoliquiritigenin, a flavonoid derived from the root of Glycyrrhiza uralensis Fisch, represents a constituent of TCM with historical significance as one of the earliest recognized herbal remedies. The primary bioactive compound found in licorice root, isoliquiritigenin, exhibits a spectrum of biological functions encompassing antiviral properties, antioxidative effects against free radicals, and mitigation of lipid peroxidation. Isoliquiritigenin suppresses cellular proliferation within nasopharyngeal carcinoma cell lines, encompassing C666-1 and CNE2. Isoliquiritigenin facilitated apoptosis in nasopharyngeal carcinoma cells by enhancing the expression of Caspase-9, Caspase-3, and Bax while inhibiting the expression of Bcl-2. Furthermore, miR-32 markedly increases in nasopharyngeal carcinoma tissues, with isoliquiritigenin demonstrating a significant capacity to decrease miR-32 expression. Elevated levels of miR-32 facilitated the proliferation and migration of nasopharyngeal carcinoma cells while concurrently inhibiting apoptosis. Notably, the administration of isoliquiritigenin significantly impedes the impact mediated by miR-32. Mechanistically, miR-32 targets large tumor suppressor 2 (LATS2) and impedes the growth of nasopharyngeal carcinoma cells by modulating the Wnt signaling pathway. Moreover, isoliquiritigenin displays similar findings in vivo experimentation and suppresses xenograft development in nude mice, concurrent with reductions in tumor volume and modulation of LATS2 and miR-32 expression. So, isoliquiritigenin possesses significant potential as an efficacious anti-nasopharyngeal carcinoma agent, demonstrating effectiveness both in vivo and in vitro through modulation of the miR-32/LATS2/Wnt pathway.78 Additionally, Peng et al conducted a study examining the efficacy of 3′, 4′, 5′, 4″-tetramethoxychalcone (TMC), a derivative of ISL, in targeting tumor growth specifically within triple-negative breast cancer (TNBC) cells. Initially, their investigation revealed that TMC prompted apoptosis in TNBC cells in a dosage-correlated fashion. The miR-374a overexpression was observed across multiple TNBC cellular models, encompassing BT549, 4T1, and MDA-MB-231 cell lines. They additionally documented that miR-374a exhibited a direct binding affinity towards the 3′-UTR region of BAX gene, resulting in the downregulation of both Bax mRNA and protein levels. Their findings demonstrated that miR-374a exerted an oncogenic influence on apoptosis within TNBC cells, thereby facilitating tumor initiation. Following a 24-hour treatment with TMC, TNBC cells exhibited a notable reduction in miR-374a expression. Additionally, TMC upregulated Bax mRNA expression, indicating its potential to influence BAX levels through regulatory mechanisms, whether at the transcriptional or post-transcriptional level. Thereby, TMC elicits apoptosis and impedes tumor initiation in TNBC by regulating the miR-374a/BAX axis.79
Targeting miRNAs-Apoptosis Regulatory Network via CelastrolCelastrol (Cel), alternatively referred to as tripterine, is a biologically active compound present in the root of TCM Tripterygium wilfordii Hook F. An increasing body of evidence suggests that Celastrol possesses therapeutic potential and displays a range of biological activities, notably demonstrating efficacy in cancer treatment.80 Celastrol demonstrated a dose-dependent inhibition of colon cancer cell viability, accompanied by the downregulation of PCNA. Additionally, Celastrol administration significantly reduced the expression of BCL-2 and miR-21, while increasing BAX expression and inducing Caspase-3 activity. Notably, overexpression of a miR-21 mimic enhanced cell viability, suppressed apoptosis, decreased BAX levels, and reduced Caspase-3 activity to some extent. These effects were subsequently reversed by Celastrol treatment. Moreover, the miR-21 mimic activated the PI3K/AKT/GSK-3β pathway, while the addition of Celastrol partially inhibited this activation. Therefore, Celastrol shows potential in inhibiting colon cancer cell proliferation and promoting apoptosis through its negative regulation of miR-21 and the PI3K/AKT/GSK-3β pathway.81 Celastrol has also demonstrated a therapeutic response in gastric cancer. It was found that Celastrol triggered apoptosis in gastric cancer cells by impeding the activation of the PI3K/Akt and NF-κB signaling pathways. IGF-1, which robustly activates Akt, restored NF-κB functionality in Celastrol-treated cells. Additionally, Celastrol significantly reduced the expression of miR-21. Moreover, the inhibition of miR-21 led to a reduction in phospho-Akt expression and NF-κB activity. Notably, miR-21 overexpression enhanced the activity of the PI3K/Akt and NF-κB pathways while reducing apoptosis in Celastrol-treated gastric cancer cells—a phenomenon that was potentially reversible with the administration of a PI3K inhibitor. Thus, the impact of Celastrol on apoptosis was attributed to the suppression of the PI3K/Akt-dependent NF-κB pathway via miR-21 inhibition.82
Targeting miRNAs-Apoptosis Regulatory Network via AloperineAloperine (ALO), found in the seeds and leaves of the Sophora alopecuroides L. in TCM, exhibits a diverse array of pharmacological effects, notably including anticancer properties.83 Han et al investigated the upstream pathway through which Aloperine mitigates CRC injury, focusing on elucidating its impact on the regulatory network involving circular RNA (circRNA), miRNAs, and mRNAs. Their findings demonstrated a notable reduction in the viability and proliferation of CRC cells following ALO treatment, alongside a marked elevation in the incidence of apoptotic cancer cell population, indicative of an inhibitory effect on tumor progression. In their study, among the subset of circRNAs exhibiting differential expression in CRC, solely circNSUN2 demonstrates dual functionality by serving as a target for miR-296-5p and being subject to regulation by Aloperine. The findings of their experimental investigation revealed that circNSUN2 silencing resulted in a reduction of proliferation and apoptosis promotion in CRC cells. Additionally, circNSUN2 overexpression counteracted the suppressive impact of Aloperine on cancerous cells. More importantly, they disclosed that ALO could inhibit circNSUN2 activation and counteract the stimulatory impact of circNSUN2 upregulation on the advancement of esophageal cancer. Through the utilization of dual luciferase report assay and subsequent rescue experiments, they confirmed the cellular regulatory impacts of circNSUN2 alongside miR-296-5p/ Signal Transducer and Activator of Transcription 3 (STAT3). They ultimately validated their hypothesis that modulation of the circNSUN2/miR-296-5p/STAT3 axis results in decreased proliferation and heightened apoptosis in CRC cells. Thereby, Aloperine, through modulation of the circNSUN2/miR-296-5p/STAT3 pathway and facilitation of apoptotic processes, exhibits a preventative effect against the malignancy progression of CRC cells.84
Targeting miRNAs-Apoptosis Regulatory Network via Pien Tze HuangPien Tze Huang (PZH) represents a widely recognized TCM formulation whose ingredients are mainly composed of Panax notoginseng (Burk). F. H. Chen. It is distinguished by a variety of pharmacological impacts, including the suppression of inflammatory processes and the regulation of cell proliferation.85,86 PZH exhibited a notable decrease in both the viability and cell density of HCT‑8/5‑FU cells in a manner dependent on dose and time. PZH demonstrated inhibitory effects on cellular viability, diminished the percentage of cells in the S-phase, and attenuated the expression of pro-proliferative proteins such as cyclin-dependent kinase and cyclin D1. Furthermore, administration of PZH prompted the condensation and fragmentation of nuclei, triggered the activation of caspase-9 and −3, and elevated the ratio of pro-apoptotic Bcl-2-associated X protein to B-cell lymphoma 2 protein. Notably, treatment with PZH resulted in the upregulation of miR-22 expression and the downregulation of c-Myc expression, which is a target gene of miR-22. So, PZH suppresses the growth of HCT-8/5-FU cells and induces apoptosis, predominantly by regulating the miR-22/c-Myc signaling pathway.87 Furthermore, the impact of PZH on both the apoptosis and proliferation of the BEL-7402 HCC cell line, along with the underlying mechanisms, has been investigated recently. PZH markedly reduced the viability, confluence, and clonogenic potential of BEL-7402 cells. It also elicited cell cycle arrest and facilitated the process of apoptosis. PZH significantly reduced the transcriptional activity of the pro-proliferative genes cyclin-dependent kinase 4 and cyclin D1, alongside a reduction in the expression levels of the anti-apoptotic gene Bcl-2. Additionally, PZH also led to an increase in the expression of a pivotal microRNA, specifically miR-16. So, PZH could efficiently impede the proliferation of cancer cells and trigger apoptosis in BEL-7402 HCC cells by upregulating the tumor suppressor miR-16. Therefore, regulating the miR-16 expression along with its target genes potentially constitutes a mechanism by which PZH exerts its anti-tumor effects.88
Targeting miRNAs-Apoptosis Regulatory Network via Spica PrunellaePrunellae Spica (PS) is the dried flower spike of the herb Prunella vulgaris L., frequently used as an herbal remedy in China, and is known for its anticancer properties.89 Fang et al examine the potential anti-tumor properties of ethanol extract derived from Spica Prunellae (referred to as EESP) in CRC. Initially, EESP exerted a dose-dependent effect on HCT-8 colon carcinoma cells, inducing apoptosis and suppressing cell growth. EESP notably increased the expression of miR-34a. At the same time, the mRNA and protein levels of its target genes, including Notch2, Notch1, and Bcl-2, significantly decreased. Since these target genes are associated with oncogenic properties and have therapeutic implications in apoptosis-related interventions for malignancies, the findings suggest that EESP may inhibit the proliferation of human colon carcinoma cells by inducing apoptosis, possibly through the modulation of Bcl-2, Notch1, and Notch2 expression levels via the activation of miR-34a.90
Targeting miRNAs-apoptosis regulatory network via Scutellaria barbata D. DonScutellaria barbata D. Don (S. barbata) is a widely recognized perennial herb employed within the contexts of traditional Chinese and Korean medicinal practices. Modern pharmacological investigations have demonstrated that S. barbata exhibits diverse biological properties, notably its efficacy in combating cancer.91 Zhang et al investigated the potential of the chloroform fraction derived from Scutellaria barbata D. Don (SB) to inhibit the proliferation of human colon cancer HCT-8 cells. Their findings revealed that ECSB treatment significantly suppressed HCT-8 cell proliferation and induced apoptosis in a dose-dependent manner. Additionally, they demonstrated that ECSB treatment led to a notable increase in miR-34a expression, while simultaneously reducing the expression levels of Jagged1, Bcl-2, and Notch1/2. Their subsequent experiments indicated that ECSB treatment significantly counteracted miR-34a silencing by introducing anti-miR oligonucleotide. Additionally, miR-34a silencing led to a notable increase in the expression of Bcl-2, Notch1/2, and Jagged1, all subsequently reversed upon ECSB administration. Therefore, ECSB suppresses the proliferation of cancer cells by inducing apoptosis and cell division. The modulation of miR‑34a expression mediated this effect. Their findings bolster the utilization of ECSB as a viable therapeutic intervention for colon cancer.92
Targeting miRNAs-Apoptosis Regulatory Network via LuteolinLuteolin, a flavonoid found in Chrysanthemum morifolium Ramat, and commonly used in TCM, has been employed for a long time due to its well-known anti-inflammatory properties. These properties hold promise for clinical applications, particularly in managing cancer patients.93,94 Luteolin suppresses cellular proliferation and prompts apoptosis in gastric cancer cell populations. The expression of miR-34a is decreased in a significant proportion of human primary gastric cancer specimens when juxtaposed with their corresponding non-neoplastic counterparts. Furthermore, miR-34a exerts direct regulatory control over Bcl-2, reducing Bcl-2 protein levels in gastric cancer cells upon miR-34a overexpression. Additionally, Luteolin enhances the expression of miR-34a while suppressing the expression of Bcl-2. Moreover, anti-miR-34a oligonucleotides (AMO) partially attenuate the Luteolin-induced decrease in Bcl-2 expression observed in gastric cancer cells. Thereby, Luteolin reduces Bcl-2 expression by elevating the expression of miR-34a to some extent. In this manner, the miR-34a pathway assumes significance in mediating Luteolin-triggered apoptosis in gastric cancer cell.95
Targeting miRNAs-Apoptosis Regulatory Network via BufalinBufalin serves as a bioactive constituent found within the TCM known as “Chan Su”, derived from the desiccated venomous secretions of the skin glands of either Bufo gargarizans or Bufo melanostictus. Bufalin has been employed as a therapeutic agent for cancer treatment.96 Bufalin could stimulate apoptosis in gastric cancer cells by upregulating BAX expression. Also, miR-298 directly targets BAX and functions as a regulator of BAX expression. Additionally, miR-298 increased the proliferation of gastric cancer cells while impeding apoptosis, whereas bufalin hindered cell proliferation and facilitated apoptosis through the reduction of miR-298. So, bufalin-associated miR-298 could indirectly impact cell proliferation and apoptosis by targeting BAX, thereby suggesting its viability as a prospective therapeutic intervention in the treatment of gastric cancer.97 In addition, the therapeutic response of bufalin has also been demonstrated in prostate cancer. Bufalin administration led to elevated miR-181a expression, with miR-181a induction triggering bufalin-induced apoptosis by downregulating Bcl-2 protein expression in PC-3 cells. Conversely, suppressing miR-181a activity can potentially diminish bufalin-triggered apoptosis in PC-3 cells. In this manner, the miR-181a pathway assumes a significant role in mediating bufalin-triggered apoptosis in gastric cancer cells, thereby indicating the potential of bufalin as a viable therapeutic intervention for gastric cancer.98 Furthermore, bufalin exerted inhibitory effects on cell proliferation and elicited apoptosis via a mitochondria-dependent pathway in Saos-2 and U-2OS osteosarcoma (OS) cell lines, accompanied by the generation of intracellular ROS. Further, bufalin significantly decreases the expression levels of miR-221. Notably, in silico and in vitro analysis identified Bcl2 binding component 3 (BBC3) as the direct molecular target of miR-221. Notably, the reversal of bufalin-induced effects on osteosarcoma cells was achieved by upregulating miR-221 using its mimetic agent (MIMIC) and suppressing BBC3 via siRNA. Thereby, Bufalin triggers mitochondria-dependent apoptosis in OS cells by reducing the expression of miR-221 and inducing the expression of BBC.99
Targeting miRNAs-Apoptosis Regulatory Network via HonokiolHonokiol (HNK) is a small biphenolic compound obtained from the bark of Magnolia officinalis Rehder & E.H. Wilson, known for its antineoplastic properties across a spectrum of cancer types. HNK suppresses proliferation and prompts apoptosis in human osteosarcoma cells in a dose-dependent fashion. HNK-mediated apoptosis is marked by an increase in the levels of proapoptotic proteins, such as Bcl-2-associated X protein, cleaved-poly (ADP-ribose) polymerase, and cleaved-caspase-3, alongside a reduction in the expression of the anti-apoptotic protein Bcl-2. HNK induced dysregulated expression of miRNAs in human OS, with miR-21 being among the most notably reduced miRNAs. HNK decreases miR-21 in a dosage-dependent fashion, and miR-21 restoration counteracted the inhibitory effects of HNK on OS cells. Also, miR-21 suppresses the expression of PTEN by directly interacting with its 3’-UTR. In conclusion, HNK effectively attenuated the PI3K/AKT signaling pathway. Nonetheless, the upregulation of miR-21 led to its reactivation. Taken together, HNK exerts its anticancer effects by modulating the miR-21/PTEN/PI3K/AKT signaling pathway, inhibiting cell proliferation and induction of apoptosis in OS cells.100
Targeting miRNAs-Apoptosis Regulatory Network via SolamargineSolamargine (SM) is a naturally occurring sugar alkaloid derived from Solanum nigrum L. It exhibits notable efficacy in impeding the onset and progression of diverse cancers through various pathways, including those related to apoptosis.101,102 Yin et al examined the potential significance of SM in hepatocellular carcinoma (HCC) treatment and elucidated its mechanisms of exerting an antitumor effect. They showed that SM suppressed the proliferation of HCC and efficiently triggered autophagy and apoptosis in HCC cells both in vivo and in vitro. In terms of mechanism, the oncogenic factor LIF exhibited abnormal elevation in HCC tissues but was diminished by SM in HCC cells. Additionally, LIF upregulation could reinstate the anti-HCC effects of SM through the miR-192-5p/CYR61/Akt signaling pathways. Additionally, SM can reprogram tumor-associated macrophages through the LIF/p-Stat3 pathway, leading to the inhibition of HCC growth and epithelial-mesenchymal transition while also modulating other immune cell populations, including T cells, dendritic cells (DCs), and myeloid-derived suppressor cells (MDSCs) within the tumor microenvironment.103
Targeting miRNAs-Apoptosis Regulatory Network via Zhoushi Qiling DecoctionThe Zhoushi Qi Ling decoction (ZQD) represents a TCM formulation capable of eliciting apoptotic pathways within malignant cells.104 Cao et al sought to assess the effectiveness of ZQD when administered alongside androgen deprivation therapy (ADT) in prostate cancer and to investigate its underlying mechanism of action. Their study enrolled a cohort comprising 151 individuals to undergo either ADT alone or a combination of ADT and ZQD treatment. Evidence showed that the survival rates of patients undergoing combined ADT with ZQD were notably superior to those undergoing ADT monotherapy. Cellular investigations also revealed a notable increase in miR-143 expression within DU145 cells after their ZQD treatment exposure. In serum samples, increased levels of miR-143 expression were identified after treatment with ADT+ZQE. Conversely, such elevated expression was not observed among patients subjected solely to ADT. In DU145 cells, administering ZQD resulted in a dose-dependent elevation of apoptosis, a process mitigated by applying anti-miR-143 treatment. Subsequent investigation into their functional characteristics revealed a specific binding region between miR-143 and Bcl-2. Additionally, it was observed that administration of ZQD resulted in decreased expression levels of Bcl-2. Moreover, they noticed that the administration of ZQD resulted in elevated caspase-3 and Bax expression levels. In this manner, administration of ZQD therapy has the potential to induce apoptosis in prostate cancer cells by increasing miR-143 expression, thus suggesting a plausible mechanism for the observed inhibitory impact of ZQD in prostate cancer patient.105
Targeting miRNAs-Apoptosis Regulatory Network via Icariinlcariin, a prenylated flavonol glycoside derived from Epimedium brevicornum Maxim., has exhibited various pharmacological properties, notably as an anti-cancer agent.106,107 Icariin exerted concurrent inhibitory effects on cell proliferation, facilitated apoptosis, and elevated caspase-3 activity within the A2780 cell line. In ovarian cancer A2780 cells, icariin led to notable reductions in miR-21 expression levels, concurrently augmenting protein expression levels of PTEN and RECK and diminishing Bcl-2 protein expression levels. Notably, miR-21 modulated the putative anticancer properties of icariin on cellular proliferation and apoptosis in A2780 ovarian cancer cells by targeting Bcl-2, RECK, and PTEN. In this manner, the involvement of miR-21 and its associated target genes could significantly influence the underlying molecular processes contributing to the anticancer properties of icariin.108
Targeting miRNAs-Apoptosis Regulatory Network via GeniposideGeniposide, derived from the traditional Chinese herbal remedy “Zhizi”, found in the fruit of Gardenia jasminoides J. Ellis.109 Prior investigations have elucidated its significant involvement in mitigating inflammation, countering oxidative stress, and exhibiting potential antineoplastic properties. Geniposide induces a dose-dependent suppression of OCI-LY7 and OCI-LY3 cell proliferation while concurrently augmenting the proportion of apoptotic cells and elevating cleaved caspase-3 and cleaved PARP levels within DLBCL cellular populations. In DLBCL tissues and cell lines, the expression of long noncoding RNA HLA complex P5 (lncRNA HCP5) increased, whereas administration of geniposide notably attenuated its expression levels. As well, HCP5 silencing led to the suppression of cell proliferation and induction of apoptosis in both OCI-LY3 and OCI-LY7 cell lines. In Vitro, and silico analysis demonstrated miR-27b-3p as a direct target of HCP5, and HCP5 gene suppression and geniposide administration significantly led to an upregulation in the expression of miR-27b-3p within DLBCL cells. Notably, the downregulation of HCP5 led to a decrease in MET protein expression within DLBCL cells, a reduction that was later reversed upon silencing miR-27b-3p. Moreover, exogenous MET administration partially mitigated the proliferative suppression and apoptotic effects of geniposide in DLBCL cells. Thereby, geniposide hinders the proliferation and prompts the apoptosis of DLBCL cells, possibly through modulation of the HCP5/miR-27b-3p/MET axis, suggesting a promising therapeutic approach for managing DLBCL.110
Targeting miRNAs-Apoptosis Regulatory Network via AmentoflavoneAmentoflavone (AMF), an inherent biflavonoid compound, finds extensive application within TCM. AMF was first extracted from the foliage of Selaginella tamariscina, Selaginella rupestris, and Ginkgo biloba. It has been extensively documented as a compound exhibiting diverse biological properties, including anticancer effects.111 Zhaohui et al sought to examine the impact of AMF, a bioactive flavonoid constituent found in Selaginella tamariscina Spring, on glioma cells, elucidating the mechanistic pathways through which it operates. Their findings indicated a notable decrease in the expression of miR-124-3p within glioma tissues compared to those of normal brain tissues. It was additionally revealed that AF reduced cell viability and induced apoptosis in both glioma cell lines in a dose-dependent fashion. Their further experimentation revealed that AF prompted apoptosis and hindered glycolytic activity within glioma cells by upregulating miR-124-3p. Notably, in their study, upregulation of miR-124-3p induced by AF occurred through the repression of DNMT1 via Sp1, subsequently triggered by AF’s activation of the ROS/AMPK signaling pathway. Thereby, AF possesses potent anti-glioma properties by inducing apoptosis and suppressing glycolysis in glioma cells through upregulating miR-124-3p.112
Targeting miRNAs-Apoptosis Regulatory Network via Oroxin BOroxin B (OB), chemically identified as Baicalein7-O-β-gentiobioside with the molecular formula C27H30O15 and a molecular weight of 594.52 Da, represents a flavonoid monomer constituent sourced from the traditional Chinese medicinal plant Oroxylum indicum (L). Vent. Emerging studies demonstrate the anti-lymphoma activity of OB, with preliminary evidence suggesting a lack of overt toxicity. Li et al endeavor to explore the anticancer properties of OB both in vitro and in vivo, elucidating its role in inducing apoptosis in HCC. They noted that OB demonstrates significant anti-hepatocellular carcinoma efficacy both in vitro and in vivo, accompanied by a notable reduction in miR-221 expression compared to the control group. They also observed a reduction in both PI3K mRNA and protein levels in groups treated with OB, compared to the control group, suggesting an upregulation of PTEN expression. Therefore, OB’s anticancer properties manifest through the facilitation of PTEN gene activity, which subsequently leads to the suppression of the PI3K/Akt signaling cascade via the reduction of miR-221
Comments (0)