Abstract
Ginsenosides in ginseng are known for their potential health benefits, including antioxidant properties and their potential to exhibit anticancer effects. Besides a various range of coding genes, ginsenosides impose their efficacy by targeting noncoding RNAs. Long noncoding RNA (
lncRNA) has gained significant attention from both basic and clinical oncology fields due to its involvement in various cancer cell activities such as proliferation, apoptosis, metastasis, and autophagy. These events can be achieved either by lncRNA alone or in association with microRNAs or proteins. This review aims to summarize the diverse activities of lncRNAs that are regulated by ginsenosides, focusing on their role in regulating target genes through signaling pathways in human diseases. We highlight the results of studies on the expression profiles of lncRNAs induced by ginsenosides in efforts to inhibit cancer cell proliferation. Finally, we discuss the potential and challenges of utilizing lncRNAs as diagnostic markers for disease treatment.
Keywords: Ginsenosides, Long noncoding RNAs, Cancer, Diagnostic markers
Graphical abstract

1. Introduction
Ginsenosides, which are bioactive compounds abundant in ginseng, have gained recognition for their potential health benefits, including antioxidant properties and anti-cancer effects [1,2]. These compounds can exert their efficacy by targeting noncoding RNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) [3,4]. LncRNAs, a class of noncoding RNAs longer than 200 nucleotides, do not code for proteins [5,6]. Instead, the transcripts themselves regulate the expression of other genes through diverse mechanisms [7,8]. These mechanisms include modulating the expression of nearby genes in a cis-acting mode [9], binding to target RNA to induce degradation or modulation [10], or regulating protein activity after forming complexes with specific proteins [11]. As of December 1st, 2023, 56,946 lncRNA genes have been identified in humans (http://lncipedia.org), with many of them being associated with cancer [12] (see Fig. 1).
Fig. 1.
The action modes of ginsenosides and lncRNAs in cancer cells. Ginsenosides have been shown to either up- or down-regulate various lncRNAs through mechanisms that are not well elucidated. One such mechanism involves the suppression of CFAP20DC-AS1 by hypermethylation at the promoter CpGs. These lncRNAs, in turn, exert their influence through several mechanisms. They can control the expression of nearby genes (cis genes), act as sponges for microRNAs, modify histones or IκB, or modulate target proteins. These diverse events collectively regulate cancer-related target genes, resulting in the alteration of cancer cell activities such as apoptosis, metastasis, the Warburg effect, and the ER stress pathway.
Despite the prevalence of lncRNAs over miRNAs in human cells, there is limited understanding of how ginsenosides regulate lncRNAs in diseases. Further research is needed to elucidate their roles in conjunction with ginsenosides during disease progression. The complex engagement of lncRNAs in various cellular processes in cancer and other diseases, including proliferation, apoptosis, metastasis, and autophagy, has generated considerable interest in their potential as therapeutic targets [[13], [14], [15]]. This review focuses on summarizing the varied activities of lncRNAs regulated by ginsenosides, emphasizing their role in modulating target genes through signaling pathways in human diseases, particularly cancer. The exploration encompasses the expression profiles of lncRNAs induced by ginsenosides and their impact on inhibiting cancer cell proliferation. Additionally, we delve into the potential of utilizing lncRNAs as diagnostic markers for disease treatment, presenting a promising avenue for the development of targeted therapies with clinical applications [16,17].
Ginsenosides, including various types like Rh2, Rh3, Rg1, and Rg3, exhibit multifaceted pharmacological activities in numerous cancers and other diseases. For instance, Rh2 demonstrates therapeutic potential in breast cancer by intricately regulating lncRNAs such as STXBP5-AS1 [18] and CFAP20DC-AS1 (formerly known as C3orf67-AS1) [19]. Similarly, Rh3 and Rg1 exhibit diverse impacts on cancer modulation, underlining their potential in personalized therapeutic interventions [20,21]. These ginsenosides also have effects on diseases including myocardial injury, neurodegenerative disease, and skin disease [[22], [23], [24], [25]]. Moreover, other ginsenosides, including Rb3, R1, Rd, Rh7, and compound K, along with non-ginsenoside molecules in ginseng extracts, contribute to the expansive range of therapeutic effects observed [[26], [27], [28], [29], [30]]. Despite notable progress, challenges persist in understanding the complete spectrum of lncRNAs influenced by ginsenosides in cancer. Further research is warranted to unravel the intricate interactions between ginsenosides and lncRNAs, with the anticipation that novel lncRNAs will be discovered, enriching our comprehension of their roles in cancer development. This review sets the stage for future investigations into the epigenetic aspects of ginsenosides, offering valuable insights for the development of targeted cancer therapies and highlighting the molecular complexity of cancer for potential personalized therapeutic interventions.
2. Ginsenodides, lncRNAs, and diseases
2.1. Rh2 and lncRNAs
Rh2 is derived from the protopanaxadiol group, which is one of the major groups of ginsenosides [31]. Rh2 is being studied for its potential to impede cancer cell growth through multiple mechanisms. It disrupts the cell cycle, inhibiting the division of cancer cells, and induces apoptosis in specific cancer cell types [32]. Additionally, Rh2 is implicated in limiting angiogenesis, the formation of blood vessels supplying nutrients to tumors, potentially reducing the blood supply to cancer cells [33]. Moreover, it may suppress the metastatic spread of cancer cells, possibly by influencing gene expression related to metastasis [34]. The compound interacts with crucial signaling pathways like PI3K/Akt and MAPK, impacting cancer cell proliferation and survival [35].
Rh2 emerges as a pivotal player in cancer modulation by intricately regulating lncRNAs (Table 1). Specifically, Rh2 demonstrates versatile therapeutic potential in breast cancer by downregulating CFAP20DC-AS1, a miR-3614-3p sponge, thereby disrupting key pathways associated with proliferation and apoptosis. The nuanced interplay between Rh2, miR-3614-3p, and their downstream targets BBX and TNFAIP3 underscores Rh2's significance in orchestrating anti-proliferative responses in cancer cells [4]. In a separate context, Rh2 induces reversible hypermethylation of the C3orf67-AS1 promoter in MCF-7 cells, leading to its downregulation. Reduced C3orf67-AS1 inhibits cell growth, and clinical data suggests its potential role in anti-proliferative effects [19]. Furthermore, microarray analysis in HepG2 cells treated with Rh2 reveals differential expression of lncRNAs, thereby suggesting potential targets for hepatocellular carcinoma therapy [36]. This underscores the broad impact of Rh2 on cancer-related pathways. In the context of bone formation, Rh2 treatment in MC3T3-E1 cells is shown to increase the expression of lncRNA H19, leading to osteopontin (OPN) overexpression. The knockdown of H19 inhibits Rh2-induced cell proliferation and reduces OPN levels through the regulation of histone acetylation on the OPN promoter. This emphasizes the significance of H19 in Rh2-mediated bone formation [37]. Additionally, in a mouse cornea alkali burn model, Rh2 eyedrops exhibit inhibitory effects on alkali-induced neovascularization and inflammatory cell infiltrations. Notably, Rh2 attenuates the alkali-induced expression of mRNAs and lncRNAs in the cornea [38]. In a distinct context, Rh2 exhibits the ability to upregulate the expression of hub miRNAs, suppress the upregulation of prognosis-associated lncRNAs, and ultimately restore the expression of cancer-related lncRNAs to precancerous levels. Additionally, these findings position Rh2 as a potential candidate for small molecule drug development in kidney renal clear cell carcinoma (KIRC) treatment, presenting a novel approach for KIRC therapy [39].
Table 1.
LncRNAs regulated by ginsenoside Rh2.
| LncRNA | Expression alteration by ginsenoside | Mode of activity of lncRNA | Cell line/Tissue | Reference |
|---|---|---|---|---|
| STXBP5-AS1 | Up | Sponges miR-4425 to suppress cancer cell proliferation | MCF-7/Breast | [18] |
| CFAP20DC-AS1 | Down | Sponge miR-3614-3p to induce apoptosis | MCF-7/Breast | [4] |
| C3orf67-AS1 | Down | Hypermethylated by Rh2 at promoter | MCF-7/Breast | [19] |
| Multiple lncRNAs | Variable | Potential targets for hepatoma therapy | HepG2/Liver | [36] |
| H19 | Up | Increases osteopontin expression | MC3T3-E1 | [37] |
| Multiple lncRNAs | Variable | Inhibit corneal neovascularization | Human umbilical vein endothelial cells | [38] |
| Multiple lncRNAs | Variable | Serve as biomarkers for cancer prognosis | 9 types of cancer | [39] |
Collectively, these diverse findings shed light on the multifaceted mechanisms by which Rh2 exerts its therapeutic effects via lncRNAs in numerous cancers, such as breast cancer and hepatocellular carcinoma, as well as in various human diseases, including bone formation and even corneal health. These insights offer hope for the advancement of interventions based on Rh2 and lncRNAs, with potential clinical implications.
2.2. Rg1 and lncRNAs
Rg1 exhibits diverse pharmacological activities, including anti-inflammatory effects, neuroprotection, cognitive enhancement, cardioprotection, potential anti-cancer properties, immunomodulation, and anti-fatigue effects, making it a subject of interest for its potential therapeutic applications in various health conditions [40,41]. In a study examining Müller cells under high-glucose conditions, it was revealed that Rg1 effectively mitigated mesenchymal activation and fibrosis by regulating the miR-2113/RP11-982M15.8/Zeb1 pathway. This was evidenced by its impact on MMP-2, MMP-9, and TIMP-2 expression (Table 2) [42]. Furthermore, Rg1 was found to boost proliferation and adipogenic differentiation in human adipose-derived stem cells (hASCs) by influencing pathways such as adipocytokine signaling and IL-17 signaling. This effect is mediated through regulatory factors FXR1 and Lnc-GAS5-AS1, highlighting Rg1's potential in promoting hASCs by modulating these pathways [43]. In high glucose-induced human retinal endothelial cells, Rg1 inhibited proliferation, migration, and angiogenesis by upregulating the lncRNA SNHG7. MiR-2116-5p had a target regulatory relationship with both lncRNA SNHG7 and SIRT3 [44]. Additionally, Rg1 reduced microglial activation and mitochondrial dysfunction to alleviate depression-like behavior via the gas5/ezh2/socs3/nrf2 axis [45]. In the cancer context, lncRNA GAS5 acts as a tumor suppressor by orchestrating the cell cycle, fostering apoptosis, and interacting with pivotal genes in the B-cell lymphoma development [46]. GAS5 also inhibits angiogenesis and disrupts the glucocorticoid receptor pathway, stressing its multifaceted role as a potential target for lung cancer therapies and a valuable biomarker for prognosis and diagnosis [47].
Table 2.
LncRNAs regulated by ginsenoside Rg1.
| LncRNA | Expression alteration by ginsenoside | Mode of activity of lncRNA | Cell line/Tissue | Reference |
|---|---|---|---|---|
| RP11-982M15.8 | Down | Sponge miR-2113 to inhibit mesenchymal activation and fibrosis | MIO-M1/Retina | [42] |
| GAS5-AS1 | Down | Suppress proliferation of adipose-derived stem cells | Human adipose-derived stem cells | [43] |
| SNHG7 | Up | Sponge miR-2116-5p | HREC/retina | [44] |
| GAS5 | Down | Stimulate depression-like behavior | Rat brain | [45] |
2.3. Rg3 and lncRNAs
Rg3 demonstrates notable anti-cancer effects across various malignancies. In breast cancer, Rg3's impact intertwines with STXBP5-AS1 and RFX3-AS1, influencing cell proliferation and metastasis-free survival (Table 3) [48]. In ovarian cancer, Rg3 targets H19, disrupting the Warburg effect and tumorigenesis [49,50]. For gemcitabine-resistant pancreatic cancer, Rg3 overcomes chemoresistance by inducing apoptosis and modulating lncRNA CASC2/PTEN signaling [51]. In gliomas, Rg3 counters lncRNA NKILA-induced hypoxia, the Warburg effect, and angiogenesis [52]. In hepatocellular carcinoma, Rg3 reduces HOTAIR levels, inhibiting proliferation, migration, and invasion [53]. In osteosarcoma, Rg3 impacts SOX21-AS1, regulating mTOR and KLF4 for cell proliferation [54]. In sepsis-induced liver injury, Rg3 enhances lncRNA TUG1 expression, activating the SIRT1/AMPK pathway and promoting autophagy [55]. Additionally, Rg3 exhibits regulation of multiple lncRNAs while managing cerebral ischemia-reperfusion injury, suggesting its potential as a neuroprotectant in rats [56]. Notably, in MCF-7 breast cancer cells, Rg3-induced hypermethylation downregulates ATXN8OS, suppressing cell proliferation and promoting apoptosis [57]. In colon cancer cells, Rg3 reduces ATXN8OS, inhibiting metastasis by down-regulating miR-15b-5p and inhibiting AXIN2 [58]. Lastly, in gallbladder cancer xenografts, Rg3 inhibits tumor growth by upregulating the ER stress-mediated signaling pathway, achieved through the upregulation of the long non-coding RNA p21 [59]. These findings collectively underscore the diverse and promising anti-cancer properties of Rg3 across various cancer types, emphasizing its significant impact on lncRNA-mediated regulatory networks.
Table 3.
LncRNAs regulated by ginsenoside Rg3.
| LncRNA | Expression alteration by ginsenoside | Mode of activity of lncRNA | Cell line/Tissue | Reference |
|---|---|---|---|---|
| ATXN8OS | Down | Sponges miR-424-5p | MCF-7/Breast | [57] |
| ATXN8OS | Down | Sponges miR-15b-5p | Colon | [58] |
| STXBP5-AS1 | Up | Increase apoptosis | MCF-7/Breast | [48] |
| RFX3-AS1 | Down | Decrease apoptosis | MCF-7/Breast | [48] |
| H19 | Down | Enhance Warburg effect | SKOV3/Ovary | [49] |
| CASC2 | Up | Activate PTEN signaling | Panc-1,SW1990/Pancreas | [51] |
| NKILA | Down | Enhance Warburg effect | LN229 et al./Glioma | [52] |
| H19 | Down | Enhance proliferation and invasion | SKOV3 and A2780)/Ovary | [50] |
| HOTAIR | Down | Enhance proliferation and invasion | SMMC-7721, SK-Hep-1/Liver | [53] |
| SOX21-AS1 | Down | Sponge miR-7-5p, miR-145-5p | 143B/Bone | [54] |
| TUG1 | Up | Sponge miR-200a-3p | Primary hepatocytes/Liver | [55] |
| Multiple lncRNAs | Variable | Involved in neuroprotection | Rat brain | [56] |
| p21 | Up | Activate ER stress pathway | GBC-SD/gall bladder | [59] |
2.4. Other ginsenosides and lncRNAs
Ginsenosides, including Rb3, R1, Rd, Rh7, and compound K (CK), show versatile therapeutic effects. For instance, ginsenoside Rd demonstrated the suppression of SCC9 cell growth and migration, inducing apoptosis and inhibiting metastasis by downregulating lncRNA H19 and miR-675-5p, resulting in elevated CDH1 and E-cadherin expression (Table 4) [60]. In cardiac scenarios, Panax notoginseng saponins (PNS), particularly R1, enhanced cardiac function, reduced fibrosis and pyroptosis, and modulated ANRIL expression [61]. ANRIL has been implicated in various diseases, particularly cardiovascular disorders, where it modulates pathways related to inflammation, oxidative stress, and vascular dysfunction. Its dysregulation is associated with increased susceptibility to atherosclerosis, coronary artery disease, and other cardiovascular conditions [62]. PNS also exhibited anti-apoptotic effects in H9C2 cardiomyocytes during oxygen-glucose depletion (OGD)-induced myocardial ischemia, with RNA-seq analysis revealing notable changes in lncRNA expression post-PNS treatment, suggesting a potential cardioprotective role through non-coding RNA regulation [63]. Notoginsenoside R1 further lowered blood pressure in spontaneously hypertensive rats by inducing nitric oxide generation through AK094457-mediated iNOS expression [64]. Moreover, R1 demonstrated regulatory effects on proliferation, apoptosis, inflammatory response, and oxidative stress in human umbilical vein endothelial cells (HUVECs) exposed to oxidized low-density lipoprotein. This was achieved through the modulation of the XIST/miR-221-3p/TRAF6 axis via the NF-κB pathway [65].
Table 4.
LncRNAs regulated by other ginsenosides.
| LncRNA (ginsenoside) | Expression alteration by ginsenoside | Mode of activity of lncRNA | Cell line/Tissue | Reference |
|---|---|---|---|---|
| H19 (Rd) | Down | Enhance migration and invasion | SCC9/Tongue | [60] |
| ANRIL (Saponin) | Down | Enhance pyroptosis | H9C2/Rat heart | [61] |
| AK094457 (R1) | Down | Enhance blood pressure | WYK/Rat blood vessel | [64] |
| XIST (R1) | Down | Sponge miR-221-3p | Human umbilical vein endothelial cells | [65] |
| Multiple lncRNAs (Rf) | Variable | Inhibit tau proteotoxicity | C. elegans | [66] |
| Multiple lncRNAs (Saponin) | Variable | Protect cardiomyocytes from apoptosis | H9C2/Rat heart | [63] |
| THOR (Compound K) | Down | Enhance growth of renal carcinoma cell | Caki-1, 768-O/Kidney | [67] |
| H19 (Rb3) | Down | Sponge miR-29b-3p | A549, H460/Lung | [68] |
| ILF3-AS1 (Rh7) | Down | Sponge miR-212 | A549, H1299/Lung | [29] |
Ginsenoside Rf altered expression levels of lncRNAs, miRNAs, and mRNAs, potentially impacting tauopathy in neurodegenerative diseases in a worm model [66]. Eight lncRNAs (MSTRG.20812.2, MSTRG.22617.2, MSTRG.28210.13, MSTRG.5728.12, MSTRG.29708.1, MSTRG.3342.25, MSTRG.3342.31, and MSTRG.8841.8) were identified as potential targets of Rf in the inhibition of tauopathy. Ginsenoside compound K demonstrated anti-tumor effects in renal cell carcinoma by inducing apoptosis and inhibiting lncRNA THOR [67]. Additionally, Rb3 exhibited therapeutic effects in smoke-induced lung injury by modulating the H19/miR-29b-3p/HMBG1 signaling pathway [68]. Furthermore, ginsenoside Rh7, through the regulation of ILF3-AS1, influenced proliferation and metastasis in non-small cell lung cancer by modulating the miR-212/SMAD1 axis [29]. Together, these findings propose the diverse therapeutic potential of ginsenosides, emphasizing their significant impact on lncRNA-mediated regulatory networks across various physiological and pathological conditions.
2.5. Non-ginsenoside molecules of ginseng and lncRNAs
While this review primarily focuses on ginsenoside-regulated lncRNAs in human diseases, the inclusion of non-ginsenoside molecules from ginseng extracts broadens the scope to encompass the wider regulatory landscape. Specifically, a tRNA-derived fragment (tRF) originated from the 3′ end of tRNAGln (UUG) in ginseng directly suppresses the MIAT long noncoding RNA, resulting in increased expression of VEGFA. Introduction of a tRF mimic demonstrates notable enhancements in cardiac function, preserving cardiomyocyte cytoskeleton integrity and mitochondrial functionality [69]. In another study, a Chinese anti-tumor medicine, ADI, whose formula incorporates ginseng, was utilized to discern target genes and regulatory networks. This investigation revealed the presence of nine lncRNA nodes, including OIP5-AS1 and FDG5-AS1, within a competitive endogenous RNA regulatory network [70]. However, the specific ginseng component responsible for inducing these lncRNAs remains unidentified.
3. Regulatory mechanism of lncRNAs dysregulated by ginsenosides
LncRNAs play a pivotal role in cellular regulation through diverse mechanisms, encompassing sponging activity, chromatin modification, transcriptional regulation, modulation of RNA stability, influence on subcellular localization, and modulation of protein activity [71,72]. A literature review delving into the regulation of lncRNAs by ginsenosides has revealed a predominant emphasis on the modulation of protein activity and sponging activity, as outlined in the following sections. Despite significant progress, the regulatory mechanisms by which ginsenosides induce the expression of specific lncRNAs remain incompletely understood. One contributing factor to this gap in knowledge is the alteration of methylation levels at the CpG site of lncRNAs, as exemplified by C3orf67-AS1 [19]. Further research is necessary to unveil the intricate molecular interactions and signaling pathways involved in the ginsenoside-mediated regulation of lncRNAs.
3.1. Transcriptional regulation
In the context of transcriptional regulation, lncRNAs exert their influence over gene expression by interacting with key molecular players such as transcriptional factors and histones [73,74]. The present section also delineates the impact of lncRNAs on genes within a signaling pathway, wherein their expression undergoes modulation. One illustrative instance of this intricate regulatory dance involves the adjustment of histone acetylation, as demonstrated in the study of lncRNA H19. The suppression of H19 resulted in a reduction of osteopontin mRNA and protein levels in Rh2-treated cells, achieved through the inhibition of histones H3 and H4 acetylation on the osteopontin promoter [37]. LncRNAs induced up- or down-regulation of various genes such as PTEN [51], E-cadherin [50], MMP2, MMP9 [53] (Rg3), SOCS3, NRF2 [45] (Rg1), and iNOS [64] (R1).
3.2. Sponging microRNAs
The term "sponging activity" refers to a regulatory mechanism by which lncRNAs act as molecular sponges for miRNAs [75]. By binding to miRNAs, lncRNAs prevent them from interacting with their mRNA targets. The sequestration of miRNAs by lncRNAs can result in increased stability and expression of the target mRNAs that would have been otherwise downregulated by the miRNAs. Dysregulation of this sponging activity can have implications in diseases, including cancer, where aberrant expression of lncRNAs and miRNAs is often observed [76,77]. Of the approximately 50 lncRNAs reviewed in the current manuscript, 10 are known to act as miRNA sponges (Table 1, Table 2, Table 3, Table 4). Rg3, for example, has been implicated in upregulating FDFT1 by reducing miR-4425 levels, thereby inhibiting ovarian cancer progression [78]. MiR-4425 has been found to be sponged by the tumor-suppressive lncRNA HCG11 in glioma, indicating a complex interplay between ginsenosides, lncRNAs, and microRNAs [79].
3.3. Protein modulation
LncRNA NKILA is upregulated by NFκB and binds strongly to NFκB/IκB. NKILA masks IκB phosphorylation motifs, thereby stabilizing the NFκB/IκB complex by inhibiting IκB phosphorylation and NFκB activation [11]. Rg3 suppresses NKILA accumulation and reverses its stimulation of the Warburg effect and angiogenesis in glioma [52]. H19, when complexed with the methyl-CpG-binding domain protein 1 (MBD1), binds to the control region of its target genes and finely tunes histone methylation [80]. As described previously, the expression of H19 is regulated by various ginsenosides, including Rg3, Rh2, and Rb3.
4. Conclusions
In summary, this review explores the intricate relationship between ginsenosides and long non-coding RNAs (lncRNAs) in cancer and other diseases. Ginsenosides like Rh2, Rh3, Rg1, and Rg3 demonstrate significant potential in regulating lncRNAs to hinder cancer progression by inhibiting cell proliferation, inducing apoptosis, and suppressing metastasis. These interactions also hold promise for interventions in cardiovascular disorders, neurodegenerative diseases, inflammation, and metabolic disorders. Highlighting the pivotal role of lncRNAs in mediating ginsenoside-induced anticancer effects, the review discusses specific lncRNAs such as CFAP20DC-AS1, C3orf67-AS1, H19, GAS5, STXBP5-AS1, and RFX3-AS1. Additionally, it explores the potential of using lncRNAs as diagnostic markers and emphasizes the need for further research to comprehend the complete spectrum of ginsenoside-influenced lncRNAs in cancer. Overall, this concise review contributes to our understanding of ginsenoside epigenetics and presents new opportunities for personalized therapeutic interventions in cancer and beyond.
Conflicts of interests
The authors declare that they have no conflicts of interests.
Acknowledgments
This work was supported by the Basic Science Research Program (NRF-2022R1A2C1003483) of the National Research Foundation of Korea, funded by the Ministry of Education, Science and Technology.
References
- 1.Ahuja A., Kim J.H., Kim J.H., Yi Y.S., Cho J.Y. Functional role of ginseng-derived compounds in cancer. J Ginseng Res. 2018;42:248–254. doi: 10.1016/j.jgr.2017.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Deng X., Wang J., Lu C., Zhou Y., Shen L., Ge A., Fan H., Liu L. Updating the therapeutic role of ginsenosides in breast cancer: a bibliometrics study to an in-depth review. Front Pharmacol. 2023;14 doi: 10.3389/fphar.2023.1226629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Fan M., Shan M., Lan X., Fang X., Song D., Luo H., Wu D. Anti-cancer effect and potential microRNAs targets of ginsenosides against breast cancer. Front Pharmacol. 2022;13 doi: 10.3389/fphar.2022.1033017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Park J.E., Ji H.W., Kim H.W., Baek M., Jung S., Kim S.J. Ginsenoside Rh2 regulates the CFAP20DC-AS1/MicroRNA-3614-3p/BBX and TNFAIP3 Axis to induce apoptosis in breast cancer cells. Am J Chin Med. 2022;50:1703–1717. doi: 10.1142/S0192415X22500720. [DOI] [PubMed] [Google Scholar]
- 5.Gupta R.A., Shah N., Wang K.C., Kim J., Horlings H.M., Wong D.J., Tsai M.C., Hung T., Argani P., Rinn J.L., et al. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010;464:1071–1076. doi: 10.1038/nature08975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Das P.K., Siddika A., Rashel K.M., Auwal A., Soha K., Rahman M.A., Pillai S., Islam F. Roles of long noncoding RNA in triple-negative breast cancer. Cancer Med. 2023;12:20365–20379. doi: 10.1002/cam4.6600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Liu L., Zhang Y., Lu J. The roles of long noncoding RNAs in breast cancer metastasis. Cell Death Dis. 2020;11:749. doi: 10.1038/s41419-020-02954-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bartonicek N., Maag J.L., Dinger M.E. Long noncoding RNAs in cancer: mechanisms of action and technological advancements. Mol Cancer. 2016;15:43. doi: 10.1186/s12943-016-0530-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Nie Y., Zhou L., Wang H., Chen N., Jia L., Wang C., Wang Y., Chen J., Wen X., Niu C., et al. Profiling the epigenetic interplay of lncRNA RUNXOR and oncogenic RUNX1 in breast cancer cells by gene in situ cis-activation. Am J Cancer Res. 2019;9:1635–1649. [PMC free article] [PubMed] [Google Scholar]
- 10.Nie W., Ge H.J., Yang X.Q., Sun X., Huang H., Tao X., Chen W.S., Li B. LncRNA-UCA1 exerts oncogenic functions in non-small cell lung cancer by targeting miR-193a-3p. Cancer Lett. 2016;371:99–106. doi: 10.1016/j.canlet.2015.11.024. [DOI] [PubMed] [Google Scholar]
- 11.Liu B., Sun L., Liu Q., Gong C., Yao Y., Lv X., Lin L., Yao H., Su F., Li D., et al. A cytoplasmic NF-κB interacting long noncoding RNA blocks IκB phosphorylation and suppresses breast cancer metastasis. Cancer Cell. 2015;27:370–381. doi: 10.1016/j.ccell.2015.02.004. [DOI] [PubMed] [Google Scholar]
- 12.Kalhori M.R., Khodayari H., Khodayari S., Vesovic M., Jackson G., Farzaei M.H., Bishayee A. Regulation of long non-coding RNAs by plant secondary metabolites: a novel anticancer therapeutic approach. Cancers. 2021;13:1274. doi: 10.3390/cancers13061274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Chen X., Zhao Y., Wang D., Lin Y., Hou J., Xu X., Wu J., Zhong L., Zhou Y., Shen J., et al. The HNF4α-bc200-FMR1-positive feedback loop promotes growth and metastasis in invasive mucinous lung adenocarcinoma. Cancer Res. 2021;81:5904–5918. doi: 10.1158/0008-5472.CAN-21-0980. [DOI] [PubMed] [Google Scholar]
- 14.Zhang C., Shen C. Identification of hub gene and lncRNA signature related to entotic cell death in cutaneous melanoma for prognostic and immune prediction. Medicine (Baltim) 2023;102 doi: 10.1097/MD.0000000000035881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Balusu S., Horré K., Thrupp N., Craessaerts K., Snellinx A., Serneels L., T'Syen D., Chrysidou I., Arranz A.M., Sierksma A., et al. MEG3 activates necroptosis in human neuron xenografts modeling Alzheimer's disease. Science. 2023;381:1176–1182. doi: 10.1126/science.abp9556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Uttam V., Rana M.K., Sharma U., Singh K., Jain A. Circulating long non-coding RNA EWSAT1 acts as a liquid biopsy marker for esophageal squamous cell carcinoma: a pilot study. Noncoding RNA Res. 2024;9:1–11. doi: 10.1016/j.ncrna.2023.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Yang Q., Wang M., Xu J., Yu D., Li Y., Chen Y., Zhang X., Zhang J., Gu J., Zhang X. LINC02159 promotes non-small cell lung cancer progression via ALYREF/YAP1 signaling. Mol Cancer. 2023;22:122. doi: 10.1186/s12943-023-01814-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Park J.E., Kim H.W., Yun S.H., Kim S.J. Ginsenoside Rh2 upregulates long noncoding RNA STXBP5-AS1 to sponge microRNA-4425 in suppressing breast cancer cell proliferation. J Ginseng Res. 2021;45:754–762. doi: 10.1016/j.jgr.2021.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Jeong D., Ham J., Park S., Kim H.W., Kim H., Ji H.W., Kim S.J. Ginsenoside Rh2 suppresses breast cancer cell proliferation by epigenetically regulating the long noncoding RNA C3orf67-AS1. Am J Chin Med. 2019;47:1643–1658. doi: 10.1142/S0192415X19500848. [DOI] [PubMed] [Google Scholar]
- 20.Cong Z., Zhao Q., Yang B., Cong D., Zhou Y., Lei X., Zhang X. Ginsenoside Rh3 inhibits proliferation and induces apoptosis of colorectal cancer cells. Pharmacology. 2020;105:329–338. doi: 10.1159/000503821. [DOI] [PubMed] [Google Scholar]
- 21.Luo Y., Wang B., Liu J., Ma F., Luo D., Zheng Z., Lu Q., Zhou W., Zheng Y., Zhang C., et al. Ginsenoside RG1 enhances the paracrine effects of bone marrow-derived mesenchymal stem cells on radiation induced intestinal injury. Aging (Albany N Y) 2020;13:1132–1152. doi: 10.18632/aging.202241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Qi Z., Yan Z., Wang Y., Ji N., Yang X., Zhang A., Li M., Xu F., Zhang J. Ginsenoside Rh2 inhibits NLRP3 inflammasome activation and improves exosomes to alleviate hypoxia-induced myocardial injury. Front Immunol. 2022;13 doi: 10.3389/fimmu.2022.883946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kim E.J., Jung I.H., Van Le T.K., Jeong J.J., Kim N.J., Kim D.H. Ginsenosides Rg5 and Rh3 protect scopolamine-induced memory deficits in mice. J Ethnopharmacol. 2013;146:294–299. doi: 10.1016/j.jep.2012.12.047. [DOI] [PubMed] [Google Scholar]
- 24.Zhou T., Zu G., Zhang X., Wang X., Li S., Gong X., Liang Z., Zhao J. Neuroprotective effects of ginsenoside Rg1 through the Wnt/β-catenin signaling pathway in both in vivo and in vitro models of Parkinson's disease. Neuropharmacology. 2016;101:480–489. doi: 10.1016/j.neuropharm.2015.10.024. [DOI] [PubMed] [Google Scholar]
- 25.Lee H., Hong Y., Tran Q., Cho H., Kim M., Kim C., Kwon S.H., Park S., Park J., Park J. A new role for the ginsenoside RG3 in antiaging via mitochondria function in ultraviolet-irradiated human dermal fibroblasts. J Ginseng Res. 2019;43:431–441. doi: 10.1016/j.jgr.2018.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Luo X., Wang H., Ji D. Carbon nanotubes (CNT)-loaded ginsenosides Rb3 suppresses the PD-1/PD-L1 pathway in triple-negative breast cancer. Aging (Albany N Y) 2021;13:17177–17189. doi: 10.18632/aging.203131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Wang H., Yan Y., Lan H., Wei N., Zheng Z., Wu L., Jaspers R.T., Wu G., Pathak J.L. Notoginsenoside R1 promotes migration, adhesin, spreading, and osteogenic differentiation of human adipose tissue-derived mesenchymal stromal cells. Molecules. 2022;27:3403. doi: 10.3390/molecules27113403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Cui T., Xiao X., Pan Z., Tang K., Zhong Y., Chen Y., Guo J., Duan S., Zhong G., Li T., et al. Harnessing the therapeutic potential of ginsenoside Rd for activating SIRT6 in treating a mouse model of nonalcoholic fatty liver disease. ACS Omega. 2023;8:29735–29745. doi: 10.1021/acsomega.3c04122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Chen X., Liu W., Liu B. Ginsenoside Rh7 suppresses proliferation, migration and invasion of NSCLC cells through targeting ILF3-AS1 mediated miR-212/SMAD1 Axis. Front Oncol. 2021;11 doi: 10.3389/fonc.2021.656132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Liu M., Zhang Y., Zhang A., Deng Y., Gao X., Wang J., Wang Y., Wang S., Liu J., Chen S., et al. Compound K is a potential clinical anticancer agent in prostate cancer by arresting cell cycle. Phytomedicine. 2023;109 doi: 10.1016/j.phymed.2022.154584. [DOI] [PubMed] [Google Scholar]
- 31.Liang J., Tang X., Wan S., Guo J., Zhao P., Lu L. Structure modification of ginsenoside Rh(2) and cytostatic activity on cancer cells. ACS Omega. 2023;8:17245–17253. doi: 10.1021/acsomega.3c01665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hwang H.J., Hong S.H., Moon H.S., Yoon Y.E., Park S.Y. Ginsenoside Rh2 sensitizes the anti-cancer effects of sunitinib by inducing cell cycle arrest in renal cell carcinoma. Sci Rep. 2022;12 doi: 10.1038/s41598-022-20075-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Huang Y., Huang H., Han Z., Li W., Mai Z., Yuan R. Ginsenoside Rh2 inhibits angiogenesis in prostate cancer by targeting CNNM1. J Nanosci Nanotechnol. 2019;19:1942–1950. doi: 10.1166/jnn.2019.16404. [DOI] [PubMed] [Google Scholar]
- 34.Hu Q.R., Huang Q.X., Hong H., Pan Y., Luo T., Li J., Deng Z.Y., Chen F. Ginsenoside Rh2 and its octyl ester derivative inhibited invasion and metastasis of hepatocellular carcinoma via the c-Jun/COX2/PGE2 pathway. Phytomedicine. 2023;121 doi: 10.1016/j.phymed.2023.155131. [DOI] [PubMed] [Google Scholar]
- 35.Li C., Gao H., Feng X., Bi C., Zhang J., Yin J. Ginsenoside Rh2 impedes proliferation and migration and induces apoptosis by regulating NF-κB, MAPK, and PI3K/Akt/mTOR signaling pathways in osteosarcoma cells. J Biochem Mol Toxicol. 2020;34 doi: 10.1002/jbt.22597. [DOI] [PubMed] [Google Scholar]
- 36.Chen W.W., Huang Y.F., Hu Z.B., Liu Y.M., Xiao H.X., Liu D.B., Zhuang Y.Z. Microarray analysis of altered long non-coding RNA expression profile in liver cancer cells treated by ginsenoside Rh2. J Asian Nat Prod Res. 2019;21:742–753. doi: 10.1080/10286020.2018.1490273. [DOI] [PubMed] [Google Scholar]
- 37.Dong B., Pang T.T. LncRNA H19 contributes to Rh2-mediated MC3T3-E1cell proliferation by regulation of osteopontin. Cell Mol Biol (Noisy-le-grand) 2017;63:1–6. doi: 10.14715/cmb/2017.63.8.1. [DOI] [PubMed] [Google Scholar]
- 38.Zhang X.P., Li K.R., Yu Q., Yao M.D., Ge H.M., Li X.M., Jiang Q., Yao J., Cao C. Ginsenoside Rh2 inhibits vascular endothelial growth factor-induced corneal neovascularization. FASEB J. 2018;32:3782–3791. doi: 10.1096/fj.201701074RR. [DOI] [PubMed] [Google Scholar]
- 39.Zhu Y., Dong S., Zhu Y., Zhao Y., Xu Y. Identification of cancer prognosis-associated lncRNAs based on the miRNA-TF co-regulatory motifs and dosage sensitivity. Mol Omics. 2019;15:361–373. doi: 10.1039/c9mo00089e. [DOI] [PubMed] [Google Scholar]
- 40.Gao Y., Li J., Wang J., Li X., Li J., Chu S., Li L., Chen N., Zhang L. Ginsenoside Rg1 prevent and treat inflammatory diseases: a review. Int Immunopharmacol. 2020;87 doi: 10.1016/j.intimp.2020.106805. [DOI] [PubMed] [Google Scholar]
- 41.Sun Y., Yang Y., Liu S., Yang S., Chen C., Lin M., Zeng Q., Long J., Yao J., Yi F., et al. New therapeutic approaches to and mechanisms of ginsenoside Rg1 against neurological diseases. Cells. 2022;11:2529. doi: 10.3390/cells11162529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Xue L.P., Fu X.L., Hu M., Zhang L.W., Li Y.D., Peng Y.L., Ding P. Rg1 inhibits high glucose-induced mesenchymal activation and fibrosis via regulating miR-2113/RP11-982M15.8/Zeb1 pathway. Biochem Biophys Res Commun. 2018;501:827–832. doi: 10.1016/j.bbrc.2018.04.055. [DOI] [PubMed] [Google Scholar]
- 43.Xu F.T., Xu Y.L., Rong Y.X., Huang D.L., Lai Z.H., Liu X.H., Yang L.H., Mo S., Wu Z.Q., Li H.M. Rg1 promotes the proliferation and adipogenic differentiation of human adipose-derived stem cells via FXR1/lnc-GAS5-AS1 pathway. Curr Stem Cell Res Ther. 2022;17:815–824. doi: 10.2174/1574888X16666211129121414. [DOI] [PubMed] [Google Scholar]
- 44.Xue L., Hu M., Li J., Li Y., Zhu Q., Zhou G., Zhang X., Zhou Y., Zhang J., Ding P. Ginsenoside Rg1 inhibits high glucose-induced proliferation, migration, and angiogenesis in retinal endothelial cells by regulating the lncRNA SNHG7/miR-2116-5p/SIRT3 Axis. J Oncol. 2022;2022 doi: 10.1155/2022/6184631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Li J., Gao W., Zhao Z., Li Y., Yang L., Wei W., Ren F., Li Y., Yu Y., Duan W., et al. Ginsenoside Rg1 reduced microglial activation and mitochondrial dysfunction to alleviate depression-like behaviour via the GAS5/EZH2/SOCS3/NRF2 Axis. Mol Neurobiol. 2022;59:2855–2873. doi: 10.1007/s12035-022-02740-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Miao Y., Chen X., Qin M., Zhou W., Wang Y., Ji Y. lncRNA GAS5, as a ceRNA, inhibits the proliferation of diffuse large B-cell lymphoma cells by regulating the miR-18a-5p/RUNX1 axis. Int J Oncol. 2021;59:94. doi: 10.3892/ijo.2021.5274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Cheng Y., Dai X., Yang T., Zhang N., Liu Z., Jiang Y. Low long noncoding RNA growth arrest-specific transcript 5 expression in the exosomes of lung cancer cells promotes tumor angiogenesis. J Oncol. 2019;2019 doi: 10.1155/2019/2476175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Ham J., Jeong D., Park S., Kim H.W., Kim H., Kim S.J. Ginsenoside Rg3 and Korean Red Ginseng extract epigenetically regulate the tumor-related long noncoding RNAs RFX3-AS1 and STXBP5-AS1. J Ginseng Res. 2019;43:625–634. doi: 10.1016/j.jgr.2019.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Zheng X., Zhou Y., Chen W., Chen L., Lu J., He F., Li X., Zhao L. Ginsenoside 20(S)-Rg3 prevents PKM2-targeting miR-324-5p from H19 sponging to antagonize the Warburg effect in ovarian cancer cells. Cell Physiol Biochem. 2018;51:1340–1353. doi: 10.1159/000495552. [DOI] [PubMed] [Google Scholar]
- 50.Zhao L., Sun W., Zheng A., Zhang Y., Fang C., Zhang P. Ginsenoside Rg3 suppresses ovarian cancer cell proliferation and invasion by inhibiting the expression of lncRNA H19. Acta Biochim Pol. 2021;68:575–582. doi: 10.18388/abp.2020_5343. [DOI] [PubMed] [Google Scholar]
- 51.Zou J., Su H., Zou C., Liang X., Fei Z. Ginsenoside Rg3 suppresses the growth of gemcitabine-resistant pancreatic cancer cells by upregulating lncRNA-CASC2 and activating PTEN signaling. J Biochem Mol Toxicol. 2020;34 doi: 10.1002/jbt.22480. [DOI] [PubMed] [Google Scholar]
- 52.Chen Z., Li S., Shen L., Wei X., Zhu H., Wang X., Yang M., Zheng X. NF-kappa B interacting long noncoding RNA enhances the Warburg effect and angiogenesis and is associated with decreased survival of patients with gliomas. Cell Death Dis. 2020;11:323. doi: 10.1038/s41419-020-2520-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Pu Z., Ge F., Wang Y., Jiang Z., Zhu S., Qin S., Dai Q., Liu H., Hua H. Ginsenoside-Rg3 inhibits the proliferation and invasion of hepatoma carcinoma cells via regulating long non-coding RNA HOX antisense intergenic. Bioengineered. 2021;12:2398–2409. doi: 10.1080/21655979.2021.1932211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zhang W., Wang Q., Du H., Jiang S. CRISPR/Cas9-mediated overexpression of long non-coding RNA SRY-box transcription factor 21 antisense divergent transcript 1 regulates the proliferation of osteosarcoma by increasing the expression of mechanistic target of rapamycin kinase and Kruppel-like factor 4. Bioengineered. 2022;13:6678–6687. doi: 10.1080/21655979.2021.1995106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Wu P., Yu X., Peng Y., Wang Q.L., Deng L.T., Xing W. Ginsenoside Rg3 alleviates septic liver injury by regulating the lncRNA TUG1/miR-200c-3p/SIRT1 axis. J Inflamm. 2021;18:31. doi: 10.1186/s12950-021-00296-2. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 56.Yang Y., He B., Yang R., Chen D., Zhang X., Li F., Shen Z., Chen P. Comprehensive analysis of lncRNA expression profiles in rats with cerebral ischemia-reperfusion injury after treatment with 20(R)-ginsenoside Rg3. J Integr Neurosci. 2022;21:16. doi: 10.31083/j.jin2101016. [DOI] [PubMed] [Google Scholar]
- 57.Kim H., Ji H.W., Kim H.W., Yun S.H., Park J.E., Kim S.J. Ginsenoside Rg3 prevents oncogenic long noncoding RNA ATXN8OS from inhibiting tumor-suppressive microRNA-424-5p in breast cancer cells. Biomolecules. 2021;11:118. doi: 10.3390/biom11010118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wang Jun, Zhao L., Gao X. Ginsenoside Rg3 induces low expression of lncRNA ATXN8OS to inhibit colon cancer metastasis. Russ J Bioorganic Chem. 2023;49:562–570. [Google Scholar]
- 59.Wu K., Huang J., Li N., Xu T., Cai W., Ye Z. Antitumor effect of ginsenoside Rg3 on gallbladder cancer by inducing endoplasmic reticulum stress-mediated apoptosis in vitro and in vivo. Oncol Lett. 2018;16:5687–5696. doi: 10.3892/ol.2018.9331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Chang L., Wang D., Kan S., Hao M., Liu H., Yang Z., Xia Q., Liu W. Ginsenoside Rd inhibits migration and invasion of tongue cancer cells through H19/miR-675-5p/CDH1 axis. J Appl Oral Sci. 2022;30 doi: 10.1590/1678-7757-2022-0144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Xu Y., Cao L., Zou W., Yu R., Shen W. Panax notoginseng saponins inhibits NLRP3 inflammasome-mediated pyroptosis by downregulating lncRNA-ANRIL in cardiorenal syndrome type 4. Chin Med. 2023;18:50. doi: 10.1186/s13020-023-00756-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kong Y., Hsieh C.H., Alonso L.C. ANRIL: a lncRNA at the CDKN2A/B locus with roles in cancer and metabolic disease. Front Endocrinol. 2018;9:405. doi: 10.3389/fendo.2018.00405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Chen S., Wu Y., Qin X., Wen P., Liu J., Yang M. Global gene expression analysis using RNA-seq reveals the new roles of Panax notoginseng Saponins in ischemic cardiomyocytes. J Ethnopharmacol. 2021;268 doi: 10.1016/j.jep.2020.113639. [DOI] [PubMed] [Google Scholar]
- 64.Yang Y., Xi P., Xie Y., Zhao C., Xu J., Jiang J. Notoginsenoside R1 reduces blood pressure in spontaneously hypertensive rats through a long non-coding RNA AK094457. Int J Clin Exp Pathol. 2015;8:2700–2709. [PMC free article] [PubMed] [Google Scholar]
- 65.Zhao J., Cui L., Sun J., Xie Z., Zhang L., Ding Z., Quan X. Notoginsenoside R1 alleviates oxidized low-density lipoprotein-induced apoptosis, inflammatory response, and oxidative stress in HUVECS through modulation of XIST/miR-221-3p/TRAF6 axis. Cell Signal. 2020;76 doi: 10.1016/j.cellsig.2020.109781. [DOI] [PubMed] [Google Scholar]
- 66.Zhang S., Wang H., Wang J., Jin W., Yan X., Chen X., Wang D., Zhao D., Wang Y., Cong D., et al. Ginsenoside Rf inhibits human tau proteotoxicity and causes specific LncRNA, miRNA and mRNA expression changes in Caenorhabditis elegans model of tauopathy. Eur J Pharmacol. 2022;922 doi: 10.1016/j.ejphar.2022.174887. [DOI] [PubMed] [Google Scholar]
- 67.Chen S., Ye H., Gong F., Mao S., Li C., Xu B., Ren Y., Yu R. Ginsenoside compound K exerts antitumour effects in renal cell carcinoma via regulation of ROS and lncRNA THOR. Oncol Rep. 2021;45:38. doi: 10.3892/or.2021.7989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Tan Y., Sun D., Chen J., Li R., Wang S. Ginsenoside Rb3 alleviates smoke-induced lung injury via the H19/miR-29b-3p/HGMB1/TLR4 signalling pathway. J Cell Mol Med. 2021;25:2725–2729. doi: 10.1111/jcmm.15844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Hu K., Yan T.M., Cao K.Y., Li F., Ma X.R., Lai Q., Liu J.C., Pan Y., Kou J.P., Jiang Z.H. A tRNA-derived fragment of ginseng protects heart against ischemia/reperfusion injury via targeting the lncRNA MIAT/VEGFA pathway. Mol Ther Nucleic Acids. 2022;29:672–688. doi: 10.1016/j.omtn.2022.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Lu S., Huang J., Zhang J., Wu C., Huang Z., Tao X., You L., Stalin A., Chen M., Li J., et al. The anti-hepatocellular carcinoma effect of Aidi injection was related to the synergistic action of cantharidin, formononetin, and isofraxidin through BIRC5, FEN1, and EGFR. J Ethnopharmacol. 2024;319 doi: 10.1016/j.jep.2023.117209. [DOI] [PubMed] [Google Scholar]
- 71.Yang Z., Xu F., Teschendorff A.E., Zhao Y., Yao L., Li J., He Y. Insights into the role of long non-coding RNAs in DNA methylation mediated transcriptional regulation. Front Mol Biosci. 2022;9 doi: 10.3389/fmolb.2022.1067406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Liu J., Zhou W.Y., Luo X.J., Chen Y.X., Wong C.W., Liu Z.X., Bo Zheng J., Yu Mo H., Chen J.Q., Li J.J., et al. Long noncoding RNA Regulating ImMune Escape regulates mixed lineage leukaemia protein-1-H3K4me3-mediated immune escape in oesophageal squamous cell carcinoma. Clin Transl Med. 2023;13 doi: 10.1002/ctm2.1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Endo I., Amatya V.J., Kushitani K., Nakagiri T., Aoe K., Takeshima Y. Long non-coding RNA LINC00152 requires EZH2 to promote mesothelioma cell proliferation, migration, and invasion. Anticancer Res. 2023;43:5367–5376. doi: 10.21873/anticanres.16740. [DOI] [PubMed] [Google Scholar]
- 74.Xu H., Ba Z., Liu C., Yu X. Long noncoding RNA DLEU1 promotes proliferation and glycolysis of gastric cancer cells via APOC1 upregulation by recruiting SMYD2 to induce trimethylation of H3K4 modification. Transl Oncol. 2023;36 doi: 10.1016/j.tranon.2023.101731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Fang J., Li K., Huang C., Xue H., Ni Q. LncRNA TTN-AS1 confers tamoxifen resistance in breast cancer via sponging miR-107 to modulate PI3K/AKT signaling pathway. Am J Transl Res. 2022;14:2267–2279. [PMC free article] [PubMed] [Google Scholar]
- 76.Lu S., Zeng L., Mo G., Lei D., Li Y., Ou G., Wu H., Sun J., Rong C., He S., et al. Long non-coding RNA SNHG17 may function as a competitive endogenous RNA in diffuse large B-cell lymphoma progression by sponging miR-34a-5p. PLoS One. 2023;18 doi: 10.1371/journal.pone.0294729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kim H.W., Baek M., Jung S., Jang S., Lee H., Yang S.H., Kwak B.S., Kim S.J. ELOVL2-AS1 suppresses tamoxifen resistance by sponging miR-1233-3p in breast cancer. Epigenetics. 2023;18 doi: 10.1080/15592294.2023.2276384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Lu J., Zhou Y., Zheng X., Chen L., Tuo X., Chen H., Xue M., Chen Q., Chen W., Li X., et al. 20(S)-Rg3 upregulates FDFT1 via reducing miR-4425 to inhibit ovarian cancer progression. Arch Biochem Biophys. 2020;693 doi: 10.1016/j.abb.2020.108569. [DOI] [PubMed] [Google Scholar]
- 79.Zhang L., Cao Y., Kou X., Che L., Zhou X., Chen G., Zhao J. Long non-coding RNA HCG11 suppresses the growth of glioma by cooperating with the miR-4425/MTA3 axis. J Gene Med. 2019;21 doi: 10.1002/jgm.3074. [DOI] [PubMed] [Google Scholar]
- 80.Monnier P., Martinet C., Pontis J., Stancheva I., Ait-Si-Ali S., Dandolo L. H19 lncRNA controls gene expression of the Imprinted Gene Network by recruiting MBD1. Proc Natl Acad Sci U S A. 2013;110:20693–20698. doi: 10.1073/pnas.1310201110. [DOI] [PMC free article] [PubMed] [Google Scholar]

