You're using an outdated browser. Please upgrade to a modern browser for the best experience.
Submitted Successfully!
Thank you for your contribution! You can also upload a video entry or images related to this topic. For video creation, please contact our Academic Video Service.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2488 2023-03-23 13:02:12 |
2 format change + 3 word(s) 2491 2023-03-24 03:52:30 |

Video Upload Options

We provide professional Academic Video Service to translate complex research into visually appealing presentations. Would you like to try it?

Confirm

Are you sure to Delete?
Yes No
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Farooqi, A.A.; Rakhmetova, V.S.; Kapanova, G.; Tashenova, G.; Tulebayeva, A.; Akhenbekova, A.; Ibekenov, O.; Turgambayeva, A.; Xu, B. Bufalin-Mediated Regulation of Cell Signaling Pathways in Cancers. Encyclopedia. Available online: https://encyclopedia.pub/entry/42478 (accessed on 17 July 2025).
Farooqi AA, Rakhmetova VS, Kapanova G, Tashenova G, Tulebayeva A, Akhenbekova A, et al. Bufalin-Mediated Regulation of Cell Signaling Pathways in Cancers. Encyclopedia. Available at: https://encyclopedia.pub/entry/42478. Accessed July 17, 2025.
Farooqi, Ammad Ahmad, Venera S. Rakhmetova, Gulnara Kapanova, Gulnara Tashenova, Aigul Tulebayeva, Aida Akhenbekova, Onlassyn Ibekenov, Assiya Turgambayeva, Baojun Xu. "Bufalin-Mediated Regulation of Cell Signaling Pathways in Cancers" Encyclopedia, https://encyclopedia.pub/entry/42478 (accessed July 17, 2025).
Farooqi, A.A., Rakhmetova, V.S., Kapanova, G., Tashenova, G., Tulebayeva, A., Akhenbekova, A., Ibekenov, O., Turgambayeva, A., & Xu, B. (2023, March 23). Bufalin-Mediated Regulation of Cell Signaling Pathways in Cancers. In Encyclopedia. https://encyclopedia.pub/entry/42478
Farooqi, Ammad Ahmad, et al. "Bufalin-Mediated Regulation of Cell Signaling Pathways in Cancers." Encyclopedia. Web. 23 March, 2023.
Bufalin-Mediated Regulation of Cell Signaling Pathways in Cancers
Edit

Bufalin is a pharmacologically active molecule isolated from the skin of the toad Bufo gargarizans or Bufo melanostictus. Bufalin has characteristically unique properties to regulate multiple molecular targets and can be used to harness multi-targeted therapeutic regimes against different cancers. There is burgeoning evidence related to functional roles of signaling cascades in carcinogenesis and metastasis. Bufalin has been reported to regulate pleiotropically a myriad of signal transduction cascades in various cancers. Importantly, bufalin mechanistically regulated JAK/STAT, Wnt/β-Catenin, mTOR, TRAIL/TRAIL-R, EGFR, and c-MET pathways. Furthermore, bufalin-mediated modulation of non-coding RNAs in different cancers has also started to gain tremendous momentum. Similarly, bufalin-mediated targeting of tumor microenvironments and tumor macrophages is an area of exciting research and we have only started to scratch the surface of the complicated nature of molecular oncology. Cell culture studies and animal models provide proof-of-concept for the impetus role of bufalin in the inhibition of carcinogenesis and metastasis.

cancer apoptosis cell signaling bufalin metastasis

1. Introduction

Recent advancements in next-generation sequencing and multi-omics analyses have demonstrated how crosstalk of different signaling cascades can result in the formation of a complex web of circuitries within cancer cells that, if fully mapped, can be utilized for more precisely targeted therapies. A wealth of information shows that intracellular signaling is dependent on a multitude of signaling pathways that have evolved for tightly orchestrated and dynamic cellular responses. Furthermore, many signaling cascades interact with each other and form multi-dimensional networks that regulate the integration of numerous inputs to generate sophisticated cellular responses. Seminal research works have uncovered key discoveries in fundamental biology and different types of cellular signaling pathways. Deregulation of transduction cascades not only promoted cancer progression but also fueled the spread of therapeutically resistant and metastatically competent cancer cells to distant organs for the development of secondary tumors [1][2][3][4][5][6].
Natural product research in preclinical studies has generated valuable literature related to inhibition of carcinogenesis and metastasis [7][8][9]. Small molecules are pharmacological tools of considerable value for mechanistic dissection of highly intricate biological processes and identification of possible therapeutic interventions. Chemoproteomic workflows have enabled additional multiplexing in research methodologies, which will be valuable for assessing target identification and compound selectivity. The metamorphosis of preclinical research has widened the avenues of effective clinical research. Mechanistic insights gleaned over decades of ground-breaking discoveries have sparked unprecedented research interests in pharmacological evaluation of natural products in the amelioration and remedy of different diseases [10][11].
Bufalin is a pharmacologically active molecule isolated from the skin of the toad Bufo gargarizans or Bufo melanostictus. A substantial volume of conceptual knowledge has been added to the rapidly evolving field of medicinal research associated with the pharmaceutical significance of bufalin. Cancer chemopreventive effects of bufalin have been reviewed previously in various useful and informative review articles [12][13][14][15][16].

2. Regulation of JAK/STAT Pathway by Bufalin

A look through a scientific lens indicates that in a burst of research activity, principally published between 1991 and 1994, the cast of JAK-STAT family members and the trajectories of the pathway were mapped to a greater extent. Many functional and structural protein studies and physiological studies on the proteins of the pathway have been reported. The Janus kinase (JAK)/signal transducers and activators of transcription (STATs) transduction pathway is an intracellular signaling cascade required for response to many extracellular ligands. Essentially, phosphorylation induces JAK activation and consequently these kinases phosphorylate intracellular components of the receptors, which allows the recruitment of STAT proteins [17][18][19][20][21]. Genome-wide analyses have yielded a number of discoveries about the biology of STAT proteins. In this section, the most recent evidence has been gathered to summarize multi-step regulation of JAK/STAT pathways by bufalin in different cancers.
Cancer-associated fibroblasts (CAFs) have a critical role in tumor microenvironment. CAF-conditioned media-treated colorectal cancer cells expressed high levels of p-STAT3 and matrix metalloproteinase-2, whereas low levels of E-cadherin were found in hyperactive STAT3-expressing cancer cells. Bufalin blocked CAF-induced invasion and metastasis of colorectal cancer cells by inactivation of the STAT3 pathway. Intraperitoneal injections of bufalin efficiently suppressed hepatic metastatic nodules in mice injected with HCT116 and CAF cells in the spleen (shown in Figure 1) [22].
Figure 1. (A) JAK/STAT signaling triggered the upregulation of Bcl2, Mcl-1, survivin, and VEGF. JAK2 phosphorylated STAT3 and promoted nuclear accumulation of STAT3 proteins. (B,C) Bufalin inhibited the activation of JAK2 and STAT3. (D) Acetyl-bufalin inhibited tumor formation in mice. Bufalin also inhibited angiogenesis and liver metastasis.

3. Regulation of AKT/mTOR Pathway by Bufalin

mTOR protein kinase occupies a central role in the nexus of many signaling cascades and plays essential roles in the regulation of different mechanisms. Protein synthesis is a resource-intensive and energy-intensive process in the rapidly growing cells. It is thus tightly controlled by mTORC1, which promotes protein synthesis by phosphorylation of 4E-BPs (eukaryotic initiation factor 4E-binding proteins) and p70S6K1 (S6 kinase 1). In its unphosphorylated state, 4E-BP1 suppressed translation by binding and sequestration of eIF4E (eukaryotic translation initiation factor 4E), an essential constituent of the eIF4F cap-binding complex [23]. mTORC1 regulated cap-dependent translation of mRNAs by direct phosphorylation of the inhibitors of eIF4E: namely, 4E-BP1 and 4E-BP2. TSC2 formed a heterodimeric complex with TSC1 and inhibited mTORC1. However, phosphorylation of TSC2 at the 1462nd threonine by AKT inhibited its GAP activity for RHEB, which therefore remained in a GTP-bound active state and activated mTORC1. AMPK inhibited mTORC1 by phosphorylation of RAPTOR at serine-792 and TSC2 at serine-1387 which promoted the inhibitory functions of TSC1-TSC2 complexes [24][25]. Here, the researchers offer an overview of current advancements in the field regarding the regulation of the AKT/mTOR pathway by bufalin.
Importantly, bufalin significantly reduced the phosphorylation levels of mTOR and S6K. Furthermore, HIF-1α levels were significantly reduced by bufalin. HIF-1α overexpression attenuated the inhibitory effects of bufalin on ovarian cancer cells. Intraperitoneal injections of bufalin proficiently induced regression of tumor xenografts in rodent models inoculated with PA-1 cells [26].
Cbl-b efficiently promoted autophagic pathway activity induced by bufalin through the inactivation of mTOR and activation of ERK1/2. mTOR has been reported to negatively regulate autophagy. Therefore, once activated by AKT/PKB, mTOR inhibited autophagy by enhancing the phosphorylation of p70S6K. Bufalin effectively reduced p-AKT, p-mTOR, and p-p70S6K (Figure 2) [27]. Together, these details indicate that inactivation of AKT/mTOR/p70S6K cascades and functionalization of the ERK pathway are involved in the activation of the autophagic pathway in bufalin-treated MGC803 cancer cells.
Figure 2. Bufalin mediated inactivation of AKT/mTOR pathway. Bufalin effectively reduced p-AKT, p-mTOR and p-p70S6K. Bufalin inhibited tumor growth by inactivation of AKT/mTOR pathway.

4. Regulation of Wnt/β-Catenin by Bufalin

In the absence of Wnt signals, degradation of β-catenin is mediated by a destruction complex consisting of adenomatous polyposis coli (APC), Axin and glycogen synthase kinase-3 (GSK-3β) proteins. Following the binding of Wnt to receptors of Frizzled and LRP families on the cell surface, β-catenin efficiently moved into the nucleus and transcriptionally regulated a myriad of gene networks. Phosphorylation of GSK-3β at serine-9 resulted in the inactivation of GSK-3β. Therefore, GSK-3β inactivation led to activation and transportation of β-catenin to the nucleus [28][29][30][31]. In this section, the researchers highlight the recent breakthroughs that have been made in the field of molecular oncology and discuss how regulation of the Wnt/β-catenin pathway by bufalin will influence ongoing basic research and the design of rationale-based clinical trials to improve the treatment options for cancer patients.
Cell cycle-related kinase (CCRK) acted as an oncogenic master modulator for the activation and nuclear translocation of β-catenin, where it formed a complex with transcriptional factor TCF. Notably, the complex binds to promoter regions of EGFR (epidermal growth factor receptor) and CCND1 (cyclin D1). Bufalin efficiently reduced the levels of CCND1, EGFR, and CCRK. It was shown that CCRK overexpression promoted tumorigenesis by activation of β-catenin/TCF signaling. Subcutaneous inoculation of PLC5 cells into the right flanks of athymic nude mice was used for the construction of the xenograft rodent model. Tumor pieces were implanted into the liver lobes of nude mice for the development of orthotopic models. Bufalin not only reduced CCRK but also decreased nuclear levels of β-catenin in the tumor tissues [32].
Bufalin effectively blocked androgen receptor-mediated transcriptional upregulation of CCRK (cell cycle-related kinase) in HepG2.2.15 and PLC5 cells. Levels of phosphorylated androgen receptor were found to be reduced by bufalin. GSK-3β phosphorylation by CCRK caused activation of β-catenin. Bufalin inhibited HBx-mediated intrahepatic tumorigenicity, and reduced the levels of p-ARSer81, CCRK, p-GSK3βSer9, and active β-catenin in tumor tissues [33].
Bufalin markedly inhibited the migratory and invasive capacities of hepatocellular carcinoma cells, and efficiently caused reduction in the levels of p-GSK3βSer9 and active β-catenin in BEL-7402 cells [34].
Deregulated expression of β-catenin resulted in the instability of the complexes formed with E-cadherin. Dissociation of β-catenin and E-cadherin resulted in a loss of epithelial characteristics and potently promoted increasingly invasive phenotypes. Bufalin interfered with nuclear transportation of β-catenin in colorectal cancer cells [35].
The recent advancements in the characterization of aberrantly activated β-catenin target gene programming in cancer cells also provide exceptional prospects for pharmacological targeting of the oncogenic Wnt/β-catenin pathway. As illustrated by the examples given in this section, comprehensive experimental evaluation of the functional effects of bufalin on the Wnt/β-catenin pathway will be advantageous.

5. Regulation of TRAIL Pathway by Bufalin

Excitingly, a maze of information in the rapidly growing field of apoptosis research has unveiled dichotomously branched pathways, consisting of extrinsic and intrinsic apoptotic pathways. Binding of TRAIL to TRAIL-R1 or TRAIL-R2 results in oligomerization of receptors on the cell membrane and initiation of apoptotic cell death. Following ligand–receptor interactions, FAS associated protein with death domain (FADD) is recruited to death domain motifs within the carboxyl terminus of death receptors. Studies have shown that death inducible signaling complex (DISC) is formed at death receptors by assembly of multi-molecular machinery consisting of FADD and pro-caspase-8, and promotes the functionalization of caspase-8 [36][37][38][39][40][41]. During intrinsic apoptosis, loss of subcellular and submitochondrial compartmentalization triggered the exit of cytochrome c, SMAC/DIABLO, and OMI/HTRA. In this section, the researchers review collected key aspects associated with bufalin-mediated regulation of TRAIL-mediated apoptotic cell death.
Intriguingly, aggregations of lipid rafts as well as redistribution of death receptors (DR4, DR5) in lipid rafts were identified in bufalin-treated MCF-7 and MDA-MB-231 cancer cells. The findings revealed that lipid raft dysfunction caused resistance against TRAIL, whereas bufalin-mediated redistribution of DR4 and DR5 within lipid rafts significantly contributed to TRAIL-mediated apoptotic death in breast cancer cells. Depletion of cholesterol by methyl-β-cyclodextrin has been a widely used approach. Clustering of DR4 and DR5 was reduced markedly in cancer cells pre-treated with methyl-β-cyclodextrin [42].
Studies have yielded convincing evidence that Cbl-b negatively regulated the TRAIL-driven pathway. Cbl-b was downregulated by bufalin in MDA-MB-231 and MCF-7 cancer cells. Essentially, bufalin upregulated the levels of DR4 and DR5 by suppression in the levels of Cbl-b. Bufalin and TRAIL-mediated activation of ERK, JNK, and p38 MAPK was found to be significantly enhanced in Cbl-b-silenced cancer cells [43].
Bufalin increased the levels of Bax, cytochrome c, Endonuclease G and AIF (apoptosis-inducing factor). Concomitantly, bufalin reduced Bcl-2 in NPC-TW 076 cells. Additionally, bufalin stimulated the expression levels of TRAIL, DR4, DR5, and FADD [44].
The TRAIL pathway contains another protein that blocks caspase activation. Importantly, c-FLIP (cellular FLICE inhibitory protein) is an inactive homologue of caspase-8 that contains a DED but lacks a catalytically active site. Bufalin upregulated the expression of DR5 in T24 cancer cells. Moreover, TRAIL and bufalin efficiently reduced the levels of c-FLIP and XIAP in T24 cancer cells (Figure 3) [45].
Figure 3. Diagrammatic representation of regulation of TRAIL-mediated apoptotic death by bufalin. Importantly, bufalin enhanced the levels of FADD and DR4/DR5. Bufalin activated death inducible signaling complex. Bufalin triggered the release of cytochrome c and SMAC/DIABLO. C-FLIP prevented the formation of DISC but bufalin prominently reduced the levels of c-FLIP. Bufalin also inhibited Bcl-2 and XIAP.

6. Regulation of Non-Coding RNAs by Bufalin

More prominently, extraordinary strides have been made in the achievement of a high-resolution view of mechanistic regulation of cell signaling pathways by non-coding RNAs in different cancers. The widespread alteration of non-coding RNAs demonstrated that deregulation of miRNAs [46][47][48], lncRNAs [49][50][51][52], and circular RNAs [53][54][55] contributed to multiple hallmarks of cancer.

7. Tumor Inhibitory Role of Bufalin: Animal Model Studies

Osimertinib is a third-generation standard-of-care therapy for EGFR mutation-positive advanced non–small-cell lung cancers. Osimertinib caused considerable reduction in the levels of Mcl-1 in HCC827 and PC-9 cells. USP9X and Ku70 have been functionally characterized as Mcl-1 deubiquitinases. Studies had shown that deubiquitinases removed polyubiquitin chains from Mcl-1 and enhanced its stability (Figure 4). Levels of Mcl-1 levels were found to be robustly increased in Ku70-overexpressing PC-9 cells. However, silencing of Ku70 led to a notable reduction in the levels of Mcl-1 and enhanced osimertinib-sensitivity in PC-9/OR cells. Moreover, combinatorial treatment with osimertinib and bufalin significantly downregulated the levels of Ku70 and Mcl-1 in tumor tissues of NSCLC xenograft mouse models [56].
Figure 4. (A) Bufalin induced dissociation of KU70 and Mcl-1 and promoted degradation of Mcl-1. (B) Bufalin enhanced the interactions of ZFP91 and E2F2. Bufalin mediated an increase in the polyubiquitination levels of E2F2. (C) Bufalin inhibited calcineurin mediated dephosphorylation and nuclear accumulation of NFAT. NFAT stimulated the expression of c-Myc. (D,E) Bufalin enhanced the expression of NKG2D in natural killer cells. Bufalin reduced the levels of ADAM9 and inhibited the shedding of MICA.

8. Regulation of Tumor Microenvironment by Bufalin

Macrophages have functional plasticity and can be polarized into two characteristically distinct phenotypes for modulation of the tumor microenvironment.
Bufalin induced the activation the NF-κB pathway and triggered upregulation of IFNγ and TNFα. However, constitutive overexpression of p50 in bone-marrow-derived macrophage (BMDMs) markedly counteracted the effects of bufalin and concomitantly reduced the expression of M1-associated genes. Importantly, the proportions of CD163+CD206+ M2 macrophages were found to be sharply increased and caused reversal of the pre-dominant effects of bufalin-primed M1 macrophages. In HCC-bearing animal models, overexpression of p50 led to remarkable impairment in the tumor-inhibitory effects of bufalin. Furthermore, the percentage of tumor-promoting M2 macrophages was found to be enhanced in mice bearing p50-overexpressing tumors. The accumulation of bufalin-induced CD4+ and CD8+ T cells in the tissue microenvironment was also reduced in mice inoculated with p50-overexpressing cancer cells [57].
Bufalin efficiently inhibited chemo-resistant cell-mediated polarization of M2 macrophages. Macrophage migration inhibitory factor (MIF) inhibited the polarization of macrophages. Bufalin blocked SRC-3-mediated transcriptional upregulation of MIF and abrogated macrophage polarization [58].
First discovered in the late 1990s, activating receptor NKG2D participates in the immunosurveillance of cytotoxic lymphocytes primarily through identification of stress-induced ligands MICA/B on the surfaces of cancer cells. Bufalin upregulated membrane bound-MICA (m-MICA) in liver cancer cells and reduced the levels of soluble s-MICA. Bufalin enhanced the expression of NKG2D in NK-92MI cells. Moreover, expression levels of inhibitory receptors (NKG2A, TIGIT and CTLA-4) were also found to be suppressed in NK-92MI cells. Bufalin reduced the levels of ADAM9 and inhibited the shedding of MICA (Figure 4) [59].

References

  1. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674.
  2. Puisieux, A.; Brabletz, T.; Caramel, J. Oncogenic roles of EMT-inducing transcription factors. Nat. Cell Biol. 2014, 16, 488–494.
  3. Dongre, A.; Weinberg, R.A. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat. Rev. Mol. Cell Biol. 2019, 20, 69–84.
  4. Rosell, R. Mediating Resistance in Oncogene-Driven Cancers. N. Engl. J. Med. 2013, 368, 1551–1552.
  5. Hidalgo, M.; Maitra, A. The Hedgehog Pathway and Pancreatic Cancer. New Engl. J. Med. 2009, 361, 2094–2096.
  6. Valastyan, S.; Weinberg, R.A. Tumor Metastasis: Molecular Insights and Evolving Paradigms. Cell 2011, 147, 275–292.
  7. Clardy, J.; Walsh, C. Lessons from natural molecules. Nature 2004, 432, 829–837.
  8. Marcus, D.M.; Grollman, A.P. Botanical Medicines—The Need for New Regulations. New Engl. J. Med. 2002, 347, 2073–2076.
  9. Mann, J. Natural products in cancer chemotherapy: Past, present and future. Nat. Rev. Cancer 2002, 2, 143–148.
  10. Rodrigues, T.; Reker, D.; Schneider, P.; Schneider, G. Counting on natural products for drug design. Nat. Chem. 2016, 8, 531–541.
  11. Koehn, F.E.; Carter, G.T. The evolving role of natural products in drug discovery. Nat. Rev. Drug Discov. 2005, 4, 206–220.
  12. Soumoy, L.; Ghanem, G.E.; Saussez, S.; Journe, F. Bufalin for an innovative therapeutic approach against cancer. Pharmacol. Res. 2022, 184, 106442.
  13. Jiang, H.-Y.; Zheng, H.-M.; Xia, C.; Li, X.; Wang, G.; Zhao, T.; Cui, X.-N.; Wang, R.-Y.; Liu, Y. The Research Progress of Bufalin in the Treatment of Hepatocellular Carcinoma. OncoTargets Ther. 2022, ume 15, 291–298.
  14. Cheng, C.-S.; Wang, J.; Chen, J.; Kuo, K.T.; Tang, J.; Gao, H.; Chen, L.; Chen, Z.; Meng, Z. New therapeutic aspects of steroidal cardiac glycosides: The anticancer properties of Huachansu and its main active constituent Bufalin. Cancer Cell Int. 2019, 19, 92.
  15. Lan, Y.; Lou, J.; Jiang, X.; Wang, X.; Xing, J.; Li, S.; Zhang, B. A research update on the anticancer effects of bufalin and its derivatives (Review). Oncol. Lett. 2019, 17, 3635–3640.
  16. Wang, J.; Xia, Y.; Zuo, Q.; Chen, T. Molecular mechanisms underlying the�antimetastatic activity of bufalin (Review). Mol. Clin. Oncol. 2018, 8, 631–636.
  17. Nelson, E.A.; Walker, S.R.; Weisberg, E.; Bar-Natan, M.; Barrett, R.; Gashin, L.B.; Terrell, S.; Klitgaard, J.L.; Santo, L.; Addorio, M.R.; et al. The STAT5 inhibitor pimozide decreases survival of chronic myelogenous leukemia cells resistant to kinase inhibitors. Blood 2011, 117, 3421–3429.
  18. Turkson, J.; Ryan, D.; Kim, J.S.; Zhang, Y.; Chen, Z.; Haura, E.; Laudano, A.; Sebti, S.; Hamilton, A.D.; Jove, R. Phosphotyrosyl Peptides Block Stat3-mediated DNA Binding Activity, Gene Regulation, and Cell Transformation. J. Biol. Chem. 2001, 276, 45443–45455.
  19. Siddiquee, K.; Zhang, S.; Guida, W.C.; Blaskovich, M.A.; Greedy, B.; Lawrence, H.R.; Yip, M.L.R.; Jove, R.; McLaughlin, M.M.; Lawrence, N.J.; et al. Selective chemical probe inhibitor of Stat3, identified through structure-based virtual screening, induces antitumor activity. Proc. Natl. Acad. Sci. USA 2007, 104, 7391–7396.
  20. Brooks, A.J.; Dai, W.; O’Mara, M.L.; Abankwa, D.; Chhabra, Y.; Pelekanos, R.A.; Gardon, O.; Tunny, K.A.; Blucher, K.M.; Morton, C.J.; et al. Mechanism of Activation of Protein Kinase JAK2 by the Growth Hormone Receptor. Science 2014, 344, 1249783.
  21. Wilmes, S.; Hafer, M.; Vuorio, J.; Tucker, J.A.; Winkelmann, H.; Löchte, S.; Stanly, T.A.; Prieto, K.D.P.; Poojari, C.; Sharma, V.; et al. Mechanism of homodimeric cytokine receptor activation and dysregulation by oncogenic mutations. Science 2021, 367, 643–652.
  22. Glassman, C.R.; Tsutsumi, N.; Saxton, R.A.; Lupardus, P.J.; Jude, K.M.; Garcia, K.C. Structure of a Janus kinase cytokine receptor complex reveals the basis for dimeric activation. Science 2022, 376, 163–169.
  23. Kim, D.-H.; Sarbassov, D.D.; Ali, S.M.; King, J.E.; Latek, R.R.; Erdjument-Bromage, H.; Tempst, P.; Sabatini, D.M. mTOR Interacts with Raptor to Form a Nutrient-Sensitive Complex that Signals to the Cell Growth Machinery. Cell 2002, 110, 163–175.
  24. Nojima, H.; Tokunaga, C.; Eguchi, S.; Oshiro, N.; Hidayat, S.; Yoshino, K.-I.; Hara, K.; Tanaka, N.; Avruch, J.; Yonezawa, K. The Mammalian Target of Rapamycin (mTOR) Partner, Raptor, Binds the mTOR Substrates p70 S6 Kinase and 4E-BP1 through Their TOR Signaling (TOS) Motif. J. Biol. Chem. 2003, 278, 15461–15464.
  25. Hara, K.; Yonezawa, K.; Kozlowski, M.T.; Sugimoto, T.; Andrabi, K.; Weng, Q.-P.; Kasuga, M.; Nishimoto, I.; Avruch, J. Regulation of eIF-4E BP1 Phosphorylation by mTOR. J. Biol. Chem. 1997, 272, 26457–26463.
  26. Su, S.; Dou, H.; Wang, Z.; Zhang, Q. Bufalin inhibits ovarian carcinoma via targeting mTOR/HIF-α pathway. Basic Clin. Pharmacol. Toxicol. 2021, 128, 224–233.
  27. Qi, H.-Y.; Qu, X.-J.; Liu, J.; Hou, K.-Z.; Fan, Y.-B.; Che, X.-F.; Liu, Y.-P. Bufalin induces protective autophagy by Cbl-b regulating mTOR and ERK signaling pathways in gastric cancer cells. Cell Biol. Int. 2019, 43, 33–43.
  28. Korinek, V.; Barker, N.; Morin, P.J.; van Wichen, D.; de Weger, R.; Kinzler, K.W.; Vogelstein, B.; Clevers, H. Constitutive transcriptional activation by a beta-catenin-Tcf complex in APC-/- colon carcinoma. Science 1997, 275, 1784–1787.
  29. Morin, P.J.; Sparks, A.B.; Korinek, V.; Barker, N.; Clevers, H.; Vogelstein, B.; Kinzler, K.W. Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science 1997, 275, 1787–1790.
  30. Suzuki, H.; Watkins, D.N.; Jair, K.-W.; Schuebel, K.E.; Markowitz, S.D.; Chen, W.D.; Pretlow, T.P.; Yang, B.; Akiyama, Y.; van Engeland, M.; et al. Epigenetic inactivation of SFRP genes allows constitutive WNT signaling in colorectal cancer. Nat. Genet. 2004, 36, 417–422.
  31. Behrens, J.; Jerchow, B.-A.; Würtele, M.; Grimm, J.; Asbrand, C.; Wirtz, R.; Kühl, M.; Wedlich, D.; Birchmeier, W. Functional Interaction of an Axin Homolog, Conductin, with β-Catenin, APC, and GSK3β. Science 1998, 280, 596–599.
  32. Yu, Z.; Feng, H.; Zhuo, Y.; Li, M.; Zhu, X.; Huang, L.; Zhang, X.; Zhou, Z.; Zheng, C.; Jiang, Y.; et al. Bufalin inhibits hepatitis B virus-associated hepatocellular carcinoma development through androgen receptor dephosphorylation and cell cycle-related kinase degradation. Cell. Oncol. 2020, 43, 1129–1145.
  33. Yu, Z.; Feng, H.; Sun, X.; Zhuo, Y.; Li, M.; Zhou, Z.; Huang, L.; Jiang, Y.; Zhu, X.; Zhang, X.; et al. Bufalin suppresses hepatocarcinogenesis by targeting β-catenin/TCF signaling via cell cycle-related kinase. Sci. Rep. 2018, 8, 3891.
  34. Gai, J.Q.; Sheng, X.; Qin, J.M.; Sun, K.; Zhao, W.; NI, L. The effect and mechanism of bufalin on regulating hepatocellular carcinoma cell invasion and metastasis via Wnt/β-catenin signaling pathway. Int. J. Oncol. 2016, 48, 338–348.
  35. Ding, L.; Yang, Y.; Lu, Q.; Qu, D.; Chandrakesan, P.; Feng, H.; Chen, H.; Chen, X.; Liao, Z.; Du, J.; et al. Bufalin Inhibits Tumorigenesis, Stemness, and Epithelial–Mesenchymal Transition in Colorectal Cancer through a C-Kit/Slug Signaling Axis. Int. J. Mol. Sci. 2022, 23, 13354.
  36. Pitti, R.M.; Marsters, S.A.; Ruppert, S.; Donahue, C.J.; Moore, A.; Ashkenazi, A. Induction of Apoptosis by Apo-2 Ligand, a New Member of the Tumor Necrosis Factor Cytokine Family. J. Biol. Chem. 1996, 271, 12687–12690.
  37. Walczak, H.; Miller, R.E.; Ariail, K.; Gliniak, B.; Griffith, T.S.; Kubin, M.; Chin, W.; Jones, J.; Woodward, A.; Le, T.; et al. Tumoricidal activity of tumor necrosis factor–related apoptosis–inducing ligand in vivo. Nat. Med. 1999, 5, 157–163.
  38. Pan, G.; O’Rourke, K.; Chinnaiyan, A.M.; Gentz, R.; Ebner, R.; Ni, J.; Dixit, V.M. The Receptor for the Cytotoxic Ligand TRAIL. Science 1997, 276, 111–113.
  39. Zhang, X.D.; Franco, A.; Myers, K.; Gray, C.; Nguyen, T.; Hersey, P. Relation of TNF-related apoptosis-inducing ligand (TRAIL) receptor and FLICE-inhibitory protein expression to TRAIL-induced apoptosis of melanoma. Cancer Res 1999, 59, 2747–2753.
  40. Gliniak, B.; Le, T. Tumor necrosis factor-related apoptosis-inducing ligand’s antitumor activity in vivo is enhanced by the chemotherapeutic agent CPT-11. Cancer Res. 1999, 59, 6153–6158.
  41. Roth, W.; Isenmann, S.; Naumann, U.; Kügler, S.; Bähr, M.; Dichgans, J.; Ashkenazi, A.; Weller, M. Locoregional Apo2L/TRAIL erad-icates intracranial human malignant glioma xenografts in athymic mice in the absence of neurotoxicity. Biochem. Biophys. Res. Commun. 1999, 265, 479–483.
  42. Yan, S.; Qu, X.; Xu, L.; Che, X.; Ma, Y.; Zhang, L.; Teng, Y.; Zou, H.; Liu, Y. Bufalin enhances TRAIL-induced apoptosis by redistributing death receptors in lipid rafts in breast cancer cells. Anti-Cancer Drugs 2014, 25, 683–689.
  43. Yan, S.; Qu, X.; Xu, C.; Zhu, Z.; Zhang, L.; Xu, L.; Song, N.; Teng, Y.; Liu, Y. Down-regulation of Cbl-b by bufalin results in up-regulation of DR4/DR5 and sensitization of TRAIL-induced apoptosis in breast cancer cells. J. Cancer Res. Clin. Oncol. 2012, 138, 1279–1289.
  44. Su, E.-Y.; Chu, Y.-L.; Chueh, F.-S.; Ma, Y.-S.; Peng, S.-F.; Huang, W.-W.; Liao, C.-L.; Huang, A.-C.; Chung, J.-G. Bufalin Induces Apoptotic Cell Death in Human Nasopharyngeal Carcinoma Cells through Mitochondrial ROS and TRAIL Pathways. Am. J. Chin. Med. 2019, 47, 237–257.
  45. Kang, K.-H.; Han, M.H.; Jeong, J.-W.; Park, C.; Lee, S.-H.; Lee, H.W.; Hong, S.H.; Choi, Y.H.; Hong, S.H. Bufalin sensitizes human bladder carcinoma cells to TRAIL-mediated apoptosis. Oncol. Lett. 2017, 14, 853–859.
  46. Volinia, S.; Calin, G.A.; Liu, C.G.; Ambs, S.; Cimmino, A.; Petrocca, F.; Visone, R.; Iorio, M.; Roldo, C.; Ferracin, M.; et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proc. Natl. Acad. Sci. USA 2006, 103, 2257–2261.
  47. Lytle, J.R.; Yario, T.A.; Steitz, J.A. Target mRNAs are repressed as efficiently by microRNA-binding sites in the 5’ UTR as in the 3’ UTR. Proc. Natl. Acad. Sci. USA 2007, 104, 9667–9672.
  48. Khraiwesh, B.; Arif, M.A.; Seumel, G.I.; Ossowski, S.; Weigel, D.; Reski, R.; Frank, W. Transcriptional Control of Gene Expression by MicroRNAs. Cell 2010, 140, 111–122.
  49. Cabili, M.N.; Dunagin, M.C.; McClanahan, P.D.; Biaesch, A.; Padovan-Merhar, O.; Regev, A.; Rinn, J.L.; Raj, A. Localization and abundance analysis of human lncRNAs at single-cell and single-molecule resolution. Genome Biol. 2015, 16, 20.
  50. Iyer, M.K.; Niknafs, Y.S.; Malik, R.; Singhal, U.; Sahu, A.; Hosono, Y.; Barrette, T.R.; Prensner, J.R.; Evans, J.R.; Zhao, S.; et al. The landscape of long noncoding RNAs in the human transcriptome. Nat Genet. 2015, 47, 199–208.
  51. Guttman, M.; Amit, I.; Garber, M.; French, C.; Lin, M.F.; Feldser, D.; Huarte, M.; Zuk, O.; Carey, B.W.; Cassady, J.P.; et al. Chromatin signature reveals over a thousand highly conserved large non-coding RNAs in mammals. Nature 2009, 458, 223–227.
  52. Chiu, H.-S.; Somvanshi, S.; Patel, E.; Chen, T.-W.; Singh, V.P.; Zorman, B.; Patil, S.L.; Pan, Y.; Chatterjee, S.S.; Sood, A.K.; et al. Pan-Cancer Analysis of lncRNA Regulation Supports Their Targeting of Cancer Genes in Each Tumor Context. Cell Rep. 2018, 23, 297–312.
  53. Memczak, S.; Jens, M.; Elefsinioti, A.; Torti, F.; Krueger, J.; Rybak, A.; Maier, L.; Mackowiak, S.D.; Gregersen, L.H.; Munschauer, M.; et al. Circular RNAs are a large class of animal RNAs with regulatory potency. Nature 2013, 495, 333–338.
  54. Hansen, T.B.; Jensen, T.I.; Clausen, B.H.; Bramsen, J.B.; Finsen, B.; Damgaard, C.K.; Kjems, J. Natural RNA circles function as efficient microRNA sponges. Nature 2013, 495, 384–388.
  55. Salzman, J.; Gawad, C.; Wang, P.L.; Lacayo, N.; Brown, P.O. Circular RNAs Are the Predominant Transcript Isoform from Hundreds of Human Genes in Diverse Cell Types. PLoS ONE 2012, 7, e30733.
  56. Cao, F.; Gong, Y.-B.; Kang, X.-H.; Lu, Z.-H.; Wang, Y.; Zhao, K.-L.; Miao, Z.-H.; Liao, M.-J.; Xu, Z.-Y. Degradation of MCL-1 by bufalin reverses acquired resistance to osimertinib in EGFR-mutant lung cancer. Toxicol. Appl. Pharmacol. 2019, 379, 114662.
  57. Yu, Z.; Li, Y.; Li, Y.; Zhang, J.; Li, M.; Ji, L.; Tang, Y.; Zheng, Y.; Sheng, J.; Han, Q.; et al. Bufalin stimulates antitumor immune response by driving tumor-infiltrating macrophage toward M1 phenotype in hepatocellular carcinoma. J. Immunother. Cancer 2022, 10, e004297.
  58. Chen, J.; Wang, H.; Jia, L.; He, J.; Li, Y.; Liu, H.; Wu, R.; Qiu, Y.; Zhan, Y.; Yuan, Z.; et al. Bufalin targets the SRC-3/MIF pathway in chemoresistant cells to regulate M2 macrophage polarization in colorectal cancer. Cancer Lett. 2021, 513, 63–74.
  59. Fu, R.; Yu, F.; Wu, W.; Liu, J.; Li, J.; Guo, F.; Xu, L.; Wang, F.; Cui, X. Bufalin enhances the killing efficacy of NK cells against hepatocellular carcinoma by inhibiting MICA shedding. Int. Immunopharmacol. 2021, 101, 108195.
More
Upload a video for this entry
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , , ,
View Times: 671
Revisions: 2 times (View History)
Update Date: 24 Mar 2023
1000/1000
Hot Most Recent
Academic Video Service