Research Progress with Atractylone as an Antitumor Agent
Abstract
:1. Introduction
2. Atractylone and Its Antitumor Activity
2.1. Anti-Liver-Cancer Activity
2.2. Anti-Colon-Cancer Activity
2.3. Anti-Glioblastoma Activity
2.4. Anti-Gastric-Cancer Activity
2.5. Activity Against Other Cancer Cells
3. The Chemical Synthesis of Atractylone
4. Discussion
Pathways and Targets | Mechanism of Action (MOA) |
---|---|
Caspase-mediated apoptosis pathway | Abnormal reduction in apoptosis is a significant factor contributing to tumorigenesis. The imbalance between proapoptotic and antiapoptotic proteins plays a crucial role. Atractylone has been found to downregulate Bcl-2 and upregulate Bax and Cleaved caspase-3, ultimately leading to intrinsic cellular apoptosis. Caspases are essential in initiating and executing apoptosis. Their reduced activity can result in decreased apoptosis and abnormal signaling pathways. The signaling pathways mediating apoptosis include extrinsic pathways mediated by death receptors and intrinsic pathways mediated by mitochondria [77]. Atractylone primarily targets the intrinsic pathway, which relies on the release of specific active substances from mitochondria. Thus, this apoptotic pathway is referred to as mitochondrial apoptosis. The intrinsic pathway is triggered upon internal stimulation, resulting in the opening of the mitochondrial permeability transition pore and loss of the mitochondrial transmembrane potential. As an outcome, several proapoptotic proteins, such as cytochrome c, are released into the cytoplasm, eventually activating caspase. Though different pathways may initiate apoptosis, cell death is induced through the execution pathway, which involves caspase-3 activation [78,79]. |
ROS | ROS is a byproduct of normal cellular metabolism, which contains derivatives comprising highly unstable oxygen free radicals, such as superoxide anion (O2•−) and hydroxyl (OH•). Mitochondria and NADPH oxidases are two significant contributors to endogenous ROS in cancer [80]. Atractylone affects hepatocellular carcinoma cells by inducing apoptosis via the mitochondrial apoptotic pathway, which characterizes a reduction in MTP and an elevation in ROS levels. The implications of increased ROS levels can be multifaceted, as they may both promote and inhibit malignant behavior. Elevated ROS levels are considered oncogenic due to their capacity to oxidize nucleic acids and cause damage to DNA, proteins, and lipids. Furthermore, Atractylone functions as a potential antioxidant by facilitating the degradation of superoxide anion (O2•−) and hydrogen peroxide (H2O2), thereby preventing oxidative damage to DNA. Additionally, Atractylone has been shown to diminish the production of inflammatory factors and oxidative stress in the gastrointestinal tract. |
MMPs | Matrix metalloproteinase-9 (MMP-9), whose expression is frequently dysregulated in cancer, promotes tumor growth, invasion, and metastasis by multiple mechanisms. MMPs have long been associated with solid tumor invasion, metastasis, and angiogenesis [81]. MMP-9 regulates tumor growth, invasion, and metastasis, including proteolytic remodeling of extracellular matrix (ECM), alteration of cell–cell and cell–ECM interactions, migration, and angiogenesis [82]. After treatment, Atractylone significantly reduces the expression of INF-γ, TNF-α, and MMP-9 in colorectal cancer cells. MMPs are zinc-dependent endopeptidases that play a crucial role in the degradation and remodeling of ECM components and basement membranes. Overexpression of MMP genes can lead to ECM remodeling, thereby enhancing cancer cells’ invasive and migratory capabilities. Conversely, inhibiting the expression of MMP-2 and MMP-9 can decrease cancer cell adhesion and metastasis [83,84]. Therefore, targeting the suppression of MMP expression presents an effective strategy for controlling cancer cell metastasis. |
AKT | AKT is a promising target and an effector of the PI3K/AKT/mTOR pathway that activates tumors. The AKT kinase family comprises the AKT1, AKT2, and AKT3 isoforms. AKT activity is controlled in an AKT-dependent manner via phosphorylation and dephosphorylation [85]. The PI3K/AKT/mTOR axis is significantly altered in cancer, and targeting this axis with multiple inhibitors can modulate a variety of cellular processes such as cell proliferation, autophagy, apoptosis, angiogenesis, EMT, and chemoresistance [86]. The PI3K/AKT/mTOR pathway is a key target for treating colorectal cancer, and after treatment with Atractylone, the expression of PI3K, AKT, and mTOR in colorectal cancer cells is significantly reduced. Aberrant activation of this pathway induces cell survival and metastasis. It can also lead cancer cells to escape from apoptosis through dysregulation of anti- and proapoptotic genes. It also possesses a central role in EMT promotion and chemoresistance [87]. Targeted therapy against the PI3K pathway may be an essential strategy in treating colorectal cancer. |
Arresting the cell cycle | The process of cell division plays a vital role in cancer progression. Cell proliferation and error-free chromosome segregation during mitosis are central events in the life cycle. Mistakes during cell division generate changes in chromosome content and alter chromosome number balances [88]. Cancer cells are characterized by their ability to proliferate and divide indefinitely, evading regulation by the cell cycle. This uncontrolled proliferation is a hallmark of dysregulated cell cycles. Cancer cells have evolved mechanisms that allow them to alter their structure and bypass signaling pathways and checkpoint controls to escape surveillance by the immune system and circumvent regulatory checkpoints in the cell cycle. These adaptations ensure genomic stability in cancer cells while promoting unrestrained growth. Consequently, targeting the cell cycle has been regarded as a promising strategy for cancer therapy [89]. One approach within cell-cycle-targeted therapy involves controlling the expression of cyclins and cyclin-dependent kinases (CDKs), or other molecules associated with abnormal checkpoint regulation, to manage cancer cell growth [90]. Atractylone has been shown to inhibit the proliferation and migration of glioblastoma (GBM) cells while inducing apoptosis through G1 phase arrest in these cell. Further research is needed to explore its significant potential in targeted therapies against cancer related to the cell cycle. |
Microbial flora | Gastrointestinal microorganisms have been identified as significant factors influencing cancer development in studies examining the effects of Atractylone on colon and gastric cancer. Inflammatory factors play a central role in the occurrence and progression of cancer. Atractylone has been shown to reduce the production of inflammatory factors and oxidative stress within the gastrointestinal tract. This is achieved by lowering tumor necrosis factor (TNF)-α and ROS levels while simultaneously increasing the expression of adhesion proteins such as claudin, ZO-1, and occludin. These actions help regulate inflammation-related dysbiosis in gut microbiota when treating gastrointestinal diseases. The effects of Atractylone primarily focus on amino acid metabolic pathways, which are beneficial for repairing gastrointestinal damage and enhancing intestinal immunity. |
Epithelial–mesenchymal transition (EMT) | During the EMT transformation process, EMT transcription factors, cytokines, related signaling pathways, and some noncoding RNAs mediate the resistance of HCCs. Atractylone can regulate the EMT process and EMT-related proteins, reduce cell-to-cell adhesion, change cell morphology, enhance cell migration and invasion ability, and significantly improve the resistance of HCCs. EMT is a switch that allows cells to transition between epithelial and mesenchymal states and is a key to the metastatic process [91]. |
TMPO-AS1 and CCDC183-AS1 | Atractylone has been shown to regulate the expression of TMPO-AS1 and CCDC183-AS1, inhibit the proliferation, invasion, and migration of HepG2 hepatocellular carcinoma cells, and improve cell apoptosis, making it a potential target for the treatment of liver cancer with Atractylone. LncRNA TMPO-AS1 can regulate the progression of the cancer cell cycle and adhesion, and further affect the prognosis of patients. CCDC183-AS1 can improve the proliferation, invasion, and migration of HepG2 cells treated with Atractylone and increase cell apoptosis [92,93,94]. |
SIRT3 | SIRT3 has always been a focus of cancer research, playing a central role in mitochondrial biology. It can promote cell survival by regulating the oxidative stress pathway while maintaining ROS levels, proliferation, and invasive phenotypes to prevent cell apoptosis and promote carcinogenesis. The mechanism of action of Atractylone on SIRT3 is still not well understood and needs further research. |
Notch 1 | The Notch 1 pathway is closely related to various signaling pathways in tumor development. Atractylone has not yet deepened research on Notch 1. It acts on the Notch1 pathway, playing a core role in the growth of GBM tumors, angiogenesis, and radiation resistance. Notch ss actively involved in a process known as epithelial-to-mesenchymal transition (EMT), in which epithelial cells obtain a mesenchymal phenotype, ultimately leading to migration. This is achieved by the interaction of Notch with transcription factors Slug, Snail, and TGF-b, which are critical for EMT [95]. Notch signaling highly regulates the formation and maintenance of cancer stem cells (CSCs), which lead to metastasis and tumorigenesis. |
Other targets | Dipeptidyl peptidase IV, retinoid acid β receptor, and cellular retinoic acid-binding protein 2 are also predictive targets for the anticancer effects of Atractylone. Specific studies have not yet been conducted, and this still needs further exploration. |
5. Materials and Methods
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Zugazagoitia, J.; Guedes, C.; Ponce, S.; Ferrer, I.; Molina-Pinelo, S.; Paz-Ares, L. Current Challenges in Cancer Treatment. Clin. Ther. 2016, 38, 1551–1566. [Google Scholar] [CrossRef] [PubMed]
- Stout, N.L.; Mina, D.S.; Lyons, K.D.; Robb, K.; Silver, J.K. A systematic review of rehabilitation and exercise recommendations in oncology guidelines. CA Cancer J. Clin. 2021, 71, 149–175. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Stanková, K.; Brown, J.S.; Dalton, W.S.; Gatenby, R.A. Optimizing Cancer Treatment Using Game Theory: A Review. JAMA Oncol. 2019, 5, 96–103. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Pérez-Herrero, E.; Fernández-Medarde, A. Advanced targeted therapies in cancer: Drug nanocarriers, the future of chemotherapy. Eur. J. Pharm. Biopharm. 2015, 93, 52–79. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.J.; Zhao, Z.Y.; Chang, L.K.; Cao, Y.; Wang, S.; Kang, C.Z.; Wang, H.Y.; Zhou, L.; Huang, L.Q.; Guo, L.P. Atractylodis Rhizoma: A review of its traditional uses, phytochemistry, pharmacology, toxicology and quality control. J. Ethnopharmacol. 2021, 266, 113415. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Jun, X.; Fu, P.; Lei, Y.; Cheng, P. Pharmacological effects of medicinal components of Atractylodes lancea (Thunb.) DC. Chin. Med. 2018, 13, 59. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Cao, Y.; Zhang, W.J.; Chang, L.K.; Kang, C.Z.; Wang, Y.F.; Xie, D.M.; Wang, S.; Guo, L.P. Comparison of transcriptome of Atractylodes lancea rhizome and exploration of genes for sesquiterpenoid biosynthesis. Zhongguo Zhong Yao Za Zhi 2022, 47, 4895–4907. (In Chinese) [Google Scholar] [CrossRef] [PubMed]
- Zhu, B.; Zhang, Q.L.; Hua, J.W.; Cheng, W.L.; Qin, L.P. The traditional uses, phytochemistry, and pharmacology of Atractylodes macrocephala Koidz.: A review. J. Ethnopharmacol. 2018, 226, 143–167. [Google Scholar] [CrossRef] [PubMed]
- Koonrungsesomboon, N.; Na-Bangchang, K.; Karbwang, J. Therapeutic potential and pharmacological activities of Atractylodes lancea (Thunb.) DC. Asian Pac. J. Trop. Med. 2014, 7, 421–428. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y.X.; Lu, W.W.; Geng, Y.C.; Yu, C.H.; Sun, H.J.; Kim, Y.J.; Zhang, G.; Kim, T. Antioxidant, Antimicrobial and Anti-Inflammatory Activities of Essential Oil Derived from the Wild Rhizome of Atractylodes macrocephala. Chem. Biodivers. 2020, 17, e2000268. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.J.; Yao, J.; Song, X.; He, L.W.; Meng, X.C. Overview of chemical composition, pharmacological effects, and clinical application of medicinal plants of Atractylodes DC. Chin. Arch. Trad. Chin. Med. 2023, 41, 151–154. [Google Scholar]
- Ma, Z.; Liu, G.; Yang, Z.; Zhang, G.; Sun, L.; Wang, M.; Ren, X. Species Differentiation and Quality Evaluation for Atractylodes Medicinal Plants by GC/MS Coupled with Chemometric Analysis. Chem. Biodivers. 2023, 20, e202300793. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Li, L.; Ran, X.; Dou, D.; Li, B.; Yang, B.; Li, W.; Koike, K.; Kuang, H. What caused the changes in the usage of Atractylodis Macrocephalae Rhizoma from ancient to current times? J. Nat. Med. 2016, 70, 36–44. [Google Scholar] [CrossRef] [PubMed]
- Wang, K.T.; Chen, L.G.; Yang, L.L.; Ke, W.M.; Chang, H.C.; Wang, C.C. Analysis of the sesquiterpenoids in processed Atractylodis Rhizoma. Chem. Pharm. Bull. 2007, 55, 50–56. [Google Scholar] [CrossRef] [PubMed]
- Hao, Y.; Zhang, X.; Lin, X.; Yang, S.; Huang, Y.; Lai, W.; Liao, X.; Liao, W.; Fu, C.; Zhang, Z. The traditional Chinese medicine processing change chemical composition and pharmacological effectiveness: Taking Atractylodes macrocephala Koidz. and honey bran-fried Atractylodes macrocephala Koidz. as examples. Phytomedicine 2024, 130, 155739. [Google Scholar] [CrossRef] [PubMed]
- Meng, Y.H.; Meng, X.Y.; Zhang, Z.P.; Liu, X.B.; Zhai, C.M. Determination of Atractylone and other four effective components in Atractylodes macrocephala and its processed products by HPLC. Chem. Eng. 2019, 33, 24–26. [Google Scholar]
- Guo, L.P.; LIU, J.Y.; JI, L.; Huang, L.Q. The naphtha composing characteristics of geoherbs of Atractylodes lancea. Zhongguo Zhong Yao Za Zhi 2002, 27, 814–819. (In Chinese) [Google Scholar] [PubMed]
- Duan, Y.; Meng, L. Research progress in multi-targeted anti-tumor natural products. Acta Pharm. Sin. 2021, 403–413. [Google Scholar]
- Farhat, J.; Alzyoud, L.; Alwahsh, M.; Al-Omari, B. Structure-Activity Relationship of Benzofuran Derivatives with Potential Anticancer Activity. Cancers 2022, 14, 2196. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Alizadeh, M.; Jalal, M.; Hamed, K.; Saber, A.; Kheirouri, S.; Pourteymour Fard Tabrizi, F.; Kamari, N. Recent Updates on Anti-Inflammatory and Antimicrobial Effects of Furan Natural Derivatives. J. Inflamm. Res. 2020, 13, 451–463. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Kim, H.K.; Yun, Y.K.; Ahn, Y.J. Toxicity of atractylon and atractylenolide III Identified in Atractylodes ovata rhizome to Dermatophagoides farinae and Dermatophagoides pteronyssinus. J. Agric. Food Chem. 2007, 55, 6027–6031. [Google Scholar] [CrossRef] [PubMed]
- Yi, Z.R.; Zhang, R.Y.; Wang, J.L.; Li, K.A.; Zhang, Y.X.; Chen, M.F.; Zhen, Y.J.; Tang, Q. Determination of volatile components in different parts of Atractylodes macrocephala Koidz. by HS-SPME-GC-MS. Nat. Prod. Res. Dev. 2022, 34, 1690–1698. [Google Scholar]
- Taniguchi, H. Liver Cancer 2.0. Int. J. Mol. Sci. 2023, 24, 17275. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Rawla, P.; Sunkara, T.; Muralidharan, P.; Raj, J.P. Update in global trends and aetiology of hepatocellular carcinoma. Contemp. Oncol. 2018, 22, 141–150. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Yang, J.D.; Hainaut, P.; Gores, G.J.; Amadou, A.; Plymoth, A.; Roberts, L.R. A global view of hepatocellular carcinoma: Trends, risk, prevention and management. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 589–604. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Cheng, Y.; Chen, T.; Yang, X.; Xue, J.; Chen, J. Atractylon induces apoptosis and suppresses metastasis in hepatic cancer cells and inhibits growth in vivo. Cancer Manag. Res. 2019, 11, 5883–5894. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Moloney, J.N.; Cotter, T.G. ROS signalling in the biology of cancer. Semin. Cell Dev. Biol. 2018, 80, 50–64. [Google Scholar] [CrossRef] [PubMed]
- Guo, X.; Wang, Y. LncRNA TMPO-AS1 promotes hepatocellular carcinoma cell proliferation, migration and invasion through sponging miR-329-3p to stimulate FOXK1-mediated AKT/mTOR signaling pathway. Cancer Med. 2020, 9, 5235–5246. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Zhu, H.; Zhang, H.; Pei, Y.; Liao, Z.; Liu, F.; Su, C.; Liu, Y.; Dong, R.; Song, J.; Zhang, X.; et al. Long non-coding RNA CCDC183-AS1 acts AS a miR-589-5p sponge to promote the progression of hepatocellular carcinoma through regulating SKP1 expression. J. Exp. Clin. Cancer Res. 2021, 40, 57. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Cheng, Y.; Ping, J.; Chen, J.; Fu, Y.; Zhao, H.; Xue, J. Molecular mechanism of atractylon in the invasion and migration of hepatic cancer cells based on high-throughput sequencing. Mol. Med. Rep. 2022, 25, 112. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Yang, X.L.; Xue, J.H.; Chen, T.Y.; Ping, J.; Hou, T.L.; Chen, Y. Effect of atractylone on the viability and apoptosis of hepatoma HepG2 cells and related mechanism. J. Clin. Hepatobiliary Dis. 2021, 37, 2589–2594. [Google Scholar]
- Guo, N.N. Experimental Study on Atractylodes Ketone-Induced Apoptosis in Human Hepatocellular Carcinoma Cells (HepG2). Bachelor’s Thesis, Shenzhen University, Shenzhen, China, 2015. (In Chinese). [Google Scholar]
- Mai, J.S.; Chen, S.; Ke, B.L.; He, Q.Q.; Wang, H.Y.; Ping, J.; Chen, J.J.; Chen, Y. Effects of atractylolone on tumor growth, epithelial mesenchymal transition and expression of apoptosis markers in hepatoma-bearing mice. Tradit. Chin. Med. Inf. 2022, 39, 28–32. (In Chinese) [Google Scholar]
- Jin, Y.Y.; Mai, J.X.; Ping, J. Effects and Mechanisms of Atractylone on Proliferation, Migration, Invasion and Apoptosis of Human Hepatocellular Carcinoma Cells MHCC97-H. New Drugs Clin. Pharmacol. Tradit. Chin. Med. 2023, 34, 1370–1376. (In Chinese) [Google Scholar]
- El-readi, M.Z.; Eid, S.; Abdelghany, A.A.; Al-Amoudi, H.S.; Efferth, T.; Wink, M. Resveratrol mediated cancer cell apoptosis, and modulation of multidrug resistance proteins and metabolic enzymes. Phytomedicine 2019, 55, 269–281. [Google Scholar] [CrossRef]
- Marin, J.J.G.; Macias, R.I.R.; Monte, M.J.; Romero, M.R.; Asensio, M.; Sanchez-Martin, A.; Cives-Losada, C.; Temprano, A.G.; Espinosa-Escudero, R.; Reviejo, M.; et al. Molecular Bases of Drug Resistance in Hepatocellular Carcinoma. Cancers 2020, 12, 1663. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Tang, L.; Chen, L.; Zhang, C. To Explore the Mechanism of Atractylon on Chemotherapy Resistance of Hepatocellular Carcinoma Cells Based on Notch1 Pathway. Her. Tradit. Chin. Med. 2023, 29, 11–16. (In Chinese) [Google Scholar] [CrossRef]
- Willyard, C. The Colon Cancer Conundrum. Nature, 2021; ahead of print. [Google Scholar] [CrossRef] [PubMed]
- Amri, R.; Bordeianou, L.G.; Berger, D.L. The conundrum of the young colon cancer patient. Surgery 2015, 158, 1696–1703. [Google Scholar] [CrossRef]
- Burris, H.A., 3rd. Overcoming acquired resistance to anticancer therapy: Focus on the PI3K/AKT/mTOR pathway. Cancer Chemother. Pharmacol. 2013, 71, 829–842. [Google Scholar] [CrossRef] [PubMed]
- Geng, W.; Liang, W.; Ye, Z.B.; Fan, Y.N. Mechanism of Atractylodes atractylone on HT-29 apoptosis in colorectal cancer cells. Chin. Pat. Med. 2018, 40, 937–940. (In Chinese) [Google Scholar]
- Mao, J.; Wang, X.; Yu, M.; Sun, C. Effects of Atractylon on Proliferation and Apoptosis of Intestinal Cancer Cells Through PI3K/AKT/mTOR Signaling Pathway. Cell Mol. Biol. 2022, 68, 153–160. [Google Scholar] [CrossRef] [PubMed]
- Malla, R.R. Microbiome Conundrum in Colon Cancer: Development, Progression, and Therapeutics. Crit. Rev. Oncog. 2020, 25, 129–139. [Google Scholar] [CrossRef] [PubMed]
- Lavelle, A.; Lennon, G.; O’Sullivan, O.; Docherty, N.; Balfe, A.; Maguire, A.; Mulcahy, H.E.; Doherty, G.; O’Donoghue, D.; Hyland, J.; et al. Spatial variation of the colonic microbiota in patients with ulcerative colitis and control volunteers. Gut 2015, 64, 1553–1561. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Wieczorska, K.; Stolarek, M.; Stec, R. The Role of the Gut Microbiome in Colorectal Cancer: Where Are We? Where Are We Going? Clin. Colorectal Cancer 2020, 19, 5–12. [Google Scholar] [CrossRef] [PubMed]
- Gkikas, K.; Svolos, V.; Hansen, R.; Russell, R.K.; Gerasimidis, K. Take-Home Messages from 20 Years of Progress in Dietary Therapy of Inflammatory Bowel Disease. Ann. Nutr. Metab. 2023, 79, 476–484. [Google Scholar] [CrossRef] [PubMed]
- Cotugno, R.; Gallotta, D.; Piaz, F.D.; Apicella, I.; De Falco, S.; Rosselli, S.; Bruno, M.; Belisario, M.A. Powerful tumor cell growth-inhibiting activity of a synthetic derivative of atractyligenin: Involvement of PI3K/Akt pathway and thioredoxin system. Biochim. Biophys. Acta 2014, 1840, 1135–1144. [Google Scholar] [CrossRef] [PubMed]
- Feng, W.; Ao, H.; Yue, S.; Peng, C. Systems pharmacology reveals the unique mechanism features of Shenzhu Capsule for treatment of ulcerative colitis in comparison with synthetic drugs. Sci. Rep. 2018, 8, 16160. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Kanneganti, T.D. Inflammatory Bowel Disease and the NLRP3 Inflammasome. N. Engl. J. Med. 2017, 377, 694–696. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; He, Y.; Wang, N.; Li, Y.; Du, Y.; He, N.; Wang, B.; Zhang, T. Atractylone in the Atractylodes macrocephala Rhizoma Essential Oil and Its Anti-Inflammatory Activity. Molecules 2023, 28, 7340. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Cheng, Y.; Ling, Z.; Li, L. The Intestinal Microbiota and Colorectal Cancer. Front. Immunol. 2020, 11, 615056. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Chokshi, C.R.; Brakel, B.A.; Tatari, N.; Savage, N.; Salim, S.K.; Venugopal, C.; Singh, S.K. Advances in Immunotherapy for Adult Glioblastoma. Cancers 2021, 13, 3400. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Colamaria, A.; Leone, A.; Fochi, N.P.; Di Napoli, V.; Giordano, G.; Landriscina, M.; Patel, K.; Carbone, F. Tumor treating fields for the treatment of glioblastoma: Current understanding and future perspectives. Surg. Neurol. Int. 2023, 14, 394. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Sun, S.; Shi, J.; Wang, X.; Huang, C.; Huang, Y.; Xu, J.; Jiang, Y.; Cao, L.; Xie, T.; Wang, Y.; et al. Atractylon inhibits the tumorigenesis of glioblastoma through SIRT3 signaling. Am. J. Cancer Res. 2022, 12, 2310–2322. [Google Scholar] [PubMed] [PubMed Central]
- Lai, C.C.; Lin, P.M.; Lin, S.F.; Hsu, C.H.; Lin, H.C.; Hu, M.L.; Hsu, C.M.; Yang, M.Y. Altered expression of SIRT gene family in head and neck squamous cell carcinoma. Tumour Biol. 2013, 34, 1847–1854. [Google Scholar] [CrossRef] [PubMed]
- Lin, Y.; Liu, X.; Tan, D.; Jiang, Z. Atractylon treatment prevents sleep-disordered breathing-induced cognitive dysfunction by suppression of chronic intermittent hypoxia-induced M1 microglial activation. Biosci. Rep. 2020, 40, BSR20192800. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Bazzoni, R.; Bentivegna, A. Role of Notch Signaling Pathway in Glioblastoma Pathogenesis. Cancers 2019, 11, 292. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Han, N.; Hu, G.; Shi, L.; Long, G.; Yang, L.; Xi, Q.; Guo, Q.; Wang, J.; Dong, Z.; Zhang, M. Notch1 ablation radiosensitizes glioblastoma cells. Oncotarget 2017, 8, 88059–88068. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Zhao, M.; Wang, Q.; Ouyang, Z.; Han, B.; Wang, W.; Wei, Y.; Wu, Y.; Yang, B. Selective fraction of Atractylodes lancea (Thunb.) DC. and its growth inhibitory effect on human gastric cancer cells. Cytotechnology 2014, 66, 201–208. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Gu, S.; Li, L.; Huang, H.; Wang, B.; Zhang, T. Antitumor, Antiviral, and Anti-Inflammatory Efficacy of Essential Oils from Atractylodes macrocephala Koidz. Produced with Different Processing Methods. Molecules 2019, 24, 2956. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Vivarelli, S.; Salemi, R.; Candido, S.; Falzone, L.; Santagati, M.; Stefani, S.; Torino, F.; Banna, G.L.; Tonini, G.; Libra, M. Gut Microbiota and Cancer: From Pathogenesis to Therapy. Cancers 2019, 11, 38. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Villéger, R.; Lopès, A.; Carrier, G.; Veziant, J.; Billard, E.; Barnich, N.; Gagnière, J.; Vazeille, E.; Bonnet, M. Intestinal Microbiota: A Novel Target to Improve Anti-Tumor Treatment? Int. J. Mol. Sci. 2019, 20, 4584. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Li, L.; Du, Y.; Wang, Y.; He, N.; Wang, B.; Zhang, T. Atractylone Alleviates Ethanol-Induced Gastric Ulcer in Rat with Altered Gut Microbiota and Metabolites. J. Inflamm. Res. 2022, 15, 4709–4723. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Wang, C.C.; Chen, L.G.; Yang, L.L. Cytotoxic activity of sesquiterpenoids from Atractylodes ovata on leukemia cell lines. Planta Med. 2002, 68, 204–208. [Google Scholar] [CrossRef] [PubMed]
- Yu, S.; Yasukawa, K.; Takido, M. Atractylodis Rhizoma extract and its component, atractylon, inhibit tumor promotion in mouse skin two-stage carcinogenesis. Phytomedicine 1994, 1, 55–58. [Google Scholar] [CrossRef] [PubMed]
- Desouki, M.M.; Doubinskaia, I.; Gius, D.; Abdulkadir, S.A. Decreased mitochondrial SIRT3 expression is a potential molecular biomarker associated with poor outcome in breast cancer. Hum. Pathol. 2014, 45, 1071–1077. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Marfe, G.; Tafani, M.; Indelicato, M.; Sinibaldi-Salimei, P.; Reali, V.; Pucci, B.; Fini, M.; Russo, M.A. Kaempferol induces apoptosis in two different cell lines via Akt inactivation, Bax and SIRT3 activation, and mitochondrial dysfunction. J. Cell Biochem. 2009, 106, 643–650. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Liu, Y.L.; Cheng, W.; Yin, X.M.; Jiang, B. The expression of SIRT3 in primary hepatocellular carcinoma and the mechanism of its tumor suppressing effects. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 978–998. [Google Scholar] [PubMed]
- Schultz, A.G.; Godfrey, J.D. An annelation approach to the synthesis of eudesmane and elemane sesquiterpene lactones. Total synthesis of dl-dihydrocallitrisin, dl-7,8-epialantolactone, dl-7,8-epiisoalantolactone, and dl-atractylon. J. Am. Chem. Soc. 1980, 102, 2414–2428. [Google Scholar] [CrossRef]
- Jonida, S.; Lúcia, S. One-Step Synthesis of Furan Rings from α-Isopropylidene Ketones Mediated by Iodine/DMSO: An Approach to Potent Bioactive Terpenes. J. Org. Chem. 2019, 84, 6886–6894. [Google Scholar]
- Singhal, R.; Choudhary, S.P.; Malik, B.; Pilania, M. I2/DMSO-mediated oxidative C-C and C-heteroatom bond formation: A sustainable approach to chemical synthesis. RSC Adv. 2024, 14, 5817–5845. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Honan, M.C. Total synthesis of (±)-atractylon and (±)-lindestrene. Tetrahedron Lett. 1985, 26, 6393–6396. [Google Scholar] [CrossRef]
- Bagal, S.K.; Adlington, R.M.; Baldwin, J.E.; Marquez, R. Dimerization of Butenolide Structures. A Biomimetic Approach to the Dimeric Sesquiterpene Lactones (±)-Biatractylolide and (±)-Biepiasterolide. J. Org. Chem. 2004, 69, 9100–9108. [Google Scholar]
- Kim, J.H.; Lee, Y.; Lee, G.; Doh, E.J.; Hong, S. Quantitative Interrelation between Atractylenolide I, II, and III in Koidzumi Rhizomes, and Evaluation of Their Oxidative Transformation Using a Biomimetic Kinetic Model. ACS Omega 2018, 3, 14833–14840. [Google Scholar] [CrossRef] [PubMed]
- Hu, C.Y.; Guo, W.Q.; Zang, S.; Gu, H.J.; Jiang, W.N.; Wang, T.Y. Poteintial targets prediction research of atractylon using Autodock. Biotechnology 2014, 24, 60–63. (In Chinese) [Google Scholar]
- Zhou, Y.; Lu, J.; Zhang, X.D.; Yi, Z.X.; Wang, B.L. Potential target prediction and forward molecular docking verification of atractylon. J. Shaoyang Univ. (Nat. Sci. Ed.) 2019, 16, 98–104. [Google Scholar]
- Kashyap, D.; Garg, V.K.; Goel, N. Intrinsic and extrinsic pathways of apoptosis: Role in cancer development and prognosis. Adv. Protein Chem. Struct. Biol. 2021, 125, 73–120. [Google Scholar] [CrossRef] [PubMed]
- Srivastava, N.; Saxena, A.K. Caspase-3 Activators as Anticancer Agents. Curr. Protein Pept. Sci. 2023, 24, 783–804. [Google Scholar] [CrossRef] [PubMed]
- Eskandari, E.; Eaves, C.J. Paradoxical roles of caspase-3 in regulating cell survival, proliferation, and tumorigenesis. J. Cell Biol. 2022, 221, e202201159. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Cheung, E.C.; Vousden, K.H. The role of ROS in tumour development and progression. Nat. Rev. Cancer 2022, 22, 280–297. [Google Scholar] [CrossRef] [PubMed]
- Das, S.; Amin, S.A.; Jha, T. Inhibitors of gelatinases (MMP-2 and MMP-9) for the management of hematological malignancies. Eur. J. Med. Chem. 2021, 223, 113623. [Google Scholar] [CrossRef] [PubMed]
- Juric, V.; O’Sullivan, C.; Stefanutti, E.; Kovalenko, M.; Greenstein, A.; Barry-Hamilton, V.; Mikaelian, I.; Degenhardt, J.; Yue, P.; Smith, V.; et al. MMP-9 inhibition promotes anti-tumor immunity through disruption of biochemical and physical barriers to T-cell trafficking to tumors. PLoS ONE 2018, 13, e0207255. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Qian, C.; Wang, J.; Qian, Y.; Hu, R.; Zou, J.; Zhu, C.; Zhu, Y.; Qi, S.; Jia, X.; Wu, L.; et al. Tumor-Cell-Surface Adherable Peptide-Drug Conjugate Prodrug Nanoparticles Inhibit Tumor Metastasis and Augment Treatment Efficacy. Nano Lett. 2020, 20, 4153–4161. [Google Scholar] [CrossRef] [PubMed]
- Huang, H. Matrix Metalloproteinase-9 (MMP-9) as a Cancer Biomarker and MMP-9 Biosensors: Recent Advances. Sensors 2018, 18, 3249. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Revathidevi, S.; Munirajan, A.K. Akt in cancer: Mediator and more. Semin. Cancer Biol. 2019, 59, 80–91. [Google Scholar] [CrossRef] [PubMed]
- Peng, Y.; Wang, Y.; Zhou, C.; Mei, W.; Zeng, C. PI3K/Akt/mTOR Pathway and Its Role in Cancer Therapeutics: Are We Making Headway? Front. Oncol. 2022, 12, 819128. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Fattahi, S.; Amjadi-Moheb, F.; Tabaripour, R.; Ashrafi, G.H.; Akhavan-Niaki, H. PI3K/AKT/mTOR signaling in gastric cancer: Epigenetics and beyond. Life Sci. 2020, 262, 118513. [Google Scholar] [CrossRef] [PubMed]
- Jamasbi, E.; Hamelian, M.; Hossain, M.A.; Varmira, K. The cell cycle, cancer development and therapy. Mol. Biol. Rep. 2022, 49, 10875–10883. [Google Scholar] [CrossRef] [PubMed]
- Thu, K.L.; Soria-Bretones, I.; Mak, T.W.; Cescon, D.W. Targeting the cell cycle in breast cancer: Towards the next phase. Cell Cycle 2018, 17, 1871–1885. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Wenzel, E.S.; Singh, A.T.K. Cell-cycle Checkpoints and Aneuploidy on the Path to Cancer. In Vivo 2018, 32, 1–5. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
- Jonckheere, S.; Adams, J.; De-Groote, D.; Campbell, K.; Berx, G.; Goossens, S. Epithelial-Mesenchymal Transition (EMT) as a Therapeutic Target. Cells Tissues Organs 2022, 211, 157–182. [Google Scholar] [CrossRef] [PubMed]
- Li, D.S.; Ainiwaer, J.L.; Sheyhiding, I.; Zhang, Z.; Zhang, L.W. Identification of key long non-coding RNAs as competing endogenous RNAs for miRNA-mRNA in lung adenocarcinoma. Eur. Rev. Med. Pharmacol. Sci. 2016, 20, 2285–2295. [Google Scholar] [PubMed]
- Mu, X.; Wu, H.; Liu, J.; Hu, X.; Wu, H.; Chen, L.; Liu, W.; Luo, S.; Zhao, Y. Long noncoding RNA TMPO-AS1 promotes lung adenocarcinoma progression and is negatively regulated by miR-383-5p. Biomed. Pharmacother. 2020, 125, 109989. [Google Scholar] [CrossRef] [PubMed]
- Huang, W.; Su, X.; Yan, W.; Kong, Z.; Wang, D.; Huang, Y.; Zhai, Q.; Zhang, X.; Wu, H.; Li, Y.; et al. Overexpression of AR-regulated lncRNA TMPO-AS1 correlates with tumor progression and poor prognosis in prostate cancer. Prostate 2018, 78, 1248–1261. [Google Scholar] [CrossRef] [PubMed]
- Vinson, K.E.; George, D.C.; Fender, A.W.; Bertrand, F.E.; Sigounas, G. The Notch pathway in colorectal cancer. Int. J. Cancer 2016, 138, 1835–1842. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Yao, Y.; Shen, G.; Luo, J.; Wang, J.; Xu, Z.; Wang, H.; Cui, L. Research Progress with Atractylone as an Antitumor Agent. Molecules 2024, 29, 5450. https://doi.org/10.3390/molecules29225450
Yao Y, Shen G, Luo J, Wang J, Xu Z, Wang H, Cui L. Research Progress with Atractylone as an Antitumor Agent. Molecules. 2024; 29(22):5450. https://doi.org/10.3390/molecules29225450
Chicago/Turabian StyleYao, Ying, Guanghuan Shen, Jianghan Luo, Jinhong Wang, Zheng Xu, Hao Wang, and Linlin Cui. 2024. "Research Progress with Atractylone as an Antitumor Agent" Molecules 29, no. 22: 5450. https://doi.org/10.3390/molecules29225450
APA StyleYao, Y., Shen, G., Luo, J., Wang, J., Xu, Z., Wang, H., & Cui, L. (2024). Research Progress with Atractylone as an Antitumor Agent. Molecules, 29(22), 5450. https://doi.org/10.3390/molecules29225450