The Role and Mechanism of Perilla frutescens in Cancer Treatment
<p>Flowchart. The antitumor effect of <span class="html-italic">Perilla frutescens</span> was verified by web pharmacology analysis of <span class="html-italic">Perilla frutescens</span>. Literature search, review, and synthesis of the literature to summarize the mechanism of action and related signaling pathways of <span class="html-italic">Perilla frutescens</span> as well as active ingredients against tumors. In the red box are cancer-related pathways in the KEGG pathway.</p> "> Figure 2
<p><span class="html-italic">Perilla</span>-active composition target plot and GO and KEGG analyses. In the red box are the signaling pathways associated with various types of cancer in the KEGG enrichment analysis.</p> "> Figure 3
<p>The chemical classes of <span class="html-italic">Perilla frutescens</span> compounds.</p> "> Figure 4
<p>Structural formulae of representative compounds: (<b>A</b>) Perillaldehyde, (<b>B</b>) Rosmaric acid, (<b>C</b>) Luteolin, and (<b>D</b>) Isoegomaketone.</p> "> Figure 5
<p>Anticancer mechanism of <span class="html-italic">Perilla frutescens</span>.</p> ">
Abstract
:1. Introduction
2. Network Diagram of Anti-Tumor Effect of Perilla frutescens
3. Active Ingredients
3.1. Alkaloids
3.2. Phenylpropane
3.3. Terpenoids
3.4. Polyphenol Compounds
3.5. Flavonoids
3.6. Anthocyanins, Coumarins, Carotenoids, and Neolignans
3.7. Fatty Acids, Tocopherols, and Phytosterols
3.8. Glucoside and Peptide
Active Ingredients | Species | References |
---|---|---|
Alkaloids | Neoechinulin A | [26] |
Benzene propane | Eleuthero | [49] |
Myricetin | [49] | |
Eugenol | [75] | |
Terpenoids | Perillone | [76] |
Perillaldehyde | [77] | |
Polyphenols | Rosmarinic acid | [78] |
Flavonoids | Luteolin | [78] |
Apigenin | [29] | |
Isoestradiol | [27] | |
Baicalin | [79] | |
Anthocyanins | Malonylstilbene | [79] |
Perillin | [80] | |
Carotenoids | Lolliolactone | [81] |
Isoxolactone | [81] | |
Neolignan | Mullein | [82] |
Gooseberry | [82] | |
Coumarins | Heptazine | [30] |
6,7-Dihydroxycoumarin | [83] | |
Fatty acids | Lauric acid | [84] |
Palm oleic acid | [85] | |
Tocopherol | Delta-tocopherol | [86] |
Gamma tocopherol | [86] | |
Beta tocopherol | [86] | |
Alpha tocopherol | [86] | |
Glucoside | Perilla lactone A | [81] |
Perillolactone B | [81] | |
Loganin | [87] | |
Phytosterols | Vegetable oil sterols | [86] |
Soysterol | [86] | |
beta-Sitosterol | [86] |
4. Anti-Cancer Compound Structural Formula
5. Anti-Cancer Effect
5.1. Cell Transfer
5.2. Apoptosis
5.3. Cell Cycle
5.4. Cell Senescence
5.5. Oxidative Stress Response and Cellular Inflammation
5.6. Cell Growth
5.7. Cell Proliferation
Type of Drug | Type of Cancer | Model | IC50 or Dose | Mechanism of Action | Reference |
---|---|---|---|---|---|
Perilla frutescens leaf extract | Colon cancer | HCT116 human colon cancer cells | Dose: 87.5–350 μg/mL | Inhibits the growth, colony formation, and adhesion of human colon and lung cancer cells and the migration of human lung cancer cells. | [100] |
Lung cancer | H1299 human non-small cell lung cancer cells | Dose: 87.5–350 μg/mL | |||
Perilla frutescens leaf extract | Triple negative breast cancer | HEK293A, MDA-MB-231, MCF10A and BT549 cells | HEK293A IC50: 584.3 μg/mL MDA-MB-231 IC50: 268.9 μg/mL MCF10A IC50: 650.8 μg/mL BT549 IC50: 307.1 μg/mL | Increased YAP phosphorylation and reduced YAP-TEAD-mediated transcriptional activity. | [102] |
IK | Prostate cancer | RC-58T/h/SA#4 cells | Dose: 10–200 ng/mL | Enhancement of tumor necrosis factor-related apoptosis-inducing ligands (TRAIL)-mediated apoptosis through upregulation of DR5 by an ROS-independent pathway. | [89] |
Perillaldehyde | Prostate cancer | RAW264.7 and PC-3 cells | Dose: 0.5–5 μM | Activation of the NF-κB pathway of nuclear factor-κB ligands and receptor activators to inhibit cancer cell-induced osteoclast formation. | [98] |
IK | Liver cancer | Huh-7 and Hep3B cells and nude mouse models of hepatocellular carcinoma | Dose: 10 nmol/L | Significantly inhibited cell viability and xenograft tumor formation in HCC cells and inhibited AKT phosphorylation, but not AKT and p38 expression. | [113] |
Perillaldehyde | Stomach cancer | MFC murine-derived cells and GC9811-P human gastric cancer cells | Dose: 0.1–5 mM | PAH activates AMPK by increasing Thr172 phosphorylation and activity; PAH increases the expression of beclin-1, LC3-II, caspase-3, and p53. | [112] |
PSO and Ros A | Lung cancer | A549 lung adenocarcinoma cells | Dose PSO: 0–400 μg/mL Dsoe Ros A: 0–40 μg/mL | PSO and Ros A scavenge TNF-α induced ROS levels, resulting in reduced expression of MnSOD, FOXO1, NF-κB, and JNK signaling pathways. | [91] |
PO | Breast cancer, colon cancer | Female SD rats | Dose: 10%PO | Alpha-linolenic acid-rich PO diet inhibits the development of breast, colon, and kidney tumors. | [114] |
PDMF | Lung cancer | Human lung adenocarcinoma A549 cells | Dose: 30–75 μg/mL | Triggering p53-driven G2/M cell cycle arrest and apoptosis. | [104] |
PDMF | Lung cancer | A549 human lung adenocarcinoma cells | Activation of the p21-p549 pathway in A53 cells; p53 is particularly important for cellular senescence | [107] | |
Ethanolic extract of Perilla frutescens (EPF) | Liver cancer | Human hepatocellular carcinoma HuH7 cells | IC50: 3.43 mg/mL | Protective effect of ethanol extract on the production of reactive oxygen species and lipid peroxidation in FeCl3–induction of HuH7 cells in a dose-dependent manner | [111] |
Perilla frutescens leaf extract | Skin tumors | - | Dose: 0.05% | Significant reduction in tumor incidence and diversity. | [115] |
IK | Melanoma | B16 melanoma cells | Dose: 10–100 μM | IK-induced apoptosis involves the production of ROS and the upregulation of Bax and Bcl-2 expression, leading to the release of cytochrome c and AIF. IK inhibits melanoma cell growth and induces apoptosis through the activation of ROS-mediated cysteinase-dependent and non-dependent pathways. | [116] |
Perilla extract | Liver cancer | Human hepatocellular carcinoma HepG2 cells | Dose: 105 μg/mL | Expression of a large number of apoptosis-related genes is regulated in a time-dependent manner. | [42] |
Perilla extract | Skin cancer | Two-stage skin carcinogenesis model in mice | Dose: 2.0 mg/mice | Part of the anti-cancer effect of perilla extract is due to RA through two separate mechanisms: inhibition of the inflammatory response and scavenging of reactive oxygen radicals. | [117] |
PO | Liver cancer | Diethylnitrosamine (DEN)-induced hepatocellular carcinoma in male F344 rats | Dose: 5% | PO enriched with n-6 and n-3 PUFA altered the membrane fatty acid composition of the liver and inhibited the development of hepatocellular carcinoma in rats. | [118] |
Luteolin | Colon cancer | HT-29 human colon cancer cells | Dose: 0–60 μmol/L | By activating caspase-3, -7, and -9, the cleavage of poly (ADP-ribose) polymerase was enhanced, the expressions of p21 (CIP1/WAF1), survivin, Mcl-1, Bcl-x(L), and Mdm-2 were decreased, and the activities of cyclin-dependent kinase (CDK)4 and CDK2 were inhibited. | [90] |
PO | Breast cancer | PhIP-induced mammary carcinogenesis model in rats | Dose: 0.1% | CFA-P may retard the development of PhIP-induced breast tumors, inhibit the formation of PhIP-DNA adducts, and reduce breast carcinogenesis in the context of post-initiation inhibition of cell proliferation. | [119] |
IK | Colon cancer | DLD1 colon cancer cells | Dose: 10–100 μM | IK treatment led to the cleavage of caspases-3, -8, and -9 in a dose- and time-dependent manner. IK treatment also led to cleavage of Bid and translocation of Bax. IK induced apoptosis via cystathione-dependent and caspase-non-dependent pathways in DLD1 cells. | [120] |
IK | Melanoma | SK-MEL-2 human melanoma Cells | Dose: 100 μM | IK-induced ROS production regulated cell growth inhibition and induced apoptosis through cysteinase-dependent and non-independent pathways by modulating PI2K/AKT signaling in SK-MEL-3 cells. Reduced protein levels of Bax and cytochrome c as well as PARP cleavage, while protein levels of Bcl-2 were increased. | [121] |
Ros A | Liver cancer | Hep G1 human liver cancer cells | IC50: 50 μM | Ros A dose-dependently attenuated aflatoxin- and hectoroxin-induced ROS production and inhibition of DNA and protein synthesis. Similarly, prevention of apoptosis by reduction of DNA fragmentation and inhibition of cysteinase-3 activation. | [122] |
EPF | Liver cancer | MDA-MB-231 human breast cancer cells | Dose: 2.5–10 μg/mL | EPF inhibits the ability of adrenergic agonists to promote cancer cell metastasis by inhibiting Src-mediated EMT. | [92] |
Breast cancer | Hep3B human hepatocellular carcinoma cells | Dose: 25–100 μg/mL |
6. Summary of Anticancer Mechanism
7. Preventative Effects
Composition | Cancer | Models | IC50 or Dose | Conclusion | Mechanisms | References |
---|---|---|---|---|---|---|
PDMF | Lung cancer | A549 human adenocarcinoma of the lung | Dose: 10–125 μM | PDMF and anti-cancer tyrosine kinase inhibitors (TKI) synergistically inhibit the proliferation of A549 cells. | Synergy | [127] |
PO | Colon cancer | Female F3 rats | Dose: 9%, 32%, 40%. | The relatively small amount of PO, accounting for 25% of total dietary fat, may have a significant beneficial effect in reducing the risk of colon cancer. | Preventive role | [125] |
PO | Colon Cancer | Male F344 rats | Dose: 3%, 6%, 12% | PO significantly reduced ras expression and AgNORs count (a biomarker of cell proliferation) in colonic mucosa. A significant increase in n-3 polyunsaturated fatty acids in the membrane phospholipid fraction and a decrease in PGE2 levels were observed in the colonic mucosa of rats fed with PO. | Preventive role | [94] |
PO | Colon cancer | Male F344 rats | Dose: 3%, 12% | β-Carotene plus PO also inhibited the number of silver-stained nucleolar organizer regions and the expression of ras mRNA (a biomarker of cell proliferation) in the colonic mucosa. | Synergy, preventive role | [128] |
PO | Colon cancer | Male F20 rats | Dose: 10%, 20% | Dietary PO significantly inhibits the development of small bowel and colon tumors in APC (min) mice. | Preventive role | [129] |
8. Summary and Outlook
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Sample Availability
References
- Torre, L.A.; Siegel, R.L.; Ward, E.M.; Jemal, A. Global Cancer Incidence and Mortality Rates and Trends—An Update. Cancer Epidemiol. Biomark. Prev. 2016, 25, 16–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hazafa, A.; Rehman, K.U.; Jahan, N.; Jabeen, Z. The Role of Polyphenol (Flavonoids) Compounds in the Treatment of Cancer Cells. Nutr. Cancer 2020, 72, 386–397. [Google Scholar] [CrossRef] [PubMed]
- Santucci, C.; Carioli, G.; Bertuccio, P.; Malvezzi, M.; Pastorino, U.; Boffetta, P.; Negri, E.; Bosetti, C.; La Vecchia, C. Progress in cancer mortality, incidence, and survival: A global overview. Eur. J. Cancer Prev. 2020, 29, 367–381. [Google Scholar] [CrossRef]
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
- Yusefi, A.R.; Bagheri Lankarani, K.; Bastani, P.; Radinmanesh, M.; Kavosi, Z. Risk Factors for Gastric Cancer: A Systematic Review. Asian Pac. J. Cancer Prev. 2018, 19, 591–603. [Google Scholar] [CrossRef]
- Roy, P.S.; Saikia, B.J. Cancer and cure: A critical analysis. Indian J. Cancer 2016, 53, 441–442. [Google Scholar] [CrossRef] [PubMed]
- Cross, D.; Burmester, J.K. Gene therapy for cancer treatment: Past, present and future. Clin. Med. Res. 2006, 4, 218–227. [Google Scholar] [CrossRef] [Green Version]
- Zaimy, M.A.; Saffarzadeh, N.; Mohammadi, A.; Pourghadamyari, H.; Izadi, P.; Sarli, A.; Moghaddam, L.K.; Paschepari, S.R.; Azizi, H.; Torkamandi, S.; et al. New methods in the diagnosis of cancer and gene therapy of cancer based on nanoparticles. Cancer Gene Ther. 2017, 24, 233–243. [Google Scholar] [CrossRef]
- Leibovici, J.; Itzhaki, O.; Huszar, M.; Sinai, J. Targeting the tumor microenvironment by immunotherapy: Part 2. Immunotherapy 2011, 3, 1385–1408. [Google Scholar] [CrossRef]
- Fatma, H.; Siddique, H.R. Research and Patents Status of Selected Phytochemicals Against Cancer: How Close and How Far? Recent Pat. Anticancer Drug Discov. 2023, 18, 428–447. [Google Scholar] [CrossRef]
- Shin, S.A.; Moon, S.Y.; Kim, W.Y.; Paek, S.M.; Park, H.H.; Lee, C.S. Structure-Based Classification and Anti-Cancer Effects of Plant Metabolites. Int. J. Mol. Sci. 2018, 19, 2651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khan, A.W.; Farooq, M.; Haseeb, M.; Choi, S. Role of Plant-Derived Active Constituents in Cancer Treatment and Their Mechanisms of Action. Cells 2022, 11, 326. [Google Scholar] [CrossRef] [PubMed]
- Kandaswami, C.; Lee, L.T.; Lee, P.P.; Hwang, J.J.; Ke, F.C.; Huang, Y.T.; Lee, M.T. The antitumor activities of flavonoids. In Vivo 2005, 19, 895–909. [Google Scholar] [PubMed]
- Mohi-Ud-Din, R.; Mir, R.H.; Sabreen, S.; Jan, R.; Pottoo, F.H.; Singh, I.P. Recent Insights into Therapeutic Potential of Plant-Derived Flavonoids against Cancer. Anticancer Agents Med. Chem. 2022, 22, 3343–3369. [Google Scholar] [CrossRef] [PubMed]
- Huang, W.Y.; Cai, Y.Z.; Zhang, Y. Natural phenolic compounds from medicinal herbs and dietary plants: Potential use for cancer prevention. Nutr. Cancer 2010, 62, 1–20. [Google Scholar] [CrossRef]
- Sharma, A.; Kaur, M.; Katnoria, J.K.; Nagpal, A.K. Polyphenols in Food: Cancer Prevention and Apoptosis Induction. Curr. Med. Chem. 2018, 25, 4740–4757. [Google Scholar] [CrossRef]
- Niedzwiecki, A.; Roomi, M.W.; Kalinovsky, T.; Rath, M. Anticancer Efficacy of Polyphenols and Their Combinations. Nutrients 2016, 8, 552. [Google Scholar] [CrossRef] [Green Version]
- Abel, S.; Riedel, S.; Gelderblom, W.C. Dietary PUFA and cancer. Proc. Nutr. Soc. 2014, 73, 361–367. [Google Scholar] [CrossRef] [PubMed]
- Zou, H.; Li, Y.; Liu, X.; Wu, Z.; Li, J.; Ma, Z. Roles of plant-derived bioactive compounds and related microRNAs in cancer therapy. Phytother. Res. 2021, 35, 1176–1186. [Google Scholar] [CrossRef] [PubMed]
- Samec, M.; Liskova, A.; Kubatka, P.; Uramova, S.; Zubor, P.; Samuel, S.M.; Zulli, A.; Pec, M.; Bielik, T.; Biringer, K.; et al. The role of dietary phytochemicals in the carcinogenesis via the modulation of miRNA expression. J. Cancer Res. Clin. Oncol. 2019, 145, 1665–1679. [Google Scholar] [CrossRef]
- Srivastava, S.K.; Arora, S.; Averett, C.; Singh, S.; Singh, A.P. Modulation of microRNAs by phytochemicals in cancer: Underlying mechanisms and translational significance. Biomed. Res. Int. 2015, 2015, 848710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pandey, A.; Bhatt, K.C. Diversity distribution and collection of genetic resources of cultivated and weedy type in Perilla frutescens (L.) Britton var. frutescens and their uses in Indian Himalaya. Genet. Resour. Crop Evol. 2008, 55, 883–892. [Google Scholar] [CrossRef]
- Yu, H.; Qiu, J.F.; Ma, L.J.; Hu, Y.J.; Li, P.; Wan, J.B. Phytochemical and phytopharmacological review of Perilla frutescens L. (Labiatae), a traditional edible-medicinal herb in China. Food Chem. Toxicol. 2017, 108, 375–391. [Google Scholar] [CrossRef] [PubMed]
- Hou, T.; Netala, V.R.; Zhang, H.; Xing, Y.; Li, H.; Zhang, Z. Perilla frutescens: A Rich Source of Pharmacological Active Compounds. Molecules 2022, 27, 3578. [Google Scholar] [CrossRef]
- Wang, P.; Jin, B.; Lian, C.; Guo, K.; Ma, C. Comparative Analysis of Polycyclic Aromatic Hydrocarbons and Halogenated Polycyclic Aromatic Hydrocarbons in Different Parts of Perilla frutescens (L.) Britt. Molecules 2022, 27, 3133. [Google Scholar] [CrossRef]
- Wang, X.F.; Li, H.; Jiang, K.; Wang, Q.Q.; Zheng, Y.H.; Tang, W.; Tan, C.H. Anti-inflammatory constituents from Perilla frutescens on lipopolysaccharide-stimulated RAW264.7 cells. Fitoterapia 2018, 130, 61–65. [Google Scholar] [CrossRef]
- Park, Y.D.; Jin, C.H.; Choi, D.S.; Byun, M.W.; Jeong, I.Y. Biological evaluation of isoegomaketone isolated from Perilla frutescens and its synthetic derivatives as anti-inflammatory agents. Arch. Pharm. Res. 2011, 34, 1277–1282. [Google Scholar] [CrossRef]
- Nam, B.; So, Y.; Kim, H.Y.; Kim, J.B.; Jin, C.H.; Han, A.R. A New Monoterpene from the Leaves of a Radiation Mutant Cultivar of Perilla frutescens var. crispa with Inhibitory Activity on LPS-Induced NO Production. Molecules 2017, 22, 1471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ha, T.J.; Lee, J.H.; Lee, M.H.; Lee, B.W.; Kwon, H.S.; Park, C.H.; Shim, K.B.; Kim, H.T.; Baek, I.Y.; Jang, D.S. Isolation and identification of phenolic compounds from the seeds of Perilla frutescens (L.) and their inhibitory activities against α-glucosidase and aldose reductase. Food Chem. 2012, 135, 1397–1403. [Google Scholar] [CrossRef] [PubMed]
- Nakajima, A.; Yamamoto, Y.; Yoshinaka, N.; Namba, M.; Matsuo, H.; Okuyama, T.; Yoshigai, E.; Okumura, T.; Nishizawa, M.; Ikeya, Y. A new flavanone and other flavonoids from green perilla leaf extract inhibit nitric oxide production in interleukin 1β-treated hepatocytes. Biosci. Biotechnol. Biochem. 2015, 79, 138–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, Y.K.; Yao, Y.Y.; Chang, Y.N. Characterization of Anthocyanins in Perilla frutescens var. acuta Extract by Advanced UPLC-ESI-IT-TOF-MSn Method and Their Anticancer Bioactivity. Molecules 2015, 20, 9155–9169. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ryu, J.H.; Son, H.J.; Lee, S.H.; Sohn, D.H. Two neolignans from Perilla frutescens and their inhibition of nitric oxide synthase and tumor necrosis factor-alpha expression in murine macrophage cell line RAW 264.7. Bioorg. Med. Chem. Lett. 2002, 12, 649–651. [Google Scholar] [CrossRef] [PubMed]
- Fujita, T.; Ohira, K.; Miyatake, K.; Nakano, Y.; Nakayama, M. Inhibitory effects of perillosides A and C, and related monoterpene glucosides on aldose reductase and their structure-activity relationships. Chem. Pharm. Bull. 1995, 43, 920–926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- You, C.X.; Yang, K.; Wu, Y.; Zhang, W.J.; Wang, Y.; Geng, Z.F.; Chen, H.P.; Jiang, H.Y.; Du, S.S.; Deng, Z.W.; et al. Chemical composition and insecticidal activities of the essential oil of Perilla frutescens (L.) Britt. aerial parts against two stored product insects. Eur. Food Res. Technol. 2014, 239, 481–490. [Google Scholar] [CrossRef]
- Oh, H.A.; Park, C.S.; Ahn, H.J.; Park, Y.S.; Kim, H.M. Effect of Perilla frutescens var. acuta Kudo and rosmarinic acid on allergic inflammatory reactions. Exp. Biol. Med. 2011, 236, 99–106. [Google Scholar] [CrossRef]
- Kamei, R.; Fujimura, T.; Matsuda, M.; Kakihara, K.; Hirakawa, N.; Baba, K.; Ono, K.; Arakawa, K.; Kawamoto, S. A flavanone derivative from the Asian medicinal herb (Perilla frutescens) potently suppresses IgE-mediated immediate hypersensitivity reactions. Biochem. Biophys. Res. Commun. 2017, 483, 674–679. [Google Scholar] [CrossRef]
- Takeda, H.; Tsuji, M.; Inazu, M.; Egashira, T.; Matsumiya, T. Rosmarinic acid and caffeic acid produce antidepressive-like effect in the forced swimming test in mice. Eur. J. Pharmacol. 2002, 449, 261–267. [Google Scholar] [CrossRef]
- Osakabe, N.; Yasuda, A.; Natsume, M.; Sanbongi, C.; Kato, Y.; Osawa, T.; Yoshikawa, T. Rosmarinic acid, a major polyphenolic component of Perilla frutescens, reduces lipopolysaccharide (LPS)-induced liver injury in D-galactosamine (D-GalN)-sensitized mice. Free Radic. Biol. Med. 2002, 33, 798–806. [Google Scholar] [CrossRef]
- Li, J.J.; Li, Z.; Gu, L.J.; Choi, K.J.; Kim, D.S.; Kim, H.K.; Sung, C.K. The promotion of hair regrowth by topical application of a Perilla frutescens extract through increased cell viability and antagonism of testosterone and dihydrotestosterone. J. Nat. Med. 2018, 72, 96–105. [Google Scholar] [CrossRef]
- Zhao, G.; Yao-Yue, C.; Qin, G.W.; Guo, L.H. Luteolin from Purple Perilla mitigates ROS insult particularly in primary neurons. Neurobiol. Aging 2012, 33, 176–186. [Google Scholar] [CrossRef]
- Tan, B.L.; Norhaizan, M.E.; Liew, W.P.; Sulaiman Rahman, H. Antioxidant and Oxidative Stress: A Mutual Interplay in Age-Related Diseases. Front. Pharmacol. 2018, 9, 1162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lin, C.S.; Kuo, C.L.; Wang, J.P.; Cheng, J.S.; Huang, Z.W.; Chen, C.F. Growth inhibitory and apoptosis inducing effect of Perilla frutescens extract on human hepatoma HepG2 cells. J. Ethnopharmacol. 2007, 112, 557–567. [Google Scholar] [CrossRef] [PubMed]
- Yamamoto, H.; Ogawa, T. Antimicrobial activity of perilla seed polyphenols against oral pathogenic bacteria. Biosci. Biotechnol. Biochem. 2002, 66, 921–924. [Google Scholar] [CrossRef]
- Wang, Z.X.; Lin, Q.Q.; Tu, Z.C.; Zhang, L. The influence of in vitro gastrointestinal digestion on the Perilla frutescens leaf extract: Changes in the active compounds and bioactivities. J. Food Biochem. 2020, 44, e13530. [Google Scholar] [CrossRef]
- Huang, J.P.; Wang, Y.J.; Tian, T.; Wang, L.; Yan, Y.; Huang, S.X. Tropane alkaloid biosynthesis: A centennial review. Nat. Prod. Rep. 2021, 38, 1634–1658. [Google Scholar] [CrossRef]
- Bharathi Priya, L.; Huang, C.Y.; Hu, R.M.; Balasubramanian, B.; Baskaran, R. An updated review on pharmacological properties of neferine—A bisbenzylisoquinoline alkaloid from Nelumbo nucifera. J. Food Biochem. 2021, 45, e13986. [Google Scholar] [CrossRef] [PubMed]
- Hu, X.; Li, D.; Chu, C.; Li, X.; Wang, X.; Jia, Y.; Hua, H.; Xu, F. Antiproliferative Effects of Alkaloid Evodiamine and Its Derivatives. Int. J. Mol. Sci. 2018, 19, 3403. [Google Scholar] [CrossRef] [Green Version]
- Ito, M.; Kiuchi, F.; Yang, L.L.; Honda, G. Perilla citriodora from Taiwan and its phytochemical characteristics. Biol. Pharm. Bull. 2000, 23, 359–362. [Google Scholar] [CrossRef]
- Lim, H.J.; Woo, K.W.; Lee, K.R.; Lee, S.K.; Kim, H.P. Inhibition of Proinflammatory Cytokine Generation in Lung Inflammation by the Leaves of Perilla frutescens and Its Constituents. Biomol. Ther. 2014, 22, 62–67. [Google Scholar] [CrossRef] [Green Version]
- Uemura, T.; Yashiro, T.; Oda, R.; Shioya, N.; Nakajima, T.; Hachisu, M.; Kobayashi, S.; Nishiyama, C.; Arimura, G.I. Intestinal Anti-Inflammatory Activity of Perillaldehyde. J. Agric. Food Chem. 2018, 66, 3443–3448. [Google Scholar] [CrossRef]
- Chu, L.; Li, C.; Li, Y.; Yu, Q.; Yu, H.; Li, C.; Meng, W.; Zhu, J.; Wang, Q.; Wang, C.; et al. Perillaldehyde Inhibition of cGAS Reduces dsDNA-Induced Interferon Response. Front. Immunol. 2021, 12, 655637. [Google Scholar] [CrossRef] [PubMed]
- Huang, R.; Wu, D.; Ji, Z.; Fan, B.; She, Y.; Zhang, X.; Duan, L.; Shen, Q. Characterization of a Group of 2,3-Oxidosqualene Cyclase Genes Involved in the Biosynthesis of Diverse Triterpenoids of Perilla frutescens. J. Agric. Food Chem. 2023, 71, 2523–2531. [Google Scholar] [CrossRef] [PubMed]
- Huang, M.T.; Ho, C.T.; Wang, Z.Y.; Ferraro, T.; Lou, Y.R.; Stauber, K.; Ma, W.; Georgiadis, C.; Laskin, J.D.; Conney, A.H. Inhibition of skin tumorigenesis by rosemary and its constituents carnosol and ursolic acid. Cancer Res. 1994, 54, 701–708. [Google Scholar] [PubMed]
- Banno, N.; Akihisa, T.; Tokuda, H.; Yasukawa, K.; Higashihara, H.; Ukiya, M.; Watanabe, K.; Kimura, Y.; Hasegawa, J.; Nishino, H. Triterpene acids from the leaves of Perilla frutescens and their anti-inflammatory and antitumor-promoting effects. Biosci. Biotechnol. Biochem. 2004, 68, 85–90. [Google Scholar] [CrossRef] [Green Version]
- Cho, J.; Tremmel, L.; Rho, O.; Camelio, A.M.; Siegel, D.; Slaga, T.J.; DiGiovanni, J. Evaluation of pentacyclic triterpenes found in Perilla frutescens for inhibition of skin tumor promotion by 12-O-tetradecanoylphorbol-13-acetate. Oncotarget 2015, 6, 39292–39306. [Google Scholar] [CrossRef] [Green Version]
- Yang, S.Y.; Hong, C.O.; Lee, G.P.; Kim, C.T.; Lee, K.W. The hepatoprotection of caffeic acid and rosmarinic acid, major compounds of Perilla frutescens, against t-BHP-induced oxidative liver damage. Food Chem. Toxicol. 2013, 55, 92–99. [Google Scholar] [CrossRef]
- Komatsu, K.I.; Takanari, J.; Maeda, T.; Kitadate, K.; Sato, T.; Mihara, Y.; Uehara, K.; Wakame, K. Perilla leaf extract prevents atopic dermatitis induced by an extract of Dermatophagoides farinae in NC/Nga mice. Asian Pac. J. Allergy Immunol. 2016, 34, 272–277. [Google Scholar] [CrossRef] [Green Version]
- Jeon, I.H.; Kim, H.S.; Kang, H.J.; Lee, H.S.; Jeong, S.I.; Kim, S.J.; Jang, S.I. Anti-inflammatory and antipruritic effects of luteolin from Perilla (P. frutescens L.) leaves. Molecules 2014, 19, 6941–6951. [Google Scholar] [CrossRef] [Green Version]
- Gaihre, Y.R.; Iwamoto, A.; Oogai, S.; Hamajima, H.; Tsuge, K.; Nagata, Y.; Yanagita, T. Perilla pomace obtained from four different varieties have different levels and types of polyphenols and anti-allergic activity. Cytotechnology 2022, 74, 341–349. [Google Scholar] [CrossRef]
- Choi, E.M. Luteolin protects osteoblastic MC3T3-E1 cells from antimycin A-induced cytotoxicity through the improved mitochondrial function and activation of PI3K/Akt/CREB. Toxicol. In Vitro 2011, 25, 1671–1679. [Google Scholar] [CrossRef]
- Fujiwara, Y.; Kono, M.; Ito, A.; Ito, M. Anthocyanins in perilla plants and dried leaves. Phytochemistry 2018, 147, 158–166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, J.; He, Y.; Yu, D.; Jin, L.; Gong, X.; Zhang, B. Perilla oil regulates intestinal microbiota and alleviates insulin resistance through the PI3K/AKT signaling pathway in type-2 diabetic KKAy mice. Food Chem. Toxicol. 2020, 135, 110965. [Google Scholar] [CrossRef]
- Park, H.; Sa, K.J.; Hyun, D.Y.; Lee, S.; Lee, J.K. Identifying SSR Markers Related to Seed Fatty Acid Content in Perilla Crop (Perilla frutescens L.). Plants 2021, 10, 1404. [Google Scholar] [CrossRef]
- Hamazaki, K. Role of Omega-3 Polyunsaturated Fatty Acids in Mental Health—Studies from Japan. J. Oleo Sci. 2019, 68, 511–515. [Google Scholar] [CrossRef]
- Thomas, S.S.; Cha, Y.S.; Kim, K.A. Protective Effect of Diet-Supplemented and Endogenously Produced Omega-3 Fatty Acids against HFD-Induced Colon Inflammation in Mice. Foods 2022, 11, 2124. [Google Scholar] [CrossRef]
- Liu, H.; Wang, F.; Liu, X.; Xie, Y.; Xia, H.; Wang, S.; Sun, G. Effects of marine-derived and plant-derived omega-3 polyunsaturated fatty acids on erythrocyte fatty acid composition in type 2 diabetic patients. Lipids Health Dis. 2022, 21, 20. [Google Scholar] [CrossRef]
- Wang, F.; Zhu, H.; Hu, M.; Wang, J.; Xia, H.; Yang, X.; Yang, L.; Sun, G. Perilla Oil Supplementation Improves Hypertriglyceridemia and Gut Dysbiosis in Diabetic KKAy Mice. Mol. Nutr. Food Res. 2018, 62, e1800299. [Google Scholar] [CrossRef] [Green Version]
- Kangwan, N.; Pratchayasakul, W.; Kongkaew, A.; Pintha, K.; Chattipakorn, N.; Chattipakorn, S.C. Perilla Seed Oil Alleviates Gut Dysbiosis, Intestinal Inflammation and Metabolic Disturbance in Obese-Insulin-Resistant Rats. Nutrients 2021, 13, 3141. [Google Scholar] [CrossRef] [PubMed]
- Kim, T.J.; Park, J.G.; Kim, H.Y.; Ha, S.H.; Lee, B.; Park, S.U.; Seo, W.D.; Kim, J.K. Metabolite Profiling and Chemometric Study for the Discrimination Analyses of Geographic Origin of Perilla (Perilla frutescens) and Sesame (Sesamum indicum) Seeds. Foods 2020, 9, 989. [Google Scholar] [CrossRef] [PubMed]
- Torri, L.; Bondioli, P.; Folegatti, L.; Rovellini, P.; Piochi, M.; Morini, G. Development of Perilla seed oil and extra virgin olive oil blends for nutritional, oxidative stability and consumer acceptance improvements. Food Chem. 2019, 286, 584–591. [Google Scholar] [CrossRef]
- Lee, H.; Sung, J.; Kim, Y.; Jeong, H.S.; Lee, J. Protective Effects of Unsaponifiable Matter from Perilla Seed Meal on UVB-induced Damages and the Underlying Mechanisms in Human Skin Fibroblasts. Antioxidants 2019, 8, 644. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, T.N.; Tam, L.T.; Pham Thi Mai, H.; Tran Thi Hong, H.; Ninh, T.N.; Cuong, D.V.; Nguyen Xuan, C.; Tran, H.Q. Antimicrobial secondary metabolites from the aerial parts of Perilla frutescens. Nat. Prod. Res. 2022, 2022, 1–9. [Google Scholar] [CrossRef]
- Liu, Y.; Li, D.; Wei, Y.; Ma, Y.; Wang, Y.; Huang, L.; Wang, Y. Hydrolyzed peptides from purple perilla (Perilla frutescens L. Britt.) seeds improve muscle synthesis and exercise performance in mice. J. Food Biochem. 2020, 44, e13461. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Hu, L.; Cai, T.; Chen, Q.; Ma, Q.; Yang, J.; Meng, C.; Hong, J. Purification and identification of two novel antioxidant peptides from perilla (Perilla frutescens L. Britton) seed protein hydrolysates. PLoS ONE 2018, 13, e0200021. [Google Scholar] [CrossRef] [Green Version]
- Ahmed, H.M.; Tavaszi-Sarosi, S. Identification and quantification of essential oil content and composition, total polyphenols and antioxidant capacity of Perilla frutescens (L.) Britt. Food Chem. 2019, 275, 730–738. [Google Scholar] [CrossRef]
- Ito, M.; Honda, G.; Sydara, K. Perilla frutescens var. frutescens in northern Laos. J. Nat. Med. 2008, 62, 251–258. [Google Scholar] [CrossRef] [PubMed]
- Bumblauskiené, L.; Jakstas, V.; Janulis, V.; Mazdzieriené, R.; Ragazinskiené, O. Preliminary analysis on essential oil composition of Perilla L. cultivated in Lithuania. Acta Pol. Pharm. 2009, 66, 409–413. [Google Scholar] [PubMed]
- Zhou, X.J.; Yan, L.L.; Yin, P.P.; Shi, L.L.; Zhang, J.H.; Liu, Y.J.; Ma, C. Structural characterisation and antioxidant activity evaluation of phenolic compounds from cold-pressed Perilla frutescens var. arguta seed flour. Food Chem. 2014, 164, 150–157. [Google Scholar] [CrossRef]
- Meng, L.; Lozano, Y.F.; Gaydou, E.M.; Li, B. Antioxidant activities of polyphenols extracted from Perilla frutescens varieties. Molecules 2008, 14, 133–140. [Google Scholar] [CrossRef] [Green Version]
- Yamazaki, M.; Nakajima, J.; Yamanashi, M.; Sugiyama, M.; Makita, Y.; Springob, K.; Awazuhara, M.; Saito, K. Metabolomics and differential gene expression in anthocyanin chemo-varietal forms of Perilla frutescens. Phytochemistry 2003, 62, 987–995. [Google Scholar] [CrossRef]
- Liu, Y.; Liu, X.-H.; Zhou, S.; Gao, H.; Li, G.-L.; Guo, W.-J.; Fang, X.-Y.; Wang, W. Perillanolides A and B, new monoterpene glycosides from the leaves of Perilla frutescens. Rev. Bras. Farmacogn. 2017, 27, 564–568. [Google Scholar] [CrossRef]
- Razgonova, M.P.; Kon’kova, N.G.; Zakharenko, A.M.; Golokhvast, K.S. Polyphenols of Perilla frutescens of the family Lamiaceae identified by tandem mass spectrometry. Vavilovskii Zhurnal Genet. Sel. 2022, 26, 637–644. [Google Scholar] [CrossRef]
- Masahiro, T.; Risa, M.; Harutaka, Y.; Kazuhiro, C. Novel Antioxidants Isolated from Perilla frutescens Britton var. crispa (Thunb.). Biosci. Biotechnol. Biochem. 1996, 60, 1093–1095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, R.L.; Zhang, J.; Mou, Z.L.; Hao, S.L.; Zhang, Z.Q. Microwave-assisted one-step extraction-derivatization for rapid analysis of fatty acids profile in herbal medicine by gas chromatography-mass spectrometry. Analyst 2012, 137, 5135–5143. [Google Scholar] [CrossRef] [PubMed]
- Asif, M. Health effects of omega-3,6,9 fatty acids: Perilla frutescens is a good example of plant oils. Orient. Pharm. Exp. Med. 2011, 11, 51–59. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.K.; Park, S.Y.; Na, J.K.; Seong, E.S.; Yu, C.Y. Metabolite profiling based on lipophilic compounds for quality assessment of perilla (Perilla frutescens) cultivars. J. Agric. Food Chem. 2012, 60, 2257–2263. [Google Scholar] [CrossRef]
- Lee, Y.H.; Kim, B.; Kim, S.; Kim, M.-S.; Kim, H.; Hwang, S.-R.; Kim, K.; Lee, J.H. Characterization of metabolite profiles from the leaves of green perilla (Perilla frutescens) by ultra high performance liquid chromatography coupled with electrospray ionization quadrupole time-of-flight mass spectrometry and screening for their antioxidant properties. J. Food Drug Anal. 2017, 25, 776–788. [Google Scholar] [CrossRef]
- Erhunmwunsee, F.; Pan, C.; Yang, K.; Li, Y.; Liu, M.; Tian, J. Recent development in biological activities and safety concerns of perillaldehyde from perilla plants: A review. Crit. Rev. Food Sci. Nutr. 2022, 62, 6328–6340. [Google Scholar] [CrossRef]
- Lee, J.H.; Cho, H.D.; Jeong, I.Y.; Lee, M.K.; Seo, K.I. Sensitization of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-resistant primary prostate cancer cells by isoegomaketone from Perilla frutescens. J. Nat. Prod. 2014, 77, 2438–2443. [Google Scholar] [CrossRef]
- Lim, D.Y.; Jeong, Y.; Tyner, A.L.; Park, J.H. Induction of cell cycle arrest and apoptosis in HT-29 human colon cancer cells by the dietary compound luteolin. Am. J. Physiol. Gastrointest. Liver Physiol. 2007, 292, G66–G75. [Google Scholar] [CrossRef] [Green Version]
- Tantipaiboonwong, P.; Chaiwangyen, W.; Suttajit, M.; Kangwan, N.; Kaowinn, S.; Khanaree, C.; Punfa, W.; Pintha, K. Molecular Mechanism of Antioxidant and Anti-Inflammatory Effects of Omega-3 Fatty Acids in Perilla Seed Oil and Rosmarinic Acid Rich Fraction Extracted from Perilla Seed Meal on TNF-α Induced A549 Lung Adenocarcinoma Cells. Molecules 2021, 26, 6757. [Google Scholar] [CrossRef]
- Jeong, J.H.; Park, H.J.; Chi, G.Y.; Choi, Y.H.; Park, S.H. An Ethanol Extract of Perilla frutescens Leaves Suppresses Adrenergic Agonist-Induced Metastatic Ability of Cancer Cells by Inhibiting Src-Mediated EMT. Molecules 2023, 28, 3414. [Google Scholar] [CrossRef]
- Park, D.D.; Yum, H.W.; Zhong, X.; Kim, S.H.; Kim, S.H.; Kim, D.H.; Kim, S.J.; Na, H.K.; Sato, A.; Miura, T.; et al. Perilla frutescens Extracts Protects against Dextran Sulfate Sodium-Induced Murine Colitis: NF-κB, STAT3, and Nrf2 as Putative Targets. Front. Pharmacol. 2017, 8, 482. [Google Scholar] [CrossRef] [Green Version]
- Onogi, N.; Okuno, M.; Komaki, C.; Moriwaki, H.; Kawamori, T.; Tanaka, T.; Mori, H.; Muto, Y. Suppressing effect of perilla oil on azoxymethane-induced foci of colonic aberrant crypts in rats. Carcinogenesis 1996, 17, 1291–1296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fares, J.; Fares, M.Y.; Khachfe, H.H.; Salhab, H.A.; Fares, Y. Molecular principles of metastasis: A hallmark of cancer revisited. Signal Transduct. Target. Ther. 2020, 5, 28. [Google Scholar] [CrossRef] [PubMed]
- Pastushenko, I.; Blanpain, C. EMT Transition States during Tumor Progression and Metastasis. Trends Cell Biol. 2019, 29, 212–226. [Google Scholar] [CrossRef] [Green Version]
- Kim-Fuchs, C.; Le, C.P.; Pimentel, M.A.; Shackleford, D.; Ferrari, D.; Angst, E.; Hollande, F.; Sloan, E.K. Chronic stress accelerates pancreatic cancer growth and invasion: A critical role for beta-adrenergic signaling in the pancreatic microenvironment. Brain Behav. Immun. 2014, 40, 40–47. [Google Scholar] [CrossRef] [Green Version]
- Lin, Z.; Huang, S.; LingHu, X.; Wang, Y.; Wang, B.; Zhong, S.; Xie, S.; Xu, X.; Yu, A.; Nagai, A.; et al. Perillaldehyde inhibits bone metastasis and receptor activator of nuclear factor-κB ligand (RANKL) signaling-induced osteoclastogenesis in prostate cancer cell lines. Bioengineered 2022, 13, 2710–2719. [Google Scholar] [CrossRef] [PubMed]
- Wong, R.S. Apoptosis in cancer: From pathogenesis to treatment. J. Exp. Clin. Cancer Res. 2011, 30, 87. [Google Scholar] [CrossRef] [Green Version]
- Kwak, Y.; Ju, J. Inhibitory activities of Perilla frutescens britton leaf extract against the growth, migration, and adhesion of human cancer cells. Nutr. Res. Pract. 2015, 9, 11–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, F.X.; Guan, K.L. The Hippo pathway: Regulators and regulations. Genes Dev. 2013, 27, 355–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, C.L.; Shin, Y.S.; Choi, S.H.; Oh, S.; Kim, K.; Jeong, H.S.; Mo, J.S. Extracts of Perilla frutescens var. Acuta (Odash.) Kudo Leaves Have Antitumor Effects on Breast Cancer Cells by Suppressing YAP Activity. Evid. Based Complement. Alternat. Med. 2021, 2021, 5619761. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Peng, Y.; Wei, W. Cell cycle on the crossroad of tumorigenesis and cancer therapy. Trends Cell Biol. 2022, 32, 30–44. [Google Scholar] [CrossRef] [PubMed]
- Abd El-Hafeez, A.A.; Fujimura, T.; Kamei, R.; Hirakawa, N.; Baba, K.; Ono, K.; Kawamoto, S. A methoxyflavanone derivative from the Asian medicinal herb (Perilla frutescens) induces p53-mediated G(2)/M cell cycle arrest and apoptosis in A549 human lung adenocarcinoma. Cytotechnology 2018, 70, 899–912. [Google Scholar] [CrossRef] [PubMed]
- Campisi, J. Aging, cellular senescence, and cancer. Annu. Rev. Physiol. 2013, 75, 685–705. [Google Scholar] [CrossRef] [Green Version]
- Jochems, F.; Thijssen, B.; De Conti, G.; Jansen, R.; Pogacar, Z.; Groot, K.; Wang, L.; Schepers, A.; Wang, C.; Jin, H.; et al. The Cancer SENESCopedia: A delineation of cancer cell senescence. Cell Rep. 2021, 36, 109441. [Google Scholar] [CrossRef]
- Maeda, A.; Fujimura, T.; Hirakawa, N.; Baba, K.; Kawamoto, S. A Methoxyflavanone from Perilla frutescens Induces Cellular Senescence in A549 Human Lung Adenocarcinoma Cells but Not in Normal Human Bronchial Epithelial Cells. Biol. Pharm. Bull. 2022, 45, 1581–1584. [Google Scholar] [CrossRef]
- Balkwill, F. Tumour necrosis factor and cancer. Nat. Rev. Cancer 2009, 9, 361–371. [Google Scholar] [CrossRef]
- Paradee, N.; Utama-Ang, N.; Uthaipibull, C.; Porter, J.B.; Garbowski, M.W.; Srichairatanakool, S. Extracts of Thai Perilla frutescens nutlets attenuate tumour necrosis factor-α-activated generation of microparticles, ICAM-1 and IL-6 in human endothelial cells. Biosci. Rep. 2020, 40, BSR20192110. [Google Scholar] [CrossRef]
- Jelic, M.D.; Mandic, A.D.; Maricic, S.M.; Srdjenovic, B.U. Oxidative stress and its role in cancer. J. Cancer Res. Ther. 2021, 17, 22–28. [Google Scholar] [CrossRef]
- Paradee, N.; Howes, M.R.; Utama-Ang, N.; Chaikitwattna, A.; Hider, R.C.; Srichairatanakool, S. A chemically characterized ethanolic extract of Thai Perilla frutescens (L.) Britton fruits (nutlets) reduces oxidative stress and lipid peroxidation in human hepatoma (HuH7) cells. Phytother. Res. 2019, 33, 2064–2074. [Google Scholar] [CrossRef]
- Zhang, Y.; Liu, S.; Feng, Q.; Huang, X.; Wang, X.; Peng, Y.; Zhao, Z.; Liu, Z. Perilaldehyde activates AMP-activated protein kinase to suppress the growth of gastric cancer via induction of autophagy. J. Cell. Biochem. 2019, 120, 1716–1725. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Huang, X.; Han, J.; Zheng, W.; Ma, W. Extract of Perilla frutescens inhibits tumor proliferation of HCC via PI3K/AKT signal pathway. Afr. J. Tradit. Complement. Altern. Med. 2013, 10, 251–257. [Google Scholar] [CrossRef] [PubMed]
- Hirose, M.; Masuda, A.; Ito, N.; Kamano, K.; Okuyama, H. Effects of dietary perilla oil, soybean oil and safflower oil on 7,12-dimethylbenz[a]anthracene (DMBA) and 1,2-dimethyl-hydrazine (DMH)-induced mammary gland and colon carcinogenesis in female SD rats. Carcinogenesis 1990, 11, 731–735. [Google Scholar] [CrossRef] [PubMed]
- Ueda, H.; Yamazaki, C.; Yamazaki, M. Inhibitory effect of Perilla leaf extract and luteolin on mouse skin tumor promotion. Biol. Pharm. Bull. 2003, 26, 560–563. [Google Scholar] [CrossRef] [Green Version]
- Kwon, S.J.; Lee, J.H.; Moon, K.D.; Jeong, I.Y.; Ahn, D.U.; Lee, M.K.; Seo, K.I. Induction of apoptosis by isoegomaketone from Perilla frutescens L. in B16 melanoma cells is mediated through ROS generation and mitochondrial-dependent, -independent pathway. Food Chem. Toxicol. 2014, 65, 97–104. [Google Scholar] [CrossRef]
- Osakabe, N.; Yasuda, A.; Natsume, M.; Yoshikawa, T. Rosmarinic acid inhibits epidermal inflammatory responses: Anticarcinogenic effect of Perilla frutescens extract in the murine two-stage skin model. Carcinogenesis 2004, 25, 549–557. [Google Scholar] [CrossRef]
- Okuno, M.; Tanaka, T.; Komaki, C.; Nagase, S.; Shiratori, Y.; Muto, Y.; Kajiwara, K.; Maki, T.; Moriwaki, H. Suppressive effect of low amounts of safflower and perilla oils on diethylnitrosamine-induced hepatocarcinogenesis in male F344 rats. Nutr. Cancer 1998, 30, 186–193. [Google Scholar] [CrossRef]
- Futakuchi, M.; Cheng, J.L.; Hirose, M.; Kimoto, N.; Cho, Y.M.; Iwata, T.; Kasai, M.; Tokudome, S.; Shirai, T. Inhibition of conjugated fatty acids derived from safflower or perilla oil of induction and development of mammary tumors in rats induced by 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP). Cancer Lett. 2002, 178, 131–139. [Google Scholar] [CrossRef]
- Cho, B.O.; Jin, C.H.; Park, Y.D.; Ryu, H.W.; Byun, M.W.; Seo, K.I.; Jeong, I.Y. Isoegomaketone induces apoptosis through caspase-dependent and caspase-independent pathways in human DLD1 cells. Biosci. Biotechnol. Biochem. 2011, 75, 1306–1311. [Google Scholar] [CrossRef] [Green Version]
- Kwon, S.J.; Lee, J.H.; Moon, K.D.; Jeong, I.Y.; Yee, S.T.; Lee, M.K.; Seo, K.I. Isoegomaketone induces apoptosis in SK-MEL-2 human melanoma cells through mitochondrial apoptotic pathway via activating the PI3K/Akt pathway. Int. J. Oncol. 2014, 45, 1969–1976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Renzulli, C.; Galvano, F.; Pierdomenico, L.; Speroni, E.; Guerra, M.C. Effects of rosmarinic acid against aflatoxin B1 and ochratoxin-A-induced cell damage in a human hepatoma cell line (Hep G2). J. Appl. Toxicol. 2004, 24, 289–296. [Google Scholar] [CrossRef] [PubMed]
- Asefy, Z.; Tanomand, A.; Hoseinnejhad, S.; Ceferov, Z.; Oshaghi, E.A.; Rashidi, M. Unsaturated fatty acids as a co-therapeutic agents in cancer treatment. Mol. Biol. Rep. 2021, 48, 2909–2916. [Google Scholar] [CrossRef] [PubMed]
- Freitas, R.D.S.; Campos, M.M. Protective Effects of Omega-3 Fatty Acids in Cancer-Related Complications. Nutrients 2019, 11, 945. [Google Scholar] [CrossRef] [Green Version]
- Narisawa, T.; Fukaura, Y.; Yazawa, K.; Ishikawa, C.; Isoda, Y.; Nishizawa, Y. Colon cancer prevention with a small amount of dietary perilla oil high in alpha-linolenic acid in an animal model. Cancer 1994, 73, 2069–2075. [Google Scholar] [CrossRef] [PubMed]
- Narisawa, T.; Takahashi, M.; Kotanagi, H.; Kusaka, H.; Yamazaki, Y.; Koyama, H.; Fukaura, Y.; Nishizawa, Y.; Kotsugai, M.; Isoda, Y.; et al. Inhibitory effect of dietary perilla oil rich in the n-3 polyunsaturated fatty acid alpha-linolenic acid on colon carcinogenesis in rats. Jpn. J. Cancer Res. 1991, 82, 1089–1096. [Google Scholar] [CrossRef]
- Abd El-Hafeez, A.A.; Fujimura, T.; Kamei, R.; Hirakawa, N.; Baba, K.; Ono, K.; Kawamoto, S. Synergistic tumor suppression by a Perilla frutescens-derived methoxyflavanone and anti-cancer tyrosine kinase inhibitors in A549 human lung adenocarcinoma. Cytotechnology 2018, 70, 913–919. [Google Scholar] [CrossRef]
- Komaki, C.; Okuno, M.; Onogi, N.; Moriwaki, H.; Kawamori, T.; Tanaka, T.; Mori, H.; Muto, Y. Synergistic suppression of azoxymethane-induced foci of colonic aberrant crypts by the combination of beta-carotene and perilla oil in rats. Carcinogenesis 1996, 17, 1897–1901. [Google Scholar] [CrossRef] [Green Version]
- Rao, C.V.; Patlolla, J.M.; Cooma, I.; Kawamori, T.; Steele, V.E. Prevention of familial adenomatous polyp development in APC min mice and azoxymethane-induced colon carcinogenesis in F344 Rats by ω-3 fatty acid rich perilla oil. Nutr. Cancer 2013, 65 (Suppl. S1), 54–60. [Google Scholar] [CrossRef]
- Sodir, N.M.; Evan, G.I. Finding cancer’s weakest link. Oncotarget 2011, 2, 1307–1313. [Google Scholar] [CrossRef] [Green Version]
- Rajabi, S.; Maresca, M.; Yumashev, A.V.; Choopani, R.; Hajimehdipoor, H. The Most Competent Plant-Derived Natural Products for Targeting Apoptosis in Cancer Therapy. Biomolecules 2021, 11, 534. [Google Scholar] [CrossRef] [PubMed]
- Wahle, K.W.; Brown, I.; Rotondo, D.; Heys, S.D. Plant phenolics in the prevention and treatment of cancer. Adv. Exp. Med. Biol. 2010, 698, 36–51. [Google Scholar] [CrossRef] [PubMed]
- Hu, Y.; Guo, N.; Yang, T.; Yan, J.; Wang, W.; Li, X. The Potential Mechanisms by which Artemisinin and Its Derivatives Induce Ferroptosis in the Treatment of Cancer. Oxid. Med. Cell. Longev. 2022, 2022, 1458143. [Google Scholar] [CrossRef] [PubMed]
- Aiello, P.; Sharghi, M.; Mansourkhani, S.M.; Ardekan, A.P.; Jouybari, L.; Daraei, N.; Peiro, K.; Mohamadian, S.; Rezaei, M.; Heidari, M.; et al. Medicinal Plants in the Prevention and Treatment of Colon Cancer. Oxid. Med. Cell. Longev. 2019, 2019, 2075614. [Google Scholar] [CrossRef] [Green Version]
- Ahmed, H.M. Ethnomedicinal, Phytochemical and Pharmacological Investigations of Perilla frutescens (L.) Britt. Molecules 2018, 24, 102. [Google Scholar] [CrossRef] [Green Version]
- Liu, S.; Jin, X.; Shang, Y.; Wang, L.; Du, K.; Chen, S.; Li, J.; He, J.; Fang, S.; Chang, Y. A comprehensive review of the botany, ethnopharmacology, phytochemistry, pharmacology, toxicity and quality control of Perillae Fructus. J. Ethnopharmacol. 2023, 304, 116022. [Google Scholar] [CrossRef]
- Wang, R.; Zhang, Q.; Feng, C.; Zhang, J.; Qin, Y.; Meng, L. Advances in the Pharmacological Activities and Effects of Perilla Ketone and Isoegomaketone. Evid. Based Complement. Alternat. Med. 2022, 2022, 8809792. [Google Scholar] [CrossRef]
- Swamy, M.K.; Sinniah, U.R.; Ghasemzadeh, A. Anticancer potential of rosmarinic acid and its improved production through biotechnological interventions and functional genomics. Appl. Microbiol. Biotechnol. 2018, 102, 7775–7793. [Google Scholar] [CrossRef]
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. |
© 2023 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
Huang, S.; Nan, Y.; Chen, G.; Ning, N.; Du, Y.; Lu, D.; Yang, Y.; Meng, F.; Yuan, L. The Role and Mechanism of Perilla frutescens in Cancer Treatment. Molecules 2023, 28, 5883. https://doi.org/10.3390/molecules28155883
Huang S, Nan Y, Chen G, Ning N, Du Y, Lu D, Yang Y, Meng F, Yuan L. The Role and Mechanism of Perilla frutescens in Cancer Treatment. Molecules. 2023; 28(15):5883. https://doi.org/10.3390/molecules28155883
Chicago/Turabian StyleHuang, Shicong, Yi Nan, Guoqing Chen, Na Ning, Yuhua Du, Doudou Lu, Yating Yang, Fandi Meng, and Ling Yuan. 2023. "The Role and Mechanism of Perilla frutescens in Cancer Treatment" Molecules 28, no. 15: 5883. https://doi.org/10.3390/molecules28155883
APA StyleHuang, S., Nan, Y., Chen, G., Ning, N., Du, Y., Lu, D., Yang, Y., Meng, F., & Yuan, L. (2023). The Role and Mechanism of Perilla frutescens in Cancer Treatment. Molecules, 28(15), 5883. https://doi.org/10.3390/molecules28155883