Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleExperimental Studies

Magnolol Suppresses ERK/NF-κB Signaling and Triggers Apoptosis Through Extrinsic/Intrinsic Pathways in Osteosarcoma

CHI-HUAN LI, MING-CHOU KU, KUN-CHING LEE, PO-FU YUEH, FEI-TING HSU, RONG-FONG LIN, CHUNG-CHI YANG, WEI-CHUN WANG, JIANN-HWA CHEN, LI-CHO HSU and YUAN-HAO LEE
Anticancer Research September 2022, 42 (9) 4403-4410; DOI: https://doi.org/10.21873/anticanres.15940
CHI-HUAN LI
1Department of Orthopedics, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
MING-CHOU KU
2Department of Orthopedics, Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
KUN-CHING LEE
3Department of Radiation Oncology, National Yang Ming Chiao Tung University Hospital, Yilan, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
PO-FU YUEH
4Institute of Traditional Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, R.O.C.;
5Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
FEI-TING HSU
5Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
RONG-FONG LIN
6Department of Optometry, Central Taiwan University of Science and Technology, Taichung, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
CHUNG-CHI YANG
7Division of Cardiovascular Medicine, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan, R.O.C.;
8Cardiovascular Division, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
WEI-CHUN WANG
9Department of Oral and Maxillofacial Surgery, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
JIANN-HWA CHEN
10Department of Emergency Medicine, Cathay General Hospital, Taipei, Taiwan, R.O.C.;
11School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: cgh08335{at}cgh.org.tw hsulc{at}ymuh.ym.edu.tw lyuanhao{at}mail.cmu.edu.tw
LI-CHO HSU
12Department of Medicine, National Yang-Ming Chiao-Tung University Hospital, Yilan, Taiwan, R.O.C.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: cgh08335{at}cgh.org.tw hsulc{at}ymuh.ym.edu.tw lyuanhao{at}mail.cmu.edu.tw
YUAN-HAO LEE
13Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan, R.O.C.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: cgh08335{at}cgh.org.tw hsulc{at}ymuh.ym.edu.tw lyuanhao{at}mail.cmu.edu.tw
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: Osteosarcoma is an aggressive primary malignant bone tumor that occurs in childhood. Although the diagnostic and treatment options have been improved, osteosarcoma confers poor prognosis. Magnolol, an active component of Magnoliae officinalis cortex, has been widely applied in herb medicine and has been shown to have multiple pharmacological activities. However, whether magnolol possesses anti-osteosarcoma capacity remains unknown. Materials and Methods: We examined magnolol is cytotoxicity, and whether it regulates apoptosis and oncogene expression using MTT, flow cytometry and Western blotting assays in osteosarcoma cells. Results: Magnolol exerted toxicity towards U-2 OS cells by inducing intrinsic/extrinsic apoptosis pathways. Additionally, treatment of U-2 OS cells with magnolol inhibited MAPK1 mitogen-activated protein kinase 1 (ERK)/Nuclear factor kappa B (NF-Embedded ImageB) signaling involved in tumor progression and reduced the expression of anti-apoptotic and metastasis-associated genes. Conclusion: Magnolol may induce apoptosis and inactivate ERK/NF-Embedded ImageB signal transduction in osteosarcoma cells.

Key Words:
  • Magnolol
  • ERK
  • NF-Embedded ImageB
  • osteosarcoma
  • apoptosis

Osteosarcoma (OS), the most common form of malignant bone tumor, often occurs in children, adolescents, and adults older than 50 years (1, 2). Chemotherapy induces DNA damage and interferes with DNA metabolism resulting in cell cycle arrest, cellular senescence, and apoptosis in cancers (3). Chemoresistance and metastasis are major causes of treatment failure in OS (4-6). Mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) is a crucial mediator of malignant phenotypes of OS, including growth, survival, angiogenesis, invasion, metastasis, and chemoresistance through upregulating of oncogenic transcriptional factors. Inhibition of ERK signaling has been reported to suppress OS but also to enhance the anti-OS efficacy of chemotherapy (7, 8). Therefore, ERK has been considered as a promising therapeutic target for the treatment of OS.

Several multikinase inhibitors have been indicated to prolong the survival of patients with relapsed and unresectable high-grade OS after failure of chemotherapy. Both sorafenib and regorafenib are oral multikinase inhibitor that show positive effects on overall survival and progression-free survival in patients with OS (9). Sorafenib and regorafenib induce apoptosis, inhibit growth, reduce anti-apoptotic signaling, and impair invasion ability in OS cells. Suppression of ERK signaling was associated with either sorafenib- or regorafenib-mediated inhibition of OS progression (10, 11).

Natural compounds isolated from medicinal plants exert anti-cancer functions in OS down-regulation of ERK activation (12-14). For instance, amentoflavone, a biflavonoid compound found in Selaginella tamariscina, blocked ERK signaling and inhibited the growth and invasion capacity of OS in vitro and in vivo (2, 15). Magnolol, a bioactive compound of the medicinal plant Magnolia officinalis, alleviated ERK-mediated osteoclastogenesis and resulted in the suppression of ovariectomy induced bone loss (16). In addition, magnolol also induced apoptosis through G0/G1 phase arrest and p53-mediated intrinsic pathway in OS cells (17). However, the anti-OS effect of magnolol has not yet been fully understood. The major goal of this study was to evaluate the anticancer effect and action mechanism of magnolol on OS cells.

Materials and Methods

Reagents and antibodies. Magnolol was purchased from Wuhan ChemFaces Biochemical Co., Ltd. (Wuhan, Hubei, PR China). Dimethyl sulfoxide (DMSO), crystal violet, and 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) were purchased from Sigma Chemical Co. (St. Louis, MO, USA). McCoy’s 5A medium, fetal bovine serum (FBS), and penicillin-streptomycin (PS) were all purchased from GIBCO®/Invitrogen Life Technologies (Carlsbad, CA, USA). Primary antibodies against myeloid cell leukemia 1 (MCL-1) (D35A5) [1:1,000, Cell Signaling Technology (CST), Danvers, MA, USA], X-linked inhibitor of apoptosis protein (XIAP, 1:1,000, CST), cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein (c-FLIP, 1:1,000, CST), matrix metallopeptidase 2 (MMP2, 1:1,000, Invitrogen), matrix metallopeptidase 9 (MMP9, PA5-13199, 1:1,000, Invitrogen), vascular endothelial growth factor (VEGF, ab46154, 1:1,000, Abcam, Cambridge, UK), urokinase-type plasminogen activator (uPA, 1:1,000, GenTex, Irvine, CA, USA), glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (D16H11) XP® (1:1,000, CST), extracellular signal-regulated kinase (ERK) Thr202/Try204 (1:1,000, CST), ERK (1:1,000, CST), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-Embedded ImageB) Ser536 (93H1) (1:1,000, CST), and NF-Embedded ImageB (D14E12) XP® (1:1,000, CST) were purchased from different companies as listed. Secondary antibodies for western blotting, including peroxidase affiniPure Goat Anti-Mouse IgG and Goat Anti-Rabbit IgG were all obtained from Jackson Immunoresearch Laboratories Inc. (West Grove, PA, USA).

Cell culture. U-2 OS cells were purchased from Bioresource Collection and Research Center (BCRC, Hsinchu, Taiwan, ROC), and maintained in complete McCoy’s 5A medium (10% Fetal Bovine Serum, 1% Penicillin-Streptomycin). Cells were subcultured following trypsinization using 1× trypsin EDTA solution C (SARTORIUS, Göttingen, Germany), collected, resuspended to adequate plates, and incubated in a 37°C humidified incubator in a 5% CO2 and 95% air mixture (2).

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. U-2 OS cells were plated in 96 well plates at 5×103 cells per well and incubated overnight. Then cells were treated with various concentrations of magnolol (0-200 μM) for 24 or 48 h. After treatment, the medium was discarded and replaced by MTT solution (0.5 mg/ml MTT in PBS) for another 2 h incubation at 37°C. The MTT solution was removed, 100 μl DMSO were added in each well and the optical density (OD) was measured at 570 mm absorbance wavelength. The blank value was defined as baseline (+/−0.01) (18).

Transwell assay (migration and invasion). U-2 OS cells were plated in 6 well plates at 5×104 cells per well and incubated overnight. Then, cells were treated with various concentrations of magnolol (0, 75, 100 μM) for 48 h. For the migration test, 8 μm pore transwells (BD Biosciences, Franklin Lakes, NJ, USA) were inserted in 24-well plate. For the invasion test, the upper inserts of transwells were coated with a matrigel mixture (matrigel:medium=1:1), placed in the 24 well plate and incubated in 37°C. The treated cells were then trypsinized, seeded at 5×104 cells (in serum-free medium) in the upper chambers of the transwells and the bottom chambers were filled with 500 μl complete medium and incubated for another 24 h. After incubation, transwells were fixed with fixation solution (methanol: acetic=3:1) and the cells were stained with 0.1% crystal violet. Bright images were captured using a light microscope. The density of migration and invasion cells were calculated using Image J software version 1.50 (National Institutes of Health, Bethesda, MD, USA) (19).

Flow cytometry. U-2 OS cells were plated in 6 well plates at a density of 5×104 cells per well and incubated overnight. Then, the cells were treated with various concentrations of magnolol (0, 75, 100, 125 μM) for 48 h. After treatment, cells were trypsinized, stained by FITC-DEVD-FMK (cleaved caspase-3), FITC-IETD-FMK (cleaved caspase-8), FITC-VAD-FMK (cleaved caspase-9), CD95 (Fas) Alexa fluor™ 488 (#13-0951-85, Invitrogen), CD178 (Fas-L) PE (#12-9919-42, Invitrogen), PARP-1 (cleaved Asp214) Alexa fluor™ 488 (#53-6668-42, Invitrogen), DiOC6 (3,3′-Dihexyloxacarbocyanine Iodide, ab189808, abcam) for MMP loss detection, and apoptosis analysis using an FITC Annexin V Apoptosis Detection Kit (BD Bioscience) for apoptosis detection, respectively. After cell staining processes, stained cells were washed by PBS and measured the fluorescence intensity by flow cytometry (NovoCyte, Agilent Technologies, ACEA US). The results were analyzed and quantified by FlowJo software (version 7.6.1; FlowJo LLC, Ashland, OR, USA) (20).

Western blotting. U-2 OS cells were plated in 6 well plates at a density of 5×104 cells per well and incubated overnight. Then, cells were treated with various concentrations of magnolol (0, 75, 100, 125 μM) for 48 h. The treated cells were lysed using RIPA lysis buffer containing proteinase and phosphatase inhibitors to collect whole cell protein on ice, and then cellular proteins were separated using 8-12% SDS-PAGE gel. The separated proteins were transferred onto polyvinylidene difluoride (PVDF) membranes and blocked with 5% non-fat milk for one hour at room temperature. The PVDF membranes were incubated with primary and secondary antibodies to detect target proteins and reacted with Immobilon Western Chemiluminescent HRP Substrate (Pierce, Rockford, IL, USA). The target protein band signals were detected using the UVP ChemiDoc-It™ image station (Analytik Jena, Jena, Germany) and quantified using the software VisionWorks (AnalytikJena) (21).

Statistical analysis. The comparison between untreated and treated group was calculated by one-way ANOVA using Microsoft excel 2016 version. A p-value less than 0.05 was considered as significant difference. Each value in this study is displayed as mean±standard error. Statistical difference symbols between groups are defined in each figure legend.

Results

Magnolol induced U-2 OS cell cytotoxicity and activated apoptosis pathways. To reveal the anti-osteosarcoma effect of magnolol, we first used the MTT assay to test the viability of human osteosarcoma U-2 OS cells after treatment with different concentrations of magnolol. It was demonstrated that magnolol significantly reduced U-2 OS cells viability at 125 and 200 μM after 24 h treatment, and markedly reduced cell viability at 75, 100, 125, and 200 μM after 48 h treatment (Figure 1A). We then further investigated whether magnolol-induced cytotoxicity is related to apoptosis induction. We stained cells with multiple cells apoptosis markers and evaluated their levels/activation by flow cytometry. As indicated in Figure 1B and C, fluorescence intensities of both cleaved-caspase-3 and cleaved-PARP-1 were increased after 75, 100, and 125 μM of magnolol treatment. Additionally, U-2 OS cells were double-stained with annexin-V (FITC) and PI (22), which is used to identify apoptosis status of cells. The result showed that 75, 100, and 125 μM of magnolol greatly induced U-2 OS annexin-V activation, which was interpreted as apoptosis induction (Figure 1D). These results indicated that magnolol may not only induce cytotoxicity of human osteosarcoma cells in a dose- and time-dependent manner, but also activated annexin-V to initiate apoptosis cell death.

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Magnolol induced cytotoxicity and activated apoptosis in U-2 OS cells. (A) Viability U-2 OS cells treated with magnolol is tested using the MTT assay. U-2 OS cells treated with 0, 75, 100, and 125 μM magnolol for 48 h are stained with (B) FITC-DEVD-FMK, (C) PARP-1 (cleaved Asp214) Alexa fluor™ 488, (D) and Annexin V Apoptosis detection kit for flow cytometry measurement (a1 p-value<0.05, a2 p-value<0.01. a3 p-value <0.005 vs. 0 μM magnolol).

Magnolol activated both the extrinsic and the intrinsic apoptosis pathways in U-2 OS cells. Next, whether magnolol may trigger the extrinsic and/or the intrinsic apoptosis pathways in U-2 OS cells were also tested. The levels of the mediator of the extrinsic apoptosis pathway, cleaved-caspase-8, were strongly increased by magnolol treatment (Figure 2A). The levels of cell death receptor and its ligand, Fas as well as Fas-L, were both effectively induced by magnolol (Figure 2B and C). However, the levels of the intrinsic pathway marker, cleaved-caspase-9, were also increased by magnolol treatment (Figure 2D). Furthermore, the mitochondrial membrane potential was lost by magnolol treatment (Figure 2E). Taken together, we found that magnolol-induced cytotoxicity of human osteosarcoma U-2 OS is correlated to the activation of the extrinsic and the intrinsic apoptosis pathways.

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Magnolol activated both the extrinsic and the intrinsic apoptosis pathways in U-2 OS cells. U-2 OS cells treated with 0, 75, 100, and 125 μM magnolol for 48h are stained with (A) FITC-IETD-FMK, (B) CD95 (Fas) Alexa fluor™ 488, (C) CD178 (Fas-L) PE, (D) FITC-VAD-FMK, and (E) DiOC6 for flow cytometry measurement (a1 p-value <0.05, a2 p-value <0.01, a3 p-value <0.005 vs. 0 μM magnolol).

Magnolol suppressed U-2 OS cell migration, invasion ability and tumor progression related protein expression. In addition, we performed western blotting to examine the regulation of various oncogene proteins in U-2 OS cells after magnolol treatment, including anti-apoptosis markers (MCL-1, XIAP, and C-FLIP), angiogenesis markers (VEGF), and migration/invasion related markers (MMP2, MMP9, and uPA). The levels of all the above-mentioned proteins were decreased by magnolol in a dose-dependent manner (Figure 3A). Furthermore, we used 8 μm transwell assays to identify whether the migration and invasion abilities of U-2 OS cells were inhibited by magnolol treatment. The results demonstrated that magnolol may inhibit U-2 OS cells migration and invasion capacity (Figure 3B). Taken together, magnolol may effectively suppress tumor progression associated protein expression and thus inhibit the growth and invasion/migration potential of U-2 OS cells.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Magnolol suppressed U-2 OS cell migration and invasion and the expression of tumor progression related proteins. (A) U-2 OS cells are treated with 0, 75, 100, and 125 μM of magnolol for 48 h and expression of several oncogenic proteins (MCL-1, XIAP, C-FLIP, MMP-2, MMP-9, VEGF, uPA) and of housekeeping gene GAPDH were examined using western blotting. (B) U-2 OS cells are pre-treated with 0, 75, and 100 μM magnolol for 48 h, and their migration/invasion abilities are examined using transwell assays (a3 p-value <0.005 vs. 0 μM magnolol).

Magnolol inhibited U-2 OS cell is associated with the inactivaiton of ERK/NF-Embedded ImageB pathway. To investigate the upstream regulator of these oncogenes, we further examined the phosphorylation of several key regulators of tumor progression. Multiple studies have demonstrated that the phosphorylation of ERK and NF-Embedded ImageB in osteosarcoma will trigger their growth. Western blot analysis indicated that magnolol greatly suppressed the phosphorylation of ERK as well as the phosphorylation of NF-Embedded ImageB (Figure 4). Hence, we suggest that magnolol inhibits U-2 OS cells growth via the inactivation of ERK/NF-Embedded ImageB pathway.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Magnolol inhibited the ERK/NF-Embedded ImageB pathway. U-2 OS cells are treated with 0, 75, 100, and 125 μM of magnolol for 48 h and the expression of ERK/NF-Embedded ImageB pathway related proteins, such as ERK (Thr202/Tyr204), ERK, NF-Embedded ImageB (Ser536), NF-Embedded ImageB and of the housekeeping gene GAPDH are assayed using western blotting.

Discussion

Anti-apoptotic proteins, such as MCL-1, XIAP, and C-FLIP, mediate acquired resistance of OS cells to chemotherapy and their inhibition sensitizes OS cells to methotrexate and cisplatin (23-25). Ideal apoptotic inducers restrain tumor growth and survival through effective induction of apoptosis and down-regulation of anti-apoptotic protein expression (26). Our results showed magnolol triggered apoptosis while reduced protein levels of MCL-1, XIAP, and C-FLIP in U-2 OS cells (Figure 1B-D and Figure 3A). Zhou et al. presented that magnolol induced apoptosis through the p53-mediated intrinsic apoptosis pathway (17). In addition to the stimulation of intrinsic apoptotic signaling, we also found that extrinsic apoptotic signaling such as activation of Fas, FasL, and cleaved-caspase-8 was effectively triggered by treatment with magnolol (Figure 2).

Metastasis-associated proteins such as MMP-9, MMP-2, and VEGF mediate extracellular matrix degradation and angiogenesis, which enhance tumor metastasis. Furthermore, their over-expression is associated with metastasis and worse survival of patients with OS (1, 20, 27, 28). Our results indicated that magnolol triggered inhibition of metastasis-associated proteins and eliminated the invasion ability of U-2 OS cells (Figure 3). Activation of NF-Embedded ImageB, an oncogenic transcription factor, may be involved in the expression of anti-apoptotic and metastasis-associated proteins. NF-Embedded ImageB is constitutively activated by upstream kinases, such as AKT and MAPKs, in cancers (29, 30). Previous studies showed that PD98059 (the ERK inhibitor) and QNZ (the NF-Embedded ImageB inhibitor) effectively reduced endogenous NF-Embedded ImageB signaling, and the above-mentioned antiapoptotic and metastasis-associated protein expression in OS cells (2, 11). Our data indicated that phosphorylation of both ERK and NF-Embedded ImageB was abolished by treatment with magnolol (Figure 4).

In conclusion, the extrinsic and intrinsic pathways participated in magnolol-induced apoptosis. In addition, magnolol also suppressed ERK/NF-Embedded ImageB-mediated anti-apoptosis and invasion/migration. We suggested that the induction of apoptosis and the suppression of ERK/NF-Embedded ImageB signaling are associated with magnolol-inhibited growth, survival, and invasion/migration of OS cells.

Acknowledgements

The Authors thank the Medical Research Core Facilities Center, and the Office of Research and Development at China Medical University (Taichung, Taiwan, ROC) for their technical support. This study was also supported by the following institutes: National Yang Ming Chiao Tung University Hospital, Yilan, Taiwan (ID: RD2022-010 and RD2022-021), Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan (ID: BRD-109028), Cathay General Hospital, Taipei, Taiwan (ID: CGH-MR-A11026).

Footnotes

  • ↵*,# These Authors contributed equally to this study.

  • Authors’ Contributions

    CHL, MCK, KCL, PFY and RFL performed all of the experiments. CHL, FTH, RFL, CCY and WCW prepared the first draft of the article. FTH, JHC and YHL participate in the design of this study, performed the literature review, and prepared the final versions of the article.

  • Conflicts of Interest

    The Authors declare no competing financial interests regarding this study.

  • Received June 23, 2022.
  • Revision received July 12, 2022.
  • Accepted July 13, 2022.
  • Copyright © 2022 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.

References

  1. ↵
    1. Chang PY,
    2. Hsieh MJ,
    3. Hsieh YS,
    4. Chen PN,
    5. Yang JS,
    6. Lo FC,
    7. Yang SF and
    8. Lu KH
    : Tricetin inhibits human osteosarcoma cells metastasis by transcriptionally repressing MMP-9 via p38 and Akt pathways. Environ Toxicol 32(8): 2032-2040, 2017. PMID: 27860196. DOI: 10.1002/tox.22380
    OpenUrlCrossRefPubMed
  2. ↵
    1. Pan PJ,
    2. Tsai JJ and
    3. Liu YC
    : Amentoflavone inhibits metastatic potential through suppression of ERK/NF-Embedded ImageB activation in osteosarcoma U2OS cells. Anticancer Res 37(9): 4911-4918, 2017. PMID: 28870912. DOI: 10.21873/anticanres.11900
    OpenUrlAbstract/FREE Full Text
  3. ↵
    1. van den Boogaard WMC,
    2. Komninos DSJ and
    3. Vermeij WP
    : Chemotherapy side-effects: Not all DNA damage is equal. Cancers (Basel) 14(3): 627, 2022. PMID: 35158895. DOI: 10.3390/cancers14030627
    OpenUrlCrossRefPubMed
  4. ↵
    1. Zhao J,
    2. Zhao Y,
    3. Ma X,
    4. Feng H and
    5. Jia L
    : Outstanding prognostic value of novel ferroptosis-related genes in chemoresistance osteosarcoma patients. Sci Rep 12(1): 5029, 2022. PMID: 35322804. DOI: 10.1038/s41598-022-09080-5
    OpenUrlCrossRefPubMed
    1. Marchandet L,
    2. Lallier M,
    3. Charrier C,
    4. Baud’huin M,
    5. Ory B and
    6. Lamoureux F
    : Mechanisms of resistance to conventional therapies for osteosarcoma. Cancers (Basel) 13(4): 683, 2021. PMID: 33567616. DOI: 10.3390/cancers13040683
    OpenUrlCrossRefPubMed
  5. ↵
    1. Yoshizawa M,
    2. Nakamura S,
    3. Sugiyama Y,
    4. Tamai S,
    5. Ishida Y,
    6. Sueyoshi M,
    7. Toda Y,
    8. Hosogi S,
    9. Yano Y and
    10. Ashihara E
    : 6-Hydroxythiobinupharidine inhibits migration of LM8 osteosarcoma cells by decreasing expression of LIM domain kinase 1. Anticancer Res 39(12): 6507-6513, 2019. PMID: 31810915. DOI: 10.21873/anticanres.13865
    OpenUrlAbstract/FREE Full Text
  6. ↵
    1. Chandhanayingyong C,
    2. Kim Y,
    3. Staples JR,
    4. Hahn C and
    5. Lee FY
    : MAPK/ERK signaling in osteosarcomas, ewing sarcomas and chondrosarcomas: Therapeutic implications and future directions. Sarcoma 2012: 404810, 2012. PMID: 22577336. DOI: 10.1155/2012/404810
    OpenUrlCrossRefPubMed
  7. ↵
    1. Noh K,
    2. Kim KO,
    3. Patel NR,
    4. Staples JR,
    5. Minematsu H,
    6. Nair K and
    7. Lee FY
    : Targeting inflammatory kinase as an adjuvant treatment for osteosarcomas. J Bone Joint Surg Am 93(8): 723-732, 2011. PMID: 21508279. DOI: 10.2106/JBJS.J.00302
    OpenUrlAbstract/FREE Full Text
  8. ↵
    1. Nakano K
    : Challenges of systemic therapy investigations for bone sarcomas. Int J Mol Sci 23(7): 3540, 2022. PMID: 35408900. DOI: 10.3390/ijms23073540
    OpenUrlCrossRefPubMed
  9. ↵
    1. Pignochino Y,
    2. Grignani G,
    3. Cavalloni G,
    4. Motta M,
    5. Tapparo M,
    6. Bruno S,
    7. Bottos A,
    8. Gammaitoni L,
    9. Migliardi G,
    10. Camussi G,
    11. Alberghini M,
    12. Torchio B,
    13. Ferrari S,
    14. Bussolino F,
    15. Fagioli F,
    16. Picci P and
    17. Aglietta M
    : Sorafenib blocks tumour growth, angiogenesis and metastatic potential in preclinical models of osteosarcoma through a mechanism potentially involving the inhibition of ERK1/2, MCL-1 and ezrin pathways. Mol Cancer 8: 118, 2009. PMID: 20003259. DOI: 10.1186/1476-4598-8-118
    OpenUrlCrossRefPubMed
  10. ↵
    1. Pan PJ,
    2. Liu YC and
    3. Hsu FT
    : Protein kinase B and extracellular signal-regulated kinase inactivation is associated with regorafenib-induced inhibition of osteosarcoma progression in vitro and in vivo. J Clin Med 8(6): 900, 2019. PMID: 31238539. DOI: 10.3390/jcm8060900
    OpenUrlCrossRefPubMed
  11. ↵
    1. Tobeiha M,
    2. Rajabi A,
    3. Raisi A,
    4. Mohajeri M,
    5. Yazdi SM,
    6. Davoodvandi A,
    7. Aslanbeigi F,
    8. Vaziri M,
    9. Hamblin MR and
    10. Mirzaei H
    : Potential of natural products in osteosarcoma treatment: Focus on molecular mechanisms. Biomed Pharmacother 144: 112257, 2021. PMID: 34688081. DOI: 10.1016/j.biopha.2021.112257
    OpenUrlCrossRefPubMed
    1. Kuan LY,
    2. Chen WL,
    3. Chen JH,
    4. Hsu FT,
    5. Liu TT,
    6. Chen WT,
    7. Wang KL,
    8. Chen WC,
    9. Liu YC and
    10. Wang WS
    : Magnolol induces apoptosis and inhibits ERK-modulated metastatic potential in hepatocellular carcinoma cells. In Vivo 32(6): 1361-1368, 2018. PMID: 30348689. DOI: 10.21873/invivo.11387
    OpenUrlAbstract/FREE Full Text
  12. ↵
    1. Chen YS,
    2. Sun R,
    3. Chen WL,
    4. Yau YC,
    5. Hsu FT,
    6. Chung JG,
    7. Tsai CJ,
    8. Hsieh CL,
    9. Chiu YM and
    10. Chen JH
    : The in vivo radiosensitizing effect of magnolol on tumor growth of hepatocellular carcinoma. In Vivo 34(4): 1789-1796, 2020. PMID: 32606148. DOI: 10.21873/invivo.11973
    OpenUrlAbstract/FREE Full Text
  13. ↵
    1. Lee YJ,
    2. Chung JG,
    3. Chien YT,
    4. Lin SS and
    5. Hsu FT
    : Suppression of ERK/NF-Embedded ImageB activation is associated with amentoflavone-inhibited osteosarcoma progression in vivo. Anticancer Res 39(7): 3669-3675, 2019. PMID: 31262893. DOI: 10.21873/anticanres.13515
    OpenUrlAbstract/FREE Full Text
  14. ↵
    1. Fei WY,
    2. Huo Q,
    3. Zhao PQ,
    4. Qin LJ and
    5. Li T
    : Magnolol prevents ovariectomy induced bone loss by suppressing osteoclastogenesis via inhibition of the nuclear factor Embedded ImageB and mitogen activated protein kinase pathways. Int J Mol Med 43(4): 1669-1678, 2019. PMID: 30816431. DOI: 10.3892/ijmm.2019.4099
    OpenUrlCrossRefPubMed
  15. ↵
    1. Zhou S,
    2. Wen H and
    3. Li H
    : Magnolol induces apoptosis in osteosarcoma cells via G0/G1 phase arrest and p53-mediated mitochondrial pathway. J Cell Biochem 120(10): 17067-17079, 2019. PMID: 31155771. DOI: 10.1002/jcb.28968
    OpenUrlCrossRefPubMed
  16. ↵
    1. Su CM,
    2. Weng YS,
    3. Kuan LY,
    4. Chen JH and
    5. Hsu FT
    : Suppression of PKCδ/NF-Embedded ImageB signaling and apoptosis induction through extrinsic/intrinsic pathways are associated magnolol-inhibited tumor progression in colorectal cancer in vitro and in vivo. Int J Mol Sci 21(10): 3527, 2020. PMID: 32429376. DOI: 10.3390/ijms21103527
    OpenUrlCrossRefPubMed
  17. ↵
    1. Yueh PF,
    2. Lee YH,
    3. Fu CY,
    4. Tung CB,
    5. Hsu FT and
    6. Lan KL
    : Magnolol induces the extrinsic/intrinsic apoptosis pathways and inhibits STAT3 signaling-mediated invasion of glioblastoma cells. Life (Basel) 11(12): 1399, 2021. PMID: 34947930. DOI: 10.3390/life11121399
    OpenUrlCrossRefPubMed
  18. ↵
    1. Wu CH,
    2. Lin KH,
    3. Fu BS,
    4. Hsu FT,
    5. Tsai JJ,
    6. Weng MC and
    7. Pan PJ
    : Sorafenib induces apoptosis and inhibits NF-Embedded ImageB-mediated anti-apoptotic and metastatic potential in osteosarcoma cells. Anticancer Res 41(3): 1251-1259, 2021. PMID: 33788716. DOI: 10.21873/anticanres.14882
    OpenUrlAbstract/FREE Full Text
  19. ↵
    1. Yueh PF,
    2. Lee YH,
    3. Chiang IT,
    4. Chen WT,
    5. Lan KL,
    6. Chen CH and
    7. Hsu FT
    : Suppression of EGFR/PKC-δ/NF-Embedded ImageB signaling associated with imipramine-inhibited progression of non-small cell lung cancer. Front Oncol 11: 735183, 2021. PMID: 34765548. DOI: 10.3389/fonc.2021.735183
    OpenUrlCrossRefPubMed
  20. ↵
    1. Pearson GW
    : Control of invasion by epithelial-to-mesenchymal transition programs during metastasis. J Clin Med 8(5): 646, 2019. PMID: 31083398. DOI: 10.3390/jcm8050646
    OpenUrlCrossRefPubMed
  21. ↵
    1. Xu W,
    2. Li Z,
    3. Zhu X,
    4. Xu R and
    5. Xu Y
    : miR-29 family inhibits resistance to methotrexate and promotes cell apoptosis by targeting COL3A1 and MCL1 in osteosarcoma. Med Sci Monit 24: 8812-8821, 2018. PMID: 30518744. DOI: 10.12659/MSM.911972
    OpenUrlCrossRefPubMed
    1. Liu XG,
    2. Xu J,
    3. Li F,
    4. Li MJ and
    5. Hu T
    : Down-regulation of miR-377 contributes to cisplatin resistance by targeting XIAP in osteosarcoma. Eur Rev Med Pharmacol Sci 22(5): 1249-1257, 2018. PMID: 29565481. DOI: 10.26355/eurrev_201803_14465
    OpenUrlCrossRefPubMed
  22. ↵
    1. Rao-Bindal K,
    2. Rao CK,
    3. Yu L and
    4. Kleinerman ES
    : Expression of c-FLIP in pulmonary metastases in osteosarcoma patients and human xenografts. Pediatr Blood Cancer 60(4): 575-579, 2013. PMID: 23255321. DOI: 10.1002/pbc.24412
    OpenUrlCrossRefPubMed
  23. ↵
    1. Pfeffer CM and
    2. Singh ATK
    : Apoptosis: a target for anticancer therapy. Int J Mol Sci 19(2): 448, 2018. PMID: 29393886. DOI: 10.3390/ijms19020448
    OpenUrlCrossRefPubMed
  24. ↵
    1. Zhang C,
    2. Wang L,
    3. Xiong C,
    4. Zhao R,
    5. Liang H and
    6. Luo X
    : The role of vascular endothelial growth factor as a prognostic and clinicopathological marker in osteosarcoma: a systematic review and meta-analysis. J Orthop Surg Res 16(1): 738, 2021. PMID: 34963495. DOI: 10.1186/s13018-021-02888-3
    OpenUrlCrossRefPubMed
  25. ↵
    1. Tsagaraki I,
    2. Phenekos C,
    3. Tsilibary E and
    4. Tzinia A
    : Calcitonin-induced NF-Embedded ImageB activation up-regulates fibronectin expression in MG63 osteosarcoma cells. Anticancer Res 33(11): 4901-4906, 2013. PMID: 24222127.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    1. Rasmi RR,
    2. Sakthivel KM and
    3. Guruvayoorappan C
    : NF-Embedded ImageB inhibitors in treatment and prevention of lung cancer. Biomed Pharmacother 130: 110569, 2020. PMID: 32750649. DOI: 10.1016/j.biopha.2020.110569
    OpenUrlCrossRefPubMed
  27. ↵
    1. Kaltschmidt C,
    2. Banz-Jansen C,
    3. Benhidjeb T,
    4. Beshay M,
    5. Förster C,
    6. Greiner J,
    7. Hamelmann E,
    8. Jorch N,
    9. Mertzlufft F,
    10. Pfitzenmaier J,
    11. Simon M,
    12. Schulte Am Esch J,
    13. Vordemvenne T,
    14. Wähnert D,
    15. Weissinger F,
    16. Wilkens L and
    17. Kaltschmidt B
    : A role for NF-Embedded ImageB in organ specific cancer and cancer stem cells. Cancers (Basel) 11(5): 655, 2019. PMID: 31083587. DOI: 10.3390/cancers11050655
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Anticancer Research: 42 (9)
Anticancer Research
Vol. 42, Issue 9
September 2022
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Magnolol Suppresses ERK/NF-κB Signaling and Triggers Apoptosis Through Extrinsic/Intrinsic Pathways in Osteosarcoma
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
1 + 0 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
Magnolol Suppresses ERK/NF-κB Signaling and Triggers Apoptosis Through Extrinsic/Intrinsic Pathways in Osteosarcoma
CHI-HUAN LI, MING-CHOU KU, KUN-CHING LEE, PO-FU YUEH, FEI-TING HSU, RONG-FONG LIN, CHUNG-CHI YANG, WEI-CHUN WANG, JIANN-HWA CHEN, LI-CHO HSU, YUAN-HAO LEE
Anticancer Research Sep 2022, 42 (9) 4403-4410; DOI: 10.21873/anticanres.15940

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
Magnolol Suppresses ERK/NF-κB Signaling and Triggers Apoptosis Through Extrinsic/Intrinsic Pathways in Osteosarcoma
CHI-HUAN LI, MING-CHOU KU, KUN-CHING LEE, PO-FU YUEH, FEI-TING HSU, RONG-FONG LIN, CHUNG-CHI YANG, WEI-CHUN WANG, JIANN-HWA CHEN, LI-CHO HSU, YUAN-HAO LEE
Anticancer Research Sep 2022, 42 (9) 4403-4410; DOI: 10.21873/anticanres.15940
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgements
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Magnolol Suppresses Osteosarcoma Progression via Apoptosis Induction and EGFR/AKT Pathway Inactivation in a U-2 OS Xenograft Model
  • MMP-2 Regulation of Emmprin on Tumour Cells and CD73 on Fibroblasts During Tumour-Stromal Interaction
  • Accessing Apoptosis Induction and Metastasis Inhibition Effect of Magnolol on Triple Negative Breast Cancer In Vitro
  • Google Scholar

More in this TOC Section

  • Pulsed Electromagnetic Field Promotes Doxorubicin-induced Apoptosis by Increasing Caspase-2 Activation in MDA-MB-231 Breast Cancer Cells
  • Fibroblast Supernatants Modulate Treatment Responses in Human Papillomavirus Positive and Negative Oropharyngeal Cancer Cell Lines
  • Impact of Interleukin-12B Genotypes on Breast Cancer Risk
Show more Experimental Studies

Similar Articles

Keywords

  • magnolol
  • ERK
  • NF-B
  • Osteosarcoma
  • apoptosis
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire