1
|
Rayamajhi A, Gyawali N, Karki D, Pérez-Caltzontzin LE, Peña-Corona SI, Cortés H, Adhikari A, Leyva-Gómez G, Uprety Y, Habtemariam S, Kiyekbayeva L, Sharifi-Rad J. Magnolol and its semi-synthetic derivatives: a comprehensive review of anti-cancer mechanisms, pharmacokinetics, and future therapeutic potential. Discov Oncol 2025; 16:683. [PMID: 40335865 PMCID: PMC12058641 DOI: 10.1007/s12672-025-02409-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/16/2025] [Indexed: 05/09/2025] Open
Abstract
In recent years, magnolol (MG), a natural active compound of polyphenolic nature, has garnered significant interest for its anti-cancer effects. Numerous studies conducted on cell lines and animal models have indicated a positive impact of administering drugs or semi-synthesized products derived from MG, including a decreased incidence of various cancers. This review aims to illustrate the underlying cellular and molecular basis of its actions. The article includes in-depth explanations of phytochemistry, semi-synthetic derivatives, bioavailability, pharmacokinetics, preclinical research, anti-tumor mechanisms, human clinical studies, toxicity, side effects, and safety. It also demonstrates that, in contrast to the wealth of synthetic medications, MG is highly effective against bladder, colon, gastric, skin, liver, lung, gallbladder, and prostate cancers. The findings of this review indicate that MG is a promising candidate as an anti-tumor agent, and future research should focus on developing new semi-synthetic derivative compounds with potential anti-tumor properties.
Collapse
Affiliation(s)
- Asmita Rayamajhi
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Nisha Gyawali
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Deepa Karki
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Luis E Pérez-Caltzontzin
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México
| | - Sheila I Peña-Corona
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México
| | - Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de Mexico, México
| | - Achyut Adhikari
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal.
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México.
| | - Yadav Uprety
- Central Department of Botany, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, Central Avenue, Chatham-Maritime, Kent, ME4 4 TB, UK
| | - Lashyn Kiyekbayeva
- Department of Pharmaceutical Technology, Pharmaceutical School, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, Iran.
- Centro de Estudios Tecnológicos y Universitarios del Golfo, Veracruz, Mexico.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
2
|
Yu K, Li X, Shi X, Li R, Zhang M. EEPD1 regulates inflammation and endothelial apoptosis in atherosclerosis through KLF4-EEPD1-ERK axis. Clin Transl Med 2025; 15:e70311. [PMID: 40268512 PMCID: PMC12017893 DOI: 10.1002/ctm2.70311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Inflammation and endothelial apoptosis are implicated in the advancement of atherosclerosis. EEPD1 holds a pivotal position in the repair of DNA damage and contributes to the progression of multiple cancers. However, the role of EEPD1 in cardiovascular diseases needs to be explored further, especially in atherosclerosis. METHODS We constructed EEPD1 and ApoE (apolipoprotein E)-deficient mice to assess how EEPD1 influences endothelial inflammation and apoptosis within atherosclerotic plaques. High-throughput RNA sequencing of human aortic endothelial cell groups treated with siCon+TNFα and siEEPD1+TNFα identified notable disparities in the MAPK pathway between groups. Chromatin immunoprecipitation and luciferase reporter assay confirmed that KLF4 directly regulates EEPD1. RESULTS Further examination of gene expression data revealed elevated EEPD1 concentrations in atherosclerotic plaques of patients, which findings were corroborated in the aortas of ApoE-/- mice. Present study demonstrated that adhesion molecule expression, endothelial apoptosis, aortic root plaques and macrophage accumulation were markedly ameliorated in EEPD1-/-ApoE-/- mice compared to WT ApoE-/- mice. Functional analysis revealed that increase in EEPD1 promotes ERK phosphorylation and significantly increases endothelial apoptosis and inflammation in atherosclerosis, which was abrogated by inhibition of ERK phosphorylation. We found KLF4 to be the transcription repressor of EEPD1 through luciferase assay and chromatin immunoprecipitation, and KLF4 inhibition abrogated the amelioration of endothelial apoptosis and inflammation caused by EEPD1 deletion. CONCLUSIONS Collectively, this study revealed that EEPD1 deletion can lead to amelioration of atherosclerosis through the KLF4-EEPD1-ERK axis. Hence, targeting EEPD1 could be a promising therapeutic strategy for patients with atherosclerosis.
Collapse
Affiliation(s)
- Kaiwen Yu
- Department of CardiologyShanghai Jiao Tong University Affiliated Chest HospitalShanghaiChina
| | - Xiang Li
- Department of CardiologyShanghai Jiao Tong University Affiliated Chest HospitalShanghaiChina
| | - Xin Shi
- Department of CardiologyShanghai Jiao Tong University Affiliated Chest HospitalShanghaiChina
| | - Ruogu Li
- Department of CardiologyShanghai Jiao Tong University Affiliated Chest HospitalShanghaiChina
| | - Min Zhang
- Department of CardiologyShanghai Jiao Tong University Affiliated Chest HospitalShanghaiChina
| |
Collapse
|
3
|
Praveen Kumar PK, Sundar H, Balakrishnan K, Subramaniam S, Ramachandran H, Kevin M, Michael Gromiha M. The Role of HSP90 and TRAP1 Targets on Treatment in Hepatocellular Carcinoma. Mol Biotechnol 2025; 67:1367-1381. [PMID: 38684604 DOI: 10.1007/s12033-024-01151-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/18/2024] [Indexed: 05/02/2024]
Abstract
Hepatocellular Carcinoma (HCC) is the predominant form of liver cancer and arises due to dysregulation of the cell cycle control machinery. Heat Shock Protein 90 (HSP90) and mitochondrial HSP90, also referred to as TRAP1 are important critical chaperone target receptors for early diagnosis and targeting HCC. Both HSP90 and TRAP1 expression was found to be higher in HCC patients. Hence, the importance of HSP90 and TRAP1 inhibitors mechanism and mitochondrial targeted delivery of those inhibitors function is widely studied. This review also focuses on importance of protein-protein interactions of HSP90 and TRAP1 targets and association of its interacting proteins in various pathways of HCC. To further elucidate the mechanism, systems biology approaches and computational biology approach studies are well explored in the association of inhibition of herbal plant molecules with HSP90 and its mitochondrial type in HCC.
Collapse
Affiliation(s)
- P K Praveen Kumar
- Department of Biotechnology, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur Tk, Tamil Nadu, 602117, India.
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India.
| | - Harini Sundar
- Department of Biotechnology, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur Tk, Tamil Nadu, 602117, India
| | - Kamalavarshini Balakrishnan
- Department of Biotechnology, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur Tk, Tamil Nadu, 602117, India
| | - Sakthivel Subramaniam
- Department of Biotechnology, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur Tk, Tamil Nadu, 602117, India
| | - Hemalatha Ramachandran
- Department of Biotechnology, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur Tk, Tamil Nadu, 602117, India
| | - M Kevin
- Department of Biotechnology, Sri Venkateswara College of Engineering, Pennalur, Sriperumbudur Tk, Tamil Nadu, 602117, India
| | - M Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, 600036, India
| |
Collapse
|
4
|
Wei H, Zhang Y, Gao M, Yang J, Wang S, Zhou X, Wei H, Xiao F. Loss of FAM172A gene prompts cell proliferation in liver regeneration. Mol Cell Biochem 2025; 480:1183-1195. [PMID: 38896202 DOI: 10.1007/s11010-024-05044-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 05/27/2024] [Indexed: 06/21/2024]
Abstract
The present study was designed to explore the function of FAM172A in liver regeneration and HCC. Mice were sacrificed after 70% partial hepatectomy (PH). RNA sequencing was performed on primary hepatocytes of WT and FAM172A-/- mice. We used HepG2 cells to construct cell lines with stably knockdown and overexpression of FAM172A. The expression of FAM172A in liver tissues was investigated by immunohistochemical staining, and we also used public database to perform survival analysis and prognostic model in HCC. Compared with WT mice after PH, normalized liver weight/body weight (LW/BW) ratio and the proliferating cell nuclear antigen (PCNA) protein level of FAM172A-/- mice elevated. The DEGs were mainly enriched in inflammatory response, tumor necrosis factor production, and wound healing. FAM172A knockdown enhanced the NFκB-TNFα and pERK-YAP1-Cyclin D1 axis. FAM172A peptide inhibited proliferation of primary hepatocytes. Moreover, the low expression of FAM172A in human HCC tissues implies a lower likelihood of survival and a valid diagnostic marker for HCC. Loss of FAM172A gene promotes cell proliferation by pERK-YAP1-Cyclin D1 and pNFκB-TNFα pathways during liver regeneration after PH. FAM172A may be a favorable diagnosis marker of HCC.
Collapse
Affiliation(s)
- Herui Wei
- Department of Gastroenterology, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Yifan Zhang
- Department of Gastroenterology, Peking University People's Hospital, Beijing, 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, 100044, China
| | - Meixin Gao
- Department of Gastroenterology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100176, China
| | - Junru Yang
- Department of Gastroenterology, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Shiwei Wang
- Department of Gastroenterology, Peking University People's Hospital, Beijing, 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, 100044, China
| | - Xingang Zhou
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Hongshan Wei
- Department of Gastroenterology, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| | - Fan Xiao
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
- Beijing Institute of Infectious Diseases, Beijing, 100015, China.
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| |
Collapse
|
5
|
Chen H, Lin Y, Chen J, Luo X, Kan Y, He Y, Zhu R, Jin J, Li D, Wang Y, Han Z. Targeting caspase-8: a new strategy for combating hepatocellular carcinoma. Front Immunol 2024; 15:1501659. [PMID: 39726605 PMCID: PMC11669555 DOI: 10.3389/fimmu.2024.1501659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024] Open
Abstract
Hepatocellular carcinoma (HCC) represents the most prevalent form of primary liver cancer and has a high mortality rate. Caspase-8 plays a pivotal role in an array of cellular signaling pathways and is essential for the governance of programmed cell death mechanisms, inflammatory responses, and the dynamics of the tumor microenvironment. Dysregulation of caspase-8 is intricately linked to the complex biological underpinnings of HCC. In this manuscript, we provide a comprehensive review of the regulatory roles of caspase-8 in apoptosis, necroptosis, pyroptosis, and PANoptosis, as well as its impact on inflammatory reactions and the intricate interplay with critical immune cells within the tumor microenvironment, such as tumor-associated macrophages, T cells, natural killer cells, and dendritic cells. Furthermore, we emphasize how caspase-8 plays pivotal roles in the development, progression, and drug resistance observed in HCC, and explore the potential of targeting caspase-8 as a promising strategy for HCC treatment.
Collapse
Affiliation(s)
- Haoran Chen
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Yumeng Lin
- Health Management Center, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Jie Chen
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Xuemei Luo
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Yubo Kan
- Sichuan Provincial Woman’s and Children’s Hospital/The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, China
| | - Yuqi He
- Department of Blood Transfusion, Lu’an People’s Hospital, the Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Renhe Zhu
- Department of Blood Transfusion, Lu’an People’s Hospital, the Affiliated Hospital of Anhui Medical University, Lu’an, China
| | - Jiahui Jin
- Department of gastroenterology, Baoji Central Hospital, Baoji, China
| | - Dongxuan Li
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Yi Wang
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Zhongyu Han
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| |
Collapse
|
6
|
Zuo W, Ma H, Bi J, Li T, Mo Y, Yu S, Wang J, Li B, Huang J, Li Y, Li L. Phosphorylation of RelA/p65 Ser536 inhibits the progression and metastasis of hepatocellular carcinoma by mediating cytoplasmic retention of NF-κB p65. Gastroenterol Rep (Oxf) 2024; 12:goae094. [PMID: 39498383 PMCID: PMC11534074 DOI: 10.1093/gastro/goae094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 08/26/2024] [Accepted: 09/06/2024] [Indexed: 11/07/2024] Open
Abstract
Background Intrahepatic and extrahepatic metastases contribute to the high recurrence rate and mortality of hepatocellular carcinoma (HCC). Constitutive activation of nuclear factor-κB (NF-κB) is a crucial feature of HCC. NF-κB p65 (p50-p65) is the most common dimeric form. Ser536 acts as an essential phosphorylation site of RelA/p65. However, the effect of RelA/p65 Ser536 phosphorylation on progression and metastases during intermediate and advanced HCC has not been reported. Methods Phosphorylation of RelA/p65 (p-p65 Ser536) and NF-κB p65 were detected by using immunohistochemical staining in HCC tissue samples. The biological effects of RelA/p65 Ser536 phosphorylation were evaluated by using xenograft and metastasis models. NF-κB p65 nuclear translocation was detected by using Western blotting. The binding of NF-κB p65 to the BCL2, SNAIL, and MMP9 promoters was detected by using chromatin immunoprecipitation. The biological effects on proliferation, migration, invasion, and epithelial-mesenchymal transition were assessed by using tetrazolium-based colorimetry, colony formation, EdU incorporation, flow cytometry, cell wound healing, and transwell assay. Results NF-κB p65 is highly expressed, while p-p65 Ser536 is not well expressed in intermediate and advanced HCC tissues. In vivo experiments demonstrated that a phosphorylation-mimetic mutant of RelA/p65 Ser536 (p65/S536D) prevents tumor progression and metastasis. In vitro experiments showed that p65/S536D inhibits proliferation, migration, and invasion. Mechanistically, RelA/p65 Ser536 phosphorylation inhibits NF-κB p65 nuclear translocation and reduces NF-κB p65 binding to the BCL2, SNAIL, and MMP9 promoters. Conclusions RelA/p65 Ser536 phosphorylation was detrimental to NF-κB p65 entry into the nucleus and inhibited HCC progression and metastasis by reducing BCL2, SNAIL, and MMP9. The phosphorylation site of RelA/p65 Ser536 has excellent potential to be a promising target for NF-κB-targeted therapy in HCC.
Collapse
Affiliation(s)
- Wentao Zuo
- College of Basic Medical, Guilin Medical University, Guilin, Guangxi, P. R. China
| | - Haoyang Ma
- College of Basic Medical, Guilin Medical University, Guilin, Guangxi, P. R. China
| | - Jianghui Bi
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, P. R. China
| | - Tiaolan Li
- College of Basic Medical, Guilin Medical University, Guilin, Guangxi, P. R. China
| | - Yifeng Mo
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, P. R. China
| | - Shiyu Yu
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, P. R. China
| | - Jia Wang
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, P. R. China
| | - Beiqing Li
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, P. R. China
| | - Jinfeng Huang
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, P. R. China
| | - Yongwen Li
- College of Pharmacy, Guilin Medical University, Guilin, Guangxi, P. R. China
| | - Li Li
- College of Basic Medical, Guilin Medical University, Guilin, Guangxi, P. R. China
| |
Collapse
|
7
|
Peng Y, Wu X, Zhang Y, Yin Y, Chen X, Zheng D, Wang J. An Overview of Traditional Chinese Medicine in the Treatment After Radical Resection of Hepatocellular Carcinoma. J Hepatocell Carcinoma 2023; 10:2305-2321. [PMID: 38143910 PMCID: PMC10743783 DOI: 10.2147/jhc.s413996] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/03/2023] [Indexed: 12/26/2023] Open
Abstract
According to the Barcelona Clinic Liver Cancer (BCLC) system, radical resection of early stage primary hepatocellular carcinoma (HCC) mainly includes liver transplantation, surgical resection, and radiofrequency ablation (RFA), which yield 5-year survival rates of about 70-79%, 41.3-69.5%, and 40-70%, respectively. The tumor-free 5-year rate for HCC patients undergoing radical resection only reach up to 13.7 months, so the prevention of recurrence after radical resection of HCC is very important for the prognosis of patients. The traditional Chinese medicine (TCM) takes the approach of multitarget and overall-regulation to treat tumors, it can also independently present the "component-target-pathway" related to a particular disease, and its systematic and holistic characteristics can provide a personalized therapy based on symptoms of the patient by treating the patient as a whole. TCM as postoperative adjuvant therapy after radical resection of HCC in Barcelona Clinic liver cancer A or B stages, and the numerous clinical trials confirmed that the efficacy of TCM in the field of HCC has a significant effect, not only improving the prognosis and quality of life but also enhancing patient survival rate. However, with the characteristics of multi-target, multi-component, and multi-pathway, the specific mechanism of Chinese medicine in the treatment of diseases is still unclear. Because of the positive pharmacological activities of TCM in combating anti-tumors, the mechanism studies of TCM have demonstrated beneficial effects on the regulation of immune function, chronic inflammation, the proliferation and metastasis of liver cancer cells, autophagy, and cell signaling pathways related to liver cancer. Therefore, this article reviews the mechanism of traditional Chinese medicine in reducing the recurrence rate of HCC after radical resection.
Collapse
Affiliation(s)
- Yichen Peng
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Hepatobiliary Department, Luzhou, People’s Republic of China
- Department of Integrated Traditional Chinese & Western Medicine, The Southwest Medical University, Luzhou, People’s Republic of China
| | - Xia Wu
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Hepatobiliary Department, Luzhou, People’s Republic of China
- Department of Integrated Traditional Chinese & Western Medicine, The Southwest Medical University, Luzhou, People’s Republic of China
| | - Yurong Zhang
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Hepatobiliary Department, Luzhou, People’s Republic of China
| | - Yue Yin
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Hepatobiliary Department, Luzhou, People’s Republic of China
| | - Xianglin Chen
- Department of Integrated Traditional Chinese & Western Medicine, The Southwest Medical University, Luzhou, People’s Republic of China
| | - Ding Zheng
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Hepatobiliary Department, Luzhou, People’s Republic of China
| | - Jing Wang
- The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Hepatobiliary Department, Luzhou, People’s Republic of China
| |
Collapse
|
8
|
Hsu FT, Liu WL, Lee SR, Jeng LB, Chen JH. Unveiling nature's potential weapon: Magnolol's role in combating bladder cancer by upregulating the miR-124 and inactivating PKC-δ/ERK axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:154947. [PMID: 37549536 DOI: 10.1016/j.phymed.2023.154947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/07/2023] [Accepted: 06/28/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND Bladder cancer (BC) is a challenging disease to manage. Researchers have been investigating the potential of magnolol, a compound derived from Magnolia officinalis, as an anti-cancer agent. However, the exact regulatory mechanism of magnolol and its impact on the NF-κB signaling pathway in BC remain unclear. MATERIALS To comprehensively evaluate its therapeutic potential, the researchers conducted a series of experiments using BC cell lines (TSGH8301, T24, and MB49) and in vivo animal models. RESULTS The results of the study demonstrated that magnolol exhibits cytotoxic effects on BC cells by activating both the extrinsic and intrinsic apoptosis signaling pathways. Additionally, the expression of anti-apoptotic genes was downregulated by magnolol treatment. The researchers also uncovered the regulatory role of PKCδ/ERK and miR-124-3p in the NF-κB pathway, which may be influenced by magnolol. Treatment with magnolol led to the inactivation of PKCδ/ERK and an increase in miR-124-3p expression, effectively inhibiting NF-κB-mediated progression of BC. Importantly, the administration of magnolol did not result in significant toxicity in normal tissues, highlighting its potential as a safe adjunctive therapy with minimal adverse effects. CONCLUSION These findings position magnolol as a promising therapeutic agent for the treatment of BC. By activating apoptosis signaling pathways and inhibiting NF-κB pathway through the upregulation of miR-124-3p and downregulation of PKCδ/ERK activation, magnolol holds promise for suppressing tumor progression and improving patient outcomes in BC. Further research and clinical trials are warranted to explore the full potential of magnolol in the future.
Collapse
Affiliation(s)
- Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C
| | - Wei-Lin Liu
- Department of Radiation Oncology, Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C
| | - Sin-Rong Lee
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C; Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei, Taiwan, R.O.C
| | - Long-Bin Jeng
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan, R.O.C; Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan, R.O.C
| | - Jiann-Hwa Chen
- Department of Emergency Medicine, Cathay General Hospital, Taipei, Taiwan, R.O.C; School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, R.O.C.
| |
Collapse
|
9
|
Liang D, Liu L, Zhao Y, Luo Z, He Y, Li Y, Tang S, Tang J, Chen N. Targeting extracellular matrix through phytochemicals: a promising approach of multi-step actions on the treatment and prevention of cancer. Front Pharmacol 2023; 14:1186712. [PMID: 37560476 PMCID: PMC10407561 DOI: 10.3389/fphar.2023.1186712] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/11/2023] [Indexed: 08/11/2023] Open
Abstract
Extracellular matrix (ECM) plays a pivotal and dynamic role in the construction of tumor microenvironment (TME), becoming the focus in cancer research and treatment. Multiple cell signaling in ECM remodeling contribute to uncontrolled proliferation, metastasis, immune evasion and drug resistance of cancer. Targeting trilogy of ECM remodeling could be a new strategy during the early-, middle-, advanced-stages of cancer and overcoming drug resistance. Currently nearly 60% of the alternative anticancer drugs are derived from natural products or active ingredients or structural analogs isolated from plants. According to the characteristics of ECM, this manuscript proposes three phases of whole-process management of cancer, including prevention of cancer development in the early stage of cancer (Phase I); prevent the metastasis of tumor in the middle stage of cancer (Phase II); provide a novel method in the use of immunotherapy for advanced cancer (Phase III), and present novel insights on the contribution of natural products use as innovative strategies to exert anticancer effects by targeting components in ECM. Herein, we focus on trilogy of ECM remodeling and the interaction among ECM, cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs), and sort out the intervention effects of natural products on the ECM and related targets in the tumor progression, provide a reference for the development of new drugs against tumor metastasis and recurrence.
Collapse
Affiliation(s)
- Dan Liang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunjie Zhao
- Key Laboratory of Marine Fishery Resources Exploitment and Utilization of Zhejiang Province, College of Pharmaceutical Science and Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Zhenyi Luo
- Graduate School, Guangxi University of Chinese Medicine, Nanning, China
| | - Yadi He
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanping Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyun Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianyuan Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Nianzhi Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Wu W, Wang Y, Xie J, Fan S. Empagliflozin: a potential anticancer drug. Discov Oncol 2023; 14:127. [PMID: 37436535 DOI: 10.1007/s12672-023-00719-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/06/2023] [Indexed: 07/13/2023] Open
Abstract
Empagliflozin, a sodium-glucose cotransporter 2 (SGLT2) inhibitor, is a highly effective and well-tolerated antidiabetic drug. In addition to hypoglycemic effects, empagliflozin has many other effects, such as being hypotensive and cardioprotective. It also has anti-inflammatory and antioxidative stress effects in diabetic nephropathy. Several studies have shown that empagliflozin has anticancer effects. SGLT2 is expressed in a variety of cancer cell lines. The SGLT2 inhibitor empagliflozin has significant inhibitory effects on certain types of tumor cells, such as inhibition of proliferation, migration and induction of apoptosis. In conclusion, empagliflozin has promising applications in cancer therapy as a drug for the treatment of diabetes and heart failure. This article provides a brief review of the anticancer effects of empagliflozin.
Collapse
Affiliation(s)
- Wenwen Wu
- School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China
| | - Yanyan Wang
- Department of Ultrasonic Medicine, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, 221000, China
| | - Jun Xie
- School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China.
| | - Shaohua Fan
- School of Life Sciences, Jiangsu Normal University, Xuzhou, Jiangsu, 221116, China.
| |
Collapse
|
11
|
Kundu M, Das S, Nandi S, Dhara D, Mandal M. Magnolol and Temozolomide exhibit a synergistic anti-glioma activity through MGMT inhibition. Biochim Biophys Acta Mol Basis Dis 2023:166782. [PMID: 37286145 DOI: 10.1016/j.bbadis.2023.166782] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 05/27/2023] [Accepted: 05/29/2023] [Indexed: 06/09/2023]
Abstract
Temozolomide (TMZ) is the leading chemotherapeutic agent used for glioma therapy due to its good oral absorption and blood-brain barrier permeability. However, its anti-glioma efficacy may be limited due to its adverse effects and resistance development. O6-Methylguanine-DNA-methyltransferase (MGMT), an enzyme associated with TMZ resistance, is activated via the NF-κB pathway, which is found to be upregulated in glioma. TMZ also upregulates NF-κB signaling like many other alkylating agents. Magnolol (MGN), a natural anti-cancer agent, has been reported to inhibit NF-κB signaling in multiple myeloma, cholangiocarcinoma, and hepatocellular carcinoma. MGN has already shown promising results in anti-glioma therapy. However, the synergistic action of TMZ and MGN has not been explored. Therefore, we investigated the effect of TMZ and MGN treatment in glioma and observed their synergistic pro-apoptotic action in both in vitro and in vivo glioma models. To explore the mechanism of this synergistic action, we found that MGN inhibits MGMT enzyme both in vitro and in vivo glioma. Next, we established the link between NF-κB signaling and MGN-induced MGMT inhibition in glioma. MGN inhibits the phosphorylation of p65, a subunit of NF-κB, and its nuclear translocation to block NF-κB pathway activation in glioma. MGN-induced NF-κB inhibition results in the transcriptional inhibition of MGMT in glioma. TMZ and MGN combinatorial treatment also impedes p65 nuclear translocation to inhibit MGMT in glioma. We observed a similar effect of TMZ and MGN treatment in the rodent glioma model. Thus, we concluded that MGN potentiates TMZ-induced apoptosis in glioma by inhibiting NF-κB pathway-mediated MGMT activation.
Collapse
Affiliation(s)
- Moumita Kundu
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Subhayan Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Suvendu Nandi
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Dibakar Dhara
- Department of Chemistry, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| |
Collapse
|
12
|
Wang Y, Li J, Xia L. Plant-derived natural products and combination therapy in liver cancer. Front Oncol 2023; 13:1116532. [PMID: 36865794 PMCID: PMC9971944 DOI: 10.3389/fonc.2023.1116532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Liver cancer is one of the malignant cancers globally and seriously endangers human health because of its high morbidity and mortality. Plant-derived natural products have been evaluated as potential anticancer drugs due to low side effects and high anti-tumor efficacy. However, plant-derived natural products also have defects of poor solubility and cumbersome extraction process. In recent years, a growing numbers of plant derived natural products have been used in combination therapy of liver cancer with conventional chemotherapeutic agents, which has improved clinical efficacy through multiple mechanisms, including inhibition of tumor growth, induction of apoptosis, suppression of angiogenesis, enhancement of immunity, reversal of multiple drug resistance and reduction of side effects. The therapeutic effects and mechanisms of plant-derived natural products and combination therapy on liver cancer are reviewed to provide references for developing anti-liver-cancer strategies with high efficacy and low side effects.
Collapse
Affiliation(s)
- Yuqin Wang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, China
| | - Jinyao Li
- *Correspondence: Jinyao Li, ; Lijie Xia,
| | - Lijie Xia
- *Correspondence: Jinyao Li, ; Lijie Xia,
| |
Collapse
|
13
|
Wang X, Liu Q, Fu Y, Ding RB, Qi X, Zhou X, Sun Z, Bao J. Magnolol as a Potential Anticancer Agent: A Proposed Mechanistic Insight. Molecules 2022; 27:molecules27196441. [PMID: 36234977 PMCID: PMC9570903 DOI: 10.3390/molecules27196441] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is a serious disease with high mortality and morbidity worldwide. Natural products have served as a major source for developing new anticancer drugs during recent decades. Magnolol, a representative natural phenolic lignan isolated from Magnolia officinali, has attracted considerable attention for its anticancer properties in recent years. Accumulating preclinical studies have demonstrated the tremendous therapeutic potential of magnolol via a wide range of pharmacological mechanisms against cancer. In this review, we summarized the latest advances in preclinical studies investigating anticancer properties of magnolol and described the important signaling pathways explaining its underlying mechanisms. Magnolol was capable of inhibiting cancer growth and metastasis against various cancer types. Magnolol exerted anticancer effects through inhibiting proliferation, inducing cell cycle arrest, provoking apoptosis, restraining migration and invasion, and suppressing angiogenesis. Multiple signaling pathways were also involved in the pharmacological actions of magnolol against cancer, such as PI3K/Akt/mTOR signaling, MAPK signaling and NF-κB signaling. Based on this existing evidence summarized in the review, we have conclusively confirmed magnolol had a multi-target anticancer effect against heterogeneous cancer disease. It is promising to develop magnolol as a drug candidate for cancer therapy in the future.
Collapse
Affiliation(s)
- Xiaofeng Wang
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Qingqing Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Yuanfeng Fu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Ren-Bo Ding
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Xingzhu Qi
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Xuejun Zhou
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Zhihua Sun
- State International Joint Research Center for Animal Health Breeding, Key Laboratory of Control and Prevention of Animal Disease of Xinjiang Production & Construction Corps, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
- Correspondence: (Z.S.); (J.B.)
| | - Jiaolin Bao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
- Correspondence: (Z.S.); (J.B.)
| |
Collapse
|
14
|
Dai Q, Ain Q, Rooney M, Song F, Zipprich A. Role of IQ Motif-Containing GTPase-Activating Proteins in Hepatocellular Carcinoma. Front Oncol 2022; 12:920652. [PMID: 35785216 PMCID: PMC9243542 DOI: 10.3389/fonc.2022.920652] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/10/2022] [Indexed: 11/21/2022] Open
Abstract
IQ motif-containing GTPase-activating proteins (IQGAPs) are a class of scaffolding proteins, including IQGAP1, IQGAP2, and IQGAP3, which govern multiple cellular activities by facilitating cytoskeletal remodeling and cellular signal transduction. The role of IQGAPs in cancer initiation and progression has received increasing attention in recent years, especially in hepatocellular carcinoma (HCC), where the aberrant expression of IQGAPs is closely related to patient prognosis. IQGAP1 and 3 are upregulated and are considered oncogenes in HCC, while IQGAP2 is downregulated and functions as a tumor suppressor. This review details the three IQGAP isoforms and their respective structures. The expression and role of each protein in different liver diseases and mainly in HCC, as well as the underlying mechanisms, are also presented. This review also provides a reference for further studies on IQGAPs in HCC.
Collapse
Affiliation(s)
- Qingqing Dai
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
- Else Kröner Graduate School for Medical Students “Jena School for Ageing Medicine (JSAM)”, Jena University Hospital, Jena, Germany
| | - Quratul Ain
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Michael Rooney
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
| | - Fei Song
- Department of Urology, Jena University Hospital, Jena, Germany
| | - Alexander Zipprich
- Department of Internal Medicine IV (Gastroenterology, Hepatology, and Infectious Diseases), Jena University Hospital, Jena, Germany
- *Correspondence: Alexander Zipprich,
| |
Collapse
|
15
|
Jiang H, Tang W, Song Y, Jin W, Du Q. Induction of Apoptosis by Metabolites of Rhei Radix et Rhizoma (Da Huang): A Review of the Potential Mechanism in Hepatocellular Carcinoma. Front Pharmacol 2022; 13:806175. [PMID: 35308206 PMCID: PMC8924367 DOI: 10.3389/fphar.2022.806175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/24/2022] [Indexed: 11/28/2022] Open
Abstract
Liver cancer is a global disease with a high mortality rate and limited treatment options. Alternations in apoptosis of tumor cells and immune cells have become an important method for detailing the underlying mechanisms of hepatocellular carcinoma (HCC). Bcl-2 family, Caspase family, Fas and other apoptosis-related proteins have also become antagonistic targets of HCC. Da Huang (Rhei Radix et Rhizoma, RR), a traditional Chinese herb, has recently demonstrated antitumor behaviors. Multiple active metabolites of RR, including emodin, rhein, physcion, aloe-emodin, gallic acid, and resveratrol, can successfully induce apoptosis and inhibit HCC. However, the underlying mechanisms of these metabolites inhibiting the occurrence and development of HCC by inducing apoptosis is complicated owing to the multi-target and multi-pathway characteristics of traditional Chinese herbs. Accordingly, this article reviews the pathways of apoptosis, the relationship between HCC and apoptosis, the role and mechanism of apoptosis induced by mitochondrial endoplasmic reticulum pathway and death receptor pathway in HCC and the mechanism of six RR metabolites inhibiting HCC by inducing apoptosis.
Collapse
Affiliation(s)
- Huanyu Jiang
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wuyinuo Tang
- Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yang Song
- Emergency Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Jin
- Emergency Department, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Quanyu Du
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
16
|
Fakhri S, Moradi SZ, Yarmohammadi A, Narimani F, Wallace CE, Bishayee A. Modulation of TLR/NF-κB/NLRP Signaling by Bioactive Phytocompounds: A Promising Strategy to Augment Cancer Chemotherapy and Immunotherapy. Front Oncol 2022; 12:834072. [PMID: 35299751 PMCID: PMC8921560 DOI: 10.3389/fonc.2022.834072] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
Background Tumors often progress to a more aggressive phenotype to resist drugs. Multiple dysregulated pathways are behind this tumor behavior which is known as cancer chemoresistance. Thus, there is an emerging need to discover pivotal signaling pathways involved in the resistance to chemotherapeutic agents and cancer immunotherapy. Reports indicate the critical role of the toll-like receptor (TLR)/nuclear factor-κB (NF-κB)/Nod-like receptor pyrin domain-containing (NLRP) pathway in cancer initiation, progression, and development. Therefore, targeting TLR/NF-κB/NLRP signaling is a promising strategy to augment cancer chemotherapy and immunotherapy and to combat chemoresistance. Considering the potential of phytochemicals in the regulation of multiple dysregulated pathways during cancer initiation, promotion, and progression, such compounds could be suitable candidates against cancer chemoresistance. Objectives This is the first comprehensive and systematic review regarding the role of phytochemicals in the mitigation of chemoresistance by regulating the TLR/NF-κB/NLRP signaling pathway in chemotherapy and immunotherapy. Methods A comprehensive and systematic review was designed based on Web of Science, PubMed, Scopus, and Cochrane electronic databases. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines were followed to include papers on TLR/NF-κB/NLRP and chemotherapy/immunotherapy/chemoresistance by phytochemicals. Results Phytochemicals are promising multi-targeting candidates against the TLR/NF-κB/NLRP signaling pathway and interconnected mediators. Employing phenolic compounds, alkaloids, terpenoids, and sulfur compounds could be a promising strategy for managing cancer chemoresistance through the modulation of the TLR/NF-κB/NLRP signaling pathway. Novel delivery systems of phytochemicals in cancer chemotherapy/immunotherapy are also highlighted. Conclusion Targeting TLR/NF-κB/NLRP signaling with bioactive phytocompounds reverses chemoresistance and improves the outcome for chemotherapy and immunotherapy in both preclinical and clinical stages.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Akram Yarmohammadi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Narimani
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Carly E. Wallace
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| |
Collapse
|
17
|
Chen X, Lin S, Lin Y, Wu S, Zhuo M, Zhang A, Zheng J, You Z. BRAF-activated WT1 contributes to cancer growth and regulates autophagy and apoptosis in papillary thyroid carcinoma. J Transl Med 2022; 20:79. [PMID: 35123502 PMCID: PMC8818187 DOI: 10.1186/s12967-022-03260-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 01/17/2022] [Indexed: 12/17/2022] Open
Abstract
Abstract
Background
Papillary thyroid carcinoma (PTC) is one of most prevalent malignant endocrine neoplasms, and it is associated with a high frequency of BRAF gene mutations, which lead to lymphatic metastasis and distant metastasis that promote tumor progression. The molecular mechanism of PTC and the role of BRAF mutation in PTC progression and development need to be further elucidated.
Methods
In this study, a comprehensive bioinformatics analysis was performed to identify the differentially expressed genes and signaling pathways in thyroid cancer patients carrying mutant BRAF. Then, we confirmed the prognostic role of WT1 in thyroid cancer patients. Immunohistochemistry was performed to measure the expression profile of WT1 in PTC tissue. Lentivirus shWT1 was transfected into BRAFV600E (mutant) PTC cells to stably inhibit WT1 expression. CCK-8, EdU, immunofluorescence, colony formation, cell migration, cell wound healing, apoptosis and autophagy assays were performed to assess the biological functions of WT1 in BRAFV600E PTC cells. RNA sequencing, immunohistochemistry and immunoblotting were performed to explore the molecular mechanism of WT1 in BRAFV600E PTC cells.
Results
The results confirmed that “epithelial cell proliferation”, “apoptosis” and “selective autophagy” were closely associated with this BRAF mutant in these thyroid cancer patients. Knocking down BRAF-activated WT1 effectively inhibited the proliferation and migration of BRAFV600E PTC cells. Silencing WT1 significantly inhibited autophagy and promoted the apoptosis of BRAFV600E PTC cells. Mechanistic investigations showed that silencing WT1 expression remarkably suppressed the AKT/mTOR and ERK/P65 signaling pathways in BRAFV600E PTC cells.
Conclusion
All these results indicate that WT1 is a promising prognostic biomarker and facilitates PTC progression and development of cells carrying the BRAFV600E mutation.
Collapse
|
18
|
Wang Y, Sun C, Huang L, Liu M, Li L, Wang X, Wang L, Sun S, Xu H, Ma G, Zhang L, Zheng J, Liu H. Magnolol-loaded Cholesteryl Biguanide Conjugate Hydrochloride Nanoparticles for Triple-negative Breast Cancer Therapy. Int J Pharm 2022; 615:121509. [PMID: 35085734 DOI: 10.1016/j.ijpharm.2022.121509] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/15/2022] [Accepted: 01/21/2022] [Indexed: 12/25/2022]
Abstract
The potential of combination therapy using nanoparticle delivery systems in improving triple-negative breast cancer treatment efficacy remains to be explored. Here, we report a novel nanoparticle system using a cholesterol biguanide conjugate hydrochloride (CBH) as both a drug and carrier to load magnolol (MAG). Poly(ethylene glycol)-poly(lactic-co-glycolic acid) (mPEG-PLGA) and aminoethyl anisamide-poly(ethylene glycol)-poly(lactic-co-glycolic acid) (AEAA-PEG-PLGA) were added to form nanoparticles. Nanoparticles accumulated most in tumor tissues when the weight ratio of AEAA-PEG-PLGA to mPEG-PLGA was 4:1. MAG and CBH exerted a synergistic inhibitory effect on 4T1 cells. An in vitro study showed that nanoparticles displayed the highest tumor cell uptake rate, highest apoptosis rate, and strongest inhibitory effect on tumor cell migration and monoclonal formation. CBH might promote nanoparticle uptake by cells and lysosomal escape. After intravenous administration to mice with 4T1 breast tumors in situ, the nanoparticles inhibited tumor growth without obvious toxicity. Western blot results showed that nanoparticles altered the levels of p53, p-AKT, and p-AMPK in the tumor tissue. Moreover, cell apoptosis was found in the same area of H&E-stained and TUNEL-stained tumors treated with the nanoparticles. Collectively, this nanoparticle system provides a novel combination drug delivery strategy for treating triple-negative breast cancer.
Collapse
Affiliation(s)
- Yanzhi Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou 450001, China.
| | - Cancan Sun
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou 450001, China; Department of Pharmacy, People's Hospital of Zhengzhou, Zhengzhou 450001, China
| | - Leaf Huang
- Division of Pharmaco-engineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mengqian Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou 450001, China
| | - Lu Li
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou 450001, China
| | - Xiping Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou 450001, China
| | - Linchao Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou 450001, China
| | - Shanshan Sun
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou 450001, China
| | - Haiwei Xu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou 450001, China
| | - Gege Ma
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou 450001, China
| | - Lei Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou 450001, China
| | - Jiaxin Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou 450001, China.
| | - Hongmin Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Henan Key Laboratory of Drug Quality Control & Evaluation, School of Pharmaceutical Sciences, Zhengzhou University, Ministry of Education of China, Zhengzhou 450001, China.
| |
Collapse
|
19
|
Su C, Lin S, Wang H, Hsu F, Chung JG, Hsu L. The inhibitory effect and mechanism of quetiapine on tumor progression in hepatocellular carcinoma in vivo. ENVIRONMENTAL TOXICOLOGY 2022; 37:92-100. [PMID: 34626444 PMCID: PMC9293313 DOI: 10.1002/tox.23380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 05/27/2023]
Abstract
Hepatocellular carcinoma (HCC) is the primary tumor of the liver and the fourth leading cause of cancer-related death. Recently, several studies indicated the anti-tumor potential of antipsychotic medicine. Quetiapine, an atypical antipsychotic, is used to treat schizophrenia, bipolar disorder, and major depressive disorder since 1997. However, whether quetiapine may show potential to suppress HCC progression and its underlying mechanism is persisting unclear. Quetiapine has been shown to induce apoptosis and inhibit invasion ability in HCC in vitro. Here, we established two different HCC (Hep3B, SK-Hep1) bearing animals to identify the treatment efficacy of quetiapine. Tumor growth, signaling transduction, and normal tissue pathology after quetiapine treatment were validated by caliper, bioluminescence image, immunohistochemistry (IHC), and hematoxylin and eosin staining, respectively. Quetiapine suppressed HCC progression in a dose-dependent manner. Extracellular signal-regulated kinases (ERKs) and Nuclear factor-κB (NF-κB) mediated downstream proteins, such as myeloid leukemia cell differentiation protein (MCL-1), cellular FLICE-inhibitory protein (C-FLIP), X-linked inhibitor of apoptosis protein (XIAP), Cyclin-D1, matrix metallopeptidase 9 (MMP-9), vascular endothelial growth factor-A (VEGF-A) and indoleamine 2,3-dioxygenase (IDO) which involved in proliferation, survival, angiogenesis, invasion and anti-tumor immunity were all decreased by quetiapine. In addition, extrinsic/intrinsic caspase-dependent and caspase-independent pathways, including cleaved caspase-3, -8, and - 9 were increased by quetiapine. In sum, the tumor inhibition that results from quetiapine may associate with ERK and NF-κB inactivation.
Collapse
Affiliation(s)
- Chun‐Min Su
- Department of SurgeryShow Chwan Memorial HospitalChanghuaTaiwan, ROC
| | - Song‐Shei Lin
- Department of Medical Imaging and Radiological SciencesCentral Taiwan University of Science and TechnologyTaichungTaiwan, ROC
| | - Hsiao‐Chia Wang
- Emergency DepartmentCathay General HospitalTaipeiTaiwan, ROC
- School of MedicineFu Jen Catholic UniversityNew Taipei CityTaiwan, ROC
| | - Fei‐Ting Hsu
- Department of Biological Science and TechnologyChina Medical UniversityTaichungTaiwan, ROC
| | - Jing Gung Chung
- Department of Biological Science and TechnologyChina Medical UniversityTaichungTaiwan, ROC
| | - Li‐Cho Hsu
- School of MedicineNational Yang‐Ming Chiao‐Tung University HospitalTaipeiTaiwan, ROC
| |
Collapse
|
20
|
Magnolol Induces the Extrinsic/Intrinsic Apoptosis Pathways and Inhibits STAT3 Signaling-Mediated Invasion of Glioblastoma Cells. Life (Basel) 2021; 11:life11121399. [PMID: 34947930 PMCID: PMC8706091 DOI: 10.3390/life11121399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/05/2021] [Accepted: 12/06/2021] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common form of malignant brain tumor, with poor prognosis; the efficacy of current standard therapy for GBM remains unsatisfactory. Magnolol, an herbal medicine from Magnolia officinalis, exhibited anticancer properties against many types of cancers. However, whether magnolol suppresses GBM progression as well as its underlying mechanism awaits further investigation. In this study, we used the MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide) assay, apoptosis marker analysis, transwell invasion and wound-healing assays to identify the effects of magnolol on GBM cells. We also validated the potential targets of magnolol on GBM with the GEPIA (Gene Expression Profiling Interactive Analysis) and Western blotting assay. Magnolol was found to trigger cytotoxicity and activate extrinsic/intrinsic apoptosis pathways in GBM cells. Both caspase-8 and caspase-9 were activated by magnolol. In addition, GEPIA data indicated the PKCδ (Protein kinase C delta)/STAT3 (Signal transducer and activator of transcription 3) signaling pathway as a potential target of GBM. Magnolol effectively suppressed the phosphorylation and nuclear translocation of STAT3 in GBM cells. Meanwhile, tumor invasion and migration ability and the associated genes, including MMP-9 (Matrix metalloproteinase-9) and uPA (Urokinase-type plasminogen activator), were all diminished by treatment with magnolol. Taken together, our results suggest that magnolol-induced anti-GBM effect may be associated with the inactivation of PKCδ/STAT3 signaling transduction.
Collapse
|
21
|
Abdelhamid AM, Saber S, Youssef ME, Gaafar AGA, Eissa H, Abd-Eldayem MA, Alqarni M, Batiha GES, Obaidullah AJ, Shahien MA, El-Ahwany E, Amin NA, Etman MA, Kaddah MMY, Abd El-Fattah EE. Empagliflozin adjunct with metformin for the inhibition of hepatocellular carcinoma progression: Emerging approach for new application. Biomed Pharmacother 2021; 145:112455. [PMID: 34844106 DOI: 10.1016/j.biopha.2021.112455] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/05/2021] [Accepted: 11/16/2021] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is on the rise worldwide, and its incidence in diabetic patients is two to three times that of non-diabetics. Current therapeutic options fail to provide considerable survival benefits to patients with HCC. There is a strong possibility that the FDA-approved antidiabetic combination of empagliflozin and metformin could show complementary effects to control HCC progression. However, their multitarget effects have not yet been studied on HCC development. Therefore, the present study aims to evaluate the antitumorigenic activity of this combination in non-diabetic mice with diethylnitrosamine-induced HCC. Empagliflozin/metformin combination prolonged survival and improved histological features of mice livers. Additionally, Empagliflozin/metformin showed anti-inflammatory potential and relieved oxidative stress. On the one hand these effects are likely attributed to the ability of metformin to inactivate NF-κB in an AMPK-dependent mechanism and on the other hand to the ability of the empagliflozin to inhibit the MAPKs, p38 and ERK1/2. Empagliflozin also showed a less robust effect on AMPK than that of metformin. Moreover, empagliflozin enhanced the autophagy inducing activity of metformin. Furthermore, empagliflozin/metformin exhibited increased apoptotic potential. Consequently, empagliflozin augmented the antitumorigenic function of metformin by exerting better control of angiogenesis, and metastasis. To conclude, our findings suggest empagliflozin as an ideal adjunct to metformin for the inhibition of HCC progression. In addition, since the incidence of hypoglycemia is minimal due to insulin-independent mechanism of action of both treatments, empagliflozin/metformin could be a promising therapeutic modality for the management of diabetic patients with HCC; and even non diabetic ones.
Collapse
Affiliation(s)
- Amir Mohamed Abdelhamid
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt.
| | - Mahmoud E Youssef
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Ahmed Gaafar Ahmed Gaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| | - Hanan Eissa
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Marwa A Abd-Eldayem
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University, New Damietta, Egypt
| | - Mohammed Alqarni
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, AlBeheira, Egypt
| | - Ahmad J Obaidullah
- Drug Exploration and Development Chair (DEDC), Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed Awad Shahien
- Department of Clinical Pharmacology, Faculty of Medicine, Damietta University, Damietta, Egypt
| | - Eman El-Ahwany
- Department of Immunology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Noha A Amin
- Department of Hematology, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mohamed Ali Etman
- Research and Development, Department of Drug Stability, Safe Pharma, Pharco Pharmaceuticals, Alexandria, Egypt
| | - Mohamed M Y Kaddah
- Pharmaceutical and Fermentation Industries Development Center, City of Scientific Research and Technological Applications, New Borg El-Arab 21934, Alexandria, Egypt
| | - Eslam E Abd El-Fattah
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| |
Collapse
|
22
|
Wu Z, Yu L, Li X, Li X. Protective Mechanism of Trimetazidine in Myocardial Cells in Myocardial Infarction Rats through ERK Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9924549. [PMID: 34651051 PMCID: PMC8510807 DOI: 10.1155/2021/9924549] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/06/2021] [Indexed: 01/21/2023]
Abstract
OBJECTIVE To study the protective effect of trimetazidine on myocardial cells in rats with myocardial infarction and explore its effect on ERK signaling pathway. METHODS 40 SD rats were randomly divided into the sham operation group, model group, low-dose group, and high-dose group (intra-abdominal injection of trimetazidine 5 mg/kg and 10 mg/kg, respectively), construction of rat myocardial infarction model by coronary artery left anterior descending artery ligation. 7 days after surgery, the survival rate and cardiac function of each group of rats were recorded. The myocardial infarct size was detected by TTC staining. The apoptosis level of rat cardiomyocytes was detected by TUNEL staining. The content of ROS in rat cardiomyocytes was detected by DCFH-DA. Western-blot was used to detection of Caspase-3, Bcl-2/Bax, and ERK signaling pathway-related proteins in myocardial tissue. RESULTS Compared with the model group, the survival rate of the rats in the low-dose group and the high-dose group was significantly increased (P < 0.01), the cardiac function was significantly improved (P < 0.01), the myocardial infarct size was significantly decreased (P < 0.01), the level of apoptosis was significantly decreased (P < 0.01), the content of ROS in cardiomyocytes was significantly decreased (P < 0.01), the protein expression of Caspase-3 and NF-κB in cardiomyocytes was significantly decreased (P < 0.01), and the expression of Bcl-2/Bax and p-ERK were significantly increased (P < 0.01). CONCLUSION Trimetazidine can activate ERK signaling pathway in cardiomyocytes of rats with myocardial infarction, increase the expression of p-ERK, decrease the content of ROS in cardiomyocytes, decrease the expression of apoptotic proteins, reduce myocardial infarct size, improve cardiac function, and increase myocardial function.
Collapse
Affiliation(s)
- Zhenjun Wu
- Cardiology Department, Tianjin Huaxing Hospital, Tianjin 300270, China
| | - Lihua Yu
- Beijing Chaoyang Integrative Medicine Emergency Medical Center, Beijing 100020, China
| | - Xinyue Li
- Cardiology Department, Xianshuigu Hospital of Jinnan District, Tianjin 300350, China
| | - Xuewen Li
- Cadre's Ward, Characteristic Medical Center of Chinese People's Armed Police Force, Tijian 300350, China
| |
Collapse
|
23
|
Chen YS, Sun R, Chen WL, Yau YC, Hsu FT, Chung JG, Tsai CJ, Hsieh CL, Chiu YM, Chen JH. The In Vivo Radiosensitizing Effect of Magnolol on Tumor Growth of Hepatocellular Carcinoma. In Vivo 2021; 34:1789-1796. [PMID: 32606148 DOI: 10.21873/invivo.11973] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND/AIM Radiation (RT) induced ERK/NF-κB in hepatocellular carcinoma (HCC) has been reported in our previous works; it weakens the toxicity of RT or triggers a radioresistance effect. Thus, combining RT with a suitable NF-κB inhibitor may sensitize HCC to RT. Magnolol, a bioactive compound, was known to have anti-inflammatory and anti-tumor functions. Here, we aimed to investigate whether magnolol may enhance anti-HCC efficacy of RT in vivo. MATERIALS AND METHODS We established a Hep3B bearing mouse to evaluate the efficacy of the combination treatment of magnolol and RT. RESULTS Most significantly, tumor volume and tumor weight inhibition was found in the combination group. Tumor immunohistochemistry staining also illustrated the suppression of RT-induced ERK/NF-κB-related proteins expression by magnolol. In addition, intrinsic apoptosis-related proteins, such as caspase-3 and -9, were markedly increased in the combination group. CONCLUSION Magnolol may effectively enhance anti-HCC ability of RT by downregulating the expression of ERK/NF-κB-related proteins and increasing the expression of apoptosis-related proteins.
Collapse
Affiliation(s)
- Yu-Shan Chen
- Department of Radiation Oncology, Show Chwan Memorial Hospital, Changhua, Taiwan, R.O.C
| | - Rou Sun
- Department of Medical Imaging, Taipei Medical University Hospital, Taipei, Taiwan, R.O.C
| | - Wei-Lung Chen
- Department of Emergency Medicine, Cathay General Hospital, Taipei, Taiwan, R.O.C.,School of Medicine, Fu Jen Catholic University, Taipei, Taiwan, R.O.C
| | - Yu-Chen Yau
- Department of Radiation Oncology, Chang Bing Show Chwan Memorial Hospital, Lukang, Taiwan, R.O.C
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C
| | - Chia-Jung Tsai
- Department of Medical Imaging and Radiological Sciences, I-Shou University, Kaohsiung, Taiwan, R.O.C
| | - Chia-Ling Hsieh
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, R.O.C. .,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Ying-Ming Chiu
- Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan, R.O.C. .,School of Medicine, China Medical University, Taichung, Taiwan, R.O.C
| | - Jiann-Hwa Chen
- Department of Emergency Medicine, Cathay General Hospital, Taipei, Taiwan, R.O.C. .,School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, R.O.C
| |
Collapse
|
24
|
Limam I, Ben Aissa-Fennira F, Essid R, Chahbi A, Kefi S, Mkadmini K, Elkahoui S, Abdelkarim M. Hydromethanolic root and aerial part extracts from Echium arenarium Guss suppress proliferation and induce apoptosis of multiple myeloma cells through mitochondrial pathway. ENVIRONMENTAL TOXICOLOGY 2021; 36:874-886. [PMID: 33393729 DOI: 10.1002/tox.23090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/19/2020] [Indexed: 06/12/2023]
Abstract
Echium arenarium Guss is a Mediterranean plant traditionally used in healing skin wound and it was reported exhibiting potent antioxidant, antibacterial, and antiparasitic activities. However, antitumoral activities of this plant have not yet been explored. Here we investigated for the first time, root (EARE) and aerial part (EAAPE) extracts of E. arenarium Guss to examine cytotoxicity and apoptosis activation pathway on U266 human multiple myeloma (MM) cell line. We demonstrated that EARE and EAAPE decreased U266 cell viability in a dose dependent manner. Based on 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, EARE was significantly two times more efficient (IC50 value 41 μg/ml) than EAAPE (IC50 value 82 μg/ml) considering 48 h of treatment. Furthermore, after 24 h of exposure to 100 μg/ml of EARE or EAAPE, cell cycle showed remarkable increase in sub-G1 population and a decrease of U266 cells proportion in G1 phase. In addition, EARE increased cell percentage in S phase. Moreover, analysis revealed that EAAPE or EARE induced apoptosis of U266 cells after 24 h of treatment. Interestingly, depolarization of mitochondrial membrane potential and activation of caspase 3/7 were demonstrated in treated U266 cells. Phytochemical analysis of E. arenarium extracts showed that EARE exhibited the highest content of total phenolic content. Interestingly, six phenolic compounds were identified. Myricitrin was the major compound in EARE, followed by luteolin 7-O-glucoside, resorcinol, polydatin, Trans-hydroxycinnamic acid, and hyperoside. These findings proved that an intrinsic mitochondria-mediated apoptosis pathway probably mediated the apoptotic effects of E. arenarium Guss extracts on U266 cells, and this will suggest several action plans to treat MM.
Collapse
Affiliation(s)
- Inès Limam
- Laboratory of oncohematology, Faculty of medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Fatma Ben Aissa-Fennira
- Laboratory of oncohematology, Faculty of medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Rim Essid
- Laboratory of Bioactive Substances, Center of Biotechnology, Ecopark of Borj Cedria, Hammam-Lif, Tunisia
| | - Ahlem Chahbi
- Laboratory of oncohematology, Faculty of medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| | - Sarra Kefi
- Laboratory of Bioactive Substances, Center of Biotechnology, Ecopark of Borj Cedria, Hammam-Lif, Tunisia
| | - Khaoula Mkadmini
- Medicinal and Aromatic Plants Laboratory, Biotechnology Center of Borj Cedria, Hammam-Lif, Tunisia
| | - Salem Elkahoui
- Laboratory of Bioactive Substances, Center of Biotechnology, Ecopark of Borj Cedria, Hammam-Lif, Tunisia
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Mohamed Abdelkarim
- Laboratory of oncohematology, Faculty of medicine of Tunis, Tunis El Manar University, Tunis, Tunisia
| |
Collapse
|
25
|
Induction of Apoptosis, Inhibition of MCL-1, and VEGF-A Expression Are Associated with the Anti-Cancer Efficacy of Magnolol Combined with Regorafenib in Hepatocellular Carcinoma. Cancers (Basel) 2021; 13:cancers13092066. [PMID: 33922992 PMCID: PMC8123296 DOI: 10.3390/cancers13092066] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
While regorafenib was approved for the treatment of advanced HCC in 2017, with a partial response and survival benefit; other combination agents to facilitate the efficacy of regorafenib still need to be explored. Magnolol is a potential natural anti-tumor compound for many types of cancers. Combination indexes calculated on the basis of both in vitro and in vivo models have indicated a synergistic effect of the combination of regorafenib and magnolol. The overexpression of the VEGF-A protein significantly diminished regorafenib's inhibition of cell viability, while the transient knockdown of VEGF-A by siRNA effectively sensitized HCC cells to regorafenib. In addition, the inhibition of MCL-1 by siRNA combined with regorafenib allowed for a significantly greater inhibition of cell growth, compared to regorafenib alone. A lower protein expression level for VEGF-A and MCL-1 was found for the combination treatment of HCC in vitro and in vivo. A superior metastasis inhibition was also found in the combination group, as compared to the single-treatment groups, using a transwell assay, wound healing assay, and Western blotting. The caspase-dependent and -independent and DNA damage effects, as determined by flow cytometry and a comet assay, were increased by the combination therapy. Taken together, magnolol sensitized HCC to regorafenib, which was correlated with the reduction of VEGF-A and MCL-1 and the induction of apoptosis.
Collapse
|
26
|
Li G, Lu Y, Fan Y, Ning Q, Li W. Enhanced oral bioavailability of magnolol via mixed micelles and nanosuspensions based on Soluplus ®-Poloxamer 188. Drug Deliv 2020; 27:1010-1017. [PMID: 32631085 PMCID: PMC7470061 DOI: 10.1080/10717544.2020.1785582] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 11/29/2022] Open
Abstract
Magnolol, known to have extensive biological activities, is the major bioactive ingredient isolated from the root and stem bark of Magnolia officinalis. However, the clinical application of magnolol is limited by poor aqueous solubility and absorption. The aim of this study is to develop novel mixed micelles and nanosuspensions composed of two biocompatible copolymers, Soluplus® and Poloxamer 188, and to improve the solubility and oral bioavailability of magnolol. The magnolol-loaded mixed micelles (MMs) and magnolol nanosuspensions (MNs) were prepared to use film hydration and antisolvent methods, respectively. The optimal MMs and MNs formulations were prepared to use magnolol, Soluplus®, and Poloxamer 188 in ratios of 1:12:5 and 2:1:1, respectively. The average particle size of MMs was 111.8 ± 14.6, and MNs was 78.53 ± 5.4 nm. The entrapment and drug loading efficiency for MMs were 89.58 ± 2.54% and 5.46 ± 0.65%, correspondingly. The drug loading efficiency of MNs was 42.50 ± 1.57%. In the in vitro release study, MMs showed a slow drug release while that of MNs was fast. The results of the Caco-2 transcellular transport study indicated that both MMs and MNs increased the permeation of magnolol. MMs and MNs markedly promoted gastrointestinal drug absorption by 2.85 and 2.27-fold, respectively, as shown in the pharmacokinetics study. These results indicated that both MMs and MNs formulations prepared with Soluplus® and Poloxamer 188 are promising drug delivery systems for improving the oral absorption of insoluble drugs in the gastrointestinal tract.
Collapse
Affiliation(s)
- Guoyuan Li
- State Key Laboratory of Natural Medicines, The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, Department of Marine Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Yuting Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Yongchun Fan
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Qing Ning
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P. R. China
| | - Weiguang Li
- State Key Laboratory of Natural Medicines, The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation of Jiangsu Province, Department of Marine Pharmacy, China Pharmaceutical University, Nanjing, P. R. China
| |
Collapse
|
27
|
GDF15 Repression Contributes to 5-Fluorouracil Resistance in Human Colon Cancer by Regulating Epithelial-Mesenchymal Transition and Apoptosis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2826010. [PMID: 33062674 PMCID: PMC7542494 DOI: 10.1155/2020/2826010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/04/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022]
Abstract
Chemotherapy based on 5-fluorouracil (5-FU) is the standard approach for colon cancer treatment, and resistance to 5-FU is a significant obstacle in the clinical treatment of colon cancer. However, the mechanisms underlying 5-FU resistance in colon cancer cells remain largely unknown. This study aimed at determining whether 5-FU-resistant colon cancer cells undergo epithelial-mesenchymal transition (EMT) and apoptosis and the role of GDF15—a member of the transforming growth factor β/bone morphogenetic protein super family and a protein known to be involved in cancer progression—in the regulation of EMT and apoptosis of these cells, along with the underlying mechanisms. In vitro apoptosis detection assay, growth inhibition assay, transwell, and wound healing experiments revealed that 5-FU-resistant colon cancer cells possessed enhanced EMT and antiapoptotic ability. These cells also showed a stronger tendency to proliferate and metastasize in vivo. Quantitative reverse transcription-PCR and western blotting revealed that 5-FU-resistant colon cancer cells expressed lower levels of growth differentiation factor 15 (GDF15) than did 5-FU-sensitive colon cancer cells. Moreover, the transient GDF15 overexpression resensitized 5-FU-resistant colon cells to 5-FU. Collectively, these findings indicate the mechanism underlying the 5-FU resistance of colon cancer cells and provide new therapeutic targets for improving the prognosis of colon cancer patients.
Collapse
|