1
|
Liu X, Su YX, Yang YM, Li RT, Zhang ZJ. The Small Molecules of Plant Origin with Anti-Glioma Activity. Int J Mol Sci 2025; 26:1942. [PMID: 40076568 PMCID: PMC11900624 DOI: 10.3390/ijms26051942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Gliomas, originating from glial cells, are prevalent and aggressive brain tumors with high recurrence rates and poor prognosis. Despite advancements in surgical, radiation, and chemotherapeutic treatments, the survival rates remain low. Current standard therapies, such as Temozolomide, have limitations due to cytotoxicity, restricted effectiveness, and severe side effects. So, the development of safer anti-glioma agents is the need of the hour. Bioactive compounds of plant origin, either natural or synthetic, have potential implications due to them actively attacking different targets with a wide range of bioactivities, including anti-glioma activities. In this review, for the first time, there is an overall overview of 51 small molecules of plant origin and seven of their synthetic derivatives, represented as anti-glioma agents in the past decades. The goal of the present review is to provide a summary to comprehend the anti-glioma effects of these compounds in addition to providing a reference for preclinical research into novel anti-glioma agents for future clinical application.
Collapse
Affiliation(s)
| | | | | | - Rong-Tao Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (X.L.); (Y.-X.S.); (Y.-M.Y.)
| | - Zhi-Jun Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (X.L.); (Y.-X.S.); (Y.-M.Y.)
| |
Collapse
|
2
|
Sheida A, Farshadi M, Mirzaei A, Najjar Khalilabad S, Zarepour F, Taghavi SP, Hosseini Khabr MS, Ravaei F, Rafiei S, Mosadeghi K, Yazdani MS, Fakhraie A, Ghattan A, Zamani Fard MM, Shahyan M, Rafiei M, Rahimian N, Talaei Zavareh SA, Mirzaei H. Potential of Natural Products in the Treatment of Glioma: Focus on Molecular Mechanisms. Cell Biochem Biophys 2024; 82:3157-3208. [PMID: 39150676 DOI: 10.1007/s12013-024-01447-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
Despite the waning of traditional treatments for glioma due to possible long-term issues, the healing possibilities of substances derived from nature have been reignited in the scientific community. These natural substances, commonly found in fruits and vegetables, are considered potential alternatives to pharmaceuticals, as they have been shown in prior research to impact pathways surrounding cancer progression, metastases, invasion, and resistance. This review will explore the supposed molecular mechanisms of different natural components, such as berberine, curcumin, coffee, resveratrol, epigallocatechin-3-gallate, quercetin, tanshinone, silymarin, coumarin, and lycopene, concerning glioma treatment. While the benefits of a balanced diet containing these compounds are widely recognized, there is considerable scope for investigating the efficacy of these natural products in treating glioma.
Collapse
Affiliation(s)
- Amirhossein Sheida
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Amirhossein Mirzaei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shakiba Najjar Khalilabad
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Zarepour
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Sadat Hosseini Khabr
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Ravaei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Sara Rafiei
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Kimia Mosadeghi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Sepehr Yazdani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Fakhraie
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Ghattan
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Masoud Zamani Fard
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Shahyan
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Rafiei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
3
|
Fan H, Xie X, Kuang X, Du J, Peng F. MicroRNAs, Key Regulators in Glioma Progression as Potential Therapeutic Targets for Chinese Medicine. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:1799-1825. [PMID: 36121713 DOI: 10.1142/s0192415x22500768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Gliomas are tumors of the primary central nervous system associated with poor prognosis and high mortality. The 5-year survival rate of patients with gliomas received surgery combined with chemotherapy or radiotherapy does not exceed 5%. Although temozolomide is commonly used in the treatment of gliomas, the development of resistance limits its use. MicroRNAs are non-coding RNAs involved in numerous processes of glioma cells, such as proliferation, migration and apoptosis. MicroRNAs regulate cell cycle, PI3K/AKT signal pathway, and target apoptosis-related genes (e.g., BCL6), angiogenesis-related genes (e.g., VEGF) and other related genes to suppress gliomas. Evidence illustrates that microRNAs can regulate the sensitivity of gliomas to temozolomide, cisplatin, and carmustine, thereby enhancing the efficacy of these agents. Moreover, traditional Chinese medicine (e.g., tanshinone IIA, xanthohumol, and curcumin) exert antiglioma effects by regulating the expression of microRNAs, and then microRNAs inhibit gliomas through influencing the process of tumors by targeting certain genes. In this paper, the mechanisms through which microRNAs regulate the sensitivity of gliomas to therapeutic drugs are described, and traditional Chinese medicine that can suppress gliomas through microRNAs are discussed. This review aims to provide new insights into the traditional Chinese medicine treatment of gliomas.
Collapse
Affiliation(s)
- Huali Fan
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China
| | - Xiaofang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Xi Kuang
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China
| | - Junrong Du
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China
| | - Fu Peng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China
| |
Collapse
|
4
|
Tabnak P, Masrouri S, Mafakheri A. Natural products in suppressing glioma progression: A focus on the role of microRNAs. Phytother Res 2022; 36:1576-1599. [PMID: 35174549 DOI: 10.1002/ptr.7414] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 12/07/2021] [Accepted: 01/29/2022] [Indexed: 11/06/2022]
Abstract
Glioma is one of the most common malignancies of the central nervous system. Due to inadequate response to the current treatments available, glioma has been at the center of recent cancer studies searching for novel treatment strategies. This has prompted an intensive search using linkage studies and preliminary evidence to gain efficient insight into the mechanisms involved in the alleviation of the pathogenesis of glioma mediated by miRNAs, a group of noncoding RNAs that affect gene expression posttranscriptionally. Dysregulated expression of miRNAs can exacerbate the malignant features of tumor cells in glioma and other cancers. Natural products can exert anticancer effects on glioma cells by stimulating the expression levels of tumor suppressor miRNAs and repressing the expression levels of oncogenic miRNAs. In this review, we aimed to collect and analyze the literature addressing the roles of natural products in the treatment of glioma, with an emphasis on their involvement in the regulation of miRNAs.
Collapse
Affiliation(s)
- Peyman Tabnak
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soroush Masrouri
- Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Asrin Mafakheri
- Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
5
|
Yao H, Li X, Pan X, Xu J, Zhao S, Su Z, Qiu S. Fraxetin exerts anticancer effect in glioma by suppressing MiR-21-3p. Drug Dev Res 2021; 83:501-511. [PMID: 34523750 DOI: 10.1002/ddr.21881] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/02/2021] [Accepted: 09/06/2021] [Indexed: 01/21/2023]
Abstract
Fraxetin (FXT) exerts anticancer function in multiple cancers, but its function on glioma was ill-defined. This article expounded the mechanism by which FXT exerts an anticancer effect in glioma. The effect of gradient concentration of FXT on the viability of glioma cell lines was determined by cell counting kit 8. Effects of FXT on proliferation, apoptosis, and cell cycle in glioma cell lines were determined by colony formation assay, flow cytometry, and Hoechst 33342 staining. Expressions of apoptosis-related gene, cycle-related gene, and glioma-related miRNAs after FXT (25 and 50 μmol/L) treatment were determined by quantitative reverse transcription polymerase chain reaction and western blot as needed. After miR-21-3p overexpression, cell viability and apoptosis of glioma cell lines treated with FXT (50 μmol/L) were tested again. Although 1 μmol/L FXT had no significant effect on cell viability, 5, 10, 25, and 50 μmol/L FXT suppressed cell viability in a concentration-dependent manner. FXT inhibited proliferation, promoted apoptosis, and induced cell cycle arrest in G0/G1 phase in glioma cell lines. These effects may be achieved by elevated expressions of Bax and cleaved caspase-3 and diminished expressions of Bcl-2, Bcl-XL, cyclin E1, cyclin D1, and cyclin-dependent kinase-6. FXT attenuated the contents of miR-21-3p and miR-455-3p, and escalated the contents of miR-124-3p and miR-7-5p. The regulation of FXT on cell viability, proliferation and apoptosis was reversed by miR-21-3p overexpression. FXT suppressed the development of glioma cells by downregulating miR-21-3p.
Collapse
Affiliation(s)
- Hanxun Yao
- Department of Neurosurgery, Huzhou Central Hospital, Huzhou, China
| | - Xiaobin Li
- Department of Neurosurgery, Huzhou Central Hospital, Huzhou, China
| | - Xuyan Pan
- Department of Neurosurgery, Huzhou Central Hospital, Huzhou, China
| | - Jie Xu
- Department of Neurosurgery, Huzhou Central Hospital, Huzhou, China
| | - Shufa Zhao
- Department of Neurosurgery, Huzhou Central Hospital, Huzhou, China
| | - Zhongzhou Su
- Department of Neurosurgery, Huzhou Central Hospital, Huzhou, China
| | - Sheng Qiu
- Department of Neurosurgery, Huzhou Central Hospital, Huzhou, China
| |
Collapse
|
6
|
Li Y, Yang Y, Feng Y, Pu J, Hou LA. Combined effects of Pseudomonas quinolone signal-based quorum quenching and graphene oxide on the mitigation of biofouling and improvement of the application potential for the thin-film composite membrane. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 760:143348. [PMID: 33162137 DOI: 10.1016/j.scitotenv.2020.143348] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 10/20/2020] [Accepted: 10/20/2020] [Indexed: 06/11/2023]
Abstract
Biofouling caused by the growth of the biofilm is the main bottleneck that limits the effective operation of thin-film composite (TFC) membrane in the forward osmosis (FO) process. This study investigated the combined effects of graphene oxide (GO) immobilized thin-film nanocomposite (TFN-S) membrane and Pseudomonas quinolone signal (PQS)-based quorum quenching on biofouling mitigation, especially under the operation of pressure-retarded osmosis (PRO) mode, and the influence of methyl anthranilate (MA) inhibitor on the composition and structure of biofilm was also evaluated. Synthetic wastewater was used as the feed solution, in which the model strain Pseudomonas aeruginosa was added to simulate biofouling. The results showed that GO modification and MA addition both efficiently mitigated flux decline and EPS secretion, but the interference of PQS pathway on biofouling control was better than GO embedding. TFN-S membrane with MA addition exhibited superior anti-biofouling performance based on the combined effects of GO and MA. The alleviated concentration polarization and enhanced hydrophilicity of the TFN-S membrane reduced the flux decline in the early stage. Additionally, the antibacterial property of GO inhibited the viability of the attached bacteria (under PRO mode) and MA further mitigated the EPS secretion and biofilm development in the later stage. In the presence of PQS inhibitor MA, live/total cells ratio was 15% and 13% higher than that of TFC membrane in FO and PRO modes, respectively. Furthermore, exogenous addition of MA led to a relatively loose biofilm structure, resulting in high membrane permeability in the biofouling formation process.
Collapse
Affiliation(s)
- Yuan Li
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing 100875, China.
| | - Yu Yang
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing 100875, China.
| | - Yuruo Feng
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing 100875, China.
| | - Jian Pu
- Institute for Future Initiatives, The University of Tokyo, Tokyo 113-8654, Japan.
| | - Li-An Hou
- State Key Laboratory of Water Environment Simulation, School of Environment, Beijing Normal University, Beijing 100875, China; Xi'an High-Tech Institute, Xi'an 710025, China.
| |
Collapse
|
7
|
Chelliah SS, Paul EAL, Kamarudin MNA, Parhar I. Challenges and Perspectives of Standard Therapy and Drug Development in High-Grade Gliomas. Molecules 2021; 26:1169. [PMID: 33671796 PMCID: PMC7927069 DOI: 10.3390/molecules26041169] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 12/18/2022] Open
Abstract
Despite their low incidence rate globally, high-grade gliomas (HGG) remain a fatal primary brain tumor. The recommended therapy often is incapable of resecting the tumor entirely and exclusively targeting the tumor leads to tumor recurrence and dismal prognosis. Additionally, many HGG patients are not well suited for standard therapy and instead, subjected to a palliative approach. HGG tumors are highly infiltrative and the complex tumor microenvironment as well as high tumor heterogeneity often poses the main challenges towards the standard treatment. Therefore, a one-fit-approach may not be suitable for HGG management. Thus, a multimodal approach of standard therapy with immunotherapy, nanomedicine, repurposing of older drugs, use of phytochemicals, and precision medicine may be more advantageous than a single treatment model. This multimodal approach considers the environmental and genetic factors which could affect the patient's response to therapy, thus improving their outcome. This review discusses the current views and advances in potential HGG therapeutic approaches and, aims to bridge the existing knowledge gap that will assist in overcoming challenges in HGG.
Collapse
Affiliation(s)
- Shalini Sundramurthi Chelliah
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
- School of Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Ervin Ashley Lourdes Paul
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
| | - Muhamad Noor Alfarizal Kamarudin
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
| | - Ishwar Parhar
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
| |
Collapse
|
8
|
Bae H, Lee JY, Song J, Song G, Lim W. Osthole interacts with an ER-mitochondria axis and facilitates tumor suppression in ovarian cancer. J Cell Physiol 2020; 236:1025-1042. [PMID: 32697363 DOI: 10.1002/jcp.29913] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 12/29/2022]
Abstract
Osthole is a natural coumarin found in a variety of plants and has been reported to have diverse biological functions, including antimicrobial, antiviral, immunomodulatory, and anticancer effects. Here, we investigated the natural derivative osthole as a promising anticancer compound against ovarian cancer and evaluated its ability to suppress and abrogate tumor progression. In addition, we found the endoplasmic reticulum-mitochondrial axis-mediated anticancer mechanisms of osthole against ES2 and OV90 ovarian cancer cells and demonstrated its calcium-dependent pharmacological potential. Mechanistically, osthole was found to target the phosphatidylinositol 3-kinase/mitogen-activated protein kinase signaling pathway to facilitate tumor suppression in ovarian cancer. Furthermore, we identified the effects of osthole in a three-dimensional tumor-formation model using the zebrafish xenograft assay, providing convincing evidence of the pharmacological effects of osthole within the anchorage-independent tumor microenvironment. These findings suggest that osthole has strong potential as a pharmacological agent for targeting ovarian cancer.
Collapse
Affiliation(s)
- Hyocheol Bae
- Department of Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jin-Young Lee
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jisoo Song
- Department of Food and Nutrition, Kookmin University, Seoul, Republic of Korea
| | - Gwonhwa Song
- Department of Biotechnology, Korea University, Seoul, Republic of Korea
| | - Whasun Lim
- Department of Food and Nutrition, Kookmin University, Seoul, Republic of Korea
| |
Collapse
|
9
|
Ye J, Sun D, Yu Y, Yu J. Osthole resensitizes CD133 + hepatocellular carcinoma cells to cisplatin treatment via PTEN/AKT pathway. Aging (Albany NY) 2020; 12:14406-14417. [PMID: 32673286 PMCID: PMC7425450 DOI: 10.18632/aging.103484] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/27/2020] [Indexed: 04/12/2023]
Abstract
The population of CD133 positive cancer cells has been reported to be responsible for drug resistance of hepatocellular carcinoma (HCC). However, the potential molecular mechanism by which CD133+ HCC cells develop drug resistance is still unclear. In this study, we found that CD133+ HepG2 and Huh7 cells were resistant to cisplatin treatment, compared to the CD133- HepG2 and Huh7 cells. However, treatment with osthole, a natural coumarin isolated from umbelliferae plant monomers, was found to resensitize CD133+ HepG2 and Huh7 cells to cisplatin treatment. In the mechanism research, we found that treatment with osthole increased the expression of PTEN. As a result, osthole inhibited the phosphorylation of AKT and Bad to decrease the amount of free Bcl-2 in CD133+ HepG2 and Huh7 cells. Finally, cisplatin-induced mitochondrial apoptosis was expanded. In conclusion, combination treatment with osthole can resensitize CD133+ HCC cells to cisplatin treatment via the PTEN/AKT pathway.
Collapse
Affiliation(s)
- Junfeng Ye
- Department of Hepato-Biliary-Pancreatic Surgery, First Hospital Jilin University, Changchun 130021, Jilin Province, China
| | - Di Sun
- Department of Colorectal and Anal Surgery, First Hospital Jilin University, Changchun 130021, Jilin Province,130021, China
| | - Ying Yu
- Department of Hepato-Biliary-Pancreatic Surgery, First Hospital Jilin University, Changchun 130021, Jilin Province, China
| | - Jinhai Yu
- Department of Gastrointestinal Surgery, First Hospital Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
10
|
Sumorek-Wiadro J, Zając A, Bądziul D, Langner E, Skalicka-Woźniak K, Maciejczyk A, Wertel I, Rzeski W, Jakubowicz-Gil J. Coumarins modulate the anti-glioma properties of temozolomide. Eur J Pharmacol 2020; 881:173207. [PMID: 32446712 DOI: 10.1016/j.ejphar.2020.173207] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/08/2020] [Accepted: 05/14/2020] [Indexed: 12/14/2022]
Abstract
In the recent years, coumarin bioactive compounds have been identified to posess anticancer properties. Therefore, the aim of the present study was to investigate for the first time the efficacy of osthole, umbelliferone, esculin, and 4-hydroxycoumarin, alone and in combination with Temozolomide, in the elimination of deadly brain tumors, anaplastic astrocytoma (AA) and glioblastoma multiforme (GBM) cells via programmed death. Our results indicated that osthole, umbelliferone, esculin, and 4-hydroxycoumarin initiated mainly apoptosis in the T98G and MOGGCCM cells. Osthole was the most effective. It also initiated autophagy in a small percentage of the cell population. The co-incubation with Temozolomide did not increase the pro-apoptotic potential of natural compounds but decreased the level of autophagy in the T98G cells. Apoptosis was associated with reduced mitochondrial membrane potential, activation of caspase 3, inhibition of Bcl-2 expression and the presence of a Bcl-2/Beclin 1. Blocking of Bcl-2 expression resulted in promotion of apoptosis, but not autophagy, in the MOGGCCM and T98G lines. It also sensitized astrocytoma cells, but not GBM, to the combined osthole and TMZ treatment, which was correlated with a reduced level of Beclin 1 and increased expression of caspase 3. Osthole and TMZ, alone and in combination, inhibited the migratory phenotype of the GBM and AA cells. In summary, our results indicated that osthole effectively eliminated glioma cells via apoptosis, what was correlated with Bcl-2/Beclin 1 complex formation. Considering the anti-migratory effect, osthole and Temozolomide display antiglioma potential but it needs further extensive studies.
Collapse
Affiliation(s)
- Joanna Sumorek-Wiadro
- Department of Functional Anatomy and Cytobiology, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland.
| | - Adrian Zając
- Department of Functional Anatomy and Cytobiology, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland.
| | - Dorota Bądziul
- Department of Biology, Institute of Medical Sciences, Medical College of Rzeszow University, Rejtana 16 C, 35-959, Rzeszów, Poland.
| | - Ewa Langner
- Department of Medical Biology, Institute of Agricultural Medicine, Jaczewskiego 2, 20-950, Lublin, Poland.
| | - Krystyna Skalicka-Woźniak
- Independent Laboratory of Natural Products Chemistry, Medical University of Lublin, Chodzki 1, 20-093, Lublin, Poland.
| | - Aleksandra Maciejczyk
- Department of Functional Anatomy and Cytobiology, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland.
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, 1st Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Staszica 16, 20-081, Lublin, Poland.
| | - Wojciech Rzeski
- Department of Functional Anatomy and Cytobiology, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland; Department of Medical Biology, Institute of Agricultural Medicine, Jaczewskiego 2, 20-950, Lublin, Poland.
| | - Joanna Jakubowicz-Gil
- Department of Functional Anatomy and Cytobiology, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland.
| |
Collapse
|
11
|
Yool AJ, Ramesh S. Molecular Targets for Combined Therapeutic Strategies to Limit Glioblastoma Cell Migration and Invasion. Front Pharmacol 2020; 11:358. [PMID: 32292341 PMCID: PMC7118801 DOI: 10.3389/fphar.2020.00358] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/10/2020] [Indexed: 12/21/2022] Open
Abstract
The highly invasive nature of glioblastoma imposes poor prospects for patient survival. Molecular evidence indicates glioblastoma cells undergo an intriguing expansion of phenotypic properties to include neuron-like signaling using excitable membrane ion channels and synaptic proteins, augmenting survival and motility. Neurotransmitter receptors, membrane signaling, excitatory receptors, and Ca2+ responses are important candidates for the design of customized treatments for cancers within the heterogeneous central nervous system. Relatively few published studies of glioblastoma multiforme (GBM) have evaluated pharmacological agents targeted to signaling pathways in limiting cancer cell motility. Transcriptomic analyses here identified classes of ion channels, ionotropic receptors, and synaptic proteins that are enriched in human glioblastoma biopsy samples. The pattern of GBM-enriched gene expression points to a major role for glutamate signaling. However, the predominant role of AMPA receptors in fast excitatory signaling throughout the central nervous system raises a challenge on how to target inhibitors selectively to cancer cells while maintaining tolerability. This review critically evaluates a panel of ligand- and voltage-gated ion channels and synaptic proteins upregulated in GBM, and the evidence for their potential roles in the pathological disease progress. Evidence suggests combinations of therapies could be more effective than single agents alone. Natural plant products used in traditional medicines for the treatment of glioblastoma contain flavonoids, terpenoids, polyphenols, epigallocatechin gallate, quinones, and saponins, which might serendipitously include agents that modulate some classes of signaling compounds highlighted in this review. New therapeutic strategies are likely to exploit evidence-based combinations of selected agents, each at a low dose, to create new cancer cell-specific therapeutics.
Collapse
Affiliation(s)
- Andrea J. Yool
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Sunita Ramesh
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- College of Science and Engineering, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
12
|
Sun Y, Yang AWH, Lenon GB. Phytochemistry, Ethnopharmacology, Pharmacokinetics and Toxicology of Cnidium monnieri (L.) Cusson. Int J Mol Sci 2020; 21:E1006. [PMID: 32028721 PMCID: PMC7037677 DOI: 10.3390/ijms21031006] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/24/2020] [Accepted: 01/27/2020] [Indexed: 01/30/2023] Open
Abstract
Cnidium monnieri (L.) Cusson (CMC) is a traditional Chinese herbal medicine that has been widely grown and used in Asia. It is also known as "She chuang zi" in China (Chinese: ), "Jashoshi" in Japan, "Sasangia" in Korea, and "Xa sang tu" in Vietnam. This study aimed to provide an up-to-date review of its phytochemistry, ethnopharmacology, pharmacokinetics, and toxicology. All available information on CMC was collected from the Encyclopedia of Traditional Chinese Medicines, PubMed, EMBASE, ScienceDirect, Scopus, Web of Science, and China Network Knowledge Infrastructure. The updated chemical structures of the compounds are those ones without chemical ID numbers or references from the previous review. A total of 429 chemical constituents have been elucidated and 56 chemical structures have been firstly identified in CMC with traceable evidence. They can be categorized as coumarins, volatile constituents, liposoluble compounds, chromones, monoterpenoid glucosides, terpenoids, glycosides, glucides, and other compounds. CMC has demonstrated impressive potential for the management of various diseases in extensive preclinical research. Since most of the studies are overly concentrated on osthole, more research is needed to investigate other chemical constituents.
Collapse
Affiliation(s)
| | | | - George Binh Lenon
- School of Health and Biomedical Sciences, RMIT University, Melbourne 3083, Australia; (Y.S.); (A.W.H.Y.)
| |
Collapse
|
13
|
Lu K, Lin J, Jiang J. Osthole inhibited cell proliferation and induced cell apoptosis through decreasing CPEB2 expression via up-regulating miR-424 in endometrial carcinoma. J Recept Signal Transduct Res 2020; 40:89-96. [PMID: 31971049 DOI: 10.1080/10799893.2019.1710846] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Objective: Endometrial carcinoma (EC) was the fourth female malignancies in developed countries. Given that the prognosis of EC is extremely poor, it is vital to investigate its pathogenesis and effective therapeutic targets. However, the mechanism of osthole in EC remains unknown.Materials and methods: Firstly, the different doses of osthole (0, 50, 100, and 200 μM) were used to treat the Ishikawa and KLE cells. The cell proliferation, apoptosis, and cell cycle were measured by cell counting kit-8 (CCK-8), Annexin V-FITC/PI, and cell cycle assays. The apoptosis-related protein levels were examined by western blot. The miR-424 levels in Ishikawa and KLE cells were assessed by quantitative RT-PCR (qRT-PCR). Also, the binding of miR-424 and cytoplasmic polyadenylation element binding protein 2 (CEPB2) was detected by the luciferase reporter assay.Results: In this study, the increasing dose of osthole inhibited proliferation and induced apoptosis of Ishikawa and KLE cells. Moreover, the increasing dose of osthole up-regulated miR-424 and down-regulated the expression of CPEB2. CPEB2 was proved to be the target gene of miR-424. Interestingly, the over-expression of CPEB2 could reverse the changes of osthole-induced proliferation and apoptosis of Ishikawa and KLE cells.Conclusions: In summary, we provided first evidences that osthole inhibited proliferation and induced apoptosis through up-regulating miR-424 to inhibit expression of CPEB2 in EC. Our findings indicated that osthole might act as a novel and potential therapeutic agent for the treatment of EC.
Collapse
Affiliation(s)
- Kena Lu
- Department of Gynecology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi Zhuang Autonomous Region, Nanning City, China
| | - Jiajing Lin
- Department of Gynecology, Liuzhou Worker's Hospital, Guangxi Zhuang Autonomous Region, Liuzhou City, China
| | - Jun Jiang
- Department of Gynecology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou City, China
| |
Collapse
|
14
|
Ashrafizadeh M, Mohammadinejad R, Samarghandian S, Yaribeygi H, Johnston TP, Sahebkar A. Anti-Tumor Effects of Osthole on Different Malignant Tissues: A Review of Molecular Mechanisms. Anticancer Agents Med Chem 2020; 20:918-931. [PMID: 32108003 DOI: 10.2174/1871520620666200228110704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/09/2019] [Accepted: 01/29/2020] [Indexed: 12/16/2022]
Abstract
Cancer management and/or treatment require a comprehensive understanding of the molecular and signaling pathways involved. Recently, much attention has been directed to these molecular and signaling pathways, and it has been suggested that a number of biomolecules/players involved in such pathways, such as PI3K/Akt, NF-kB, STAT, and Nrf2 contribute to the progression, invasion, proliferation, and metastasis of malignant cells. Synthetic anti-tumor agents and chemotherapeutic drugs have been a mainstay in cancer therapy and are widely used to suppress the progression and, hopefully, halt the proliferation of malignant cells. However, these agents have some undesirable side-effects and, therefore, naturally-occurring compounds with high potency and fewer side-effects are now of great interest. Osthole is a plant-derived chemical compound that can inhibit the proliferation of malignant cells and provide potent anti-cancer effects in various tissues. Therefore, in this review, we presented the main findings concerning the potential anti-tumor effects of osthole and its derivatives and described possible molecular mechanisms by which osthole may suppress malignant cell proliferation in different tissues.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Saeed Samarghandian
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Habib Yaribeygi
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, United States
| | | |
Collapse
|
15
|
Agarwal S, Muniyandi P, Maekawa T, Kumar DS. Vesicular systems employing natural substances as promising drug candidates for MMP inhibition in glioblastoma: A nanotechnological approach. Int J Pharm 2018; 551:339-361. [PMID: 30236647 DOI: 10.1016/j.ijpharm.2018.09.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 09/14/2018] [Accepted: 09/15/2018] [Indexed: 12/16/2022]
Abstract
Glioblastoma multiforme (GBM), one of the most lethal Brain tumors, characterized by its high invasive nature and increased mortality rates forms a major bottleneck in transport of therapeutics across the Blood Brain Barrier (BBB). Matrix metalloproteinases (MMPs) are classified as enzymes, which are found to be up regulated in the Glioma tumor microenvironment and thus can be considered as a target for inhibition for curbing GBM. Many chemotherapeutics and techniques have been employed for inhibiting MMPs till now but all of them failed miserably and were withdrawn in clinical trials due to their inability in restricting the tumor growth or increasing the overall survival rates. Thus, the quest for finding the suitable MMP inhibitor is still on and there is a critical need for identification of novel compounds which can alter the BBB permeability, restrain tumor growth and prevent tumor recurrence. Currently, naturally derived substances are gaining widespread attention as tumor inhibitors and many studies have been reported by far highlighting their importance in restricting MMP expression thus serving as chemotherapeutics for cancer due to their minimal toxicity. These substances may serve as probable candidates for inhibiting MMP expression in GBM. However, targeting and delivering the inhibitor to its target site is an issue that needs to be overcome in order to attain maximum specificity and sustained release. The birth of nanotechnology served as a boon in delivering drugs to the most complicated areas thus paving way for Nano drug delivery. An efficient Nano carrier with ability to cross the BBB and competently kill the Glioma cells forms the prerequisite for GBM chemotherapy. Vesicular drug delivery systems are one such class of carriers, which have the capacity to release the drug at a predetermined rate at the target site thus minimizing any undesirable side effects. Exploiting vesicular systems as promising Nano drug carriers to formulate naturally derived substances, that can bypass the BBB and act as an inhibitor against MMPs in GBM is the main theme of this review.
Collapse
Affiliation(s)
- Srishti Agarwal
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama 350-8585, Japan
| | - Priyadharshni Muniyandi
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama 350-8585, Japan
| | - Toru Maekawa
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama 350-8585, Japan
| | - D Sakthi Kumar
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama 350-8585, Japan.
| |
Collapse
|
16
|
Noruzi S, Azizian M, Mohammadi R, Hosseini SA, Rashidi B, Mohamadi Y, Nesaei A, Seiri P, Sahebkar A, Salarinia R, Aghdam AM, Mirzaei H. Micro-RNAs as critical regulators of matrix metalloproteinases in cancer. J Cell Biochem 2018; 119:8694-8712. [PMID: 30132957 DOI: 10.1002/jcb.27182] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 05/24/2018] [Indexed: 12/25/2022]
Abstract
Metastasis is known to be one of the important factors associated with cancer-related deaths worldwide. Several cellular and molecular targets are involved in the metastasis process. Among these targets, matrix metalloproteinases (MMPs) play central roles in promoting cancer metastasis. MMPs could contribute toward tumor growth, angiogenesis, migration, and invasion via degradation of the extracellular matrix and activation of pre-pro-growth factors. Therefore, identification of various cellular and molecular pathways that affect MMPs could contribute toward a better understanding of the metastatic pathways involved in various tumors. Micro-RNAs are important targets that could affect MMPs. Multiple lines of evidence have indicated that deregulation of various micro-RNAs, including miR-9, Let-7, miR-10b, and miR-15b, affects metastasis of tumor cells via targeting MMPs.
Collapse
Affiliation(s)
- Somaye Noruzi
- Department of Medical Biotechnology and Molecular Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnourd, Iran
| | - Mitra Azizian
- Department of Clinical Biochemistry, Ftabaculty of Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Rezvan Mohammadi
- Department of Medical Biotechnology and Molecular Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnourd, Iran
| | - Seyede Atefe Hosseini
- Department of Medical Biotechnology and Molecular Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnourd, Iran
| | - Bahman Rashidi
- Department of Anatomical Sciences, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yousef Mohamadi
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Anatomy, Faculty of medicine, Qom University of Medical Sciences, Qom, Iran
| | - Abolfazl Nesaei
- Department of Basic Sciences, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Parvaneh Seiri
- Department of Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Salarinia
- Department of Medical Biotechnology and Molecular Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnourd, Iran
| | - Arad Mobasher Aghdam
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Department of Biomaterials, Tissue Engineering and Nanotechnology, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
17
|
Vengoji R, Macha MA, Batra SK, Shonka NA. Natural products: a hope for glioblastoma patients. Oncotarget 2018; 9:22194-22219. [PMID: 29774132 PMCID: PMC5955138 DOI: 10.18632/oncotarget.25175] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 03/28/2018] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive malignant tumors with an overall dismal survival averaging one year despite multimodality therapeutic interventions including surgery, radiotherapy and concomitant and adjuvant chemotherapy. Few drugs are FDA approved for GBM, and the addition of temozolomide (TMZ) to standard therapy increases the median survival by only 2.5 months. Targeted therapy appeared promising in in vitro monolayer cultures, but disappointed in preclinical and clinical trials, partly due to the poor penetration of drugs through the blood brain barrier (BBB). Cancer stem cells (CSCs) have intrinsic resistance to initial chemoradiation therapy (CRT) and acquire further resistance via deregulation of many signaling pathways. Due to the failure of classical chemotherapies and targeted drugs, research efforts focusing on the use of less toxic agents have increased. Interestingly, multiple natural compounds have shown antitumor and apoptotic effects in TMZ resistant and p53 mutant GBM cell lines and also displayed synergistic effects with TMZ. In this review, we have summarized the current literature on natural products or product analogs used to modulate the BBB permeability, induce cell death, eradicate CSCs and sensitize GBM to CRT.
Collapse
Affiliation(s)
- Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Muzafar A. Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Otolaryngology/Head and Neck Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Eppley Institute for Research in Cancer and Allied Diseases and Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Nicole A. Shonka
- Eppley Institute for Research in Cancer and Allied Diseases and Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
18
|
Shokoohinia Y, Jafari F, Mohammadi Z, Bazvandi L, Hosseinzadeh L, Chow N, Bhattacharyya P, Farzaei MH, Farooqi AA, Nabavi SM, Yerer MB, Bishayee A. Potential Anticancer Properties of Osthol: A Comprehensive Mechanistic Review. Nutrients 2018; 10:E36. [PMID: 29301373 PMCID: PMC5793264 DOI: 10.3390/nu10010036] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/15/2017] [Accepted: 12/29/2017] [Indexed: 01/13/2023] Open
Abstract
Cancer is caused by uncontrolled cell proliferation which has the potential to occur in different tissues and spread into surrounding and distant tissues. Despite the current advances in the field of anticancer agents, rapidly developing resistance against different chemotherapeutic drugs and significantly higher off-target effects cause millions of deaths every year. Osthol is a natural coumarin isolated from Apiaceaous plants which has demonstrated several pharmacological effects, such as antineoplastic, anti-inflammatory and antioxidant properties. We have attempted to summarize up-to-date information related to pharmacological effects and molecular mechanisms of osthol as a lead compound in managing malignancies. Electronic databases, including PubMed, Cochrane library, ScienceDirect and Scopus were searched for in vitro, in vivo and clinical studies on anticancer effects of osthol. Osthol exerts remarkable anticancer properties by suppressing cancer cell growth and induction of apoptosis. Osthol's protective and therapeutic effects have been observed in different cancers, including ovarian, cervical, colon and prostate cancers as well as chronic myeloid leukemia, lung adenocarcinoma, glioma, hepatocellular, glioblastoma, renal and invasive mammary carcinoma. A large body of evidence demonstrates that osthol regulates apoptosis, proliferation and invasion in different types of malignant cells which are mediated by multiple signal transduction cascades. In this review, we set spotlights on various pathways which are targeted by osthol in different cancers to inhibit cancer development and progression.
Collapse
Affiliation(s)
- Yalda Shokoohinia
- Pharmaceutical Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 67146, Iran.
- Department of Pharmacognosy and Biotechnology, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 67146, Iran.
| | - Fataneh Jafari
- Pharmaceutical Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 67146, Iran.
| | - Zeynab Mohammadi
- Students Research Committee, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 67146, Iran.
| | - Leili Bazvandi
- Students Research Committee, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 67146, Iran.
| | - Leila Hosseinzadeh
- Pharmaceutical Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 67146, Iran.
| | - Nicholas Chow
- Department of Clinical and Administrative Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA.
| | | | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah 67146, Iran.
| | - Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore 54000, Pakistan.
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran.
| | - Mükerrem Betül Yerer
- Department of Pharmacology, Faculty of Pharmacy, University of Erciyes, 38039 Kayseri, Turkey.
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL 33169, USA.
| |
Collapse
|
19
|
Zhu X, Song X, Xie K, Zhang X, He W, Liu F. Osthole induces apoptosis and suppresses proliferation via the PI3K/Akt pathway in intrahepatic cholangiocarcinoma. Int J Mol Med 2017; 40:1143-1151. [PMID: 28902342 PMCID: PMC5593451 DOI: 10.3892/ijmm.2017.3113] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 07/28/2017] [Indexed: 12/20/2022] Open
Abstract
Osthole is a natural coumarin isolated from Umbelliferae plant monomers. Previous research has indicated that osthole exerts a wide variety of biological effects, acting as anti-seizure, anti-osteoporosis and anti-inflammation. However, the regulatory effect and related molecular mechanism of osthole in intrahepatic cholangiocarcinoma (ICC) remain unknown. In the present study, the authors found that osthole inhibited ICC cell lines in a dose- and time-dependent manner. Osthole also significantly induced mitochondrial-dependent apoptosis by upregulating Bax, cleaved caspase-3, cleaved caspase-9, and cleaved poly ADP-ribose polymerase expression, and by downregulating Bcl-2 expression. Moreover, the levels of p-Akt and PI3K were significantly decreased, while total Akt protein levels were unchanged. Following transfection with wild-type-Akt and constitutively active (CA)-Akt plasmids, the effects of osthole were decreased. Osthole was also able to suppress tumor growth in vivo. Together, these data demonstrated that osthole induces mitochondrial-dependent apoptosis via the PI3K/Akt pathway, suggesting that osthole may represent a novel and effective agent for the treatment of ICC.
Collapse
Affiliation(s)
- Xingyang Zhu
- Department of General Surgery, The First Affiliated Hospital of Medical University of Anhui, Hefei, Anhui 230022, P.R. China
| | - Xiaoling Song
- Department of Cardiothoracic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200082, P.R. China
| | - Kun Xie
- Department of General Surgery, The First Affiliated Hospital of Medical University of Anhui, Hefei, Anhui 230022, P.R. China
| | - Xue Zhang
- Department of General Surgery, The First Affiliated Hospital of Medical University of Anhui, Hefei, Anhui 230022, P.R. China
| | - Wei He
- Department of General Surgery, The First Affiliated Hospital of Medical University of Anhui, Hefei, Anhui 230022, P.R. China
| | - Fubao Liu
- Department of General Surgery, The First Affiliated Hospital of Medical University of Anhui, Hefei, Anhui 230022, P.R. China
- Correspondence to: Professor Fubao Liu, Department of General Surgery, The First Affiliated Hospital of Medical University of Anhui, 218 Jixi Road, Hefei, Anhui 230022, P.R. China, E-mail:
| |
Collapse
|
20
|
Zhao X, Chen R, Liu M, Feng J, Chen J, Hu K. Remodeling the blood-brain barrier microenvironment by natural products for brain tumor therapy. Acta Pharm Sin B 2017; 7:541-553. [PMID: 28924548 PMCID: PMC5595291 DOI: 10.1016/j.apsb.2017.07.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/08/2017] [Accepted: 07/12/2017] [Indexed: 12/23/2022] Open
Abstract
Brain tumor incidence shows an upward trend in recent years; brain tumors account for 5% of adult tumors, while in children, this figure has increased to 70%. Moreover, 20%-30% of malignant tumors will eventually metastasize into the brain. Both benign and malignant tumors can cause an increase in intracranial pressure and brain tissue compression, leading to central nervous system (CNS) damage which endangers the patients' lives. Despite the many approaches to treating brain tumors and the progress that has been made, only modest gains in survival time of brain tumor patients have been achieved. At present, chemotherapy is the treatment of choice for many cancers, but the special structure of the blood-brain barrier (BBB) limits most chemotherapeutic agents from passing through the BBB and penetrating into tumors in the brain. The BBB microenvironment contains numerous cell types, including endothelial cells, astrocytes, peripheral cells and microglia, and extracellular matrix (ECM). Many chemical components of natural products are reported to regulate the BBB microenvironment near brain tumors and assist in their treatment. This review focuses on the composition and function of the BBB microenvironment under both physiological and pathological conditions, and the current research progress in regulating the BBB microenvironment by natural products to promote the treatment of brain tumors.
Collapse
Affiliation(s)
- Xiao Zhao
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Rujing Chen
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mei Liu
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jianfang Feng
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jun Chen
- Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai 201203, China
| | - Kaili Hu
- Murad Research Center for Modernized Chinese Medicine, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
21
|
Lin ZK, Liu J, Jiang GQ, Tan G, Gong P, Luo HF, Li HM, Du J, Ning Z, Xin Y, Wang ZY. Osthole inhibits the tumorigenesis of hepatocellular carcinoma cells. Oncol Rep 2017; 37:1611-1618. [DOI: 10.3892/or.2017.5403] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 01/09/2017] [Indexed: 11/06/2022] Open
|