1
|
Lee SJ, Pak SW, Kim WI, Park SH, Cho YK, Ko JW, Kim TW, Kim JS, Kim JC, Lim JO, Shin IS. Silibinin Suppresses Inflammatory Responses Induced by Exposure to Asian Sand Dust. Antioxidants (Basel) 2024; 13:1187. [PMID: 39456441 PMCID: PMC11505622 DOI: 10.3390/antiox13101187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024] Open
Abstract
Asian sand dust (ASD), generated from the deserts of China and Mongolia, affects Korea and Japan during spring and autumn, causing harmful effects on various bio-organs, including the respiratory system, due to its irritants such as fine dust, chemicals, and toxic materials. Here, we investigated the therapeutic effects of silibinin against ASD-induced airway inflammation using mouse macrophage-like cell line RAW264.7 and a murine model. ASD was intranasally administered to mice three times a week and silibinin was administered for 6 days by oral gavage. In ASD-stimulated RAW264.7 cells, silibinin treatment decreased tumor necrosis factor-α production and reduced the expression of p-p65NF-κB, p-p38, and cyclooxygenase (COX)-2, while increasing heme oxygenase (HO)-1 expression. In ASD-exposed mice, silibinin administration reduced inflammatory cell count and cytokines in bronchoalveolar lavage fluid and decreased inflammatory cell infiltration in lung tissue. Additionally, silibinin lowered oxidative stress, as evidenced by decreased 8-hydroxy-2'-deoxyguanosin (8-OHdG) expression and increased HO-1 expression. The expression of inflammatory-related proteins, including p-p65NF-κB, COX-2, and p-p38, was markedly reduced by silibinin administration. Overall, silibinin treatment reduced the expression of p-p65NF-κB, COX-2, and p-p38 in response to ASD exposure, while increasing HO-1 expression both in vitro and in vivo. These findings suggest that silibinin mitigates pulmonary inflammation caused by ASD exposure by reducing inflammatory signaling and oxidative stress, indicating its potential as a therapeutic agent for ASD-induced pulmonary inflammation.
Collapse
Affiliation(s)
- Se-Jin Lee
- BK21 FOUR Program, College of Veterinary Medicine, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju 61186, Republic of Korea; (S.-J.L.); (S.-W.P.); (W.-I.K.); (S.-H.P.); (J.-S.K.); (J.-C.K.)
| | - So-Won Pak
- BK21 FOUR Program, College of Veterinary Medicine, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju 61186, Republic of Korea; (S.-J.L.); (S.-W.P.); (W.-I.K.); (S.-H.P.); (J.-S.K.); (J.-C.K.)
| | - Woong-Il Kim
- BK21 FOUR Program, College of Veterinary Medicine, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju 61186, Republic of Korea; (S.-J.L.); (S.-W.P.); (W.-I.K.); (S.-H.P.); (J.-S.K.); (J.-C.K.)
| | - Sin-Hyang Park
- BK21 FOUR Program, College of Veterinary Medicine, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju 61186, Republic of Korea; (S.-J.L.); (S.-W.P.); (W.-I.K.); (S.-H.P.); (J.-S.K.); (J.-C.K.)
| | - Young-Kwon Cho
- College of Health Sciences, Cheongju University, 298 Daesung-ro, Sangdang-gu, Cheongju-si 28503, Republic of Korea;
| | - Je-Won Ko
- BK21 FOUR Program, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Daejeon 34134, Republic of Korea; (J.-W.K.); (T.-W.K.)
| | - Tae-Won Kim
- BK21 FOUR Program, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Daejeon 34134, Republic of Korea; (J.-W.K.); (T.-W.K.)
| | - Joong-Sun Kim
- BK21 FOUR Program, College of Veterinary Medicine, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju 61186, Republic of Korea; (S.-J.L.); (S.-W.P.); (W.-I.K.); (S.-H.P.); (J.-S.K.); (J.-C.K.)
| | - Jong-Choon Kim
- BK21 FOUR Program, College of Veterinary Medicine, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju 61186, Republic of Korea; (S.-J.L.); (S.-W.P.); (W.-I.K.); (S.-H.P.); (J.-S.K.); (J.-C.K.)
| | - Je-Oh Lim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, 177 Geonjae-ro, Naju-si 58245, Republic of Korea
| | - In-Sik Shin
- BK21 FOUR Program, College of Veterinary Medicine, Chonnam National University, 77 Yong-bong-ro, Buk-gu, Gwangju 61186, Republic of Korea; (S.-J.L.); (S.-W.P.); (W.-I.K.); (S.-H.P.); (J.-S.K.); (J.-C.K.)
| |
Collapse
|
2
|
Sharma A, Wairkar S. Flavonoids for treating pulmonary fibrosis: Present status and future prospects. Phytother Res 2024; 38:4406-4423. [PMID: 38986681 DOI: 10.1002/ptr.8285] [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] [Received: 12/27/2023] [Revised: 06/08/2024] [Accepted: 06/18/2024] [Indexed: 07/12/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with an unknown underlying cause. There is no complete cure for IPF; however, two anti-fibrotic agents (Nintedanib and pirfenidone) are approved by the USFDA to extend the patient's life span. Therefore, alternative therapies supporting the survival of fibrotic patients have been studied in recent literature. The abundance of phenolic compounds, particularly flavonoids, has gathered attention due to their potential health benefits. Various flavonoids, like naringin, quercetin, baicalin, baicalein, puerarin, silymarin, and kaempferol, exhibit anti-inflammatory and anti-oxidant properties, which help decrease lung fibrosis. Various databases, including PubMed, EBSCO, ProQuest, and Scopus, as well as particular websites, such as the World Health Organisation and the National Institutes of Health, were used to conduct a literature search. Several mechanisms of action of flavonoids are reported with the help of in vivo and cell line studies emphasizing their ability to modulate oxidative stress, inflammation, and fibrotic processes in the lungs. They are reported for the restoration of biomarkers like hydroxyproline, cytokines, superoxide dismutase, malondialdehyde and others associated with IPF and for modulating various pathways responsible for the progression of pulmonary fibrosis. Yet, flavonoids have some drawbacks, such as poor solubility, challenging drug loading, stability issues, and scarce bioavailability. Therefore, novel formulations of flavonoids are explored, including liposomes, solid lipid microparticles, polymeric nanoparticles, nanogels, and nanocrystals, to enhance the therapeutic efficacy of flavonoids in pulmonary fibrosis. This review focuses on the role of flavonoids in mitigating idiopathic pulmonary fibrosis, their mode of action and novel formulations.
Collapse
Affiliation(s)
- Anju Sharma
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, Maharashtra, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, Mumbai, Maharashtra, India
| |
Collapse
|
3
|
Arghidash F, Javid-Naderi MJ, Gheybi F, Gholamhosseinian H, Kesharwani P, Sahebkar A. Exploring the multifaceted effects of silymarin on melanoma: Focusing on the role of lipid-based nanocarriers. J Drug Deliv Sci Technol 2024; 99:105950. [DOI: 10.1016/j.jddst.2024.105950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Li P, Wang D, Yang X, Liu C, Li X, Zhang X, Liu K, Zhang Y, Zhang M, Wang C, Wang R. Anti-Tumor Activity and Mechanism of Silibinin Based on Network Pharmacology and Experimental Verification. Molecules 2024; 29:1901. [PMID: 38675723 PMCID: PMC11054111 DOI: 10.3390/molecules29081901] [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: 03/24/2024] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Silibinin is a flavonoid compound extracted from the seeds of Silybum marianum (L.) Gaertn. It has the functions of liver protection, blood-lipid reduction and anti-tumor effects. However, the potential molecular mechanism of silibinin against tumors is still unknown. This study aimed to assess the anti-tumor effects of silibinin in adenoid cystic carcinoma (ACC2) cells and Balb/c nude mice, and explore its potential mechanism based on network pharmacology prediction and experimental verification. A total of 347 targets interacting with silibinin were collected, and 75 targets related to the tumor growth process for silibinin were filtrated. Based on the PPI analysis, CASP3, SRC, ESR1, JAK2, PRKACA, HSPA8 and CAT showed stronger interactions with other factors and may be the key targets of silibinin for treating tumors. The predicted target proteins according to network pharmacology were verified using Western blot analysis in ACC2 cells and Balb/c nude mice. In the pharmacological experiment, silibinin was revealed to significantly inhibit viability, proliferation, migration and induce the apoptosis of ACC2 cells in vitro, as well as inhibit the growth and development of tumor tissue in vivo. Western blot analysis showed that silibinin affected the expression of proteins associated with cell proliferation, migration and apoptosis, such as MMP3, JNK, PPARα and JAK. The possible molecular mechanism involved in cancer pathways, PI3K-Akt signaling pathway and viral carcinogenesis pathway via the inhibition of CASP3, MMP3, SRC, MAPK10 and CDK6 and the activation of PPARα and JAK. Overall, our results provided insight into the pharmacological mechanisms of silibinin in the treatment of tumors. These results offer a support for the anti-tumor uses of silibinin.
Collapse
Affiliation(s)
- Peihai Li
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Dexu Wang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Xueliang Yang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Changyu Liu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Xiaobin Li
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Xuanming Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Kechun Liu
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Yun Zhang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| | - Mengqi Zhang
- Key Laboratory of Novel Food Resources Processing, Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Processing Technology of Shandong Province, Institute of Agro-Food Science and Technology, Shandong Academy of Agricultural Sciences, Jinan 250100, China
| | - Changyun Wang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Rongchun Wang
- Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; (P.L.); (D.W.)
| |
Collapse
|
5
|
Surai PF, Surai A, Earle-Payne K. Silymarin and Inflammation: Food for Thoughts. Antioxidants (Basel) 2024; 13:98. [PMID: 38247522 PMCID: PMC10812610 DOI: 10.3390/antiox13010098] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/07/2024] [Accepted: 01/13/2024] [Indexed: 01/23/2024] Open
Abstract
Inflammation is a vital defense mechanism, creating hostile conditions for pathogens, preventing the spread of tissue infection and repairing damaged tissues in humans and animals. However, when inflammation resolution is delayed or compromised as a result of its misregulation, the process proceeds from the acute phase to chronic inflammation, leading to the development of various chronic illnesses. It is proven that redox balance disturbances and oxidative stress are among major factors inducing NF-κB and leading to over-inflammation. Therefore, the anti-inflammatory properties of various natural antioxidants have been widely tested in various in vitro and in vivo systems. Accumulating evidence indicates that silymarin (SM) and its main constituent silibinin/silybin (SB) have great potential as an anti-inflammation agent. The main anti-inflammatory mechanism of SM/SB action is attributed to the inhibition of TLR4/NF-κB-mediated signaling pathways and the downregulated expression of pro-inflammatory mediators, including TNF-α, IL-1β, IL-6, IL-12, IL-23, CCL4, CXCL10, etc. Of note, in the same model systems, SM/SB was able to upregulate anti-inflammatory cytokines (IL-4, IL-10, IL-13, TGF-β, etc.) and lipid mediators involved in the resolution of inflammation. The inflammatory properties of SM/SB were clearly demonstrated in model systems based on immune (macrophages and monocytes) and non-immune (epithelial, skin, bone, connective tissue and cancer) cells. At the same time, the anti-inflammatory action of SM/SB was confirmed in a number of in vivo models, including toxicity models, nonalcoholic fatty liver disease, ischemia/reperfusion models, stress-induced injuries, ageing and exercising models, wound healing and many other relevant model systems. It seems likely that the anti-inflammatory activities of SM/SB are key elements on the health-promoting properties of these phytochemicals.
Collapse
Affiliation(s)
- Peter F. Surai
- Vitagene and Health Research Centre, Bristol BS4 2RS, UK
- Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria
- Faculty of Agricultural and Environmental Sciences, Szent Istvan University, H-2103 Gödöllo, Hungary
- Biochemistry and Physiology Department, Saint-Petersburg State University of Veterinary Medicine, 196084 St. Petersburg, Russia
- Faculty of Veterinary Medicine, Sumy National Agrarian University, 40021 Sumy, Ukraine
- Faculty of Technology of Grain and Grain Business, Odessa National Technological University, 65039 Odessa, Ukraine
| | | | - Katie Earle-Payne
- NHS Greater Glasgow and Clyde, Renfrewshire Health and Social Care Centre, 10 Ferry Road, Renfrew PA4 8RU, UK
| |
Collapse
|
6
|
Gupta J, Jalil AT, Riyad Muedii ZAH, Aminov Z, Alsaikhan F, Ramírez-Coronel AA, Ramaiah P, Farhood B. The Radiosensitizing Potentials of Silymarin/Silibinin in Cancer: A Systematic Review. Curr Med Chem 2024; 31:6992-7014. [PMID: 37921180 DOI: 10.2174/0109298673248404231006052436] [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] [Received: 02/09/2023] [Revised: 07/10/2023] [Accepted: 09/11/2023] [Indexed: 11/04/2023]
Abstract
INTRODUCTION Although radiotherapy is one of the main cancer treatment modalities, exposing healthy organs/tissues to ionizing radiation during treatment and tumor resistance to ionizing radiation are the chief challenges of radiotherapy that can lead to different adverse effects. It was shown that the combined treatment of radiotherapy and natural bioactive compounds (such as silymarin/silibinin) can alleviate the ionizing radiation-induced adverse side effects and induce synergies between these therapeutic modalities. In the present review, the potential radiosensitization effects of silymarin/silibinin during cancer radiation exposure/radiotherapy were studied. METHODS According to the PRISMA guideline, a systematic search was performed for the identification of relevant studies in different electronic databases of Google Scholar, PubMed, Web of Science, and Scopus up to October 2022. We screened 843 articles in accordance with a predefined set of inclusion and exclusion criteria. Seven studies were finally included in this systematic review. RESULTS Compared to the control group, the cell survival/proliferation of cancer cells treated with ionizing radiation was considerably less, and silymarin/silibinin administration synergistically increased ionizing radiation-induced cytotoxicity. Furthermore, there was a decrease in the tumor volume, weight, and growth of ionizing radiation-treated mice as compared to the untreated groups, and these diminutions were predominant in those treated with radiotherapy plus silymarin/ silibinin. Furthermore, the irradiation led to a set of biochemical and histopathological changes in tumoral cells/tissues, and the ionizing radiation-induced alterations were synergized following silymarin/silibinin administration (in most cases). CONCLUSION In most cases, silymarin/silibinin administration could sensitize the cancer cells to ionizing radiation through an increase of free radical formation, induction of DNA damage, increase of apoptosis, inhibition of angiogenesis and metastasis, etc. However, suggesting the use of silymarin/silibinin during radiotherapeutic treatment of cancer patients requires further clinical studies.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, U.P., India
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | | | - Zafar Aminov
- Department of Public Health and Healthcare Management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, 103 Makhtumkuli Str., Tashkent, Uzbekistan
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Andrés Alexis Ramírez-Coronel
- Psychometry and Ethology Laboratory, Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Catholic University of Cuenca, Cuenca, Ecuador
- Epidemiology and Biostatistics Research Group, CES University, Medellin, Colombia
- Educational Statistics Research Group (GIEE), National University of Education, Cuenca, Ecuador
| | | | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
7
|
Pham ND, Nguyen THN, Vu NBD, Tran TNM, Pham BN, Le HS, Vo KH, Le XC, Tran LBH, Nguyen MH. Comparison of the radioprotective effects of the liposomal forms of five natural radioprotectants in alleviating the adverse effects of ionising irradiation on human lymphocytes and skin cells in radiotherapy. J Microencapsul 2023; 40:613-629. [PMID: 37815151 DOI: 10.1080/02652048.2023.2268705] [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] [Received: 02/12/2023] [Accepted: 10/02/2023] [Indexed: 10/11/2023]
Abstract
This study aims to evaluate the radioprotective effects of liposomes encapsulating curcumin (Lip-CUR), silibinin (Lip-SIL), α-tocopherol (Lip-TOC), quercetin (Lip-QUE) and resveratrol (Lip-RES) in alleviating the adverse effects of ionising irradiation on human lymphoctyes and skin cells in radiotherapy. Liposomes encapsulating the above natural radioprotectants (Lip-NRPs) were prepared by the film hydration method combined with sonication. Their radioprotective effects for the cells against X-irradiation was evaluated using trypan-blue assay and γ-H2AX assay. All prepared Lip-NRPs had a mean diameter less than 240 nm, polydispersity index less than 0.32, and zeta potential more than -23 mV. Among them, the radioprotective effect of Lip-RES was lowest, while that of Lip-QUE was highest. Lip-SIL also exhibited a high radioprotective effect despite its low DPPH-radical scavenging activity (12.9%). The radioprotective effects of Lip-NRPs do not solely depend on the free radical scavenging activity of NRPs but also on their ability to activate cellular mechanisms.
Collapse
Affiliation(s)
- Ngoc-Duy Pham
- Laboratory of Tissue Engineering and Biomedical Materials, University of Science, Ho Chi Minh City, Vietnam
- Vietnam National University, Ho Chi Minh City, Vietnam
- Center of Radiation Technology and Biotechnology, Nuclear Research Institute, Da Lat, Vietnam
| | | | - Ngoc-Bich-Dao Vu
- Center of Radiation Technology and Biotechnology, Nuclear Research Institute, Da Lat, Vietnam
| | - Thi-Ngoc-Mai Tran
- Center of Radiation Technology and Biotechnology, Nuclear Research Institute, Da Lat, Vietnam
| | - Bao-Ngoc Pham
- Center of Radiation Technology and Biotechnology, Nuclear Research Institute, Da Lat, Vietnam
| | - Hoang-Sinh Le
- VN-UK Institute for Research and Executive Education, The University of Danang, Da Nang, Vietnam
| | - Kim-Hai Vo
- Department of Health of Lam-Dong Province, Da Lat, Vietnam
| | - Xuan-Cuong Le
- Center of Radiation Technology and Biotechnology, Nuclear Research Institute, Da Lat, Vietnam
| | - Le-Bao-Ha Tran
- Laboratory of Tissue Engineering and Biomedical Materials, University of Science, Ho Chi Minh City, Vietnam
| | - Minh-Hiep Nguyen
- Center of Radiation Technology and Biotechnology, Nuclear Research Institute, Da Lat, Vietnam
| |
Collapse
|
8
|
Zhang Y, Huang Y, Li Z, Wu H, Zou B, Xu Y. Exploring Natural Products as Radioprotective Agents for Cancer Therapy: Mechanisms, Challenges, and Opportunities. Cancers (Basel) 2023; 15:3585. [PMID: 37509245 PMCID: PMC10377328 DOI: 10.3390/cancers15143585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/04/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023] Open
Abstract
Radiotherapy is an important cancer treatment. However, in addition to killing tumor cells, radiotherapy causes damage to the surrounding cells and is toxic to normal tissues. Therefore, an effective radioprotective agent that prevents the deleterious effects of ionizing radiation is required. Numerous synthetic substances have been shown to have clear radioprotective effects. However, most of these have not been translated for use in clinical applications due to their high toxicity and side effects. Many medicinal plants have been shown to exhibit various biological activities, including antioxidant, anti-inflammatory, and anticancer activities. In recent years, new agents obtained from natural products have been investigated by radioprotection researchers, due to their abundance of sources, high efficiency, and low toxicity. In this review, we summarize the mechanisms underlying the radioprotective effects of natural products, including ROS scavenging, promotion of DNA damage repair, anti-inflammatory effects, and the inhibition of cell death signaling pathways. In addition, we systematically review natural products with radioprotective properties, including polyphenols, polysaccharides, alkaloids, and saponins. Specifically, we discuss the polyphenols apigenin, genistein, epigallocatechin gallate, quercetin, resveratrol, and curcumin; the polysaccharides astragalus, schisandra, and Hohenbuehelia serotina; the saponins ginsenosides and acanthopanax senticosus; and the alkaloids matrine, ligustrazine, and β-carboline. However, further optimization through structural modification, improved extraction and purification methods, and clinical trials are needed before clinical translation. With a deeper understanding of the radioprotective mechanisms involved and the development of high-throughput screening methods, natural products could become promising novel radioprotective agents.
Collapse
Affiliation(s)
- Yi Zhang
- Division of Thoracic Oncology, Cancer Center, Department of Radiation Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ying Huang
- College of Management, Sichuan Agricultural University, Chengdu 611130, China
| | - Zheng Li
- Division of Thoracic Oncology, Cancer Center, Department of Radiation Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanyou Wu
- Zhongshan Ophthalmic Center, State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou 510060, China
| | - Bingwen Zou
- Division of Thoracic Oncology, Cancer Center, Department of Radiation Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong Xu
- Division of Thoracic Oncology, Cancer Center, Department of Radiation Oncology, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
Prades-Sagarra È, Yaromina A, Dubois LJ. Polyphenols as Potential Protectors against Radiation-Induced Adverse Effects in Patients with Thoracic Cancer. Cancers (Basel) 2023; 15:cancers15092412. [PMID: 37173877 PMCID: PMC10177176 DOI: 10.3390/cancers15092412] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Radiotherapy is one of the standard treatment approaches used against thoracic cancers, occasionally combined with chemotherapy, immunotherapy and molecular targeted therapy. However, these cancers are often not highly sensitive to standard of care treatments, making the use of high dose radiotherapy necessary, which is linked with high rates of radiation-induced adverse effects in healthy tissues of the thorax. These tissues remain therefore dose-limiting factors in radiation oncology despite recent technological advances in treatment planning and delivery of irradiation. Polyphenols are metabolites found in plants that have been suggested to improve the therapeutic window by sensitizing the tumor to radiotherapy, while simultaneously protecting normal cells from therapy-induced damage by preventing DNA damage, as well as having anti-oxidant, anti-inflammatory or immunomodulatory properties. This review focuses on the radioprotective effect of polyphenols and the molecular mechanisms underlying these effects in the normal tissue, especially in the lung, heart and esophagus.
Collapse
Affiliation(s)
- Èlia Prades-Sagarra
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ludwig J Dubois
- The M-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
10
|
Obrador E, Salvador-Palmer R, Villaescusa JI, Gallego E, Pellicer B, Estrela JM, Montoro A. Nuclear and Radiological Emergencies: Biological Effects, Countermeasures and Biodosimetry. Antioxidants (Basel) 2022; 11:1098. [PMID: 35739995 PMCID: PMC9219873 DOI: 10.3390/antiox11061098] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022] Open
Abstract
Atomic and radiological crises can be caused by accidents, military activities, terrorist assaults involving atomic installations, the explosion of nuclear devices, or the utilization of concealed radiation exposure devices. Direct damage is caused when radiation interacts directly with cellular components. Indirect effects are mainly caused by the generation of reactive oxygen species due to radiolysis of water molecules. Acute and persistent oxidative stress associates to radiation-induced biological damages. Biological impacts of atomic radiation exposure can be deterministic (in a period range a posteriori of the event and because of destructive tissue/organ harm) or stochastic (irregular, for example cell mutation related pathologies and heritable infections). Potential countermeasures according to a specific scenario require considering basic issues, e.g., the type of radiation, people directly affected and first responders, range of doses received and whether the exposure or contamination has affected the total body or is partial. This review focuses on available medical countermeasures (radioprotectors, radiomitigators, radionuclide scavengers), biodosimetry (biological and biophysical techniques that can be quantitatively correlated with the magnitude of the radiation dose received), and strategies to implement the response to an accidental radiation exposure. In the case of large-scale atomic or radiological events, the most ideal choice for triage, dose assessment and victim classification, is the utilization of global biodosimetry networks, in combination with the automation of strategies based on modular platforms.
Collapse
Affiliation(s)
- Elena Obrador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Rosario Salvador-Palmer
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Juan I. Villaescusa
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain; (J.I.V.); (A.M.)
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| | - Eduardo Gallego
- Energy Engineering Department, School of Industrial Engineering, Polytechnic University of Madrid, 28040 Madrid, Spain;
| | - Blanca Pellicer
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - José M. Estrela
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Alegría Montoro
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain; (J.I.V.); (A.M.)
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| |
Collapse
|
11
|
Fakhri S, Piri S, Moradi SZ, Khan H. Phytochemicals Targeting Oxidative Stress, Interconnected Neuroinflammatory, and Neuroapoptotic Pathways Following Radiation. Curr Neuropharmacol 2022; 20:836-856. [PMID: 34370636 PMCID: PMC9881105 DOI: 10.2174/1570159x19666210809103346] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/19/2021] [Accepted: 06/28/2021] [Indexed: 11/22/2022] Open
Abstract
The radiation for therapeutic purposes has shown positive effects in different contexts; however, it can increase the risk of many age-related and neurodegenerative diseases such as Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), and Parkinson's disease (PD). These different outcomes highlight a dose-response phenomenon called hormesis. Prevailing studies indicate that high doses of radiation could play several destructive roles in triggering oxidative stress, neuroapoptosis, and neuroinflammation in neurodegeneration. However, there is a lack of effective treatments in combating radiation-induced neurodegeneration, and the present drugs suffer from some drawbacks, including side effects and drug resistance. Among natural entities, polyphenols are suggested as multi-target agents affecting the dysregulated pathogenic mechanisms in neurodegenerative disease. This review discusses the destructive effects of radiation on the induction of neurodegenerative diseases by dysregulating oxidative stress, apoptosis, and inflammation. We also describe the promising effects of polyphenols and other candidate phytochemicals in preventing and treating radiation-induced neurodegenerative disorders, aiming to find novel/potential therapeutic compounds against such disorders.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;,Address correspondence to these author at the Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; E-mail: Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan; E-mail:
| | - Sana Piri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;,These authors have contributed equally to this work.
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;,Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran;,These authors have contributed equally to this work.
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan,Address correspondence to these author at the Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran; E-mail: Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan; E-mail:
| |
Collapse
|
12
|
Khazdair MR, Boskabady MH. Possible treatment with medicinal herbs and their ingredients of lung disorders induced by sulfur mustard exposures: a review. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:54191-54208. [PMID: 34382165 DOI: 10.1007/s11356-021-15697-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 07/24/2021] [Indexed: 06/13/2023]
Abstract
Chemical warfare (CW) agents are toxic synthetic chemicals that affect human's health, and sulfur mustard (SM) is a well-known chemical weapon that caused deaths of victims. The lung is the main target of SM exposure, and there are no definitive therapeutic modalities for lung injury induced by this agent. The possible therapeutic effects of medicinal plants and their active ingredients on lung injury induced by SM were reviewed in this article until the end of June 2021. Medicinal plants including Crocus sativus, Curcuma longa, Thymus vulgaris, Nigella sativa, and Zataria multiflora and also natural compounds showed therapeutic potential in improving of various features of lung injury induced by SM and other related chemical agents. Several studies showed therapeutic effects of some medicinal plants and natural products on lung inflammation, oxidative stress, and immune responses in experimental studies in SM-induced lung injury. In addition, clinical studies also showed the effect of medicinal plants and natural compounds on respiratory symptoms, pulmonary function tests (PFTs), and inflammatory markers. The therapeutic effects of medicinal plants and natural products on lung disorder induced by SM and related chemical agents were shown through amelioration of various features of lung injury.
Collapse
Affiliation(s)
- Mohammad Reza Khazdair
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Hossein Boskabady
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
13
|
Ali SA, Saifi MA, Godugu C, Talla V. Silibinin alleviates silica-induced pulmonary fibrosis: Potential role in modulating inflammation and epithelial-mesenchymal transition. Phytother Res 2021; 35:5290-5304. [PMID: 34250649 DOI: 10.1002/ptr.7210] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/25/2022]
Abstract
Pulmonary fibrosis (PF) is a devastating interstitial lung disease resulting from indefinite causes with very few limited, those too ineffective therapeutic options. Earlier evidence reported inflammation and epithelial-mesenchymal transition (EMT) are the major threats in PF. The present study was aimed to examine the anti-fibrotic activity of silibinin (SB) in PF. PF was induced by administering oropharyngeal 1.5 mg/mice silica on day 1, followed by treatment with and without oral SB for 14 days. Lung injury was assessed by x-ray analysis on day 14 and all the animals were sacrificed on day 15. The results showed that silica remarkably altered the histoarchitecture and induced the expression of inflammatory components in BALF and pulmonary tissue. Immunoblotting investigation quantified the expression of TGF-β, p-smad2/3, collagen-I, fibronectin, and α-SMA in the pulmonary tissue. To this end, treatment with SB alleviated inflammatory components, including IL-1β, IL-6, and TNF-α in the fibrotic tissue. Moreover, SB harnessed the tissue architecture, improved diffusive scattering of x-ray signals, and modulated epithelial-mesenchymal phenotypic alterations, including TGF-β, p-smad2/3, and collagen-I. Altogether, the significant reduction of inflammatory signaling, collagen deposition, and epithelial-mesenchymal transdifferentiation by SB suggested that it could be used as a potential therapeutic candidate to treat pulmonary inflammation and fibrosis.
Collapse
Affiliation(s)
- Syed Afroz Ali
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-(NIPER), Hyderabad, India.,Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research-(NIPER), Hyderabad, India
| | - Mohd Aslam Saifi
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research-(NIPER), Hyderabad, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research-(NIPER), Hyderabad, India
| | - Venu Talla
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-(NIPER), Hyderabad, India
| |
Collapse
|
14
|
Adnan M, Rasul A, Shah MA, Hussain G, Asrar M, Riaza A, Sarfraza I, Hussaina A, Khorsandid K, Laie NS, Hussaina SM. Radioprotective Role of Natural Polyphenols: From Sources to Mechanisms. Anticancer Agents Med Chem 2021; 22:30-39. [PMID: 33874875 DOI: 10.2174/1871520621666210419095829] [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: 08/16/2020] [Revised: 09/22/2020] [Accepted: 10/19/2020] [Indexed: 11/22/2022]
Abstract
The identification and development of radioprotective agents has emerged as a subject matter of research during recent years due to the growing usage of ionizing radiation in different areas of human life. Previous work on synthetic radioprotectors has achieved limited progress because of the numerous issues associated with toxicity. Compounds extracted from plants have potential to serve as lead candidates for developing ideal radioprotectors due to their low cost, safety and selectivity. Polyphenols are the most abundant and commonly dispersed group of biologically active molecules possessing broad range of pharmacological activities. Polyphenols have displayed efficacy for radioprotection during various investigations and can be administered at high doses with lesser toxicity. Detoxification of free radicals, modulating inflammatory responses, DNA repair, stimulation of hematopoietic recovery, and immune functions are the main mechanisms for radiation protection with polyphenols. Epicatechin, epigallocatechin-3-gallate, apigenin, caffeic acid phenylethylester, and silibinin provide cytoprotection together with the suppression of many pro-inflammatory cytokines owing to their free radical scavenging, anti-oxidant, and anti-inflammatory properties. Curcumin, resveratrol, quercetin, gallic acid, and rutin's radioprotective properties are regulated primarily by direct or indirect decline in cellular stress. Thus, polyphenols may serve as potential candidates for radioprotection in the near future, however, extensive investigations are still required to better understand their protection mechanisms.
Collapse
Affiliation(s)
- Muhammad Adnan
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000. Pakistan
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000. Pakistan
| | - Muhammad A Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University Faisalabad 38000. Pakistan
| | - Ghulam Hussain
- Neurochemical biology and Genetics Laboratory, Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad 38000. Pakistan
| | - Muhammad Asrar
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000. Pakistan
| | - Ammara Riaza
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000. Pakistan
| | - Iqra Sarfraza
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000. Pakistan
| | - Arif Hussaina
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000. Pakistan
| | - Khatereh Khorsandid
- Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran. Iran
| | - Ngit S Laie
- Institute for Research in Molecular Medicine Universiti Sains Malaysia, Pulau Pinang. Malaysia
| | - Syed M Hussaina
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000. Pakistan
| |
Collapse
|
15
|
Silibinin and SARS-CoV-2: Dual Targeting of Host Cytokine Storm and Virus Replication Machinery for Clinical Management of COVID-19 Patients. J Clin Med 2020; 9:jcm9061770. [PMID: 32517353 PMCID: PMC7356916 DOI: 10.3390/jcm9061770] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/25/2020] [Accepted: 06/05/2020] [Indexed: 01/08/2023] Open
Abstract
COVID-19, the illness caused by infection with the novel coronavirus SARS-CoV-2, is a rapidly spreading global pandemic in urgent need of effective treatments. Here we present a comprehensive examination of the host- and virus-targeted functions of the flavonolignan silibinin, a potential drug candidate against COVID-19/SARS-CoV-2. As a direct inhibitor of STAT3—a master checkpoint regulator of inflammatory cytokine signaling and immune response—silibinin might be expected to phenotypically integrate the mechanisms of action of IL-6-targeted monoclonal antibodies and pan-JAK1/2 inhibitors to limit the cytokine storm and T-cell lymphopenia in the clinical setting of severe COVID-19. As a computationally predicted, remdesivir-like inhibitor of RNA-dependent RNA polymerase (RdRp)—the central component of the replication/transcription machinery of SARS-CoV-2—silibinin is expected to reduce viral load and impede delayed interferon responses. The dual ability of silibinin to target both the host cytokine storm and the virus replication machinery provides a strong rationale for the clinical testing of silibinin against the COVID-19 global public health emergency. A randomized, open-label, phase II multicentric clinical trial (SIL-COVID19) will evaluate the therapeutic efficacy of silibinin in the prevention of acute respiratory distress syndrome in moderate-to-severe COVID-19-positive onco-hematological patients at the Catalan Institute of Oncology in Catalonia, Spain.
Collapse
|
16
|
Lim JO, Shin NR, Seo YS, Nam HH, Ko JW, Jung TY, Lee SJ, Kim HJ, Cho YK, Kim JC, Lee IC, Kim JS, Shin IS. Silibinin Attenuates Silica Dioxide Nanoparticles-Induced Inflammation by Suppressing TXNIP/MAPKs/AP-1 Signaling. Cells 2020; 9:cells9030678. [PMID: 32164364 PMCID: PMC7140632 DOI: 10.3390/cells9030678] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/07/2020] [Accepted: 03/08/2020] [Indexed: 12/22/2022] Open
Abstract
Silica dioxide nanoparticles (SiONPs) have been applied to several fields, such as drug delivery and gene therapy. However, SiONPs are a constituent of fine dust and can induce excessive inflammatory responses in the lungs via the airways. Silibinin, a major component of silymarin, has been known for its anti-oxidant and anti-inflammatory effects. In the present study, we explored the protective effects of silibinin against SiONPs-induced airway inflammation and explored its underlying mechanism of action, focusing on thioredoxin-interacting protein (TXNIP)/mitogen-activated protein kinases (MAPKs) in vitro and in vivo. In SiONPs-stimulated NCI-H292 airway epithelial cells, silibinin treatment effectively suppressed the elevation of the mRNA expression of tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and IL-1β, which was accompanied by the reduction in the expression of TXNIP, MAPKs, and activator protein-1 (AP-1). In SiONPs-treated mice, silibinin administration inhibited the increase in inflammatory cell counts and proinflammatory mediators, and it alleviated airway inflammation by SiONPs exposure. In addition, silibinin administration effectively suppressed the elevation of TXNIP/MAPKs/AP-1 signaling by SiONPs exposure. Taken together, silibinin effectively inhibited SiONPs-induced inflammatory responses, and this effect was closely related to the inhibition of TXNIP/MAPK/AP-1 signaling. These results suggested that silibinin might be useful for reducing pulmonary inflammation induced by SiONPs.
Collapse
Affiliation(s)
- Je-Oh Lim
- College of Veterinary Medicine (BK21 Plus Project Team), Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| | - Na-Rae Shin
- Research Institute of Radiation & Medical Science, Korea Institute of Radiation & Medical Sciences, Seoul 01812, Korea
| | - Yun-Soo Seo
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Geonjae-ro 177, Naju-si, Jeollanam-do 58245, Korea
| | - Hyeon-Hwa Nam
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Geonjae-ro 177, Naju-si, Jeollanam-do 58245, Korea
| | - Je-Won Ko
- College of Veterinary Medicine (BK21 Plus Project Team), Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| | - Tae-Yang Jung
- College of Veterinary Medicine (BK21 Plus Project Team), Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| | - Se-Jin Lee
- College of Veterinary Medicine (BK21 Plus Project Team), Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| | - Ha-Jung Kim
- College of Veterinary Medicine (BK21 Plus Project Team), Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| | - Young-Kwon Cho
- College of Health Sciences, Cheongju University, 298 Daesung-ro, Sangdang-gu, Cheongju-si, Chungbuk 28503, Korea
| | - Jong-Choon Kim
- College of Veterinary Medicine (BK21 Plus Project Team), Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| | - In-Chul Lee
- Functional Biomaterial Research Center, Jeonbuk Branch, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si, Jeonbuk 56212, Korea
| | - Joong-Sun Kim
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Geonjae-ro 177, Naju-si, Jeollanam-do 58245, Korea
| | - In-Sik Shin
- College of Veterinary Medicine (BK21 Plus Project Team), Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Korea
| |
Collapse
|
17
|
Liu K, Zhou S, Liu J, Wang Y, Zhu F, Liu M. Silibinin attenuates high-fat diet-induced renal fibrosis of diabetic nephropathy. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3117-3126. [PMID: 31695328 PMCID: PMC6718242 DOI: 10.2147/dddt.s209981] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022]
Abstract
Aim Diabetic nephropathy (DN) is the leading cause of end-stage renal disease. Silibinin is a flavonoid compound which has medicinal value. Previous studies revealed that silibinin exhibited an anti-fibrotic effect. However, whether silibinin could attenuate high-fat diet (HFD)-induced renal fibrosis remains unclear. Therefore, this study aimed to explore the molecular mechanism by which silibinin regulated renal fibrosis induced by HFD. Methods In the present study, human renal glomerular endothelial cells (HRGECs) were treated with various concentrations of silibinin. Then, cell viability and apoptosis were measured by MTT assay and flow cytometry, respectively. In addition, HRGECs were exposed to 100 nM TGF-β1 for mimicking in vitro renal fibrosis. The expressions of collagen I, fibronectin, and α-SMA were detected by reverse transcription-quantitative polymerase
chain reaction and Western blot. Protein levels of p-IκB and p-p65 were examined by Western blot; meanwhile, level of NF-κB was measured by immunofluorescence staining. Furthermore, HFD-induced mouse model of renal fibrosis was established. The mouse body weight, fasting glucose, kidney weight/body weight, microalbuminuria, kidney histopathology, and fibrotic area were measured to assess the severity of renal fibrosis. Results Low concentration of silibinin (≤50 μM) had no cytotoxicity, while high concentration of silibinin (≥75 μM) exhibited significant cytotoxicity. Additionally, TGF-β1 increased the expressions of collagen I, fibronectin, α-SMA, p-IκB, and p-p65 and decreased the level NF-κB, while these effects were notably reversed by 50 μM silibinin. Moreover, both 50 and 100 mg/kg silibinin greatly decreased HFD-induced the upregulation of kidney weight/body weight, microalbuminuria, and fibrotic area. 100 mg/kg silibinin markedly reduced collagen I, fibronectin, and p-p65 expressions in mice renal tissues. Conclusion Silibinin was able to attenuate renal fibrosis in vitro and in vivo via inhibition of NF-κB. These data suggested that silibinin may serve as a potential agent to alleviate the renal fibrosis of DN.
Collapse
Affiliation(s)
- Kun Liu
- Department of Nephrology, Jining No. 1 People's Hospital, Jining 272000, Shandong, People's Republic of China
| | - Shiju Zhou
- Department of Nephrology, Jining No. 1 People's Hospital, Jining 272000, Shandong, People's Republic of China
| | - Jinyan Liu
- Department of Nephrology, Jining No. 1 People's Hospital, Jining 272000, Shandong, People's Republic of China.,Department of Nephrology, Jining Medical University, Jining 272000, Shandong, People's Republic of China
| | - Yingying Wang
- Department of Nephrology, Jining No. 1 People's Hospital, Jining 272000, Shandong, People's Republic of China
| | - Fengxian Zhu
- Department of Nephrology, Jining No. 1 People's Hospital, Jining 272000, Shandong, People's Republic of China
| | - Man Liu
- Department of Nephrology, Jining No. 1 People's Hospital, Jining 272000, Shandong, People's Republic of China
| |
Collapse
|
18
|
Reactive Oxygen Species Drive Epigenetic Changes in Radiation-Induced Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4278658. [PMID: 30881591 PMCID: PMC6381575 DOI: 10.1155/2019/4278658] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 12/06/2018] [Accepted: 12/12/2018] [Indexed: 12/14/2022]
Abstract
Radiation-induced fibrosis (RIF) develops months to years after initial radiation exposure. RIF occurs when normal fibroblasts differentiate into myofibroblasts and lay down aberrant amounts of extracellular matrix proteins. One of the main drivers for developing RIF is reactive oxygen species (ROS) generated immediately after radiation exposure. Generation of ROS is known to induce epigenetic changes and cause differentiation of fibroblasts to myofibroblasts. Several antioxidant compounds have been shown to prevent radiation-induced epigenetic changes and the development of RIF. Therefore, reviewing the ROS-linked epigenetic changes in irradiated fibroblast cells is essential to understand the development and prevention of RIF.
Collapse
|
19
|
Fischer N, Seo EJ, Efferth T. Prevention from radiation damage by natural products. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 47:192-200. [PMID: 30166104 DOI: 10.1016/j.phymed.2017.11.005] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 10/20/2017] [Accepted: 11/12/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND Radiotherapy is a mainstay of cancer treatment since decades. Ionizing radiation (IR) is used for destruction of cancer cells and shrinkage of tumors. However, the increase of radioresistance in cancer cells and radiation toxicity to normal tissues are severe concerns. The exposure to radiation generates intracellular reactive oxygen species (ROS), which leads to DNA damage by lipid peroxidation, removal of thiol groups from cellular and membrane proteins, strand breaks and base alterations. HYPOTHESIS Plants have to deal with radiation-induced damage (UV-light of sun, other natural radiation sources). Therefore, it is worth speculating that radioprotective mechanisms have evolved during evolution of life. We hypothesize that natural products from plants may also protect from radiation damage caused as adverse side effects of cancer radiotherapy. METHODS The basis of this systematic review, we searched the relevant literature in the PubMed database. RESULTS Flavonoids, such as genistein, epigallocatechin-3-gallate, epicatechin, apigenin and silibinin mainly act as antioxidant, free radical scavenging and anti-inflammatory compounds, thus, providing cytoprotection in addition to downregulation of several pro-inflammatory cytokines. Comparable effects have been found in phenylpropanoids, especially caffeic acid phenylethylester, curcumin, thymol and zingerone. Besides, resveratrol and quercetin are the most important cytoprotective polyphenols. Their radioprotective effects are mediated by a wide range of mechanisms mainly leading to direct or indirect reduction of cellular stress. Ascorbic acid is broadly used as antioxidant, but it has also shown activity in reducing cellular damage after irradiation mainly due to its antioxidant capabilities. The metal ion chelator, gallic acid, represents another natural product attenuating cellular damage caused by radiation. CONCLUSIONS Some secondary metabolites from plants reveal radioprotective features against cellular damage caused by irradiation. These results warrant further analysis to develop phytochemicals as radioprotectors for clinical use.
Collapse
Affiliation(s)
- Nicolas Fischer
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Ean-Jeong Seo
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
20
|
Zhou L, Zhang X, Li H, Niu C, Yu D, Yang G, Liang X, Wen X, Li M, Cui J. Validating the pivotal role of the immune system in low-dose radiation-induced tumor inhibition in Lewis lung cancer-bearing mice. Cancer Med 2018; 7:1338-1348. [PMID: 29479834 PMCID: PMC5911597 DOI: 10.1002/cam4.1344] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 10/18/2017] [Accepted: 12/26/2017] [Indexed: 12/17/2022] Open
Abstract
Although low-dose radiation (LDR) possesses the two distinct functions of inducing hormesis and adaptive responses, which result in immune enhancement and tumor inhibition, its clinical applications have not yet been elucidated. The major obstacle that hinders the application of LDR in the clinical setting is that the mechanisms underlying induction of tumor inhibition are unclear, and the risks associated with LDR are still unknown. Thus, to overcome this obstacle and elucidate the mechanisms mediating the antitumor effects of LDR, in this study, we established an in vivo lung cancer model to investigate the participation of the immune system in LDR-induced tumor inhibition and validated the pivotal role of the immune system by impairing immunity with high-dose radiation (HDR) of 1 Gy. Additionally, the LDR-induced adaptive response of the immune system was also observed by sequential HDR treatment in this mouse model. We found that LDR-activated T cells and natural killer cells and increased the cytotoxicity of splenocytes and the infiltration of T cells in the tumor tissues. In contrast, when immune function was impaired by HDR pretreatment, LDR could not induce tumor inhibition. However, when LDR was administered before HDR, the immunity could be protected from impairment, and tumor growth could be inhibited to some extent, indicating the induction of the immune adaptive response by LDR. Therefore, we demonstrated that immune enhancement played a key role in LDR-induced tumor inhibition. These findings emphasized the importance of the immune response in tumor radiotherapy and may help promote the application of LDR as a novel approach in clinical practice.
Collapse
MESH Headings
- Animals
- Biomarkers
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/metabolism
- Carcinoma, Lewis Lung/pathology
- Carcinoma, Lewis Lung/radiotherapy
- Cytokines/metabolism
- Disease Models, Animal
- Female
- Immune System/radiation effects
- Immunohistochemistry
- Immunomodulation/radiation effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Mice
- Radiation Dosage
- Radiation, Ionizing
- Spleen/cytology
- Spleen/immunology
- Spleen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Burden/radiation effects
Collapse
Affiliation(s)
- Lei Zhou
- Cancer CenterThe First Hospital of Jilin UniversityChangchun130021China
| | - Xiaoying Zhang
- Cancer CenterThe First Hospital of Jilin UniversityChangchun130021China
| | - Hui Li
- Cancer CenterThe First Hospital of Jilin UniversityChangchun130021China
| | - Chao Niu
- Cancer CenterThe First Hospital of Jilin UniversityChangchun130021China
| | - Dehai Yu
- Cancer CenterThe First Hospital of Jilin UniversityChangchun130021China
| | - Guozi Yang
- Department of Radiation‐OncologyThe First Hospital of Jilin UniversityChangchun130021China
| | - Xinyue Liang
- Cancer CenterThe First Hospital of Jilin UniversityChangchun130021China
| | - Xue Wen
- Cancer CenterThe First Hospital of Jilin UniversityChangchun130021China
| | - Min Li
- Cancer CenterThe First Hospital of Jilin UniversityChangchun130021China
| | - Jiuwei Cui
- Cancer CenterThe First Hospital of Jilin UniversityChangchun130021China
| |
Collapse
|
21
|
Najafi M, Motevaseli E, Shirazi A, Geraily G, Rezaeyan A, Norouzi F, Rezapoor S, Abdollahi H. Mechanisms of inflammatory responses to radiation and normal tissues toxicity: clinical implications. Int J Radiat Biol 2018; 94:335-356. [PMID: 29504497 DOI: 10.1080/09553002.2018.1440092] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 01/23/2018] [Accepted: 01/31/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE Cancer treatment is one of the most challenging diseases in the present era. Among a few modalities for cancer therapy, radiotherapy plays a pivotal role in more than half of all treatments alone or combined with other cancer treatment modalities. Management of normal tissue toxicity induced by radiation is one of the most important limiting factors for an appropriate radiation treatment course. The evaluation of mechanisms of normal tissue toxicity has shown that immune responses especially inflammatory responses play a key role in both early and late side effects of exposure to ionizing radiation (IR). DNA damage and cell death, as well as damage to some organelles such as mitochondria initiate several signaling pathways that result in the response of immune cells. Massive cell damage which is a common phenomenon following exposure to a high dose of IR cause secretion of a lot of inflammatory mediators including cytokines and chemokines. These mediators initiate different changes in normal tissues that may continue for a long time after irradiation. In this study, we reviewed the mechanisms of inflammatory responses to IR that are involved in normal tissue toxicity and considered as the most important limiting factors in radiotherapy. Also, we introduced some agents that have been proposed for management of these responses. CONCLUSIONS The early inflammation during the radiation treatment is often a limiting factor in radiotherapy. In addition to the limiting factors, chronic inflammatory responses may increase the risk of second primary cancers through continuous free radical production, attenuation of tumor suppressor genes, and activation of oncogenes. Moreover, these effects may influence non-irradiated tissues through a mechanism named bystander effect.
Collapse
Affiliation(s)
- Masoud Najafi
- a Radiology and Nuclear Medicine Department, School of Paramedical Sciences , Kermanshah University of Medical Science , Kermanshah , Iran
| | - Elahe Motevaseli
- b Department of Molecular Medicine, School of Advanced Technologies in Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Alireza Shirazi
- c Department of Medical Physics and Biomedical Engineering, Faculty of Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Ghazale Geraily
- c Department of Medical Physics and Biomedical Engineering, Faculty of Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Abolhasan Rezaeyan
- d Department of Medical Physics, School of Medicine , Iran University of Medical Sciences , Tehran , Iran
| | - Farzad Norouzi
- e Science and Research Branch , Azad University , Tehran , Iran
| | - Saeed Rezapoor
- f Department of Radiology, Faculty of Paramedical Sciences , Tehran University of Medical Sciences , Tehran , Iran
| | - Hamid Abdollahi
- d Department of Medical Physics, School of Medicine , Iran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
22
|
Yahyapour R, Amini P, Rezapour S, Cheki M, Rezaeyan A, Farhood B, Shabeeb D, Musa AE, Fallah H, Najafi M. Radiation-induced inflammation and autoimmune diseases. Mil Med Res 2018; 5:9. [PMID: 29554942 PMCID: PMC5859747 DOI: 10.1186/s40779-018-0156-7] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 03/02/2018] [Indexed: 12/22/2022] Open
Abstract
Currently, ionizing radiation (IR) plays a key role in the agricultural and medical industry, while accidental exposure resulting from leakage of radioactive sources or radiological terrorism is a serious concern. Exposure to IR has various detrimental effects on normal tissues. Although an increased risk of carcinogenesis is the best-known long-term consequence of IR, evidence has shown that other diseases, particularly diseases related to inflammation, are common disorders among irradiated people. Autoimmune disorders are among the various types of immune diseases that have been investigated among exposed people. Thyroid diseases and diabetes are two autoimmune diseases potentially induced by IR. However, the precise mechanisms of IR-induced thyroid diseases and diabetes remain to be elucidated, and several studies have shown that chronic increased levels of inflammatory cytokines after exposure play a pivotal role. Thus, cytokines, including interleukin-1(IL-1), tumor necrosis factor (TNF-α) and interferon gamma (IFN-γ), play a key role in chronic oxidative damage following exposure to IR. Additionally, these cytokines change the secretion of insulin and thyroid-stimulating hormone(TSH). It is likely that the management of inflammation and oxidative damage is one of the best strategies for the amelioration of these diseases after a radiological or nuclear disaster. In the present study, we reviewed the evidence of radiation-induced diabetes and thyroid diseases, as well as the potential roles of inflammatory responses. In addition, we proposed that the mitigation of inflammatory and oxidative damage markers after exposure to IR may reduce the incidence of these diseases among individuals exposed to radiation.
Collapse
Affiliation(s)
- Rasoul Yahyapour
- School of Medicine, Jiroft University of Medical Sciences, Jiroft, Zip code: 8813833435, Iran
| | - Peyman Amini
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Zip code: 1417613151, Iran
| | - Saeed Rezapour
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Zip code: 1417613151, Iran
| | - Mohsen Cheki
- Department of Radiologic Technology, Faculty of Paramedicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Zip code: 6135715794, Iran
| | - Abolhasan Rezaeyan
- Department of Medical Physics, School of Medicine, Iran University of Medical Sciences, Tehran, Zip code: 1449614535, Iran
| | - Bagher Farhood
- Departments of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Zip code: 3715835155, Iran
| | - Dheyauldeen Shabeeb
- Department of Medical Physics and Biomedical Engineering, Faculty of Medicine, Tehran University of Medical Sciences (International Campus), Tehran, Zip code: 1417613151, Iran.,Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
| | - Ahmed Eleojo Musa
- Research center for molecular and cellular imaging, Tehran University of Medical Sciences, Tehran, Zip code: 1417613151, Iran
| | - Hengameh Fallah
- Department of Chemistry, Faculty of Science, Islamic Azad University, Arak, Zip code: 3836119131, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Science, Kermanshah, Zip code: 6714869914, Iran.
| |
Collapse
|
23
|
Yang J, Chen J, He J, Li J, Shi J, Cho WC, Liu X. Wnt signaling as potential therapeutic target in lung cancer. Expert Opin Ther Targets 2016; 20:999-1015. [PMID: 26882052 DOI: 10.1517/14728222.2016.1154945] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 02/12/2016] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Wingless-type (Wnt) signaling is tightly regulated at multiple cellular levels and is dysregulated in lung cancer. Therefore, it offers therapeutic targets for developing novel agents for lung cancer treatment. AREAS COVERED In this article, we discuss the role of the Wnt signaling pathway in lung cancer, highlighting the aberrant activation of Wnt in lung cancer stem cells and its implication in resistance to radiotherapy, chemotherapy and targeted therapy. We also expound the regulatory roles of microRNAs in Wnt signaling, as well as the potential of the Wnt pathway to provide biomarkers and therapeutic targets in lung cancer. The potential use of small molecule and biological inhibitors targeting the Wnt pathway for lung cancer therapy and prevention is also discussed. EXPERT OPINION Wnt signaling plays an important role in the development and metastasis of lung cancer; the pathway provides targets to develop agents towards for cancer prevention and therapy. A number of clinical trials have shown the effectiveness of Wnt pathway inhibitors in epithelial tumors. However, the side effects should be considered. Nevertheless, the results from clinical studies suggest that inhibitors targeting the Wnt signaling show promise against lung cancer.
Collapse
Affiliation(s)
- Jiali Yang
- a Ningxia Key laboratory of Clinical and Pathogenic Microbiology , Center of Laboratory Medicine of General Hospital at Ningxia Medical University , Yinchuan , Ningxia 750004 , China
| | - Juan Chen
- b Department of Pulmonary and Critical Care Medicine , General Hospital, Ningxia Medical University , Yinchuan , Ningxia , China
| | - Jinxi He
- c Department of Thoracic Surgery , General Hospital, Ningxia Medical University , Yinchuan , Ningxia , China
| | - Jing Li
- c Department of Thoracic Surgery , General Hospital, Ningxia Medical University , Yinchuan , Ningxia , China
| | - Juan Shi
- a Ningxia Key laboratory of Clinical and Pathogenic Microbiology , Center of Laboratory Medicine of General Hospital at Ningxia Medical University , Yinchuan , Ningxia 750004 , China
| | - William C Cho
- d Department of Clinical Oncology , Queen Elizabeth Hospital , Kowloon , Hong Kong
| | - Xiaoming Liu
- a Ningxia Key laboratory of Clinical and Pathogenic Microbiology , Center of Laboratory Medicine of General Hospital at Ningxia Medical University , Yinchuan , Ningxia 750004 , China
- e Human Stem Cell Institute, General Hospital, Ningxia Medical University , Yinchuan , Ningxia , China
| |
Collapse
|
24
|
Emerging targets for radioprotection and radiosensitization in radiotherapy. Tumour Biol 2016; 37:11589-11609. [DOI: 10.1007/s13277-016-5117-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 06/09/2016] [Indexed: 01/12/2023] Open
|