1
|
Hou B, Wang X, He Z, Liu H. Integrative approach using network pharmacology, bioinformatics, and experimental methods to explore the mechanism of cantharidin in treating colorectal cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6745-6761. [PMID: 38507104 DOI: 10.1007/s00210-024-03041-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 03/06/2024] [Indexed: 03/22/2024]
Abstract
Cantharidin, a terpenoid produced by blister beetles, has been used in traditional Chinese medicine to treat various ailments and cancers. However, its biological activity, impact, and anticancer mechanisms remain unclear. The Cantharidin chemical gene connections were identified using various databases. The GSE21815 dataset was used to collect the gene expression information. Differential gene analysis and gene ontology analyses were performed. Gene set enrichment analysis was used to assess the activation of disease pathways. Weighted gene co-expression network analysis and differential analysis were used to identify illness-associated genes, examine differential genes, and discover therapeutic targets via protein-protein interactions. MCODE analysis of major subgroup networks was used to identify critical genes influenced by Cantharidin, examine variations in the expression of key clustered genes in colorectal cancer vs. control samples, and describe the subject operators. Single-cell GSE188711 dataset was preprocessed to investigate Cantharidin's therapeutic targets and signaling pathways in colorectal cancer. Single-cell RNA sequencing was utilized to identify 22 cell clusters and marker genes for two different cell types in each cluster. The effects of different Cantharidin concentrations on colorectal cancer cells were studied in vitro. One hundred and ninety-seven Cantharidin-associated target genes and 480 critical genes implicated in the development of the illness were identified. Cantharidin significantly inhibited the proliferation and migration of HCT116 cells and promoted apoptosis at certain concentrations. Patients on current therapy develop inherent and acquired resistance. Our study suggests that Cantharidin may play an anti-CRC role by modulating immune function.
Collapse
Affiliation(s)
- Benchao Hou
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiaomin Wang
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, No. 1688, Meiling Avenue, Wanli District, Nanchang, 330004, Jiangxi, China
| | - Zhijian He
- Department of Radiation Oncology, Jiangxi Cancer Hospital, 519 Beijing East Road, Qingshanhu District, Nanchang, 330029, Jiangxi, China.
| | - Haiyun Liu
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, No. 1688, Meiling Avenue, Wanli District, Nanchang, 330004, Jiangxi, China.
| |
Collapse
|
2
|
Cui H, Wang Y, Zhou T, Qu L, Zhang X, Wang Y, Han M, Yang S, Ren X, Wang G, Gang X. Targeting DGAT1 inhibits prostate cancer cells growth by inducing autophagy flux blockage via oxidative stress. Oncogene 2024; 43:136-150. [PMID: 37973951 DOI: 10.1038/s41388-023-02878-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 10/04/2023] [Accepted: 10/23/2023] [Indexed: 11/19/2023]
Abstract
Impaired macroautophagy/autophagy flux has been implicated in the treatment of prostate cancer (PCa). However, the mechanism underlying autophagy dysregulation in PCa remains unknown. In the current study, we investigated the role of diacylglycerol acyltransferases 1 (DGAT1) and its potential effects on cellular energy homeostasis and autophagy flux in PCa. The results of immunohistochemical staining suggested that DGAT1 expression was positively corrected with tumor stage and node metastasis, indicating DGAT1 is an important factor involved in the development and progression of PCa. Furthermore, targeting DGAT1 remarkably inhibited cell proliferation in vitro and suppressed PCa growth in xenograft models by triggering severe oxidative stress and subsequently autophagy flux blockage. Mechanically, DGAT1 promoted PCa progression by maintaining cellular energy homeostasis, preserving mitochondrial function, protecting against reactive oxygen species, and subsequently promoting autophagy flux via regulating lipid droplet formation. Moreover, we found that fenofibrate exhibits as an upstream regulator of DGAT1. Fenofibrate performed its anti-PCa effect involved the aforementioned mechanisms, and partially dependent on the regulation of DGAT1. Collectively. These findings indicate that DGAT1 regulates PCa lipid droplets formation and is essential for PCa progression. Targeting DGAT1 might be a promising method to control the development and progression of PCa. Schematic representation of DGAT1 affects autophagy flux by regulating lipid homeostasis and maintaining mitochondrial function in prostate cancer (PCa). PCa is characterized up-regulation of DGAT1, leading to the translocation of free fatty acids into lipid droplets, thereby preventing PCa cell from lipotoxicity. Inhibition of DGAT1 suppresses growth of PCa by inducing oxidative stress and subsequently autophagy flux blockage. Further, the current results revealed that fenofibrate exhibits as an upstream regulator of DGAT1, and fenofibrate plays an anti-PCa role partially dependent on the regulation of DGAT1, suggesting a potential therapeutic approach to ameliorate this refractory tumor.
Collapse
Affiliation(s)
- Haiying Cui
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Yao Wang
- Department of Orthopedics, The Second Hospital Jilin University, Changchun, 130021, Jilin Province, China
| | - Tong Zhou
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Limei Qu
- Department of Pathology, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Institute of Immunology, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Yingdi Wang
- Department of Urology, Jilin Oncological Hospital, Changchun, 130021, Jilin Province, China
| | - Mingyue Han
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Shuo Yang
- Department of Clinical Nutrition, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Xinhua Ren
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China.
| | - Xiaokun Gang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China.
| |
Collapse
|
3
|
He T, Duan C, Feng W, Ao J, Lu D, Li X, Zhang J. Bibliometric Analysis and Systemic Review of Cantharidin Research Worldwide. Curr Pharm Biotechnol 2024; 25:1585-1601. [PMID: 39034837 DOI: 10.2174/0113892010244101231024111850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 08/25/2023] [Accepted: 09/19/2023] [Indexed: 07/23/2024]
Abstract
BACKGROUND Cantharidin (CTD), a natural toxic compound from blister beetle Mylabris, has been used for cancer treatment for millenary. CTD and its analogs have become mainstream adjuvant drugs with radiotherapy and chemotherapy in clinical applications. However, the detailed pharmacology mechanism of CTD was not fully elucidated. METHODS Publications of CTD were collected from the Web of Science Core Collection database from 1991 to 2023 using CiteSpace, VOSviewer, and Scimago Graphica software. RESULTS A total of 1,611 publications of CTD were mainly published in China and the United States. The University of Newcastle has published the most researches. Mcclusey, Adam, Sakoff, Jennette, and Zhang, Yalin had the most CTD publications with higher H. Notably, CTD researches were mainly published in Bioorganic & Medicinal Chemistry Letters and the Journal of Biological Chemistry. Cluster profile results revealed that protein phosphatase 2A (PP2A), human gallbladder carcinoma, Aidi injection, and cell apoptosis were the hotspots. Concentration on the pharmacology function of PP2A subunit regulation, hepatotoxicity, nephrotoxicity, and cardiotoxicity mechanism should be strengthened in the future. CONCLUSION Bibliometric analysis combined with a systemic review of CTD research first revealed that PP2A and CTD analogs were the knowledge base of CTD, and PP2A subunit regulation and toxic mechanism could be the frontiers of CTD.
Collapse
Affiliation(s)
- Tianmu He
- School of Basic Medicine, Zunyi medical University, Zunyi 550025, China
- School of Basic Medicine, Guizhou Medical University, Guiyang 563000, Guizhou, China
| | - Cancan Duan
- School of Pharmacy and Key Laboratory of Basic Pharmacology Ministry Education and Joint International Research Laboratory of Ethnomedicine, Ministry of Education, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Wenzhong Feng
- School of Pharmacy and Key Laboratory of Basic Pharmacology Ministry Education and Joint International Research Laboratory of Ethnomedicine, Ministry of Education, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Jingwen Ao
- School of Pharmacy and Key Laboratory of Basic Pharmacology Ministry Education and Joint International Research Laboratory of Ethnomedicine, Ministry of Education, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Dingyang Lu
- School of Pharmacy and Key Laboratory of Basic Pharmacology Ministry Education and Joint International Research Laboratory of Ethnomedicine, Ministry of Education, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Xiaofei Li
- School of Basic Medicine, Zunyi medical University, Zunyi 550025, China
- School of Basic Medicine, Guizhou Medical University, Guiyang 563000, Guizhou, China
| | - Jianyong Zhang
- School of Pharmacy and Key Laboratory of Basic Pharmacology Ministry Education and Joint International Research Laboratory of Ethnomedicine, Ministry of Education, Zunyi Medical University, Zunyi 563000, Guizhou, China
| |
Collapse
|
4
|
Yan J, Gao YM, Deng XL, Wang HS, Shi GT. Integrative analysis of the molecular signature of target genes involved in the antitumor effects of cantharidin on hepatocellular carcinoma. BMC Cancer 2023; 23:1161. [PMID: 38017425 PMCID: PMC10685469 DOI: 10.1186/s12885-023-11594-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 10/31/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Cantharidin (CTD) is the active ingredient of Chinese medicine, which has been traditionally used in multiple cancers treatment, especially in hepatocellular carcinoma (HCC). However, a comprehensive analysis of the CTD-related molecular mechanism is still necessary to understand its functions in HCC treatment. This study aimed to reveal the novel molecular targets and regulatory networks of CTD in HCC. METHODS A model of H22 tumour-bearing mice was constructed, and the function of CTD in tumour growth was evaluated. An integrated approach of CTD associated transcriptional profiling and biological systems analysis was used to identify key regulators involved in antitumour pathways. The identified differential expression patterns were supported by the results of Gene Ontology (GO) term and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyse, and by protein-protein interaction (PPI) network construction. The relationships between gene expression and tumour immunity were evaluated using Tumour Immune Estimation Resource (TIMER). Prognostic value was analyzed with Kaplan-Meier plotter. RESULTS In the present study, the therapeutic effect of CTD on HCC was evaluated in vivo. We obtained the CTD-related transcriptional profiles, systematically and intuitively illustrated its possible pharmacological mechanisms in HCC through multiple targets and signalling pathways. These results revealed that the CTD-related differentially expressed genes were involved in autophagy, transcription factors (TFs) related transcriptional regulation, fatty acid metabolism and immune response in HCC. We found that MAPT, TOP2A, CENPF and MEFV were hub genes of CTD targets involved in autophagy regulation. Totally, 14 TFs have been confirmed to be critical for transcriptional regulation, and 33 TF targets were identified as the hub genes in transcriptional mis-regulation pathway in cancer. These TFs were associated with the immune response and immune cell infiltration. In addition, the downregulated genes were significantly enriched in metabolic regulation pathways, especially fatty acid metabolism after CTD treatment. Furthermore, the network of CTD associated miRNAs with these fatty acid metabolism-related targets was constructed in HCC. CONCLUSIONS Taken together, our results comprehensively elucidated that CTD could act on multiple targets in HCC therapy, affecting autophagy, transcriptional regulation, the immune response and fatty acid metabolism. Our results provide a foundation for the study of the molecular mechanistic of CTD and its clinical application in the treatment of HCC.
Collapse
Affiliation(s)
- Jia Yan
- School of Basic medical, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yu Min Gao
- School of Public health, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xiu Ling Deng
- School of Basic medical, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.
| | - Hai Sheng Wang
- School of Basic medical, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.
| | - Gui Tao Shi
- Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.
| |
Collapse
|
5
|
Inhibition of MZF1/c-MYC Axis by Cantharidin Impairs Cell Proliferation in Glioblastoma. Int J Mol Sci 2022; 23:ijms232314727. [PMID: 36499054 PMCID: PMC9740304 DOI: 10.3390/ijms232314727] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
Myeloid zinc finger 1 (MZF1), also known as zinc finger protein 42, is a zinc finger transcription factor, belonging to the Krüppel-like family that has been implicated in several types of malignancies, including glioblastoma multiforme (GBM). MZF1 is reportedly an oncogenic gene that promotes tumor progression. Moreover, higher expression of MZF1 has been associated with a worse overall survival rate among patients with GBM. Thus, MZF1 may be a promising target for therapeutic interventions. Cantharidin (CTD) has been traditionally used in Chinese medicine to induce apoptosis and inhibit cancer cell proliferation; however, the mechanism by which CTD inhibits cell proliferation remains unclear. In this study, we found that the expression of MZF1 was higher in GBM tissues than in adjacent normal tissues and low-grade gliomas. Additionally, the patient-derived GBM cells and GBM cell lines presented higher levels of MZF1 than normal human astrocytes. We demonstrated that CTD had greater anti-proliferative effects on GBM than a derivative of CTD, norcantharidin (NCTD). MZF1 expression was strongly suppressed by CTD treatment. Furthermore, MZF1 enhanced the proliferation of GBM cells and upregulated the expression of c-MYC, whereas these effects were reversed by CTD treatment. The results of our study suggest that CTD may be a promising therapeutic agent for patients with GBM and suggest a promising direction for further investigation.
Collapse
|
6
|
Strategies for Solubility and Bioavailability Enhancement and Toxicity Reduction of Norcantharidin. Molecules 2022; 27:molecules27227740. [PMID: 36431851 PMCID: PMC9693198 DOI: 10.3390/molecules27227740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/01/2022] [Accepted: 11/06/2022] [Indexed: 11/12/2022] Open
Abstract
Cantharidin (CTD) is the main active ingredient isolated from Mylabris, and norcantharidin (NCTD) is a demethylated derivative of CTD, which has similar antitumor activity to CTD and lower toxicity than CTD. However, the clinical use of NCTD is limited due to its poor solubility, low bioavailability, and toxic effects on normal cells. To overcome these shortcomings, researchers have explored a number of strategies, such as chemical structural modifications, microsphere dispersion systems, and nanodrug delivery systems. This review summarizes the structure-activity relationship of NCTD and novel strategies to improve the solubility and bioavailability of NCTD as well as reduce the toxicity. This review can provide evidence for further research of NCTD.
Collapse
|
7
|
Yu C, Li Y, Chen G, Wu C, Wang X, Zhang Y. Bioactive constituents of animal-derived traditional Chinese medicinal materials for breast cancer: opportunities and challenges. J Zhejiang Univ Sci B 2022; 23:547-563. [PMID: 35794685 PMCID: PMC9264107 DOI: 10.1631/jzus.b2101019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/20/2022] [Indexed: 11/11/2022]
Abstract
Breast cancer is globally the most common invasive cancer in women and remains one of the leading causes of cancer-related deaths. Surgery, radiotherapy, chemotherapy, immunotherapy, and endocrine therapy are currently the main treatments for this cancer type. However, some breast cancer patients are prone to drug resistance related to chemotherapy or immunotherapy, resulting in limited treatment efficacy. Consequently, traditional Chinese medicinal materials (TCMMs) as natural products have become an attractive source of novel drugs. In this review, we summarized the current knowledge on the active components of animal-derived TCMMs, including Ophiocordycepssinensis-derived cordycepin, the aqueous and ethanolic extracts of O.sinensis, norcantharidin (NCTD), Chansu, bee venom, deer antlers, Ostreagigas, and scorpion venom, with reference to marked anti-breast cancer effects due to regulating cell cycle arrest, proliferation, apoptosis, metastasis, and drug resistance. In future studies, the underlying mechanisms for the antitumor effects of these components need to be further investigated by utilizing multi-omics technologies. Furthermore, large-scale clinical trials are necessary to validate the efficacy of bioactive constituents alone or in combination with chemotherapeutic drugs for breast cancer treatment.
Collapse
Affiliation(s)
- Chaochao Yu
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yi Li
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Guopeng Chen
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Chaoyan Wu
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Xiuping Wang
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yingwen Zhang
- Department of Integrated Chinese and Western Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
8
|
Chillappagari S, Schwarz J, Kesireddy V, Knoell J, Korfei M, Hoetzenecker K, Schmitz ML, Behl C, Bellusci S, Guenther A, Mahavadi P. Therapeutic induction of Bcl2-associated athanogene 3-mediated autophagy in idiopathic pulmonary fibrosis. Clin Transl Med 2022; 12:e935. [PMID: 35834635 PMCID: PMC9282656 DOI: 10.1002/ctm2.935] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 05/25/2022] [Accepted: 06/02/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Exaggerated fibroblast proliferation is a well-known feature in idiopathic pulmonary fibrosis (IPF) which may be - in part - due to insufficient autophagy, a lysosome dependent cellular surveillance pathway. Bcl2-associated athanogene 3 (BAG3) is a pivotal co-chaperone of the autophagy pathway. Here, we studied whether therapeutic modulation of BAG3-mediated autophagy can rescue insufficient autophagy and impact IPF fibroblast proliferation. METHODS Primary interstitial fibroblasts or precision cut lung slices (PCLS) of IPF lungs were treated with (1) the antifibrotic drug pirfenidone (Pirf), (2) the demethylating agent 5-azacytidine (Aza), (3) the BAG3 modulator cantharidin (Ctd). Autophagy flux was measured following pretreatment with the autophagy inhibitors or by GFP-RFP-LC3B transfection followed by drug treatments. Proliferation was measured by 5-bromo-2'-deoxyuridine assay. BAG3, filamin C (FLNC), proliferating-cell-nuclear-antigen (PCNA), collagen1A1 (COL1A1) and autophagy proteins were assessed by immunoblotting or immunofluorescence. Loss of function experiments were performed by siRNA mediated knockdown of BAG3. RESULTS In comparison with healthy donors, increased BAG3 protein was observed in IPF lung homogenates and IPF fibroblasts. In addition, the substrate of BAG3-mediated autophagy, FLNC, was increased in IPF fibroblasts, implying insufficient activation of BAG3-dependent autophagy. Therapeutic modulation of this pathway using Aza and Ctd alone or in combination with the IPF therapy drug Pirf rescued the insufficient BAG3-mediated autophagy and decreased fibroblast proliferation. Such effects were observed upon therapeutic modulation of BAG3 but not upon knock down of BAG3 per se in IPF fibroblasts. Similarly, PCLS of IPF patients showed a significant decrease in collagen deposition in response to these drugs, either alone or in a more potent form in combination with Pirf. CONCLUSIONS Our study reveals that repurposing drugs that modulate autophagy regulating proteins render therapeutic benefits in IPF. Fine tuning of this pathway may hence signify a promising therapeutic strategy to ameliorate antifibrotic properties and augment the efficacy of current IPF therapy.
Collapse
Affiliation(s)
- Shashipavan Chillappagari
- Department of Internal Medicine, Justus-Liebig University (JLU) Giessen, Giessen, Hessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Hessen, Germany
- Department of Biochemistry, Faculty of Medicine, JLU Giessen, Giessen, Hessen, Germany
| | - Julian Schwarz
- Department of Internal Medicine, Justus-Liebig University (JLU) Giessen, Giessen, Hessen, Germany
| | - Vidyasagar Kesireddy
- Department of Internal Medicine, Justus-Liebig University (JLU) Giessen, Giessen, Hessen, Germany
| | - Jessica Knoell
- Department of Internal Medicine, Justus-Liebig University (JLU) Giessen, Giessen, Hessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Hessen, Germany
| | - Martina Korfei
- Department of Internal Medicine, Justus-Liebig University (JLU) Giessen, Giessen, Hessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Hessen, Germany
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Vienna General Hospital, Vienna, Austria
- European IPF Network and European IPF Registry, Giessen, Germany
| | - M Lienhard Schmitz
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Hessen, Germany
- Department of Biochemistry, Faculty of Medicine, JLU Giessen, Giessen, Hessen, Germany
- Member of the Cardio-Pulmonary Institute (CPI), JLU Giessen, Giessen, Germany
| | - Christian Behl
- Institute of Pathobiochemistry, The Autophagy Lab, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Saverio Bellusci
- Department of Internal Medicine, Justus-Liebig University (JLU) Giessen, Giessen, Hessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Hessen, Germany
- Member of the Cardio-Pulmonary Institute (CPI), JLU Giessen, Giessen, Germany
| | - Andreas Guenther
- Department of Internal Medicine, Justus-Liebig University (JLU) Giessen, Giessen, Hessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Hessen, Germany
- European IPF Network and European IPF Registry, Giessen, Germany
- Member of the Cardio-Pulmonary Institute (CPI), JLU Giessen, Giessen, Germany
- Lung Clinic, Agaplesion Evangelisches Krankenhaus Mittelhessen, Giessen, Germany
| | - Poornima Mahavadi
- Department of Internal Medicine, Justus-Liebig University (JLU) Giessen, Giessen, Hessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Centre for Lung Research (DZL), Giessen, Hessen, Germany
| |
Collapse
|
9
|
Role of Induced Programmed Cell Death in the Chemopreventive Potential of Apigenin. Int J Mol Sci 2022; 23:ijms23073757. [PMID: 35409117 PMCID: PMC8999072 DOI: 10.3390/ijms23073757] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 02/01/2023] Open
Abstract
The flavonoid apigenin (4′,5,7-trihydroxyflavone), which is one of the most widely distributed phytochemicals in the plant kingdom, is one of the most thoroughly investigated phenolic components. Previous studies have attributed the physiological effects of apigenin to its anti-allergic, antibacterial, antidiabetic, anti-inflammatory, antioxidant, antiviral, and blood-pressure-lowering properties, and its documented anticancer properties have been attributed to the induction of apoptosis and autophagy, the inhibition of inflammation, angiogenesis, and cell proliferation, and the regulation of cellular responses to oxidative stress and DNA damage. The most well-known mechanism for the compound’s anticancer effects in human cancer cell lines is apoptosis, followed by autophagy, and studies have also reported that apigenin induces novel cell death mechanisms, such as necroptosis and ferroptosis. Therefore, the aim of this paper is to review the therapeutic potential of apigenin as a chemopreventive agent, as well as the roles of programmed cell death mechanisms in the compound’s chemopreventive properties.
Collapse
|
10
|
杨 健, 曾 妍, 吴 小, 王 志. [Effect of DR5-mediated docetaxel-loaded lipid microbubble combined with ultrasoundtargeted microbubble destruction on HepG2 cell proliferation and apoptosis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:1220-1225. [PMID: 34549714 PMCID: PMC8527229 DOI: 10.12122/j.issn.1673-4254.2021.08.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To investigate the effect of DR5-mediated docetaxel-targeted lipid microbubbles (MBs) combined with ultrasound-targeted microbubble destruction on apoptosis and expressions of Bcl-2, nuclear factor-κB(NF-κB), caspase-8, and DR5 in human HepG2 cells. METHODS HepG2 cells were treated with docetaxel at its 50% inhibitory concentration (IC50) of 5 nmol/L, docetaxel combined with ultrasound, blank MBs, blank MBs combined with ultrasound (0.5 W/cm2 for 45 s), drugloaded lipid MBs (DLLM), DLLM combined with ultrasound, DR5-mediated DLLM (DR5-DLLM), or DR5-DLLM combined with ultrasound.After the treatments, the cells were further cultured for 24 h, and CCK-8 assay, TUNEL staining and flow cytometry were used to assess cell proliferation, apoptosis, and cell cycle changes; the changes in mRNA and protein expression levels of Bcl-2, NF-κB, caspase-8, and DR5 were detected with RT-qPCR and Western blotting. RESULTS Among all the treatments, DR5-DLLM combined with ultrasound produced the strongest effects to inhibit the proliferation (P < 0.001), promote apoptosis (P < 0.001), and cause G2/M cell cycle arrest (P < 0.001) in HepG2 cells.The combined treatment with DR5-DLLM and ultrasound also significantly downregulated Bcl-2 and NF-κB (P < 0.001) and upregulated DR5 and caspase-8 expressions (P < 0.001) at both the mRNA and protein levels. CONCLUSION DR5-DLLM combined with ultrasound-targeted microbubble destruction can induce G2/M cell cycle arrest, proliferation inhibition and apoptosis in HepG2 cells by downregulating Bcl-2 and NF-κB and upregulating DR5 and caspase-8 expressions, indicating its value as a novel ultrasoundtargeted therapy for liver cancer.
Collapse
Affiliation(s)
- 健 杨
- 重庆医科大学附属第一医院消化内科, 重庆 400016Department of Gastroenterology, First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - 妍 曾
- 重庆医科大学附属第二医院精神心理科, 重庆 400010Department of Psychology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - 小翎 吴
- 重庆医科大学附属第二医院消化内科, 重庆 400010Department of Gastroenterology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - 志刚 王
- 重庆医科大学超声影像学研究所, 重庆 400010Institue of Ultrasound Imaging, Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
11
|
Razeghian E, Suksatan W, Sulaiman Rahman H, Bokov DO, Abdelbasset WK, Hassanzadeh A, Marofi F, Yazdanifar M, Jarahian M. Harnessing TRAIL-Induced Apoptosis Pathway for Cancer Immunotherapy and Associated Challenges. Front Immunol 2021; 12:699746. [PMID: 34489946 PMCID: PMC8417882 DOI: 10.3389/fimmu.2021.699746] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/05/2021] [Indexed: 01/04/2023] Open
Abstract
The immune cytokine tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has attracted rapidly evolving attention as a cancer treatment modality because of its competence to selectively eliminate tumor cells without instigating toxicity in vivo. TRAIL has revealed encouraging promise in preclinical reports in animal models as a cancer treatment option; however, the foremost constraint of the TRAIL therapy is the advancement of TRAIL resistance through a myriad of mechanisms in tumor cells. Investigations have documented that improvement of the expression of anti-apoptotic proteins and survival or proliferation involved signaling pathways concurrently suppressing the expression of pro-apoptotic proteins along with down-regulation of expression of TRAILR1 and TRAILR2, also known as death receptor 4 and 5 (DR4/5) are reliable for tumor cells resistance to TRAIL. Therefore, it seems that the development of a therapeutic approach for overcoming TRAIL resistance is of paramount importance. Studies currently have shown that combined treatment with anti-tumor agents, ranging from synthetic agents to natural products, and TRAIL could result in induction of apoptosis in TRAIL-resistant cells. Also, human mesenchymal stem/stromal cells (MSCs) engineered to generate and deliver TRAIL can provide both targeted and continued delivery of this apoptosis-inducing cytokine. Similarly, nanoparticle (NPs)-based TRAIL delivery offers novel platforms to defeat barricades to TRAIL therapeutic delivery. In the current review, we will focus on underlying mechanisms contributed to inducing resistance to TRAIL in tumor cells, and also discuss recent findings concerning the therapeutic efficacy of combined treatment of TRAIL with other antitumor compounds, and also TRAIL-delivery using human MSCs and NPs to overcome tumor cells resistance to TRAIL.
Collapse
Affiliation(s)
- Ehsan Razeghian
- Human Genetics Division, Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Suleimanyah, Suleimanyah, Iraq
- Department of Medical Laboratory Sciences, Komar University of Science and Technology, Sulaimaniyah, Iraq
| | - Dmitry O. Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow, Russia
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Faroogh Marofi
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahboubeh Yazdanifar
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Mostafa Jarahian
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
12
|
Zinnah KMA, Park SY. Sensitizing TRAIL‑resistant A549 lung cancer cells and enhancing TRAIL‑induced apoptosis with the antidepressant amitriptyline. Oncol Rep 2021; 46:144. [PMID: 34080659 PMCID: PMC8185507 DOI: 10.3892/or.2021.8095] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/16/2021] [Indexed: 12/20/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a cytokine with the potential to induce cancer cell-specific apoptosis with minimal toxicity to normal cells. Therefore, the resistance of certain cancer cells to TRAIL is a major concern and agents that can either enhance TRAIL capabilities or overcome TRAIL resistance are necessary for the development of cancer treatments. The present study investigated whether the antidepressant drug amitriptyline could sensitize TRAIL-resistant A549 lung cancer cells and enhance TRAIL-induced apoptosis. Antidepressants are usually prescribed to cancer patients to relieve emotional distress, such as depression or dysthymia. The present study revealed for the first time, to the best of our knowledge, that amitriptyline increased death receptor (DR) 4 and 5 expression, a requirement for TRAIL-induced cell death. Genetic inhibitors of DR4 and DR5 significantly reduced amitriptyline-enhanced TRAIL-mediated apoptosis. Additionally, the present study explored whether blocking autophagy increased DR4 and DR5 expression. Blocking autophagy flux with the final stage autophagy inhibitor chloroquine (CQ) also upregulated DR4 and DR5 expression. TRAIL in combination with amitriptyline or CQ significantly increased the expression of apoptosis-indicator proteins cleaved caspase-8 and caspase-3. The expression levels of LC3-II and p62 were significantly higher in amitriptyline-treated cells, which confirmed that amitriptyline blocks autophagy by inhibiting the fusion of autophagosomes with lysosomes. Overall, the present results contributed to understanding the mechanism responsible for the synergistic anticancer effect of amitriptyline and TRAIL and also presented a novel mechanism involved in DR4 and DR5 upregulation.
Collapse
Affiliation(s)
- K M A Zinnah
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk 54596, Republic of Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Jeonbuk National University, Iksan, Jeonbuk 54596, Republic of Korea
| |
Collapse
|
13
|
Xu JL, Yuan L, Tang YC, Xu ZY, Xu HD, Cheng XD, Qin JJ. The Role of Autophagy in Gastric Cancer Chemoresistance: Friend or Foe? Front Cell Dev Biol 2020; 8:621428. [PMID: 33344463 PMCID: PMC7744622 DOI: 10.3389/fcell.2020.621428] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer is the third most common cause of cancer-related death worldwide. Drug resistance is the main inevitable and vital factor leading to a low 5-year survival rate for patients with gastric cancer. Autophagy, as a highly conserved homeostatic pathway, is mainly regulated by different proteins and non-coding RNAs (ncRNAs) and plays dual roles in drug resistance of gastric cancer. Thus, targeting key regulatory nodes in the process of autophagy by small molecule inhibitors or activators has become one of the most promising strategies for the treatment of gastric cancer in recent years. In this review, we provide a systematic summary focusing on the relationship between autophagy and chemotherapy resistance in gastric cancer. We comprehensively discuss the roles and molecular mechanisms of multiple proteins and the emerging ncRNAs including miRNAs and lncRNAs in the regulation of autophagy pathways and gastric cancer chemoresistance. We also summarize the regulatory effects of autophagy inhibitor and activators on gastric cancer chemoresistance. Understanding the vital roles of autophagy in gastric cancer chemoresistance will provide novel opportunities to develop promising therapeutic strategies for gastric cancer.
Collapse
Affiliation(s)
- Jing-Li Xu
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, China.,Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China.,The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yuan
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, China.,Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China.,The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan-Cheng Tang
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tsai, Hong Kong, China
| | - Zhi-Yuan Xu
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, China.,Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Han-Dong Xu
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, China.,Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China.,The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiang-Dong Cheng
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, China.,Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jiang-Jiang Qin
- Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Hangzhou, China.,Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
14
|
Maiese A, De Matteis A, Bolino G, Turillazzi E, Frati P, Fineschi V. Hypo-Expression of Flice-Inhibitory Protein and Activation of the Caspase-8 Apoptotic Pathways in the Death-Inducing Signaling Complex Due to Ischemia Induced by the Compression of the Asphyxiogenic Tool on the Skin in Hanging Cases. Diagnostics (Basel) 2020; 10:diagnostics10110938. [PMID: 33198065 PMCID: PMC7696535 DOI: 10.3390/diagnostics10110938] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 11/16/2022] Open
Abstract
The FLICE-inhibitory protein (c-FLIPL) (55 kDa) is expressed in numerous tissues and most abundantly in the kidney, skeletal muscles and heart. The c-FLIPL has a region of homology with caspase-8 at the carboxy-terminal end which allows the molecule to assume a tertiary structure similar to that of caspases-8 and -10. Consequently, c-FLIPL acts as a negative inhibitor of caspase-8, preventing the processing and subsequent release of the pro-apoptotic molecule active form. The c-FLIP plays as an inhibitor of apoptosis induced by a variety of agents, such as tumor necrosis factor (TNF), T cell receptor (TCR), TNF-related apoptosis inducing ligand (TRAIL), Fas and death receptor (DR). Increased expression of c-FLIP has been found in many human malignancies and shown to be involved in resistance to CD95/Fas and TRAIL receptor-induced apoptosis. We wanted to verify an investigative protocol using FLIP to make a differential diagnosis between skin sulcus with vitality or non-vital skin sulcus in hanged subjects and those undergoing simulated hanging (suspension of the victim after murder). The study group consisted of 21 cases who died from suicidal hanging. The control group consisted of traumatic or natural deaths, while a third group consisted of simulated hanging cases. The reactions to the Anti-FLIP Antibody (Abcam clone-8421) was scored for each section with a semi-quantitative method by means of microscopic observation carried out with confocal microscopy and three-dimensional reconstruction. The results obtained allow us to state that the skin reaction to the FLIP is extremely clear and precise, allowing a diagnosis of unequivocal vitality and a very objective differentiation with the post-mortal skin sulcus.
Collapse
Affiliation(s)
- Aniello Maiese
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126 Pisa PI, Italy; (A.M.); (E.T.)
- IRCCS (Istituto di Ricerca e Cura a Carattere Scientifico) Neuromed Mediterranean Neurological Institute, Via Atinense 18, 86077 Pozzilli IS, Italy;
| | - Alessandra De Matteis
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome RM, Italy; (A.D.M.); (G.B.)
| | - Giorgio Bolino
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome RM, Italy; (A.D.M.); (G.B.)
| | - Emanuela Turillazzi
- Department of Surgical Pathology, Medical, Molecular and Critical Area, Institute of Legal Medicine, University of Pisa, 56126 Pisa PI, Italy; (A.M.); (E.T.)
| | - Paola Frati
- IRCCS (Istituto di Ricerca e Cura a Carattere Scientifico) Neuromed Mediterranean Neurological Institute, Via Atinense 18, 86077 Pozzilli IS, Italy;
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome RM, Italy; (A.D.M.); (G.B.)
| | - Vittorio Fineschi
- IRCCS (Istituto di Ricerca e Cura a Carattere Scientifico) Neuromed Mediterranean Neurological Institute, Via Atinense 18, 86077 Pozzilli IS, Italy;
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome RM, Italy; (A.D.M.); (G.B.)
- Correspondence:
| |
Collapse
|
15
|
Ashrafizadeh M, Bakhoda MR, Bahmanpour Z, Ilkhani K, Zarrabi A, Makvandi P, Khan H, Mazaheri S, Darvish M, Mirzaei H. Apigenin as Tumor Suppressor in Cancers: Biotherapeutic Activity, Nanodelivery, and Mechanisms With Emphasis on Pancreatic Cancer. Front Chem 2020; 8:829. [PMID: 33195038 PMCID: PMC7593821 DOI: 10.3389/fchem.2020.00829] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 08/05/2020] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer is the most lethal malignancy of the gastrointestinal tract. Due to its propensity for early local and distant spread, affected patients possess extremely poor prognosis. Currently applied treatments are not effective enough to eradicate all cancer cells, and minimize their migration. Besides, these treatments are associated with adverse effects on normal cells and organs. These therapies are not able to increase the overall survival rate of patients; hence, finding novel adjuvants or alternatives is so essential. Up to now, medicinal herbs were utilized for therapeutic goals. Herbal-based medicine, as traditional biotherapeutics, were employed for cancer treatment. Of them, apigenin, as a bioactive flavonoid that possesses numerous biological properties (e.g., anti-inflammatory and anti-oxidant effects), has shown substantial anticancer activity. It seems that apigenin is capable of suppressing the proliferation of cancer cells via the induction of cell cycle arrest and apoptosis. Besides, apigenin inhibits metastasis via down-regulation of matrix metalloproteinases and the Akt signaling pathway. In pancreatic cancer cells, apigenin sensitizes cells in chemotherapy, and affects molecular pathways such as the hypoxia inducible factor (HIF), vascular endothelial growth factor (VEGF), and glucose transporter-1 (GLUT-1). Herein, the biotherapeutic activity of apigenin and its mechanisms toward cancer cells are presented in the current review to shed some light on anti-tumor activity of apigenin in different cancers, with an emphasis on pancreatic cancer.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Mohammad Reza Bakhoda
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Bahmanpour
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khandan Ilkhani
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| | - Pooyan Makvandi
- Centre for Micro-BioRobotics, Istituto Italiano di Tecnologia, Pisa, Italy.,Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Samaneh Mazaheri
- Department of Analytical Chemistry, Faculty of Chemistry, University of Kashan, Kashan, Iran
| | - Maryam Darvish
- Department of Medical Biotechnology, Faculty of Medicine, Arak University of Medical Science, Arak, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|