1
|
Sharma A, Balde A, Nazeer RA. A review on animal venom-based matrix metalloproteinase modulators and their therapeutic implications. Int Immunopharmacol 2025; 157:114703. [PMID: 40300352 DOI: 10.1016/j.intimp.2025.114703] [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: 01/18/2025] [Revised: 04/03/2025] [Accepted: 04/19/2025] [Indexed: 05/01/2025]
Abstract
Matrix Metalloproteinases (MMPs) belong to a family of proteolytic enzymes that degrade extracellular matrix components, such as collagen, elastin, laminin, and fibronectin. They also play a part in tissue remodeling by cleaving and rejoining the tissue proteins. Cancer, neurodegenerative disorders, cardiovascular diseases, arthritis, and chronic inflammatory conditions are just some of the diseases that can start or get worse when different MMPs are not working properly. Venomous Animals such as honeybees, toads, snakes, spiders, scorpions, jellyfish, and sea anemones contain venom-secreting glands, which help them defend against predators and immobilize their prey. The molecules that come from animal venom are a complicated mix of bioactive molecules, such as peptides, enzymes, proteins, and small organic compounds that do a number of biological things. Venom-derived molecules have been found to modulate MMP. These venoms and their components target specific signaling pathways, modifying MMP expression levels to either induce inflammation or exhibit anti-inflammatory effects. In this review, we study and explore different MMPs, such as MMP1, MMP2, MMP3, MMP7, MMP8, and MMP9, and their roles in the progression of certain diseases. We also look at different types of molecules derived from marine and land animal venom that are used as MMP modulators. We look at how they work by targeting specific signaling pathways to change MMPs and how they might be used as a medicine to stop diseases by decreasing MMPs.
Collapse
Affiliation(s)
- Ansumaan Sharma
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India.
| |
Collapse
|
2
|
Gao X, Bian T, Wei X, Xu M, Ren J, Zhao Y, Zheng Y, Li X. Exploration of mechanism of Bufalin and Bufalin derivatives in hepatocellular carcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156686. [PMID: 40220402 DOI: 10.1016/j.phymed.2025.156686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/11/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Bufalin, a promising candidate for the treatment of hepatocellular carcinoma (HCC), has garnered interest from researchers in exploring its pharmacological mechanisms. The traditional single-target research method makes it difficult to elucidate the molecular mechanism systematically, and there are still limitations. PURPOSE To explore the synergistic mechanisms of Bufalin and its derivatives in the treatment of HCC through quantitative and integrated analysis of big data, this study provides a reference for clinical screening of anti-liver cancer drugs and accelerates the research and development of new drugs. METHODS The literature on the anti-liver cancer activity of Bufalin was searched, the research trends and hotspots were analyzed, and the hot targets were identified. Potential targets and signaling pathways were predicted and compared with established targets. To evaluate the affinity of Bufalin and its derivatives with key targets and validate the results of network pharmacology. The structure-activity relationships and pathways were reviewed, and key pharmacophore and potential pathways were identified. RESULTS Analysis of 302 literatures showed that Na+/K+-ATPase was a hot target in the anti-liver cancer mechanism of Bufalin. 76 potential targets are primarily associated with pathways such as PI3K/AKT/mTOR, Hedgehog, and AMPK/mTOR, which play roles in regulating key targets like phosphoinositide 3-kinase (PIK3), adenosine monophosphate-activated protein kinase (AMPK), and epidermal growth factor receptor (EGFR). Bufalin derivatives have a higher affinity with key targets. The cdocker interaction energy (CIE) of BF211, compound 1, and compound 2 with PI3K were -48.0722 kcal/mol, -50.8791 kcal/mol, and -59.7326 kcal/mol, respectively, which were significantly lower than Bufalin (-26.0859 kcal/mol). CONCLUSIONS Bufalin and its derivatives have significant anti-HCC activity and show good potential for drug development. Their mechanism of action involves a complex network of AMPK targets and PI3K/AKT/mTOR signaling pathways. In the future, specific inhibitors should be developed against these targets and pathways as a new therapeutic strategy for HCC. Among them, compounds 2, 7, and 8 have a higher affinity for potential targets than Bufalin and its other derivatives and have higher research value. In addition, α-pyrone is the key pharmacophore of Bufalin, and structural modification of the C3 position can significantly regulate its cytotoxicity and solubility. Therefore, optimization of the structure of the C3 position is expected to reduce the toxicity and improve the water solubility of Bufalin, overcoming the limitations of its clinical application and providing a safer and more effective solution for the treatment of HCC.
Collapse
Affiliation(s)
- Xinchen Gao
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Tianrun Bian
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xinyu Wei
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Mengsi Xu
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jiali Ren
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ying Zhao
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yanchao Zheng
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xiankuan Li
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
3
|
Ding Y, Qin J, Zhang M, Wu H, Liu C, Guo Y, Wu W, Jiang Y, Zhang C, Ma Y, Chen X, Lu J, Liu K, Dong Z, Zhao J, Qiao Y. SRPK1 Activation Facilitates Gli3 S664 Phosphorylation and Promotes Metastasis in Esophageal Squamous Cell Carcinoma. Mol Carcinog 2025. [PMID: 40222042 DOI: 10.1002/mc.23913] [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: 07/15/2024] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 04/15/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) stands out as one of the most malignant digestive tumors, with its prognosis marred by frequent metastasis and recurrence. However, the mechanism behind ESCC metastasis remains elusive, impeding therapeutic advancements. SRPK1 emerges as an independent prognostic marker for ESCC patients. Our research illuminates the consequential role of SRPK1, where its genetic knockout led to decreased levels of transcription factors Snail and Slug, concomitant with an enhanced expression of the cell-to-cell adhesion protein E-cadherin. Conversely, reintroducing an overexpression of SRPK1 reversed the effects, highlighting its essential role in ESCC metastasis. Through bioinformatics analysis, we identified a correlation between SRPK1 and Gli3. Furthermore, increased levels of Gli3 and its phosphorylated form, p-Gli3S664, were detected in ESCC tissues, which are implicated in promoting ESCC metastasis. Notably, our research confirmed that SRPK1 promotes migration, invasion, and epithelial-mesenchymal transition (EMT) of ESCC cells through phosphorylates Gli3 at ser 664. Additional investigations reveal that dihydroartemisinin (DHA) effectively impedes ESCC cell metastasis by suppressing SRPK1-mediated phosphorylation of Gli3S664 both in vitro and in vivo. Consequently, our study underscores the pivotal role of the SRPK1-p-Gli3S664 axis in ESCC metastasis and suggests DHA as a promising candidate for preventing ESCC metastasis by targeting this axis.
Collapse
Affiliation(s)
- Yongwei Ding
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Shaoxing People Hospital, Shaoxing, Zhejiang, China
- Henan Provincial Cooperative Innovation Center for Cancer Chemo-Prevention, Zhengzhou, China
| | - Jiace Qin
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Mengjia Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Huiting Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Chang Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaping Guo
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Provincial Cooperative Innovation Center for Cancer Chemo-Prevention, Zhengzhou, China
| | - Wenjie Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Provincial Cooperative Innovation Center for Cancer Chemo-Prevention, Zhengzhou, China
| | - Yanan Jiang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Provincial Cooperative Innovation Center for Cancer Chemo-Prevention, Zhengzhou, China
| | - Chenjuan Zhang
- Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanying Ma
- Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinghuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Provincial Cooperative Innovation Center for Cancer Chemo-Prevention, Zhengzhou, China
| | - Jing Lu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Provincial Cooperative Innovation Center for Cancer Chemo-Prevention, Zhengzhou, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Provincial Cooperative Innovation Center for Cancer Chemo-Prevention, Zhengzhou, China
- The China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Ziming Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Provincial Cooperative Innovation Center for Cancer Chemo-Prevention, Zhengzhou, China
| | - Jimin Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Provincial Cooperative Innovation Center for Cancer Chemo-Prevention, Zhengzhou, China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| | - Yan Qiao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Henan Provincial Cooperative Innovation Center for Cancer Chemo-Prevention, Zhengzhou, China
| |
Collapse
|
4
|
Li Y, Zhou L, Sun K, Guo R, Li Z, Wen Q, Fu G, Yang S. Integrated network pharmacology, proteomics, molecular docking, and experiments in vivo and in vitro to explore the efficacy and potential mechanism of bufalin against hepatocellular carcinoma angiogenesis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119589. [PMID: 40057142 DOI: 10.1016/j.jep.2025.119589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 02/16/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bufalin is a potent bioactive compound extracted from the venom of toads such as Bufo gargarizans. It has rich pharmacological effects, and its traditional applications mainly include anti-cancer, anti-inflammatory and analgesic, especially in cancer treatment, which has been a hot topic of research. Prior research has suggested that bufalin may have anti-tumor angiogenic effects. However, the efficacy and mechanism of bufalin inhibiting hepatocellular carcinoma (HCC) angiogenesis have yet to be further investigated. AIM OF THE STUDY An extensive detailed strategy via network pharmacology, proteomics, histopathological analysis, molecular docking, in vitro experiments, and in vivo magnetic resonance imaging (MRI) examinations were adopted to investigate the efficacy and mechanisms of bufalin against HCC angiogenesis. MATERIALS AND METHODS Micro-vessel density (MVD) and intravoxel incoherent motion (IVIM) perfusion-related parameters based on magnetic resonance diffusion-weighted imaging were used to identify the effect of bufalin against HCC angiogenesis. Potential bufalin and HCC targets were gathered from appropriate databases. The STRING database was used to construct the target protein interaction networks. The "clusterprofiler" package (version 4.2.2) in R was applied to conduct the target-related Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment and Gene Ontology (GO) analysis. Network pharmacology and proteomics were integrated to identify key targets and pathways related to bufalin against HCC angiogenesis. Molecular docking and Western Blot were utilized to validate the findings. RESULTS Analysis through IVIM and MVD showed that bufalin could inhibit HCC angiogenesis in nude mice models. A total of 159 common targets of bufalin and HCC were identified by network pharmacology. GO analysis revealed that these targets focused on multiple angiogenesis-related biological processes, including endothelial cell proliferation and migration, sprouting angiogenesis, and regulation of angiogenesis. The KEGG enrichment results suggested that bufalin could regulate multiple signaling pathways to inhibit HCC angiogenesis, including VEGF, MAPK, PI3K-Akt, mTOR, and HIF-1 signaling pathways. MAPK1, MAPK14, PRKCA, EIF4E, and APEX1 might be critical targets in regulating the above pathways. The molecular docking and Western blot analysis verified the effects of bufalin on target proteins. CONCLUSION This study demonstrated that bufalin might inhibit HCC angiogenesis by regulating multiple targets and pathways. These findings offer theoretical insights and experimental foundations for the clinical application and commercial development of bufalin in the treatment of HCC.
Collapse
Affiliation(s)
- Yuanchao Li
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhi-Jiang Road, Shanghai, 200071, People's Republic of China
| | - Lingwei Zhou
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhi-Jiang Road, Shanghai, 200071, People's Republic of China
| | - Kang Sun
- Department of Laboratory, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhi-Jiang Road, Shanghai, 200071, People's Republic of China
| | - Ran Guo
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhi-Jiang Road, Shanghai, 200071, People's Republic of China
| | - Zehua Li
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhi-Jiang Road, Shanghai, 200071, People's Republic of China
| | - Qingqing Wen
- GE Healthcare, MR Research, Beijing, PK, People's Republic of China
| | - Guifeng Fu
- GE Healthcare, MR Research, Beijing, PK, People's Republic of China
| | - Shuohui Yang
- Department of Radiology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhi-Jiang Road, Shanghai, 200071, People's Republic of China.
| |
Collapse
|
5
|
Zheng J, Wang S, Xia L, Sun Z, Chan KM, Bernards R, Qin W, Chen J, Xia Q, Jin H. Hepatocellular carcinoma: signaling pathways and therapeutic advances. Signal Transduct Target Ther 2025; 10:35. [PMID: 39915447 PMCID: PMC11802921 DOI: 10.1038/s41392-024-02075-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/18/2024] [Accepted: 11/14/2024] [Indexed: 02/09/2025] Open
Abstract
Liver cancer represents a major global health concern, with projections indicating that the number of new cases could surpass 1 million annually by 2025. Hepatocellular carcinoma (HCC) constitutes around 90% of liver cancer cases and is primarily linked to factors incluidng aflatoxin, hepatitis B (HBV) and C (HCV), and metabolic disorders. There are no obvious symptoms in the early stage of HCC, which often leads to delays in diagnosis. Therefore, HCC patients usually present with tumors in advanced and incurable stages. Several signaling pathways are dis-regulated in HCC and cause uncontrolled cell propagation, metastasis, and recurrence of HCC. Beyond the frequently altered and therapeutically targeted receptor tyrosine kinase (RTK) pathways in HCC, pathways involved in cell differentiation, telomere regulation, epigenetic modification and stress response also provide therapeutic potential. Investigating the key signaling pathways and their inhibitors is pivotal for achieving therapeutic advancements in the management of HCC. At present, the primary therapeutic approaches for advanced HCC are tyrosine kinase inhibitors (TKI), immune checkpoint inhibitors (ICI), and combination regimens. New trials are investigating combination therapies involving ICIs and TKIs or anti-VEGF (endothelial growth factor) therapies, as well as combinations of two immunotherapy regimens. The outcomes of these trials are expected to revolutionize HCC management across all stages. Here, we provide here a comprehensive review of cellular signaling pathways, their therapeutic potential, evidence derived from late-stage clinical trials in HCC and discuss the concepts underlying earlier clinical trials, biomarker identification, and the development of more effective therapeutics for HCC.
Collapse
Affiliation(s)
- Jiaojiao Zheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Siying Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Lei Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Zhen Sun
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Kui Ming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, PR China
| | - René Bernards
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wenxin Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jinhong Chen
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, PR China.
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Haojie Jin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
6
|
Liaghat M, Ferdousmakan S, Mortazavi SH, Yahyazadeh S, Irani A, Banihashemi S, Seyedi Asl FS, Akbari A, Farzam F, Aziziyan F, Bakhtiyari M, Arghavani MJ, Zalpoor H, Nabi-Afjadi M. The impact of epithelial-mesenchymal transition (EMT) induced by metabolic processes and intracellular signaling pathways on chemo-resistance, metastasis, and recurrence in solid tumors. Cell Commun Signal 2024; 22:575. [PMID: 39623377 PMCID: PMC11610171 DOI: 10.1186/s12964-024-01957-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024] Open
Abstract
The intricate cellular process, known as the epithelial-mesenchymal transition (EMT), significantly influences solid tumors development. Changes in cell shape, metabolism, and gene expression linked to EMT facilitate tumor cell invasion, metastasis, drug resistance, and recurrence. So, a better understanding of the intricate processes underlying EMT and its role in tumor growth may lead to the development of novel therapeutic approaches for the treatment of solid tumors. This review article focuses on the signals that promote EMT and metabolism, the intracellular signaling pathways leading to EMT, and the network of interactions between EMT and cancer cell metabolism. Furthermore, the functions of EMT in treatment resistance, recurrence, and metastasis of solid cancers are covered. Lastly, treatment approaches that focus on intracellular signaling networks and metabolic alterations brought on by EMT will be discussed.
Collapse
Affiliation(s)
- Mahsa Liaghat
- Department of Medical Laboratory Sciences, Faculty of Medical Sciences, Kazerun Branch, Islamic Azad University, Kazerun, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Saeid Ferdousmakan
- Department of Pharmacy Practice, Nargund College of Pharmacy, Bangalore, 560085, India
| | | | - Sheida Yahyazadeh
- Department of Immunology, Faculty of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Asrin Irani
- Department of Biology, Faculty of Basic Sciences, University of Guilan, Rasht, Iran
| | - Sara Banihashemi
- Department of Bioscience, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | | | - Abdullatif Akbari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farnoosh Farzam
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Aziziyan
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maryam Bakhtiyari
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Mohammad Javad Arghavani
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Hamidreza Zalpoor
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran.
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
7
|
Guo H, Hu Z, Yang X, Yuan Z, Wang M, Chen C, Xie L, Gao Y, Li W, Bai Y, Lin C. Smad4 regulates TGF-β1-mediated hedgehog activation to promote epithelial-to-mesenchymal transition in pancreatic cancer cells by suppressing Gli1 activity. Comput Struct Biotechnol J 2024; 23:1189-1200. [PMID: 38525105 PMCID: PMC10957521 DOI: 10.1016/j.csbj.2024.03.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 03/10/2024] [Accepted: 03/10/2024] [Indexed: 03/26/2024] Open
Abstract
Pancreatic cancer (PC) is an aggressive and metastatic gastrointestinal tumor with a poor prognosis. Persistent activation of the TGF-β/Smad signaling induces PC cell (PCC) invasion and infiltration via epithelial-to-mesenchymal transition (EMT). Hedgehog signaling is a crucial pathway for the development of PC via the transcription factors Gli1/2/3. This study aimed to investigate the underlying molecular mechanisms of action of hedgehog activation in TGF-β1-triggered EMT in PCCs (PANC-1 and BxPc-3). In addition, overexpression and shRNA techniques were used to evaluate the role of Smad4 in TGF-β1-treated PCCs. Our data showed that TGF-β1 promoted PCC invasion and infiltration via Smad2/3-dependent EMT. Hedgehog-Gli signaling axis in PCCs was activated upon TGF-β1 stimulation. Inhibition of hedgehog with cyclopamine effectively antagonized TGF-β1-induced EMT, thereby suggesting that the hedgehog signaling may act as a downstream cascade signaling of TGF-β1. As a key protein that assists the nuclear translocation of Smad2/3, Smad4 was highly expressed in PANC-1 cells, but not in BxPc-3 cells. Conversely, Gli1 expression was low in PANC-1 cells, but high in BxPc-3 cells. Furthermore, knockdown of Smad4 in PANC-1 cells by shRNA inhibited TGF-β1-mediated EMT and collagen deposition. Overexpression of Smad4 did not affect TGF-β1-mediated EMT due to the lack of significant increase in nuclear expression of Smad4. Importantly, Gli1 activity was upregulated by Smad4 knockdown in PANC-1 cells and downregulated by Smad4 overexpression in BxPc-3 cells, indicating that Gli1 may be a negative target protein downstream of Smad4. Thus, Smad4 regulates TGF-β1-mediated hedgehog activation to promote EMT in PCCs by suppressing Gli1 activity.
Collapse
Affiliation(s)
- Hangcheng Guo
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
- Sichuan Mianyang 404 Hospital, Mianyang 621000, China
| | - Zujian Hu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Xuejia Yang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Ziwei Yuan
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Mengsi Wang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Chaoyue Chen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Lili Xie
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Yuanyuan Gao
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Wangjian Li
- Department of Urology, The Central Hospital Affiliated to Shaoxing University, Shaoxing 312030, China
| | - Yongheng Bai
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
- National Key Clinical Specialty (General Surgery), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Chunjing Lin
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
- Medicine and Health Care Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
8
|
Tian H, Zhao F, Yue BS, Zhai BT. Combinational Antitumor Strategies Based on the Active Ingredients of Toad Skin and Toad Venom. Drug Des Devel Ther 2024; 18:3549-3594. [PMID: 39139676 PMCID: PMC11321342 DOI: 10.2147/dddt.s469832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/25/2024] [Indexed: 08/15/2024] Open
Abstract
A multidrug combination strategy is an important mean to improve the treatment of cancer and is the mainstream scheme of clinical cancer treatment. The active ingredients of traditional Chinese medicine, represented by toad skin and toad venom, have the advantages of high efficiency, low toxicity, wide action and multiple targets and have become ideal targets in combined treatment strategies for tumors in recent years. Toad skin and toad venom are traditional Chinese animal medicines derived from Bufo bufo gargarizans Cantor or Bufo melanostictus Schneider that have shown excellent therapeutic effects on the treatment of various cancers and cancer pain as adjuvant antitumor drugs in clinical practice. The involved mechanisms include inducing apoptosis, arresting the cell cycle, inhibiting cell proliferation, migration and invasion, inhibiting tumor angiogenesis, reversing the multidrug resistance of tumor cells, and regulating multiple signaling pathways and targets. Moreover, a multidrug combination strategy based on a nanodelivery system can realize the precise loading of the active ingredients of toad skin or toad venom and other antitumor drugs and carry drugs to overcome physiological and pathological barriers, complete efficient enrichment in tumor tissues, and achieve targeted delivery to tumor cells and the controlled release of drugs, thus enhancing antitumor efficacy and reducing toxicity and side effects. This article reviewed the clinical efficacy and safety of the combination of toad skin and toad venom with chemotherapeutic drugs, targeted drugs, analgesics and other drugs; evaluated the effects and mechanisms of the combination of toad skin and toad venom with chemotherapy, targeted therapy, radiotherapy or hyperthermia, traditional Chinese medicine, signaling pathway inhibitors and other therapies in cell and animal models; and summarized the codelivery strategies for the active ingredients of toad skin and toad venom with chemotherapeutic drugs, small-molecule targeted drugs, monoclonal antibodies, active ingredients of traditional Chinese medicine, and photodynamic and photothermal therapeutic drugs to provide a basis for the rational drug use of toad skin and toad venom in the clinic and the development of novel drug delivery systems.
Collapse
Affiliation(s)
- Huan Tian
- Department of Pharmacy, Xi’an Hospital of Traditional Chinese Medicine, Xi’an, People’s Republic of China
| | - Feng Zhao
- Department of Pharmacy, Xi’an Hospital of Traditional Chinese Medicine, Xi’an, People’s Republic of China
| | - Bao-Sen Yue
- Department of Pharmacy, Xi’an Hospital of Traditional Chinese Medicine, Xi’an, People’s Republic of China
| | - Bing-Tao Zhai
- College of Pharmacy, Shaanxi University of Chinese Medicine, Xi’an, People’s Republic of China
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Xi’an, People’s Republic of China
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Xi’an, People’s Republic of China
- Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Xi’an, People’s Republic of China
| |
Collapse
|
9
|
Mhaidly N, Barake N, Trelcat A, Journe F, Saussez S, Descamps G. Bufalin Suppresses Head and Neck Cancer Development by Modulating Immune Responses and Targeting the β-Catenin Signaling Pathway. Cancers (Basel) 2024; 16:2739. [PMID: 39123466 PMCID: PMC11311268 DOI: 10.3390/cancers16152739] [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: 06/26/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Bufalin, a cardiotonic steroid derived from the Chinese toad (Bufo gargarizans), has demonstrated potent anticancer properties across various cancer types, positioning it as a promising therapeutic candidate. However, comprehensive mechanistic studies specific to head and neck cancers have been lacking. Our study aimed to bridge this gap by investigating bufalin's mechanisms of action in head and neck cancer cells. Using several methods, such as Western blotting, immunofluorescence, and flow cytometry, we observed bufalin's dose-dependent reduction in cell viability, disruption of cell membrane integrity, and inhibition of colony formation in both HPV-positive and HPV-negative cell lines. Bufalin induces apoptosis through the modulation of apoptosis-related proteins, mitochondrial function, and reactive oxygen species production. It also arrests the cell cycle at the G2/M phase and attenuates cell migration while affecting epithelial-mesenchymal transition markers and targeting pivotal signaling pathways, including Wnt/β-catenin, EGFR, and NF-κB. Additionally, bufalin exerted immunomodulatory effects by polarizing macrophages toward the M1 phenotype, bolstering antitumor immune responses. These findings underscore bufalin's potential as a multifaceted therapeutic agent against head and neck cancers, targeting essential pathways involved in proliferation, apoptosis, cell cycle regulation, metastasis, and immune modulation. Further research is warranted to validate these mechanisms and optimize bufalin's clinical application.
Collapse
Affiliation(s)
- Nour Mhaidly
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| | - Noura Barake
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| | - Anne Trelcat
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| | - Fabrice Journe
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium;
| | - Sven Saussez
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| | - Géraldine Descamps
- Department of Human Anatomy and Experimental Oncology, Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars, 8, 7000 Mons, Belgium; (N.M.); (N.B.); (A.T.); (S.S.)
| |
Collapse
|
10
|
Wang Y, Zhang A, Li Q, Liu C. Modulating pancreatic cancer microenvironment: The efficacy of Huachansu in mouse models via TGF-β/Smad pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117872. [PMID: 38325667 DOI: 10.1016/j.jep.2024.117872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/07/2024] [Accepted: 02/04/2024] [Indexed: 02/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huachansu (HCS) is a traditional Chinese medicine obtained from the dried skin glands of Bufo gargarizans and clinical uses of HCS have been approved in China to treat malignant tumors. The traditional Chinese medicine theory states that HCS relieves patients with cancer by promoting blood circulation to remove blood stasis. Clinical observation found that local injection of HCS given to pancreatic cancer patients can significantly inhibit tumor progression and assist in enhancing the efficacy of chemotherapy. However, the material basis and underlying mechanism have not yet been elucidated. AIM OF THE STUDY To investigate the therapeutic potential of HCS for the treatment of pancreatic cancer in in situ transplanted tumor nude mouse model. Furthermore, this study sought to elucidate the molecular mechanisms underlying its efficacy and assess the impact of HCS on the microenvironment of pancreatic cancer. To identify the antitumor effect of HCS in in situ transplanted tumor nude mouse model and determine the Chemopreventive mechanism of HCS on tumor microenvironment (TME). METHODS Using the orthotopic transplantation nude mouse model with fluorescently labeled pancreatic cancer cell lines SW1990 and pancreatic stellate cells (PSCs), we examined the effect of HCS on the pancreatic ductal adenocarcinoma (PDAC) microenvironment based on the transforming growth factor β (TGF-β)/Smad pathway. The expression of TGF-β, smad2, smad3, smad4, collagen type-1 genes and proteins in nude mouse model were detected by qRT-PCR and Western blot. RESULTS HCS significantly reduced tumor growth rate, increased the survival rate, and ameliorated the histopathological changes in the pancreas. It was found that HCS concentration-dependently reduced the expression of TGF-β1 and collagen type-1 genes and proteins, decreased the expression of Smad2 and Smad3 genes, and downregulated the phosphorylation level of Smad2/3. Additionally, the gene and protein expression of Smad4 were promoted by HCS. Further, the promoting effect gradually enhanced with the rise of HCS concentration. CONCLUSIONS The results demonstrated HCS could regulate the activity of the TGF-β/Smad pathway in PDAC, improved the microenvironment of PDAC and delayed tumor progression. This study not only indicated that the protective mechanism of HCS on PDAC might be attributed partly to the inhibition of cytokine production and the TGF-β/Smad pathway, but also provided evidence for HCS as a potential medicine for PDAC treatment.
Collapse
Affiliation(s)
- Yuehui Wang
- Beijing University of Chinese Medicine, Beijing, 100105, China.
| | - Arun Zhang
- Beijing University of Chinese Medicine, Beijing, 100105, China.
| | - Quanwang Li
- Oncology Department, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China.
| | - Chuanbo Liu
- Oncology Department, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China.
| |
Collapse
|
11
|
Jeng KS, Chang CF, Tsang YM, Sheen IS, Jeng CJ. Reappraisal of the Roles of the Sonic Hedgehog Signaling Pathway in Hepatocellular Carcinoma. Cancers (Basel) 2024; 16:1739. [PMID: 38730691 PMCID: PMC11083695 DOI: 10.3390/cancers16091739] [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: 04/05/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
HCC remains one of the leading causes of cancer-related death globally. The main challenges in treatments of hepatocellular carcinoma (HCC) primarily arise from high rates of postoperative recurrence and the limited efficacy in treating advanced-stage patients. Various signaling pathways involved in HCC have been reported. Among them, the Sonic hedgehog (SHH) signaling pathway is crucial. The presence of SHH ligands is identified in approximately 60% of HCC tumor tissues, including tumor nests. PTCH-1 and GLI-1 are detected in more than half of HCC tissues, while GLI-2 is found in over 84% of HCC tissues. The SHH signaling pathway (including canonical and non-canonical) is involved in different aspects of HCC, including hepatocarcinogenesis, tumor growth, tumor invasiveness, progression, and migration. The SHH signaling pathway also contributes to recurrence, metastasis, modulation of the cancer microenvironment, and sustaining cancer stem cells. It also affects the resistance of HCC cells to chemotherapy, target therapy, and radiotherapy. Reappraisal of the roles of the SHH signaling pathway in HCC may trigger some novel therapies for HCC.
Collapse
Affiliation(s)
- Kuo-Shyang Jeng
- Department of Surgery, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Chiung-Fang Chang
- Department of Medical Research, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan;
| | - Yuk-Ming Tsang
- Department of Imaging Medicine, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan;
| | - I-Shyan Sheen
- Department of Gastroenterology & Hepatology, Linkou Chang Memorial Hospital, Chang Gung Medical Foundation, Taoyuan City 333, Taiwan;
| | - Chi-Juei Jeng
- Graduate Institude of Clinical Medicine, National Taiwan University, College of Medicine, Taipei City 10617, Taiwan;
| |
Collapse
|
12
|
Sun Y, Li X, Cheng H, Wang S, Zhou D, Ding J, Ma F. Drug resistance and new therapies in gallbladder cancer. Drug Discov Ther 2023; 17:220-229. [PMID: 37587052 DOI: 10.5582/ddt.2023.01013] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
Gallbladder cancer (GBC) is a highly aggressive malignancy, which poses significant challenges for timely diagnosis, resulting in a dismal prognosis. Chemotherapy serves as a primary treatment option in cases where surgery is not feasible. However, the emergence of chemoresistance poses a significant challenge to the effectiveness of chemotherapy, ultimately resulting in a poor prognosis. Despite extensive research on mechanisms of chemotherapeutic resistance in oncology, the underlying mechanisms of chemoresistance in GBC remain poorly understood. In this review, we present the findings from the last decade on the molecular mechanisms of chemotherapeutic resistance in GBC. We hope that these insights may provide novel therapeutic and experimental targets for further investigations into this lethal disease.
Collapse
Affiliation(s)
- Yuxin Sun
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxuan Li
- Qingdao University, Qingdao, Shandong, China
| | - Haihong Cheng
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shouhua Wang
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Zhou
- Department of General Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Ding
- Department of Biliary and Pancreatic Surgery, Shanghai Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fei Ma
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Pediatric Research, Shanghai, China
| |
Collapse
|
13
|
Bufalin-Mediated Regulation of Cell Signaling Pathways in Different Cancers: Spotlight on JAK/STAT, Wnt/β-Catenin, mTOR, TRAIL/TRAIL-R, and Non-Coding RNAs. Molecules 2023; 28:molecules28052231. [PMID: 36903477 PMCID: PMC10004807 DOI: 10.3390/molecules28052231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 03/04/2023] Open
Abstract
The renaissance of research into natural products has unequivocally and paradigmatically shifted our knowledge about the significant role of natural products in cancer chemoprevention. Bufalin is a pharmacologically active molecule isolated from the skin of the toad Bufo gargarizans or Bufo melanostictus. Bufalin has characteristically unique properties to regulate multiple molecular targets and can be used to harness multi-targeted therapeutic regimes against different cancers. There is burgeoning evidence related to functional roles of signaling cascades in carcinogenesis and metastasis. Bufalin has been reported to regulate pleiotropically a myriad of signal transduction cascades in various cancers. Importantly, bufalin mechanistically regulated JAK/STAT, Wnt/β-Catenin, mTOR, TRAIL/TRAIL-R, EGFR, and c-MET pathways. Furthermore, bufalin-mediated modulation of non-coding RNAs in different cancers has also started to gain tremendous momentum. Similarly, bufalin-mediated targeting of tumor microenvironments and tumor macrophages is an area of exciting research and we have only started to scratch the surface of the complicated nature of molecular oncology. Cell culture studies and animal models provide proof-of-concept for the impetus role of bufalin in the inhibition of carcinogenesis and metastasis. Bufalin-related clinical studies are insufficient and interdisciplinary researchers require detailed analysis of the existing knowledge gaps.
Collapse
|
14
|
Soumoy L, Ghanem GE, Saussez S, Journe F. Bufalin for an innovative therapeutic approach against cancer. Pharmacol Res 2022; 184:106442. [PMID: 36096424 DOI: 10.1016/j.phrs.2022.106442] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/30/2022] [Accepted: 09/07/2022] [Indexed: 11/19/2022]
Abstract
Bufalin is an endogenous cardiotonic steroid, first discovered in toad venom but also found in the plasma of healthy humans, with anti-tumour activities in different cancer types. The current review is focused on its mechanisms of action and highlights its very large spectrum of effects both in vitro and in vivo. All leads to the conclusion that bufalin mediates its effects by affecting all the hallmarks of cancer and seems restricted to cancer cells avoiding side effects. Bufalin decreases cancer cell proliferation by acting on the cell cycle and inducing different mechanisms of cell death including apoptosis, necroptosis, autophagy and senescence. Bufalin also moderates metastasis formation by blocking migration and invasion as well as angiogenesis and by inducing a phenotype switch towards differentiation and decreasing cancer cell stemness. Regarding its various mechanisms of action in cancer cells, bufalin blocks overactivated signalling pathways and modifies cell metabolism. Moreover, bufalin gained lately a huge interest in the field of drug resistance by both reversing various drug resistance mechanisms and affecting the immune microenvironment. Together, these data support bufalin as a quite promising new anti-cancer drug candidate.
Collapse
Affiliation(s)
- Laura Soumoy
- Laboratory of Human Anatomy & Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000 Mons, Belgium.
| | - Ghanem E Ghanem
- Laboratory of Clinical and Experimental Oncology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium
| | - Sven Saussez
- Laboratory of Human Anatomy & Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000 Mons, Belgium
| | - Fabrice Journe
- Laboratory of Human Anatomy & Experimental Oncology, Faculty of Medicine and Pharmacy, University of Mons (UMONS), 7000 Mons, Belgium; Laboratory of Clinical and Experimental Oncology, Institut Jules Bordet, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium.
| |
Collapse
|
15
|
Ning Z, Zhao Y, Yan X, Hua Y, Meng Z. Flower-like Composite Material Delivery of Co-Packaged Lenvatinib and Bufalin Prevents the Migration and Invasion of Cholangiocarcinoma. NANOMATERIALS 2022; 12:nano12122048. [PMID: 35745387 PMCID: PMC9230555 DOI: 10.3390/nano12122048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/20/2022] [Indexed: 02/01/2023]
Abstract
The co-delivery of multiple drugs using nanocarriers has been recognized as a promising strategy for cancer treatment to enhance therapeutic efficacy. In this study, a monodisperse mesoporous silica nanoparticle (mSiO2) is prepared and functionalized into high-efficiency loaded Lenvatinib and Bufalin for targeted delivery to Cholangiocarcinoma (CCA). mSiO2 was synthesized on solid silica nanoparticles by oil–water interface method, and highly monodisperse mSiO2 with uniform morphology was obtained. mSiO2 was then sequentially modified by polyethylene glycol (PEG) and the targeting molecule folic acid (FA). mSiO2-FA was designed as co-delivery system for Lenvatinib (Le) and Bufalin (Bu) to increase drug availability and highly target tumor cells. Compared with unfunctionalized mSiO2, mSiO2-FA can more efficiently enter human CCA cell lines (9810 cells) and enhance intracellular drug delivery. Moreover, drug-loaded mSiO2-FA (Le/Bu@mSiO2-FA) significantly inhibited the viability, migration and invasion of 9810 cells. In vivo, the nanocomplex significantly reduced the tumor load in CCA tumor-bearing mouse models compared to Le or Bu alone. The current work provides a useful strategy for highly targeted and multidrug-resistance reversal therapy for CCA.
Collapse
|
16
|
Jiang HY, Zheng HM, Xia C, Li X, Wang G, Zhao T, Cui XN, Wang RY, Liu Y. The Research Progress of Bufalin in the Treatment of Hepatocellular Carcinoma. Onco Targets Ther 2022; 15:291-298. [PMID: 35345394 PMCID: PMC8957335 DOI: 10.2147/ott.s333233] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 03/07/2022] [Indexed: 11/23/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest cancers in the world with a five-year survival rate of less than 20%. Nonetheless, selecting an appropriate therapeutic agent to inhibit the development of hepatoma cells is still a challenge. Bufalin, a component of the traditional Chinese medicine Chansu, has been shown to inhibit the proliferation, invasion and metastasis of HCC through various signaling pathways. In addition, bufalin and sorafenib demonstrate a synergistic effect in cancer therapeutics. This review highlighted on several focal signaling pathways involved in the inhibitory effects of bufalin on HCC and its synergistic mechanisms with sorafenib. The immunotherapy effect of bufalin has also been discussed as a novel property.
Collapse
Affiliation(s)
- Han-Yu Jiang
- Department of Oncology, The Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Hui-Min Zheng
- Department of Oncology, The Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Cheng Xia
- Department of Oncology, The Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Xiang Li
- Department of Oncology, The Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Gang Wang
- Department of Oncology, The Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Tong Zhao
- Department of Oncology, The Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China
| | - Xiao-Nan Cui
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People's Republic of China
| | - Ruo-Yu Wang
- Department of Oncology, The Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China.,The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian, People's Republic of China
| | - Ying Liu
- Department of Oncology, The Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China.,The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian, People's Republic of China
| |
Collapse
|
17
|
Zheng X, Liu X, Lei Y, Wang G, Liu M. Glypican-3: A Novel and Promising Target for the Treatment of Hepatocellular Carcinoma. Front Oncol 2022; 12:824208. [PMID: 35251989 PMCID: PMC8889910 DOI: 10.3389/fonc.2022.824208] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/18/2022] [Indexed: 02/05/2023] Open
Abstract
Glypican-3 (GPC3) is a membrane-associated proteoglycan that is specifically up-regulated in hepatocellular carcinoma (HCC) although rarely or not expressed in normal liver tissues, making it a perfect diagnostic and treatment target for HCC. Several GPC3-based clinical trials are ongoing and recently several innovative GPC3-targeted therapeutic methods have emerged with exciting results, including GPC3 vaccine, anti-GPC3 immunotoxin, combined therapy with immune checkpoint blockades (ICBs), and chimeric antigen receptor (CAR) T or NK cells. Here, we review the value of GPC3 in the diagnosis and prognosis of HCC, together with its signaling pathways, with a specific focus on GPC3-targeted treatments of HCC and some prospects for the future GPC3-based therapeutic strategies in HCC.
Collapse
Affiliation(s)
- Xiufeng Zheng
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Xun Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Yanna Lei
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Gang Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Ming Liu
- Department of Abdominal Oncology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Traditional Chinese medicine for prevention and treatment of hepatocellular carcinoma: A focus on epithelial-mesenchymal transition. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2021; 19:469-477. [PMID: 34538644 DOI: 10.1016/j.joim.2021.08.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 04/21/2021] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent malignant cancers worldwide. Epithelial-mesenchymal transition (EMT), which endows epithelial cells with mesenchymal properties, plays an important role in the early stages of metastasis. Conventional cancer therapies have promising effects, but issues remain, such as high rates of metastasis and drug resistance. Thus, exploring and evaluating new therapies is an urgent need. Traditional Chinese medicines (TCMs) have been acknowledged for their multi-target and coordinated intervention effects against HCC. Accumulating evidence indicates that TCM can inhibit the malignancy of cells and the progression of EMT in HCC. However, studies on the effects of TCM on EMT in HCC are scarce. In this review, we summarized recent developments in anti-EMT TCMs and formulae, focusing on their underlying pharmacological mechanisms, to provide a foundation for further research on the exact mechanisms through which TCM affects EMT in HCC.
Collapse
|
19
|
Zou D, Song J, Deng M, Ma Y, Yang C, Liu J, Wang S, Wen Z, Tang Y, Qu X, Zhang Y. Bufalin inhibits peritoneal dissemination of gastric cancer through endothelial nitric oxide synthase-mitogen-activated protein kinases signaling pathway. FASEB J 2021; 35:e21601. [PMID: 33913201 DOI: 10.1096/fj.202002780r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/19/2021] [Accepted: 04/01/2021] [Indexed: 01/11/2023]
Abstract
Peritoneal dissemination threatens the survival of patients with gastric cancer (GC). Bufalin is an extract of traditional Chinese medicine, which has been proved to have anticancer effect. The target of bufalin in suppressing gastric cancer peritoneal dissemination (GCPD) and the underlying mechanism are still unclear. In this research, GC cell line MGC-803 and high-potential peritoneal dissemination cell line MKN-45P were treated with bufalin or L-NAME. Malignant biological behavior and protein level of GC cell lines were detected with MTT, wound healing, transwell, adhesion, and western blotting. Bioinformatics analysis and patient tissues were used to verify the role of endothelial nitric oxide synthase (NOS3) in GC. Mice model was used to assess the effect of bufalin and role of NOS3 in vivo. We found that bufalin inhibited the proliferation, invasion, and migration in GC cell lines. NOS3, which was an independent prognostic factor of GC patients, was predicted to be a potential target of bufalin. Further experiments proved that bufalin reduced the phosphorylation of NOS3, thereby inhibiting the mitogen-activated protein kinase (MAPK) signaling pathway, and ultimately suppressed GCPD by inhibiting EMT process. In conclusion, NOS3 was a potential therapeutic target and prognostic biomarker of GC. Bufalin could suppress GCPD through NOS3-MAPK signaling pathway, which provided more evidence support for intraperitoneal perfusion of bufalin to treat GCPD.
Collapse
Affiliation(s)
- Dan Zou
- The First Laboratory of Cancer Institute, the First Hospital of China Medical University, Shenyang, China
| | - Jincheng Song
- The First Laboratory of Cancer Institute, the First Hospital of China Medical University, Shenyang, China.,Department of Oncology, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Mingming Deng
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yanju Ma
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, Shenyang, China
| | - Chunjiao Yang
- The First Laboratory of Cancer Institute, the First Hospital of China Medical University, Shenyang, China
| | - Jiaqing Liu
- The First Laboratory of Cancer Institute, the First Hospital of China Medical University, Shenyang, China
| | - Song Wang
- The First Laboratory of Cancer Institute, the First Hospital of China Medical University, Shenyang, China
| | - Zhenpeng Wen
- The First Laboratory of Cancer Institute, the First Hospital of China Medical University, Shenyang, China
| | - Yu Tang
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Insititute, Shenyang, China
| | - Xiujuan Qu
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, China
| | - Ye Zhang
- The First Laboratory of Cancer Institute, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
20
|
Mir IH, Guha S, Behera J, Thirunavukkarasu C. Targeting molecular signal transduction pathways in hepatocellular carcinoma and its implications for cancer therapy. Cell Biol Int 2021; 45:2161-2177. [PMID: 34270844 DOI: 10.1002/cbin.11670] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/22/2021] [Accepted: 07/11/2021] [Indexed: 12/27/2022]
Abstract
Hepatocellular carcinoma is a substantial health concern. It is currently the third dominating cause of mortality associated with cancer worldwide. The development of hepatocellular carcinoma is an intricate process that encompasses the impairment of genetic, epigenetic, and signal transduction mechanisms contributing to an aberrant metabolic system, enabling tumorigenesis. Throughout the past decade, research has led to the revelation of molecular pathways implicated in the progression of this notorious disorder. The altered signal transduction pathways, such as the mitogen-activated protein kinase pathway, phosphoinositol 3-kinase/protein kinase B/mammalian target of rapamycin pathway, WNT/β-catenin pathway, hepatocyte growth factor/c-MET pathway, and just another kinase/signal transducers and activators of transcription signaling pathway is of much therapeutic significance, as targeting them may avail to revert, retard or avert hepatocarcinogenesis. The present review article sums up the contemporary knowledge of such signaling mechanisms, including their therapeutic targets and betokens that novel and efficacious therapies can be developed only by the keen understanding of their character in hepatocarcinogenesis. In additament, we address the role of consequential therapeutic agents and preclinical nondrug therapies known for combating hepatocarcinogenesis.
Collapse
Affiliation(s)
- Ishfaq Hassan Mir
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | - Shreyoshi Guha
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | - Jajnasenee Behera
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | | |
Collapse
|
21
|
Song J, Zou D, Zhao X, Chen Y, Lv F, Wang S, Sui D, Han Q, Yang C, Wang X, Liu B, Deng M, Zhang Y. Bufalin inhibits human diffuse large B-cell lymphoma tumorigenesis by inducing cell death through the Ca2+/NFATC1/cMYC pathway. Carcinogenesis 2021; 42:303-314. [PMID: 33124657 DOI: 10.1093/carcin/bgaa108] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 09/22/2020] [Accepted: 10/13/2020] [Indexed: 01/01/2023] Open
Abstract
The 5-year survival rate of diffuse large B-cell lymphoma (DLBCL) can reach 60%. However, nearly half of patients undergo relapse/refractory issues with a survival period of less than 2 years. New therapeutic approaches are therefore needed to improve chemotherapy efficacy and patient survival. Bufalin (BF), isolated from the traditional Chinese medicine Chansu, has been reported to play an anticancer role in multiple cancer cell types. However, there are few reports of the effects of BF on the growth of DLBCL. In the present study, we demonstrated that BF exerts antitumor activity in DLBCL cells, both in vitro and in vivo. Treatment of DLBCL cells with BF resulted in increased proliferation and apoptosis in a dose- and time-dependent manner. Daily intraperitoneal injection of 1.5 mg/kg BF significantly delayed DLBCL xenograft growth in NOD/SCID mice without affecting body weight. Bioinformatics analysis showed that BF may regulate NFATC1 protein and affect expression of its downstream gene, cMYC. Our results suggest that BF can attenuate NFATC1 translocation by reducing the intracellular calcium concentration; BF may also have a low synergistic effect with cyclosporin A. In conclusion, we demonstrated that BF exerts antitumor activity that is mediated at least in part by the Ca2+/NFATC1/cMYC pathway. Our findings suggest that BF can be effectively applied as a novel potential therapeutic agent for DLBCL.
Collapse
Affiliation(s)
- Jincheng Song
- The First Laboratory of Cancer Institute, The First Affiliated Hospital of China Medical University, Shenyang, PR China.,Department of Lymphoma, Lymphoma and Myeloma Diagnosis and Treatment Center, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, PR China
| | - Dan Zou
- The First Laboratory of Cancer Institute, The First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Xiaoxuan Zhao
- Department of Dermatology, Dalian Dermatosis Hospital, Dalian, Liaoning, PR China
| | - Yang Chen
- The First Laboratory of Cancer Institute, The First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Fei Lv
- The First Laboratory of Cancer Institute, The First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Song Wang
- The First Laboratory of Cancer Institute, The First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Dan Sui
- The First Laboratory of Cancer Institute, The First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Qiuyue Han
- The First Laboratory of Cancer Institute, The First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Chunjiao Yang
- The First Laboratory of Cancer Institute, The First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Ximing Wang
- The First Laboratory of Cancer Institute, The First Affiliated Hospital of China Medical University, Shenyang, PR China
| | - Bofang Liu
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Mingming Deng
- Department of Respiratory and Infectious Disease of Geriatrics, The First Hospital of China Medical University, Shenyang, PR China
| | - Ye Zhang
- The First Laboratory of Cancer Institute, The First Affiliated Hospital of China Medical University, Shenyang, PR China
| |
Collapse
|
22
|
Sheng X, Zhu P, Zhao Y, Zhang J, Li H, Zhao H, Qin J. Effect of PI3K/AKT/mTOR Signaling Pathway on Regulating and Controlling the Anti-Invasion and Metastasis of Hepatoma Cells by Bufalin. Recent Pat Anticancer Drug Discov 2021; 16:54-65. [PMID: 33530915 DOI: 10.2174/1574892816666210201120324] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 01/30/2023]
Abstract
BACKGROUND Autophagy plays a "double-edged sword" in the process of tumorigenesis, development and metastasis. OBJECTIVE In this study, we explored the effect of PI3K/AKT/mTOR autophagy-related signaling pathway on regulating and controlling the invasion and metastasis of liver cancer cells by Bufalin. METHODS The cell counting, migration, adhesion and invasion assay were used to evaluate the effect of Bufalin on cell proliferation, invasion and metastasis. The protein expression of PI3K/AKT/ mTOR signaling pathway were detected by the Western Blotting technique. RESULTS After inhibiting autophagy of HCC-LM3 cells, the inhibitory effect of Bufalin on adhesion, migration and invasion of HCC-LM3 cells was significantly enhanced. Synergistic inhibition was strongest when different autophagy inhibitors were combined with 3MA and CQ. After inhibiting autophagy, Bufalin significantly inhibited the protein expression of P-AKT, Cyclin D1, MMP- 2, MMP-9 and VEGF in HCC-LM3 cells. The protein expression of PTEN and E-Cadherin in HCC-LM3 cells was significantly increased. CONCLUSION The present study shows that the anti-tumor effect of Bufalin mainly inhibit proliferation, extracellular matrix degradation and angiogenesis of HCC by influencing autophagy. These findings confirm the capability of Bufalin in inhibiting metastasis of HCC and in parallel to current patents, could be applied as a novel therapeutic strategy in the prevention of metastasis of HCC.
Collapse
Affiliation(s)
- Xia Sheng
- Department of Pathology, Affiliated to Minhang Hospital, Fudan University, Xinsong Rd. 170, Minhang District, Shanghai 201100, China
| | - Pengfei Zhu
- Department of General Surgery, Affiliated to the Third Hospital, Naval Medical University, North Moyu Rd. 700, Jiangding District, Shanghai 201805, China
| | - Yi Zhao
- Department of General Surgery, Affiliated to the Third Hospital, Naval Medical University, North Moyu Rd. 700, Jiangding District, Shanghai 201805, China
| | - Jinwei Zhang
- Department of General Surgery, Affiliated to the Third Hospital, Naval Medical University, North Moyu Rd. 700, Jiangding District, Shanghai 201805, China
| | - Haijia Li
- Department of General Surgery, Affiliated to the Third Hospital, Naval Medical University, North Moyu Rd. 700, Jiangding District, Shanghai 201805, China
| | - Huan Zhao
- Department of General Surgery, Affiliated to the Third Hospital, Naval Medical University, North Moyu Rd. 700, Jiangding District, Shanghai 201805, China
| | - Jianmin Qin
- Department of General Surgery, Affiliated to the Third Hospital, Naval Medical University, North Moyu Rd. 700, Jiangding District, Shanghai 201805, China
| |
Collapse
|
23
|
Yang H, Liu Y, Zhao MM, Guo Q, Zheng XK, Liu D, Zeng KW, Tu PF. Therapeutic potential of targeting membrane-spanning proteoglycan SDC4 in hepatocellular carcinoma. Cell Death Dis 2021; 12:492. [PMID: 33990545 PMCID: PMC8121893 DOI: 10.1038/s41419-021-03780-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022]
Abstract
Syndecan-4 (SDC4) functions as a major endogenous membrane-associated receptor and widely regulates cytoskeleton, cell adhesion, and cell migration in human tumorigenesis and development, which represents a charming anti-cancer therapeutic target. Here, SDC4 was identified as a direct cellular target of small-molecule bufalin with anti-hepatocellular carcinoma (HCC) activity. Mechanism studies revealed that bufalin directly bond to SDC4 and selectively increased SDC4 interaction with substrate protein DEAD-box helicase 23 (DDX23) to induce HCC genomic instability. Meanwhile, pharmacological promotion of SDC4/DDX23 complex formation also inactivated matrix metalloproteinases (MMPs) and augmented p38/JNK MAPKs phosphorylation, which are highly associated with HCC proliferation and migration. Notably, specific knockdown of SDC4 or DDX23 markedly abolished bufalin-dependent inhibition of HCC proliferation and migration, indicating SDC4/DDX23 signaling axis is highly involved in the HCC process. Our results indicate that membrane-spanning proteoglycan SDC4 is a promising druggable target for HCC, and pharmacological regulation of SDC4/DDX23 signaling axis with small-molecule holds great potential to benefit HCC patients.
Collapse
Affiliation(s)
- Heng Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yang Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Mei-Mei Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Qiang Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xi-Kang Zheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Dan Liu
- Proteomics Laboratory, Medical and Healthy Analytical Center, Peking University Health Science Center, Beijing, 100191, China
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Peng-Fei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
24
|
Li FJ, Hu JH, Ren X, Zhou CM, Liu Q, Zhang YQ. Toad venom: A comprehensive review of chemical constituents, anticancer activities, and mechanisms. Arch Pharm (Weinheim) 2021; 354:e2100060. [PMID: 33887066 DOI: 10.1002/ardp.202100060] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 12/31/2022]
Abstract
Toad venom, a traditional natural medicine, has been used for hundreds of years in China for treating different diseases. Many studies have been performed to elucidate the cardiotonic and analgesic activities of toad venom. Until the last decade, an increasing number of studies have documented that toad venom is a source of lead compound(s) for the development of potential cancer treatment drugs. Research has shown that toad venom contains 96 types of bufadienolide monomers and 23 types of indole alkaloids, such as bufalin, cinobufagin, arenobufagin, and resibufogenin, which exhibit a wide range of anticancer activities in vitro and, in particular, in vivo for a range of cancers. The main antitumor mechanisms are likely to be apoptosis or/and autophagy induction, cell cycle arrest, cell metastasis suppression, reversal of drug resistance, or growth inhibition of cancer cells. This review summarizes the chemical constituents of toad venom, analyzing their anticancer activities and molecular mechanisms for cancer treatments. We also outline the importance of further studies regarding the material basis and anticancer mechanisms of toad venom.
Collapse
Affiliation(s)
- Fang-Jie Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing-Hong Hu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Provincial Collaborative Innovation Center for Quality Control and Construction of the Whole Industrial Chain of Traditional Chinese Medicine, Jinan, China
| | - Xin Ren
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cheng-Mei Zhou
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qian Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Provincial Collaborative Innovation Center for Quality Control and Construction of the Whole Industrial Chain of Traditional Chinese Medicine, Jinan, China
| | - Yong-Qing Zhang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.,Shandong Provincial Collaborative Innovation Center for Quality Control and Construction of the Whole Industrial Chain of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
25
|
Li H, Zhou L, Zhou J, Li Q, Ji Q. Underlying mechanisms and drug intervention strategies for the tumour microenvironment. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:97. [PMID: 33722297 PMCID: PMC7962349 DOI: 10.1186/s13046-021-01893-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/24/2021] [Indexed: 02/08/2023]
Abstract
Cancer occurs in a complex tissue environment, and its progression depends largely on the tumour microenvironment (TME). The TME has a highly complex and comprehensive system accompanied by dynamic changes and special biological characteristics, such as hypoxia, nutrient deficiency, inflammation, immunosuppression and cytokine production. In addition, a large number of cancer-associated biomolecules and signalling pathways are involved in the above bioprocesses. This paper reviews our understanding of the TME and describes its biological and molecular characterization in different stages of cancer development. Furthermore, we discuss in detail the intervention strategies for the critical points of the TME, including chemotherapy, targeted therapy, immunotherapy, natural products from traditional Chinese medicine, combined drug therapy, etc., providing a scientific basis for cancer therapy from the perspective of key molecular targets in the TME.
Collapse
Affiliation(s)
- Haoze Li
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lihong Zhou
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jing Zhou
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qi Li
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China. .,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Qing Ji
- Department of Medical Oncology and Cancer Institute, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China. .,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
26
|
Zhang X, Zhao X, Liu K, Che Y, Qiu X, Qu Y, Sun X, Song J. Bufalin: A Systematic Review of Research Hotspots and Antitumor Mechanisms by Text Mining and Bioinformatics. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:1633-1650. [PMID: 33148004 DOI: 10.1142/s0192415x20500810] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Bufalin is an anticancer drug extract from traditional Chinese medicine. Several articles about bufalin have been published. However, the literature on bufalin has not yet been systematically studied. This study aimed to identify the study status and knowledge structures of bufalin and to summarize the antitumor mechanism. Data were retrieved and downloaded from the PubMed database. The softwares of BICOMB, gCLUTO, Ucinet 6.0, and NetDraw2.084 were used to analyze these publications. The bufalin related genes were recognized and tagged by ABNER software. Then these BF-related genes were performed by Gene Ontology (GO) enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways analysis, and protein-protein interaction (PPI) network analysis. A total of 474 papers met the search criteria from 2000 to 2019. By biclustering clustering analysis, the 50 high-frequency main MeSH terms/subheadings were classified into 5 clusters. The clusters of drug therapy and the mechanism of bufalin were hotspot topics. A total of 50 genes were identified as BF-related genes. PPI network analysis showed that inducing apoptosis was the main effect of bufalin, and apoptosis-related gene Caspase 3 was the most reported by people. Bufalin could inhibit the proliferation, invasion, and metastasis of cancer cells through multiple signaling pathways, such as PI3K/AKT, Hedgehog, MAPK/JNK, Wnt/[Formula: see text]-catenin, TGF-[Formula: see text]/Smad, Integrin signaling pathway, and NF-KB signaling pathway via KEGG analysis. Through the quantitative analysis of bufalin literature, we revealed the research status and hot spots in this field and provided some guidance for further research.
Collapse
Affiliation(s)
- Xian Zhang
- Lymphoma and Myeloma Diagnosis and Treatment Center, The Second Affiliated Hospital of Dalian Medical University Dalian, Liaoning 116023, P. R. China
| | - Xiaoxuan Zhao
- Department of Gastrointestinal Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P. R. China.,Department of Dermatology, Dalian Dermatology Hospital, Dalian, Liaoning 116023, P. R. China
| | - Kaili Liu
- Lymphoma and Myeloma Diagnosis and Treatment Center, The Second Affiliated Hospital of Dalian Medical University Dalian, Liaoning 116023, P. R. China
| | - Yuxuan Che
- Lymphoma and Myeloma Diagnosis and Treatment Center, The Second Affiliated Hospital of Dalian Medical University Dalian, Liaoning 116023, P. R. China
| | - Xun Qiu
- Lymphoma and Myeloma Diagnosis and Treatment Center, The Second Affiliated Hospital of Dalian Medical University Dalian, Liaoning 116023, P. R. China
| | - Yanjun Qu
- Department of Gastrointestinal Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P. R. China
| | - Xiuhua Sun
- Lymphoma and Myeloma Diagnosis and Treatment Center, The Second Affiliated Hospital of Dalian Medical University Dalian, Liaoning 116023, P. R. China
| | - Jincheng Song
- Department of Gastrointestinal Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P. R. China
| |
Collapse
|
27
|
Ke B, Wang XN, Liu N, Li B, Wang XJ, Zhang RP, Liang H. Sonic Hedgehog/Gli1 Signaling Pathway Regulates Cell Migration and Invasion via Induction of Epithelial-to-mesenchymal Transition in Gastric Cancer. J Cancer 2020; 11:3932-3943. [PMID: 32328197 PMCID: PMC7171499 DOI: 10.7150/jca.42900] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/23/2020] [Indexed: 02/06/2023] Open
Abstract
Background: The aberrant activation of the Sonic hedgehog (Shh) signaling pathway is involved in progression of several types of cancer, including gastric cancer (GC). However, it remains uncertain whether it also plays a critical role in promoting cancer initiation and progression by inducing epithelial-to-mesenchymal transition (EMT) in GC. Thus, the aim of the present study was to determine whether the Shh pathway is involved in GC, and to investigate the function of the Shh pathway in the induction of EMT in GC. Materials and methods: The expression levels of Shh pathway members and EMT markers were examined in GC tissues by immunohistochemistry. The association between these factors and patient clinicopathological characteristics was analyzed. In addition, Gli-antagonist 61 (GANT61) was used to block Shh/Gli1 pathway activity, and recombinant Shh proteins (N-Shh) were used to activate the Shh pathway in GC cells. Wound healing and Transwell invasion and migration assays were performed to assess the effects of the Shh pathway on the migration and invasion of GC cells in vitro. Furthermore, western blot analysis was used to examine the changes in protein expression. Results: The results demonstrated that these Shh/Gli1 pathway members were upregulated in GC tissues, and that Gli1 upregulation was associated with tumor progression and a poor prognosis. Gli1 expression was negatively associated with E-cadherin (E-Cad) expression, and positively with Vimentin (VIM) expression in GC specimens. Further analysis revealed that when the Shh/Gli1 pathway was activated, the migratory and invasive abilities of GC cells were enhanced, and the expression levels of Gli1 and VIM were increased, while E-Cad expression was decreased. Opposite results were observed when the Shh/Gli1 pathway was blocked by GANT61. Conclusions: The present study indicated that the Shh/Gli1 pathway exhibits an abnormal activation pattern in GC with possible predictive and prognostic significance. The Shh/Gli1 pathway may promote the migratory and invasive potential of GC cells by inducing EMT. The Shh/Gli1 pathway can thus be considered as a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Bin Ke
- Department of Gastric Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin, 300060, P.R. China
| | - Xiao-Na Wang
- Department of Gastric Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin, 300060, P.R. China
| | - Ning Liu
- Department of Gastric Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin, 300060, P.R. China
| | - Bin Li
- Department of Gastric Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin, 300060, P.R. China
| | - Xue-Jun Wang
- Department of Gastric Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin, 300060, P.R. China
| | - Ru-Peng Zhang
- Department of Gastric Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin, 300060, P.R. China
| | - Han Liang
- Department of Gastric Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin, 300060, P.R. China
| |
Collapse
|
28
|
Qian L, Su H, Wang G, Li B, Shen G, Gao Q. Anti-tumor Activity of Bufalin by Inhibiting c-MET Mediated MEK/ERK and PI3K/AKT Signaling Pathways in Gallbladder Cancer. J Cancer 2020; 11:3114-3123. [PMID: 32231716 PMCID: PMC7097950 DOI: 10.7150/jca.38393] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 02/06/2020] [Indexed: 12/30/2022] Open
Abstract
Gallbladder cancer is one of the most common malignant tumors in the biliary tract. In recent years, the chemotherapy treatment for gallbladder carcinoma has exhibited obvious characteristics of drug resistance and insensitivity, and one of the main factors is the existence of cancer stem cells. Here in this study, the effect of Bufalin on gallbladder cancer (GBC-SD) cells and the related mechanism were studied. The results indicated that Bufalin could inhibit the growth of gallbladder carcinoma both in vivo and in vitro. According to the biological behavior analysis, Bufalin induced apoptosis, inhibited the propagation, migration and invasion of GBC-SD cells, and blocked cell cycle at the G2/M stage. Besides, Bufalin inhibited the tumor sphere formation capability of gallbladder carcinoma in matrigel, reduced the expression of multiple stemness-associated proteins, including Oct4, Sox2 and the stem cell-surface marker proteins CD133 and CD44. Western blot assay showed that Bufalin inhibited MEK/ERK and PI3-K/AKT signaling pathways by inhibiting the expression of p-c-Met, which in turn affected the expression of apoptosis-related protein Mcl-1, and the invasion-associated proteins E-cadherin, MMP9 and Snail. Bufalin was found to have an inhibitory effect on the GBC-SD cell growth and reduce the self-renewal and characteristic of gallbladder cancer stem cells. It enhanced the chemotherapeutic sensitivity and reduced the metastasis of gallbladder carcinoma. In conclusion, Bufalin can be used as a new promising anticancer drug for gallbladder cancer patients who are resistant to traditional chemotherapy.
Collapse
Affiliation(s)
- Liqiang Qian
- Department of General Surgery, Suzhou Ninth People's Hospital, Suzhou, China
| | - Haoyuan Su
- Department of General Surgery, Suzhou Ninth People's Hospital, Suzhou, China
| | - Gang Wang
- Department of General Surgery, Suzhou Ninth People's Hospital, Suzhou, China
| | - Bin Li
- Department of General Surgery, Suzhou Ninth People's Hospital, Suzhou, China
| | - Genhai Shen
- Department of General Surgery, Suzhou Ninth People's Hospital, Suzhou, China
| | - Quangen Gao
- Department of General Surgery, Suzhou Ninth People's Hospital, Suzhou, China
| |
Collapse
|
29
|
Dimri M, Satyanarayana A. Molecular Signaling Pathways and Therapeutic Targets in Hepatocellular Carcinoma. Cancers (Basel) 2020; 12:cancers12020491. [PMID: 32093152 PMCID: PMC7072513 DOI: 10.3390/cancers12020491] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/14/2020] [Accepted: 02/18/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a complex biological process and is often diagnosed at advanced stages with no effective treatment options. With advances in tumor biology and molecular genetic profiling, several different signaling pathways and molecular mechanisms have been identified as responsible for initiating and promoting HCC. Targeting these critical pathways, which include the receptor tyrosine kinase pathways, the Ras mitogen-activated protein kinase (Ras/Raf/MAPK), the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR), the Wnt/β-catenin signaling pathway, the ubiquitin/proteasome degradation and the hedgehog signaling pathway has led to the identification of novel therapeutics for HCC treatment. In this review, we elaborated on our current understanding of the signaling pathways involved in the development and initiation of HCC and anticipate the potential targets for therapeutic drug development.
Collapse
|
30
|
Han M, Yang G, Lin Q, Yang Y, Zhang H, Su Y. Determination of Endogenous Bufalin in Serum of Patients With Hepatocellular Carcinoma Based on HPLC-MS/MS. Front Oncol 2020; 9:1572. [PMID: 32039033 PMCID: PMC6989541 DOI: 10.3389/fonc.2019.01572] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/30/2019] [Indexed: 12/15/2022] Open
Abstract
Bufalin is a cardiotonic steroid and a key active ingredient of the Chinese medicine ChanSu. It has significant anti-tumor activity against many malignancies, including hepatocellular carcinoma (HCC). Previous studies have shown that human bodies contain an endogenous bufalin-like substance. This study aimed to confirm whether the endogenous bufalin-like substances is bufalin and further detect the differences between HCC and control groups of endogenous bufalin concentration by the high-performance liquid chromatography coupled tandem mass spectrometry (HPLC-MS/MS). The results confirmed the endogenous bufalin-like substance is bufalin. Totally, 227 serum samples were collected: 54 from HCC patients and 173 from healthy volunteers constituting a control group. Both the test group and the control group contained bufalin in serum, revealing that bufalin is indeed an endogenous substance. The bufalin concentration was 1.3 nM in HCC patients and 5.7 nM in normal people (P < 0.0001). These results indicate that human bodies contain endogenous bufalin, and it may be negatively correlated with the incidence of HCC.
Collapse
Affiliation(s)
- Mengfei Han
- Department of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Geliang Yang
- Department of Traditional Chinese Medicine and Acupuncture, The Second Medical Centre, Chinese People Liberation Army General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Qing Lin
- Fuzhou Traditional Chinese Hospital, Fuzhou, China
| | - Yanlong Yang
- Department of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Huiqing Zhang
- Department of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Yonghua Su
- Department of Traditional Chinese Medicine, Changhai Hospital, Naval Medical University, Shanghai, China.,Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
31
|
Zhan X, Wu H, Wu H, Wang R, Luo C, Gao B, Chen Z, Li Q. Metabolites from Bufo gargarizans (Cantor, 1842): A review of traditional uses, pharmacological activity, toxicity and quality control. JOURNAL OF ETHNOPHARMACOLOGY 2020; 246:112178. [PMID: 31445132 DOI: 10.1016/j.jep.2019.112178] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 08/17/2019] [Accepted: 08/21/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bufo gargarizans (Cantor, 1842) (BGC), a traditional medicinal animal distributed in many provinces of China, is well known for the pharmaceutical value of Chansu and Chanpi. As traditional Chinese medicines (TCMs), Chansu and Chanpi, with their broad-spectrum of therapeutic applications, have long been applied to detoxification, anti-inflammation, analgesia, etc. OVERARCHING OBJECTIVE: We critically analyzed the current evidence for the traditional uses, chemical profiles, pharmacological activity, toxicity and quality control of BGC (Bufonidae family) to provide a scientific basis for future in-depth studies and perspectives for the discovery of potential drug candidates. METHODOLOGY All of the available information on active constituents and TCMs derived from BGC was obtained using the keywords "Bufo gargarizans", "Chansu", "Chanpi", "Huachansu", or "Cinobufacini" through different electronic databases, including PubMed, Web of Science, Chinese National Knowledge Infrastructure (CNKI), the Wanfang Database, and Pharmacopoeia of China. In addition, Chinese medicine books from different times were used to elucidate the traditional uses of BGC. Electronic databases, including the "IUCN Red List of Threatened Species", "American Museum of Natural History" and "AmphibiaWeb Species Lists", were used to validate the scientific name of BGC. RESULTS To date, about 118 bufadienolide monomers and 11 indole alkaloids have been identified from BGC in total. The extracts and isolated compounds exhibit a wide range of in vitro and in vivo pharmacological effects. The literature search demonstrated that the ethnomedicinal uses of BGC, such as detoxification, anti-inflammation and the ability to reduce swelling and pain associated with infections, are correlated with its modern pharmacological activities, including antitumor, immunomodulation and attenuation of cancer-derived pain. Bufadienolides and indole alkaloids have been regarded as the main active substances in BGC, among which bufadienolides have significant antitumor activity. Furthermore, the cardiotoxicity of bufadienolides was discussed, and the main molecular mechanism involves in the inhibition of Na+/K+-ATPase. Besides, with the development of modern analytical techniques, the quality control methods of BGC-derived TCMs are being improved constantly. CONCLUSIONS An increasing number of reports suggest that BGC can be regarded as an excellent source for exploring the potential antitumor constituents. However, the future antitumor research of BGC needs to follow the standard pharmacology guidelines, so as to provide comprehensive pharmacological information and aid the reproducibility of the data. Besides, to ensure the efficacy and safety of BGC-derived TCMs, it is vital to construct a comprehensive quality evaluation model on the basis of clarifying pharmacodynamic-related and toxicity-related compositions.
Collapse
Affiliation(s)
- Xiang Zhan
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230038, China; Scientific Research & Experiment Center, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Huan Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230038, China; Scientific Research & Experiment Center, Anhui University of Chinese Medicine, Hefei, 230038, China; Anhui China Resources Jin Chan Pharmaceutical Co., Ltd., Huaibei, 235000, China.
| | - Hong Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Rong Wang
- Anhui China Resources Jin Chan Pharmaceutical Co., Ltd., Huaibei, 235000, China
| | - Chuan Luo
- Anhui China Resources Jin Chan Pharmaceutical Co., Ltd., Huaibei, 235000, China
| | - Bo Gao
- Anhui China Resources Jin Chan Pharmaceutical Co., Ltd., Huaibei, 235000, China
| | - Zhiwu Chen
- Basic Medical College, Anhui Medical University, Hefei, 230032, China
| | - Qinglin Li
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui Province Key Laboratory of R&D of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230038, China; Scientific Research & Experiment Center, Anhui University of Chinese Medicine, Hefei, 230038, China; Anhui China Resources Jin Chan Pharmaceutical Co., Ltd., Huaibei, 235000, China.
| |
Collapse
|
32
|
Si A, Wang L, Miao K, Zhang R, Ji H, Lei Z, Cheng Z, Fang X, Hao B. miR-219 regulates liver cancer stem cell expansion via E-cadherin pathway. Cell Cycle 2019; 18:3550-3561. [PMID: 31724462 PMCID: PMC6927721 DOI: 10.1080/15384101.2019.1691762] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 10/03/2019] [Accepted: 10/06/2019] [Indexed: 02/06/2023] Open
Abstract
Liver cancer stem cells contribute to tumorigenesis, progression, recurrence and drug resistance of hepatocellular carcinoma (HCC). However, the underlying mechanism for the propagation of liverCSCs is not fully understood yet. Here we show that miR-219 is upregulated in liver CSCs. Knockdown of miR-219 attenuates the self-renewal and tumorigenicity of liver CSCs. Conversely, miR-219 overexpressing enhances the self-renewal and tumorigenicity of liver CSCs.Mechanistically,miR-219 downregulates E-cadherin via itsmRNA 3'UTR in liver CSCs. The correlation between miR-219 and E-cadherin is validated in human HCC tissues. Furthermore, the miR-219 expression determines the responses of hepatoma cells to sorafenib treatment. Our findings indicate that miR-219 plays a critical role in liver CSCs expansion and sorafenib response, rendering miR-219 as an optimal target for the prevention and intervention of HCC.Abbreviations: HCC: Hepatocellular carcinoma; CSCs: cancer stem cells; DMEM: Dulbecco's modified Eagle's medium; FBS: fetal bovine serum; OS: overall survival.
Collapse
Affiliation(s)
- Anfeng Si
- Department of Surgical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Longqi Wang
- Department of General Surgery I, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Kun Miao
- Oncology Department Ward, Tianchang People’s Hospital, Anhui, China
| | - Rongrong Zhang
- Department of General Surgery III, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Huiyu Ji
- Department of Surgical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Zhengqing Lei
- Department of General Surgery, the Affiliated Zhongda Hospital, Southeast University, Nanjing, China
| | - Zhangjun Cheng
- Department of General Surgery, the Affiliated Zhongda Hospital, Southeast University, Nanjing, China
| | - Xiangchun Fang
- Department of Surgical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Baobing Hao
- Department of Surgical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
33
|
Ueno M, Kawai M, Park JY, Hirono S, Okada KI, Hayami S, Wang HJ, Yamaue H. Prognostic Analysis of Hepatocellular Carcinoma With Hepatitis C Virus Infection Using Epithelial-Mesenchymal Transition Gene Profiles. J Surg Res 2019; 245:302-308. [PMID: 31421377 DOI: 10.1016/j.jss.2019.07.077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/24/2019] [Accepted: 07/19/2019] [Indexed: 01/18/2023]
Abstract
BACKGROUND Epithelial-mesenchymal transition genes have prognostic influence on hepatocellular carcinoma (HCC). Previously, the following four epithelial-mesenchymal transition-related genes were considered to be significantly influential: E-cadherin (CDH1), inhibitor of DNA binding 2 (ID2), matrix metalloproteinase 9 (MMP9), and transcription factor 3 (TCF3). A prognostic prediction model, NRISK4 = (-0.333 × [CDH1] - 0.400 × [ID2] + 0.339 × [MMP9] + 0.387 × [TCF3]) was constructed, but from patients with HCC with predominantly hepatitis B virus infection. We therefore aim to validate if this model also fits patients with HCC and hepatitis C virus (HCV) infection. METHODS We collected HCC tissue samples from 67 patients with HCV infection. Discrimination of the NRISK4 was re-estimated using receiver operating curve analysis and we redefined the appropriate cutoff value. Using this cutoff value, patients were divided into two groups (high/low risk patients) and we compared their clinicopathological factors and prognosis. RESULTS Area under the curve of NRISK4 prediction was 0.70 and an appropriate cutoff value was 3.19 in this cohort. Patients were divided into high- (n = 25) and low-risk (n = 42) patients for prognosis. There were no significant differences in tumor factors between the two groups. Cancer-specific survival rates at 5 y after surgery on high- and low-risk patients were 45% and 68%, respectively (P = 0.02). At 2 y after surgery, recurrence rates were 68% and 37% among high- and low-risk patients, respectively (P = 0.01). Aggressive recurrences were highly observed in the high-risk patients (P = 0.01). CONCLUSIONS NRISK4 model could also successfully validate prognosis of patients with HCC with HCV infection similarly to in the previous report of patients with hepatitis B virus infection, especially in the early period after surgery.
Collapse
Affiliation(s)
- Masaki Ueno
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Manabu Kawai
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | | | - Seiko Hirono
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Ken-Ichi Okada
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Shinya Hayami
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan
| | - Hee-Jung Wang
- Department of Surgery, Ajou University School of Medicine, Suwon, Korea
| | - Hiroki Yamaue
- Second Department of Surgery, Wakayama Medical University, Wakayama, Japan.
| |
Collapse
|
34
|
An W, Lai H, Zhang Y, Liu M, Lin X, Cao S. Apoptotic Pathway as the Therapeutic Target for Anticancer Traditional Chinese Medicines. Front Pharmacol 2019; 10:758. [PMID: 31354479 PMCID: PMC6639427 DOI: 10.3389/fphar.2019.00758] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/11/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer is a leading cause of morbidity and mortality worldwide. Apoptosis is a process of programmed cell death and it plays a vital role in human development and tissue homeostasis. Mounting evidence indicates that apoptosis is closely related to the survival of cancer and it has emerged as a key target for the discovery and development of novel anticancer drugs. Various studies indicate that targeting the apoptotic signaling pathway by anticancer drugs is an important mechanism in cancer therapy. Therefore, numerous novel anticancer agents have been discovered and developed from traditional Chinese medicines (TCMs) by targeting the cellular apoptotic pathway of cancer cells and shown clinically beneficial effects in cancer therapy. This review aims to provide a comprehensive discussion for the role, pharmacology, related biology, and possible mechanism(s) of a number of important anticancer TCMs and their derivatives mainly targeting the cellular apoptotic pathway. It may have important clinical implications in cancer therapy.
Collapse
Affiliation(s)
- Weixiao An
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Pharmacy, Nanchong Central Hospital, Nanchong, China
| | - Honglin Lai
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,Department of Pharmacy, Affliated Hospital of Traditional Chinese Medicine, Southwest Medical University, Luzhou, China
| | - Yangyang Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Minghua Liu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiukun Lin
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Shousong Cao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
35
|
Cheng CS, Wang J, Chen J, Kuo KT, Tang J, Gao H, Chen L, Chen Z, Meng Z. New therapeutic aspects of steroidal cardiac glycosides: the anticancer properties of Huachansu and its main active constituent Bufalin. Cancer Cell Int 2019; 19:92. [PMID: 31011289 PMCID: PMC6458819 DOI: 10.1186/s12935-019-0806-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/27/2019] [Indexed: 12/20/2022] Open
Abstract
Aim of the review In the past decade, increasing research attention investigated the novel therapeutic potential of steroidal cardiac glycosides in cancer treatment. Huachansu and its main active constituent Bufalin have been studied in vitro, in vivo and clinical studies. This review aims to summarize the multi-target and multi-pathway pharmacological effects of Bufalin and Huachansu in the last decade, with the aim of providing a more comprehensive view and highlighting the recently discovered molecular mechanisms. Results Huachansu and its major derivative, Bufalin, had been found to possess anti-cancer effects in a variety of cancer cell lines both in vitro and in vivo. The underlying anti-cancer molecular mechanisms mainly involved anti-proliferation, apoptosis induction, anti-metastasis, anti-angiogenesis, epithelial-mesenchymal transition inhibition, anti-inflammation, Na+/K+-ATPase activity targeting, the steroid receptor coactivator family inhibitions, etc. Moreover, the potential side-effects and toxicities of the toad extract, Huachansu, and Bufalin, including hematological, gastrointestinal, mucocutaneous and cardiovascular adverse reactions, were reported in animal studies and clinic trails. Conclusions Further research is needed to elucidate the potential drug-drug interactions and multi-target interaction of Bufalin and Huachansu. Large-scale clinical trials are warranted to translate the knowledge of the anticancer actions of Bufalin and Huachansu into clinical applications as effective and safe treatment options for cancer patients in the future.
Collapse
Affiliation(s)
- Chien-Shan Cheng
- 1Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,3School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Jiaqiang Wang
- 2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China.,Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433 China.,5Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China
| | - Jie Chen
- 3School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China.,6Department of Orthopaedics, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Kuei Ting Kuo
- 3School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR China
| | - Jian Tang
- 1Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Huifeng Gao
- 1Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Lianyu Chen
- 1Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Zhen Chen
- 1Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Zhiqiang Meng
- 1Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032 China.,2Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| |
Collapse
|
36
|
Wu D, Zhou WY, Lin XT, Fang L, Xie CM. Bufalin induces apoptosis via mitochondrial ROS-mediated caspase-3 activation in HCT-116 and SW620 human colon cancer cells. Drug Chem Toxicol 2019; 42:444-450. [PMID: 30777466 DOI: 10.1080/01480545.2018.1512611] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Bufalin has been reported to kill various types of cancer including human colorectal cancer. Our previous study demonstrated that bufalin induced cell death via autophagy in HT-29 and Caco-2 colon cancer cells, but the action of bufalin remains unclear. This study was conducted to investigate the role of bufalin in other colon cancer HCT-116 and SW620 cells as well as its potential mechanism. METHODS The effect of bufalin in HCT-116 and SW620 colon cancer cells was detected by assessing cell viability and cell death. Apoptotic cells were analyzed by Western blot and trypan blue dye exclusion assay. Mitochondrial ROS production was analyzed by flow cytometry after DCFDA and DHR-123 staining. The potential mechanism was investigated via pharmacological inhibitors. RESULTS Bufalin had high potency against HCT-116 and SW620 cells with IC50 values of 12.823 ± 1.792 nM and 26.303 ± 2.498 nM in HCT-116 and SW620 cells, respectively. Bufalin decreased cell viability, increased cell death as well as caspase-3 downstream target (cleaved PARP) accumulation, and these actions were significantly blocked by pan-caspase inhibitor zVAD-FMK. Mechanistically, ROS production, but neither the NAD(P)H oxidase, AMPK, ERK nor p38, is responsible for bufalin-induced apoptotic cell death. Moreover, bufalin-induced ROS generation is derived from mitochondria. CONCLUSION Bufalin significantly induces apoptosis in HCT-116 and SW620 colon cancer cells via mitochondrial ROS-mediated caspase-3 activation. We believe that our novel findings will greatly alter our current understanding on the anti-cancer mechanism of bufalin in colon cancer cells and will pave the way for further exploiting the clinical application.
Collapse
Affiliation(s)
- Di Wu
- a Institute of Hepatobiliary Surgery, Southwest Hospital , Third Military Medical University (Army Medical University) , Chongqing , China
| | - Wen-Yi Zhou
- b Essence Securities Co., Ltd. , Chongqing , China
| | - Xiao-Tong Lin
- a Institute of Hepatobiliary Surgery, Southwest Hospital , Third Military Medical University (Army Medical University) , Chongqing , China
| | - Lei Fang
- a Institute of Hepatobiliary Surgery, Southwest Hospital , Third Military Medical University (Army Medical University) , Chongqing , China
| | - Chuan-Ming Xie
- a Institute of Hepatobiliary Surgery, Southwest Hospital , Third Military Medical University (Army Medical University) , Chongqing , China
| |
Collapse
|
37
|
Huang H, Fu S, Liu D. Detection and Analysis of the Hedgehog Signaling Pathway-Related Long Non-Coding RNA (lncRNA) Expression Profiles in Keloid. Med Sci Monit 2018; 24:9032-9044. [PMID: 30543583 PMCID: PMC6301256 DOI: 10.12659/msm.911159] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/26/2018] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Hedgehog (Hh) signaling pathway-related genes have important roles in several physiological and disease processes that involve cell proliferation. Long non-coding region RNAs (lncRNAs) have a regulatory role on gene expression. Keloid is characterized by excessive proliferation of scar tissue following trauma. The aims of this study were to evaluate the Hh signaling pathway in keloid skin tissues and its downstream gene expression and lncRNAs, compared with normal skin. MATERIAL AND METHODS Four pairs of keloids and adjacent normal skin epidermis underwent total RNA extraction. Gene chip high-throughput real-time quantitative polymerase chain reaction (qPCR) was used to examine the differential expression profiles of the Hh signaling pathway-related lncRNAs and mRNAs in the human keloid and normal skin. The differentially expressed mRNAs were analyzed by Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) to identify their biological roles. RESULTS In keloid tissue, differential expression of 33 mRNAs and 30 lncRNAs relating to the Hh pathway, were verified by gene chip qPCR. The results of GO and KEGG analysis showed that the upregulated mRNAs were involved in cell proliferation, cell growth, and tissue repair, and down-regulated mRNAs were involved in apoptosis. The lncRNA, AC073257.2, affected cell keloid growth and proliferation by its upstream target the GLI2 gene at the transcriptional level. The lncRNA, HNF1A-AS1, affected cell keloid growth and proliferation by its neighboring target gene, HNF1A. CONCLUSIONS Differential expression occurred in Hh signaling pathway-related lncRNAs and mRNAs, which may provide further insight into the development of keloid.
Collapse
Affiliation(s)
- Heping Huang
- Institute of Burns, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
- Department of Plastic and Aesthetic Surgery, Jingxi Maternal and Child Health Hospital, Nanchang, Jiangxi, P.R. China
| | - Shangfeng Fu
- Institute of Burns, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| | - Dewu Liu
- Institute of Burns, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, P.R. China
| |
Collapse
|
38
|
Wang J, Xia Y, Zuo Q, Chen T. Molecular mechanisms underlying the antimetastatic activity of bufalin. Mol Clin Oncol 2018; 8:631-636. [PMID: 29732152 DOI: 10.3892/mco.2018.1591] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/09/2018] [Indexed: 12/14/2022] Open
Abstract
Bufalin is a monomer compound extract from Chansu, which is a traditional Chinese medicine obtained from the skin and parotid venom glands of toads, such as Bufo bufo gargarizans Cantor and Bufo melanostictus Schneider. Chansu had been used in traditional Chinese medicine for >1,000 years due to its cardiac, anti-inflammatory and anticancer properties. Previous studies identified bufalin as the main anticancer compound of Chansu, and recent evidence has corroborated its anticancer properties. Bufalin inhibits cancer cell proliferation, induces cell cycle arrest, induces cancer cell apoptosis, inhibits neovascularization, induces cell differentiation, inhibits cancer metastasis and invasion, and enhances chemotherapeutic drug sensitivity. However, the function and mechanism of bufalin in metastatic cancer cells have not yet been expounded. The aim of the present review was to discuss the recent progress and prospects of bufalin in the prevention of cancer metastasis, particularly in inhibiting epithelial-to-mesenchymal transition.
Collapse
Affiliation(s)
- Jie Wang
- Department of Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Yue Xia
- Department of Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Qingshong Zuo
- Department of Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| | - Teng Chen
- Department of Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, P.R. China
| |
Collapse
|