1
|
Elkotamy MS, Elgohary MK, Alkabbani MA, Binjubair FA, Alanazi MM, Alsulaimany M, Al-Rashood ST, Ghabbour HA, Abdel-Aziz HA. Design, synthesis and biological evaluation of pyrazolo[3,4- b]pyridine derivatives as dual CDK2/PIM1 inhibitors with potent anti-cancer activity and selectivity. J Biomol Struct Dyn 2025:1-25. [PMID: 40079180 DOI: 10.1080/07391102.2025.2475233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/31/2025] [Indexed: 03/14/2025]
Abstract
The discovery of novel, selective inhibitors targeting CDK2 and PIM1 kinases, which regulate cell survival, proliferation, and treatment resistance, is crucial for advancing cancer therapy. This study reports the design, synthesis, and biological evaluation of three novel pyrazolo[3,4-b]pyridine derivatives (6a-c), confirmed via spectral analyses. These compounds were assessed for anti-cancer activity against breast, colon, liver, and cervical cancers using the MTT assay. Among the tested compounds, 6b exhibited superior efficacy, with higher selectivity indices for HCT-116 (15.05) and HepG2 (9.88) compared to the reference drug staurosporine. Mechanistic studies revealed that 6b induced apoptosis (63.04-fold increase) and arrested the cell cycle at the G0-G1 phase, highlighting its anti-proliferative effects. In an in-vivo solid Ehrlich carcinoma (SEC) mouse model, compound 6b significantly reduced tumor weight and volume, exceeding the efficacy of doxorubicin. Additionally, 6b potently inhibited CDK2 and PIM1 kinases (IC50: 0.27 and 0.67 µM, respectively) and reduced tumor-promoting TNF-alpha expression, as confirmed by histopathological and immunohistochemical studies. Computational analyses, including molecular docking, molecular dynamics simulations, and DFT calculations, provided insights into the binding stability and interaction mechanisms of 6b with CDK2 and PIM1, while in-silico pharmacokinetic and toxicity evaluations confirmed its favorable drug-like profile and safety. This study highlights compound 6b as a promising dual CDK2/PIM1 inhibitor with potent anti-cancer activity and selectivity, paving the way for its further optimization and development as a lead molecule in cancer therapy.
Collapse
Affiliation(s)
- Mahmoud S Elkotamy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Egypt
| | - Mohamed K Elgohary
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Egypt
| | | | - Faizah A Binjubair
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Manal M Alanazi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Marwa Alsulaimany
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Sara T Al-Rashood
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hazem A Ghabbour
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
| | - Hatem A Abdel-Aziz
- Applied Organic Chemistry Department, National Research Center, Dokki, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| |
Collapse
|
2
|
Yang X, Liu C, Lei Y, Liu Z, Zhu B, Zhao D. PIM1 signaling in immunoinflammatory diseases: an emerging therapeutic target. Front Immunol 2024; 15:1443784. [PMID: 39372407 PMCID: PMC11449710 DOI: 10.3389/fimmu.2024.1443784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024] Open
Abstract
PIM1, the proviral integration site for Moloney murine leukemia virus, is a member of the serine/threonine protein kinase family. It is involved in many biological events, such as cell survival, cell cycle progression, cell proliferation, and cell migration, and has been widely studied in malignant diseases. However, recent studies have shown that PIM1 plays a prominent role in immunoinflammatory diseases, including autoimmune uveitis, inflammatory bowel disease, asthma, and rheumatoid arthritis. PIM1 can function in inflammatory signal transduction by phosphorylating multiple inflammatory protein substrates and mediating macrophage activation and T lymphocyte cell specification, thus participating in the development of multiple immunoinflammatory diseases. Moreover, the inhibition of PIM1 has been demonstrated to ameliorate certain immunoinflammatory disorders. Based on these studies, we suggest PIM1 as a potential therapeutic target for immunoinflammatory diseases and a valid candidate for future research. Herein, for the first time, we provide a detailed review that focuses on the roles of PIM1 in the pathogenesis of immunoinflammatory diseases.
Collapse
Affiliation(s)
- Xue Yang
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chunming Liu
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yuxi Lei
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhi Liu
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Bin Zhu
- Department of Pediatrics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Dongchi Zhao
- Department of Pediatrics, Children’s Digital Health and Data Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
3
|
Rong J, Wang Q, Li T, Qian J, Cheng J. Glucose metabolism in glioma: an emerging sight with ncRNAs. Cancer Cell Int 2024; 24:316. [PMID: 39272133 PMCID: PMC11395608 DOI: 10.1186/s12935-024-03499-8] [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: 03/05/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Glioma is a primary brain tumor that grows quickly, has an unfavorable prognosis, and can spread intracerebrally. Glioma cells rely on glucose as the major energy source, and glycolysis plays a critical role in tumorigenesis and progression. Substrate utilization shifts throughout glioma progression to facilitate energy generation and biomass accumulation. This metabolic reprogramming promotes glioma cell proliferation and metastasis and ultimately decreases the efficacy of conventional treatments. Non-coding RNAs (ncRNAs) are involved in several glucose metabolism pathways during tumor initiation and progression. These RNAs influence cell viability and glucose metabolism by modulating the expression of key genes of the glycolytic pathway. They can directly or indirectly affect glycolysis in glioma cells by influencing the transcription and post-transcriptional regulation of oncogenes and suppressor genes. In this review, we discussed the role of ncRNAs in the metabolic reprogramming of glioma cells and tumor microenvironments and their abnormal expression in the glucometabolic pathway in glioma. In addition, we consolidated the existing theoretical knowledge to facilitate the use of this emerging class of biomarkers as biological indicators and potential therapeutic targets for glioma.
Collapse
Affiliation(s)
- Jun Rong
- Department of Neurosurgery, Xuancheng People's Hospital, The Affiliated Xuancheng Hospital of Wannan Medical College, Xuancheng, People's Republic of China
| | - Qifu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital), WuHu, People's Republic of China
| | - Tingzheng Li
- Department of Neurosurgery, Xuancheng Central Hospital, Xuancheng, People's Republic of China
| | - Jin Qian
- Department of Neurosurgery, Xuancheng People's Hospital, The Affiliated Xuancheng Hospital of Wannan Medical College, Xuancheng, People's Republic of China.
| | - Jinchao Cheng
- Department of Neurosurgery, Xuancheng Central Hospital, Xuancheng, People's Republic of China.
| |
Collapse
|
4
|
Zhao R, Yin F, Fredimoses M, Zhao J, Fu X, Xu B, Liang M, Chen H, Liu K, Lei M, Laster KV, Li Z, Kundu JK, Dong Z, Lee MH. Targeting FGFR1 by β,β-dimethylacrylalkannin suppresses the proliferation of colorectal cancer in cellular and xenograft models. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155612. [PMID: 38669968 DOI: 10.1016/j.phymed.2024.155612] [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: 01/08/2024] [Revised: 03/05/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) continues to be a major global health challenge, ranking as a top cause of cancer-related mortality. Alarmingly, the five-year survival rate for CRC patients hovers around a mere 10-30 %. The disruption of fibroblast growth factor receptor (FGFRs) signaling pathways is significantly implicated in the onset and advancement of CRC, presenting a promising target for therapeutic intervention in CRC management. Further investigation is essential to comprehensively elucidate FGFR1's function in CRC and to create potent therapies that specifically target FGFR1. PURPOSE This study aims to demonstrate the oncogenic role of FGFR1 in colorectal cancer and to explore the potential of β,β-dimethylacrylalkannin (β,β-DMAA) as a therapeutic option to inhibit FGFR1. METHODS In this research, we employed a comprehensive suite of techniques including tissue array, kinase profiling, computational docking, knockdown assay to predict and explore the inhibitor of FGFR1. Furthermore, we utilized kinase assay, pull-down, cell proliferation tests, and Patient derived xenograft (PDX) mouse models to further investigate a novel FGFR1 inhibitor and its impact on the growth of CRC. RESULTS In our research, we discovered that FGFR1 protein is markedly upregulated in colorectal cancer tissues, suggesting a significant role in regulating cellular proliferation, particularly in patients with colorectal cancer. Furthermore, we conducted a computational docking, kinase profiling analysis, simulation and identified that β,β-DMAA could directly bind with FGFR1 within ATP binding pocket domain. Cell-based assays confirmed that β,β-DMAA effectively inhibited the proliferation of colon cancer cells and also triggered cell cycle arrest, apoptosis, and altered FGFR1-mediated signaling pathways. Moreover, β,β-DMAA effectively attenuated the development of PDX tumors in mice that were FGFR1-positive, with no notable toxicity observed. In summary, our study highlights the pivotal role of FGFR1 in colorectal cancer, suggesting that inhibiting FGFR1 activity could be a promising strategy for therapeutic intervention. We present strong evidence that targeting FGFR1 with β,β-DMAA is a viable approach for the management of colorectal cancer. Given its low toxicity and high efficacy, β,β-DMAA, as an FGFR1 inhibitor, warrants further investigation in clinical settings for the treatment of FGFR1-positive tumors.
Collapse
Affiliation(s)
- Ran Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000, China
| | - Fanxiang Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; Translational Medical Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | | | - Jianhua Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Xiaorong Fu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Beibei Xu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Mengrui Liang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | - Hanyong Chen
- The Hormel Institute, University of Minnesota, Austin, MN55912, USA
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou 450000, China
| | - Mingjuan Lei
- China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China
| | | | - Zhi Li
- Department of General Surgery, the Affiliated Tumor Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Joydeb Kumar Kundu
- Li Ka Shing Applied Virology Institute, University of Alberta, Edmonton AB T6G 2R3, Canada
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000, China.
| | - Mee-Hyun Lee
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou 450000, China; College of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea.
| |
Collapse
|
5
|
Cui XL, Nie J, Zhu H, Kowitwanich K, Beadell AV, West-Szymanski DC, Zhang Z, Dougherty U, Kwesi A, Deng Z, Li Y, Meng D, Roggin K, Barry T, Owyang R, Fefferman B, Zeng C, Gao L, Zhao CWT, Malina Y, Wei J, Weigert M, Kang W, Goel A, Chiu BCH, Bissonnette M, Zhang W, Chen M, He C. LABS: linear amplification-based bisulfite sequencing for ultrasensitive cancer detection from cell-free DNA. Genome Biol 2024; 25:157. [PMID: 38877540 PMCID: PMC11177480 DOI: 10.1186/s13059-024-03262-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/29/2024] [Indexed: 06/16/2024] Open
Abstract
Methylation-based liquid biopsies show promises in detecting cancer using circulating cell-free DNA; however, current limitations impede clinical application. Most assays necessitate substantial DNA inputs, posing challenges. Additionally, underrepresented tumor DNA fragments may go undetected during exponential amplification steps of traditional sequencing methods. Here, we report linear amplification-based bisulfite sequencing (LABS), enabling linear amplification of bisulfite-treated DNA fragments in a genome-wide, unbiased fashion, detecting cancer abnormalities with sub-nanogram inputs. Applying LABS to 100 patient samples revealed cancer-specific patterns, copy number alterations, and enhanced cancer detection accuracy by identifying tissue-of-origin and immune cell composition.
Collapse
Affiliation(s)
- Xiao-Long Cui
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ji Nie
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Houxiang Zhu
- Department of Medicine, The University of Chicago, Chicago, IL, USA
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA
| | - Krissana Kowitwanich
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Alana V Beadell
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Diana C West-Szymanski
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Zhou Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Akushika Kwesi
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Zifeng Deng
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Yan Li
- Department of Medicine, The University of Chicago, Chicago, IL, USA
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA
| | - Danqing Meng
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Kevin Roggin
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Teresa Barry
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Ryan Owyang
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Ben Fefferman
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Chang Zeng
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lu Gao
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Carolyn W T Zhao
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Yuri Malina
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Jiangbo Wei
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA
| | - Melanie Weigert
- Department of Obstetrics and Gynecology/Section of Gynecologic Oncology, The University of Chicago, Chicago, IL, USA
| | - Wenjun Kang
- Department of Medicine, The University of Chicago, Chicago, IL, USA
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA
| | - Ajay Goel
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Brian C-H Chiu
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Marc Bissonnette
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Wei Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Mengjie Chen
- Department of Medicine, The University of Chicago, Chicago, IL, USA.
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA.
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL, USA.
- Howard Hughes Medical Institute, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
6
|
Jin F, Lin Y, Yuan W, Wu S, Yang M, Ding S, Liu J, Chen Y. Recent advances in c-Met-based dual inhibitors in the treatment of cancers. Eur J Med Chem 2024; 272:116477. [PMID: 38733884 DOI: 10.1016/j.ejmech.2024.116477] [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: 03/25/2024] [Revised: 05/03/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
The cellular-mesenchymal epithelial transition factor (c-Met) is a receptor tyrosine kinase (RTK) located on the 7q31 locus encoding the Met proto-oncogene and plays a critical role in regulating cell proliferation, metastasis, differentiation, and apoptosis through various signaling pathways. However, its aberrant activation and overexpression have been implicated in many human cancers. Therefore, c-Met is a promising target for cancer treatment. However, the anticancer effect of selective single-targeted drugs is limited due to the complexity of the signaling system and the involvement of different proteins and enzymes. After inhibiting one pathway, signal molecules can be transmitted through other pathways, resulting in poor efficacy of single-targeted drug therapy. Dual inhibitors that simultaneously block c-Met and another factor can significantly improve efficacy and overcome some of the shortcomings of single-target inhibitors, including drug resistance. In this review, We introduced c-Met kinase and the synergism between c-Met and other anti-tumor targets, then dual-target inhibitors based on c-Met for the treatment of cancers were summarized and their design concepts and structure-activity relationships (SARs) were discussed elaborately, providing a valuable insight for the further development of novel c-Met-based dual inhibitors.
Collapse
Affiliation(s)
- Fanqi Jin
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China
| | - Yihan Lin
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China
| | - Weidong Yuan
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China
| | - Shuang Wu
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China
| | - Min Yang
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China
| | - Shi Ding
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China; API Engineering Technology Research Center of Liaoning Province, Shenyang, Liaoning, 110036, PR China; Small Molecular Targeted Drug R&D Engineering Research Center of Liaoning Province, Shenyang, Liaoning, 110036, PR China
| | - Ju Liu
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China; API Engineering Technology Research Center of Liaoning Province, Shenyang, Liaoning, 110036, PR China; Small Molecular Targeted Drug R&D Engineering Research Center of Liaoning Province, Shenyang, Liaoning, 110036, PR China.
| | - Ye Chen
- College of Pharmacy of Liaoning University, Shenyang, Liaoning, 110036, PR China; API Engineering Technology Research Center of Liaoning Province, Shenyang, Liaoning, 110036, PR China; Small Molecular Targeted Drug R&D Engineering Research Center of Liaoning Province, Shenyang, Liaoning, 110036, PR China.
| |
Collapse
|
7
|
Chen X, Qian J, Liang S, Qian J, Luo W, Shi Y, Zhu H, Hu X, Wu G, Li X, Liang G. Hyperglycemia activates FGFR1 via TLR4/c-Src pathway to induce inflammatory cardiomyopathy in diabetes. Acta Pharm Sin B 2024; 14:1693-1710. [PMID: 38572108 PMCID: PMC10985127 DOI: 10.1016/j.apsb.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/11/2023] [Accepted: 01/05/2024] [Indexed: 04/05/2024] Open
Abstract
Protein tyrosine kinases (RTKs) modulate a wide range of pathophysiological events in several non-malignant disorders, including diabetic complications. To find new targets driving the development of diabetic cardiomyopathy (DCM), we profiled an RTKs phosphorylation array in diabetic mouse hearts and identified increased phosphorylated fibroblast growth factor receptor 1 (p-FGFR1) levels in cardiomyocytes, indicating that FGFR1 may contribute to the pathogenesis of DCM. Using primary cardiomyocytes and H9C2 cell lines, we discovered that high-concentration glucose (HG) transactivates FGFR1 kinase domain through toll-like receptor 4 (TLR4) and c-Src, independent of FGF ligands. Knocking down the levels of either TLR4 or c-Src prevents HG-activated FGFR1 in cardiomyocytes. RNA-sequencing analysis indicates that the elevated FGFR1 activity induces pro-inflammatory responses via MAPKs-NFκB signaling pathway in HG-challenged cardiomyocytes, which further results in fibrosis and hypertrophy. We then generated cardiomyocyte-specific FGFR1 knockout mice and showed that a lack of FGFR1 in cardiomyocytes prevents diabetes-induced cardiac inflammation and preserves cardiac function in mice. Pharmacological inhibition of FGFR1 by a selective inhibitor, AZD4547, also prevents cardiac inflammation, fibrosis, and dysfunction in both type 1 and type 2 diabetic mice. These studies have identified FGFR1 as a new player in driving DCM and support further testing of FGFR1 inhibitors for possible cardioprotective benefits.
Collapse
Affiliation(s)
- Xiong Chen
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Department of Wound Repair, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Jinfu Qian
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Shiqi Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Jianchang Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Wu Luo
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Yujuan Shi
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Hong Zhu
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiang Hu
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Gaojun Wu
- Department of Cardiology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Xiaokun Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Department of Wound Repair, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
| | - Guang Liang
- Department of Endocrinology, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 311399, China
| |
Collapse
|
8
|
Nian C, Gan X, Liu Q, Wu Y, Kong M, Zhang P, Jin M, Dong Z, Li W, Wang L, He W, Li X, Wu J. Synthesis and Anti-gastric Cancer Activity by Targeting FGFR1 Pathway of Novel Asymmetric Bis-chalcone Compounds. Curr Med Chem 2024; 31:6521-6541. [PMID: 38847254 DOI: 10.2174/0109298673298420240530093525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/03/2024] [Accepted: 04/19/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Bis-chalcone compounds with symmetrical structures, either isolated from natural products or chemically synthesized, have multiple pharmacological activities. Asymmetric Bis-chalcone compounds have not been reported before, which might be attributed to the synthetic challenges involved, and it remains unknown whether these compounds possess any potential pharmacological activities. AIMS The aim of this study is to investigate the synthesis route of asymmetric bis-chalcone compounds and identify potential candidates with efficient anti-tumor activity. METHODS The two-step structural optimization of the bis-chalcone compounds was carried out sequentially, guided by the screening of the compounds for their growth inhibitory activity against gastric cancer cells by MTT assay. The QSAR model of compounds was established through random forest (RF) algorithm. The activities of the optimal compound J3 on growth inhibition, apoptosis, and apoptosis-inducing protein expression in gastric cancer cells were investigated sequentially by colony formation assay, flow cytometry, and western blotting. Further, the inhibitory effects of J3 on the FGFR1 signaling pathway were explored by Western Blotting, shRNA, and MTT assays. Finally, the in vivo anti-tumor activity and mechanism of J3 were studied through nude mice xenograft assay, western blotting. RESULTS 27 asymmetric bis-chalcone compounds, including two types (N and J) were sequentially designed and synthesized. Some N-class compounds have good inhibitory activity on the growth of gastric cancer cells. The vast majority of J-class compounds optimized on the basis of N3 exhibit excellent inhibitory activity on gastric cancer cell growth. We established a QSAR model (R2 = 0.851627) by applying random forest algorithms. The optimal compound J3, which had better activity, concentration-dependently inhibited the formation of gastric cancer cell colonies and led to cell apoptosis by inducing the expression of the pro-apoptotic protein cleaved PARP in a dose-dependent manner. J3 may exert anti-gastric cancer effects by inhibiting the activation of FGFR1/ERK pathway. Moreover, at a dose of 10 mg/kg/day, J3 inhibited tumor growth in nude mice by nearly 70% in vivo with no significant toxic effect on body weight and organs. CONCLUSION In summary, this study outlines a viable method for the synthesis of novel asymmetric bischalcone compounds. Furthermore, the compound J3 demonstrates substantial promise as a potential candidate for an anti-tumor drug.
Collapse
Affiliation(s)
- Chunhui Nian
- The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xin Gan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Department of Pharmacy, Ezhou Central Hospital, Ezhou, Hubei, 436000, China
| | - Qunpeng Liu
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, Zhejiang, 325035, China
| | - Yuna Wu
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University; Wenzhou, 325027, China
| | - Miaomiao Kong
- The 1st affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Peiqin Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Mingming Jin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zhaojun Dong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wulan Li
- The 1st Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Ledan Wang
- The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Wenfei He
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, Zhejiang, 325035, China
| | - Jianzhang Wu
- The Second Affiliated Hospital and Yuying Children's Hospital of the Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
- The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University; Wenzhou, 325027, China
| |
Collapse
|
9
|
Zhang W, Huang Z, Xiao Z, Wang H, Liao Q, Deng Z, Wu D, Wang J, Li Y. NF-κB downstream miR-1262 disturbs colon cancer cell malignant behaviors by targeting FGFR1. Acta Biochim Biophys Sin (Shanghai) 2023; 55:1819-1832. [PMID: 37867436 PMCID: PMC10686795 DOI: 10.3724/abbs.2023235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/23/2023] [Indexed: 10/24/2023] Open
Abstract
Despite substantial advancements in screening, surgery, and chemotherapy, colorectal cancer remains the second most lethal form of the disease. Nuclear factor kappa B (NF-κB) signaling is a critical driver facilitating the malignant transformation of chronic inflammatory bowel diseases. In this study, deregulated miRNAs that could play a role in colon cancer are analyzed and investigated for specific functions in vitro using cancer cells and in vivo using a subcutaneous xenograft model. miRNA downstream targets are analyzed, and predicted binding and regulation are verified. miR-1262, an antitumor miRNA, is downregulated in colon cancer tissue samples and cell lines. miR-1262 overexpression suppresses colon cancer malignant behaviors in vitro and tumor development and metastasis in a subcutaneous xenograft model and a lung metastasis mouse model in vivo. miR-1262 directly targets fibroblast growth factor receptor 1 (FGFR1) and inhibits FGFR1 expression. FGFR1 overexpression shows oncogenic functions through the regulation of cancer cell proliferation, invasion, and migration; when cotransfected, lv-FGFR1 partially attenuates the antitumor effects of agomir-1262. NF-κB binds to the miR-1262 promoter region and inhibits transcription activity. The NF-κB inhibitor CAPE exerts antitumor effects; miR-1262 inhibition partially reverses CAPE effects on colon cancer cells. Conclusively, miR-1262 serves as an antitumor miRNA in colon cancer by targeting FGFR1. The NF-κB/miR-1262/FGFR1 axis modulates colon cancer cell phenotypes, including proliferation, invasion, and migration.
Collapse
Affiliation(s)
- Weilin Zhang
- The Second School of Clinical MedicineSouthern Medical UniversityGuangzhou510080China
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People’s HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
- Department of General SurgeryHunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University)Changsha410005China
| | - Zhongcheng Huang
- Department of General SurgeryHunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University)Changsha410005China
| | - Zhigang Xiao
- Department of General SurgeryHunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University)Changsha410005China
| | - Hui Wang
- Department of Cardiovascular MedicineHunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University)Changsha410005China
| | - Qianchao Liao
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People’s HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| | - Zhengru Deng
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People’s HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| | - Deqing Wu
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People’s HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| | - Junjiang Wang
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People’s HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| | - Yong Li
- The Second School of Clinical MedicineSouthern Medical UniversityGuangzhou510080China
- Department of Gastrointestinal SurgeryDepartment of General SurgeryGuangdong Provincial People’s HospitalGuangdong Academy of Medical SciencesGuangzhou510080China
| |
Collapse
|
10
|
Xiao Z, Wang Z, Zhang T, Liu Y, Si M. Bidirectional Mendelian randomization analysis of the genetic association between primary lung cancer and colorectal cancer. J Transl Med 2023; 21:722. [PMID: 37840123 PMCID: PMC10577972 DOI: 10.1186/s12967-023-04612-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND With the development and popularization of low-dose chest CT technology, the diagnosis and survival rates of patients with early lung cancer (LC) have significantly improved. The occurrence of colorectal cancer (CRC) as the second primary cancer (SPC) in primary lung cancer (PLC) survivors has become an essential factor affecting the prognosis of early LC. This study explored the potential association between PLC and CRC genetically, laying a foundation for developing SPC-CRC prevention strategies after primary early LC. METHODS Based on a two-sample bidirectional Mendelian randomization (MR) design, this study systematically screened genetic instrumental variables (IVs) based on the genome-wide association studies (GWAS) of PLC and CRC, applied inverse variance weighted (IVW) as the main method to assess the incidence association between the two cancers, and used a variety of other MR methods for supplementary analysis. Finally, the Genetic Risk Scores (GRS) method was used for secondary analysis to verify the results robustness further. RESULTS From LC to CRC forward MR analysis, 20 genetic IVs of overall LC, 15 genetic IVs of squamous cell lung carcinoma (LUSC), and 10 genetic IVs of adenocarcinoma of the lung (LUAD) were screened. In the reverse MR analysis from CRC to LC, 47 genetic IVs for overall CRC, 37 for colon cancer, and 25 for rectal cancer were screened. The IVW method and a variety of MR methods all found that overall LC and CRC were significantly associated at the genetic level. Subgroup analysis also showed that LUSC was associated with CRC. And the results of the GRS method were consistent with those of the main analysis, confirming the robustness of the study. Our MR study found an association between LC and CRC, with an increased risk of SPC-CRC following PLC, especially LUSC. Our study provides an essential basis for the precise prevention of SPC-CRC after PLC, suggesting that we should pay more attention to the population with a history of PLC in clinical work, and pay close attention to the incidence of SPC-CRC, and carry out intervention and treatment as soon as possible.
Collapse
Affiliation(s)
- Zhihan Xiao
- Department of Cardiothoracic Surgery, Wuhu Second People's Hospital, Wuhu, China
| | - Zichen Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tongyu Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi Liu
- Department of Digestive System, Anqing Municipal Hospital, Anqing, China.
| | - Mingxuan Si
- Department of Thoracic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
11
|
Michalkova R, Kello M, Cizmarikova M, Bardelcikova A, Mirossay L, Mojzis J. Chalcones and Gastrointestinal Cancers: Experimental Evidence. Int J Mol Sci 2023; 24:ijms24065964. [PMID: 36983038 PMCID: PMC10059739 DOI: 10.3390/ijms24065964] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/10/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Colorectal (CRC) and gastric cancers (GC) are the most common digestive tract cancers with a high incidence rate worldwide. The current treatment including surgery, chemotherapy or radiotherapy has several limitations such as drug toxicity, cancer recurrence or drug resistance and thus it is a great challenge to discover an effective and safe therapy for CRC and GC. In the last decade, numerous phytochemicals and their synthetic analogs have attracted attention due to their anticancer effect and low organ toxicity. Chalcones, plant-derived polyphenols, received marked attention due to their biological activities as well as for relatively easy structural manipulation and synthesis of new chalcone derivatives. In this study, we discuss the mechanisms by which chalcones in both in vitro and in vivo conditions suppress cancer cell proliferation or cancer formation.
Collapse
Affiliation(s)
- Radka Michalkova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Martin Kello
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Martina Cizmarikova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Annamaria Bardelcikova
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Ladislav Mirossay
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| | - Jan Mojzis
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia
| |
Collapse
|
12
|
Zhao G, Ren Y, Yan J, Zhang T, Lu P, Lei J, Rao H, Kang X, Cao Z, Peng F, Peng C, Rao C, Li Y. Neoprzewaquinone A Inhibits Breast Cancer Cell Migration and Promotes Smooth Muscle Relaxation by Targeting PIM1 to Block ROCK2/STAT3 Pathway. Int J Mol Sci 2023; 24:ijms24065464. [PMID: 36982538 PMCID: PMC10051292 DOI: 10.3390/ijms24065464] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Salvia miltiorrhiza Bunge (Danshen) has been widely used to treat cancer and cardiovascular diseases in Chinese traditional medicine. Here, we found that Neoprzewaquinone A (NEO), an active component of S. miltiorrhiza, selectively inhibits PIM1. We showed that NEO potently inhibits PIM1 kinase at nanomolar concentrations and significantly suppresses the growth, migration, and Epithelial-Mesenchymal Transition (EMT) in the triple-negative breast cancer cell line, MDA-MB-231 in vitro. Molecular docking simulations revealed that NEO enters the PIM1 pocket, thereby triggering multiple interaction effects. Western blot analysis revealed that both NEO and SGI-1776 (a specific PIM1 inhibitor), inhibited ROCK2/STAT3 signaling in MDA-MB-231 cells, indicating that PIM1 kinase modulates cell migration and EMT via ROCK2 signaling. Recent studies indicated that ROCK2 plays a key role in smooth muscle contraction, and that ROCK2 inhibitors effectively control the symptoms of high intraocular pressure (IOP) in glaucoma patients. Here, we showed that NEO and SGI-1776 significantly reduce IOP in normal rabbits and relax pre-restrained thoracic aortic rings in rats. Taken together, our findings indicated that NEO inhibits TNBC cell migration and relaxes smooth muscles mainly by targeting PIM1 and inhibiting ROCK2/STAT3 signaling, and that PIM1 may be an effective target for IOP and other circulatory diseases.
Collapse
Affiliation(s)
- Guiying Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yali Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jie Yan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tingrui Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Peng Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jieting Lei
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Huanan Rao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xin Kang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhixing Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Fu Peng
- West China School of Pharmacy, Sichuan University, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chaolong Rao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica with Medical and Edible Values, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Correspondence: (C.R.); (Y.L.)
| | - Yuzhi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Correspondence: (C.R.); (Y.L.)
| |
Collapse
|