1
|
Zheng S, Xue C, Xue T, Li S, Zao X, Li X, Cao X, Chen Y, Qi W, Wang W, Zhang P, Ye Y. Research Progress of Chinese Medicine in Treating Chronic Liver Disease by Regulating Autophagy. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:2053-2077. [PMID: 39614413 DOI: 10.1142/s0192415x24500794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
In recent years, rising living standards and an accelerated lifestyle have led to an increase in the incidence of chronic liver disease. Modern medicine has yet to fully develop effective methods for preventing and treating these conditions due to their complex pathogenesis. Autophagy, a cellular process that maintains homeostasis by removing abnormal proteins, has emerged as a promising therapeutic target for chronic liver diseases. These diseases include liver fibrosis, liver cirrhosis, non-alcoholic steatohepatitis, chronic hepatitis B, and hepatocellular carcinoma. Chinese medicine, with its multi-component, multi-target, and multi-pathway approach, offers unique advantages in treating these conditions, especially given the unclear etiology of chronic liver diseases. Recent research demonstrates that Chinese medicine - comprising single herbs, herbal combinations, and proprietary formulas - can effectively regulate autophagy, thereby providing therapeutic and preventive benefits for chronic liver diseases. This paper reviews recent studies, categorizes various chronic liver diseases, and examines the impact of active ingredients and compound formulas from Chinese medicine on autophagy. These insights are crucial for slowing the progression of chronic liver diseases and pave the way for the future application of Chinese medicine in preventing and managing these conditions through autophagy modulation.
Collapse
Affiliation(s)
- Shihao Zheng
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100007, P. R. China
- Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Chengyuan Xue
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100007, P. R. China
- Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Tianyu Xue
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, P. R. China
| | - Size Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100007, P. R. China
- Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Xiaobin Zao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100007, P. R. China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Xiaoke Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100007, P. R. China
- Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Xu Cao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100007, P. R. China
- Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Yu Chen
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Wenying Qi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100007, P. R. China
- Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Wei Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100007, P. R. China
- Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Peng Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, P. R. China
| | - Yongan Ye
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100007, P. R. China
- Liver Diseases Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, P. R. China
| |
Collapse
|
2
|
Xu L, Yang J, Cao X, Chen J, Liu Z, Cai L, Yu Y, Huang H. Sequential system based on ferritin delivery system and cell therapy for modulating the pathological microenvironment and promoting recovery. Int J Pharm 2024; 664:124607. [PMID: 39159856 DOI: 10.1016/j.ijpharm.2024.124607] [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: 05/19/2024] [Revised: 08/10/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
The vicious crosstalk among capillarization of hepatic sinusoidal endothelial cells (LSECs), activation of hepatic stellate cells (aHSCs), and hepatocyte damage poses a significant impediment to the successful treatment of liver fibrosis. In this study, we propose a sequential combination therapy aimed at disrupting the malignant crosstalk and reshaping the benign microenvironment while repairing damaged hepatocytes to achieve effective treatment of liver fibrosis. Firstly, H-subunit apoferrin (Ferritin) was adopted to load platycodonin D (PLD) and MnO2, forming ferritin@MnO2/PLD (FMP) nanoparticles, which exploited the high affinity of ferritin for the highly expressed transferrin receptor 1 (TfR1) to achieve the precise targeted delivery of FMP in the liver. Upon PLD intervention, restoration of the fenestration pores in capillarized LSECs was facilitated by modulating the phosphatidyl inositol 3-kinase/protein kinase B (PI3K/AKT) and Kruppel Like Factor 2 (KLF2) signaling pathways both in vitro and in vivo, enabling efficient entry of FMP into the Disse space. Subsequently, FMP NPs effectively inhibited HSC activation by modulating the TLR2/TLR4/NF-κB-p65 signaling pathway. Moreover, FMP NPs efficiently scavenged reactive oxygen species (ROS) and mitigated the expression of inflammatory mediators, thereby reshaping the microenvironment to support hepatocyte repair. Finally, administration of bone marrow mesenchymal stem cells (BMMSCs) was employed to promote the regeneration and functional recovery of damaged hepatocytes. In conclusion, the combined sequential therapy involving FMP and BMMSCs effectively attenuated liver fibrosis induced by CCl4 administration, resulting in significant amelioration of the fibrotic condition. The therapeutic strategy outlined in this study underscores the significance of disrupting the deleterious cellular interactions and remodeling the microenvironment, thereby presenting a promising avenue for clinical intervention in liver fibrosis.
Collapse
Affiliation(s)
- Lixing Xu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Jie Yang
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China; Department of Pharmacy, Haimen People's Hospital, Nantong 226100, China
| | - Xinyu Cao
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Jiayi Chen
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Zhikuan Liu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Liangliang Cai
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China; Department of Pharmacy, Affiliated Hospital of Nantong University, Pharmacy School of Nantong University, Nantong 226001, China.
| | - Yanyan Yu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China.
| | - Haiqin Huang
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China.
| |
Collapse
|
3
|
Qu J, Wang L, Li Y, Li X. Liver sinusoidal endothelial cell: An important yet often overlooked player in the liver fibrosis. Clin Mol Hepatol 2024; 30:303-325. [PMID: 38414375 PMCID: PMC11261236 DOI: 10.3350/cmh.2024.0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 02/29/2024] Open
Abstract
Liver sinusoidal endothelial cells (LSECs) are liver-specific endothelial cells with the highest permeability than other mammalian endothelial cells, characterized by the presence of fenestrae on their surface, the absence of diaphragms and the lack of basement membrane. Located at the interface between blood and other liver cell types, LSECs mediate the exchange of substances between the blood and the Disse space, playing a crucial role in maintaining substance circulation and homeostasis of multicellular communication. As the initial responders to chronic liver injury, the abnormal LSEC activation not only changes their own physicochemical properties but also interrupts their communication with hepatic stellate cells and hepatocytes, which collectively aggravates the process of liver fibrosis. In this review, we have comprehensively updated the various pathways by which LSECs were involved in the initiation and aggravation of liver fibrosis, including but not limited to cellular phenotypic change, the induction of capillarization, decreased permeability and regulation of intercellular communications. Additionally, the intervention effects and latest regulatory mechanisms of anti-fibrotic drugs involved in each aspect have been summarized and discussed systematically. As we studied deeper into unraveling the intricate role of LSECs in the pathophysiology of liver fibrosis, we unveil a promising horizon that pave the way for enhanced patient outcomes.
Collapse
Affiliation(s)
- Jiaorong Qu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Le Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yufei Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
4
|
Zhai S, Wang R, Wang J, Xu X, Niu L, Guo M, Zhang Y, Shi Y, Tang X. Curcumol: a review of its pharmacology, pharmacokinetics, drug delivery systems, structure-activity relationships, and potential applications. Inflammopharmacology 2024; 32:1659-1704. [PMID: 38520574 DOI: 10.1007/s10787-024-01447-6] [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: 08/29/2023] [Accepted: 02/16/2024] [Indexed: 03/25/2024]
Abstract
Curcumol (Cur), a guaiane-type sesquiterpenoid hemiketal, is an important and representative bioactive component extracted from the essential oil of the rhizomes of Curcumae rhizoma which is also known as "Ezhu" in traditional Chinese medicine. Recently, Cur has received considerable attention from the research community due to its favorable pharmacological activities, including anti-cancer, hepatoprotective, anti-inflammatory, anti-viral, anti-convulsant, and other activities, and has also exerted therapeutic effect on various cancers, liver diseases, inflammatory diseases, and infectious diseases. Pharmacokinetic studies have shown that Cur is rapidly distributed in almost all organs of rats after intragastric administration with high concentrations in the small intestine and colon. Several studies focusing on structure-activity relationship (SAR) of Cur have shown that some Cur derivatives, chemically modified at C-8 or C-14, exhibited more potent anti-cancer activity and lower toxicity than Cur itself. This review aims to comprehensively summarize the latest advances in the pharmacological and pharmacokinetic properties of Cur in the last decade with a focus on its anti-cancer and hepatoprotective potentials, as well as the research progress in drug delivery system and potential applications of Cur to date, to provide researchers with the latest information, to highlighted the limitations of relevant research at the current stage and the aspects that should be addressed in future research. Our results indicate that Cur and its derivatives could serve as potential novel agents for the treatment of a variety of diseases, particularly cancer and liver diseases.
Collapse
Affiliation(s)
- Sicheng Zhai
- School of Medical Engineering, Haojing College of Shaanxi University of Science & Technology, Tongyi Avenue, Xi Xian New District, Xianyang City, 712046, Shaanxi Province, People's Republic of China
| | - Rui Wang
- School of Medical Engineering, Haojing College of Shaanxi University of Science & Technology, Tongyi Avenue, Xi Xian New District, Xianyang City, 712046, Shaanxi Province, People's Republic of China
| | - Jingyuan Wang
- Key Laboratory of Basic and New Drug Research in Chinese Medicine, School of Pharmacy, Shaanxi University of Chinese Medicine, Shiji Avenue, Xi Xian New District, Xianyang City, 712046, Shaanxi Province, People's Republic of China
| | - Xiangdong Xu
- School of Medical Engineering, Haojing College of Shaanxi University of Science & Technology, Tongyi Avenue, Xi Xian New District, Xianyang City, 712046, Shaanxi Province, People's Republic of China
| | - Le Niu
- School of Medical Engineering, Haojing College of Shaanxi University of Science & Technology, Tongyi Avenue, Xi Xian New District, Xianyang City, 712046, Shaanxi Province, People's Republic of China
| | - Min Guo
- School of Medical Engineering, Haojing College of Shaanxi University of Science & Technology, Tongyi Avenue, Xi Xian New District, Xianyang City, 712046, Shaanxi Province, People's Republic of China
| | - Yongling Zhang
- School of Medical Engineering, Haojing College of Shaanxi University of Science & Technology, Tongyi Avenue, Xi Xian New District, Xianyang City, 712046, Shaanxi Province, People's Republic of China
| | - Yajun Shi
- Key Laboratory of Basic and New Drug Research in Chinese Medicine, School of Pharmacy, Shaanxi University of Chinese Medicine, Shiji Avenue, Xi Xian New District, Xianyang City, 712046, Shaanxi Province, People's Republic of China.
| | - Xuexue Tang
- School of Medical Engineering, Haojing College of Shaanxi University of Science & Technology, Tongyi Avenue, Xi Xian New District, Xianyang City, 712046, Shaanxi Province, People's Republic of China.
| |
Collapse
|
5
|
Sun D, Du X, Cao X, Wu B, Li S, Zhao Y, Liu T, Xu L, Huang H. Neutrophil-Based Bionic Delivery System Breaks Through the Capillary Barrier of Liver Sinusoidal Endothelial Cells and Inhibits the Activation of Hepatic Stellate Cells. Mol Pharm 2024; 21:2043-2057. [PMID: 38471114 DOI: 10.1021/acs.molpharmaceut.4c00173] [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] [Indexed: 03/14/2024]
Abstract
The capillarization of hepatic sinusoids resulting from the activation of hepatic stellate cells poses a significant challenge, impeding the effective delivery of therapeutic agents to the Disse space for liver fibrosis treatment. Therefore, overcoming these barriers and achieving efficient drug delivery to activated hepatic stellate cells (aHSCs) are pressing challenge. In this study, we developed a synergistic sequential drug delivery approach utilizing neutrophil membrane hybrid liposome@atorvastatin/amlisentan (NCM@AtAm) and vitamin A-neutrophil membrane hybrid liposome @albumin (VNCM@Bai) nanoparticles (NPs) to breach the capillary barrier for targeted HSC cell delivery. Initially, NCM@AtAm NPs were successfully directed to the site of hepatic fibrosis through neutrophil-mediated inflammatory targeting, resulting in the normalization of liver sinusoidal endothelial cells (LSECs) and restoration of fenestrations under the combined influence of At and Am. Elevated tissue levels of the p-Akt protein and endothelial nitric oxide synthase (eNOS) indicated the normalization of LSECs following treatment with At and Am. Subsequently, VNCM@Bai NPs traversed the restored LSEC fenestrations to access the Disse space, facilitating the delivery of Bai into aHSCs under vitamin A guidance. Lastly, both in vitro and in vivo results demonstrated the efficacy of Bai in inhibiting HSC cell activation by modulating the PPAR γ/TGF-β1 and STAT1/Smad7 signaling pathways, thereby effectively treating liver fibrosis. Overall, our designed synergistic sequential delivery system effectively overcomes the barrier imposed by LSECs, offering a promising therapeutic strategy for liver fibrosis treatment in clinical settings.
Collapse
Affiliation(s)
- Dan Sun
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Xiao Du
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing Medical Center for Clinical Pharmacy, Nanjing 210008, China
| | - Xinyu Cao
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Bingyu Wu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Shanshan Li
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui Province 233030,China
| | - Yongmei Zhao
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead, NSW 2145, Australia
| | - Lixing Xu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Haiqin Huang
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| |
Collapse
|
6
|
Lowe KO, Tanase CE, Maghami S, Fisher LE, Ghaemmaghami AM. Inflammatory Network of Liver Fibrosis and How It Can Be Targeted Therapeutically. IMMUNO 2023; 3:375-408. [DOI: 10.3390/immuno3040023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Liver fibrosis is a complex, dynamic process associated with a broad spectrum of chronic liver diseases and acute liver failure, characterised by the dysregulated intrahepatic production of extracellular matrix proteins replacing functional liver cells with scar tissue. Fibrosis progresses due to an interrelated cycle of hepatocellular injury, triggering a persistent wound-healing response. The accumulation of scar tissue and chronic inflammation can eventually lead to cirrhosis and hepatocellular carcinoma. Currently, no therapies exist to directly treat or reverse liver fibrosis; hence, it remains a substantial global disease burden. A better understanding of the intricate inflammatory network that drives the initiation and maintenance of liver fibrosis to enable the rationale design of new intervention strategies is required. This review clarifies the most current understanding of the hepatic fibrosis cellular network with a focus on the role of regulatory T cells, and a possible trajectory for T cell immunotherapy in fibrosis treatment. Despite good progress in elucidating the role of the immune system in liver fibrosis, future work to better define the function of different immune cells and their mediators at different fibrotic stages is needed, which will enhance the development of new therapies.
Collapse
Affiliation(s)
- Kirstin O. Lowe
- School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | | | - Susan Maghami
- Hull York Medical School, University of York, York YO10 5DD, UK
| | - Leanne E. Fisher
- School of Life Sciences, University of Nottingham, Nottingham NG7 2RD, UK
| | | |
Collapse
|
7
|
Duan X, Zhao T, Wang J, Wang J, Zheng Y. Curcumol targets glutaminase 1 to regulate glutamine metabolism and induce senescence of hepatic stellate cells. Eur J Integr Med 2023; 62:102278. [DOI: 10.1016/j.eujim.2023.102278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
|
8
|
Li Y, Lu Y, Nian M, Sheng Q, Zhang C, Han C, Dou X, Ding Y. Therapeutic potential and mechanism of Chinese herbal medicines in treating fibrotic liver disease. Chin J Nat Med 2023; 21:643-657. [PMID: 37777315 DOI: 10.1016/s1875-5364(23)60443-1] [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: 12/07/2022] [Indexed: 10/02/2023]
Abstract
Liver fibrosis is a pathological condition characterized by replacement of normal liver tissue with scar tissue, and also the leading cause of liver-related death worldwide. During the treatment of liver fibrosis, in addition to antiviral therapy or removal of inducers, there remains a lack of specific and effective treatment strategies. For thousands of years, Chinese herbal medicines (CHMs) have been widely used to treat liver fibrosis in clinical setting. CHMs are effective for liver fibrosis, though its mechanisms of action are unclear. In recent years, many studies have attempted to determine the possible mechanisms of action of CHMs in treating liver fibrosis. There have been substantial improvements in the experimental investigation of CHMs which have greatly promoted the understanding of anti-liver fibrosis mechanisms. In this review, the role of CHMs in the treatment of liver fibrosis is described, based on studies over the past decade, which has addressed the various mechanisms and signaling pathways that mediate therapeutic efficacy. Among them, inhibition of stellate cell activation is identified as the most common mechanism. This article provides insights into the research direction of CHMs, in order to expand its clinical application range and improve its effectiveness.
Collapse
Affiliation(s)
- Yanwei Li
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Yunrui Lu
- Liaoning University of Traditional Chinese Medicine, Shenyang 110000, China
| | - Mozuo Nian
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Qiuju Sheng
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Chong Zhang
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Chao Han
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Xiaoguang Dou
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang 110000, China
| | - Yang Ding
- Department of Infectious Diseases, Shengjing Hospital of China Medical University, Shenyang 110000, China.
| |
Collapse
|
9
|
Li Y, Zhou Y, Xia S, Chen L, Yang T, Zhao D, Zhang Z, Shao J, Xu X, Zhang F, Zheng S. Blockade of KLF5/LDH-A feedback loop contributes to Curcumol inhibition of sinusoidal endothelial cell glycolysis and mitigation of liver fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154759. [PMID: 37031640 DOI: 10.1016/j.phymed.2023.154759] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/14/2023] [Accepted: 03/08/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND LSECs (Liver sinusoidal endothelial cells) are the portal of liver, their pathological angiogenesis plays a constructive role in etiopathogenesis of liver fibrosis by affecting liver tissue repair and inflammatory drive. Although intervention in angiogenesis can effectively inhibit abnormal activation of LSEC, no effective drugs have been found to treat liver fibrosis. PURPOSE We investigated the effect of the natural compound Curcumol on LSEC angiogenesis and elucidated the novel underlying mechanism, expecting to provide a scientific basis for exploring potential therapeutic drugs for liver fibrosis. METHODS Various cellular and molecular assays, as well as genetic assays, were used to detect pathological angiogenesis and changes in glycolysis levels in cultured rat LSECs and mouse liver fibrosis models. RESULTS Transcription factor KLF5 is able to influence the angiogenic properties of LSEC by regulating the glycolytic process, and affect the expression of LDH-A by transcriptionally binding to its promoter. In our study, we were surprised to find that LDH-A (the final step of glycolysis) has a strong regulatory effect on the glycolytic process of LSEC. Through in-depth study, we found that LDH-A could affect the transcriptional activity of KLF5, thus forming a positive feedback loop. Curcumol could break this positive feedback loop and inhibit the glycolysis-dependent angiogenic nature of LSEC, thus alleviating liver fibrosis. Curcumol reduced extracellular matrix (ECM) deposition, attenuated pathological angiogenesis in LSEC, and decreased the level of CCl4-induced liver fibrosis in mice. CONCLUSION Our results demonstrated the great utilization potentiality of KLF5 in liver fibrosis, and the innovative discovery that LDH-A regulates the glycolytic process and forms a malignant feedback loop by exerting non-enzymatic effects. It also reveals the prospect of Curcumol-regulated KLF5/LDH-A feedback loop in the treatment of liver fibrosis, providing a new option for the future medicine of liver fibrosis.
Collapse
Affiliation(s)
- Yang Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuanyuan Zhou
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Siwei Xia
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Li Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ting Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Danli Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xuefen Xu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
10
|
Hao M, Yao Z, Zhao M, Chen Z, Wang P, Sang X, Yang Q, Wang K, Han X, Cao G. Active ingredients screening and pharmacological mechanism research of curcumae rhizoma-sparganii rhizoma herb pair ameliorates liver fibrosis based on network pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2023; 305:116111. [PMID: 36592822 DOI: 10.1016/j.jep.2022.116111] [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: 07/24/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Curcumae Rhizoma-Sparganii Rhizoma (CR-SR) is a classic herbal pair to promote blood circulation and remove blood stasis in ancient China. However, the molecular mechanism is still unclear. AIM OF STUDY To screen out the anti-liver fibrosis active ingredients in CR-SR. Moreover, preliminary exploration the molecular mechanism of CR-SR to ameliorates liver fibrosis. MATERIALS AND METHODS In this research, plant taxonomy has been confirmed in the "The Plant List" database (www.theplantlist.org). The chemical components of CR-SR were analysed by ultra-performance liquid chromatography-quadrupole/time-of-flight mass spectrometry (UPLC-Q/TOF-MS). "Component-Target-Pathway-Disease" network of CR-SR components were built by network pharmacology. Then, the interaction between primary components and predicted protein targets based on network pharmacology were validated by molecular docking. The pharmacological actions of CR-SR were verified by blood biochemical indexes, histopathologic examination of CCL4 induced rats' model. The core protein targets were verified by Western blot. The effects of screened active components by molecular autodocking were verified by HSC-T6 cell experiment. RESULTS The result shows that 57 chemical constituents in CR-SR herbal pair were identified by UPLC-Q/TOF-MS, in which, 27 compounds were closely connected with liver fibrosis related protein targets. 55 protein targets screened out by "component-target-pathway-disease network" maybe the underlying targets for CR-SR to cure liver fibrosis. Moreover, the 55 protein targets are mainly related to RNA transcription, apoptosis, and signal transduction. The molecular autodocking predicted that ten components can bond well with PTGS2 and RELA protein targets. The blood biochemical indexes, histopathologic examination of CCL4 induced rats experiment showed that CR-SR has well intervention effect of liver fibrosis. The Western blot analysis indicated that CR-SR could significantly inhibit RELA, PTGS2, IL-6, SRC, and AKT1 protein expression to exert the anti-fibrosis effect. The HSC-T6 cell experiment indicated that both formononetin (FNT) and curdione could significantly inhibit the activation of HSC and reduce the expression of PTGS2, and p-AKT1 which was accordance with the molecular autodocking results. CONCLUSION This study proved the molecular mechanism of CR-SR multi-component and multi-target anti-liver fibrosis effect through mass spectrometry, network pharmacology, and western blotting technology. The research provides a theoretical evidence for the development and utilization of CR-SR herbal pair.
Collapse
Affiliation(s)
- Min Hao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Zhouhui Yao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Mengting Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Ziyan Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Pingping Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Xianan Sang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Qiao Yang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Kuilong Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Xin Han
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Gang Cao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| |
Collapse
|
11
|
Zheng Y, Zhao T, Wang J, Jiang R, Huang J, Li W, Wang J. Curcumol alleviates liver fibrosis through inducing autophagy and ferroptosis in hepatic stellate cells. FASEB J 2022; 36:e22665. [PMID: 36398583 DOI: 10.1096/fj.202200933rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/30/2022] [Accepted: 11/08/2022] [Indexed: 11/19/2022]
Abstract
To explore the effect of curcumol on autophagy and ferroptosis of hepatic stellate cells, and to clarify the molecular mechanism of its anti-hepatic fibrosis. In the present study, we report that curcumol promotes the death of activated HSCs and reduces the deposition of extracellular matrix. Interestingly, curcumol treatment can trigger ferroptosis to eliminate activated HSCs characterized by iron overload, lipid ROS accumulation, glutathione depletion, and lipid peroxidation. Curcumol promotes HSC autophagy, which may be the key mechanism for its induction of ferroptosis. It is worth noting that the upregulation of nuclear receptor coactivator 4 (NCOA4) may play a key molecular mechanism. NCOA4 mediates the release of iron ions and induces the occurrence of ferroptosis. Overall, curcumol promotes autophagy in hepatic stellate cells, mediates the degradation of NCOA4 and FTH1 complexes, releases iron ions, leads to iron overload, and induces ferroptosis, which may be an important mechanism for its anti-hepatic fibrosis effect.
Collapse
Affiliation(s)
- Yang Zheng
- Department of Medicine, Faculty of Chinese Medicine Science, Guangxi University of Chinese Medicine, Nanning, China
| | - Tiejian Zhao
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Jiaru Wang
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Ruizhu Jiang
- Department of Medicine, Faculty of Chinese Medicine Science, Guangxi University of Chinese Medicine, Nanning, China
| | - Jinbiao Huang
- Department of Medicine, Faculty of Chinese Medicine Science, Guangxi University of Chinese Medicine, Nanning, China
| | - Weimin Li
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Jiahui Wang
- Department of Medicine, Faculty of Chinese Medicine Science, Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
12
|
Li Z, Hao E, Cao R, Lin S, Zou L, Huang T, Du Z, Hou X, Deng J. Analysis on internal mechanism of zedoary turmeric in treatment of liver cancer based on pharmacodynamic substances and pharmacodynamic groups. CHINESE HERBAL MEDICINES 2022; 14:479-493. [PMID: 36405057 PMCID: PMC9669400 DOI: 10.1016/j.chmed.2022.06.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 03/04/2022] [Accepted: 06/27/2022] [Indexed: 11/30/2022] Open
Abstract
Zedoary tumeric (Curcumae Rhizoma, Ezhu in Chinese) has a long history of application and has great potential in the treatment of liver cancer. The antiliver cancer effect of zedoary tumeric depends on the combined action of multiple pharmacodynamic substances. In order to clarify the specific mechanism of zedoary tumeric against liver cancer, this paper first analyzes the mechanism of its single pharmacodynamic substance against liver cancer, and then verifies the joint anti liver cancer mechanism of its "pharmacodynamic group". By searching the research on the antihepatoma effect of active components of zedoary tumeric in recent years, we found that pharmacodynamic substances, including curcumol, zedoarondiol, curcumenol, curzerenone, curdione, curcumin, germacrone, β-elemene, can act on multi-target and multi-channel to play an antihepatoma role. For example, curcumin can regulate miR, GLO1, CD133, VEGF, YAP, LIN28B, GPR81, HCAR-1, P53 and PI3K/Akt/mTOR, HSP70/TLR4 and NF-κB. Wnt/TGF/EMT, Nrf2/Keap1, JAK/STAT and other pathways play an antihepatoma role. Network pharmacological analysis showed that the core targets of the "pharmacodynamic group" for anti-life cancer are AKT1, EGFR, MAPK8, etc, and the core pathways are neuroactive live receiver interaction, nitrogen metabolism, HIF-1 signaling pathway, etc. At the same time, by comparing and analyzing the relationship between the specific mechanisms of pharmacodynamic substance and "pharmacodynamic group", it is found that they have great reference significance in target, pathway, biological function, determination of core pharmacodynamic components, formation of core target protein interaction, in-depth research of single pharmacodynamic substance, increasing curative effect and so on. By analyzing the internal mechanism of zedoary tumeric pharmacodynamic substance and "pharmacodynamic group" in the treatment of liver cancer, this paper intends to provide some ideas and references for the deeper pharmacological research of zedoary tumeric and the relationship between pharmacodynamic substance and "pharmacodynamic group".
Collapse
Affiliation(s)
- Zeyu Li
- China ASEAN Joint Laboratory for International Cooperation in Traditional Medicine Research, Nanning 530200, China
- Guangxi Scientific Experiment Center of Traditional Chinese Medicine, Nanning 530000, China
- Guangxi Key Laboratory of Pharmacodynamics of Traditional Chinese Medicine, Nanning 530000, China
| | - Erwei Hao
- China ASEAN Joint Laboratory for International Cooperation in Traditional Medicine Research, Nanning 530200, China
- Guangxi Scientific Experiment Center of Traditional Chinese Medicine, Nanning 530000, China
- Guangxi Key Laboratory of Pharmacodynamics of Traditional Chinese Medicine, Nanning 530000, China
| | - Rui Cao
- China ASEAN Joint Laboratory for International Cooperation in Traditional Medicine Research, Nanning 530200, China
- Guangxi Scientific Experiment Center of Traditional Chinese Medicine, Nanning 530000, China
- Guangxi Key Laboratory of Pharmacodynamics of Traditional Chinese Medicine, Nanning 530000, China
| | - Si Lin
- China ASEAN Joint Laboratory for International Cooperation in Traditional Medicine Research, Nanning 530200, China
- Guangxi Scientific Experiment Center of Traditional Chinese Medicine, Nanning 530000, China
- Guangxi Key Laboratory of Pharmacodynamics of Traditional Chinese Medicine, Nanning 530000, China
| | - Linghui Zou
- China ASEAN Joint Laboratory for International Cooperation in Traditional Medicine Research, Nanning 530200, China
- Guangxi Scientific Experiment Center of Traditional Chinese Medicine, Nanning 530000, China
- Guangxi Key Laboratory of Pharmacodynamics of Traditional Chinese Medicine, Nanning 530000, China
| | - Tianyan Huang
- China ASEAN Joint Laboratory for International Cooperation in Traditional Medicine Research, Nanning 530200, China
- Guangxi Scientific Experiment Center of Traditional Chinese Medicine, Nanning 530000, China
- Guangxi Key Laboratory of Pharmacodynamics of Traditional Chinese Medicine, Nanning 530000, China
| | - Zhengcai Du
- China ASEAN Joint Laboratory for International Cooperation in Traditional Medicine Research, Nanning 530200, China
- Guangxi Scientific Experiment Center of Traditional Chinese Medicine, Nanning 530000, China
- Guangxi Key Laboratory of Pharmacodynamics of Traditional Chinese Medicine, Nanning 530000, China
| | - Xiaotao Hou
- China ASEAN Joint Laboratory for International Cooperation in Traditional Medicine Research, Nanning 530200, China
- Guangxi Scientific Experiment Center of Traditional Chinese Medicine, Nanning 530000, China
- Guangxi Key Laboratory of Pharmacodynamics of Traditional Chinese Medicine, Nanning 530000, China
| | - Jiagang Deng
- China ASEAN Joint Laboratory for International Cooperation in Traditional Medicine Research, Nanning 530200, China
- Guangxi Scientific Experiment Center of Traditional Chinese Medicine, Nanning 530000, China
- Guangxi Key Laboratory of Pharmacodynamics of Traditional Chinese Medicine, Nanning 530000, China
| |
Collapse
|
13
|
Zhou BW, Liu HM, Jia XH. The Role and Mechanisms of Traditional Chinese Medicine for Airway Inflammation and Remodeling in Asthma: Overview and Progress. Front Pharmacol 2022; 13:917256. [PMID: 35910345 PMCID: PMC9335520 DOI: 10.3389/fphar.2022.917256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/09/2022] [Indexed: 11/27/2022] Open
Abstract
Asthma as an individual disease has blighted human health for thousands of years and is still a vital global health challenge at present. Though getting much progress in the utilization of antibiotics, mucolytics, and especially the combination of inhaled corticosteroids (ICS) and long-acting β-agonists (LABA), we are confused about the management of asthmatic airway inflammation and remodeling, which directly threatens the quality of life for chronic patients. The blind addition of ICS will not benefit the remission of cough, wheeze, or sputum, but to increase the risk of side effects. Thus, it is necessary to explore an effective therapy to modulate asthmatic inflammation and airway remodeling. Traditional Chinese Medicine (TCM) has justified its anti-asthma effect in clinical practice but its underlying mechanism and specific role in asthma are still unknown. Some animal studies demonstrated that the classic formula, direct exacts, and natural compounds isolated from TCM could significantly alleviate airway structural alterations and exhibit the anti-inflammatory effects. By investigating these findings and data, we will discuss the possible pathomechanism underlined airway inflammation and remodeling in asthma and the unique role of TCM in the treatment of asthma through regulating different signaling pathways.
Collapse
Affiliation(s)
- Bo-wen Zhou
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hua-man Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xin-hua Jia
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Xin-hua Jia,
| |
Collapse
|
14
|
Zheng Y, Wang J, Zhao T, Wang L, Wang J. Modulation of the VEGF/AKT/eNOS signaling pathway to regulate liver angiogenesis to explore the anti-hepatic fibrosis mechanism of curcumol. JOURNAL OF ETHNOPHARMACOLOGY 2021; 280:114480. [PMID: 34358654 DOI: 10.1016/j.jep.2021.114480] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 07/20/2021] [Accepted: 07/30/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Curcuma is a common Chinese herbal medicine that is used in the clinical treatment of chronic liver disease. Studies have found that curcumol is the main active ingredient of curcuma and has good hepatoprotective and anti-inflammatory effects. However, there are few reports on the molecular mechanism underlying the anti-liver fibrosis effect of curcumol. AIM To explore the effect of curcumol on liver angiogenesis, and to reveal the mechanism of curcumol against liver fibrosis. MATERIALS AND METHODS We used liver collagenase perfusion combined with Percoll density gradient sedimentation to separate primary liver sinusoidal endothelial cells, and then applied a leptin-activated cell pathological model. The cells were divided into four treatment groups as follows: blank group, model group, curcumol group, and solafini group. MTT was used to detect the cell proliferation rate in each group, and RT-PCR and western blotting were used to detect the expressions of VEGF, AKT, eNOS, CD31, and vWF. A fluorescent probe was used to detect NO expression, and scanning electron microscopy was used to observe changes in the cell fenestration structure. Angiogenesis assays were used to observe blood vessel formation in each group. RESULTS The results of the MTT test found that the proliferation rate of each group was higher. The results of the molecular biology tests found that curcumol inhibited the activity of the VEGF/AKT/eNOS pathway, thereby increasing fenestration of sinusoidal endothelial cells and inhibiting liver angiogenesis. These differences were statistically significant compared with the model group. CONCLUSIONS Curcumol inhibits the activity of the VEGF/AKT/eNOS signaling pathway, regulates the structure of hepatic sinusoidal endothelial cells, and inhibits liver angiogenesis, which together may explain its anti-liver fibrosis mechanism.
Collapse
Affiliation(s)
- Yang Zheng
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese, Medicine, Nanning, Guangxi, 530222, China
| | - Jiaru Wang
- College of Nursing,Guangdong Medical University, Dongguan, Guangdong, 523000, China
| | - Tiejian Zhao
- Department of Physiology, College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530222, China
| | - Lei Wang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese, Medicine, Nanning, Guangxi, 530222, China
| | - Jiahui Wang
- Department of Medicine, Faculty of Chinese Medicine Science Guangxi University of Chinese, Medicine, Nanning, Guangxi, 530222, China.
| |
Collapse
|
15
|
Sheng W, Xu W, Ding J, Li L, You X, Wu Y, He Q. Curcumol inhibits the malignant progression of prostate cancer and regulates the PDK1/AKT/mTOR pathway by targeting miR‑9. Oncol Rep 2021; 46:246. [PMID: 34590156 PMCID: PMC8493056 DOI: 10.3892/or.2021.8197] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/12/2021] [Indexed: 02/07/2023] Open
Abstract
Curcumol has been reported to exert anti-tumor activity, but its intrinsic molecular mechanism in prostate cancer remains to be elucidated. The present study aimed to analyze the effect of curcumol on prostate cancer and identify its possible internal regulatory pathway using in vitro cell culture and in vivo tumor model experiments. The cytotoxicity of curcumol was detected using a Cell Counting Kit-8 assay and it was found that curcumol had no obvious toxicity or side effects on RWPE-1 cells. Wound healing, Transwell and flow cytometry assays demonstrated that curcumol could affect the activity of PC3 cells. The luciferase reporter assay also indicated that microRNA (miR)-9 could directly target pyruvate dehydrogenase kinase 1 (PDK1). After PC3 cells were transfected with miR-9 inhibitor or treated with curcumol, the expression levels of the PDK1/AKT/mTOR signaling pathway-related proteins [PDK1, phosphorylated (p)-AKT and p-mTOR] were increased or decreased, respectively. Next, the prostate cancer cell xenograft model was established. Tumor size and the expression levels of PDK1/AKT/mTOR signaling pathway-related factors were altered following treatment with curcumol. The in vitro and in vivo experiments collectively demonstrated that curcumol could inhibit the PDK1/AKT/mTOR signaling pathway by upregulating the expression level of miR-9. The present study found that curcumol regulates the PDK1/AKT/mTOR signaling pathway via miR-9 and affects the development of prostate cancer. These findings could provide a possible scientific insight for research into treatments for prostate cancer.
Collapse
Affiliation(s)
- Wen Sheng
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Wenjing Xu
- Dermatology Department, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410021, P.R. China
| | - Jin Ding
- Andrology Clinic, Shenzhen Baoan Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong 518133, P.R. China
| | - Ling Li
- Medical Basic Teaching Experiment Center, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Xujun You
- Andrology Clinic, Shenzhen Baoan Hospital of Traditional Chinese Medicine, Shenzhen, Guangdong 518133, P.R. China
| | - Yongrong Wu
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Qinghu He
- Andrology Laboratory, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| |
Collapse
|
16
|
Jia S, Guo P, Lu J, Huang X, Deng L, Jin Y, Zhao L, Fan X. Curcumol Ameliorates Lung Inflammation and Airway Remodeling via Inhibiting the Abnormal Activation of the Wnt/β-Catenin Pathway in Chronic Asthmatic Mice. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:2641-2651. [PMID: 34188444 PMCID: PMC8232843 DOI: 10.2147/dddt.s292642] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 05/24/2021] [Indexed: 12/14/2022]
Abstract
Background Curcumol exhibits anti-inflammatory effect, but its effect on chronic asthma lacked research. Therefore, this study explored the role of curcumol in asthma. Methods A chronic asthmatic mice model was established by ovalbumin induction. After treatment with curcumol, airway resistance in mice was detected by forced oscillation technique. The histopathological features of airway tissues, pulmonary inflammation, and inflammation cell recruitment in the bronchoalveolar lavage fluid (BALF) of mice were detected by hematoxylin-eosin staining. Collagen deposition in the airways of mice was examined by Masson staining. The secretion of ovalbumin-IgE, IL-4, IL-5, IL-13 in mouse serum and VEGFA secretion in BALF were analyzed by ELISA. Finally, the expressions of β-catenin, Wnt5a, VEGFA, TGF-β1, Fibronectin, and MMP-9 in mice lung tissues were determined by Western blot or immunohistochemical. Results Curcumol attenuated airway hyperresponsiveness, airway remodeling, and pulmonary inflammation in chronic asthmatic mice. Curcumol relieved collagen deposition in airway tissues, inflammation cell recruitment in BALF, and reduced the up-regulation of serum ovalbumin-IgE, IL-4, IL-5, and IL-13 and BALF VEGFA in chronic asthmatic mice. In addition, curcumol attenuated the up-regulated expressions of β-catenin, Wnt5a, VEGFA, TGF-β1, Fibronectin, and MMP-9 in the lung tissues of chronic asthmatic mice, but curcumol treatment did not show such effects on healthy mice. Conclusion Our findings revealed that curcumol could ameliorate lung inflammation and airway remodeling by inhibiting the abnormal activation of the Wnt/β-catenin pathway in chronic asthmatic mice, indicating that curcumol could be used as a novel anti-asthma drug for basic and clinical research.
Collapse
Affiliation(s)
- Shanshan Jia
- Department of Respiration, Hengdian Wenrong's Hospital, Dongyang, People's Republic of China
| | - Pin Guo
- Lithotriptic Section, Dongyang Red Cross Hospital, Dongyang City, People's Republic of China
| | - Junhua Lu
- Department of Respiration, Hengdian Wenrong's Hospital, Dongyang, People's Republic of China
| | - Xujun Huang
- Intensive Care Unit, Hengdian Wenrong's Hospital, Dongyang, People's Republic of China
| | - Laming Deng
- Department of Emergency, Hengdian Wenrong's Hospital, Dongyang, People's Republic of China
| | - Yan Jin
- Department of Nursing, Hengdian Wenrong's Hospital, Dongyang, People's Republic of China
| | - Lanyan Zhao
- Department of Respiration, Dongyang People's Hospital, Dongyang City, People's Republic of China
| | - Xiaofang Fan
- School of Basic Medicine, Wenzhou Medical University, Wenzhou City, People's Republic of China
| |
Collapse
|
17
|
Chen Y, Liao W, Zhu Z, Chen J, Yang Q, Zheng Y, Zhang X, Limsila B, Lu M, Fu S, Li R. Essential oil from the raw and vinegar-processed Rhizoma Curcumae ameliorate CCl 4-induced liver fibrosis: integrating network pharmacology and molecular mechanism evaluation. Food Funct 2021; 12:4199-4220. [PMID: 33870974 DOI: 10.1039/d0fo03052j] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Liver fibrosis, caused by multiple chronic liver injuries, is a known contributor to cirrhosis and even liver cancer. As a Traditional Chinese Medicine (TCM), Rhizoma curcumae has been extensively used in the treatment of liver fibrosis with satisfying therapeutic effects; however, its mechanism is unclear. The essential oil is the main bioactive component. The purpose of this study was to investigate the chemical profile and the pharmacological mechanisms of the essential oil of Rhizoma curcumae (EORC) against liver fibrosis by combining network pharmacology and transcriptomic technologies. A total of 37 active compounds were identified using the GC/MS system and literature mining, and the corresponding putative targets were predicted. Then, network pharmacology method was applied to identify the 168 candidate targets of EORC-alleviated liver fibrosis. String database and Cytoscape software were used to build the herb-compound-target network and protein-protein interactions (PPIs) network. Functional and pathway enrichment analysis indicated that EORC significantly influenced TGF-β1/Smads and PI3K/AKT pathways. Experimentally, we verified that EORC attenuated the severity and pathological changes during liver fibrosis progression based on the CCl4-induced liver fibrosis rat model. Transcriptomic technologies demonstrated that EORC ameliorated liver fibrosis partially by regulating the TGF-β1/Smads and PI3K/AKT pathways. In addition, the effect of vinegar-processed EORC was more significant than that of the raw one. Therefore, EORC can alleviate the severity of liver fibrosis through mechanisms predicted by network pharmacology and provide a basis for the further understanding of the application of EORC in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Yi Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Wan Liao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Zongping Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Jiao Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Qingsong Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Yongfeng Zheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Xinjie Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Boonjai Limsila
- Institute of Thai-Chinese Medicine Department of Thai Traditional and Alternative Medicines, Ministry of Public Health, Bangkok 11000, Thailand
| | - Meigui Lu
- Huachiew TCM Hospital, Bangkok 10100, Thailand
| | - Shu Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| | - Rui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China.
| |
Collapse
|