1
|
Wang X, Liu Q, Cheng P, Yang T, Zhao T, Liu M, Dai E, Sha W, Yuan J, Rong J, Qu H, Zhou H. LuQi formula ameliorates pressure overload-induced heart failure by regulating macrophages and regulatory T cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156527. [PMID: 40118747 DOI: 10.1016/j.phymed.2025.156527] [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: 09/02/2024] [Revised: 02/11/2025] [Accepted: 02/15/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Inflammatory macrophages in failing myocardium secrete CCL17, which targets CCR4 in immunosuppressive Tregs and inhibits the intracellular second messenger ARRB2-mediated cardiac chemotaxis. Traditional Chinese medicine (TCM) LuQi formula (LQF) is safe and effective in treating heart failure (HF). This study aims to elucidate the cardioprotective mechanism of LQF through its modulation of cardiac macrophages and Tregs. METHODS In vivo, the HF mouse model was established via transverse aortic constriction (TAC), with the superagonistic anti-CD28 monoclonal antibody (CD28-SA)-induced Tregs expansion as a positive control. Proteomics analysis elucidated the core link of LQF in anti-HF. In vitro, bone marrow-derived macrophages (BMDMs) were isolated, and Naive CD4+T cells were sorted and stimulated to differentiate into Tregs. The pharmacological mechanism of LQF was confirmed through histological and molecular biology experiments. RESULTS Proteomics reveals that LQF modulates the immune microenvironment of failing myocardium. We revealed that LQF inhibited cardiac inflammatory macrophage infiltration and NF-κB (p50, p65)/CCL17 axis expression, and promoted cardiac Tregs recruitment against HF, with the comparable efficacy of CD-SA28-induced Tregs expansion. Mechanistically, LQF inhibited the NF-κB activator 1-induced NF-κB (p50, p65)/CCL17 axis overexpression, and JSH-23 (NF-κB Inhibitor) abolished NF-κB (p50, p65)/CCL17 axis expression in inflammatory macrophages. Furthermore, the inhibition of CCL17 expression in inflammatory macrophages by LQF was found to be mediated by NF-κB (p50, p65). LQF concentration-dependently promoted Tregs CD73/Foxp3 axis expression, enhancing Tregs immunosuppressive function. LQF activated CCR4-ARRB2 complex and CCR4/ARRB2 axis expression in Tregs. Although AZD2098 (CCR4 Inhibitor) blocked CCR4 expression and CCR4-ARRB2 complex, LQF promoted ARRB2-mediated Tregs cardiac chemotaxis independent of the CCR4. We revealed that NF-κB p50SEP337-CCL17, NF-κB p65SEP536-CCL17, and CCR4-ARRB2 highly bound subunit interface targets. Molecular docking analysis demonstrated that the LQF's active ingredients exhibit good binding affinity with the NF-κB (p50, p65) /CCL17 axis in macrophages and Foxp3 in Tregs. CONCLUSION LQF has the potential to enhance the cardiac immune microenvironment and effectively prevent and treat HF by modulating both innate and adaptive immune responses. It achieves this by inhibiting the infiltration of inflammatory macrophages, suppressing the NF-κB (p50, p65)/CCL17 axis, and promoting Tregs recruitment. The active ingredients of LQF provide valuable candidate compounds for developing new anti-HF drugs. Furthermore, CD-28SA-induced Tregs expansion showed cardioprotective effects in TAC-induced non-ischemic HF models.
Collapse
Affiliation(s)
- Xinting Wang
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qian Liu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Peipei Cheng
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tianshu Yang
- Department of Cardiovascular Disease, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, ShangHai 200040, China
| | - Tingyao Zhao
- Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200050, China
| | - Meng Liu
- Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200050, China
| | - Enrui Dai
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wanjing Sha
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jinfeng Yuan
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jingfeng Rong
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Huiyan Qu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hua Zhou
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
2
|
Zhao XP, Duan L, Zhao QR, Lv X, Tian NY, Yang SL, Dong K. NLRP3 inflammasome as a therapeutic target in doxorubicin-induced cardiotoxicity: role of phytochemicals. Front Pharmacol 2025; 16:1567312. [PMID: 40313623 PMCID: PMC12043718 DOI: 10.3389/fphar.2025.1567312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025] Open
Abstract
Doxorubicin (DOX) has received widespread attention as a broad-spectrum antitumor drug. However, it has been a recognized challenge that long-term DOX injections can lead to severe cardiotoxicity. There are numerous interventions to DOX-induced cardiotoxicity, and the most cost-effective is phytochemicals. It has been reported that phytochemicals have complex and diverse biological properties, facilitating the mitigation of DOX-induced cardiotoxicity. DOX-induced cardiotoxicity has numerous pathological mechanisms, and the nod-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome-mediated cardiomyocyte pyroptosis is one of them. This review initially presents an overview of the pathological mechanisms that underlie cardiotoxicity induced by DOX. Subsequently, we present a comprehensive elucidation of the structure and activation of the NLRP3 inflammasome. Finally, we provide a detailed summary of phytochemicals that can mitigate DOX-induced cardiotoxicity by influencing the expression of the NLRP3 inflammasome in cardiomyocytes.
Collapse
Affiliation(s)
- Xiao-Peng Zhao
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Lian Duan
- China Volleyball College, Beijing Sport University, Beijing, China
- College of Physical Education, Yanshan University, Qinhuangdao, China
| | - Qian-Ru Zhao
- Shenyang Sports Research and Medical Center, Shenyang Sports Development Center, Shenyang, China
| | - Xing Lv
- Department of Rehabilitation, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Nai-Yuan Tian
- College of Physical Education, Yanshan University, Qinhuangdao, China
| | - Sheng-Lei Yang
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Kun Dong
- College of Physical Education, Yanshan University, Qinhuangdao, China
| |
Collapse
|
3
|
Gong G, Liu Y, Zhang Z, Zheng Y. Therapeutic Targets and Molecular Mechanisms of Calycosin in the Treatment of Depression: Insights From Chronic Mild Stress Animal Models. CNS Neurosci Ther 2025; 31:e70353. [PMID: 40260589 PMCID: PMC12012568 DOI: 10.1111/cns.70353] [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: 10/11/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Depression is a complex psychiatric disorder with limited therapeutic options and various side effects. Calycosin, a bioactive compound derived from Astragalus membranaceus, possesses multiple pharmacological properties. This study aimed to investigate the antidepressant effects of calycosin in chronic mild stress (CMS) animal models of depression and to elucidate its underlying mechanisms. METHODS The antidepressant effects of calycosin were assessed in vivo using CMS animal models of depression, including the grooming frequency test, sucrose intake test, tail suspension test, and open field test. Neurogenic effects were evaluated by measuring the levels of BDNF, GDNF, and NGF in isolated hippocampus tissues. The hepatoprotective effects were assessed by measuring liver enzyme levels. The molecular mechanisms underlying calycosin's antidepressant effects were explored in vitro using PC12 cells. RESULTS Calycosin exhibited potent antidepressant-like activities in CMS animal models of depression. Treatment with calycosin significantly alleviated depressive symptoms and improved neurogenic effects. Additionally, calycosin displayed hepatoprotective effects by modulating liver enzymes in vitro. The antidepressant effects of calycosin are mediated by the stimulation of the TrkB-MEK-Erk1/2-CREB signaling pathway. CONCLUSION In conclusion, calycosin shows promise as a novel therapeutic agent for depression due to its potent antidepressant-like activities and diverse pharmacological properties. Further studies are warranted to elucidate the exact molecular targets of calycosin and to assess its efficacy and safety in clinical settings.
Collapse
Affiliation(s)
- Guowei Gong
- Department of BioengineeringZunyi Medical UniversityZhuhaiGuangdongChina
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food EngineeringHanshan Normal UniversityChaozhouGuangdongChina
| | - Yaqun Liu
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food EngineeringHanshan Normal UniversityChaozhouGuangdongChina
| | - Zhenxia Zhang
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food EngineeringHanshan Normal UniversityChaozhouGuangdongChina
| | - Yuzhong Zheng
- Guangdong Key Laboratory for Functional Substances in Medicinal Edible Resources and Healthcare Products, School of Life Sciences and Food EngineeringHanshan Normal UniversityChaozhouGuangdongChina
| |
Collapse
|
4
|
Seck I, Ndoye SF, Kapchoup MVK, Nguemo F, Ciss I, Ba LA, Ba A, Sokhna S, Seck M. Effects of plant extracts and derivatives on cardiac K +, Nav, and Ca v channels: a review. Nat Prod Res 2025; 39:1213-1240. [PMID: 38586947 DOI: 10.1080/14786419.2024.2337112] [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: 11/14/2023] [Revised: 02/29/2024] [Accepted: 03/24/2024] [Indexed: 04/09/2024]
Abstract
Natural products (NPs) are endless sources of compounds for fighting against several pathologies. Many dysfunctions, including cardiovascular disorders, such as cardiac arrhythmias have their modes of action regulation of the concentration of electrolytes inside and outside the cell targeting ion channels. Here, we highlight plant extracts and secondary metabolites' effects on the treatment of related cardiac pathologies on hERG, Nav, and Cav of cardiomyocytes. The natural product's pharmacology of expressed receptors like alpha-adrenergic receptors causes an influx of Ca2+ ions through receptor-operated Ca2+ ion channels. We also examine the NPs associated with cardiac contractions such as myocardial contractility by reducing the L-type calcium current and decreasing the intracellular calcium transient, inhibiting the K+ induced contractions, decreasing amplitude of myocyte shortening and showed negative ionotropic and chronotropic effects due to decreasing cytosolic Ca2+. We examine whether the NPs block potassium channels, particular the hERG channel and regulatory effects on Nav1.7.
Collapse
Affiliation(s)
- Insa Seck
- Laboratoire de Chimie de Coordination Organique, Université Cheikh Anta Diop de Dakar, Dakar, Senegal
- Laboratoire de Chimie Organique et Thérapeutique, Université Cheikh Anta Diop de Dakar, Dakar, Senegal
| | - Samba Fama Ndoye
- Laboratoire de Chimie Organique et Thérapeutique, Université Cheikh Anta Diop de Dakar, Dakar, Senegal
| | | | - Filomain Nguemo
- Institute of Neurophysiology, University of Cologne, Cologne, Germany
| | - Ismaila Ciss
- Laboratoire de Chimie Organique et Thérapeutique, Université Cheikh Anta Diop de Dakar, Dakar, Senegal
| | - Lalla Aicha Ba
- Laboratoire de Chimie Organique et Thérapeutique, Université Cheikh Anta Diop de Dakar, Dakar, Senegal
| | - Abda Ba
- Laboratoire de Chimie Organique et Thérapeutique, Université Cheikh Anta Diop de Dakar, Dakar, Senegal
| | - Seynabou Sokhna
- Laboratoire de Chimie Organique et Thérapeutique, Université Cheikh Anta Diop de Dakar, Dakar, Senegal
| | - Matar Seck
- Laboratoire de Chimie Organique et Thérapeutique, Université Cheikh Anta Diop de Dakar, Dakar, Senegal
| |
Collapse
|
5
|
Yang X, Pan Y, Cai L, Wang W, Zhai X, Zhang Y, Wu Q, Chen J, Zhang C, Wang Y. Calycosin Ameliorates Neuroinflammation via TLR4-Mediated Signal Following Cerebral Ischemia/Reperfusion Injury in vivo and in vitro. J Inflamm Res 2024; 17:10711-10727. [PMID: 39677283 PMCID: PMC11645956 DOI: 10.2147/jir.s480262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 11/29/2024] [Indexed: 12/17/2024] Open
Abstract
Background Cerebral ischemia-reperfusion injury (CIRI) is a key pathophysiological process that leads to stroke mortality, with TLR4-mediated inflammation playing a crucial role. Our previous research highlighted the neuroprotective effects of the phytoestrogen calycosin on CIRI, although the precise mechanism remains unclear. This study aimed to explore the effects of calycosin on the HMGB1/TLR4/NF-κB signaling pathway in rat models of CIRI, both in vivo and in vitro. Methods In vivo, a rat CIRI model was established using middle cerebral artery occlusion (MCAO), inducing ischemia for 1.5 h followed by 24 h of reperfusion. Calycosin was administered intraperitoneally 1 h after ischemia. Neurological deficits and brain infarct volumes were evaluated. Histological changes and key protein expressions around the ischemic penumbra were assessed by H&E staining and immunofluorescence. In vitro, primary neurons and PC12 cells were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) to mimic CIRI. Cell viability was measured using a CCK8 assay, and alterations in HMGB1/TLR4/NF-κB pathway components were analyzed using qRT-PCR, Western blotting, and ELISA. Results In the MCAO rat model, calycosin significantly reduced neurological deficits and infarct sizes, and improved brain tissue damage following reperfusion. Similarly, in the OGD/R model, calycosin attenuated neuronal injury in PC12 cells and in primary neurons. Additionally, calycosin inhibited LPS-induced activation of the HMGB1/TLR4/NF-κB signaling pathway in PC12 cells. Both in vitro and in vivo studies have shown that calycosin effectively downregulates HMGB1 and TLR4 expression, decreases NF-κB and IκB phosphorylation, and reduces the secretion of inflammatory cytokines such as IL-6 and IL-18. Conclusion These findings suggest that calycosin mitigates cerebral ischemia-reperfusion injury and neuroinflammation by inhibiting the HMGB1/TLR4/NF-κB signaling pathway, thereby providing neuroprotection.
Collapse
Affiliation(s)
- Xin Yang
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Yanjin Pan
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Le Cai
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Wenbo Wang
- Department of Neurosurgery, Nanxishan Hospital of Guangxi Zhuang Autonomous Region, Guilin, 541002, People’s Republic of China
| | - Xiaoya Zhai
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Yuhui Zhang
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Qiguang Wu
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Jian Chen
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People’s Republic of China
| | - Chong Zhang
- Department of Neurology, The Second Affiliated Hospital of Guilin Medical University, Guilin, 541199, People’s Republic of China
- Guangxi Medical and Health Key Cultivation Discipline Construction Project, Guilin, 541199, People’s Republic of China
| | - Yong Wang
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199, People’s Republic of China
- Guangxi Medical and Health Key Cultivation Discipline Construction Project, Guilin, 541199, People’s Republic of China
- Department of Physiology, Guilin Medical University, Guilin, 541199, People’s Republic of China
| |
Collapse
|
6
|
Xu H, Wu K, Guo C, Zhong G. Calycosin action against atherosclerosis: integrating network pharmacology and in-silico investigation. Acta Cardiol 2024; 79:566-574. [PMID: 38771335 DOI: 10.1080/00015385.2024.2356902] [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: 10/26/2023] [Revised: 04/11/2024] [Accepted: 05/12/2024] [Indexed: 05/22/2024]
Abstract
Atherosclerosis, caused by lipid deposit in the arterial wall for narrowing the arteries, is an increased risk factor of developing heart failure. Presently, clinical first-line drug therapy can be found with side effects, and thus new substitute medication should be developed needfully. Calycosin is one of the most bioactive products refined from natural plant, and it exerts promising cardiovascular protective effect. However, the pharmacological mechanisms of calycosin against atherosclerosis have not been elaborated. In this study, a systematic network pharmacology combined with molecular docking analysis was used to reveal the interaction activity and biological target in calycosin against atherosclerosis. We screened all preparative targets linked to calycosin and atherosclerosis from the available public databases. These results indicated total 409 putative targets in calycosin action, 71 of which were interacted with atherosclerosis. Further biological docking analysis suggested that calycosin displayed the powerful binding affinities with target proteins, including interleukin-6 (IL6) and mitogen-activated protein kinase 3 (MAPK3) MAPK3. Then enrichment findings revealed that calycosin action to treat atherosclerosis might be related to inhibition of inflammatory reaction and oxidative stress through modulating nucleolus transcription factor for improving lipid metabolism. In conclusion, the anti-atherosclerotic targets and molecular mechanisms in calycosin action were revealed systematically through preclinical evaluation. And calycosin may be a potential natural compound for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Hongyuan Xu
- Cardiology Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Cardiology Department, Guigang City People's Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, Guangxi, China
| | - Kunpeng Wu
- Neurosurgery Department, The People's Hospital of Laibin City, Laibin, Guangxi, China
| | - Chao Guo
- Department of Clinical Pharmacy, Guigang City People's Hospital, The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, Guangxi, China
| | - Guoqiang Zhong
- Cardiology Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
7
|
Wang Q, Jiang Y, Wei N, Li J, Zhang M, Chen L. Comparative pharmacokinetics of four bioactive components in normal and chronic heart failure rats after oral administration of Qiangxin Lishui Prescription by microdialysis combined with ultra-high-performance liquid chromatography. J Sep Sci 2023; 46:e2300518. [PMID: 37853838 DOI: 10.1002/jssc.202300518] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/07/2023] [Accepted: 10/06/2023] [Indexed: 10/20/2023]
Abstract
Qiangxin Lishui Prescription (QLP) has been clinically applied for treating heart failure with remarkable curative effects. A multi-component pharmacokinetic research is very necessary for determining active substances in it. This study aims to profile the traits and differences in the pharmacokinetics of salvianolic acid B, astragaloside IV, calycosin-7-O-β-D-glucoside and kaempferol in QLP between normal and chronic heart failure (CHF) rats by microdialysis combined with ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). Sensitive, selective, and online microdialysis combined with the UHPLC-MS/MS method was successfully established and applied to study the pharmacokinetics of QLP. The pathological condition of CHF could lead to the enhancement of systematic exposure and reduction of the metabolic rate of four bioactive components for better bioavailability and therapeutic efficacy. The pharmacokinetic results will provide data support for the clinical application of QLP.
Collapse
Affiliation(s)
- Qin Wang
- Department of Pharmacy, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Yong Jiang
- Department of Pharmacy, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Nina Wei
- Department of Pharmacy, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Jindong Li
- Department of Pharmacy, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Mei Zhang
- Department of Pharmacy, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| | - Linwei Chen
- Department of Pharmacy, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, China
| |
Collapse
|
8
|
Chang L, Zhang A, Liu W, Cao P, Dong L, Gao X. Calycosin inhibits hepatocyte apoptosis in acute liver failure by suppressing the TLR4/NF-κB pathway: An in vitro study. Immun Inflamm Dis 2023; 11:e935. [PMID: 37506138 PMCID: PMC10336678 DOI: 10.1002/iid3.935] [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: 03/08/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Acute liver failure (ALF) is a serious liver disease that is difficult to treat owing to its unclear pathogenesis. This study aimed to investigate the roles and molecular mechanisms of calycosin (CA) in ALF. METHODS In this study, the roles and mechanism of CA in ALF were explored using an in vitro lipopolysaccharide (LPS)-induced ALF cell model. Additionally, 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyltetrazolium bromide assay was used to assess the effect of CA on the activity of LPS-induced L02 human liver epithelial cells, and flow cytometry was used to detect apoptosis in L02 cells. Expression levels of apoptosis-related genes, Bax and Bcl-2, were measured using reverse transcription-quantitative polymerase chain reaction and Western blot analysis. Expression levels of inflammatory factors in LPS-induced L02 cells were measured using an enzyme-linked immunosorbent assay. Additionally, the effect of CA on ALF was inhibited via transfection of a toll-like receptor 4 (TLR4)-plasmid to elucidate the relationship between CA and TLR4/nuclear factor (NF)-κB signaling pathway in ALF. RESULTS CA had no toxic effects on L02 cells, but enhanced the activity of LPS-induced L02 cells in a dose-dependent manner. Apoptosis and inflammatory factor release was increased in ALF, activating the TLR4/NF-κB signaling pathway. However, CA treatment inhibited the apoptosis and release of inflammatory factors. Further mechanistic studies revealed that the upregulation of TLR4 expression reversed the alleviating effects of CA on inflammation and apoptosis in LPS-induced L02 cells. CONCLUSION CA alleviates inflammatory damage in LPS-induced L02 cells by inhibiting the TLR4/NF-κB pathway and may be a promising therapeutic agent for ALF treatment.
Collapse
Affiliation(s)
- Le Chang
- Gastroenterology Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Aiqing Zhang
- Gastroenterology Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Wenjuan Liu
- Gastroenterology Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Ping Cao
- Gastroenterology Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Lixian Dong
- Gastroenterology Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Xiaoxue Gao
- Gastroenterology Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalThird Hospital of Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
9
|
Alleviation of liver cirrhosis and associated portal-hypertension by Astragalus species in relation to their UPLC-MS/MS metabolic profiles: a mechanistic study. Sci Rep 2022; 12:11884. [PMID: 35831335 PMCID: PMC9279505 DOI: 10.1038/s41598-022-15958-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/01/2022] [Indexed: 11/08/2022] Open
Abstract
Liver cirrhosis is a late-stage liver disease characterized by excessive fibrous deposition triggering portal-hypertension (PH); the prime restrainer for cirrhosis-related complications. Remedies that can dually oppose hepatic fibrosis and lower PH, may prevent progression into decompensated-cirrhosis. Different Astragalus-species members have shown antifibrotic and diuretic actions with possible subsequent PH reduction. However, A.spinosus and A.trigonus were poorly tested for eliciting these actions. Herein, A.spinosus and A.trigonus roots and aerial parts extracts were subjected to comprehensive metabolic-fingerprinting using UHPLC-MS/MS resulting in 56 identified phytoconstituents, followed by chemometric untargeted analysis that revealed variable metabolic profiles exemplified by different species and organ types. Consequently, tested extracts were in-vivo evaluated for potential antifibrotic/anticirrhotic activity by assessing specific markers. The mechanistic prospective to induce diuresis was investigated by analyzing plasma aldosterone and renal-transporters gene-expression. Serum apelin and dimethylarginine-dimethylaminohydrolase-1 were measured to indicate the overall effect on PH. All extracts amended cirrhosis and PH to varying extents and induced diuresis via different mechanisms. Further, An OPLS model was built to generate a comprehensive metabolic-profiling of A.spinosus and A.trigonus secondary-metabolites providing a chemical-based evidence for their efficacious consistency. In conclusion, A.spinosus and A.trigonus organs comprised myriad pharmacologically-active constituents that act synergistically to ameliorate cirrhosis and associated PH.
Collapse
|