1
|
Zhang M, Barroso E, Peña L, Rada P, Valverde ÁM, Wahli W, Palomer X, Vázquez-Carrera M. PPARβ/δ attenuates hepatic fibrosis by reducing SMAD3 phosphorylation and p300 levels via AMPK in hepatic stellate cells. Biomed Pharmacother 2024; 179:117303. [PMID: 39153437 DOI: 10.1016/j.biopha.2024.117303] [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: 06/05/2024] [Revised: 08/04/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024] Open
Abstract
The role of peroxisome proliferator-activated receptor (PPAR)β/δ in hepatic fibrosis remains a subject of debate. Here, we examined the effects of a PPARβ/δ agonist on the pathogenesis of liver fibrosis and the activation of hepatic stellate cells (HSCs), the main effector cells in liver fibrosis, in response to the pro-fibrotic stimulus transforming growth factor-β (TGF-β). The PPARβ/δ agonist GW501516 completely prevented glucose intolerance and peripheral insulin resistance, blocked the accumulation of collagen in the liver, and attenuated the expression of inflammatory and fibrogenic genes in mice fed a choline-deficient high-fat diet (CD-HFD). The antifibrogenic effect of GW501516 observed in the livers CD-HFD-fed mice could occur through an action on HSCs since primary HSCs isolated from Ppard-/- mice showed increased mRNA levels of the profibrotic gene Col1a1. Moreover, PPARβ/δ activation abrogated TGF-β1-mediated cell migration (an indicator of cell activation) in LX-2 cells (immortalized activated human HSCs). Likewise, GW501516 attenuated the phosphorylation of the main downstream intracellular protein target of TGF-β1, suppressor of mothers against decapentaplegic (SMAD)3, as well as the levels of the SMAD3 co-activator p300 via the activation of AMP-activated protein kinase (AMPK) and the subsequent inhibition of extracellular signal-regulated kinase-1/2 (ERK1/2) in LX-2 cells. Overall, these findings uncover a new mechanism by which the activation of AMPK by a PPARβ/δ agonist reduces TGF-β1-mediated activation of HSCs and fibrosis via the reduction of both SMAD3 phosphorylation and p300 levels.
Collapse
Affiliation(s)
- Meijian Zhang
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain.
| | - Lucía Peña
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain
| | - Patricia Rada
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC/UAM), Madrid, Spain
| | - Ángela M Valverde
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC/UAM), Madrid, Spain
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, Lausanne CH-1015, Switzerland; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; ToxAlim (Research Center in Food Toxicology), INRAE, UMR1331, Toulouse Cedex F-31300, France
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain.
| |
Collapse
|
2
|
Wang M, Wang Y, Li X, Yin Y, Zhang X, Wu S, Wang H, Zhao Y. Effects of Dietary Ursolic Acid on Growth Performance and Intestinal Health of Largemouth Bass ( Micropterus salmoides). Animals (Basel) 2024; 14:2492. [PMID: 39272277 PMCID: PMC11394043 DOI: 10.3390/ani14172492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
This study aimed to investigate the effects of ursolic acid (UA) on the growth performance and intestinal health of largemouth bass (Micropterus salmoides). Four diets were formulated with UA supplementation at 0, 250, 500, and 1000 mg/kg, defined as the control (CON), UA250, UA500, and UA1000, respectively. After an 8-week feeding experiment, the results showed that, in the UA500 group, the final body weight (FBW), weight gain rate (WGR), and specific growth rate (SGR) increased, and the feed conversion ratio (FCR) and hepatosomatic index decreased. Total superoxide dismutase (T-SOD) activity exhibited a significant increase, and malondialdehyde (MDA) content decreased. An intestinal histological analysis revealed an improvement in the intestinal structural integrity of the UA500 group. The mRNA relative expression levels of physical barrier-related genes [occludin, claudin-1, and zonula occluden-1 (zo-1)] were upregulated. The mRNA relative expression of interlenkin 10 (il-10) increased, and the mRNA relative expression of interlenkin 1β (il-1β) and tumor necrosis factor-α (tnf-α) significantly decreased. The abundance of Firmicutes and Proteobacteria decreased, and the abundance of Tenericutes increased. The abundance of Mycoplasma, Cyanobium, and Staphylococcus decreased, while the abundance of Clostridium increased. In conclusion, dietary supplementation of UA significantly enhanced the growth performance and antioxidant capacity of largemouth bass while improving intestinal barrier function through its influence on the abundance of intestinal flora, such as Tenericutes, Firmicutes, and Mycoplasma. Optimal dietary UA levels for largemouth bass were determined to be between 498 and 520 mg/kg based on quadratic regression analyses of WGR, SGR, and FCR or T-SOD and MDA content.
Collapse
Affiliation(s)
- Min Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Yongfang Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Xiang Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Yue Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Xiwen Zhang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Shuang Wu
- Fisheries College, Hunan Agricultural University, Changsha 410128, China
| | - Hongquan Wang
- Fisheries College, Hunan Agricultural University, Changsha 410128, China
| | - Yurong Zhao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
3
|
Zhao M, Wu F, Tang Z, Yang X, Liu Y, Wang F, Chen B. Anti-inflammatory and antioxidant activity of ursolic acid: a systematic review and meta-analysis. Front Pharmacol 2023; 14:1256946. [PMID: 37841938 PMCID: PMC10568483 DOI: 10.3389/fphar.2023.1256946] [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: 07/12/2023] [Accepted: 09/19/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction: There is currently evidence suggesting that ursolic acid may exert a favorable influence on both anti-inflammatory and antioxidant impact. Nevertheless, the anti-inflammatory and antioxidant activities of ursolic acid have not been systematically evaluated. Consequently, this study aims to conduct a systematic review and meta-analysis regarding the impact of ursolic acid on markers of inflammatory and antioxidant activity in both animal models and in vitro systems. Methods: The search encompassed databases such as PubMed, Web of Science, Google Scholar, and ScienceDirect, up until May 2023. All eligible articles in English were included in the analysis. Standard mean difference (SMD) was pooled using a random-effects model, and the included studies underwent a thorough assessment for potential bias. Results: The final review comprised 31 articles. In disease-model related studies, animal experiments have consistently shown that ursolic acid significantly reduced the levels of inflammatory parameters IL-1β, IL-6 and TNF-α in mouse tissues. In vitro studies have similarly showed that ursolic acid significantly reduced the levels of inflammatory parameters IL-1β, IL-6, IL-8 and TNF-α. Our results showed that ursolic acid could significantly elevate SOD and GSH levels, while significantly reducing MDA levels in animal tissues. The results of in vitro studies shown that ursolic acid significantly increased the level of GSH and decreased the level of MDA. Discussion: Findings from both animal and in vitro studies suggest that ursolic acid decreases inflammatory cytokine levels, elevates antioxidant enzyme levels, and reduces oxidative stress levels (graphical abstract). This meta-analysis furnishes compelling evidence for the anti-inflammatory and antioxidant properties of ursolic acid.
Collapse
Affiliation(s)
- Man Zhao
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Fengyang Wu
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Zhaohong Tang
- Hebei Research Institute of Microbiology Co., Ltd., Baoding, China
| | - Xinyu Yang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Yanhua Liu
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Fengxia Wang
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| | - Baojiang Chen
- College of Animal Science and Technology, Hebei Agricultural University, Baoding, China
| |
Collapse
|
4
|
Deng J, Qin L, Zhou Z. Network Pharmacology and Molecular Docking Reveal the Mechanism of Isodon ternifolius (D. Don) Kudo Against Liver Fibrosis. Drug Des Devel Ther 2023; 17:2335-2351. [PMID: 37576085 PMCID: PMC10416792 DOI: 10.2147/dddt.s412818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/18/2023] [Indexed: 08/15/2023] Open
Abstract
Aim Many studies have demonstrated the hepatoprotective or anti-fibrotic effects of Isodon ternifolius, but its pharmacological basis and mechanism remain unclear. In this study, we used in vitro models to validate the predicted results and revealed the potential mechanism of action and active ingredients through network pharmacology methods and molecular docking. Methods The chemical components of Isodon ternifolius were identified by literatures. Potential targets of Isodon ternifolius were predicted by Swiss Target Prediction. The disease targets were collected through the databases of Gene Card. Common targets of Isodon ternifolius and liver fibrosis were obtained by online tool Venny 2.1. PPI protein interaction network was obtained using String database, and target protein interaction network was drawn using Cytoscape software. Signaling pathway enrichment analysis was performed on drug-disease targets with of DAVID database. Results Twenty-one potential active ingredients and 298 potential targets were predicted by Swiss Target Prediction platform. Ninety pathways related to liver fibrosis were obtained by KEGG enrichment. The TLR4, MAPK and PI3K-Akt signaling pathways are mostly associated with liver fibrosis. Molecular docking techniques were used to validate the core target proteins TNF, Akt1, MAPK1, EGFR and TLR4 binding to the ingredients of Isodon ternifolius, which showed that a multitude of ingredients of Isodon ternifolius were able to bind to the above target proteins, especially 2α-hydroxy oleanolic acid and (-)-Lambertic acid. Our experimental validation results showed that Isodon ternifolius inhibited the activation of PI3K-Akt and ERK1/2 signaling pathways. Conclusion Through a network pharmacology approach and in vitro cell assay, we predicted and validated the active compounds of Isodon ternifolius and its potential targets for LF treatment. The results suggest that the mechanism of Isodon ternifolius treating LF by inhibiting angiogenesis may be related to the ERK1/2 and PI3K/Akt signaling pathways.
Collapse
Affiliation(s)
- Jiasheng Deng
- School of Pharmacy, Guangxi University of Chinese Medicine, Nanning, Guangxi, People’s Republic of China
| | - Le Qin
- Department of Pharmacy, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, People’s Republic of China
| | - Zhipin Zhou
- Department of Pharmacy, Liuzhou People’s Hospital, Liuzhou, Guangxi, People’s Republic of China
| |
Collapse
|
5
|
Abstract
Liver fibrosis has a high incidence worldwide and is the common pathological basis of many chronic liver diseases. Liver fibrosis is caused by the excessive deposition of extracellular matrix and concomitant collagen accumulation in livers and can lead to the development of liver cirrhosis and even liver cancer. A large number of studies have provided evidence that liver fibrosis can be blocked or even reversed by appropriate medical interventions. However, the antifibrosis drugs with ideal clinical efficacy are still insufficient. The edible plant-derived natural compounds have been reported to exert effective antifibrotic effects with few side-effects, representing a kind of promising source for the treatment of liver fibrosis. In this article, we reviewed the current progress of the natural compounds derived from dietary plants in the treatment of liver fibrosis, including phenolic compounds (capsaicin, chlorogenic acid, curcumin, ellagic acid, epigallocatechin-3-gallate, resveratrol, sinapic acid, syringic acid, vanillic acid and vitamin E), flavonoid compounds (genistein, hesperidin, hesperetin, naringenin, naringin and quercetin), sulfur-containing compounds (S-allylcysteine, ergothioneine, lipoic acid and sulforaphane) and other compounds (betaine, caffeine, cucurbitacin B, lycopene, α-mangostin, γ-mangostin, ursolic acid, vitamin C and yangonin). The pharmacological effects and related mechanisms of these compounds in in-vivo and in-vitro models of liver fibrosis are focused.
Collapse
|
6
|
Ren G, Xu G, Li R, Xie H, Cui Z, Wang L, Zhang C. Modulation of Bleomycin-induced Oxidative Stress and Pulmonary Fibrosis by Ginkgetin in Mice via AMPK. Curr Mol Pharmacol 2023; 16:217-227. [PMID: 35249515 DOI: 10.2174/1874467215666220304094058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 12/20/2021] [Accepted: 12/29/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Ginkgetin, a flavonoid extracted from Ginkgo biloba, has been shown to exhibit broad anti-inflammatory, anticancer, and antioxidative bioactivity. Moreover, the extract of Ginkgo folium has been reported on attenuating bleomycin-induced pulmonary fibrosis, but the anti-fibrotic effects of ginkgetin are still unclear. This study was intended to investigate the protective effects of ginkgetin against experimental pulmonary fibrosis and its underlying mechanism. METHODS In vivo, bleomycin (5 mg/kg) in 50 μL saline was administrated intratracheally in mice. One week after bleomycin administration, ginkgetin (25 or 50 mg/kg) or nintedanib (40 mg/kg) was administrated intragastrically daily for 14 consecutive days. In vitro, the AMPK-siRNA transfection in primary lung fibroblasts further verified the regulatory effect of ginkgetin on AMPK. RESULTS Administration of bleomycin caused characteristic histopathology structural changes with elevated lipid peroxidation, pulmonary fibrosis indexes, and inflammatory mediators. The bleomycin- induced alteration was normalized by ginkgetin intervention. Moreover, this protective effect of ginkgetin (20 mg/kg) was equivalent to that of nintedanib (40 mg/kg). AMPK-siRNA transfection in primary lung fibroblasts markedly blocked TGF-β1-induced myofibroblasts transdifferentiation and abolished oxidative stress. CONCLUSION All these results suggested that ginkgetin exerted ameliorative effects on bleomycininduced oxidative stress and lung fibrosis mainly through an AMPK-dependent manner.
Collapse
Affiliation(s)
- Guoqing Ren
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, P.R. China
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Gonghao Xu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Renshi Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, P.R. China
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Haifeng Xie
- Chengdu Biopurify Phytochemicals Ltd., Chengdu, P.R. China
| | - Zhengguo Cui
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, P.R. China
- Department of Environmental Health, 23-3 Matsuoka Shimoaizuki, Eiheiji,Fukui 910-1193, Japan
| | - Lei Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Chaofeng Zhang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, P.R. China
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Medicines, China Pharmaceutical University, Nanjing, P.R. China
| |
Collapse
|
7
|
Hyun MK, Kim DH, Park CH, Noh SG, Choi S, Lee JY, Choi JH, Park D, Choi YJ, Chung HY. Protective mechanisms of loquat leaf extract and ursolic acid against diabetic pro-inflammation. J Mol Med (Berl) 2022; 100:1455-1464. [PMID: 35962799 DOI: 10.1007/s00109-022-02243-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 07/14/2022] [Accepted: 08/04/2022] [Indexed: 10/15/2022]
Abstract
The pharmacological effectiveness of loquat leaf extract (LE) and its important component, ursolic acid (UA), in the treatment of diabetes mellitus, has been well established in traditional medicine; however, the mechanism underlying their action is still unclear. We evaluated the protective effects of LE and UA against hyperglycemia-induced advanced glycation end product (AGE) formations and hepatic pro-inflammation. Oral administration of UA and LE at a dose of 50 mg/kg/day for 15 days yielded no significant hypoglycemic effect in diabetic db/db mice. UA and LE suppressed hepatic oxidative stress and AGE formation in diabetic mice, and this was followed by the downregulated mitogen-activated protein kinase signaling and nuclear factor κ B (NF-κB) activity. To identify the molecular target of LE and UA, a docking simulation was performed, and this predicted UA to bind to liver kinase B1 (LKB1), an upstream of AMP-activated protein kinase (AMPK)/transcription factor forkhead box O3 (FOXO3) axis. UA reversed the high-glucose-induced downregulation of LKB1-AMPK1-FOXO3 activation and antioxidant gene transcription. These findings demonstrated the antioxidant and anti-inflammatory effects of UA and LE against hyperglycemia-induced hepatic inflammation. Furthermore, we speculate that the LKB1/AMPK/FOXO3 pathway is a potential target responsible for these beneficial effects of LE and UA.
Collapse
Affiliation(s)
- Min Kyung Hyun
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro, 63 Beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea.,Department of Medicinal Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration, Eumseong, 369-873, Republic of Korea
| | - Dae Hyun Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro, 63 Beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea
| | - Chan Hum Park
- Department of Medicinal Crop Research, National Institute of Horticultural and Herbal Science, Rural Development Administration, Eumseong, 369-873, Republic of Korea
| | - Sang Gyun Noh
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro, 63 Beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea
| | - Sihyun Choi
- Department of Biopharmaceutical Engineering, College of Science and Technology, Dongguk University Gyeongju, 123 Dongdaero, Gyeongju, 38066, Gyeongbuk, Republic of Korea
| | - Jae Yong Lee
- Department of Biopharmaceutical Engineering, College of Science and Technology, Dongguk University Gyeongju, 123 Dongdaero, Gyeongju, 38066, Gyeongbuk, Republic of Korea
| | - Ji Hye Choi
- Department of Biopharmaceutical Engineering, College of Science and Technology, Dongguk University Gyeongju, 123 Dongdaero, Gyeongju, 38066, Gyeongbuk, Republic of Korea
| | - Duhyeon Park
- Department of Biopharmaceutical Engineering, College of Science and Technology, Dongguk University Gyeongju, 123 Dongdaero, Gyeongju, 38066, Gyeongbuk, Republic of Korea
| | - Yeon Ja Choi
- Department of Biopharmaceutical Engineering, College of Science and Technology, Dongguk University Gyeongju, 123 Dongdaero, Gyeongju, 38066, Gyeongbuk, Republic of Korea.
| | - Hae Young Chung
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro, 63 Beon-gil, Geumjeong-gu, Busan, 46241, Republic of Korea.
| |
Collapse
|
8
|
Chen XF, Wang Y, Ji S, Sun X, Feng Q, Yu H, Liu C. Hepatoprotective Efficacy and Interventional Mechanism of Qijia Rougan Decoction in Liver Fibrosis. Front Pharmacol 2022; 13:911250. [PMID: 35846987 PMCID: PMC9283647 DOI: 10.3389/fphar.2022.911250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/24/2022] [Indexed: 12/26/2022] Open
Abstract
Liver fibrosis is a leading contributor to chronic liver diseases such as cirrhosis and liver cancer, which pose a serious health threat worldwide, and there are no effective drugs to treat it. Qijia Rougan decoction was modified from Sanjiasan, a traditional Chinese medicine (TCM) described in the “Wenyilun” manuscript. Qijia Rougan decoction possesses hepatoprotective and antifibrotic effects for clinical applications. However, its underlying mechanisms remain largely unknown. In this study, fibrotic rats induced by carbon tetrachloride (CCl4) were treated with two doses of Qijia Rougan decoction. Histopathological and serum biochemical analyses were carried out to assess liver structure and function, respectively. High-performance liquid chromatography (HPLC) coupled with mass spectrometry (MS) was performed to identify bioactive compositions in Qijia Rougan decoction. Transcriptome analysis using mRNA-sequencing (mRNA-Seq) was used to explore the underlying mechanisms and verified by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. Qijia Rougan decoction significantly attenuated CCl4-induced hepatic fibrotic injury, supported by promoted liver function and improved liver fibrosis. Eight main representative components originating from raw materials in the Qijia Rougan decoction were found to possess an antifibrotic role. Mechanistically, Qijia Rougan decoction regulated biological processes such as oxidation–reduction, fatty acid metabolism, cell adhesion, and transforming growth factor beta (TGFβ) signaling. We determined that Qijia Rougan decoction reversed the expression of inflammatory cytokines and inhibited the activation of fibrosis-related TGFβ signaling. It also reversed the deterioration of liver structure and function in rats induced by CCl4. Overall, Qijia Rougan decoction significantly mediated metabolism-associated processes, inhibited inflammatory reactions, and repressed fibrosis-related TGFβ signaling, which prevented liver fibrosis deterioration. Our study deepens our understanding of TCM in the diagnosis and treatment of liver fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Han Yu
- *Correspondence: Han Yu, ; Chao Liu,
| | - Chao Liu
- *Correspondence: Han Yu, ; Chao Liu,
| |
Collapse
|
9
|
Song Y, Zhao F, Ma W, Li G. Hotspots and trends in liver kinase B1 research: A bibliometric analysis. PLoS One 2021; 16:e0259240. [PMID: 34735498 PMCID: PMC8568265 DOI: 10.1371/journal.pone.0259240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 10/15/2021] [Indexed: 11/29/2022] Open
Abstract
Introduction In the past 22 years, a large number of publications have reported that liver kinase B1 (LKB1) can regulate a variety of cellular processes and play an important role in many diseases. However, there is no systematic bibliometric analysis on the publications of LKB1 to reveal the research hotspots and future direction. Methods Publications were retrieved from the Web of Science Core Collection (WoSCC), Scopus, and PubMed databases. CiteSpace and VOSviewer were used to analysis the top countries, institutions, authors, source journals, discipline categories, references, and keywords. Results In the past 22 years, the number of LKB1 publications has increased gradually by year. The country, institution, author, journals that have published the most articles and cited the most frequently were the United States, Harvard University, Prof. Benoit Viollet, Journal of Biochemistry and Plos One. The focused research hotspot was the molecular functions of LKB1. The emerging hotspots and future trends are the clinical studies about LKB1 and co-mutated genes as biomarkers in tumors, especially in lung adenocarcinoma. Conclusions Our research could provide knowledge base, frontiers, emerging hotspots and future trends associated with LKB1 for researchers in this field, and contribute to finding potential cooperation possibilities.
Collapse
Affiliation(s)
- Yaowen Song
- Department of Radiotherapy Oncology, The First Affiliated Hospital of China Medical University, Shenyan, China
| | - Fangkun Zhao
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Ma
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Guang Li
- Department of Radiotherapy Oncology, The First Affiliated Hospital of China Medical University, Shenyan, China
- * E-mail:
| |
Collapse
|
10
|
Cloves Regulate Na +-K +-ATPase to Exert Antioxidant Effect and Inhibit UVB Light-Induced Skin Damage in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5197919. [PMID: 34471465 PMCID: PMC8405327 DOI: 10.1155/2021/5197919] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/10/2021] [Indexed: 12/15/2022]
Abstract
The purpose of this study was to observe the effect of cloves (Syzygium aromaticum (L.) Merr. & L.M. Perry) on the mouse skin using a UVB-induced skin injury mouse model. The serum, liver, and skin indexes of mice were determined by kits, H&E tissue staining, and qPCR assay. The compound composition of cloves was determined by HPLC. The results showed that cloves increased the activity of Na+-K+-ATPase in the skin and then maintained the sodium and potassium pump in the damaged skin muscle membrane. Cloves alleviated the oxidative stress injury induced by UVB irradiation by normalizing the related oxidative stress indexes (T-SOD, CAT, AGEs, and H2O2) in serum and skin. Inhibition of the proinflammatory cytokines TNF-α, IL-1β, and IL-6 and increased activation of anti-inflammatory cytokines IL-4 and IL-10 occurred after treatment with cloves, which ultimately reduced the inflammatory damage to the body. Further results showed that cloves upregulate SOD1, SOD2, CAT, GSH, IL-10, IκB-α, AMPK, SIRT1, LKB1, PGC-1α, APPL1, and FoxO1 and downregulate NF-κB p65, TNF-α, IL-6, and mTOR mRNA expression in the skin tissues of UVB-damaged mice. The results of composition analysis showed that the five most abundant compounds in cloves are rutin, isoquercitrin, ferulic acid, dihydroquercetin, and quercitrin. Cloves regulate the skin sarcomembrane Na+-K+-ATPase through these five compounds, and because they regulate the oxidation, inflammation, and ATP energy consumption of the body, they subsequently protect the skin from UVB damage.
Collapse
|
11
|
Li SX, Li C, Pang XR, Zhang J, Yu GC, Yeo AJ, Lavin MF, Shao H, Jia Q, Peng C. Metformin Attenuates Silica-Induced Pulmonary Fibrosis by Activating Autophagy via the AMPK-mTOR Signaling Pathway. Front Pharmacol 2021; 12:719589. [PMID: 34434111 PMCID: PMC8381252 DOI: 10.3389/fphar.2021.719589] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
Long-term exposure to crystalline silica particles leads to silicosis characterized by persistent inflammation and progressive fibrosis in the lung. So far, there is no specific treatment to cure the disease other than supportive care. In this study, we examined the effects of metformin, a prescribed drug for type || diabetes on silicosis and explored the possible mechanisms in an established rat silicosis model in vivo, and an in vitro co-cultured model containing human macrophages cells (THP-1) and human bronchial epithelial cells (HBEC). Our results showed that metformin significantly alleviated the inflammation and fibrosis of lung tissues of rats exposed to silica particles. Metformin significantly reduced silica particle-induced inflammatory cytokines including transforming growth factor-β1 (TGF-β1), tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) in rat lung tissue and HBEC culture supernatant. The protein levels of Vimentin and α-smooth muscle actin (α-SMA) were significantly decreased by metfomin while expression level of E-cadherin (E-Cad) increased. Besides, metformin increased the expression levels of phosphorylated adenosine 5′-monophosphate (AMP)-activated protein kinase (p-AMPK), microtubule-associated protein (MAP) light chain 3B (LC3B) and Beclin1 proteins, and reduced levels of phosphorylated mammalian target of rapamycin (p-mTOR) and p62 proteins in vivo and in vitro. These results suggest that metformin could inhibit silica-induced pulmonary fibrosis by activating autophagy through the AMPK-mTOR pathway.
Collapse
Affiliation(s)
- Shu-Xian Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Chao Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xin-Ru Pang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Juan Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Gong-Chang Yu
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Abrey J Yeo
- University of Queensland Centre for Clinical Research (UQCCR), Brisbane, QLD, Australia
| | - Martin F Lavin
- University of Queensland Centre for Clinical Research (UQCCR), Brisbane, QLD, Australia
| | - Hua Shao
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Cheng Peng
- Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
12
|
Ursolic Acid and Related Analogues: Triterpenoids with Broad Health Benefits. Antioxidants (Basel) 2021; 10:antiox10081161. [PMID: 34439409 PMCID: PMC8388988 DOI: 10.3390/antiox10081161] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/04/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022] Open
Abstract
Ursolic acid (UA) is a well-studied natural pentacyclic triterpenoid found in herbs, fruit and a number of traditional Chinese medicinal plants. UA has a broad range of biological activities and numerous potential health benefits. In this review, we summarize the current data on the bioavailability and pharmacokinetics of UA and review the literature on the biological activities of UA and its closest analogues in the context of inflammation, metabolic diseases, including liver and kidney diseases, obesity and diabetes, cardiovascular diseases, cancer, and neurological disorders. We end with a brief overview of UA’s main analogues with a special focus on a newly discovered naturally occurring analogue with intriguing biological properties and potential health benefits, 23-hydroxy ursolic acid.
Collapse
|
13
|
Wang H, Wang Z, Wang Y, Li X, Yang W, Wei S, Shi C, Qiu J, Ni M, Rao J, Cheng F. miRNA-130b-5p promotes hepatic stellate cell activation and the development of liver fibrosis by suppressing SIRT4 expression. J Cell Mol Med 2021; 25:7381-7394. [PMID: 34272822 PMCID: PMC8335697 DOI: 10.1111/jcmm.16766] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/07/2021] [Accepted: 06/15/2021] [Indexed: 12/14/2022] Open
Abstract
Liver fibrosis is a progressive disease accompanied by the deposition of extracellular matrix (ECM). Numerous reports have demonstrated that alterations in the expression of microRNAs (miRNAs) are related to liver disease. However, the effect of individual miRNAs on liver fibrosis has not been studied. Hepatic stellate cells (HSCs), being responsible for producing ECM, exert an important influence on liver fibrosis. Then, microarray analysis of non‐activated and activated HSCs induced by transforming growth factor β1 (TGF‐β1) showed that miR‐130b‐5p expression was strongly up‐regulated during HSC activation. Moreover, the progression of liver fibrosis had a close connection with the expression of miR‐130b‐5p in different liver fibrosis mouse models. Then, we identified that there were specific binding sites between miR‐130b‐5p and the 3′ UTR of Sirtuin 4 (SIRT4) via a luciferase reporter assay. Knockdown of miR‐130b‐5p increased SIRT4 expression and ameliorated liver fibrosis in mice transfected with antagomiR‐130b‐5p oligos. In general, our results suggested that miR‐130b‐5p promoted HSC activation by targeting SIRT4, which participates in the AMPK/TGF‐β/Smad2/3 signalling pathway. Hence, regulating miR‐130b‐5p maybe serve as a crucial therapeutic treatment for hepatic fibrosis.
Collapse
Affiliation(s)
- Hao Wang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Living Donor Liver Transplantation, National Health Commission (NHC), Nanjing, China
| | - Zeng Wang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Living Donor Liver Transplantation, National Health Commission (NHC), Nanjing, China
| | - Yirui Wang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Living Donor Liver Transplantation, National Health Commission (NHC), Nanjing, China
| | - Xiangcheng Li
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Living Donor Liver Transplantation, National Health Commission (NHC), Nanjing, China
| | - Wenjie Yang
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Living Donor Liver Transplantation, National Health Commission (NHC), Nanjing, China
| | - Song Wei
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Living Donor Liver Transplantation, National Health Commission (NHC), Nanjing, China.,School of Medical, Southeast University, Nanjing, China
| | - Chengyu Shi
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Living Donor Liver Transplantation, National Health Commission (NHC), Nanjing, China
| | - Jiannan Qiu
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Living Donor Liver Transplantation, National Health Commission (NHC), Nanjing, China
| | - Ming Ni
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Living Donor Liver Transplantation, National Health Commission (NHC), Nanjing, China
| | - Jianhua Rao
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Living Donor Liver Transplantation, National Health Commission (NHC), Nanjing, China
| | - Feng Cheng
- Hepatobiliary/Liver Transplantation Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China.,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China.,Key Laboratory of Living Donor Liver Transplantation, National Health Commission (NHC), Nanjing, China
| |
Collapse
|
14
|
Tian S, Yang X, Wang J, Luo J, Guo H. 1,25-(OH) 2D 3 ameliorates renal interstitial fibrosis in UUO rats through the AMPKα/mTOR pathway. J Int Med Res 2021; 49:300060520981360. [PMID: 33530801 PMCID: PMC7871069 DOI: 10.1177/0300060520981360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Objective To investigate the effects of 1,25(OH)2D3 on renal fibrosis associated with the AMP-activated protein kinase (AMPK)α/mechanistic target of rapamycin (mTOR) signalling pathway in a rat model of unilateral ureteral obstruction (UUO). Methods A total of 54 male Sprague Dawley rats were randomly divided into three groups: sham-operation group, UUO group, and UUO plus calcitriol (3 ng/100 g) group. Renal tissue was excised for histological examination by immunohistochemistry and Western blot, and for gene expression analysis using real-time polymerase chain reaction. Results 1,25(OH)2D3 enhanced AMPKα levels, inhibited mTOR levels and slowed the development of interstitial fibrosis in kidney tissue. Compared with the UUO plus calcitriol group, UUO rats demonstrated more severe renal damage characterized by marked tubular atrophy, interstitial fibrosis and significant induction of fibrogenic transforming growth factor-β1 and increased extra-cellular matrix proteins (α-smooth muscle actin and collagen type III), and decreased E-cadherin. Conclusion Treatment with 1,25(OH)2D3 altered the AMPKα/mTOR signalling pathway to suppress excessive fibroblast activation observed in UUO rats. This may serve as a novel mechanism to ameliorate renal dysfunction and fibrotic lesions.
Collapse
Affiliation(s)
- Shasha Tian
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaopeng Yang
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jianwu Wang
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jing Luo
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hui Guo
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.,Department of Nephrology, Shenzhen Baoan Shiyan People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
15
|
Ren C, Kong D, Ning C, Xing H, Cheng Y, Zhang Y, Lu Y, Li N, Chen X, Zhao D. Improved Pharmacokinetic Characteristics of Ursolic Acid in Rats Following Intratracheal Instillation and Nose-Only Inhalation Exposure. J Pharm Sci 2021; 110:905-913. [PMID: 33049258 DOI: 10.1016/j.xphs.2020.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/16/2022]
Abstract
Ursolic acid (UA) is a common pentacyclic triterpene phytochemical with various pharmacological activities. However, UA is classified as a class IV drug in BCS system and its development as an oral drug is limited. Pulmonary delivery is an effective way to improve the bioavailability of drugs with low absorption. In this study, the differences in pharmacokinetic behaviors of UA after pulmonary and oral administration was explored in rats. Compared with oral administration, the plasma concentration of UA increased rapidly after pulmonary administration, and the bioavailability increased about 80 times. UA instantly accumulated in the lungs after pulmonary administration, and the pulmonary AUC0-t/dose increased by 114 times compared to oral dosing. Incubation experiments showed that the metabolism of UA in rat lung microsomes was significantly reduced compared with that in liver microsomes, in which the clearance rate of phase I and phase II metabolism was reduced by 14.7 times and 1.4 times respectively. These results indicated that pulmonary administration could improve the bioavailability of UA and reduce its metabolism. This study not only provides a preferable route of administration for the application of UA but also offers new insights for the development of phytochemical drug candidates with poor pharmacokinetic properties.
Collapse
Affiliation(s)
- Chang Ren
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Dexuan Kong
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Chen Ning
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Han Xing
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yujie Cheng
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yongjie Zhang
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yang Lu
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ning Li
- National Experimental Teaching Demonstration Center of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xijing Chen
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Di Zhao
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
16
|
Acín S, Muñoz DL, Guillen A, Soscue D, Castaño A, Echeverri F, Balcazar N. Triterpene-enriched fractions from Eucalyptus tereticornis ameliorate metabolic alterations in a mouse model of diet-induced obesity. JOURNAL OF ETHNOPHARMACOLOGY 2021; 265:113298. [PMID: 32860892 DOI: 10.1016/j.jep.2020.113298] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 08/11/2020] [Accepted: 08/16/2020] [Indexed: 06/11/2023]
Abstract
ETNOPHARMACOLOGICAL RELEVANCE Eucalyptus tereticornis Sm. (Eu) is a plant species used in traditional medicine to treat diabetes mellitus. Eu leaf extracts have been shown to regulate immuno-metabolic activities that are associated with obesity and insulin resistance. OBE100 and OBE104 are two natural Eu extracts that are rich in pentacyclic triterpenes. The major compounds identified in OBE100 are ursolic acid (UA), oleanolic acid (OA), and ursolic acid lactone (UAL), and the major compounds identified in OBE104 are UA and OA. AIM OF THE STUDY This study aimed to investigate the effects of two extracts from Eu leaves with different triterpene composition in a nutritional animal model of prediabetes. METHODS A mouse model of diet-induced obesity was used to analyze the effects of the OBE100 and OBE104 treatments on metabolic markers and gene expression in liver and visceral adipose tissue. RESULTS Treating the prediabetic mouse model with OBE100 and OBE104 increased glucose tolerance. However, only the Eu extract that contained three triterpenes reduced mouse body weight, hepatic and adipose fat content, and plasma lipid levels. OBE100 treatment also led to decreased hepatic mRNA levels of PPARA, CPT1A, and SERBP1. In visceral adipose tissue, OBE100 treatment reduced expression of PPARA and ACACA and increased UCP1 expression. CONCLUSIONS These results suggest that developing a new multitargeting bioactive compound from the natural extract from Eu may help combat obesity and diabetes. Treatment with OBE100 had better effects than OBE104 in a diet-induced obesity mouse model, suggesting that the OBE100 extract, which contains three triterpenes, may be beneficial in combating obesity.
Collapse
Affiliation(s)
- Sergio Acín
- Molecular Genetics Group, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia; Department of Physiology and Biochemistry, School of Medicine, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia
| | - Diana Lorena Muñoz
- Department of Physiology and Biochemistry, School of Medicine, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia
| | - Alis Guillen
- Molecular Genetics Group, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia
| | - Duberney Soscue
- Molecular Genetics Group, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia
| | - Adriana Castaño
- Group of Organic Natural Product Chemistry, Faculty of Natural and Exact Sciences, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia
| | - Fernando Echeverri
- Group of Organic Natural Product Chemistry, Faculty of Natural and Exact Sciences, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia
| | - Norman Balcazar
- Molecular Genetics Group, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia; Department of Physiology and Biochemistry, School of Medicine, Universidad de Antioquia, Calle 70, N° 52-21, A.A, 1226, Medellin, Colombia.
| |
Collapse
|
17
|
Naß J, Efferth T. Ursolic acid ameliorates stress and reactive oxygen species in C. elegans knockout mutants by the dopamine Dop1 and Dop3 receptors. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 81:153439. [PMID: 33352493 DOI: 10.1016/j.phymed.2020.153439] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/16/2020] [Accepted: 12/06/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Depression and stress-related disorders are leading causes of death worldwide. Standard treatments elevating serotonin or noradrenaline levels are not sufficiently effective and cause adverse side effects. A connection between dopamine pathways and stress-related disorders has been suggested. Compounds derived from herbal medicine could be a promising alternative. We examined the neuroprotective effects of ursolic acid (UA) by focusing on dopamine signalling. METHODS Trolox equivalent capacity assay was used to determine the antioxidant activities of UA in vitro. C. elegans N2 wildtype and dopamine receptor-knockout mutants (dop1-deficient RB665 and dop3-deficient LX703 strains) were used as in vivo models. H2DCFDA and acute juglone assays were applied to determine the antioxidant activity in dependency of dopamine pathways in vivo. Stress was assessed by heat and acute osmotic stress assays. The influence of UA on overall survival was analyzed by a life span assay. The dop1 and dop3 mRNA expression was determined by real time RT-PCR. We also examined the binding affinity of UA towards C. elegans Dop1 and Dop3 receptors as well as human dopamine receptors D1 and D3 by molecular docking. RESULTS Antioxidant activity assays showed that UA exerts strong antioxidant activity. UA increased resistance towards oxidative, osmotic and heat stress. Additionally, UA increased life span of nematodes. Moreover, dop1 and dop3 gene expression was significantly enhanced upon UA treatment. Docking analysis revealed stronger binding affinity of UA to C. elegans and human dopamine receptors than the natural ligand, dopamine. Binding to Dop1 was stronger than to Dop3. CONCLUSION UA reduced stress-dependent ROS generation and acted through Dop1 and to a lesser extent through Dop3 to reduce stress and prolong life span in C. elegans. These results indicate that UA could be a promising lead compound for the development of new antidepressant medications.
Collapse
MESH Headings
- Animals
- Antioxidants/pharmacology
- Caenorhabditis elegans/drug effects
- Caenorhabditis elegans/genetics
- Caenorhabditis elegans/physiology
- Caenorhabditis elegans Proteins/chemistry
- Caenorhabditis elegans Proteins/genetics
- Caenorhabditis elegans Proteins/metabolism
- Dopamine/metabolism
- Gene Knockout Techniques
- Humans
- Longevity/drug effects
- Molecular Docking Simulation
- Mutation
- Reactive Oxygen Species/metabolism
- Receptors, Dopamine D1/chemistry
- Receptors, Dopamine D1/genetics
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D2/chemistry
- Receptors, Dopamine D2/genetics
- Receptors, Dopamine D2/metabolism
- Receptors, Dopamine D3/chemistry
- Receptors, Dopamine D3/metabolism
- Signal Transduction/drug effects
- Stress, Physiological/drug effects
- Stress, Physiological/genetics
- Triterpenes/chemistry
- Triterpenes/pharmacology
- Ursolic Acid
Collapse
Affiliation(s)
- Janine Naß
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany.
| |
Collapse
|
18
|
Lee NR, Meng RY, Rah SY, Jin H, Ray N, Kim SH, Park BH, Kim SM. Reactive Oxygen Species-Mediated Autophagy by Ursolic Acid Inhibits Growth and Metastasis of Esophageal Cancer Cells. Int J Mol Sci 2020; 21:E9409. [PMID: 33321911 PMCID: PMC7764507 DOI: 10.3390/ijms21249409] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Ursolic acid (UA) possesses various pharmacological activities, such as antitumorigenic and anti-inflammatory effects. In the present study, we investigated the mechanisms underlying the effects of UA against esophageal squamous cell carcinoma (ESCC) (TE-8 cells and TE-12 cells). The cell viability assay showed that UA decreased the viability of ESCC in a dose-dependent manner. In the soft agar colony formation assay, the colony numbers and size were reduced in a dose-dependent manner after UA treatment. UA caused the accumulation of vacuoles and LC3 puncta, a marker of autophagosome, in a dose-dependent manner. Autophagy induction was confirmed by measuring the expression levels of LC3 and p62 protein in ESCC cells. UA increased LC3-II protein levels and decreased p62 levels in ESCC cells. When autophagy was hampered using 3-methyladenine (3-MA), the effect of UA on cell viability was reversed. UA also significantly inhibited protein kinase B (Akt) activation and increased p-Akt expression in a dose-dependent manner in ESCC cells. Accumulated LC3 puncta by UA was reversed after wortmannin treatment. LC3-II protein levels were also decreased after treatment with Akt inhibitor and wortmannin. Moreover, UA treatment increased cellular reactive oxygen species (ROS) levels in ESCC in a time- and dose-dependent manner. Diphenyleneiodonium (an ROS production inhibitor) blocked the ROS and UA induced accumulation of LC3-II levels in ESCC cells, suggesting that UA-induced cell death and autophagy are mediated by ROS. Therefore, our data indicate that UA inhibits the growth of ESCC cells by inducing ROS-dependent autophagy.
Collapse
Affiliation(s)
- Na-Ri Lee
- Division of Hematology and Oncology, Jeonbuk National University Medical School, Jeonju 54907, Korea;
- Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju 54907, Korea;
- Research Institute of Clinical Medicine, Biomedical Research Institute of Jeonbuk National University Medical School, Jeonju 54907, Korea
| | - Ruo Yu Meng
- Department of Physiology and Institute of Medical Science, Jeonbuk National University Medical School, Jeonju 54907, Korea; (R.Y.M.); (N.R.)
| | - So-Young Rah
- Department of Biochemistry, Jeonbuk National University Medical School, Jeonju 54907, Korea; (S.-Y.R.); (B.H.P.)
| | - Hua Jin
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China;
| | - Navin Ray
- Department of Physiology and Institute of Medical Science, Jeonbuk National University Medical School, Jeonju 54907, Korea; (R.Y.M.); (N.R.)
| | - Seong-Hun Kim
- Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju 54907, Korea;
| | - Byung Hyun Park
- Department of Biochemistry, Jeonbuk National University Medical School, Jeonju 54907, Korea; (S.-Y.R.); (B.H.P.)
| | - Soo Mi Kim
- Department of Physiology and Institute of Medical Science, Jeonbuk National University Medical School, Jeonju 54907, Korea; (R.Y.M.); (N.R.)
| |
Collapse
|
19
|
Therapeutic effects of Chlorella vulgaris on carbon tetrachloride induced liver fibrosis by targeting Hippo signaling pathway and AMPK/FOXO1 axis. Mol Biol Rep 2020; 48:117-126. [PMID: 33296068 DOI: 10.1007/s11033-020-05978-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 11/03/2020] [Indexed: 12/27/2022]
Abstract
This study was conducted to present the mechanism of the therapeutic effects of Chlorella vulgaris extract (CV) on the carbon tetrachloride (CCl4) induced liver fibrosis model. Primarily, the mechanism of antioxidant effects of CV were investigated via measuring the expression of forkhead box protein O1 (FOXO1) and phosphorylated 5' adenosine monophosphate-activated protein kinase (p-AMPK) as upstream regulators of superoxide dismutase (SOD) and catalase (CAT). Subsequently, we investigated the regulatory effect of CV treatment on the yes-associated protein (YAP) and transcriptional coactivators with a PDZ-binding motif (TAZ) as fibrogenic factors. Male Wistar rats received CCl4 and olive oil solution 1 ml/kg intraperitoneally for 12 weeks, twice weekly. CV 50 and 100 mg/kg were administered on a daily basis by gavage in the last 4 weeks. Ultimately, liver marker enzymes and hepatic hydroxyproline content were measured. The activity of SOD and CAT and the expression of YAP, TAZ, FOXO1, SOD, and CAT were analyzed. Finally, the protein levels of YAP, TAZ, and p-AMPK were detected. CV administration decreased liver marker enzymes and hydroxyproline content significantly. The expression and protein levels of YAP and TAZ reduced by CV treatment. Furthermore, the augmentation of expression and function of CAT and SOD by CV treatment was followed by an increase in the expression of FOXO1 and protein level of p-AMPK. Our data revealed that the stimulation of expression and function of SOD and CAT by CV treatment could be mediated by FOXO1/p-AMPK axis. Moreover, anti-fibrotic effect of CV might be associated with its inhibitory effect on the hepatic expression of YAP and TAZ. Chlorella vulgaris treatment ameliorates liver fibrosis via two cellular mechanisms. A) Likely, Chlorella vulgaris treatment increases gene expression of enzymatic antioxidants superoxide dismutase (SOD) and catalase (CAT) via upregulating its upstream regulatory elements i.e. phosphorylated 5' adenosine monophosphate-activated protein kinase (p-AMPK) and forkhead box protein O1 (FOXO1). These possible regulatory effects maybe lead to reduce reactive oxygen species level (ROS). B) Chlorella vulgaris treatment decreases hepatic protein level and gene expression of key elements of Hippo signaling pathway i.e. Yes-associated protein (YAP) and Transcriptional coactivators with a PDZ-binding motif (TAZ). Figure created with BioRender ( https://biorender.com ). ROS: Reactive oxygen species, YAP: Yes-associated protein, TAZ: Transcriptional coactivators with a PDZ-binding motif, FOXO1: Fork head Box O1, AMPK: 5' adenosine monophosphate activated protein kinase, SOD: Superoxide dismutase, CAT: Catalase, P: Phosphate group.
Collapse
|
20
|
Synthesis and anti-inflammatory activity of saponin derivatives of δ-oleanolic acid. Eur J Med Chem 2020; 209:112932. [PMID: 33131725 DOI: 10.1016/j.ejmech.2020.112932] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/04/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023]
Abstract
Pentacyclic triterpenes (PTs) are the active ingredients of many medicinal herbs and pharmaceutical formulations, and are well-known for their anti-inflammatory activity. On the other hand, anti-inflammatory effects of AMP-activated protein kinase (AMPK) have recently drawn much attention. In this study, we found that a variety of naturally occurring PTs sapogenins and saponins could stimulate the phosphorylation of AMPK, and identified δ-oleanolic acid (10) as a potent AMPK activator. Based on these findings, 23 saponin derivatives of δ-oleanolic acid were synthesized in order to find more potent anti-inflammatory agents with improved pharmacokinetic properties. The results of cellular assays showed that saponin 29 significantly inhibited LPS-induced secretion of pro-inflammatory factors TNF-α and IL-6 in THP1-derived macrophages. Preliminary mechanistic studies showed that 29 stimulated the phosphorylation of AMPK and acetyl-CoA carboxylase (ACC). The bioavailability of 29 was significantly improved in comparison with its aglycon. More importantly, 29 showed significant anti-inflammatory and liver-protective effects in LPS/D-GalN-induced fulminant hepatic failure mice. Taken together, PTs saponins hold promise as therapeutic agents for inflammatory diseases.
Collapse
|
21
|
Zhao P, Saltiel AR. From overnutrition to liver injury: AMP-activated protein kinase in nonalcoholic fatty liver diseases. J Biol Chem 2020; 295:12279-12289. [PMID: 32651233 PMCID: PMC7443502 DOI: 10.1074/jbc.rev120.011356] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/24/2020] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver diseases (NAFLDs), especially nonalcoholic steatohepatitis (NASH), have become a major cause of liver transplant and liver-associated death. However, the pathogenesis of NASH is still unclear. Currently, there is no FDA-approved medication to treat this devastating disease. AMP-activated protein kinase (AMPK) senses energy status and regulates metabolic processes to maintain homeostasis. The activity of AMPK is regulated by the availability of nutrients, such as carbohydrates, lipids, and amino acids. AMPK activity is increased by nutrient deprivation and inhibited by overnutrition, inflammation, and hypersecretion of certain anabolic hormones, such as insulin, during obesity. The repression of hepatic AMPK activity permits the transition from simple steatosis to hepatocellular death; thus, activation might ameliorate multiple aspects of NASH. Here we review the pathogenesis of NAFLD and the impact of AMPK activity state on hepatic steatosis, inflammation, liver injury, and fibrosis during the transition of NAFL to NASH and liver failure.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Alan R Saltiel
- Department of Medicine, University of California San Diego, La Jolla, California, USA; Department of Pharmacology, University of California San Diego, La Jolla, California, USA.
| |
Collapse
|
22
|
McCallum ML, Pru CA, Smith AR, Kelp NC, Foretz M, Viollet B, Du M, Pru JK. A functional role for AMPK in female fertility and endometrial regeneration. Reproduction 2020; 156:501-513. [PMID: 30328345 DOI: 10.1530/rep-18-0372] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a highly conserved heterotrimeric complex that acts as an intracellular energy sensor. Based on recent observations of AMPK expression in all structures of the female reproductive system, we hypothesized that AMPK is functionally required for maintaining fertility in the female. This hypothesis was tested by conditionally ablating the two catalytic alpha subunits of AMPK, Prkaa1 and Prkaa2, using Pgr-cre mice. After confirming the presence of PRKAA1, PRKAA2 and the active phospho-PRKAA1/2 in the gravid uterus by immunohistochemistry, control (Prkaa1/2 fl/fl ) and double conditional knockout mice (Prkaa1/2 d/d ) were placed into a six-month breeding trial. While the first litter size was comparable between Prkaa1/2 fl/fl and Prkaa1/2 d/d female mice (P = 0.8619), the size of all subsequent litters was dramatically reduced in Prkaa1/2 d/d female mice (P = 0.0015). All Prkaa1/2 d/d female mice experienced premature reproductive senescence or dystocia by the fourth parity. This phenotype manifested despite no difference in estrous cycle length, ovarian histology in young and old nulliparous or multiparous animals, mid-gestation serum progesterone levels or uterine expression of Esr1 or Pgr between Prkaa1/2 fl/fl and Prkaa1/2 d/d female mice suggesting that the hypothalamic-pituitary-ovary axis remained unaffected by PRKAA1/2 deficiency. However, an evaluation of uterine histology from multiparous animals identified extensive endometrial fibrosis and disorganized stromal-glandular architecture indicative of endometritis, a condition that causes subfertility or infertility in most mammals. Interestingly, Prkaa1/2 d/d female mice failed to undergo artificial decidualization. Collectively, these findings suggest that AMPK plays an essential role in endometrial regeneration following parturition and tissue remodeling that accompanies decidualization.
Collapse
Affiliation(s)
- Melissa L McCallum
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Cindy A Pru
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Andrea R Smith
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Nicole C Kelp
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - Marc Foretz
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Benoit Viollet
- INSERM, U1016, Institut Cochin, Paris, France.,CNRS, UMR 8104, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Min Du
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| | - James K Pru
- Department of Animal Sciences, Center for Reproductive Biology, Washington State University, Pullman, Washington, USA
| |
Collapse
|
23
|
Kong DL, Kong FY, Liu XY, Yan C, Cui J, Tang RX, Zheng KY. Soluble egg antigen of Schistosoma japonicum induces pyroptosis in hepatic stellate cells by modulating ROS production. Parasit Vectors 2019; 12:475. [PMID: 31610797 PMCID: PMC6791022 DOI: 10.1186/s13071-019-3729-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 09/23/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Inflammation-induced dysfunction of hepatic stellate cells (HSCs) is involved in schistosomiasis-associated liver fibrosis, and soluble egg antigen (SEA) is a crucial pathogen-associated molecular pattern associated with liver injury in schistosomiasis. In addition, numerous studies have shown that caspase-1-mediated pyroptosis participates in the development of multiple inflammation-related diseases. However, whether pyroptotic cell death of HSCs is involved in SEA-mediated liver damage is not well understood. METHODS Primary cultured HSCs and Schistosoma japonicum-infected mouse liver tissue were analysed for histological changes and caspase-1 activation, and the role of pyroptosis in the mechanisms underlying SEA-induced HSC death was investigated. Accumulation of reactive oxygen species (ROS) in infected livers and SEA-stimulated HSCs was measured by flow cytometry and immunofluorescence. RESULTS Caspase-1 activity was elevated in both liver tissues and HSCs of S. japonicum-infected mice. Furthermore, SEA stimulation increased the proportion of pyroptotic HSCs, as shown by lactate dehydrogenase (LDH) release assays and by flow cytometric analysis of propidium iodide (PI) and caspase-1 double staining in cells. In addition, ROS generation was elevated in infected liver tissues and SEA-stimulated HSCs, and ROS inhibition downregulated SEA-induced caspase-1 activation and pyroptosis in HSCs. CONCLUSIONS Our present study demonstrates that pyroptotic cell death in HSCs induced by SEA via ROS-mediated caspase-1 activation may serve as a significant mechanism to initiate the inflammatory response and thereby exacerbate liver injury during S. japonicum infection.
Collapse
Affiliation(s)
- De-Long Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Fan-Yun Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Xiang-Ye Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Chao Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Jie Cui
- Department of Physiology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Ren-Xian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China.
| | - Kui-Yang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China.
| |
Collapse
|
24
|
Habtemariam S. Antioxidant and Anti-inflammatory Mechanisms of Neuroprotection by Ursolic Acid: Addressing Brain Injury, Cerebral Ischemia, Cognition Deficit, Anxiety, and Depression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8512048. [PMID: 31223427 PMCID: PMC6541953 DOI: 10.1155/2019/8512048] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/27/2019] [Accepted: 05/08/2019] [Indexed: 12/19/2022]
Abstract
Ursolic acid (UA) is a pentacyclic triterpene which is found in common herbs and medicinal plants that are reputed for a variety of pharmacological effects. Both as an active principle of these plants and as a nutraceutical ingredient, the pharmacology of UA in the CNS and other organs and systems has been extensively reported in recent years. In this communication, the antioxidant and anti-inflammatory axis of UA's pharmacology is appraised for its therapeutic potential in some common CNS disorders. Classic examples include the traumatic brain injury (TBI), cerebral ischemia, cognition deficit, anxiety, and depression. The pharmacological efficacy for UA is demonstrated through the therapeutic principle of one drug → multitargets → one/many disease(s). Both specific enzymes and receptor targets along with diverse pharmacological effects associated with oxidative stress and inflammatory signalling are scrutinised.
Collapse
Affiliation(s)
- Solomon Habtemariam
- Pharmacognosy Research Laboratories & Herbal Analysis Services, University of Greenwich, Central Avenue, Chatham-Maritime, Kent ME4 4TB, UK
| |
Collapse
|
25
|
AMPK agonist AICAR ameliorates portal hypertension and liver cirrhosis via NO pathway in the BDL rat model. J Mol Med (Berl) 2019; 97:423-434. [PMID: 30721324 PMCID: PMC6394556 DOI: 10.1007/s00109-019-01746-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 01/07/2019] [Accepted: 01/16/2019] [Indexed: 02/06/2023]
Abstract
Recent studies have indicated that the Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) pathway is closely involved in liver fibrosis and other fibrotic diseases. However, whether targeting the AMPK pathway can rescue liver fibrosis and its complications, such as portal hypertension, is unknown. This study aimed to explore the therapeutic value of AICAR (5-aminoimidazole-4-carboxyamide ribonucleoside), an agonist of the AMPK pathway, on liver fibrosis and portal hypertension in bile duct ligation (BDL) rats. In vitro experiments showed that the gene expression levels of TGF-b, a-SMA, and collagen 1 in primary rat hepatic stellate cells (HSCs) were significantly decreased after AICAR treatment. The p-eNOS expression and nitric oxide (NO) production were increased by AICAR administration in sinusoidal endothelial cells (SECs). For in vivo animal studies, AICAR acutely decreased portal pressure in the BDL and CCL4 fibrotic rats, but not in the partial portal vein ligation (PVL) rats, without changes in systemic hemodynamics. It was also observed by using intravital fluorescence microscopy that AICAR led to sinusoidal vasodilation in situ experiment. We propose that the relevant mechanisms may be related to the activation of the AMPK/NO pathway in SECs and that this activation promoted NO production in the liver, thereby promoting hepatic sinusoid microcirculation and decreased intrahepatic resistance. The results were verified using the NO inhibitor L-NAME. Chronic AICAR treatment also showed profound beneficial effects on the BDL model rats. The hemodynamic condition was greatly improved, but the positive effect could be partially blocked by L-NAME. Moreover, AICAR also decreased hepatic fibrogenesis in the BDL rats. KEY MESSAGES: Acute and chronic use of AICAR could alleviate portal pressure without changing systemic hemodynamics. AICAR induced sinusoidal vasodilation by improving NO bioavailability and ameliorating endothelial dysfunction in vivo and in vitro. AICAR could alleviate liver cirrhosis in the BDL model rats.
Collapse
|
26
|
Tsogbadrakh B, Ryu H, Ju KD, Lee J, Yun S, Yu KS, Kim HJ, Ahn C, Oh KH. AICAR, an AMPK activator, protects against cisplatin-induced acute kidney injury through the JAK/STAT/SOCS pathway. Biochem Biophys Res Commun 2019; 509:680-686. [PMID: 30616891 DOI: 10.1016/j.bbrc.2018.12.159] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 12/23/2018] [Indexed: 12/19/2022]
Abstract
Cisplatin causes acute kidney injury (AKI) through proximal tubular injury. We investigated the protective effect of the adenosine monophosphate protein kinase (AMPK) activator 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) against cisplatin-induced AKI. We investigated whether the AMP-kinase activator AICAR ameliorates cisplatin-induced AKI through the JAK/STAT/SOCS pathway. Male Sprague-Dawley (SD) rats were randomly divided into four groups: control, AICAR, cisplatin, and cisplatin + AICAR. As appropriate to their treatment group, the rats were injected with a single dose of cisplatin (7 mg/kg, i.p.). AICAR was administered to the rats at 100 mg/kg i.p. daily. Blood urea nitrogen (BUN) and serum creatinine were measured. Renal damage was analyzed in sections stained with hematoxylin and eosin (H&E). Renal tissues were also examined by immunohistochemistry and western blot for p-AMPK, Kim-1, cleaved caspase 3, and JAK/STAT/SOCS. For in vitro studies, NRK-52E normal rat kidney cells were treated with cisplatin and/or AICAR. By western blot, we confirmed the expression of p-AMPK and the JAK/STAT/SOCS pathway in NRK-52E cells. AICAR was protective against cisplatin-induced acute tubular injury by up-regulating p-AMPK expression in NRK-52E cells. Protein expression levels of JAK2/STAT1 were markedly ameliorated in NRK-52E cells by AICAR. The protective mechanism of AICAR may be associated with suppression of the JAK2/STAT1 pathway and up-regulation of SOCS1, an inhibitor of the JAK2/STAT1 pathway. The present study demonstrates the protective effects of AICAR against cisplatin-induced AKI and shows a new renoprotective mechanism through the JAK2/STAT1/SOCS1 pathway and apoptosis inhibition. This study suggests that activation of the AMPK activator AICAR might ameliorate cisplatin-induced AKI.
Collapse
Affiliation(s)
- Bodokhsuren Tsogbadrakh
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Hyunjin Ryu
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyung Don Ju
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jinho Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sohyun Yun
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kyung-Sang Yu
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
| | - Hyo Jin Kim
- Department of Internal Medicine, Dongkuk University, Kyungju, Republic of Korea
| | - Curie Ahn
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea; Transplantation Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Grouix B, Sarra-Bournet F, Leduc M, Simard JC, Hince K, Geerts L, Blais A, Gervais L, Laverdure A, Felton A, Richard J, Ouboudinar J, Gagnon W, Leblond FA, Laurin P, Gagnon L. PBI-4050 Reduces Stellate Cell Activation and Liver Fibrosis through Modulation of Intracellular ATP Levels and the Liver Kinase B1/AMP-Activated Protein Kinase/Mammalian Target of Rapamycin Pathway. J Pharmacol Exp Ther 2018; 367:71-81. [PMID: 30093459 DOI: 10.1124/jpet.118.250068] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/12/2018] [Indexed: 12/30/2022] Open
Abstract
Hepatic fibrosis is a major cause of morbidity and mortality for which there is currently no effective therapy. We previously showed that 2-(3-pentylphenyl)acetic acid (PBI-4050) is a dual G protein-coupled receptor GPR40 agonist/GPR84 antagonist that exerts antifibrotic, anti-inflammatory, and antiproliferative action. We evaluated PBI-4050 for the treatment of liver fibrosis in vivo and elucidated its mechanism of action on human hepatic stellate cells (HSCs). The antifibrotic effect of PBI-4050 was evaluated in carbon tetrachloride (CCl4)- and bile duct ligation-induced liver fibrosis rodent models. Treatment with PBI-4050 suppressed CCl4-induced serum aspartate aminotransferase levels, inflammatory marker nitric oxide synthase, epithelial to mesenchymal transition transcription factor Snail, and multiple profibrotic factors. PBI-4050 also decreased GPR84 mRNA expression in CCl4-induced injury, while restoring peroxisome proliferator-activated receptor γ (PPARγ) to the control level. Collagen deposition and α-smooth muscle actin (α-SMA) protein levels were also attenuated by PBI-4050 treatment in the bile duct ligation rat model. Transforming growth factor-β-activated primary HSCs were used to examine the effect of PBI-4050 and its mechanism of action in vitro. PBI-4050 inhibited HSC proliferation by arresting cells in the G0/G1 cycle phase. Subsequent analysis demonstrated that PBI-4050 signals through a reduction of intracellular ATP levels, activation of liver kinase B1 (LKB1) and AMP-activated protein kinase (AMPK), and blockade of mammalian target of rapamycin (mTOR), resulting in reduced protein and mRNA levels of α-SMA and connective tissue growth factor and restored PPARγ mRNA expression. Our findings suggest that PBI-4050 may exert antifibrotic activity in the liver through a novel mechanism of action involving modulation of intracellular ATP levels and the LKB1/AMPK/mTOR pathway in stellate cells, and PBI-4050 may be a promising agent for treating liver fibrosis.
Collapse
Affiliation(s)
| | | | - Martin Leduc
- Prometic BioSciences Inc., Laval, Québec, Canada
| | | | - Kathy Hince
- Prometic BioSciences Inc., Laval, Québec, Canada
| | | | | | | | | | | | | | | | | | | | | | - Lyne Gagnon
- Prometic BioSciences Inc., Laval, Québec, Canada
| |
Collapse
|
28
|
Retraction: Berberine Improves Kidney Function in Diabetic Mice via AMPK Activation. PLoS One 2017; 12:e0190562. [PMID: 29284063 PMCID: PMC5746262 DOI: 10.1371/journal.pone.0190562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
29
|
A Novel Phenylchromane Derivative Increases the Rate of Glucose Uptake in L6 Myotubes and Augments Insulin Secretion from Pancreatic Beta-Cells by Activating AMPK. Pharm Res 2017; 34:2873-2890. [DOI: 10.1007/s11095-017-2271-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/24/2017] [Indexed: 01/04/2023]
|
30
|
Jiang S, Li T, Yang Z, Yi W, Di S, Sun Y, Wang D, Yang Y. AMPK orchestrates an elaborate cascade protecting tissue from fibrosis and aging. Ageing Res Rev 2017; 38:18-27. [PMID: 28709692 DOI: 10.1016/j.arr.2017.07.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 07/07/2017] [Accepted: 07/10/2017] [Indexed: 01/10/2023]
Abstract
Fibrosis is a common process characterized by excessive extracellular matrix (ECM) accumulation after inflammatory injury, which is also a crucial cause of aging. The process of fibrosis is involved in the pathogenesis of most diseases of the heart, liver, kidney, lung, and other organs/tissues. However, there are no effective therapies for this pathological alteration. Annually, fibrosis represents a huge financial burden for the USA and the world. 5'-AMP-activated protein kinase (AMPK) is a pivotal energy sensor that alleviates or delays the process of fibrogenesis. In this review, we first present basic background information on AMPK and fibrogenesis and describe the protective roles of AMPK in three fibrogenic phases. Second, we analyze the protective action of AMPK during fibrosis in myocardial, hepatic, renal, pulmonary, and other organs/tissues. Third, we present a comprehensive discussion of AMPK during fibrosis and draw a conclusion. This review highlights recent advances, vital for basic research and clinical drug design, in the regulation of AMPK during fibrosis.
Collapse
Affiliation(s)
- Shuai Jiang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Aerospace Medicine, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhi Yang
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Yang Sun
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, 127 Changle West Road, Xi'an 710032, China
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
31
|
Liang Z, Li T, Jiang S, Xu J, Di W, Yang Z, Hu W, Yang Y. AMPK: a novel target for treating hepatic fibrosis. Oncotarget 2017; 8:62780-62792. [PMID: 28977988 PMCID: PMC5617548 DOI: 10.18632/oncotarget.19376] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 07/08/2017] [Indexed: 12/19/2022] Open
Abstract
Fibrosis is a common process of excessive extracellular matrix (ECM) accumulation following inflammatory injury. Fibrosis is involved in the pathogenesis of almost all liver diseases for which there is no effective treatment. 5'-AMP-activated protein kinase (AMPK) is a cellular energy sensor that can ameliorate the process of hepatic fibrogenesis. Given the existing evidence, we first introduce the basic background of AMPK and hepatic fibrosis and the actions of AMPK in hepatic fibrosis. Second, we discuss the three phases of hepatic fibrosis and potential drugs that target AMPK. Third, we analyze possible anti-fibrosis mechanisms and other benefits of AMPK on the liver. Finally, we summarize and briefly explain the current objections to targeting AMPK. This review may aid clinical and basic research on AMPK, which may be a novel drug candidate for hepatic fibrosis.
Collapse
Affiliation(s)
- Zhenxing Liang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Tian Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an 710069, China.,Department of Biomedical Engineering, The Fourth Military Medical University, Xi'an 710032, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, Xi'an 710032, China
| | - Jing Xu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wencheng Di
- Department of Cardiology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Zhi Yang
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi'an 710032, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, Xi'an 710032, China
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi'an 710069, China.,Department of Biomedical Engineering, The Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
32
|
Liqustri lucidi Fructus inhibits hepatic injury and functions as an antioxidant by activation of AMP-activated protein kinase in vivo and in vitro. Chem Biol Interact 2017; 262:57-68. [DOI: 10.1016/j.cbi.2016.11.031] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 11/14/2016] [Accepted: 11/30/2016] [Indexed: 12/27/2022]
|
33
|
Taurine zinc solid dispersions enhance bile-incubated L02 cell viability and improve liver function by inhibiting ERK2 and JNK phosphorylation during cholestasis. Toxicology 2016; 366-367:10-9. [PMID: 27501764 DOI: 10.1016/j.tox.2016.07.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 07/20/2016] [Accepted: 07/25/2016] [Indexed: 12/31/2022]
Abstract
Dietary intakes of taurine and zinc are associated with decreased risk of liver disease. In this study, solid dispersions (SDs) of a taurine zinc complex on hepatic injury were examined in vitro using the immortalized human hepatocyte cell line L02 and in a rat model of bile duct ligation. Sham-operated and bile duct ligated Sprague-Dawley rats were treated with the vehicle alone or taurine zinc (40, 80, 160mg/kg) for 17days. Bile duct ligation significantly increased blood lipid levels, and promoted hepatocyte apoptosis, inflammation and compensatory biliary proliferation. In vitro, incubation with bile significantly reduced L02 cell viability; this effect was significantly attenuated by pretreatment with SP600125 (a JNK inhibitor) and enhanced when co-incubated with taurine zinc SDs. In vivo, administration of taurine zinc SDs decreased serum alanine aminotransferase and aspartate aminotransferase activities in a dose-dependent manner and attenuated the increases in serum total bilirubin, total cholesterol and low density lipoprotein cholesterol levels after bile duct ligation. Additionally, taurine zinc SDs downregulated the expression of interleukin-1β and inhibited the phosphorylation of Jun N-terminal kinase (JNK) and extracellular signal-regulated kinase2 (ERK2) in the liver after bile duct ligation. Moreover, taurine zinc SDs had more potent blood lipid regulatory and anti-apoptotic effects than the physical mixture of taurine and zinc acetate. Therefore, we speculate that taurine zinc SDs protect liver function at least in part via a mechanism linked to reduce phosphorylation of JNK and ERK2, which suppresses inflammation, apoptosis and cholangiocyte proliferation during cholestasis.
Collapse
|
34
|
Kang JW, Hong JM, Lee SM. Melatonin enhances mitophagy and mitochondrial biogenesis in rats with carbon tetrachloride-induced liver fibrosis. J Pineal Res 2016; 60:383-93. [PMID: 26882442 DOI: 10.1111/jpi.12319] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 02/09/2016] [Indexed: 12/14/2022]
Abstract
Liver fibrosis leads to liver cirrhosis and failure, and no effective treatment is currently available. Growing evidence supports a link between mitochondrial dysfunction and liver fibrogenesis and mitochondrial quality control-based therapy has emerged as a new therapeutic target. We investigated the protective mechanisms of melatonin against mitochondrial dysfunction-involved liver fibrosis, focusing on mitophagy and mitochondrial biogenesis. Rats were treated with carbon tetrachloride (CCl4) dissolved in olive oil (0.5 mL/kg, twice a week, i.p.) for 8 wk. Melatonin was administered orally at 2.5, 5, and 10 mg/kg once a day. Chronic CCl4 exposure induced collagen deposition, hepatocellular damage, and oxidative stress, and melatonin attenuated these increases. Increases in mRNA and protein expression levels of transforming growth factor β1 and α-smooth muscle actin in response to CCl4 were attenuated by melatonin. Melatonin attenuated hallmarks of mitochondrial dysfunction, such as mitochondrial swelling and glutamate dehydrogenase release. Chronic CCl4 exposure impaired mitophagy and mitochondrial biogenesis, and melatonin attenuated this impairment, as indicated by increases in mitochondrial DNA and in protein levels of PTEN-induced putative kinase 1 (PINK1); Parkin; peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC-1α); nuclear respiratory factor 1 (NRF1); and transcription factor A, mitochondrial (TFAM). CCl4-mediated decreases in mitochondrial fission- and fusion-related proteins, such as dynamin-related protein 1 (DRP1) and mitofusin 2, were also attenuated by melatonin. Moreover, melatonin induced AMP-activated protein kinase (AMPK) phosphorylation. These results suggest that melatonin protects against liver fibrosis via upregulation of mitophagy and mitochondrial biogenesis, and may be useful as an anti-fibrotic treatment.
Collapse
Affiliation(s)
- Jung-Woo Kang
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Korea
| | - Jeong-Min Hong
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Korea
| | - Sun-Mee Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Korea
| |
Collapse
|
35
|
Grahame Hardie D. Regulation of AMP-activated protein kinase by natural and synthetic activators. Acta Pharm Sin B 2016; 6:1-19. [PMID: 26904394 PMCID: PMC4724661 DOI: 10.1016/j.apsb.2015.06.002] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/28/2015] [Indexed: 12/11/2022] Open
Abstract
The AMP-activated protein kinase (AMPK) is a sensor of cellular energy status that is almost universally expressed in eukaryotic cells. While it appears to have evolved in single-celled eukaryotes to regulate energy balance in a cell-autonomous manner, during the evolution of multicellular animals its role has become adapted so that it also regulates energy balance at the whole body level, by responding to hormones that act primarily on the hypothalamus. AMPK monitors energy balance at the cellular level by sensing the ratios of AMP/ATP and ADP/ATP, and recent structural analyses of the AMPK heterotrimer that have provided insight into the complex mechanisms for these effects will be discussed. Given the central importance of energy balance in diseases that are major causes of morbidity or death in humans, such as type 2 diabetes, cancer and inflammatory disorders, there has been a major drive to develop pharmacological activators of AMPK. Many such activators have been described, and the various mechanisms by which these activate AMPK will be discussed. A particularly large class of AMPK activators are natural products of plants derived from traditional herbal medicines. While the mechanism by which most of these activate AMPK has not yet been addressed, I will argue that many of them may be defensive compounds produced by plants to deter infection by pathogens or grazing by insects or herbivores, and that many of them will turn out to be inhibitors of mitochondrial function.
Collapse
Affiliation(s)
- David Grahame Hardie
- Division of Cell Signaling & Immunology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, UK
| |
Collapse
|
36
|
Domitrović R, Potočnjak I. A comprehensive overview of hepatoprotective natural compounds: mechanism of action and clinical perspectives. Arch Toxicol 2015; 90:39-79. [DOI: 10.1007/s00204-015-1580-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 08/11/2015] [Indexed: 12/22/2022]
|
37
|
Chu X, He X, Shi Z, Li C, Guo F, Li S, Li Y, Na L, Sun C. Ursolic acid increases energy expenditure through enhancing free fatty acid uptake and β-oxidation via an UCP3/AMPK-dependent pathway in skeletal muscle. Mol Nutr Food Res 2015; 59:1491-503. [PMID: 25944715 DOI: 10.1002/mnfr.201400670] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Revised: 04/16/2015] [Accepted: 04/21/2015] [Indexed: 12/12/2022]
Abstract
SCOPE Ursolic acid (UA) is a triterpenoid compound with multifold biological functions. Our previous studies have reported that UA protects against high-fat diet-induced obesity and improves insulin resistance (IR). However, the potential mechanisms are still undefined. Free fatty acid (FFA) metabolism in skeletal muscle plays a central role in obesity and IR. Therefore, in this study, we investigated the effect and the potential mechanisms of UA on skeletal muscle FFA metabolism. METHODS AND RESULTS In diet-induced obese rats, 0.5% UA supplementation for 6 weeks markedly reduced body weight, increased energy expenditure, decreased FFA level in serum and skeletal muscle and triglyceride content in skeletal muscle. In vitro, the data provided directly evidence that UA significantly increased fluorescently labeled FFA uptake and (3) H-labeled palmitic acid β-oxidation. UA-activated AMP-activated protein kinase (AMPK) and downstream targets were involved in the increase of FFA catabolism. Moreover, upregulated uncoupling protein 3 (UCP3) by UA contributed to AMPK activation via elevating adenosine monophosphate/adenosine triphosphate ratio. CONCLUSION UA increases FFA burning through enhancing skeletal muscle FFA uptake and β-oxidation via an UCP3/AMPK-dependent pathway, which provides a novel perspective on the biological function of UA against obesity and IR.
Collapse
Affiliation(s)
- Xia Chu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Xuan He
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Zhiping Shi
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Chunjuan Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Fuchuan Guo
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Songtao Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Ying Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
- Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| | - Lixin Na
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, China
- Research Institute of Food, Nutrition and Health, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, China
| |
Collapse
|
38
|
Abstract
Numerous studies have found that NADPH oxidase (NOX) plays a key role in the development of hepatic fibrosis. The activation of NOX in hepatic stellate cells (HSCs) is followed by production of reactive oxygen species and activation of PI3K/Akt and P38MAPK signaling pathways, which in turn causes the excessive expression of type I collagen and its deposition in the liver. Ursolic acid (UA) is a Chinese herbal monomer that widely exists in multiple medicinal plants. Studies have found that UA markedly ameliorates liver fibrosis in experimental animals, and reduces hepatocyte necrosis. In this paper, we review the recent advances in understanding the antifibrotic effect of UA in the liver and the related mechanisms.
Collapse
|
39
|
Abstract
Liver fibrosis is the pathological consequence of chronic liver diseases, where an excessive deposition of extracellular matrix (ECM) proteins occurs, concomitantly with the processes of repair and regeneration. It is characterized by increased production of matrix proteins, in particular collagens, and decreased matrix remodelling. The principal source of ECM accumulation is myofibroblasts (MFB). Most fibrogenic MFB are endogenous to the liver, coming from hepatic stellate cells (HSC) and portal fibroblasts. Dysregulated inflammatory responses have been associated with most (if not all) hepatotoxic insults and chronic oxidative stress play a role during the initial liver inflammatory phase and its progression to fibrosis. Redox-regulated processes are responsible for activation of HSC to MFB, as well as maintenance of the MFB function. Increased oxidative stress also induces hepatocyte apoptosis, which contributes to increase the liver injury and to transdifferentiate HSC to MFB, favouring the fibrogenic process. Mitochondria and other redox-active enzymes can generate superoxide and hydrogen peroxide as a by-product in liver cells. Moreover, accumulating evidence indicates that NADPH oxidases (NOXs), which play a critical role in the inflammatory response, may contribute to reactive oxygen species (ROS) production during liver fibrosis, being important players in HSC activation and hepatocyte apoptosis. Based on the knowledge of the pathogenic role of ROS, different strategies to prevent or reverse the oxidative damage have been developed to be used as therapeutic tools in liver fibrosis. This review will update all these concepts, highlighting the relevance of redox biology in chronic fibrogenic liver pathologies. Oxidative stress is a major cause for initiation/progression of liver fibrosis. Redox-regulated processes activate hepatic stellate cells to myofibroblasts. Increased oxidative stress induces hepatocyte apoptosis. NOX inhibitors are considered as a new strategy to prevent/reverse liver fibrosis. NADPH oxidases (NOX) have been involved in liver fibrogenic responses.
Collapse
Affiliation(s)
- Eva Crosas-Molist
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain
| | - Isabel Fabregat
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain; Department of Physiological Sciences II, University of Barcelona, L'Hospitalet, Barcelona, Spain.
| |
Collapse
|
40
|
Ma J, Yu H, Liu J, Chen Y, Wang Q, Xiang L. Metformin attenuates hyperalgesia and allodynia in rats with painful diabetic neuropathy induced by streptozotocin. Eur J Pharmacol 2015; 764:599-606. [PMID: 26054810 DOI: 10.1016/j.ejphar.2015.06.010] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 05/16/2015] [Accepted: 06/04/2015] [Indexed: 11/28/2022]
Abstract
Painful diabetic neuropathy is a common complication of diabetes mellitus, which often makes the patients suffer from severe hyperalgesia and allodynia. Thus far, the treatment of painful diabetic neuropathy remains unsatisfactory. Metformin, which is the first-line drug for type-2 diabetes, has been proved to attenuate hyperexcitability in sensory neurons linked to chemotherapy-induced neuropathic pain, highlighting its potential in alleviating pain related with painful diabetic neuropathy. The present study was designed to investigate the potential beneficial effect of metformin on hyperalgesia and allodynia in diabetic rats. The mechanical sensitivity, heat nociception, and cold allodynia were examined. The levels of malondialdehyde, superoxide dismutase, and advanced glycation end-products in the blood were measured. The expression of adenosine monophosphate-activated protein kinase (AMPK) phosphorylation and AMPK target genes were examined in the sciatic nerves of the animals. It was found that metformin was capable of attenuating diabetes-induced mechanical hyperalgesia, heat hyperalgesia and cold allodynia. In addition, metformin was capable of decreasing malondialdehyde and glycation end-products levels in blood, as well as increasing superoxide dismutas activity, indicating the inhibitory effect of metformin against diabetes-induced oxidative stress. Further studies showed that metformin could activate AMPK and increase the AMPK target genes in sciatic nerves in diabetic rats. In conclusion, metformin is able to attenuate diabetes-induced hyperalgesia and allodynia, which might be associated its anti-oxidative effect through AMPK pathway. Metformin might be used as an effective drug, especially with fewer side effects, for abnormal sensation in painful diabetic neuropathy.
Collapse
Affiliation(s)
- Junxiong Ma
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Rescue Center of Severe Wound and Trauma of Chinese PLA, Shenyang 110016, Liaoning, China
| | - Hailong Yu
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Rescue Center of Severe Wound and Trauma of Chinese PLA, Shenyang 110016, Liaoning, China
| | - Jun Liu
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Rescue Center of Severe Wound and Trauma of Chinese PLA, Shenyang 110016, Liaoning, China
| | - Yu Chen
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Rescue Center of Severe Wound and Trauma of Chinese PLA, Shenyang 110016, Liaoning, China
| | - Qi Wang
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Rescue Center of Severe Wound and Trauma of Chinese PLA, Shenyang 110016, Liaoning, China
| | - Liangbi Xiang
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Rescue Center of Severe Wound and Trauma of Chinese PLA, Shenyang 110016, Liaoning, China.
| |
Collapse
|
41
|
Jia Y, Kim S, Kim J, Kim B, Wu C, Lee JH, Jun HJ, Kim N, Lee D, Lee SJ. Ursolic acid improves lipid and glucose metabolism in high-fat-fed C57BL/6J mice by activating peroxisome proliferator-activated receptor alpha and hepatic autophagy. Mol Nutr Food Res 2015; 59:344-54. [PMID: 25418615 DOI: 10.1002/mnfr.201400399] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 10/16/2014] [Accepted: 10/17/2014] [Indexed: 12/19/2022]
Abstract
SCOPE This study investigated metabolic effects of ursolic acid (UA), a peroxisome proliferation-activated receptor (PPAR)-α activator, in vivo. METHODS AND RESULTS High-fat diet (HFD)-fed C57BL/6J mice were orally administered UA (50 or 200 mg/kg body weight) for 8 wk. UA reduced liver and adipose tissue mass, adipocyte size, and plasma leptin concentrations, plasma triglyceride and low-density-lipoprotein cholesterol concentrations, while it elevated the high-density-lipoprotein cholesterol and adiponectin concentrations significantly compared with controls. UA induced the expression of PPARα and its responsive genes involved in fatty acid uptake and β-oxidation in the livers, whereas genes involved in lipogenesis, including sterol regulatory element-binding proteins-1c, were downregulated. UA administration improved glucose tolerance and insulin sensitivity significantly compared with the HFD-fed control livers. UA administration also activated hepatic autophagy as assessed by the expression of microtubule-associated protein 1A/1B-light chain 3 (LC3)-II and other key proteins in the autophagy pathway. CONCLUSION Our findings suggest that UA ameliorates lipid and glucose metabolism in HFD-fed mice primarily by the activation of PPARα and induction of the hepatic autophagy pathway. Thus, intake of UA in the diet or in an isolated form may ameliorate lipid and glucose metabolism.
Collapse
Affiliation(s)
- Yaoyao Jia
- Department of Biotechnology, Graduate School of Life Sciences & Biotechnology, Korea University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|