1
|
Yu HC, Bai QR, Guo JJ, Chen MY, Wang L, Tong FC, Zhang SL, Wu J. Elucidating hydroxysafflor yellow A's multi-target mechanisms against alcoholic liver disease through integrative pharmacology. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155956. [PMID: 39216301 DOI: 10.1016/j.phymed.2024.155956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 07/09/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Alcoholic liver disease (ALD) significantly contributes to global liver-related morbidity and mortality. Natural products play a crucial role in the prevention and treatment of ALD. Hydroxysafflor yellow A (HSYA), a unique and primary component of Safflower (Carthamus tinctorius l.), exhibits diverse pharmacological activities. However, the impact and mechanism of HSYA on ALD have not been fully elucidated. PURPOSE The purpose of this study was to employ an integrative pharmacology approach to assess the multi-targeted mechanism of HSYA against ALD. METHODS Network pharmacology and molecular docking techniques were used to analyze the potential therapeutic signaling pathways and targets of HSYA against ALD. An ALD model in zebrafish larvae was established. Larvae were pretreated with HSYA and then exposed to ethanol. Liver injury was measured by fluorescence expression analysis in the liver-specific transgenic zebrafish line Tg (fabp10a:DsRed) and liver tissue H&E staining. Liver steatosis was determined by whole-mount oil red O staining and TG level. Additionally, an ethanol-induced hepatocyte injury model was established in vitro to observe hepatocyte damage (cell viability, ALT level), lipid accumulation (oil red O staining, TC and TG), and oxidative stress (ROS, MDA, GPx and SOD) in HepG2 cells treated with or without HSYA. Finally, qRT-PCR combined with network pharmacology and molecular docking was employed to validate the effects of HSYA on targets. RESULTS HSYA exhibited a significant, dose-dependent improvement in ethanol-induced liver injury in zebrafish larvae and HepG2 cells. Network pharmacology analysis revealed that HSYA may exert pharmacological effects against ALD through 341 potential targets. These targets are involved in various signaling pathways, including lipid metabolism and atherosclerosis, PI3K-Akt signaling pathway, MAPK signaling pathway, and ALD itself. Molecular docking studies displayed that HSYA had a strong binding affinity toward the domains of IL1B, IL6, TNF, PPARA, PPARG, HMGCR and ADH5. qRT-PCR assays demonstrated that HSYA effectively reversed the ethanol-induced aberrant gene expression of SREBF1, FASN, ACACA, CPT1A, PPARA, IL1B, IL6, TNFα, ADH5, and ALDH2 in vivo and in vitro. CONCLUSION This study offers a comprehensive investigation into the anti-ALD mechanisms of HSYA using an integrative pharmacology approach. The potential targets of HSYA may be implicated in enhancing ethanol catabolism, reducing lipid accumulation, mitigating oxidative stress, and inhibiting inflammatory response.
Collapse
Affiliation(s)
- Hai-Chuan Yu
- School of Medical Technology, Xinxiang Medical University, NO. 601 Jinsui Avenue, Xinxiang, Henan 453003, China.
| | - Qi-Rong Bai
- School of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Jiao-Jie Guo
- School of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Meng-Yao Chen
- School of Medical Technology, Xinxiang Medical University, NO. 601 Jinsui Avenue, Xinxiang, Henan 453003, China
| | - Lin Wang
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Fang-Chao Tong
- School of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Shuang-Ling Zhang
- School of Medical Technology, Xinxiang Medical University, NO. 601 Jinsui Avenue, Xinxiang, Henan 453003, China
| | - Jiao Wu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
2
|
Rabelo ACS, Andrade AKDL, Costa DC. The Role of Oxidative Stress in Alcoholic Fatty Liver Disease: A Systematic Review and Meta-Analysis of Preclinical Studies. Nutrients 2024; 16:1174. [PMID: 38674865 PMCID: PMC11055095 DOI: 10.3390/nu16081174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/05/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Alcoholic Fatty Liver Disease (AFLD) is characterized by the accumulation of lipids in liver cells owing to the metabolism of ethanol. This process leads to a decrease in the NAD+/NADH ratio and the generation of reactive oxygen species. A systematic review and meta-analysis were conducted to investigate the role of oxidative stress in AFLD. A total of 201 eligible manuscripts were included, which revealed that animals with AFLD exhibited elevated expression of CYP2E1, decreased enzymatic activity of antioxidant enzymes, and reduced levels of the transcription factor Nrf2, which plays a pivotal role in the synthesis of antioxidant enzymes. Furthermore, animals with AFLD exhibited increased levels of lipid peroxidation markers and carbonylated proteins, collectively contributing to a weakened antioxidant defense and increased oxidative damage. The liver damage in AFLD was supported by significantly higher activity of alanine and aspartate aminotransferase enzymes. Moreover, animals with AFLD had increased levels of triacylglycerol in the serum and liver, likely due to reduced fatty acid metabolism caused by decreased PPAR-α expression, which is responsible for fatty acid oxidation, and increased expression of SREBP-1c, which is involved in fatty acid synthesis. With regard to inflammation, animals with AFLD exhibited elevated levels of pro-inflammatory cytokines, including TNF-a, IL-1β, and IL-6. The heightened oxidative stress, along with inflammation, led to an upregulation of cell death markers, such as caspase-3, and an increased Bax/Bcl-2 ratio. Overall, the findings of the review and meta-analysis indicate that ethanol metabolism reduces important markers of antioxidant defense while increasing inflammatory and apoptotic markers, thereby contributing to the development of AFLD.
Collapse
Affiliation(s)
- Ana Carolina Silveira Rabelo
- Postgraduate Program in Biological Sciences, Federal University of Ouro Preto, Ouro Preto 35402-163, Brazil
- Department of Biochemistry, Federal University of Alfenas, Alfenas 37130-001, Brazil
| | | | - Daniela Caldeira Costa
- Postgraduate Program in Biological Sciences, Federal University of Ouro Preto, Ouro Preto 35402-163, Brazil
| |
Collapse
|
3
|
Huang S, Wang HL. Salvianolic acid A improves nerve regeneration and repairs nerve defects in rats with brain injury by downregulating miR-212-3p-mediated SOX7. Kaohsiung J Med Sci 2023; 39:1222-1232. [PMID: 37987200 DOI: 10.1002/kjm2.12779] [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: 04/21/2023] [Revised: 09/22/2023] [Accepted: 10/10/2023] [Indexed: 11/22/2023] Open
Abstract
This study was to probe the protective effects and mechanisms of salvianolic acid A (SAA) on cerebral ischemia-reperfusion injury (CIRI). The middle cerebral artery occlusion model (MCAO) was established in rats. Rats' behavior, neurological deficits, brain injury, inflammation, and apoptosis in the brain tissue were evaluated. The inflammatory response and apoptosis of PC12 cells induced by oxygen glucose deprivation/reperfusion (OGD/R) were detected. SAA-mediated changes in miR-212-3p, SOX7, and Wnt/β-catenin pathway were determined, and the targeting relationship between miR-212-3p and SOX7 was clarified. SAA alleviated the neurological deficits and brain injury of MCAO rats and inhibited the inflammatory response and apoptosis of OGD/R-conditioned PC-12 cells. SAA upregulated miR-212-3p, Wnt3a, and β-catenin, whereas inhibited SOX7 levels. Silencing miR-212-3p counteracted the protective effect of SAA in the context of CIRI. SOX7 was a target protein of miR-212-3p. Silencing SOX7 based on SAA and miR-212-3p knockdown suppressed OGD/R-induced inflammation and apoptosis and increased Wnt3a and β-catenin levels in PC12 cells. SAA can improve the brain and nervous system injury caused by cerebral ischemia-reperfusion by upregulating miR-212-3p, thereby inhibiting SOX7 and activating the Wnt/βcatenin signaling pathway.
Collapse
Affiliation(s)
- Shuai Huang
- Department of Rehabilitation Medicine, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong City, Jiangsu Province, China
| | - Hong-Liang Wang
- Department of Neurology, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong City, Jiangsu Province, China
| |
Collapse
|
4
|
Han W, Li H, Jiang H, Xu H, Lin Y, Chen J, Bi C, Liu Z. Progress in the mechanism of autophagy and traditional Chinese medicine herb involved in alcohol-related liver disease. PeerJ 2023; 11:e15977. [PMID: 37727691 PMCID: PMC10506582 DOI: 10.7717/peerj.15977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 08/07/2023] [Indexed: 09/21/2023] Open
Abstract
Alcohol-related liver disease (ALD) is chronic liver damage caused by long-term heavy drinking with, extremely complicated pathogenesis. The current studies speculated that excessive alcohol and its metabolites are the major causes of liver cell toxicity. Autophagy is evolutionarily conserved in eukaryotes and aggravates alcoholic liver damage, through various mechanisms, such as cellular oxidative stress, inflammation, mitochondrial damage and lipid metabolism disorders. Therefore, autophagy plays an critical role in the occurrence and development of ALD. Some studies have shown that traditional Chinese medicine extracts improve the histological characteristics of ALD, as reflected in the improvement of oxidative stress and lipid droplet clearance, which might be achieved by inducing autophagy. This article reviews the mechanisms of quercetin, baicalin, glycycoumarin, salvianolic acid A, resveratrol, ginsenoside rg1, and dihydromyricetin inducing autophagy and their participation in the inhibition of ALD. The regulation of autophagy in ALD by these traditional Chinese medicine extracts provides novel ideas for the treatment of the disease; however, its molecular mechanism needs to be elucidated further.
Collapse
Affiliation(s)
- Wenwen Han
- Department of Medical Laboratory, School of Medicine, Shaoxing University, Shaoxing, Zhejiang Province, China
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang Province, China
| | - Haiyu Li
- Department of Medical Laboratory, School of Medicine, Shaoxing University, Shaoxing, Zhejiang Province, China
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang Province, China
| | - Hanqi Jiang
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang Province, China
| | - Hang Xu
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang Province, China
| | - Yifeng Lin
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang Province, China
| | - Jiahuan Chen
- Department of Medical Laboratory, School of Medicine, Shaoxing University, Shaoxing, Zhejiang Province, China
| | - Chenchen Bi
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang Province, China
| | - Zheng Liu
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang Province, China
| |
Collapse
|
5
|
Ding Q, Zhang G, Wang Y, Xu L, Wu M, Zhou Y, Xu T, Meng X, Huang C, Zhang L. β-catenin ISGylation promotes lipid deposition and apoptosis in ethanol-stimulated liver injury models. Redox Rep 2022; 27:239-248. [PMID: 36259544 PMCID: PMC9586657 DOI: 10.1080/13510002.2022.2109360] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background The restoration of the Wnt/β-catenin pathway to alleviate alcoholic fatty liver disease (AFLD) progression is under study as a new strategy for alcoholic liver disease (ALD) treatment. Recent studies have indicated that interferon-stimulated gene 15 (ISG15) can covalently bind to β-catenin by HECT E3 ubiquitin ligase 5 (HERC5), leading to ISG degradation and downregulation of β-catenin levels. However, the relationship between β-catenin and the ISG15 system in AFLD remains unclear. Methods Here, we explored the roles of the ISG15 system in β-catenin activation and in the pathogenesis of alcohol-induced liver injury and steatosis. Results In this study, HERC5 silencing upregulated β-catenin protein expression and inhibited lipid metabolism disorders and cell apoptosis. Reduced β-catenin protein expression, increased lipid metabolism disorders, and cell apoptosis were detected in cells induced with HERC5 overexpression, which was reversible with the reactive oxygen species (ROS) inhibitor. All the above results were statistically analyzed. Thus, these observations demonstrate that β-catenin ISGylation is a prominent regulator of ALD pathology, which works by regulating ROS to induce lipid metabolism disorders and cell apoptosis. Conclusion Our findings provided the mechanism involved in the β-catenin ISGylation, allowing for future studies on the prevention or amelioration of liver injury in ALD.
Collapse
Affiliation(s)
- Qi Ding
- Anhui No.2 Provincial People's Hospital, Hefei, People's Republic of China
| | - Guodong Zhang
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Yang Wang
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Lei Xu
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Meifei Wu
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Yiwen Zhou
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Tao Xu
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Xiaoming Meng
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Cheng Huang
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| | - Lei Zhang
- School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,Key Laboratory of major autoimmune disease, Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, People's Republic of China.,The Key Laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
6
|
Yu X, Li Z, Bai R, Tang F. Transcriptional factor 3 binds to sirtuin 1 to activate the Wnt/β-catenin signaling in cervical cancer. Bioengineered 2022; 13:12516-12531. [PMID: 35587604 PMCID: PMC9275895 DOI: 10.1080/21655979.2022.2076481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Transcriptional factor 3 (TCF3, also termed E2A), first reported to exert crucial functions during lymphocyte development, has been revealed to participate in the pathogenesis of human cancers. The aim of this work was to investigate the function of TCF3 in cervical cancer (CC) and the molecular interactions. The bioinformatics prediction suggested that TCF3 was highly expressed in CC and linked to poor prognosis. Increased TCF3 expression was identified in CC cell lines, and its downregulation reduced proliferation and migration of CC cells in vitro as well as growth of xenograft tumors in vivo. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses showed that the TCF-3-related genes and genes showed differential expression between CC and normal tissues were mainly enriched in the Wnt/β-catenin pathway. TCF3 bound to sirtuin 1 (SIRT1) promoter for transcriptional activation, and SIRT1 promoted deacetylation and nuclear translocation of β-catenin in CC. SIRT1 overexpression blocked the role of TCF3 silencing and restored cell proliferation in vitro and tumor growth in vivo. Treatment with XAV-939, a β-catenin inhibitor, significantly suppressed the cell proliferation and tumor growth induced by SIRT1 overexpression. In conclusion, this study demonstrates that TCF3 augments progression of CC by activating SIRT1-mediated β-catenin signaling.
Collapse
Affiliation(s)
- Xiao Yu
- Department of Gynecological Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan, P.R. China
| | - Zhaoshuo Li
- Department of Cerebrovascular Disease, Henan Provincial People's Hospital, Henan, P.R. China
| | - Ruihua Bai
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Henan, P.R. China
| | - Fuxiang Tang
- Department of Gynecology, The Second Affiliated Hospital of Zhengzhou University, Henan, P.R. China
| |
Collapse
|
7
|
Konrath EL, Arbo MD, Arbo BD, Hort MA, Elisabetsky E, Leal MB. Plants with Anti-Addictive Potential. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1308:185-215. [PMID: 33861445 DOI: 10.1007/978-3-030-64872-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Drug addiction is prevalent among individuals of modern society, being a major cause of disability and premature loss of life. Although the drug addiction have profound social, economical and health impact in the world population, its management remains a challenge as available pharmacological treatments remains ineffective for most people. The limited efficacy and adverse effects have led to a search for alternative therapies to treat drug addiction. In this context, natural products are an important source for new chemical substances with a potential therapeutic applicability. Therefore, this chapter will present data obtained after an extensive literature search regarding the use of medicinal plants as a pharmacological alternative for drug addiction treatment.
Collapse
Affiliation(s)
- Eduardo Luis Konrath
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Marcelo Dutra Arbo
- Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Bruno Dutra Arbo
- Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Mariana Appel Hort
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande (FURG), Rio Grande, Rio Grande do Sul, Brazil
| | - Elaine Elisabetsky
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Mirna Bainy Leal
- Departamento de Farmacologia, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
8
|
Li S, Qian Q, Ying N, Lai J, Feng L, Zheng S, Jiang F, Song Q, Chai H, Dou X. Activation of the AMPK-SIRT1 pathway contributes to protective effects of Salvianolic acid A against lipotoxicity in hepatocytes and NAFLD in mice. Front Pharmacol 2020; 11:560905. [PMID: 33328983 PMCID: PMC7734334 DOI: 10.3389/fphar.2020.560905] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/30/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Salvianolic acid A (Sal A), a natural polyphenol compound extracted from Radix Salvia miltiorrhiza (known as Danshen in China), possesses a variety of potential pharmacological activities. The aim of this study is to determine mechanisms of hepatoprotective effects of Sal A against lipotoxicity both in cultured hepatocytes and in a mouse model of fatty liver disease. Methods: High-fat and high-carbohydrate diet (HFCD)-fed C57BL/6J mice were employed to establish hepatic lipotoxicity in a mouse model. Two doses of Sal A were administered every other day via intraperitoneal injection (20 and 40 mg/kg BW, respectively). After a 10-week intervention, liver injury was detected by immunohistochemical and biochemical analyses. For in vitro studies, we used HepG2, a human hepatoma cell line, and exposed them to palmitic acid to induce lipotoxicity. The protective effects of Sal A on palmitic acid-induced lipotoxicity were examined in Sal A-pretreated HepG2 cells. Results: Sal A treatments attenuated body weight gain, liver injury, and hepatic steatosis in mice exposed to HFCD. Sal A pretreatments ameliorated palmitic acid-induced cell death but did not reverse effects of HFCD- or palmitate-induced activations of JNK, ERK1/2, and PKA. Induction of p38 phosphorylation was significantly reversed by Sal A in HFCD-fed mice but not in palmitate-treated HepG2 cells. However, Sal A rescued hepatic AMP-activated protein kinase (AMPK) suppression and sirtuin 1 (SIRT1) downregulation by both HFCD feeding in mice and exposure to palmitate in HepG2 cells. Sal A dose-dependently up-regulated p-AMPK and SIRT1 protein levels. Importantly, siRNA silencing of either AMPK or SIRT1 gene expression abolished the protective effects of Sal A on lipotoxicity. Moreover, while AMPK silencing blocked Sal A-induced SIRT1, silencing of SIRT1 had no effect on Sal A-triggered AMPK activation, suggesting SIRT1 upregulation by Sal A is mediated by AMPK activation. Conclusion: Our data uncover a novel mechanism for hepatoprotective effects of Sal A against lipotoxicity both in livers from HFCD-fed mice and palmitic acid-treated hepatocytes.
Collapse
Affiliation(s)
- Songtao Li
- College of Basic Medicine and Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qianyu Qian
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Na Ying
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianfei Lai
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Luyan Feng
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Sitong Zheng
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fusheng Jiang
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qing Song
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hui Chai
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaobing Dou
- Molecular Medicine Institute, Zhejiang Chinese Medical University, Hangzhou, China
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
9
|
Zhou F, Teng L, Liu Y, Ma Y, Chen W, Bi L. Elaboration of the Comprehensive Metabolic Profile of Salvianolic Acid A in Vivo and in Vitro Using UFLC-Q/TOF-MS. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:12199-12207. [PMID: 31595753 DOI: 10.1021/acs.jafc.9b04131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Salvianolic acid A (Sal A) has a wide range of pharmacological activities. To date, there have been no systematic and detailed metabolite research data of Sal A after oral administration in vitro and in vivo. In this study, a rapid and systematic method based on ultrafast liquid chromatography-quadrupole-time-of-flight mass spectrometry was developed to detect metabolites of Sal A in vitro (human liver microsome, human intestinal microbiota, artificial gastric, and intestinal juice) and in vivo (urine, plasma, feces, and various organs collected after oral administration of Sal A to normal rats and pseudo-germ-free rats). A total of 26 metabolites of Sal A were characterized. These metabolites were formed through extensive metabolic reactions, such as hydroxylation, hydrogenation, and glucuronidation reactions. This study provides novel possibility for exploring the potential biological mechanism of Sal A, and aids the promotion of clinical application.
Collapse
Affiliation(s)
- Fuqiong Zhou
- Central Laboratory, Affiliated Nanjing Hospital of Chinese Medicine , Nanjing University of Chinese Medicine , 157 Daming Road , Nanjing , Jiangsu 210012 , China
- School of Preclinical Medicine , Nanjing University of Chinese Medicine , 138 Xianlin Road , Nanjing , Jiangsu 210023 , China
| | - Linxin Teng
- School of Preclinical Medicine , Nanjing University of Chinese Medicine , 138 Xianlin Road , Nanjing , Jiangsu 210023 , China
| | - Yu Liu
- School of Preclinical Medicine , Nanjing University of Chinese Medicine , 138 Xianlin Road , Nanjing , Jiangsu 210023 , China
| | - Yanxia Ma
- School of Preclinical Medicine , Nanjing University of Chinese Medicine , 138 Xianlin Road , Nanjing , Jiangsu 210023 , China
| | - Weiping Chen
- School of Preclinical Medicine , Nanjing University of Chinese Medicine , 138 Xianlin Road , Nanjing , Jiangsu 210023 , China
| | - Lei Bi
- School of Preclinical Medicine , Nanjing University of Chinese Medicine , 138 Xianlin Road , Nanjing , Jiangsu 210023 , China
| |
Collapse
|
10
|
Luo Y, Feng Y, Song L, He GQ, Li S, Bai SS, Huang YJ, Li SY, Almutairi MM, Shi HL, Wang Q, Hong M. A network pharmacology-based study on the anti-hepatoma effect of Radix Salviae Miltiorrhizae. Chin Med 2019; 14:27. [PMID: 31406500 PMCID: PMC6685170 DOI: 10.1186/s13020-019-0249-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/26/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Radix Salviae Miltiorrhizae (RSM), a well-known traditional Chinese medicine, has been shown to inhibit tumorigenesis in various human cancers. However, the anticancer effects of RSM on human hepatocellular carcinoma (HCC) and the underlying mechanisms of action remain to be fully elucidated. METHODS In this study, we aimed to elucidate the underlying molecular mechanisms of RSM in the treatment of HCC using a network pharmacology approach. In vivo and in vitro experiments were also performed to validate the therapeutic effects of RSM on HCC. RESULTS In total, 62 active compounds from RSM and 72 HCC-related targets were identified through network pharmacological analysis. RSM was found to play a critical role in HCC via multiple targets and pathways, especially the EGFR and PI3K/AKT signaling pathways. In addition, RSM was found to suppress HCC cell proliferation, and impair cancer cell migration and invasion in vitro. Flow cytometry analysis revealed that RSM induced cell cycle G2/M arrest and apoptosis, and western blot analysis showed that RSM up-regulated the expression of BAX and down-regulated the expression of Bcl-2 in MHCC97-H and HepG2 cells. Furthermore, RSM administration down-regulated the expression of EGFR, PI3K, and p-AKT proteins, whereas the total AKT level was not altered. Finally, the results of our in vivo experiments confirmed the therapeutic effects of RSM on HCC in nude mice. CONCLUSIONS We provide an integrative network pharmacology approach, in combination with in vitro and in vivo experiments, to illustrate the underlying therapeutic mechanisms of RSM action on HCC.
Collapse
Affiliation(s)
- Yi Luo
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Yu Feng
- Department of Traumatology, General Hospital of Ningxia Medical University, Yinchuan, 750004 China
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lei Song
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Gan-Qing He
- Department of Gastroenterology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 501260 China
| | - Sha Li
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Sha-Sha Bai
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Yu-Jie Huang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Si-Ying Li
- Department of Pharmacology & Toxicology, University of Kansas, Lawrence, KS USA
| | | | - Hong-Lian Shi
- Department of Pharmacology & Toxicology, University of Kansas, Lawrence, KS USA
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| | - Ming Hong
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, 510405 China
| |
Collapse
|
11
|
Li X, Liu Y, Yue W, Tan Y, Wang H, Zhang L, Chen J. A Compound of Chinese Herbs Protects against Alcoholic Liver Fibrosis in Rats via the TGF- β1/Smad Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:9121347. [PMID: 31118972 PMCID: PMC6500606 DOI: 10.1155/2019/9121347] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/11/2019] [Accepted: 03/31/2019] [Indexed: 02/06/2023]
Abstract
Alcoholic liver fibrosis (ALF) has become a major public health concern owing to its health impacts and the lack of effective treatment strategies for the disease. In this study, we investigated the effect of a compound composed of Chinese herbs Pueraria lobata (Willd.), Salvia miltiorrhiza, Schisandra chinensis, and Silybum marianum on ALF. An ALF model was established. Rats were fed with modified Lieber-Decarli alcohol liquid diet and injected with trace CCl4 at late stage. The rats were then treated with several doses of the compound. Biochemical and fibrosis-relevant parameters were measured from the sera obtained from the rats. Liver tissues were obtained for hematoxylin and eosin and Masson's trichrome staining. Matrix metalloproteinase-13 and tissue inhibitor of metalloproteinase-1 were determined by immunohistochemistry assays. The mRNA and protein expression levels of transforming growth factor-β1 (TGF-β1), Smad2, Smad3, and Smad7 on the livers were also measured by quantitative polymerase chain reaction and Western blot. Results showed that the compound treatment alleviated pathological lesions in the liver, decreased the serum levels of hyaluronan, laminin, and hydroxyproline, and diminished the expression of hepatic tissue inhibitor of metalloproteinase-1. Compound treatment also increased hepatic matrix metalloproteinase-13 expression and inhibited the TGF-β1/Smad signaling pathway. In conclusion, the compound has a protective effect against ALF in rats, and an underlying mechanism is involved in the TGF-β1/Smad signaling pathway.
Collapse
Affiliation(s)
- Xiaomeng Li
- West China School of Public Health and West China fourth Hospital, Sichuan University, Chengdu, China
- Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Yunjie Liu
- West China School of Public Health and West China fourth Hospital, Sichuan University, Chengdu, China
- Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Wuyang Yue
- West China School of Public Health and West China fourth Hospital, Sichuan University, Chengdu, China
- Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Yuefeng Tan
- West China School of Public Health and West China fourth Hospital, Sichuan University, Chengdu, China
- Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - He Wang
- West China School of Public Health and West China fourth Hospital, Sichuan University, Chengdu, China
- Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Lishi Zhang
- West China School of Public Health and West China fourth Hospital, Sichuan University, Chengdu, China
- Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Jinyao Chen
- West China School of Public Health and West China fourth Hospital, Sichuan University, Chengdu, China
- Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|