1
|
Tang S, Zhang Y, Botchway BOA, Wang X, Huang M, Liu X. Epigallocatechin-3-Gallate Inhibits Oxidative Stress Through the Keap1/Nrf2 Signaling Pathway to Improve Alzheimer Disease. Mol Neurobiol 2025; 62:3493-3507. [PMID: 39299981 DOI: 10.1007/s12035-024-04498-6] [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: 04/23/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
Alzheimer disease (AD) is a common neurodegenerative disease with an intricate pathophysiological mechanism. Oxidative stress has been shown in several investigations as a significant factor in AD progression. For instance, studies have confirmed that oxidative stress inhibition may considerably improve AD symptoms, with potent antioxidants being touted as a possible interventional strategy in the search for AD treatment. Epigallocatechin-3-gallate (EGCG) acts as a natural catechin that has antioxidant effect. It activates the kelch-like epichlorohydrin-associated proteins (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway to inhibit oxidative stress. The Keap1/Nrf2 signal pathway is not only an upstream signaling target for a variety of antioxidant enzymes, but also minimizes high levels of reactive oxygen species. This report analyzes the antioxidant effect of EGCG in AD, elaborates its specific mechanism of action, and provides a theoretical basis for its clinical application in AD.
Collapse
Affiliation(s)
- Shi Tang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, China
| | - Yong Zhang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, China
| | - Benson O A Botchway
- Bupa Cromwell Hospital, Kensington, London, UK
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia, Cyprus
| | - Xichen Wang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, China
| | - Min Huang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, China
| | - Xuehong Liu
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, China.
| |
Collapse
|
2
|
Gao G, Lin C, Li R, Xie X, Luo HB. Epigallocatechin-3-gallate inhibits the collagen accumulation of oral submucous fibrosis induced by arecoline. Front Pharmacol 2025; 16:1540559. [PMID: 39959427 PMCID: PMC11825517 DOI: 10.3389/fphar.2025.1540559] [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: 12/06/2024] [Accepted: 01/17/2025] [Indexed: 02/18/2025] Open
Abstract
Objective Oral submucous fibrosis (OSF) is a chronic oral mucosal disease, which exerts a profound impact on patients' daily life and currently lacks efficacious therapeutic interventions. Epigallocatechin-3-gallate (EGCG), the abundant polyphenol found in green tea, exhibits remarkable anti-fibrotic effects on the skin. However, the research on OSF regarding EGCG is relatively limited. Purpose We aimed to investigate the potential therapeutic effect of EGCG against OSF using an arecoline (ARE) -induced rat model and primary rat oral fibroblasts. Methods Primary rat oral mucosal fibroblasts (ROMF) were isolated and identified. Optimal ARE concentrations were established using the Cell Counting Kit-8. The impact of ARE on extracellular matrix (ECM)-related protein expression was assessed through RT-qPCR and Western blot techniques. Similarly, the effects of EGCG on ARE-induced ECM changes in ROMF were evaluated. The study also established an OSF model in Sprague-Dawley rats, induced by ARE, with pathological changes characterized using HE and Masson's staining, further assessing the impact of ARE on ECM-related protein expression in rat oral tissues through RT-qPCR and Western blot methods. Results EGCG effectively suppressed the ARE-induced ECM components while concurrently improving the OSF pathological process in vitro and in vivo. Conclusion The results indicate that the natural product EGCG effectively suppressed the increased ECM components induced by ARE and concurrently improved the OSF pathological process, indicating that EGCG could be potentially a novel anti-fibrotic candidate drug for the treatment of OSF.
Collapse
Affiliation(s)
- Ge Gao
- Key Laboratory of Tropical Biological Resources of Ministry of Education and Hainan Engineering Research Center for Drug Screening and Evaluation, School of Pharmaceutical Sciences, Hainan University, Haikou, China
- School of Life and Health Sciences, Hainan University, Haikou, China
| | - Caipeng Lin
- Key Laboratory of Tropical Biological Resources of Ministry of Education and Hainan Engineering Research Center for Drug Screening and Evaluation, School of Pharmaceutical Sciences, Hainan University, Haikou, China
- School of Life and Health Sciences, Hainan University, Haikou, China
| | - Ruibo Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education and Hainan Engineering Research Center for Drug Screening and Evaluation, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Xi Xie
- Key Laboratory of Tropical Biological Resources of Ministry of Education and Hainan Engineering Research Center for Drug Screening and Evaluation, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Hai-Bin Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education and Hainan Engineering Research Center for Drug Screening and Evaluation, School of Pharmaceutical Sciences, Hainan University, Haikou, China
- Song Li’s Academician Workstation of Hainan University (School of Pharmaceutical Sciences), Sanya, China
| |
Collapse
|
3
|
Lin Y, Zhang Y, Li Y, Xu Q, Zhang Y, Chen T, Wang J, Li J, Gong J, Chen Z, Yang Q, Li X. EGCG suppressed activation of hepatic stellate cells by regulating the PLCE1/IP 3/Ca 2+ pathway. Eur J Nutr 2024; 63:3255-3268. [PMID: 39325099 DOI: 10.1007/s00394-024-03504-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024]
Abstract
(-)-Epigallocatechin-3-O-gallate (EGCG), one of the green tea catechins, exhibits significant antioxidant properties that play an essential role in various diseases. However, the functional role and underlying mechanism of EGCG in stimulating of hepatic stellate cells (HSCs) remain unexplored in transcriptomics sequencing studies. The present study suggests that oral administration of EGCG at a dosage of 200 mg/kg/day for a duration of four weeks exhibits significant therapeutic potential in a murine model of liver fibrosis induced by CCl4. The activation of HSCs in vitro was dose-dependently inhibited by EGCG. The sequencing analysis data reveled that EGCG exerted a regulatory effect on the calcium signal in mouse HSCs, resulting in a decrease in calcium ion concentration. Further analysis revealed that EGCG inhibited the expression of phospholipase C epsilon-1 (PLCE1) and inositol 1, 4, 5-trisphosphate (IP3) in activated mouse HSCs. Additionally, EGCG contributes to the reduction the concentration of calcium ions by regulating PLCE1. After the knockdown of PLCE1, free calcium ion concentrations decreased, resulting in the inhibition of both cell proliferation and migration. Interestingly, the expression of PLCE1 and cytosolic calcium levels were regulated by reactive oxygen species(ROS). Furthermore, our findings suggest that ROS might inhibit the expression of PLCE1 by inhibiting TFEB, a transcription activator involved in the nuclear translocation process. Our study provided novel evidence regarding the regulatory effects of EGCG on activated HSCs (aHSCs) in mice by the calcium signaling pathway, emphasizing the crucial role of PLCE1 within the calcium signaling network of HSCs. The proposition was also made that PLCE1 holds promise as a novel therapeutic target for murine liver fibrosis.
Collapse
Affiliation(s)
- Ying Lin
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, No. 1838, North of Guangzhou Avenue, Guangzhou City, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yan Zhang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, No. 1838, North of Guangzhou Avenue, Guangzhou City, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yang Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, No. 1838, North of Guangzhou Avenue, Guangzhou City, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qihan Xu
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, No. 1838, North of Guangzhou Avenue, Guangzhou City, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yijie Zhang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, No. 1838, North of Guangzhou Avenue, Guangzhou City, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Tingting Chen
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, No. 1838, North of Guangzhou Avenue, Guangzhou City, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jun Wang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, No. 1838, North of Guangzhou Avenue, Guangzhou City, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jierui Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, No. 1838, North of Guangzhou Avenue, Guangzhou City, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jiacheng Gong
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, No. 1838, North of Guangzhou Avenue, Guangzhou City, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zhuoer Chen
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, No. 1838, North of Guangzhou Avenue, Guangzhou City, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qiaomu Yang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, No. 1838, North of Guangzhou Avenue, Guangzhou City, Guangdong Province, 510515, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, No. 1838, North of Guangzhou Avenue, Guangzhou City, Guangdong Province, 510515, China.
| |
Collapse
|
4
|
Amer J, Salhab A, Hussini E, Shweiki R, Zahran I, Far M. Osteopontin neutralization increases vitamin D receptors on NKT cells and ameliorates liver fibrosis by promoting their activity. Front Pharmacol 2024; 15:1484278. [PMID: 39654627 PMCID: PMC11625552 DOI: 10.3389/fphar.2024.1484278] [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: 08/21/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction and Aims Vitamin D has an immunomodulatory property influencing the activity of NKT cells. We aimed to study the impact of osteopontin (OPN), a key driver of fibrosis, on NKT cells' vitamin D receptor (VDR) and activity alterations. Methods Liver fibrosis was induced in BALB/C mice with carbon-tetrachloride (CCl4) for 8 weeks with either vitamin D [100 ng/kg] or InVivoMAb anti-mouse OPN [100 μg/kg] 2X/week started at week-4 of CCl4. The liver injury profile of serum ALT, AST, and inflammatory cytokines were evaluated. Histopathological findings were assessed via H&E staining and Sirius-Red staining. Fibrotic genes of αSMA, CREBP, and collagen III were assessed using RT-PCR. Fast blood sugar, insulin, liver cholesterol, and triglyceride were evaluated. Liver tissue-resident (tr)-NKT cells were obtained for VDR expressions, molecular pathways of p-STAT1 and P-STAT-5, and activation markers of CD107a and NKp46 using flow cytometry. Results Following vitamin D treatment, H&E staining revealed reduced microvascular and macrovascular steatosis, while Sirius-Red staining showed less fibrosis accumulation in liver fibrosis mice than in untreated counterparts. Results were associated with a significant decrease in serum cytokines of IL-β/IL-6/IL-4/OPN/TNF-α and serum AST and ALT by 2-fold and 3-fold, respectively. Fibrotic markers showed an average 1.3-fold decrease in αSMA, CREB, and Col-III in liver fibrosis mice following vitamin D treatment. Quantitated liver cholesterol and triglycerides, serum insulin, and fasting blood sugar ameliorated their levels following vitamin D treatment in liver fibrosis mice. OPN-neutralizing antibody over-expressed VDR on trNKT cells and increased CD107a and NKp46 activities of 3.1 and 3.5 folds, respectively, associated with increasing in p-STAT1 and p-STAT5 phosphorylation. These results were accompanied with a decrease in hepatic-stellate-cell activation markers of αSMA, Col-III, and desmin. Conclusion VDR expressions affect trNKT cells activity and could modulate progressions of liver fibrosis. Using an OPN-neutralizing antibody exhibited an antifibrotic effect by alleviating the liver injury profile through NKT cells. It is also suggested as an immunomodulatory target of liver fibrosis.
Collapse
Affiliation(s)
- Johnny Amer
- Department of Allied and Applied Medical Sciences, Division of anatomy, Biochemistry and Genetics, An-Najah National University, Nablus, Palestine
| | - Ahmad Salhab
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Enas Hussini
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Rasha Shweiki
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Iman Zahran
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Mohammad Far
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| |
Collapse
|
5
|
Baek JS, Lee JH, Kim JH, Cho SS, Kim YS, Yang JH, Shin EJ, Kang HG, Kim SJ, Ahn SG, Park EY, Baek DJ, Yim SK, Kang KW, Ki SH, Kim KM. An inducible sphingosine kinase 1 in hepatic stellate cells potentiates liver fibrosis. Biochem Pharmacol 2024; 229:116520. [PMID: 39236934 DOI: 10.1016/j.bcp.2024.116520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 08/19/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
Hepatic stellate cells (HSCs) play a role in hepatic fibrosis and sphingosine kinase (SphK) is involved in biological processes. As studies on the regulatory mechanisms and functions of SphK in HSCs during liver fibrosis are currently limited, this study aimed to elucidate the regulatory mechanism and connected pathways of SphK upon HSC activation. The expression of SphK1 was higher in HSCs than in hepatocytes, and upregulated in activated primary HSCs. SphK1 was also increased in liver homogenates of carbon tetrachloride-treated or bile duct ligated mice and in transforming growth factor-β (TGF-β)-treated LX-2 cells. TGF-β-mediated SphK1 induction was due to Smad3 signaling in LX-2 cells. SphK1 modulation altered the expression of liver fibrogenesis-related genes. This SphK1-mediated profibrogenic effect was dependent on SphK1/sphingosine-1-phosphate/sphingosine-1-phosphate receptor signaling through ERK. Epigallocatechin gallate blocked TGF-β-induced SphK1 expression and hepatic fibrogenesis by attenuating Smad and MAPK activation. SphK1 induced by TGF-β facilitates HSC activation and liver fibrogenesis, which is reversed by epigallocatechin gallate. Accordingly, SphK1 and related signal transduction may be utilized to treat liver fibrosis.
Collapse
Affiliation(s)
- Jin Sol Baek
- MRC-OSTRC, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju 61452, South Korea
| | - Ji Hyun Lee
- MRC-OSTRC, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju 61452, South Korea
| | - Ji Hye Kim
- MRC-OSTRC, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju 61452, South Korea
| | - Sam Seok Cho
- MRC-OSTRC, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju 61452, South Korea; Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju 61452, Republic of Korea; Institute of Well-Aging Medicare & Chosun University G-LAMP Project Group, Chosun University, Gwangju 61452, Republic of Korea
| | - Yun Seok Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Department of Pharmacy, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ji Hye Yang
- College of Korean Medicine, Dongshin University, Naju, Jeollanam-do 58245, Republic of Korea
| | - Eun Jin Shin
- Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju 61452, Republic of Korea; Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-associated Disorder Control Technology, Chosun University, Gwangju 61452, Republic of Korea; Institute of Well-Aging Medicare & Chosun University G-LAMP Project Group, Chosun University, Gwangju 61452, Republic of Korea
| | - Hyeon-Gu Kang
- Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju 61452, Republic of Korea; Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-associated Disorder Control Technology, Chosun University, Gwangju 61452, Republic of Korea
| | - Seok-Jun Kim
- Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju 61452, Republic of Korea; Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-associated Disorder Control Technology, Chosun University, Gwangju 61452, Republic of Korea; Institute of Well-Aging Medicare & Chosun University G-LAMP Project Group, Chosun University, Gwangju 61452, Republic of Korea
| | - Sang-Gun Ahn
- Department of Pathology, School of Dentistry, Chosun University, Gwangju 61452, Republic of Korea
| | - Eun Young Park
- College of Pharmacy, Mokpo National University, Muan-gun, Jeollanam-do 58554, Republic of Korea
| | - Dong Jae Baek
- College of Pharmacy, Mokpo National University, Muan-gun, Jeollanam-do 58554, Republic of Korea
| | - Sung-Kun Yim
- Marine Biotechnology Research Center, Jeonnam Bioindustry Foundation, 21-7, Nonggongdanji 4Gil, Wando-eup, Wando-gun, Jeollanam-do 59108, Republic of Korea
| | - Keon Wook Kang
- Department of Pharmacy, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sung Hwan Ki
- MRC-OSTRC, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju 61452, South Korea
| | - Kyu Min Kim
- Department of Biomedical Science, College of Natural Science, Chosun University, Gwangju 61452, Republic of Korea; Department of Integrative Biological Sciences & BK21 FOUR Educational Research Group for Age-associated Disorder Control Technology, Chosun University, Gwangju 61452, Republic of Korea; Institute of Well-Aging Medicare & Chosun University G-LAMP Project Group, Chosun University, Gwangju 61452, Republic of Korea.
| |
Collapse
|
6
|
George J, Lu Y, Tsuchishima M, Tsutsumi M. Cellular and molecular mechanisms of hepatic ischemia-reperfusion injury: The role of oxidative stress and therapeutic approaches. Redox Biol 2024; 75:103258. [PMID: 38970988 PMCID: PMC11279328 DOI: 10.1016/j.redox.2024.103258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024] Open
Abstract
Ischemia-reperfusion (IR) or reoxygenation injury is the paradoxical exacerbation of cellular impairment following restoration of blood flow after a period of ischemia during surgical procedures or other conditions. Acute interruption of blood supply to the liver and subsequent reperfusion can result in hepatocyte injury, apoptosis, and necrosis. Since the liver requires a continuous supply of oxygen for many biochemical reactions, any obstruction of blood flow can rapidly lead to hepatic hypoxia, which could quickly progress to absolute anoxia. Reoxygenation results in the increased generation of reactive oxygen species and oxidative stress, which lead to the enhanced production of proinflammatory cytokines, chemokines, and other signaling molecules. Consequent acute inflammatory cascades lead to significant impairment of hepatocytes and nonparenchymal cells. Furthermore, the expression of several vascular growth factors results in the heterogeneous closure of numerous hepatic sinusoids, which leads to reduced oxygen supply in certain areas of the liver even after reperfusion. Therefore, it is vital to identify appropriate therapeutic modalities to mitigate hepatic IR injury and subsequent tissue damage. This review covers all the major aspects of cellular and molecular mechanisms underlying the pathogenesis of hepatic ischemia-reperfusion injury, with special emphasis on oxidative stress, associated inflammation and complications, and prospective therapeutic approaches.
Collapse
Affiliation(s)
- Joseph George
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA; Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan; Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Ishikawa, 920-0293, Japan.
| | - Yongke Lu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Mutsumi Tsuchishima
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Mikihiro Tsutsumi
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan; Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Ishikawa, 920-0293, Japan
| |
Collapse
|
7
|
Ciceu A, Fenyvesi F, Hermenean A, Ardelean S, Dumitra S, Puticiu M. Advancements in Plant-Based Therapeutics for Hepatic Fibrosis: Molecular Mechanisms and Nanoparticulate Drug Delivery Systems. Int J Mol Sci 2024; 25:9346. [PMID: 39273295 PMCID: PMC11394827 DOI: 10.3390/ijms25179346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/21/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Chronic liver injuries often lead to hepatic fibrosis, a condition characterized by excessive extracellular matrix accumulation and abnormal connective tissue hyperplasia. Without effective treatment, hepatic fibrosis can progress to cirrhosis or hepatocellular carcinoma. Current treatments, including liver transplantation, are limited by donor shortages and high costs. As such, there is an urgent need for effective therapeutic strategies. This review focuses on the potential of plant-based therapeutics, particularly polyphenols, phenolic acids, and flavonoids, in treating hepatic fibrosis. These compounds have demonstrated anti-fibrotic activities through various signaling pathways, including TGF-β/Smad, AMPK/mTOR, Wnt/β-catenin, NF-κB, PI3K/AKT/mTOR, and hedgehog pathways. Additionally, this review highlights the advancements in nanoparticulate drug delivery systems that enhance the pharmacokinetics, bioavailability, and therapeutic efficacy of these bioactive compounds. Methodologically, this review synthesizes findings from recent studies, providing a comprehensive analysis of the mechanisms and benefits of these plant-based treatments. The integration of novel drug delivery systems with plant-based therapeutics holds significant promise for developing effective treatments for hepatic fibrosis.
Collapse
Affiliation(s)
- Alina Ciceu
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania
| | - Ferenc Fenyvesi
- Department of Molecular and Nanopharmaceutics, Faculty of Pharmacy, University of Debrecen, 4032 Debrecen, Hungary
| | - Anca Hermenean
- "Aurel Ardelean" Institute of Life Sciences, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania
| | - Simona Ardelean
- Faculty of Pharmacy, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania
| | - Simona Dumitra
- Faculty of Medicine, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania
| | - Monica Puticiu
- Faculty of Medicine, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania
| |
Collapse
|
8
|
Winiarska-Mieczan A, Jachimowicz-Rogowska K, Kwiecień M, Borsuk-Stanulewicz M, Tomczyk-Warunek A, Stamirowska-Krzaczek E, Purwin C, Stryjecka M, Tomaszewska M. Regular Consumption of Green Tea as an Element of Diet Therapy in Drug-Induced Liver Injury (DILI). Nutrients 2024; 16:2837. [PMID: 39275155 PMCID: PMC11396919 DOI: 10.3390/nu16172837] [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: 08/07/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/16/2024] Open
Abstract
The liver is a highly metabolically active organ, and one of the causes of its dysfunction is the damage caused by drugs and their metabolites as well as dietary supplements and herbal preparations. A common feature of such damage is drugs, which allows it to be defined as drug-induced liver injury (DILI). In this review, we analysed available research findings in the global literature regarding the effects of green tea and/or its phenolic compounds on liver function in the context of protective action during prolonged exposure to xenobiotics. We focused on the direct detoxifying action of epigallocatechin gallate (EGCG) in the liver, the impact of EGCG on gut microbiota, and the influence of microbiota on liver health. We used 127 scientific research publications published between 2014 and 2024. Improving the effectiveness of DILI detection is essential to enhance the safety of patients at risk of liver damage and to develop methods for assessing the potential hepatotoxicity of a drug during the research phase. Often, drugs cannot be eliminated, but appropriate nutrition can strengthen the body and liver, which may mitigate adverse changes resulting from DILI. Polyphenols are promising owing to their strong antioxidant and anti-inflammatory properties as well as their prebiotic effects. Notably, EGCG is found in green tea. The results of the studies presented by various authors are very promising, although not without uncertainties. Therefore, future research should focus on elucidating the therapeutic and preventive mechanisms of polyphenols in the context of liver health through the functioning of gut microbiota affecting overall health, with particular emphasis on epigenetic pathways.
Collapse
Affiliation(s)
- Anna Winiarska-Mieczan
- Institute of Animal Nutrition and Bromatology, Department of Bromatology and Nutrition Physiology, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland
- Institute of Human Nutrition and Agriculture, The University College of Applied Sciences in Chełm, Pocztowa 54, 22-100 Chełm, Poland
| | - Karolina Jachimowicz-Rogowska
- Institute of Animal Nutrition and Bromatology, Department of Bromatology and Nutrition Physiology, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland
| | - Małgorzata Kwiecień
- Institute of Animal Nutrition and Bromatology, Department of Bromatology and Nutrition Physiology, University of Life Sciences in Lublin, Akademicka 13, 20-950 Lublin, Poland
| | - Marta Borsuk-Stanulewicz
- Department of Animal Nutrition and Feed Science, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland
| | - Agnieszka Tomczyk-Warunek
- Laboratory of Locomotor Systems Research, Department of Rehabilitation and Physiotherapy, Medical University of Lublin, Jaczewskiego 8, 20-954 Lublin, Poland
| | - Ewa Stamirowska-Krzaczek
- Institute of Human Nutrition and Agriculture, The University College of Applied Sciences in Chełm, Pocztowa 54, 22-100 Chełm, Poland
| | - Cezary Purwin
- Department of Animal Nutrition and Feed Science, University of Warmia and Mazury in Olsztyn, 10-719 Olsztyn, Poland
| | - Małgorzata Stryjecka
- Institute of Human Nutrition and Agriculture, The University College of Applied Sciences in Chełm, Pocztowa 54, 22-100 Chełm, Poland
| | - Marzena Tomaszewska
- Institute of Human Nutrition and Agriculture, The University College of Applied Sciences in Chełm, Pocztowa 54, 22-100 Chełm, Poland
| |
Collapse
|
9
|
Zhao B, Liu K, Liu X, Li Q, Li Z, Xi J, Xie F, Li X. Plant-derived flavonoids are a potential source of drugs for the treatment of liver fibrosis. Phytother Res 2024; 38:3122-3145. [PMID: 38613172 DOI: 10.1002/ptr.8193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 02/28/2024] [Accepted: 03/10/2024] [Indexed: 04/14/2024]
Abstract
Liver fibrosis is a dynamic pathological process that can be triggered by any chronic liver injury. If left unaddressed, it will inevitably progress to the severe outcomes of liver cirrhosis or even hepatocellular carcinoma. In the past few years, the prevalence and fatality of hepatic fibrosis have been steadily rising on a global scale. As a result of its intricate pathogenesis, the quest for pharmacological interventions targeting liver fibrosis has remained a formidable challenge. Currently, no pharmaceuticals are exhibiting substantial clinical efficacy in the management of hepatic fibrosis. Hence, it is of utmost importance to expedite the development of novel therapeutics for the treatment of this condition. Various research studies have revealed the ability of different natural flavonoid compounds to alleviate or reverse hepatic fibrosis through a range of mechanisms, which are related to the regulation of liver inflammation, oxidative stress, synthesis and secretion of fibrosis-related factors, hepatic stellate cells activation, and proliferation, and extracellular matrix synthesis and degradation by these compounds. This review summarizes the progress of research on different sources of natural flavonoids with inhibitory effects on liver fibrosis over the last decades. The anti-fibrotic effects of natural flavonoids have been increasingly studied, making them a potential source of drugs for the treatment of liver fibrosis due to their good efficacy and biosafety.
Collapse
Affiliation(s)
- Bolin Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kai Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xing Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiuxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhibei Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingjing Xi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fan Xie
- Hospital of Chengdu University of Traditional Chinese Medicine 610032, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Saito T, Tsuchishima M, Tsutsumi M, George J. Molecular pathogenesis of metabolic dysfunction-associated steatotic liver disease, steatohepatitis, hepatic fibrosis and liver cirrhosis. J Cell Mol Med 2024; 28:e18491. [PMID: 38894579 PMCID: PMC11187936 DOI: 10.1111/jcmm.18491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/24/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is characterized by intense deposition of fat globules in the hepatic parenchyma that could potentially progress to liver cirrhosis and hepatocellular carcinoma. Here, we evaluated a rat model to study the molecular pathogenesis of the spectrum of MASLD and to screen therapeutic agents. SHRSP5/Dmcr rats were fed a high-fat and cholesterol (HFC) diet for a period of 12 weeks and evaluated for the development of steatosis (MASLD), steatohepatitis, fibrosis and cirrhosis. A group of animals were sacrificed at the end of the 4th, 6th, 8th and 12th weeks from the beginning of the experiment, along with the control rats that received normal diet. Blood and liver samples were collected for biochemical and histopathological evaluations. Immunohistochemical staining was performed for α-SMA and Collagen Type I. Histopathological examinations demonstrated steatosis at the 4th week, steatohepatitis with progressive fibrosis at the 6th week, advanced fibrosis with bridging at the 8th week and cirrhosis at the 12th week. Biochemical markers and staining for α-SMA and Collagen Type I demonstrated the progression of steatosis to steatohepatitis, hepatic fibrosis and liver cirrhosis in a stepwise manner. Control animals fed a normal diet did not show any biochemical or histopathological alterations. The results of the present study clearly demonstrated that the HFC diet-induced model of steatosis, steatohepatitis, hepatic fibrosis and cirrhosis is a feasible, quick and appropriate animal model to study the molecular pathogenesis of the spectrum of MASLD and to screen potent therapeutic agents.
Collapse
Affiliation(s)
- Takashi Saito
- Department of HepatologyKanazawa Medical UniversityUchinadaIshikawaJapan
| | | | - Mikihiro Tsutsumi
- Department of HepatologyKanazawa Medical UniversityUchinadaIshikawaJapan
- Center for Regenerative MedicineKanazawa Medical University HospitalUchinadaIshikawaJapan
| | - Joseph George
- Department of HepatologyKanazawa Medical UniversityUchinadaIshikawaJapan
- Center for Regenerative MedicineKanazawa Medical University HospitalUchinadaIshikawaJapan
| |
Collapse
|
11
|
Yamagata M, Tsuchishima M, Saito T, Tsutsumi M, George J. Therapeutic implication of human placental extract to prevent liver cirrhosis in rats with metabolic dysfunction-associated steatohepatitis. Clin Sci (Lond) 2024; 138:327-349. [PMID: 38381799 DOI: 10.1042/cs20230533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 02/14/2024] [Accepted: 02/21/2024] [Indexed: 02/23/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is always accompanied with hepatic fibrosis that could potentially progress to liver cirrhosis and hepatocellular carcinoma. Employing a rat model, we evaluated the role of human placental extract (HPE) to arrest the progression of hepatic fibrosis to cirrhosis in patients with MASH. SHRSP5/Dmcr rats were fed with a high-fat and high-cholesterol diet for 4 weeks and evaluated for the development of steatosis. The animals were divided into control and treated groups and received either saline or HPE (3.6 ml/kg body weight) subcutaneously thrice a week. A set of animals were killed at the end of 6th, 8th, and 12th weeks from the beginning of the experiment. Serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), hepatic malondialdehyde (MDA), and glutathione content were measured. Immunohistochemical staining was performed for α-smooth muscle actin (α-SMA), 4-hydroxy-2-nonenal (4-HNE), collagen type I, and type III. Control rats depicted progression of liver fibrosis at 6 weeks, advanced fibrosis and bridging at 8 weeks, and cirrhosis at 12 weeks, which were significantly decreased in HPE-treated animals. Treatment with HPE maintained normal levels of MDA and glutathione in the liver. There was marked decrease in the staining intensity of α-SMA, 4-HNE, and collagen type I and type III in HPE treated rats compared with control animals. The results of the present study indicated that HPE treatment mediates immunotropic, anti-inflammatory, and antioxidant responses and attenuates hepatic fibrosis and early cirrhosis. HPE depicts therapeutic potential to arrest the progression of MASH towards cirrhosis.
Collapse
Affiliation(s)
- Mitsuyoshi Yamagata
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Mutsumi Tsuchishima
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Takashi Saito
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
| | - Mikihiro Tsutsumi
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
- Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Ishikawa 920-0293, Japan
| | - Joseph George
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293, Japan
- Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Ishikawa 920-0293, Japan
| |
Collapse
|
12
|
Azeredo PDS, Fan D, Murphy EA, Carver WE. Potential of Plant-Derived Compounds in Preventing and Reversing Organ Fibrosis and the Underlying Mechanisms. Cells 2024; 13:421. [PMID: 38474385 PMCID: PMC10930795 DOI: 10.3390/cells13050421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/15/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Increased production of extracellular matrix is a necessary response to tissue damage and stress. In a normal healing process, the increase in extracellular matrix is transient. In some instances; however, the increase in extracellular matrix can persist as fibrosis, leading to deleterious alterations in organ structure, biomechanical properties, and function. Indeed, fibrosis is now appreciated to be an important cause of mortality and morbidity. Extensive research has illustrated that fibrosis can be slowed, arrested or even reversed; however, few drugs have been approved specifically for anti-fibrotic treatment. This is in part due to the complex pathways responsible for fibrogenesis and the undesirable side effects of drugs targeting these pathways. Natural products have been utilized for thousands of years as a major component of traditional medicine and currently account for almost one-third of drugs used clinically worldwide. A variety of plant-derived compounds have been demonstrated to have preventative or even reversal effects on fibrosis. This review will discuss the effects and the underlying mechanisms of some of the major plant-derived compounds that have been identified to impact fibrosis.
Collapse
Affiliation(s)
- Patrícia dos Santos Azeredo
- Laboratory of Atherosclerosis, Thrombosis and Cell Therapy, Institute of Biology, State University of Campinas—UNICAMP Campinas, Campinas 13083-970, Brazil;
| | - Daping Fan
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA;
| | - E. Angela Murphy
- Department of Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC 29209, USA;
| | - Wayne E. Carver
- Department of Cell Biology and Anatomy, School of Medicine, University of South Carolina, Columbia, SC 29209, USA;
| |
Collapse
|
13
|
Li D, Cao D, Sun Y, Cui Y, Zhang Y, Jiang J, Cao X. The roles of epigallocatechin gallate in the tumor microenvironment, metabolic reprogramming, and immunotherapy. Front Immunol 2024; 15:1331641. [PMID: 38348027 PMCID: PMC10859531 DOI: 10.3389/fimmu.2024.1331641] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/15/2024] [Indexed: 02/15/2024] Open
Abstract
Cancer, a disease that modern medicine has not fully understood and conquered, with its high incidence and mortality, deprives countless patients of health and even life. According to global cancer statistics, there were an estimated 19.3 million new cancer cases and nearly 10 million cancer deaths in 2020, with the age-standardized incidence and mortality rates of 201.0 and 100.7 per 100,000, respectively. Although remarkable advancements have been made in therapeutic strategies recently, the overall prognosis of cancer patients remains not optimistic. Consequently, there are still many severe challenges to be faced and difficult problems to be solved in cancer therapy today. Epigallocatechin gallate (EGCG), a natural polyphenol extracted from tea leaves, has received much attention for its antitumor effects. Accumulating investigations have confirmed that EGCG can inhibit tumorigenesis and progression by triggering apoptosis, suppressing proliferation, invasion, and migration, altering tumor epigenetic modification, and overcoming chemotherapy resistance. Nevertheless, its regulatory roles and biomolecular mechanisms in the immune microenvironment, metabolic microenvironment, and immunotherapy remain obscure. In this article, we summarized the most recent updates about the effects of EGCG on tumor microenvironment (TME), metabolic reprogramming, and anti-cancer immunotherapy. The results demonstrated EGCG can promote the anti-cancer immune response of cytotoxic lymphocytes and dendritic cells (DCs), attenuate the immunosuppression of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs), and inhibit the tumor-promoting functions of tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), and various stromal cells including cancer-associated fibroblasts (CAFs), endothelial cells (ECs), stellate cells, and mesenchymal stem/stromal cells (MSCs). Additionally, EGCG can suppress multiple metabolic reprogramming pathways, including glucose uptake, aerobic glycolysis, glutamine metabolism, fatty acid anabolism, and nucleotide synthesis. Finally, EGCG, as an immunomodulator and immune checkpoint blockade, can enhance immunotherapeutic efficacy and may be a promising candidate for antitumor immunotherapy. In conclusion, EGCG plays versatile regulatory roles in TME and metabolic reprogramming, which provides novel insights and combined therapeutic strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Dongming Li
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Donghui Cao
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Yuanlin Sun
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yingnan Cui
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yangyu Zhang
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Jing Jiang
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, China
| | - Xueyuan Cao
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Hu Q, Zhang W, Wei F, Huang M, Shu M, Song D, Wen J, Wang J, Nian Q, Ma X, Zeng J, Zhao Y. Human diet-derived polyphenolic compounds and hepatic diseases: From therapeutic mechanisms to clinical utilization. Phytother Res 2024; 38:280-304. [PMID: 37871899 DOI: 10.1002/ptr.8043] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/12/2023] [Accepted: 10/01/2023] [Indexed: 10/25/2023]
Abstract
This review focuses on the potential ameliorative effects of polyphenolic compounds derived from human diet on hepatic diseases. It discusses the molecular mechanisms and recent advancements in clinical applications. Edible polyphenols have been found to play a therapeutic role, particularly in liver injury, liver fibrosis, NAFLD/NASH, and HCC. In the regulation of liver injury, polyphenols exhibit anti-inflammatory and antioxidant effects, primarily targeting the TGF-β, NF-κB/TLR4, PI3K/AKT, and Nrf2/HO-1 signaling pathways. In the regulation of liver fibrosis, polyphenolic compounds effectively reverse the fibrotic process by inhibiting the activation of hepatic stellate cells (HSC). Furthermore, polyphenolic compounds show efficacy against NAFLD/NASH by inhibiting lipid oxidation and accumulation, mediated through the AMPK, SIRT, and PPARγ pathways. Moreover, several polyphenolic compounds exhibit anti-HCC activity by suppressing tumor cell proliferation and metastasis. This inhibition primarily involves blocking Akt and Wnt signaling, as well as inhibiting the epithelial-mesenchymal transition (EMT). Additionally, clinical trials and nutritional evidence support the notion that certain polyphenols can improve liver disease and associated metabolic disorders. However, further fundamental research and clinical trials are warranted to validate the efficacy of dietary polyphenols.
Collapse
Affiliation(s)
- Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Wenwen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Feng Wei
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meilan Huang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mengyao Shu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianxia Wen
- School of Food and Bioengineering, Xihua University, Chengdu, China
| | - Jundong Wang
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qing Nian
- Department of Blood Transfusion, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanling Zhao
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
15
|
Tang Z, Xia Z, Wang X, Liu Y. The critical role of osteopontin (OPN) in fibrotic diseases. Cytokine Growth Factor Rev 2023; 74:86-99. [PMID: 37648616 DOI: 10.1016/j.cytogfr.2023.08.007] [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: 07/18/2023] [Revised: 08/22/2023] [Accepted: 08/22/2023] [Indexed: 09/01/2023]
Abstract
Fibrosis is a pathological condition characterized by the excessive deposition of extracellular matrix components in tissues and organs, leading to progressive architectural remodelling and contributing to the development of various diseases. Osteopontin (OPN), a highly phosphorylated glycoprotein, has been increasingly recognized for its involvement in the progression of tissue fibrosis. This review provides a comprehensive overview of the genetic and protein structure of OPN and focuses on our current understanding of the role of OPN in the development of fibrosis in the lungs and other tissues. Additionally, special attention is given to the potential of OPN as a biomarker and a novel therapeutic target in the treatment of fibrosis.
Collapse
Affiliation(s)
- Ziyi Tang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zijing Xia
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiangpeng Wang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Rheumatology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100000, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; Rare Diseases Center, Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
16
|
Niu W, Zhu M, Wang M, Zhang G, Zheng C, Bao Y, Li Y, Zhang N, Wang J, He H, Wang Y. Discovery and development of benzene sulfonamide derivatives as anti-hepatic fibrosis agents. Bioorg Med Chem Lett 2023; 88:129290. [PMID: 37080476 DOI: 10.1016/j.bmcl.2023.129290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/14/2023] [Accepted: 04/15/2023] [Indexed: 04/22/2023]
Abstract
A novel benzene sulfonamide compound named IMB16-4 exhibits excellent anti-hepatic fibrosis activity in a recent study. To develop potential anti-hepatic fibrosis agents, a series of benzene sulfonamide derivatives were designed and synthesized based on the scaffold of the lead compound IMB16-4. As it turned out, most of the derivatives displayed potential anti-hepatic fibrosis activity, among which, compounds 11a, 11b, 11d, 13a, 36b, and 47b exhibited inhibition rates of 42.3%, 48.7%, 42.4%, 40.0%, 39.4%, and 49.3%, respectively, which were equivalent to the control IMB16-4 with an inhibition rate of 35.9%, Costunolide with an inhibition rate of 45.4%, and much more potent than that of Epigallocatechin gallate (EGCG) with an inhibition rate of 25.3%. Especially, compounds 46a, 46b, and 46c exhibited excellent anti-hepatic fibrosis activity with inhibition rates of 61.7%, 54.8%, and 60.7%, which were almost 1.5-fold inhibition rates of IMB16-4. In addition, compounds 46a, 46b, and 46c exhibited remarkable inhibitory activity in the gene expression of COL1A1, MMP-2, and the protein expression of COL1A1, FN, α-SMA, and TIMP-1 by inhibiting the JAK1-STAT1/3 pathway. These findings furnished valuable inspiration for the further development of anti-hepatic fibrosis agents.
Collapse
Affiliation(s)
- Weiping Niu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Mei Zhu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Minghua Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Guoning Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Chenghong Zheng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yunyang Bao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yiming Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Na Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Juxian Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Hongwei He
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China
| | - Yucheng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
17
|
Sheng Y, Sun Y, Tang Y, Yu Y, Wang J, Zheng F, Li Y, Sun Y. Catechins: Protective mechanism of antioxidant stress in atherosclerosis. Front Pharmacol 2023; 14:1144878. [PMID: 37033663 PMCID: PMC10080012 DOI: 10.3389/fphar.2023.1144878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 03/15/2023] [Indexed: 04/11/2023] Open
Abstract
Tea has long been valued for its health benefits, especially its potential to prevent and treat atherosclerosis (AS). Abnormal lipid metabolism and oxidative stress are major factors that contribute to the development of AS. Tea, which originated in China, is believed to help prevent AS. Research has shown that tea is rich in catechins, which is considered a potential source of natural antioxidants. Catechins are the most abundant antioxidants in green tea, and are considered to be the main compound responsible for tea's antioxidant activity. The antioxidant properties of catechins are largely dependent on the structure of molecules, and the number and location of hydroxyl groups or their substituents. As an exogenous antioxidant, catechins can effectively eliminate lipid peroxidation products. They can also play an antioxidant role indirectly by activating the endogenous antioxidant system by regulating enzyme activity and signaling pathways. In this review, we summarized the preventive effect of catechin in AS, and emphasized that improving the antioxidant effect and lipid metabolism disorders of catechins is the key to managing AS.
Collapse
Affiliation(s)
| | - Yizhuo Sun
- *Correspondence: Fengjie Zheng, ; Yuhang Li, ; Yan Sun,
| | | | | | | | - Fengjie Zheng
- *Correspondence: Fengjie Zheng, ; Yuhang Li, ; Yan Sun,
| | - Yuhang Li
- *Correspondence: Fengjie Zheng, ; Yuhang Li, ; Yan Sun,
| | - Yan Sun
- *Correspondence: Fengjie Zheng, ; Yuhang Li, ; Yan Sun,
| |
Collapse
|
18
|
Zhou Z, Li K, Guo J, Wang Y, Wei Y, Duan J, Chen M, Shi L, Hu W. Green Tea Catechin EGCG Ameliorates Thioacetamide-Induced Hepatic Encephalopathy in Rats via Modulation of the Microbiota-Gut-Liver Axis. Mol Nutr Food Res 2022; 67:e2200821. [PMID: 36573265 DOI: 10.1002/mnfr.202200821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 11/23/2022] [Indexed: 12/28/2022]
Abstract
SCOPE Existing research suggests that (-)-epigallocatechin-3-gallate (EGCG), which is a natural tea catechin active substance, can protect against liver injury. However, its mechanism for hepatic encephalopathy (HE) treatment is still unclear. In this study, the role of EGCG in the amelioration of HE rats and the effect on the microbiota-gut-liver axis are mainly analyzed. METHODS AND RESULTS Thioacetamide (TAA) is employed to induce the HE model in rats. The results of open field test show that EGCG restores locomotor activity and exploratory behavior. Histological and biochemical results demonstrate that EGCG ameliorates brain and liver damage, decreases the expression of pro-inflammatory cytokines, and increases the activity of antioxidant enzymes. Meanwhile, EGCG modulates the Nrf2 pathway and TLR4/NF-κB pathway to mitigate TAA-induced oxidative stress and inflammatory responses. Immunohistochemistry reveals protection of the intestinal barrier by EGCG upregulating the expression of occludin and zonula occludens-1. Furthermore, serum levels of ammonia and LPS are reduced. 16S rRNA analysis shows that EGCG treatment increases the abundance of beneficial bacteria (e.g., Bifidobacterium, Lactobacillus, and Limosilactobacillus). CONCLUSION The above results reveal that EGCG has anti-oxidative stress and anti-inflammatory effects, and ameliorates the condition through the microbiota-gut-liver axis, with potential for the treatment of HE.
Collapse
Affiliation(s)
- Zhengming Zhou
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ke Li
- Department of Clinical Nutrition, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jiankui Guo
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunfeng Wang
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yaoyao Wei
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Juan Duan
- Department of Nutrition and Food Hygiene, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Muxi Chen
- Department of Clinical Nutrition, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lei Shi
- Department of Clinical Nutrition, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wen Hu
- Department of Clinical Nutrition, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
19
|
Nomura M, George J, Hashizume C, Saito T, Ueda Y, Ishigaki Y, Tsuchishima M, Tsutsumi M. Surgical implantation of human adipose derived stem cells attenuates experimentally induced hepatic fibrosis in rats. Mol Med 2022; 28:143. [PMID: 36447136 PMCID: PMC9706981 DOI: 10.1186/s10020-022-00566-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 11/03/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are multipotent stromal cells and could exert hepatoprotective effects against acute liver injury, steatohepatitis, and fibrogenesis. Here, we evaluated the effects of human adipose derived stem cells (hADSCs) to attenuate experimentally induced hepatic fibrosis and early cirrhosis in rats. METHODS Hepatic fibrosis was induced by intraperitoneal injections of CCl4 (0.1 ml/100 g body weight) twice a week for 8 weeks. hADSCs were isolated and cultured on polyethylene discs coated with hydroxyapatite and 2 cm diameter disc was surgically implanted on the right lateral lobe of the liver. Discs implanted without hADSCs served as control. The animals were injected again with CCl4 once a week for another 8 weeks. All the animals were sacrificed at the end of 16th week. RESULTS Serial administrations of CCl4 resulted in well developed fibrosis and early cirrhosis at 8th week which maintained until the 16th week. Animals treated with hADSC discs depicted over 50% decrease of collagen with significant increase in serum albumin and total protein levels. Immunohistochemical staining for TGF-β1, α-smooth muscle actin, and collagen type I and type III demonstrated marked decrease compared to the animals without hADSC treatment. CONCLUSIONS Treatment with hADSCs improved liver functions, markedly reduced hepatic fibrosis and early cirrhosis. Various pleiotropic and paracrine factors secreted from the hADSCs seem to serve as reparative functions in the attenuation of liver cirrhosis. The data demonstrated that treatment with hADSCs can be successfully used as a potent therapeutic method to prevent progression of hepatic fibrosis and related adverse events.
Collapse
Affiliation(s)
- Masateru Nomura
- grid.411998.c0000 0001 0265 5359Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293 Japan
| | - Joseph George
- grid.411998.c0000 0001 0265 5359Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293 Japan ,grid.510345.60000 0004 6004 9914Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Ishikawa 920-0293 Japan
| | - Chieko Hashizume
- grid.411998.c0000 0001 0265 5359Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293 Japan
| | - Takashi Saito
- grid.411998.c0000 0001 0265 5359Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293 Japan
| | - Yoshimichi Ueda
- grid.411998.c0000 0001 0265 5359Department of Pathology II, Kanazawa Medical University, Uchinada, Ishikawa 920-0293 Japan
| | - Yasuhito Ishigaki
- grid.510345.60000 0004 6004 9914Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Ishikawa 920-0293 Japan ,grid.411998.c0000 0001 0265 5359Medical Research Institute, Kanazawa Medical University, Uchinada, Ishikawa 920-0293 Japan
| | - Mutsumi Tsuchishima
- grid.411998.c0000 0001 0265 5359Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293 Japan
| | - Mikihiro Tsutsumi
- grid.411998.c0000 0001 0265 5359Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293 Japan ,grid.510345.60000 0004 6004 9914Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Ishikawa 920-0293 Japan
| |
Collapse
|
20
|
Wang JR, Song XH, Li LY, Gao SJ, Shang FH, Zhang XM, Yang Y. Metabolomic analysis reveals dynamic changes in secondary metabolites of Sophora japonica L. during flower maturation. FRONTIERS IN PLANT SCIENCE 2022; 13:916410. [PMID: 35991425 PMCID: PMC9386383 DOI: 10.3389/fpls.2022.916410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
Sophora japonica L. is widely consumed in China because of its medicinal and nutritional value. Its quality is greatly affected by the accumulation of metabolites, which varies with the stage of flower development. However, changes in the characteristics of the secondary metabolites during flower maturity remain unclear. Ultra-high-performance liquid chromatography coupled with electrospray ionization-triple quadrupole-linear ion trap mass spectrometry (UPLC-ESI-QTRAP-MS/MS) revealed dynamic changes in the secondary metabolites of S. japonica during the five flower-maturity stages. We monitored 331 metabolites and screened 164. The differential metabolites showed seven trends during flower maturation, with flavonoids and phenolic acids having the most varied expressions. Flower buds (S2-S3) are rich in flavonoids and are thus suitable for use in high-quality medicine or industrial extraction. Our study provides an empirical basis for the informed harvesting of S. japonica based on its mode of utilization.
Collapse
Affiliation(s)
- Ji-Rui Wang
- Three Grade Laboratory of Chinese Medicine Chemistry, Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Chongqing Sub-Center of National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Chongqing, China
| | - Xu-Hong Song
- Three Grade Laboratory of Chinese Medicine Chemistry, Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Chongqing Sub-Center of National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Chongqing, China
| | - Long-Yun Li
- Three Grade Laboratory of Chinese Medicine Chemistry, Chongqing Academy of Chinese Materia Medica, Chongqing, China
- Chongqing Sub-Center of National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Chongqing, China
| | - Si-Jia Gao
- Three Grade Laboratory of Chinese Medicine Chemistry, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Fang-Hong Shang
- Three Grade Laboratory of Chinese Medicine Chemistry, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Xiao-Mei Zhang
- Three Grade Laboratory of Chinese Medicine Chemistry, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| | - Yong Yang
- Three Grade Laboratory of Chinese Medicine Chemistry, Chongqing Academy of Chinese Materia Medica, Chongqing, China
| |
Collapse
|