1
|
Szternel Ł, Sobucki B, Wieprzycka L, Krintus M, Panteghini M. Golgi protein 73 in liver fibrosis. Clin Chim Acta 2025; 565:119999. [PMID: 39401651 DOI: 10.1016/j.cca.2024.119999] [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: 09/11/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/17/2024]
Abstract
Golgi protein 73 (GP73) is implicated in key pathogenic processes, particularly those related to inflammation and fibrogenesis. In the last years, its measurement has emerged as a promising biomarker for detection of liver fibrosis (LF), a common consequence of chronic liver disease that can progress to cirrhosis and eventually hepatocellular carcinoma. GP73 concentrations in blood appear significantly increased in LF patients, correlating with disease severity, making this biomarker a possible non-invasive alternative for detecting and monitoring this condition regardless of etiology. Understanding the molecular mechanisms involving GP73 expression could also lead to new therapeutic strategies aimed at modulating its synthesis or function to prevent or reverse LF. Despite its clinical potential, GP73 as a LF biomarker faces several challenges. The lack of demonstrated comparability among different assays as well as the lack of knowledge of individual variability can make difficult the result interpretation. Further research is therefore needed focusing on robust clinical validation of GP73 as a LF biomarker. Addressing analytical, biological, and clinical limitations will be critical to exploiting its potential for improving detection and monitoring of advanced LF.
Collapse
Affiliation(s)
- Łukasz Szternel
- Department of Laboratory Medicine, Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Poland
| | - Bartłomiej Sobucki
- Department of Laboratory Medicine, Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Poland
| | - Laura Wieprzycka
- Department of Laboratory Medicine, Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Poland
| | - Magdalena Krintus
- Department of Laboratory Medicine, Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Poland.
| | - Mauro Panteghini
- Department of Laboratory Medicine, Nicolaus Copernicus University in Torun, Collegium Medicum in Bydgoszcz, Poland
| |
Collapse
|
2
|
Sadri M, Shafaghat Z, Roozbehani M, Hoseinzadeh A, Mohammadi F, Arab FL, Minaeian S, Fard SR, Faraji F. Effects of Probiotics on Liver Diseases: Current In Vitro and In Vivo Studies. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10431-z. [PMID: 39739162 DOI: 10.1007/s12602-024-10431-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2024] [Indexed: 01/02/2025]
Abstract
Various types of liver or hepatic diseases cause the death of about 2 million people worldwide every year, of which 1 million die from the complications of cirrhosis and another million from hepatocellular carcinoma and viral hepatitis. Currently, the second most common solid organ transplant is the liver, and the current rate represents less than 10% of global transplant requests. Hence, finding new approaches to treat and prevent liver diseases is essential. In liver diseases, the interaction between the liver, gut, and immune system is crucial, and probiotics positively affect the human microbiota. Probiotics are a non-toxic and biosafe alternative to synthetic chemical compounds. Health promotion by lowering cholesterol levels, stimulating host immunity, the natural gut microbiota, and other functions are some of the activities of probiotics, and their metabolites, including bacteriocins, can exert antimicrobial effects against a broad range of pathogenic bacteria. The present review discusses the available data on the results of preclinical and clinical studies on the effects of probiotic administration on different types of liver diseases.
Collapse
Affiliation(s)
- Maryam Sadri
- Department of Immunology, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Shafaghat
- Department of Immunology, Iran University of Medical Sciences, Tehran, Iran
| | - Mona Roozbehani
- Vaccine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Akram Hoseinzadeh
- Cancer Research Center, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Fatemeh Mohammadi
- Department of Immunology, School of Medicine, Mashhad University of Medicine Sciences, Mashhad, Iran
| | - Fahimeh Lavi Arab
- Department of Immunology, School of Medicine, Mashhad University of Medicine Sciences, Mashhad, Iran
| | - Sara Minaeian
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medicine Sciences, Tehran, Iran
| | - Soheil Rahmani Fard
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medicine Sciences, Tehran, Iran
| | - Fatemeh Faraji
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medicine Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Wei B, Huang J, Zhang Y, Hu X, Ma C, Li Y, Chen P. Restoration of RECK expression attenuates liver fibrosis induced by carbon tetrachloride through the Nrf2-MMP9 axis. Int Immunopharmacol 2024; 143:113475. [PMID: 39476567 DOI: 10.1016/j.intimp.2024.113475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/08/2024] [Accepted: 10/20/2024] [Indexed: 11/28/2024]
Abstract
Liver fibrosis is a reversible process that can be delayed or even reversed through appropriate intervention during its development. The protein RECK, encoded by the Reck gene, regulates matrix metalloproteinase (MMP) activity and plays a crucial role in extracellular matrix (ECM) degradation and remodeling. Reduced RECK expression is found in various fibrotic tissues. However, the impact of restoring RECK expression on the development and progression of liver fibrosis has not yet been determined. This study found that the restoration of RECK expression attenuated TGF-β1-induced hepatic stellate cell (HSC) activation and mitigated carbon tetrachloride (CCl4)-induced acute liver injury. In a mouse model of liver fibrosis induced by CCl4, restoration of RECK expression reduced the degree of fibrosis, collagen deposition, and level of oxidative stress. RECK competes with Nrf2 for binding to Keap1, resulting in a decrease in the degradation of Nrf2 by Keap1 and an increase in the accumulation of Nrf2 in the cytoplasm. Under oxidative stress conditions, Nrf2 can be translocated to the nucleus for expression, initiating an antioxidant stress response, furthermore, Nrf2 can also activate MMP-9 and degrade the over-deposited collagen, thereby achieving the effect of alleviating liver fibrosis. Our study reveals a novel mechanism by which restoration of RECK expression ameliorates liver fibrosis, providing a promising target for combating liver fibrosis.
Collapse
Affiliation(s)
- Bizhen Wei
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China
| | - Jing Huang
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China; Department of Respiratory and Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yu Zhang
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China
| | - Xiuxiu Hu
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China
| | - Cao Ma
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China; Department of Pathology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yiping Li
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China.
| | - Pingsheng Chen
- Department of Pathology, School of Medicine, Southeast University, Nanjing, China; Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| |
Collapse
|
4
|
Wei Z, Liu J, Wang N, Wei K. Kidney function mediates the association of per- and poly-fluoroalkyl substances (PFAS) and heavy metals with hepatic fibrosis risk. ENVIRONMENTAL RESEARCH 2024; 263:120092. [PMID: 39357638 DOI: 10.1016/j.envres.2024.120092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/26/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
Heavy metals and per- and polyfluoroalkyl substances (PFAS) are significantly associated with the risk of hepatic fibrosis. However, the potential mediating effect of kidney function in the relationship between heavy metals, PFAS, and hepatic fibrosis risk remains unexplored. This research gap limits the development of hepatic fibrosis prevention and treatment strategies. To address this, this study conducts a cross-sectional analysis based on data from 10,870 participants in NHANES 2005-2018 to explore the relationship between heavy metals, PFAS, and the risk of hepatic fibrosis, as well as the mediating effect of kidney function. Participants with a Fibrosis-4 index <1.45 are defined as not having hepatic fibrosis in this study. Results from generalized linear regression models and weighted quantile sum regression models indicate that both individual and combined exposures to heavy metals and PFAS are positively associated with the risk of hepatic fibrosis. Nonlinear exposure-response functions suggest that there may be a threshold for the relationship between heavy metals (except mercury) and PFAS with the risk of hepatic fibrosis. Furthermore, heavy metals and PFAS increase the risk of kidney function impairment. After stratification by kidney function stage, the relationship between heavy metals (except lead) and proteinuria is not significant, while PFAS show a significant negative association with proteinuria. The decline in kidney function has a significant mediating effect in the relationship between heavy metals and PFAS and the risk of hepatic fibrosis, with mediation effect proportions all above 20%. The findings suggest that individual or combined exposure to heavy metals and PFAS does not increase the risk of hepatic fibrosis until a certain threshold is reached, and the mediating role of declining kidney function is very important. These results highlight the need to consider kidney function in the context of hepatic fibrosis risk assessment and management.
Collapse
Affiliation(s)
- Zhengqi Wei
- School of Public Health, Guilin Medical University, Guilin, Guangxi, 541199, China
| | - Jincheng Liu
- Huazhong University of Science and Technology Tongji Medical College, Wuhan, Hubei, 430000, China
| | - Na Wang
- School of Public Health, Guilin Medical University, Guilin, Guangxi, 541199, China.
| | - Keke Wei
- Huazhong University of Science and Technology Tongji Medical College, Wuhan, Hubei, 430000, China.
| |
Collapse
|
5
|
Liu X, Cai Y, Zhang Y, Zhang H, Tian S, Gong Y, Song Q, Chen X, Ma X, Wen Y, Chen Y, Zeng J. Artesunate: A potential drug for the prevention and treatment from hepatitis to hepatocellular carcinoma. Pharmacol Res 2024; 210:107526. [PMID: 39617278 DOI: 10.1016/j.phrs.2024.107526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/14/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Liver cancer represents a multifactorial, multistage, and intricately progressive malignancy. Over the past decade, artesunate (ART), initially renowned for its anti-malarial efficacy, has been the focus of over 3000 studies uncovering its diverse pharmacological actions, including anti-inflammatory, immunoregulatory, metabolic regulatory, anti-fibrotic, and anti-cancer properties. This review highlights ART's role in the multistep progression from hepatitis to cancer and its underlying regulatory mechanisms, revealing signal transducer and activator of transcription 3 (STAT3) and ferroptosis (a novel form of programmed cell death) as promising therapeutic targets. ART demonstrates efficacy in inhibiting hepatitis virus infections, modulating inflammation, and facilitating recovery from inflammatory processes. During stages of hepatic fibrosis or cirrhosis, ART reverses fibrotic and cirrhotic changes by suppressing hepatic stellate cell activity, regulating inflammatory pathways, inhibiting hematopoietic stem cell proliferation, and inducing ferroptosis. Additionally, ART hinders hepatocellular carcinoma (HCC) cell proliferation, invasion, and metastasis, induces apoptosis and autophagy, combats drug resistance, and enhances chemosensitivity. Collectively, ART exhibits multi-step actions across multiple targets and signaling pathways, highlighting its potential as a clinical candidate for the prevention and treatment of liver cancer, from hepatitis and hepatic fibrosis to advanced HCC.
Collapse
Affiliation(s)
- Xinyue Liu
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China; Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Yilin Cai
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China; Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Yuanhao Zhang
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China
| | - Hetian Zhang
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China
| | - Sisi Tian
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China; Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Yuxia Gong
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China; Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Qinmei Song
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China; Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Xiaotong Chen
- School of Clinical Medicine, Chengdu University of Chinese Medicine, Chengdu 610075, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yu Chen
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| |
Collapse
|
6
|
Cen X, Wang W, Hong S, Wang Q, Wang N, Mo L, Li J, Li J. Integrated microbiome and metabolomic analyses revealed the antifibrotic effect of vanillic acid on thioacetamide-induced liver fibrosis in mice. Food Funct 2024; 15:11780-11794. [PMID: 39545308 DOI: 10.1039/d4fo02309a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Vanillic acid (VA) is a natural phenolic acid compound that is widely found in various foods and medicinal plants, with a remarkable antifibrotic effect observed in animal studies, but its exact antifibrotic mechanism remains unclear. Herein, hepatic function, fibrotic index, and histopathological, microbiome, and metabolomic methods were used to investigate the potential mechanisms behind the improvement effect of vanillic acid against thioacetamide (TAA)-induced liver fibrosis in mice. Our results showed that VA reversed TAA-induced liver fibrosis manifested a decrease in collagen fiber deposition, serum transaminase, serum hepatic fibrotic index, and liver inflammation indicator levels. When analyzed, TAA injection mainly increased the abundance of Akkermansia and Roseburia and significantly reduced the abundance of Anaerotruncus. VA reversed these changes back to normal levels to varying degrees. Metabolomic profiling demonstrated that VA treatment was efficacious in modulating several key liver metabolites involved in neuroactive ligand-receptor interaction, prolactin signaling pathway, estrogen signaling pathway, and glutathione metabolism. Conclusively, VA may ameliorate liver damage and suppress the fibrogenesis caused by thioacetamide by correcting intestinal microbiota disorders and promoting normal hepatic metabolism. This research provides a novel perspective on vanillic acid as a dietary supplement for hepatic fibrosis improvement.
Collapse
Affiliation(s)
- Xiaofeng Cen
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, China.
- Guangxi Health Commission Key Laboratory of Entire Lifecycle Health and Care, School of Public Health, Guilin Medical University, 541199, Guilin, China
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, 541199, Guilin, China
| | - Wei Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, China.
- Guangxi Health Commission Key Laboratory of Entire Lifecycle Health and Care, School of Public Health, Guilin Medical University, 541199, Guilin, China
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, 541199, Guilin, China
| | - Siyan Hong
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, China.
- Guangxi Health Commission Key Laboratory of Entire Lifecycle Health and Care, School of Public Health, Guilin Medical University, 541199, Guilin, China
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, 541199, Guilin, China
| | - Qin Wang
- Guangxi Health Commission Key Laboratory of Entire Lifecycle Health and Care, School of Public Health, Guilin Medical University, 541199, Guilin, China
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, 541199, Guilin, China
| | - Na Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, China.
- Guangxi Health Commission Key Laboratory of Entire Lifecycle Health and Care, School of Public Health, Guilin Medical University, 541199, Guilin, China
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, 541199, Guilin, China
| | - Ling Mo
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, China.
- Guangxi Health Commission Key Laboratory of Entire Lifecycle Health and Care, School of Public Health, Guilin Medical University, 541199, Guilin, China
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, 541199, Guilin, China
| | - Jingjing Li
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, China.
- Guangxi Health Commission Key Laboratory of Entire Lifecycle Health and Care, School of Public Health, Guilin Medical University, 541199, Guilin, China
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, 541199, Guilin, China
| | - Jingwen Li
- Department of Nutrition and Food Hygiene, School of Public Health, Guilin Medical University, Guilin, China.
- Guangxi Health Commission Key Laboratory of Entire Lifecycle Health and Care, School of Public Health, Guilin Medical University, 541199, Guilin, China
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, School of Public Health, Guilin Medical University, 541199, Guilin, China
| |
Collapse
|
7
|
Sokal-Dembowska A, Jarmakiewicz-Czaja S, Filip R. Flavonoids and Their Role in Preventing the Development and Progression of MAFLD by Modifying the Microbiota. Int J Mol Sci 2024; 25:11187. [PMID: 39456969 PMCID: PMC11508831 DOI: 10.3390/ijms252011187] [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: 09/25/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
With the increasing prevalence and serious health consequences of metabolic-associated fatty liver disease (MAFLD), early diagnosis and intervention are key to effective treatment. Recent studies highlight the important role of dietary factors, including the use of flavonoids, in improving liver health. These compounds possess anti-inflammatory, antioxidant, and liver-protective properties. Flavonoids have been shown to affect the gut microbiota, which plays a key role in liver function and disease progression. Therefore, their role in preventing the development and progression of MAFLD through modulation of the microbiome seems to be of interest. This narrative review aims to consolidate the current evidence on the effects of selected flavonoids on MAFLD progression, their potential mechanisms of action, and the implications for the development of personalized dietary interventions for the management of liver disease.
Collapse
Affiliation(s)
- Aneta Sokal-Dembowska
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland.; (S.J.-C.)
| | - Sara Jarmakiewicz-Czaja
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland.; (S.J.-C.)
| | - Rafał Filip
- Institute of Medicine, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
- Department of Gastroenterology with IBD Unit, Clinical Hospital No. 2, 35-301 Rzeszow, Poland
| |
Collapse
|
8
|
Liu X, Xia N, Yu Q, Jin M, Wang Z, Fan X, Zhao W, Li A, Jiang Z, Zhang L. Silybin Meglumine Mitigates CCl 4-Induced Liver Fibrosis and Bile Acid Metabolism Alterations. Metabolites 2024; 14:556. [PMID: 39452937 PMCID: PMC11509150 DOI: 10.3390/metabo14100556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Altered patterns of bile acids (BAs) are frequently present in liver fibrosis, and BAs function as signaling molecules to initiate inflammatory responses. Silybin meglumine (SLB-M) is widely used in treating various liver diseases including liver fibrosis. However, research on its effects on bile acid (BA) metabolism is limited. This study investigated the therapeutic effects of SLB-M on liver fibrosis and BA metabolism in a CCl4-induced murine model. METHODS A murine liver fibrosis model was induced by CCl4. Fibrosis was evaluated using HE, picrosirius red, and Masson's trichrome staining. Liver function was assessed by serum and hepatic biochemical markers. Bile acid (BA) metabolism was analyzed using LC-MS/MS. Bioinformatics analyses, including PPI network, GO, and KEGG pathway analyses, were employed to explore molecular mechanisms. Gene expression alterations in liver tissue were examined via qRT-PCR. RESULTS SLB-M treatment resulted in significant histological improvements in liver tissue, reducing collagen deposition and restoring liver architecture. Biochemically, SLB-M not only normalized serum liver enzyme levels (ALT, AST, TBA, and GGT) but also mitigated disruptions in both systemic and hepatic BA metabolism by increased unconjugated BAs like cholic acid and chenodeoxycholic acid but decreased conjugated BAs including taurocholic acid and taurodeoxycholic acid, compared to that in CCl4-induced murine model. Notably, SLB-M efficiently improved the imbalance of BA homeostasis in liver caused by CCl4 via activating Farnesoid X receptor. CONCLUSIONS These findings underscore SLB-M decreased inflammatory response, reconstructed BA homeostasis possibly by regulating key pathways, and gene expressions in BA metabolism.
Collapse
Affiliation(s)
- Xiaoxin Liu
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Ninglin Xia
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Qinwei Yu
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Ming Jin
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Zifan Wang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Xue Fan
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Wen Zhao
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Anqin Li
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Zhenzhou Jiang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
| | - Luyong Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (X.L.); (N.X.); (Q.Y.); (M.J.); (Z.W.); (X.F.); (W.Z.); (A.L.)
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
9
|
Obmann VC, Ardoino M, Klaus J, Catucci D, Berzigotti A, Montani M, Peters A, Todorski I, Wagner B, Zbinden L, Gräni C, Ebner L, Heverhagen JT, Christe A, Huber AT. MRI Extracellular Volume Fraction in Liver Fibrosis-A Comparison of Different Time Points and Blood Pool Measurements. J Magn Reson Imaging 2024; 60:1678-1688. [PMID: 38553860 DOI: 10.1002/jmri.29259] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/12/2024] [Accepted: 01/14/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Extracellular volume (ECV) correlates with the degree of liver fibrosis. PURPOSE To analyze the performance of liver MRI-based ECV evaluations with different blood pool measurements at different time points. STUDY TYPE Prospective. SAMPLE 73 consecutive patients (n = 31 females, mean age 56 years) with histopathology-proven liver fibrosis. FIELD STRENGTH/SEQUENCE 3T acquisition within 90 days of biopsy, including shortened modified look-locker inversion recovery T1 mapping. ASSESSMENT Polygonal regions of interest were manually drawn in the liver, aorta, vena cava, and in the main, left and right portal vein on four slices before and after Gd-DOTA administration at 5/10/15 minutes. ECV was calculated 1) on one single slice on portal bifurcation level, and 2) averaged over all four slices. STATISTICAL TESTS Parameters were compared between patients with fibrosis grades F0-2 and F3-F4 with the Mann-Whitney U and fishers exact test. ROC analysis was used to assess the performance of the parameters to predict F3-4 fibrosis. A P-value <0.05 was considered statistically significant. RESULTS ECV was significantly higher in F3-4 fibrosis (35.4% [33.1%-37.6%], 36.1% [34.2%-37.5%], and 37.0% [34.8%-39.2%] at 5/10/15 minutes) than in patients with F0-2 fibrosis (33.3% [30.8%-34.8%], 33.7% [31.6%-34.7%] and 34.9% [32.2%-36.0%]; AUC = 0.72-0.75). Blood pool T1 relaxation times in the aorta and vena cava were longer on the upper vs. lower slices at 5 minutes, but not at 10/15 minutes. AUC values were similar when measured on a single slice (AUC = 0.69-0.72) or based on blood pool measurements in the cava or portal vein (AUC = 0.63-0.67 and AUC = 0.65-0.70). DATA CONCLUSION Liver ECV is significantly higher in F3-4 fibrosis compared to F0-2 fibrosis with blood pool measurements performed in the aorta, inferior vena cava, and portal vein at 5, 10, and 15 minutes. However, a smaller variability was observed for blood pool measurements between slices at 15 minutes. LEVEL OF EVIDENCE 1 TECHNICAL EFFICACY: Stage 3.
Collapse
Affiliation(s)
- Verena Carola Obmann
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Liver Elastography Center, Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, Bern, Switzerland
| | - Marie Ardoino
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Jeremias Klaus
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Institute of Forensic Medicine, University of Bern, Bern, Switzerland
| | - Damiano Catucci
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Annalisa Berzigotti
- Department of Visceral Surgery and Medicine, Inselspital Bern University Hospital, University of Bern, Bern, Switzerland
| | - Matteo Montani
- Department of Pathology, University of Bern, Bern, Switzerland
| | - Alan Peters
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Inga Todorski
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Benedikt Wagner
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Lukas Zbinden
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| | - Christoph Gräni
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Lukas Ebner
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Radiology and Nuclear Medicine (RUN), Luzerner Kantonsspital, University of Lucerne, Switzerland, Lucerne, Switzerland
| | - Johannes Thomas Heverhagen
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
| | - Andreas Christe
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Liver Elastography Center, Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, Bern, Switzerland
| | - Adrian Thomas Huber
- Department of Diagnostic, Interventional and Pediatric Radiology, Inselspital Bern, University Hospital, University of Bern, Bern, Switzerland
- Liver Elastography Center, Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, Bern, Switzerland
- Radiology and Nuclear Medicine (RUN), Luzerner Kantonsspital, University of Lucerne, Switzerland, Lucerne, Switzerland
| |
Collapse
|
10
|
Diwan R, Gaytan SL, Bhatt HN, Pena-Zacarias J, Nurunnabi M. Liver fibrosis pathologies and potentials of RNA based therapeutics modalities. Drug Deliv Transl Res 2024; 14:2743-2770. [PMID: 38446352 DOI: 10.1007/s13346-024-01551-8] [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] [Accepted: 02/14/2024] [Indexed: 03/07/2024]
Abstract
Liver fibrosis (LF) occurs when the liver tissue responds to injury or inflammation by producing excessive amounts of scar tissue, known as the extracellular matrix. This buildup stiffens the liver tissue, hinders blood flow, and ultimately impairs liver function. Various factors can trigger this process, including bloodborne pathogens, genetic predisposition, alcohol abuse, non-steroidal anti-inflammatory drugs, non-alcoholic steatohepatitis, and non-alcoholic fatty liver disease. While some existing small-molecule therapies offer limited benefits, there is a pressing need for more effective treatments that can truly cure LF. RNA therapeutics have emerged as a promising approach, as they can potentially downregulate cytokine levels in cells responsible for liver fibrosis. Researchers are actively exploring various RNA-based therapeutics, such as mRNA, siRNA, miRNA, lncRNA, and oligonucleotides, to assess their efficacy in animal models. Furthermore, targeted drug delivery systems hold immense potential in this field. By utilizing lipid nanoparticles, exosomes, nanocomplexes, micelles, and polymeric nanoparticles, researchers aim to deliver therapeutic agents directly to specific biomarkers or cytokines within the fibrotic liver, increasing their effectiveness and reducing side effects. In conclusion, this review highlights the complex nature of liver fibrosis, its underlying causes, and the promising potential of RNA-based therapeutics and targeted delivery systems. Continued research in these areas could lead to the development of more effective and personalized treatment options for LF patients.
Collapse
Affiliation(s)
- Rimpy Diwan
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA
- Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX, 79968, USA
| | - Samantha Lynn Gaytan
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA
- Department of Interdisciplinary Health Sciences, College of Health Sciences, The University of Texas El Paso, El Paso, Texas, 79968, USA
| | - Himanshu Narendrakumar Bhatt
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA
- Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX, 79968, USA
| | - Jacqueline Pena-Zacarias
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA
- Department of Biological Sciences, College of Science, The University of Texas El Paso, El Paso, Texas, 79968, USA
| | - Md Nurunnabi
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas El Paso, El Paso, TX, 79902, USA.
- Department of Biomedical Engineering, College of Engineering, The University of Texas El Paso, El Paso, TX, 79968, USA.
- Department of Interdisciplinary Health Sciences, College of Health Sciences, The University of Texas El Paso, El Paso, Texas, 79968, USA.
- Border Biomedical Research Center, The University of Texas El Paso, El Paso, TX, 79968, USA.
| |
Collapse
|
11
|
Sánchez-López CM, González-Arce A, Ramírez-Toledo V, Bernal D, Marcilla A. Unraveling new players in helminth pathology: extracellular vesicles from Fasciola hepatica and Dicrocoelium dendriticum exert different effects on hepatic stellate cells and hepatocytes. Int J Parasitol 2024; 54:617-634. [PMID: 38925265 DOI: 10.1016/j.ijpara.2024.06.002] [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: 12/27/2023] [Revised: 03/02/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024]
Abstract
Fasciola hepatica and Dicrocoelium dendriticum are parasitic trematodes residing in the bile ducts of mammalian hosts, causing, in some cases, impairment of liver function and hepatic fibrosis. Previous studies have shown that extracellular vesicles released by F. hepatica (FhEVs) and D. dendriticum (DdEVs) induce a distinct phenotype in human macrophages, but there is limited information on the effect of parasitic EVs on liver cells, which interact directly with the worms in natural infections. In this study, we isolated FhEVs and DdEVs by size exclusion chromatography and labeled them with a lipophilic fluorescent dye to analyze their uptake by human hepatic stellate cells (HSC) and hepatocytes, important cell types in liver pathology, using synthetic liposomes as internal labeling and uptake control. We analyzed EV uptake and the proteome profiles after the treatment with EVs for both cell types. Our results reveal that EVs establish unique and specific interactions with stellate cells and hepatocytes, suggesting a different role of EVs derived from each parasite, depending on the migration route to reach their final niche. FhEVs have a cytostatic effect on HSCs, but induce the extracellular matrix secretion and elicit anti-inflammatory responses in hepatocytes. DdEVs have a more potent anti-proliferative effect than FhEVs and trigger a global inflammatory response, increasing the levels of NF-κB and other inflammatory mediators in both cell types. These interactions may have a major influence on the progression of the disease, serving to generate conditions that may favor the establishment of the helminths in the host.
Collapse
Affiliation(s)
- Christian M Sánchez-López
- Área de Parasitología, Departament de Farmacia i Tecnologia Farmacèutica i Parasitologia. Universitat de València, Burjassot, Valencia, Spain; Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research IIS La Fe-Universitat de València, Valencia, Spain
| | - Aránzazu González-Arce
- Área de Parasitología, Departament de Farmacia i Tecnologia Farmacèutica i Parasitologia. Universitat de València, Burjassot, Valencia, Spain
| | | | - Dolores Bernal
- Departament de Bioquímica i Biologia Molecular, Facultat de Ciències Biològiques, Universitat de València, Burjassot, Valencia, Spain.
| | - Antonio Marcilla
- Área de Parasitología, Departament de Farmacia i Tecnologia Farmacèutica i Parasitologia. Universitat de València, Burjassot, Valencia, Spain; Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research IIS La Fe-Universitat de València, Valencia, Spain.
| |
Collapse
|
12
|
Xu X, Wu Y, Zhao Y, Liu A, Yi C, Zhang A, Wang X. Inhibition of Macrophage Pyroptosis─A New Therapeutic Strategy to Alleviate T-2 Toxin-Induced Subacute Liver Injury by Directly Competing with the Key Target. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:18670-18681. [PMID: 39112929 DOI: 10.1021/acs.jafc.4c03340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Multiple compounds are related to the development of liver injury, such as toxins, drugs, and environmental pollutants. Although there are reports that the T-2 toxin can cause liver injury, its toxic mechanism remains unclear, which further impedes the development of effective antidotes. In this study, CRISPR-Cas9 genome-wide screening technology was used to identify transformation-related protein 53 inducible nuclear protein 1 (trp53inp1) as a toxic target of the T-2 toxin. Mechanism studies have shown that the T-2 toxin induced pyroptosis of macrophages (J774A.1 cells) by activating the trp53inp1/NF-κB/NLRP3/GSDMD-N pathway, leading to a subacute liver injury. Also, the new drug berberine (BER) identified through virtual screening significantly alleviated the subacute liver injury by competitively binding trp53inp1 via His224; the effect was better than those of the positive control drugs N-acetylcysteine (NAC) and disulfiram (DSF). In summary, the above results indicate that trp53inp1 is a key target for T-2 toxin to induce subacute liver injury and that inhibiting macrophage pyroptosis is a new method for treating liver injury. In addition, this study provides a new method and strategy for the discovery of key disease targets and the search for effective drugs.
Collapse
Affiliation(s)
- Xiaoqing Xu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan , Hubei 430070, China
- MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan , Hubei 430070, China
| | - Yue Wu
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan , Hubei 430070, China
| | - Yongxia Zhao
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan , Hubei 430070, China
- MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan , Hubei 430070, China
| | - Aimei Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning 437100, P.R. China
| | - Chenyang Yi
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan , Hubei 430070, China
| | - Anding Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan , Hubei 430070, China
- Guangdong Provincial Key Laboratory of Research on the Technology of Pig-breeding and Pig-disease prevention, Guangzhou, Guangdong 510000, China
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan , Hubei 430070, China
- MAO Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan , Hubei 430070, China
| |
Collapse
|
13
|
Buakaew W, Krobthong S, Yingchutrakul Y, Khamto N, Sutana P, Potup P, Thongsri Y, Daowtak K, Ferrante A, Léon C, Usuwanthim K. In Vitro Investigation of the Anti-Fibrotic Effects of 1-Phenyl-2-Pentanol, Identified from Moringa oleifera Lam., on Hepatic Stellate Cells. Int J Mol Sci 2024; 25:8995. [PMID: 39201682 PMCID: PMC11354330 DOI: 10.3390/ijms25168995] [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: 06/15/2024] [Revised: 08/17/2024] [Accepted: 08/17/2024] [Indexed: 09/03/2024] Open
Abstract
Liver fibrosis, characterized by excessive extracellular matrix deposition, is driven by activated hepatic stellate cells (HSCs). Due to the limited availability of anti-fibrotic drugs, the research on therapeutic agents continues. Here we have investigated Moringa oleifera Lam. (MO), known for its various bioactive properties, for anti-fibrotic effects. This study has focused on 1-phenyl-2-pentanol (1-PHE), a compound derived from MO leaves, and its effects on LX-2 human hepatic stellate cell activation. TGF-β1-stimulated LX-2 cells were treated with MO extract or 1-PHE, and the changes in liver fibrosis markers were assessed at both gene and protein levels. Proteomic analysis and molecular docking were employed to identify potential protein targets and signaling pathways affected by 1-PHE. Treatment with 1-PHE downregulated fibrosis markers, including collagen type I alpha 1 chain (COL1A1), collagen type IV alpha 1 chain (COL4A1), mothers against decapentaplegic homologs 2 and 3 (SMAD2/3), and matrix metalloproteinase-2 (MMP2), and reduced the secretion of matrix metalloproteinase-9 (MMP-9). Proteomic analysis data showed that 1-PHE modulates the Wnt/β-catenin pathway, providing a possible mechanism for its effects. Our results suggest that 1-PHE inhibits the TGF-β1 and Wnt/β-catenin signaling pathways and HSC activation, indicating its potential as an anti-liver-fibrosis agent.
Collapse
Affiliation(s)
- Watunyoo Buakaew
- Department of Microbiology, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand
- Cellular and Molecular Immunology Research Unit (CMIRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Sucheewin Krobthong
- Center of Excellence in Natural Products Chemistry (CENP), Department of Chemistry Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
- National Center for Genetic Engineering and Biotechnology, NSTDA, Pathum Thani 12120, Thailand
| | - Yodying Yingchutrakul
- National Center for Genetic Engineering and Biotechnology, NSTDA, Pathum Thani 12120, Thailand
| | - Nopawit Khamto
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pornsuda Sutana
- Cellular and Molecular Immunology Research Unit (CMIRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Pachuen Potup
- Cellular and Molecular Immunology Research Unit (CMIRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Yordhathai Thongsri
- Cellular and Molecular Immunology Research Unit (CMIRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Krai Daowtak
- Cellular and Molecular Immunology Research Unit (CMIRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| | - Antonio Ferrante
- Department of Immunopathology, South Australia (SA) Pathology, Women's and Children's Hospital, Adelaide, SA 5006, Australia
- The Adelaide Medical School, The School of Biological Science and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia
| | - Catherine Léon
- INSERM, UMR_S1255, Université de Strasbourg, Etablissement Français du Sang-GEST, 67000 Strasbourg, France
| | - Kanchana Usuwanthim
- Cellular and Molecular Immunology Research Unit (CMIRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand
| |
Collapse
|
14
|
Fan Z, Yin B, Chen X, Yang G, Zhang W, Ye X, Han H, Li M, Shu M, Liu R. Comprehensive analysis of paraspeckle-associated gene modules unveils prognostic signatures and immunological relevance in multi-cancers. Discov Oncol 2024; 15:345. [PMID: 39133261 PMCID: PMC11319543 DOI: 10.1007/s12672-024-01188-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 07/23/2024] [Indexed: 08/13/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide, characterized by high rates of angiogenesis and immune evasion. Paraspeckle genes, involved in gene regulation and RNA metabolism, have recently been linked to tumor progression. This study aims to elucidate the relationship between paraspeckle genes and HCC prognosis, focusing on SFPQ, DDX39B, and UBAP2. METHODS We analyzed HCC (LIHC) and prostate cancer (PRAD) samples from the TCGA database to explore the correlation between paraspeckle genes and angiogenesis. We conducted unsupervised clustering, risk scoring, and survival analysis to identify distinct patient groups and their clinical outcomes. Gene expression data were used to perform differential analysis and Gene Ontology (GO) enrichment. RESULTS Our analysis identified significant correlations between paraspeckle genes and angiogenesis across multiple cancer types. Elevated expression levels of SFPQ, DDX39B, and UBAP2 were associated with poor prognosis in HCC patients, and all of them has statistical significance. Unsupervised clustering of HCC samples based on paraspeckle gene expression revealed two distinct clusters, with high-risk patients exhibiting stronger immune suppression and tumor immune evasion. GO enrichment highlighted critical pathways related to angiogenesis and immune regulation. Additionally, a risk scoring model based on these genes effectively distinguished high-risk and low-risk patient groups, providing valuable prognostic insights. CONCLUSION This study demonstrates that SFPQ, DDX39B, and UBAP2 are significantly associated with poor prognosis in HCC, likely due to their roles in promoting angiogenesis and immune suppression. These findings highlight the potential of paraspeckle genes as prognostic biomarkers and therapeutic targets, offering new avenues for personalized treatment strategies in HCC. Further research into their functional mechanisms and clinical applicability is crucial for advancing HCC treatment and improving patient outcomes.
Collapse
Affiliation(s)
- Zhuoyang Fan
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bowen Yin
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Key Laboratory of Medical Molecular Virology, (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaochen Chen
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guowei Yang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Zhang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaodan Ye
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, 200032, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hong Han
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Ming Li
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Minfeng Shu
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
- Key Laboratory of Medical Molecular Virology, (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Rong Liu
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Interventional Radiology, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, 361015, China.
| |
Collapse
|
15
|
Blank V, Karlas T, Anderegg U, Wiegand J, Arnold J, Bundalian L, Le Duc GD, Körner C, Ebert T, Saalbach A. Thy-1 restricts steatosis and liver fibrosis in steatotic liver disease. Liver Int 2024; 44:2075-2090. [PMID: 38702958 DOI: 10.1111/liv.15956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/26/2024] [Accepted: 04/16/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND AND AIMS Steatotic liver disease (SLD) is generally considered to represent a hepatic manifestation of metabolic syndrome and includes a disease spectrum comprising isolated steatosis, metabolic dysfunction-associated steatohepatitis, liver fibrosis and ultimately cirrhosis. A better understanding of the detailed underlying pathogenic mechanisms of this transition is crucial for the design of new and efficient therapeutic interventions. Thymocyte differentiation antigen (Thy-1, also known as CD90) expression on fibroblasts controls central functions relevant to fibrogenesis, including proliferation, apoptosis, cytokine responsiveness, and myofibroblast differentiation. METHODS The impact of Thy-1 on the development of SLD and progression to fibrosis was investigated in high-fat diet (HFD)-induced SLD wild-type and Thy-1-deficient mice. In addition, the serum soluble Thy-1 (sThy-1) concentration was analysed in patients with metabolic dysfunction-associated SLD stratified according to steatosis, inflammation, or liver fibrosis using noninvasive markers. RESULTS We demonstrated that Thy-1 attenuates the development of fatty liver and the expression of profibrogenic genes in the livers of HFD-induced SLD mice. Mechanistically, Thy-1 directly inhibits the profibrotic activation of nonparenchymal liver cells. In addition, Thy-1 prevents palmitic acid-mediated amplification of the inflammatory response of myeloid cells, which might indirectly contribute to the pronounced development of liver fibrosis in Thy-1-deficient mice. Serum analysis of patients with metabolically associated steatotic liver disease syndrome revealed that sThy-1 expression is correlated with liver fibrosis status, as assessed by liver stiffness, the Fib4 score, and the NAFLD fibrosis score. CONCLUSION Our data strongly suggest that Thy-1 may function as a fibrosis-protective factor in mouse and human SLD.
Collapse
Affiliation(s)
- Valentin Blank
- Division of Gastroenterology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
- Division of Interdisciplinary Ultrasound, Department of Internal Medicine I - Gastroenterology and Pneumology, University Hospital Halle, Halle, Germany
| | - Thomas Karlas
- Division of Gastroenterology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| | - Ulf Anderegg
- Department of Dermatology, Venereology and Allergology, University of Leipzig Medical Center, Leipzig, Germany
| | - Johannes Wiegand
- Division of Hepatology, Department of Medicine II, Leipzig University Medical Center, Leipzig, Germany
| | - Josi Arnold
- Department of Dermatology, Venereology and Allergology, University of Leipzig Medical Center, Leipzig, Germany
| | - Linnaeus Bundalian
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Gabriela-Diana Le Duc
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Christiane Körner
- Division of Hepatology, Clinic of Oncology, Gastroenterology, Hepatology, and Pneumology, University Hospital Leipzig, Leipzig, Germany
| | - Thomas Ebert
- Division of Endocrinology, Department of Medicine III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Anja Saalbach
- Department of Dermatology, Venereology and Allergology, University of Leipzig Medical Center, Leipzig, Germany
| |
Collapse
|
16
|
Nasr HE, Hegazy AM, El-Shaer NO, El-Shafey RS, Elgendy SA, Elnoury HA, Gazzar WBE, Mohammed LA. Ameliorative effects of sildenafil against carbon tetrachloride induced hepatic fibrosis in rat model through downregulation of osteopontin gene expression. Sci Rep 2024; 14:16902. [PMID: 39043726 PMCID: PMC11266717 DOI: 10.1038/s41598-024-67305-1] [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: 06/06/2024] [Accepted: 07/10/2024] [Indexed: 07/25/2024] Open
Abstract
The liver carries out many essential tasks, such as synthesising cholesterol, controlling the body's storage of glycogen, and detoxifying metabolites, in addition to performing, and regulating homeostasis. Hepatic fibrosis is a pathological state characterized by over accumulation of extracellular matrix (ECM) including collagen fibers. Sildenafil (a selective inhibitor of type 5 phosphodiesterase) has anti-inflammatory, antioxidant and anti-apoptotic properties. It is commonly used to treat erectile dysfunction in male. The purpose of the current investigation was to evaluate sildenafil's hepatoprotective potential against liver fibrosis in rats that was caused by carbon tetrachloride (CCl4). Liver enzymes and oxidative markers as well as profibrotic genes were determined. The findings showed that sildenafil alleviates the hepatic dysfunctions caused by CCl4 by restoring normal levels of ALT, AST, and GGT as well as by restoring the antioxidant status demonstrated by increased glutathione (GSH), and catalase. In addition, a significantly down-regulated the mRNA expressions of profibrotic genes [collagen-1α, IL-1β, osteopontin (OPN), and transforming growth factor-β (TGF-β)]. Additionally, sildenafil lessens the periportal fibrosis between hepatic lobules, congestion and dilatation in the central vein, and the inflammatory cell infiltrations. As a result, it is hypothesized that sildenafil may be helpful in the management of hepatotoxicity brought on by CCl4 through suppressing OPN.
Collapse
Affiliation(s)
- Hend Elsayed Nasr
- Department of Medical Biochemistery and Molecular Biology, Faculty of Medicine, Benha University, Benha, 13518, Egypt
| | - Ahmed Medhat Hegazy
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, 13736, Qalyubia, Egypt.
| | - Noha Osama El-Shaer
- Department of Physiology, Faculty of Medicine, Benha University, Benha, 13518, Egypt
| | - Rabab Shaban El-Shafey
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha, 13518, Egypt
| | - Salwa A Elgendy
- Deparment of Pharmacology, Faculty of Medicine, Benha University, Benha, 13518, Egypt
| | - Heba A Elnoury
- Deparment of Pharmacology, Faculty of Medicine, Benha University, Benha, 13518, Egypt
| | - Walaa Bayoumie El Gazzar
- Department of Medical Biochemistery and Molecular Biology, Faculty of Medicine, Benha University, Benha, 13518, Egypt
| | - Lina Abdelhady Mohammed
- Department of Medical Biochemistery and Molecular Biology, Faculty of Medicine, Benha University, Benha, 13518, Egypt
| |
Collapse
|
17
|
Buakaew W, Krobthong S, Yingchutrakul Y, Potup P, Thongsri Y, Daowtak K, Ferrante A, Usuwanthim K. Investigating the Antifibrotic Effects of β-Citronellol on a TGF-β1-Stimulated LX-2 Hepatic Stellate Cell Model. Biomolecules 2024; 14:800. [PMID: 39062514 PMCID: PMC11274813 DOI: 10.3390/biom14070800] [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: 06/08/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Liver fibrosis, a consequence of chronic liver damage or inflammation, is characterized by the excessive buildup of extracellular matrix components. This progressive condition significantly raises the risk of severe liver diseases like cirrhosis and hepatocellular carcinoma. The lack of approved therapeutics underscores the urgent need for novel anti-fibrotic drugs. Hepatic stellate cells (HSCs), key players in fibrogenesis, are promising targets for drug discovery. This study investigated the anti-fibrotic potential of Citrus hystrix DC. (KL) and its bioactive compound, β-citronellol (β-CIT), in a human HSC cell line (LX-2). Cells exposed to TGF-β1 to induce fibrogenesis were co-treated with crude KL extract and β-CIT. Gene expression was analyzed by real-time qRT-PCR to assess fibrosis-associated genes (ACTA2, COL1A1, TIMP1, SMAD2). The release of matrix metalloproteinase 9 (MMP-9) was measured by ELISA. Proteomic analysis and molecular docking identified potential signaling proteins and modeled protein-ligand interactions. The results showed that both crude KL extract and β-CIT suppressed HSC activation genes and MMP-9 levels. The MAPK signaling pathway emerged as a potential target of β-CIT. This study demonstrates the ability of KL extract and β-CIT to inhibit HSC activation during TGF-β1-induced fibrogenesis, suggesting a promising role of β-CIT in anti-hepatic fibrosis therapies.
Collapse
Affiliation(s)
- Watunyoo Buakaew
- Department of Microbiology, Faculty of Medicine, Srinakharinwirot University, Bangkok 10110, Thailand;
- Cellular and Molecular Immunology Research Unit (CMIRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand; (P.P.); (Y.T.); (K.D.)
| | - Sucheewin Krobthong
- Center of Excellence in Natural Products Chemistry (CENP), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand;
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand;
| | - Yodying Yingchutrakul
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency (NSTDA), Pathum Thani 12120, Thailand;
| | - Pachuen Potup
- Cellular and Molecular Immunology Research Unit (CMIRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand; (P.P.); (Y.T.); (K.D.)
| | - Yordhathai Thongsri
- Cellular and Molecular Immunology Research Unit (CMIRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand; (P.P.); (Y.T.); (K.D.)
| | - Krai Daowtak
- Cellular and Molecular Immunology Research Unit (CMIRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand; (P.P.); (Y.T.); (K.D.)
| | - Antonio Ferrante
- Department of Immunopathology, South Australia (SA) Pathology, Women’s and Children’s Hospital, Adelaide, SA 5006, Australia;
- The Adelaide Medical School, The School of Biological Science and the Robinson Research Institute, University of Adelaide, Adelaide, SA 5000, Australia
| | - Kanchana Usuwanthim
- Cellular and Molecular Immunology Research Unit (CMIRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok 65000, Thailand; (P.P.); (Y.T.); (K.D.)
| |
Collapse
|
18
|
Xu X, Feng J, Wang X, Zeng X, Luo Y, He X, Yang M, Lv T, Feng Z, Bao L, Zhao L, Huang D, Huang Y. Mitochondrial GRIM19 Loss Induces Liver Fibrosis through NLRP3/IL33 Activation via Reactive Oxygen Species/NF-кB Signaling. J Clin Transl Hepatol 2024; 12:539-550. [PMID: 38974954 PMCID: PMC11224902 DOI: 10.14218/jcth.2023.00562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/24/2024] [Accepted: 05/11/2024] [Indexed: 07/09/2024] Open
Abstract
Background and Aims Hepatic fibrosis (HF) is a critical step in the progression of hepatocellular carcinoma (HCC). Gene associated with retinoid-IFN-induced mortality 19 (GRIM19), an essential component of mitochondrial respiratory chain complex I, is frequently attenuated in various human cancers, including HCC. Here, we aimed to investigate the potential relationship and underlying mechanism between GRIM19 loss and HF pathogenesis. Methods GRIM19 expression was evaluated in normal liver tissues, hepatitis, hepatic cirrhosis, and HCC using human liver disease spectrum tissue microarrays. We studied hepatocyte-specific GRIM19 knockout mice and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein-9 (Cas9) lentivirus-mediated GRIM19 gene-editing in murine hepatocyte AML12 cells in vitro and in vivo. We performed flow cytometry, immunofluorescence, immunohistochemistry, western blotting, and pharmacological intervention to uncover the potential mechanisms underlying GRIM19 loss-induced HF. Results Mitochondrial GRIM19 was progressively downregulated in chronic liver disease tissues, including hepatitis, cirrhosis, and HCC tissues. Hepatocyte-specific GRIM19 heterozygous deletion induced spontaneous hepatitis and subsequent liver fibrogenesis in mice. In addition, GRIM19 loss caused chronic liver injury through reactive oxygen species (ROS)-mediated oxidative stress, resulting in aberrant NF-кB activation via an IKK/IкB partner in hepatocytes. Furthermore, GRIM19 loss activated NLRP3-mediated IL33 signaling via the ROS/NF-кB pathway in hepatocytes. Intraperitoneal administration of the NLRP3 inhibitor MCC950 dramatically alleviated GRIM19 loss-driven HF in vivo. Conclusions The mitochondrial GRIM19 loss facilitates liver fibrosis through NLRP3/IL33 activation via ROS/NF-кB signaling, providing potential therapeutic approaches for earlier HF prevention.
Collapse
Affiliation(s)
- Xiaohui Xu
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
- Department of Cardiology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Key Cardiovascular Specialty, Laboratory of Children’s Important Organ Development and Diseases of Chongqing Municipal Health Commission, Chongqing, China
| | - Jinmei Feng
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
- Department of Laboratory Medicine, Chongqing Western Hospital, Chongqing, China
| | - Xin Wang
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Institute for Viral Hepatitis, Chongqing Medical University, Chongqing, China
| | - Xin Zeng
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Ying Luo
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Xinyu He
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Meihua Yang
- Departments of Neurology, Epilepsy Center, Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, MO, USA
| | - Tiewei Lv
- Department of Cardiology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Key Cardiovascular Specialty, Laboratory of Children’s Important Organ Development and Diseases of Chongqing Municipal Health Commission, Chongqing, China
| | - Zijuan Feng
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Liming Bao
- Department of Clinical Pathology and Laboratory Medicine, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Li Zhao
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Daochao Huang
- Institute of Pediatrics, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Yi Huang
- Department of Cardiology, Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Key Cardiovascular Specialty, Laboratory of Children’s Important Organ Development and Diseases of Chongqing Municipal Health Commission, Chongqing, China
- Departments of Medicine (Oncology), Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
19
|
Sharma S, Gali S, Kundu A, Park JH, Kim JS, Kim HS. Tenovin-1, a Selective SIRT1/2 Inhibitor, Attenuates High-fat Diet-induced Hepatic Fibrosis via Inhibition of HSC Activation in ZDF Rats. Int J Biol Sci 2024; 20:3334-3352. [PMID: 38993557 PMCID: PMC11234213 DOI: 10.7150/ijbs.97304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/21/2024] [Indexed: 07/13/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) increases the risk of non-alcoholic fatty liver disease (NAFLD) progression to advanced stages, especially upon high-fat diet (HFD). HFD-induced hepatic fibrosis can be marked by oxidative stress, inflammation, and activation of hepatic stellate cells. Sirtuin 1/2 (SIRT1/2), NAD-dependent class III histone deacetylases, are involved in attenuation of fibrosis. In our conducted research, TGF-β1-activated LX-2 cells, free fatty acid (FFA)-treated simultaneous co-culture (SCC) cells, and HFD-induced hepatic fibrosis in Zucker diabetic fatty (ZDF) rats, a widely used animal model in the study of metabolic syndromes, were used to evaluate the protective effect of Tenovin-1, a SIRT1/2 inhibitor. ZDF rats were divided into chow diet, HFD, and HFD + Tenovin-1 groups. Tenovin-1 reduced hepatic damage, inhibited inflammatory cell infiltration, micro/ macro-vesicular steatosis and prevented collagen deposition HFD-fed rats. Tenovin-1 reduced serum biochemical parameters, triglyceride (TG) and malondialdehyde (MDA) levels but increased glutathione, catalase, and superoxide dismutase levels. Tenovin-1 mitigated proinflammatory cytokines IL-6, IL-1β, TNFα and fibrosis biomarkers in HFD rats, TGF-β1-activated LX-2 and FFA treated SCC cells. Additionally, Tenovin-1 suppressed SIRT1/2 expression and inhibited JNK-1 and STAT3 phosphorylation in HFD rats and FFA-treated SCC cells. In conclusion, Tenovin-1 attenuates hepatic fibrosis by stimulating antioxidants and inhibiting inflammatory cytokines under HFD conditions in diabetic rats.
Collapse
Affiliation(s)
- Swati Sharma
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
| | - Sreevarsha Gali
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
| | - Amit Kundu
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
- Department of Pharmacology, GITAM School of Pharmacy, GITAM (Deemed to be University), Rushikonda, Visakhapatnam-530045, Andhra Pradesh, India
| | - Jae Hyeon Park
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
| | - Jae-Sung Kim
- Department of Surgery, Washington University in St. Louis, St. Louis, MO, 63110, USA
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, 2066, Seobu-ro, Jangan-gu, Suwon 26419, Republic of Korea
| |
Collapse
|
20
|
Taychaworaditsakul W, Sawong S, Intatham S, Chansakaow S, Kunnaja P, Chewonarin T, Jaijoy K, Wittayapraparat A, Yusuk P, Sireeratawong S. The Ameliorative Effect of Litsea martabanica (Kurz) Hook. f. Leaf Water Extract on Chlorpyrifos-Induced Toxicity in Rats and Its Antioxidant Potentials. Foods 2024; 13:1695. [PMID: 38890923 PMCID: PMC11172329 DOI: 10.3390/foods13111695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
Litsea martabanica root's antioxidant and acetylcholinesterase (AChE) activity showed promise as a pesticide detoxification agent in our previous study. In addition to its root, leaves can help alleviate pesticide exposure, although there is limited scientific evidence supporting their efficacy. However, the use of roots in several countries, such as Thailand, could contribute to environmental degradation, as highland communities traditionally used leaves instead of roots. This study aims to evaluate the antioxidant activity and anti-pesticide potential of water extract from L. martabanica leaves through in vitro and in vivo investigations. In the in vitro study, L. martabanica water extract and its fractions demonstrated antioxidant activity and induced apoptosis in hepatic satellite cells. In the in vivo study, treatment with the leaf extract led to increased AChE activity, decreased malondialdehyde (MDA) levels, increased superoxide dismutase (SOD) levels, and reduced glutathione in chlorpyrifos-exposed rats. Histopathological examination revealed that chlorpyrifos-treated rats exhibited liver cell damage, while treatment with the water extract of L. martabanica exhibited a protective effect on the liver. In conclusion, L. martabanica water extract exhibited antioxidant activity, enhanced AChE activity, and improved histopathological abnormalities in the liver.
Collapse
Affiliation(s)
- Weerakit Taychaworaditsakul
- Clinical Research Center for Food and Herbal Product Trials and Development (CR-FAH), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.T.); (S.S.); (S.I.)
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Suphunwadee Sawong
- Clinical Research Center for Food and Herbal Product Trials and Development (CR-FAH), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.T.); (S.S.); (S.I.)
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Supaporn Intatham
- Clinical Research Center for Food and Herbal Product Trials and Development (CR-FAH), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.T.); (S.S.); (S.I.)
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sunee Chansakaow
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Phraepakaporn Kunnaja
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Teera Chewonarin
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Kanjana Jaijoy
- McCormick Faculty of Nursing, Payap University, Chiang Mai 50000, Thailand;
| | - Absorn Wittayapraparat
- Highland Research and Development Institute (Public Organization), Chiang Mai 50200, Thailand; (A.W.); (P.Y.)
| | - Pedcharada Yusuk
- Highland Research and Development Institute (Public Organization), Chiang Mai 50200, Thailand; (A.W.); (P.Y.)
| | - Seewaboon Sireeratawong
- Clinical Research Center for Food and Herbal Product Trials and Development (CR-FAH), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.T.); (S.S.); (S.I.)
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
21
|
Zarlashat Y, Abbas S, Ghaffar A. Hepatocellular Carcinoma: Beyond the Border of Advanced Stage Therapy. Cancers (Basel) 2024; 16:2034. [PMID: 38893154 PMCID: PMC11171154 DOI: 10.3390/cancers16112034] [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: 03/31/2024] [Revised: 04/27/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the deadliest emergent health issue around the globe. The stronger oncogenic effect, proteins, and weakened immune response are precisely linked with a significant prospect of developing HCC. Several conventional systemic therapies, antiangiogenic therapy, and immunotherapy techniques have significantly improved the outcomes for early-, intermediate-, and advanced-stage HCC patients, giving new hope for effective HCC management and prolonged survival rates. Innovative therapeutic approaches beyond conventional treatments have altered the landscape of managing HCC, particularly focusing on targeted therapies and immunotherapies. The advancement in HCC treatment suggested by the Food and Drug Administration is multidimensional treatment options, including multikinase inhibitors (sorafenib, lenvatinib, regorafenib, ramucirumab, and cabozantinib) and immune checkpoint inhibitors (atezolizumab, pembrolizumab, durvalumab, tremelimumab, ipilimumab, and nivolumab), in monotherapy and in combination therapy to increase life expectancy of HCC patients. This review highlights the efficacy of multikinase inhibitors and immune checkpoint inhibitors in monotherapy and combination therapy through the analysis of phase II, and III clinical trials, targeting the key molecular pathways involved in cellular signaling and immune response for the prospective treatment of advanced and unresectable HCC and discusses the upcoming combinations of immune checkpoint inhibitors-tyrosine kinase inhibitors and immune checkpoint inhibitors-vascular endothelial growth factor inhibitors. Finally, the hidden challenges with pharmacological therapy for HCC, feasible solutions for the future, and implications of possible presumptions to develop drugs for HCC treatment are reported.
Collapse
Affiliation(s)
- Yusra Zarlashat
- Department of Biochemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Shakil Abbas
- Gomal Center of Biotechnology and Biochemistry (GCBB), Gomal University, Dera Ismail Khan 29050, Pakistan;
| | - Abdul Ghaffar
- Department of Biochemistry, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| |
Collapse
|
22
|
Bozhkov AI, Akzhyhitov RA, Bilovetska SG, Ivanov EG, Dobrianska NI, Bondar AY. The Effect of Retinol Acetate on Liver Fibrosis Depends on the Temporal Features of the Development of Pathology. J Clin Exp Hepatol 2024; 14:101338. [PMID: 38264572 PMCID: PMC10801314 DOI: 10.1016/j.jceh.2023.101338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 12/19/2023] [Indexed: 01/25/2024] Open
Abstract
Background The effect of vitamin A on the manifestations of liver fibrosis is controversial and establishing the causes of its multidirectional influence is an urgent problem. In the work, the functional characteristics of the liver with Cu-induced fibrosis were determined after the restoration of vitamin A to the control level at the F0/F1 stage. Methods In animals with liver fibrosis, classical indicators of physiology, functional activity of the liver, histological, and hematological characteristics were determined; the content of calcium and ROS was determined in bone marrow cells. Results It was shown that in the liver with Cu-induced fibrosis, the restoration of vitamin A content to control values after per os injections of a retinol acetate solution at a dose of 0.10 mg (300 IU)/100 g of body weight in the early stages of this pathology development (Fо/F1) was accompanied by: a decrease in the number of immunocompetent cells in the bloodstream to control values; normalization of the amount of calcium ions and ROS in bone marrow cells; restoration to the control level of activity of alkaline phosphatase; an increase in the number of binuclear hepatocytes; and restoration of the dynamics of body weight growth in experimental animals, even against the background of the ongoing action of the hepatotoxic factor. Conclusion We came to the conclusion that the multidirectional action of vitamin A, which occurs in liver fibrosis, depends not only on the concentration of vitamin A in the liver but also on temporal characteristics of cellular and metabolic links involved in the adaptive response formation. It was suggested that knowledge of the initial temporal metabolic characteristics and the amount of vitamin A in the liver, taking into account the stages of fibrosis development, can be an effective way to restore the altered homeostatic parameters of the body.
Collapse
Affiliation(s)
- Anatoly I. Bozhkov
- Biology Research Institute V. N. Karazin Kharkiv National University, 4 Svobody Sq., Kharkiv, 61022, Ukraine
| | - Rustam A. Akzhyhitov
- Biology Research Institute V. N. Karazin Kharkiv National University, 4 Svobody Sq., Kharkiv, 61022, Ukraine
| | - Svitlana G. Bilovetska
- Biology Research Institute V. N. Karazin Kharkiv National University, 4 Svobody Sq., Kharkiv, 61022, Ukraine
| | - Evgeny G. Ivanov
- Biology Research Institute V. N. Karazin Kharkiv National University, 4 Svobody Sq., Kharkiv, 61022, Ukraine
| | - Nataliia I. Dobrianska
- Biology Research Institute V. N. Karazin Kharkiv National University, 4 Svobody Sq., Kharkiv, 61022, Ukraine
| | - Anastasiia Yu Bondar
- Biology Research Institute V. N. Karazin Kharkiv National University, 4 Svobody Sq., Kharkiv, 61022, Ukraine
| |
Collapse
|
23
|
Habibi P, Falamarzi K, Ebrahimi ND, Zarei M, Malekpour M, Azarpira N. GDF11: An emerging therapeutic target for liver diseases and fibrosis. J Cell Mol Med 2024; 28:e18140. [PMID: 38494851 PMCID: PMC10945076 DOI: 10.1111/jcmm.18140] [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: 09/17/2023] [Revised: 01/07/2024] [Accepted: 01/16/2024] [Indexed: 03/19/2024] Open
Abstract
Growth differentiation factor 11 (GDF11), also known as bone morphogenetic protein 11 (BMP11), has been identified as a key player in various biological processes, including embryonic development, aging, metabolic disorders and cancers. GDF11 has also emerged as a critical component in liver development, injury and fibrosis. However, the effects of GDF11 on liver physiology and pathology have been a subject of debate among researchers due to conflicting reported outcomes. While some studies suggest that GDF11 has anti-aging properties, others have documented its senescence-inducing effects. Similarly, while GDF11 has been implicated in exacerbating liver injury, it has also been shown to have the potential to reduce liver fibrosis. In this narrative review, we present a comprehensive report of recent evidence elucidating the diverse roles of GDF11 in liver development, hepatic injury, regeneration and associated diseases such as non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), liver fibrosis and hepatocellular carcinoma. We also explore the therapeutic potential of GDF11 in managing various liver pathologies.
Collapse
Affiliation(s)
- Pardis Habibi
- Student Research CommitteeShiraz University of Medical SciencesShirazIran
- Transplant Research CenterShiraz University of Medical SciencesShirazIran
| | - Kimia Falamarzi
- Student Research CommitteeShiraz University of Medical SciencesShirazIran
- Transplant Research CenterShiraz University of Medical SciencesShirazIran
| | | | - Mohammad Zarei
- Renal Division, Brigham & Women's HospitalHarvard Medical SchoolBostonMassachusettsUSA
- John B. Little Center for Radiation SciencesHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Mahdi Malekpour
- Student Research CommitteeShiraz University of Medical SciencesShirazIran
- Transplant Research CenterShiraz University of Medical SciencesShirazIran
| | - Negar Azarpira
- Transplant Research CenterShiraz University of Medical SciencesShirazIran
| |
Collapse
|
24
|
AL-ADL MENNA, YOUSSEF MAGDYM, EL-SEBAIE AHMED, REFAAT SHERIF, EL-SAID AFAF. The role of polymorphic cytochrome P450 gene (CYP2B6) in B-chronic lymphocytic leukemia (B-CLL) incidence and outcome among Egyptian patients. Oncol Res 2024; 32:785-797. [PMID: 38560574 PMCID: PMC10972729 DOI: 10.32604/or.2024.047021] [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: 10/22/2023] [Accepted: 01/17/2024] [Indexed: 04/04/2024] Open
Abstract
Cytochromes P450 (CYPs) play a prominent role in catalyzing phase I xenobiotic biotransformation and account for about 75% of the total metabolism of commercially available drugs, including chemotherapeutics. The gene expression and enzyme activity of CYPs are variable between individuals, which subsequently leads to different patterns of susceptibility to carcinogenesis by genotoxic xenobiotics, as well as differences in the efficacy and toxicity of clinically used drugs. This research aimed to examine the presence of the CYP2B6*9 polymorphism and its possible association with the incidence of B-CLL in Egyptian patients, as well as the clinical outcome after receiving cyclophosphamide chemotherapy. DNA was isolated from whole blood samples of 100 de novo B-CLL cases and also from 100 sex- and age-matched healthy individuals. The presence of the CYP2B6*9 (G516T) polymorphism was examined by PCR-based allele specific amplification (ASA). Patients were further indicated for receiving chemotherapy, and then they were followed up. The CYP2B6*9 variant indicated a statistically significant higher risk of B-CLL under different genetic models, comprising allelic (T-allele vs. G-allele, OR = 4.8, p < 0.001) and dominant (GT + TT vs. GG, OR = 5.4, p < 0.001) models. Following cyclophosphamide chemotherapy, we found that the patients with variant genotypes (GT + TT) were less likely to achieve remission compared to those with the wild-type genotype (GG), with a response percentage of (37.5% vs. 83%, respectively). In conclusion, our findings showed that the CYP2B6*9 (G516T) polymorphism is associated with B-CLL susceptibility among Egyptian patients. This variant greatly affected the clinical outcome and can serve as a good therapeutic marker in predicting response to cyclophosphamide treatment.
Collapse
Affiliation(s)
- MENNA AL-ADL
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Mansoura University, Mansoura, 35511, Egypt
| | - MAGDY M. YOUSSEF
- Division of Biochemistry, Department of Chemistry, Faculty of Science, Mansoura University, Mansoura, 35511, Egypt
| | - AHMED EL-SEBAIE
- Hematology Unit, Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura, 35511, Egypt
| | - SHERIF REFAAT
- Medical Oncology Unit, Oncology Center Mansoura University, Mansoura, 35511, Egypt
| | - AFAF EL-SAID
- Department of Genetics, Mansoura University Children’s Hospital, Mansoura, 35511, Egypt
| |
Collapse
|
25
|
Rajendran P, Renu K, Abdallah BM, Ali EM, Veeraraghavan VP, Sivalingam K, Rustagi Y, Abdelsalam SA, Ibrahim RIH, Al-Ramadan SY. Nimbolide: promising agent for prevention and treatment of chronic diseases (recent update). Food Nutr Res 2024; 68:9650. [PMID: 38571915 PMCID: PMC10989234 DOI: 10.29219/fnr.v68.9650] [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: 04/28/2023] [Revised: 10/09/2023] [Accepted: 10/16/2023] [Indexed: 04/05/2024] Open
Abstract
Background Nimbolide, a bioactive compound derived from the neem tree, has garnered attention as a potential breakthrough in the prevention and treatment of chronic diseases. Recent updates in research highlight its multifaceted pharmacological properties, demonstrating anti-inflammatory, antioxidant, and anticancer effects. With a rich history in traditional medicine, nimbolide efficacy in addressing the molecular complexities of conditions such as cardiovascular diseases, diabetes, and cancer positions it as a promising candidate for further exploration. As studies progress, the recent update underscores the growing optimism surrounding nimbolide as a valuable tool in the ongoing pursuit of innovative therapeutic strategies for chronic diseases. Methods The comprehensive search of the literature was done until September 2020 on the MEDLINE, Embase, Scopus and Web of Knowledge databases. Results Most studies have shown the Nimbolide is one of the most potent limonoids derived from the flowers and leaves of neem (Azadirachta indica), which is widely used to treat a variety of human diseases. In chronic diseases, nimbolide reported to modulate the key signaling pathways, such as Mitogen-activated protein kinases (MAPKs), Wingless-related integration site-β (Wnt-β)/catenin, NF-κB, PI3K/AKT, and signaling molecules, such as transforming growth factor (TGF-β), Matrix metalloproteinases (MMPs), Vascular Endothelial Growth Factor (VEGF), inflammatory cytokines, and epithelial-mesenchymal transition (EMT) proteins. Nimbolide has anti-inflammatory, anti-microbial, and anti-cancer properties, which make it an intriguing compound for research. Nimbolide demonstrated therapeutic potential for osteoarthritis, rheumatoid arthritis, cardiovascular, inflammation and cancer. Conclusion The current review mainly focused on understanding the molecular mechanisms underlying the therapecutic effects of nimbolide in chronic diseases.
Collapse
Affiliation(s)
- Peramaiyan Rajendran
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Basem M. Abdallah
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
| | - Enas M. Ali
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
- Department of Botany and Microbiology, Faculty of Science, Cairo University, Cairo, Egypt
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| | - Kalaiselvi Sivalingam
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, USA
| | - Yashika Rustagi
- Centre for Cancer Genomics, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Salaheldin Abdelraouf Abdelsalam
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
- Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
| | - Rashid Ismael Hag Ibrahim
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, Saudi Arabia
- Department of Botany, Faculty of Science, University of Khartoum, Sudan
| | - Saeed Yaseen Al-Ramadan
- Department of Anatomy, College of Veterinary Medicine, King Faisal University, Al-Ahsa, Saudi Arabia
| |
Collapse
|
26
|
Wang W, Li K, Xiao W. The pharmacological role of Ginsenoside Rg3 in liver diseases: A review on molecular mechanisms. J Ginseng Res 2024; 48:129-139. [PMID: 38465219 PMCID: PMC10920009 DOI: 10.1016/j.jgr.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/23/2023] [Accepted: 11/10/2023] [Indexed: 03/12/2024] Open
Abstract
Liver diseases are a significant global health burden and are among the most common diseases. Ginssennoside Rg3 (Rg3), which is one of the most abundant ginsenosides, has been found to have significant preventive and therapeutic effects against various types of diseases with minimal side effects. Numerous studies have demonstrated the significant preventive and therapeutic effects of Rg3 on various liver diseases such as viral hepatitis, acute liver injury, nonalcoholic liver diseases (NAFLD), liver fibrosis and hepatocellular carcinoma (HCC). The underlying molecular mechanism behind these effects is attributed to apoptosis, autophagy, antioxidant, anti-inflammatory activities, and the regulation of multiple signaling pathways. This review provides a comprehensive description of the potential molecular mechanisms of Rg3 in the development of liver diseases. The article focuses on the regulation of apoptosis, oxidative stress, autophagy, inflammation, and other related factors. Additionally, the review discusses combination therapy and liver targeting strategy, which can accelerate the translation of Rg3 from bench to bedside. Overall, this article serves as a valuable reference for researchers and clinicians alike.
Collapse
Affiliation(s)
- Wenhong Wang
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Ke Li
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| | - Weihua Xiao
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
- The Key Lab of Exercise and Health Sciences of Ministry of Education, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
27
|
Xiang W, Yin G, Liu H, Wei J, Yu X, Xie Y, Zhang L, XueTang, Jiang W, Lu N. Arctium lappa L. polysaccharides enhanced the therapeutic effects of nasal ectomesenchymal stem cells against liver fibrosis by inhibiting the Wnt/β-catenin pathway. Int J Biol Macromol 2024; 261:129670. [PMID: 38280697 DOI: 10.1016/j.ijbiomac.2024.129670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/14/2024] [Accepted: 01/20/2024] [Indexed: 01/29/2024]
Abstract
The oxidative microenvironment in fibrotic livers often diminishes the effectiveness of mesenchymal stem cells (MSCs)-based therapy. Recent research suggests that pharmacological pre-treatment could enhance the therapeutic performance of MSCs. In this study, we assessed the impact of Arctium lappa L. polysaccharides (ALP) on the biological properties of nasal ectomesenchymal stem cells (EMSCs) and investigated the augmenting effect of ALP pretreatment on EMSCs (ALP-EMSCs) for the treatment of liver fibrosis. ALP treatment demonstrated multiple biological impacts on EMSC functions regarding liver fibrosis: firstly, it maintained the stemness of the cells while boosting the EMSCs' paracrine effects; secondly, it increased the expression of anti-inflammatory and antioxidant factors; thirdly, it inhibited the activation of hepatic stellate cells (HSCs) and liver collagen build-up by modulating the Wnt/β-catenin signaling pathways. Collectively, these effects helped to halt the progression of liver fibrosis. Therefore, the use of ALP-EMSCs presents an innovative and promising approach for treating hepatic fibrosis in clinical scenarios.
Collapse
Affiliation(s)
- Wen Xiang
- School of Medicine, Nankai University, Tianjin, China; Department of Liver Transplantation, Tianjin First Central Hospital, Tianjin, China; Tianjin Key Laboratory of Molecular and Treatment of Liver Cancer, Tianjin First Center Hospital, Tianjin, China
| | - Guoliang Yin
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Haoming Liu
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jiayi Wei
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xinghui Yu
- School of Medicine, Nankai University, Tianjin, China; Department of Liver Transplantation, Tianjin First Central Hospital, Tianjin, China; Tianjin Key Laboratory of Molecular and Treatment of Liver Cancer, Tianjin First Center Hospital, Tianjin, China
| | - Yan Xie
- Department of Liver Transplantation, Tianjin First Central Hospital, Tianjin, China; Tianjin Key Laboratory of Molecular and Treatment of Liver Cancer, Tianjin First Center Hospital, Tianjin, China
| | - Li Zhang
- Department of Liver Transplantation, Tianjin First Central Hospital, Tianjin, China; Tianjin Key Laboratory of Molecular and Treatment of Liver Cancer, Tianjin First Center Hospital, Tianjin, China
| | - XueTang
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wentao Jiang
- School of Medicine, Nankai University, Tianjin, China; Department of Liver Transplantation, Tianjin First Central Hospital, Tianjin, China; Tianjin Key Laboratory of Molecular and Treatment of Liver Cancer, Tianjin First Center Hospital, Tianjin, China.
| | - Naiyan Lu
- School of Food Science and Technology, Jiangnan University, Wuxi, China.
| |
Collapse
|
28
|
徐 小, 冯 金, 罗 颖, 何 昕, 臧 金, 黄 道. [Adeno-associated virus-mediated hepatocyte-specific NDUFA13 overexpression protects against CCl 4-induced liver fibrosis in mice by inhibiting hepatic NLRP3 activation]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:201-209. [PMID: 38501404 PMCID: PMC10954519 DOI: 10.12122/j.issn.1673-4254.2024.02.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Indexed: 03/20/2024]
Abstract
OBJECTIVE To investigate the protective effect of NDUFA13 protein against acute liver injury and liver fibrosis in mice and explore the possible mechanisms. METHODS BALB/C mice (7 to 8 weeks old) were divided into normal group, CCl4 group, CCl4+AAV-NC group and CCl4+AAV-NDU13 group (n=18). Mouse models of liver fibrosis were established by intraperitoneal injection of CCl4 twice a week for 3, 5 or 7 weeks, and the recombinant virus AAV8-TBG-NC or AAV8-TBG-NDUFA13 was injected via the tail vein 7-10 days prior to CCl4 injection. After the treatments, pathological changes in the liver of the mice were observed using HE and Masson staining. Hepatic expression levels of NDUFA13 and α-SMA were detected with Western blotting, and the coexpression of NDUFA13 and NLRP3, TNF-α and IL-1β, and α-SMA and collagen Ⅲ was analyzed with immunofluorescence assay. RESULTS HE and Masson staining showed deranged liver architecture, necrotic hepatocytes and obvious inflammatory infiltration and collagen fiber deposition in mice with CCl4 injection (P < 0.001). NDUFA13 expression markedly decreased in CCl4-treated mice (P < 0.001), while a significant reduction in inflammatory aggregation and fibrosis was observed in mice with AAV-mediated NDUFA13 overexpression (P < 0.001). In CCl4+AAV-NDU13 group, immunofluorescence assay revealed markedly weakened activation of NLRP3 inflammasomes (P < 0.001), significantly decreased TNF-α and IL-1β secretion (P < 0.001), and inhibited hepatic stellate cell activation (P < 0.05) and collagen formation in the liver (P < 0.001). CONCLUSION Mitochondrial NDUFA13 overexpression in hepatocytes protects against CCl4- induced liver fibrosis in mice by inhibiting activation of NLRP3 signaling.
Collapse
Affiliation(s)
- 小惠 徐
- 重庆医科大学附属儿童医院儿科研究所//国家儿童健康与疾病临床医学研究中心//儿童发育疾病研究教育部重点实验室,重庆 400014Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- 重庆医科大学附属儿童医院心内科//国家儿童健康与疾病临床医学研究中心//儿童发育疾病研究教育部重点实验室//国家临床心血管内科重点专科//重庆市卫生健康委儿童重要器官发育与疾病重点实验室,重庆 400014Department of Cardiology, Children's Hospital of Chongqing Medical University, Key Laboratory of Children's Important Organ Development and Diseases of Chongqing Municipal Health Commission, National Clinical Key Cardiovascular Specialty, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
| | - 金梅 冯
- 重庆医科大学附属儿童医院儿科研究所//国家儿童健康与疾病临床医学研究中心//儿童发育疾病研究教育部重点实验室,重庆 400014Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
- 重庆市九龙坡区第二人民医院检验科,重庆 400052Department of Laboratory Medicine, Second People's Hospital of Jiulongpo District, Chongqing 400052, China
| | - 颖 罗
- 重庆医科大学附属儿童医院儿科研究所//国家儿童健康与疾病临床医学研究中心//儿童发育疾病研究教育部重点实验室,重庆 400014Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
| | - 昕觎 何
- 重庆医科大学附属儿童医院儿科研究所//国家儿童健康与疾病临床医学研究中心//儿童发育疾病研究教育部重点实验室,重庆 400014Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
| | - 金宝 臧
- 重庆医科大学附属儿童医院儿科研究所//国家儿童健康与疾病临床医学研究中心//儿童发育疾病研究教育部重点实验室,重庆 400014Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
| | - 道超 黄
- 重庆医科大学附属儿童医院儿科研究所//国家儿童健康与疾病临床医学研究中心//儿童发育疾病研究教育部重点实验室,重庆 400014Institute of Pediatrics, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing 400014, China
| |
Collapse
|
29
|
Mekala S, Sukumar G, Chawla S, Geesala R, Prashanth J, Reddy BJM, Mainkar P, Das A. Therapeutic Potential of Benzimidazoisoquinoline Derivatives in Alleviating Murine Hepatic Fibrosis. Chem Biodivers 2024; 21:e202301429. [PMID: 38221801 DOI: 10.1002/cbdv.202301429] [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: 09/15/2023] [Revised: 01/09/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
Short Title: Benzimidazoisoquinoline derivatives as potent antifibrotics Hepatic fibrosis is a pathological condition of liver disease with an increasing number of cases worldwide. Therapeutic strategies are warranted to target the activated hepatic stellate cells (HSCs), the collagen-producing cells, an effective strategy for controlling the disease progression. Benzimidazoisoquinoline derivatives were synthesized as hybrid molecules by the combination of benzimidazoles and isoquinolines to evaluate their anti-fibrotic potential using an in-vitro and in-vivo model of hepatic fibrosis. A small library of benzimidazoisoquinoline derivatives (1-17 and 18-21) was synthesized from 2-aryl benzimidazole and acetylene functionalities through C-H and N-H activation. Compounds (10 and its recently synthesized derivatives 18-21) depicted a significant decrease in PDGF-BB and/or TGFβ-induced proliferation (1.7-1.9 -fold), migration (3.5-5.0 -fold), and fibrosis-related gene expressions in HSCs. These compounds could revert the hepatic damage caused by chronic exposure to hepatotoxicants, ethanol, and/or carbon tetrachloride as evident from the histological, biochemical, and molecular analysis. Anti-fibrotic effect of the compounds was supported by the decrease in the malondialdehyde level, collagen deposition, and gene expression levels of fibrosis-related markers such as α-SMA, COL1α1, PDGFRβ, and TGFRIIβ in the preclinical models of hepatic fibrosis. In conclusion, the synthesized benzimidazoisoquinoline derivatives (compounds 18, 19, 20, and 21) possess anti-fibrotic therapeutic potential against liver fibrosis.
Collapse
Affiliation(s)
- Sowmya Mekala
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, TS-500 007, INDIA
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, INDIA
| | - Genji Sukumar
- Department of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, TS-500007, INDIA
- Department of Chemistry, Adikavi Nannaya University, Rajamahendravaram, AP-533 296, INDIA
| | - Shilpa Chawla
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, TS-500 007, INDIA
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, INDIA
| | - Ramasatyaveni Geesala
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, TS-500 007, INDIA
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, INDIA
| | - Jupally Prashanth
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, INDIA
- Centre for X-ray Crystallography, Department of Analytical & Structural Chemistry, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, TS-500 007, INDIA
| | - B Jagan Mohan Reddy
- Department of Chemistry, Adikavi Nannaya University, Rajamahendravaram, AP-533 296, INDIA
| | - Prathama Mainkar
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, INDIA
- Department of Organic Synthesis and Process Chemistry, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, TS-500007, INDIA
| | - Amitava Das
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, TS-500 007, INDIA
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, INDIA
| |
Collapse
|
30
|
Sinha S, Hassan N, Schwartz RE. Organelle stress and alterations in interorganelle crosstalk during liver fibrosis. Hepatology 2024; 79:482-501. [PMID: 36626634 DOI: 10.1097/hep.0000000000000012] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/03/2022] [Indexed: 01/12/2023]
Abstract
The synchronous functioning and quality control of organelles ensure cell survival and function and are essential for maintaining homeostasis. Prolonged exposure to stressors (viruses, bacteria, parasitic infections, alcohol, drugs) or genetic mutations often disrupt the functional integrity of organelles which plays a critical role in the initiation and progression of several diseases including chronic liver diseases. One of the most important pathologic consequences of chronic liver diseases is liver fibrosis, characterized by tissue scarring due to the progressive accumulation of extracellular matrix components. Left untreated, fibrosis may advance to life-threatening complications such as cirrhosis, hepatic decompensation, and HCC, which collectively accounts for ∼1 million deaths per year worldwide. Owing to the lack of treatment options that can regress or reverse cirrhosis, liver transplantation is currently the only available treatment for end-stage liver disease. However, the limited supply of usable donor organs, adverse effects of lifelong immunosuppressive regimes, and financial considerations pose major challenges and limit its application. Hence, effective therapeutic strategies are urgently needed. An improved understanding of the organelle-level regulation of fibrosis can help devise effective antifibrotic therapies focused on reducing organelle stress, limiting organelle damage, improving interorganelle crosstalk, and restoring organelle homeostasis; and could be a potential clinical option to avoid transplantation. This review provides a timely update on the recent findings and mechanisms covering organelle-specific dysfunctions in liver fibrosis, highlights how correction of organelle functions opens new treatment avenues and discusses the potential challenges to clinical application.
Collapse
Affiliation(s)
- Saloni Sinha
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | | | | |
Collapse
|
31
|
Tanabe KK, Zahrieh D, Strand CA, Hoshida Y, Flotte TJ, Della’Zanna G, Umar A, Chavin KD, Cleary S, Kubota N, Llovet JM, Patel T, Siegel C, Limburg PJ. Epidermal Growth Factor Receptor Inhibition With Erlotinib in Liver: Dose De-Escalation Pilot Trial as an Initial Step in a Chemoprevention Strategy. GASTRO HEP ADVANCES 2024; 3:426-439. [PMID: 39131140 PMCID: PMC11307768 DOI: 10.1016/j.gastha.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 01/19/2024] [Indexed: 08/13/2024]
Abstract
Background and Aims Effective approaches for prevention of hepatocellular carcinoma (HCC) will have a significant impact on HCC-related mortality. There are strong preclinical data and rationale to support targeting epidermal growth factor receptor (EGFR) for HCC chemoprevention. Small molecule inhibitors of EGFR have been Food and Drug Administration-approved for cancer therapy, which provides an opportunity to repurpose one of these drugs for chemoprevention of HCC. Unfortunately, the frequency of side effects associated with administration of these drugs at oncology doses renders them ineffective for chemoprevention. This clinical trial assesses whether lower doses of one of these inhibitors, erlotinib, still engages EGFR in the liver to block signaling (eg, EGFR phosphorylation). The objective of this clinical trial was determination of a safe and minimum effective dose of erlorinib for which ≥ 50% reduction phospho-EGFR immunohistochemical staining in the liver was observed. Methods Forty six participants were preregistered and 25 participants were registered in this multicenter trial. By dose de-escalation trial design, cohorts of participants received a 7-day course of erlotinib 75 mg/day, 50 mg/day or 25 mg/day with liver tissue acquisition prior to and after erlotinib. Results A ≥50% reduction phospho-EGFR immunohistochemical staining in the liver was observed in a minimum of 40% of participants (predetermined threshhold) at each of the dose levels. Erlotinib was very well tolerated with few side effects observed, particularly at the dose of 25 mg/day. Favorable modulation of the Prognostic Liver Signature was observed in participants who received erlotinib. Conclusion These data support the selection of erlotinib doses as low as 25 mg/day of for a longer intervention to assess for evidence of efficacy as an HCC chemoprevention drug (ClinicalTrials.govNCT02273362).
Collapse
Affiliation(s)
- Kenneth K. Tanabe
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - David Zahrieh
- Division of Clinical Trial and Biostatistics, Mayo Clinic, Rochester, New York
| | - Carrie A. Strand
- Division of Clinical Trial and Biostatistics, Mayo Clinic, Rochester, New York
| | - Yujin Hoshida
- Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Thomas J. Flotte
- Mayo Clinic Pathology Research Core, Mayo Clinic, Rochester, New York
| | - Gary Della’Zanna
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Asad Umar
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Kenneth D. Chavin
- Department of Surgery, UH Cleveland Medical Center and Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Sean Cleary
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, New York
| | - Naoto Kubota
- Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Josep M. Llovet
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Liver Unit, Translational Research in Hepatic Oncology, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Tushar Patel
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida
| | | | - Paul J. Limburg
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, New York
| |
Collapse
|
32
|
Salama YA, Hassan HM, El-Gayar AM, Abdel-Rahman N. Combined quercetin and simvastatin attenuate hepatic fibrosis in rats by modulating SphK1/NLRP3 pathways. Life Sci 2024; 337:122349. [PMID: 38128755 DOI: 10.1016/j.lfs.2023.122349] [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: 09/15/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023]
Abstract
Liver fibrosis involves several signalling pathways working in concert regulating the deposition of extracellular matrix. In this study, we evaluated the effect of quercetin and simvastatin alone and their combination on the treatment of experimentally induced hepatic fibrosis in rats. To decipher the potential mechanisms involved, liver fibrosis was induced in rats by administration of 40 % carbon tetrachloride (CCl4) (1 μl/g rat, i.p., twice weekly) for 6 weeks. Quercetin (50 mg/kg, orally), simvastatin (40 mg/kg, orally) either individually or combined were administered for another 4 weeks. The three treatment groups ameliorated hepatic dysfunction and altered parameters of sphingolipid and pyroptosis pathways. Yet, the combined group showed a more pronounced effect. Treatments lowered serum levels of GOT, GPT, ALP and elevated albumin and total protein levels. Histopathological and electron microscope examination of liver tissue revealed diminished fibrosis and inflammation. Protein expression levels of α-SMA, IL-1β, PPAR-γ, TGF-β1, caspase-1 and caspase-3 expression in liver tissues were reduced. Additionally, hepatic mRNA levels of SphK1 and NLRP3 decreased after treatment. Furthermore, the three groups lowered MDA levels and elevated total antioxidant capacity, GSH and Nrf2 expression levels. Treatments downregulated sphingolipid pathway and NLRP3-mediated pyroptosis and stimulated an anti-apoptotic, anti-proliferative and antioxidant activity. This suggests that targeting the SphK1/NLRP3 pathway could be a prospective therapeutic strategy against liver fibrosis.
Collapse
Affiliation(s)
- Yasmin A Salama
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, 35516, Egypt; Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt
| | - Hanan M Hassan
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt
| | - Amal M El-Gayar
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, 35516, Egypt
| | - Noha Abdel-Rahman
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, 35516, Egypt.
| |
Collapse
|
33
|
Moore B, Jolly J, Izumiyama M, Kawai E, Ravasi T, Ryu T. Tissue-specific transcriptional response of post-larval clownfish to ocean warming. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 908:168221. [PMID: 37923256 DOI: 10.1016/j.scitotenv.2023.168221] [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: 05/24/2023] [Revised: 10/24/2023] [Accepted: 10/28/2023] [Indexed: 11/07/2023]
Abstract
Anthropogenically driven climate change is predicted to increase average sea surface temperatures, as well as the frequency and intensity of marine heatwaves in the future. This increasing temperature is predicted to have a range of negative physiological impacts on multiple life-stages of coral reef fish. Nevertheless, studies of early-life stages remain limited, and tissue-specific transcriptomic studies of post-larval coral reef fish are yet to be conducted. Here, in an aquaria-based study we investigate the tissue-specific (brain, liver, muscle, and digestive tract) transcriptomic response of post-larval (20 dph) Amphiprion ocellaris to temperatures associated with future climate change (+3 °C). Additionally, we utilized metatranscriptomic sequencing to investigate how the microbiome of the digestive tract changes at +3 °C. Our results show that the transcriptional response to elevated temperatures is highly tissue-specific, as the number of differentially expressed genes (DEGs) and gene functions varied amongst the brain (102), liver (1785), digestive tract (380), and muscle (447). All tissues displayed DEGs associated with thermal stress, as 23 heat-shock protein genes were upregulated in all tissues. Our results indicate that post-larval clownfish may experience liver fibrosis-like symptoms at +3 °C as genes associated with extracellular matrix structure, oxidative stress, inflammation, glucose transport, and metabolism were all upregulated. We also observe a shift in the digestive tract microbiome community structure, as Vibrio sp. replace Escherichia coli as the dominant bacteria. This shift is coupled with the dysregulation of various genes involved in immune response in the digestive tract. Overall, this study highlights post-larval clownfish will display tissue-specific transcriptomic responses to future increases in temperature, with many potentially harmful pathways activated at +3 °C.
Collapse
Affiliation(s)
- Billy Moore
- Marine Climate Change Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Jeffrey Jolly
- Marine Climate Change Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Michael Izumiyama
- Marine Climate Change Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Erina Kawai
- Marine Climate Change Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Timothy Ravasi
- Marine Climate Change Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Taewoo Ryu
- Marine Climate Change Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan.
| |
Collapse
|
34
|
Pringle TA, Ramon-Gil E, Leslie J, Oakley F, Wright MC, Knight JC, Luli S. Synthesis and preclinical evaluation of a 89Zr-labelled human single chain antibody for non-invasive detection of hepatic myofibroblasts in acute liver injury. Sci Rep 2024; 14:633. [PMID: 38182623 PMCID: PMC10770171 DOI: 10.1038/s41598-023-50779-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: 05/16/2023] [Accepted: 12/25/2023] [Indexed: 01/07/2024] Open
Abstract
Synaptophysin is expressed on fibrogenic hepatic myofibroblasts. C1-3 is a single chain human antibody (scAb) that binds specifically to synaptophysin on hepatic myofibroblasts, providing a targeting vector for novel in vivo imaging agents of chronic liver disease. C1-3 and a negative control scAb, CSBD9, were radiolabelled with zirconium-89 via desferrioxamine chelation to enable non-invasive molecular imaging with positron emission tomography (PET). DFO-scAb conjugates were characterised by gel electrophoresis (SDS-PAGE) and MALDI-TOF spectrometry, and 89Zr-labelled with high radiolabelling efficiency (99%). [89Zr]Zr-DFO-C1-3 exhibited high in vitro stability (> 99%) in mouse and human sera over 3 days at 25 and 37 °C. Activated hepatic myofibroblasts incubated with [89Zr]Zr-DFO-C1-3 displayed significantly higher internalised activity (59.46%, P = 0.001) compared to the [89Zr]Zr-DFO-CSBD9 control, indicating synaptophysin-mediated uptake and high binding specificity of [89Zr]Zr-DFO-C1-3. Mice with CCl4-induced acute liver damage exhibited significantly higher liver uptake of [89Zr]Zr-DFO-C1-3, compared to controls, confirmed by both Cerenkov imaging and ex vivo gamma counting (4.41 ± 0.19%ID/g, P < 0.0001). CCl4-induced liver damage and the number of hepatic myofibroblasts was confirmed by αSMA staining of liver sections. These findings indicate that [89Zr]Zr-DFO-C1-3 has promising utility as a PET imaging agent for non-invasive detection of hepatic myofibroblasts following acute liver injury.
Collapse
Affiliation(s)
- Toni A Pringle
- School of Natural and Environmental Sciences, Newcastle University, Bedson Building, Newcastle upon Tyne, NE1 7RU, UK
| | - Erik Ramon-Gil
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Newcastle Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - Jack Leslie
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Newcastle Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Newcastle Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - Matthew C Wright
- Liver Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - James C Knight
- School of Natural and Environmental Sciences, Newcastle University, Bedson Building, Newcastle upon Tyne, NE1 7RU, UK.
- Newcastle Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK.
| | - Saimir Luli
- Newcastle Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.
- Newcastle Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK.
- Preclinical In Vivo Imaging, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.
- Medical School, Newcastle University, 4th Floor William Leech Building, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
35
|
Zhang C, Xi Y, Zhang Y, He P, Su X, Fan F, Wu M, Kong X, Shi Y. Genetic association analysis of dietary intake and idiopathic pulmonary fibrosis: a two-sample mendelian randomization study. BMC Pulm Med 2024; 24:15. [PMID: 38178024 PMCID: PMC10768076 DOI: 10.1186/s12890-023-02831-8] [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: 09/26/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND IPF is a complex lung disease whose aetiology is not fully understood, but diet may have an impact on its development and progression. Therefore, we investigated the potential causal connection between dietary intake and IPF through TSMR to offer insights for early disease prevention recommendations. METHODS The study incorporated 29 dietary exposure factors, oily fish intake, bacon intake, processed meat intake, poultry intake, beef intake, pork intake, lamb/mutton intake, non-oily fish intake, fresh fruit intake, cooked vegetable intake, baked bean intake, fresh tomato intake, tinned tomato intake, salad/raw vegetable intake, Fresh fruit intake, coffee intake, tea intake, water intake, red wine intake, average weekly beer plus cider intake, alcoholic drinks per week, cereal intake, bread intake, whole-wheat intake, whole-wheat cereal intake, cheese intake, yogurt intake, salt added to food and whole egg intake. The study explored the causal link between diet and IPF using TSMR analysis, predominantly the IVW method, and performed sensitivity analyses to validate the results. RESULT The study revealed that consuming oily fish, yogurt, and dried fruits had a protective effect against IPF, whereas the consumption of alcoholic beverages and beef was linked to an increased risk of IPF. CONCLUSION In this MR study, it was discovered that the consumption of oily fish, yogurt, and dried fruits exhibited a protective effect against IPF, whereas the intake of alcoholic beverages and beef was associated with an elevated risk of IPF. These findings underscore the significance of making informed and timely dietary decisions in IPF prevention.
Collapse
Affiliation(s)
- Chenwei Zhang
- Department of Respiratory and Critical Care Medicine, Shanxi Medical University Affiliated First Hospital, Taiyuan, 030000, China
| | - Yujia Xi
- Department of Urology, The Second Hospital of Shanxi Medical University, Taiyuan, 030000, China
| | - Yukai Zhang
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan, 030000, China
| | - Peiyun He
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan, 030000, China
| | - Xuesen Su
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan, 030000, China
| | - Fangfang Fan
- Department of Respiratory and Critical Care Medicine, Shanxi Medical University Affiliated First Hospital, Taiyuan, 030000, China
| | - Min Wu
- Department of Respiratory and Critical Care Medicine, Shanxi Medical University Affiliated First Hospital, Taiyuan, 030000, China
| | - Xiaomei Kong
- Department of Respiratory and Critical Care Medicine, Shanxi Medical University Affiliated First Hospital, Taiyuan, 030000, China.
- NHC Key Laboratory of Pneumoconiosis, Taiyuan, 030000, China.
| | - Yiwei Shi
- Department of Respiratory and Critical Care Medicine, Shanxi Medical University Affiliated First Hospital, Taiyuan, 030000, China.
- NHC Key Laboratory of Pneumoconiosis, Taiyuan, 030000, China.
| |
Collapse
|
36
|
Odeghe E, Oyeleke G, Odofin M, Duguru M, Davwar P, Nyam D, Lesi O, Okeke E, Adelabu H, Odukoya O, Akanmu A, Adeyemo W, Abdulkareem F, Imade G, Joyce B, Khan I, Chandler A, Sagay A, Murphy R, Hou L, Hawkins C. Hepatitis B and C Virus Co-Infection and Their Association With Liver Disease in Persons With HIV in Nigeria. J Int Assoc Provid AIDS Care 2024; 23:23259582241292511. [PMID: 39469965 PMCID: PMC11528674 DOI: 10.1177/23259582241292511] [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: 05/15/2024] [Revised: 09/11/2024] [Accepted: 09/28/2024] [Indexed: 10/30/2024] Open
Abstract
PLAIN LANGUAGE SUMMARY Hepatitis B and C infection and liver disease in people with HIV infection in NigeriaPeople living with human immunodeficiency virus (HIV) infection who have hepatitis B or C infection have a higher chance of developing advanced liver disease than those who do not have either of the hepatitis infections. This finding highlights the importance of awareness, screening, and treatment of the hepatitis viruses in HIV programs in order to reduce the risk of liver disease in this population.
Collapse
Affiliation(s)
- Emuobor Odeghe
- Department of Medicine, University of Lagos/Lagos University Teaching Hospital, Lagos, Nigeria
| | - Ganiyat Oyeleke
- Department of Medicine, University of Lagos/Lagos University Teaching Hospital, Lagos, Nigeria
| | - Mayowa Odofin
- Department of Haematology and Blood Transfusion, University of Lagos/Lagos University Teaching Hospital, Lagos, Nigeria
| | - Mary Duguru
- Department of Medicine, University of Jos/Jos University Teaching Hospital, Plateau, Nigeria
| | - Pantong Davwar
- Department of Medicine, University of Jos/Jos University Teaching Hospital, Plateau, Nigeria
| | - David Nyam
- Department of Medicine, University of Jos/Jos University Teaching Hospital, Plateau, Nigeria
| | - Olufunmilayo Lesi
- Department of Medicine, University of Lagos/Lagos University Teaching Hospital, Lagos, Nigeria
| | - Edith Okeke
- Department of Medicine, University of Jos/Jos University Teaching Hospital, Plateau, Nigeria
| | - Hameed Adelabu
- Department of Haematology and Blood Transfusion, University of Lagos/Lagos University Teaching Hospital, Lagos, Nigeria
| | - Oluwakemi Odukoya
- Department of Community Health and Primary Care, University of Lagos/Lagos University Teaching Hospital, Lagos, Nigeria
| | - Alani Akanmu
- Department of Haematology and Blood Transfusion, University of Lagos/Lagos University Teaching Hospital, Lagos, Nigeria
| | - Wasiu Adeyemo
- Department of Oral and Maxillofacial Surgery, University of Lagos/Lagos University Teaching Hospital, Lagos, Nigeria
| | - Fatimah Abdulkareem
- Department of Anatomic and Molecular Pathology, University of Lagos/Lagos University Teaching Hospital, Lagos, Nigeria
| | - Godwin Imade
- Department of Obstetrics and Gynaecology, University of Jos/Jos University Teaching Hospital, Plateau, Nigeria
| | - Brian Joyce
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Imran Khan
- School of Professional Studies, Northwestern University, Evanston, IL, USA
| | - Ariel Chandler
- School of Professional Studies, Northwestern University, Evanston, IL, USA
| | - Atiene Sagay
- Department of Obstetrics and Gynaecology, University of Jos/Jos University Teaching Hospital, Plateau, Nigeria
| | - Robert Murphy
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Claudia Hawkins
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
37
|
Radwan AM, Fatoh SA, Massoud A, Tousson E. Effectiveness of curcumin nanoparticles in rat liver fibrosis caused by thioacetamide. ENVIRONMENTAL TOXICOLOGY 2024; 39:388-397. [PMID: 37782692 DOI: 10.1002/tox.23984] [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: 02/24/2023] [Revised: 08/03/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023]
Abstract
Although curcumin possesses anti-inflammatory, antioxidant, and cytoprotective qualities, its low absorption limits its medicinal uses. Before examining how curcumin influenced rats' liver fibrosis when thioacetamide (TAA) was produced, the current study employed nanoparticles (NPs) to improve curcumin bioavailability. Sixty mature rats were separated into six groups (Group 1, control; Group 2, curcumin; Group 3, curcumin nanoparticles; Group 4, TAA; Group 5, TAA + curcumin; Group 6, TAA + curcumin NPs). TAA administration caused considerable increases in serum liver enzymes associated with a remarkable depletion in the levels of albumin and total protein relative to the control. In addition, a significant elevation in malonaldehyde (MDA) level with a significant depletion in the antioxidant enzymes activity was detected. Also, TAA had a significant effect on the inflammation markers represented by the elevation in tumor necrosis factor (TNFα) and DNA damage. Administration of curcumin or curcumin NPs in TAA-intoxicated rats significantly (p < .001, p < .0001) alleviates liver injury by correcting antioxidant status, inflammatory markers, and oxidative stress. The results of comparing TAA-intoxicated rats treated with curcumin NPs to TAA-intoxicated rats treated with bulk curcumin revealed that the ameliorative effect of nanocurcumin was stronger. These observations concluded that nanoparticle formulation can increase curcumin bioavailability and solubility, enhancing its antioxidant and anti-inflammatory efficiency, resulting in greater potential against thioacetamide-induced hepatotoxicity in rats.
Collapse
Affiliation(s)
- Aliaa M Radwan
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Shahenda A Fatoh
- Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Ahmed Massoud
- Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Ehab Tousson
- Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
38
|
Verma S, Ishteyaque S, Washimkar KR, Verma S, Nilakanth Mugale M. Mitochondrial-mediated nuclear remodeling and macrophage polarizations: A key switch from liver fibrosis to HCC progression. Exp Cell Res 2024; 434:113878. [PMID: 38086504 DOI: 10.1016/j.yexcr.2023.113878] [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: 07/28/2023] [Revised: 10/24/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023]
Abstract
Liver fibrosis is a significant health burden worldwide and has emerged as the leading cause of Hepatocellular carcinoma (HCC) incidence. Mitochondria are the dynamic organelles that regulate the differentiation, survival, and polarization of macrophages. Nuclear-DNA-associated proteins, micro-RNAs, as well as macrophage polarization are essential for maintaining intracellular and extra-cellular homeostasis in the liver parenchyma. Dysregulated mitochondrial coding genes (ETS complexes I, II, III, IV, and V), non-coding RNAs (mitomiRs), and nuclear alteration lead to the production of reactive oxygen species (ROS) and inflammation which are implicated in the transition of liver fibrosis into HCC. Recent findings indicated the protecting effect of E74-like factor 3/peroxisome proliferator-activated receptor-γ (Elf-3/PPAR-γ). HDAR-y inhibits the deacetylation of PPAR-y and maintains the PPAR-y pathway. Elf-3 plays a tumor suppressive role through epithelial-mesenchymal transition-related gene and zinc finger E-box binding homeobox 2 (ZEB-2) domain. Additionally, the development of HCC includes the PI3K/Akt/mTOR and transforming Growth Factor β (TGF-β) pathway that promotes the Epithelial-mesenchymal transition (EMT) through Smad/Snail/Slug signaling cascade. In contrast, the TLR2/NOX2/autophagy axis promotes M2 polarization in HCC. Thus, a thorough understanding of the mitochondrial and nuclear reciprocal relationship related to macrophage polarization could provide new research opportunities concerning diseases with a significant impact on liver parenchyma towards developing liver fibrosis or liver cancer. Moreover, this knowledge can be used to develop new therapeutic strategies to treat liver diseases.
Collapse
Affiliation(s)
- Shobhit Verma
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sharmeen Ishteyaque
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kaveri R Washimkar
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Smriti Verma
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Madhav Nilakanth Mugale
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute (CSIR-CDRI), Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
39
|
Sharma N, Sistla R, Andugulapati SB. Yohimbine ameliorates liver inflammation and fibrosis by regulating oxidative stress and Wnt/β-catenin pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155182. [PMID: 37952411 DOI: 10.1016/j.phymed.2023.155182] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/17/2023] [Accepted: 11/01/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND AND PURPOSE Chronic liver injury, caused by various aetiologies, causes recurrent tissue damage, culminating in decreased liver regenerative ability and resulting in fibrosis followed by cirrhosis. In this study, the anti-fibrotic activity of Yohimbine hydrochloride (YHC) was investigated using various in vitro models and in vivo models. METHODS To assess the anti-inflammatory, antioxidant, and anti-fibrotic effects of YHC, lipopolysaccharide or TGF-β induced differentiation or lipid-induced oxidative-stress models were employed using HLECs, HSC-LX2, and HepG2 cells. Further, thioacetamide (TAA) induced hepatic inflammation/fibrosis models were utilized to validate the YHC's anti-fibrotic activity in rats. RESULTS Inflammation/differentiation experiments in HLECs and HSC-LX2 revealed that YHC treatment significantly (p < 0.001) mitigated the lipopolysaccharide or TGF-β induced upregulation of inflammatory and fibrotic markers expression respectively. In addition, YHC dose-dependently reduced the TGF-β induced migration and palmitic acid-induced oxidative stress in HepG2 cells. Further, TAA administration (5 weeks) in vivo rat model showed increased inflammatory marker levels/expression, oxidative stress, and pathological abnormalities. Additionally, TAA administration (9 weeks) elevated the fibrotic marker expression, collagen deposition in liver tissues, and shortened longevity in rats. Treatment with YHC dose-dependently mitigated the TAA-induced abnormalities in both inflammation and fibrosis models and improved the survival of the rats. Further mechanistic approaches revealed that TAA administration elevated the JNK, Wnt components and β-catenin expression in hepatic stellate cells and animal tissues. Further treatment with YHC significantly modulated the JNK/Wnt/β-catenin signaling. Moreover, the β-catenin nuclear translocation results showed that β-catenin levels were significantly elevated in the nuclear fraction of TAA control samples and reduced in YHC-treated samples. CONCLUSION Yohimbine treatment significantly improved inflammation and fibrosis by inhibiting differentiation, oxidative stress, and collagen deposition by partly modulating the JNK/Wnt/β-catenin pathway. These results might serve as a foundation for proposing yohimbine as a potential lead compound for liver fibrosis.
Collapse
Affiliation(s)
- Nidhi Sharma
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad-500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh-201 002, India
| | - Ramakrishna Sistla
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad-500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh-201 002, India
| | - Sai Balaji Andugulapati
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Hyderabad-500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh-201 002, India.
| |
Collapse
|
40
|
Yang P, Jin L, Liao J, Jin K, Shao X, Li C, Qian J, Cheng J, Yu D, Guo R, Xu X, Lu X, Fan X. Revealing spatial multimodal heterogeneity in tissues with SpaTrio. CELL GENOMICS 2023; 3:100446. [PMID: 38116121 PMCID: PMC10726534 DOI: 10.1016/j.xgen.2023.100446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/28/2023] [Accepted: 10/26/2023] [Indexed: 12/21/2023]
Abstract
Capturing and depicting the multimodal tissue information of tissues at the spatial scale remains a significant challenge owing to technical limitations in single-cell multi-omics and spatial transcriptomics sequencing. Here, we developed a computational method called SpaTrio that can build spatial multi-omics data by integrating these two datasets through probabilistic alignment and enabling further analysis of gene regulation and cellular interactions. We benchmarked SpaTrio using simulation datasets and demonstrated its accuracy and robustness. Next, we evaluated SpaTrio on biological datasets and showed that it could detect topological patterns of cells and modalities. SpaTrio has also been applied to multiple sets of actual data to uncover spatially multimodal heterogeneity, understand the spatiotemporal regulation of gene expression, and resolve multimodal communication among cells. Our data demonstrated that SpaTrio could accurately map single cells and reconstruct the spatial distribution of various biomolecules, providing valuable multimodal insights into spatial biology.
Collapse
Affiliation(s)
- Penghui Yang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314103, China
| | - Lijun Jin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314103, China
| | - Jie Liao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314103, China
| | - Kaiyu Jin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xin Shao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314103, China
| | - Chengyu Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314103, China
| | - Jingyang Qian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314103, China
| | - Junyun Cheng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Dingyi Yu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Rongfang Guo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiao Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiaoyan Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Jinhua Institute of Zhejiang University, Jinhua 321016 China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China.
| | - Xiaohui Fan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314103, China; Jinhua Institute of Zhejiang University, Jinhua 321016 China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China.
| |
Collapse
|
41
|
Cuesta ÁM, Palao N, Bragado P, Gutierrez-Uzquiza A, Herrera B, Sánchez A, Porras A. New and Old Key Players in Liver Cancer. Int J Mol Sci 2023; 24:17152. [PMID: 38138981 PMCID: PMC10742790 DOI: 10.3390/ijms242417152] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Liver cancer represents a major health problem worldwide with growing incidence and high mortality, hepatocellular carcinoma (HCC) being the most frequent. Hepatocytes are likely the cellular origin of most HCCs through the accumulation of genetic alterations, although hepatic progenitor cells (HPCs) might also be candidates in specific cases, as discussed here. HCC usually develops in a context of chronic inflammation, fibrosis, and cirrhosis, although the role of fibrosis is controversial. The interplay between hepatocytes, immune cells and hepatic stellate cells is a key issue. This review summarizes critical aspects of the liver tumor microenvironment paying special attention to platelets as new key players, which exert both pro- and anti-tumor effects, determined by specific contexts and a tight regulation of platelet signaling. Additionally, the relevance of specific signaling pathways, mainly HGF/MET, EGFR and TGF-β is discussed. HGF and TGF-β are produced by different liver cells and platelets and regulate not only tumor cell fate but also HPCs, inflammation and fibrosis, these being key players in these processes. The role of C3G/RAPGEF1, required for the proper function of HGF/MET signaling in HCC and HPCs, is highlighted, due to its ability to promote HCC growth and, regulate HPC fate and platelet-mediated actions on liver cancer.
Collapse
Affiliation(s)
- Ángel M. Cuesta
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (Á.M.C.); (N.P.); (P.B.); (A.G.-U.); (B.H.); (A.S.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Nerea Palao
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (Á.M.C.); (N.P.); (P.B.); (A.G.-U.); (B.H.); (A.S.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Paloma Bragado
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (Á.M.C.); (N.P.); (P.B.); (A.G.-U.); (B.H.); (A.S.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Alvaro Gutierrez-Uzquiza
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (Á.M.C.); (N.P.); (P.B.); (A.G.-U.); (B.H.); (A.S.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Blanca Herrera
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (Á.M.C.); (N.P.); (P.B.); (A.G.-U.); (B.H.); (A.S.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD-ISCIII), 28040 Madrid, Spain
| | - Aránzazu Sánchez
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (Á.M.C.); (N.P.); (P.B.); (A.G.-U.); (B.H.); (A.S.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD-ISCIII), 28040 Madrid, Spain
| | - Almudena Porras
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain; (Á.M.C.); (N.P.); (P.B.); (A.G.-U.); (B.H.); (A.S.)
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| |
Collapse
|
42
|
Ruiz de Galarreta M, Arriazu E, Pérez de Obanos MP, Ansorena E, Iraburu MJ. Antifibrogenic and apoptotic effects of Ocoxin in cultured rat hepatic stellate cells. J Physiol Biochem 2023; 79:881-890. [PMID: 35239161 PMCID: PMC10635942 DOI: 10.1007/s13105-022-00878-5] [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: 11/02/2021] [Accepted: 01/25/2022] [Indexed: 10/18/2022]
Abstract
Ocoxin is a nutritional supplement that has been shown to exert antioxidant and immunomodulatory responses in patients with chronic hepatitis C. The present work aimed to determine the effects of Ocoxin on activated hepatic stellate cells (HSC), the cell type mainly responsible for collagen deposition in the fibrotic liver. Ocoxin was found to reduce the survival of a cell line of immortalized non-tumoral rat HSC in a dose-response fashion and to diminish collagen type I levels. This latter effect was observed even at doses not affecting cell survival, pointing to an antifibrogenic action for the supplement. The decrease in viability exerted by Ocoxin on HSC correlated with an increase in histone-associated fragments in the cytoplasm and with increased activity of caspase-3, indicating the induction of apoptosis. To determine the molecular mechanisms mediating Ocoxin-induced apoptosis, the activation of members of the MAPK family was analyzed. Incubation of HSC with Ocoxin caused a transient and dramatic enhancement on ERK, JNK, and p38 MAPK phosphorylation levels. Using specific inhibitors for these enzymes, p38 MAPK was identified as a key mediator of the apoptotic effect of Ocoxin on HSC.
Collapse
Affiliation(s)
| | - Elena Arriazu
- Department of Biochemistry and Genetics, University of Navarra, 31008, Pamplona, Spain
| | | | - Eduardo Ansorena
- Department of Biochemistry and Genetics, University of Navarra, 31008, Pamplona, Spain
| | - María J Iraburu
- Department of Biochemistry and Genetics, University of Navarra, 31008, Pamplona, Spain.
| |
Collapse
|
43
|
Shu Y, He Y, Ye G, Liu X, Huang J, Zhang Q, Tian D, Wang T, Shu J. Curcumin inhibits the activity and induces apoptosis of activated hepatic stellate cell by suppressing autophagy. J Cell Biochem 2023; 124:1764-1778. [PMID: 37909649 DOI: 10.1002/jcb.30487] [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: 02/01/2023] [Revised: 09/09/2023] [Accepted: 09/25/2023] [Indexed: 11/03/2023]
Abstract
Curcumin, a kind of natural compound, has been previously proven to inhibit the autophagy in hepatic stellate cells (HSCs) and induce their apoptosis. However, it is not clear whether the enhanced apoptosis of activated HSCs (aHSCs) caused by curcumin depends on autophagy inhibition. We aim to verify this hypothesis and explore the potential mechanisms in this study. Immortalized human HSC line LX-2 was used as an experimental specimen and pretreated with transforming growth factor β1(TGF-β1) for 24 h to activate it before drug application. The levels of autophagy, apoptosis, cell activity, lipid metabolism, and the activity of the PI3K/Akt/mTOR signal pathway were evaluated by multiple methods, such as Western blotting, mcherry-EGFP-LC3B adenoviruses transfection, immunofluorescence, Nile Red staining, flow cytometry among others. Our results showed that rapamycin, an autophagy activator, could partly offset the effects of curcumin on autophagy and apoptosis of LX-2 cells, while 3-Methyladenine (3-MA), an autophagy inhibitor, could enhance these effects. Furthermore, curcumin could promote the activity of the PI3K/Akt/mTOR signal pathway in LX-2 cells, while PI3K inhibitor could partly offset this effect and increase the autophagy level. Overall, we demonstrated that curcumin could inhibit the activity and promote LX-2 cells apoptosis by suppressing autophagy by activating the PI3K/Akt/mTOR signal pathway. In addition, lipid recovery and energy deprivation due to autophagy inhibition may be the exact mechanism by which curcumin attenuates the pro-fibrotic activity of LX-2.
Collapse
Affiliation(s)
- Yongxiang Shu
- Department of Gastroenterology, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Yajun He
- Department of Clinical laboratory, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Guorong Ye
- Department of Gastroenterology, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Xuyou Liu
- Department of Gastroenterology, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Jiahuang Huang
- Department of Gastroenterology, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Qinghui Zhang
- Department of Gastroenterology, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Da Tian
- Department of Gastroenterology, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Tengyan Wang
- Department of Gastroenterology, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Jianchang Shu
- Department of Gastroenterology, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
44
|
Chen W, Sun Y, Chen S, Ge X, Zhang W, Zhang N, Wu X, Song Z, Han H, Desert R, Yan X, Yang A, Das S, Athavale D, Nieto N, You H. Matrisome gene-based subclassification of patients with liver fibrosis identifies clinical and molecular heterogeneities. Hepatology 2023; 78:1118-1132. [PMID: 37098756 PMCID: PMC10524702 DOI: 10.1097/hep.0000000000000423] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 03/27/2023] [Indexed: 04/27/2023]
Abstract
BACKGROUND AIMS Excessive deposition and crosslinking of extracellular matrix increases liver density and stiffness, promotes fibrogenesis, and increases resistance to fibrinolysis. An emerging therapeutic opportunity in liver fibrosis is to target the composition of the extracellular matrix or block pathogenic communication with surrounding cells. However, the type and extent of extracellular changes triggering liver fibrosis depend on the underlying etiology. Our aim was to unveil matrisome genes not dependent on etiology, which are clinically relevant to liver fibrosis. APPROACH RESULTS We used transcriptomic profiles from liver fibrosis cases of different etiologies to identify and validate liver fibrosis-specific matrisome genes (LFMGs) and their clinical and biological relevance. Dysregulation patterns and cellular landscapes of LFMGs were further explored in mouse models of liver fibrosis progression and regression by bulk and single-cell RNA sequencing. We identified 35 LFMGs, independent of etiology, representing an LFMG signature defining liver fibrosis. Expression of the LFMG signature depended on histological severity and was reduced in regressive livers. Patients with liver fibrosis, even with identical pathological scores, could be subclassified into LFMG Low and LFMG High , with distinguishable clinical, cellular, and molecular features. Single-cell RNA sequencing revealed that microfibrillar-associated protein 4 + activated HSC increased in LFMG High patients and were primarily responsible for the LFMG signature expression and dysregulation. CONCLUSIONS The microfibrillar-associated protein 4 + -activated HSC-derived LFMG signature classifies patients with liver fibrosis with distinct clinical and biological characteristics. Our findings unveil hidden information from liver biopsies undetectable using traditional histologic assessments.
Collapse
Affiliation(s)
- Wei Chen
- Beijing Clinical Research Institute, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Yameng Sun
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Shuyan Chen
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Xiaodong Ge
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Wen Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Ning Zhang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Xiaoning Wu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Zhuolun Song
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Hui Han
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Romain Desert
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Xuzhen Yan
- Beijing Clinical Research Institute, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Aiting Yang
- Beijing Clinical Research Institute, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| | - Sukanta Das
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Dipti Athavale
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
| | - Natalia Nieto
- Department of Pathology, University of Illinois at Chicago, 840 S. Wood St., Suite 130 CSN, MC 847, Chicago, IL 60612, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, 840 S. Wood St., Suite 1020N, MC 787, Chicago, IL 60612, USA
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China
| |
Collapse
|
45
|
Mahli A, Thasler WE, Biendl M, Hellerbrand C. Hop-derived Humulinones Reveal Protective Effects in in vitro Models of Hepatic Steatosis, Inflammation and Fibrosis. PLANTA MEDICA 2023; 89:1138-1146. [PMID: 37343573 DOI: 10.1055/a-2103-3230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/23/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is emerging as leading cause of liver disease worldwide. Specific pharmacologic therapy for NAFLD is a major unmet medical need. Recently, iso-alpha acids, hop-derived bitter compounds in beer, have been shown to beneficially affect NAFLD pathology. Humulinones are further hop derived bitter acids particularly found in modern styles of beer. So far, biological effects of humulinones have been unknown. Here, we investigated the effect of humulinones in in vitro models for hepatic steatosis, inflammation and fibrosis. Humulinones dose-dependently inhibited fatty acid induced lipid accumulation in primary human hepatocytes. Humulinones reduced the expression of fatty acid uptake transporter CD36 and key enzymes of (de novo) lipid synthesis. Conversely, humulinones increased the expression of FABP1, CPT1 and ACOX1, indicative for increased lipid combustion. Furthermore, humulinones ameliorated steatosis induced pro-inflammatory gene expression. Furthermore, humulinones significantly reduced the expression of pro-inflammatory and pro-fibrogenic factors in control as well as lipopolysaccharide treated activated hepatic stellate cells, which play a key role in hepatic fibrosis. In conclusion, humulinones beneficially affect different pathophysiological steps of NAFLD. Our data suggest humulinones as promising therapeutic agents for the prevention and treatment of NAFLD.
Collapse
Affiliation(s)
- Abdo Mahli
- Institute of Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
| | - Wolfgang E Thasler
- Human Tissue and Cell Research-Services GmbH, Planegg/Martinsried, Germany
| | - Martin Biendl
- Hopsteiner, Hallertauer Hopfenveredelung GmbH, Mainburg, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
46
|
Sztolsztener K, Konstantynowicz-Nowicka K, Pędzińska-Betiuk A, Chabowski A. Concentration-Dependent Attenuation of Pro-Fibrotic Responses after Cannabigerol Exposure in Primary Rat Hepatocytes Cultured in Palmitate and Fructose Media. Cells 2023; 12:2243. [PMID: 37759466 PMCID: PMC10526512 DOI: 10.3390/cells12182243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Hepatic fibrosis is a consequence of liver injuries, in which the overproduction and progressive accumulation of extracellular matrix (ECM) components with the simultaneous failure of matrix turnover mechanisms are observed. The aim of this study was to investigate the concentration-dependent influence of cannabigerol (CBG, Cannabis sativa L. component) on ECM composition with respect to transforming growth factor beta 1 (TGF-β1) changes in primary hepatocytes with fibrotic changes induced by palmitate and fructose media. Cells were isolated from male Wistar rats' livers in accordance with the two-step collagenase perfusion technique. This was followed by hepatocytes incubation with the presence or absence of palmitate with fructose and/or cannabigerol (at concentrations of 1, 5, 10, 15, 25, 30 µM) for 48 h. The expression of ECM mRNA genes and proteins was determined using PCR and Western blot, respectively, whereas media ECM level was evaluated using ELISA. Our results indicated that selected low concentrations of CBG caused a reduction in TGF-β1 mRNA expression and secretion into media. Hepatocyte exposure to cannabigerol at low concentrations attenuated collagen 1 and 3 deposition. The protein and/or mRNA expressions and MMP-2 and MMP-9 secretion were augmented using CBG. Considering the mentioned results, low concentrations of cannabigerol treatment might expedite fibrosis regression and promote regeneration.
Collapse
Affiliation(s)
- Klaudia Sztolsztener
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland; (K.K.-N.); (A.C.)
| | | | - Anna Pędzińska-Betiuk
- Department of Experimental Physiology and Pathophysiology, Medical University of Bialystok, 15-089 Bialystok, Poland;
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland; (K.K.-N.); (A.C.)
| |
Collapse
|
47
|
Jain I, Brougham-Cook A, Underhill GH. Effect of distinct ECM microenvironments on the genome-wide chromatin accessibility and gene expression responses of hepatic stellate cells. Acta Biomater 2023; 167:278-292. [PMID: 37343907 PMCID: PMC10527607 DOI: 10.1016/j.actbio.2023.06.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/25/2023] [Accepted: 06/14/2023] [Indexed: 06/23/2023]
Abstract
Hepatic stellate cells (HSCs) are one of the primary drivers of liver fibrosis in non-alcoholic fatty liver disease. Although HSC activation in liver disease is associated with changes in extracellular matrix (ECM) deposition and remodeling, it remains unclear how ECM regulates the phenotypic state transitions of HSCs. Using high-throughput cellular microarrays, coupled with genome-wide ATAC and RNA sequencing within engineered ECM microenvironments, we investigated the effect of ECM and substrate stiffness on chromatin accessibility and resulting gene expression in activated primary human HSCs. Cell microarrays demonstrated the cooperative effects of stiffness and ECM composition on H3K4 and H3K9 methylation/acetylation. ATAC sequencing revealed higher chromatin accessibility in HSCs on 1kPa compared to 25kPa substrates for all ECM conditions. Gene set enrichment analysis using RNA sequencing data of HSCs in defined ECM microenvironments demonstrated higher enrichment of NAFLD and fibrosis-related genes in pre-activated HSCs on 1kPa relative to 25kPa. Overall, these findings are indicative of a microenvironmental adaptation response in HSCs, and the acquisition of a persistent activation state. Combined ATAC/RNA sequencing analyses enabled identification of candidate regulatory factors, including HSD11B1 and CEBPb. siRNA-mediated knockdown of HSD11b1 and CEBPb demonstrated microenvironmental controlled reduction in fibrogenic markers in HSCs. STATEMENT OF SIGNIFICANCE: Hepatic stellate cells (HSCs) are one of the primary drivers of liver fibrosis in non-alcoholic fatty liver disease. Although HSC activation in liver disease is associated with changes in extracellular matrix (ECM) deposition and remodeling, it remains unclear how ECM regulates the phenotypic state transitions of HSCs. Using high-throughput cellular microarrays, coupled with genome-wide ATAC and RNA sequencing within engineered ECM microenvironments, we investigated the effect of ECM and substrate stiffness on chromatin accessibility and resulting gene expression in activated primary human HSCs. Overall, these findings were indicative of a microenvironmental adaptation response in HSCs, and the acquisition of a persistent activation state. Combined ATAC/RNA sequencing analyses enabled identification of candidate regulatory factors, including HSD11B1 and CEBPb. siRNA-mediated knockdown of HSD11b1 and CEBPb demonstrated microenvironmental controlled reduction in fibrogenic markers in HSCs.
Collapse
Affiliation(s)
- Ishita Jain
- University of Illinois at Urbana Champaign, Urbana, USA
| | | | | |
Collapse
|
48
|
Allameh A, Niayesh-Mehr R, Aliarab A, Sebastiani G, Pantopoulos K. Oxidative Stress in Liver Pathophysiology and Disease. Antioxidants (Basel) 2023; 12:1653. [PMID: 37759956 PMCID: PMC10525124 DOI: 10.3390/antiox12091653] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/15/2023] [Accepted: 08/20/2023] [Indexed: 09/29/2023] Open
Abstract
The liver is an organ that is particularly exposed to reactive oxygen species (ROS), which not only arise during metabolic functions but also during the biotransformation of xenobiotics. The disruption of redox balance causes oxidative stress, which affects liver function, modulates inflammatory pathways and contributes to disease. Thus, oxidative stress is implicated in acute liver injury and in the pathogenesis of prevalent infectious or metabolic chronic liver diseases such as viral hepatitis B or C, alcoholic fatty liver disease, non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). Moreover, oxidative stress plays a crucial role in liver disease progression to liver fibrosis, cirrhosis and hepatocellular carcinoma (HCC). Herein, we provide an overview on the effects of oxidative stress on liver pathophysiology and the mechanisms by which oxidative stress promotes liver disease.
Collapse
Affiliation(s)
- Abdolamir Allameh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 1411713116, Iran; (A.A.); (R.N.-M.); (A.A.)
| | - Reyhaneh Niayesh-Mehr
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 1411713116, Iran; (A.A.); (R.N.-M.); (A.A.)
| | - Azadeh Aliarab
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 1411713116, Iran; (A.A.); (R.N.-M.); (A.A.)
| | - Giada Sebastiani
- Chronic Viral Illness Services, McGill University Health Center, Montreal, QC H4A 3J1, Canada;
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Kostas Pantopoulos
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| |
Collapse
|
49
|
Li H, Wang H, Yang A, Xue M, Wang J, Lv Q, Liu J, Hu L, Zhang Y, Wang X. Gypenosides Synergistically Reduce the Extracellular Matrix of Hepatic Stellate Cells and Ameliorate Hepatic Fibrosis in Mice. Molecules 2023; 28:5448. [PMID: 37513321 PMCID: PMC10386501 DOI: 10.3390/molecules28145448] [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: 06/25/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Liver fibrosis resulting from chronic liver damage is becoming one of the major threats to health worldwide. Active saponin constituents isolated from Gynostemma pentaphyllum were found to possess a protective effect in liver diseases. Here, we obtained a naturally abundant gypenoside, XLVI, and evaluated its liver protection activity in both animal and cellular models. The results showed that it ameliorated acute and chronic liver injuries and lightened the process of fibrogenesis in vivo. XLVI can inhibit TGF-β-induced activation of hepatic stellate cells and ECM deposition in vitro. The underlying mechanism study verified that it upregulated the protein expression of protein phosphatase 2C alpha and strengthened the vitality of the phosphatase together with a PP2Cα agonist gypenoside NPLC0393. These results shed new light on the molecular mechanisms and the potential therapeutic function of the traditional herb Gynostemma pentaphyllum in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Han Li
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hanghang Wang
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Aiping Yang
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Mingzhen Xue
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Junyang Wang
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qi Lv
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jian Liu
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lihong Hu
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yinan Zhang
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiachang Wang
- Jiangsu Key Laboratory for Functional Substances of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Fujian Province Key Laboratory for the Development of Bioactive Material from Marine Algae, College of Oceanology and Food Science, Quanzhou Normal University, Quanzhou 362000, China
| |
Collapse
|
50
|
Abdelfattah AM, Mahmoud SS, El-Wafaey DI, Abdelgeleel HM, Abdelhamid AM. Diacerein ameliorates cholestasis-induced liver fibrosis in rat via modulating HMGB1/RAGE/NF-κB/JNK pathway and endoplasmic reticulum stress. Sci Rep 2023; 13:11455. [PMID: 37454204 PMCID: PMC10349817 DOI: 10.1038/s41598-023-38375-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023] Open
Abstract
Diacerein is an interleukin (IL)-1β inhibitor approved for osteoarthritis. This study aimed to investigate the potential anti-fibrotic effect of diacerein against bile duct ligation (BDL)-induced liver fibrosis. Forty male Wistar rats were divided into: sham-operated group, BDL group, and BDL groups treated with diacerein at 10, 30, and 50 mg/kg/day starting two days before surgery and continued for 4 weeks. Diacerein decreased the hepatic injury markers and alleviated oxidative stress triggered by BDL by reducing hepatic malondialdehyde (MDA) and increasing hepatic superoxide dismutase (SOD) levels. Diacerein mitigated BDL-induced inflammation via lowering hepatic levels and mRNA expression of high mobility group box 1 (HMGB1), nuclear factor-κB (NF-κB), and IL-1β. The hepatic gene expression of Advanced Glycation End products Receptor (RAGE) gene and immunohistochemical expression of some ER stress markers, e.g., glucose-regulated protein 78 (GRP78), inositol-requiring enzyme 1 (IRE1α), protein kinase RNA-like endoplasmic reticulum kinase (PERK), CCAAT/enhancer-binding protein homologous protein (CHOP), and phosphorylated c-Jun N-terminal kinase protein contents were lowered by diacerein. Furthermore, diacerein suppressed the hepatic levels of fibrogenic mediators, e.g., Transforming growth factor β1 (TGF-β1), α- smooth muscle actin (α-SMA), collagen 1, and hydroxyproline, as well as the apoptotic caspase 3 and BAX immunostaining in BDL rats. The histopathological abnormalities induced by BDL significantly improved. Our study demonstrated that diacerein exhibited an antifibrotic effect by inhibiting HMGB1/RAGE/NF-κB/JNK pathway, and ER stress. Better protection was observed with increasing the dose.
Collapse
Affiliation(s)
| | - Shireen Sami Mahmoud
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, Sharkia, Egypt
| | - Dalia Ibrahim El-Wafaey
- Human Anatomy and Embryology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | - Amira Mohamed Abdelhamid
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, Sharkia, Egypt.
| |
Collapse
|