1
|
Zhang Z, Wu G, Yang J, Liu X, Chen Z, Liu D, Huang Y, Yang F, Luo W. Integrated network pharmacology, transcriptomics and metabolomics to explore the material basis and mechanism of Danggui-Baishao herb pair for treating hepatic fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118834. [PMID: 39299362 DOI: 10.1016/j.jep.2024.118834] [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: 06/23/2024] [Revised: 09/13/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Danggui-Baishao herb pair (DB) is commonly used as Chinese herbal formulas for treating hepatic fibrosis (HF). However, there are few research on the combined application of the two drugs in treating HF, and the precise mechanisms and fundamental components of DB in addressing HF are still unclear. AIM OF THE STUDY The intention of this research is to identify the molecular foundation and functional targets of DB to elucidate the mechanisms for treating HF. METHODS The ingredients absorbed from DB in rat plasma were analyzed using UPLC-QE-MS. Therapeutic efficacy of DB in a rat model of CCl4-induced HF assessed using biochemical indices, pathological tissue observations, immunohistochemical and western blotting. An integrated strategy of transcriptomics, metabolomics, and network pharmacology was then utilized to explain the possible material basis and mechanisms of DB for treating HF. Western blotting was carried out to verify the critical mechanism. RESULTS DB reduced the level of liver function and inflammation related indicators in CCl4-induced HF (P < 0.05 or P < 0.01), as well as ameliorated pathological histological changes, and reduced the expressions of collagen type I (Col-I) and α-smooth muscle actin (α-SMA). Nineteen ingredients absorbed from DB were identified. Comprehensive investigations of transcriptomics, metabolomics, and network pharmacology revealed that DB modulated the PI3K/Akt/NF-κB signaling pathway to ameliorate fibrosis induced by CCl4 in HF rats. According to the molecular docking results, core tagets were highly favored by kaempferol, benzoylpaeoniflorin, albiflorin, paeoniflorin, and levistilide A. CONCLUSIONS The possible mechanisms for DB treatment of HF include decreasing the activity of hepatic stellate cells (HSCs), decreasing collagen synthesis and deposition, attenuating the hepatic inflammatory response, inhibiting hepatocyte apoptosis, and increasing the level of niacinamide (NAM), thus exerting its anti-HF effect.
Collapse
Affiliation(s)
- Zhihong Zhang
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730101, China; Gansu Key Laboratory of Pharmacology and Toxicology of Traditional Chinese Medicine, Lanzhou, 730000, China
| | - Guotai Wu
- Gansu Key Laboratory of Pharmacology and Toxicology of Traditional Chinese Medicine, Lanzhou, 730000, China; Long Yao Industry Innovation Research Institute, Lanzhou, 730000, China
| | - Jie Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xuxia Liu
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730101, China
| | - Zhengjun Chen
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730101, China
| | - Dongling Liu
- Long Yao Industry Innovation Research Institute, Lanzhou, 730000, China
| | - Yan Huang
- Department of Pharmacy, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Fude Yang
- School of Pharmacy, Gansu University of Chinese Medicine, Lanzhou, 730101, China.
| | - Wenrong Luo
- Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, 730050, China.
| |
Collapse
|
2
|
Li Q, Zhu K, Huang L, Niu X, Li L, Gao L, Xia Z. Polystyrene microplastics induce liver fibrosis and lipid deposition in mice through three hub genes revealed by the RNA-seq. Sci Rep 2025; 15:2583. [PMID: 39833454 DOI: 10.1038/s41598-025-86810-5] [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: 10/06/2023] [Accepted: 01/14/2025] [Indexed: 01/22/2025] Open
Abstract
Nano- and microplastics (NMPs) have become a serious global environmental threat that causes damage to mammalian organs. In this work, we investigated the potential molecular mechanism underlying the development of liver fibrosis induced by long-term exposure to three different sized polystyrene (PS)-NMPs (80 nm, 0.5 µm and 5 µm) in mice. Liver fibrosis levels were evaluated in mice after chronic exposure to PS-NMPs. Liver inflammation was mainly increased in chronic exposure to 80 nm and 0.5 µm PS-NMPs. Liver lipid deposition was significantly enhanced after PS-NMPs exposure. However, oxidative stress was not changed under PS-NMPs exposure. GO enrichment and KEGG pathway analyses revealed that the DEGs and shared DEGs were mainly enriched in the metabolism of lipids. The mRNA expression levels of genes related to fatty acid oxidation, synthesis and transport were dramatically induced by PS-NMPs exposure. Four hub genes, Acot3, Abcc3, Nr1i3 and Fmo2, were identified by CytoHubba analysis of shared DEGs. The mRNA expression levels of three hub genes, Acot3, Abcc3 and Nr1i3, were significantly augmented under chronic PS-NMPs exposure. Our results suggest that Acot3, Abcc3 and Nr1i3 are potential molecules involved in the development of liver fibrosis under chronic exposure to PS-NMPs.
Collapse
Affiliation(s)
- Qingwen Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Kai Zhu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lizhi Huang
- School of Civil Engineering, Wuhan University, Wuhan, 430072, China
| | - Xuan Niu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lili Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Likun Gao
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, 518020, China.
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
3
|
Sultana M, Islam MA, Khairnar R, Kumar S. A guide to pathophysiology, signaling pathways, and preclinical models of liver fibrosis. Mol Cell Endocrinol 2025; 598:112448. [PMID: 39755140 DOI: 10.1016/j.mce.2024.112448] [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: 10/28/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 01/06/2025]
Abstract
Liver fibrosis is potentially a reversible form of liver disease that evolved from the early stage of liver scarring as a consequence of chronic liver injuries. Recurrent injuries in the liver without any appropriate medication cause the injuries to get intense and deeper, which gradually leads to the progression of irreversible cirrhosis or carcinoma. Unfortunately, there are no approved treatment strategies for reversing hepatic fibrosis, making it one of the significant risk factors for developing advanced liver disorders and liver disease-associated mortality. Consequently, the interpretation of the fundamental mechanisms, etiology, and pathogenesis is crucial for identifying the potential therapeutic target as well as evaluating novel anti-fibrotic therapy. However, despite innumerable research, the functional mechanism and disease characteristics are still obscure. To accelerate the understanding of underlying disease pathophysiology, molecular pathways and disease progression mechanism, it is crucial to mimic human liver disease through the formation of precise disease models. Although various in vitro and in vivo liver fibrotic models have emerged and developed already, a perfect clinical model replicating human liver diseases is yet to be established, which is one of the major challenges in discovering proper therapeutics. This review paper will shed light on pathophysiology, signaling pathways, preclinical models of liver fibrosis, and their limitations.
Collapse
Affiliation(s)
- Mehonaz Sultana
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Md Asrarul Islam
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Rhema Khairnar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Sunil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| |
Collapse
|
4
|
Zhao L, Tang H, Cheng Z. Pharmacotherapy of Liver Fibrosis and Hepatitis: Recent Advances. Pharmaceuticals (Basel) 2024; 17:1724. [PMID: 39770566 PMCID: PMC11677259 DOI: 10.3390/ph17121724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/05/2024] [Accepted: 12/17/2024] [Indexed: 01/03/2025] Open
Abstract
Liver fibrosis is a progressive scarring process primarily caused by chronic inflammation and injury, often closely associated with viral hepatitis, alcoholic liver disease, metabolic dysfunction-associated steatotic liver disease (MASLD), drug-induced liver injury, and autoimmune liver disease (AILD). Currently, there are very few clinical antifibrotic drugs available, and effective targeted therapy is lacking. Recently, emerging antifibrotic drugs and immunomodulators have shown promising results in animal studies, and some have entered clinical research phases. This review aims to systematically review the molecular mechanisms underlying liver fibrosis, focusing on advancements in drug treatments for hepatic fibrosis. Furthermore, since liver fibrosis is a progression or endpoint of many diseases, it is crucial to address the etiological treatment and secondary prevention for liver fibrosis. We will also review the pharmacological treatments available for common hepatitis leading to liver fibrosis.
Collapse
Affiliation(s)
- Liangtao Zhao
- Hepato-Pancreato-Biliary Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China;
| | - Haolan Tang
- School of Medicine, Southeast University, Nanjing 210009, China;
| | - Zhangjun Cheng
- Hepato-Pancreato-Biliary Center, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China;
| |
Collapse
|
5
|
Luo W, Wu H, Yang Z, Lan T, Wu L, Huang Y. Machine Learning and Experimental Validation Identified Ferroptosis Signature and Innovative Biomarkers (ESR1 and GSTZ1) in Liver Fibrosis. J Inflamm Res 2024; 17:10313-10332. [PMID: 39649424 PMCID: PMC11625426 DOI: 10.2147/jir.s490258] [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: 08/06/2024] [Accepted: 11/28/2024] [Indexed: 12/10/2024] Open
Abstract
Background Targeting ferroptosis is an effective approach to mitigate hepatic fibrosis, yet no reports exist on the ferroptosis signature in liver fibrosis. This study aimed to explore ferroptosis characteristics in this disease. Methods RNAseq data from GSE6764, GSE188604 and Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC) were downloaded. Multiple machine learning methods, including Weighted Gene Co-expression Network Analysis (WGCNA), Random Forest (RF) and Support Vector Machine (SVM), were used to identify core genes in liver fibrosis and ferroptosis. WGCNA can pinpoint modules linked to clinical traits, aiding in discovering diagnostic and progression molecules in complex diseases. RF and SVM are often utilized for WGCNA validation to boost result accuracy. Carbon tetrachloride (CCl4) was used to establish a mouse liver fibrosis model to validate core gene expression, which was also assessed in test and validation GEO datasets. Finally, the diagnostic role of the core genes in liver fibrosis and hepatocellular carcinoma (HCC) was also investigated using ROC analysis. Results Multiple machine learning methods screened nine core genes, including IL1B, GSTZ1, LIFR, SLC25A37, PTGS2, MT1G, HSPB1, ESR1, and PHGDH. In vivo experimental validation, RT-PCR showed ESR1 and GSTZ1 were significantly under-expressed in the liver fibrosis group compared to the normal group. Simultaneously, in GSE6764 and GSE188604, ESR1 and GSTZ1 were also identified as protective genes for liver fibrosis. More in-depth research found that ESR1 and GSTZ1 exhibited a good diagnostic performance both in liver fibrosis and HCC, suggesting that a persistent decrease in ESR1 and GSTZ1 in patients might signal the progression from hepatic fibrosis to HCC. Conclusion The present study is the first to report the ferroptosis signature in liver fibrosis and identifies two novel biomarkers, ESR1 and GSTZ1, providing new insights for the diagnosis and treatment of liver fibrosis in the future.
Collapse
Affiliation(s)
- Wen Luo
- Department of Gastrointestinal Surgery, Liuzhou Workers Hospital, Liuzhou, People’s Republic of China
| | - Hongwen Wu
- Department of Pharmacy, Liuzhou Workers Hospital, Liuzhou, People’s Republic of China
| | - Zhijie Yang
- Department of Pharmacy, Liuzhou Workers Hospital, Liuzhou, People’s Republic of China
| | - Tian Lan
- Department of Pharmacy, Liuzhou Workers Hospital, Liuzhou, People’s Republic of China
| | - Liya Wu
- Department of Neurology, Liuzhou Workers Hospital, Liuzhou, People’s Republic of China
| | - Yushen Huang
- Department of Pharmacy, Liuzhou Workers Hospital, Liuzhou, People’s Republic of China
| |
Collapse
|
6
|
Pilling D, Martinez TC, Gomer RH. Inhibition of CCl4-induced liver inflammation and fibrosis by a NEU3 inhibitor. PLoS One 2024; 19:e0308060. [PMID: 39570922 PMCID: PMC11581222 DOI: 10.1371/journal.pone.0308060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 09/26/2024] [Indexed: 11/24/2024] Open
Abstract
Sialic acids are located on the ends of many glycoconjugates and are cleaved off by enzymes called sialidases (neuraminidases). Upregulation of neuraminidase 3 (NEU3) is associated with intestinal inflammation and colitis, neuroinflammation, and lung fibrosis. Genetic ablation of NEU3 or pharmacological inhibition of NEU3 reduces lung fibrosis in mice. To determine if inhibiting NEU3 can inhibit liver fibrosis in the commonly-used CCl4 model, in this report, we examined the effects of injections of the NEU3 inhibitor 2-acetyl pyridine (2AP). 2AP inhibited CCl4-induced weight loss in female but not male mice. 2AP attenuated CCl4-induced liver inflammation and fibrosis in male and female mice, but did not affect CCl4-induced steatosis. After CCl4 treatment, female but not male mice had significant increases in liver neutrophils, and 2AP attenuated this response. 2AP also reversed CCl4-induced liver desialylation and CCl4-induced increased expression of NEU3. Patients with pulmonary fibrosis have increased desialylation of some serum proteins, and elevated serum levels of NEU3. We find that sera from patients with nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) have elevated desialylation of a serum protein and patients with NAFLD have increased levels of NEU3. These data suggest that elevated levels of NEU3 may be associated with liver inflammation and fibrosis, and that in mice this is ameliorated by injections of a NEU3 inhibitor.
Collapse
Affiliation(s)
- Darrell Pilling
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Trevor C. Martinez
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| | - Richard H. Gomer
- Department of Biology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
7
|
Elmorsy EA, Saber S, Hamad RS, Abdel-Reheim MA, Nadwa EH, Alibrahim AOE, Alkhamiss AS, AlSalloom AA, Mohamed EA, Nour-El-Din M, Amer MM, Abdel-Hamed MR, Mohamed NB, Abozaid L, Mostafa-Hedeab G, Ahmed SS, Taha HH, Khalifa AK. Modulating the HSP90 control over NFκB/NLRP3/Caspase-1 axis is a new therapeutic target in the management of liver fibrosis: Insights into the role of TAS-116 (Pimitespib). Life Sci 2024; 354:122966. [PMID: 39147320 DOI: 10.1016/j.lfs.2024.122966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/06/2024] [Accepted: 08/10/2024] [Indexed: 08/17/2024]
Abstract
Aberrant activation of the NLRP3 inflammasome is recognized to induce a chronic inflammatory response in the liver, ultimately leading to hepatic fibrosis. HSP90 is suggested to regulate NLRP3 activation and its downstream signaling. This study is the first to explore the potential therapeutic role of pimitespib in mitigating liver fibrosis in rats. The results of the study revealed that pimitespib effectively suppressed hepatic inflammation and fibrogenesis by modulating HSP90's control over the NFκB/NLRP3/caspase-1 axis. In vitro experiments demonstrated that pimitespib reduced LDH levels and increased hepatocyte survival, whereas in vivo, it prolonged the survival of rats with hepatic fibrosis. Additionally, pimitespib exhibited improvements in the function and microscopic characteristics of rat livers. Pimitespib effectively inhibited NFκB, which serves as the priming signal for NLRP3 activation. Pimitespib's inhibitory effect on NLRP3, identified as an HSP90 client protein, plays a central role in the observed anti-fibrotic effect. The simultaneous inhibition of both priming and activation signals of NLRP3 by pimitespib led to a reduction in caspase-1 activity and subsequent suppression of the N-terminal fragment of gasdermin D, ultimately constraining hepatocyte pyroptotic cell death. These diverse effects were associated with a decrease in the transcription of inflammatory mediators IL-1β, IL-18, and TNF-α, as well as the fibrogenic mediators TGF-β, TIMP-1, PDGF-BB, and Col1a1. Moreover, pimitespib induced the expression of HSP70, which could further contribute to the repression of fibrosis development. In summary, our findings provide an evolutionary perspective on managing liver fibrosis, positioning pimitespib as a promising candidate for anti-inflammatory and antifibrotic therapy.
Collapse
Affiliation(s)
- Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia; Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt.
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 11152, Egypt.
| | - Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia; Central Laboratory, Theodor Bilharz Research Institute, Giza 12411, Egypt.
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Eman Hassan Nadwa
- Department of Pharmacology and Therapeutics, College of Medicine, Jouf University, Sakaka, Saudi Arabia; Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt.
| | - Alaa Oqalaa E Alibrahim
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia.
| | - Abdullah S Alkhamiss
- Department of Pathology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - A A AlSalloom
- Department of Pathology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Enas A Mohamed
- Department of Anatomy, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia; Department of Anatomy, Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - M Nour-El-Din
- Department of Anatomy, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Maha M Amer
- Department of Anatomy, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia; Department of Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Mohamed R Abdel-Hamed
- Department of Anatomy, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia; Department of Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| | - Nahla B Mohamed
- Department of Pathology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Lobaina Abozaid
- Department of Pathology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Gomaa Mostafa-Hedeab
- Department of Pharmacology and Therapeutics, College of Medicine, Jouf University, Sakaka, Saudi Arabia; Pharmacology Department, Faculty of Medicine, Beni-Suef University, Beni Suef, Egypt.
| | - Syed Suhail Ahmed
- Department of Microbiology and Immunology, College of Medicine, Qassim University, Buraidah 51452, Saudi Arabia.
| | - Hagir Hussein Taha
- Department of Basic Medical Sciences, Unaizah College of Medicine and Medical Sciences, Qassim University, Unaizah 51452, Saudi Arabia.
| | - Amira Karam Khalifa
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt; Department of Medical Pharmacology, Faculty of Medicine, Nahda University, New Beni Suef 62521, Egypt.
| |
Collapse
|
8
|
Zhao J, Yue P, Mi N, Li M, Fu W, Zhang X, Gao L, Bai M, Tian L, Jiang N, Lu Y, Ma H, Dong C, Zhang Y, Zhang H, Zhang J, Ren Y, Suzuki A, Wong PF, Tanaka K, Rerknimitr R, Junger HH, Cheung TT, Melloul E, Demartines N, Leung JW, Yao J, Yuan J, Lin Y, Schlitt HJ, Meng W. Biliary fibrosis is an important but neglected pathological feature in hepatobiliary disorders: from molecular mechanisms to clinical implications. MEDICAL REVIEW (2021) 2024; 4:326-365. [PMID: 39135601 PMCID: PMC11317084 DOI: 10.1515/mr-2024-0029] [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: 03/27/2024] [Accepted: 06/06/2024] [Indexed: 08/15/2024]
Abstract
Fibrosis resulting from pathological repair secondary to recurrent or persistent tissue damage often leads to organ failure and mortality. Biliary fibrosis is a crucial but easily neglected pathological feature in hepatobiliary disorders, which may promote the development and progression of benign and malignant biliary diseases through pathological healing mechanisms secondary to biliary tract injuries. Elucidating the etiology and pathogenesis of biliary fibrosis is beneficial to the prevention and treatment of biliary diseases. In this review, we emphasized the importance of biliary fibrosis in cholangiopathies and summarized the clinical manifestations, epidemiology, and aberrant cellular composition involving the biliary ductules, cholangiocytes, immune system, fibroblasts, and the microbiome. We also focused on pivotal signaling pathways and offered insights into ongoing clinical trials and proposing a strategic approach for managing biliary fibrosis-related cholangiopathies. This review will offer a comprehensive perspective on biliary fibrosis and provide an important reference for future mechanism research and innovative therapy to prevent or reverse fibrosis.
Collapse
Affiliation(s)
- Jinyu Zhao
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Ping Yue
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Ningning Mi
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Matu Li
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Wenkang Fu
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xianzhuo Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Long Gao
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Mingzhen Bai
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Liang Tian
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Ningzu Jiang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yawen Lu
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Haidong Ma
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Chunlu Dong
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yong Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Hengwei Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Jinduo Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yanxian Ren
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Azumi Suzuki
- Department of Gastroenterology, Hamamatsu Medical Center, Hamamatsu, Japan
| | - Peng F. Wong
- Department of Vascular Surgery, The James Cook University Hospital, Middlesbrough, UK
| | - Kiyohito Tanaka
- Department of Gastroenterology, Kyoto Second Red Cross Hospital, Kyoto, Japan
| | - Rungsun Rerknimitr
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn, Bangkok, Thailand
- Excellence Center for Gastrointestinal Endoscopy, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Henrik H. Junger
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Tan T. Cheung
- Department of Surgery, The University of Hong Kong, Hong Kong, China
| | - Emmanuel Melloul
- Department of Visceral Surgery, Lausanne University Hospital CHUV, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Nicolas Demartines
- Department of Visceral Surgery, Lausanne University Hospital CHUV, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Joseph W. Leung
- Division of Gastroenterology and Hepatology, UC Davis Medical Center and Sacramento VA Medical Center, Sacramento, CA, USA
| | - Jia Yao
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou, China
| | - Jinqiu Yuan
- Clinical Research Center, Big Data Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Yanyan Lin
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Hans J. Schlitt
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Wenbo Meng
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
9
|
Leaker BD, Wang Y, Tam J, Anderson RR. Analysis of culture and RNA isolation methods for precision-cut liver slices from cirrhotic rats. Sci Rep 2024; 14:15349. [PMID: 38961190 PMCID: PMC11222550 DOI: 10.1038/s41598-024-66235-2] [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: 09/11/2023] [Accepted: 06/28/2024] [Indexed: 07/05/2024] Open
Abstract
Precision-cut liver slices (PCLS) are increasingly used as a model to investigate anti-fibrotic therapies. However, many studies use PCLS from healthy animals treated with pro-fibrotic stimuli in culture, which reflects only the early stages of fibrosis. The effects of different culture conditions on PCLS from cirrhotic animals has not been well characterized and there is no consensus on optimal methods. In this study, we report a method for the collection and culture of cirrhotic PCLS and compare the effect of common culture conditions on viability, function, and gene expression. Additionally, we compared three methods of RNA isolation and identified a protocol with high yield and purity. We observed significantly increased albumin production when cultured with insulin-transferrin-selenium and dexamethasone, and when incubated on a rocking platform. Culturing with insulin-transferrin-selenium and dexamethasone maintained gene expression closer to the levels in fresh slices. However, despite stable viability and function up to 4 days, we found significant changes in expression of key genes by day 2. Interestingly, we also observed that cirrhotic PCLS maintain viability in culture longer than slices from healthy animals. Due to the influence of matrix stiffness on fibrosis and hepatocellular function, it is important to evaluate prospective anti-fibrotic therapies in a platform that preserves tissue biomechanics. PCLS from cirrhotic animals represent a promising tool for the development of treatments for chronic liver disease.
Collapse
Affiliation(s)
- Ben D Leaker
- Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Cambridge, MA, USA.
- Wellman Center for Photomedicine, Massachusetts General Hospital, Thier Research Building, MGH, 55 Blossom Street, Boston, MA, USA.
| | - Yongtao Wang
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Liver Center, Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Joshua Tam
- Wellman Center for Photomedicine, Massachusetts General Hospital, Thier Research Building, MGH, 55 Blossom Street, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - R Rox Anderson
- Wellman Center for Photomedicine, Massachusetts General Hospital, Thier Research Building, MGH, 55 Blossom Street, Boston, MA, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Hu Y, Geng Q, Wang L, Wang Y, Huang C, Fan Z, Kong D. Research progress and application of liver organoids for disease modeling and regenerative therapy. J Mol Med (Berl) 2024; 102:859-874. [PMID: 38802517 PMCID: PMC11213763 DOI: 10.1007/s00109-024-02455-3] [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: 09/18/2023] [Revised: 04/19/2024] [Accepted: 05/08/2024] [Indexed: 05/29/2024]
Abstract
The liver is a major metabolic organ of the human body and has a high incidence of diseases. In recent years, the annual incidence of liver disease has increased, seriously endangering human life and health. The study of the occurrence and development mechanism of liver diseases, discovery of new therapeutic targets, and establishment of new methods of medical treatment are major issues related to the national economy and people's livelihood. The development of stable and effective research models is expected to provide new insights into the pathogenesis of liver diseases and the search for more effective treatment options. Organoid technology is a new in vitro culture system, and organoids constructed by human cells can simulate the morphological structure, gene expression, and glucose and lipid metabolism of organs in vivo, providing a new model for related research on liver diseases. This paper reviews the latest research progress on liver organoids from the establishment of cell sources and application of liver organoids and discusses their application potential in the field of liver disease research.
Collapse
Affiliation(s)
- Yang Hu
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, China
| | - Qiao Geng
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
| | - Lu Wang
- Department of Angioenterology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, 157 Daming Avenue, Nanjing, 210022, Jiangsu, China
| | - Yi Wang
- Department of Angioenterology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, 157 Daming Avenue, Nanjing, 210022, Jiangsu, China
| | - Chuyue Huang
- Department of Angioenterology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, 157 Daming Avenue, Nanjing, 210022, Jiangsu, China
| | - Zhimin Fan
- Department of Angioenterology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, 157 Daming Avenue, Nanjing, 210022, Jiangsu, China.
| | - Desong Kong
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210022, Jiangsu, China.
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China.
| |
Collapse
|
11
|
Parsa S, Dousti M, Mohammadi N, Abedanzadeh M, Dehdari Ebrahimi N, Dara M, Sani M, Nekouee M, Abolmaali SS, Sani F, Azarpira N. The effects of simvastatin-loaded nanoliposomes on human multilineage liver fibrosis microtissue. J Cell Mol Med 2024; 28:e18529. [PMID: 38984945 PMCID: PMC11234647 DOI: 10.1111/jcmm.18529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/09/2024] [Accepted: 06/23/2024] [Indexed: 07/11/2024] Open
Abstract
In this in vitro study, for the first time, we evaluate the effects of simvastatin-loaded liposome nanoparticles (SIM-LipoNPs) treatment on fibrosis-induced liver microtissues, as simvastatin (SIM) has shown potential benefits in the non-alcoholic fatty liver disease process. We developed multicellular liver microtissues composed of hepatic stellate cells, hepatoblastoma cells and human umbilical vein endothelial cells. The microtissues were supplemented with a combination of palmitic acid and oleic acid to develop fibrosis models. Subsequently, various groups of microtissues were exposed to SIM and SIM-LipoNPs at doses of 5 and 10 mg/mL. The effectiveness of the treatments was evaluated by analysing cell viability, production of reactive oxygen species (ROS) and nitric oxide (NO), the expression of Kruppel-like factor (KLF) 2, and pro-inflammatory cytokines (interleukin(IL)-1 α, IL-1 β, IL-6 and tumour necrosis factor-α), and the expression of collagen I. Our results indicated that SIM-LipoNPs application showed promising results. SIM-LipoNPs effectively amplified the SIM-klf2-NO pathway at a lower dosage compatible with a high dosage of free SIM, which also led to reduced oxidative stress by decreasing ROS levels. SIM-LipoNPs administration also resulted in a significant reduction in pro-inflammatory cytokines and Collagen I mRNA levels, as a marker of fibrosis. In conclusion, our study highlights the considerable therapeutic potential of using SIM-LipoNPs to prevent liver fibrosis progress, underscoring the remarkable properties of SIM-LipoNPs in activating the KLF2-NO pathway and anti-oxidative and anti-inflammatory response.
Collapse
Affiliation(s)
- Shima Parsa
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Dousti
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasim Mohammadi
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mozhgan Abedanzadeh
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mahintaj Dara
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Sani
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Muhammad Nekouee
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farnaz Sani
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
12
|
Xiao L, Sunniya H, Li J, Kakar MU, Dai R, Li B. Isolation and purification of polysaccharides from Bupleurum marginatum Wall.ex DC and their anti-liver fibrosis activities. Front Pharmacol 2024; 15:1342638. [PMID: 38576476 PMCID: PMC10991770 DOI: 10.3389/fphar.2024.1342638] [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: 11/22/2023] [Accepted: 01/23/2024] [Indexed: 04/06/2024] Open
Abstract
Bupleurum marginatum Wall.ex DC [Apiaceae] (BM)is widely grown in southwestern China, and the whole plant is used as Traditional Chinese Medicine (TCM). Polysaccharides are main natural products in lots of TCM and have been studied for their effects of reducing oxidative stress, anti-inflammation and immune regulation. Herein, we investigated the extraction techniques of Bupleurum marginatum Wall.ex DC polysaccharides (BMP), the identification of their key components, and their ability to inhibit liver fibrosis in both cellular and animal models. Component identification indicated that monosaccharides in BMP mainly consisted of glucose, galactose, mannose, rhamnose, arabinose, and xylose. In vivo analysis revealed that BMP provided significant protective effects on N-Nitroso dimethylamine (NDMA)-induced liver fibrosis rats through reducing hepatomegaly, reducing tissue inflammation, and reducing collagen deposition. BMP also improved the hepatobiliary system and liver metabolism in accord to reduce the serum levels of ALT, AST, ALP, r-GT, and TBIL. In addition, BMP could reduce the level of inflammation and fibrosis through inhibition of IL-1β and TGF-β1. Cellular studies showed that the BMP could provide therapeutic effects on lipopolysaccharide (LPS)-induced cellular fibrosis model, and could reduce the level of inflammation and fibrosis by decreasing the level of TGF-β1, IL-1β, and TNF-α. Our study demonstrated that BMP may provide a new therapy strategy of liver injury and liver fibrosis.
Collapse
Affiliation(s)
- Li Xiao
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, China
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Hafsa Sunniya
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Jingyi Li
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, China
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Mohib Ullah Kakar
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Rongji Dai
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, China
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Bo Li
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, China
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
13
|
Petrenko O, Königshofer P, Brusilovskaya K, Hofer BS, Bareiner K, Simbrunner B, Jühling F, Baumert TF, Lupberger J, Trauner M, Kauschke SG, Pfisterer L, Simon E, Rendeiro AF, de Rooij LP, Schwabl P, Reiberger T. Transcriptomic signatures of progressive and regressive liver fibrosis and portal hypertension. iScience 2024; 27:109301. [PMID: 38469563 PMCID: PMC10926212 DOI: 10.1016/j.isci.2024.109301] [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: 09/11/2023] [Revised: 12/10/2023] [Accepted: 02/16/2024] [Indexed: 03/13/2024] Open
Abstract
Persistent liver injury triggers a fibrogenic program that causes pathologic remodeling of the hepatic microenvironment (i.e., liver fibrosis) and portal hypertension. The dynamics of gene regulation during liver disease progression and early regression remain understudied. Here, we generated hepatic transcriptome profiles in two well-established liver disease models at peak fibrosis and during spontaneous regression after the removal of the inducing agents. We linked the dynamics of key disease readouts, such as portal pressure, collagen area, and transaminase levels, to differentially expressed genes, enabling the identification of transcriptomic signatures of progressive vs. regressive liver fibrosis and portal hypertension. These candidate biomarkers (e.g., Tcf4, Mmp7, Trem2, Spp1, Scube1, Islr) were validated in RNA sequencing datasets of patients with cirrhosis and portal hypertension, and those cured from hepatitis C infection. Finally, deconvolution identified major cell types and suggested an association of macrophage and portal hepatocyte signatures with portal hypertension and fibrosis area.
Collapse
Affiliation(s)
- Oleksandr Petrenko
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna 1090, Austria
- Christian Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna 1090, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | - Philipp Königshofer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna 1090, Austria
- Christian Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna 1090, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | - Ksenia Brusilovskaya
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna 1090, Austria
- Christian Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna 1090, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | - Benedikt S. Hofer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna 1090, Austria
- Christian Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna 1090, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | - Katharina Bareiner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna 1090, Austria
| | - Benedikt Simbrunner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna 1090, Austria
- Christian Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna 1090, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | - Frank Jühling
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hepatiques UMR_S1110, Strasbourg 67000, France
| | - Thomas F. Baumert
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hepatiques UMR_S1110, Strasbourg 67000, France
- Service d’hépato-gastroentérologie, Hôpitaux Universitaires de Strasbourg, Strasbourg 67000, France
- Institut Universitaire de France (IUF), 75005 Paris, France
| | - Joachim Lupberger
- Université de Strasbourg, Inserm, Institut de Recherche sur les Maladies Virales et Hepatiques UMR_S1110, Strasbourg 67000, France
- Service d’hépato-gastroentérologie, Hôpitaux Universitaires de Strasbourg, Strasbourg 67000, France
- Institut Universitaire de France (IUF), 75005 Paris, France
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna 1090, Austria
| | - Stefan G. Kauschke
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co.KG, 88397 Biberach an der Riss, Germany
| | - Larissa Pfisterer
- Department of CardioMetabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co.KG, 88397 Biberach an der Riss, Germany
| | - Eric Simon
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharma GmbH & Co.KG, 88397 Biberach an der Riss, Germany
| | - André F. Rendeiro
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | - Laura P.M.H. de Rooij
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | - Philipp Schwabl
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna 1090, Austria
- Christian Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna 1090, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna 1090, Austria
- Christian Doppler Laboratory for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna 1090, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria
| |
Collapse
|
14
|
Liu H, Fu Y, Guo D, Li S, Jin Y, Zhang A, Wu C. TMM: A comprehensive CAD system for hepatic fibrosis 5-grade METAVIR staging based on liver MRI. Med Phys 2024; 51:2032-2043. [PMID: 37734071 DOI: 10.1002/mp.16700] [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: 02/22/2023] [Revised: 05/16/2023] [Accepted: 05/26/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Precise staging of hepatic fibrosis with MRI is necessary as it can assist precision medicine. Computer aided diagnosis (CAD) system with distinguishing radiomics features and radiologists domain knowledge is expected to obtain 5-grade meta-analysis of histological data in viral hepatitis (METAVIR) staging. PURPOSE This study aims to obtain the precise staging of hepatic fibrosis based on MRI as it predicts the risk of future liver-related morbidity and the need for treatment, monitoring and surveillance. Based on METAVIR score, fibrosis can be divided into five stages. Most previous researches focus on binary classification, such as cirrhosis versus non-cirrhosis, early versus advanced fibrosis, and substantial fibrosis or not. In this paper, a comprehensive CAD system TMM is proposed to precisely class hepatic fibrosis into five stages for precision medicine instead of the common binary classification. METHODS We propose a novel hepatic fibrosis staging CAD system TMM which includes three modules, Two-level Image Statistical Radiomics Feature (TISRF), Monotonic Error Correcting Output Codes (MECOC) and Monotone Multiclassification with Deep Forest (MMDF). TISRF extracts radiomics features for distinguishing different hepatic fibrosis stages. MECOC is proposed to encode monotonic multiclass by making full use of the progressive severity of hepatic fibrosis and increase the fault tolerance and error correction ability. MMDF combines multiple Deep Forest network to ensure the final five-class classification, which can achieve more precise classification than the common binary classification. The performance of the proposed hepatic fibrosis CAD system is tested on the hepatic data collected from our rabbits models of fibrosis. RESULTS A total of 140 regions of interest (ROI) are selected from MRI T1W of liver fibrosis models in 35 rabbits with F0(n = 16), F1(n = 28), F2(n = 29), F3(n = 44) and F4(n = 23). The performance is evaluated by five-fold cross-validation. TMM can achieve the highest total accuracy of 72.14% for five fibrosis stages compared with other popular classifications. To make a comprehensive comparison, a binary classification experiment have been carried out. CONCLUSIONS T1WI can obtain precise staging of hepatic fibrosis with the help of comprehensive CAD including radiomics features extraction inspired by radiologists, monotonic multiclass according to the severity of hepatic fibrosis, and deep learning classification.
Collapse
Affiliation(s)
- Hui Liu
- School of Biomedical Engineering, Dalian University of Technology & Liaoning Key Laboratory of Integrated Circuit and Biomedical Electronic System, Dalian, China
| | - Yaqing Fu
- School of Biomedical Engineering, Dalian University of Technology & Liaoning Key Laboratory of Integrated Circuit and Biomedical Electronic System, Dalian, China
| | - Dongmei Guo
- Department of Radiology Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Shuo Li
- Department of Radiology Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yilin Jin
- School of Biomedical Engineering, Dalian University of Technology & Liaoning Key Laboratory of Integrated Circuit and Biomedical Electronic System, Dalian, China
| | - Aoran Zhang
- School of Biomedical Engineering, Dalian University of Technology & Liaoning Key Laboratory of Integrated Circuit and Biomedical Electronic System, Dalian, China
| | - Chengjun Wu
- School of Biomedical Engineering, Dalian University of Technology & Liaoning Key Laboratory of Integrated Circuit and Biomedical Electronic System, Dalian, China
| |
Collapse
|
15
|
Liang C, Murray S, Li Y, Lee R, Low A, Sasaki S, Chiang AWT, Lin WJ, Mathews J, Barnes W, Lewis NE. LipidSIM: Inferring mechanistic lipid biosynthesis perturbations from lipidomics with a flexible, low-parameter, Markov modeling framework. Metab Eng 2024; 82:110-122. [PMID: 38311182 PMCID: PMC11163374 DOI: 10.1016/j.ymben.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/03/2024] [Accepted: 01/21/2024] [Indexed: 02/10/2024]
Abstract
Lipid metabolism is a complex and dynamic system involving numerous enzymes at the junction of multiple metabolic pathways. Disruption of these pathways leads to systematic dyslipidemia, a hallmark of many pathological developments, such as nonalcoholic steatohepatitis and diabetes. Recent advances in computational tools can provide insights into the dysregulation of lipid biosynthesis, but limitations remain due to the complexity of lipidomic data, limited knowledge of interactions among involved enzymes, and technical challenges in standardizing across different lipid types. Here, we present a low-parameter, biologically interpretable framework named Lipid Synthesis Investigative Markov model (LipidSIM), which models and predicts the source of perturbations in lipid biosynthesis from lipidomic data. LipidSIM achieves this by accounting for the interdependency between the lipid species via the lipid biosynthesis network and generates testable hypotheses regarding changes in lipid biosynthetic reactions. This feature allows the integration of lipidomics with other omics types, such as transcriptomics, to elucidate the direct driving mechanisms of altered lipidomes due to treatments or disease progression. To demonstrate the value of LipidSIM, we first applied it to hepatic lipidomics following Keap1 knockdown and found that changes in mRNA expression of the lipid pathways were consistent with the LipidSIM-predicted fluxes. Second, we used it to study lipidomic changes following intraperitoneal injection of CCl4 to induce fast NAFLD/NASH development and the progression of fibrosis and hepatic cancer. Finally, to show the power of LipidSIM for classifying samples with dyslipidemia, we used a Dgat2-knockdown study dataset. Thus, we show that as it demands no a priori knowledge of enzyme kinetics, LipidSIM is a valuable and intuitive framework for extracting biological insights from complex lipidomic data.
Collapse
Affiliation(s)
- Chenguang Liang
- Department of Bioengineering, University of California, La Jolla, CA, 92093, USA
| | - Sue Murray
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, 92010, USA
| | - Yang Li
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, 92010, USA
| | - Richard Lee
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, 92010, USA
| | - Audrey Low
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, 92010, USA
| | - Shruti Sasaki
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, 92010, USA
| | - Austin W T Chiang
- Department of Pediatrics, University of California, La Jolla, CA, 92093, USA
| | - Wen-Jen Lin
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404333, Taiwan
| | - Joel Mathews
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, 92010, USA
| | - Will Barnes
- Ionis Pharmaceuticals, Inc., Carlsbad, CA, 92010, USA
| | - Nathan E Lewis
- Department of Bioengineering, University of California, La Jolla, CA, 92093, USA; Department of Pediatrics, University of California, La Jolla, CA, 92093, USA.
| |
Collapse
|
16
|
Manco M, Ammirata G, Petrillo S, De Giorgio F, Fontana S, Riganti C, Provero P, Fagoonee S, Altruda F, Tolosano E. FLVCR1a Controls Cellular Cholesterol Levels through the Regulation of Heme Biosynthesis and Tricarboxylic Acid Cycle Flux in Endothelial Cells. Biomolecules 2024; 14:149. [PMID: 38397386 PMCID: PMC10887198 DOI: 10.3390/biom14020149] [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: 12/21/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Feline leukemia virus C receptor 1a (FLVCR1a), initially identified as a retroviral receptor and localized on the plasma membrane, has emerged as a crucial regulator of heme homeostasis. Functioning as a positive regulator of δ-aminolevulinic acid synthase 1 (ALAS1), the rate-limiting enzyme in the heme biosynthetic pathway, FLVCR1a influences TCA cycle cataplerosis, thus impacting TCA flux and interconnected metabolic pathways. This study reveals an unexplored link between FLVCR1a, heme synthesis, and cholesterol production in endothelial cells. Using cellular models with manipulated FLVCR1a expression and inducible endothelial-specific Flvcr1a-null mice, we demonstrate that FLVCR1a-mediated control of heme synthesis regulates citrate availability for cholesterol synthesis, thereby influencing cellular cholesterol levels. Moreover, alterations in FLVCR1a expression affect membrane cholesterol content and fluidity, supporting a role for FLVCR1a in the intricate regulation of processes crucial for vascular development and endothelial function. Our results underscore FLVCR1a as a positive regulator of heme synthesis, emphasizing its integration with metabolic pathways involved in cellular energy metabolism. Furthermore, this study suggests that the dysregulation of heme metabolism may have implications for modulating lipid metabolism. We discuss these findings in the context of FLVCR1a's potential heme-independent function as a choline importer, introducing additional complexity to the interplay between heme and lipid metabolism.
Collapse
Affiliation(s)
- Marta Manco
- Molecular Biotechnology Center “Guido Tarone”, Via Nizza 52, 10126 Torino, Italy; (M.M.); (G.A.); (S.P.); (F.D.G.); (S.F.); (C.R.); (S.F.); (F.A.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, 3000 Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Giorgia Ammirata
- Molecular Biotechnology Center “Guido Tarone”, Via Nizza 52, 10126 Torino, Italy; (M.M.); (G.A.); (S.P.); (F.D.G.); (S.F.); (C.R.); (S.F.); (F.A.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Sara Petrillo
- Molecular Biotechnology Center “Guido Tarone”, Via Nizza 52, 10126 Torino, Italy; (M.M.); (G.A.); (S.P.); (F.D.G.); (S.F.); (C.R.); (S.F.); (F.A.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Francesco De Giorgio
- Molecular Biotechnology Center “Guido Tarone”, Via Nizza 52, 10126 Torino, Italy; (M.M.); (G.A.); (S.P.); (F.D.G.); (S.F.); (C.R.); (S.F.); (F.A.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Simona Fontana
- Molecular Biotechnology Center “Guido Tarone”, Via Nizza 52, 10126 Torino, Italy; (M.M.); (G.A.); (S.P.); (F.D.G.); (S.F.); (C.R.); (S.F.); (F.A.)
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Chiara Riganti
- Molecular Biotechnology Center “Guido Tarone”, Via Nizza 52, 10126 Torino, Italy; (M.M.); (G.A.); (S.P.); (F.D.G.); (S.F.); (C.R.); (S.F.); (F.A.)
- Department of Oncology, University of Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Paolo Provero
- Department of Neurosciences “Rita Levi Montalcini”, University of Torino, Corso Massimo D’Azeglio 52, 10126 Torino, Italy;
- Center for Omics Sciences, Ospedale San Raffaele IRCCS, Via Olgettina 60, 20132 Milan, Italy
| | - Sharmila Fagoonee
- Molecular Biotechnology Center “Guido Tarone”, Via Nizza 52, 10126 Torino, Italy; (M.M.); (G.A.); (S.P.); (F.D.G.); (S.F.); (C.R.); (S.F.); (F.A.)
- Institute of Biostructure and Bioimaging, CNR c/o Molecular Biotechnology Center “Guido Tarone”, 10126 Torino, Italy
| | - Fiorella Altruda
- Molecular Biotechnology Center “Guido Tarone”, Via Nizza 52, 10126 Torino, Italy; (M.M.); (G.A.); (S.P.); (F.D.G.); (S.F.); (C.R.); (S.F.); (F.A.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Emanuela Tolosano
- Molecular Biotechnology Center “Guido Tarone”, Via Nizza 52, 10126 Torino, Italy; (M.M.); (G.A.); (S.P.); (F.D.G.); (S.F.); (C.R.); (S.F.); (F.A.)
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| |
Collapse
|
17
|
Mao J, Tan L, Tian C, Wang W, Zhang H, Zhu Z, Li Y. Research progress on rodent models and its mechanisms of liver injury. Life Sci 2024; 337:122343. [PMID: 38104860 DOI: 10.1016/j.lfs.2023.122343] [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: 09/21/2023] [Revised: 11/22/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
The liver is the most important organ for biological transformation in the body and is crucial for maintaining the body's vital activities. Liver injury is a serious pathological condition that is commonly found in many liver diseases. It has a high incidence rate, is difficult to cure, and is prone to recurrence. Liver injury can cause serious harm to the body, ranging from mild to severe fatty liver disease. If the condition continues to worsen, it can lead to liver fibrosis and cirrhosis, ultimately resulting in liver failure or liver cancer, which can seriously endanger human life and health. Therefore, establishing an rodent model that mimics the pathogenesis and severity of clinical liver injury is of great significance for better understanding the pathogenesis of liver injury patients and developing more effective clinical treatment methods. The author of this article summarizes common chemical liver injury models, immune liver injury models, alcoholic liver injury models, drug-induced liver injury models, and systematically elaborates on the modeling methods, mechanisms of action, pathways of action, and advantages or disadvantages of each type of model. The aim of this study is to establish reliable rodent models for researchers to use in exploring anti-liver injury and hepatoprotective drugs. By creating more accurate theoretical frameworks, we hope to provide new insights into the treatment of clinical liver injury diseases.
Collapse
Affiliation(s)
- Jingxin Mao
- Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Lihong Tan
- Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing 400030, China
| | - Cheng Tian
- Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing 400030, China
| | - Wenxiang Wang
- Chongqing Three Gorges Medical College, Chongqing 404120, China
| | - Hao Zhang
- Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing 400030, China
| | - Zhaojing Zhu
- Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing 400030, China
| | - Yan Li
- Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing 400030, China.
| |
Collapse
|
18
|
Ma L, Yin L, Zhu H, Li J, Wang D. Two-dimensional vascularized liver organoid on extracellular matrix with defined stiffness for modeling fibrotic and normal tissues. J Tissue Eng 2024; 15:20417314241268344. [PMID: 39130682 PMCID: PMC11316963 DOI: 10.1177/20417314241268344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/18/2024] [Indexed: 08/13/2024] Open
Abstract
Antifibrotic drug screening requires evaluating the inhibitory effects of drug candidates on fibrotic cells while minimizing any adverse effects on normal cells. It is challenging to create organ-specific vascularized organoids that accurately model fibrotic and normal tissues for drug screening. Our previous studies have established methods for culturing primary microvessels and epithelial cells from adult tissues. In this proof-of-concept study, we used rats as a model organism to create a two-dimensional vascularized liver organoid model that comprised primary microvessels, epithelia, and stellate cells from adult livers. To provide appropriate substrates for cell culture, we engineered ECMs with defined stiffness to mimic the different stages of fibrotic tissues and normal tissues. We examined the effects of two TGFβ signaling inhibitors, A83-01 and pirfenidone, on the vascularized liver organoids on the stiff and soft ECMs. We found that A83-01 inhibited fibrotic markers while promoting epithelial genes of hepatocytes and cholangiocytes. However, it inhibited microvascular genes on soft ECM, indicating a detrimental effect on normal tissues. Furthermore, A83-01 significantly promoted the expression of markers of stem cells and cancers, increasing the potential risk of it being a carcinogen. In contrast, pirfenidone, an FDA-approved compound for antifibrotic treatments, did not significantly affect all the genes examined on soft ECM. Although pirfenidone had minor effects on most genes, it did reduce the expression of collagens, the major components of fibrotic tissues. These results explain why pirfenidone can slow fibrosis progression with minor side effects in clinical trials. In conclusion, our study presents a method for creating vascularized liver organoids that can accurately mimic fibrotic and normal tissues for drug screening. Our findings provide valuable insights into the potential risks and benefits of using A83-01 and pirfenidone as antifibrotic drugs. This method can be applied to other organs to create organ-specific vascularized organoids for drug development.
Collapse
Affiliation(s)
- Lei Ma
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, China
| | - Lin Yin
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, China
| | - Hai Zhu
- Department of Urology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jing Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, China
| | - Dong Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
19
|
Li K, Ren K, Du S, Gao X, Yu J. Development of Liver Cancer Organoids: Reproducing Tumor Microenvironment and Advancing Research for Liver Cancer Treatment. Technol Cancer Res Treat 2024; 23:15330338241285097. [PMID: 39363866 PMCID: PMC11456184 DOI: 10.1177/15330338241285097] [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: 04/26/2024] [Revised: 07/26/2024] [Accepted: 08/30/2024] [Indexed: 10/05/2024] Open
Abstract
Liver cancer a leading cause of cancer-related deaths worldwide, yet understanding of its development mechanism remains limited, and treatment barriers present substantial challenges. Owing to the heterogeneity of tumors, traditional 2D culture models are inadequate for capturing the complexity and diversity of tumor biology and understanding of the disease. Organoids have garnered considerable attention because of their ability to self-renew and develop functional structures in vitro that closely resemble those of human organs. This review explores the history of liver organoids, their cellular origins, techniques of constructing tumor microenvironments that recapitulate liver cancer organoids, and the biological and clinical applications of liver and liver cancer organoids and explores the current challenges related to liver cancer organoid applications and potentially valuable solutions, with the aim of facilitating the construction of in vitro clinical models of liver cancer therapeutic research.
Collapse
Affiliation(s)
- Kangkang Li
- Department of Hepato-Biliary-Pancreatic Surgery, Fuyang Hospital Affiliataed Bengbu Medical College, Fuyang, Anhui province, China, 236000
| | - Kuiwu Ren
- Department of Hepato-Biliary-Pancreatic Surgery, Fuyang Hospital Affiliataed Bengbu Medical College, Fuyang, Anhui province, China, 236000
| | - Sen Du
- Department of Hepato-Biliary-Pancreatic Surgery, Fuyang Hospital Affiliataed Bengbu Medical College, Fuyang, Anhui province, China, 236000
| | - Xiang Gao
- Department of Hepato-Biliary-Pancreatic Surgery, Fuyang People's Hospital of Anhui Medical University, Fuyang, Anhui province, China, 236000
| | - Jiangtao Yu
- Department of Hepato-Biliary-Pancreatic Surgery, Fuyang Hospital Affiliataed Bengbu Medical College, Fuyang, Anhui province, China, 236000
- Department of Hepato-Biliary-Pancreatic Surgery, Fuyang People's Hospital of Anhui Medical University, Fuyang, Anhui province, China, 236000
| |
Collapse
|
20
|
Qiu L, Ma Z, Sun J, Wu Z, Wang M, Wang S, Zhao Y, Liang S, Hu M, Li Y. Establishment of a Spontaneous Liver Fibrosis Model in NOD/SCID Mice Induced by Natural Aging. BIOLOGY 2023; 12:1493. [PMID: 38132319 PMCID: PMC10740877 DOI: 10.3390/biology12121493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Liver fibrosis, a critical pathological feature of chronic liver diseases, arises from a multitude of pathogenic factors. Consequently, establishing an appropriate animal model to simulate liver fibrosis holds immense significance for comprehending its underlying pathogenesis. Despite the numerous methodologies available for generating liver fibrosis models, they often deviate substantially from the spontaneous age-related liver fibrosis process. In this study, compared with young (12 weeks) and middle-aged NOD/SCID mice (32 weeks), there were a large number of fibrous septum and collagen in the liver tissue of old NOD/SCID mice (43 weeks, 43 W). Immunohistochemical analysis unequivocally indicated heightened α-SMA content within the liver tissue of the 43 W mice, thereby underscoring aging's role in triggering the epithelial-to-mesenchymal transition. In addition, SA-β-gal staining as well as P21 expression were increased, and SIRT1 and SIRT3 expression were decreased in 43 W mice. A comprehensive evaluation encompassing transmission electron microscopy and fluorescence quantitative analysis elucidated compromised mitochondrial function and reduced antioxidant capacity in hepatocytes of the 43 W mice. Furthermore, the aging process activated the pro-fibrotic TGF-β-SMAD pathway, concurrently inducing hepatocellular inflammation. The results of the present study not only validate the successful construction of a spontaneous liver fibrosis mouse model through natural aging induction but also provide initial insights into the mechanisms underpinning age-induced liver fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Min Hu
- Yunnan Key Laboratory for Basic Research on Bone and Joint Diseases & Yunnan Stem Cell Translational Research Center, Kunming University, Kunming 650214, China; (L.Q.); (Z.M.); (J.S.); (Z.W.); (M.W.); (S.W.); (Y.Z.); (S.L.)
| | - Yanjiao Li
- Yunnan Key Laboratory for Basic Research on Bone and Joint Diseases & Yunnan Stem Cell Translational Research Center, Kunming University, Kunming 650214, China; (L.Q.); (Z.M.); (J.S.); (Z.W.); (M.W.); (S.W.); (Y.Z.); (S.L.)
| |
Collapse
|
21
|
Azizsoltani A, Hatami B, Zali MR, Mahdavi V, Baghaei K, Alizadeh E. Obeticholic acid-loaded exosomes attenuate liver fibrosis through dual targeting of the FXR signaling pathway and ECM remodeling. Biomed Pharmacother 2023; 168:115777. [PMID: 37913732 DOI: 10.1016/j.biopha.2023.115777] [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: 07/23/2023] [Revised: 10/12/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023] Open
Abstract
End-stage of liver fibrosis as a precancerous state could lead to cirrhosis and hepatocellular carcinoma which liver transplantation is the only effective treatment. Previous studies have indicated that farnesoid X receptor (FXR) agonists, such as obeticholic acid (OCA) protect against hepatic injuries. However, free OCA administration results in side effects in clinical trials that could be alleviated by applying bio carriers such as MSC-derived exosomes (Exo) with the potential to mimic the biological regenerative effect of their parent cells, as proposed in this study. Loading OCA into the Exo was conducted via water bath sonication. Ex vivo bio distribution studies validated the Exo-loaded OCA more permanently accumulated in the liver. Using CCL4-induced liver fibrosis, we proposed whether Exo isolated from human Warton's Jelly mesenchymal stem cells loaded with a minimal dosage of OCA can facilitate liver recovery. Notably, Exo-loaded OCA exerted additive anti-fibrotic efficacy on histopathological features in CCL4-induced fibrotic mice. Compared to baseline, Exo-mediated delivery OCA results in marked improvements in the fibrotic-related indicators as well as serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) concentrations. Accordingly, the synergistic impact of Exo-loaded OCA as a promising approach is associated with the inactivation of hepatic stellate cells (HSCs), extracellular matrix (ECM) remodeling, and Fxr-Cyp7a1 cascade on CCL4-induced liver fibrosis mice. In conclusion, our data confirmed the additive protective effects of Exo-loaded OCA in fibrotic mice, which suggests a valuable therapeutic strategy to combat liver fibrosis. Furthermore, the use of Exo for accurate drug delivery to the liver tissue can be inspiring.
Collapse
Affiliation(s)
- Arezou Azizsoltani
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behzad Hatami
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahideh Mahdavi
- Iranian Research Institute of Plant Protection (IRIPP), Agricultural Research, Education and Extension Organization (AREEO), Tehran, Iran
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Effat Alizadeh
- Drug Applied Research Center and Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
22
|
Dong Y, Wang X, Xu L, Li X, Dai H, Mao X, Chu Y, Yuan X, Liu H. Development of a Chimeric Protein BiPPB-mIFNγ-tTβRII for Improving the Anti-Fibrotic Activity in Vivo by Targeting Fibrotic Liver and Dual Inhibiting the TGF-β1/Smad Signaling Pathway. Protein J 2023; 42:753-765. [PMID: 37690089 DOI: 10.1007/s10930-023-10147-z] [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] [Accepted: 08/02/2023] [Indexed: 09/12/2023]
Abstract
Excessive production of transforming growth factor β1 (TGF-β1) in activated hepatic stellate cells (aHSCs) promotes liver fibrosis by activating the TGF-β1/Smad signaling pathway. Thus, specifically inhibiting the pro-fibrotic activity of TGF-β1 in aHSCs is an ideal strategy for treating liver fibrosis. Overexpression of platelet-derived growth factor β receptor (PDGFβR) has been demonstrated on the surface of aHSCs relative to normal cells in liver fibrosis. Interferon-gamma peptidomimetic (mIFNγ) and truncated TGF-β receptor type II (tTβRII) inhibit the TGF-β1/Smad signaling pathway by different mechanisms. In this study, we designed a chimeric protein by the conjugation of (1) mIFNγ and tTβRII coupled via plasma protease-cleavable linker sequences (FNPKTP) to (2) PDGFβR-recognizing peptide (BiPPB), namely BiPPB-mIFNγ-tTβRII. This novel protein BiPPB-mIFNγ-tTβRII was effectively prepared using Escherichia coli expression system. The active components BiPPB-mIFNγ and tTβRII were slowly released from BiPPB-mIFNγ-tTβRII by hydrolysis using the plasma protease thrombin in vitro. Moreover, BiPPB-mIFNγ-tTβRII highly targeted to fibrotic liver tissues, markedly ameliorated liver morphology and fibrotic responses in chronic liver fibrosis mice by both inhibiting the phosphorylation of Smad2/3 and inducing the expression of Smad7. Meanwhile, BiPPB-mIFNγ-tTβRII markedly reduced the deposition of collagen fibrils and expression of fibrosis-related proteins in acute liver fibrosis mice. Furthermore, BiPPB-mIFNγ-tTβRII showed a good safety performance in both liver fibrosis mice. Taken together, BiPPB-mIFNγ-tTβRII improved the in vivo anti-liver fibrotic activity due to its high fibrotic liver-targeting potential and the dual inhibition of the TGF-β1/Smad signaling pathway, which may be a potential candidate for targeting therapy on liver fibrosis.
Collapse
Affiliation(s)
- Yixin Dong
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, 157011, Mudanjiang, PR China
| | - Xiaohua Wang
- Laboratory of Pathogenic Microbiology and Immunology, Mudanjiang Medical University, 157011, Mudanjiang, PR China
| | - Liming Xu
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, 157011, Mudanjiang, PR China
| | - Xin Li
- Department of Pediatrics, Hongqi Hospital Affiliated to Mudanjiang Medical University, 157011, Mudanjiang, PR China
| | - Haibing Dai
- Department of Biology, Mudanjiang Medical University, 157011, Mudanjiang, PR China
| | - Xu Mao
- Department of Pharmacology, Mudanjiang Medical University, 157011, Mudanjiang, PR China
| | - Yanhui Chu
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, 157011, Mudanjiang, PR China
| | - Xiaohuan Yuan
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, 157011, Mudanjiang, PR China.
| | - Haifeng Liu
- Heilongjiang Province Key Laboratory for Anti-fibrosis Biotherapy, Mudanjiang Medical University, 157011, Mudanjiang, PR China.
- Laboratory of Pathogenic Microbiology and Immunology, Mudanjiang Medical University, 157011, Mudanjiang, PR China.
| |
Collapse
|
23
|
Septiana WL, Ayudyasari W, Gunardi H, Pawitan JA, Balachander GM, Yu H, Antarianto RD. Liver organoids cocultured on decellularized native liver scaffolds as a bridging therapy improves survival from liver failure in rabbits. In Vitro Cell Dev Biol Anim 2023; 59:747-763. [PMID: 38110841 DOI: 10.1007/s11626-023-00817-8] [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: 07/18/2023] [Accepted: 09/28/2023] [Indexed: 12/20/2023]
Abstract
The present study aimed to develop viable liver organoids using decellularized native liver scaffolds and evaluate the efficacy of human liver organoid transplantation in a rabbit model of cirrhosis. Liver organoids were formed by coculture of hepatocyte-like cells derived from the human-induced pluripotent stem cells with three other cell types. Twelve 3-mo-old New Zealand White Rabbits underwent a sham operation, bile duct ligation, or biliary duct ligation followed by liver organoid transplantation. Liver organoid structure and function before and after transplantation were evaluated using histological and molecular analyses. A survival analysis using the Kaplan-Meier method was performed to determine the cumulative probability of survival according to liver organoid transplantation with significantly greater overall survival observed in rabbits that underwent liver organoid transplantation (P = 0.003, log-rank test). The short-term group had higher hepatic expression levels of ALB and CYP3A mRNA and lower expression levels of AST mRNA compared to the long-term group. The short-term group also had lower collagen deposition in liver tissues. Transplantation of human liver organoids cocultured in decellularized native liver scaffold into rabbits that had undergone bile duct ligation improved short-term survival and hepatic function. The results of the present study highlight the potential of liver organoid transplantation as a bridging therapy in liver failure; however, rejection and poor liver organoid function may limit the long-term efficacy of this therapeutic approach.
Collapse
Affiliation(s)
- Wahyunia Likhayati Septiana
- Program Doktor Ilmu Biomedik, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Department of Histology, Faculty of Medicine, Universitas Gunadarma, Depok, Indonesia
| | - Wulan Ayudyasari
- Department of Surgery, Fakultas Kedokteran Universitas Indonesia, Jakarta, Indonesia
| | - Hardian Gunardi
- Department of Surgery, Fakultas Kedokteran Universitas Indonesia, Jakarta, Indonesia
| | - Jeanne Adiwinata Pawitan
- Department of Histology, Fakultas Kedokteran Universitas Indonesia, Jl Salemba Raya No 6. Jakarta Pusat 10430, Jakarta, Indonesia
- Stem Cell and Tissue Engineering Research Cluster, (IMERI) Indonesian Medical Education and Research Institute, Jakarta, Indonesia
- Integrated Service Unit of Stem Cell Medical Technology (IPT TK Sel Punca), Dr. Cipto Mangunkusumo General Hospital (RSCM), Jakarta, Indonesia
| | - Gowri Manohari Balachander
- Department of Physiology, The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, MD9-04-11, 2 Medical Drive, Singapore, 117593, Singapore
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, India, 221005
| | - Hanry Yu
- Department of Physiology, The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, MD9-04-11, 2 Medical Drive, Singapore, 117593, Singapore
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, India, 221005
- Institute of Bioengineering & Bioimaging, A*STAR, 31 Biopolis Way, #07-01, Singapore, 138669, Singapore
- CAMP, Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, Level 4 Enterprise Wing, Singapore, 138602, Singapore
- Mechanobiology Institute, National University of Singapore, T-Lab, #05-01, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Radiana Dhewayani Antarianto
- Department of Histology, Fakultas Kedokteran Universitas Indonesia, Jl Salemba Raya No 6. Jakarta Pusat 10430, Jakarta, Indonesia.
- Stem Cell and Tissue Engineering Research Cluster, (IMERI) Indonesian Medical Education and Research Institute, Jakarta, Indonesia.
| |
Collapse
|
24
|
Abdalla N, Abo-ElMatty DM, Saleh S, Ghattas MH, Omar NN. Empagliflozin suppresses hedgehog pathway, alleviates ER stress, and ameliorates hepatic fibrosis in rats. Sci Rep 2023; 13:19046. [PMID: 37923828 PMCID: PMC10624673 DOI: 10.1038/s41598-023-46288-5] [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: 07/27/2023] [Accepted: 10/30/2023] [Indexed: 11/06/2023] Open
Abstract
Worldwide mortality from hepatic fibrosis remains high, due to hepatocellular carcinoma and end stage liver failure. The progressive nature of hepatic fibrosis from inflammation to cicatrized tissues warrants subtle intervention with pharmacological agents that hold potential. Empagliflozin (Empa), a novel hypoglycemic drug with antioxidant and anti-inflammatory properties, has lately been proposed to have additional antifibrotic activities. In the current study, we examined the antifibrotic effect of the Empa through modulating the activity of hepatic stellate cells by hedgehog (Hh) pathway. We also assessed the markers of inflammatory response and endoplasmic reticulum (ER) stress. Male Albino rats were treated with either CCl4 (0.4 mg/kg twice/week) and/or Empa (10 mg/kg/day) for eight weeks. In this study, CCl4 rats had active Hh signaling as indicated by overexpression of Patched 1, Smoothened and Glioblastoma-2. CCl4 induced ER stress as CHOP expression was upregulated and ERAD was downregulated. CCl4-induced inflammatory response was demonstrated through increased levels of TNF-α, IL-6 and mRNA levels of IL-17 while undetectable expression of IL-10. Conversely, Empa elicited immunosuppression, suppressed the expression of Hh markers, and reversed markers of ER stress. In conclusion, Empa suppressed CCl4-induced Hh signaling and proinflammatory response, meanwhile embraced ER stress in the hepatic tissues, altogether provided hepatoprotection.
Collapse
Affiliation(s)
- Nourihan Abdalla
- Department of Biochemistry, Faculty of Pharmacy, Modern University for Technology and Information, Mokattam, Cairo, 11585, Egypt
| | - Dina M Abo-ElMatty
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Sami Saleh
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Maivel H Ghattas
- Department of Medical Biochemistry, Faculty of Medicine, Port Said University, Port Said, Egypt.
| | - Nesreen Nabil Omar
- Department of Biochemistry, Faculty of Pharmacy, Modern University for Technology and Information, Mokattam, Cairo, 11585, Egypt
| |
Collapse
|
25
|
Kandhi R, Yeganeh M, Yoshimura A, Menendez A, Ramanathan S, Ilangumaran S. Hepatic stellate cell-intrinsic role of SOCS1 in controlling hepatic fibrogenic response and the pro-inflammatory macrophage compartment during liver fibrosis. Front Immunol 2023; 14:1259246. [PMID: 37860002 PMCID: PMC10582746 DOI: 10.3389/fimmu.2023.1259246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Introduction Hepatic stellate cells (HSC) become activated, differentiate to myofibroblasts and produce extracellular fibrillar matrix during liver fibrosis. The hepatic fibrogenic response is orchestrated by reciprocal interactions between HSCs and macrophages and their secreted products. SOCS1 can regulate several cytokines and growth factors implicated in liver fibrosis. Here we investigated the role of SOCS1 in regulating HSC activation. Methods Mice lacking SOCS1 in HSCs (Socs1ΔHSC) were generated by crossing Socs1fl/fl and LratCre mice. Liver fibrosis was induced by carbon tetrachloride and evaluated by Sirius red staining, hydroxyproline content and immunostaining of myofibroblasts. Gene expression of pro-fibrogenic factors, cytokines, growth factors and chemokines were quantified by RT-qPCR. The phenotype and the numbers of intrahepatic leukocyte subsets were studied by flow cytometry. The impact of fibrosis on the development of diethyl nitrosamine-induced hepatocellular carcinoma was evaluated. Results Socs1ΔHSC mice developed more severe liver fibrosis than control Socs1fl/fl mice that was characterized by increased collagen deposition and myofibroblast differentiation. Socs1ΔHSC mice showed a significant increase in the expression of smooth muscle actin, collagens, matrix metalloproteases, cytokines, growth factors and chemokines in the liver following fibrosis induction. The fibrotic livers of Socs1ΔHSC mice displayed heightened inflammatory cell infiltration with increased proportion and numbers of Ly6ChiCCR2+ pro-inflammatory macrophages. This macrophage population contained elevated numbers of CCR2+CX3CR1+ cells, suggesting impaired transition towards restorative macrophages. Fibrosis induction following exposure to diethyl nitrosamine resulted in more numerous and larger liver tumor nodules in Socs1ΔHSC mice than in Socs1fl/fl mice. Discussion Our findings indicate that (i) SOCS1 expression in HSCs is a critical to control liver fibrosis and development of hepatocaellular carcinoma, and (ii) attenuation of HSC activation by SOCS1 regulates pro-inflammatory macrophage recruitment and differentiation during liver fibrosis.
Collapse
Affiliation(s)
- Rajani Kandhi
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mehdi Yeganeh
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Alfredo Menendez
- Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Sheela Ramanathan
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Subburaj Ilangumaran
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
26
|
Ma L, Wu Q, Tam PKH. The Current Proceedings of PSC-Based Liver Fibrosis Therapy. Stem Cell Rev Rep 2023; 19:2155-2165. [PMID: 37490204 DOI: 10.1007/s12015-023-10592-4] [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] [Accepted: 07/12/2023] [Indexed: 07/26/2023]
Abstract
Liver fibrosis was initially considered to be an irreversible process which will eventually lead to the occurrence of liver cancer. So far there has been no effective therapeutic approach to treat liver fibrosis although scientists have put tremendous efforts into the underlying mechanisms of this disease. Therefore, in-depth research on novel and safe treatments of liver fibrosis is of great significance to human health. Pluripotent stem cells (PSCs) play important roles in the study of liver fibrosis due to their unique features in self-renewal ability, pluripotency, and paracrine function. This article mainly reviews the applications of PSCs in the study of liver fibrosis in recent years. We discuss the role of PSC-derived liver organoids in the study of liver fibrosis, and the latest research advances on the differentiation of PSCs into hepatocytes or macrophages. We also highlight the importance of exosomes of PSCs for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Li Ma
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Qiang Wu
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China.
| | - Paul Kwong-Hang Tam
- Faculty of Medicine, Macau University of Science and Technology, Taipa, China.
| |
Collapse
|
27
|
Abubakr S, Hazem NM, Sherif RN, Elhawary AA, Botros KG. Correlation between SDF-1α, CD34 positive hematopoietic stem cells and CXCR4 expression with liver fibrosis in CCl4 rat model. BMC Gastroenterol 2023; 23:323. [PMID: 37730560 PMCID: PMC10512633 DOI: 10.1186/s12876-023-02932-y] [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: 04/22/2023] [Accepted: 08/25/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND One of the most frequent disorders is liver fibrosis. An improved understanding of the different events during the process of liver fibrosis & its reversibility could be helpful in its staging and in finding potential therapeutic agents. AIM The goal of this research was to evaluate the relationship among CD34 + HPSCs, SDF-1α, and CXCR4 receptor expression with the percentage of the area of hepatic fibrosis. MATERIALS AND METHODS Thirty-six male Sprague-Dawley rats were separated into the control group, liver injury group & spontaneous reversion group. The liver injury was induced by using 2 ml/kg CCl4 twice a week. Flow cytometric examination of CD34 + cells in the blood & liver was performed. Bone marrow & liver samples were taken for evaluation of the SDF-1α mRNA by PCR. Liver specimens were stained for histopathological and CXCR4 immuno-expression evaluation. RESULTS In the liver injury group, the hepatic enzymes, fibrosis area percentage, CXCR4 receptor expression in the liver, CD34 + cells in the blood and bone marrow & the level SDF-1α in the liver and its concentration gradient were statistically significantly elevated with the progression of the liver fibrosis. On the contrary, SDF-1α in the bone marrow was statistically significantly reduced with the development of liver fibrosis. During the spontaneous reversion group, all the studied parameters apart from SDF-1α in the bone marrow were statistically substantially decreased compared with the liver injury group. We found a statistically substantial positive correlation between fibrosis area and all of the following: liver enzymes, CXCR4 receptor expression in the liver, CD34 + cells in the blood and liver, and SDF- 1α in the liver and its concentration gradient. In conclusion, in CCl4 rat model, the fibrosis area is significantly correlated with many parameters in the blood, bone marrow, and liver, which can be used during the process of follow-up during the therapeutic interventions.
Collapse
Affiliation(s)
- Sara Abubakr
- Human Anatomy & Embryology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Noha M Hazem
- Medical Biochemistry and Molecular Biology Department, Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Algomhoria Street, Mansoura, 35516, Egypt.
- Pathological Sciences Department, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia.
| | - R N Sherif
- Human Anatomy & Embryology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Adel Abdelmohdy Elhawary
- Human Anatomy & Embryology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Kamal G Botros
- Human Anatomy & Embryology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
28
|
Bala S, Zhuang Y, Nagesh PT, Catalano D, Zivny A, Wang Y, Xie J, Gao G, Szabo G. Therapeutic inhibition of miR-155 attenuates liver fibrosis via STAT3 signaling. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:413-427. [PMID: 37547286 PMCID: PMC10403732 DOI: 10.1016/j.omtn.2023.07.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 07/11/2023] [Indexed: 08/08/2023]
Abstract
Most chronic liver diseases progress to liver fibrosis, which, when left untreated, can lead to cirrhosis and hepatocellular carcinoma. MicroRNA (miRNA)-targeted therapeutics have become attractive approaches to treat diseases. In this study, we investigated the therapeutic effect of miR-155 inhibition in the bile duct ligation (BDL) mouse model of liver fibrosis and evaluated the role of miR-155 in chronic liver fibrosis using miR-155-deficient (miR-155 knockout [KO]) mice. We found increased hepatic miR-155 expression in patients with cirrhosis and in the BDL- and CCl4-induced mouse models of liver fibrosis. Liver fibrosis was significantly reduced in miR-155 KO mice after CCl4 administration or BDL. To assess the therapeutic potential of miR-155 inhibition, we administered an rAAV8-anti-miR-155 tough decoy in vivo that significantly reduced liver damage and fibrosis in BDL. BDL-induced protein levels of transforming growth factor β (TGF-β), p-SMAD2/3, and p-STAT3 were attenuated in anti-miR-155-treated compared with control mice. Hepatic stellate cells from miR-155 KO mice showed attenuation in activation and mesenchymal marker expression. In vitro, miR-155 gain- and loss-of-function studies revealed that miR-155 regulates activation of stellate cells partly via STAT3 signaling. Our study suggests that miR-155 is the key regulator of liver fibrosis and might be a potential therapeutic target to attenuate fibrosis progression.
Collapse
Affiliation(s)
- Shashi Bala
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Yuan Zhuang
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | | | - Donna Catalano
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Adam Zivny
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Yanbo Wang
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Jun Xie
- Horae Gene Therapy Center, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Gyongyi Szabo
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| |
Collapse
|
29
|
Nishi K, Yagi H, Ohtomo M, Nagata S, Udagawa D, Tsuchida T, Morisaku T, Kitagawa Y. A thioacetamide-induced liver fibrosis model for pre-clinical studies in microminipig. Sci Rep 2023; 13:14996. [PMID: 37696857 PMCID: PMC10495379 DOI: 10.1038/s41598-023-42144-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/06/2023] [Indexed: 09/13/2023] Open
Abstract
Drug-induced liver fibrosis models are used in normal and immunosuppressed small animals for transplantation and regenerative medicine to improve liver fibrosis. Although large animal models are needed for pre-clinical studies, they are yet to be established owing to drug sensitivity in animal species and difficulty in setting doses. In this study, we evaluated liver fibrosis by administering thioacetamide (TA) to normal microminipig and thymectomized microminipig; 3 times for 1 week (total duration: 8 weeks). The pigs treated with TA showed elevated blood cytokine levels and a continuous liver injury at 8 weeks. RNA-seq of the liver showed increased expression of fibrosis-related genes after TA treatment. Histopathological examination showed degenerative necrosis of hepatocytes around the central vein, and revealed fibrogenesis and hepatocyte proliferation. TA treatment caused CD3-positive T cells and macrophages scattered within the hepatic lobule to congregate near the center of the lobule and increased αSMA-positive cells. Thymectomized pigs showed liver fibrosis similar to that of normal pigs, although the clinical signs tended to be milder. This model is similar to pathogenesis of liver fibrosis reported in other animal models. Therefore, it is expected to contribute to research as a drug discovery and pre-clinical transplantation models.
Collapse
Affiliation(s)
- Kotaro Nishi
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan
| | - Hiroshi Yagi
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan.
| | - Mana Ohtomo
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan
| | - Shogo Nagata
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan
| | - Daisuke Udagawa
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan
| | - Tomonori Tsuchida
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan
| | - Toshinori Morisaku
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, 35, Shinano-machi, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
30
|
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: 58] [Impact Index Per Article: 29.0] [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
|
31
|
Sani F, Soufi Zomorrod M, Azarpira N, Soleimani M. The Effect of Mesenchymal Stem Cell-Derived Exosomes and miR17-5p Inhibitor on Multicellular Liver Fibrosis Microtissues. Stem Cells Int 2023; 2023:8836452. [PMID: 37576406 PMCID: PMC10421706 DOI: 10.1155/2023/8836452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 08/15/2023] Open
Abstract
Background Although several studies have been conducted on modeling human liver disease, it is still challenging to mimic nonalcoholic fatty liver disease in vitro. Here, we aimed to develop a fibrotic liver microtissue composed of hepatocytes, hepatic stellate, and endothelial cells. In addition, the therapeutic effects of umbilical cord mesenchymal stem cell-derived exosomes (UC-MSC-EXO) and anti-miR17-5p as new antifibrotic drugs were investigated. Methods To create an effective preclinical fibrosis model, multicellular liver microtissues (MLMs) consisting of HepG2, LX2, and HUVECs were cultured and supplemented with a mixture of palmitic acid and oleic acid for 96 hr. Then, MLMs were exposed to UC-MSC-EXO and anti-miR17-5p in different groups. The results of cell viability, reactive oxygen species (ROS) production, liver enzyme levels, inflammation, and histopathology were analyzed to assess the treatment efficacy. Furthermore, the expression of collagen I (COL I) and α-smooth muscle actin (α-SMA) as critical matrix components, transforming growth factor beta (TGF-β), and miR-17-5p were measured. Results Free fatty acid supplementation causes fibrosis in MLMs. Our results demonstrated that UC-MSC-EXO and anti-miR17-5p attenuated TGF-β1, interleukin-1β, and interleukin-6 in all experimental groups. According to the suppression of the TGF-β1 pathway, LX2 activation was inhibited, reducing extracellular matrix proteins, including COL I and α-SMA. Also, miR-17-5p expression was elevated in fibrosis conditions. Furthermore, we showed that our treatments decreased alanine aminotransferase and aspartate aminotransferase, and increased albumin levels in the culture supernatant. We also found that both MSC-EXO and MSC-EXO + anti-miR17-5p treatments could reduce ROS production. Conclusion Our findings indicated that anti-miR17-5p and MSC-EXO might be promising therapeutic options for treating liver fibrosis. Furthermore, EXO + anti-miR had the best effects on boosting the fibrotic markers. Therefore, we propose this novel MLM model to understand fibrosis mechanisms better and develop new drugs.
Collapse
Affiliation(s)
- Farnaz Sani
- Hematology Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mina Soufi Zomorrod
- Hematology Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Khalili Street P.O. Box 7193711351, Shiraz, Iran
| | - Masoud Soleimani
- Hematology Department, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Martinez-Lopez S, Angel-Gomis E, Sanchez-Ardid E, Pastor-Campos A, Picó J, Gomez-Hurtado I. The 3Rs in Experimental Liver Disease. Animals (Basel) 2023; 13:2357. [PMID: 37508134 PMCID: PMC10376896 DOI: 10.3390/ani13142357] [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: 06/14/2023] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Patients with cirrhosis present multiple physiological and immunological alterations that play a very important role in the development of clinically relevant secondary complications to the disease. Experimentation in animal models is essential to understand the pathogenesis of human diseases and, considering the high prevalence of liver disease worldwide, to understand the pathophysiology of disease progression and the molecular pathways involved, due to the complexity of the liver as an organ and its relationship with the rest of the organism. However, today there is a growing awareness about the sensitivity and suffering of animals, causing opposition to animal research among a minority in society and some scientists, but also about the attention to the welfare of laboratory animals since this has been built into regulations in most nations that conduct animal research. In 1959, Russell and Burch published the book "The Principles of Humane Experimental Technique", proposing that in those experiments where animals were necessary, everything possible should be done to try to replace them with non-sentient alternatives, to reduce to a minimum their number, and to refine experiments that are essential so that they caused the least amount of pain and distress. In this review, a comprehensive summary of the most widely used techniques to replace, reduce, and refine in experimental liver research is offered, to assess the advantages and weaknesses of available experimental liver disease models for researchers who are planning to perform animal studies in the near future.
Collapse
Affiliation(s)
- Sebastian Martinez-Lopez
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
- Departamento de Medicina Clínica, Universidad Miguel Hernández, 03550 Sant Joan, Spain
| | - Enrique Angel-Gomis
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
- Departamento de Medicina Clínica, Universidad Miguel Hernández, 03550 Sant Joan, Spain
| | - Elisabet Sanchez-Ardid
- CIBERehd, Instituto de Salud Carlos III, 28220 Madrid, Spain
- Servicio de Patología Digestiva, Institut de Recerca IIB-Sant Pau, Hospital de Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Alberto Pastor-Campos
- Oficina de Investigación Responsable, Universidad Miguel Hernández, 03202 Elche, Spain
| | - Joanna Picó
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
| | - Isabel Gomez-Hurtado
- Instituto ISABIAL, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain
- Departamento de Medicina Clínica, Universidad Miguel Hernández, 03550 Sant Joan, Spain
- CIBERehd, Instituto de Salud Carlos III, 28220 Madrid, Spain
| |
Collapse
|
33
|
Van Campenhout R, Caufriez A, Tabernilla A, Maerten A, De Boever S, Sanz-Serrano J, Kadam P, Vinken M. Pannexin1 channels in the liver: an open enemy. Front Cell Dev Biol 2023; 11:1220405. [PMID: 37492223 PMCID: PMC10363690 DOI: 10.3389/fcell.2023.1220405] [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: 05/10/2023] [Accepted: 06/23/2023] [Indexed: 07/27/2023] Open
Abstract
Pannexin1 proteins form communication channels at the cell plasma membrane surface, which allow the transfer of small molecules and ions between the intracellular compartment and extracellular environment. In this way, pannexin1 channels play an important role in various cellular processes and diseases. Indeed, a plethora of human pathologies is associated with the activation of pannexin1 channels. The present paper reviews and summarizes the structure, life cycle, regulation and (patho)physiological roles of pannexin1 channels, with a particular focus on the relevance of pannexin1 channels in liver diseases.
Collapse
|
34
|
Sitbon A, Delmotte PR, Goumard C, Turco C, Gautheron J, Conti F, Aoudjehane L, Scatton O, Monsel A. Therapeutic potentials of mesenchymal stromal cells-derived extracellular vesicles in liver failure and marginal liver graft rehabilitation: a scoping review. Minerva Anestesiol 2023; 89:690-706. [PMID: 37079286 DOI: 10.23736/s0375-9393.23.17265-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023]
Abstract
Liver failure includes distinct subgroups of diseases: Acute liver failure (ALF) without preexisting cirrhosis, acute-on-chronic liver failure (ACLF) (severe form of cirrhosis associated with organ failures and excess mortality), and liver fibrosis (LF). Inflammation plays a key role in ALF, LF, and more specifically in ACLF for which we have currently no treatment other than liver transplantation (LT). The increasing incidence of marginal liver grafts and the shortage of liver grafts require us to consider strategies to increase the quantity and quality of available liver grafts. Mesenchymal stromal cells (MSCs) have shown beneficial pleiotropic properties with limited translational potential due to the pitfalls associated with their cellular nature. MSC-derived extracellular vesicles (MSC-EVs) are innovative cell-free therapeutics for immunomodulation and regenerative purposes. MSC-EVs encompass further advantages: pleiotropic effects, low immunogenicity, storage stability, good safety profile, and possibility of bioengineering. Currently, no human studies explored the impact of MSC-EVs on liver disease, but several preclinical studies highlighted their beneficial effects. In ALF and ACLF, data showed that MSC-EVs attenuate hepatic stellate cells activation, exert antioxidant, anti-inflammatory, anti-apoptosis, anti-ferroptosis properties, and promote regeneration of the liver, autophagy, and improve metabolism through mitochondrial function recovery. In LF, MSC-EVs demonstrated anti-fibrotic properties associated with liver tissue regeneration. Normothermic-machine perfusion (NMP) combined with MSC-EVs represents an attractive therapy to improve liver regeneration before LT. Our review suggests a growing interest in MSC-EVs in liver failure and gives an appealing insight into their development to rehabilitate marginal liver grafts through NMP.
Collapse
Affiliation(s)
- Alexandre Sitbon
- Multidisciplinary Intensive Care Unit, Department of Anesthesiology and Critical Care, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne University, Paris, France -
- UMRS-938, Research Center of Saint-Antoine (CRSA), Sorbonne University, Paris, France -
| | - Pierre-Romain Delmotte
- Multidisciplinary Intensive Care Unit, Department of Anesthesiology and Critical Care, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne University, Paris, France
| | - Claire Goumard
- UMRS-938, Research Center of Saint-Antoine (CRSA), Sorbonne University, Paris, France
- Department of Digestive, Hepatobiliary Surgery and Liver Transplantation, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne University, Paris, France
| | - Célia Turco
- UMRS-938, Research Center of Saint-Antoine (CRSA), Sorbonne University, Paris, France
- Liver Transplantation Unit, Department of Digestive and Oncologic Surgery, University Hospital of Besançon, Besançon, France
| | - Jérémie Gautheron
- UMRS-938, Research Center of Saint-Antoine (CRSA), Sorbonne University, Paris, France
| | - Filomena Conti
- UMRS-938, Research Center of Saint-Antoine (CRSA), Sorbonne University, Paris, France
- Department of Digestive, Hepatobiliary Surgery and Liver Transplantation, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne University, Paris, France
- IHU-Innovation of Cardiometabolism and Nutrition (ICAN), INSERM, Sorbonne University, Paris, France
| | - Lynda Aoudjehane
- UMRS-938, Research Center of Saint-Antoine (CRSA), Sorbonne University, Paris, France
- IHU-Innovation of Cardiometabolism and Nutrition (ICAN), INSERM, Sorbonne University, Paris, France
| | - Olivier Scatton
- UMRS-938, Research Center of Saint-Antoine (CRSA), Sorbonne University, Paris, France
- Department of Digestive, Hepatobiliary Surgery and Liver Transplantation, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne University, Paris, France
| | - Antoine Monsel
- Multidisciplinary Intensive Care Unit, Department of Anesthesiology and Critical Care, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (APHP), Sorbonne University, Paris, France
- INSERM UMRS-959 Immunology-Immunopathology-Immunotherapy (I3), Sorbonne University, Paris, France
| |
Collapse
|
35
|
Karri K, Waxman DJ. Dysregulation of murine long noncoding single-cell transcriptome in nonalcoholic steatohepatitis and liver fibrosis. RNA (NEW YORK, N.Y.) 2023; 29:977-1006. [PMID: 37015806 PMCID: PMC10275269 DOI: 10.1261/rna.079580.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 03/09/2023] [Indexed: 06/18/2023]
Abstract
LncRNAs comprise a heterogeneous class of RNA-encoding genes typified by low expression, nuclear enrichment, high tissue-specificity, and functional diversity, but the vast majority remain uncharacterized. Here, we assembled the mouse liver noncoding transcriptome from >2000 bulk RNA-seq samples and discovered 48,261 liver-expressed lncRNAs, a majority novel. Using these lncRNAs as a single-cell transcriptomic reference set, we elucidated lncRNA dysregulation in mouse models of high fat diet-induced nonalcoholic steatohepatitis and carbon tetrachloride-induced liver fibrosis. Trajectory inference analysis revealed lncRNA zonation patterns across the liver lobule in each major liver cell population. Perturbations in lncRNA expression and zonation were common in several disease-associated liver cell types, including nonalcoholic steatohepatitis-associated macrophages, a hallmark of fatty liver disease progression, and collagen-producing myofibroblasts, a central feature of liver fibrosis. Single-cell-based gene regulatory network analysis using bigSCale2 linked individual lncRNAs to specific biological pathways, and network-essential regulatory lncRNAs with disease-associated functions were identified by their high network centrality metrics. For a subset of these lncRNAs, promoter sequences of the network-defined lncRNA target genes were significantly enriched for lncRNA triplex formation, providing independent mechanistic support for the lncRNA-target gene linkages predicted by the gene regulatory networks. These findings elucidate liver lncRNA cell-type specificities, spatial zonation patterns, associated regulatory networks, and temporal patterns of dysregulation during hepatic disease progression. A subset of the liver disease-associated regulatory lncRNAs identified have human orthologs and are promising candidates for biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Kritika Karri
- Department of Biology, Boston University, Boston, Massachusetts 02215, USA
- Bioinformatics Program, Boston University, Boston, Massachusetts 02215, USA
| | - David J Waxman
- Department of Biology, Boston University, Boston, Massachusetts 02215, USA
- Bioinformatics Program, Boston University, Boston, Massachusetts 02215, USA
| |
Collapse
|
36
|
Lee YS, Seki E. In Vivo and In Vitro Models to Study Liver Fibrosis: Mechanisms and Limitations. Cell Mol Gastroenterol Hepatol 2023; 16:355-367. [PMID: 37270060 PMCID: PMC10444957 DOI: 10.1016/j.jcmgh.2023.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/26/2023] [Accepted: 05/26/2023] [Indexed: 06/05/2023]
Abstract
Liver fibrosis is a common result of liver injury owing to various kinds of chronic liver diseases. A deeper understanding of the pathophysiology of liver fibrosis and identifying potential therapeutic targets of liver fibrosis is important because liver fibrosis may progress to advanced liver diseases, such as cirrhosis and hepatocellular carcinoma. Despite numerous studies, the underlying mechanisms of liver fibrosis remain unclear. Mechanisms of the development and progression of liver fibrosis differ according to etiologies. Therefore, appropriate liver fibrosis models should be selected according to the purpose of the study and the type of underlying disease. Many in vivo animal and in vitro models have been developed to study liver fibrosis. However, there are no perfect preclinical models for liver fibrosis. In this review, we summarize the current in vivo and in vitro models for studying liver fibrosis and highlight emerging in vitro models, including organoids and liver-on-a-chip models. In addition, we discuss the mechanisms and limitations of each model.
Collapse
Affiliation(s)
- Young-Sun Lee
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California; Department of Internal Medicine, Korea University College of Medicine, Seoul, South Korea
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
37
|
Wang C, Liu Y, Gong L, Xue X, Fu K, Ma C, Li Y. Phillygenin Ameliorates Carbon Tetrachloride-Induced Liver Fibrosis: Suppression of Inflammation and Wnt/β-Catenin Signaling Pathway. Inflammation 2023:10.1007/s10753-023-01826-1. [PMID: 37219693 DOI: 10.1007/s10753-023-01826-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/18/2023] [Accepted: 04/23/2023] [Indexed: 05/24/2023]
Abstract
Liver fibrosis (LF) is caused by the chronic wound healing response to liver injury from various origins. Among the causes, inflammatory response is the central trigger of LF. Phillygenin (PHI) is a lignan derived from Forsythia suspensa, which has significant anti-inflammatory properties. However, the effect of PHI on improving LF and the underlying mechanism have rarely been studied. In this study, we used carbon tetrachloride (CCl4) to establish a mouse model of LF. Through histological analysis of liver tissue, and measurement of the levels of hepatocyte damage markers (ALT, AST, TBIL, TBA) and four indicators of LF (Col IV, HA, LN, PC-III) in serum, it was shown that PHI improved liver function and reduced the progress of LF. Subsequently, the detection of fibrogenic biomarkers in liver tissue showed that PHI inhibited the activation of hepatic stellate cells (HSCs). Next, the expression of inflammatory markers in liver tissue/serum was detected by immunohistochemistry, RT-qPCR, and ELISA, suggesting that PHI inhibited inflammation during LF. Similarly, in vitro experiments also confirmed that PHI could inhibit lipopolysaccharide-induced inflammatory responses in RAW264.7 cells, which showed strong anti-inflammatory effects. In addition, the results of network pharmacology, molecular docking, RT-qPCR and western blot confirmed that PHI could alleviate CCl4-induced LF by inhibiting the Wnt/β-catenin pathway. In conclusion, our research showed that PHI curbed LF through inhibition of HSC activation and collagen accumulation via inhibiting multiple profibrogenic factors, modulating a variety of inflammatory factors, and suppressing the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yanfang Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Lihong Gong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Xinyan Xue
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Ke Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Cheng Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, Sichuan, China.
| |
Collapse
|
38
|
Tong H, Wang L, Zhang K, Shi J, Wu Y, Bao Y, Wang C. S100A6 Activates Kupffer Cells via the p-P38 and p-JNK Pathways to Induce Inflammation, Mononuclear/macrophage Infiltration Sterile Liver Injury in Mice. Inflammation 2023; 46:534-554. [PMID: 36484925 DOI: 10.1007/s10753-022-01750-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/17/2022] [Accepted: 09/30/2022] [Indexed: 12/13/2022]
Abstract
Noninfectious liver injury, including the effects of chemical material, drugs and diet, is a major cause of liver diseases worldwide. In chemical and drugs-induced liver injury, innate inflammatory responses are mediated by extracellular danger signals. The S100 protein can act as danger signals, which can promote the migration and chemotaxis of immune cells, promote the release of various inflammatory cytokines, and regulate the body's inflammatory and immune responses. However, the role of S100A6 in inflammatory response in chemical and drugs-induced sterile liver injury remains unclear. We constructed the model of sterile liver injury induced by carbon tetrachloride (CCl4)/Paracetamol (APAP) and performed RNA sequencing (RNA-seq) on the liver tissues after injury (days 2 and 5). We analyzed inflammatory protein secretion in the liver tissue supernatant by enzyme-linked immunosorbent assay (ELISA), determined the inflammation response by bioinformatic analysis during sterile liver injury, and assessed mononuclear/macrophage infiltration by immunohistochemistry and flow cytometry. Immunohistochemistry was used to analyze the location of S100A6. We conducted inflammatory factor expression analysis and molecular mechanistic studies in Kupffer cells (KCs) induced by S100A6 using quantitative reverse transcription-polymerase chain reaction (qRT-PCR), ELISA, and western blot in vitro experiments. We performed chemokine CCL2 expression analysis and molecular mechanism studies using the same method. We used a Transwell assay to show the infiltration of mononuclear/macrophage. We here observed that aggravated inflammatory response was shown in CCl4 and APAP-administrated mice, as evidenced by enhanced production of inflammatory cytokines (TNF-α, IL-1β), and elevated mononuclear/macrophage infiltration and activation of immunity. The expression of S100A6 was significantly increased on day 2 after sterile liver injury, which is primarily produced by injured liver cells. Mechanistic studies established that S100A6 activates Kupffer cells (KCs) via the p-P38, p-JNK and P65 pathways to induce inflammation in vitro. Furthermore, TNF-α can stimulate liver cells via the p-P38 and p-JNK pathways to produce CCL2 and promote the infiltration of mononuclear/macrophage. In summary, we showed that S100A6 plays an important role in regulating inflammation, thus influencing sterile liver injury. Our findings provide novel evidence that S100A6 can as a danger signal that contributes to pro-inflammatory activation through p-P38 and p-JNK pathways in CCl4 and APAP-induced sterile liver injury in mice. In addition, the inflammatory factor TNF-α induces a large amount of CCL2 production in normal liver cells surrounding the injured area through a paracrine action, which is chemotactic for blood mononuclear/macrophage infiltration.
Collapse
Affiliation(s)
- He Tong
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Li Wang
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, Inner Mongolia, China
| | - Kefan Zhang
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Jing Shi
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Yongshuai Wu
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China
| | - Yulong Bao
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, Inner Mongolia, China.
| | - Changshan Wang
- School of Life Science, Inner Mongolia University, Yu Quan District, Xin Lin Guo Le Nan Road 49, Hohhot, 010020, Inner Mongolia, China.
| |
Collapse
|
39
|
Ghobrial DK, El-Nikhely N, Sheta E, Ragab HM, Rostom SAF, Saeed H, Wahid A. The Role of Pyrazolo[3,4-d]pyrimidine-Based Kinase Inhibitors in The Attenuation of CCl4-Induced Liver Fibrosis in Rats. Antioxidants (Basel) 2023; 12:antiox12030637. [PMID: 36978885 PMCID: PMC10045301 DOI: 10.3390/antiox12030637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Liver Fibrosis can be life-threatening if left untreated as it may lead to serious, incurable complications. The common therapeutic approach is to reverse the fibrosis while the intervention is still applicable. Celecoxib was shown to exhibit some antifibrotic properties in the induced fibrotic liver in rats. The present study aimed to investigate the possible antifibrotic properties in CCl4-induced liver fibrosis in male Sprague–Dawley rats compared to celecoxib of three novel methoxylated pyrazolo[3,4-d]pyrimidines. The three newly synthesized compounds were proved to be safe candidates. They showed a therapeutic effect against severe CCl4-induced fibrosis but at different degrees. The three compounds were able to partially reverse hepatic architectural distortion and reduce the fibrotic severity by showing antioxidant properties reducing MDA with increasing GSH and SOD levels, remodeling the extracellular matrix proteins and liver enzymes balance, and reducing the level of proinflammatory (TNF-α and IL-6) and profibrogenic (TGF-β) cytokines. The results revealed that the dimethoxy-analog exhibited the greatest activity in all the previously mentioned parameters compared to celecoxib and the other two analogs which could be attributed to the different methoxylation patterns of the derivatives. Collectively, the dimethoxy-derivative could be considered a safe promising antifibrotic candidate.
Collapse
Affiliation(s)
- Diana K. Ghobrial
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria 21526, Egypt
- Correspondence: (D.K.G.); (A.W.)
| | - Nefertiti El-Nikhely
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria 21526, Egypt
| | - Eman Sheta
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria 21321, Egypt
| | - Hanan M. Ragab
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21500, Egypt
| | - Sherif A. F. Rostom
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21500, Egypt
| | - Hesham Saeed
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria 21526, Egypt
| | - Ahmed Wahid
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21500, Egypt
- Correspondence: (D.K.G.); (A.W.)
| |
Collapse
|
40
|
Santana JG, Shewarega A, Nam D, Kahl V, Madoff DC, Zhang X, Chapiro J. Experimental VX2 Rabbit Liver Tumor Model in Carbon Tetrachloride-Induced Cirrhosis of the Liver. J Vasc Interv Radiol 2023; 34:404-408.e1. [PMID: 36473611 PMCID: PMC11037556 DOI: 10.1016/j.jvir.2022.11.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/09/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Liver cirrhosis is a major underlying factor in the development of hepatocellular carcinoma. Currently, there is an unmet need for midsize experimental vertebrate models that would offer reproducible implantable liver tumors in a cirrhotic liver background. This study establishes a protocol for a syngeneic rabbit model of VX2 liver cancer with underlying liver cirrhosis induced using carbon tetrachloride (CCl4). Male New Zealand white rabbits (n = 3) received CCl4 by intragastric administration once weekly. Concentrations started at 5% v/v CCl4 dissolved in olive oil. CCl4 dosing was progressively increased every week by 2.5% v/v increments for the duration of treatment (16 weeks total). VX2 tumors were then orthotopically implanted into the left hepatic lobe and allowed to grow for 3 weeks. Cross-sectional imaging confirmed the presence of hepatic tumors. Gross and histopathological evaluations showed reproducible tumor growth in the presence of liver cirrhosis in all animals.
Collapse
Affiliation(s)
- Jessica G Santana
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; Department of Biomedical Engineering, Yale School of Medicine, New Haven, Connecticut
| | - Annabella Shewarega
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; Department of Radiology, Charité-Universitätsmedizin Berlin, Berlin Institute of Health, Berlin, Germany
| | - David Nam
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Vinzent Kahl
- Department of Radiology, Charité-Universitätsmedizin Berlin, Berlin Institute of Health, Berlin, Germany
| | - David C Madoff
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut
| | - Xuchen Zhang
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Julius Chapiro
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut.
| |
Collapse
|
41
|
AbouSamra MM, Elgohary R, Mansy SS. Innovated pirfenidone loaded lecithin nanocapsules for targeting liver fibrosis: Formulation, characterization and in vivo study. Int J Pharm 2023; 631:122539. [PMID: 36572266 DOI: 10.1016/j.ijpharm.2022.122539] [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: 08/29/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022]
Abstract
Increasing interest in developing antifibrotic therapies became a paramount priority due to the globally raised incidence of deaths secondary to hepatic cirrhosis. This work deals with the development of innovative antifibrotic pirfenidone -loaded lecithin core nanocapsules. This with the intention to target the liver and to increase the drug bioavailability, reducing drug liver toxicity, and studying the associated hepatic microenvironment changes. PFD-loaded lecithin nanocapsules (PFD-LENCs) were prepared using the natural lipoid S45 for its dual benefits of being both a lipid and an amphiphilic surfactant. The selected formulation exhibited in vitro sustained drug release up to 24 h compared to free PFD, which is consistent with the studied pharmacokinetic profile. The studied cytotoxicity of PFD as well as PFD-LENCs exhibited negligible cytotoxicity in normal oral epithelial cells. For exploring the capability of the PFD-LENCs in reaching the liver; in vivo tracing using CLSM, in vivo biodistribution to the vital organs were conducted and electron microscopic examination for depicting nanoparticles in liver tissue was performed. Results revealed the capability of the prepared fluorescent LENC2 in reaching the liver, PFD-LENCs detection in the Disse space of the liver and the significant accumulation of PFD-LENCs in liver tissue compared to the other tested organs. The assessment of the necro-inflammatory, antioxidant and the anti-fibrotic effect of PFD-LENCs (50 & 100 mg/kg) exhibited a significant decrease of liver enzymes, TNF-α, TGF-β, Col-1, α-SMA, and TIMP-1, and a significant increase of catalase enzyme and MMP2 compared to free PFD. EM studies, revealed often detection of dendritic cells in PFD-LENCs (100 mg/kg) treated mice and abnormal collagen structure which can represent an adjunct contribution to the antifibrotic mechanism of PFD-LENCs. In conclusion, the development of this innovative PFD loaded lecithin nanocapsules achieved a targeting ability to the liver, controlled drug release, thereby increase the PFD therapeutic value in downregulating hepatic fibrosis in adjunct with the reduction of liver toxicity.
Collapse
Affiliation(s)
- Mona M AbouSamra
- Pharmaceutical Technology Department, National Research Centre, Egypt.
| | - Rania Elgohary
- Narcotics, Ergogenics and Poisons Department, National Research Centre, Egypt
| | - Soheir S Mansy
- Electron Microscopy Research Department, Theodor Bilharz Research Institute, Cairo, Egypt
| |
Collapse
|
42
|
Co-Treatments of Gardeniae Fructus and Silymarin Ameliorates Excessive Oxidative Stress-Driven Liver Fibrosis by Regulation of Hepatic Sirtuin1 Activities Using Thioacetamide-Induced Mice Model. Antioxidants (Basel) 2022; 12:antiox12010097. [PMID: 36670959 PMCID: PMC9854785 DOI: 10.3390/antiox12010097] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 12/27/2022] [Accepted: 12/28/2022] [Indexed: 01/04/2023] Open
Abstract
Gardeniae Fructus (GF, the dried ripe fruits of Gardenia jasminoides Ellis) has traditionally been used to treat various diseases in East Asian countries, such as liver disease. Silymarin is a well-known medicine used to treat numerous liver diseases globally. The present study was purposed to evaluate the synergistic effects of GF and silymarin on the thioacetamide (TAA)-induced liver fibrosis of a mouse model. Mice were orally administered with distilled water, GF (100 mg/kg, GF 100), silymarin (100 mg/kg, Sily 100), and GF and silymarin mixtures (50 and 100 mg/kg, GS 50 and 100). The GS group showed remarkable amelioration of liver injury in the serum levels and histopathology by observing the inflamed cell infiltrations and decreases in necrotic bodies through the liver tissue. TAA caused liver tissue oxidation, which was evidenced by the abnormal statuses of lipid peroxidation and deteriorations in the total glutathione in the hepatic protein levels; moreover, the immunohistochemistry supported the increases in the positive signals against 4-hydroxyneal and 8-OHdG through the liver tissue. These alterations corresponded well to hepatic inflammation by an increase in F4/80 positive cells and increases in pro-inflammatory cytokines in the hepatic protein levels; however, administration with GS, especially the high dose group, not only remarkably reduced oxidative stress and DNA damage in the liver cells but also considerably diminished pro-inflammatory cytokines, which were driven by Kupffer cell activations, as compared with each of the single treatment groups. The pharmacological properties of GS prolonged liver fibrosis by the amelioration of hepatic stellate cells’ (HSCs’) activation that is dominantly expressed by huge extracellular matrix (ECM) molecules including α-smooth muscle actin, and collagen type1 and 3, respectively. We further figured out that GS ameliorated HSCs activated by the regulation of Sirtuin 1 (Sirt1) activities in the hepatic protein levels, and this finding excellently reenacted the transforming growth factor-β-treated LX-2-cells-induced cell death signals depending on the Sirt1 activities. Future studies need to reveal the pharmacological roles of GS on the specific cell types during the liver fibrosis condition.
Collapse
|
43
|
Chae YJ, Heo H, Woo CW, Kim ST, Kwon JI, Choi MY, Sung YS, Kim KW, Kim JK, Choi Y, Woo DC. Preclinical Long-term Magnetic Resonance Imaging Study of Silymarin Liver-protective Effects. J Clin Transl Hepatol 2022; 10:1167-1175. [PMID: 36381105 PMCID: PMC9634766 DOI: 10.14218/jcth.2021.00499] [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: 11/04/2021] [Revised: 02/09/2022] [Accepted: 03/17/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS Efficacy evaluations with preclinical magnetic resonance imaging (MRI) are uncommon, but MRI in the preclinical phase of drug development provides information that is useful for longitudinal monitoring. The study aim was to monitor the protective effectiveness of silymarin with multiparameter MRI and biomarkers in a thioacetamide (TAA)-induced model of liver injury in rats. Correlation analysis was conducted to assess compare the monitoring of liver function by MRI and biomarkers. METHODS TAA was injected three times a week for 8 weeks to generate a disease model (TAA group). In the TAA and silymarin-treated (TAA-SY) groups, silymarin was administered three times weekly from week 4. MR images were acquired at 0, 2, 4, 6, and 8 weeks in the control, TAA, and TAA-SY groups. RESULTS The area under the curve to maximum time (AUCtmax) and T2* values of the TAA group decreased over the study period, but the serological markers of liver abnormality increased significantly more than those in the control group. In the TAA-SY group, MRI and serological biomarkers indicated attenuation of liver function as in the TAA group. However, pattern changes were observed from week 6 to comparable levels in the control group with silymarin treatment. Negative correlations between either AUCtmax or T2* values and the serological biomarkers were observed. CONCLUSIONS Silymarin had hepatoprotective effects on TAA-induced liver injury and demonstrated the usefulness of multiparametric MRI to evaluate efficacy in preclinical studies of liver drug development.
Collapse
Affiliation(s)
- Yeon Ji Chae
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hwon Heo
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chul-Woong Woo
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Sang-Tae Kim
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Jae-Im Kwon
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Monica Young Choi
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yu Sub Sung
- Clinical Research Center, Asan Medical Center, Seoul, Republic of Korea
| | - Kyung Won Kim
- Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong Kon Kim
- Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yoonseok Choi
- Medical Research Institute, Gangneung Asan Hospital, Gangneung-si, Gangwon-do, Republic of Korea
- Correspondence to: Dong Cheol Woo, Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea. ORCID: https://orcid.org/0000-0001-8202-015X. Tel: +82-2-3010-4155, Fax: +82-10-5559-7102, E-mail: ; Yoonseok Choi, Medical Research Institute, Gangneung Asan Hospital, University of Ulsan College of Medicine 38, Bangdong-gil, Sacheon-myeon, Gangneung-si, Gangwon-do 25440, Korea. ORCID: https://orcid.org/0000-0002-8478-2999. Tel: +82-33-610-4799, Fax: +82-33-610-3089, E-mail:
| | - Dong-Cheol Woo
- Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
- Correspondence to: Dong Cheol Woo, Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center 88, Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Korea. ORCID: https://orcid.org/0000-0001-8202-015X. Tel: +82-2-3010-4155, Fax: +82-10-5559-7102, E-mail: ; Yoonseok Choi, Medical Research Institute, Gangneung Asan Hospital, University of Ulsan College of Medicine 38, Bangdong-gil, Sacheon-myeon, Gangneung-si, Gangwon-do 25440, Korea. ORCID: https://orcid.org/0000-0002-8478-2999. Tel: +82-33-610-4799, Fax: +82-33-610-3089, E-mail:
| |
Collapse
|
44
|
The m 6A methyltransferase Mettl3 deficiency attenuates hepatic stellate cell activation and liver fibrosis. Mol Ther 2022; 30:3714-3728. [PMID: 35923112 PMCID: PMC9734030 DOI: 10.1016/j.ymthe.2022.07.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/21/2022] [Accepted: 07/30/2022] [Indexed: 12/14/2022] Open
Abstract
Activation of hepatic stellate cells (HSCs) is a central driver of liver fibrosis. Previous investigations have identified various altered epigenetic landscapes during the cellular progression of HSC activation. N6-methyladenosine (m6A) is the most abundant internal RNA modification in eukaryotic cells and is dynamically regulated under various physiological and pathophysiological conditions. However, the functional role of Mettl3-mediated m6A in liver fibrosis remains elusive. Here, we found that the HSC-specific knockout of m6A methyltransferase Mettl3 suppressed HSC activation and significantly alleviated liver fibrosis. Multi-omics analysis of HSCs showed that Mettl3 depletion reduced m6A deposition on mRNA transcripts of Lats2 (a central player of the Hippo/YAP signaling pathway) and slowed down their degradation. Elevated Lats2 increased phosphorylation of the downstream transcription factor YAP, suppressed YAP nuclear translocation, and decreased pro-fibrotic gene expression. Overexpressing YAP mutant resistant to phosphorylation by Lats2 partially rescued the activation and pro-fibrotic gene expression of Mettl3-deficient HSCs. Our study revealed that disruption of Mettl3 in HSCs mitigated liver fibrosis by controlling the Hippo/YAP signaling pathway, providing potential therapeutic strategies to alleviate liver fibrosis by targeting epitranscriptomic machinery.
Collapse
|
45
|
Kartasheva-Ebertz D, Gaston J, Lair-Mehiri L, Mottez E, Buivan TP, Massault PP, Scatton O, Gaujoux S, Vaillant JC, Pol S, Lagaye S. IL-17A in Human Liver: Significant Source of Inflammation and Trigger of Liver Fibrosis Initiation. Int J Mol Sci 2022; 23:ijms23179773. [PMID: 36077175 PMCID: PMC9456490 DOI: 10.3390/ijms23179773] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
IL-17A is considered to guide liver inflammation and fibrosis. From twenty-two human liver samples of different fibrosis stages (F0 to F4), IL-17A, IL-22, and TGFβ1 protein expression in liver tissue lysates were analyzed. Ten paired samples of liver tissue (F0–F1 stage) and blood from the same patient were used to analyze intrahepatic and blood T-lymphoid IL-17A+ cells by flow cytometry. The analyses have been performed regardless of pathology, considering the stage of fibrosis. Human liver tissue was used for the primary human liver slice cultures, followed by subsequent cytokine stimulation and fibrotic markers’ analysis by ELISA. IL-17A production in human liver tissue was significantly higher in the early fibrotic stage compared with the advanced stage. Th17 T cells and, to a lesser extent, MAIT cells were the main sources of IL-17A in both compartments, the liver and the blood. Moreover, the presence of liver Th17IL-17A+INFγ+ cells was detected in the liver. IL-17A stimulation of human liver slice culture increased the expression of profibrotic and pro-inflammatory markers. IL-17A, secreted by Th17 and MAIT cells in the liver, triggered fibrosis by inducing the expression of IL-6 and profibrotic markers and could be a target for antifibrotic treatment. Further amplitude studies are needed to confirm the current results.
Collapse
Affiliation(s)
- Daria Kartasheva-Ebertz
- Institut Pasteur, Immunobiologie des Cellules Dendritiques, INSERM U1223, F-75015 Paris, France
- Université de Paris Cité, F-75005 Paris, France
- Correspondence:
| | - Jesintha Gaston
- Institut Pasteur, Immunobiologie des Cellules Dendritiques, INSERM U1223, F-75015 Paris, France
- Université de Paris Cité, F-75005 Paris, France
| | - Loriane Lair-Mehiri
- Department of Hepatology and Addictology, AP-HP, Groupe Hospitalier Cochin, Université de Paris, F-75014 Paris, France
| | - Estelle Mottez
- Institut Pasteur, Centre de Recherche Translationnelle, F-75015 Paris, France
| | - Tan-Phuc Buivan
- Institut Pasteur, Centre de Recherche Translationnelle, F-75015 Paris, France
| | - Pierre-Philippe Massault
- Department of Digestive Surgery, AP-HP, Groupe Hospitalier Cochin, Université de Paris, F-75014 Paris, France
| | - Olivier Scatton
- Department of Hepato-Biliary and Pancreatic Surgery and Liver Transplantation, AP-HP Pitié-Salpêtrière Hospital, Medecine Sorbonne Université, F-75013 Paris, France
| | - Sebastien Gaujoux
- Department of Hepato-Biliary and Pancreatic Surgery and Liver Transplantation, AP-HP Pitié-Salpêtrière Hospital, Medecine Sorbonne Université, F-75013 Paris, France
| | - Jean-Christophe Vaillant
- Department of Hepato-Biliary and Pancreatic Surgery and Liver Transplantation, AP-HP Pitié-Salpêtrière Hospital, Medecine Sorbonne Université, F-75013 Paris, France
| | - Stanislas Pol
- Institut Pasteur, Immunobiologie des Cellules Dendritiques, INSERM U1223, F-75015 Paris, France
- Université de Paris Cité, F-75005 Paris, France
- Department of Hepatology and Addictology, AP-HP, Groupe Hospitalier Cochin, Université de Paris, F-75014 Paris, France
| | - Sylvie Lagaye
- Institut Pasteur, Immunobiologie des Cellules Dendritiques, INSERM U1223, F-75015 Paris, France
- Centre de Recherche (CDR) Saint-Antoine, INSERM—UMR_S 938/Sorbonne Université, F-75012 Paris, France
| |
Collapse
|
46
|
Computational simulation of liver fibrosis dynamics. Sci Rep 2022; 12:14112. [PMID: 35982187 PMCID: PMC9388486 DOI: 10.1038/s41598-022-18123-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/05/2022] [Indexed: 11/08/2022] Open
Abstract
Liver fibrosis is a result of homeostasis breakdown caused by repetitive injury. The accumulation of collagens disrupts liver structure and function, which causes serious consequences such as cirrhosis. Various mathematical simulation models have been developed to understand these complex processes. We employed the agent-based modelling (ABM) approach and implemented inflammatory processes in central venous regions. Collagens were individually modelled and visualised depending on their origin: myofibroblast and portal fibroblast. Our simulation showed that the administration of toxic compounds induced accumulation of myofibroblast-derived collagens in central venous regions and portal fibroblast-derived collagens in portal areas. Subsequently, these collagens were bridged between central-central areas and spread all over areas. We confirmed the consistent dynamic behaviour of collagen formulation in our simulation and from histological sections obtained via in vivo experiments. Sensitivity analyses identified dead hepatocytes caused by inflammation and the ratio of residential liver cells functioned as a cornerstone for the initiation and progression of liver fibrosis. The validated mathematical model demonstrated here shows virtual experiments that are complementary to biological experiments, which contribute to understanding a new mechanism of liver fibrosis.
Collapse
|
47
|
Liu Q, Zeng A, Liu Z, Wu C, Song L. Liver organoids: From fabrication to application in liver diseases. Front Physiol 2022; 13:956244. [PMID: 35923228 PMCID: PMC9340459 DOI: 10.3389/fphys.2022.956244] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/30/2022] [Indexed: 12/12/2022] Open
Abstract
As the largest internal organ, the liver is the key hub for many physiological processes. Previous research on the liver has been mainly conducted on animal models and cell lines, in which not only there are deficiencies in species variability and retention of heritable material, but it is also difficult for primary hepatocytes to maintain their metabolic functions after in vitro expansion. Because of the increased burden of liver disease worldwide, there is a growing demand for 3D in vitro liver models—Liver Organoids. Based on the type of initiation cells, the liver organoid can be classified as PSC-derived or ASC-derived. Liver organoids originated from ASC or primary sclerosing cholangitis, which are co-cultured in matrix gel with components such as stromal cells or immune cells, and eventually form three-dimensional structures in the presence of cytokines. Liver organoids have already made progress in drug screening, individual medicine and disease modeling with hereditary liver diseases, alcoholic or non-alcoholic liver diseases and primary liver cancer. In this review, we summarize the generation process of liver organoids and the current clinical applications, including disease modeling, drug screening and individual medical treatment, which provide new perspectives for liver physiology and disease research.
Collapse
Affiliation(s)
- Qianglin Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Anqi Zeng
- Institute of Translational Pharmacology and Clinical Application, Sichuan Academy of Chinese Medical Science, Chengdu, China
| | - Zibo Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chunjie Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Chunjie Wu, ; Linjiang Song,
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Chunjie Wu, ; Linjiang Song,
| |
Collapse
|
48
|
Ghanim M, Amer J, Salhab A, Jaradat N. Ecballium elaterium improved stimulatory effects of tissue-resident NK cells and ameliorated liver fibrosis in a thioacetamide mice model. Biomed Pharmacother 2022; 150:112942. [PMID: 35429743 DOI: 10.1016/j.biopha.2022.112942] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/26/2022] [Accepted: 04/06/2022] [Indexed: 11/30/2022] Open
Abstract
Ecballium elaterium (EE), widely used plant in Mediterranean medicine, showed anticancer activity. This study aimed to investigate EE effects on liver fibrosis in an animal model of thioacetamide (TAA). Intraperitoneal administration of TAA was performed twice weekly for four weeks in C57BL6J mice. Livers were extracted and serum were evaluated for inflammatory markers (H&E staining, ALT, AST, ALP), pro-inflammatory cytokines, fibrosis (Sirius red staining, Masson's trichrome, α-smooth muscle actin and collagen III), and metabolic (cholesterol, triglyceride, C-peptide, and fasting-blood-sugar) profiles. Glutathione, glutathione peroxidase, and catalase liver antioxidant markers were assessed. Tissue-resident NK cells from mice livers were functionally assessed for activating receptors and cytotoxicity. Compared to vehicle-treated mice, the TAA-induced liver injury showed attenuation in the histopathology outcome following EE treatment. In addition, EE-treated mice resulted in decreased serum levels of ALT, AST, and ALP, associated with a decrease in IL-20, TGF-β, IL-17, IL-22 and MCP-1 concentrations. Moreover, EE-treated mice exhibited improved lipid profile of cholesterol, triglycerides, C-peptide, and FBS. EE treatment maintained GSH, GPX, and CAT liver antioxidant activity and led to elevated counts of tissue-resident NK (trNK) cells in the TAA-mice. Consequently, trNK demonstrated an increase in CD107a and IFN-γ with improved potentials to kill activated hepatic-stellate cells in an in vitro assay. EE exhibited antifibrotic and antioxidative effects, increased the number of trNK cells, and improved metabolic outcomes. This plant extract could be a targeted therapy for patients with advanced liver injury.
Collapse
Affiliation(s)
- Mustafa Ghanim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, P.O. Box 7, Nablus, Palestine.
| | - Johnny Amer
- Department of Allied and Applied Medical Sciences, Division of Anatomy Biochemistry and Genetics, An-Najah National University, P.O. Box 7, Nablus, Palestine.
| | - Ahmad Salhab
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, An-Najah National University, P.O. Box 7, Nablus, Palestine
| | - Nidal Jaradat
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, P.O. Box 7, Nablus, Palestine
| |
Collapse
|
49
|
Zahmatkesh E, Othman A, Braun B, Aspera R, Ruoß M, Piryaei A, Vosough M, Nüssler A. In vitro modeling of liver fibrosis in 3D microtissues using scalable micropatterning system. Arch Toxicol 2022; 96:1799-1813. [PMID: 35366062 DOI: 10.1007/s00204-022-03265-7] [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: 12/09/2021] [Accepted: 02/23/2022] [Indexed: 11/02/2022]
Abstract
Liver fibrosis is the late consequence of chronic liver inflammation which could eventually lead to cirrhosis, and liver failure. Among various etiological factors, activated hepatic stellate cells (aHSCs) are the major players in liver fibrosis. To date, various in vitro liver fibrosis models have been introduced to address biological and medical questions. Availability of traditional in vitro models could not fully recapitulate complicated pathology of liver fibrosis. The purpose of this study was to develop a simple and robust model to investigate the role of aHSCs on the progression of epithelial to mesenchymal transition (EMT) in hepatocytes during liver fibrogenesis. Therefore, we applied a micropatterning approach to generate 3D co-culture microtissues consisted of HepaRG and human umbilical cord endothelial cells (HUVEC) which co-cultured with inactivated LX-2 cells or activated LX-2 cells, respectively, as normal or fibrotic liver models in vitro. The result indicated that the activated LX-2 cells could induce EMT in HepaRG cells through activation of TGF-β/SMAD signaling pathway. Besides, in the fibrotic microtissue, physiologic function of HepaRG cells attenuated compared to the control group, e.g., metabolic activity and albumin secretion. Moreover, our results showed that after treatment with Galunisertib, the fibrogenic properties decreased, in the term of gene and protein expression. In conclusion, it is proposed that aHSCs could lead to EMT in hepatocytes during liver fibrogenesis. Furthermore, the scalable micropatterning approach could provide enough required liver microtissues to prosper our understanding of the mechanisms involved in the progression of liver fibrosis as well as high throughput (HT) drug screening.
Collapse
Affiliation(s)
- Ensieh Zahmatkesh
- Department of Traumatology, Siegfried Weller Institute, University of Tübingen, BG Tübingen, 72076, Tübingen, Germany.,Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Amnah Othman
- Department of Traumatology, Siegfried Weller Institute, University of Tübingen, BG Tübingen, 72076, Tübingen, Germany
| | - Bianca Braun
- Department of Traumatology, Siegfried Weller Institute, University of Tübingen, BG Tübingen, 72076, Tübingen, Germany
| | - Romina Aspera
- Department of Traumatology, Siegfried Weller Institute, University of Tübingen, BG Tübingen, 72076, Tübingen, Germany
| | - Marc Ruoß
- Department of Traumatology, Siegfried Weller Institute, University of Tübingen, BG Tübingen, 72076, Tübingen, Germany
| | - Abbas Piryaei
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, 1665659911, Iran
| | - Andreas Nüssler
- Department of Traumatology, Siegfried Weller Institute, University of Tübingen, BG Tübingen, 72076, Tübingen, Germany.
| |
Collapse
|
50
|
Li N, Shan S, Li XQ, Chen TT, Qi M, Zhang SN, Wang ZY, Zhang LL, Wei W, Sun WY. G Protein-Coupled Receptor Kinase 2 as Novel Therapeutic Target in Fibrotic Diseases. Front Immunol 2022; 12:822345. [PMID: 35111168 PMCID: PMC8801426 DOI: 10.3389/fimmu.2021.822345] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/27/2021] [Indexed: 11/13/2022] Open
Abstract
G protein-coupled receptor kinase 2 (GRK2), an important subtype of GRKs, specifically phosphorylates agonist-activated G protein-coupled receptors (GPCRs). Besides, current research confirms that it participates in multiple regulation of diverse cells via a non-phosphorylated pathway, including interacting with various non-receptor substrates and binding partners. Fibrosis is a common pathophysiological phenomenon in the repair process of many tissues due to various pathogenic factors such as inflammation, injury, drugs, etc. The characteristics of fibrosis are the activation of fibroblasts leading to myofibroblast proliferation and differentiation, subsequent aggerate excessive deposition of extracellular matrix (ECM). Then, a positive feedback loop is occurred between tissue stiffness caused by ECM and fibroblasts, ultimately resulting in distortion of organ architecture and function. At present, GRK2, which has been described as a multifunctional protein, regulates copious signaling pathways under pathophysiological conditions correlated with fibrotic diseases. Along with GRK2-mediated regulation, there are diverse effects on the growth and apoptosis of different cells, inflammatory response and deposition of ECM, which are essential in organ fibrosis progression. This review is to highlight the relationship between GRK2 and fibrotic diseases based on recent research. It is becoming more convincing that GRK2 could be considered as a potential therapeutic target in many fibrotic diseases.
Collapse
Affiliation(s)
- Nan Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Shan Shan
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Xiu-Qin Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Ting-Ting Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Meng Qi
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Sheng-Nan Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Zi-Ying Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Ling-Ling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Wu-Yi Sun
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| |
Collapse
|