151
|
Torre P, Motta BM, Sciorio R, Masarone M, Persico M. Inflammation and Fibrogenesis in MAFLD: Role of the Hepatic Immune System. Front Med (Lausanne) 2021; 8:781567. [PMID: 34957156 PMCID: PMC8695879 DOI: 10.3389/fmed.2021.781567] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic (dysfunction)-associated fatty liver disease (MAFLD) is the definition recently proposed to better circumscribe the spectrum of conditions long known as non-alcoholic fatty liver disease (NAFLD) that range from simple steatosis without inflammation to more advanced liver diseases. The progression of MAFLD, as well as other chronic liver diseases, toward cirrhosis, is driven by hepatic inflammation and fibrogenesis. The latter, result of a "chronic wound healing reaction," is a dynamic process, and the understanding of its underlying pathophysiological events has increased in recent years. Fibrosis progresses in a microenvironment where it takes part an interplay between fibrogenic cells and many other elements, including some cells of the immune system with an underexplored or still unclear role in liver diseases. Some therapeutic approaches, also acting on the immune system, have been probed over time to evaluate their ability to improve inflammation and fibrosis in NAFLD, but to date no drug has been approved to treat this condition. In this review, we will focus on the contribution of the liver immune system in the progression of NAFLD, and on therapies under study that aim to counter the immune substrate of the disease.
Collapse
Affiliation(s)
- Pietro Torre
- Internal Medicine and Hepatology Unit, Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, University of Salerno, Salerno, Italy
| | - Benedetta Maria Motta
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Roberta Sciorio
- Internal Medicine and Hepatology Unit, Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, University of Salerno, Salerno, Italy
| | - Mario Masarone
- Internal Medicine and Hepatology Unit, Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, University of Salerno, Salerno, Italy
| | - Marcello Persico
- Internal Medicine and Hepatology Unit, Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, University of Salerno, Salerno, Italy
| |
Collapse
|
152
|
Xiao P, Li M, Zhou M, Zhao X, Wang C, Qiu J, Fang Q, Jiang H, Dong H, Zhou R. TTP protects against acute liver failure by regulating CCL2 and CCL5 through m6A RNA methylation. JCI Insight 2021; 6:149276. [PMID: 34877932 PMCID: PMC8675193 DOI: 10.1172/jci.insight.149276] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 10/27/2021] [Indexed: 01/02/2023] Open
Abstract
Tristetraprolin (TTP), an important immunosuppressive protein regulating mRNA decay through recognition of the AU-rich elements (AREs) within the 3′-UTRs of mRNAs, participates in the pathogenesis of liver diseases. However, whether TTP regulates mRNA stability through other mechanisms remains poorly understood. Here, we report that TTP was upregulated in acute liver failure (ALF), resulting in decreased mRNA stabilities of CCL2 and CCL5 through promotion of N6-methyladenosine (m6A) mRNA methylation. Overexpression of TTP could markedly ameliorate hepatic injury in vivo. TTP regulated the mRNA stabilization of CCL2 and CCL5. Interestingly, increased m6A methylation in CCL2 and CCL5 mRNAs promoted TTP-mediated RNA destabilization. Moreover, induction of TTP upregulated expression levels of WT1 associated protein, methyltransferase like 14, and YT521-B homology N6-methyladenosine RNA binding protein 2, which encode enzymes regulating m6A methylation, resulting in a global increase of m6A methylation and amelioration of liver injury due to enhanced degradation of CCL2 and CCL5. These findings suggest a potentially novel mechanism by which TTP modulates mRNA stabilities of CCL2 and CCL5 through m6A RNA methylation, which is involved in the pathogenesis of ALF.
Collapse
Affiliation(s)
- Pingping Xiao
- Hubei Province Key Laboratory of Allergy and Immunology and.,Department of Parasitology, School of Basic Medical Sciences, Wuhan University, Wuhan, China.,School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Mingxuan Li
- Hubei Province Key Laboratory of Allergy and Immunology and
| | - Mengsi Zhou
- Hubei Province Key Laboratory of Allergy and Immunology and
| | - Xuejun Zhao
- Hubei Province Key Laboratory of Allergy and Immunology and.,Department of Parasitology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Cheng Wang
- Hubei Province Key Laboratory of Allergy and Immunology and
| | - Jinglin Qiu
- School of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Qian Fang
- Hubei Province Key Laboratory of Allergy and Immunology and.,Department of Parasitology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Hong Jiang
- Department of Parasitology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Huifen Dong
- Hubei Province Key Laboratory of Allergy and Immunology and.,Department of Parasitology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Rui Zhou
- Hubei Province Key Laboratory of Allergy and Immunology and.,Department of Parasitology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
153
|
Ren L, Li J, Liu L, Wu W, Zhao D, Zhang K, Xin X, Yang L, Yin L. Resolving hepatic fibrosis via suppressing oxidative stress and an inflammatory response using a novel hyaluronic acid modified nanocomplex. Biomater Sci 2021; 9:8259-8269. [PMID: 34761752 DOI: 10.1039/d1bm01499d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Hepatic fibrosis remains a serious threat to human health globally and there are no effective antifibrotic pharmacotherapeutic strategies, to date. Upon the activation of hepatic stellate cells, excess deposition of the extracellular matrix occurs, acting as a trigger that generates reactive oxygen species and an inflammatory response, thereby exacerbating the development of hepatic fibrosis and inflammation. In this study, we incorporated an idea that targets key pathways for developing novel anti-fibrosis nanomedicine. Previous studies have reported the potential of LY294002 (LY) as a PI3K/Akt inhibitor that suppresses the HSC activation and fibrosis development; however, its poor water solubility impedes further investigation. Moreover, the proliferation of HSC, severe oxidative stress and inflammatory conditions could be undermined by oridonin (ORD) treatment. Herein, we developed an HA-ORD/LY-Lips nanocomplex, where LY294002 was encapsulated into liposomes to prepare LY-Lips while ORD was conjugated with a hyaluronic acid (HA) polymer acting as a prodrug HA-ORD. The complex exerts great potential in improving the liver-targeted drug release. We adopted a series of in vitro and in vivo evaluations which demonstrate that HA-ORD/LY-Lips can significantly avert activation of hepatic stellate cells via scavenging reactive oxygen species and suppressing an inflammatory response. Our work implements a proof of concept strategy for fibrosis treatment based on the dual antioxidative and anti-inflammatory mechanisms, which may be applicable to treat liver fibrosis associated with a dysregulated inflammatory microenvironment.
Collapse
Affiliation(s)
- Lianjie Ren
- Jiangsu Province Engineering Research Center for R&D and Evaluation of Intelligent Drugs and Key Functional Excipients, China Pharmaceutical University, Nanjing 210009, China. .,Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China. .,Center for Drug Evaluation, NMPA, Beijing 100022, China
| | - Jingjing Li
- Jiangsu Province Engineering Research Center for R&D and Evaluation of Intelligent Drugs and Key Functional Excipients, China Pharmaceutical University, Nanjing 210009, China. .,Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Lisha Liu
- Jiangsu Province Engineering Research Center for R&D and Evaluation of Intelligent Drugs and Key Functional Excipients, China Pharmaceutical University, Nanjing 210009, China. .,Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Wantao Wu
- Jiangsu Province Engineering Research Center for R&D and Evaluation of Intelligent Drugs and Key Functional Excipients, China Pharmaceutical University, Nanjing 210009, China. .,Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Di Zhao
- Jiangsu Province Engineering Research Center for R&D and Evaluation of Intelligent Drugs and Key Functional Excipients, China Pharmaceutical University, Nanjing 210009, China. .,Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Kai Zhang
- Jiangsu Province Engineering Research Center for R&D and Evaluation of Intelligent Drugs and Key Functional Excipients, China Pharmaceutical University, Nanjing 210009, China. .,Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Xiaofei Xin
- Jiangsu Province Engineering Research Center for R&D and Evaluation of Intelligent Drugs and Key Functional Excipients, China Pharmaceutical University, Nanjing 210009, China. .,Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Lei Yang
- Jiangsu Province Engineering Research Center for R&D and Evaluation of Intelligent Drugs and Key Functional Excipients, China Pharmaceutical University, Nanjing 210009, China. .,Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Lifang Yin
- Jiangsu Province Engineering Research Center for R&D and Evaluation of Intelligent Drugs and Key Functional Excipients, China Pharmaceutical University, Nanjing 210009, China. .,Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China. .,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.,Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
154
|
Su Q, Kim SY, Adewale F, Zhou Y, Aldler C, Ni M, Wei Y, Burczynski ME, Atwal GS, Sleeman MW, Murphy AJ, Xin Y, Cheng X. Single-cell RNA transcriptome landscape of hepatocytes and non-parenchymal cells in healthy and NAFLD mouse liver. iScience 2021; 24:103233. [PMID: 34755088 PMCID: PMC8560975 DOI: 10.1016/j.isci.2021.103233] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global health-care problem with limited therapeutic options. To obtain a cellular resolution of pathogenesis, 82,168 single-cell transcriptomes (scRNA-seq) across different NAFLD stages were profiled, identifying hepatocytes and 12 other non-parenchymal cell (NPC) types. scRNA-seq revealed insights into the cellular and molecular mechanisms of the disease. We discovered a dual role for hepatic stellate cells in gene expression regulation and in the potential to trans-differentiate into myofibroblasts. We uncovered distinct expression profiles of Kupffer cells versus monocyte-derived macrophages during NAFLD progression. Kupffer cells showed stronger immune responses, while monocyte-derived macrophages demonstrated a capability for differentiation. Three chimeric NPCs were identified including endothelial-chimeric stellate cells, hepatocyte-chimeric endothelial cells, and endothelial-chimeric Kupffer cells. Our work identified unanticipated aspects of mouse with NAFLD at the single-cell level and advanced the understanding of cellular heterogeneity in NAFLD livers.
Collapse
Affiliation(s)
- Qi Su
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, USA
| | - Sun Y. Kim
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, USA
| | - Funmi Adewale
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, USA
| | - Ye Zhou
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, USA
| | - Christina Aldler
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, USA
| | - Min Ni
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, USA
| | - Yi Wei
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, USA
| | - Michael E. Burczynski
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, USA
| | - Gurinder S. Atwal
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, USA
| | - Mark W. Sleeman
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, USA
| | - Andrew J. Murphy
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, USA
| | - Yurong Xin
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, USA
| | - Xiping Cheng
- Regeneron Pharmaceuticals, Inc., 777 Old Saw Mill River Road, Tarrytown, New York 10591, USA
| |
Collapse
|
155
|
Ge S, Yang W, Chen H, Yuan Q, Liu S, Zhao Y, Zhang J. MyD88 in Macrophages Enhances Liver Fibrosis by Activation of NLRP3 Inflammasome in HSCs. Int J Mol Sci 2021; 22:ijms222212413. [PMID: 34830293 PMCID: PMC8622429 DOI: 10.3390/ijms222212413] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/04/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic liver disease mediated by the activation of hepatic stellate cells (HSCs) leads to liver fibrosis. The signal adaptor MyD88 of Toll-like receptor (TLR) signaling is involved during the progression of liver fibrosis. However, the specific role of MyD88 in myeloid cells in liver fibrosis has not been thoroughly investigated. In this study, we used a carbon tetrachloride (CCl4)-induced mouse fibrosis model in which MyD88 was selectively depleted in myeloid cells. MyD88 deficiency in myeloid cells attenuated liver fibrosis in mice and decreased inflammatory cell infiltration. Furthermore, deficiency of MyD88 in macrophages inhibits the secretion of CXC motif chemokine 2 (CXCL2), which restrains the activation of HSCs characterized by NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome activation. Moreover, targeting CXCL2 by CXCR2 inhibitors attenuated the activation of HSCs and reduced liver fibrosis. Thus, MyD88 may represent a potential candidate target for the prevention and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Shuang Ge
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning 530021, China; (S.G.); (W.Y.)
| | - Wei Yang
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning 530021, China; (S.G.); (W.Y.)
| | - Haiqiang Chen
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China; (H.C.); (Q.Y.); (S.L.)
| | - Qi Yuan
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China; (H.C.); (Q.Y.); (S.L.)
| | - Shi Liu
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China; (H.C.); (Q.Y.); (S.L.)
| | - Yongxiang Zhao
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning 530021, China; (S.G.); (W.Y.)
- Correspondence: (Y.Z.); (J.Z.)
| | - Jinhua Zhang
- National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-Targeting Theranostics, Guangxi Medical University, Nanning 530021, China; (S.G.); (W.Y.)
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China; (H.C.); (Q.Y.); (S.L.)
- Correspondence: (Y.Z.); (J.Z.)
| |
Collapse
|
156
|
Yue H, Wang P, Zhang L, Ning D, Cai W, Wang Y, Wang J. Sialoglycoproteins isolated from the eggs of Carassius auratus alleviates CCL4-induced liver injury via downregulation of the IRE-α/NF-κB signaling pathway. J Food Biochem 2021; 45:e13964. [PMID: 34730246 DOI: 10.1111/jfbc.13964] [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: 07/12/2021] [Revised: 09/18/2021] [Accepted: 09/23/2021] [Indexed: 11/28/2022]
Abstract
Chemical liver injury is a common cause of liver disease primarily characterized by oxidative stress and inflammation. Sialoglycoproteins isolated from the eggs of Carassius auratus (Ca-SGP) have been proved to exhibit the antioxidant effect. However, the effect of Ca-SGP on liver injury remains unclear. Thus, this study was aimed to determine the effect of Ca-SGP on CCL4-induced chronic chemical liver injury and explore the underlying molecular mechanism. Results showed that Ca-SGP mitigated the elevated levels of serum alanine aminotransferase and aspartate aminotransferase, inhibited the systemic oxidative stress, and reduced the levels of pro-inflammatory factors TNF-α and IL-1β. Histologic results showed that Ca-SGP supplements alleviated hepatocyte necrosis and liver macrophage infiltration. Further, Ca-SGP supplement decreased endoplasmic reticulum stress-related proteins expression, including BiP, IRE-α, p-IRE-α, and TRAF2, and further inhibited the trigger of the NF-κB pathway. In summary, Ca-SGP might be a novel agent for liver injury treatment, and its potential mechanism was related to the inhibition of liver inflammation induced by the endoplasmic reticulum. PRACTICAL APPLICATION: The fish egg is an important by-product in fish processing. Carassius auratus is a common freshwater fish with large catches and low prices. However, the eggs of C. auratus are usually direct discard or processed into salted roe products, and the quality and value of these salted products are unsatisfactory. In this current study, we confirmed that sialoglycoproteins isolated from the C. auratus eggs have the potential for the treatment of liver injury and determined that its mechanism is related to the endoplasmic reticulum and inflammation, which put forward a new idea for solving the by-product of fish processing.
Collapse
Affiliation(s)
- Hao Yue
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Peng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Lei Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Ding Ning
- Malvern College Qingdao, Qingdao, China
| | - Weizhen Cai
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Yanchao Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Jingfeng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| |
Collapse
|
157
|
Lefebvre MN, Surette FA, Anthony SM, Vijay R, Jensen IJ, Pewe LL, Hancox LS, Van Braeckel-Budimir N, van de Wall S, Urban SL, Mix MR, Kurup SP, Badovinac VP, Butler NS, Harty JT. Expeditious recruitment of circulating memory CD8 T cells to the liver facilitates control of malaria. Cell Rep 2021; 37:109956. [PMID: 34731605 PMCID: PMC8628427 DOI: 10.1016/j.celrep.2021.109956] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/08/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
Circulating memory CD8 T cell trafficking and protective capacity during liver-stage malaria infection remains undefined. We find that effector memory CD8 T cells (Tem) infiltrate the liver within 6 hours after malarial or bacterial infections and mediate pathogen clearance. Tem recruitment coincides with rapid transcriptional upregulation of inflammatory genes in Plasmodium-infected livers. Recruitment requires CD8 T cell-intrinsic LFA-1 expression and the presence of liver phagocytes. Rapid Tem liver infiltration is distinct from recruitment to other non-lymphoid tissues in that it occurs both in the absence of liver tissue resident memory "sensing-and-alarm" function and ∼42 hours earlier than in lung infection by influenza virus. These data demonstrate relevance for Tem in protection against malaria and provide generalizable mechanistic insights germane to control of liver infections.
Collapse
Affiliation(s)
- Mitchell N Lefebvre
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Medical Scientist Training Program, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA
| | - Fionna A Surette
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA; Department of Microbiology and Immunology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Scott M Anthony
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Rahul Vijay
- Department of Microbiology and Immunology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Isaac J Jensen
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA
| | - Lecia L Pewe
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Lisa S Hancox
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | | | - Stephanie van de Wall
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Stina L Urban
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Madison R Mix
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Medical Scientist Training Program, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA
| | - Samarchith P Kurup
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA; Department of Microbiology and Immunology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - Noah S Butler
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA; Department of Microbiology and Immunology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA
| | - John T Harty
- Department of Pathology, University of Iowa, Carver College of Medicine, Iowa City, IA 52246, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52246, USA.
| |
Collapse
|
158
|
De Muynck K, Vanderborght B, Van Vlierberghe H, Devisscher L. The Gut-Liver Axis in Chronic Liver Disease: A Macrophage Perspective. Cells 2021; 10:2959. [PMID: 34831182 PMCID: PMC8616442 DOI: 10.3390/cells10112959] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic liver disease (CLD) is a growing health concern which accounts for two million deaths per year. Obesity, alcohol overconsumption, and progressive cholestasis are commonly characterized by persistent low-grade inflammation and advancing fibrosis, which form the basis for development of end-stage liver disease complications, including hepatocellular carcinoma. CLD pathophysiology extends to the intestinal tract and is characterized by intestinal dysbiosis, bile acid dysregulation, and gut barrier disruption. In addition, macrophages are key players in CLD progression and intestinal barrier breakdown. Emerging studies are unveiling macrophage heterogeneity and driving factors of their plasticity in health and disease. To date, in-depth investigation of how gut-liver axis disruption impacts the hepatic and intestinal macrophage pool in CLD pathogenesis is scarce. In this review, we give an overview of the role of intestinal and hepatic macrophages in homeostasis and gut-liver axis disruption in progressive stages of CLD.
Collapse
Affiliation(s)
- Kevin De Muynck
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences, Liver Research Center Ghent, Ghent University, 9000 Ghent, Belgium; (K.D.M.); (B.V.)
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Liver Research Center Ghent, Ghent University, 9000 Ghent, Belgium;
| | - Bart Vanderborght
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences, Liver Research Center Ghent, Ghent University, 9000 Ghent, Belgium; (K.D.M.); (B.V.)
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Liver Research Center Ghent, Ghent University, 9000 Ghent, Belgium;
| | - Hans Van Vlierberghe
- Hepatology Research Unit, Department of Internal Medicine and Pediatrics, Liver Research Center Ghent, Ghent University, 9000 Ghent, Belgium;
| | - Lindsey Devisscher
- Gut-Liver Immunopharmacology Unit, Department of Basic and Applied Medical Sciences, Liver Research Center Ghent, Ghent University, 9000 Ghent, Belgium; (K.D.M.); (B.V.)
| |
Collapse
|
159
|
The immune niche of the liver. Clin Sci (Lond) 2021; 135:2445-2466. [PMID: 34709406 DOI: 10.1042/cs20190654] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 09/17/2021] [Accepted: 10/08/2021] [Indexed: 12/19/2022]
Abstract
The liver is an essential organ that is critical for the removal of toxins, the production of proteins, and the maintenance of metabolic homeostasis. Behind each liver functional unit, termed lobules, hides a heterogeneous, complex, and well-orchestrated system. Despite parenchymal cells being most commonly associated with the liver's primary functionality, it has become clear that it is the immune niche of the liver that plays a central role in maintaining both local and systemic homeostasis by propagating hepatic inflammation and orchestrating its resolution. As such, the immunological processes that are at play in healthy and diseased livers are being investigated thoroughly in order to understand the underpinnings of inflammation and the potential avenues for restoring homeostasis. This review highlights recent advances in our understanding of the immune niche of the liver and provides perspectives for how the implementation of new transcriptomic, multimodal, and spatial technologies can uncover the heterogeneity, plasticity, and location of hepatic immune populations. Findings from these technologies will further our understanding of liver biology and create a new framework for the identification of therapeutic targets.
Collapse
|
160
|
Tiwari-Heckler S, Yee EU, Yalcin Y, Park J, Nguyen DHT, Gao W, Csizmadia E, Afdhal N, Mukamal KJ, Robson SC, Lai M, Schwartz RE, Jiang ZG. Adenosine deaminase 2 produced by infiltrative monocytes promotes liver fibrosis in nonalcoholic fatty liver disease. Cell Rep 2021; 37:109897. [PMID: 34706243 PMCID: PMC8606247 DOI: 10.1016/j.celrep.2021.109897] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 07/19/2021] [Accepted: 10/06/2021] [Indexed: 02/07/2023] Open
Abstract
Elevated circulating activity of adenosine deaminase 2 (ADA2) is associated with liver fibrosis in nonalcoholic fatty liver disease (NAFLD). In the liver of NAFLD patients, ADA2-positive portal macrophages are significantly associated with the degree of liver fibrosis. These liver macrophages are CD14- and CD16-positive and co-express chemokine receptors CCR2, CCR5, and CXCR3, indicating infiltrative monocyte origin. Human circulatory monocytes release ADA2 upon macrophage differentiation in vitro. When stimulated by recombinant human ADA2 (rhADA2), human monocyte-derived macrophages demonstrate upregulation of pro-inflammatory and pro-fibrotic genes, including PDGF-B, a key pro-fibrotic cytokine. This PDGF-B upregulation is reproduced by inosine, the enzymatic product of ADA2, but not adenosine, and is abolished by E359N, a loss-of-function mutation in ADA2. Finally, rhADA2 also stimulates PDGF-B production from Kupffer cells in primary human liver spheroids. Together, these data suggest that infiltrative monocytes promote fibrogenesis in NAFLD via ADA2-mediated autocrine/paracrine signaling culminating in enhanced PDGF-B production.
Collapse
Affiliation(s)
- Shilpa Tiwari-Heckler
- Department of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Gastroenterology, University Hospital Heidelberg, Heidelberg, Germany
| | - Eric U Yee
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 11794, USA
| | - Yusuf Yalcin
- Department of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jiwoon Park
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, NY, USA
| | - Duc-Huy T Nguyen
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, NY, USA
| | - Wenda Gao
- Antagen Institute for Biomedical Research, Boston, MA 02118, USA
| | - Eva Csizmadia
- Department of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Nezam Afdhal
- Department of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Kenneth J Mukamal
- Division of General Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Simon C Robson
- Department of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston MA 02215, USA
| | - Michelle Lai
- Department of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Robert E Schwartz
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, NY, USA.
| | - Z Gordon Jiang
- Department of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
161
|
Troutman TD, Kofman E, Glass CK. Exploiting dynamic enhancer landscapes to decode macrophage and microglia phenotypes in health and disease. Mol Cell 2021; 81:3888-3903. [PMID: 34464593 PMCID: PMC8500948 DOI: 10.1016/j.molcel.2021.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/19/2021] [Accepted: 07/30/2021] [Indexed: 12/24/2022]
Abstract
The development and functional potential of metazoan cells is dependent on combinatorial roles of transcriptional enhancers and promoters. Macrophages provide exceptionally powerful model systems for investigation of mechanisms underlying the activation of cell-specific enhancers that drive transitions in cell fate and cell state. Here, we review recent advances that have expanded appreciation of the diversity of macrophage phenotypes in health and disease, emphasizing studies of liver, adipose tissue, and brain macrophages as paradigms for other tissue macrophages and cell types. Studies of normal tissue-resident macrophages and macrophages associated with cirrhosis, obese adipose tissue, and neurodegenerative disease illustrate the major roles of tissue environment in remodeling enhancer landscapes to specify the development and functions of distinct macrophage phenotypes. We discuss the utility of quantitative analysis of environment-dependent changes in enhancer activity states as an approach to discovery of regulatory transcription factors and upstream signaling pathways.
Collapse
Affiliation(s)
- Ty D Troutman
- Department of Medicine, University of California, San Diego, San Diego, CA, USA; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Eric Kofman
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, USA; Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, San Diego, CA, USA
| | - Christopher K Glass
- Department of Medicine, University of California, San Diego, San Diego, CA, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
162
|
Song X, Gao X, Wang Y, Raja R, Zhang Y, Yang S, Li M, Yao Z, Wei L. HCV Core Protein Induces Chemokine CCL2 and CXCL10 Expression Through NF-κB Signaling Pathway in Macrophages. Front Immunol 2021; 12:654998. [PMID: 34531848 PMCID: PMC8438213 DOI: 10.3389/fimmu.2021.654998] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022] Open
Abstract
HCV core protein is the first structural protein synthesized during hepatitis C virus (HCV) infection and replication. It is released from virus infected liver cells and mediates multiple functions to affect host cell response. The innate immune response is the first line of defense against viral infection. After HCV infection, Kupffer cells (KCs) which are liver macrophages play an important role in host innate immune response. Kupffer cells act as phagocytes and release different cytokines and chemokines to counter viral infection and regulate inflammation and fibrosis in liver. Earlier, we have demonstrated that HCV core protein interacts with gC1qR and activates MAPK, NF-κB and PI3K/AKT pathways in macrophages. In this study, we explored the effect of HCV core protein on CCL2 and CXCL10 expression in macrophages and the signaling pathways involved. Upon silencing of gC1qR, we observed a significant decrease expression of CCL2 and CXCL10 in macrophages in the presence of HCV core protein. Inhibiting NF-κB pathway, but not P38, JNK, ERK and AKT pathways greatly reduced the expression of CCL2 and CXCL10. Therefore, our results indicate that interaction of HCV core protein with gC1qR could induce CCL2 and CXCL10 secretion in macrophages via NF-κB signaling pathway. These findings may shed light on the understanding of how leukocytes migrate into the liver and exaggerate host-derived immune responses and may provide novel therapeutic targets in HCV chronic inflammation.
Collapse
Affiliation(s)
- Xiaotian Song
- Department of Immunology, Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| | - Xue Gao
- Department of Immunology, Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| | - Yadong Wang
- Department of Infectious Diseases, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Rameez Raja
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Yaoyu Zhang
- Department of Immunology, Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| | - Shulin Yang
- Department of Immunology, Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| | - Miao Li
- Department of Immunology, Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| | - Zhiyan Yao
- Department of Immunology, Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| | - Lin Wei
- Department of Immunology, Hebei Medical University, Shijiazhuang, China.,Key Laboratory of Immune Mechanism and Intervention on Serious Disease in Hebei Province, Shijiazhuang, China
| |
Collapse
|
163
|
Slusarczyk P, Mleczko-Sanecka K. The Multiple Facets of Iron Recycling. Genes (Basel) 2021; 12:genes12091364. [PMID: 34573346 PMCID: PMC8469827 DOI: 10.3390/genes12091364] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 12/13/2022] Open
Abstract
The production of around 2.5 million red blood cells (RBCs) per second in erythropoiesis is one of the most intense activities in the body. It continuously consumes large amounts of iron, approximately 80% of which is recycled from aged erythrocytes. Therefore, similar to the “making”, the “breaking” of red blood cells is also very rapid and represents one of the key processes in mammalian physiology. Under steady-state conditions, this important task is accomplished by specialized macrophages, mostly liver Kupffer cells (KCs) and splenic red pulp macrophages (RPMs). It relies to a large extent on the engulfment of red blood cells via so-called erythrophagocytosis. Surprisingly, we still understand little about the mechanistic details of the removal and processing of red blood cells by these specialized macrophages. We have only started to uncover the signaling pathways that imprint their identity, control their functions and enable their plasticity. Recent findings also identify other myeloid cell types capable of red blood cell removal and establish reciprocal cross-talk between the intensity of erythrophagocytosis and other cellular activities. Here, we aimed to review the multiple and emerging facets of iron recycling to illustrate how this exciting field of study is currently expanding.
Collapse
|
164
|
Nanoparticles to Target and Treat Macrophages: The Ockham's Concept? Pharmaceutics 2021; 13:pharmaceutics13091340. [PMID: 34575416 PMCID: PMC8469871 DOI: 10.3390/pharmaceutics13091340] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/15/2021] [Accepted: 08/19/2021] [Indexed: 12/19/2022] Open
Abstract
Nanoparticles are nanomaterials with three external nanoscale dimensions and an average size ranging from 1 to 1000 nm. Nanoparticles have gained notoriety in technological advances due to their tunable physical, chemical, and biological characteristics. However, the administration of functionalized nanoparticles to living beings is still challenging due to the rapid detection and blood and tissue clearance by the mononuclear phagocytic system. The major exponent of this system is the macrophage. Regardless the nanomaterial composition, macrophages can detect and incorporate foreign bodies by phagocytosis. Therefore, the simplest explanation is that any injected nanoparticle will be probably taken up by macrophages. This explains, in part, the natural accumulation of most nanoparticles in the spleen, lymph nodes, and liver (the main organs of the mononuclear phagocytic system). For this reason, recent investigations are devoted to design nanoparticles for specific macrophage targeting in diseased tissues. The aim of this review is to describe current strategies for the design of nanoparticles to target macrophages and to modulate their immunological function involved in different diseases with special emphasis on chronic inflammation, tissue regeneration, and cancer.
Collapse
|
165
|
Wang T, Ma C. The hepatic macrophage pool in NASH. Cell Mol Immunol 2021; 18:2059-2060. [PMID: 33976405 PMCID: PMC8322417 DOI: 10.1038/s41423-021-00690-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 11/08/2022] Open
Affiliation(s)
- Tixiao Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province, and Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College of Shandong University, Jinan, Shandong, China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province, and Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College of Shandong University, Jinan, Shandong, China.
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
166
|
Li X, Hollingshead N, Lampert S, Truong CD, Li W, Niu J, Crispe IN, Soysa R. A conserved pathway of transdifferentiation in murine Kupffer cells. Eur J Immunol 2021; 51:2452-2463. [PMID: 34324208 DOI: 10.1002/eji.202049124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 06/07/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022]
Abstract
Abundant long-lived liver-resident macrophages, termed Kupffer cells, are activated during chronic liver injury. They secrete both pro-inflammatory and pro-fibrotic cytokines, which act on hepatic stellate cells causing their transdifferentiation into myofibroblasts that deposit collagen. In other tissues, wound-associated macrophages go further, and transdifferentiate into fibrocytes, secreting collagen themselves. We tested Kupffer cells for this property in two experimental models: mixed non-parenchymal cell culture, and precision-cut liver slice culture. Using the Emr1-Cre transgene as a driver and the RiboTag transgene as a reporter, we found that Kupffer cells undergo transdifferentiation under these circumstances. Over time, they lose the expression of both Kupffer cell-specific and macrophage-specific genes and the transcription factors that control their expression, and they begin to express multiple genes and proteins characteristic of either myofibroblasts or tissue fibroblasts. These effects were strongly conserved between non-parenchymal cell culture and liver tissue slice culture, arguing that such transdifferentiation is a conserved function of Kupffer cells. We conclude that in addition to supporting fibrosis through an action on stellate cells, Kupffer cells also participate in liver fibrosis through transdifferentiation into fibrocytes.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Nicole Hollingshead
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA
| | - Sarah Lampert
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA
| | - Camtu D Truong
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA
| | - Wanyu Li
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Junqi Niu
- Department of Hepatology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Ian N Crispe
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA.,Department of Immunology, University of Washington, Seattle, USA
| | - Radika Soysa
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA
| |
Collapse
|
167
|
Han J, Zhang X. Complement Component C3: A Novel Biomarker Participating in the Pathogenesis of Non-alcoholic Fatty Liver Disease. Front Med (Lausanne) 2021; 8:653293. [PMID: 34395461 PMCID: PMC8358116 DOI: 10.3389/fmed.2021.653293] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is currently the most common cause of chronic liver disorder worldwide. The pathological spectrum of NAFLD ranges from simple steatosis to non-alcoholic steatohepatitis (NASH) that induces progressive liver cirrhosis and eventually hepatocellular carcinoma (HCC). However, the molecular mechanisms driving the transformation of NASH are obscure. There is a compelling need for understanding the pathogenic mechanisms of NASH, and thereby providing new insight into mechanism-based therapy. Currently, several studies reported that complement system, an innate immune system, played an important role in the pathogenesis of NAFLD, which was also proved by our recent study. Complement component 3 (C3), a protein of the innate immune system, plays a hub role in the complement system. Herein, we present a review on the role and molecular mechanism of C3 in NASH as well as its implication in NASH diagnosis and treatment.
Collapse
Affiliation(s)
- Juqiang Han
- Institute of Liver Disease, The 7th Medical Centre of Chinese People Liberation Army General Hospital, Beijing, China.,The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiang Zhang
- The Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
168
|
Dynamin-related protein 1 deficiency accelerates lipopolysaccharide-induced acute liver injury and inflammation in mice. Commun Biol 2021; 4:894. [PMID: 34290349 PMCID: PMC8295278 DOI: 10.1038/s42003-021-02413-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 06/30/2021] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial fusion and fission, which are strongly related to normal mitochondrial function, are referred to as mitochondrial dynamics. Mitochondrial fusion defects in the liver cause a non-alcoholic steatohepatitis-like phenotype and liver cancer. However, whether mitochondrial fission defect directly impair liver function and stimulate liver disease progression, too, is unclear. Dynamin-related protein 1 (DRP1) is a key factor controlling mitochondrial fission. We hypothesized that DRP1 defects are a causal factor directly involved in liver disease development and stimulate liver disease progression. Drp1 defects directly promoted endoplasmic reticulum (ER) stress, hepatocyte death, and subsequently induced infiltration of inflammatory macrophages. Drp1 deletion increased the expression of numerous genes involved in the immune response and DNA damage in Drp1LiKO mouse primary hepatocytes. We administered lipopolysaccharide (LPS) to liver-specific Drp1-knockout (Drp1LiKO) mice and observed an increased inflammatory cytokine expression in the liver and serum caused by exaggerated ER stress and enhanced inflammasome activation. This study indicates that Drp1 defect-induced mitochondrial dynamics dysfunction directly regulates the fate and function of hepatocytes and enhances LPS-induced acute liver injury in vivo.
Collapse
|
169
|
Eudy BJ, McDermott CE, Liu X, da Silva RP. Targeted and untargeted metabolomics provide insight into the consequences of glycine-N-methyltransferase deficiency including the novel finding of defective immune function. Physiol Rep 2021; 8:e14576. [PMID: 32951289 PMCID: PMC7507444 DOI: 10.14814/phy2.14576] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 01/27/2023] Open
Abstract
Fatty liver disease is increasing along with the prevalence of obesity and type‐2 diabetes. Hepatic fibrosis is a major health complication for which there are no efficacious treatment options available. A better understanding of the fundamental mechanisms that contribute to the accumulation of fibrosis is needed. Glycine‐N‐methyltransferase (GNMT) is a critical enzyme in one‐carbon metabolism that serves to regulate methylation and remethylation reactions. GNMT knockout (GNMT‐/‐) mice display spontaneous hepatic fibrosis and later develop hepatocellular carcinoma. Previous literature supports the idea that hypermethylation as a consequence of GNMT deletion contributes to the hepatic phenotype observed. However, limited metabolomic information is available and the underlying mechanisms that contribute to hepatic fibrogenesis in GNMT‐/‐ mice are still incomplete. Therefore, our goals were to use dietary intervention to determine whether increased lipid load exacerbates steatosis and hepatic fibrosis in this model and to employ both targeted and untargeted metabolomics to further understand the metabolic consequences of GNMT deletion. We find that GNMT mice fed high‐fat diet do not accumulate more lipid or fibrosis in the liver and are in fact resistant to weight gain. Metabolomics analysis confirmed that pan‐hypermethylation occurs in GNMT mice resulting in a depletion of nicotinamide intermediate metabolites. Further, there is a disruption in tryptophan catabolism that prevents adequate immune cell activation in the liver. The chronic cellular damage cannot be appropriately cleared due to a lack of immune checkpoint activation. This mouse model is an excellent example of how a disruption in small molecule metabolism can significantly impact immune function.
Collapse
Affiliation(s)
- Brandon J Eudy
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, USA
| | - Caitlin E McDermott
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, USA
| | - Xiuli Liu
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Robin P da Silva
- Department of Food Science and Human Nutrition, University of Florida, Gainesville, FL, USA
| |
Collapse
|
170
|
Pose E, Coll M, Martínez‐Sánchez C, Zeng Z, Surewaard BGJ, Català C, Velasco‐de Andrés M, Lozano JJ, Ariño S, Fuster D, Niñerola‐Bazán A, Graupera I, Muñoz É, Lozano F, Sancho‐Bru P, Kubes P, Ginès P. Programmed Death Ligand 1 Is Overexpressed in Liver Macrophages in Chronic Liver Diseases, and Its Blockade Improves the Antibacterial Activity Against Infections. Hepatology 2021; 74:296-311. [PMID: 33219516 PMCID: PMC8362175 DOI: 10.1002/hep.31644] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 10/12/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIMS Bacterial infections are common and severe in cirrhosis, but their pathogenesis is poorly understood. Dysfunction of liver macrophages may play a role, but information about their function in cirrhosis is limited. Our aims were to investigate the specific profile and function of liver macrophages in cirrhosis and their contribution to infections. Macrophages from human cirrhotic livers were characterized phenotypically by transcriptome analysis and flow cytometry; function was assessed in vivo by single photon emission computerized tomography in patients with cirrhosis. Serum levels of specific proteins and expression in peripheral monocytes were determined by ELISA and flow cytometry. In vivo phagocytic activity of liver macrophages was measured by spinning disk intravital microscopy in a mouse model of chronic liver injury. APPROACH AND RESULTS Liver macrophages from patients with cirrhosis overexpressed proteins related to immune exhaustion, such as programmed death ligand 1 (PD-L1), macrophage receptor with collagenous structure (MARCO), and CD163. In vivo phagocytic activity of liver macrophages in patients with cirrhosis was markedly impaired. Monocytes from patients with cirrhosis showed overexpression of PD-L1 that paralleled disease severity, correlated with its serum levels, and was associated with increased risk of infections. Blockade of PD-L1 with anti-PD-L1 antibody caused a shift in macrophage phenotype toward a less immunosuppressive profile, restored liver macrophage in vivo phagocytic activity, and reduced bacterial dissemination. CONCLUSION Liver cirrhosis is characterized by a remarkable impairment of phagocytic function of macrophages associated with an immunosuppressive transcriptome profile. The programmed cell death receptor 1/PD-L1 axis plays a major role in the impaired activity of liver macrophages. PD-L1 blockade reverses the immune suppressive profile and increases antimicrobial activity of liver macrophages in cirrhosis.
Collapse
Affiliation(s)
- Elisa Pose
- Liver UnitHospital ClínicBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)BarcelonaSpain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Mar Coll
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)BarcelonaSpain
| | | | - Zhutian Zeng
- Snyder Institute for Chronic Diseases, University of CalgaryCalgaryABCanada
| | | | - Cristina Català
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | | | - Juan José Lozano
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)BarcelonaSpain
| | - Sílvia Ariño
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - David Fuster
- Nuclear Medicine Department, Hospital ClínicUniversity of BarcelonaBarcelonaSpain,Centro Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER‐bbn)BarcelonaSpain
| | - Aida Niñerola‐Bazán
- Nuclear Medicine Department, Hospital ClínicUniversity of BarcelonaBarcelonaSpain,Centro Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER‐bbn)BarcelonaSpain
| | - Isabel Graupera
- Liver UnitHospital ClínicBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)BarcelonaSpain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Érica Muñoz
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Francisco Lozano
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain,Immunology UnitHospital ClínicBarcelonaSpain,Biomedicine DepartmentUniversity of BarcelonaBarcelonaSpain
| | - Pau Sancho‐Bru
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain
| | - Paul Kubes
- Snyder Institute for Chronic Diseases, University of CalgaryCalgaryABCanada
| | - Pere Ginès
- Liver UnitHospital ClínicBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)BarcelonaSpain,Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)BarcelonaSpain,Medicine Department, Faculty of MedicineUniversity of BarcelonaBarcelonaSpain
| |
Collapse
|
171
|
Ding W, Duan Y, Qu Z, Feng J, Zhang R, Li X, Sun D, Zhang X, Lu Y. Acidic Microenvironment Aggravates the Severity of Hepatic Ischemia/Reperfusion Injury by Modulating M1-Polarization Through Regulating PPAR-γ Signal. Front Immunol 2021; 12:697362. [PMID: 34234785 PMCID: PMC8255974 DOI: 10.3389/fimmu.2021.697362] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/28/2021] [Indexed: 01/22/2023] Open
Abstract
Hepatic injury induced by ischemia and reperfusion (HIRI) is a major clinical problem after liver resection or transplantation. The polarization of macrophages plays an important role in regulating the severity of hepatic ischemia/reperfusion injury. Recent evidence had indicated that the ischemia induces an acidic microenvironment by causing increased anaerobic glycolysis and accumulation of lactic acid. We hypothesize that the acidic microenvironment might cause the imbalance of intrahepatic immunity which aggravated HIRI. The hepatic ischemia/reperfusion injury model was established to investigate the effect of the acidic microenvironment to liver injury. Liposomes were used to deplete macrophages in vivo. Macrophages were cultured under low pH conditions to analyze the polarization of macrophages in vitro. Activation of the PPAR-γ signal was determined by Western blot. PPAR-γ agonist GW1929 was administrated to functionally test the role of PPAR-γ in regulating macrophage-mediated effects in the acidic microenvironment during HIRI. We demonstrate that acidic microenvironment aggravated HIRI while NaHCO3 reduced liver injury through neutralizing the acid, besides, liposome abolished the protective ability of NaHCO3 through depleting the macrophages. In vivo and vitro experiment showed that acidic microenvironment markedly promoted M1 polarization but inhibited M2 polarization of macrophage. Furthermore, the mechanistic study proved that the PPAR-γ signal was suppressed during the polarization of macrophages under pH = 6.5 culture media. The addition of PPAR-γ agonist GW1929 inhibited M1 polarization under acidic environment and reduced HIRI. Our results indicate that acidic microenvironment is a key regulator in HIRI which promoted M1 polarization of macrophages through regulating PPAR-γ. Conversely, PPAR-γ activation reduced liver injury, which provides a novel therapeutic concept to prevent HIRI.
Collapse
Affiliation(s)
- Wei Ding
- Hepatopancreatobiliary Surgery Department, The Third Affiliated Hospital of Soochow University, Changzhou First People’s Hospital, Changzhou, China
- General Surgery Department, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China
| | - Yunfei Duan
- Hepatopancreatobiliary Surgery Department, The Third Affiliated Hospital of Soochow University, Changzhou First People’s Hospital, Changzhou, China
| | - Zhen Qu
- Hepatopancreatobiliary Surgery Department, The Third Affiliated Hospital of Soochow University, Changzhou First People’s Hospital, Changzhou, China
| | - Jiawei Feng
- Hepatopancreatobiliary Surgery Department, The Third Affiliated Hospital of Soochow University, Changzhou First People’s Hospital, Changzhou, China
| | - Rongsheng Zhang
- Hepatobiliary Surgery Department, Nanjing Eight One Hospital, Nanjing, China
| | - Xiaodong Li
- Hepatopancreatobiliary Surgery Department, The Third Affiliated Hospital of Soochow University, Changzhou First People’s Hospital, Changzhou, China
| | - Donglin Sun
- Hepatopancreatobiliary Surgery Department, The Third Affiliated Hospital of Soochow University, Changzhou First People’s Hospital, Changzhou, China
| | - Xiaoying Zhang
- Hepatopancreatobiliary Surgery Department, The Third Affiliated Hospital of Soochow University, Changzhou First People’s Hospital, Changzhou, China
| | - Yunjie Lu
- Hepatopancreatobiliary Surgery Department, The Third Affiliated Hospital of Soochow University, Changzhou First People’s Hospital, Changzhou, China
| |
Collapse
|
172
|
Ahmed O, Robinson MW, O'Farrelly C. Inflammatory processes in the liver: divergent roles in homeostasis and pathology. Cell Mol Immunol 2021; 18:1375-1386. [PMID: 33864004 PMCID: PMC8166849 DOI: 10.1038/s41423-021-00639-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/10/2021] [Indexed: 02/07/2023] Open
Abstract
The hepatic immune system is designed to tolerate diverse harmless foreign moieties to maintain homeostasis in the healthy liver. Constant priming and regulation ensure that appropriate immune activation occurs when challenged by pathogens and tissue damage. Failure to accurately discriminate, regulate, or effectively resolve inflammation offsets this balance, jeopardizing overall tissue health resulting from an either overly tolerant or an overactive inflammatory response. Compelling scientific and clinical evidence links dysregulated hepatic immune and inflammatory responses upon sterile injury to several pathological conditions in the liver, particularly nonalcoholic steatohepatitis and ischemia-reperfusion injury. Murine and human studies have described interactions between diverse immune repertoires and nonhematopoietic cell populations in both physiological and pathological activities in the liver, although the molecular mechanisms driving these associations are not clearly understood. Here, we review the dynamic roles of inflammatory mediators in responses to sterile injury in the context of homeostasis and disease, the clinical implications of dysregulated hepatic immune activity and therapeutic developments to regulate liver-specific immunity.
Collapse
Affiliation(s)
- Ola Ahmed
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Mark W Robinson
- Department of Biology, Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Ireland
| | - Cliona O'Farrelly
- School of Medicine, Trinity College Dublin, Dublin, Ireland.
- School of Biochemistry & Immunology, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
173
|
Activated platelets contribute to the progression of hepatocellular carcinoma by altering the tumor environment. Life Sci 2021; 277:119612. [PMID: 33991548 DOI: 10.1016/j.lfs.2021.119612] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/30/2021] [Accepted: 04/30/2021] [Indexed: 12/22/2022]
Abstract
AIM Hepatocellular carcinoma (HCC) is a primary liver cancer that usually develops in a background of chronic liver disease and prolonged inflammation. A major contributor in the complex molecular pathogenesis of HCC is the highly intertwined cross-talk between the tumor and the surrounding stromal cells, such as hepatic stellate cells, endothelial cells, macrophages and other immune cells. These tumor-stroma interactions actively fuel tumor growth and modulate the hepatic microenvironment to benefit tumor invasion and disease progression. Platelets have been reported to interact with different cell types in the tumor microenvironment, including tumor cells, stellate cells and macrophages. MATERIALS AND METHODS Mice were treated with hepatocarcinogenic compound diethylnitrosamine for 25 weeks to induce HCC in the background of fibrosis and inflammation. From week 10, anti-platelet drug Clopidogrel was added to the drinking water and mice were given ad libitum access. KEY FINDINGS In this study, we show that activated platelets promote tumor cell proliferation and contribute to the adverse tumor-stroma cross-talk that fuels tumor progression. We also show that inhibiting platelet activation with the P2Y12-inhibitor Clopidogrel decreases the number of tumors in a chemically induced mouse model for HCC. SIGNIFICANCE These results suggest an important role for platelets in the pathogenesis of HCC and that the use of anti-platelet drugs may be therapeutically relevant for patients with liver cancer.
Collapse
|
174
|
Maretti-Mira AC, Golden-Mason L, Salomon MP, Kaplan MJ, Rosen HR. Cholesterol-Induced M4-Like Macrophages Recruit Neutrophils and Induce NETosis. Front Immunol 2021; 12:671073. [PMID: 34012454 PMCID: PMC8126646 DOI: 10.3389/fimmu.2021.671073] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/20/2021] [Indexed: 01/11/2023] Open
Abstract
The liver is the central organ for cholesterol synthesis and homeostasis. The effects of dietary cholesterol on hepatic injury, mainly of oxidized low-density lipoproteins (OxLDL), are not fully understood. Here, we show that the degree of cholesterol oxidation had different impacts on the global gene expression of human M2-like macrophages, with highly oxidized LDL causing the most dramatic changes. M2-like macrophages and Kupffer cells undergo M4-like polarization, decreasing the expression of important markers, such as IL10, MRC1, and CD163. These cells also displayed functional changes, with reduced phagocytic capacity, increased neutrophil recruitment, and more effective neutrophil extracellular traps (NETs) induction. Our findings provide a link between LDL oxidation and modification of peripheral and liver macrophage function.
Collapse
Affiliation(s)
- Ana C Maretti-Mira
- Department of Medicine, Keck School of Medicine, Gastroenterology & Hepatology, Research Center for Liver Disease, University of Southern California (USC), Los Angeles, CA, United States
| | - Lucy Golden-Mason
- Department of Medicine, Keck School of Medicine, Gastroenterology & Hepatology, Research Center for Liver Disease, University of Southern California (USC), Los Angeles, CA, United States
| | - Matthew P Salomon
- Department of Medicine, Keck School of Medicine, Gastroenterology & Hepatology, Research Center for Liver Disease, University of Southern California (USC), Los Angeles, CA, United States
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Hugo R Rosen
- Department of Medicine, Keck School of Medicine, Gastroenterology & Hepatology, Research Center for Liver Disease, University of Southern California (USC), Los Angeles, CA, United States
| |
Collapse
|
175
|
Qin N, Xu G, Wang Y, Zhan X, Gao Y, Wang Z, Fu S, Shi W, Hou X, Wang C, Li R, Liu Y, Wang J, Zhao H, Xiao X, Bai Z. Bavachin enhances NLRP3 inflammasome activation induced by ATP or nigericin and causes idiosyncratic hepatotoxicity. Front Med 2021; 15:594-607. [PMID: 33909257 DOI: 10.1007/s11684-020-0809-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 06/03/2020] [Indexed: 12/31/2022]
Abstract
Psoraleae Fructus (PF) is a well-known traditional herbal medicine in China, and it is widely used for osteoporosis, vitiligo, and other diseases in clinical settings. However, liver injury caused by PF and its preparations has been frequently reported in recent years. Our previous studies have demonstrated that PF could cause idiosyncratic drug-induced liver injury (IDILI), but the mechanism underlying its hepatotoxicity remains unclear. This paper reports that bavachin isolated from PF enhances the specific stimuli-induced activation of the NLRP3 inflammasome and leads to hepatotoxicity. Bavachin boosts the secretion of IL-1β and caspase-1 caused by ATP or nigericin but not those induced by poly(I:C), monosodium urate crystal, or intracellular lipopolysaccharide. Bavachin does not affect AIM2 or NLRC4 inflammasome activation. Mechanistically, bavachin specifically increases the production of nigericin-induced mitochondrial reactive oxygen species among the most important upstream events in the activation of the NLRP3 inflammasome. Bavachin increases the levels of aspartate transaminase and alanine aminotransferase in serum and hepatocyte injury accompanied by the secretion of IL-1β via a mouse model of lipopolysaccharide-mediated susceptibility to IDILI. These results suggest that bavachin specifically enhances the ATP- or nigericin-induced activation of the NLRP3 inflammasome. Bavachin also potentially contributes to PF-induced idiosyncratic hepatotoxicity. Moreover, bavachin and PF should be evaded among patients with diseases linked to the ATP- or nigericin-mediated activation of the NLRP3 inflammasome, which may be a dangerous factor for liver injury.
Collapse
Affiliation(s)
- Nan Qin
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.,Department of Pharmacy, Nantong Hospital of Traditional Chinese Medicine, Nantong, 226300, China
| | - Guang Xu
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Yan Wang
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.,School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Xiaoyan Zhan
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Yuan Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Zhilei Wang
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Shubin Fu
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Wei Shi
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Xiaorong Hou
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Chunyu Wang
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Ruisheng Li
- Research Center for Clinical and Translational Medicine, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Yan Liu
- Research Center for Clinical and Translational Medicine, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Jiabo Wang
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Haiping Zhao
- School of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330004, China.
| | - Xiaohe Xiao
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China. .,Integrative Medical Centre, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Zhaofang Bai
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
176
|
Kim H, Lee DS, An TH, Park HJ, Kim WK, Bae KH, Oh KJ. Metabolic Spectrum of Liver Failure in Type 2 Diabetes and Obesity: From NAFLD to NASH to HCC. Int J Mol Sci 2021; 22:ijms22094495. [PMID: 33925827 PMCID: PMC8123490 DOI: 10.3390/ijms22094495] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023] Open
Abstract
Liver disease is the spectrum of liver damage ranging from simple steatosis called as nonalcoholic fatty liver disease (NAFLD) to hepatocellular carcinoma (HCC). Clinically, NAFLD and type 2 diabetes coexist. Type 2 diabetes contributes to biological processes driving the severity of NAFLD, the primary cause for development of chronic liver diseases. In the last 20 years, the rate of non-viral NAFLD/NASH-derived HCC has been increasing rapidly. As there are currently no suitable drugs for treatment of NAFLD and NASH, a class of thiazolidinediones (TZDs) drugs for the treatment of type 2 diabetes is sometimes used to improve liver failure despite the risk of side effects. Therefore, diagnosis, prevention, and treatment of the development and progression of NAFLD and NASH are important issues. In this review, we will discuss the pathogenesis of NAFLD/NASH and NAFLD/NASH-derived HCC and the current promising pharmacological therapies of NAFLD/NASH. Further, we will provide insights into "adipose-derived adipokines" and "liver-derived hepatokines" as diagnostic and therapeutic targets from NAFLD to HCC.
Collapse
Affiliation(s)
- Hyunmi Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (H.-J.P.); (W.K.K.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Da Som Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (H.-J.P.); (W.K.K.)
| | - Tae Hyeon An
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (H.-J.P.); (W.K.K.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Hyun-Ju Park
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (H.-J.P.); (W.K.K.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (H.-J.P.); (W.K.K.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (H.-J.P.); (W.K.K.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34141, Korea
- Correspondence: (K.-H.B.); (K.-J.O.); Tel.: +82-42-860-4268 (K.-H.B.); +82-42-879-8265 (K.-J.O.)
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (H.-J.P.); (W.K.K.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34141, Korea
- Correspondence: (K.-H.B.); (K.-J.O.); Tel.: +82-42-860-4268 (K.-H.B.); +82-42-879-8265 (K.-J.O.)
| |
Collapse
|
177
|
Fathi F, Sanei B, Ganjalikhani Hakemi M, Saidi RF, Rezaei A. Liver Resection Promotes (Regulates) Proinflammatory Cytokines in Patients with Hepatocellular Carcinoma. Can J Gastroenterol Hepatol 2021; 2021:5593655. [PMID: 33987145 PMCID: PMC8093046 DOI: 10.1155/2021/5593655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Several animal studies have shown the roles of cytokines in regulating liver regeneration following liver resection (LR), which is a type of surgery designed to remove cancerous tumors from the liver. This study investigated how the expressions and serum levels of some pro- and anti-inflammatory cytokines in patients with hepatocellular carcinoma (HCC) were changed during LR. METHODS Liver tissues from 15 patients with HCC were collected and the levels of interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), IL-1α, IL-1 β, IL-10, and transforming growth factor-beta1 (TGF-β1) were assessed using real-time PCR assay at different times before and after LR. The serum values of TNF-α and IL-6 were also measured by ELISA. RESULTS After 60 and 90 minutes of LR, IL-6 gene expression was significantly increased (P < 0.001 - 0.05). The same trend was also observed in TNF-α expression after 90 minutes of LR (P < 0.01). No significant changes were observed in the expressions of IL-1α, IL-1β, IL-10, and TGF-β1 before and after LR. In addition, LR had significant effects on TNF-α and IL-6 serum levels (P < 0.05 - 0.0001). CONCLUSION Our data provided further evidence to reveal that IL-6 and TNF-α cytokines are critical to improve liver regeneration.
Collapse
Affiliation(s)
- Farshid Fathi
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Behnam Sanei
- Department of Hepatobiliary & Pancreatic Surgery and Liver Transplantation, Al-Zahra Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Reza F. Saidi
- Division of Transplant Services, Department of Surgery, SUNY Upstate Medical University Syracuse, Syracuse, NY 13210, USA
| | - Abbas Rezaei
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
178
|
Zimmermann A, Hänsel R, Gemünden K, Kegel-Hübner V, Babel J, Bläker H, Matz-Soja M, Seehofer D, Damm G. In Vivo and In Vitro Characterization of Primary Human Liver Macrophages and Their Inflammatory State. Biomedicines 2021; 9:biomedicines9040406. [PMID: 33918803 PMCID: PMC8070551 DOI: 10.3390/biomedicines9040406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 11/16/2022] Open
Abstract
Liver macrophages (LMs) play a central role in acute and chronic liver pathologies. Investigation of these processes in humans as well as the development of diagnostic tools and new therapeutic strategies require in vitro models that closely resemble the in vivo situation. In our study, we sought to gain further insight into the role of LMs in different liver pathologies and into their characteristics after isolation from liver tissue. For this purpose, LMs were characterized in human liver tissue sections using immunohistochemistry and bioinformatic image analysis. Isolated cells were characterized in suspension using FACS analyses and in culture using immunofluorescence staining and laser scanning microscopy as well as functional assays. The majority of our investigated liver tissues were characterized by anti-inflammatory LMs which showed a homogeneous distribution and increased cell numbers in correlation with chronic liver injuries. In contrast, pro-inflammatory LMs appeared as temporary and locally restricted reactions. Detailed characterization of isolated macrophages revealed a complex disease dependent pattern of LMs consisting of pro- and anti-inflammatory macrophages of different origins, regulatory macrophages and monocytes. Our study showed that in most cases the macrophage pattern can be transferred in adherent cultures. The observed exceptions were restricted to LMs with pro-inflammatory characteristics.
Collapse
Affiliation(s)
- Andrea Zimmermann
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.Z.); (R.H.); (K.G.); (V.K.-H.); (J.B.); (D.S.)
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - René Hänsel
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.Z.); (R.H.); (K.G.); (V.K.-H.); (J.B.); (D.S.)
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), Leipzig University, 04107 Leipzig, Germany
| | - Kilian Gemünden
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.Z.); (R.H.); (K.G.); (V.K.-H.); (J.B.); (D.S.)
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - Victoria Kegel-Hübner
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.Z.); (R.H.); (K.G.); (V.K.-H.); (J.B.); (D.S.)
| | - Jonas Babel
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.Z.); (R.H.); (K.G.); (V.K.-H.); (J.B.); (D.S.)
| | - Hendrik Bläker
- Institute for Pathology, University Hospital, Leipzig University, 04103 Leipzig, Germany;
| | - Madlen Matz-Soja
- Rudolf-Schönheimer-Institute of Biochemistry, Leipzig University, 04103 Leipzig, Germany;
- Department for Hepatology, University Hospital, Leipzig University, 04103 Leipzig, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.Z.); (R.H.); (K.G.); (V.K.-H.); (J.B.); (D.S.)
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.Z.); (R.H.); (K.G.); (V.K.-H.); (J.B.); (D.S.)
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany
- Correspondence: ; Tel.: +49-341-9739656
| |
Collapse
|
179
|
Traum D, Wang YJ, Schwarz KB, Schug J, Wong DK, Janssen HLA, Terrault NA, Khalili M, Wahed AS, Murray KF, Rosenthal P, Ling SC, Rodriguez-Baez N, Sterling RK, Lau DT, Block TM, Feldman MD, Furth EE, Lee WM, Kleiner DE, Lok AS, Kaestner KH, Chang KM. Highly multiplexed 2-dimensional imaging mass cytometry analysis of HBV-infected liver. JCI Insight 2021; 6:146883. [PMID: 33621209 PMCID: PMC8119221 DOI: 10.1172/jci.insight.146883] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/18/2021] [Indexed: 02/06/2023] Open
Abstract
Studies of human hepatitis B virus (HBV) immune pathogenesis are hampered by limited access to liver tissues and technologies for detailed analyses. Here, utilizing imaging mass cytometry (IMC) to simultaneously detect 30 immune, viral, and structural markers in liver biopsies from patients with hepatitis B e antigen+ (HBeAg+) chronic hepatitis B, we provide potentially novel comprehensive visualization, quantitation, and phenotypic characterizations of hepatic adaptive and innate immune subsets that correlated with hepatocellular injury, histological fibrosis, and age. We further show marked correlations between adaptive and innate immune cell frequencies and phenotype, highlighting complex immune interactions within the hepatic microenvironment with relevance to HBV pathogenesis.
Collapse
Affiliation(s)
- Daniel Traum
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.,Medical Research, The Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
| | - Yue J Wang
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.,Biomedical Sciences, College of Medicine, Florida State University, Tallahasee, Florida, USA
| | | | - Jonathan Schug
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - David Kh Wong
- Toronto Centre for Liver Disease, University of Toronto, Toronto, Ontario, Canada
| | - Harry LA Janssen
- Toronto Centre for Liver Disease, University of Toronto, Toronto, Ontario, Canada
| | - Norah A Terrault
- Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Mandana Khalili
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Abdus S Wahed
- University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Karen F Murray
- Cleveland Clinic Pediatric Institute, Cleveland, Ohio, USA
| | | | - Simon C Ling
- The Hospital for Sick Children and Department of Paediatrics and University of Toronto, Toronto, Canada
| | - Norberto Rodriguez-Baez
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Richard K Sterling
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Daryl Ty Lau
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | - Michael D Feldman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Elizabeth E Furth
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - William M Lee
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - David E Kleiner
- Laboratory of Pathology, National Cancer Institute, Bethesda, Maryland, USA
| | - Anna S Lok
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kyong-Mi Chang
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.,Medical Research, The Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
180
|
Singanayagam A, Triantafyllou E. Macrophages in Chronic Liver Failure: Diversity, Plasticity and Therapeutic Targeting. Front Immunol 2021; 12:661182. [PMID: 33868313 PMCID: PMC8051585 DOI: 10.3389/fimmu.2021.661182] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic liver injury results in immune-driven progressive fibrosis, with risk of cirrhosis development and impact on morbidity and mortality. Persistent liver cell damage and death causes immune cell activation and inflammation. Patients with advanced cirrhosis additionally experience pathological bacterial translocation, exposure to microbial products and chronic engagement of the immune system. Bacterial infections have a high incidence in cirrhosis, with spontaneous bacterial peritonitis being the most common, while the subsequent systemic inflammation, organ failure and immune dysregulation increase the mortality risk. Tissue-resident and recruited macrophages play a central part in the development of inflammation and fibrosis progression. In the liver, adipose tissue, peritoneum and intestines, diverse macrophage populations exhibit great phenotypic and functional plasticity determined by their ontogeny, epigenetic programming and local microenvironment. These changes can, at different times, promote or ameliorate disease states and therefore represent potential targets for macrophage-directed therapies. In this review, we discuss the evidence for macrophage phenotypic and functional alterations in tissue compartments during the development and progression of chronic liver failure in different aetiologies and highlight the potential of macrophage modulation as a therapeutic strategy for liver disease.
Collapse
Affiliation(s)
- Arjuna Singanayagam
- Infection and Immunity Clinical Academic Group, St. George’s University Hospitals NHS Foundation Trust, London, United Kingdom
| | - Evangelos Triantafyllou
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| |
Collapse
|
181
|
Xue R, Qiu J, Wei S, Liu M, Wang Q, Wang P, Sha B, Wang H, Shi Y, Zhou J, Rao J, Lu L. Lycopene alleviates hepatic ischemia reperfusion injury via the Nrf2/HO-1 pathway mediated NLRP3 inflammasome inhibition in Kupffer cells. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:631. [PMID: 33987329 PMCID: PMC8106004 DOI: 10.21037/atm-20-7084] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Lycopene is a naturally occurring carotenoid found in many fruits and vegetables, which has antioxidant effects. Although lycopene’s protective effect has been observed on ischemia reperfusion (IR) injury in different organs, the effect of lycopene on Kupffer cells (KCs) has not been clearly elucidated in IR-induced acute hepatic inflammatory injury. Methods Mice were administered with either olive oil (10 mL/kg body weight) as the control or lycopene (20 mg/kg body weight) by gavage for 2 weeks before undergoing hepatic IR injury. Results In this study, we observed that the levels of aspartate aminotransferases (AST), alanine aminotransferase (ALT), and the percentages of hepatocellular apoptosis in mice pretreated with lycopene were significantly lower than control mice. Lycopene inhibited F4/80+ macrophage and Ly6G+ neutrophil accumulation, which further decreased the levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin 6 (IL-6). Interestingly, lycopene induced increased autophagy in KCs, which was evidenced by elevated autophagosomes and the increased protein level of LC3B. In these KCs, lycopene-induced upregulation of autophagy inhibited NOD-like receptor family pyrin domain-containing 3 protein (NLRP3) inflammasome activation, which was demonstrated by the reduced mRNA and protein levels of NLRP3, cleaved caspase-1, an apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and IL-1β. Furthermore, 3-methyladenine, an autophagy inhibitor, abolished lycopene’s inhibitory effect on the NLRP3 inflammasome in KCs, which led to increased hepatic IR injury. Intriguingly, we identified that the protein levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1) were elevated in KCs isolated from IR-stressed mice pretreated with lycopene. Nrf2-siRNA or HO-1-siRNA could block the autophagy activation enhanced by lycopene in KCs, resulting in the activation of the NLRP3 inflammasome and aggravated hepatic IR injury. Conclusions Our findings demonstrated that lycopene promoted Nrf2/HO-1 pathway activation and further suppressed the NLRP3 inflammasome via enhancing KC autophagy, which alleviated hepatic IR injury.
Collapse
Affiliation(s)
- Rong Xue
- School of Medicine, Southeast University, Nanjing, China.,Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University, Nanjing, China & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Jiannan Qiu
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University, Nanjing, China & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,The Affiliated Cancer Hospital (Jiangsu Cancer Hospital), Nanjing Medical University, Nanjing, China
| | - Song Wei
- School of Medicine, Southeast University, Nanjing, China.,The Affiliated Cancer Hospital (Jiangsu Cancer Hospital), Nanjing Medical University, Nanjing, China
| | - Mu Liu
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University, Nanjing, China & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,The Affiliated Cancer Hospital (Jiangsu Cancer Hospital), Nanjing Medical University, Nanjing, China
| | - Qi Wang
- School of Medicine, Southeast University, Nanjing, China.,The Affiliated Cancer Hospital (Jiangsu Cancer Hospital), Nanjing Medical University, Nanjing, China
| | - Peng Wang
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University, Nanjing, China & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,The Affiliated Cancer Hospital (Jiangsu Cancer Hospital), Nanjing Medical University, Nanjing, China
| | - Bowen Sha
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University, Nanjing, China & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,The Affiliated Cancer Hospital (Jiangsu Cancer Hospital), Nanjing Medical University, Nanjing, China
| | - Hao Wang
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University, Nanjing, China & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,The Affiliated Cancer Hospital (Jiangsu Cancer Hospital), Nanjing Medical University, Nanjing, China
| | - Yong Shi
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University, Nanjing, China & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,The Affiliated Cancer Hospital (Jiangsu Cancer Hospital), Nanjing Medical University, Nanjing, China
| | - Jinren Zhou
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University, Nanjing, China & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,The Affiliated Cancer Hospital (Jiangsu Cancer Hospital), Nanjing Medical University, Nanjing, China
| | - Jianhua Rao
- Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University, Nanjing, China & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China
| | - Ling Lu
- School of Medicine, Southeast University, Nanjing, China.,Hepatobiliary Center of The First Affiliated Hospital, Nanjing Medical University, Nanjing, China & Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, China.,The Affiliated Cancer Hospital (Jiangsu Cancer Hospital), Nanjing Medical University, Nanjing, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, China.,Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
182
|
Zivko C, Fuhrmann G, Luciani P. Liver-derived extracellular vesicles: A cell by cell overview to isolation and characterization practices. Biochim Biophys Acta Gen Subj 2021; 1865:129559. [DOI: 10.1016/j.bbagen.2020.129559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/16/2020] [Accepted: 02/11/2020] [Indexed: 02/08/2023]
|
183
|
Rockey DC, Friedman SL. Fibrosis Regression After Eradication of Hepatitis C Virus: From Bench to Bedside. Gastroenterology 2021; 160:1502-1520.e1. [PMID: 33529675 PMCID: PMC8601597 DOI: 10.1053/j.gastro.2020.09.065] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/01/2020] [Accepted: 09/06/2020] [Indexed: 02/06/2023]
Abstract
Hepatitis C virus (HCV) infection and its complications have been the major cause of cirrhosis and its complications for several decades in the Western world. Until recently, treatment for HCV with interferon-based regimens was associated with moderate success but was difficult to tolerate. More recently, however, an arsenal of novel and highly effective direct-acting antiviral (DAA) drugs has transformed the landscape by curing HCV in a broad range of patients, including those with established advanced fibrosis, cirrhosis, comorbidities, and even those with complications of cirrhosis. Fibrosis is a dynamic process comprising both extracellular matrix deposition, as well as its degradation. With almost universal sustained virologic response (SVR) (ie, elimination of HCV), it is timely to explore whether HCV eradication can reverse fibrosis and cirrhosis. Indeed, fibrosis in several types of liver disease is reversible, including HCV. However, we do not know with certainty in whom fibrosis regression can be expected after HCV elimination, how quickly it occurs, and whether antifibrotic therapies will be indicated in those with persistent cirrhosis. This review summarizes the evidence for reversibility of fibrosis and cirrhosis after HCV eradication, its impact on clinical outcomes, and therapeutic prospects for directly promoting fibrosis regression in patients whose fibrosis persists after SVR.
Collapse
Affiliation(s)
- Don C Rockey
- The Medical University of South Carolina, Charleston, South Carolina.
| | - Scott L Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
184
|
Chang B, Huang A, Saxena R, Sun Y, Liu S, Zhou G, Li B, Teng G, Zhao J, Zhang W, Jiang Y, Han S, Yang Z, Zhao J, Zou Z, Liangpunsakul S. Hepatic Histopathology Among Excessive Drinkers Without Advanced Liver Disease. Alcohol Alcohol 2021; 56:669-677. [PMID: 33765150 DOI: 10.1093/alcalc/agab017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/07/2021] [Accepted: 02/23/2021] [Indexed: 11/13/2022] Open
Abstract
AIMS Alcohol-associated liver disease represents a spectrum of histopathological changes from steatosis to advanced fibrosis and cirrhosis. The major goals of this retrospective study were to characterize the histologic features in patients with excessive alcohol use who presented with an abnormal hepatic panel and/or abnormal radiographic imaging and did not meet the clinical diagnosis of alcoholic hepatitis or cirrhosis. METHODS We performed a retrospective study to describe hepatic histology of 62 and 83 excessive drinkers with normal and abnormal serum aspartate transaminase, respectively. The types of inflammatory cells in the liver were characterized by immunohistochemistry for CD4, CD8, CD20, CD68 and myeloperoxidase. RESULTS Among 62 patients with aspartate aminotransferase (AST) ≤ 50 U/L, 37% had histological evidence of steatosis. Of these, we found evidence of hepatocyte ballooning (21%), lobular inflammation (50%), portal inflammation (52%) and fibrosis (14%). For those with AST > 50 U/L, the presence of hepatic steatosis, lobular inflammation and portal inflammation was observed in 29, 60 and 69% of patients, respectively. Fibrosis was found in 33%, four with bridging fibrosis, and one with cirrhosis. We observed the aggregation of CD68+ macrophages, rather than normally distributed with minimal neutrophilic infiltration. Lobular and portal lymphocytic infiltrations are primarily CD8+ T cells. CONCLUSION Abnormal hepatic histopathology occurs in excessive drinkers with normal transaminase activity. Future studies to determine the diagnostic modalities to detect such abnormalities and to better understand its clinical implications and long-term outcome are needed.
Collapse
Affiliation(s)
- Binxia Chang
- The Center for Non-Infectious Liver Disease, Institute of Alcoholic liver Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Ang Huang
- The Center for Non-Infectious Liver Disease, Institute of Alcoholic liver Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Romil Saxena
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yin Sun
- The Center for Non-Infectious Liver Disease, Institute of Alcoholic liver Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Shuhong Liu
- Department of Pathology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Guangde Zhou
- Department of Pathology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Baosen Li
- The Center for Non-Infectious Liver Disease, Institute of Alcoholic liver Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Guangju Teng
- The Center for Non-Infectious Liver Disease, Institute of Alcoholic liver Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Jun Zhao
- The Center for Non-Infectious Liver Disease, Institute of Alcoholic liver Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Wei Zhang
- The Center for Non-Infectious Liver Disease, Institute of Alcoholic liver Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Yanchao Jiang
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Sen Han
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Zhihong Yang
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jingmin Zhao
- Department of Pathology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Zhengsheng Zou
- The Center for Non-Infectious Liver Disease, Institute of Alcoholic liver Disease, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| |
Collapse
|
185
|
3-B-RUT, a derivative of RUT, protected against alcohol-induced liver injury by attenuating inflammation and oxidative stress. Int Immunopharmacol 2021; 95:107471. [PMID: 33756231 DOI: 10.1016/j.intimp.2021.107471] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 12/25/2022]
Abstract
Alcoholic liver disease (ALD) is the most common chronic liver disease worldwide. Currently, there is no definitive treatment for alcohol-induced liver injury (ALI). Inflammatory response and oxidative stress play a crucial role in ALI. Cyclooxygenase 2 (COX-2) can be induced by inflammation and it has been reported that the enhanced expression of COX-2 in alcoholic liver injury. Rutaecarpine (RUT) was extracted from evodia rutaecarpa. RUT has a wide range of pharmacological activities. In order to increase its anti-inflammatory activity, our group introduced sulfonyl group to synthesized the 3-[2-(trifluoromethoxy)benzenesulfonamide]-rutaecarpine (3-B-RUT). In this study, we explored the protective effect of 3-B-RUT on alcoholic liver injury in vivo and in vitro and preliminarily explore its mechanism. Mice ALI model was established according to the chronic-plus-binge ethanol model. Results showed that 3-B-RUT (20 μg/kg) attenuated alcohol-induced liver injury and suppressed liver inflammation and oxidative stress, and the effect was comparable to RUT (20 mg/kg). In vitro results are consistent with in vivo results. Mechanistically, the 3-B-RUT might suppress inflammatory response and oxidative stress by regulating activation of NF-κB/COX-2 pathway. In summary, 3-B-RUT, a derivative of RUT, may be a promising clinical candidate for ALI treatment.
Collapse
|
186
|
Characterization of the inflammatory microenvironment and hepatic macrophage subsets in experimental hepatocellular carcinoma models. Oncotarget 2021; 12:562-577. [PMID: 33796224 PMCID: PMC7984829 DOI: 10.18632/oncotarget.27906] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/15/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide. HCC typically develops on a background of chronic inflammation and fibrosis with tumor associated macrophages (TAMs) playing an important role in chronic inflammation-induced HCC and progression. However, the liver harbors unique macrophages, resident liver Kupffer cells (KCs) and monocyte-derived macrophages (Mo-Mφ), and their contribution to HCC and to the population of TAMs is incompletely known. Here, we characterized the tumor microenvironment and the proportion and transcriptional profile of hepatic macrophages (Mφ) in two commonly used HCC mouse models. A gradually increased expression of inflammatory, immune regulatory, fibrotic and cell proliferation pathways and markers was observed during diethylnitrosamine (DEN)- and non-alcoholic steatohepatitis (NASH)-induced HCC development. The transcriptional phenotypes of isolated hepatic Mφ subsets were clearly distinct and shifted during HCC development, with mixed pro-inflammatory and tumor-promoting expression profiles. There were marked differences between the models as well, with Mφ in NASH-HCC exhibiting a more immunomodulatory phenotype, in conjunction with an upregulation of lipid metabolism genes. Our data show that at least some infiltrated macrophages display expression of pro-tumoral markers, and that Kupffer cells are part of the population of TAMs and enhance tumor progression. These insights are useful to further unravel sequential pathogenic events during hepatocarcinogenesis and direct future development of new treatment strategies for HCC.
Collapse
|
187
|
Xiang X, Wang J, Lu D, Xu X. Targeting tumor-associated macrophages to synergize tumor immunotherapy. Signal Transduct Target Ther 2021; 6:75. [PMID: 33619259 PMCID: PMC7900181 DOI: 10.1038/s41392-021-00484-9] [Citation(s) in RCA: 520] [Impact Index Per Article: 130.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/30/2020] [Accepted: 10/15/2020] [Indexed: 02/08/2023] Open
Abstract
The current treatment strategies in advanced malignancies remain limited. Notably, immunotherapies have raised hope for a successful control of these advanced diseases, but their therapeutic responses are suboptimal and vary considerably among individuals. Tumor-associated macrophages (TAMs) are a major component of the tumor microenvironment (TME) and are often correlated with poor prognosis and therapy resistance, including immunotherapies. Thus, a deeper understanding of the complex roles of TAMs in immunotherapy regulation could provide new insight into the TME. Furthermore, targeting of TAMs is an emerging field of interest due to the hope that these strategies will synergize with current immunotherapies. In this review, we summarize recent studies investigating the involvement of TAMs in immune checkpoint inhibition, tumor vaccines and adoptive cell transfer therapies, and discuss the therapeutic potential of targeting TAMs as an adjuvant therapy in tumor immunotherapies.
Collapse
Affiliation(s)
- Xiaonan Xiang
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, China
| | - Jianguo Wang
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, China
| | - Di Lu
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, CAMS, Hangzhou, China.
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
188
|
Jensen BAH, Holm JB, Larsen IS, von Burg N, Derer S, Sonne SB, Pærregaard SI, Damgaard MV, Indrelid SA, Rivollier A, Agrinier AL, Sulek K, Arnoldussen YJ, Fjære E, Marette A, Angell IL, Rudi K, Treebak JT, Madsen L, Åkesson CP, Agace W, Sina C, Kleiveland CR, Kristiansen K, Lea TE. Lysates of Methylococcus capsulatus Bath induce a lean-like microbiota, intestinal FoxP3 +RORγt +IL-17 + Tregs and improve metabolism. Nat Commun 2021; 12:1093. [PMID: 33597537 PMCID: PMC7889900 DOI: 10.1038/s41467-021-21408-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/27/2021] [Indexed: 02/08/2023] Open
Abstract
Interactions between host and gut microbial communities are modulated by diets and play pivotal roles in immunological homeostasis and health. We show that exchanging the protein source in a high fat, high sugar, westernized diet from casein to whole-cell lysates of the non-commensal bacterium Methylococcus capsulatus Bath is sufficient to reverse western diet-induced changes in the gut microbiota to a state resembling that of lean, low fat diet-fed mice, both under mild thermal stress (T22 °C) and at thermoneutrality (T30 °C). Concomitant with microbiota changes, mice fed the Methylococcus-based western diet exhibit improved glucose regulation, reduced body and liver fat, and diminished hepatic immune infiltration. Intake of the Methylococcu-based diet markedly boosts Parabacteroides abundances in a manner depending on adaptive immunity, and upregulates triple positive (Foxp3+RORγt+IL-17+) regulatory T cells in the small and large intestine. Collectively, these data point to the potential for leveraging the use of McB lysates to improve immunometabolic homeostasis.
Collapse
MESH Headings
- Animals
- Diet
- Forkhead Transcription Factors/immunology
- Forkhead Transcription Factors/metabolism
- Homeostasis/immunology
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Intestine, Large/immunology
- Intestine, Large/metabolism
- Intestine, Large/microbiology
- Intestine, Small/immunology
- Intestine, Small/metabolism
- Intestine, Small/microbiology
- Male
- Methylococcus capsulatus/chemistry
- Methylococcus capsulatus/immunology
- Mice, Inbred C57BL
- Microbiota/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Obesity/immunology
- Proteins/immunology
- Proteins/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Mice
Collapse
Affiliation(s)
- Benjamin A H Jensen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Laval University, Laval, QC, Canada.
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Jacob B Holm
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Clinical Microbiomics, Copenhagen, Denmark
| | - Ida S Larsen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Laval University, Laval, QC, Canada
| | - Nicole von Burg
- Mucosal Immunology, Department of Health Technology, Technical University of Denmark, Copenhagen, Denmark
| | - Stefanie Derer
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Si B Sonne
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Simone I Pærregaard
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Mucosal Immunology, Department of Health Technology, Technical University of Denmark, Copenhagen, Denmark
| | - Mads V Damgaard
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Integrative Metabolism and Environmental Influences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stine A Indrelid
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Oslo, Norway
| | - Aymeric Rivollier
- Mucosal Immunology, Department of Health Technology, Technical University of Denmark, Copenhagen, Denmark
| | - Anne-Laure Agrinier
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Laval University, Laval, QC, Canada
| | - Karolina Sulek
- Novo Nordisk Foundation Center for Basic Metabolic Research, Integrative Metabolism and Environmental Influences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yke J Arnoldussen
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Oslo, Norway
| | - Even Fjære
- Institute of Marine Research, Bergen, Norway
| | - André Marette
- Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Laval University, Laval, QC, Canada
| | - Inga L Angell
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Oslo, Norway
| | - Knut Rudi
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Oslo, Norway
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Integrative Metabolism and Environmental Influences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lise Madsen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Institute of Marine Research, Bergen, Norway
| | - Caroline Piercey Åkesson
- Department of Anatomy and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Oslo, Norway
| | - William Agace
- Mucosal Immunology, Department of Health Technology, Technical University of Denmark, Copenhagen, Denmark
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Christian Sina
- Institute of Nutritional Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Charlotte R Kleiveland
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Oslo, Norway
| | - Karsten Kristiansen
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
- Institute of Metagenomics, BGI-Shenzhen, Shenzhen, P.R. China.
| | - Tor E Lea
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Oslo, Norway.
| |
Collapse
|
189
|
Du Y, Paglicawan L, Soomro S, Abunofal O, Baig S, Vanarsa K, Hicks J, Mohan C. Epigallocatechin-3-Gallate Dampens Non-Alcoholic Fatty Liver by Modulating Liver Function, Lipid Profile and Macrophage Polarization. Nutrients 2021; 13:599. [PMID: 33670347 PMCID: PMC7918805 DOI: 10.3390/nu13020599] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 12/17/2022] Open
Abstract
Epigallocatechin-3-gallate (EGCG) has been shown to attenuate obesity, fatty liver disease, hepatic inflammation and lipid profiles. Here, we validate the efficacy of EGCG in a murine model of non-alcoholic fatty liver disease (NAFLD) and extend the mechanistic insights. NAFLD was induced in mice by a high-fat diet (HFD) with 30% fructose. EGCG was administered at a low dose (25 mg/kg/day, EGCG-25) or high dose (50 mg/kg/day, EGCG-50) for 8 weeks. In HFD-fed mice, EGCG attenuated body and liver weight by ~22% and 47%, respectively, accompanied by ~47% reduction in hepatic triglyceride (TG) accumulation and ~38% reduction in serum cholesterol, resonating well with previous reports in the literature. In EGCG-treated mice, the hepatic steatosis score and the non-alcoholic steatohepatitis activity score were both reduced by ~50% and ~57%, respectively, accompanied by improvements in hepatic inflammation grade. Liver enzymes were improved ~2-3-fold following EGCG treatment, recapitulating previous reports. Hepatic flow cytometry demonstrated that EGCG-fed mice had lower Ly6C+, MHCII+ and higher CD206+, CD23+ hepatic macrophage infiltration, indicating that EGCG impactedM1/M2 macrophage polarization. Our study further validates the salubrious effects of EGCG on NAFLD and sheds light on a novel mechanistic contribution of EGCG, namely hepatic M1-to-M2 macrophage polarization. These findings offer further support for the use of EGCG in human NAFLD.
Collapse
Affiliation(s)
- Yong Du
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (L.P.); (S.S.); (O.A.); (S.B.); (K.V.)
| | - Laura Paglicawan
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (L.P.); (S.S.); (O.A.); (S.B.); (K.V.)
| | - Sanam Soomro
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (L.P.); (S.S.); (O.A.); (S.B.); (K.V.)
| | - Omar Abunofal
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (L.P.); (S.S.); (O.A.); (S.B.); (K.V.)
| | - Sahar Baig
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (L.P.); (S.S.); (O.A.); (S.B.); (K.V.)
| | - Kamala Vanarsa
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (L.P.); (S.S.); (O.A.); (S.B.); (K.V.)
| | - John Hicks
- Department of Pathology, Texas Children’s Hospital, Houston, TX 77030, USA;
| | - Chandra Mohan
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA; (L.P.); (S.S.); (O.A.); (S.B.); (K.V.)
| |
Collapse
|
190
|
Inhibition of androgen/AR signaling inhibits diethylnitrosamine (DEN) induced tumour initiation and remodels liver immune cell networks. Sci Rep 2021; 11:3646. [PMID: 33574348 PMCID: PMC7878907 DOI: 10.1038/s41598-021-82252-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
A promotional role for androgen receptor (AR) signaling in hepatocellular carcinogenesis is emerging. In pre-clinical models, including diethylnitrosamine- (DEN-) induced hepatocellular carcinoma (HCC), anti-androgen therapies delay hepatocarcinogenesis. However, pharmacologic anti-androgen therapy in advanced HCC patients fails, suggesting that AR plays a role in HCC onset. This study aims to characterize AR expression and function throughout DEN-induced liver inflammation and carcinogenesis and evaluate the efficacy of prophylactic AR antagonism to prevent hepatocarcinogenesis. We demonstrate that pharmacologic AR antagonism with enzalutamide inhibits hepatocellular carcinogenesis. With enzalutamide treatment, we observe decreased CYP2E1 expression, reducing DEN-induced hepatocyte death and DNA ethyl-adducts. AR protein expression analyses show that DEN causes an initial upregulation of AR in portal fibroblasts and leukocytes, but not hepatocytes, suggesting that hepatocyte-autonomous AR signaling is not essential for DEN-induced carcinogenesis. Ablating androgen signaling by surgical castration reduced pre-carcinogen Kupffer cell populations but did not alter DEN-mediated immune cell recruitment nor AR expression. In this study, we identified that anti-androgen interventions modulate mutagenic DNA adducts, tumour initiation, and immune cell composition. Additionally, we find that AR expression in hepatocytes is not present during nor required for early DEN-mediated carcinogenesis.
Collapse
|
191
|
Hastings KL, Green MD, Gao B, Ganey PE, Roth RA, Burleson GR. Beyond Metabolism: Role of the Immune System in Hepatic Toxicity. Int J Toxicol 2021; 39:151-164. [PMID: 32174281 DOI: 10.1177/1091581819898399] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The liver is primarily thought of as a metabolic organ; however, the liver is also an important mediator of immunological functions. Key perspectives on this emerging topic were presented in a symposium at the 2018 annual meeting of the American College of Toxicology entitled "Beyond metabolism: Role of the immune system in hepatic toxicity." Viral hepatitis is an important disease of the liver for which insufficient preventive vaccines exist. Host immune responses inadequately clear these viruses and often potentiate immunological inflammation that damages the liver. In addition, the liver is a key innate immune organ against bacterial infection. Hepatocytes and immune cells cooperatively control systemic and local bacterial infections. Conversely, bacterial infection can activate multiple types of immune cells and pathways to cause hepatocyte damage and liver injury. Finally, the immune system and specifically cytokines and drugs can interact in idiosyncratic drug-induced liver injury. This rare disease can result in a disease spectrum that ranges from mild to acute liver failure. The immune system plays a role in this disease spectrum.
Collapse
Affiliation(s)
| | | | - Bin Gao
- Laboratory of Liver Diseases, NIH, Bethesda, MD, USA
| | - Patricia E Ganey
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Robert A Roth
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Gary R Burleson
- BRT-Burleson Research Technologies, Inc, Morrisville, NC, USA
| |
Collapse
|
192
|
Hogg C, Panir K, Dhami P, Rosser M, Mack M, Soong D, Pollard JW, Jenkins SJ, Horne AW, Greaves E. Macrophages inhibit and enhance endometriosis depending on their origin. Proc Natl Acad Sci U S A 2021; 118:e2013776118. [PMID: 33536334 PMCID: PMC8017702 DOI: 10.1073/pnas.2013776118] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Macrophages are intimately involved in the pathophysiology of endometriosis, a chronic inflammatory disorder characterized by the growth of endometrial-like tissue (lesions) outside the uterus. By combining genetic and pharmacological monocyte and macrophage depletion strategies we determined the ontogeny and function of macrophages in a mouse model of induced endometriosis. We demonstrate that lesion-resident macrophages are derived from eutopic endometrial tissue, infiltrating large peritoneal macrophages (LpM) and monocytes. Furthermore, we found endometriosis to trigger continuous recruitment of monocytes and expansion of CCR2+ LpM. Depletion of eutopic endometrial macrophages results in smaller endometriosis lesions, whereas constitutive inhibition of monocyte recruitment significantly reduces peritoneal macrophage populations and increases the number of lesions. Reprogramming the ontogeny of peritoneal macrophages such that embryo-derived LpM are replaced by monocyte-derived LpM decreases the number of lesions that develop. We propose a putative model whereby endometrial macrophages are "proendometriosis" while newly recruited monocyte-derived macrophages, possibly in LpM form, are "antiendometriosis." These observations highlight the importance of monocyte-derived macrophages in limiting disease progression.
Collapse
Affiliation(s)
- Chloe Hogg
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Kavita Panir
- Centre for Early Life, Warwick Medical School, University of Warwick, CV2 2DX Coventry, United Kingdom
| | - Priya Dhami
- Centre for Early Life, Warwick Medical School, University of Warwick, CV2 2DX Coventry, United Kingdom
| | - Matthew Rosser
- Centre for Early Life, Warwick Medical School, University of Warwick, CV2 2DX Coventry, United Kingdom
| | - Matthias Mack
- Department of Internal Medicine II-Nephrology, University Hospital Regensburg, 93042 Regensburg, Germany
| | - Daniel Soong
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Jeffrey W Pollard
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Stephen J Jenkins
- Centre for Inflammation Research, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Andrew W Horne
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Erin Greaves
- Centre for Early Life, Warwick Medical School, University of Warwick, CV2 2DX Coventry, United Kingdom;
| |
Collapse
|
193
|
Xie M, Chia RH, Li D, Teo FX, Krueger C, Sabapathy K. Functional interaction between macrophages and hepatocytes dictate the outcome of liver fibrosis. Life Sci Alliance 2021; 4:4/4/e202000803. [PMID: 33514653 PMCID: PMC7893818 DOI: 10.26508/lsa.202000803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/07/2021] [Accepted: 01/15/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocytes and liver-resident macrophages known as Kupffer cells (KCs) are key cell types involved in liver fibrosis. The transcription factor c-Jun plays a fundamental role in regulating hepatocyte and macrophage functions. We have examined c-Jun's role in the functional interaction of these cells during liver fibrosis induced by carbon tetrachloride. While hepatocyte-specific c-jun deletion led to increased fibrosis, the opposite outcome was observed when c-jun was deleted in both hepatocytes and KCs. Molecular analyses revealed compromised cytokine gene expression as the apical event related to the phenotype. Yet, purified hepatocytes from both mouse cohorts showed similar defects in cytokine gene expression. However, we noted increased macrophage infiltration in the absence of c-Jun in hepatocytes, which when chemically depleted, reversed the phenotype. Consistently, c-jun deletion in KCs alone also led to reduced fibrosis and cytokine gene expression. By contrast, c-jun deletion in hepatocytes and KCs did not affect the resolution phase after fibrotic injury. These data together demonstrate a pro-fibrogenic role for c-Jun in hepatocytes and KCs that functionally interact to regulate liver fibrosis.
Collapse
Affiliation(s)
- Min Xie
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, Singapore
| | - Ren Hui Chia
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dan Li
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, Singapore
| | - Fanny Xueting Teo
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, Singapore
| | - Christian Krueger
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, Singapore
| | - Kanaga Sabapathy
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, Singapore .,Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Institute of Molecular and Cellular Biology, Singapore, Singapore
| |
Collapse
|
194
|
Wang H, Zhang CS, Fang BB, Hou J, Li WD, Li ZD, Li L, Bi XJ, Li L, Abulizi A, Shao YM, Lin RY, Wen H. Dual Role of Hepatic Macrophages in the Establishment of the Echinococcus multilocularis Metacestode in Mice. Front Immunol 2021; 11:600635. [PMID: 33488594 PMCID: PMC7820908 DOI: 10.3389/fimmu.2020.600635] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022] Open
Abstract
Echinococcus multilocularis larvae, predominantly located in the liver, cause a tumor-like parasitic disease, alveolar echinococcosis (AE), that is characterized by increased infiltration of various immune cells, including macrophages, around the lesion that produces an "immunosuppressive" microenvironment, favoring its persistent infection. However, the role of hepatic macrophages in the host defense against E. multilocularis infection remains poorly defined. Using human liver tissues from patients with AE and a hepatic experimental mouse model of E. multilocularis, we investigated the phenotype and function of hepatic macrophages during the parasite infection. In the present study, we found that a large number of CD68+ macrophages accumulated around the metacestode lesion in the liver of human AE samples and that both S100A9+ proinflammatory (M1 phenotype) and CD163+ anti-inflammatory (M2 phenotype) macrophages were significantly higher in close liver tissue (CLT) than in distant liver tissue (DLT), whereas M2 macrophages represent the dominant macrophage population. Furthermore, E. multilocularis-infected mice exhibited a massive increase in macrophage (F4/80+) infiltration in the liver as early as day 5, and the infiltrated macrophages were mainly monocyte-derived macrophages (CD11bhi F4/80int MoMFs) that preferentially differentiated into the M1 phenotype (iNOS+) at the early stage of E. multilocularis infection and then polarized to anti-inflammatory macrophages of the M2 phenotype (CD206+) at the chronic stage of infection. We further showed that elimination of macrophages by treatment of mice with clodronate-liposomes before E. multilocularis infection impaired worm expulsion and was accompanied by a reduction in liver fibrosis, yielding a high parasite burden. These results suggest that hepatic macrophages may play a dual role in the establishment and development of E. multilocularis metacestodes in which early larvae clearance is promoted by M1 macrophages while persistent metacestode infection is favored by M2 macrophages.
Collapse
Affiliation(s)
- Hui Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Basic Medical College, Xinjiang Medical University, Urumqi, China.,Branch of The First Affiliated Hospital of Xinjiang Medical University, Changji, China
| | - Chuan-Shan Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Basic Medical College, Xinjiang Medical University, Urumqi, China.,Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Bin-Bin Fang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jiao Hou
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wen-Ding Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhi-De Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Liang Li
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiao-Juan Bi
- Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Liang Li
- Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Abuduaini Abulizi
- Department of Hepatic Hydatid and Hepatobiliary Surgery, Digestive and Vascular Surgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ying-Mei Shao
- Department of Hepatic Hydatid and Hepatobiliary Surgery, Digestive and Vascular Surgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ren-Yong Lin
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hao Wen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, WHO Collaborating Centre on Prevention and Case Management of Echinococcosis, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
195
|
Liang XY, Hong FF, Yang SL. Astragaloside IV Alleviates Liver Inflammation, Oxidative Stress and Apoptosis to Protect Against Experimental Non-Alcoholic Fatty Liver Disease. Diabetes Metab Syndr Obes 2021; 14:1871-1883. [PMID: 33953586 PMCID: PMC8089473 DOI: 10.2147/dmso.s304817] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Non-alcoholic fatty liver disease (NAFLD) is the main form of chronic liver disease in the world. Astragaloside IV (ASIV) has been tested in experimental models of different diseases. The purpose of this study was to evaluate the effect and protective mechanism of ASIV on NAFLD. METHODS Lipopolysaccharide (LPS)- and palmitate acid (PA)-induced RAW264.7 cells and LO2 cells were used as a NAFLD model. The mice NAFLD model was evaluated by hematoxylin-eosin staining (HE staining), and aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels. Liver lipid metabolism was evaluated by triglyceride (TG) and total cholesterol (TC) kits and oil red O staining. Oxidative stress indicators were examined through biochemical methods. Inflammatory factors were explored through enzyme-linked immuno sorbent assay (ELISA), real-time quantitative PCR and oxidative stress indicator kits. The expression levels of 5-LO (5-lipoxygenase) and leukotriene A4 hydrolase (LTA4H) were checked by real-time quantitative PCR and Western blotting. Apoptosis was detected by Annexin V-FITC/PI cell apoptosis detection kit. RESULTS Our results showed that in vivo ASIV significantly reduced liver tissue damage, and serum AST, ALT and serum TG levels in NAFLD mice. In vitro, ASIV reduced cell supernatant TG and TC content increased by PA treatment, and significantly decreased the accumulation of intracellular lipid droplets induced by PA treatment. Additionally, ASIV reduced reactive oxygen species (ROS) and malondialdehyde (MDA) levels, and restored glutathione peroxidase (GSH-Px) levels in PA-treated LO2 cell supernatant. Furthermore, ASIV inhibited the production of proinflammatory cytokines (IL-6 and TNF-α) in RAW264.7 cells induced by LPS. We also found that ASIV downregulated the expression of 5-LO and LTB4 (leukotriene B4) in NAFLD mice. Moreover, ASIV restored apoptotic protein (Bax and Bcl-2) expression in PA-treated LO2 cells. CONCLUSION ASIV may reduce liver steatosis, hepatocyte oxidative stress and apoptosis, and decrease liver inflammation, thereby attenuating the progression of NAFLD and thus might be of therapeutic interest.
Collapse
Affiliation(s)
- Xiao-yu Liang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Fen-Fang Hong
- Experimental Teaching Center, Nanchang University, Nanchang, 330031, People’s Republic of China
- Fen-Fang Hong Experimental Teaching Center, Nanchang University, Nanchang, 330031, People’s Republic of ChinaTel +86 18970965319 Email
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang, 330006, People’s Republic of China
- Correspondence: Shu-Long Yang Department of Physiology, College of Medicine, Nanchang University, Nanchang, 330006, People’s Republic of ChinaTel +86 13576291532 Email
| |
Collapse
|
196
|
Su SB, Qin SY, Xian XL, Huang FF, Huang QL, ZhangDi HJ, Jiang HX. Interleukin-22 regulating Kupffer cell polarization through STAT3/Erk/Akt crosstalk pathways to extenuate liver fibrosis. Life Sci 2021; 264:118677. [PMID: 33129875 DOI: 10.1016/j.lfs.2020.118677] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/14/2020] [Accepted: 10/24/2020] [Indexed: 12/19/2022]
Abstract
AIMS Interleukin (IL)-22 activates multiple signaling pathways to exert anti-inflammatory effects, but few studies have examined whether and how IL-22 may shift macrophage polarization between M1 (pro-inflammatory) and M2 (anti-inflammatory) states and thereby influence the progression of hepatic fibrosis. MAIN METHODS Utilized CCl4 to induce liver fibrosis in mice, detected the role of IL-22 in inhibiting liver fibrosis by regulating Kupffer cells (KCs) polarization in vivo and in vitro. U937 cells were used to confirm the mechanism of IL-22 regulating macrophage polarization via the STAT3/Erk/Akt pathways. Human liver specimens were collected to verify the correlation between the levels of IL-22 and KCs during liver fibrogenesis. KEY FINDINGS During CCl4-induced liver fibrosis progression in mice, adding exogenous IL-22 significantly inhibited pro-fibrogenic and macrophage phenotype-altering factors secreted by M1-KCs, and it increased the number of M2-KCs. In co-cultures of hepatic stellate cells and KCs from mice treated with IL-22, a high M2/M1-KCs ratio inhibited collagen production and stellate cell activation. These results suggest that IL-22 can increase the ratio of M2-KCs to M1-KCs and thereby attenuate the progression of liver fibrosis. Mechanistic studies in vitro showed that IL-22 promoted polarization of lipopolysaccharide-treated U937 macrophages from M1 to M2. The cytokine exerted these effects by activating the STAT3 pathway while suppressing Erk1/2 and Akt pathways. Furthermore, immunofluorescent staining in human liver specimens confirmed that IL-22 levels positively correlated with the number of M2-KCs during liver fibrogenesis. SIGNIFICANCE IL-22 regulates the STAT3/Erk/Akt to increase the M2/M1-KCs ratio and thereby slow liver fibrogenesis.
Collapse
Affiliation(s)
- Si-Biao Su
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Province, China
| | - Shan-Yu Qin
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Province, China
| | - Xiao-Long Xian
- Graduate School of Guangxi Medical University, Nanning 530021, Guangxi Province, China
| | - Fei-Fei Huang
- Graduate School of Guangxi Medical University, Nanning 530021, Guangxi Province, China
| | - Qiu-Lan Huang
- Graduate School of Guangxi Medical University, Nanning 530021, Guangxi Province, China
| | - Han-Jing ZhangDi
- Graduate School of Guangxi Medical University, Nanning 530021, Guangxi Province, China
| | - Hai-Xing Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Province, China.
| |
Collapse
|
197
|
Feng D, Gao B. From basic liver immunology to therapeutic opportunities for liver diseases. Cell Mol Immunol 2021; 18:1-3. [PMID: 33339875 PMCID: PMC7852661 DOI: 10.1038/s41423-020-00607-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 11/27/2020] [Indexed: 02/06/2023] Open
Affiliation(s)
- Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
198
|
Cell therapy for advanced liver diseases: Repair or rebuild. J Hepatol 2021; 74:185-199. [PMID: 32976865 DOI: 10.1016/j.jhep.2020.09.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 08/18/2020] [Accepted: 09/14/2020] [Indexed: 12/15/2022]
Abstract
Advanced liver disease presents a significant worldwide health and economic burden and accounts for 3.5% of global mortality. When liver disease progresses to organ failure the only effective treatment is liver transplantation, which necessitates lifelong immunosuppression and carries associated risks. Furthermore, the shortage of suitable donor organs means patients may die waiting for a suitable transplant organ. Cell therapies have made their way from animal studies to a small number of early clinical trials. Herein, we review the current state of cell therapies for liver disease and the mechanisms underpinning their actions (to repair liver tissue or rebuild functional parenchyma). We also discuss cellular therapies that are on the clinical horizon and challenges that must be overcome before routine clinical use is a possibility.
Collapse
|
199
|
Pilling D, Karhadkar TR, Gomer RH. A CD209 ligand and a sialidase inhibitor differentially modulate adipose tissue and liver macrophage populations and steatosis in mice on the Methionine and Choline-Deficient (MCD) diet. PLoS One 2020; 15:e0244762. [PMID: 33378413 PMCID: PMC7773271 DOI: 10.1371/journal.pone.0244762] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is associated with obesity and type 2 diabetes and is characterized by the accumulation of fat in the liver (steatosis). NAFLD can transition into non-alcoholic steatohepatitis (NASH), with liver cell injury, inflammation, and an increased risk of fibrosis. We previously found that injections of either 1866, a synthetic ligand for the lectin receptor CD209, or DANA, a sialidase inhibitor, can inhibit inflammation and fibrosis in multiple animal models. The methionine and choline-deficient (MCD) diet is a model of NASH which results in the rapid induction of liver steatosis and inflammation. In this report, we show that for C57BL/6 mice on a MCD diet, injections of both 1866 and DANA reversed MCD diet-induced decreases in white fat, decreases in adipocyte size, and white fat inflammation. However, these effects were not observed in type 2 diabetic db/db mice on a MCD diet. In db/db mice on a MCD diet, 1866 decreased liver steatosis, but these effects were not observed in C57BL/6 mice. There was no correlation between the ability of 1866 or DANA to affect steatosis and the effects of these compounds on the density of liver macrophage cells expressing CLEC4F, CD64, F4/80, or Mac2. Together these results indicate that 1866 and DANA modulate adipocyte size and adipose tissue macrophage populations, that 1866 could be useful for modulating steatosis, and that changes in the local density of 4 different liver macrophages cell types do not correlate with effects on liver steatosis.
Collapse
Affiliation(s)
- Darrell Pilling
- Department of Biology, Texas A&M University, College Station, TX, United States of America
| | - Tejas R Karhadkar
- Department of Biology, Texas A&M University, College Station, TX, United States of America
| | - Richard H Gomer
- Department of Biology, Texas A&M University, College Station, TX, United States of America
| |
Collapse
|
200
|
Buberci R, Paydas S. Elevated Monocyte Count and Loss of Renal Function in Renal Transplant Patients. Transplant Proc 2020; 52:3080-3084. [DOI: 10.1016/j.transproceed.2020.02.176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/15/2020] [Accepted: 02/23/2020] [Indexed: 12/18/2022]
|