1
|
Feng X, Feng B, Zhou J, Yang J, Pan Q, Yu J, Shang D, Li L, Cao H. Mesenchymal stem cells alleviate mouse liver fibrosis by inhibiting pathogenic function of intrahepatic B cells. Hepatology 2025; 81:1211-1227. [PMID: 38546278 PMCID: PMC11902620 DOI: 10.1097/hep.0000000000000831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/09/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND AND AIMS The immunomodulatory characteristics of mesenchymal stem cells (MSCs) make them a promising therapeutic approach for liver fibrosis (LF). Here, we postulated that MSCs could potentially suppress the pro-fibrotic activity of intrahepatic B cells, thereby inhibiting LF progression. APPROACH AND RESULTS Administration of MSCs significantly ameliorated LF as indicated by reduced myofibroblast activation, collagen deposition, and inflammation. The treatment efficacy of MSCs can be attributed to decreased infiltration, activation, and pro-inflammatory cytokine production of intrahepatic B cells. Single-cell RNA sequencing revealed a distinct intrahepatic B cell atlas, and a subtype of naive B cells (B-II) was identified, which were markedly abundant in fibrotic liver, displaying mature features with elevated expression of several proliferative and inflammatory genes. Transcriptional profiling of total B cells revealed that intrahepatic B cells displayed activation, proliferation, and pro-inflammatory gene profile during LF. Fibrosis was attenuated in mice ablated with B cells (μMT) or in vivo treatment with anti-CD20. Moreover, fibrosis was recapitulated in μMT after adoptive transfer of B cells, which in turn could be rescued by MSC injection, validating the pathogenic function of B cells and the efficacy of MSCs on B cell-promoted LF progression. Mechanistically, MSCs could inhibit the proliferation and cytokine production of intrahepatic B cells through exosomes, regulating the Mitogen-activated protein kinase and Nuclear factor kappa B signaling pathways. CONCLUSIONS Intrahepatic B cells serve as a target of MSCs, play an important role in the process of MSC-induced amelioration of LF, and may provide new clues for revealing the novel mechanisms of MSC action.
Collapse
Affiliation(s)
- Xudong Feng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, China
| | - Bing Feng
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, China
| | - Jiahang Zhou
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, China
| | - Jinfeng Yang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, China
| | - Qiaoling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, China
| | - Jiong Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, China
| | - Dandan Shang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan City, China
| | - Lanjuan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan City, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou City, China
- National Medical Center for Infectious Diseases, Hangzhou City, China
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, China
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou City, China
- Key Laboratory of Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases of Zhejiang Province, Hangzhou City, China
| |
Collapse
|
2
|
Zheng L, Wu J, Hu H, Cao H, Xu N, Chen K, Wen B, Wang H, Yuan H, Xie L, Jiang Y, Li Z, Liang C, Yuan J, Li Z, Yuan X, Xiao W, Wang J. Single-cell RNA transcriptome landscape of murine liver following systemic administration of mesoporous silica nanoparticles. J Control Release 2023; 361:427-442. [PMID: 37487929 DOI: 10.1016/j.jconrel.2023.07.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/28/2023] [Accepted: 07/22/2023] [Indexed: 07/26/2023]
Abstract
Due to the unique physicochemical properties, mesoporous silica nanoparticles (MONs) have been widely utilized in biomedical fields for drug delivery, gene therapy, disease diagnosis and imaging. With the extensive applications and large-scale production of MONs, the potential effects of MONs on human health are gaining increased attention. To better understand the cellular and molecular mechanisms underlying the effects of MONs on the mouse liver, we profiled the transcriptome of 63,783 single cells from mouse livers following weekly intravenous administration of MONs for 2 weeks. The results showed that the proportion of endothelial cells and CD4+ T cells was increased, whereas that of Kupffer cells was decreased, in a dose-dependent manner after MONs treatment in the mouse liver. We also observed that the proportion of inflammation-related Kupffer cell subtype and wound healing-related hepatocyte subtype were elevated, but the number of hepatocytes with detoxification characteristics was reduced after MONs treatment. The cell-cell communication network revealed that there was more crosstalk between cholangiocytes and Kupffer cells, liver capsular macrophages, hepatic stellate cells, and endothelial cells following MONs treatment. Furthermore, we identified key ligand-receptor pairs between crucial subtypes after MONs treatment that are known to promote liver fibrosis. Collectively, our study explored the effects of MONs on mouse liver at a single-cell level and provides comprehensive information on the potential hepatotoxicity of MONs.
Collapse
Affiliation(s)
- Liuhai Zheng
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, Guangdong 510632, China
| | - Jiangpeng Wu
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Hong Hu
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Hua Cao
- Department of Oncology, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Nan Xu
- Division of Thyroid surgery, Department of General Surgery, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Kun Chen
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Bowen Wen
- College of Natural Resources and Environment, South China Agricultural University, 483 Wushan Road, Guangzhou, Guangdong 510642, China
| | - Huifang Wang
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, Guangdong 510632, China
| | - Haitao Yuan
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, Guangdong 510632, China
| | - Lulin Xie
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Yuke Jiang
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Zhifen Li
- School of Chemistry and Chemical Engineering, Shanxi Datong University, Xing Yun Street, Pingcheng District, Datong, Shanxi Province 037009, PR China
| | - Cailing Liang
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Jimin Yuan
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Zhijie Li
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China.
| | - Xiaopeng Yuan
- Department of Laboratory Medicine, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China.
| | - Wei Xiao
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China.
| | - Jigang Wang
- Department of Breast Surgery, Department of General Surgery, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China.
| |
Collapse
|
3
|
Biliary NIK promotes ductular reaction and liver injury and fibrosis in mice. Nat Commun 2022; 13:5111. [PMID: 36042192 PMCID: PMC9427946 DOI: 10.1038/s41467-022-32575-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
Abstract
Excessive cholangiocyte expansion (ductular reaction) promotes liver disease progression, but the underlying mechanism is poorly understood. Here we identify biliary NF-κB-inducing kinase (NIK) as a pivotal regulator of ductular reaction. NIK is known to activate the noncanonical IKKα/NF-κB2 pathway and regulate lymphoid tissue development. We find that cholangiocyte NIK is upregulated in mice with cholestasis induced by bile duct ligation (BDL), 5-diethoxycarbonyl-1,4-dihydrocollidine (DDC), or α-naphtyl-isothiocyanate (ANIT). DDC, ANIT, or BDL induces ductular reaction, liver injury, inflammation, and fibrosis in mice. Cholangiocyte-specific deletion of NIK, but not IKKα, blunts these pathological alterations. NIK inhibitor treatment similarly ameliorates DDC-induced ductular reaction, liver injury, and fibrosis. Biliary NIK directly increases cholangiocyte proliferation while suppressing cholangiocyte death, and it also promotes secretion of cholangiokines from cholangiocytes. Cholangiokines stimulate liver macrophages and hepatic stellate cells, augmenting liver inflammation and fibrosis. These results unveil a NIK/ductular reaction axis and a NIK/cholangiokine axis that promote liver disease progression. Excessive expansion of cholangiocytes in the liver leads to ductular reaction and liver disease. Here, the authors show that genetic ablation, or pharmacological inhibition, of biliary NIK blocks ductular reaction, liver inflammation, and liver fibrosis in mice by modulating secretion of cholangiokines that mediate liver inflammation and fibrosis.
Collapse
|
4
|
Tao L, Ren X, Zhai W, Chen Z. Progress and Prospects of Non-Canonical NF-κB Signaling Pathway in the Regulation of Liver Diseases. Molecules 2022; 27:molecules27134275. [PMID: 35807520 PMCID: PMC9268066 DOI: 10.3390/molecules27134275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/04/2023] Open
Abstract
Non-canonical nuclear factor kappa B (NF-κB) signaling pathway regulates many physiological and pathological processes, including liver homeostasis and diseases. Recent studies demonstrate that non-canonical NF-κB signaling pathway plays an essential role in hyperglycemia, non-alcoholic fatty liver disease, alcoholic liver disease, liver regeneration, liver injury, autoimmune liver disease, viral hepatitis, and hepatocellular carcinoma. Small-molecule inhibitors targeting to non-canonical NF-κB signaling pathway have been developed and shown promising results in the treatment of liver injuries. Here, the recent advances and future prospects in understanding the roles of the non-canonical NF-κB signaling pathways in the regulation of liver diseases are discussed.
Collapse
Affiliation(s)
- Li Tao
- Emergency Department, 305 Hospital of People’s Liberation Army, Beijing 100017, China; (L.T.); (W.Z.)
| | - Xiaomeng Ren
- College of Pharmaceutical and Biology Engineering, Shenyang University of Chemical Technology, Shenyang 110142, China
- Correspondence: (X.R.); (Z.C.); Tel.: +86-45186402029 (Z.C.)
| | - Wenhui Zhai
- Emergency Department, 305 Hospital of People’s Liberation Army, Beijing 100017, China; (L.T.); (W.Z.)
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
- Correspondence: (X.R.); (Z.C.); Tel.: +86-45186402029 (Z.C.)
| |
Collapse
|
5
|
Rui L, Lin JD. Reprogramming of Hepatic Metabolism and Microenvironment in Nonalcoholic Steatohepatitis. Annu Rev Nutr 2022; 42:91-113. [PMID: 35584814 PMCID: PMC10122183 DOI: 10.1146/annurev-nutr-062220-105200] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD), a spectrum of metabolic liver disease associated with obesity, ranges from relatively benign hepatic steatosis to nonalcoholic steatohepatitis (NASH). The latter is characterized by persistent liver injury, inflammation, and liver fibrosis, which collectively increase the risk for end-stage liver diseases such as cirrhosis and hepatocellular carcinoma. Recent work has shed new light on the pathophysiology of NAFLD/NASH, particularly the role of genetic, epigenetic, and dietary factors and metabolic dysfunctions in other tissues in driving excess hepatic fat accumulation and liver injury. In parallel, single-cell RNA sequencing studies have revealed unprecedented details of the molecular nature of liver cell heterogeneity, intrahepatic cross talk, and disease-associated reprogramming of the liver immune and stromal vascular microenvironment. This review covers the recent advances in these areas, the emerging concepts of NASH pathogenesis, and potential new therapeutic opportunities. Expected final online publication date for the Annual Review of Nutrition, Volume 42 is August 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Liangyou Rui
- Department of Molecular and Integrated Physiology and Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA;
| | - Jiandie D Lin
- Life Sciences Institute and Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA;
| |
Collapse
|
6
|
Tao X, Zhang R, Du R, Yu T, Yang H, Li J, Wang Y, Liu Q, Zuo S, Wang X, Lazarus M, Zhou L, Wang B, Yu Y, Shen Y. EP3 enhances adhesion and cytotoxicity of NK cells toward hepatic stellate cells in a murine liver fibrosis model. J Exp Med 2022; 219:213141. [PMID: 35420633 DOI: 10.1084/jem.20212414] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/07/2022] [Accepted: 03/09/2022] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells exhibit antifibrotic properties in liver fibrosis (LF) by suppressing activated hepatic stellate cell (HSC) populations. Prostaglandin E2 (PGE2) plays a dual role in innate and adaptive immunity. Here, we found that E-prostanoid 3 receptor (EP3) was markedly downregulated in NK cells from liver fibrosis mice and patients with liver cirrhosis. NK cell-specific deletion of EP3 aggravated hepatic fibrogenesis in mouse models of LF. Loss of EP3 selectively reduced the cytotoxicity of the CD27+CD11b+ double positive (DP) NK subset against activated HSCs. Mechanistically, deletion of EP3 impaired the adhesion and cytotoxicity of DP NK cells toward HSCs through modulation of Itga4-VCAM1 binding. EP3 upregulated Itga4 expression in NK cells through promoting Spic nuclear translocation via PKC-mediated phosphorylation of Spic at T191. Activation of EP3 by sulprostone alleviated CCL4-induced liver fibrosis in mice. Thus, EP3 is required for adhesion and cytotoxicity of NK cells toward HSCs and may serve as a therapeutic target for the management of LF.
Collapse
Affiliation(s)
- Xixi Tao
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Rui Zhang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ronglu Du
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Tingting Yu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hui Yang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Jiwen Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yuhong Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qian Liu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shengkai Zuo
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xi Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba City, Ibaraki, Japan
| | - Lu Zhou
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Ying Yu
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yujun Shen
- Department of Pharmacology, Tianjin Key Laboratory of Inflammatory Biology, Center for Cardiovascular Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
7
|
Targeted inhibition of the immunoproteasome blocks endothelial MHC class II antigen presentation to CD4 + T cells in chronic liver injury. Int Immunopharmacol 2022; 107:108639. [PMID: 35219165 DOI: 10.1016/j.intimp.2022.108639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/13/2022] [Accepted: 02/16/2022] [Indexed: 02/08/2023]
Abstract
Chronic or overwhelming liver injury is frequently associated with fibrosis, which is the main histological characteristic of non-alcoholic steatohepatitis (NASH). Currently, there is no effective treatment for liver fibrosis. Adaptive immunity is one of the perpetrators of liver inflammation and involves the antigen-specific activation of lymphocytes. Targeting adaptive immunity has been proposed as a novel therapeutic approach for NASH. In this study, we demonstrated that liver endothelial cells contribute to MHC class II (MHC-II) antigen presentation to CD4+ T cells after chronic liver injury. In human cirrhotic liver samples, we observed an increased expression of endothelial MHC-II and of the antigen presentation-associated protein LMP7, which is one of the proteolytically active subunits of the immunoproteasome. In a CCl4-induced chronic injury model or a diet- and chemical-induced NASH model, endothelial MHC-II and LMP7 expression was induced to increase. PR-957, a selective inhibitor of the immunoproteasome, inhibited MHC-II expression in endothelial cells and CD4+ T cell response after chronic liver injury. In vitro experiment demonstrated PR-957 also reversed IFN-γ-induced upregulation of MHC-II in endothelial cells. Furthermore, PR-957 treatment or CD4+ T cell depletion in chronic liver injury alleviated liver fibrosis and reduced inflammation, as indicated by the downregulation of inflammatory response markers (F4/80, IL-1, IL-6 and IL-18). In conclusion, targeted inhibition of the immunoproteasome blocks endothelial MHC-II antigen presentation to CD4+ T cells in chronic liver injury. In this regard, the PR-957 inhibitor is a promising candidate for the development of future therapies against NASH.
Collapse
|
8
|
Ding K, Li X, Ren X, Ding N, Tao L, Dong X, Chen Z. GBP5 promotes liver injury and inflammation by inducing hepatocyte apoptosis. FASEB J 2021; 36:e22119. [PMID: 34958688 DOI: 10.1096/fj.202101448r] [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: 09/15/2021] [Revised: 12/02/2021] [Accepted: 12/09/2021] [Indexed: 12/13/2022]
Abstract
Liver injury is the first step in causing fibrosis, cirrhosis, and liver cancer, leading to mortality. However, the drivers of progressive liver injury are still incompletely defined. Here, we identify GBP5 as a major factor causing liver injury and inflammation. We show that the expression of GBP5 is abnormally elevated in the damaged liver, and its expression depends at least partially on the NF-κB-inducing kinase (NIK)/NF-κB2 signaling pathway. Knockout of Gbp5 ameliorates D-galactosamine/lipopolysaccharide (GalN/LPS)-induced liver injury and inflammation. Conversely, liver-specific overexpression of GBP5 induces liver injury and inflammation. Mechanistically, GBP5 induces hepatocyte apoptosis through the activation of both calpain/caspase 12/caspase 3 and TNFα/caspase 8/caspase 3 signaling pathways. Inhibition of either calpain activity or caspase 3 prevents GBP5-induced cell death. Our data demonstrate that GBP5 expression is induced by toxins or the NIK signaling pathway, which promotes both extrinsic and intrinsic apoptosis signaling pathways and further induces liver injury, providing a novel drug target for the treatment of liver injury and inflammation.
Collapse
Affiliation(s)
- Kaixin Ding
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xinzhi Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xiaomeng Ren
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, China.,Shenyang University of Chemical Technology, Shenyang, China
| | - Na Ding
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Li Tao
- 305 Hospital of People's Liberation Army, Beijing, China
| | - Xue Dong
- Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, China
| | - Zheng Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
9
|
Zhong X, Zhang Z, Shen H, Xiong Y, Shah YM, Liu Y, Fan X, Rui L. Hepatic NF-κB-Inducing Kinase and Inhibitor of NF-κB Kinase Subunit α Promote Liver Oxidative Stress, Ferroptosis, and Liver Injury. Hepatol Commun 2021; 5:1704-1720. [PMID: 34558831 PMCID: PMC8485893 DOI: 10.1002/hep4.1757] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/20/2021] [Accepted: 05/11/2021] [Indexed: 12/14/2022] Open
Abstract
Drug-induced hepatotoxicity limits development of new effective medications. Drugs and numerous endogenous/exogenous agents are metabolized/detoxified by hepatocytes, during which reactive oxygen species (ROS) are generated as a by-product. ROS has broad adverse effects on liver function and integrity, including damaging hepatocyte proteins, lipids, and DNA and promoting liver inflammation and fibrosis. ROS in concert with iron overload drives ferroptosis. Hepatic nuclear factor kappa B (NF-κB)-inducing kinase (NIK) is aberrantly activated in a broad spectrum of liver disease. NIK phosphorylates and activates inhibitor of NF-κB kinase subunit alpha (IKKα), and the hepatic NIK/IKKα cascade suppresses liver regeneration. However, the NIK/IKKα pathway has not been explored in drug-induced liver injury. Here, we identify hepatic NIK as a previously unrecognized mediator for acetaminophen (APAP)-induced acute liver failure. APAP treatment increased both NIK transcription and NIK protein stability in primary hepatocytes as well as in liver in mice. Hepatocyte-specific overexpression of NIK augmented APAP-induced liver oxidative stress in mice and increased hepatocyte death and mortality in a ROS-dependent manner. Conversely, hepatocyte-specific ablation of NIK or IKKα mitigated APAP-elicited hepatotoxicity and mortality. NIK increased lipid peroxidation and cell death in APAP-stimulated primary hepatocytes. Pretreatment with antioxidants or ferroptosis inhibitors blocked NIK/APAP-induced hepatocyte death. Conclusion: We unravel a previously unrecognized NIK/IKKα/ROS/ferroptosis axis engaged in liver disease progression.
Collapse
Affiliation(s)
- Xiao Zhong
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
- Department of Infectious DiseasesHunan Key Laboratory of Viral HepatitisXiangya HospitalCentral South UniversityChangshaChina
| | - Zhiguo Zhang
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Hong Shen
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Yi Xiong
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Yatrik M. Shah
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
| | - Yong Liu
- College of Life SciencesInstitute for Advanced StudiesWuhan UniversityWuhanChina
| | - Xue‐Gong Fan
- Department of Infectious DiseasesHunan Key Laboratory of Viral HepatitisXiangya HospitalCentral South UniversityChangshaChina
| | - Liangyou Rui
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborMIUSA
- Division of Gastroenterology and HepatologyDepartment of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborMIUSA
| |
Collapse
|
10
|
Halkowycz P, Grimshaw CE, Keung W, Tanis P, Proffitt C, Peacock K, de Jong R, Sabat M, Banerjee U, Ermolieff J. Biochemical and Cellular Profile of NIK Inhibitors with Long Residence Times. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2021; 26:676-683. [PMID: 33084478 DOI: 10.1177/2472555220964450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Two different signaling pathways lead to the activation of the transcription factor NF-κB, initiating distinct biological responses: The canonical NF-κB pathway activation has been implicated in host immunity and inflammatory responses, whereas the noncanonical pathway activation has been involved in lymphoid organ development and B-cell maturation, as well as in the development of chronic inflammatory diseases and some hematologic cancers. The NF-κB-inducing kinase (NIK) is a cytoplasmic Ser/Thr kinase and is a key regulator of the noncanonical pathway. NIK activation results in the processing of the p100 subunit to p52, leading to the formation of the RelB/p52 complex and noncanonical pathway activation. Because of its role in the development of lymphoid malignancies, this kinase has always been considered as an attractive target for the treatment of certain types of cancers and immune diseases. We at Takeda have pursued a drug discovery program to identify small-molecule inhibitors against NIK. This report provides an overview of the data generated from our screening campaign using a small fragment library. Most importantly, we also provide a kinetic analysis of published compounds and chemical series developed at Takeda that are associated with a slow tight-binding mechanism and excellent cellular potency.
Collapse
Affiliation(s)
- Petro Halkowycz
- Medicinal Chemistry-In Vitro Pharmacology Gastrointestinal, Takeda Pharmaceutical, San Diego, CA, USA
| | | | | | - Paul Tanis
- Medicinal Chemistry CNS, Takeda Pharmaceutical, San Diego, CA, USA
| | - Chris Proffitt
- Gastrointestinal-Immunology, Takeda Pharmaceutical, San Diego, CA, USA
| | | | - Ron de Jong
- Ron de Jong Consulting, LLC, San Diego, CA, USA
| | - Mark Sabat
- Medicinal Chemistry-In Vitro Pharmacology Gastrointestinal, Takeda Pharmaceutical, San Diego, CA, USA
| | - Urmi Banerjee
- CNS-In Vitro Pharmacology, Takeda Pharmaceutical, San Diego, CA, USA
| | - Jacques Ermolieff
- Medicinal Chemistry-In Vitro Pharmacology Gastrointestinal, Takeda Pharmaceutical, San Diego, CA, USA
| |
Collapse
|
11
|
Fas/FasL mediates NF-κBp65/PUMA-modulated hepatocytes apoptosis via autophagy to drive liver fibrosis. Cell Death Dis 2021; 12:474. [PMID: 33980818 PMCID: PMC8115181 DOI: 10.1038/s41419-021-03749-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/30/2022]
Abstract
Fas/Fas ligand (FasL)-mediated cell apoptosis involves a variety of physiological and pathological processes including chronic hepatic diseases, and hepatocytes apoptosis contributes to the development of liver fibrosis following various causes. However, the mechanism of the Fas/FasL signaling and hepatocytes apoptosis in liver fibrogenesis remains unclear. The Fas/FasL signaling and hepatocytes apoptosis in liver samples from both human sections and mouse models were investigated. NF-κBp65 wild-type mice (p65f/f), hepatocytes specific NF-κBp65 deletion mice (p65Δhepa), p53-upregulated modulator of apoptosis (PUMA) wild-type (PUMA-WT) and PUMA knockout (PUMA-KO) littermate models, and primary hepatic stellate cells (HSCs) were also used. The mechanism underlying Fas/FasL-regulated hepatocytes apoptosis to drive HSCs activation in fibrosis was further analyzed. We found Fas/FasL promoted PUMA-mediated hepatocytes apoptosis via regulating autophagy signaling and NF-κBp65 phosphorylation, while inhibition of autophagy or PUMA deficiency attenuated Fas/FasL-modulated hepatocytes apoptosis and liver fibrosis. Furthermore, NF-κBp65 in hepatocytes repressed PUMA-mediated hepatocytes apoptosis via regulating the Bcl-2 family, while NF-κBp65 deficiency in hepatocytes promoted PUMA-mediated hepatocytes apoptosis and enhanced apoptosis-linked inflammatory response, which contributed to the activation of HSCs and liver fibrogenesis. These results suggest that Fas/FasL contributes to NF-κBp65/PUMA-modulated hepatocytes apoptosis via autophagy to enhance liver fibrogenesis, and this network could be a potential therapeutic target for liver fibrosis.
Collapse
|
12
|
Xiao W, He Z, Luo W, Feng D, Wang Y, Tang T, Yang A, Luo J. BYHWD Alleviates Inflammatory Response by NIK-Mediated Repression of the Noncanonical NF-κB Pathway During ICH Recovery. Front Pharmacol 2021; 12:632407. [PMID: 34025405 PMCID: PMC8138445 DOI: 10.3389/fphar.2021.632407] [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: 11/23/2020] [Accepted: 04/19/2021] [Indexed: 01/24/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is a life-threatening type of stroke that lacks effective treatments. The inflammatory response following ICH is a vital response that affects brain repair and organism recovery. The nuclear factor κB (NF-κB) signaling pathway is considered one of the most important inflammatory response pathways and one of its response pathways, the noncanonical NF-κB signaling pathway, is known to be associated with persistent effect and chronic inflammation. NF-κB–inducing kinase (NIK) via the noncanonical NF-κB signaling plays a key role in controlling inflammation. Here, we investigated potential effects of the traditional Chinese medicine formula Buyang Huanwu Decoction (BYHWD) on inflammatory response in a rat model of ICH recovery by inhibiting the NIK-mediated the noncanonical NF-κB signaling pathway. In the first part, rats were randomly divided into three groups: the sham group, the ICH group, and the BYHWD group. ICH was induced in rats by injecting collagenase (type VII) into the right globus pallidus of rats' brain. For the BYHWD group, rats were administered BYHWD (4.36 g/kg) once a day by intragastric administration until they were sacrificed. Neurological function was evaluated in rats by a modified neurological severity score (mNSS), the corner turn test, and the foot-fault test. The cerebral edema showed the degree of inflammatory response by sacrificed brain water content. Western blot and real-time quantitative reverse transcription PCR tested the activity of inflammatory response and noncanonical NF-κB signaling. In the second part, siRNA treatment and assessment of inflammation level as well as alterations in the noncanonical NF-κB signaling were performed to determine whether the effect of BYHWD on inflammatory response was mediated by suppression of NIK via the noncanonical NF-κB signaling pathway. We show that BYHWD treated rats exhibited: (i) better health conditions and better neural functional recovery; (ii) decreased inflammatory cytokine and the edema; (iii) reduced expression of NIK, a key protein in unregulated the noncanonical NF-κB signaling pathways; (iv) when compared with pretreated rats with NIK targeting (NIK siRNAs), showed the same effect of inhibiting the pathway and decreased inflammatory cytokine. BYHWD can attenuate the inflammatory response during ICH recovery in rats by inhibiting the NIK-mediated noncanonical NF-κB signaling pathway.
Collapse
Affiliation(s)
- Wei Xiao
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Zehui He
- Department of Orthopedics, Movement System Injury and Repair Research Center, Xiangya Hospital, Central South University, Changsha, China
| | - Weikang Luo
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Dandan Feng
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Wang
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Tao Tang
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Ali Yang
- Department of Neurology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiekun Luo
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative Medicine, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
13
|
Chen Q, Lu X, Zhang X. Noncanonical NF-κB Signaling Pathway in Liver Diseases. J Clin Transl Hepatol 2021; 9:81-89. [PMID: 33604258 PMCID: PMC7868705 DOI: 10.14218/jcth.2020.00063] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/04/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
The noncanonical NF-κB signaling pathway is an important branch of NF-κB signaling. It is involved in regulating multiple important biological processes, including inflammation and host immune response. A central adaptor protein of the noncanonical NF-κB pathway is NF-κB-inducing kinase (NIK), which activates the downstream kinase IKKα to process p100 to p52, thereby forming the RelB/p52 heterodimer to initiate the expression of target genes. Currently, many specific inhibitors and monoclonal antibodies targeting or triggering this pathway are being developed and tested for various diseases, including cancers, autoimmune diseases, and virus infection. Given that aberrant activation of the noncanonical NF-κB pathway is frequently observed in various liver diseases, targeting this pathway may be a promising therapeutic strategy to alleviate liver inflammation. Moreover, activation of this pathway may contribute to the antiviral immune response and promote the clearance of persistent hepatotropic virus infection. Here, we review the role of the noncanonical NF-κB pathway in the occurrence and development of different liver diseases, and discuss the potency and application of modulating the noncanonical NF-κB pathway for treatment of these liver diseases.
Collapse
Affiliation(s)
- Qianhui Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou,Guangdong, China
| | - Xinyu Lu
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou,Guangdong, China
| | - Xiaoyong Zhang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou,Guangdong, China
- Hepatology Unit, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
- Correspondence to: Xiaoyong Zhang, Hepatology Unit and Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Guangzhou, Guangdong 510515, China. Tel: +86-20-62787830, E-mail:
| |
Collapse
|
14
|
Wang Y, Yang Z, Wei Y, Li X, Li S. Apolipoprotein A4 regulates the immune response in carbon tetrachloride-induced chronic liver injury in mice. Int Immunopharmacol 2021; 90:107222. [PMID: 33276196 DOI: 10.1016/j.intimp.2020.107222] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/02/2020] [Accepted: 11/16/2020] [Indexed: 12/19/2022]
Abstract
This article explores the role of ApoA4 in a CCl4-induced chronic liver injury (CLI) mouse model. C57BL/6J mice (WT) and ApoA4 knock-out (KO) mice were divided into CCl4 CLI (WT-CCl4 and KO-CCl4) and olive oil solvent control groups (WT-Veh and KO-Veh). Some of the KO-CCl4 mice were additionally treated with recombinant mouse ApoA4 and primary mouse T lymphocyte injections. After 6 weeks, histological analyses, biochemical and superoxide dismutase (SOD) and malondialdehyde (MDA) assays, flow cytometry of immune cells and qRT-PCR analyses were performed. KO mice after treatment with CCl4 showed reduced hepatic SOD and enhanced serum MDA activities leading to worsening liver injury and fibrosis compared with WT-CCl4, accompanied by enhanced hepatic alpha smooth muscle actin (α-SMA), tissue inhibitor of metalloproteinases-1 (TIMP-1) and collagen type I alpha 1 chain (COL1A1) transcriptions, elevated macrophage M1 levels, enhanced tumor necrosis factor-alpha (TNF-α), Interleukin 6 (IL-6) and C-C Motif Chemokine Ligand 5 (CCL5), but reduced Interleukin 10 (IL-10), monocyte chemotactic protein 1 (MCP-1), C-C Motif Chemokine Receptor 2 (CCR2), C-X3-C Motif Chemokine Receptor 1 (CX3CR1) and C-X-C Motif Chemokine Ligand 9 (CXCL9) transcription, as well as reduced CD3+, CD4+ and CD8+ T cell percentages in hepatic tissue, blood cells and spleen. In addition, CD11b+CD115+, CD11b+/Ly6Chigh, CD11b+/LyC6- and CD11b+/Ly6Cint cells were enhanced, which partly reversed by ApoA4 protein and T cell injections. In conclusion, we propose that ApoA4 might be involved in liver protection via inhibiting fibrotic mediators and inflammatory cytokines, suppression of pro-inflammatory hepatic M1 cell invasion and regulation of CD8+ T and CD4+ T lymphocytes.
Collapse
Affiliation(s)
- Yinan Wang
- Bio-evidence Science Academy (BSA), Xi'an Jiaotong University (XJTU), No. 205 Zhuque Street, Xi'an 710061, China; Key laboratory of Ministry of Public Health for Forensic Sciences, No. 205 Zhuque Street, Xi'an 710061, China
| | - Ziyu Yang
- Bio-evidence Science Academy (BSA), Xi'an Jiaotong University (XJTU), No. 205 Zhuque Street, Xi'an 710061, China; Key laboratory of Ministry of Public Health for Forensic Sciences, No. 205 Zhuque Street, Xi'an 710061, China
| | - Yang Wei
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, Institute of Digestive Diseases. The Second Affiliated Hospital, Xi'an Jiaotong University, No. 157 West 5th Road, Xi'an 710004, China
| | - Xiaoming Li
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medical Institute, Institute of Digestive Diseases. The Second Affiliated Hospital, Xi'an Jiaotong University, No. 157 West 5th Road, Xi'an 710004, China.
| | - Shengbin Li
- Bio-evidence Science Academy (BSA), Xi'an Jiaotong University (XJTU), No. 205 Zhuque Street, Xi'an 710061, China; Key laboratory of Ministry of Public Health for Forensic Sciences, No. 205 Zhuque Street, Xi'an 710061, China.
| |
Collapse
|
15
|
Liang Q, Hu Y, Zhang M, Lin C, Zhang W, Li Y, Zhu P, Xue P, Chen Y, Li Q, Wang K. The T Cell Receptor Immune Repertoire Protects the Liver From Reconsitution. Front Immunol 2020; 11:584979. [PMID: 33391261 PMCID: PMC7775400 DOI: 10.3389/fimmu.2020.584979] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
Aberrant immune cell infiltrates and microcircumstances represent characteristic features of liver fibrosis. In this study, we profiled the transcriptomes of intrahepatic CD45+ immune cells, from mice, using single-cell RNA sequencing (scRNA-seq) technology to understand the landscape of intrahepatic immune cells during the pathogenesis of fibrosis. Analysis of approximately 10,000 single-cell transcriptomes revealed an increase in dendritic cells (DCs), macrophages, and neutrophils and a decrease in T and natural killer T (NKT) cells. In addition, we report changes in the transcriptomes of diverse immune cell types, implying a deteriorating intrahepatic immune microcircumstance. Furthermore, we uncovered a novel fibrosis-associated CD8 T (Ccl5+, Ccl4+) and CD4 T (mt-Co1+) cell subpopulation, which infiltrates fibrotic liver and is characterized by abnormal activation or inactivation as well as a TCR decline. The results from scRNA-seq and bulk immune repertoire sequencing (IR-seq) revealed an obvious decline in T cell receptor (TCR) clonotypes combined with shrinking VJ and VDJ segment usage, as well as lower complementarity-determining region 3 (CDR3) amino acid (AA) diversity from fibrotic liver. Interestingly, a deficiency of TCR IR (TcrbKO mice) led to a deterioration of liver fibrosis, coupled with activation of hepatic stellate cells (HSCs) induced by the upregulation of macrophage and γδ T cell distribution in fibrotic TcrbKO livers. Our findings reveal the landscape and dynamics of single immune cells in liver fibrosis, and clarify the protective role of TCR IR in response to chronic liver injury.
Collapse
Affiliation(s)
- Qing Liang
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Yudi Hu
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Meina Zhang
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Chunjie Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, China
| | - Wei Zhang
- Department of Pathology, The 971 Hospital of People's Liberation Army Navy, Qingdao, China
| | - Ying Li
- Department of Pathology, Qingdao Municipal Hospital, Qingdao, China
| | - Ping Zhu
- Department of Gynaecology and Obstetrics, The 971 Hospital of People's Liberation Army Navy, Qingdao, China
| | - Pengxin Xue
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Yujie Chen
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Qiyuan Li
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen, China
| | - Kejia Wang
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
16
|
β-arrestin2 deficiency protects against hepatic fibrosis in mice and prevents synthesis of extracellular matrix. Cell Death Dis 2020; 11:389. [PMID: 32439968 PMCID: PMC7242363 DOI: 10.1038/s41419-020-2596-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 11/17/2022]
Abstract
Hepatic fibrosis is a disease of the wound-healing response following chronic liver injury, and activated hepatic stellate cells (HSCs) play a crucial role in the progression of hepatic fibrosis. β-arrestin2 functions as a multiprotein scaffold to coordinate complex signal transduction networks. Although β-arrestin2 transduces diverse signals in cells, little is known about its involvement in the regulation of liver fibrosis. Our current study utilized a porcine serum-induced liver fibrosis model and found increased expression of β-arrestin2 in hepatic tissues with the progression of hepatic fibrosis, which was positively correlated with collagen levels. Furthermore, changes in human fibrotic samples were also observed. We next used β-arrestin2−/− mice to demonstrate that β-arrestin2 deficiency ameliorates CCl4-induced liver fibrosis and decreases collagen deposition. The in vitro depletion and overexpression experiments showed that decreased β-arrestin2 inhibited HSCs collagen production and elevated TβRIII expression, thus downregulating the TGF-β1 pathway components Smad2, Smad3 and Akt. These findings suggest that β-arrestin2 deficiency ameliorates liver fibrosis in mice, and β-arrestin2 may be a potential treatment target in hepatic fibrosis.
Collapse
|
17
|
Ge W, Gao Y, Zhao Y, Yang Y, Sun Q, Yang X, Xu X, Zhang J. Decreased T-cell mediated hepatic injury in concanavalin A-treated PLRP2-deficient mice. Int Immunopharmacol 2020; 85:106604. [PMID: 32428799 DOI: 10.1016/j.intimp.2020.106604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022]
Abstract
Concanavalin A (Con A) activates innate immunity and causes liver damage mediated by cytotoxic T lymphocytes (CTL) in mice. The Pancreatic lipase-related protein 2 (PLRP2) is induced by interleukin (IL)-4 in vitro in CTLs and associated with CTL functions. We examined the role of PLRP2 in a mouse model of Con A-induced T cell-mediated hepatitis. PLRP2-knockout and wild-type (WT) mice were inoculated with 20 mg/kg Con A. Mice lacking PLRP2 reduced Con A-induced hepatitis, which was manifested by a decrease in serum aminotransferase and histopathological assessment. The expression and secretion of cytokines including tumor necrosis factor-alpha (TNF-α), interferon (IFN)-γ, IL-6, and IL-1β were suppressed in Con A-treated PLRP2-knockout mice. In PLRP2 knockout mice, Con A-induced liver chemokines and adhesion molecules (such as MIP-1α, MIP-1β, ICAM-1 and MCP-1) were also down regulated. In the WT liver treated with Con A, the number of T cells (CD4+ and CD8+) and macrophages (CD11b+ F4/80+) increased significantly, while the lack of PLRP2 reduced the number of T cells in the liver, but had no effect on macrophages. The shift of the metabolic profiles was impaired in Con A-treated PLRP2-knockout mice compared to WT mice. In conclusion, these results indicate that PLRP2 deficiency reduces T-cell mediated Con A-induced hepatitis, and suggest PLRP2 is a potential target of anti-inflammatory and immunomodulatory drugs to treat immune-mediated hepatitis.
Collapse
Affiliation(s)
- Wenhao Ge
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Yan Gao
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Yang Zhao
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Yunxia Yang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Qi Sun
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Xiao Yang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Xi Xu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China
| | - Jianfa Zhang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing, China.
| |
Collapse
|
18
|
Shen H, Ji Y, Xiong Y, Kim H, Zhong X, Jin MG, Shah YM, Omary MB, Liu Y, Qi L, Rui L. Medullary thymic epithelial NF-kB-inducing kinase (NIK)/IKKα pathway shapes autoimmunity and liver and lung homeostasis in mice. Proc Natl Acad Sci U S A 2019; 116:19090-19097. [PMID: 31481626 PMCID: PMC6754592 DOI: 10.1073/pnas.1901056116] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Aberrant T cell development is a pivotal risk factor for autoimmune disease; however, the underlying molecular mechanism of T cell overactivation is poorly understood. Here, we identified NF-κB-inducing kinase (NIK) and IkB kinase α (IKKα) in thymic epithelial cells (TECs) as essential regulators of T cell development. Mouse TEC-specific ablation of either NIK or IKKα resulted in severe T cell-mediated inflammation, injury, and fibrosis in the liver and lung, leading to premature death within 18 d of age. NIK or IKKα deficiency abrogated medullary TEC development, and led to breakdown of central tolerance, production of autoreactive T cells, and fatal autoimmune destruction in the liver and lung. TEC-specific ablation of NIK or IKKα also impaired thymic T cell development from the double-negative through the double-positive stages and inhibited peripheral B cell development. These results unravel a hitherto unrecognized essential role of TEC-intrinsic NIK and IKKα pathways in autoimmunity and T cell-instigated chronic liver and lung diseases.
Collapse
Affiliation(s)
- Hong Shen
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Yewei Ji
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Yi Xiong
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Hana Kim
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Xiao Zhong
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Michelle G Jin
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - M Bishr Omary
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Yong Liu
- College of Life Sciences, The Institute for Advanced Studies, Wuhan University, 430072 Wuhan, China
| | - Ling Qi
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Liangyou Rui
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109;
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
19
|
Pan Z, Tian Y, Cao C, Niu G. The Emerging Role of YAP/TAZ in Tumor Immunity. Mol Cancer Res 2019; 17:1777-1786. [PMID: 31308148 DOI: 10.1158/1541-7786.mcr-19-0375] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/03/2019] [Accepted: 07/10/2019] [Indexed: 11/16/2022]
Abstract
Yes-associated protein (YAP)/WW domain-containing transcription regulator 1 (TAZ) is an important transcriptional regulator and effector of the Hippo signaling pathway that has emerged as a critical determinant of malignancy in many human tumors. YAP/TAZ expression regulates the cross-talk between immune cells and tumor cells in the tumor microenvironment through its influence on T cells, myeloid-derived suppressor cells, and macrophages. However, the mechanisms underlying these effects are poorly understood. An improved understanding of the role of YAP/TAZ in tumor immunity is essential for exploring innovative tumor treatments and making further breakthroughs in antitumor immunotherapy. This review primarily focuses on the role of YAP/TAZ in immune cells, their interactions with tumor cells, and how this impacts on tumorigenesis, progression, and therapy resistance.
Collapse
Affiliation(s)
- Zhaoji Pan
- Xuzhou Central Hospital, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou, Jiangsu, P.R. China
| | - Yiqing Tian
- Xinyi People's Hospital, Xinyi, Xuzhou, Jiangsu, P.R. China.
| | - Chengsong Cao
- Xuzhou Central Hospital, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou, Jiangsu, P.R. China
| | - Guoping Niu
- Xuzhou Central Hospital, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou, Jiangsu, P.R. China
| |
Collapse
|
20
|
Valiño-Rivas L, Vaquero JJ, Sucunza D, Gutierrez S, Sanz AB, Fresno M, Ortiz A, Sanchez-Niño MD. NIK as a Druggable Mediator of Tissue Injury. Trends Mol Med 2019; 25:341-360. [PMID: 30926358 DOI: 10.1016/j.molmed.2019.02.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/07/2019] [Accepted: 02/13/2019] [Indexed: 12/15/2022]
Abstract
NF-κB-inducing kinase (NIK, MAP3K14) is best known as the apical kinase that triggers non-canonical NF-κB activation and by its role in the immune system. Recent data indicate a role for NIK expressed by non-lymphoid cells in cancer, kidney disease, liver injury, glucose homeostasis, osteosarcopenia, vascular calcification, hematopoiesis, and endothelial function. The spectrum of NIK-associated disease now ranges from immunodeficiency (when NIK is defective) to autoimmunity, cancer, sterile inflammation, fibrosis, and metabolic disease when NIK is overactive. The development of novel small-molecule NIK inhibitors has paved the way to test NIK targeting to treat disease in vivo, and may eventually lead to NIK targeting in the clinic. In addition, NIK activators are being explored for specific conditions such as myeloid leukemia.
Collapse
Affiliation(s)
- Lara Valiño-Rivas
- Department of Nephrology and Hypertension, Instituto de Investigación Sanitaria (IIS) Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid (UAM), Red de Investigación Renal (REDINREN), and Fundación Renal Íñigo Álvarez de Toledo (FRIAT), Madrid, Spain
| | - Juan José Vaquero
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcala and REDINREN, Madrid, Spain
| | - David Sucunza
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcala and REDINREN, Madrid, Spain
| | - Sara Gutierrez
- Departamento de Química Orgánica y Química Inorgánica, Universidad de Alcala and REDINREN, Madrid, Spain
| | - Ana B Sanz
- Department of Nephrology and Hypertension, Instituto de Investigación Sanitaria (IIS) Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid (UAM), Red de Investigación Renal (REDINREN), and Fundación Renal Íñigo Álvarez de Toledo (FRIAT), Madrid, Spain
| | - Manuel Fresno
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas de la UAM, Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, Instituto de Investigación Sanitaria (IIS) Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid (UAM), Red de Investigación Renal (REDINREN), and Fundación Renal Íñigo Álvarez de Toledo (FRIAT), Madrid, Spain; These authors contributed equally.
| | - Maria Dolores Sanchez-Niño
- Department of Nephrology and Hypertension, Instituto de Investigación Sanitaria (IIS) Fundación Jiménez Díaz, School of Medicine, Universidad Autónoma de Madrid (UAM), Red de Investigación Renal (REDINREN), and Fundación Renal Íñigo Álvarez de Toledo (FRIAT), Madrid, Spain; These authors contributed equally.
| |
Collapse
|
21
|
Ramakrishnan SK, Zhang H, Ma X, Jung I, Schwartz AJ, Triner D, Devenport SN, Das NK, Xue X, Zeng MY, Hu Y, Mortensen RM, Greenson JK, Cascalho M, Wobus CE, Colacino JA, Nunez G, Rui L, Shah YM. Intestinal non-canonical NFκB signaling shapes the local and systemic immune response. Nat Commun 2019; 10:660. [PMID: 30737385 PMCID: PMC6368617 DOI: 10.1038/s41467-019-08581-8] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 01/21/2019] [Indexed: 12/13/2022] Open
Abstract
Microfold cells (M-cells) are specialized cells of the intestine that sample luminal microbiota and dietary antigens to educate the immune cells of the intestinal lymphoid follicles. The function of M-cells in systemic inflammatory responses are still unclear. Here we show that epithelial non-canonical NFkB signaling mediated by NFkB-inducing kinase (NIK) is highly active in intestinal lymphoid follicles, and is required for M-cell maintenance. Intestinal NIK signaling modulates M-cell differentiation and elicits both local and systemic IL-17A and IgA production. Importantly, intestinal NIK signaling is active in mouse models of colitis and patients with inflammatory bowel diseases; meanwhile, constitutive NIK signaling increases the susceptibility to inflammatory injury by inducing ectopic M-cell differentiation and a chronic increase of IL-17A. Our work thus defines an important function of non-canonical NFkB and M-cells in immune homeostasis, inflammation and polymicrobial sepsis.
Collapse
Affiliation(s)
| | - Huabing Zhang
- Department of Molecular & Integrative Physiology, University of Michigan, Michigan, MI, 48109, USA
| | - Xiaoya Ma
- Department of Molecular & Integrative Physiology, University of Michigan, Michigan, MI, 48109, USA
| | - Inkyung Jung
- Department of Molecular & Integrative Physiology, University of Michigan, Michigan, MI, 48109, USA
| | - Andrew J Schwartz
- Department of Molecular & Integrative Physiology, University of Michigan, Michigan, MI, 48109, USA
| | - Daniel Triner
- Department of Molecular & Integrative Physiology, University of Michigan, Michigan, MI, 48109, USA
| | - Samantha N Devenport
- Department of Molecular & Integrative Physiology, University of Michigan, Michigan, MI, 48109, USA
| | - Nupur K Das
- Department of Molecular & Integrative Physiology, University of Michigan, Michigan, MI, 48109, USA
| | - Xiang Xue
- Department of Molecular & Integrative Physiology, University of Michigan, Michigan, MI, 48109, USA
| | - Melody Y Zeng
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
- Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yinling Hu
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Richard M Mortensen
- Department of Molecular & Integrative Physiology, University of Michigan, Michigan, MI, 48109, USA
| | - Joel K Greenson
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Marilia Cascalho
- Transplantation Biology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Christiane E Wobus
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Justin A Colacino
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Gabriel Nunez
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
- Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Liangyou Rui
- Department of Molecular & Integrative Physiology, University of Michigan, Michigan, MI, 48109, USA
- Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yatrik M Shah
- Department of Molecular & Integrative Physiology, University of Michigan, Michigan, MI, 48109, USA.
- Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
22
|
Ronis MJ, Gomez-Acevedo H, Shankar K, Sharma N, Blackburn M, Singhal R, Mercer KE, Badger TM. EB 2017 Article: Soy protein isolate feeding does not result in reproductive toxicity in the pre-pubertal rat testis. Exp Biol Med (Maywood) 2019; 243:695-707. [PMID: 29763383 DOI: 10.1177/1535370218771333] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The isoflavone phytoestrogens found in the soy protein isolate used in soy infant formulas have been shown to have estrogenic actions in the developing male reproductive tract resulting in reproductive toxicity. However, few studies have examined potential estrogenicity of soy protein isolate as opposed to that of pure isoflavones. In this study, we fed weanling male Sprague-Dawley rats a semi-purified diet with casein or soy protein isolate as the sole protein source from postnatal day 21 to 33. Additional groups were fed casein or soy protein isolate and treated s.c. with 10 µg/kg/d estradiol via osmotic minipump. Estradiol treatment reduced testis, prostate weights, and serum androgen concentrations ( P < 0.05). Soy protein isolate had no effect. Estradiol up-regulated 489 and down-regulated 1237 testicular genes >1.5-fold ( P < 0.05). In contrast, soy protein isolate only significantly up-regulated expression of 162 genes and down-regulated 16 genes. The top 30 soy protein isolate-up-regulated genes shared 93% concordance with estradiol up-regulated genes. There was little overlap between soy protein isolate down-regulated genes and those down-regulated by estradiol treatment. Functional annotation analysis revealed significant differences in testicular biological processes affected by estradiol or soy protein isolate. Estradiol had major actions on genes involved in reproductive processes including down-regulation of testicular steroid synthesis and expression of steroid receptor activated receptor (Star) and cytochrome P450 17α-hydroxylase/(Cyp17a1). In contrast, soy protein isolate primarily affected pathways associated with macromolecule modifications including ubiquitination and histone methylation. Our results indicate that rather than acting as a weak estrogen in the developing testis, soy protein isolate appears to act as a selective estrogen receptor modulator with little effect on reproductive processes. Impact statement Soy protein isolate (SPI) is the sole protein used to make soy-based infant formulas. SPI contains phytoestrogens, which are structurally similar to estradiol. These phytoestrogens, daidzein, genistein, and equol, fit the definition of endocrine-disrupting compounds, and at high concentrations, have estrogenic actions resulting in reproductive toxicity in the developing male, when provided as isolated chemicals. However, few animal studies have examined the potential estrogenicity of SPI as opposed to pure isoflavones. In this study, SPI feeding did not elicit an estrogenic response in the testis nor any adverse outcomes including reduced testicular growth, or androgen production during early development in rats when compared to those receiving estradiol. These findings are consistent with emerging data showing no differences in reproductive development in males and female children that received breast milk, cow's milk formula, or soy infant formula during the postnatal feeding period.
Collapse
Affiliation(s)
- Martin Jj Ronis
- 1 Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center - New Orleans, LA 70112, USA
| | - Horacio Gomez-Acevedo
- 2 Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Kartik Shankar
- 3 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,4 Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
| | - Neha Sharma
- 4 Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
| | | | - Rohit Singhal
- 4 Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
| | - Kelly E Mercer
- 3 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,4 Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
| | - Thomas M Badger
- 3 Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.,4 Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
| |
Collapse
|
23
|
Xiong Y, Torsoni AS, Wu F, Shen H, Liu Y, Zhong X, Canet MJ, Shah YM, Omary MB, Liu Y, Rui L. Hepatic NF-kB-inducing kinase (NIK) suppresses mouse liver regeneration in acute and chronic liver diseases. eLife 2018; 7:e34152. [PMID: 30070632 PMCID: PMC6078493 DOI: 10.7554/elife.34152] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 07/28/2018] [Indexed: 12/24/2022] Open
Abstract
Reparative hepatocyte replication is impaired in chronic liver disease, contributing to disease progression; however, the underlying mechanism remains elusive. Here, we identify Map3k14 (also known as NIK) and its substrate Chuk (also called IKKα) as unrecognized suppressors of hepatocyte replication. Chronic liver disease is associated with aberrant activation of hepatic NIK pathways. We found that hepatocyte-specific deletion of Map3k14 or Chuk substantially accelerated mouse hepatocyte proliferation and liver regeneration following partial-hepatectomy. Hepatotoxin treatment or high fat diet feeding inhibited the ability of partial-hepatectomy to stimulate hepatocyte replication; remarkably, inactivation of hepatic NIK markedly increased reparative hepatocyte proliferation under these liver disease conditions. Mechanistically, NIK and IKKα suppressed the mitogenic JAK2/STAT3 pathway, thereby inhibiting cell cycle progression. Our data suggest that hepatic NIK and IKKα act as rheostats for liver regeneration by restraining overgrowth. Pathological activation of hepatic NIK or IKKα likely blocks hepatocyte replication, contributing to liver disease progression.
Collapse
Affiliation(s)
- Yi Xiong
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborUnited States
| | - Adriana Souza Torsoni
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborUnited States
- Laboratory of Metabolic Disorders, School of Applied SciencesUniversity of CampinasLimeiraBrazil
| | - Feihua Wu
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborUnited States
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese MedicineChina Pharmaceutical UniversityNanjingChina
| | - Hong Shen
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborUnited States
| | - Yan Liu
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborUnited States
| | - Xiao Zhong
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborUnited States
| | - Mark J Canet
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborUnited States
| | - Yatrik M Shah
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborUnited States
| | - M Bishr Omary
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborUnited States
| | - Yong Liu
- College of Life Sciences, Institute for Advanced StudiesWuhan UniversityWuhanChina
| | - Liangyou Rui
- Department of Molecular and Integrative PhysiologyUniversity of Michigan Medical SchoolAnn ArborUnited States
- Department of Internal MedicineUniversity of Michigan Medical SchoolAnn ArborUnited States
| |
Collapse
|