1
|
Tabei Y, Nakajima Y. IL-1β-activated PI3K/AKT and MEK/ERK pathways coordinately promote induction of partial epithelial-mesenchymal transition. Cell Commun Signal 2024; 22:392. [PMID: 39118068 PMCID: PMC11308217 DOI: 10.1186/s12964-024-01775-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a cellular process in embryonic development, wound healing, organ fibrosis, and cancer metastasis. Previously, we and others have reported that proinflammatory cytokine interleukin-1β (IL-1β) induces EMT. However, the exact mechanisms, especially the signal transduction pathways, underlying IL-1β-mediated EMT are not yet completely understood. Here, we found that IL-1β stimulation leads to the partial EMT-like phenotype in human lung epithelial A549 cells, including the gain of mesenchymal marker (vimentin) and high migratory potential, without the complete loss of epithelial marker (E-cadherin). IL-1β-mediated partial EMT induction was repressed by PI3K inhibitor LY294002, indicating that the PI3K/AKT pathway plays a significant role in the induction. In addition, ERK1/2 inhibitor FR180204 markedly inhibited the IL-1β-mediated partial EMT induction, demonstrating that the MEK/ERK pathway was also involved in the induction. Furthermore, we found that the activation of the PI3K/AKT and MEK/ERK pathways occurred downstream of the epidermal growth factor receptor (EGFR) pathway and the IL-1 receptor (IL-1R) pathway, respectively. Our findings suggest that the PI3K/AKT and MEK/ERK pathways coordinately promote the IL-1β-mediated partial EMT induction. The inhibition of not one but both pathways is expected yield clinical benefits by preventing partial EMT-related disorders such as organ fibrosis and cancer metastasis.
Collapse
Affiliation(s)
- Yosuke Tabei
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 2217-14 Hayashi-Cho, Takamatsu, Kagawa, 761-0395, Japan.
| | - Yoshihiro Nakajima
- Health and Medical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 2217-14 Hayashi-Cho, Takamatsu, Kagawa, 761-0395, Japan
| |
Collapse
|
2
|
Gutierrez AH, Mazariegos MS, Alemany S, Nevzorova YA, Cubero FJ, Sanz-García C. Tumor progression locus 2 (TPL2): A Cot-plicated progression from inflammation to chronic liver disease. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166660. [PMID: 36764206 DOI: 10.1016/j.bbadis.2023.166660] [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: 10/26/2022] [Revised: 01/24/2023] [Accepted: 01/30/2023] [Indexed: 02/10/2023]
Abstract
The cytoplasmic protein tumor progression locus 2 (TPL2), also known as cancer Osaka thyroid (Cot), or MAP3K8, is thought to have a significant role in a variety of cancers and illnesses and it is a key component in the activation pathway for the expression of inflammatory mediators. Despite the tight connection between inflammation and TPL2, its function has not been extensively studied in chronic liver disease (CLD), a major cause of morbidity and mortality worldwide. Here, we analyze more in detail the significance of TPL2 in CLD to shed light on the pathological and molecular transduction pattern of TPL2 during the progression of CLD. This might result in important advancements and enable progress in the diagnosis and treatment of CLD.
Collapse
Affiliation(s)
- Alejandro H Gutierrez
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, 28040 Madrid, Spain
| | - Marina S Mazariegos
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, 28040 Madrid, Spain
| | - Susana Alemany
- Department of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049 Madrid, Spain; Biomedicine Unit (Unidad Asociada al CSIC), Universidad de Las Palmas de Gran Canaria, 35001 Las Palmas, Spain
| | - Yulia A Nevzorova
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28029 Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28007 Madrid, Spain
| | - Carlos Sanz-García
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, 28040 Madrid, Spain.
| |
Collapse
|
3
|
Labiano I, Agirre-Lizaso A, Olaizola P, Echebarria A, Huici-Izagirre M, Olaizola I, Esparza-Baquer A, Sharif O, Hijona E, Milkiewicz P, Milkiewicz M, González-Romero F, Aspichueta P, Monte MJ, Marin JJG, Vucur M, Luedde T, Marzioni M, Mann DA, Bujanda L, Rodrigues PM, Banales JM, Perugorria MJ. TREM-2 plays a protective role in cholestasis by acting as a negative regulator of inflammation. J Hepatol 2022; 77:991-1004. [PMID: 35750136 DOI: 10.1016/j.jhep.2022.05.044] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 05/19/2022] [Accepted: 05/22/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUND & AIMS Inflammation, particularly that mediated by bacterial components translocating from the gut to the liver and binding to toll-like receptors (TLRs), is central to cholestatic liver injury. The triggering receptor expressed on myeloid cells-2 (TREM-2) inhibits TLR-mediated signaling and exerts a protective role in hepatocellular injury and carcinogenesis. This study aims to evaluate the role of TREM-2 in cholestasis. METHODS TREM-2 expression was analyzed in the livers of patients with primary biliary cholangitis (PBC) or primary sclerosing cholangitis (PSC), and in mouse models of cholestasis. Wild-type (WT) and Trem-2 deficient (Trem-2-/-) mice were subjected to experimental cholestasis and gut sterilization. Primary cultured Kupffer cells were incubated with lipopolysaccharide and/or ursodeoxycholic acid (UDCA) and inflammatory responses were analyzed. RESULTS TREM-2 expression was upregulated in the livers of patients with PBC or PSC, and in murine models of cholestasis. Compared to WT, the response to bile duct ligation (BDL)-induced obstructive cholestasis or alpha-naphtylisothiocyanate (ANIT)-induced cholestasis was exacerbated in Trem-2-/- mice. This was characterized by enhanced necroptotic cell death, inflammatory responses and biliary expansion. Antibiotic treatment partially abrogated the effects observed in Trem-2-/- mice after BDL. Experimental overexpression of TREM-2 in the liver of WT mice downregulated ANIT-induced IL-33 expression and neutrophil recruitment. UDCA regulated Trem-1 and Trem-2 expression in primary cultured mouse Kupffer cells and dampened inflammatory gene transcription via a TREM-2-dependent mechanism. CONCLUSIONS TREM-2 acts as a negative regulator of inflammation during cholestasis, representing a novel potential therapeutic target. LAY SUMMARY Cholestasis (the reduction or cessation of bile flow) causes liver injury. This injury is exacerbated when gut-derived bacterial components interact with receptors (specifically Toll-like receptors or TLRs) on liver-resident immune cells, promoting inflammation. Herein, we show that the anti-inflammatory receptor TREM-2 dampens TLR-mediated signaling and hence protects against cholestasis-induced liver injury. Thus, TREM-2 could be a potential therapeutic target in cholestasis.
Collapse
Affiliation(s)
- Ibone Labiano
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), Donostia-San Sebastian, Spain
| | - Aloña Agirre-Lizaso
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), Donostia-San Sebastian, Spain
| | - Paula Olaizola
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), Donostia-San Sebastian, Spain
| | - Anne Echebarria
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), Donostia-San Sebastian, Spain
| | - Maider Huici-Izagirre
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), Donostia-San Sebastian, Spain
| | - Irene Olaizola
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), Donostia-San Sebastian, Spain
| | - Aitor Esparza-Baquer
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), Donostia-San Sebastian, Spain
| | - Omar Sharif
- Institute for Vascular Biology, Center for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Elizabeth Hijona
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), Donostia-San Sebastian, Spain; CIBERehd, Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Department of Nursing, Faculty of Medicine and Nursing, University of the Basque Country, UPV/EHU, Donostia-San Sebastian, Spain
| | - Piotr Milkiewicz
- Liver and Internal Medicine Unit, Department of General, Transplant and Liver Surgery of the Medical University of Warsaw, Warsaw, Poland; Translational Medicine Group, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Malgorzata Milkiewicz
- Department of Medical Biology, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Francisco González-Romero
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, UPV/EHU, Leioa, Spain
| | - Patricia Aspichueta
- CIBERehd, Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country, UPV/EHU, Leioa, Spain; Biocruces Health Research Institute, Barakaldo, Spain
| | - Maria J Monte
- CIBERehd, Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain
| | - Jose J G Marin
- CIBERehd, Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain
| | - Mihael Vucur
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany
| | - Marco Marzioni
- Clinic of Gastroenterology and Hepatology, Università Politecnica delle Marche, Ancona, Italy
| | - Derek A Mann
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4th Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK; Department of Gastroenterology and Hepatology, School of Medicine, Koç University, Istanbul, Turkey; Fibrofind Ltd, William Leech Building, Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Luis Bujanda
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), Donostia-San Sebastian, Spain; CIBERehd, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Pedro M Rodrigues
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), Donostia-San Sebastian, Spain; CIBERehd, Instituto de Salud Carlos III (ISCIII), Madrid, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jesus M Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), Donostia-San Sebastian, Spain; CIBERehd, Instituto de Salud Carlos III (ISCIII), Madrid, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain.
| | - Maria J Perugorria
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), Donostia-San Sebastian, Spain; CIBERehd, Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Department of Medicine, Faculty of Medicine and Nursing, University of the Basque Country, UPV/EHU, Leioa, Spain.
| |
Collapse
|
4
|
Blair L, Pattison MJ, Chakravarty P, Papoutsopoulou S, Bakiri L, Wagner EF, Smale S, Ley SC. TPL-2 Inhibits IFN-β Expression via an ERK1/2-TCF-FOS Axis in TLR4-Stimulated Macrophages. THE JOURNAL OF IMMUNOLOGY 2022; 208:941-954. [PMID: 35082159 PMCID: PMC9012084 DOI: 10.4049/jimmunol.2100213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 10/20/2021] [Indexed: 12/29/2022]
Abstract
TPL-2 activation of ERK1/2 regulates gene expression in TLR-stimulated macrophages. TPL-2 regulates transcription via ERK1/2 phosphorylation of ternary complex factors. TPL-2 inhibits Ifnb1 transcription via ternary complex factor–induced Fos mRNA expression.
TPL-2 kinase plays an important role in innate immunity, activating ERK1/2 MAPKs in myeloid cells following TLR stimulation. We investigated how TPL-2 controls transcription in TLR4-stimulated mouse macrophages. TPL-2 activation of ERK1/2 regulated expression of genes encoding transcription factors, cytokines, chemokines, and signaling regulators. Bioinformatics analysis of gene clusters most rapidly induced by TPL-2 suggested that their transcription was mediated by the ternary complex factor (TCF) and FOS transcription factor families. Consistently, TPL-2 induced ERK1/2 phosphorylation of the ELK1 TCF and the expression of TCF target genes. Furthermore, transcriptomic analysis of TCF-deficient macrophages demonstrated that TCFs mediate approximately half of the transcriptional output of TPL-2 signaling, partially via induced expression of secondary transcription factors. TPL-2 signaling and TCFs were required for maximal TLR4-induced FOS expression. Comparative analysis of the transcriptome of TLR4-stimulated Fos−/− macrophages indicated that TPL-2 regulated a significant fraction of genes by controlling FOS expression levels. A key function of this ERK1/2-TCF-FOS pathway was to mediate TPL-2 suppression of type I IFN signaling, which is essential for host resistance against intracellular bacterial infection.
Collapse
Affiliation(s)
- Louise Blair
- Immune Cell Signalling Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Michael J Pattison
- Immune Cell Signalling Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Probir Chakravarty
- Bioinformatics and Biostatistics Technology Platform, The Francis Crick Institute, London, United Kingdom
| | | | - Latifa Bakiri
- Laboratory of Genes and Disease, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Erwin F Wagner
- Laboratory of Genes and Disease, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Laboratory of Genes and Disease, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Stephen Smale
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA; and
| | - Steven C Ley
- Immune Cell Signalling Laboratory, The Francis Crick Institute, London, United Kingdom;
- Department of Immunology and Inflammation, Imperial College London, London, United Kingdom
| |
Collapse
|
5
|
Hao J, Shen C, Wei N, Yan M, Zhang X, Xu G, Zhang D, Hou J, Cao W, Jin Y, Zhang K, Zheng H, Liu X. Foot-and-Mouth Disease Virus Capsid Protein VP1 Antagonizes TPL2-Mediated Activation of the IRF3/IFN-β Signaling Pathway to Facilitate the Virus Replication. Front Immunol 2021; 11:580334. [PMID: 33488582 PMCID: PMC7821752 DOI: 10.3389/fimmu.2020.580334] [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: 07/22/2020] [Accepted: 11/18/2020] [Indexed: 11/13/2022] Open
Abstract
Foot-and-mouth disease (FMD) is a severe, highly contagious viral disease of cloven-hoofed animals. In order to establish an infection, the FMD virus (FMDV) needs to counteract host antiviral responses. Tumor progression locus 2 (TPL2), a mitogen-activated protein kinase, can regulate innate and adaptive immunity; however, its exact mechanisms underlying TPL2-mediated regulation of the pathogenesis of FMDV infection remain unknown. In this study, we confirmed that TPL2 could inhibit FMDV replication in vitro and in vivo. The virus replication increased in Tpl2-deficient suckling mice in association with reduced expression of interferon-stimulated genes interferon-α (IFN-α) and myxovirus resistance (MX2) and significantly reduced expression of C-X-C motif chemokine ligand 10 (CXCL10), interferon regulatory factor 3 (IRF3), and IRF7, while the phosphorylation of IRF3 was not detected. Moreover, the interactions between TPL2 and VP1 were also confirmed. The overexpression of TPL2 promoted IRF3-mediated dose-dependent activation of the IFN-β signaling pathway in association with interactions between IRF3 and TPL2. VP1 also inhibited phosphorylation of TPL2 at Thr290, while Thr290 resulted as the key functional site associated with the TPL2-mediated antiviral response. Taken together, this study indicated that FMDV capsid protein VP1 antagonizes TPL2-mediated activation of the IRF3/IFN-β signaling pathway for immune escape and facilitated virus replication.
Collapse
Affiliation(s)
- Junhong Hao
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, Lanzhou, China
| | - Chaochao Shen
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, Lanzhou, China
| | - Nannan Wei
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, Lanzhou, China
| | - Minghao Yan
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, Lanzhou, China
| | - Xuegang Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, Lanzhou, China
| | - Guowei Xu
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, Lanzhou, China
| | - Dajun Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, Lanzhou, China
| | - Jing Hou
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, Lanzhou, China
| | - Weijun Cao
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, Lanzhou, China
| | - Ye Jin
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, Lanzhou, China
| | - Keshan Zhang
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, Lanzhou, China
| | - Haixue Zheng
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, Lanzhou, China
| | - Xiangtao Liu
- State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute of Chinese Academy of Agriculture Science, Lanzhou, China
| |
Collapse
|
6
|
Moran-Salvador E, Garcia-Macia M, Sivaharan A, Sabater L, Zaki MY, Oakley F, Knox A, Page A, Luli S, Mann J, Mann DA. Fibrogenic Activity of MECP2 Is Regulated by Phosphorylation in Hepatic Stellate Cells. Gastroenterology 2019; 157:1398-1412.e9. [PMID: 31352003 PMCID: PMC6853276 DOI: 10.1053/j.gastro.2019.07.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 07/12/2019] [Accepted: 07/17/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Methyl-CpG binding protein 2, MECP2, which binds to methylated regions of DNA to regulate transcription, is expressed by hepatic stellate cells (HSCs) and is required for development of liver fibrosis in mice. We investigated the effects of MECP2 deletion from HSCs on their transcriptome and of phosphorylation of MECP2 on HSC phenotype and liver fibrosis. METHODS We isolated HSCs from Mecp2-/y mice and wild-type (control) mice. HSCs were activated in culture and used in array analyses of messenger RNAs and long noncoding RNAs. Kyoto Encyclopedia of Genes and Genomes pathway analyses identified pathways regulated by MECP2. We studied mice that expressed a mutated form of Mecp2 that encodes the S80A substitution, MECP2S80, causing loss of MECP2 phosphorylation at serine 80. Liver fibrosis was induced in these mice by administration of carbon tetrachloride, and liver tissues and HSCs were collected and analyzed. RESULTS MECP2 deletion altered expression of 284 messenger RNAs and 244 long noncoding RNAs, including those that regulate DNA replication; are members of the minichromosome maintenance protein complex family; or encode CDC7, HAS2, DNA2 (a DNA helicase), or RPA2 (a protein that binds single-stranded DNA). We found that MECP2 regulates the DNA repair Fanconi anemia pathway in HSCs. Phosphorylation of MECP2S80 and its putative kinase, HAS2, were induced during transdifferentiation of HSCs. HSCs from MECP2S80 mice had reduced proliferation, and livers from these mice had reduced fibrosis after carbon tetrachloride administration. CONCLUSIONS In studies of mice with disruption of Mecp2 or that expressed a form of MECP2 that is not phosphorylated at S80, we found phosphorylation of MECP2 to be required for HSC proliferation and induction of fibrosis. In HSCs, MECP2 regulates expression of genes required for DNA replication and repair. Strategies to inhibit MECP2 phosphorylation at S80 might be developed for treatment of liver fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jelena Mann
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom.
| | | |
Collapse
|
7
|
Xu J, Pei S, Wang Y, Liu J, Qian Y, Huang M, Zhang Y, Xiao Y. Tpl2 Protects Against Fulminant Hepatitis Through Mobilization of Myeloid-Derived Suppressor Cells. Front Immunol 2019; 10:1980. [PMID: 31481966 PMCID: PMC6710335 DOI: 10.3389/fimmu.2019.01980] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/05/2019] [Indexed: 12/18/2022] Open
Abstract
Myeloid derived suppressor cells (MDSC) in the liver microenvironment protects against the inflammation-induced liver injury in fulminant hepatitis (FH). However, the molecular mechanism through which MDSC is recruited into the inflamed liver remain elusive. Here we identified a protein kinase Tpl2 as a critical mediator of MDSC recruitment into liver during the pathogenesis of Propionibacterium acnes/LPS-induced FH. Loss of Tpl2 dramatically suppressed MDSC mobilization into liver, leading to exaggerated local inflammation and increased FH-induced mortality. Mechanistically, although the protective effect of Tpl2 for FH-induced mortality was dependent on the presence of MDSC, Tpl2 neither directly targeted myeloid cells nor T cells to regulate FH pathogenesis, but functioned in hepatocytes to mediate the induction of MDSC-attracting chemokine CXCL1 and CXCL2 through modulating IL-25 (also known as IL-17E) signaling. As a consequence, increased MDSC in the inflamed liver specifically restrained the local proliferation of infiltrated pathogenic CD4+ T cells, and thus protected against the inflammation-induced acute liver failure. Together, our findings established Tpl2 as a critical mediator of MDSC recruitment and highlighted the therapeutic potential of Tpl2 for the treatment of FH.
Collapse
Affiliation(s)
- Jing Xu
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Medical College of Soochow University, Soochow University, Suzhou, China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Siyu Pei
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Junli Liu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Youcun Qian
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mingzhu Huang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yanyun Zhang
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Medical College of Soochow University, Soochow University, Suzhou, China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
8
|
ERK Pathway in Activated, Myofibroblast-Like, Hepatic Stellate Cells: A Critical Signaling Crossroad Sustaining Liver Fibrosis. Int J Mol Sci 2019; 20:ijms20112700. [PMID: 31159366 PMCID: PMC6600376 DOI: 10.3390/ijms20112700] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/21/2019] [Accepted: 05/30/2019] [Indexed: 12/12/2022] Open
Abstract
Fibrogenic progression of chronic liver disease, whatever the etiology, is characterized by persistent chronic parenchymal injury, chronic activation of inflammatory response, and sustained activation of liver fibrogenesis, and of pathological wound healing response. A critical role in liver fibrogenesis is played by hepatic myofibroblasts (MFs), a heterogeneous population of α smooth-muscle actin—positive cells that originate from various precursor cells through a process of activation and transdifferentiation. In this review, we focus the attention on the role of extracellular signal-regulated kinase (ERK) signaling pathway as a critical one in modulating selected profibrogenic phenotypic responses operated by liver MFs. We will also analyze major therapeutic antifibrotic strategies developed in the last two decades in preclinical studies, some translated to clinical conditions, designed to interfere directly or indirectly with the Ras/Raf/MEK/ERK signaling pathway in activated hepatic MFs, but that also significantly increased our knowledge on the biology and pathobiology of these fascinating profibrogenic cells.
Collapse
|
9
|
Dong X, Liu J, Xu Y, Cao H. Role of macrophages in experimental liver injury and repair in mice. Exp Ther Med 2019; 17:3835-3847. [PMID: 31007731 PMCID: PMC6468932 DOI: 10.3892/etm.2019.7450] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 12/06/2018] [Indexed: 02/06/2023] Open
Abstract
Liver macrophages make up the largest proportion of tissue macrophages in the host and consist of two dissimilar groups: Kupffer cells (KCs) and monocyte-derived macrophages (MoMø). As the liver is injured, KCs sense the injury and initiate inflammatory cascades mediated by the release of inflammatory cytokines and chemokines. Subsequently, inflammatory monocytes accumulate in the liver via chemokine-chemokine receptor interactions, resulting in massive inflammatory MoMø infiltration. When live r injury ceases, restorative macrophages, derived from recruited inflammatory monocytes (lymphocyte antigen 6 complex, locus Chi monocytes), promote the resolution of hepatic damage and fibrosis. Consequently, a large number of studies have assessed the mechanisms by which liver macrophages exert their opposing functions at different time-points during liver injury. The present review primarily focuses on the diverse functions of macrophages in experimental liver injury, fibrosis and repair in mice and illustrates how macrophages may be targeted to treat liver disease.
Collapse
Affiliation(s)
- Xiaotian Dong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Jingqi Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Yanping Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Hongcui Cao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
10
|
Carneros D, Santamaría EM, Larequi E, Vélez-Ortiz JM, Reboredo M, Mancheño U, Perugorria MJ, Navas P, Romero-Gómez M, Prieto J, Hervás-Stubbs S, Bustos M. Cardiotrophin-1 is an anti-inflammatory cytokine and promotes IL-4-induced M2 macrophage polarization. FASEB J 2019; 33:7578-7587. [PMID: 30892966 DOI: 10.1096/fj.201801563r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Macrophages play a central role in tissue remodeling, repair, and resolution of inflammation. Macrophage polarization to M1 or M2 activation status may determine the progression or resolution of the inflammatory response. We have previously reported that cardiotrophin-1 (CT-1) displays both cytoprotective and metabolic activities. The role of CT-1 in inflammation remains poorly understood. Here, we employed recombinant CT-1 (rCT-1) and used CT-1-null mice and myeloid-specific CT-1 transgenic mice to investigate whether CT-1 might play a role in the modulation of the inflammatory response. We observed that CT-1 deficiency was associated with enhanced release of inflammatory mediators and with stronger activation of NF-κB in response to LPS, whereas the inflammatory response was attenuated in CT-1 transgenic mice or by administering rCT-1 to wild-type animals prior to LPS challenge. We found that CT-1 promoted IL-6 expression only by nonhematopoietic cells, whereas LPS up-regulated IL-6 expression in both hematopoietic and nonhematopoietic cells. Notably, rCT-1 inhibited LPS-mediated soluble IL-6R induction. Using IL-6-/- mice, we showed that rCT-1 inhibited LPS-induced TNF-α and IFN-γ response in an IL-6-independent manner. Importantly, we demonstrated that CT-1 primes macrophages for IL-4-dependent M2 polarization by inducing IL-4 receptor expression. Mechanistic analyses showed that the signal transducer and activator of transcription 3-suppressor of cytokine signaling 3 axis mediates this effect. Our findings support the notion that CT-1 is a critical regulator of inflammation and suggest that rCT-1 could be a molecule with potential therapeutic application in inflammatory conditions.-Carneros, D., Santamaría, E. M., Larequi, E., Vélez-Ortiz, J. M., Reboredo, M., Mancheño, U., Perugorria, M. J., Navas, P., Romero-Gómez, M., Prieto, J., Hervás-Stubbs, S., Bustos, M. Cardiotrophin-1 is an anti-inflammatory cytokine and promotes IL-4-induced M2 macrophage polarization.
Collapse
Affiliation(s)
- David Carneros
- Institute of Biomedicine of Seville (IBiS), Consejo Superior de Investigaciones Científicas (CSIC)-University of Seville-Virgen del Rocio University Hospital, Seville, Spain
| | - Eva M Santamaría
- Division of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Centro de Investigatión Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Eduardo Larequi
- Division of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Jose Miguel Vélez-Ortiz
- Institute of Biomedicine of Seville (IBiS), Consejo Superior de Investigaciones Científicas (CSIC)-University of Seville-Virgen del Rocio University Hospital, Seville, Spain
| | - Mercedes Reboredo
- Division of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Centro de Investigatión Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Uxua Mancheño
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - María Jesús Perugorria
- Centro de Investigatión Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain.,Biodonostia Health Research Institute, Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Plácido Navas
- Centro Andaluz de Biología del Desarrollo (CABD), University of Pablo de Olavide-Consejo Superior de Investigaciones Científicas (CSIC), Seville, Spain.,Centro de Investigación Biomédica en Red Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Seville, Spain
| | - Manuel Romero-Gómez
- Institute of Biomedicine of Seville (IBiS), Consejo Superior de Investigaciones Científicas (CSIC)-University of Seville-Virgen del Rocio University Hospital, Seville, Spain.,Centro de Investigatión Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Prieto
- Division of Hepatology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Centro de Investigatión Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Hervás-Stubbs
- Division of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.,Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Matilde Bustos
- Institute of Biomedicine of Seville (IBiS), Consejo Superior de Investigaciones Científicas (CSIC)-University of Seville-Virgen del Rocio University Hospital, Seville, Spain
| |
Collapse
|
11
|
Perugorria MJ, Esparza-Baquer A, Oakley F, Labiano I, Korosec A, Jais A, Mann J, Tiniakos D, Santos-Laso A, Arbelaiz A, Gawish R, Sampedro A, Fontanellas A, Hijona E, Jimenez-Agüero R, Esterbauer H, Stoiber D, Bujanda L, Banales JM, Knapp S, Sharif O, Mann DA. Non-parenchymal TREM-2 protects the liver from immune-mediated hepatocellular damage. Gut 2019; 68:533-546. [PMID: 29374630 PMCID: PMC6580759 DOI: 10.1136/gutjnl-2017-314107] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 12/26/2017] [Accepted: 12/28/2017] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Liver injury impacts hepatic inflammation in part via Toll-like receptor (TLR) signalling. Triggering receptor expressed on myeloid cells 2 (TREM-2) modulates TLR4-mediated inflammation in bone marrow (BM)-derived macrophages but its function in liver injury is unknown. Here we hypothesised that the anti-inflammatory effects of TREM-2 on TLR signalling may limit hepatic injury. DESIGN TREM-2 expression was analysed in livers of humans with various forms of liver injury compared with control individuals. Acute and chronic liver injury models were performed in wild type and Trem-2-/- mice. Primary liver cells from both genotypes of mice were isolated for in vitro experiments. RESULTS TREM-2 was expressed on non-parenchymal hepatic cells and induced during liver injury in mice and man. Mice lacking TREM-2 exhibited heightened liver damage and inflammation during acute and repetitive carbon tetrachloride and acetaminophen (APAP) intoxication, the latter of which TREM-2 deficiency was remarkably associated with worsened survival. Liver damage in Trem-2-/- mice following chronic injury and APAP challenge was associated with elevated hepatic lipid peroxidation and macrophage content. BM transplantation experiments and cellular reactive oxygen species assays revealed effects of TREM-2 in the context of chronic injury depended on both immune and resident TREM-2 expression. Consistent with effects of TREM-2 on inflammation-associated injury, primary hepatic macrophages and hepatic stellate cells lacking TREM-2 exhibited augmented TLR4-driven proinflammatory responses. CONCLUSION Our data indicate that by acting as a natural brake on inflammation during hepatocellular injury, TREM-2 is a critical regulator of diverse types of hepatotoxic injury.
Collapse
Affiliation(s)
- Maria J Perugorria
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- CIBERehd, Instituto de Salud Carlos III, San Sebastián, Spain
| | - Aitor Esparza-Baquer
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), San Sebastian, Spain
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Ibone Labiano
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), San Sebastian, Spain
| | - Ana Korosec
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Alexander Jais
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Jelena Mann
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Dina Tiniakos
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Alvaro Santos-Laso
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), San Sebastian, Spain
| | - Ander Arbelaiz
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), San Sebastian, Spain
| | - Riem Gawish
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Ana Sampedro
- Hepatology Programme, CIMA, University of Navarra, Pamplona, Spain
| | | | - Elizabeth Hijona
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), San Sebastian, Spain
- CIBERehd, Instituto de Salud Carlos III, San Sebastián, Spain
| | - Raul Jimenez-Agüero
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), San Sebastian, Spain
| | - Harald Esterbauer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Dagmar Stoiber
- Institute of Pharmacology, Center of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Luis Bujanda
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), San Sebastian, Spain
- CIBERehd, Instituto de Salud Carlos III, San Sebastián, Spain
| | - Jesus María Banales
- Department of Liver and Gastrointestinal Diseases, Biodonostia Research Institute, Donostia University Hospital, University of the Basque Country (UPV-EHU), San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- CIBERehd, Instituto de Salud Carlos III, San Sebastián, Spain
| | - Sylvia Knapp
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
| | - Omar Sharif
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Derek A Mann
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
12
|
Gong J, Fang C, Zhang P, Wang PX, Qiu Y, Shen LJ, Zhang L, Zhu XY, Tian S, Li F, Wang Z, Huang Z, Wang A, Zhang XD, She ZG. Tumor Progression Locus 2 in Hepatocytes Potentiates Both Liver and Systemic Metabolic Disorders in Mice. Hepatology 2019; 69:524-544. [PMID: 29381809 DOI: 10.1002/hep.29820] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/25/2018] [Indexed: 12/27/2022]
Abstract
Tumor progression locus 2 (TPL2), a serine/threonine kinase, has been regarded as a potentially interesting target for the treatment of various diseases with an inflammatory component. However, the function of TPL2 in regulating hepatocyte metabolism and liver inflammation during the progression of nonalcoholic fatty liver disease (NAFLD) is poorly understood. Here, we report that TPL2 protein expression was significantly increased in fatty liver from diverse species, including humans, monkeys, and mice. Further investigations revealed that compared to wild-type (WT) littermates, hepatocyte-specific TPL2 knockout (HKO) mice exhibited improved lipid and glucose imbalance, reserved insulin sensitivity, and alleviated inflammation in response to high-fat diet (HFD) feeding. Overexpression of TPL2 in hepatocytes led to the opposite phenotype. Regarding the mechanism, we found that mitogen-activated protein kinase kinase 7 (MKK7) was the specific substrate of TPL2 for c-Jun N-terminal kinase (JNK) activation. TPL2-MKK7-JNK signaling in hepatocytes represents a promising drugable target for treating NAFLD and associated metabolic disorders. Conclusion: In hepatocytes, TPL2 acts as a key mediator that promotes both liver and systemic metabolic disturbances by specifically increasing MKK7-JNK activation.
Collapse
Affiliation(s)
- Jun Gong
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
| | - Chun Fang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Peng Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Pi-Xiao Wang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Yixing Qiu
- Lab of Animal Models and Functional Genomics (LAMFG), College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
- TCM and Ethnomedicine Innovation & Development Laboratory, Sino-Pakistan TCM Research Center, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Li-Jun Shen
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Li Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xue-Yong Zhu
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Song Tian
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Feng Li
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
| | - Zhihua Wang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zan Huang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Aibing Wang
- Lab of Animal Models and Functional Genomics (LAMFG), College of Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Xiao-Dong Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhi-Gang She
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Institute of Model Animals of Wuhan University, Wuhan, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
13
|
TPL-2 restricts Ccl24-dependent immunity to Heligmosomoides polygyrus. PLoS Pathog 2017; 13:e1006536. [PMID: 28759611 PMCID: PMC5560741 DOI: 10.1371/journal.ppat.1006536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 08/17/2017] [Accepted: 07/17/2017] [Indexed: 11/19/2022] Open
Abstract
TPL-2 (COT, MAP3K8) kinase activates the MEK1/2-ERK1/2 MAPK signaling pathway in innate immune responses following TLR, TNFR1 and IL-1R stimulation. TPL-2 contributes to type-1/Th17-mediated autoimmunity and control of intracellular pathogens. We recently demonstrated TPL-2 reduces severe airway allergy to house dust mite by negatively regulating type-2 responses. In the present study, we found that TPL-2 deficiency resulted in resistance to Heligmosomoides polygyrus infection, with accelerated worm expulsion, reduced fecal egg burden and reduced worm fitness. Using co-housing experiments, we found resistance to infection in TPL-2 deficient mice (Map3k8-/-) was independent of microbiota alterations in H. polygyrus infected WT and Map3k8-/-mice. Additionally, our data demonstrated immunity to H. polygyrus infection in TPL-2 deficient mice was not due to dysregulated type-2 immune responses. Genome-wide analysis of intestinal tissue from infected TPL-2-deficient mice identified elevated expression of genes involved in chemotaxis and homing of leukocytes and cells, including Ccl24 and alternatively activated genes. Indeed, Map3k8-/-mice had a significant influx of eosinophils, neutrophils, monocytes and Il4GFP+ T cells. Conditional knockout experiments demonstrated that specific deletion of TPL-2 in CD11c+ cells, but not Villin+ epithelial cells, LysM+ myeloid cells or CD4+ T cells, led to accelerated resistance to H. polygyrus. In line with a central role of CD11c+ cells, CD11c+ CD11b+ cells isolated from TPL-2-deficient mice had elevated Ccl24. Finally, Ccl24 neutralization in TPL-2 deficient mice significantly decreased the expression of Arg1, Retnla, Chil3 and Ear11 correlating with a loss of resistance to H. polygyrus. These observations suggest that TPL-2-regulated Ccl24 in CD11c+CD11b+ cells prevents accelerated type-2 mediated immunity to H. polygyrus. Collectively, this study identifies a previously unappreciated role for TPL-2 controlling immune responses to H. polygyrus infection by restricting Ccl24 production.
Collapse
|
14
|
Abstract
Understanding the underlying molecular mechanisms of liver fibrosis is important to develop effective therapy. Herein, we show that focal-adhesion-kinse (FAK) plays a key role in promoting hepatic stellate cells (HSCs) activation in vitro and liver fibrosis progression in vivo. FAK activation is associated with increased expression of α-smooth muscle actin (α-SMA) and collagen in fibrotic live tissues. Transforming growth factor beta-1 (TGF-β1) induces FAK activation in a time and dose dependent manner. FAK activation precedes the α-SMA expression in HSCs. Inhibition of FAK activation blocks the α-SMA and collagen expression, and inhibits the formation of stress fibers in TGF-β1 treated HSCs. Furthermore, inhibition of FAK activation significantly reduces HSC migration and small GTPase activation, and induces apoptotic signaling in TGF-β1 treated HSCs. Importantly, FAK inhibitor attenuates liver fibrosis in vivo and significantly reduces collagen and α-SMA expression in an animal model of liver fibrosis. These data demonstrate that FAK plays an essential role in HSC activation and liver fibrosis progression, and FAK signaling pathway could be a potential target for liver fibrosis.
Collapse
|
15
|
Boldrini L, Giordano M, Servadio A, Niccoli C, Bertoglio P, Lucchi M, Melfi F, Mussi A, Fontanini G. Prognostic role of TPL2 in early‑stage non‑small cell lung cancer. Mol Med Rep 2017; 15:3451-3458. [PMID: 28393206 PMCID: PMC5436291 DOI: 10.3892/mmr.2017.6430] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 11/09/2016] [Indexed: 01/10/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for ~70% of all lung cancer-associated mortalities worldwide. The serine/threonine protein kinase tumor progression locus 2 [TPL2/MAP3 kinase 8 (MAP3K8)] may impact oncogenic events; however the role of TPL2 in lung carcinogenesis remains unclear. The present study was focused on the potential prognostic role of TPL2 in 101 patients with early-stage NSCLC. Since TPL2 is a potential target of miR-21, the association between TPL2 and miR-21 expression was also examined. TPL2 and miR-21 mRNA expression was quantified using reverse transcription quantitative polymerase chain reaction (RT-qPCR). TPL2 protein levels were evaluated by immunohistochemistry (IHC). The present study identified that the mRNA expression of TPL2 was low in 52/101 (51%) cases and high in 49/101 (49%) cases. IHC analysis of TPL2 protein expression often demonstrated identical mRNA results. No statistically significant associations were observed between the mRNA expression of TPL2 and the predominant clinicopathological characteristics of the patients with NSCLC, as well as identifying no association between TPL2 and miR-21. TPL2 mRNA expression was significantly higher in patients with NSCLC with good prognosis (disease-free interval P=0.009; overall survival P=0.024), when compared with those of poor prognosis. Focusing on the difference in mRNA expression of TPL2 among the adenocarcinomas in affected patients, TPL2 was more highly expressed in lepidic adenocarcinomas compared with in the other subtypes (P=0.012). The present study is the first examination, to the best of our knowledge, of TPL2 in early-stage NSCLC in relation to miR-21, and in different adenocarcinoma subtypes. Future studies must clarify the mechanism by which TPL2 is involved in lung carcinogenesis due to its important translational implications.
Collapse
Affiliation(s)
- Laura Boldrini
- Department of Surgical, Medical, Molecular Pathology and Critical Care, University of Pisa, I‑56126 Pisa, Italy
| | - Mirella Giordano
- Department of Surgical, Medical, Molecular Pathology and Critical Care, University of Pisa, I‑56126 Pisa, Italy
| | - Adele Servadio
- Department of Surgical, Medical, Molecular Pathology and Critical Care, University of Pisa, I‑56126 Pisa, Italy
| | - Cristina Niccoli
- Department of Surgical, Medical, Molecular Pathology and Critical Care, University of Pisa, I‑56126 Pisa, Italy
| | - Pietro Bertoglio
- Department of Surgical, Medical, Molecular Pathology and Critical Care, University of Pisa, I‑56126 Pisa, Italy
| | - Marco Lucchi
- Department of Surgical, Medical, Molecular Pathology and Critical Care, University of Pisa, I‑56126 Pisa, Italy
| | - Franca Melfi
- Unit of Thoracic Surgery, Azienda Ospedaliero‑Universitaria Pisana, I‑56126 Pisa, Italy
| | - Alfredo Mussi
- Department of Surgical, Medical, Molecular Pathology and Critical Care, University of Pisa, I‑56126 Pisa, Italy
| | - Gabriella Fontanini
- Department of Surgical, Medical, Molecular Pathology and Critical Care, University of Pisa, I‑56126 Pisa, Italy
| |
Collapse
|
16
|
Sanz-Garcia C, Sánchez Á, Contreras-Jurado C, Cales C, Barranquero C, Muñoz M, Merino R, Escudero P, Sanz MJ, Osada J, Aranda A, Alemany S. Map3k8 Modulates Monocyte State and Atherogenesis in ApoE-/- Mice. Arterioscler Thromb Vasc Biol 2016; 37:237-246. [PMID: 27856455 DOI: 10.1161/atvbaha.116.308528] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 11/07/2016] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Map3k8 (Cot/Tpl2) activates the MKK1/2-ERK1/2, MAPK pathway downstream from interleukin-1R, tumor necrosis factor-αR, NOD-2R (nucleotide-binding oligomerization domain-like 2R), adiponectinR, and Toll-like receptors. Map3k8 plays a key role in innate and adaptive immunity and influences inflammatory processes by modulating the functions of different cell types. However, its role in atherogenesis remains unknown. In this study, we analyzed the role of this kinase in this pathology. APPROACH AND RESULTS We show here that Map3k8 deficiency results in smaller numbers of Ly6ChighCD11clow and Ly6ClowCD11chigh monocytes in ApoE-/- mice fed a high-fat diet (HFD). Map3k8-/-ApoE-/- monocytes displayed high rates of apoptosis and reduced amounts of Nr4a1, a transcription factor known to modulate apoptosis in Ly6ClowCD11chigh monocytes. Map3k8-/-ApoE-/- splenocytes and macrophages showed irregular patterns of cytokine and chemokine expression. Map3k8 deficiency altered cell adhesion and migration in vivo and decreased CCR2 expression, a determinant chemokine receptor for monocyte mobilization, on circulating Ly6ChighCD11clow monocytes. Map3k8-/-ApoE-/- mice fed an HFD showed decreased cellular infiltration in the atherosclerotic plaque, with low lipid content. Lesions had similar size after Map3k8+/+ApoE-/- bone marrow transplant into Map3k8-/-ApoE-/- and Map3k8+/+ApoE-/- mice fed an HFD, whereas smaller plaques were observed after the transplantation of bone marrow lacking both ApoE and Map3k8. CONCLUSIONS Map3k8 decreases apoptosis of monocytes and enhances CCR2 expression on Ly6ChighCD11clow monocytes of ApoE-/- mice fed an HFD. These findings explain the smaller aortic lesions in ApoE-/- mice with Map3k8-/-ApoE-/- bone marrow cells fed an HFD, supporting further studies of Map3k8 as an antiatherosclerotic target.
Collapse
Affiliation(s)
- Carlos Sanz-Garcia
- From the Instituto de Investigaciones Biomédicas "Alberto Sols" Madrid, Consejo Superior de Investigaciones Científicas (CSIC-UAM) y Unidad de Biomedicina (UA, CSIC), University of Las Palmas de Gran Canaria, España (C.S.-G., Á.S., C.C.-J., C.C., A.A., S.A.); Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, IISA, University of Zaragoza, España (C.B., J.O.); Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC), Santander, España (M.M., R.M.); and Departmento de Farmacologia, Facultad de Medicina, University of Valencia, INCLIVA, España (P.E., M.-J.S.)
| | - Ángela Sánchez
- From the Instituto de Investigaciones Biomédicas "Alberto Sols" Madrid, Consejo Superior de Investigaciones Científicas (CSIC-UAM) y Unidad de Biomedicina (UA, CSIC), University of Las Palmas de Gran Canaria, España (C.S.-G., Á.S., C.C.-J., C.C., A.A., S.A.); Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, IISA, University of Zaragoza, España (C.B., J.O.); Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC), Santander, España (M.M., R.M.); and Departmento de Farmacologia, Facultad de Medicina, University of Valencia, INCLIVA, España (P.E., M.-J.S.)
| | - Constanza Contreras-Jurado
- From the Instituto de Investigaciones Biomédicas "Alberto Sols" Madrid, Consejo Superior de Investigaciones Científicas (CSIC-UAM) y Unidad de Biomedicina (UA, CSIC), University of Las Palmas de Gran Canaria, España (C.S.-G., Á.S., C.C.-J., C.C., A.A., S.A.); Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, IISA, University of Zaragoza, España (C.B., J.O.); Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC), Santander, España (M.M., R.M.); and Departmento de Farmacologia, Facultad de Medicina, University of Valencia, INCLIVA, España (P.E., M.-J.S.)
| | - Carmela Cales
- From the Instituto de Investigaciones Biomédicas "Alberto Sols" Madrid, Consejo Superior de Investigaciones Científicas (CSIC-UAM) y Unidad de Biomedicina (UA, CSIC), University of Las Palmas de Gran Canaria, España (C.S.-G., Á.S., C.C.-J., C.C., A.A., S.A.); Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, IISA, University of Zaragoza, España (C.B., J.O.); Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC), Santander, España (M.M., R.M.); and Departmento de Farmacologia, Facultad de Medicina, University of Valencia, INCLIVA, España (P.E., M.-J.S.)
| | - Cristina Barranquero
- From the Instituto de Investigaciones Biomédicas "Alberto Sols" Madrid, Consejo Superior de Investigaciones Científicas (CSIC-UAM) y Unidad de Biomedicina (UA, CSIC), University of Las Palmas de Gran Canaria, España (C.S.-G., Á.S., C.C.-J., C.C., A.A., S.A.); Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, IISA, University of Zaragoza, España (C.B., J.O.); Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC), Santander, España (M.M., R.M.); and Departmento de Farmacologia, Facultad de Medicina, University of Valencia, INCLIVA, España (P.E., M.-J.S.)
| | - Marta Muñoz
- From the Instituto de Investigaciones Biomédicas "Alberto Sols" Madrid, Consejo Superior de Investigaciones Científicas (CSIC-UAM) y Unidad de Biomedicina (UA, CSIC), University of Las Palmas de Gran Canaria, España (C.S.-G., Á.S., C.C.-J., C.C., A.A., S.A.); Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, IISA, University of Zaragoza, España (C.B., J.O.); Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC), Santander, España (M.M., R.M.); and Departmento de Farmacologia, Facultad de Medicina, University of Valencia, INCLIVA, España (P.E., M.-J.S.)
| | - Ramón Merino
- From the Instituto de Investigaciones Biomédicas "Alberto Sols" Madrid, Consejo Superior de Investigaciones Científicas (CSIC-UAM) y Unidad de Biomedicina (UA, CSIC), University of Las Palmas de Gran Canaria, España (C.S.-G., Á.S., C.C.-J., C.C., A.A., S.A.); Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, IISA, University of Zaragoza, España (C.B., J.O.); Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC), Santander, España (M.M., R.M.); and Departmento de Farmacologia, Facultad de Medicina, University of Valencia, INCLIVA, España (P.E., M.-J.S.)
| | - Paula Escudero
- From the Instituto de Investigaciones Biomédicas "Alberto Sols" Madrid, Consejo Superior de Investigaciones Científicas (CSIC-UAM) y Unidad de Biomedicina (UA, CSIC), University of Las Palmas de Gran Canaria, España (C.S.-G., Á.S., C.C.-J., C.C., A.A., S.A.); Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, IISA, University of Zaragoza, España (C.B., J.O.); Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC), Santander, España (M.M., R.M.); and Departmento de Farmacologia, Facultad de Medicina, University of Valencia, INCLIVA, España (P.E., M.-J.S.)
| | - Maria-Jesús Sanz
- From the Instituto de Investigaciones Biomédicas "Alberto Sols" Madrid, Consejo Superior de Investigaciones Científicas (CSIC-UAM) y Unidad de Biomedicina (UA, CSIC), University of Las Palmas de Gran Canaria, España (C.S.-G., Á.S., C.C.-J., C.C., A.A., S.A.); Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, IISA, University of Zaragoza, España (C.B., J.O.); Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC), Santander, España (M.M., R.M.); and Departmento de Farmacologia, Facultad de Medicina, University of Valencia, INCLIVA, España (P.E., M.-J.S.)
| | - Jesús Osada
- From the Instituto de Investigaciones Biomédicas "Alberto Sols" Madrid, Consejo Superior de Investigaciones Científicas (CSIC-UAM) y Unidad de Biomedicina (UA, CSIC), University of Las Palmas de Gran Canaria, España (C.S.-G., Á.S., C.C.-J., C.C., A.A., S.A.); Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, IISA, University of Zaragoza, España (C.B., J.O.); Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC), Santander, España (M.M., R.M.); and Departmento de Farmacologia, Facultad de Medicina, University of Valencia, INCLIVA, España (P.E., M.-J.S.)
| | - Ana Aranda
- From the Instituto de Investigaciones Biomédicas "Alberto Sols" Madrid, Consejo Superior de Investigaciones Científicas (CSIC-UAM) y Unidad de Biomedicina (UA, CSIC), University of Las Palmas de Gran Canaria, España (C.S.-G., Á.S., C.C.-J., C.C., A.A., S.A.); Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, IISA, University of Zaragoza, España (C.B., J.O.); Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC), Santander, España (M.M., R.M.); and Departmento de Farmacologia, Facultad de Medicina, University of Valencia, INCLIVA, España (P.E., M.-J.S.)
| | - Susana Alemany
- From the Instituto de Investigaciones Biomédicas "Alberto Sols" Madrid, Consejo Superior de Investigaciones Científicas (CSIC-UAM) y Unidad de Biomedicina (UA, CSIC), University of Las Palmas de Gran Canaria, España (C.S.-G., Á.S., C.C.-J., C.C., A.A., S.A.); Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, IISA, University of Zaragoza, España (C.B., J.O.); Instituto de Biomedicina y Biotecnología de Cantabria (CSIC-UC), Santander, España (M.M., R.M.); and Departmento de Farmacologia, Facultad de Medicina, University of Valencia, INCLIVA, España (P.E., M.-J.S.).
| |
Collapse
|
17
|
Kannan Y, Perez-Lloret J, Li Y, Entwistle LJ, Khoury H, Papoutsopoulou S, Mahmood R, Mansour NR, Ching-Cheng Huang S, Pearce EJ, Pedro S. de Carvalho L, Ley SC, Wilson MS. TPL-2 Regulates Macrophage Lipid Metabolism and M2 Differentiation to Control TH2-Mediated Immunopathology. PLoS Pathog 2016; 12:e1005783. [PMID: 27487182 PMCID: PMC4972396 DOI: 10.1371/journal.ppat.1005783] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 06/30/2016] [Indexed: 01/05/2023] Open
Abstract
Persistent TH2 cytokine responses following chronic helminth infections can often lead to the development of tissue pathology and fibrotic scarring. Despite a good understanding of the cellular mechanisms involved in fibrogenesis, there are very few therapeutic options available, highlighting a significant medical need and gap in our understanding of the molecular mechanisms of TH2-mediated immunopathology. In this study, we found that the Map3 kinase, TPL-2 (Map3k8; Cot) regulated TH2-mediated intestinal, hepatic and pulmonary immunopathology following Schistosoma mansoni infection or S. mansoni egg injection. Elevated inflammation, TH2 cell responses and exacerbated fibrosis in Map3k8–/–mice was observed in mice with myeloid cell-specific (LysM) deletion of Map3k8, but not CD4 cell-specific deletion of Map3k8, indicating that TPL-2 regulated myeloid cell function to limit TH2-mediated immunopathology. Transcriptional and metabolic assays of Map3k8–/–M2 macrophages identified that TPL-2 was required for lipolysis, M2 macrophage activation and the expression of a variety of genes involved in immuno-regulatory and pro-fibrotic pathways. Taken together this study identified that TPL-2 regulated TH2-mediated inflammation by supporting lipolysis and M2 macrophage activation, preventing TH2 cell expansion and downstream immunopathology and fibrosis. Chronic helminth infections can cause significant morbidity and organ damage in their definitive mammalian hosts. Managing this collateral damage can reduce morbidity and preserve vital tissues for normal organ function. One particular consequence of some chronic helminth infections is the deposition of fibrotic scar tissue, following immune responses directed towards helminth material. In this study we tested the role of a particular signalling kinase, TPL-2, and identified that it critically regulated the magnitude of fibrotic scarring following infection. Using several murine models with genetic deletions of TPL-2 in either all cells or specific deletion in subsets of immune cells (Map3k8–/–Map3k8fl/fl) we identified that expression of TPL-2 in myeloid cells was essential to prevent severe immune-mediated pathology. Using genome-wide analyses and metabolic assays, we discovered that TPL-2 was required for normal lipid metabolism and appropriate activation of myeloid cells / macrophages to limit fibrosis. These results revealed a previously unappreciated role for TPL-2 in preventing severe pathology following infection. Thus, activating this pathway may limit immune mediated pathology following chronic helminth infection. More broadly, this pathway is being targeted to treat inflammatory diseases and cancer [1, 2]. This study would suggest that caution should be taken to prevent untoward co-morbidities and fibrosis-related pathologies in patients when targeting TPL-2.
Collapse
Affiliation(s)
- Yashaswini Kannan
- Allergy and Anti-Helminth Immunity Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Jimena Perez-Lloret
- Allergy and Anti-Helminth Immunity Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Yanda Li
- Allergy and Anti-Helminth Immunity Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Lewis J. Entwistle
- Allergy and Anti-Helminth Immunity Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Hania Khoury
- Mycobacterial Metabolism and Antibiotic Research Laboratory, The Francis Crick Institute, London, United Kingdom
| | | | - Radma Mahmood
- Experimental Histopathology, Mill Hill Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Nuha R. Mansour
- Department of Infection and Immunity, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Stanley Ching-Cheng Huang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Edward J. Pearce
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Luiz Pedro S. de Carvalho
- Mycobacterial Metabolism and Antibiotic Research Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Steven C. Ley
- Immune Cell Signaling Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Mark S. Wilson
- Allergy and Anti-Helminth Immunity Laboratory, The Francis Crick Institute, London, United Kingdom
- * E-mail:
| |
Collapse
|
18
|
Kannan Y, Li Y, Coomes SM, Okoye IS, Pelly VS, Sriskantharajah S, Gückel E, Webb L, Czieso S, Nikolov N, MacDonald AS, Ley SC, Wilson MS. Tumor progression locus 2 reduces severe allergic airway inflammation by inhibiting Ccl24 production in dendritic cells. J Allergy Clin Immunol 2016; 139:655-666.e7. [PMID: 27484038 PMCID: PMC5292997 DOI: 10.1016/j.jaci.2016.05.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 05/01/2016] [Accepted: 05/23/2016] [Indexed: 12/16/2022]
Abstract
Background The molecular and cellular pathways driving the pathogenesis of severe asthma are poorly defined. Tumor progression locus 2 (TPL-2) (COT, MAP3K8) kinase activates the MEK1/2-extracellular-signal regulated kinase 1/2 MAP kinase signaling pathway following Toll-like receptor, TNFR1, and IL-1R stimulation. Objective TPL-2 has been widely described as a critical regulator of inflammation, and we sought to investigate the role of TPL-2 in house dust mite (HDM)-mediated allergic airway inflammation. Methods A comparative analysis of wild-type and Map3k8−/− mice was conducted. Mixed bone marrow chimeras, conditional knockout mice, and adoptive transfer models were also used. Differential cell counts were performed on the bronchoalveolar lavage fluid, followed by histological analysis of lung sections. Flow cytometry and quantitative PCR was used to measure type 2 cytokines. ELISA was used to assess the production of IgE, type 2 cytokines, and Ccl24. RNA sequencing was used to characterize dendritic cell (DC) transcripts. Results TPL-2 deficiency led to exacerbated HDM-induced airway allergy, with increased airway and tissue eosinophilia, lung inflammation, and IL-4, IL-5, IL-13, and IgE production. Increased airway allergic responses in Map3k8−/− mice were not due to a cell-intrinsic role for TPL-2 in T cells, B cells, or LysM+ cells but due to a regulatory role for TPL-2 in DCs. TPL-2 inhibited Ccl24 expression in lung DCs, and blockade of Ccl24 prevented the exaggerated airway eosinophilia and lung inflammation in mice given HDM-pulsed Map3k8−/− DCs. Conclusions TPL-2 regulates DC-derived Ccl24 production to prevent severe type 2 airway allergy in mice.
Collapse
Affiliation(s)
- Yashaswini Kannan
- Francis Crick Institute, Mill Hill Laboratory, the Ridgeway, London, United Kingdom
| | - Yanda Li
- Francis Crick Institute, Mill Hill Laboratory, the Ridgeway, London, United Kingdom
| | - Stephanie M Coomes
- Francis Crick Institute, Mill Hill Laboratory, the Ridgeway, London, United Kingdom
| | - Isobel S Okoye
- Francis Crick Institute, Mill Hill Laboratory, the Ridgeway, London, United Kingdom
| | - Victoria S Pelly
- Francis Crick Institute, Mill Hill Laboratory, the Ridgeway, London, United Kingdom
| | | | - Eva Gückel
- Francis Crick Institute, Mill Hill Laboratory, the Ridgeway, London, United Kingdom
| | - Lauren Webb
- Faculty of Life Sciences, Manchester Collaborative Centre for Inflammation Research, the University of Manchester, Manchester, United Kingdom
| | - Stephanie Czieso
- Francis Crick Institute, Mill Hill Laboratory, the Ridgeway, London, United Kingdom
| | - Nikolay Nikolov
- Francis Crick Institute, Mill Hill Laboratory, the Ridgeway, London, United Kingdom
| | - Andrew S MacDonald
- Faculty of Life Sciences, Manchester Collaborative Centre for Inflammation Research, the University of Manchester, Manchester, United Kingdom
| | - Steven C Ley
- Francis Crick Institute, Mill Hill Laboratory, the Ridgeway, London, United Kingdom.
| | - Mark S Wilson
- Francis Crick Institute, Mill Hill Laboratory, the Ridgeway, London, United Kingdom.
| |
Collapse
|
19
|
Luli S, Di Paolo D, Perri P, Brignole C, Hill SJ, Brown H, Leslie J, Marshall H, Wright MC, Mann DA, Ponzoni M, Oakley F. A new fluorescence-based optical imaging method to non-invasively monitor hepatic myofibroblasts in vivo. J Hepatol 2016; 65:75-83. [PMID: 27067455 PMCID: PMC4914605 DOI: 10.1016/j.jhep.2016.03.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 03/03/2016] [Accepted: 03/23/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Currently, staging of fibrosis in preclinical rodent liver fibrosis models is achieved histologically. Many animals are used at multiple time-points to assess disease progression or therapeutic responses. Hepatic myofibroblasts promote liver fibrosis therefore quantifying these cells in vivo could assess disease or predict therapeutic responses in mice. We fluorescently labelled a single chain antibody (C1-3) that binds hepatic myofibroblasts to monitor fibrogenesis in vivo. METHODS CCl4 was used to induce acute liver injury in WT and cRel(-/-) mice. Bile duct ligation was used to model chronic fibrosis. Hepatic myofibroblasts were depleted using a liposome-drug delivery system or chemically with sulfasalazine. An IVIS® spectrum visualised fluorophore-conjugated C1-3 in vivo. RESULTS IVIS detection of fluorescently labelled-C1-3 but not a control antibody discriminates between fibrotic and non-fibrotic liver in acute and chronic liver fibrosis models. cRel(-/-) mice have a fibro-protective phenotype and IVIS signal is reduced in CCl4 injured cRel(-/-) mice compared to wild-type. In vivo imaging of fluorescently labelled-C1-3 successfully predicts reductions in hepatic myofibroblast numbers in fibrotic liver disease in response to therapy. CONCLUSIONS We report a novel fluorescence imaging method to assess murine hepatic myofibroblast numbers in vivo during liver fibrosis and after therapy. We also describe a novel liposomal antibody targeting system to selectively deliver drugs to hepatic myofibroblasts in vivo. C1-3 binds human hepatic myofibroblast therefore imaging labelled-C1-3 could be used for clinical studies in man to help stage fibrosis, demonstrate efficacy of drugs that promote hepatic myofibroblast clearance or predict early therapeutic responses. LAY SUMMARY In response to damage and injury scars develop in the liver and the main cell that makes the scar tissue is the hepatic myofibroblast (HM). C1-3 is an antibody fragment that binds to the scar forming HM. We have fluorescently labelled C1-3 and given it to mice that have either normal or scarred livers (which contain HM) and then used a machine called an in vivo imaging system (IVIS) that takes pictures of different wavelengths of light, to visualise the antibody binding to HM inside the living mouse. Using fluorescently labelled C1-3 we can assess HM numbers in the injured liver and monitor response to therapy. We have also used C1-3 to target drugs encapsulated in lipid carriers (liposomes) to the HM to kill the HM and reduce the liver disease.
Collapse
Affiliation(s)
- Saimir Luli
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Daniela Di Paolo
- Experimental Therapy Unit, Laboratory of Oncology, Istituto Giannina Gaslini, Genoa, Italy
| | - Patrizia Perri
- Experimental Therapy Unit, Laboratory of Oncology, Istituto Giannina Gaslini, Genoa, Italy
| | - Chiara Brignole
- Experimental Therapy Unit, Laboratory of Oncology, Istituto Giannina Gaslini, Genoa, Italy
| | - Stephen J. Hill
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Helen Brown
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Jack Leslie
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - H.L. Marshall
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Matthew C. Wright
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Derek A. Mann
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Mirco Ponzoni
- Experimental Therapy Unit, Laboratory of Oncology, Istituto Giannina Gaslini, Genoa, Italy
| | - Fiona Oakley
- Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne NE2 4HH, UK.
| |
Collapse
|
20
|
Yan C, Shen LP, Ma R, Li B, Li XY, Hua H, Zhang B, Yu Q, Wang YG, Tang RX, Zheng KY. Characterization and identification of differentially expressed microRNAs during the process of the peribiliary fibrosis induced by Clonorchis sinensis. INFECTION GENETICS AND EVOLUTION 2016; 43:321-8. [PMID: 27267304 DOI: 10.1016/j.meegid.2016.06.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 05/26/2016] [Accepted: 06/03/2016] [Indexed: 12/16/2022]
Abstract
Clonorchis sinensis (C. sinensis) infection can lead to biliary fibrosis. MicroRNAs (miRNAs) play important roles in regulation of genes expression in the liver diseases. However, the differential expression of miRNAs that probably regulates the portal fibrogenesis caused by C. sinensis has not yet been investigated. Hepatic miRNAs expression profiles from C. sinensis-infected mice at different time-points were analyzed by miRNA microarray and validated by quantitative real-time PCR (qRT-PCR). 349 miRNAs were differentially expressed in the liver of the C. sinensis-infected mice at 2, 8 or 16weeks post infection (p.i.), compared with those at 0week p.i., and there were 143 down-regulated and 206 up-regulated miRNAs among them. These all dysregulated miRNAs were potentially involved in the pathological processes of clonorchiasis by regulation of cancer-related signaling pathway, TGF-β signaling pathway, MAPK signaling pathway, Toll-like receptor signaling pathway, PI3K /AKT signaling pathway, etc. 169 of these dysregulated miRNAs were predicted to be involved in the TGF/Smads signaling pathway which plays an important role in the biliary fibrosis caused by C. sinensis. Additionally, miRNA-32, miRNA-34a, miRNA-125b and miRNA-497 were negatively correlated with Smad7 expression, indicating these miRNAs may specifically down-regulate Smad7 expression and participate in regulation of biliary fibrosis caused by C. sinensis. The results of the present study for the first time demonstrated that miRNAs were differentially expressed in the liver of mice infected by C. sinensis, and these miRNAs may play important roles in regulation of peribiliary fibrosis caused by C. sinensis, which may provide possible therapeutic targets for clonorchiasis.
Collapse
Affiliation(s)
- Chao Yan
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, People's Republic of China
| | - Li-Ping Shen
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, People's Republic of China
| | - Rui Ma
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, People's Republic of China
| | - Bo Li
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, People's Republic of China
| | - Xiang-Yang Li
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, People's Republic of China
| | - Hui Hua
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, People's Republic of China
| | - Bo Zhang
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, People's Republic of China
| | - Qian Yu
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, People's Republic of China
| | - Yu-Gang Wang
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, People's Republic of China
| | - Ren-Xian Tang
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, People's Republic of China.
| | - Kui-Yang Zheng
- Department of Pathogenic Biology and Immunology, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, People's Republic of China.
| |
Collapse
|
21
|
Shaker ME, Trawick BN, Mehal WZ. The novel TLR9 antagonist COV08-0064 protects from ischemia/reperfusion injury in non-steatotic and steatotic mice livers. Biochem Pharmacol 2016; 112:90-101. [PMID: 27157410 DOI: 10.1016/j.bcp.2016.05.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/04/2016] [Indexed: 12/27/2022]
Abstract
Ischemia/reperfusion (I/R) injury constitutes a major reason for failure of liver surgeries and transplantation. I/R injury is more severe in steatotic livers and limits their use in transplantation. Here, we present a novel and selective Toll-like receptor 9 (TLR9) antagonist COV08-0064 and test its potential to protect from I/R-induced injury in normal and steatotic livers. The in vivo effects of COV08-0064 pretreatment were investigated on normal chow diet (NCD) and high fat diet (HFD)-fed mice subjected to segmental (70%) warm hepatic I/R. Also, the in vitro effects of COV08-0064 were elucidated in murine macrophages and dendritic cells. Mice on a HFD had pronouncedly greater hepatic I/R injury than mice on a NCD. COV08-0064-pretreatment to both NCD and HFD-fed mice reduced hepatic I/R injury. COV08-0064-pretreatment was associated with less production of the liver inflammatory cytokines and mediators TNF-α, IL-1β, IL-6, NLRP3, iNOS and MCP-1. These manifestations were preceded with inhibition of JNK and ERK phosphorylation and TLR9 cleavage in the liver. COV08-0064 enhanced the hepatic expression of the endogenous anti-inflammatory cytokines IL-10 and IL-1Ra at the early phase I/R injury. In vitro, COV08-0064 selectively blocked mRNA upregulation of TNF-α, IL-1β, NLRP3 and MCP-1 in macrophages and IFN-β mRNA in dendritic cells induced by the TLR9 agonist CpG-ODN. These effects were concordant with inhibition of JNK, ERK, IκBα and IKKα/β phosphorylation. In conclusion, TLR9 signaling inhibition by COV08-0064 may be an effective approach in liver surgeries including transplantation to limit I/R-injury and overcome the shortages in the donor pool by incorporating steatotic livers.
Collapse
Affiliation(s)
- Mohamed E Shaker
- Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, CT 06520, USA; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Bobby N Trawick
- Center for Organic Chemistry, Mallinckrodt Pharmaceuticals, St. Louis, MO 63147, USA
| | - Wajahat Z Mehal
- Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, CT 06520, USA; Section of Digestive Diseases, Department of Veterans Affairs Connecticut Healthcare, West Haven, CT 06516, USA.
| |
Collapse
|
22
|
Vyrla D, Nikolaidis G, Oakley F, Perugorria MJ, Tsichlis PN, Mann DA, Eliopoulos AG. TPL2 Kinase Is a Crucial Signaling Factor and Mediator of NKT Effector Cytokine Expression in Immune-Mediated Liver Injury. THE JOURNAL OF IMMUNOLOGY 2016; 196:4298-310. [DOI: 10.4049/jimmunol.1501609] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 02/26/2016] [Indexed: 12/26/2022]
|
23
|
Stice CP, Hussain S, Liu C, Ausman LM, Wang XD, Greenberg AS. Deletion of tumor progression locus 2 attenuates alcohol-induced hepatic inflammation. Hepatobiliary Surg Nutr 2016; 5:29-37. [PMID: 26904554 DOI: 10.3978/j.issn.2304-3881.2015.12.11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND The pathogenesis of alcoholic liver disease (ALD) involves the interaction of several inflammatory signaling pathways. Tumor progression locus 2 (TPL2), also known as Cancer Osaka Thyroid (COT) and MAP3K8, is a serine-threonine kinase that functions as a critical regulator of inflammatory pathways by up-regulating production of inflammatory cytokines. The present study aims to fill the gap in knowledge regarding the involvement of TPL2 in the mechanism of alcohol-induced hepatic inflammation. METHODS Male TPL2(-/-) knockout (TPL2KO) mice and TPL2(+/+) wild-type (WT) mice were group pair-fed with Lieber-DeCarli liquid ethanol diet (EtOH diet, 27% energy from EtOH) or control diet (ctrl diet) for 4 weeks. Both histological and molecular biomarkers involved in the induction of hepatic inflammation by alcohol consumption were examined. RESULTS Consumption of the EtOH diet in WT mice lead to a significant induction of TPL2 mRNA expression as compared with WT mice fed ctrl diet. A significant induction in inflammatory foci and steatosis was also observed in WT mice fed EtOH diet. The deletion of TPL2 significantly reduced inflammatory foci in the liver of mice consuming both ctrl and EtOH diets as compared to their respective WT controls. This reduction was associated with suppression of hepatic inflammatory gene expression of interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and interleukin-1β (IL-1β) and macrophage marker F4/80. In addition, histological analysis of livers revealed that TPL2 deletion resulted in reduced steatosis in both ctrl (significant) and EtOH (non-significant) diet-fed mice as compared to their respective WT controls. CONCLUSIONS The demonstration that TPL2 deletion attenuates alcohol-induced hepatic inflammation provides evidence of a novel role for TPL2 in the pathogenesis of ALD.
Collapse
Affiliation(s)
- Camilla P Stice
- 1 Nutrition and Cancer Biology Laboratory, 2 Obesity and Metabolism Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Sajid Hussain
- 1 Nutrition and Cancer Biology Laboratory, 2 Obesity and Metabolism Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Chun Liu
- 1 Nutrition and Cancer Biology Laboratory, 2 Obesity and Metabolism Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Lynne M Ausman
- 1 Nutrition and Cancer Biology Laboratory, 2 Obesity and Metabolism Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Xiang-Dong Wang
- 1 Nutrition and Cancer Biology Laboratory, 2 Obesity and Metabolism Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| | - Andrew S Greenberg
- 1 Nutrition and Cancer Biology Laboratory, 2 Obesity and Metabolism Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
| |
Collapse
|
24
|
Wang M, Zheng W, Zhu X, Xu J, Cai B, Zhang Y, Zheng F, Zhou L, Yang Z, Zhang X, Wang C, Nie S, Zhu J. A Human Anti-Toll Like Receptor 4 Fab Fragment Inhibits Lipopolysaccharide-Induced Pro-Inflammatory Cytokines Production in Macrophages. PLoS One 2016; 11:e0146856. [PMID: 26785354 PMCID: PMC4718644 DOI: 10.1371/journal.pone.0146856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 12/21/2015] [Indexed: 01/01/2023] Open
Abstract
The results of clinical and experimental studies suggest that endotoxin/toll-like receptor 4 (TLR4)-mediated proinflammatory and profibrotic signaling activation is critical in the development of hepatic fibrosis. However, studies examining the role of specific TLR4 inhibitor are still lacking. The present study was aimed to prepare a human anti-TLR4 Fab fragment, named hTLR4-Fab01, and to explore its immune activity. We screened the positive clone of anti-human TLR4 phagemid from a human phage-display antibody library using recombinant TLR4 protein, which was used as template cDNA for the amplification of variable regions of the heavy (VH) chain and light chain (VL), then coupled with highly conserved regions of the heavy chain domain 1 (CH1) and the light chain (CL), respectively. Thus, the prokaryotic expression vector pETDuet-1 of hTLR4-Fab01 was constructed and transformed into Escherichia coli (E. coli) BL21. The characteristic of hTLR4-Fab01 was examined by SDS-PAGE, Western blotting, ELISA, affinity and kinetics assay. Further, our data demonstrate that hTLR4-Fab01 could specifically bind to TLR4, and its treatment obviously attenuated the proinflammatory effect, characterized by less LPS-induced TNF-α, IL-1, IL-6 and IL-8 production in human macrophages. In conclusion, we have successfully prepared the hTLR4-Fab01 with efficient activity for blocking LPS-induced proinflammatory cytokines production, suggesting that the hTLR4-Fab01 may be a potential candidate for the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Maorong Wang
- Institute of Liver Disease, Nanjing Jingdu Hospital, Nanjing 210002, China
- Anhui Medical University Affiliated with Bayi Clinical College, Nanjing 210002, China
- * E-mail: (MW); (JZ)
| | - Wenkai Zheng
- Institute of Liver Disease, Nanjing Jingdu Hospital, Nanjing 210002, China
| | - Xuhui Zhu
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Jing Xu
- Institute of Liver Disease, Nanjing Jingdu Hospital, Nanjing 210002, China
| | - Binggang Cai
- Anhui Medical University Affiliated with Bayi Clinical College, Nanjing 210002, China
| | - Yiqing Zhang
- Institute of Liver Disease, Nanjing Jingdu Hospital, Nanjing 210002, China
| | - Feng Zheng
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China
| | - Linfu Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhiguo Yang
- Institute of Liver Disease, Nanjing Jingdu Hospital, Nanjing 210002, China
| | - Xin Zhang
- Institute of Liver Disease, Nanjing Jingdu Hospital, Nanjing 210002, China
| | - Changjun Wang
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China
| | - Shinan Nie
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, China
| | - Jin Zhu
- Huadong Medical Institute of Biotechniques, Nanjing 210002, China
- Department of Pathology, Key Laboratory of Antibody Technique of the Ministry of Health, NJMU, Nanjing 210029, China
- * E-mail: (MW); (JZ)
| |
Collapse
|
25
|
Cartwright T, Perkins ND, L Wilson C. NFKB1: a suppressor of inflammation, ageing and cancer. FEBS J 2016; 283:1812-22. [PMID: 26663363 DOI: 10.1111/febs.13627] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/23/2015] [Accepted: 12/08/2015] [Indexed: 12/18/2022]
Abstract
The pleiotropic consequences of nuclear factor of kappa light polypeptide gene enhancer in B-cells (NF-κB) pathway activation result from the combinatorial effects of the five subunits that form the homo- and heterodimeric NF-κB complexes. Although biochemical and gene knockout studies have demonstrated overlapping and distinct functions for these proteins, much is still not known about the mechanisms determining context-dependent functions, the formation of different dimer complexes and transcriptional control in response to diverse stimuli. Here we discuss recent results that reveal that the nuclear factor of kappa light polypeptide gene enhancer in B-cells 1 (NFKB1) (p105/p50) subunit is an important regulator of NF-κB activity in vivo. These effects are not restricted to being a dimer partner for other NF-κB subunits. Rather p50 homodimers have a critical role as suppressors of the NF-κB response, while the p105 precursor has a variety of NF-κB-independent functions. The importance of Nfkb1 function can be seen in mouse models, where Nfkb1(-/-) mice display increased inflammation and susceptibility to certain forms of DNA damage, leading to cancer, and a rapid ageing phenotype. In humans, low expression of Kip1 ubiquitination-promoting complex 1 (KPC1), a ubiquitin ligase required for p105 to p50 processing, was shown to correlate with a reduction in p50 and glioblastoma incidence. Therefore, while the majority of research in this field has focused on the upstream signalling pathways leading to NF-κB activation or the function of other NF-κB subunits, such as RelA (p65), these data demonstrate a critical role for NFKB1, potentially revealing new strategies for targeting this pathway in inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Tyrell Cartwright
- Fibrosis Laboratory, Institute of Cellular Medicine, Newcastle University, UK
| | - Neil D Perkins
- Institute for Cell and Molecular Biosciences, Faculty of Medical Sciences, Newcastle University, UK
| | - Caroline L Wilson
- Fibrosis Laboratory, Institute of Cellular Medicine, Newcastle University, UK
| |
Collapse
|
26
|
Piao RL, Xiu M, Brigstock DR, Gao RP. An immortalized rat pancreatic stellate cell line RP-2 as a new cell model for evaluating pancreatic fibrosis, inflammation and immunity. Hepatobiliary Pancreat Dis Int 2015; 14:651-9. [PMID: 26663014 DOI: 10.1016/s1499-3872(15)60415-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND Pancreatic stellate cells (PSCs) play a critical role in the pathogenesis of pancreatic fibrosis and have emerging functions as progenitor cells, immune cells or intermediaries in pancreatic exocrine secretion. Increasing evidence has shown that desmin as an exclusive cytoskeleton marker of PSC is only expressed in part of these cells. This study was to establish a desmin-positive PSC cell line and evaluate its actions on pancreatic fibrosis, inflammation and immunity. METHODS The presence of cytoskeletal proteins, integrin α5β1 or TLR4, was determined by immunocytochemistry while the production of desmin, collagen I, MMP-1, MMP-2, TIMP-2, or CD14 was evaluated by Western blotting. The levels of desmin, collagen I, IL-1 and IL-6 mRNA were determined by real-time quantitative PCR. The secretion of cytokines was detected by ELISA. Cell function was assessed using adhesion, migration, or proliferation assays. RESULTS A stable activated rat PSC cell line (designated as RP-2) was established by RSV promoter/enhancer-driven SV40 large T antigen expression. RP-2 cells retained typical PSC properties, exhibited a myofibroblast-like phenotype and persistently produced desmin. The cells produced collagen I protein, matrix metalloproteinases and inhibitors thereof. RP-2 cells demonstrated typical PSC functions, including proliferation, adherence, and migration, the latter two of which occurred in response to fibronectin and were mediated by integrin α5β1. TLR4 and its response genes including proinflammatory cytokines (IL-1, IL-6, TNF-alpha) and chemotactic cytokines (MCP-1, MIP-1α, Rantes) were produced by RP-2 cells and activated by LPS. LPS-induced IL-1 or IL-6 mRNA expression in this cell line was fully blocked with MyD88 inhibitor. CONCLUSION RP-2 cells provide a novel tool for analyzing the properties and functions of PSCs in the pathogenesis of fibrosis, inflammation and immunity in the pancreas.
Collapse
Affiliation(s)
- Rong-Li Piao
- Department of Hepatic-biliary-pancreatic Medicine, the First Hospital of Jilin University, Changchun 130021, China.
| | | | | | | |
Collapse
|
27
|
Kawada N. Cytoglobin as a Marker of Hepatic Stellate Cell-derived Myofibroblasts. Front Physiol 2015; 6:329. [PMID: 26617531 PMCID: PMC4643130 DOI: 10.3389/fphys.2015.00329] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/30/2015] [Indexed: 12/13/2022] Open
Abstract
Myofibroblasts play important roles in inflammation, fibrosis and tumorigenesis in chronically inflamed liver. Liver myofibroblasts originate from hepatic stellate cells, portal fibroblasts or mesothelial cells, and they are localized in and around fibrotic septum and portal tracts. Liver myofibroblasts are the source of extracellular matrix materials, including type I collagen and multiple fibrogenic growth factors, such as transforming growth factor-β and vascular endothelial growth factor. Although a detailed characterization of the function of individual myofibroblasts has not been conducted, owing to the lack of appropriate cell markers, recent lineage-tracing technology has revealed the limited contribution of myofibroblasts that are derived from portal fibroblasts to various types of liver fibrosis, as compared with the contribution of hepatic stellate cells. In addition, cytoglobin, which is the fourth globin in mammals and function as a local gas sensor, provides a new perspective on the involvement of stellate cells in fibrosis and carcinogenesis, possibly through its anti-oxidative properties and is a promising new marker that discriminates between myofibroblasts derived from stellate cells and those from portal fibroblasts.
Collapse
Affiliation(s)
- Norifumi Kawada
- Department of Hepatology, Graduate School of Medicine, Osaka City University Osaka, Japan
| |
Collapse
|
28
|
Kawada N, Parola M. Interactions of Stellate Cells with Other Non-Parenchymal Cells. STELLATE CELLS IN HEALTH AND DISEASE 2015:185-207. [DOI: 10.1016/b978-0-12-800134-9.00012-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
29
|
Yang JJ, Tao H, Hu W, Liu LP, Shi KH, Deng ZY, Li J. MicroRNA-200a controls Nrf2 activation by target Keap1 in hepatic stellate cell proliferation and fibrosis. Cell Signal 2014; 26:2381-9. [PMID: 25049078 DOI: 10.1016/j.cellsig.2014.07.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 07/09/2014] [Indexed: 01/19/2023]
Abstract
Hepatic fibrosis is a common final pathological process in the progression of liver disease, which is primarily due to oxidative stress. Nrf2 is known to coordinate induction of genes that encode antioxidant enzymes. Moreover, Nrf2 expression is largely regulated through the association of Nrf2 with Keap1, which results in cytoplasmic Nrf2 degradation. Conversely, little is known concerning the regulation of Keap1 expression. Although the function of miRNA-200a controls Keap1 gene expression has been discussed in many cancers and fibrotic diseases, its role in hepatic fibrosis is still poorly understood. By using miRNA mimic, we observed miRNA-200a silencing in activated hepatic stellate cell and demonstrated that upon re-expression, miRNA-200a targets the Keap1, and leading to Keap1 mRNA degradation. We find that treatment with miRNA-200a mimics, restored miRNA-200a expression and reduced Keap1 levels. This reduction in Keap1 levels corresponded with Nrf2 nuclear translocation and activation of Nrf2-dependent NQO1 gene transcription. Moreover, we found that Nrf2 activation inhibited the TGF-β1-independent growth of hepatic stellate cell. Finally, our study demonstrates that miRNA-200a regulates the Keap1/Nrf2 pathway in hepatic stellate cell and fibrosis, and we find that epigenetic therapy can restore miRNA-200a regulation of Keap1 expression, therefore reactivating the Nrf2-dependent antioxidant pathway in liver fibrosis.
Collapse
Affiliation(s)
- Jing-Jing Yang
- Department of Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Hui Tao
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Wei Hu
- Department of Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Li-Ping Liu
- Department of Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Kai-Hu Shi
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Zi-Yu Deng
- Department of Scientific and Educational, The Second Hospital of Anhui Medical University, China
| | - Jun Li
- School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
30
|
Tsubota A, Mogushi K, Aizaki H, Miyaguchi K, Nagatsuma K, Matsudaira H, Kushida T, Furihata T, Tanaka H, Matsuura T. Involvement of MAP3K8 and miR-17-5p in poor virologic response to interferon-based combination therapy for chronic hepatitis C. PLoS One 2014; 9:e97078. [PMID: 24819603 PMCID: PMC4018277 DOI: 10.1371/journal.pone.0097078] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 04/14/2014] [Indexed: 12/14/2022] Open
Abstract
Despite advances in chronic hepatitis C treatment, a proportion of patients respond poorly to treatment. This study aimed to explore hepatic mRNA and microRNA signatures involved in hepatitis C treatment resistance. Global hepatic mRNA and microRNA expression profiles were compared using microarray data between treatment responses. Quantitative real-time polymerase chain reaction validated the gene signatures from 130 patients who were infected with hepatitis C virus genotype 1b and treated with pegylated interferon-alpha and ribavirin combination therapy. The correlation between mRNA and microRNA was evaluated using in silico analysis and in vitro siRNA and microRNA inhibition/overexpression experiments. Multivariate regression analysis identified that the independent variables IL28B SNP rs8099917, hsa-miR-122-5p, hsa-miR-17-5p, and MAP3K8 were significantly associated with a poor virologic response. MAP3K8 and miR-17-5p expression were inversely correlated with treatment response. Furthermore, miR-17-5p repressed HCV production by targeting MAP3K8. Collectively, the data suggest that several molecules and the inverse correlation between mRNA and microRNA contributed to a host genetic refractory hepatitis C treatment response.
Collapse
Affiliation(s)
- Akihito Tsubota
- Institute of Clinical Medicine and Research (ICMR), Jikei University School of Medicine, Kashiwa, Chiba, Japan
- Division of Gastroenterology and Hepatology, Kashiwa Hospital, The Jikei University School of Medicine, Kashiwa, Chiba, Japan
- * E-mail:
| | - Kaoru Mogushi
- Department of Bioinformatics, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Hideki Aizaki
- Department of Virology II, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Ken Miyaguchi
- Department of Bioinformatics, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Keisuke Nagatsuma
- Institute of Clinical Medicine and Research (ICMR), Jikei University School of Medicine, Kashiwa, Chiba, Japan
- Division of Gastroenterology and Hepatology, Kashiwa Hospital, The Jikei University School of Medicine, Kashiwa, Chiba, Japan
| | - Hiroshi Matsudaira
- Institute of Clinical Medicine and Research (ICMR), Jikei University School of Medicine, Kashiwa, Chiba, Japan
- Division of Gastroenterology and Hepatology, Kashiwa Hospital, The Jikei University School of Medicine, Kashiwa, Chiba, Japan
| | - Tatsuya Kushida
- National Bioscience Database Center, Japan Science and Technology Agency, Chiyoda-ku, Tokyo, Japan
| | - Tomomi Furihata
- Laboratory of Pharmacology and Toxicology, Graduate School of Pharmaceutical Science, Chiba University, Chiba, Japan
| | - Hiroshi Tanaka
- Department of Bioinformatics, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Tomokazu Matsuura
- Department of Laboratory Medicine, Jikei University School of Medicine, Minato-ku, Tokyo, Japan
| |
Collapse
|
31
|
Ceppo F, Berthou F, Jager J, Dumas K, Cormont M, Tanti JF. Implication of the Tpl2 kinase in inflammatory changes and insulin resistance induced by the interaction between adipocytes and macrophages. Endocrinology 2014; 155:951-64. [PMID: 24424060 DOI: 10.1210/en.2013-1815] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Adipose tissue inflammation is associated with the development of insulin resistance. In obese adipose tissue, lipopolysaccharides (LPSs) and saturated fatty acids trigger inflammatory factors that mediate a paracrine loop between adipocytes and macrophages. However, the inflammatory signaling proteins underlying this cross talk remain to be identified. The mitogen-activated protein kinase kinase kinase tumor progression locus 2 (Tpl2) is activated by inflammatory stimuli, including LPS, and its expression is up-regulated in obese adipose tissue, but its role in the interaction between adipocytes and macrophages remains ill-defined. To assess the implication of Tpl2 in the cross talk between these 2 cell types, we used coculture system and conditioned medium (CM) from macrophages. Pharmacological inhibition of Tpl2 in the coculture markedly reduced lipolysis and cytokine production and prevented the decrease in adipocyte insulin signaling. Tpl2 knockdown in cocultured adipocytes reduced lipolysis but had a weak effect on cytokine production and did not prevent the alteration of insulin signaling. By contrast, Tpl2 silencing in cocultured macrophages resulted in a marked inhibition of cytokine production and prevented the alteration of adipocyte insulin signaling. Further, when Tpl2 was inhibited in LPS-activated macrophages, the produced CM did not alter adipocyte insulin signaling and did not induce an inflammatory response in adipocytes. By contrast, Tpl2 silencing in adipocytes did not prevent the deleterious effects of a CM from LPS-activated macrophages. Together, these data establish that Tpl2, mainly in macrophages, is involved in the cross talk between adipocytes and macrophages that promotes inflammatory changes and alteration of insulin signaling in adipocytes.
Collapse
Affiliation(s)
- Franck Ceppo
- INSERM Unité 1065/Centre Méditerranéen de Médecine Moléculaire (C3M) and Université de Nice Sophia Antipolis (F.C., F.B., J.J., K.D, M.C., J.-F.T.), F-06204, Nice, France
| | | | | | | | | | | |
Collapse
|
32
|
Martel G, Rousseau S. TPL2 signalling: from Toll-like receptors-mediated ERK1/ERK2 activation to Cystic Fibrosis lung disease. Int J Biochem Cell Biol 2014; 52:146-51. [PMID: 24530836 DOI: 10.1016/j.biocel.2014.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 01/30/2014] [Accepted: 02/02/2014] [Indexed: 01/20/2023]
Abstract
Cystic Fibrosis (CF) is the most common lethal genetic recessive disorder, with a carrier frequency of 1 in 27 among North American Caucasians. Mitogen-activated protein kinases (MAPKs) and pro-inflammatory cytokines have crucial functions in the innate immune response of epithelial cells. They determine the inflammation status and the host response to pathogenic infections. However, in CF, bacterial-driven inflammation leads to tissue destruction, reduction in lung function and mortality. Recognition of invading pathogens is mediated in part by Toll-like receptors (TLR) activation of intracellular signalling cascade leading to cytokines' synthesis. The protein kinase Tumour Progression Locus 2 (TPL2) is a key molecule in relaying inflammatory stimuli to ERK1/ERK2 MAPKs. In this review, we summarized the recent findings on TPL2 signalling and how TPL2 can contribute to the excessive inflammation found in CF. Pharmacologically targeting this kinase could have a significant benefit for CF patients dealing with chronic bacterial infections such as Pseudomonas aeruginosa. This article is part of a Directed Issue entitled: Cystic Fibrosis: From o-mics to cell biology, physiology, and therapeutic advances.
Collapse
Affiliation(s)
- Guy Martel
- Meakins-Christie Laboratories, McGill University Health Centre Research Institute, Montreal, Canada
| | - Simon Rousseau
- Meakins-Christie Laboratories, McGill University Health Centre Research Institute, Montreal, Canada.
| |
Collapse
|
33
|
Feng M, Wang Q, Wang H, Wang M, Guan W, Lu L. Adoptive transfer of hepatic stellate cells ameliorates liver ischemia reperfusion injury through enriching regulatory T cells. Int Immunopharmacol 2014; 19:267-74. [PMID: 24444778 DOI: 10.1016/j.intimp.2014.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 12/08/2013] [Accepted: 01/07/2014] [Indexed: 01/23/2023]
Abstract
Our previous study indicated that adoptive transferred regulatory T cells (Tregs) attenuated liver ischemia reperfusion injury (IRI). Recent studies demonstrated that hepatic stellate cells (HSCs) were producers of induced Tregs (iTregs) via retinoic acid. This study aimed to investigate the role of adoptive transferred HSCs in liver IRI. Mice were treated with gradient doses of HSCs before surgery at 24h or 72h. The levels of serum aminotransferases and hepatic cytokines were evaluated after reperfusion. Meanwhile, hepatic Tregs and their subsets were analyzed by flow cytometry. We found that adoptive transferred HSCs attenuated liver IRI. Administration of HSCs expanded the number of hepatic iTregs and natural Tregs (nTregs) after reperfusion. In addition, we found that the increased Tregs were almost Helios-Tregs before surgery. These Helios-Tregs were considered as iTregs and protected liver from IRI partially. Furthermore, adoptive transferred HSCs stabilized nTregs and prevented nTregs from reducing after reperfusion. These nTregs also attenuated liver IRI partially. Depletion of Tregs abolished the protective effect of HSCs. Thus, we conclude that adoptive transferred HSCs ameliorate liver IRI in Tregs-dependent manner.
Collapse
Affiliation(s)
- Min Feng
- Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Quanrongzi Wang
- Radiology Department, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China; Key Laboratory of Living Donor Liver Transplantation of Ministry of Public Health, Nanjing 210029, China
| | - Hao Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Meng Wang
- Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Wenxian Guan
- Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | - Ling Lu
- Key Laboratory of Living Donor Liver Transplantation of Ministry of Public Health, Nanjing 210029, China
| |
Collapse
|
34
|
The transcriptomic response of rat hepatic stellate cells to endotoxin: implications for hepatic inflammation and immune regulation. PLoS One 2013; 8:e82159. [PMID: 24349206 PMCID: PMC3857241 DOI: 10.1371/journal.pone.0082159] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 10/26/2013] [Indexed: 12/13/2022] Open
Abstract
With their location in the perisinusoidal space of Disse, hepatic stellate cells (HSCs) communicate with all of the liver cell types both by physical association (cell body as well as cytosolic processes penetrating into sinusoids through the endothelial fenestrations) and by producing several cytokines and chemokines. Bacterial lipopolysaccharide (LPS), circulating levels of which are elevated in liver diseases and transplantation, stimulates HSCs to produce increased amounts of cytokines and chemokines. Although recent research provides strong evidence for the role of HSCs in hepatic inflammation and immune regulation, the number of HSC-elaborated inflammatory and immune regulatory molecules may be much greater then known at the present time. Here we report time-dependent changes in the gene expression profile of inflammatory and immune-regulatory molecules in LPS-stimulated rat HSCs, and their validation by biochemical analyses. LPS strongly up-regulated LPS-response elements (TLR2 and TLR7) but did not affect TLR4 and down-regulated TLR9. LPS also up-regulated genes in the MAPK, NFκB, STAT, SOCS, IRAK and interferon signaling pathways, numerous CC and CXC chemokines and IL17F. Interestingly, LPS modulated genes related to TGFβ and HSC activation in a manner that would limit their activation and fibrogenic activity. The data indicate that LPS-stimulated HSCs become a major cell type in regulating hepatic inflammatory and immunological responses by altering expression of numerous relevant genes, and thus play a prominent role in hepatic pathophysiology including liver diseases and transplantation.
Collapse
|
35
|
Hirschhorn J, Mohanty S, Bhat NR. The role of tumor progression locus 2 protein kinase in glial inflammatory response. J Neurochem 2013; 128:919-26. [PMID: 24188160 DOI: 10.1111/jnc.12522] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 10/29/2013] [Accepted: 10/30/2013] [Indexed: 01/10/2023]
Abstract
Tumor progression locus 2 (Tpl2)/cancer Osaka thyroid kinase is a newer member of MAP3K family that is now known for its essential role in tumor necrosis factor-aplha (TNFα) expression in macrophages, but its pro-inflammatory signaling, if any, in glia is unknown. When cultures of murine microglia and astrocytes were exposed to lipopolysaccharide, there was a rapid activation (i.e., phosphorylation) of Tpl2 in parallel to the activation of down-stream effector MAPKs, that is, extracellular signal regulated kinase (ERK), p38 MAPK and C-Jun N-terminal kinase (JNK). Pre-incubation of the cultures with a Tpl2 inhibitor selectively suppressed the activation of the primary down-stream target, that is, ERK relative to p38 MAPK and JNK. That Tpl2 activation was functionally involved in glial inflammatory response was indicated by a reduced release of the cytokines, i.e. TNFα and the expression of inducible nitric oxide synthase in the presence of the kinase inhibitor. Furthermore, over-expression of a wild-type Tpl2 construct in C-6 glia resulted in an enhanced transcriptional activation of inducible nitric oxide synthase, while transfection with a dominant negative form of Tpl-2 had the opposite effect. The findings assign an important pro-inflammatory signaling function for Tpl2 pathway in glial cells.
Collapse
Affiliation(s)
- Joshua Hirschhorn
- Department of Neurosciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | |
Collapse
|
36
|
Sanz-Garcia C, Ferrer-Mayorga G, González-Rodríguez Á, Valverde AM, Martín-Duce A, Velasco-Martín JP, Regadera J, Fernández M, Alemany S. Sterile inflammation in acetaminophen-induced liver injury is mediated by Cot/tpl2. J Biol Chem 2013; 288:15342-51. [PMID: 23572518 DOI: 10.1074/jbc.m112.439547] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Cot/tpl2 (MAP3K8) activates MKK1/2-Erk1/2 following stimulation of the Toll-like/IL-1 receptor superfamily. Here, we investigated the role of Cot/tpl2 in sterile inflammation and drug-induced liver toxicity. Cot/tpl2 KO mice exhibited reduced hepatic injury after acetaminophen challenge, as evidenced by decreased serum levels of both alanine and aspartate aminotransferases, decreased hepatic necrosis, and increased survival relative to Wt mice. Serum levels of both alanine and aspartate aminotransferases were also lower after intraperitoneal injection of acetaminophen in mice expressing an inactive form of Cot/tpl2 compared with Wt mice, suggesting that Cot/tpl2 activity contributes to acetaminophen-induced liver injury. Furthermore, Cot/tpl2 deficiency reduced neutrophil and macrophage infiltration in the liver of mice treated with acetaminophen, as well as their hepatic and systemic levels of IL-1α. Intraperitoneal injection of damage-associated molecular patterns from necrotic hepatocytes also impaired the recruitment of leukocytes and decreased the levels of several cytokines in the peritoneal cavity in Cot/tpl2 KO mice compared with Wt counterparts. Moreover, similar activation profiles of intracellular pathways were observed in Wt macrophages stimulated with Wt or Cot/tpl2 KO damage-associated molecular patterns. However, upon stimulation with damage-associated molecular patterns, the activation of Erk1/2 and JNK was deficient in Cot/tpl2 KO macrophages compared with their Wt counterparts; an effect accompanied by weaker release of several cytokines, including IL-1α, an important component in the development of sterile inflammation. Taken together, these findings indicate that Cot/tpl2 contributes to acetaminophen-induced liver injury, providing some insight into the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Carlos Sanz-Garcia
- Instituto Investigaciones Biomédicas Alberto Sols, CISC-UAM, 28029 Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Fullard N, Moles A, O'Reilly S, van Laar JM, Faini D, Diboll J, Reynolds NJ, Mann DA, Reichelt J, Oakley F. The c-Rel subunit of NF-κB regulates epidermal homeostasis and promotes skin fibrosis in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:2109-20. [PMID: 23562440 DOI: 10.1016/j.ajpath.2013.02.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 02/11/2013] [Accepted: 02/12/2013] [Indexed: 10/27/2022]
Abstract
The five subunits of transcription factor NF-κB have distinct biological functions. NF-κB signaling is important for skin homeostasis and aging, but the contribution of individual subunits to normal skin biology and disease is unclear. Immunohistochemical analysis of the p50 and c-Rel subunits within lesional psoriatic and systemic sclerosis skin revealed abnormal epidermal expression patterns, compared with healthy skin, but RelA distribution was unaltered. The skin of Nfkb1(-/-) and c-Rel(-/-) mice is structurally normal, but epidermal thickness and proliferation are significantly reduced, compared with wild-type mice. We show that the primary defect in both Nfkb1(-/-) and c-Rel(-/-) mice is within keratinocytes that display reduced proliferation both in vitro and in vivo. However, both genotypes can respond to proliferative stress, with 12-O-tetradecanoylphorbol-13-acetate-induced epidermal hyperproliferation and closure rates of full-thickness skin wounds being equivalent to those of wild-type controls. In a model of bleomycin-induced skin fibrosis, Nfkb1(-/-) and c-Rel(-/-) mice displayed opposite phenotypes, with c-Rel(-/-) mice being protected and Nfkb1(-/-) developing more fibrosis than wild-type mice. Taken together, our data reveal a role for p50 and c-Rel in regulating epidermal proliferation and homeostasis and a profibrogenic role for c-Rel in the skin, and identify a link between epidermal c-Rel expression and systemic sclerosis. Modulating the actions of these subunits could be beneficial for treating hyperproliferative or fibrogenic diseases of the skin.
Collapse
Affiliation(s)
- Nicola Fullard
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|